51
|
Kim YE, Hosp F, Frottin F, Ge H, Mann M, Hayer-Hartl M, Hartl FU. Soluble Oligomers of PolyQ-Expanded Huntingtin Target a Multiplicity of Key Cellular Factors. Mol Cell 2016; 63:951-64. [PMID: 27570076 DOI: 10.1016/j.molcel.2016.07.022] [Citation(s) in RCA: 152] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 06/17/2016] [Accepted: 07/21/2016] [Indexed: 11/17/2022]
Abstract
Huntington's disease is one of several neurodegenerative disorders characterized by the aggregation of polyglutamine (polyQ)-expanded mutant protein. How polyQ aggregation leads to cellular dysfunction is not well understood. Here, we analyzed aberrant protein interactions of soluble oligomers and insoluble inclusions of mutant huntingtin using in-cell single molecule fluorescence spectroscopy and quantitative proteomics. We find that the interactome of soluble oligomers is highly complex, with an enrichment of RNA-binding proteins as well as proteins functioning in ribosome biogenesis, translation, transcription, and vesicle transport. The oligomers frequently target proteins containing extended low-complexity sequences, potentially interfering with key cellular pathways. In contrast, the insoluble inclusions are less interactive and associate strongly with protein quality control components, such as Hsp40 chaperones and factors of the ubiquitin-proteasome system. Our results suggest a "multiple hit" model for the pathogenic effects of mutant huntingtin, with soluble forms engaging more extensively in detrimental interactions than insoluble aggregates.
Collapse
Affiliation(s)
- Yujin E Kim
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82159 Martinsried, Germany
| | - Fabian Hosp
- Department of Proteomics & Signal Transduction, Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82159 Martinsried, Germany
| | - Frédéric Frottin
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82159 Martinsried, Germany
| | - Hui Ge
- Novartis Institutes for Biomedical Research, No. 2 BoYun Road, Pudong, Shanghai 201203, China
| | - Matthias Mann
- Department of Proteomics & Signal Transduction, Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82159 Martinsried, Germany
| | - Manajit Hayer-Hartl
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82159 Martinsried, Germany.
| | - F Ulrich Hartl
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82159 Martinsried, Germany; Munich Cluster for Systems Neurology, Adolf-Butenandt-Institut, Ludwig-Maximilians-Universität München, Schillerstrasse 44, 80336 München, Germany.
| |
Collapse
|
52
|
Kratter IH, Zahed H, Lau A, Tsvetkov AS, Daub AC, Weiberth KF, Gu X, Saudou F, Humbert S, Yang XW, Osmand A, Steffan JS, Masliah E, Finkbeiner S. Serine 421 regulates mutant huntingtin toxicity and clearance in mice. J Clin Invest 2016; 126:3585-97. [PMID: 27525439 PMCID: PMC5004962 DOI: 10.1172/jci80339] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 06/30/2016] [Indexed: 01/17/2023] Open
Abstract
Huntington's disease (HD) is a progressive, adult-onset neurodegenerative disease caused by a polyglutamine (polyQ) expansion in the N-terminal region of the protein huntingtin (HTT). There are no cures or disease-modifying therapies for HD. HTT has a highly conserved Akt phosphorylation site at serine 421, and prior work in HD models found that phosphorylation at S421 (S421-P) diminishes the toxicity of mutant HTT (mHTT) fragments in neuronal cultures. However, whether S421-P affects the toxicity of mHTT in vivo remains unknown. In this work, we used murine models to investigate the role of S421-P in HTT-induced neurodegeneration. Specifically, we mutated the human mHTT gene within a BAC to express either an aspartic acid or an alanine at position 421, mimicking tonic phosphorylation (mHTT-S421D mice) or preventing phosphorylation (mHTT-S421A mice), respectively. Mimicking HTT phosphorylation strongly ameliorated mHTT-induced behavioral dysfunction and striatal neurodegeneration, whereas neuronal dysfunction persisted when S421 phosphorylation was blocked. We found that S421 phosphorylation mitigates neurodegeneration by increasing proteasome-dependent turnover of mHTT and reducing the presence of a toxic mHTT conformer. These data indicate that S421 is a potent modifier of mHTT toxicity and offer in vivo validation for S421 as a therapeutic target in HD.
Collapse
Affiliation(s)
- Ian H. Kratter
- Gladstone Institute of Neurological Disease and the Taube/Koret Center for Neurodegenerative Disease Research, San Francisco, California, USA
- Biomedical Sciences Graduate Program and
- Medical Scientist Training Program, UCSF, San Francisco, California, USA
| | - Hengameh Zahed
- Gladstone Institute of Neurological Disease and the Taube/Koret Center for Neurodegenerative Disease Research, San Francisco, California, USA
- Biomedical Sciences Graduate Program and
- Medical Scientist Training Program, UCSF, San Francisco, California, USA
| | - Alice Lau
- Department of Psychiatry and Human Behavior, University of California, Irvine, Irvine, California, USA
| | - Andrey S. Tsvetkov
- Department of Neurobiology and Anatomy, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Aaron C. Daub
- Gladstone Institute of Neurological Disease and the Taube/Koret Center for Neurodegenerative Disease Research, San Francisco, California, USA
- Medical Scientist Training Program, UCSF, San Francisco, California, USA
- Graduate Program in Bioengineering, UCSF, San Francisco, California, USA
| | - Kurt F. Weiberth
- Gladstone Institute of Neurological Disease and the Taube/Koret Center for Neurodegenerative Disease Research, San Francisco, California, USA
- Biomedical Sciences Graduate Program and
| | - Xiaofeng Gu
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Biobehavioral Sciences, Brain Research Institute, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Frédéric Saudou
- Université Grenoble Alpes, Grenoble Institut des Neurosciences, Grenoble, France
- Inserm, U1216, Grenoble, France
- Centre Hospitalier Universitaire Grenoble, Grenoble, France
| | - Sandrine Humbert
- Université Grenoble Alpes, Grenoble Institut des Neurosciences, Grenoble, France
- Inserm, U1216, Grenoble, France
- Centre Hospitalier Universitaire Grenoble, Grenoble, France
| | - X. William Yang
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Biobehavioral Sciences, Brain Research Institute, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Alex Osmand
- Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee, USA
| | - Joan S. Steffan
- Department of Psychiatry and Human Behavior, University of California, Irvine, Irvine, California, USA
| | - Eliezer Masliah
- Departments of Neurosciences and Pathology, UCSD, La Jolla, California, USA
| | - Steven Finkbeiner
- Gladstone Institute of Neurological Disease and the Taube/Koret Center for Neurodegenerative Disease Research, San Francisco, California, USA
- Biomedical Sciences Graduate Program and
- Departments of Neurology and Physiology, UCSF, San Francisco, California, USA
| |
Collapse
|
53
|
Li L, Liu H, Dong P, Li D, Legant WR, Grimm JB, Lavis LD, Betzig E, Tjian R, Liu Z. Real-time imaging of Huntingtin aggregates diverting target search and gene transcription. eLife 2016; 5. [PMID: 27484239 PMCID: PMC4972539 DOI: 10.7554/elife.17056] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 07/07/2016] [Indexed: 01/21/2023] Open
Abstract
The presumptive altered dynamics of transient molecular interactions in vivo contributing to neurodegenerative diseases have remained elusive. Here, using single-molecule localization microscopy, we show that disease-inducing Huntingtin (mHtt) protein fragments display three distinct dynamic states in living cells - 1) fast diffusion, 2) dynamic clustering and 3) stable aggregation. Large, stable aggregates of mHtt exclude chromatin and form 'sticky' decoy traps that impede target search processes of key regulators involved in neurological disorders. Functional domain mapping based on super-resolution imaging reveals an unexpected role of aromatic amino acids in promoting protein-mHtt aggregate interactions. Genome-wide expression analysis and numerical simulation experiments suggest mHtt aggregates reduce transcription factor target site sampling frequency and impair critical gene expression programs in striatal neurons. Together, our results provide insights into how mHtt dynamically forms aggregates and disrupts the finely-balanced gene control mechanisms in neuronal cells.
