1
|
D'Egidio F, Castelli V, d'Angelo M, Ammannito F, Quintiliani M, Cimini A. Brain incoming call from glia during neuroinflammation: Roles of extracellular vesicles. Neurobiol Dis 2024; 201:106663. [PMID: 39251030 DOI: 10.1016/j.nbd.2024.106663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/11/2024] Open
Abstract
The functionality of the central nervous system (CNS) relies on the connection, integration, and the exchange of information among neural cells. The crosstalk among glial cells and neurons is pivotal for a series of neural functions, such as development of the nervous system, electric conduction, synaptic transmission, neural circuit establishment, and brain homeostasis. Glial cells are crucial players in the maintenance of brain functionality in physiological and disease conditions. Neuroinflammation is a common pathological process in various brain disorders, such as neurodegenerative diseases, and infections. Glial cells, including astrocytes, microglia, and oligodendrocytes, are the main mediators of neuroinflammation, as they can sense and respond to brain insults by releasing pro-inflammatory or anti-inflammatory factors. Recent evidence indicates that extracellular vesicles (EVs) are pivotal players in the intercellular communication that underlies physiological and pathological processes. In particular, glia-derived EVs play relevant roles in modulating neuroinflammation, either by promoting or inhibiting the activation of glial cells and neurons, or by facilitating the clearance or propagation of pathogenic proteins. The involvement of EVs in neurodegenerative diseases such as Alzheimer's Disease (AD), Parkinson's Disease (PD), Huntington's Disease (HD), and Multiple Sclerosis (MS)- which share hallmarks such as neuroinflammation and oxidative stress to DNA damage, alterations in neurotrophin levels, mitochondrial impairment, and altered protein dynamics- will be dissected, showing how EVs act as pivotal cell-cell mediators of toxic stimuli, thereby propagating degeneration and cell death signaling. Thus, this review focuses on the EVs secreted by microglia, astrocytes, oligodendrocytes and in neuroinflammatory conditions, emphasizing on their effects on neurons and on central nervous system functions, considering both their beneficial and detrimental effects.
Collapse
Affiliation(s)
- Francesco D'Egidio
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Italy 67100, Via Vetoio - Coppito1, Building "Renato Ricamo"
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Italy 67100, Via Vetoio - Coppito1, Building "Renato Ricamo"
| | - Michele d'Angelo
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Italy 67100, Via Vetoio - Coppito1, Building "Renato Ricamo".
| | - Fabrizio Ammannito
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Italy 67100, Via Vetoio - Coppito1, Building "Renato Ricamo"
| | - Massimiliano Quintiliani
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Italy 67100, Via Vetoio - Coppito1, Building "Renato Ricamo"
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, Italy 67100, Via Vetoio - Coppito1, Building "Renato Ricamo"
| |
Collapse
|
2
|
Risby-Jones G, Lee JD, Woodruff TM, Fung JN. Sex differences in Huntington's disease from a neuroinflammation perspective. Front Neurol 2024; 15:1384480. [PMID: 38915800 PMCID: PMC11194371 DOI: 10.3389/fneur.2024.1384480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 05/28/2024] [Indexed: 06/26/2024] Open
Abstract
Huntington's disease (HD) is a debilitating neurodegenerative condition characterized by motor, cognitive and psychiatric abnormalities. Immune dysregulation, prominently featuring increased immune activity, plays a significant role in HD pathogenesis. In addition to the central nervous system (CNS), systemic innate immune activation and inflammation are observed in HD patients, exacerbating the effects of the Huntingtin (HTT) gene mutation. Recent attention to sex differences in HD symptom severity underscores the need to consider gender as a biological variable in neurodegenerative disease research. Understanding sex-specific immune responses holds promise for elucidating HD pathophysiology and informing targeted treatment strategies to mitigate cognitive and functional decline. This perspective will highlight the importance of investigating gender influence in HD, particularly focusing on sex-specific immune responses predisposing individuals to disease.
Collapse
Affiliation(s)
- Grace Risby-Jones
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, Australia
| | - John D. Lee
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, Australia
| | - Trent M. Woodruff
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, Australia
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
| | - Jenny N. Fung
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, Australia
| |
Collapse
|
3
|
Pérot JB, Brouillet E, Flament J. The contribution of preclinical magnetic resonance imaging and spectroscopy to Huntington's disease. Front Aging Neurosci 2024; 16:1306312. [PMID: 38414634 PMCID: PMC10896846 DOI: 10.3389/fnagi.2024.1306312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 01/24/2024] [Indexed: 02/29/2024] Open
Abstract
Huntington's disease is an inherited disorder characterized by psychiatric, cognitive, and motor symptoms due to degeneration of medium spiny neurons in the striatum. A prodromal phase precedes the onset, lasting decades. Current biomarkers include clinical score and striatal atrophy using Magnetic Resonance Imaging (MRI). These markers lack sensitivity for subtle cellular changes during the prodromal phase. MRI and MR spectroscopy offer different contrasts for assessing metabolic, microstructural, functional, or vascular alterations in the disease. They have been used in patients and mouse models. Mouse models can be of great interest to study a specific mechanism of the degenerative process, allow better understanding of the pathogenesis from the prodromal to the symptomatic phase, and to evaluate therapeutic efficacy. Mouse models can be divided into three different constructions: transgenic mice expressing exon-1 of human huntingtin (HTT), mice with an artificial chromosome expressing full-length human HTT, and knock-in mouse models with CAG expansion inserted in the murine htt gene. Several studies have used MRI/S to characterized these models. However, the multiplicity of modalities and mouse models available complicates the understanding of this rich corpus. The present review aims at giving an overview of results obtained using MRI/S for each mouse model of HD, to provide a useful resource for the conception of neuroimaging studies using mouse models of HD. Finally, despite difficulties in translating preclinical protocols to clinical applications, many biomarkers identified in preclinical models have already been evaluated in patients. This review also aims to cover this aspect to demonstrate the importance of MRI/S for studying HD.
Collapse
Affiliation(s)
- Jean-Baptiste Pérot
- Laboratoire des Maladies Neurodégénératives, Molecular Imaging Research Center, Commissariat à l’Energie Atomique et aux Energies Alternatives, Centre National de la Recherche Scientifique, Université Paris-Saclay, Fontenay-aux-Roses, France
- Institut du Cerveau – Paris Brain Institute – ICM, Sorbonne Université, Paris, France
| | - Emmanuel Brouillet
- Laboratoire des Maladies Neurodégénératives, Molecular Imaging Research Center, Commissariat à l’Energie Atomique et aux Energies Alternatives, Centre National de la Recherche Scientifique, Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Julien Flament
- Laboratoire des Maladies Neurodégénératives, Molecular Imaging Research Center, Commissariat à l’Energie Atomique et aux Energies Alternatives, Centre National de la Recherche Scientifique, Université Paris-Saclay, Fontenay-aux-Roses, France
| |
Collapse
|
4
|
Hanrahan J, Locke DP, Cahill LS. Magnetic Resonance Imaging to Detect Structural Brain Changes in Huntington's Disease: A Review of Data from Mouse Models. J Huntingtons Dis 2024; 13:279-299. [PMID: 39213087 PMCID: PMC11494634 DOI: 10.3233/jhd-240045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/14/2024] [Indexed: 09/04/2024]
Abstract
Structural magnetic resonance imaging (MRI) is a powerful tool to visualize 3D neuroanatomy and assess pathology and disease progression in neurodegenerative disorders such as Huntington's disease (HD). The development of mouse models of HD that reproduce many of the psychiatric, motor and cognitive impairments observed in human HD has improved our understanding of the disease and provided opportunities for testing novel therapies. Similar to the clinical scenario, MRI of mouse models of HD demonstrates onset and progression of brain pathology. Here, we provided an overview of the articles that used structural MRI in mouse models of HD to date, highlighting the differences between studies and models and describing gaps in the current state of knowledge and recommendations for future studies.
Collapse
Affiliation(s)
- Jenna Hanrahan
- Department of Chemistry, Memorial University of Newfoundland, St. John’s, Newfoundland and Labrador, Canada
| | - Drew P. Locke
- Department of Chemistry, Memorial University of Newfoundland, St. John’s, Newfoundland and Labrador, Canada
| | - Lindsay S. Cahill
- Department of Chemistry, Memorial University of Newfoundland, St. John’s, Newfoundland and Labrador, Canada
- Discipline of Radiology, Memorial University of Newfoundland, St. John’s, Newfoundland and Labrador, Canada
| |
Collapse
|
5
|
Binda CS, Lelos MJ, Rosser AE, Massey TH. Using gene or cell therapies to treat Huntington's disease. HANDBOOK OF CLINICAL NEUROLOGY 2024; 205:193-215. [PMID: 39341655 DOI: 10.1016/b978-0-323-90120-8.00014-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Huntington's disease is caused by a CAG repeat expansion in the first exon of the HTT gene, leading to the production of gain-of-toxic-function mutant huntingtin protein species and consequent transcriptional dysregulation and disrupted cell metabolism. The brunt of the disease process is borne by the striatum from the earliest disease stages, with striatal atrophy beginning approximately a decade prior to the onset of neurologic signs. Although the expanded CAG repeat in the HTT gene is necessary and sufficient to cause HD, other genes can influence the age at onset of symptoms and how they progress. Many of these modifier genes have roles in DNA repair and are likely to modulate the stability of the CAG repeat in somatic cells. Currently, there are no disease-modifying treatments for HD that can be prescribed to patients and few symptomatic treatments, but there is a lot of interest in therapeutics that can target the pathogenic pathways at the DNA and RNA levels, some of which have reached the stage of human studies. In contrast, cell therapies aim to replace key neural cells lost to the disease process and/or to support the host vulnerable striatum by direct delivery of cells to the brain. Ultimately it may be possible to combine gene and cell therapies to both slow disease processes and provide some level of neural repair. In this chapter we consider the current status of these therapeutic strategies along with their prospects and challenges.
Collapse
Affiliation(s)
- Caroline S Binda
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom; UK Dementia Research Institute at Cardiff, Cardiff University, Cardiff, United Kingdom
| | - Mariah J Lelos
- Brain Repair Group, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Anne E Rosser
- Brain Repair Group, School of Biosciences, Cardiff University, Cardiff, United Kingdom; BRAIN Unit, Neuroscience and Mental Health Research Institute, Cardiff, United Kingdom.
| | - Thomas H Massey
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom; UK Dementia Research Institute at Cardiff, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
6
|
Nittari G, Roy P, Martinelli I, Bellitto V, Tomassoni D, Traini E, Tayebati SK, Amenta F. Rodent Models of Huntington's Disease: An Overview. Biomedicines 2023; 11:3331. [PMID: 38137552 PMCID: PMC10741199 DOI: 10.3390/biomedicines11123331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
Huntington's disease (HD) is an autosomal-dominant inherited neurological disorder caused by a genetic mutation in the IT15 gene. This neurodegenerative disorder is caused by a polyglutamine repeat expansion mutation in the widely expressed huntingtin (HTT) protein. HD is characterized by the degeneration of basal ganglia neurons and progressive cell death in intrinsic neurons of the striatum, accompanied by dementia and involuntary abnormal choreiform movements. Animal models have been extensively studied and have proven to be extremely valuable for therapeutic target evaluations. They reveal the hallmark of the age-dependent formation of aggregates or inclusions consisting of misfolded proteins. Animal models of HD have provided a therapeutic strategy to treat HD by suppressing mutant HTT (mHTT). Transgenic animal models have significantly increased our understanding of the molecular processes and pathophysiological mechanisms underlying the HD behavioral phenotype. Since effective therapies to cure or interrupt the course of the disease are not yet available, clinical research will have to make use of reliable animal models. This paper reviews the main studies of rodents as HD animal models, highlighting the neurological and behavioral differences between them. The choice of an animal model depends on the specific aspect of the disease to be investigated. Toxin-based models can still be useful, but most experimental hypotheses depend on success in a genetic model, whose choice is determined by the experimental question. There are many animal models showing similar HD symptoms or pathologies. They include chemical-induced HDs and genetic HDs, where cell-free and cell culture, lower organisms (such as yeast, Drosophila, C. elegans, zebrafish), rodents (mice, rats), and non-human primates are involved. These models provide accessible systems to study molecular pathogenesis and test potential treatments. For developing more effective pharmacological treatments, better animal models must be available and used to evaluate the efficacy of drugs.
Collapse
Affiliation(s)
- Giulio Nittari
- School of Medicinal and Health Products Sciences, University of Camerino, Via Madonna Delle Carceri, 9, 62032 Camerino, Italy; (G.N.); (P.R.); (I.M.); (V.B.); (E.T.); (S.K.T.)
| | - Proshanta Roy
- School of Medicinal and Health Products Sciences, University of Camerino, Via Madonna Delle Carceri, 9, 62032 Camerino, Italy; (G.N.); (P.R.); (I.M.); (V.B.); (E.T.); (S.K.T.)
| | - Ilenia Martinelli
- School of Medicinal and Health Products Sciences, University of Camerino, Via Madonna Delle Carceri, 9, 62032 Camerino, Italy; (G.N.); (P.R.); (I.M.); (V.B.); (E.T.); (S.K.T.)
| | - Vincenzo Bellitto
- School of Medicinal and Health Products Sciences, University of Camerino, Via Madonna Delle Carceri, 9, 62032 Camerino, Italy; (G.N.); (P.R.); (I.M.); (V.B.); (E.T.); (S.K.T.)
| | - Daniele Tomassoni
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032 Camerino, Italy;
| | - Enea Traini
- School of Medicinal and Health Products Sciences, University of Camerino, Via Madonna Delle Carceri, 9, 62032 Camerino, Italy; (G.N.); (P.R.); (I.M.); (V.B.); (E.T.); (S.K.T.)
| | - Seyed Khosrow Tayebati
- School of Medicinal and Health Products Sciences, University of Camerino, Via Madonna Delle Carceri, 9, 62032 Camerino, Italy; (G.N.); (P.R.); (I.M.); (V.B.); (E.T.); (S.K.T.)
| | - Francesco Amenta
- School of Medicinal and Health Products Sciences, University of Camerino, Via Madonna Delle Carceri, 9, 62032 Camerino, Italy; (G.N.); (P.R.); (I.M.); (V.B.); (E.T.); (S.K.T.)
| |
Collapse
|
7
|
Shing K, Sapp E, Boudi A, Liu S, Seeley C, Marchionini D, DiFiglia M, Kegel-Gleason KB. Early whole-body mutant huntingtin lowering averts changes in proteins and lipids important for synapse function and white matter maintenance in the LacQ140 mouse model. Neurobiol Dis 2023; 187:106313. [PMID: 37777020 PMCID: PMC10731584 DOI: 10.1016/j.nbd.2023.106313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/25/2023] [Accepted: 09/27/2023] [Indexed: 10/02/2023] Open
Abstract
Expansion of a triplet repeat tract in exon 1 of the HTT gene causes Huntington's disease (HD). The mutant HTT protein (mHTT) has numerous aberrant interactions with diverse, pleiomorphic effects. Lowering mHTT is a promising approach to treat HD, but it is unclear when lowering should be initiated, how much is necessary, and what duration should occur to achieve benefits. Furthermore, the effects of mHTT lowering on brain lipids have not been assessed. Using a mHtt-inducible mouse model, we analyzed mHtt lowering initiated at different ages and sustained for different time-periods. mHTT protein in cytoplasmic and synaptic compartments of the striatum was reduced 38-52%; however, there was minimal lowering of mHTT in nuclear and perinuclear regions where aggregates formed at 12 months of age. Total striatal lipids were reduced in 9-month-old LacQ140 mice and preserved by mHtt lowering. Subclasses important for white matter structure and function including ceramide (Cer), sphingomyelin (SM), and monogalactosyldiacylglycerol (MGDG), contributed to the reduction in total lipids. Phosphatidylinositol (PI), phosphatidylserine (PS), and bismethyl phosphatidic acid (BisMePA) were also changed in LacQ140 mice. Levels of all subclasses except ceramide were preserved by mHtt lowering. mRNA expression profiling indicated that a transcriptional mechanism contributes to changes in myelin lipids, and some but not all changes can be prevented by mHtt lowering. Our findings suggest that early and sustained reduction in mHtt can prevent changes in levels of select striatal proteins and most lipids, but a misfolded, degradation-resistant form of mHTT hampers some benefits in the long term.
