1
|
Berg MJ, Veeranna, Rosa CM, Kumar A, Mohan PS, Stavrides P, Marchionini DM, Yang DS, Nixon RA. Pathobiology of the autophagy-lysosomal pathway in the Huntington's disease brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.29.596470. [PMID: 38854113 PMCID: PMC11160756 DOI: 10.1101/2024.05.29.596470] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Accumulated levels of mutant huntingtin protein (mHTT) and its fragments are considered contributors to the pathogenesis of Huntington's disease (HD). Although lowering mHTT by stimulating autophagy has been considered a possible therapeutic strategy, the role and competence of autophagy-lysosomal pathway (ALP) during HD progression in the human disease remains largely unknown. Here, we used multiplex confocal and ultrastructural immunocytochemical analyses of ALP functional markers in relation to mHTT aggresome pathology in striatum and the less affected cortex of HD brains staged from HD2 to HD4 by Vonsattel neuropathological criteria compared to controls. Immunolabeling revealed the localization of HTT/mHTT in ALP vesicular compartments labeled by autophagy-related adaptor proteins p62/SQSTM1 and ubiquitin, and cathepsin D (CTSD) as well as HTT-positive inclusions. Although comparatively normal at HD2, neurons at later HD stages exhibited progressive enlargement and clustering of CTSD-immunoreactive autolysosomes/lysosomes and, ultrastructurally, autophagic vacuole/lipofuscin granules accumulated progressively, more prominently in striatum than cortex. These changes were accompanied by rises in levels of HTT/mHTT and p62/SQSTM1, particularly their fragments, in striatum but not in the cortex, and by increases of LAMP1 and LAMP2 RNA and LAMP1 protein. Importantly, no blockage in autophagosome formation and autophagosome-lysosome fusion was detected, thus pinpointing autophagy substrate clearance deficits as a basis for autophagic flux declines. The findings collectively suggest that upregulated lysosomal biogenesis and preserved proteolysis maintain autophagic clearance in early-stage HD, but failure at advanced stages contributes to progressive HTT build-up and potential neurotoxicity. These findings support the prospect that ALP stimulation applied at early disease stages, when clearance machinery is fully competent, may have therapeutic benefits in HD patients.
Collapse
|
2
|
Herrera F, Branco-Santos J, Outeiro T. Threonine 3 regulates Serine 13/16 phosphorylation in the huntingtin exon 1. ACTA ACUST UNITED AC 2019. [DOI: 10.19185/matters.201905000005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
3
|
Branco-Santos J, Herrera F, Poças GM, Pires-Afonso Y, Giorgini F, Domingos PM, Outeiro TF. Protein phosphatase 1 regulates huntingtin exon 1 aggregation and toxicity. Hum Mol Genet 2017; 26:3763-3775. [DOI: 10.1093/hmg/ddx260] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 06/30/2017] [Indexed: 12/31/2022] Open
|
4
|
Putkhao K, Kocerha J, Cho IK, Yang J, Parnpai R, Chan AWS. Pathogenic cellular phenotypes are germline transmissible in a transgenic primate model of Huntington's disease. Stem Cells Dev 2013. [PMID: 23190281 DOI: 10.1089/scd.2012.0469] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
A transgenic primate model for Huntington's Disease (HD) first reported by our group that (HD monkeys) carry the mutant Huntingtin (HTT) gene with expanded polyglutamine (CAG) repeats and, develop chorea, dystonia, and other involuntary motor deficiencies similar to HD [ 1 ]. More recently, we have found that longitudinal magnetic resonance imaging of the HD monkey brain revealed significant atrophy in regions associated with cognitive deficits symptomatic in HD patients, providing the first animal model which replicates clinical phenotypes of diagnosed humans. Here we report germline transmission of the pathogenic mutant HTT in HD monkey by the production of embryos and subsequent derivation of HD monkey embryonic stem cells (rHD-ESCs) using HD monkey sperm. rHD-ESCs inherit mutant HTT and green fluorescent protein (GFP) genes through the gametes of HD monkey. rHD-ESCs express mutant HTT and form intranuclear inclusion, a classical cellular feature of HD. Notably, mosaicism of the pathogenic polyQ region in the sperm as well as derived ESCs were also observed, consistent with intraindividual and intergenerational reports of mosaic CAG repeats [ 2 , 3 ]and CAG expansion in HD patients [ 4-7 ]. The confirmation of transgene inheritability and development of pathogenic HD phenotype in derived rHD-ESCs reported in this study is a milestone in the pursuit of a transgenic primate model with inherited mutant HTT for development of novel disease biomarkers and therapeutics.
Collapse
|
5
|
Kumar R. Role of androgen receptor polyQ chain elongation in Kennedy's disease and use of natural osmolytes as potential therapeutic targets. IUBMB Life 2012; 64:879-84. [PMID: 23024039 DOI: 10.1002/iub.1088] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Accepted: 08/10/2012] [Indexed: 02/05/2023]
Abstract
Instability of CAG triplet repeat encoding polyglutamine (polyQ) stretches in the gene for target protein has been implicated as a putative mechanism in several inherited neurodegenerative diseases. Expansion of polyQ chain length in the androgen receptor (AR) causes spinal and bulbar muscular atrophy (SBMA) or Kennedy's disease. Although the mechanisms underlying gain-of-neurotoxic function are not completely understood, suggested pathological mechanisms of SBMA involve the formation of AR nuclear and cytoplasmic aggregates, a characteristic feature of patients with SBMA. The fact that certain AR coactivators are sequestered into the nuclear inclusions in SBMA possibly through protein-protein interactions supports the notion that AR transcriptional dysregulation may be a potential pathological mechanism leading to SBMA. AR conformational states associated with aberrant polyQ tract also modulate the interaction of AR with several coactivators. In many cases, such diseases can be treated through protein replacement therapy; however, because recombinant proteins do not cross the blood-brain barrier, the effectiveness of such therapies is limited in case of neurodegenerative diseases that warrant alternative therapeutic approaches. Among different approaches, inhibiting protein aggregation with small molecules that can stimulate protein folding and reverse aggregation are the most promising ones. Thus, naturally occurring osmolytes or "chemical chaperones" that can easily cross the blood-brain barrier and stabilize the functional form of a mutated protein by shifting the folding equilibrium away from degradation and/or aggregation is a useful therapeutic approach. In this review, we discuss the role of polyQ chain length extension in the pathophysiology of SBMA and the use of osmolytes as potential therapeutic tool.
Collapse
Affiliation(s)
- Raj Kumar
- Department of Basic Sciences, The Commonwealth Medical College, Scranton, PA 18509, USA.
| |
Collapse
|
6
|
Singer D, Zauner T, Genz M, Hoffmann R, Zuchner T. Synthesis of pathological and nonpathological human exon 1 huntingtin. J Pept Sci 2010; 16:358-63. [PMID: 20552561 DOI: 10.1002/psc.1252] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Huntington's disease (HD) is a neurodegenerative disorder that affects approximately 1 in 10 000 individuals. The underlying gene mutation was identified as a CAG-triplet repeat expansion in the gene huntingtin. The CAG sequence codes for glutamine, and in HD, an expansion of the polyglutamine (poly-Q) stretch above 35 glutamine residues results in pathogenicity. It has been demonstrated in various animal models that only the expression of exon 1 huntingtin, a 67-amino acid-long polypeptide plus a variable poly-Q stretch, is sufficient to cause full HD-like pathology. Therefore, a deeper understanding of exon 1 huntingtin, its structure, aggregation mechanism and interaction with other proteins is crucial for a better understanding of the disease. Here, we describe the synthesis of a 109-amino acid-long exon 1 huntingtin peptide including a poly-Q stretch of 42 glutamines. This microwave-assisted solid phase peptide synthesis resulted in milligram amounts of peptide with high purity. We also synthesized a nonpathogenic version of exon 1 huntingtin (90-amino acid long including a poly-Q stretch of 23 glutamine residues) using the same strategy. In circular dichroism spectroscopy, both polypeptides showed weak alpha-helical properties with the longer peptide showing a higher helical degree. These model peptides have great potential for further biomedical analyses, e.g. for large-scale pre-screenings for aggregation inhibitors, further structural analyses as well as protein-protein interaction studies.