Collapse
Affiliation(s)
- Li Li
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States.,LKS Bio-medical and Health Sciences Center, CIRM Center of Excellence, University of California, Berkeley, United States
| | - Hui Liu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Peng Dong
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Dong Li
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Wesley R Legant
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Jonathan B Grimm
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Luke D Lavis
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States.,Transcription Imaging Consortium, Howard Hughes Medical Institute, Ashburn, United States
| | - Eric Betzig
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Robert Tjian
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States.,LKS Bio-medical and Health Sciences Center, CIRM Center of Excellence, University of California, Berkeley, United States.,Transcription Imaging Consortium, Howard Hughes Medical Institute, Ashburn, United States
| | - Zhe Liu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States.,Transcription Imaging Consortium, Howard Hughes Medical Institute, Ashburn, United States
| |
Collapse
|
54
|
Comparison of mHTT Antibodies in Huntington's Disease Mouse Models Reveal Specific Binding Profiles and Steady-State Ubiquitin Levels with Disease Development. PLoS One 2016; 11:e0155834. [PMID: 27196694 PMCID: PMC4873140 DOI: 10.1371/journal.pone.0155834] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 05/04/2016] [Indexed: 11/19/2022] Open
Abstract
Huntington's disease (HD) cellular pathology is characterised by the aggregation of mutant huntingtin (mHTT) protein into inclusion bodies. The present paper compared the sensitivity of five widely used mHTT antibodies (S830; MW8; EM48; 1C2; ubiquitin) against mice from five commonly used HD mouse models (R6/1; YAC128; HdhQ92; B6 HdhQ150; B6 x129/Ola HdhQ150) at two ages to determine: the most sensitive antibodies for each model; whether mHTT antibody binding differed depending on aggregation stage (diffuse versus frank inclusion); the role of ubiquitin during aggregation as the ubiquitin proteosome system has been implicated in disease development. The models demonstrated unique profiles of antibody binding even when the models varied only by background strain (HdhQ150). MW8 was highly sensitive for detecting frank inclusions in all lines whereas EM48, ubiquitin and 1C2 demonstrated consistent staining in all models irrespective of age or form of mHTT. MW8 and S830 were the most sensitive antibodies with 1C2 the least. Ubiquitin levels were stable for each model regardless of age. Ubiquitin was particularly sensitive in young YAC128 mice that demonstrate an absence of inclusions until ~12 months of age suggesting high affinity to mHTT in its diffuse form. The data indicate that generalisations across models regarding the quantification of aggregations may not be valid and that mHTT antibody binding is unique to the mouse model and sensitive to changes in inclusion development.
Collapse
|
55
|
Yang H, Hu HY. Sequestration of cellular interacting partners by protein aggregates: implication in a loss-of-function pathology. FEBS J 2016; 283:3705-3717. [PMID: 27016044 DOI: 10.1111/febs.13722] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 03/11/2016] [Accepted: 03/24/2016] [Indexed: 01/09/2023]
Abstract
Protein misfolding and aggregation are a hallmark of several neurodegenerative diseases (NDs). However, how protein aggregation leads to cytotoxicity and neurodegeneration is still controversial. Emerging evidence demonstrates that sequestration of cellular-interacting partners by protein aggregates contributes to the pathogenesis of these diseases. Here, we review current research on sequestration of cellular proteins by protein aggregates and its relation to proteinopathies. Based on different interaction modes, we classify these protein sequestrations into four types: protein coaggregation, domain/motif-mediated sequestration, RNA-assisted sequestration, and sequestration of molecular chaperones. Thus, the cellular essential proteins and/or RNA hijacked by protein aggregates may lose their biological functions, consequently resulting in cytotoxicity and neurodegeneration. We have proposed a hijacking model recapitulating the sequestration process and the loss-of-function pathology of ND.
Collapse
Affiliation(s)
- Hui Yang
- State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hong-Yu Hu
- State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
56
|
Strong MK, Southwell AL, Yonan JM, Hayden MR, Macgregor GR, Thompson LM, Steward O. Age-Dependent Resistance to Excitotoxicity in Htt CAG140 Mice and the Effect of Strain Background. J Huntingtons Dis 2016; 1:221-41. [PMID: 23833693 DOI: 10.3233/jhd-129005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Mouse strain background can influence vulnerability to excitotoxic neuronal cell death and potentially modulate phenotypes in transgenic mouse models of human disease. Evidence supports a contribution of excitotoxicity to the selective death of medium spiny neurons in Huntington's disease (HD). Here, we assess whether strain differences in excitotoxic vulnerability influence striatal cell death in a knock-in mouse model of HD. Previous studies that evaluated resistance to excitotoxic lesions in several mouse models of HD had variable outcomes. In the present study, we directly compare one model on two different background strains to test the contribution of strain to excitotoxicity-mediated neurodegeneration. Mice of the FVB/N strain, which are highly vulnerable to excitotoxicity, become extremely resistant to quinolinic acid-induced striatal neurodegeneration with age, when carrying a huntingtin (Htt) allele expressing a HD transgene (CAG140). The resistance is much greater than the age-dependent resistance that has been previously reported in YAC128 mice. By 12 months of age, both heterozygous and homozygous FVB.CAG140 mice displayed virtually complete resistance to quinolinic acid-induced striatal neurodegeneration. A similar resistance develops in CAG140 mice on a C57BL/6N background although the effect size is smaller because C57BL/6N mice are already resistant due to genetic background. In a direct comparison with the YAC128 mice, FVB.CAG140 mice have greater resistance. FVB.CAG140 mice are also resistant to neurodegeneration following kainic acid-induced status epilepticus suggesting the existence of a common cellular mechanism that provides protection against multiple types of excitotoxic insult. These findings establish FVB.CAG140 mice as a useful model to investigate the cellular and molecular mechanisms that confer neuroprotection against excitotoxicity.
Collapse
Affiliation(s)
- Melissa K Strong
- Department of Anatomy and Neurobiology, University of California Irvine, Irvine, CA, USA
| | | | | | | | | | | | | |
Collapse
|
57
|
Bushart DD, Murphy GG, Shakkottai VG. Precision medicine in spinocerebellar ataxias: treatment based on common mechanisms of disease. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:25. [PMID: 26889478 DOI: 10.3978/j.issn.2305-5839.2016.01.06] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Spinocerebellar ataxias (SCAs) are a heterogeneous group of dominantly inherited neurodegenerative disorders affecting the cerebellum and its associated pathways. There are no available symptomatic or disease-modifying therapies available for any of the over 30 known causes of SCA. In order to develop precise treatments for SCAs, two strategies can be employed: (I) the use of gene-targeting strategies to silence disease-causing mutant protein expression; and (II) the identification and targeting of convergent mechanisms of disease across SCAs as a basis for treatment. Gene targeting strategies include RNA interference and antisense oligonucleotides designed to silence mutant genes in order to prevent mutant protein expression. These therapies can be precise, but delivery is difficult and many disease-causing mutations remain unknown. Emerging evidence suggests that several common disease mechanisms may exist across SCAs. Disrupted protein homeostasis, RNA toxicity, abnormal synaptic signaling, altered intracellular calcium handling, and altered Purkinje neuron membrane excitability are all disease mechanisms which are seen in multiple etiologies of SCA and could potentially be targeted for treatment. Clinical trials with drugs such as riluzole, a potassium channel activator, show promise for multiple SCAs and suggest that convergent disease mechanisms do exist and can be targeted. Precise treatment of SCAs may be best achieved through pharmacologic agents targeting specific disrupted pathways.