Collapse
Affiliation(s)
- Kai Shing
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Ellen Sapp
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Adel Boudi
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Sophia Liu
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Connor Seeley
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | | | - Marian DiFiglia
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | | |
Collapse
|
8
|
Ferrari Bardile C, Radulescu CI, Pouladi MA. Oligodendrocyte pathology in Huntington's disease: from mechanisms to therapeutics. Trends Mol Med 2023; 29:802-816. [PMID: 37591764 DOI: 10.1016/j.molmed.2023.07.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/18/2023] [Accepted: 07/20/2023] [Indexed: 08/19/2023]
Abstract
Oligodendrocytes (OLGs), highly specialized glial cells that wrap axons with myelin sheaths, are critical for brain development and function. There is new recognition of the role of OLGs in the pathogenesis of neurodegenerative diseases (NDDs), including Huntington's disease (HD), a prototypic NDD caused by a polyglutamine tract expansion in huntingtin (HTT), which results in gain- and loss-of-function effects. Clinically, HD is characterized by a constellation of motor, cognitive, and psychiatric disturbances. White matter (WM) structures, representing myelin-rich regions of the brain, are profoundly affected in HD, and recent findings reveal oligodendroglia dysfunction as an early pathological event. Here, we focus on mechanisms that underlie oligodendroglial deficits and dysmyelination in the progression of the disease, highlighting the pathogenic contributions of mutant HTT (mHTT). We also discuss potential therapeutic implications involving these molecular pathways.
Collapse
Affiliation(s)
- Costanza Ferrari Bardile
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, Djavad Mowafaghian Centre for Brain Health, British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | - Carola I Radulescu
- UK Dementia Research Institute, Imperial College London, London, W12 0NN, UK
| | - Mahmoud A Pouladi
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, Djavad Mowafaghian Centre for Brain Health, British Columbia Children's Hospital Research Institute, University of British Columbia, Vancouver, BC V5Z 4H4, Canada.
| |
Collapse
|
9
|
Manivannan A, Foley LM, Hitchens TK, Rattray I, Bates GP, Modo M. Ex vivo 100 μm isotropic diffusion MRI-based tractography of connectivity changes in the end-stage R6/2 mouse model of Huntington's disease. NEUROPROTECTION 2023; 1:66-83. [PMID: 37745674 PMCID: PMC10516267 DOI: 10.1002/nep3.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 11/08/2022] [Indexed: 09/26/2023]
Abstract
Background Huntington's disease is a progressive neurodegenerative disorder. Brain atrophy, as measured by volumetric magnetic resonance imaging (MRI), is a downstream consequence of neurodegeneration, but microstructural changes within brain tissue are expected to precede this volumetric decline. The tissue microstructure can be assayed non-invasively using diffusion MRI, which also allows a tractographic analysis of brain connectivity. Methods We here used ex vivo diffusion MRI (11.7 T) to measure microstructural changes in different brain regions of end-stage (14 weeks of age) wild type and R6/2 mice (male and female) modeling Huntington's disease. To probe the microstructure of different brain regions, reduce partial volume effects and measure connectivity between different regions, a 100 μm isotropic voxel resolution was acquired. Results Although fractional anisotropy did not reveal any difference between wild-type controls and R6/2 mice, mean, axial, and radial diffusivity were increased in female R6/2 mice and decreased in male R6/2 mice. Whole brain streamlines were only reduced in male R6/2 mice, but streamline density was increased. Region-to-region tractography indicated reductions in connectivity between the cortex, hippocampus, and thalamus with the striatum, as well as within the basal ganglia (striatum-globus pallidus-subthalamic nucleus-substantia nigra-thalamus). Conclusions Biological sex and left/right hemisphere affected tractographic results, potentially reflecting different stages of disease progression. This proof-of-principle study indicates that diffusion MRI and tractography potentially provide novel biomarkers that connect volumetric changes across different brain regions. In a translation setting, these measurements constitute a novel tool to assess the therapeutic impact of interventions such as neuroprotective agents in transgenic models, as well as patients with Huntington's disease.
Collapse
Affiliation(s)
- Ashwinee Manivannan
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Lesley M. Foley
- Animal Imaging Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - T. Kevin Hitchens
- Animal Imaging Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ivan Rattray
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, Huntington’s Disease Centre and UK Dementia Research Institute at UCL, University College London, London, UK
| | - Gillian P. Bates
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, Huntington’s Disease Centre and UK Dementia Research Institute at UCL, University College London, London, UK
| | - Michel Modo
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
10
|
Shing K, Sapp E, Boudi A, Liu S, Seeley C, Marchionini D, DiFiglia M, Kegel-Gleason KB. Early whole-body mutant huntingtin lowering averts changes in proteins and lipids important for synapse function and white matter maintenance in the LacQ140 mouse model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.26.525697. [PMID: 36747614 PMCID: PMC9900921 DOI: 10.1101/2023.01.26.525697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Expansion of a triplet repeat tract in exon1 of the HTT gene causes Huntington's disease (HD). The mutant HTT protein (mHTT) has numerous aberrant interactions with diverse, pleiomorphic effects. No disease modifying treatments exist but lowering mutant huntingtin (mHTT) by gene therapy is a promising approach to treat Huntington's disease (HD). It is not clear when lowering should be initiated, how much lowering is necessary and for what duration lowering should occur to achieve benefits. Furthermore, the effects of mHTT lowering on brain lipids have not been assessed. Using a mHtt-inducible mouse model we analyzed whole body mHtt lowering initiated at different ages and sustained for different time-periods. Subcellular fractionation (density gradient ultracentrifugation), protein chemistry (gel filtration, western blot, and capillary electrophoresis immunoassay), liquid chromatography and mass spectrometry of lipids, and bioinformatic approaches were used to test effects of mHTT transcriptional lowering. mHTT protein in cytoplasmic and synaptic compartments of the caudate putamen, which is most affected in HD, was reduced 38-52%. Little or no lowering of mHTT occurred in nuclear and perinuclear regions where aggregates formed at 12 months of age. mHtt transcript repression partially or fully preserved select striatal proteins (SCN4B, PDE10A). Total lipids in striatum were reduced in LacQ140 mice at 9 months and preserved by early partial mHtt lowering. The reduction in total lipids was due in part to reductions in subclasses of ceramide (Cer), sphingomyelin (SM), and monogalactosyldiacylglycerol (MGDG), which are known to be important for white matter structure and function. Lipid subclasses phosphatidylinositol (PI), phosphatidylserine (PS), and bismethyl phosphatidic acid (BisMePA) were also changed in LacQ140 mice. Levels of all subclasses other than ceramide were preserved by early mHtt lowering. Pathway enrichment analysis of RNAseq data imply a transcriptional mechanism is responsible in part for changes in myelin lipids, and some but not all changes can be rescued by mHTT lowering. Our findings suggest that early and sustained reduction in mHtt can prevent changes in levels of select striatal proteins and most lipids but a misfolded, degradation-resistant form of mHTT hampers some benefits in the long term.
Collapse
Affiliation(s)
- Kai Shing
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129
| | - Ellen Sapp
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129
| | - Adel Boudi
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129
| | - Sophia Liu
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129
| | - Connor Seeley
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129
| | | | - Marian DiFiglia
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129
| | | |
Collapse
|
11
|
Villanueva CB, Stephensen HJT, Mokso R, Benraiss A, Sporring J, Goldman SA. Astrocytic engagement of the corticostriatal synaptic cleft is disrupted in a mouse model of Huntington's disease. Proc Natl Acad Sci U S A 2023; 120:e2210719120. [PMID: 37279261 PMCID: PMC10268590 DOI: 10.1073/pnas.2210719120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 04/25/2023] [Indexed: 06/08/2023] Open
Abstract
Astroglial dysfunction contributes to the pathogenesis of Huntington's disease (HD), and glial replacement can ameliorate the disease course. To establish the topographic relationship of diseased astrocytes to medium spiny neuron (MSN) synapses in HD, we used 2-photon imaging to map the relationship of turboRFP-tagged striatal astrocytes and rabies-traced, EGFP-tagged coupled neuronal pairs in R6/2 HD and wild-type (WT) mice. The tagged, prospectively identified corticostriatal synapses were then studied by correlated light electron microscopy followed by serial block-face scanning EM, allowing nanometer-scale assessment of synaptic structure in 3D. By this means, we compared the astrocytic engagement of single striatal synapses in HD and WT brains. R6/2 HD astrocytes exhibited constricted domains, with significantly less coverage of mature dendritic spines than WT astrocytes, despite enhanced engagement of immature, thin spines. These data suggest that disease-dependent changes in the astroglial engagement and sequestration of MSN synapses enable the high synaptic and extrasynaptic levels of glutamate and K+ that underlie striatal hyperexcitability in HD. As such, these data suggest that astrocytic structural pathology may causally contribute to the synaptic dysfunction and disease phenotype of those neurodegenerative disorders characterized by network overexcitation.
Collapse
Affiliation(s)
- Carlos Benitez Villanueva
- Center for Translational Neuromedicine, University of Copenhagen, Faculty of Health and Medical Sciences, Copenhagen N2200, Denmark
| | - Hans J. T. Stephensen
- Center for Translational Neuromedicine, University of Copenhagen, Faculty of Health and Medical Sciences, Copenhagen N2200, Denmark
- Department of Computer Science, University of Copenhagen, Faculty of Science, Copenhagen N2200, Denmark
| | - Rajmund Mokso
- Faculty of Engineering, Division of Solid Mechanics, Lund University, Lund22100, Sweden
| | - Abdellatif Benraiss
- Center for Translational Neuroscience, Department of Neurology, University of Rochester Medical Center, Rochester, NY14642
| | - Jon Sporring
- Department of Computer Science, University of Copenhagen, Faculty of Science, Copenhagen N2200, Denmark
| | - Steven A. Goldman
- Center for Translational Neuromedicine, University of Copenhagen, Faculty of Health and Medical Sciences, Copenhagen N2200, Denmark
- Center for Translational Neuroscience, Department of Neurology, University of Rochester Medical Center, Rochester, NY14642
| |
Collapse
|
12
|
Pepe G, Lenzi P, Capocci L, Marracino F, Pizzati L, Scarselli P, Di Pardo A, Fornai F, Maglione V. Treatment with the Glycosphingolipid Modulator THI Rescues Myelin Integrity in the Striatum of R6/2 HD Mice. Int J Mol Sci 2023; 24:ijms24065956. [PMID: 36983032 PMCID: PMC10053002 DOI: 10.3390/ijms24065956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/16/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Huntington's disease is one of the most common dominantly inherited neurodegenerative disorders caused by an expansion of a polyglutamine (polyQ) stretch in the N-terminal region of huntingtin (Htt). Among all the molecular mechanisms, affected by the mutation, emerging evidence proposes glycosphingolipid dysfunction as one of the major determinants. High levels of sphingolipids have been found to localize in the myelin sheaths of oligodendrocytes, where they play an important role in myelination stability and functions. In this study, we investigated any potential existing link between sphingolipid modulation and myelin structure by performing both ultrastructural and biochemical analyses. Our findings demonstrated that the treatment with the glycosphingolipid modulator THI preserved myelin thickness and the overall structure and reduced both area and diameter of pathologically giant axons in the striatum of HD mice. These ultrastructural findings were associated with restoration of different myelin marker protein, such as myelin-associated glycoprotein (MAG), myelin basic protein (MBP) and 2', 3' Cyclic Nucleotide 3'-Phosphodiesterase (CNP). Interestingly, the compound modulated the expression of glycosphingolipid biosynthetic enzymes and increased levels of GM1, whose elevation has been extensively reported to be associated with reduced toxicity of mutant Htt in different HD pre-clinical models. Our study further supports the evidence that the metabolism of glycosphingolipids may represent an effective therapeutic target for the disease.
Collapse
Affiliation(s)
- Giuseppe Pepe
- IRCCS Neuromed, Via Dell'elettronica, 86077 Pozzilli, Italy
| | - Paola Lenzi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy
| | - Luca Capocci
- IRCCS Neuromed, Via Dell'elettronica, 86077 Pozzilli, Italy
| | | | | | | | - Alba Di Pardo
- IRCCS Neuromed, Via Dell'elettronica, 86077 Pozzilli, Italy
| | - Francesco Fornai
- IRCCS Neuromed, Via Dell'elettronica, 86077 Pozzilli, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy
| | | |
Collapse
|
13
|
Burtscher J, Pepe G, Maharjan N, Riguet N, Di Pardo A, Maglione V, Millet GP. Sphingolipids and impaired hypoxic stress responses in Huntington disease. Prog Lipid Res 2023; 90:101224. [PMID: 36898481 DOI: 10.1016/j.plipres.2023.101224] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/20/2023] [Accepted: 03/05/2023] [Indexed: 03/11/2023]
Abstract
Huntington disease (HD) is a debilitating, currently incurable disease. Protein aggregation and metabolic deficits are pathological hallmarks but their link to neurodegeneration and symptoms remains debated. Here, we summarize alterations in the levels of different sphingolipids in an attempt to characterize sphingolipid patterns specific to HD, an additional molecular hallmark of the disease. Based on the crucial role of sphingolipids in maintaining cellular homeostasis, the dynamic regulation of sphingolipids upon insults and their involvement in cellular stress responses, we hypothesize that maladaptations or blunted adaptations, especially following cellular stress due to reduced oxygen supply (hypoxia) contribute to the development of pathology in HD. We review how sphingolipids shape cellular energy metabolism and control proteostasis and suggest how these functions may fail in HD and in combination with additional insults. Finally, we evaluate the potential of improving cellular resilience in HD by conditioning approaches (improving the efficiency of cellular stress responses) and the role of sphingolipids therein. Sphingolipid metabolism is crucial for cellular homeostasis and for adaptations following cellular stress, including hypoxia. Inadequate cellular management of hypoxic stress likely contributes to HD progression, and sphingolipids are potential mediators. Targeting sphingolipids and the hypoxic stress response are novel treatment strategies for HD.