Collapse
Affiliation(s)
- David Singer
- Institute of Bioanalytical Chemistry, Center for Biotechnology and Biomedicine, Leipzig University, Leipzig, Germany
| | | | | | | | | |
Collapse
|
7
|
Monkey hybrid stem cells develop cellular features of Huntington's disease. BMC Cell Biol 2010; 11:12. [PMID: 20132560 PMCID: PMC2833146 DOI: 10.1186/1471-2121-11-12] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2009] [Accepted: 02/05/2010] [Indexed: 12/31/2022] Open
Abstract
Background Pluripotent stem cells that are capable of differentiating into different cell types and develop robust hallmark cellular features are useful tools for clarifying the impact of developmental events on neurodegenerative diseases such as Huntington's disease. Additionally, a Huntington's cell model that develops robust pathological features of Huntington's disease would be valuable for drug discovery research. Results To test this hypothesis, a pluripotent Huntington's disease monkey hybrid cell line (TrES1) was established from a tetraploid Huntington's disease monkey blastocyst generated by the fusion of transgenic Huntington's monkey skin fibroblast and a wild-type non-transgenic monkey oocyte. The TrES1 developed key Huntington's disease cellular pathological features that paralleled neural development. It expressed mutant huntingtin and stem cell markers, was capable of differentiating to neural cells, and developed teratoma in severely compromised immune deficient (SCID) mice. Interestingly, the expression of mutant htt, the accumulation of oligomeric mutant htt and the formation of intranuclear inclusions paralleled neural development in vitro , and even mutant htt was ubiquitously expressed. This suggests the development of Huntington's disease cellular features is influenced by neural developmental events. Conclusions Huntington's disease cellular features is influenced by neural developmental events. These results are the first to demonstrate that a pluripotent stem cell line is able to mimic Huntington's disease progression that parallels neural development, which could be a useful cell model for investigating the developmental impact on Huntington's disease pathogenesis.
Collapse
|
8
|
|
9
|
Mihm MJ, Amann DM, Schanbacher BL, Altschuld RA, Bauer JA, Hoyt KR. Cardiac dysfunction in the R6/2 mouse model of Huntington's disease. Neurobiol Dis 2006; 25:297-308. [PMID: 17126554 PMCID: PMC1850107 DOI: 10.1016/j.nbd.2006.09.016] [Citation(s) in RCA: 113] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2006] [Revised: 09/14/2006] [Accepted: 09/21/2006] [Indexed: 11/19/2022] Open
Abstract
Recent evidence suggests that mutant huntingtin protein-induced energetic perturbations contribute to neuronal dysfunction in Huntington's disease (HD). Given the ubiquitous expression of huntingtin, other cell types with high energetic burden may be at risk for HD-related dysfunction. Early-onset cardiovascular disease is the second leading cause of death in HD patients; a direct role for mutant huntingtin in this phenomenon remains unevaluated. Here we tested the hypothesis that expression of mutant huntingtin is sufficient to induce cardiac dysfunction, using a well-described transgenic model of HD (line R6/2). R6/2 mice developed cardiac dysfunction by 8 weeks of age, progressing to severe failure at 12 weeks, assessed by echocardiography. Limited evidence of cardiac remodeling (e.g. hypertrophy, fibrosis, apoptosis, beta(1) adrenergic receptor downregulation) was observed. Immunogold electron microscopy demonstrated significant elevations in nuclear and mitochondrial polyglutamine presence in the R6/2 myocyte. Significant alterations in mitochondrial ultrastructure were seen, consistent with metabolic stress. Increased cardiac lysine acetylation and protein nitration were observed and were each significantly associated with impairments in cardiac performance. These data demonstrate that mutant huntingtin expression has potent cardiotoxic effects; cardiac failure may be a significant complication of this important experimental model of HD. Investigation of the potential cardiotropic effects of mutant huntingtin in humans may be warranted.
Collapse
Affiliation(s)
- Michael J. Mihm
- Center for Cardiovascular Medicine, Columbus Children’s Research Institute, 700 Children’s Drive, Columbus, Ohio 43205
- Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210
| | - Deborah M. Amann
- Center for Cardiovascular Medicine, Columbus Children’s Research Institute, 700 Children’s Drive, Columbus, Ohio 43205
| | - Brandon L. Schanbacher
- Center for Cardiovascular Medicine, Columbus Children’s Research Institute, 700 Children’s Drive, Columbus, Ohio 43205
| | - Ruth A. Altschuld
- The Ohio State University Biophysics Program and Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio 43210
| | - John Anthony Bauer
- Center for Cardiovascular Medicine, Columbus Children’s Research Institute, 700 Children’s Drive, Columbus, Ohio 43205
- Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, Ohio 43210
- * Corresponding Authors: Division of Pharmacology, College of Pharmacy, The Ohio State University, 412 Riffe Building, 496 West 12 Avenue, Columbus, OH 43221, Phone: (614) 292-6636, e-mail: (J.A.B) and (K.R.H)
| | - Kari R. Hoyt
- Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210
- * Corresponding Authors: Division of Pharmacology, College of Pharmacy, The Ohio State University, 412 Riffe Building, 496 West 12 Avenue, Columbus, OH 43221, Phone: (614) 292-6636, e-mail: (J.A.B) and (K.R.H)
| |
Collapse
|
10
|
Arango M, Holbert S, Zala D, Brouillet E, Pearson J, Régulier E, Thakur AK, Aebischer P, Wetzel R, Déglon N, Néri C. CA150 expression delays striatal cell death in overexpression and knock-in conditions for mutant huntingtin neurotoxicity. J Neurosci 2006; 26:4649-59. [PMID: 16641246 PMCID: PMC6674076 DOI: 10.1523/jneurosci.5409-05.2006] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Transcriptional dysregulation caused by expanded polyglutamines (polyGlns) in huntingtin (htt) may be central to cell-autonomous mechanisms for neuronal cell death in Huntington's disease (HD) pathogenesis. We hypothesized that these mechanisms may involve the dysfunction of the transcriptional regulator CA150, a putative modifier of onset age in HD, because it binds to htt and accumulates in an HD grade-dependent manner in striatal and cortical neurons. Consistently, we report herein that CA150 expression rescues striatal cell death in lentiviral overexpression (rats) and knock-in (mouse cells) conditions for mutant htt neurotoxicity. In both systems, rescue was dependent on the (Gln-Ala)38 repeat normally found in CA150. We excluded the possibility that rescue may be caused by the (Gln-Ala)38 repeat interacting with polyGlns and, by doing so, blocking mutant htt toxicity. In contrast, we found the (Gln-Ala)38 repeat is required for the nuclear restriction of exogenous CA150, suggesting that rescue requires nuclear CA150. Additionally, we found the (Gln-Ala)38 repeat was dispensable for CA150 transcriptional repression ability, suggesting further that CA150 localization is critical to rescue. Finally, rescue was associated with increased neuritic aggregation, with no reduction of nuclear inclusions, suggesting the solubilization and nuclear export of mutant htt. Together, our data indicate that mutant htt may induce CA150 dysfunction in striatal neurons and suggest that the restoration of nuclear protein cooperativity may be neuroprotective.