Collapse
Affiliation(s)
- David D Bushart
- 1 Department of Molecular & Integrative Physiology, 2 Molecular & Behavioral Neuroscience Institute, 3 Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Geoffrey G Murphy
- 1 Department of Molecular & Integrative Physiology, 2 Molecular & Behavioral Neuroscience Institute, 3 Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Vikram G Shakkottai
- 1 Department of Molecular & Integrative Physiology, 2 Molecular & Behavioral Neuroscience Institute, 3 Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
58
|
Lewerenz J, Maher P. Chronic Glutamate Toxicity in Neurodegenerative Diseases-What is the Evidence? Front Neurosci 2015; 9:469. [PMID: 26733784 PMCID: PMC4679930 DOI: 10.3389/fnins.2015.00469] [Citation(s) in RCA: 508] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 11/24/2015] [Indexed: 12/13/2022] Open
Abstract
Together with aspartate, glutamate is the major excitatory neurotransmitter in the brain. Glutamate binds and activates both ligand-gated ion channels (ionotropic glutamate receptors) and a class of G-protein coupled receptors (metabotropic glutamate receptors). Although the intracellular glutamate concentration in the brain is in the millimolar range, the extracellular glutamate concentration is kept in the low micromolar range by the action of excitatory amino acid transporters that import glutamate and aspartate into astrocytes and neurons. Excess extracellular glutamate may lead to excitotoxicity in vitro and in vivo in acute insults like ischemic stroke via the overactivation of ionotropic glutamate receptors. In addition, chronic excitotoxicity has been hypothesized to play a role in numerous neurodegenerative diseases including amyotrophic lateral sclerosis, Alzheimer's disease and Huntington's disease. Based on this hypothesis, a good deal of effort has been devoted to develop and test drugs that either inhibit glutamate receptors or decrease extracellular glutamate. In this review, we provide an overview of the different pathways that are thought to lead to an over-activation of the glutamatergic system and glutamate toxicity in neurodegeneration. In addition, we summarize the available experimental evidence for glutamate toxicity in animal models of neurodegenerative diseases.
Collapse
Affiliation(s)
- Jan Lewerenz
- Department of Neurology, Ulm UniversityUlm, Germany
| | - Pamela Maher
- Cellular Neurobiology Laboratory, Salk Institute for Biological StudiesLa Jolla, CA, USA
| |
Collapse
|
59
|
van Maarschalkerweerd A, Pedersen MN, Peterson H, Nilsson M, Nguyen T, Skamris T, Rand K, Vetri V, Langkilde AE, Vestergaard B. Formation of covalent di-tyrosine dimers in recombinant α-synuclein. INTRINSICALLY DISORDERED PROTEINS 2015; 3:e1071302. [PMID: 28232892 PMCID: PMC5314896 DOI: 10.1080/21690707.2015.1071302] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 07/02/2015] [Accepted: 07/02/2015] [Indexed: 11/02/2022]
Abstract
Parkinson's disease is associated with fibril deposition in the diseased brain. Misfolding events of the intrinsically disordered synaptic protein α-synuclein are suggested to lead to the formation of transient oligomeric and cytotoxic species. The etiology of Parkinson's disease is further associated with mitochondrial dysfunction and formation of reactive oxygen species. Oxidative stress causes chemical modification of native α-synuclein, plausibly further influencing misfolding events. Here, we present evidence for the spontaneous formation of covalent di-tyrosine α-synuclein dimers in standard recombinant protein preparations, induced without extrinsic oxidative or nitrative agents. The dimers exhibit no secondary structure but advanced SAXS studies reveal an increased structural definition, resulting in a more hydrophobic micro-environment than the highly disordered monomer. Accordingly, monomers and dimers follow distinct fibrillation pathways.
Collapse
Affiliation(s)
| | - M N Pedersen
- Department of Drug Design and Pharmacology; University of Copenhagen ; Copenhagen, Denmark
| | - H Peterson
- Department of Drug Design and Pharmacology; University of Copenhagen ; Copenhagen, Denmark
| | - M Nilsson
- Department of Drug Design and Pharmacology; University of Copenhagen ; Copenhagen, Denmark
| | - Ttt Nguyen
- Department of Pharmacy; University of Copenhagen ; Copenhagen, Denmark
| | - T Skamris
- Department of Drug Design and Pharmacology; University of Copenhagen ; Copenhagen, Denmark
| | - K Rand
- Department of Pharmacy; University of Copenhagen ; Copenhagen, Denmark
| | - V Vetri
- Dipartimento di Fisica e Chimica; Universitá di Palermo ; Palermo, Italy
| | - A E Langkilde
- Department of Drug Design and Pharmacology; University of Copenhagen ; Copenhagen, Denmark
| | - B Vestergaard
- Department of Drug Design and Pharmacology; University of Copenhagen ; Copenhagen, Denmark
| |
Collapse
|
60
|
Sun X, Li PP, Zhu S, Cohen R, Marque LO, Ross CA, Pulst SM, Chan HYE, Margolis RL, Rudnicki DD. Nuclear retention of full-length HTT RNA is mediated by splicing factors MBNL1 and U2AF65. Sci Rep 2015. [PMID: 26218986 PMCID: PMC4517393 DOI: 10.1038/srep12521] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Huntington's disease (HD) is caused by a CAG repeat expansion in the huntingtin (HTT) gene. Recent evidence suggests that HD is a consequence of multimodal, non-mutually exclusive mechanisms of pathogenesis that involve both HTT protein- and HTT RNA-triggered mechanisms. Here we provide further evidence for the role of expanded HTT (expHTT) RNA in HD by demonstrating that a fragment of expHTT is cytotoxic in the absence of any translation and that the extent of cytotoxicity is similar to the cytotoxicity of an expHTT protein fragment encoded by a transcript of similar length and with a similar repeat size. In addition, full-length (FL) expHTT is retained in the nucleus. Overexpression of the splicing factor muscleblind-like 1 (MBNL1) increases nuclear retention of expHTT and decreases the expression of expHTT protein in the cytosol. The splicing and nuclear export factor U2AF65 has the opposite effect, decreasing expHTT nuclear retention and increasing expression of expHTT protein. This suggests that MBNL1 and U2AF65 play a role in nuclear export of expHTT RNA.
Collapse
Affiliation(s)
- Xin Sun
- 1] Department of Psychiatry and Behavioral Sciences, Division of Neurobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA [2] Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong, China
| | - Pan P Li
- Department of Psychiatry and Behavioral Sciences, Division of Neurobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Shanshan Zhu
- Department of Psychiatry and Behavioral Sciences, Division of Neurobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Rachael Cohen
- Department of Psychiatry and Behavioral Sciences, Division of Neurobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Leonard O Marque
- Department of Psychiatry and Behavioral Sciences, Division of Neurobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Christopher A Ross
- 1] Department of Psychiatry and Behavioral Sciences, Division of Neurobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA [2] Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA [3] Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA [4] Program of Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Stefan M Pulst
- Department of Neurology, University of Utah, Salt Lake City, Utah, USA
| | - Ho Yin Edwin Chan
- Laboratory of Drosophila Research, School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
| | - Russell L Margolis
- 1] Department of Psychiatry and Behavioral Sciences, Division of Neurobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA [2] Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA [3] Program of Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Dobrila D Rudnicki
- 1] Department of Psychiatry and Behavioral Sciences, Division of Neurobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA [2] Program of Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
61
|
Kim J, Waldvogel HJ, Faull RLM, Curtis MA, Nicholson LFB. The RAGE receptor and its ligands are highly expressed in astrocytes in a grade-dependant manner in the striatum and subependymal layer in Huntington's disease. J Neurochem 2015; 134:927-42. [DOI: 10.1111/jnc.13178] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 05/04/2015] [Accepted: 05/13/2015] [Indexed: 11/30/2022]
Affiliation(s)
- Joanne Kim
- Department of Anatomy with Radiology; Faculty of Medical and Health Science (FMHS); University of Auckland; Auckland New Zealand
- Centre for Brain Research; Faculty of Medical and Health Science (FMHS); University of Auckland; Auckland New Zealand
| | - Henry J. Waldvogel
- Department of Anatomy with Radiology; Faculty of Medical and Health Science (FMHS); University of Auckland; Auckland New Zealand
- Centre for Brain Research; Faculty of Medical and Health Science (FMHS); University of Auckland; Auckland New Zealand
| | - Richard L. M. Faull
- Department of Anatomy with Radiology; Faculty of Medical and Health Science (FMHS); University of Auckland; Auckland New Zealand
- Centre for Brain Research; Faculty of Medical and Health Science (FMHS); University of Auckland; Auckland New Zealand
| | - Maurice A. Curtis
- Department of Anatomy with Radiology; Faculty of Medical and Health Science (FMHS); University of Auckland; Auckland New Zealand
- Centre for Brain Research; Faculty of Medical and Health Science (FMHS); University of Auckland; Auckland New Zealand
| | - Louise F. B. Nicholson
- Department of Anatomy with Radiology; Faculty of Medical and Health Science (FMHS); University of Auckland; Auckland New Zealand
- Centre for Brain Research; Faculty of Medical and Health Science (FMHS); University of Auckland; Auckland New Zealand
| |
Collapse
|
62
|
Abstract
In this review, we explore the similarities and differences in the behavioural neurobiology found in the mouse models of Huntington's disease (HD) and the human disease state. The review is organised with a comparative focus on the functional domains of motor control, cognition and behavioural disturbance (akin to psychiatric disturbance in people) and how our knowledge of the underlying physiological changes that are manifest in the HD mouse lines correspond to those seen in the HD clinical population. The review is framed in terms of functional circuitry and neurotransmitter systems and how abnormalities in these systems impact on the behavioural readouts across the mouse lines and how these may correspond to the deficits observed in people. In addition, interpretational issues associated with the data from animal studies are discussed.