Collapse
Affiliation(s)
- Johannes Burtscher
- Institute of Sport Sciences, University of Lausanne, 1015 Lausanne, Switzerland; Department of Biomedical Sciences, University of Lausanne, 1005 Lausanne, Switzerland.
| | - Giuseppe Pepe
- IRCCS Neuromed, Via Dell'Elettronica, 86077 Pozzilli, Italy
| | - Niran Maharjan
- Department of Neurology, Center for Experimental Neurology, Inselspital University Hospital, 3010 Bern, Switzerland; Department for Biomedical Research (DBMR), University of Bern, 3010 Bern, Switzerland
| | | | - Alba Di Pardo
- IRCCS Neuromed, Via Dell'Elettronica, 86077 Pozzilli, Italy
| | | | - Grégoire P Millet
- Institute of Sport Sciences, University of Lausanne, 1015 Lausanne, Switzerland; Department of Biomedical Sciences, University of Lausanne, 1005 Lausanne, Switzerland
| |
Collapse
|
14
|
Farzana F, McConville MJ, Renoir T, Li S, Nie S, Tran H, Hannan AJ, Hatters DM, Boughton BA. Longitudinal spatial mapping of lipid metabolites reveals pre-symptomatic changes in the hippocampi of Huntington's disease transgenic mice. Neurobiol Dis 2023; 176:105933. [PMID: 36436748 DOI: 10.1016/j.nbd.2022.105933] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/16/2022] [Accepted: 11/23/2022] [Indexed: 11/26/2022] Open
Abstract
In Huntington's disease (HD), a key pathological feature includes the development of inclusion-bodies of fragments of the mutant huntingtin protein in the neurons of the striatum and hippocampus. To examine the molecular changes associated with inclusion-body formation, we applied MALDI-mass spectrometry imaging and deuterium pulse labelling to determine lipid levels and synthesis rates in the hippocampus of a transgenic mouse model of HD (R6/1 line). The R6/1 HD mice lacked inclusions in the hippocampus at 6 weeks of age (pre-symptomatic), whereas inclusions were pervasive by 16 weeks of age (symptomatic). Hippocampal subfields (CA1, CA3 and DG), which formed the highest density of inclusion formation in the mouse brain showed a reduction in the relative abundance of neuron-enriched lipids that have roles in neurotransmission, synaptic plasticity, neurogenesis, and ER-stress protection. Lipids involved in the adaptive response to ER stress (phosphatidylinositol, phosphatidic acid, and ganglioside classes) displayed increased rates of synthesis in HD mice relative to WT mice at all the ages examined, including prior to the formation of the inclusion bodies. Our findings, therefore, support a role for ER stress occurring pre-symptomatically and potentially contributing to pathological mechanisms underlying HD.
Collapse
Affiliation(s)
- Farheen Farzana
- Florey Institute of Neuroscience & Mental Health, The University of Melbourne, Victoria 3010, Australia; Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria 3010, Australia
| | - Malcolm J McConville
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria 3010, Australia; Metabolomics Australia, The University of Melbourne, Victoria 3010, Australia
| | - Thibault Renoir
- Florey Institute of Neuroscience & Mental Health, The University of Melbourne, Victoria 3010, Australia
| | - Shanshan Li
- Florey Institute of Neuroscience & Mental Health, The University of Melbourne, Victoria 3010, Australia
| | - Shuai Nie
- Melbourne Mass Spectrometry and Proteomics Facility, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria 3010, Australia
| | - Harvey Tran
- Florey Institute of Neuroscience & Mental Health, The University of Melbourne, Victoria 3010, Australia
| | - Anthony J Hannan
- Florey Institute of Neuroscience & Mental Health, The University of Melbourne, Victoria 3010, Australia.
| | - Danny M Hatters
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria 3010, Australia.
| | - Berin A Boughton
- School of Biosciences, The University of Melbourne, Victoria 3010, Australia; Australian National Phenome Centre, Murdoch University, Murdoch 6150, Western Australia, Australia.
| |
Collapse
|
15
|
Cvetanovic M, Gray M. Contribution of Glial Cells to Polyglutamine Diseases: Observations from Patients and Mouse Models. Neurotherapeutics 2023; 20:48-66. [PMID: 37020152 PMCID: PMC10119372 DOI: 10.1007/s13311-023-01357-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2023] [Indexed: 04/07/2023] Open
Abstract
Neurodegenerative diseases are broadly characterized neuropathologically by the degeneration of vulnerable neuronal cell types in a specific brain region. The degeneration of specific cell types has informed on the various phenotypes/clinical presentations in someone suffering from these diseases. Prominent neurodegeneration of specific neurons is seen in polyglutamine expansion diseases including Huntington's disease (HD) and spinocerebellar ataxias (SCA). The clinical manifestations observed in these diseases could be as varied as the abnormalities in motor function observed in those who have Huntington's disease (HD) as demonstrated by a chorea with substantial degeneration of striatal medium spiny neurons (MSNs) or those with various forms of spinocerebellar ataxia (SCA) with an ataxic motor presentation primarily due to degeneration of cerebellar Purkinje cells. Due to the very significant nature of the degeneration of MSNs in HD and Purkinje cells in SCAs, much of the research has centered around understanding the cell autonomous mechanisms dysregulated in these neuronal cell types. However, an increasing number of studies have revealed that dysfunction in non-neuronal glial cell types contributes to the pathogenesis of these diseases. Here we explore these non-neuronal glial cell types with a focus on how each may contribute to the pathogenesis of HD and SCA and the tools used to evaluate glial cells in the context of these diseases. Understanding the regulation of supportive and harmful phenotypes of glia in disease could lead to development of novel glia-focused neurotherapeutics.
Collapse
Affiliation(s)
- Marija Cvetanovic
- Department of Neuroscience, Institute for Translational Neuroscience, University of Minnesota, Minneapolis, USA
| | - Michelle Gray
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
16
|
Lim RG, Al-Dalahmah O, Wu J, Gold MP, Reidling JC, Tang G, Adam M, Dansu DK, Park HJ, Casaccia P, Miramontes R, Reyes-Ortiz AM, Lau A, Hickman RA, Khan F, Paryani F, Tang A, Ofori K, Miyoshi E, Michael N, McClure N, Flowers XE, Vonsattel JP, Davidson S, Menon V, Swarup V, Fraenkel E, Goldman JE, Thompson LM. Huntington disease oligodendrocyte maturation deficits revealed by single-nucleus RNAseq are rescued by thiamine-biotin supplementation. Nat Commun 2022; 13:7791. [PMID: 36543778 PMCID: PMC9772349 DOI: 10.1038/s41467-022-35388-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 11/30/2022] [Indexed: 12/24/2022] Open
Abstract
The complexity of affected brain regions and cell types is a challenge for Huntington's disease (HD) treatment. Here we use single nucleus RNA sequencing to investigate molecular pathology in the cortex and striatum from R6/2 mice and human HD post-mortem tissue. We identify cell type-specific and -agnostic signatures suggesting oligodendrocytes (OLs) and oligodendrocyte precursors (OPCs) are arrested in intermediate maturation states. OL-lineage regulators OLIG1 and OLIG2 are negatively correlated with CAG length in human OPCs, and ATACseq analysis of HD mouse NeuN-negative cells shows decreased accessibility regulated by OL maturation genes. The data implicates glucose and lipid metabolism in abnormal cell maturation and identify PRKCE and Thiamine Pyrophosphokinase 1 (TPK1) as central genes. Thiamine/biotin treatment of R6/1 HD mice to compensate for TPK1 dysregulation restores OL maturation and rescues neuronal pathology. Our insights into HD OL pathology spans multiple brain regions and link OL maturation deficits to abnormal thiamine metabolism.
Collapse
Affiliation(s)
- Ryan G Lim
- UCI MIND, University of California Irvine, Irvine, CA, USA
| | - Osama Al-Dalahmah
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Jie Wu
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Maxwell P Gold
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Guomei Tang
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Miriam Adam
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - David K Dansu
- Advanced Science Research Center at the City University of New York, New York, NY, USA
| | - Hye-Jin Park
- Advanced Science Research Center at the City University of New York, New York, NY, USA
| | - Patrizia Casaccia
- Advanced Science Research Center at the City University of New York, New York, NY, USA
| | | | - Andrea M Reyes-Ortiz
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA
| | - Alice Lau
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, CA, USA
| | - Richard A Hickman
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Fatima Khan
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Fahad Paryani
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Alice Tang
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Kenneth Ofori
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Emily Miyoshi
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA
| | - Neethu Michael
- Department of Pathology, University of California Irvine, Irvine, CA, USA
| | - Nicolette McClure
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA
| | - Xena E Flowers
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York City, New York, NY, USA
| | - Jean Paul Vonsattel
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York City, New York, NY, USA
| | - Shawn Davidson
- Lewis-Sigler Institute for Integrative Genomics, Princeton, NJ, USA
| | - Vilas Menon
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Vivek Swarup
- UCI MIND, University of California Irvine, Irvine, CA, USA
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA
| | - Ernest Fraenkel
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - James E Goldman
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA.
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York City, New York, NY, USA.
| | - Leslie M Thompson
- UCI MIND, University of California Irvine, Irvine, CA, USA.
- Department of Biological Chemistry, University of California Irvine, Irvine, CA, USA.
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, CA, USA.
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA.
- Sue and Bill Gross Stem Cell Center University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
17
|
Sun Y, Tong H, Yang T, Liu L, Li XJ, Li S. Insights into White Matter Defect in Huntington's Disease. Cells 2022; 11:3381. [PMID: 36359783 PMCID: PMC9656068 DOI: 10.3390/cells11213381] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/21/2022] [Accepted: 10/21/2022] [Indexed: 08/05/2023] Open
Abstract
Huntington's disease (HD) is an autosomal-dominant inherited progressive neurodegenerative disorder. It is caused by a CAG repeat expansion in the Huntingtin gene that is translated to an expanded polyglutamine (PolyQ) repeat in huntingtin protein. HD is characterized by mood swings, involuntary movement, and cognitive decline in the late disease stage. HD patients often die 15-20 years after disease onset. Currently, there is no cure for HD. Due to the striking neuronal loss in HD, most studies focused on the investigation of the predominantly neuronal degeneration in specific brain regions. However, the pathology of the white matter area in the brains of HD patients was also reported by clinical imaging studies, which showed white matter abnormalities even before the clinical onset of HD. Since oligodendrocytes form myelin sheaths around the axons in the brain, white matter lesions are likely attributed to alterations in myelin and oligodendrocyte-associated changes in HD. In this review, we summarized the evidence for white matter, myelin, and oligodendrocytes alterations that were previously observed in HD patients and animal models. We also discussed potential mechanisms for white matter changes and possible treatment to prevent glial dysfunction in HD.
Collapse
|
18
|
Bocci T, Baloscio D, Ferrucci R, Briscese L, Priori A, Sartucci F. Interhemispheric Connectivity in Idiopathic Cervical Dystonia and Spinocerebellar Ataxias: A Transcranial Magnetic Stimulation Study. Clin EEG Neurosci 2022; 53:460-466. [PMID: 32938220 DOI: 10.1177/1550059420957487] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND RATIONALE Hyperkinetic movement disorders represent a heterogeneous group of diseases, different from a genetic and clinical perspective. In the past, neurophysiological approaches provided different, sometimes contradictory findings, pointing to an impaired cortical inhibition as a common electrophysiological marker. Our aim was to evaluate changes in interhemispheric communication in patients with idiopathic cervical dystonia (ICD) and spinocerebellar ataxias (SCAs). MATERIALS AND METHODS Eleven patients with ICD, 7 with genetically confirmed SCA2 or SCA3, and 10 healthy volunteers were enrolled. The onset latency and duration of the ipsilateral silent period (iSPOL and iSPD, respectively), as well as the so-called transcallosal conduction time (TCT), were then recorded from the abductor pollicis brevis of the right side using an 8-shaped focal coil with wing diameters of 70 mm; all these parameters were evaluated and compared among groups. In SCAs, changes in neurophysiological measures were also correlated to the mutational load. RESULTS iSPD was significantly shorter in patients with SCA2 and SCA3, when compared both to control and ICD (P < .0001); iSPOL and TCT were prolonged in SCAs patients (P < .001). Changes in iSPD, iSPOL, and TCT in SCAs are significantly correlated with the mutational load (P = .01, P = .02, and P = .002, respectively). DISCUSSION This is the first study to assess changes in interhemispheric communication in patients with SCAs and ICD, using a transcranial magnetic stimulation protocol. Together with previous data in Huntington's disease, we suggest that these changes may underlie, at least in part, a common disease mechanism of polyglutamine disorders.