Collapse
|
11
|
Aguzzi A, Raeber AJ. Transgenic models of neurodegeneration. Neurodegeneration: of (transgenic) mice and men. Brain Pathol 2006; 8:695-7. [PMID: 9804378 PMCID: PMC8098419 DOI: 10.1111/j.1750-3639.1998.tb00195.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Affiliation(s)
- A Aguzzi
- Institute of Neuropathology, Department of Pathology, University Hospital, Zurich, Switzerland.
| | | |
Collapse
|
12
|
Wang W, Duan W, Igarashi S, Morita H, Nakamura M, Ross CA. Compounds blocking mutant huntingtin toxicity identified using a Huntington's disease neuronal cell model. Neurobiol Dis 2006; 20:500-8. [PMID: 15908226 DOI: 10.1016/j.nbd.2005.03.026] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2005] [Revised: 03/27/2005] [Accepted: 03/31/2005] [Indexed: 10/25/2022] Open
Abstract
Neuronal cell death in HD is believed to be largely a dominant cell-autonomous effect of the mutant huntingtin protein. We previously developed an inducible PC12 cell model which expresses an N-terminal huntingtin fragment with an expanded poly Q repeat (N63-148Q) under the control of the tet-off system. In order to evaluate the ability of compounds to protect against mutant huntingtin toxicity in our model, we measured LDH released by dead cells into the medium. We have now screened the library of 1040 compounds from the NINDS Custom Collection as part of a National Institute of Neurological Disorders and Stroke (NINDS) collaborative project. Each positive compound was tested at 3-8 concentrations. Five compounds significantly attenuated mutant huntingtin (htt)-induced LDH release without affecting the expression level of huntingtin and independent of effect on aggregates. We also tested a broad spectrum caspase inhibitor Z-VAD-fmk and previously proposed candidate compounds. This cell model can provide a method to screen potential therapeutic compounds for treating Huntington's disease.
Collapse
Affiliation(s)
- Wenfei Wang
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD 21205-2109, USA
| | | | | | | | | | | |
Collapse
|
13
|
Charvin D, Vanhoutte P, Pagès C, Borrelli E, Borelli E, Caboche J. Unraveling a role for dopamine in Huntington's disease: the dual role of reactive oxygen species and D2 receptor stimulation. Proc Natl Acad Sci U S A 2005; 102:12218-23. [PMID: 16103364 PMCID: PMC1189314 DOI: 10.1073/pnas.0502698102] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Huntington's disease (HD), an inherited neurodegenerative disorder, results from an abnormal polyglutamine extension in the N-terminal region of the huntingtin protein. This mutation leads to protein aggregation and neurotoxicity. Despite its widespread expression in the brain and body, mutated huntingtin causes selective degeneration of striatal projection neurons. In the present study, we investigate the role of dopamine (DA) in this preferential vulnerability. Using primary cultures of striatal neurons transiently expressing GFP-tagged-exon 1 of mutated huntingtin, we show that low doses of DA (100 microM) act synergistically with mutated huntingtin to activate the proapoptotic transcription factor c-Jun. Surprisingly, DA also increases aggregate formation of mutated huntingtin in all cellular compartments, including neurites, soma, and nuclei. DA-dependent potentiation of c-Jun activation was reversed by ascorbate, a reactive oxygen species (ROS) scavenger, and SP-600125, a selective inhibitor of the c-Jun N-terminal kinase (JNK) pathway. By contrast, DA effects on aggregate formation were reversed by a selective D2 receptor antagonist and reproduced by a D2 agonist. Similarly, striatal neurons from D2 knockout mice showed no effect of DA on aggregate formation. Blocking ROS production, JNK activation, or D2 receptor stimulation significantly reversed DA aggravation of mutated huntingtin-induced striatal death. The combined treatment with the ROS scavenger and D2 antagonist totally reversed DA's effects on mutated huntingtin-induced striatal death. Thus, the present results provide insights into the cellular mechanisms that govern striatal vulnerability in HD and strongly support a dual role of JNK activation and D2 receptor signaling in this process.
Collapse
Affiliation(s)
- Delphine Charvin
- Unité Mixte de Recherche 7102, Signalisation Neuronale et Régulations Géniques, 9 Quai Saint Bernard, 75005 Paris, France
| | | | | | | | | | | |
Collapse
|
14
|
Miller TW, Zhou C, Gines S, MacDonald ME, Mazarakis ND, Bates GP, Huston JS, Messer A. A human single-chain Fv intrabody preferentially targets amino-terminal Huntingtin's fragments in striatal models of Huntington's disease. Neurobiol Dis 2005; 19:47-56. [PMID: 15837560 DOI: 10.1016/j.nbd.2004.11.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2004] [Revised: 10/29/2004] [Accepted: 11/09/2004] [Indexed: 12/01/2022] Open
Abstract
Amino-terminal fragments of huntingtin (htt) appear to result from proteolytic processing of the full-length protein in Huntington's disease (HD), and fragments containing pathological expansions of polyglutamine elicit toxicity in model systems. Such fragments are sequestered into insoluble aggregates, which may initially serve a cellular protective mechanism, while soluble fragments and/or oligomers may be a more acute toxic species. Agents which enhance mutant htt clearance have shown therapeutic potential in animal models of HD. Here, we present the first evidence of an htt-specific single-chain Fv intrabody (C4) that selectively targets the soluble fraction of amino-terminal htt fragments. Our findings suggest that the C4 intrabody binds weakly, but does not alter the levels of endogenous, full-length htt. C4 appears to decrease the steady-state levels of amino-terminal htt fragments by binding to non-aggregated, but not aggregated, htt species. Intrabodies may be used as potential curative agents, and as drug discovery tools, for HD and other misfolded protein disorders.
Collapse
Affiliation(s)
- Todd W Miller
- Division of Genetic Disorders Wadsworth Center, New York State Department of Health, and Department of Biomedical Sciences, University at Albany, NY 12208, USA
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Protein misfolding and cellular defense mechanisms in neurodegenerative diseases. NEURODEGENER DIS 2005. [DOI: 10.1017/cbo9780511544873.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
16
|
Fox SH, Nieves A, Bergeron C, Lang AE. Pure cerebello-olivary degeneration of Marie, Foix, and Alajouanine presenting with progressive cerebellar ataxia, cognitive decline, and chorea. Mov Disord 2003; 18:1550-4. [PMID: 14673899 DOI: 10.1002/mds.10606] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Parenchymatous cerebellar cortical atrophy (CCA) usually presents with a "pure" cerebellar ataxia. We describe a patient with a sporadic, late-onset progressive cerebellar ataxia plus cognitive decline and chorea who had CCA at post mortem. We discuss this unique case in the current context of classification of idiopathic cerebellar ataxia.
Collapse
Affiliation(s)
- Susan H Fox
- Walton Centre for Neurology and Neurosurgery, Liverpool. United Kingdom.
| | | | | | | |
Collapse
|
17
|
Meriin AB, Zhang X, Miliaras NB, Kazantsev A, Chernoff YO, McCaffery JM, Wendland B, Sherman MY. Aggregation of expanded polyglutamine domain in yeast leads to defects in endocytosis. Mol Cell Biol 2003; 23:7554-65. [PMID: 14560003 PMCID: PMC207640 DOI: 10.1128/mcb.23.21.7554-7565.2003] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2003] [Revised: 07/15/2003] [Accepted: 07/25/2003] [Indexed: 11/20/2022] Open
Abstract
The role of aggregation of abnormal proteins in cellular toxicity is of general importance for understanding many neurological disorders. Here, using a yeast model, we demonstrate that mutations in many proteins involved in endocytosis and actin function dramatically enhance the toxic effect of polypeptides with an expanded polyglutamine (polyQ) domain. This enhanced cytotoxicity required polyQ aggregation and was dependent on the yeast protein Rnq1 in its prion form. In wild-type cells, expression of expanded polyQ followed by its aggregation led to specific and acute inhibition of endocytosis, which preceded growth inhibition. Some components of the endocytic machinery were efficiently recruited into the polyQ aggregates. Furthermore, in cells with polyQ aggregates, cortical actin patches were delocalized and actin was recruited into the polyQ aggregates. Aggregation of polyQ in mammalian HEK293 cells also led to defects in endocytosis. Therefore, it appears that inhibition of endocytosis is a direct consequence of polyQ aggregation and could significantly contribute to cytotoxicity.