Collapse
Affiliation(s)
- Simon P Brooks
- Brain Repair Group, Division of Neuroscience, Cardiff University School of Bioscience, Museum Avenue, Cardiff, Wales, UK,
| | | |
Collapse
|
63
|
Hoffner G, Djian P. Polyglutamine Aggregation in Huntington Disease: Does Structure Determine Toxicity? Mol Neurobiol 2014; 52:1297-1314. [PMID: 25336039 DOI: 10.1007/s12035-014-8932-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 10/09/2014] [Indexed: 01/14/2023]
Abstract
Huntington disease is a dominantly inherited disease of the central nervous system. The mutational expansion of polyglutamine beyond a critical length produces a toxic gain of function in huntingtin and results in neuronal death. In the course of the disease, expanded huntingtin is proteolyzed, becomes abnormally folded, and accumulates in oligomers, fibrils, and microscopic inclusions. The aggregated forms of the expanded protein are structurally diverse. Structural heterogeneity may explain why polyglutamine-containing aggregates could paradoxically be either toxic or neuroprotective. When defined, the toxic structures could then specifically be targeted by prophylactic or therapeutic drugs aimed at inhibiting polyglutamine aggregation.
Collapse
Affiliation(s)
- Guylaine Hoffner
- Laboratoire de Physiologie Cérébrale, Centre National de la Recherche Scientifique, Université Paris Descartes, 45 rue des Saints Pères, 75006, Paris, France
| | - Philippe Djian
- Laboratoire de Physiologie Cérébrale, Centre National de la Recherche Scientifique, Université Paris Descartes, 45 rue des Saints Pères, 75006, Paris, France.
| |
Collapse
|
64
|
van Maarschalkerweerd A, Vetri V, Langkilde AE, Foderà V, Vestergaard B. Protein/lipid coaggregates are formed during α-synuclein-induced disruption of lipid bilayers. Biomacromolecules 2014; 15:3643-54. [PMID: 25210839 DOI: 10.1021/bm500937p] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Amyloid formation is associated with neurodegenerative diseases such as Parkinson's disease (PD). Significant α-synuclein (αSN) deposition in lipid-rich Lewy bodies is a hallmark of PD. Nonetheless, an unraveling of the connection between neurodegeneration and amyloid fibrils, including the molecular mechanisms behind potential amyloid-mediated toxic effects, is still missing. Interaction between amyloid aggregates and the lipid cell membrane is expected to play a key role in the disease progress. Here, we present experimental data based on hybrid analysis of two-photon-microscopy, solution small-angle X-ray scattering and circular dichroism data. Data show in real time changes in liposome morphology and stability upon protein addition and reveal that membrane disruption mediated by amyloidogenic αSN is associated with dehydration of anionic lipid membranes and stimulation of protein secondary structure. As a result of membrane fragmentation, soluble αSN:-lipid coaggregates are formed, hence, suggesting a novel molecular mechanism behind PD amyloid cytotoxicity.
Collapse
Affiliation(s)
- Andreas van Maarschalkerweerd
- Department of Drug Design and Pharmacology, University of Copenhagen , Universitetsparken 2, 2100 Copenhagen, Denmark
| | | | | | | | | |
Collapse
|
65
|
From pathways to targets: understanding the mechanisms behind polyglutamine disease. BIOMED RESEARCH INTERNATIONAL 2014; 2014:701758. [PMID: 25309920 PMCID: PMC4189765 DOI: 10.1155/2014/701758] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 09/03/2014] [Indexed: 12/27/2022]
Abstract
The history of polyglutamine diseases dates back approximately 20 years to the discovery of a polyglutamine repeat in the androgen receptor of SBMA followed by the identification of similar expansion mutations in Huntington's disease, SCA1, DRPLA, and the other spinocerebellar ataxias. This common molecular feature of polyglutamine diseases suggests shared mechanisms in disease pathology and neurodegeneration of disease specific brain regions. In this review, we discuss the main pathogenic pathways including proteolytic processing, nuclear shuttling and aggregation, mitochondrial dysfunction, and clearance of misfolded polyglutamine proteins and point out possible targets for treatment.
Collapse
|
66
|
Ghilan M, Bostrom CA, Hryciw BN, Simpson JM, Christie BR, Gil-Mohapel J. YAC128 Huntington׳s disease transgenic mice show enhanced short-term hippocampal synaptic plasticity early in the course of the disease. Brain Res 2014; 1581:117-28. [DOI: 10.1016/j.brainres.2014.06.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2014] [Revised: 05/02/2014] [Accepted: 06/07/2014] [Indexed: 01/31/2023]
|
67
|
Prion-like proteins sequester and suppress the toxicity of huntingtin exon 1. Proc Natl Acad Sci U S A 2014; 111:12085-90. [PMID: 25092318 DOI: 10.1073/pnas.1412504111] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Expansions of preexisting polyglutamine (polyQ) tracts in at least nine different proteins cause devastating neurodegenerative diseases. There are many unique features to these pathologies, but there must also be unifying mechanisms underlying polyQ toxicity. Using a polyQ-expanded fragment of huntingtin exon-1 (Htt103Q), the causal protein in Huntington disease, we and others have created tractable models for investigating polyQ toxicity in yeast cells. These models recapitulate key pathological features of human diseases and provide access to an unrivalled genetic toolbox. To identify toxicity modifiers, we performed an unbiased overexpression screen of virtually every protein encoded by the yeast genome. Surprisingly, there was no overlap between our modifiers and those from a conceptually identical screen reported recently, a discrepancy we attribute to an artifact of their overexpression plasmid. The suppressors of Htt103Q toxicity recovered in our screen were strongly enriched for glutamine- and asparagine-rich prion-like proteins. Separated from the rest of the protein, the prion-like sequences of these proteins were themselves potent suppressors of polyQ-expanded huntingtin exon-1 toxicity, in both yeast and human cells. Replacing the glutamines in these sequences with asparagines abolished suppression and converted them to enhancers of toxicity. Replacing asparagines with glutamines created stronger suppressors. The suppressors (but not the enhancers) coaggregated with Htt103Q, forming large foci at the insoluble protein deposit in which proteins were highly immobile. Cells possessing foci had fewer (if any) small diffusible oligomers of Htt103Q. Until such foci were lost, cells were protected from death. We discuss the therapeutic implications of these findings.
Collapse
|
68
|
Lin XP, Feng L, Xie CG, Chen DB, Pei Z, Liang XL, Xie QY, Li XH, Pan SY. Valproic acid attenuates the suppression of acetyl histone H3 and CREB activity in an inducible cell model of Machado-Joseph disease. Int J Dev Neurosci 2014; 38:17-22. [PMID: 25068645 DOI: 10.1016/j.ijdevneu.2014.07.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Revised: 07/15/2014] [Accepted: 07/16/2014] [Indexed: 11/26/2022] Open
Abstract
Machado-Joseph disease (MJD) is caused by a (CAG)n trinucleotide repeat expansion that is translated into an abnormally long polyglutamine tract. This disease is considered the most common form of spinocerebellar ataxia (SCA). In the present study, we developed stable inducible cell lines (PC12Tet-On-Ataxin-3-Q28/84) expressing ataxin-3 with either normal or abnormal CAG repeats under doxycycline control. The expression of acetyl histone H3 and the induction of c-Fos in response to cAMP were strongly suppressed in cells expressing the protein with the expanded polyglutamine tract. Treatment with valproic acid, a histone deacetylase inhibitor (HDACi), attenuated mutant ataxin-3-induced cell toxicity and suppression of acetyl histone H3, phosphorylated cAMP-responsive element binding protein (p-CREB) as well as c-Fos expression. These results indicate that VPA can stimulate the up-regulation of gene transcription through hyperacetylation. Thus, VPA might have a therapeutic effect on MJD.