Collapse
Affiliation(s)
- Tommaso Bocci
- "Aldo Ravelli" Center for Neurotechnology and Experiental Brain Therapeutics, Department of Health Sciences, University of Milan & ASST Santi Paolo e Carlo, Milan, Italy
| | - Davide Baloscio
- Section of Neurophysiopathology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Roberta Ferrucci
- "Aldo Ravelli" Center for Neurotechnology and Experiental Brain Therapeutics, Department of Health Sciences, University of Milan & ASST Santi Paolo e Carlo, Milan, Italy
| | - Lucia Briscese
- Severe Acquired Brain Injuries Unit, Cisanello University Hospital, Pisa, Italy
| | - Alberto Priori
- "Aldo Ravelli" Center for Neurotechnology and Experiental Brain Therapeutics, Department of Health Sciences, University of Milan & ASST Santi Paolo e Carlo, Milan, Italy
| | - Ferdinando Sartucci
- Section of Neurophysiopathology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
19
|
Casella C, Chamberland M, Laguna PL, Parker GD, Rosser AE, Coulthard E, Rickards H, Berry SC, Jones DK, Metzler‐Baddeley C. Mutation-related magnetization-transfer, not axon density, drives white matter differences in premanifest Huntington disease: Evidence from in vivo ultra-strong gradient MRI. Hum Brain Mapp 2022; 43:3439-3460. [PMID: 35396899 PMCID: PMC9248323 DOI: 10.1002/hbm.25859] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 03/07/2022] [Accepted: 03/27/2022] [Indexed: 11/10/2022] Open
Abstract
White matter (WM) alterations have been observed in Huntington disease (HD) but their role in the disease-pathophysiology remains unknown. We assessed WM changes in premanifest HD by exploiting ultra-strong-gradient magnetic resonance imaging (MRI). This allowed to separately quantify magnetization transfer ratio (MTR) and hindered and restricted diffusion-weighted signal fractions, and assess how they drove WM microstructure differences between patients and controls. We used tractometry to investigate region-specific alterations across callosal segments with well-characterized early- and late-myelinating axon populations, while brain-wise differences were explored with tract-based cluster analysis (TBCA). Behavioral measures were included to explore disease-associated brain-function relationships. We detected lower MTR in patients' callosal rostrum (tractometry: p = .03; TBCA: p = .03), but higher MTR in their splenium (tractometry: p = .02). Importantly, patients' mutation-size and MTR were positively correlated (all p-values < .01), indicating that MTR alterations may directly result from the mutation. Further, MTR was higher in younger, but lower in older patients relative to controls (p = .003), suggesting that MTR increases are detrimental later in the disease. Finally, patients showed higher restricted diffusion signal fraction (FR) from the composite hindered and restricted model of diffusion (CHARMED) in the cortico-spinal tract (p = .03), which correlated positively with MTR in the posterior callosum (p = .033), potentially reflecting compensatory mechanisms. In summary, this first comprehensive, ultra-strong gradient MRI study in HD provides novel evidence of mutation-driven MTR alterations at the premanifest disease stage which may reflect neurodevelopmental changes in iron, myelin, or a combination of these.
Collapse
Affiliation(s)
- Chiara Casella
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of PsychologyCardiff UniversityCardiffUK
- Department of Perinatal Imaging and Health, School of Biomedical Engineering & Imaging SciencesKing's College London, St Thomas' HospitalLondonUK
| | - Maxime Chamberland
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of PsychologyCardiff UniversityCardiffUK
- Donders Institute for Brain, Cognition and BehaviorRadboud UniversityNijmegenThe Netherlands
| | - Pedro L. Laguna
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of PsychologyCardiff UniversityCardiffUK
| | - Greg D. Parker
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of PsychologyCardiff UniversityCardiffUK
| | - Anne E. Rosser
- Department of Neurology and Psychological MedicineHayden Ellis BuildingCardiffUK
- School of BiosciencesCardiff UniversityCardiffUK
| | | | - Hugh Rickards
- Birmingham and Solihull Mental Health NHS Foundation TrustBirminghamUK
- Institute of Clinical Sciences, College of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
| | - Samuel C. Berry
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of PsychologyCardiff UniversityCardiffUK
| | - Derek K. Jones
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of PsychologyCardiff UniversityCardiffUK
| | - Claudia Metzler‐Baddeley
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of PsychologyCardiff UniversityCardiffUK
| |
Collapse
|
20
|
Environmental stimulation in Huntington disease patients and animal models. Neurobiol Dis 2022; 171:105725. [DOI: 10.1016/j.nbd.2022.105725] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 04/03/2022] [Accepted: 04/08/2022] [Indexed: 01/07/2023] Open
|
21
|
Karayel-Basar M, Uras I, Kiris I, Sahin B, Akgun E, Baykal AT. Spatial proteomic alterations detected via MALDI-MS imaging implicate neuronal loss in a Huntington's disease mouse (YAC128) brain. Mol Omics 2022; 18:336-347. [PMID: 35129568 DOI: 10.1039/d1mo00440a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder that occurs with the increase of CAG trinucleotide repeats in the huntingtin gene. To understand the mechanisms of HD, powerful proteomics techniques, such as liquid chromatography-tandem mass spectrometry (LC-MS/MS) were employed. However, one major drawback of these methods is loss of the region-specific quantitative information of the proteins due to analysis of total tissue lysates. Matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) is a MS-based label-free technique that works directly on tissue sections and gathers m/z values with their respective regional information. In this study, we established a data processing protocol that includes several software programs and methods to determine spatial protein alterations between the brain samples of a 12 month-old YAC128 HD mouse model and their non-transgenic littermates. 22 differentially expressed proteins were revealed with their respective regional information, and possible relationships of several proteins were discussed. As a validation of the MALDI-MSI analysis, a differentially expressed protein (GFAP) was verified using immunohistochemical staining. Furthermore, since several proteins detected in this study have previously been associated with neuronal loss, neuronal loss in the cortical region was demonstrated using an anti-NeuN immunohistochemical staining method. In conclusion, the findings of this research have provided insights into the spatial proteomic changes between HD transgenic and non-transgenic littermates and therefore, we suggest that MALDI-MSI is a powerful technique to determine spatial proteomic alterations between biological samples, and the data processing that we present here can be employed as a complementary tool for the data analysis.
Collapse
Affiliation(s)
- Merve Karayel-Basar
- Department of Medical Biochemistry and Molecular Biology, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Irep Uras
- Department of Medical Biochemistry and Molecular Biology, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Irem Kiris
- Department of Medical Biochemistry and Molecular Biology, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Betul Sahin
- Acibadem Labmed Clinical Laboratories, R&D Center, Istanbul, Turkey
| | - Emel Akgun
- Department of Medical Biochemistry and Molecular Biology, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Ahmet Tarik Baykal
- Department of Medical Biochemistry, Faculty of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| |
Collapse
|
22
|
Chen JF, Wang F, Huang NX, Xiao L, Mei F. Oligodendrocytes and Myelin: Active players in Neurodegenerative brains? Dev Neurobiol 2022; 82:160-174. [PMID: 35081276 DOI: 10.1002/dneu.22867] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 11/10/2022]
Abstract
Oligodendrocytes (OLs) are a major type of glial cells in the central nervous system that generate multiple myelin sheaths to wrap axons. Myelin ensures fast and efficient propagation of action potentials along axons and supports neurons with nourishment. The decay of OLs and myelin has been implicated in age-related neurodegenerative diseases and these changes are generally considered as an inevitable result of neuron loss and axon degeneration. Noticeably, OLs and myelin undergo dynamic changes in healthy adult brains, that is, newly formed OLs are continuously added throughout life from the differentiation of oligodendrocyte precursor cells (OPCs) and the pre-existing myelin sheaths may undergo degeneration or remodeling. Increasing evidence has shown that changes in OLs and myelin are present in the early stages of neurodegenerative diseases, and even prior to significant neuronal loss and functional deficits. More importantly, oligodendroglia-specific manipulation, by either deletion of the disease gene or enhancement of myelin renewal, can alleviate functional impairments in neurodegenerative animal models. These findings underscore the possibility that OLs and myelin are not passively but actively involved in neurodegenerative diseases and may play an important role in modulating neuronal function and survival. In this review, we summarize recent work characterizing OL and myelin changes in both healthy and neurodegenerative brains and discuss the potential of targeting oligodendroglial cells in treating neurodegenerative diseases. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Jing-Fei Chen
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University, Chongqing, 400038, China
| | - Fei Wang
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University, Chongqing, 400038, China
| | - Nan-Xing Huang
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University, Chongqing, 400038, China
| | - Lan Xiao
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University, Chongqing, 400038, China
| | - Feng Mei
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University, Chongqing, 400038, China
| |
Collapse
|
23
|
Haas E, Incebacak RD, Hentrich T, Huridou C, Schmidt T, Casadei N, Maringer Y, Bahl C, Zimmermann F, Mills JD, Aronica E, Riess O, Schulze-Hentrich JM, Hübener-Schmid J. A Novel SCA3 Knock-in Mouse Model Mimics the Human SCA3 Disease Phenotype Including Neuropathological, Behavioral, and Transcriptional Abnormalities Especially in Oligodendrocytes. Mol Neurobiol 2022; 59:495-522. [PMID: 34716557 PMCID: PMC8786755 DOI: 10.1007/s12035-021-02610-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 10/19/2021] [Indexed: 12/28/2022]
Abstract
Spinocerebellar ataxia type 3 is the most common autosomal dominant inherited ataxia worldwide, caused by a CAG repeat expansion in the Ataxin-3 gene resulting in a polyglutamine (polyQ)-expansion in the corresponding protein. The disease is characterized by neuropathological, phenotypical, and specific transcriptional changes in affected brain regions. So far, there is no mouse model available representing all the different aspects of the disease, yet highly needed for a better understanding of the disease pathomechanisms. Here, we characterized a novel Ataxin-3 knock-in mouse model, expressing a heterozygous or homozygous expansion of 304 CAACAGs in the murine Ataxin-3 locus using biochemical, behavioral, and transcriptomic approaches. We compared neuropathological, and behavioral features of the new knock-in model with the in SCA3 research mostly used YAC84Q mouse model. Further, we compared transcriptional changes found in cerebellar samples of the SCA3 knock-in mice and post-mortem human SCA3 patients. The novel knock-in mouse is characterized by the expression of a polyQ-expansion in the murine Ataxin-3 protein, leading to aggregate formation, especially in brain regions known to be vulnerable in SCA3 patients, and impairment of Purkinje cells. Along these neuropathological changes, the mice showed a reduction in body weight accompanied by gait and balance instability. Transcriptomic analysis of cerebellar tissue revealed age-dependent differential expression, enriched for genes attributed to myelinating oligodendrocytes. Comparing these changes with those found in cerebellar tissue of SCA3 patients, we discovered an overlap of differentially expressed genes pointing towards similar gene expression perturbances in several genes linked to myelin sheaths and myelinating oligodendrocytes.
Collapse
Affiliation(s)
- Eva Haas
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
- Centre for Rare Diseases, University of Tübingen, Tübingen, Germany
| | - Rana D Incebacak
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
- Centre for Rare Diseases, University of Tübingen, Tübingen, Germany
| | - Thomas Hentrich
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
- Centre for Rare Diseases, University of Tübingen, Tübingen, Germany
| | - Chrisovalantou Huridou
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
- Centre for Rare Diseases, University of Tübingen, Tübingen, Germany
| | - Thorsten Schmidt
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
- Centre for Rare Diseases, University of Tübingen, Tübingen, Germany
| | - Nicolas Casadei
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
- Centre for Rare Diseases, University of Tübingen, Tübingen, Germany
- DFG NGS Competence Center Tübingen, Tübingen, Germany
| | - Yacine Maringer
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
- Centre for Rare Diseases, University of Tübingen, Tübingen, Germany
| | - Carola Bahl
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
- Centre for Rare Diseases, University of Tübingen, Tübingen, Germany
| | - Frank Zimmermann
- Interfaculty Biomedical Facility (IBF) Biotechnology lab, University of Heidelberg, Heidelberg, Germany
| | - James D Mills
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Olaf Riess
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
- Centre for Rare Diseases, University of Tübingen, Tübingen, Germany
- DFG NGS Competence Center Tübingen, Tübingen, Germany
| | - Julia M Schulze-Hentrich
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
- Centre for Rare Diseases, University of Tübingen, Tübingen, Germany
| | - Jeannette Hübener-Schmid
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany.
- Centre for Rare Diseases, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
24
|
Zhang X, Huang N, Xiao L, Wang F, Li T. Replenishing the Aged Brains: Targeting Oligodendrocytes and Myelination? Front Aging Neurosci 2021; 13:760200. [PMID: 34899272 PMCID: PMC8656359 DOI: 10.3389/fnagi.2021.760200] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 11/02/2021] [Indexed: 11/13/2022] Open
Abstract
Aging affects almost all the aspects of brain functions, but the mechanisms remain largely undefined. Increasing number of literatures have manifested the important role of glial cells in regulating the aging process. Oligodendroglial lineage cell is a major type of glia in central nervous system (CNS), composed of mature oligodendrocytes (OLs), and oligodendroglia precursor cells (OPCs). OLs produce myelin sheaths that insulate axons and provide metabolic support to meet the energy demand. OPCs maintain the population throughout lifetime with the abilities to proliferate and differentiate into OLs. Increasing evidence has shown that oligodendroglial cells display active dynamics in adult and aging CNS, which is extensively involved in age-related brain function decline in the elderly. In this review, we summarized present knowledge about dynamic changes of oligodendroglial lineage cells during normal aging and discussed their potential roles in age-related functional decline. Especially, focused on declined myelinogenesis during aging and underlying mechanisms. Clarifying those oligodendroglial changes and their effects on neurofunctional decline may provide new insights in understanding aging associated brain function declines.
Collapse
Affiliation(s)
- Xi Zhang
- Department of Histology and Embryology, Army Medical University (Third Military Medical University), Chongqing, China
- Department of Ophthalmology, The General Hospital of Western Theater Command, Chengdu, China
| | - Nanxin Huang
- Department of Histology and Embryology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Lan Xiao
- Department of Histology and Embryology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Fei Wang
- Department of Histology and Embryology, Army Medical University (Third Military Medical University), Chongqing, China
| | - Tao Li
- Department of Histology and Embryology, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
25
|
Kim C, Yousefian-Jazi A, Choi SH, Chang I, Lee J, Ryu H. Non-Cell Autonomous and Epigenetic Mechanisms of Huntington's Disease. Int J Mol Sci 2021; 22:12499. [PMID: 34830381 PMCID: PMC8617801 DOI: 10.3390/ijms222212499] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/10/2021] [Accepted: 11/15/2021] [Indexed: 02/06/2023] Open
Abstract
Huntington's disease (HD) is a rare neurodegenerative disorder caused by an expansion of CAG trinucleotide repeat located in the exon 1 of Huntingtin (HTT) gene in human chromosome 4. The HTT protein is ubiquitously expressed in the brain. Specifically, mutant HTT (mHTT) protein-mediated toxicity leads to a dramatic degeneration of the striatum among many regions of the brain. HD symptoms exhibit a major involuntary movement followed by cognitive and psychiatric dysfunctions. In this review, we address the conventional role of wild type HTT (wtHTT) and how mHTT protein disrupts the function of medium spiny neurons (MSNs). We also discuss how mHTT modulates epigenetic modifications and transcriptional pathways in MSNs. In addition, we define how non-cell autonomous pathways lead to damage and death of MSNs under HD pathological conditions. Lastly, we overview therapeutic approaches for HD. Together, understanding of precise neuropathological mechanisms of HD may improve therapeutic approaches to treat the onset and progression of HD.