Collapse
Affiliation(s)
- Anatoli B Meriin
- Department of Biochemistry, Boston University School of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Abstract
There is growing evidence that reduced neurotrophic support is a significant factor in the pathogenesis of neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD) and amyotrophic lateral sclerosis (ALS). In this review we discuss the structure and functions of neurotrophins such as nerve growth factor, and the role of these proteins and their tyrosine kinase (Trk) receptors in the aetiology and therapy of such diseases. Neurotrophins regulate development and the maintenance of the vertebrate nervous system. In the mature nervous system they affect neuronal survival and also influence synaptic function and plasticity. The neurotrophins are able to bind to two different receptors: all bind to a common receptor p75NTR, and each also binds to one of a family of Trk receptors. By dimerization of the Trk receptors, and subsequent transphosphorylation of the intracellular kinase domain, signalling pathways are activated. We discuss here the structure and function of the neurotrophins and how they have been, or may be, used therapeutically in AD, PD, Huntington's diseases, ALS and peripheral neuropathy. Neurotrophins are central to many aspects of nervous system function. However they have not truly fulfilled their therapeutic potential in clinical trials because of the difficulties of protein delivery and pharmacokinetics in the nervous system. With the recent elucidation of the structure of the neurotrophins bound to their receptors it will now be possible, using a combination of in silico technology and novel screening techniques, to develop small molecule mimetics with much improved pharmacotherapeutic profiles.
Collapse
Affiliation(s)
- D Dawbarn
- University of Bristol, Bristol Royal Infirmary, Bristol, UK.
| | | |
Collapse
|
19
|
Agius LM. Dysregulatory dysequilibrium of gene transcription and of nuclear transport in polyglutamine neuro-degeneration. Med Hypotheses 2003; 60:869-73. [PMID: 12699716 DOI: 10.1016/s0306-9877(03)00068-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Polyglutamine neurodegeneration as an essential expansion mutation of the CAG-trinucleotide repeat encoding glutamine would appear to constitute an integral process of aggregation/accumulation that self-propagates a secondary process of possible nuclear sequestration. Within such a scheme of progressive expansion of polyglutamine stretches in strict parallel correlation with increased CAG trinucleotide repeats in genes such as ataxin-7 and its messenger RNA, it would appear that a fundamental relationship of accumulation directly inducing biophysical disruption between nuclear/nucleolar and cytoplasmic protein machineries would constitute a dysfunctional dysequilibrium accounting for self-progressive neuronal degeneration with atrophy of the cerebral cortex and ganglia such as the caudate, that is limited often to specific population groups of neurons. It is for example in terms of Huntington's disease as an autosomal dominant disorder with high penetrance on a background of onset of dementia mainly in the fourth and fifth decades of life that one might conceive of polyglutamine neurodegeneration as fundamentally a developmental disturbance affecting neuronal maturation that accounts for abnormal neurophysiological and biochemical aspects of interaction of nucleus with cytoplasm. Polyglutamine expansion and trinucleotide repeats as both progressive processes of accumulation and synthesis would constitute a complex interplay of inducing and induced effects that both contribute in probably multiple ways to the self-progressive nature of a nuclear sequestration process.
Collapse
Affiliation(s)
- Lawrence M Agius
- Department of Pathology, St. Luke's Hospital, University of Malta, Malta.
| |
Collapse
|
20
|
Apostol BL, Kazantsev A, Raffioni S, Illes K, Pallos J, Bodai L, Slepko N, Bear JE, Gertler FB, Hersch S, Housman DE, Marsh JL, Thompson LM. A cell-based assay for aggregation inhibitors as therapeutics of polyglutamine-repeat disease and validation in Drosophila. Proc Natl Acad Sci U S A 2003; 100:5950-5. [PMID: 12730384 PMCID: PMC156307 DOI: 10.1073/pnas.2628045100] [Citation(s) in RCA: 151] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The formation of polyglutamine-containing aggregates and inclusions are hallmarks of pathogenesis in Huntington's disease that can be recapitulated in model systems. Although the contribution of inclusions to pathogenesis is unclear, cell-based assays can be used to screen for chemical compounds that affect aggregation and may provide therapeutic benefit. We have developed inducible PC12 cell-culture models to screen for loss of visible aggregates. To test the validity of this approach, compounds that inhibit aggregation in the PC12 cell-based screen were tested in a Drosophila model of polyglutamine-repeat disease. The disruption of aggregation in PC12 cells strongly correlates with suppression of neuronal degeneration in Drosophila. Thus, the engineered PC12 cells coupled with the Drosophila model provide a rapid and effective method to screen and validate compounds.
Collapse
Affiliation(s)
- Barbara L Apostol
- Department of Psychiatry and Human Behavior, Gillespie 2121, University of California, Irvine, CA 92697-4260, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Steffan JS, Thompson LM. Targeting aggregation in the development of therapeutics for the treatment of Huntington's disease and other polyglutamine repeat diseases. Expert Opin Ther Targets 2003; 7:201-13. [PMID: 12667098 DOI: 10.1517/14728222.7.2.201] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Huntington's disease (HD) is one of a number of familial polyglutamine (polyQ) repeat diseases. These neurodegenerative disorders are caused by expression of otherwise unrelated proteins that contain an expansion of a polyQ tract, rendering them toxic to specific subsets of vulnerable neurons. These expanded repeats have an inherent propensity to aggregate; insoluble neuronal nuclear and cytoplasmic polyQ aggregates or inclusions are hallmarks of the disorders [1,2]. In HD, inclusions in diseased brains often precede onset of symptoms, and have been proposed to be involved in pathogenicity [3-5]. Various strategies to block the process of aggregation have been developed in an effort to create drugs that decrease neurotoxicity. A discussion of the effect of antibodies, caspase inhibitors, chemical inhibitors, heat-shock proteins, suppressor peptides and transglutaminase inhibitors upon aggregation and disease is presented.
Collapse
Affiliation(s)
- Joan S Steffan
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA 92697-4260, USA.
| | | |
Collapse
|
22
|
Fan X, Rouleau GA. Progress in understanding the pathogenesis of oculopharyngeal muscular dystrophy. Can J Neurol Sci 2003; 30:8-14. [PMID: 12619777 DOI: 10.1017/s0317167100002365] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Oculopharyngeal muscular dystrophy (OPMD) is an adult-onset disorder characterized by progressive eyelid drooping (ptosis), swallowing difficulties (dysphagia), and proximal limb weakness. The autosomal dominant form of this disease is caused by expansions of a (GCG)6 repeat to (GCG)8-13 in the PABPN1 gene. These mutations lead to the expansion of a polyalanine stretch from 10 to 12-17 alanines in the N-terminal domain of PABPN1. Mutated PABPN1 (mPABPN1) induces the formation of muscle intranuclear inclusions that are thought to be the hallmark of this disease. In this review, we discuss: 1) OPMD genetics and PABPN I function studies; 2) diseases caused by polyalanine expansions and cellular polyalanine toxicity; 3) mPABPN1-induced intranuclear inclusion toxicity; 4) role of oligomerization of mPABPNI in the formation and toxicity of OPMD intranuclear inclusions and; 5) recruitment of subcellular components to the OPMD inclusions. We present a potential molecular mechanism for OPMD pathogenesis that accounts for these observations.