Collapse
Affiliation(s)
- X P Lin
- Department of Huiqiao Building, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - L Feng
- Department of Neurological Intensive Care Unit, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - C G Xie
- Department of Neurology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - D B Chen
- Department of Neurology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Z Pei
- Department of Neurology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - X L Liang
- Department of Neurology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Q Y Xie
- Department of Hyperbaric Oxygen Therapy, Guangzhou General Hospital of Guangzhou Military Area Command of Chinese PLA, Guangzhou, Guangdong Province, China
| | - X H Li
- Department of Neurology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China.
| | - S Y Pan
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
69
|
Ribeiro FM, Camargos ERDS, de Souza LC, Teixeira AL. Animal models of neurodegenerative diseases. BRAZILIAN JOURNAL OF PSYCHIATRY 2014; 35 Suppl 2:S82-91. [PMID: 24271230 DOI: 10.1590/1516-4446-2013-1157] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The prevalence of neurodegenerative diseases, such as Alzheimer's disease (AD) and Parkinson's disease (PD), increases with age, and the number of affected patients is expected to increase worldwide in the next decades. Accurately understanding the etiopathogenic mechanisms of these diseases is a crucial step for developing disease-modifying drugs able to preclude their emergence or at least slow their progression. Animal models contribute to increase the knowledge on the pathophysiology of neurodegenerative diseases. These models reproduce different aspects of a given disease, as well as the histopathological lesions and its main symptoms. The purpose of this review is to present the main animal models for AD, PD, and Huntington's disease.
Collapse
Affiliation(s)
- Fabíola Mara Ribeiro
- Neurobiochemistry Laboratory, Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo HorizonteMG, Brazil
| | | | | | | |
Collapse
|
70
|
Wolfe KJ, Ren HY, Trepte P, Cyr DM. Polyglutamine-rich suppressors of huntingtin toxicity act upstream of Hsp70 and Sti1 in spatial quality control of amyloid-like proteins. PLoS One 2014; 9:e95914. [PMID: 24828240 PMCID: PMC4020751 DOI: 10.1371/journal.pone.0095914] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 04/01/2014] [Indexed: 11/30/2022] Open
Abstract
Protein conformational maladies such as Huntington Disease are characterized by accumulation of intracellular and extracellular protein inclusions containing amyloid-like proteins. There is an inverse correlation between proteotoxicity and aggregation, so facilitated protein aggregation appears cytoprotective. To define mechanisms for protective protein aggregation, a screen for suppressors of nuclear huntingtin (Htt103Q) toxicity was conducted. Nuclear Htt103Q is highly toxic and less aggregation prone than its cytosolic form, so we identified suppressors of cytotoxicity caused by Htt103Q tagged with a nuclear localization signal (NLS). High copy suppressors of Htt103Q-NLS toxicity include the polyQ-domain containing proteins Nab3, Pop2, and Cbk1, and each suppresses Htt toxicity via a different mechanism. Htt103Q-NLS appears to inactivate the essential functions of Nab3 in RNA processing in the nucleus. Function of Pop2 and Cbk1 is not impaired by nuclear Htt103Q, as their respective polyQ-rich domains are sufficient to suppress Htt103Q toxicity. Pop2 is a subunit of an RNA processing complex and is localized throughout the cytoplasm. Expression of just the Pop2 polyQ domain and an adjacent proline-rich stretch is sufficient to suppress Htt103Q toxicity. The proline-rich domain in Pop2 resembles an aggresome targeting signal, so Pop2 may act in trans to positively impact spatial quality control of Htt103Q. Cbk1 accumulates in discrete perinuclear foci and overexpression of the Cbk1 polyQ domain concentrates diffuse Htt103Q into these foci, which correlates with suppression of Htt toxicity. Protective action of Pop2 and Cbk1 in spatial quality control is dependent upon the Hsp70 co-chaperone Sti1, which packages amyloid-like proteins into benign foci. Protein:protein interactions between Htt103Q and its intracellular neighbors lead to toxic and protective outcomes. A subset of polyQ-rich proteins buffer amyloid toxicity by funneling toxic aggregation intermediates to the Hsp70/Sti1 system for spatial organization into benign species.
Collapse
Affiliation(s)
- Katie J. Wolfe
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Hong Yu Ren
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Philipp Trepte
- Neuroproteomics, Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| | - Douglas M. Cyr
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
71
|
|
72
|
Miettinen MS, Monticelli L, Nedumpully-Govindan P, Knecht V, Ignatova Z. Stable polyglutamine dimers can contain β-hairpins with interdigitated side chains-but not α-helices, β-nanotubes, β-pseudohelices, or steric zippers. Biophys J 2014; 106:1721-8. [PMID: 24739171 PMCID: PMC4008795 DOI: 10.1016/j.bpj.2014.02.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 02/03/2014] [Accepted: 02/25/2014] [Indexed: 10/25/2022] Open
Abstract
A common thread connecting nine fatal neurodegenerative protein aggregation diseases is an abnormally expanded polyglutamine tract found in the respective proteins. Although the structure of this tract in the large mature aggregates is increasingly well described, its structure in the small early aggregates remains largely unknown. As experimental evidence suggests that the most toxic species along the aggregation pathway are the small early ones, developing strategies to alleviate disease pathology calls for understanding the structure of polyglutamine peptides in the early stages of aggregation. Here, we present a criterion, grounded in available experimental data, that allows for using kinetic stability of dimers to assess whether a given polyglutamine conformer can be on the aggregation path. We then demonstrate that this criterion can be assessed using present-day molecular dynamics simulations. We find that although the α-helical conformer of polyglutamine is very stable, dimers of α-helices lack the kinetic stability necessary to support further oligomerization. Dimers of steric zipper, β-nanotube, and β-pseudohelix conformers are also too short-lived to initiate aggregation. The β-hairpin-containing conformers, instead, invariably form very stable dimers when their side chains are interdigitated. Combining these findings with the implications of recent solid-state NMR data on mature fibrils, we propose a possible pathway for the initial stages of polyglutamine aggregation, in which β-hairpin-containing conformers act as templates for fibril formation.
Collapse
Affiliation(s)
- Markus S Miettinen
- Fachbereich Physik, Freie Universität Berlin, Berlin, Germany; Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany.
| | - Luca Monticelli
- Institut National de la santé et de la recherche medicale, Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Paris, France; Institut National de la Transfusion Sanguine, Paris, France
| | | | - Volker Knecht
- Department of Theory and Bio-Systems, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Zoya Ignatova
- Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany.
| |
Collapse
|
73
|
Kim S, Kim KT. Therapeutic Approaches for Inhibition of Protein Aggregation in Huntington's Disease. Exp Neurobiol 2014; 23:36-44. [PMID: 24737938 PMCID: PMC3984955 DOI: 10.5607/en.2014.23.1.36] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 12/19/2013] [Accepted: 12/20/2013] [Indexed: 11/19/2022] Open
Abstract
Huntington's disease (HD) is a late-onset and progressive neurodegenerative disorder that is caused by aggregation of mutant huntingtin protein which contains expanded-polyglutamine. The molecular chaperones modulate the aggregation in early stage and known for the most potent protector of neurodegeneration in animal models of HD. Over the past decades, a number of studies have demonstrated molecular chaperones alleviate the pathogenic symptoms by polyQ-mediated toxicity. Moreover, chaperone-inducible drugs and anti-aggregation drugs have beneficial effects on symptoms of disease. Here, we focus on the function of molecular chaperone in animal models of HD, and review the recent therapeutic approaches to modulate expression and turn-over of molecular chaperone and to develop anti-aggregation drugs.