Collapse
Affiliation(s)
- Chaebin Kim
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Korea; (C.K.); (A.Y.-J.); (S.-H.C.)
| | - Ali Yousefian-Jazi
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Korea; (C.K.); (A.Y.-J.); (S.-H.C.)
| | - Seung-Hye Choi
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Korea; (C.K.); (A.Y.-J.); (S.-H.C.)
| | - Inyoung Chang
- Department of Biology, Boston University, Boston, MA 02215, USA;
| | - Junghee Lee
- Boston University Alzheimer’s Disease Research Center, Boston University, Boston, MA 02118, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
- VA Boston Healthcare System, Boston, MA 02130, USA
| | - Hoon Ryu
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Korea; (C.K.); (A.Y.-J.); (S.-H.C.)
| |
Collapse
|
26
|
Kim A, Lalonde K, Truesdell A, Gomes Welter P, Brocardo PS, Rosenstock TR, Gil-Mohapel J. New Avenues for the Treatment of Huntington's Disease. Int J Mol Sci 2021; 22:ijms22168363. [PMID: 34445070 PMCID: PMC8394361 DOI: 10.3390/ijms22168363] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 12/11/2022] Open
Abstract
Huntington’s disease (HD) is a neurodegenerative disorder caused by a CAG expansion in the HD gene. The disease is characterized by neurodegeneration, particularly in the striatum and cortex. The first symptoms usually appear in mid-life and include cognitive deficits and motor disturbances that progress over time. Despite being a genetic disorder with a known cause, several mechanisms are thought to contribute to neurodegeneration in HD, and numerous pre-clinical and clinical studies have been conducted and are currently underway to test the efficacy of therapeutic approaches targeting some of these mechanisms with varying degrees of success. Although current clinical trials may lead to the identification or refinement of treatments that are likely to improve the quality of life of those living with HD, major efforts continue to be invested at the pre-clinical level, with numerous studies testing novel approaches that show promise as disease-modifying strategies. This review offers a detailed overview of the currently approved treatment options for HD and the clinical trials for this neurodegenerative disorder that are underway and concludes by discussing potential disease-modifying treatments that have shown promise in pre-clinical studies, including increasing neurotropic support, modulating autophagy, epigenetic and genetic manipulations, and the use of nanocarriers and stem cells.
Collapse
Affiliation(s)
- Amy Kim
- Island Medical Program and Faculty of Medicine, University of British Columbia, Victoria, BC V8P 5C2, Canada; (A.K.); (K.L.)
| | - Kathryn Lalonde
- Island Medical Program and Faculty of Medicine, University of British Columbia, Victoria, BC V8P 5C2, Canada; (A.K.); (K.L.)
| | - Aaron Truesdell
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada;
- Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada
| | - Priscilla Gomes Welter
- Neuroscience Graduate Program, Federal University of Santa Catarina, Florianópolis 88040-900, Brazil; (P.G.W.); (P.S.B.)
| | - Patricia S. Brocardo
- Neuroscience Graduate Program, Federal University of Santa Catarina, Florianópolis 88040-900, Brazil; (P.G.W.); (P.S.B.)
| | - Tatiana R. Rosenstock
- Institute of Cancer and Genomic Science, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK;
- Department of Pharmacology, University of São Paulo, São Paulo 05508-000, Brazil
| | - Joana Gil-Mohapel
- Island Medical Program and Faculty of Medicine, University of British Columbia, Victoria, BC V8P 5C2, Canada; (A.K.); (K.L.)
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada;
- Correspondence: ; Tel.: +1-250-472-4597; Fax: +1-250-472-5505
| |
Collapse
|
27
|
Dansu DK, Sauma S, Casaccia P. Oligodendrocyte progenitors as environmental biosensors. Semin Cell Dev Biol 2021; 116:38-44. [PMID: 33092959 PMCID: PMC8053729 DOI: 10.1016/j.semcdb.2020.09.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/23/2020] [Accepted: 09/27/2020] [Indexed: 01/10/2023]
Abstract
The past decade has seen an important revision of the traditional concept of the role and function of glial cells. From "passive support" for neurons, oligodendrocyte lineage cells are now recognized as metabolic exchangers with neurons, a cellular interface with blood vessels and responders to gut-derived metabolites or changes in the social environment. In the developing brain, the differentiation of neonatal oligodendrocyte progenitors (nOPCs) is required for normal brain function. In adulthood, the differentiation of adult OPCs (aOPCs) serves an important role in learning, behavioral adaptation and response to myelin injury. Here, we propose the concept of OPCs as environmental biosensors, which "sense" chemical and physical stimuli over time and adjust to the new challenges by modifying their epigenome and consequent transcriptome. Because epigenetics defines the ability of the cell to "adapt" gene expression to changes in the environment, we propose a model of OPC differentiation resulting from time-dependent changes of the epigenomic landscape in response to declining mitogens, raising hormone levels, neuronal activity, changes in space constraints or stiffness of the extracellular matrix. We propose that the intrinsically different functional properties of aOPCs compared to nOPCs result from the accrual of "epigenetic memories" of distinct events, which are "recorded" in the nuclei of OPCs as histone and DNA marks, defining a "unique epigenomic landscape" over time.
Collapse
Affiliation(s)
- David K Dansu
- Graduate Program in Biochemistry, Graduate Center of the City University of New York, New York, NY, USA; Neuroscience Initiative, Advanced Science Research Center, The Graduate Center of the City University of New York, New York, NY, USA
| | - Sami Sauma
- Graduate Program in Biology, Graduate Center of the City University of New York, New York, NY, USA; Neuroscience Initiative, Advanced Science Research Center, The Graduate Center of the City University of New York, New York, NY, USA
| | - Patrizia Casaccia
- Graduate Program in Biochemistry, Graduate Center of the City University of New York, New York, NY, USA; Graduate Program in Biology, Graduate Center of the City University of New York, New York, NY, USA; Neuroscience Initiative, Advanced Science Research Center, The Graduate Center of the City University of New York, New York, NY, USA.
| |
Collapse
|
28
|
In Vivo Expression of Reprogramming Factor OCT4 Ameliorates Myelination Deficits and Induces Striatal Neuroprotection in Huntington's Disease. Genes (Basel) 2021; 12:genes12050712. [PMID: 34068799 PMCID: PMC8150572 DOI: 10.3390/genes12050712] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 04/27/2021] [Accepted: 05/01/2021] [Indexed: 12/26/2022] Open
Abstract
White matter atrophy has been shown to precede the massive loss of striatal GABAergic neurons in Huntington’s disease (HD). This study investigated the effects of in vivo expression of reprogramming factor octamer-binding transcription factor 4 (OCT4) on neural stem cell (NSC) niche activation in the subventricular zone (SVZ) and induction of cell fate specific to the microenvironment of HD. R6/2 mice randomly received adeno-associated virus 9 (AAV9)-OCT4, AAV9-Null, or phosphate-buffered saline into both lateral ventricles at 4 weeks of age. The AAV9-OCT4 group displayed significantly improved behavioral performance compared to the control groups. Following AAV9-OCT4 treatment, the number of newly generated NSCs and oligodendrocyte progenitor cells (OPCs) significantly increased in the SVZ, and the expression of OPC-related genes and glial cell-derived neurotrophic factor (GDNF) significantly increased. Further, amelioration of myelination deficits in the corpus callosum was observed through electron microscopy and magnetic resonance imaging, and striatal DARPP32+ GABAergic neurons significantly increased in the AAV9-OCT4 group. These results suggest that in situ expression of the reprogramming factor OCT4 in the SVZ induces OPC proliferation, thereby attenuating myelination deficits. Particularly, GDNF released by OPCs seems to induce striatal neuroprotection in HD, which explains the behavioral improvement in R6/2 mice overexpressing OCT4.
Collapse
|
29
|
Casella C, Kleban E, Rosser AE, Coulthard E, Rickards H, Fasano F, Metzler-Baddeley C, Jones DK. Multi-compartment analysis of the complex gradient-echo signal quantifies myelin breakdown in premanifest Huntington's disease. Neuroimage Clin 2021; 30:102658. [PMID: 33865029 PMCID: PMC8079666 DOI: 10.1016/j.nicl.2021.102658] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 03/25/2021] [Accepted: 03/30/2021] [Indexed: 12/04/2022]
Abstract
White matter (WM) alterations have been identified as a relevant pathological feature of Huntington's disease (HD). Increasing evidence suggests that WM changes in this disorder are due to alterations in myelin-associated biological processes. Multi-compartmental analysis of the complex gradient-echo MRI signal evolution in WM has been shown to quantify myelin in vivo, therefore pointing to the potential of this technique for the study of WM myelin changes in health and disease. This study first characterized the reproducibility of metrics derived from the complex multi-echo gradient-recalled echo (mGRE) signal across the corpus callosum in healthy participants, finding highest reproducibility in the posterior callosal segment. Subsequently, the same analysis pipeline was applied in this callosal region in a sample of premanifest HD patients (n = 19) and age, sex and education matched healthy controls (n = 21). In particular, we focused on two myelin-associated derivatives: i. the myelin water signal fraction (fm), a parameter dependent on myelin content; and ii. The difference in frequency between myelin and intra-axonal water pools (Δω), a parameter dependent on the ratio between the inner and the outer axonal radii. fm was found to be lower in HD patients (β = -0.13, p = 0.03), while Δω did not show a group effect. Performance in tests of working memory, executive function, social cognition and movement was also assessed, and a greater age-related decline in executive function was detected in HD patients (β = -0.06, p = 0.006), replicating previous evidence of executive dysfunction in HD. Finally, the correlation between fm, executive function, and proximity to disease onset was explored in patients, and a positive correlation between executive function and fm was detected (r = 0.542; p = 0.02). This study emphasises the potential of complex mGRE signal analysis for aiding understanding of HD pathogenesis and progression. Moreover, expanding on evidence from pathology and animal studies, it provides novel in vivo evidence supporting myelin breakdown as an early feature of HD.
Collapse
Affiliation(s)
- Chiara Casella
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Maindy Road, Cardiff, CF 24 4HQ, UK.
| | - Elena Kleban
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Maindy Road, Cardiff, CF 24 4HQ, UK
| | - Anne E Rosser
- Department of Neurology and Psychological Medicine, Hayden Ellis Building, Maindy Road, Cardiff CF24 4HQ, UK; School of Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3AX, UK
| | | | - Hugh Rickards
- Birmingham and Solihull Mental Health NHS Foundation Trust, 50 Summer Hill Road, Birmingham B1 3RB, UK; Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Fabrizio Fasano
- Siemens Healthcare Ltd, Camberly, UK; Siemens Healthcare GmbH, Erlangen, Germany
| | - Claudia Metzler-Baddeley
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Maindy Road, Cardiff, CF 24 4HQ, UK
| | - Derek K Jones
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Maindy Road, Cardiff, CF 24 4HQ, UK
| |
Collapse
|
30
|
Braz SO, Dinca DM, Gourdon G, Gomes-Pereira M. Real Time Videomicroscopy and Semiautomated Analysis of Brain Cell Culture Models of Trinucleotide Repeat Expansion Diseases. Methods Mol Biol 2020; 2056:217-240. [PMID: 31586351 DOI: 10.1007/978-1-4939-9784-8_14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Proper brain function requires the coordinated and intricate interaction between neuronal and glial cells. Like many other neurological conditions, trinucleotide repeat expansion disorders are likely initiated by the synergistic combination of abnormalities hitting different brain cell types, which ultimately disrupt brain function and lead to the onset of neurological symptoms. Understanding how trinucleotide repeat expansions affect the phenotypes and physiology of neurons and glia is fundamental to improve our understanding of disease mechanisms in the brain and shape the design of future therapeutic interventions.Here we describe a protocol for semiautomated videomicroscopy analysis of cultured brain cells, maintained under suitable and controlled conditions. Through real-time monitoring of basic cell phenotypes (such as proliferation, cell morphology, differentiation, and migration) this method provides an accurate primary assessment of the impact of the repeat expansion on the physiology of neurons and glia. The versatility of the system, the automated image acquisition and the semiautomated processing of the data collected allow rapid phenotypic analysis of individual cell types, as well as the investigation of cell-cell interactions. The stability of the acquisition system provides reproducible and robust results. The raw data can be easily exported to other software to perform more sophisticated imaging analysis and statistical tests. In summary, the methods described offer versatile, reproducible, and time-effective means to dissect the impact of the repeat expansion on different brain cell types and on intercellular interactions.
Collapse
Affiliation(s)
- Sandra O Braz
- Laboratory CTGDM, Inserm UMR1163, Paris, France.,Institut Imagine, Université Paris Descartes- Sorbonne Paris Cité, Paris, France
| | - Diana M Dinca
- Laboratory CTGDM, Inserm UMR1163, Paris, France.,Institut Imagine, Université Paris Descartes- Sorbonne Paris Cité, Paris, France
| | - Geneviève Gourdon
- Laboratory CTGDM, Inserm UMR1163, Paris, France.,Institut Imagine, Université Paris Descartes- Sorbonne Paris Cité, Paris, France.,Centre de Recherche en Myologie (CRM), Inserm UMRS974, Association Institut de Myologie, Sorbonne Université, Paris, France
| | - Mário Gomes-Pereira
- Laboratory CTGDM, Inserm UMR1163, Paris, France. .,Institut Imagine, Université Paris Descartes- Sorbonne Paris Cité, Paris, France. .,Centre de Recherche en Myologie (CRM), Inserm UMRS974, Association Institut de Myologie, Sorbonne Université, Paris, France.
| |
Collapse
|
31
|
Casella C, Bourbon-Teles J, Bells S, Coulthard E, Parker GD, Rosser A, Jones DK, Metzler-Baddeley C. Drumming Motor Sequence Training Induces Apparent Myelin Remodelling in Huntington's Disease: A Longitudinal Diffusion MRI and Quantitative Magnetization Transfer Study. J Huntingtons Dis 2020; 9:303-320. [PMID: 32894249 PMCID: PMC7836062 DOI: 10.3233/jhd-200424] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Background: Impaired myelination may contribute to Huntington’s disease (HD) pathogenesis. Objective: This study assessed differences in white matter (WM) microstructure between HD patients and controls, and tested whether drumming training stimulates WM remodelling in HD. Furthermore, it examined whether training-induced microstructural changes are related to improvements in motor and cognitive function. Methods: Participants undertook two months of drumming exercises. Working memory and executive function were assessed before and post-training. Changes in WM microstructure were investigated with diffusion tensor magnetic resonance imaging (DT-MRI)-based metrics, the restricted diffusion signal fraction (Fr) from the composite hindered and restricted model of diffusion (CHARMED) and the macromolecular proton fraction (MPF) from quantitative magnetization transfer (qMT) imaging. WM pathways linking putamen and supplementary motor areas (SMA-Putamen), and three segments of the corpus callosum (CCI, CCII, CCIII) were studied using deterministic tractography. Baseline MPF differences between patients and controls were assessed with tract-based spatial statistics. Results: MPF was reduced in the mid-section of the CC in HD subjects at baseline, while a significantly greater change in MPF was detected in HD patients relative to controls in the CCII, CCIII, and the right SMA-putamen post-training. Further, although patients improved their drumming and executive function performance, such improvements did not correlate with microstructural changes. Increased MPF suggests training-induced myelin changes in HD. Conclusion: Though only preliminary and based on a small sample size, these results suggest that tailored behavioural stimulation may lead to neural benefits in early HD, that could be exploited for delaying disease progression.