Collapse
Affiliation(s)
- Xueping Fan
- Center for Research in Neuroscience, McGill University, and the McGill University Health Center, Montreal, Quebec, Canada
| | | |
Collapse
|
23
|
Abstract
Huntington's disease (HD) results from polyglutamine expansion in huntingtin (htt), a protein with several consensus caspase cleavage sites. Despite the identification of htt fragments in the brain, it has not been shown conclusively that htt is cleaved by caspases in vivo. Furthermore, no study has addressed when htt cleavage occurs with respect to the onset of neurodegeneration. Using antibodies that detect only caspase-cleaved htt, we demonstrate that htt is cleaved in vivo specifically at the caspase consensus site at amino acid 552. We detect caspase-cleaved htt in control human brain as well as in HD brains with early grade neuropathology, including one homozygote. Cleaved htt is also seen in wild-type and HD transgenic mouse brains before the onset of neurodegeneration. These results suggest that caspase cleavage of htt may be a normal physiological event. However, in HD, cleavage of mutant htt would release N-terminal fragments with the potential for increased toxicity and accumulation caused by the presence of the expanded polyglutamine tract. Furthermore, htt fragments were detected most abundantly in cortical projection neurons, suggesting that accumulation of expanded htt fragments in these neurons may lead to corticostriatal dysfunction as an early event in the pathogenesis of HD.
Collapse
|
24
|
Wellington CL, Ellerby LM, Gutekunst CA, Rogers D, Warby S, Graham RK, Loubser O, van Raamsdonk J, Singaraja R, Yang YZ, Gafni J, Bredesen D, Hersch SM, Leavitt BR, Roy S, Nicholson DW, Hayden MR. Caspase cleavage of mutant huntingtin precedes neurodegeneration in Huntington's disease. J Neurosci 2002; 22:7862-72. [PMID: 12223539 PMCID: PMC6758089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2023] Open
Abstract
Huntington's disease (HD) results from polyglutamine expansion in huntingtin (htt), a protein with several consensus caspase cleavage sites. Despite the identification of htt fragments in the brain, it has not been shown conclusively that htt is cleaved by caspases in vivo. Furthermore, no study has addressed when htt cleavage occurs with respect to the onset of neurodegeneration. Using antibodies that detect only caspase-cleaved htt, we demonstrate that htt is cleaved in vivo specifically at the caspase consensus site at amino acid 552. We detect caspase-cleaved htt in control human brain as well as in HD brains with early grade neuropathology, including one homozygote. Cleaved htt is also seen in wild-type and HD transgenic mouse brains before the onset of neurodegeneration. These results suggest that caspase cleavage of htt may be a normal physiological event. However, in HD, cleavage of mutant htt would release N-terminal fragments with the potential for increased toxicity and accumulation caused by the presence of the expanded polyglutamine tract. Furthermore, htt fragments were detected most abundantly in cortical projection neurons, suggesting that accumulation of expanded htt fragments in these neurons may lead to corticostriatal dysfunction as an early event in the pathogenesis of HD.
Collapse
Affiliation(s)
- Cheryl L Wellington
- Centre for Molecular Medicine and Therapeutics, British Columbia Children's and Women's Hospital, Vancouver, British Columbia, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Meriin AB, Zhang X, He X, Newnam GP, Chernoff YO, Sherman MY. Huntington toxicity in yeast model depends on polyglutamine aggregation mediated by a prion-like protein Rnq1. J Cell Biol 2002; 157:997-1004. [PMID: 12058016 PMCID: PMC2174031 DOI: 10.1083/jcb.200112104] [Citation(s) in RCA: 302] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The cause of Huntington's disease is expansion of polyglutamine (polyQ) domain in huntingtin, which makes this protein both neurotoxic and aggregation prone. Here we developed the first yeast model, which establishes a direct link between aggregation of expanded polyQ domain and its cytotoxicity. Our data indicated that deficiencies in molecular chaperones Sis1 and Hsp104 inhibited seeding of polyQ aggregates, whereas ssa1, ssa2, and ydj1-151 mutations inhibited expansion of aggregates. The latter three mutants strongly suppressed the polyQ toxicity. Spontaneous mutants with suppressed aggregation appeared with high frequency, and in all of them the toxicity was relieved. Aggregation defects in these mutants and in sis1-85 were not complemented in the cross to the hsp104 mutant, demonstrating an unusual type of inheritance. Since Hsp104 is required for prion maintenance in yeast, this suggested a role for prions in polyQ aggregation and toxicity. We screened a set of deletions of nonessential genes coding for known prions and related proteins and found that deletion of the RNQ1 gene specifically suppressed aggregation and toxicity of polyQ. Curing of the prion form of Rnq1 from wild-type cells dramatically suppressed both aggregation and toxicity of polyQ. We concluded that aggregation of polyQ is critical for its toxicity and that Rnq1 in its prion conformation plays an essential role in polyQ aggregation leading to the toxicity.
Collapse
|
26
|
Shimohata T, Sato A, Burke JR, Strittmatter WJ, Tsuji S, Onodera O. Expanded polyglutamine stretches form an 'aggresome'. Neurosci Lett 2002; 323:215-8. [PMID: 11959423 DOI: 10.1016/s0304-3940(02)00162-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
To understand the pathogenetic mechanisms underlying polyglutamine (polyQ) diseases, we investigated the mechanisms of the formation of aggregate bodies containing expanded polyQ stretches, focusing on dentatorubral-pallidoluysian atrophy (DRPLA). We demonstrated that the expression of a truncated DRPLA protein containing expanded polyQ stretches in COS-7 cells resulted in the formation of perinuclear aggregate bodies that are co-localized with gamma-tubulin, a protein marker for the microtubules-organizing center (MTOC). A collapsed vimentin network surrounded these aggregate bodies. Furthermore, disruption of the microtubules (MTs) with nocodazole resulted in the formation of small aggregate bodies that were scattered throughout the cytoplasm. These findings suggest that the truncated DRPLA proteins containing expanded polyQ stretches unfold and form small aggregate bodies in the cell periphery. These aggregates move on MTs to the MTOC, where they remain as distinct 'aggresomes''.
Collapse
Affiliation(s)
- Takayoshi Shimohata
- Department of Neurology, Brain Research Institute, Niigata University, 1-757 Asahimachi, Niigata 951-8181, Japan
| | | | | | | | | | | |
Collapse
|
27
|
Kazantsev A, Walker HA, Slepko N, Bear JE, Preisinger E, Steffan JS, Zhu YZ, Gertler FB, Housman DE, Marsh JL, Thompson LM. A bivalent Huntingtin binding peptide suppresses polyglutamine aggregation and pathogenesis in Drosophila. Nat Genet 2002; 30:367-76. [PMID: 11925563 DOI: 10.1038/ng864] [Citation(s) in RCA: 125] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Huntington disease is caused by the expansion of a polyglutamine repeat in the Huntingtin protein (Htt) that leads to degeneration of neurons in the central nervous system and the appearance of visible aggregates within neurons. We have developed and tested suppressor polypeptides that bind mutant Htt and interfere with the process of aggregation in cell culture. In a Drosophila model, the most potent suppressor inhibits both adult lethality and photoreceptor neuron degeneration. The appearance of aggregates in photoreceptor neurons correlates strongly with the occurrence of pathology, and expression of suppressor polypeptides delays and limits the appearance of aggregates and protects photoreceptor neurons. These results suggest that targeting the protein interactions leading to aggregate formation may be beneficial for the design and development of therapeutic agents for Huntington disease.