Collapse
Affiliation(s)
- Sangjune Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Korea
| | - Kyong-Tai Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Korea. ; Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang 790-784, Korea
| |
Collapse
|
74
|
Hoffner G, Djian P. Monomeric, oligomeric and polymeric proteins in huntington disease and other diseases of polyglutamine expansion. Brain Sci 2014; 4:91-122. [PMID: 24961702 PMCID: PMC4066239 DOI: 10.3390/brainsci4010091] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 02/06/2014] [Accepted: 02/18/2014] [Indexed: 01/03/2023] Open
Abstract
Huntington disease and other diseases of polyglutamine expansion are each caused by a different protein bearing an excessively long polyglutamine sequence and are associated with neuronal death. Although these diseases affect largely different brain regions, they all share a number of characteristics, and, therefore, are likely to possess a common mechanism. In all of the diseases, the causative protein is proteolyzed, becomes abnormally folded and accumulates in oligomers and larger aggregates. The aggregated and possibly the monomeric expanded polyglutamine are likely to play a critical role in the pathogenesis and there is increasing evidence that the secondary structure of the protein influences its toxicity. We describe here, with special attention to huntingtin, the mechanisms of polyglutamine aggregation and the modulation of aggregation by the sequences flanking the polyglutamine. We give a comprehensive picture of the characteristics of monomeric and aggregated polyglutamine, including morphology, composition, seeding ability, secondary structure, and toxicity. The structural heterogeneity of aggregated polyglutamine may explain why polyglutamine-containing aggregates could paradoxically be either toxic or neuroprotective.
Collapse
Affiliation(s)
- Guylaine Hoffner
- Génétique moléculaire et défense antivirale, Centre National de la Recherche Scientifique, Université Paris Descartes, 45 rue des Saints Pères, 75006 Paris, France.
| | - Philippe Djian
- Génétique moléculaire et défense antivirale, Centre National de la Recherche Scientifique, Université Paris Descartes, 45 rue des Saints Pères, 75006 Paris, France.
| |
Collapse
|
75
|
Wan J, Savas JN, Roth AF, Sanders SS, Singaraja RR, Hayden MR, Yates JR, Davis NG. Tracking brain palmitoylation change: predominance of glial change in a mouse model of Huntington's disease. CHEMISTRY & BIOLOGY 2013; 20:1421-34. [PMID: 24211138 PMCID: PMC3880188 DOI: 10.1016/j.chembiol.2013.09.018] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Revised: 08/28/2013] [Accepted: 09/08/2013] [Indexed: 11/25/2022]
Abstract
Protein palmitoylation, a reversible lipid modification of proteins, is widely used in the nervous system, with dysregulated palmitoylation being implicated in a variety of neurological disorders. Described below is ABE/SILAM, a proteomic strategy that couples acyl-biotinyl exchange (ABE) purification of palmitoyl-proteins to whole animal stable isotope labeling (SILAM) to provide an accurate tracking of palmitoylation change within rodent disease models. As a first application, we have used ABE/SILAM to look at Huntington's disease (HD), profiling palmitoylation change in two HD-relevant mouse mutants: the transgenic HD model mouse YAC128 and the hypomorphic Hip14-gt mouse, which has sharply reduced expression for HIP14 (Zdhhc17), a palmitoyl-transferase implicated in the HD disease process. Rather than mapping to the degenerating neurons themselves, the biggest disease changes instead map to astrocytes and oligodendrocytes (i.e., the supporting glial cells).
Collapse
Affiliation(s)
- Junmei Wan
- Department of Pharmacology, Wayne State University, Detroit, MI 48201, USA
| | - Jeffrey N. Savas
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Amy F. Roth
- Department of Pharmacology, Wayne State University, Detroit, MI 48201, USA
| | - Shaun S. Sanders
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia V5Z 4H4 Canada
| | - Roshni R. Singaraja
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia V5Z 4H4 Canada
| | - Michael R. Hayden
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia V5Z 4H4 Canada
| | - John R. Yates
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Nicholas G. Davis
- Department of Pharmacology, Wayne State University, Detroit, MI 48201, USA
| |
Collapse
|
76
|
Bard J, Wall MD, Lazari O, Arjomand J, Munoz-Sanjuan I. Advances in huntington disease drug discovery: novel approaches to model disease phenotypes. ACTA ACUST UNITED AC 2013; 19:191-204. [PMID: 24196395 DOI: 10.1177/1087057113510320] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Huntington disease is a monogenic, autosomal dominant, progressive neurodegenerative disorder caused by a trinucleotide CAG repeat expansion in exon 1 of the huntingtin (HTT) gene; age of onset of clinical symptoms inversely correlates with expanded CAG repeat length. HD leads to extensive degeneration of the basal ganglia, hypothalamic nuclei, and selected cortical areas, and a wide range of molecular mechanisms have been implicated in disease pathology in animal or cellular models expressing mutated HTT (mHTT) proteins, either full-length or amino-terminal fragments. However, HD cellular models that recapitulate the slow progression of the disease have not been available due to the toxicity of overexpressed exogenous mHTT or to limitations with using primary cells for long-term studies. Most investigations of the effects of mHTT relied on cytotoxicity or aggregation end points in heterologous systems or in primary embryonic neuroglial cultures derived from HD mouse models. More innovative approaches are currently under active investigation, including screening using electrophysiological endpoints, as well as the recent use of primary blood mononuclear cells and of human embryonic stem cells derived from a variety of HD research participants. Here we describe how these cellular systems are being used to investigate HD biology as well as to identify mechanisms with therapeutic potential.
Collapse
Affiliation(s)
- Jonathan Bard
- 1CHDI Management/CHDI Foundation, Princeton, NJ, and Los Angeles, CA, USA
| | | | | | | | | |
Collapse
|
77
|
|
78
|
Krench M, Littleton JT. Modeling Huntington disease in Drosophila: Insights into axonal transport defects and modifiers of toxicity. Fly (Austin) 2013; 7:229-36. [PMID: 24022020 DOI: 10.4161/fly.26279] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Huntington disease (HD) is an inherited neurodegenerative disorder caused by a polyglutamine (polyQ) expansion in the huntingtin (Htt) gene. Despite years of research, there is no treatment that extends life for patients with the disorder. Similarly, little is known about which cellular pathways that are altered by pathogenic Huntingtin (Htt) protein expression are correlated with neuronal loss. As part of a longstanding effort to gain insights into HD pathology, we have been studying the protein in the context of the fruitfly Drosophila melanogaster. We generated transgenic HD models in Drosophila by engineering flies that carry a 12-exon fragment of the human Htt gene with or without the toxic trinucleotide repeat expansion. We also created variants with a monomeric red fluorescent protein (mRFP) tag fused to Htt that allows in vivo imaging of Htt protein localization and aggregation. While wild-type Htt remains diffuse throughout the cytoplasm of cells, pathogenic Htt forms insoluble aggregates that accumulate in neuronal soma and axons. Aggregates can physically block transport of numerous organelles along the axon. We have also observed that aggregates are formed quickly, within just a few hours of mutant Htt expression. To explore mechanisms of neurodegeneration in our HD model, we performed in vivo and in vitro screens to search for modifiers of viability and pathogenic Htt aggregation. Our results identified several novel candidates for HD therapeutics that can now be tested in mammalian models of HD. Furthermore, these experiments have highlighted the complex relationship between aggregates and toxicity that exists in HD.
Collapse
Affiliation(s)
- Megan Krench
- The Picower Institute for Learning and Memory; Department of Brain and Cognitive Sciences; Department of Biology; Massachusetts Institute of Technology; Cambridge, MA USA
| | - J Troy Littleton
- The Picower Institute for Learning and Memory; Department of Brain and Cognitive Sciences; Department of Biology; Massachusetts Institute of Technology; Cambridge, MA USA
| |
Collapse
|
79
|
Tsvetkov AS, Arrasate M, Barmada S, Ando DM, Sharma P, Shaby BA, Finkbeiner S. Proteostasis of polyglutamine varies among neurons and predicts neurodegeneration. Nat Chem Biol 2013; 9:586-92. [PMID: 23873212 DOI: 10.1038/nchembio.1308] [Citation(s) in RCA: 142] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Accepted: 06/25/2013] [Indexed: 01/28/2023]
Abstract
In polyglutamine (polyQ) diseases, only certain neurons die, despite widespread expression of the offending protein. PolyQ expansion may induce neurodegeneration by impairing proteostasis, but protein aggregation and toxicity tend to confound conventional measurements of protein stability. Here, we used optical pulse labeling to measure effects of polyQ expansions on the mean lifetime of a fragment of huntingtin, the protein that causes Huntington's disease, in living neurons. We show that polyQ expansion reduced the mean lifetime of mutant huntingtin within a given neuron and that the mean lifetime varied among neurons, indicating differences in their capacity to clear the polypeptide. We found that neuronal longevity is predicted by the mean lifetime of huntingtin, as cortical neurons cleared mutant huntingtin faster and lived longer than striatal neurons. Thus, cell type-specific differences in turnover capacity may contribute to cellular susceptibility to toxic proteins, and efforts to bolster proteostasis in Huntington's disease, such as protein clearance, could be neuroprotective.