Collapse
Affiliation(s)
- Chiara Casella
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Maindy Road, Cardiff, UK
| | - Jose Bourbon-Teles
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Maindy Road, Cardiff, UK
| | - Sonya Bells
- The Hospital for Sick Children, Neurosciences and Mental Health, Toronto, Canada
| | | | - Greg D Parker
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Maindy Road, Cardiff, UK
| | - Anne Rosser
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, UK.,Department of Neurology and Psychological Medicine, Hayden Ellis Building, Cardiff, UK
| | - Derek K Jones
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Maindy Road, Cardiff, UK.,Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Victoria, Australia
| | - Claudia Metzler-Baddeley
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Maindy Road, Cardiff, UK
| |
Collapse
|
32
|
Casella C, Lipp I, Rosser A, Jones DK, Metzler‐Baddeley C. A Critical Review of White Matter Changes in Huntington's Disease. Mov Disord 2020; 35:1302-1311. [PMID: 32537844 PMCID: PMC9393936 DOI: 10.1002/mds.28109] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/07/2020] [Accepted: 04/30/2020] [Indexed: 12/20/2022] Open
Abstract
Huntington's disease is a genetic neurodegenerative disorder. White matter alterations have recently been identified as a relevant pathophysiological feature of Huntington's disease, but their etiology and role in disease pathogenesis and progression remain unclear. Increasing evidence suggests that white matter changes in this disorder are attributed to alterations in myelin-associated biological processes. This review first discusses evidence from neurochemical studies lending support to the demyelination hypothesis of Huntington's disease, demonstrating aberrant myelination and changes in oligodendrocytes in the Huntington's brain. Next, evidence from neuroimaging studies is reviewed, the limitations of the described methodologies are discussed, and suggested interpretations of findings from published studies are challenged. Although our understanding of Huntington's associated pathological changes in the brain will increasingly rely on neuroimaging techniques, the shortcomings of these methodologies must not be forgotten. Advances in magnetic resonance imaging techniques and tissue modeling will enable a better in vivo, longitudinal characterization of the biological properties of white matter microstructure. This in turn will facilitate identification of disease-related biomarkers and the specification of outcome measures in clinical trials. © 2020 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Chiara Casella
- Cardiff University Brain Research Imaging CentreSchool of Psychology, Cardiff UniversityCardiffUnited Kingdom
| | - Ilona Lipp
- Department of NeurophysicsMax Planck Institute for Human Cognitive and Brain SciencesLeipzigGermany
| | - Anne Rosser
- School of BiosciencesCardiff UniversityCardiffUnited Kingdom
| | - Derek K Jones
- Cardiff University Brain Research Imaging CentreSchool of Psychology, Cardiff UniversityCardiffUnited Kingdom
- Mary MacKillop Institute for Health ResearchAustralian Catholic UniversityMelbourneVictoriaAustralia
| | - Claudia Metzler‐Baddeley
- Cardiff University Brain Research Imaging CentreSchool of Psychology, Cardiff UniversityCardiffUnited Kingdom
| |
Collapse
|
33
|
Cheong RY, Gabery S, Petersén Å. The Role of Hypothalamic Pathology for Non-Motor Features of Huntington's Disease. J Huntingtons Dis 2020; 8:375-391. [PMID: 31594240 PMCID: PMC6839491 DOI: 10.3233/jhd-190372] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Huntington’s disease (HD) is a fatal genetic neurodegenerative disorder. It has mainly been considered a movement disorder with cognitive symptoms and these features have been associated with pathology of the striatum and cerebral cortex. Importantly, individuals with the mutant huntingtin gene suffer from a spectrum of non-motor features often decades before the motor disorder manifests. These symptoms and signs include a range of psychiatric symptoms, sleep problems and metabolic changes with weight loss particularly in later stages. A higher body mass index at diagnosis is associated with slower disease progression. The common psychiatric symptom of apathy progresses with the disease. The fact that non-motor features are present early in the disease and that they show an association to disease progression suggest that unravelling the underlying neurobiological mechanisms may uncover novel targets for early disease intervention and better symptomatic treatment. The hypothalamus and the limbic system are important brain regions that regulate emotion, social cognition, sleep and metabolism. A number of studies using neuroimaging, postmortem human tissue and genetic manipulation in animal models of the disease has collectively shown that the hypothalamus and the limbic system are affected in HD. These findings include the loss of neuropeptide-expressing neurons such as orexin (hypocretin), oxytocin, vasopressin, somatostatin and VIP, and increased levels of SIRT1 in distinct nuclei of the hypothalamus. This review provides a summary of the results obtained so far and highlights the potential importance of these changes for the understanding of non-motor features in HD.
Collapse
Affiliation(s)
- Rachel Y Cheong
- Translational Neuroendocrine Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Sanaz Gabery
- Translational Neuroendocrine Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Åsa Petersén
- Translational Neuroendocrine Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
34
|
Wilton DK, Stevens B. The contribution of glial cells to Huntington's disease pathogenesis. Neurobiol Dis 2020; 143:104963. [PMID: 32593752 DOI: 10.1016/j.nbd.2020.104963] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 05/07/2020] [Accepted: 06/10/2020] [Indexed: 12/20/2022] Open
Abstract
Glial cells play critical roles in the normal development and function of neural circuits, but in many neurodegenerative diseases, they become dysregulated and may contribute to the development of brain pathology. In Huntington's disease (HD), glial cells both lose normal functions and gain neuropathic phenotypes. In addition, cell-autonomous dysfunction elicited by mutant huntingtin (mHTT) expression in specific glial cell types is sufficient to induce both pathology and Huntington's disease-related impairments in motor and cognitive performance, suggesting that these cells may drive the development of certain aspects of Huntington's disease pathogenesis. In support of this imaging studies in pre-symptomatic HD patients and work on mouse models have suggested that glial cell dysfunction occurs at a very early stage of the disease, prior to the onset of motor and cognitive deficits. Furthermore, selectively ablating mHTT from specific glial cells or correcting for HD-induced changes in their transcriptional profile rescues some HD-related phenotypes, demonstrating the potential of targeting these cells for therapeutic intervention. Here we review emerging research focused on understanding the involvement of different glial cell types in specific aspects of HD pathogenesis. This work is providing new insight into how HD impacts biological functions of glial cells in the healthy brain as well as how HD induced dysfunction in these cells might change the way they integrate into biological circuits.
Collapse
Affiliation(s)
- Daniel K Wilton
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Beth Stevens
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Stanley Center, Broad Institute, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
35
|
Jiang M, Zhang X, Liu H, LeBron J, Alexandris A, Peng Q, Gu H, Yang F, Li Y, Wang R, Hou Z, Arbez N, Ren Q, Dong JL, Whela E, Wang R, Ratovitski T, Troncoso JC, Mori S, Ross CA, Lim J, Duan W. Nemo-like kinase reduces mutant huntingtin levels and mitigates Huntington's disease. Hum Mol Genet 2020; 29:1340-1352. [PMID: 32242231 PMCID: PMC7254850 DOI: 10.1093/hmg/ddaa061] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/15/2020] [Accepted: 03/30/2020] [Indexed: 11/12/2022] Open
Abstract
Nemo-like kinase (NLK), an evolutionarily conserved serine/threonine kinase, is highly expressed in the brain, but its function in the adult brain remains not well understood. In this study, we identify NLK as an interactor of huntingtin protein (HTT). We report that NLK levels are significantly decreased in HD human brain and HD models. Importantly, overexpression of NLK in the striatum attenuates brain atrophy, preserves striatal DARPP32 levels and reduces mutant HTT (mHTT) aggregation in HD mice. In contrast, genetic reduction of NLK exacerbates brain atrophy and loss of DARPP32 in HD mice. Moreover, we demonstrate that NLK lowers mHTT levels in a kinase activity-dependent manner, while having no significant effect on normal HTT protein levels in mouse striatal cells, human cells and HD mouse models. The NLK-mediated lowering of mHTT is associated with enhanced phosphorylation of mHTT. Phosphorylation defective mutation of serine at amino acid 120 (S120) abolishes the mHTT-lowering effect of NLK, suggesting that S120 phosphorylation is an important step in the NLK-mediated lowering of mHTT. A further mechanistic study suggests that NLK promotes mHTT ubiquitination and degradation via the proteasome pathway. Taken together, our results indicate a protective role of NLK in HD and reveal a new molecular target to reduce mHTT levels.
Collapse
Affiliation(s)
- Mali Jiang
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xiaoyan Zhang
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hongshuai Liu
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jared LeBron
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Athanasios Alexandris
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Qi Peng
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hao Gu
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Fanghan Yang
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yuchen Li
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ruiling Wang
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zhipeng Hou
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nicolas Arbez
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Qianwei Ren
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jen-Li Dong
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Emma Whela
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ronald Wang
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tamara Ratovitski
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Juan C Troncoso
- Division of Neuropathology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Susumu Mori
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christopher A Ross
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Janghoo Lim
- Departments of Genetics and of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| | - Wenzhen Duan
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
36
|
Simanaviciute U, Ahmed J, Brown RE, Connor-Robson N, Farr TD, Fertan E, Gambles N, Garland H, Morton AJ, Staiger JF, Skillings EA, Trueman RC, Wade-Martins R, Wood NI, Wong AA, Grant RA. Recommendations for measuring whisker movements and locomotion in mice with sensory, motor and cognitive deficits. J Neurosci Methods 2020; 331:108532. [PMID: 31785300 DOI: 10.1016/j.jneumeth.2019.108532] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 10/30/2019] [Accepted: 11/25/2019] [Indexed: 01/02/2023]
Abstract
BACKGROUND Previous studies have measured whisker movements and locomotion to characterise mouse models of neurodegenerative disease. However, these studies have always been completed in isolation, and do not involve standardized procedures for comparisons across multiple mouse models and background strains. NEW METHOD We present a standard method for conducting whisker movement and locomotion studies, by carrying out qualitative scoring and quantitative measurement of whisker movements from high-speed video footage of mouse models of Amyotrophic Lateral Sclerosis, Huntington's disease, Parkinson's disease, Alzheimer's disease, Cerebellar Ataxia, Somatosensory Cortex Development and Ischemic stroke. RESULTS Sex, background strain, source breeder and genotype all affected whisker movements. All mouse models, apart from Parkinson's disease, revealed differences in whisker movements during locomotion. R6/2 CAG250 Huntington's disease mice had the strongest behavioural phenotype. Robo3R3-5-CKO and RIM-DKOSert mouse models have abnormal somatosensory cortex development and revealed significant changes in whisker movements during object exploration. COMPARISON WITH EXISTING METHOD(S) Our results have good agreement with past studies, which indicates the robustness and reliability of measuring whisking. We recommend that differences in whisker movements of mice with motor deficits can be captured in open field arenas, but that mice with impairments to sensory or cognitive functioning should also be filmed investigating objects. Scoring clips qualitatively before tracking will help to structure later analyses. CONCLUSIONS Studying whisker movements provides a quantitative measure of sensing, motor control and exploration. However, the effect of background strain, sex and age on whisker movements needs to be better understood.
Collapse
Affiliation(s)
- Ugne Simanaviciute
- Department of Natural Sciences, Manchester Metropolitan University, Manchester, M1 5GD, UK; School of Biological Sciences, Manchester University, Manchester, M13 9PL, UK
| | - Jewel Ahmed
- School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Richard E Brown
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, B3H 4R2, Canada
| | - Natalie Connor-Robson
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK
| | - Tracy D Farr
- School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Emre Fertan
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, B3H 4R2, Canada
| | - Nikki Gambles
- Department of Natural Sciences, Manchester Metropolitan University, Manchester, M1 5GD, UK; Public Health Institute, Liverpool John Moores University, Liverpool, L2 2QP, UK
| | - Huw Garland
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3DY, UK
| | - A Jennifer Morton
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3DY, UK
| | - Jochen F Staiger
- Institute for Neuroanatomy, University Medical Center, Göttingen, 37075, Germany
| | - Elizabeth A Skillings
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3DY, UK
| | - Rebecca C Trueman
- School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Richard Wade-Martins
- Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK
| | - Nigel I Wood
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3DY, UK
| | - Aimee A Wong
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, B3H 4R2, Canada
| | - Robyn A Grant
- Department of Natural Sciences, Manchester Metropolitan University, Manchester, M1 5GD, UK.
| |
Collapse
|
37
|
Radulescu CI, Garcia-Miralles M, Sidik H, Bardile CF, Yusof NABM, Lee HU, Ho EXP, Chu CW, Layton E, Low D, De Sessions PF, Pettersson S, Ginhoux F, Pouladi MA. Reprint of: Manipulation of microbiota reveals altered callosal myelination and white matter plasticity in a model of Huntington disease. Neurobiol Dis 2020; 135:104744. [PMID: 31931139 DOI: 10.1016/j.nbd.2020.104744] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 02/02/2019] [Accepted: 02/20/2019] [Indexed: 12/19/2022] Open
Abstract
Structural and molecular myelination deficits represent early pathological features of Huntington disease (HD). Recent evidence from germ-free (GF) animals suggests a role for microbiota-gut-brain bidirectional communication in the regulation of myelination. In this study, we aimed to investigate the impact of microbiota on myelin plasticity and oligodendroglial population dynamics in the mixed-sex BACHD mouse model of HD. Ultrastructural analysis of myelin in the corpus callosum revealed alterations of myelin thickness in BACHD GF compared to specific-pathogen free (SPF) mice, whereas no differences were observed between wild-type (WT) groups. In contrast, myelin compaction was altered in all groups when compared to WT SPF animals. Levels of myelin-related proteins were generally reduced, and the number of mature oligodendrocytes was decreased in the prefrontal cortex under GF compared to SPF conditions, regardless of genotype. Minor differences in commensal bacteria at the family and genera levels were found in the gut microbiota of BACHD and WT animals housed in standard living conditions. Our findings indicate complex effects of a germ-free status on myelin-related characteristics, and highlight the adaptive properties of myelination as a result of environmental manipulation.