Collapse
Affiliation(s)
- Aleksey Kazantsev
- Department of Biology, Center for Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Karpuj MV, Becher MW, Springer JE, Chabas D, Youssef S, Pedotti R, Mitchell D, Steinman L. Prolonged survival and decreased abnormal movements in transgenic model of Huntington disease, with administration of the transglutaminase inhibitor cystamine. Nat Med 2002; 8:143-9. [PMID: 11821898 DOI: 10.1038/nm0202-143] [Citation(s) in RCA: 279] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
An expanded polyglutamine domain in huntingtin underlies the pathogenic events in Huntington disease (HD), characterized by chorea, dementia and severe weight loss, culminating in death. Transglutaminase (TGase) may be critical in the pathogenesis, via cross-linking huntingtin. Administration of the TGase competitive inhibitor, cystamine, to transgenic mice expressing exon 1 of huntingtin containing an expanded polyglutamine repeat, altered the course of their HD-like disease. Cystamine given intraperitoneally entered brain where it inhibited TGase activity. When treatment began after the appearance of abnormal movements, cystamine extended survival, reduced associated tremor and abnormal movements and ameliorated weight loss. Treatment did not influence the appearance or frequency of neuronal nuclear inclusions. Unexpectedly, cystamine treatment increased transcription of one of the two genes shown to be neuroprotective for polyglutamine toxicity in Drosophila, dnaj (also known as HDJ1 and Hsp40 in humans and mice, respectively). Inhibition of TGase provides a new treatment strategy for HD and other polyglutamine diseases.
Collapse
Affiliation(s)
- Marcela V Karpuj
- Department of Neurological Sciences, Stanford University, Stanford, California, USA
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Lowe J, Mayer J, Landon M, Layfield R. Ubiquitin and the molecular pathology of neurodegenerative diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2001; 487:169-86. [PMID: 11403157 DOI: 10.1007/978-1-4615-1249-3_14] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
Ubiquitin plays a central role in normal cellular function as well as in disease. It is possible to group ubiquitin-immunostained structures into several main groups, the most distinctive being the ubiquitin/intermediate filament/alphaB crystallin family of inclusions that seem to represent a general cellular response to abnormal proteins recently termed the aggresomal response. While ubiquitin immunohistochemistry is a very useful technique for detecting pathological changes and inclusion bodies in the nervous system this alone is not enough to classify inclusions, and a panel of antibodies is recommended to clarify any findings made by screening tissues with anti-ubiquitin. Several mechanistic possibilities now exist to explain the accumulation of ubiquitinated proteins in cells of the nervous system, understanding of which should lead to new therapeutic advances in the group of chronic neurodegenerative diseases.
Collapse
Affiliation(s)
- J Lowe
- School of Biomedical Sciences, University of Nottingham Medical School, UK.
| | | | | | | |
Collapse
|
30
|
Hirabayashi M, Inoue K, Tanaka K, Nakadate K, Ohsawa Y, Kamei Y, Popiel AH, Sinohara A, Iwamatsu A, Kimura Y, Uchiyama Y, Hori S, Kakizuka A. VCP/p97 in abnormal protein aggregates, cytoplasmic vacuoles, and cell death, phenotypes relevant to neurodegeneration. Cell Death Differ 2001; 8:977-84. [PMID: 11598795 DOI: 10.1038/sj.cdd.4400907] [Citation(s) in RCA: 217] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2001] [Accepted: 05/07/2001] [Indexed: 11/09/2022] Open
Abstract
Neuronal cell death, abnormal protein aggregates, and cytoplasmic vacuolization are major pathologies observed in many neurodegenerative disorders such as the polyglutamine (polyQ) diseases, prion disease, Alzheimer disease, and the Lewy body diseases, suggesting common mechanisms underlying neurodegeneration. Here, we have identified VCP/p97, a member of the AAA+ family of ATPase proteins, as a polyQ-interacting protein in vitro and in vivo, and report on its characterization. Endogenous VCP co-localized with expanded polyQ (ex-polyQ) aggregates in cultured cells expressing ex-polyQ, with nuclear inclusions in Huntington disease patient brains, and with Lewy bodies in patient samples. Moreover, the expression of VCP mutants with mutations in the 2nd ATP binding domain created cytoplasmic vacuoles, followed by cell death. Very similar vacuoles were also induced by ex-polyQ expression or proteasome inhibitor treatment. These results suggest that VCP functions not only as a recognition factor for abnormally folded proteins but also as a pathological effector for several neurodegenerative phenotypes. VCP may thus be an ideal molecular target for the treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- M Hirabayashi
- The Fourth Department, Osaka Bioscience Institute, Osaka 565-0874, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Meriin AB, Mabuchi K, Gabai VL, Yaglom JA, Kazantsev A, Sherman MY. Intracellular aggregation of polypeptides with expanded polyglutamine domain is stimulated by stress-activated kinase MEKK1. J Cell Biol 2001; 153:851-64. [PMID: 11352944 PMCID: PMC2192371 DOI: 10.1083/jcb.153.4.851] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Abnormal proteins, which escape chaperone-mediated refolding or proteasome-dependent degradation, aggregate and form inclusion bodies (IBs). In several neurodegenerative diseases, such IBs can be formed by proteins with expanded polyglutamine (polyQ) domains (e.g., huntingtin). This work studies the regulation of intracellular IB formation using an NH2-terminal fragment of huntingtin with expanded polyQ domain. We demonstrate that the active form of MEKK1, a protein kinase that regulates several stress-activated signaling cascades, stimulates formation of the IBs. This function of MEKK1 requires kinase activity, as the kinase-dead mutant of MEKK1 cannot stimulate this process. Exposure of cells to UV irradiation or cisplatin, both of which activate MEKK1, also augmented the formation of IBs. The polyQ-containing huntingtin fragment exists in cells in two distinct forms: (a) in a discrete soluble complex, and (b) in association with insoluble fraction. MEKK1 strongly stimulated recruitment of polyQ polypeptides into the particulate fraction. Notably, a large portion of the active form of MEKK1 was associated with the insoluble fraction, concentrating in discrete sites, and polyQ-containing IBs always colocalized with them. We suggest that MEKK1 is involved in a process of IB nucleation. MEKK1 also stimulated formation of IBs with two abnormal polypeptides lacking the polyQ domain, indicating that this kinase has a general effect on protein aggregation.
Collapse
Affiliation(s)
- Anatoli B. Meriin
- Boston Biomedical Research Institute, Watertown, Massachusetts 02472
| | - Katsuhide Mabuchi
- Boston Biomedical Research Institute, Watertown, Massachusetts 02472
| | - Vladimir L. Gabai
- Boston Biomedical Research Institute, Watertown, Massachusetts 02472
| | - Julia A. Yaglom
- Boston Biomedical Research Institute, Watertown, Massachusetts 02472
| | - Alex Kazantsev
- Center for Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | | |
Collapse
|
32
|
Kusakabe M, Mangiarini L, Laywell ED, Bates GP, Yoshiki A, Hiraiwa N, Inoue J, Steindler DA. Loss of cortical and thalamic neuronal tenascin-C expression in a transgenic mouse expressing exon 1 of the human Huntington disease gene. J Comp Neurol 2001; 430:485-500. [PMID: 11169482 DOI: 10.1002/1096-9861(20010219)430:4<485::aid-cne1045>3.0.co;2-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
A transgenic mouse containing the first exon of the human Huntington's disease (HD) gene has revealed a variety of behavioral and pathophysiological anomalies reminiscent of certain aspects of human Huntington's disease (HD). The present study has found that expression of the extracellular matrix glycoprotein tenascin-C appears to be unaffected in astroglial cells in wild-type and R6/2 transgenic mice that express the mutant huntingtin protein but that it is conspicuously absent in two neuronal populations within the cerebral cortex and thalamus of the R6/2 mice. Loss of tenascin-C expression begins between the fourth and eighth postnatal weeks, coincidental with the onset of abnormal behavioral phenotype and the appearance of intranuclear inclusion bodies and neuropil aggregates. By 12 weeks, R6/2 mice exhibit a complete absence of tenascin-C neuronal immunolabeling, a disappearance of cRNA probe-positive neurons across discrete cytoarchitectonic regions of the dorsal thalamus (e.g., the ventromedial, parafascicular, lateral posterior, and posterior thalamic groups) and frontal cortex, and an accompanying thalamic astrogliosis. The loss of neuronal tenascin-C expression includes structures that are known to send converging excitatory axonal projections to the caudate-putamen, the structure that is most at risk for neurodegeneration in HD. Altered neuronal expression of tenascin-C in R6/2 mice implicates altered transcriptional activities of the mutant huntingtin protein. The abnormal biochemistry and possibly abnormal activity of thalamostriate and corticostriate projection neurons may also affect abnormal neuronal activities in their primary connectional target, the neostriatum, which is severely compromised in HD.