Collapse
Affiliation(s)
- Andrey S Tsvetkov
- 1] Gladstone Institute of Neurological Disease, San Francisco, California, USA. [2] Taube-Koret Center for Neurodegenerative Disease Research, San Francisco, California, USA
| | | | | | | | | | | | | |
Collapse
|
80
|
Suppressing aberrant GluN3A expression rescues synaptic and behavioral impairments in Huntington's disease models. Nat Med 2013; 19:1030-8. [PMID: 23852340 PMCID: PMC3936794 DOI: 10.1038/nm.3246] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 05/20/2013] [Indexed: 02/08/2023]
Abstract
Huntington's disease is caused by an expanded polyglutamine repeat in the huntingtin protein (HTT), but the pathophysiological sequence of events that trigger synaptic failure and neuronal loss are not fully understood. Alterations in N-methyl-D-aspartate (NMDA)-type glutamate receptors (NMDARs) have been implicated. Yet, it remains unclear how the HTT mutation affects NMDAR function, and direct evidence for a causative role is missing. Here we show that mutant HTT redirects an intracellular store of juvenile NMDARs containing GluN3A subunits to the surface of striatal neurons by sequestering and disrupting the subcellular localization of the endocytic adaptor PACSIN1, which is specific for GluN3A. Overexpressing GluN3A in wild-type mouse striatum mimicked the synapse loss observed in Huntington's disease mouse models, whereas genetic deletion of GluN3A prevented synapse degeneration, ameliorated motor and cognitive decline and reduced striatal atrophy and neuronal loss in the YAC128 Huntington's disease mouse model. Furthermore, GluN3A deletion corrected the abnormally enhanced NMDAR currents, which have been linked to cell death in Huntington's disease and other neurodegenerative conditions. Our findings reveal an early pathogenic role of GluN3A dysregulation in Huntington's disease and suggest that therapies targeting GluN3A or pathogenic HTT-PACSIN1 interactions might prevent or delay disease progression.
Collapse
|
81
|
Arribat Y, Bonneaud N, Talmat-Amar Y, Layalle S, Parmentier ML, Maschat F. A huntingtin peptide inhibits polyQ-huntingtin associated defects. PLoS One 2013; 8:e68775. [PMID: 23861941 PMCID: PMC3701666 DOI: 10.1371/journal.pone.0068775] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 06/06/2013] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Huntington's disease (HD) is caused by the abnormal expansion of the polyglutamine tract in the human Huntingtin protein (polyQ-hHtt). Although this mutation behaves dominantly, huntingtin loss of function also contributes to HD pathogenesis. Indeed, wild-type Huntingtin plays a protective role with respect to polyQ-hHtt induced defects. METHODOLOGY/PRINCIPAL FINDINGS The question that we addressed here is what part of the wild-type Huntingtin is responsible for these protective properties. We first screened peptides from the Huntingtin protein in HeLa cells and identified a 23 aa peptide (P42) that inhibits polyQ-hHtt aggregation. P42 is part of the endogenous Huntingtin protein and lies within a region rich in proteolytic sites that plays a critical role in the pathogenesis process. Using a Drosophila model of HD, we tested the protective properties of this peptide on aggregation, as well as on different polyQ-hHtt induced neuronal phenotypes: eye degeneration (an indicator of cell death), impairment of vesicular axonal trafficking, and physiological behaviors such as larval locomotion and adult survival. Together, our results demonstrate high protective properties for P42 in vivo, in whole animals. These data also demonstrate a specific role of P42 on Huntington's disease model, since it has no effect on other models of polyQ-induced diseases, such as spinocerebellar ataxias. CONCLUSIONS/SIGNIFICANCE Altogether our data show that P42, a 23 aa-long hHtt peptide, plays a protective role with respect to polyQ-hHtt aggregation as well as cellular and behavioral dysfunctions induced by polyQ-hHtt in vivo. Our study also confirms the correlation between polyQ-hHtt aggregation and neuronal defects. Finally, these results strongly suggest a therapeutic potential for P42, specific of Huntington's disease.
Collapse
Affiliation(s)
- Yoan Arribat
- Institut de Génomique Fonctionnelle (IGF), CNRS-UMR5203, INSERM-U661, University of Montpellier, Montpellier, France
| | - Nathalie Bonneaud
- Institut de Génomique Fonctionnelle (IGF), CNRS-UMR5203, INSERM-U661, University of Montpellier, Montpellier, France
| | - Yasmina Talmat-Amar
- Institut de Génomique Fonctionnelle (IGF), CNRS-UMR5203, INSERM-U661, University of Montpellier, Montpellier, France
| | - Sophie Layalle
- Institut de Génomique Fonctionnelle (IGF), CNRS-UMR5203, INSERM-U661, University of Montpellier, Montpellier, France
| | - Marie-Laure Parmentier
- Institut de Génomique Fonctionnelle (IGF), CNRS-UMR5203, INSERM-U661, University of Montpellier, Montpellier, France
- * E-mail: (FM); (MLP)
| | - Florence Maschat
- Institut de Génomique Fonctionnelle (IGF), CNRS-UMR5203, INSERM-U661, University of Montpellier, Montpellier, France
- * E-mail: (FM); (MLP)
| |
Collapse
|
82
|
Chort A, Alves S, Marinello M, Dufresnois B, Dornbierer JG, Tesson C, Latouche M, Baker DP, Barkats M, El Hachimi KH, Ruberg M, Janer A, Stevanin G, Brice A, Sittler A. Interferon beta induces clearance of mutant ataxin 7 and improves locomotion in SCA7 knock-in mice. Brain 2013; 136:1732-45. [DOI: 10.1093/brain/awt061] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
83
|
Wang ZM, Lashuel HA. Discovery of a Novel Aggregation Domain in the Huntingtin Protein: Implications for the Mechanisms of Htt Aggregation and Toxicity. Angew Chem Int Ed Engl 2013. [DOI: 10.1002/ange.201206561] [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]
|
84
|
Tsvetkov AS, Ando DM, Finkbeiner S. Longitudinal imaging and analysis of neurons expressing polyglutamine-expanded proteins. Methods Mol Biol 2013; 1017:1-20. [PMID: 23719904 DOI: 10.1007/978-1-62703-438-8_1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Misfolded proteins have been implicated in most of the major neurodegenerative diseases, and identifying drugs and pathways that protect neurons from the toxicity of misfolded proteins is of paramount importance. We invented a form of automated imaging and analysis called robotic microscopy that is well suited to the study of neurodegeneration. It enables the monitoring of large cohorts of individual neurons over their lifetimes as they undergo neurodegeneration. With automated analysis, multiple endpoints in neurons can be measured, including survival. Statistical approaches, typically reserved for engineering and clinical medicine, can be applied to these data in an unbiased fashion to discover whether factors contribute positively or negatively to neuronal fate and to quantify the importance of their contribution. Ultimately, multivariate dynamic models can be constructed from these data, which can provide a systems-level understanding of the neurodegenerative disease process and guide the rationale for the development of therapies.