Collapse
Affiliation(s)
- Carola I Radulescu
- Translational Laboratory in Genetic Medicine (TLGM), Agency for Science, Technology and Research (A*STAR), 138648, Singapore; Department of Psychology, The University of Sheffield, S1 2LT, UK
| | - Marta Garcia-Miralles
- Translational Laboratory in Genetic Medicine (TLGM), Agency for Science, Technology and Research (A*STAR), 138648, Singapore
| | - Harwin Sidik
- Translational Laboratory in Genetic Medicine (TLGM), Agency for Science, Technology and Research (A*STAR), 138648, Singapore
| | - Costanza Ferrari Bardile
- Translational Laboratory in Genetic Medicine (TLGM), Agency for Science, Technology and Research (A*STAR), 138648, Singapore
| | - Nur Amirah Binte Mohammad Yusof
- Translational Laboratory in Genetic Medicine (TLGM), Agency for Science, Technology and Research (A*STAR), 138648, Singapore
| | - Hae Ung Lee
- Lee Kong Chian School of Medicine, Nanyang Technological University, 637551, Singapore
| | - Eliza Xin Pei Ho
- GIS Efficient Rapid Microbial Sequencing, Genome Institute of Singapore, A*STAR, 138672, Singapore
| | - Collins Wenhan Chu
- GIS Efficient Rapid Microbial Sequencing, Genome Institute of Singapore, A*STAR, 138672, Singapore
| | - Emma Layton
- GIS Efficient Rapid Microbial Sequencing, Genome Institute of Singapore, A*STAR, 138672, Singapore
| | - Donovan Low
- Singapore Immunology Network, A*STAR, 138648, Singapore
| | - Paola Florez De Sessions
- GIS Efficient Rapid Microbial Sequencing, Genome Institute of Singapore, A*STAR, 138672, Singapore
| | - Sven Pettersson
- Lee Kong Chian School of Medicine, Nanyang Technological University, 637551, Singapore; Singapore Centre for Environmental Life Sciences Engineering, 60 Nanyang Drive, 637551, Singapore
| | | | - Mahmoud A Pouladi
- Translational Laboratory in Genetic Medicine (TLGM), Agency for Science, Technology and Research (A*STAR), 138648, Singapore; Department of Medicine, National University of Singapore, 117597, Singapore; Department of Physiology, National University of Singapore, 117597, Singapore.
| |
Collapse
|
38
|
Creus-Muncunill J, Ehrlich ME. Cell-Autonomous and Non-cell-Autonomous Pathogenic Mechanisms in Huntington's Disease: Insights from In Vitro and In Vivo Models. Neurotherapeutics 2019; 16:957-978. [PMID: 31529216 PMCID: PMC6985401 DOI: 10.1007/s13311-019-00782-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Huntington's disease (HD) is an autosomal dominant disorder caused by an expansion in the trinucleotide CAG repeat in exon-1 in the huntingtin gene, located on chromosome 4. When the number of trinucleotide CAG exceeds 40 repeats, disease invariably is manifested, characterized by motor, cognitive, and psychiatric symptoms. The huntingtin (Htt) protein and its mutant form (mutant huntingtin, mHtt) are ubiquitously expressed but although multiple brain regions are affected, the most vulnerable brain region is the striatum. Striatal medium-sized spiny neurons (MSNs) preferentially degenerate, followed by the cortical pyramidal neurons located in layers V and VI. Proposed HD pathogenic mechanisms include, but are not restricted to, excitotoxicity, neurotrophic support deficits, collapse of the protein degradation mechanisms, mitochondrial dysfunction, transcriptional alterations, and disorders of myelin. Studies performed in cell type-specific and regionally selective HD mouse models implicate both MSN cell-autonomous properties and cell-cell interactions, particularly corticostriatal but also with non-neuronal cell types. Here, we review the intrinsic properties of MSNs that contribute to their selective vulnerability and in addition, we discuss how astrocytes, microglia, and oligodendrocytes, together with aberrant corticostriatal connectivity, contribute to HD pathophysiology. In addition, mHtt causes cell-autonomous dysfunction in cell types other than MSNs. These findings have implications in terms of therapeutic strategies aimed at preventing neuronal dysfunction and degeneration.
Collapse
Affiliation(s)
- Jordi Creus-Muncunill
- Department of Neurology, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, NY, 10029, USA
| | - Michelle E Ehrlich
- Department of Neurology, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, NY, 10029, USA.
| |
Collapse
|
39
|
Naphade S, Tshilenge KT, Ellerby LM. Modeling Polyglutamine Expansion Diseases with Induced Pluripotent Stem Cells. Neurotherapeutics 2019; 16:979-998. [PMID: 31792895 PMCID: PMC6985408 DOI: 10.1007/s13311-019-00810-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Polyglutamine expansion disorders, which include Huntington's disease, have expanded CAG repeats that result in polyglutamine expansions in affected proteins. How this specific feature leads to distinct neuropathies in 11 different diseases is a fascinating area of investigation. Most proteins affected by polyglutamine expansions are ubiquitously expressed, yet their mechanisms of selective neurotoxicity are unknown. Induced pluripotent stem cells have emerged as a valuable tool to model diseases, understand molecular mechanisms, and generate relevant human neural and glia subtypes, cocultures, and organoids. Ideally, this tool will generate specific neuronal populations that faithfully recapitulate specific polyglutamine expansion disorder phenotypes and mimic the selective vulnerability of a given disease. Here, we review how induced pluripotent technology is used to understand the effects of the disease-causing polyglutamine protein on cell function, identify new therapeutic targets, and determine how polyglutamine expansion affects human neurodevelopment and disease. We will discuss ongoing challenges and limitations in our use of induced pluripotent stem cells to model polyglutamine expansion diseases.
Collapse
Affiliation(s)
- Swati Naphade
- Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA, 94945, USA
| | | | - Lisa M Ellerby
- Buck Institute for Research on Aging, 8001 Redwood Blvd, Novato, CA, 94945, USA.
| |
Collapse
|
40
|
Radulescu CI, Garcia-Miralles M, Sidik H, Bardile CF, Yusof NABM, Lee HU, Ho EXP, Chu CW, Layton E, Low D, De Sessions PF, Pettersson S, Ginhoux F, Pouladi MA. Manipulation of microbiota reveals altered callosal myelination and white matter plasticity in a model of Huntington disease. Neurobiol Dis 2019; 127:65-75. [DOI: 10.1016/j.nbd.2019.02.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 02/02/2019] [Accepted: 02/20/2019] [Indexed: 01/08/2023] Open
|
41
|
Tereshchenko A, Magnotta V, Epping E, Mathews K, Espe-Pfeifer P, Martin E, Dawson J, Duan W, Nopoulos P. Brain structure in juvenile-onset Huntington disease. Neurology 2019; 92:e1939-e1947. [PMID: 30971481 PMCID: PMC6511077 DOI: 10.1212/wnl.0000000000007355] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 12/27/2018] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To assess brain morphometry in a sample of patients with juvenile-onset Huntington disease (JOHD) and several mouse models of Huntington disease (HD) that likely represent the human JOHD phenotype. METHODS Despite sharing the mutation in the Huntingtin gene, adult-onset HD characteristically presents as a hyperkinetic motor disorder, while JOHD typically presents as a hypokinetic motor disease. The University of Iowa Kids-JHD program enrolls individuals 5 to 25 years of age who have already received the clinical diagnosis. A total of 19 children with juvenile HD (JHD) (mean CAG = 72) were studied. Patients with JHD were compared to healthy controls (n = 234) using a cross-sectional study design. Volumetric data from structural MRI was compared between groups. In addition, we used the same procedure to evaluate brain morphology of R6/2, zQ175, HdhQ250 HD mice models. RESULTS Participants with JHD had substantially reduced intracranial volumes. After controlling for the small intracranial volume size, the volumes of subcortical regions (caudate, putamen, globus pallidus, and thalamus) and of cortical white matter were significantly decreased in patients with JHD. However, the cerebellum was proportionately enlarged in the JHD sample. The cerebral cortex was largely unaffected. Likewise, HD mice had a lower volume of striatum and a higher volume of cerebellum, mirroring the human MRI results. CONCLUSIONS The primary pathology of JOHD extends beyond changes in the striatal volume. Brain morphology in both mice and human patients with JHD shows proportional cerebellar enlargement. This pattern of brain changes may explain the unique picture of hypokinetic motor symptoms in JHD, which is not seen in the hyperkinetic chorea-like phenotype of adult-onset HD.
Collapse
Affiliation(s)
- Alexander Tereshchenko
- From the Departments of Psychiatry (A.T., E.E., V.M., P.E.-P., E.M.), Radiology (V.M.), Neurology (K.M., P.E.-P.), and Pediatrics (K.M.), University of Iowa Hospitals and Clinics, Iowa City; Department of Biostatistics (J.D.), University of Iowa College of Public Health, Iowa City; and Department of Psychiatry and Behavioral Sciences (W.D.), Johns Hopkins University, Baltimore, MD
| | - Vincent Magnotta
- From the Departments of Psychiatry (A.T., E.E., V.M., P.E.-P., E.M.), Radiology (V.M.), Neurology (K.M., P.E.-P.), and Pediatrics (K.M.), University of Iowa Hospitals and Clinics, Iowa City; Department of Biostatistics (J.D.), University of Iowa College of Public Health, Iowa City; and Department of Psychiatry and Behavioral Sciences (W.D.), Johns Hopkins University, Baltimore, MD
| | - Eric Epping
- From the Departments of Psychiatry (A.T., E.E., V.M., P.E.-P., E.M.), Radiology (V.M.), Neurology (K.M., P.E.-P.), and Pediatrics (K.M.), University of Iowa Hospitals and Clinics, Iowa City; Department of Biostatistics (J.D.), University of Iowa College of Public Health, Iowa City; and Department of Psychiatry and Behavioral Sciences (W.D.), Johns Hopkins University, Baltimore, MD
| | - Katherine Mathews
- From the Departments of Psychiatry (A.T., E.E., V.M., P.E.-P., E.M.), Radiology (V.M.), Neurology (K.M., P.E.-P.), and Pediatrics (K.M.), University of Iowa Hospitals and Clinics, Iowa City; Department of Biostatistics (J.D.), University of Iowa College of Public Health, Iowa City; and Department of Psychiatry and Behavioral Sciences (W.D.), Johns Hopkins University, Baltimore, MD
| | - Patricia Espe-Pfeifer
- From the Departments of Psychiatry (A.T., E.E., V.M., P.E.-P., E.M.), Radiology (V.M.), Neurology (K.M., P.E.-P.), and Pediatrics (K.M.), University of Iowa Hospitals and Clinics, Iowa City; Department of Biostatistics (J.D.), University of Iowa College of Public Health, Iowa City; and Department of Psychiatry and Behavioral Sciences (W.D.), Johns Hopkins University, Baltimore, MD
| | - Erin Martin
- From the Departments of Psychiatry (A.T., E.E., V.M., P.E.-P., E.M.), Radiology (V.M.), Neurology (K.M., P.E.-P.), and Pediatrics (K.M.), University of Iowa Hospitals and Clinics, Iowa City; Department of Biostatistics (J.D.), University of Iowa College of Public Health, Iowa City; and Department of Psychiatry and Behavioral Sciences (W.D.), Johns Hopkins University, Baltimore, MD
| | - Jeffrey Dawson
- From the Departments of Psychiatry (A.T., E.E., V.M., P.E.-P., E.M.), Radiology (V.M.), Neurology (K.M., P.E.-P.), and Pediatrics (K.M.), University of Iowa Hospitals and Clinics, Iowa City; Department of Biostatistics (J.D.), University of Iowa College of Public Health, Iowa City; and Department of Psychiatry and Behavioral Sciences (W.D.), Johns Hopkins University, Baltimore, MD
| | - Wenzhen Duan
- From the Departments of Psychiatry (A.T., E.E., V.M., P.E.-P., E.M.), Radiology (V.M.), Neurology (K.M., P.E.-P.), and Pediatrics (K.M.), University of Iowa Hospitals and Clinics, Iowa City; Department of Biostatistics (J.D.), University of Iowa College of Public Health, Iowa City; and Department of Psychiatry and Behavioral Sciences (W.D.), Johns Hopkins University, Baltimore, MD
| | - Peg Nopoulos
- From the Departments of Psychiatry (A.T., E.E., V.M., P.E.-P., E.M.), Radiology (V.M.), Neurology (K.M., P.E.-P.), and Pediatrics (K.M.), University of Iowa Hospitals and Clinics, Iowa City; Department of Biostatistics (J.D.), University of Iowa College of Public Health, Iowa City; and Department of Psychiatry and Behavioral Sciences (W.D.), Johns Hopkins University, Baltimore, MD.
| |
Collapse
|
42
|
Rangel-Barajas C, Rebec GV. Overview of Huntington's Disease Models: Neuropathological, Molecular, and Behavioral Differences. ACTA ACUST UNITED AC 2019; 83:e47. [PMID: 30040221 DOI: 10.1002/cpns.47] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Transgenic mouse models of Huntington's disease (HD), a neurodegenerative condition caused by a single gene mutation, have been transformative in their ability to reveal the molecular processes and pathophysiological mechanisms underlying the HD behavioral phenotype. Three model categories have been generated depending on the genetic context in which the mutation is expressed: truncated, full-length, and knock-in. No single model, however, broadly replicates the behavioral symptoms and massive neuronal loss that occur in human patients. The disparity between model and patient requires careful consideration of what each model has to offer when testing potential treatments. Although the translation of animal data to the clinic has been limited, each model can make unique contributions toward an improved understanding of the neurobehavioral underpinnings of HD. Thus, conclusions based on data obtained from more than one model are likely to have the most success in the search for new treatment targets. © 2018 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Claudia Rangel-Barajas
- Program in Neuroscience, Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana
| | - George V Rebec
- Program in Neuroscience, Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana
| |
Collapse
|
43
|
Teo RTY, Ferrari Bardile C, Tay YL, Yusof NABM, Kreidy CA, Tan LJ, Pouladi MA. Impaired Remyelination in a Mouse Model of Huntington Disease. Mol Neurobiol 2019; 56:6873-6882. [PMID: 30937636 DOI: 10.1007/s12035-019-1579-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 03/20/2019] [Indexed: 01/26/2023]
Abstract
White matter (WM) abnormalities are a well-established feature of Huntington disease (HD), although their nature is not fully understood. Here, we asked whether remyelination as a measure of WM plasticity is impaired in a model of HD. Using the cuprizone assay, we examined demyelination and remyelination responses in YAC128 HD mice. Treatment with 0.2% cuprizone (CPZ) for 6 weeks resulted in significant reduction in mature (GSTπ-positive) oligodendrocyte counts and FluoroMyelin staining in the corpus callosum, leading to similar demyelination states in YAC128 and wild-type (WT) mice. Six weeks following cessation of CPZ, we observed robust remyelination in WT mice as indicated by an increase in mature oligodendrocyte counts and FluoroMyelin staining. In contrast, YAC128 mice exhibited an impaired remyelination response. The increase in mature oligodendrocyte counts in YAC128 HD mice following CPZ cessation was lower than that of WT. Furthermore, there was no increase in FluoroMyelin staining compared to the demyelinated state in YAC128 mice. We confirmed these findings using electron microscopy where the CPZ-induced reduction in myelinated axons was reversed following CPZ cessation in WT but not YAC128 mice. Our findings demonstrate that remyelination is impaired in YAC128 mice and suggest that WM plasticity may be compromised in HD.