Collapse
Affiliation(s)
- M Kusakabe
- Division of Experimental Animal Research, RIKEN, Tsukuba, Japan
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Sherman MY, Goldberg AL. Cellular defenses against unfolded proteins: a cell biologist thinks about neurodegenerative diseases. Neuron 2001; 29:15-32. [PMID: 11182078 DOI: 10.1016/s0896-6273(01)00177-5] [Citation(s) in RCA: 729] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- M Y Sherman
- Boston Biomedical Research Institute, 64 Grove Street, Watertown, MA 02472, USA.
| | | |
Collapse
|
34
|
Abstract
Invertebrate models of several human neurodegenerative diseases have recently been described. These models faithfully replicate key neuropathological features of the human disorders. Because the basic cell biology of the nervous system is very similar in vertebrates and invertebrates, the sophisticated and rapid genetic analysis feasible in Drosophila and C. elegans promises significant insight into human neurodegenerative syndromes. In addition, the short lifespan, small size, and ease of culturing make worms and flies ideal for drug testing.
Collapse
Affiliation(s)
- M B Feany
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
35
|
Orr HT. The ins and outs of a polyglutamine neurodegenerative disease: spinocerebellar ataxia type 1 (SCA1). Neurobiol Dis 2000; 7:129-34. [PMID: 10860780 DOI: 10.1006/nbdi.2000.0305] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Polyglutamine neurodegenerative disorders are characterized by the expansion of a glutamine tract within the mutant disease-causing protein. Expression of the mutant protein induces a progressive loss of neuronal function and the subsequent neurodegeneration of a set of neurons characteristic to each disease. Spinocerebellar ataxia type 1 (SCA1) is one polyglutamine disease where various experimental model systems, in particular transgenic mice, have been utilized to dissect the molecular and cellular events important for disease. This review summarizes these findings and places them in a context of potential future research directions.
Collapse
Affiliation(s)
- H T Orr
- Department of Genetics, University of Minnesota, Minneapolis, Minnesota, 55455, USA.
| |
Collapse
|
36
|
Walker LC, LeVine H. The cerebral proteopathies: neurodegenerative disorders of protein conformation and assembly. Mol Neurobiol 2000; 21:83-95. [PMID: 11327151 DOI: 10.1385/mn:21:1-2:083] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The abnormal assembly and deposition of specific proteins in the brain is the probable cause of most neurodegenerative disease afflicting the elderly. These "cerebral proteopathies" include Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), prion diseases, and a variety of other disorders. Evidence is accumulating that the anomalous aggregation of the proteins, and not a loss of protein function, is central to the pathogenesis of these diseases. Thus, therapeutic strategies that reduce the production, accumulation, or polymerization of pathogenic proteins might be applicable to a wide range of some of the most devastating diseases of old age.
Collapse
Affiliation(s)
- L C Walker
- Neuroscience Therapeutics, Pfizer Ann Arbor Laboratories, MI 48105, USA
| | | |
Collapse
|
37
|
Yang LY, Arnold AP. BDNF regulation of androgen receptor expression in axotomized SNB motoneurons of adult male rats. Brain Res 2000; 852:127-39. [PMID: 10661504 DOI: 10.1016/s0006-8993(99)02225-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) prevents the axotomy-induced loss of androgen receptor-like immunoreactivity (AR-LI) in the spinal nucleus of the bulbocavernosus (SNB) motoneurons of adult male rats. In this report, we investigated the dose-response effect of BDNF on androgen receptor expression in axotomized SNB motoneurons, and examined whether delayed application of BDNF to the cut SNB axons can completely reverse the axotomy-induced loss of androgen receptor expression. We also used autoradiography to test whether axotomy decreases the ability of SNB motoneurons to accumulate androgens. SNB motoneurons were axotomized bilaterally and BDNF or PBS was applied to the proximal ends of the axons. The percentage of SNB motoneurons expressing medium or high AR-LI was the major measure of androgen receptor expression. AR-LI was significantly higher on the BDNF-treated side than on the contralateral side treated with phosphate-buffered saline (PBS) for all three doses of BDNF (1.45, 2.9, and 5.8 mg/ml) and was higher than in rats treated bilaterally with PBS. Moreover, AR-LI at the highest dose of BDNF was not different from that in intact SNB motoneurons. Delayed application of BDNF to the axotomized SNB motoneurons restored the AR-LI to the intact level. The AR-LI decreased by axotomy started to increase significantly 4 days after BDNF application and returned to the intact level by 10 days. Furthermore, axotomy significantly decreased the percentage of SNB motoneurons to accumulate tritiated testosterone or its metabolites. In conclusion, our data demonstrate that BDNF completely prevents and reverses the axotomy-induced loss of AR-LI. Moreover, decrease of AR-LI by axotomy reflects the decrease in the ability of SNB motoneurons to accumulate androgens.
Collapse
Affiliation(s)
- L Y Yang
- Department of Physiological Science and Laboratory of Neuroendocrinology of the Brain Research Institute, Unitcersiry of California, Los Angeles 90095-1527, USA.
| | | |
Collapse
|
38
|
Rich T, Assier E, Skepper J, Segard HB, Allen RL, Charron D, Trowsdale J. Disassembly of nuclear inclusions in the dividing cell--a novel insight into neurodegeneration. Hum Mol Genet 1999; 8:2451-9. [PMID: 10556293 DOI: 10.1093/hmg/8.13.2451] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Spinocerebellar ataxias and Huntington's disease are examples of neurodegenerative diseases caused by a trinucleotide repeat expansion. One hallmark of such diseases is the formation of inclusion bodies (IBs) within neuronal tissue. Although these inclusions may play a pivotal role in the disease process, the reasons underlying their specific accumulation remain obscure. By studying intranuclear IBs in dividing cells we demonstrate for the first time that inclusions such as those of ataxin-1 disperse during mitosis, thus reducing the nuclear aggregate burden. IBs reform in the interphase nucleus. By high-resolution confocal microscopy we also show that inclusions comprise ordered structures capable of homotypic interactions. Unlike those of a non-pathologic protein, ataxin-1 inclusions were shown to be capable of non-specific protein sequestration. Our studies indicate that the specific accumulation of inclusions in terminally differentiated cells such as neurons is a direct consequence of their inability to divide and therefore provides a key to explaining their persistence in neurodegenerative disease.
Collapse
Affiliation(s)
- T Rich
- Department of Pathology, Division of Immunology, University of Cambridge, Cambridge CB2 1QP, UK.
| | | | | | | | | | | | | |
Collapse
|
39
|
Mangiarini L, Sathasivam K, Bates GP. Molecular Pathology of Huntington's Disease: Animal Models and Nuclear Mechanisms. Neuroscientist 1999. [DOI: 10.1177/107385849900500613] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Huntington's disease (HD) is a late-onset neurodegenerative disorder caused by a polyglutamine expansion in huntingtin, a protein of unknown function. Transgenic models expressing a portion or full-length human huntingtin have been generated to unravel the mechanism through which the mutation causes the symptoms and the selective cell death characteristic of HD. We review here advances in the understanding of HD made possible by transgenic models and the means by which they implicate polyglutamine aggregation in the pathology of triplet repeat disorders.