Collapse
Affiliation(s)
- Roy Tang Yi Teo
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research, Singapore (A*STAR), 8A Biomedical Grove, Immunos, Level 5, Singapore, 138648, Singapore
| | - Costanza Ferrari Bardile
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research, Singapore (A*STAR), 8A Biomedical Grove, Immunos, Level 5, Singapore, 138648, Singapore
| | - Yi Lin Tay
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research, Singapore (A*STAR), 8A Biomedical Grove, Immunos, Level 5, Singapore, 138648, Singapore
| | - Nur Amirah Binte Mohammad Yusof
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research, Singapore (A*STAR), 8A Biomedical Grove, Immunos, Level 5, Singapore, 138648, Singapore
| | - Charbel A Kreidy
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research, Singapore (A*STAR), 8A Biomedical Grove, Immunos, Level 5, Singapore, 138648, Singapore
| | - Liang Juin Tan
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research, Singapore (A*STAR), 8A Biomedical Grove, Immunos, Level 5, Singapore, 138648, Singapore
| | - Mahmoud A Pouladi
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research, Singapore (A*STAR), 8A Biomedical Grove, Immunos, Level 5, Singapore, 138648, Singapore.
- Department of Medicine, National University of Singapore, Singapore, 117597, Singapore.
- Department of Physiology, National University of Singapore, Singapore, 117597, Singapore.
| |
Collapse
|
44
|
Colwell CS, Ghiani CA. Potential Circadian Rhythms in Oligodendrocytes? Working Together Through Time. Neurochem Res 2019; 45:591-605. [PMID: 30906970 DOI: 10.1007/s11064-019-02778-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 03/14/2019] [Indexed: 12/15/2022]
Abstract
Oligodendrocytes (OL) are the only myelinating cells of the central nervous system thus interferences, either environmental or genetic, with their maturation or function have devastating consequences. Albeit so far neglected, one of the less appreciated, nevertheless possible, regulators of OL maturation and function is the circadian cycle. Yet, disruptions in these rhythms are unfortunately becoming a common "disorder" in the today's world. The temporal patterning of behaviour and physiology is controlled by a circadian timing system based in the anterior hypothalamus. At the molecular level, circadian rhythms are generated by a transcriptional/translational feedback system that regulates transcription and has a major impact on cellular function(s). Fundamental cellular properties/functions in most cell types vary with the daily circadian cycle: OL are unlikely an exception! To be clear, the presence of circadian oscillators or the cell-specific function(s) of the circadian clock in OL has yet to be defined. Furthermore, we wish to entertain the idea of links between the "thin" evidence on OL intrinsic circadian rhythms and their interjection(s) at different stages of lineage progression as well as in supporting/regulating OL crucial function: myelination. Individuals with intellectual and developmental syndromes as well as neurodegenerative diseases present with a disrupted sleep/wake cycle; hence, we raise the possibility that these disturbances in timing can contribute to the loss of white matter observed in these disorders. Preclinical and clinical work in this area is needed for a better understanding of how circadian rhythms influence OL maturation and function(s), to aid the development of new therapeutic strategies and standards of care for these patients.
Collapse
Affiliation(s)
- Christopher S Colwell
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA.,Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Cristina A Ghiani
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA. .,Department of Pathology & Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA. .,Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
45
|
Osipovitch M, Asenjo Martinez A, Mariani JN, Cornwell A, Dhaliwal S, Zou L, Chandler-Militello D, Wang S, Li X, Benraiss SJ, Agate R, Lampp A, Benraiss A, Windrem MS, Goldman SA. Human ESC-Derived Chimeric Mouse Models of Huntington's Disease Reveal Cell-Intrinsic Defects in Glial Progenitor Cell Differentiation. Cell Stem Cell 2018; 24:107-122.e7. [PMID: 30554964 DOI: 10.1016/j.stem.2018.11.010] [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: 11/18/2017] [Revised: 09/04/2018] [Accepted: 11/07/2018] [Indexed: 12/31/2022]
Abstract
Huntington's disease (HD) is characterized by hypomyelination and neuronal loss. To assess the basis for myelin loss in HD, we generated bipotential glial progenitor cells (GPCs) from human embryonic stem cells (hESCs) derived from mutant Huntingtin (mHTT) embryos or normal controls and performed RNA sequencing (RNA-seq) to assess mHTT-dependent changes in gene expression. In human GPCs (hGPCs) derived from 3 mHTT hESC lines, transcription factors associated with glial differentiation and myelin synthesis were sharply downregulated relative to normal hESC GPCs; NKX2.2, OLIG2, SOX10, MYRF, and their downstream targets were all suppressed. Accordingly, when mHTT hGPCs were transplanted into hypomyelinated shiverer mice, the resultant glial chimeras were hypomyelinated; this defect could be rescued by forced expression of SOX10 and MYRF by mHTT hGPCs. The mHTT hGPCs also manifested impaired astrocytic differentiation and developed abnormal fiber architecture. White matter involution in HD is thus a product of the cell-autonomous, mHTT-dependent suppression of glial differentiation.
Collapse
Affiliation(s)
- Mikhail Osipovitch
- Center for Translational Neuromedicine, University of Copenhagen Faculty of Health and Medical Science, 2200 Copenhagen N, Denmark
| | - Andrea Asenjo Martinez
- Center for Translational Neuromedicine, University of Copenhagen Faculty of Health and Medical Science, 2200 Copenhagen N, Denmark
| | - John N Mariani
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 10021, USA
| | - Adam Cornwell
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 10021, USA
| | - Simrat Dhaliwal
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 10021, USA
| | - Lisa Zou
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 10021, USA
| | - Devin Chandler-Militello
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 10021, USA
| | - Su Wang
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 10021, USA
| | - Xiaojie Li
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 10021, USA
| | - Sarah-Jehanne Benraiss
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 10021, USA
| | - Robert Agate
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 10021, USA
| | - Andrea Lampp
- Center for Translational Neuromedicine, University of Copenhagen Faculty of Health and Medical Science, 2200 Copenhagen N, Denmark
| | - Abdellatif Benraiss
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 10021, USA
| | - Martha S Windrem
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 10021, USA
| | - Steven A Goldman
- Center for Translational Neuromedicine, University of Copenhagen Faculty of Health and Medical Science, 2200 Copenhagen N, Denmark; Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 10021, USA; Neuroscience Center, Rigshospitalet, Copenhagen, Denmark.
| |
Collapse
|
46
|
Zhao Y, Sun X, Qi X. Inhibition of Drp1 hyperactivation reduces neuropathology and behavioral deficits in zQ175 knock-in mouse model of Huntington's disease. Biochem Biophys Res Commun 2018; 507:319-323. [PMID: 30449600 DOI: 10.1016/j.bbrc.2018.11.031] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 10/31/2018] [Accepted: 11/05/2018] [Indexed: 01/13/2023]
Abstract
Mitochondrial dysfunction manifests in the pathogenesis of Huntington's disease (HD), a fatal and inherited neurodegenerative disease. Dynamin-related protein 1 (Drp1) is the primary component of mitochondrial fission and becomes hyperactivated in various models of HD. We previously reported that inhibition of Drp1 hyperactivation by P110, a rationally designed peptide inhibitor of Drp1-Fis1 interaction, is protective in the HD R6/2 mouse model, which expresses a fragment of mutant Huntingtin (mHtt). In this study, we expand our work to test the effect of P110 treatment in HD knock-in (zQ175 KI) mice that express full-length mtHtt and exhibit progressive disease symptoms, reminiscent of human HD. We find that subcutaneously sustained treatment with P110 reduces movement deficits of mice. Moreover, the treatment attenuates striatal neuronal loss, microglial hyperactivity and white matter disorganization in zQ175 KI mice. These findings provide an additional line of evidence that inhibition of Drp1 hyperactivation is sufficient to reduce HD-associated neuropathology and behavioral deficits. We propose that manipulation of Drp1 hyperactivation might be a useful strategy to develop therapeutics for treating HD.
Collapse
Affiliation(s)
- Yuanyuan Zhao
- Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Xiaoyan Sun
- Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Xin Qi
- Department of Physiology & Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
| |
Collapse
|
47
|
Laquinimod Treatment Improves Myelination Deficits at the Transcriptional and Ultrastructural Levels in the YAC128 Mouse Model of Huntington Disease. Mol Neurobiol 2018; 56:4464-4478. [DOI: 10.1007/s12035-018-1393-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 10/11/2018] [Indexed: 10/28/2022]
|
48
|
Early postnatal behavioral, cellular, and molecular changes in models of Huntington disease are reversible by HDAC inhibition. Proc Natl Acad Sci U S A 2018; 115:E8765-E8774. [PMID: 30150378 PMCID: PMC6140493 DOI: 10.1073/pnas.1807962115] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
In Huntington disease (HD) gene carriers the disease-causing mutant Huntingtin (mHTT) is already present during early developmental stages, but, surprisingly, HD patients develop clinical symptoms only many years later. While a developmental role of Huntingtin has been described, so far new therapeutic approaches targeting those early neurodevelopmental processes are lacking. Here, we show that behavioral, cellular, and molecular changes associated with mHTT in the postnatal period of genetic animal models of HD can be reverted using low-dose treatment with a histone deacetylation inhibitor. Our findings support a neurodevelopmental basis for HD and provide proof of concept that pre-HD symptoms, including aberrant neuronal differentiation, are reversible by early therapeutic intervention in vivo. Huntington disease (HD) is an autosomal dominant neurodegenerative disorder caused by expanded CAG repeats in the huntingtin gene (HTT). Although mutant HTT is expressed during embryonic development and throughout life, clinical HD usually manifests later in adulthood. A number of studies document neurodevelopmental changes associated with mutant HTT, but whether these are reversible under therapy remains unclear. Here, we identify very early behavioral, molecular, and cellular changes in preweaning transgenic HD rats and mice. Reduced ultrasonic vocalization, loss of prepulse inhibition, and increased risk taking are accompanied by disturbances of dopaminergic regulation in vivo, reduced neuronal differentiation capacity in subventricular zone stem/progenitor cells, and impaired neuronal and oligodendrocyte differentiation of mouse embryo-derived neural stem cells in vitro. Interventional treatment of this early phenotype with the histone deacetylase inhibitor (HDACi) LBH589 led to significant improvement in behavioral changes and markers of dopaminergic neurotransmission and complete reversal of aberrant neuronal differentiation in vitro and in vivo. Our data support the notion that neurodevelopmental changes contribute to the prodromal phase of HD and that early, presymptomatic intervention using HDACi may represent a promising novel treatment approach for HD.
Collapse
|
49
|
Croce KR, Yamamoto A. A role for autophagy in Huntington's disease. Neurobiol Dis 2018; 122:16-22. [PMID: 30149183 DOI: 10.1016/j.nbd.2018.08.010] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 08/10/2018] [Accepted: 08/23/2018] [Indexed: 12/19/2022] Open
Abstract
The lysosome-mediated degradation pathway known as macroautophagy is the most versatile means through which cells can eliminate and recycle unwanted materials. Through both selective and non-selective means, macroautophagy can degrade a wide range of cargoes from bulk cytosol to organelles and aggregated proteins. Although studies of disorders such as Parkinson's disease and Amyotrophic Lateral Sclerosis suggest that autophagic and lysosomal dysfunction directly contributes to disease, this had not been the case for the polyglutamine disorder Huntington's disease (HD), for which there was little indication of a disruption in the autophagic-lysosomal system. This supported the possibility of targeting autophagy as a much needed therapeutic approach to combat this disease. Possibly challenging this view, however, are a recent set of studies suggesting that the protein affected in Huntington's disease, huntingtin, might mechanistically contribute to macroautophagy. In this review, we will explore how autophagy might impact or be impacted by HD pathogenesis, and whether a therapeutic approach centering on autophagy may be possible for this yet incurable disease.
Collapse
Affiliation(s)
- Katherine R Croce
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, United States
| | - Ai Yamamoto
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, United States; Department of Neurology, Columbia University, New York, NY 10032, United States.
| |
Collapse
|
50
|
Simmons DA. Modulating Neurotrophin Receptor Signaling as a Therapeutic Strategy for Huntington's Disease. J Huntingtons Dis 2018; 6:303-325. [PMID: 29254102 PMCID: PMC5757655 DOI: 10.3233/jhd-170275] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Huntington’s disease (HD) is an autosomal dominant neurodegenerative disorder caused by CAG repeat expansions in the IT15 gene which encodes the huntingtin (HTT) protein. Currently, no treatments capable of preventing or slowing disease progression exist. Disease modifying therapeutics for HD would be expected to target a comprehensive set of degenerative processes given the diverse mechanisms contributing to HD pathogenesis including neuroinflammation, excitotoxicity, and transcription dysregulation. A major contributor to HD-related degeneration is mutant HTT-induced loss of neurotrophic support. Thus, neurotrophin (NT) receptors have emerged as therapeutic targets in HD. The considerable overlap between NT signaling networks and those dysregulated by mutant HTT provides strong theoretical support for this approach. This review will focus on the contributions of disrupted NT signaling in HD-related neurodegeneration and how targeting NT receptors to augment pro-survival signaling and/or to inhibit degenerative signaling may combat HD pathologies. Therapeutic strategies involving NT delivery, peptidomimetics, and the targeting of specific NT receptors (e.g., Trks or p75NTR), particularly with small molecule ligands, are discussed.
Collapse
Affiliation(s)
- Danielle A Simmons
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|