Collapse
Affiliation(s)
- Laura Mangiarini
- Medical and Molecular Genetics GKT School of Medicine King's College Guy's Hospital London, UK
| | - Kirupa Sathasivam
- Medical and Molecular Genetics GKT School of Medicine King's College Guy's Hospital London, UK
| | - Gillian P. Bates
- Medical and Molecular Genetics GKT School of Medicine King's College Guy's Hospital London, UK
| |
Collapse
|
40
|
Davies SW, Sathasivam K, Hobbs C, Doherty P, Mangiarini L, Scherzinger E, Wanker EE, Bates GP. Detection of polyglutamine aggregation in mouse models. Methods Enzymol 1999; 309:687-701. [PMID: 10507055 DOI: 10.1016/s0076-6879(99)09045-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- S W Davies
- Department of Anatomy and Developmental Biology, University College London, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Schulz JB, Dichgans J. Molecular pathogenesis of movement disorders: are protein aggregates a common link in neuronal degeneration? Curr Opin Neurol 1999; 12:433-9. [PMID: 10555832 DOI: 10.1097/00019052-199908000-00010] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Abnormal protein aggregation has been postulated to explain the molecular basis for many neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease and prion diseases, as well as trinucleotide repeat disorders. The recent findings that mutations in alpha-synuclein lead to autosomal-dominant, early-onset Parkinson's disease in some families and that alpha-synuclein is found in Lewy bodies of all Parkinson's disease patients prompted the hypothesis that the pathophysiology of all Parkinson's disease patients starts with an abnormal folding of alpha-synuclein, producing excessive aggregation that overwhelms the antiaggregation mechanisms of the cell. The genetics of Parkinson's disease and polyglutamine repeat disorders and the evidence of abnormal processing and aggregation of the respective target proteins for the aetiology and pathogenesis in these diseases are reviewed.
Collapse
Affiliation(s)
- J B Schulz
- Department of Neurology and Medical School, University of Tübingen, Germany
| | | |
Collapse
|
42
|
Sathasivam K, Hobbs C, Mangiarini L, Mahal A, Turmaine M, Doherty P, Davies SW, Bates GP. Transgenic models of Huntington's disease. Philos Trans R Soc Lond B Biol Sci 1999; 354:963-9. [PMID: 10434294 PMCID: PMC1692600 DOI: 10.1098/rstb.1999.0447] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Huntington's disease (HD) is an inherited neurodegenerative disorder caused by a CAG-polyglutamine repeat expansion. A mouse model of this disease has been generated by the introduction of exon 1 of the human HD gene carrying highly expanded CAG repeats into the mouse germ line (R6 lines). Transgenic mice develop a progressive neurological phenotype with a movement disorder and weight loss similar to that in HD. We have previously identified neuronal inclusions in the brains of these mice that have subsequently been established as the pathological hallmark of polyglutamine disease. Inclusions are present before symptoms, which in turn occur long before any selective neuronal cell death can be identified. We have extended the search for inclusions to skeletal muscle, which, like brain, contains terminally differentiated cells. We have conducted an investigation into the skeletal muscle atrophy that occurs in the R6 lines, (i) to provide possible insights into the muscle bulk loss observed in HD patients, and (ii) to conduct a parallel analysis into the consequence of inclusion formation to that being performed in brain. The identification of inclusions in skeletal muscle might be additionally useful in monitoring the ability of drugs to prevent inclusion formation in vivo.
Collapse
Affiliation(s)
- K Sathasivam
- GKT Medical and Dental School, King's College, Guy's Hospital, London, UK
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Davies SW, Turmaine M, Cozens BA, Raza AS, Mahal A, Mangiarini L, Bates GP. From neuronal inclusions to neurodegeneration: neuropathological investigation of a transgenic mouse model of Huntington's disease. Philos Trans R Soc Lond B Biol Sci 1999; 354:971-9. [PMID: 10434295 PMCID: PMC1692612 DOI: 10.1098/rstb.1999.0448] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Huntington's disease (HD) is an inherited progressive neurodegenerative disease caused by the expansion of a polyglutamine repeat sequence within a novel protein. Recent work has shown that abnormal intranuclear inclusions of aggregated mutant protein within neurons is a characteristic feature shared by HD and several other diseases involving glutamine repeat expansion. This suggests that in each of the these disorders the affected nerve cells degenerate as a result of these abnormal inclusions. A transgenic mouse model of HD has been generated by introducing exon 1 of the HD gene containing a highly expanded CAG sequence into the mouse germline. These mice develop widespread neuronal intranuclear inclusions and neurodegeneration specifically within those areas of the brain known to degenerate in HD. We have investigated the sequence of pathological changes that occur after the formation of nuclear inclusions and that precede neuronal cell death in these cells. Although the relation between inclusion formation and neurodegeneration has recently been questioned, a full characterization of the pathways linking protein aggregation and cell death will resolve some of these controversies and will additionally provide new targets for potential therapies.
Collapse
MESH Headings
- Animals
- Brain/metabolism
- Gene Expression Regulation
- Humans
- Huntingtin Protein
- Huntington Disease/genetics
- Huntington Disease/metabolism
- In Situ Hybridization
- Mice
- Mice, Inbred C57BL
- Mice, Inbred CBA
- Mice, Transgenic
- Nerve Tissue Proteins/genetics
- Nuclear Proteins/genetics
- Receptors, AMPA/metabolism
- Receptors, GABA-A/metabolism
- Receptors, GABA-B/metabolism
- Receptors, Kainic Acid/metabolism
- Receptors, Metabotropic Glutamate/metabolism
- Receptors, N-Methyl-D-Aspartate/metabolism
- Receptors, Neurotransmitter/genetics
- Receptors, Neurotransmitter/metabolism
Collapse
Affiliation(s)
- S W Davies
- Department of Anatomy and Developmental Biology, University College London, UK.
| | | | | | | | | | | | | |
Collapse
|
44
|
Sathasivam K, Hobbs C, Turmaine M, Mangiarini L, Mahal A, Bertaux F, Wanker EE, Doherty P, Davies SW, Bates GP. Formation of polyglutamine inclusions in non-CNS tissue. Hum Mol Genet 1999; 8:813-22. [PMID: 10196370 DOI: 10.1093/hmg/8.5.813] [Citation(s) in RCA: 216] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Huntington's disease (HD) is one of a class of inherited progressive neurodegenerative disorders that are caused by a CAG/polyglutamine repeat expansion. We have previously generated mice that are transgenic for exon 1 of the HD gene carrying highly expanded CAG repeats which develop a progressive movement disorder and weight loss with similarities to HD. Neuronal inclusions composed of the exon 1 protein and ubiquitin are present in specific brain regions prior to onset of the phenotype, which in turn occurs long before specific neurodegeneration can be detected. In this report we have extended the search for polyglutamine inclusions to non-neuronal tissues. Outside the central nervous system (CNS), inclusions were identified in a variety of post-mitotic cells. This is consistent with a concentration-dependent nucleation and aggregation model of inclusion formation and indicates that brain-specific factors are not necessary for this process. To possibly gain insights into the wasting that is observed in the human disease, we have conducted a detailed analysis of the timing and progression of inclusion formation in skeletal muscle and an investigation into the cause of the severe muscle atrophy that occurs in the mouse model. The formation of inclusions in non-CNS tissues will be particularly useful with respect to in vivo monitoring of pharmaceutical agents selected for their ability to prevent polyglutamine aggregation in vitro, without the requirement that the agent can cross the blood-brain barrier in the first instance.
Collapse
Affiliation(s)
- K Sathasivam
- Medical and Molecular Genetics, GKT Medical and Dental School, King's College, 8th Floor, Guy's Tower, Guy's Hospital, London SE1 9RT, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|