Collapse
|
85
|
Wang ZM, Lashuel HA. Discovery of a Novel Aggregation Domain in the Huntingtin Protein: Implications for the Mechanisms of Htt Aggregation and Toxicity. Angew Chem Int Ed Engl 2012; 52:562-7. [DOI: 10.1002/anie.201206561] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Indexed: 11/11/2022]
|
86
|
Figiel M, Szlachcic WJ, Switonski PM, Gabka A, Krzyzosiak WJ. Mouse models of polyglutamine diseases: review and data table. Part I. Mol Neurobiol 2012; 46:393-429. [PMID: 22956270 PMCID: PMC3461215 DOI: 10.1007/s12035-012-8315-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2012] [Accepted: 07/29/2012] [Indexed: 12/23/2022]
Abstract
Polyglutamine (polyQ) disorders share many similarities, such as a common mutation type in unrelated human causative genes, neurological character, and certain aspects of pathogenesis, including morphological and physiological neuronal alterations. The similarities in pathogenesis have been confirmed by findings that some experimental in vivo therapy approaches are effective in multiple models of polyQ disorders. Additionally, mouse models of polyQ diseases are often highly similar between diseases with respect to behavior and the features of the disease. The common features shared by polyQ mouse models may facilitate the investigation of polyQ disorders and may help researchers explore the mechanisms of these diseases in a broader context. To provide this context and to promote the understanding of polyQ disorders, we have collected and analyzed research data about the characterization and treatment of mouse models of polyQ diseases and organized them into two complementary Excel data tables. The data table that is presented in this review (Part I) covers the behavioral, molecular, cellular, and anatomic characteristics of polyQ mice and contains the most current knowledge about polyQ mouse models. The structure of this data table is designed in such a way that it can be filtered to allow for the immediate retrieval of the data corresponding to a single mouse model or to compare the shared and unique aspects of many polyQ models. The second data table, which is presented in another publication (Part II), covers therapeutic research in mouse models by summarizing all of the therapeutic strategies employed in the treatment of polyQ disorders, phenotypes that are used to examine the effects of the therapy, and therapeutic outcomes.
Collapse
Affiliation(s)
- Maciej Figiel
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland.
| | | | | | | | | |
Collapse
|
87
|
Kraut DA, Israeli E, Schrader EK, Patil A, Nakai K, Nanavati D, Inobe T, Matouschek A. Sequence- and species-dependence of proteasomal processivity. ACS Chem Biol 2012; 7:1444-53. [PMID: 22716912 DOI: 10.1021/cb3001155] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The proteasome is the degradation machine at the center of the ubiquitin-proteasome system and controls the concentrations of many proteins in eukaryotes. It is highly processive so that substrates are degraded completely into small peptides, avoiding the formation of potentially toxic fragments. Nonetheless, some proteins are incompletely degraded, indicating the existence of factors that influence proteasomal processivity. We have quantified proteasomal processivity and determined the underlying rates of substrate degradation and release. We find that processivity increases with species complexity over a 5-fold range between yeast and mammalian proteasome, and the effect is due to slower but more persistent degradation by proteasomes from more complex organisms. A sequence stretch that has been implicated in causing incomplete degradation, the glycine-rich region of the NFκB subunit p105, reduces the proteasome's ability to unfold its substrate, and polyglutamine repeats such as found in Huntington's disease reduce the processivity of the proteasome in a length-dependent manner.
Collapse
Affiliation(s)
- Daniel A. Kraut
- Department of Molecular Biosciences, Northwestern University, Evanston, Illinois 60208,
United States
- Department of Chemistry, Villanova University, Villanova, Pennsylvania 19085,
United States
| | - Eitan Israeli
- Department of Molecular Biosciences, Northwestern University, Evanston, Illinois 60208,
United States
| | - Erin K. Schrader
- Department of Molecular Biosciences, Northwestern University, Evanston, Illinois 60208,
United States
| | - Ashwini Patil
- Human Genome
Center, The Institute
of Medical Science, The University of Tokyo, 4-6-1 Shirokane-dai, Minato-ku, Tokyo, Japan
| | - Kenta Nakai
- Human Genome
Center, The Institute
of Medical Science, The University of Tokyo, 4-6-1 Shirokane-dai, Minato-ku, Tokyo, Japan
| | - Dhaval Nanavati
- Proteomics Core Facility, Chemistry
of Life Processes Institute, Northwestern University, Evanston, Illinois 60208, United States
| | - Tomonao Inobe
- Frontier Research Core for Life
Sciences, University of Toyama, 3190 Gofuku,
Toyama-shi, Toyama 930-8555, Japan
| | - Andreas Matouschek
- Department of Molecular Biosciences, Northwestern University, Evanston, Illinois 60208,
United States
| |
Collapse
|
88
|
Zheng S, Ghitani N, Blackburn JS, Liu JP, Zeitlin SO. A series of N-terminal epitope tagged Hdh knock-in alleles expressing normal and mutant huntingtin: their application to understanding the effect of increasing the length of normal Huntingtin's polyglutamine stretch on CAG140 mouse model pathogenesis. Mol Brain 2012; 5:28. [PMID: 22892315 PMCID: PMC3499431 DOI: 10.1186/1756-6606-5-28] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Accepted: 08/09/2012] [Indexed: 12/19/2022] Open
Abstract
Background Huntington’s disease (HD) is an autosomal dominant neurodegenerative disease that is caused by the expansion of a polyglutamine (polyQ) stretch within Huntingtin (htt), the protein product of the HD gene. Although studies in vitro have suggested that the mutant htt can act in a potentially dominant negative fashion by sequestering wild-type htt into insoluble protein aggregates, the role of the length of the normal htt polyQ stretch, and the adjacent proline-rich region (PRR) in modulating HD mouse model pathogenesis is currently unknown. Results We describe the generation and characterization of a series of knock-in HD mouse models that express versions of the mouse HD gene (Hdh) encoding N-terminal hemaglutinin (HA) or 3xFlag epitope tagged full-length htt with different polyQ lengths (HA7Q-, 3xFlag7Q-, 3xFlag20Q-, and 3xFlag140Q-htt) and substitution of the adjacent mouse PRR with the human PRR (3xFlag20Q- and 3xFlag140Q-htt). Using co-immunoprecipitation and immunohistochemistry analyses, we detect no significant interaction between soluble full-length normal 7Q- htt and mutant (140Q) htt, but we do observe N-terminal fragments of epitope-tagged normal htt in mutant htt aggregates. When the sequences encoding normal mouse htt’s polyQ stretch and PRR are replaced with non-pathogenic human sequence in mice also expressing 140Q-htt, aggregation foci within the striatum, and the mean size of htt inclusions are increased, along with an increase in striatal lipofuscin and gliosis. Conclusion In mice, soluble full-length normal and mutant htt are predominantly monomeric. In heterozygous knock-in HD mouse models, substituting the normal mouse polyQ and PRR with normal human sequence can exacerbate some neuropathological phenotypes.
Collapse
Affiliation(s)
- Shuqiu Zheng
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA 22908, Box 801392, USA
| | | | | | | | | |
Collapse
|
89
|
Gong B, Kielar C, Morton AJ. Temporal separation of aggregation and ubiquitination during early inclusion formation in transgenic mice carrying the Huntington's disease mutation. PLoS One 2012; 7:e41450. [PMID: 22848498 PMCID: PMC3404089 DOI: 10.1371/journal.pone.0041450] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Accepted: 06/26/2012] [Indexed: 12/01/2022] Open
Abstract
Abnormal insoluble ubiqitinated protein aggregates are found in the brains of Huntington’s disease (HD) patients and in mice transgenic for the HTT mutation. Here, we describe the earliest stages of visible NII formation in brains of R6/2 mice killed between 2 and 6 weeks of age. We found that huntingtin-positive aggregates formed rapidly (within 24–48 hours) in a spatiotemporal manner similar to that we described previously for ubiquitinated inclusions. However, in most neurons, aggregates are not ubiquitinated when they first form. It has always been assumed that mutant huntingtin is recognised as ‘foreign’ and consequently ubiquitinated and targeted for degradation by the ubiquitin-proteasome system pathway. Our data, however, suggest that aggregation and ubiquitination are separate processes, and that mutant huntingtin fragment is not recognized as ‘abnormal’ by the ubiquitin-proteasome system before aggregation. Rather, mutant Htt appears to aggregate before it is ubiquitinated, and then either aggregated huntingtin is ubiquitinated or ubiquitinated proteins are recruited into aggregates. Our findings have significant implications for the role of the ubiquitin-proteasome system in the formation of aggregates, as they suggest that this system is not involved until after the first aggregates form.
Collapse
Affiliation(s)
- Belvin Gong
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
- UC Davis/NIH NeuroMab Facility, Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, California, United States of America
| | - Catherine Kielar
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - A. Jennifer Morton
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
- * E-mail:
| |
Collapse
|
90
|
Guerriero CJ, Brodsky JL. The delicate balance between secreted protein folding and endoplasmic reticulum-associated degradation in human physiology. Physiol Rev 2012; 92:537-76. [PMID: 22535891 DOI: 10.1152/physrev.00027.2011] [Citation(s) in RCA: 314] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Protein folding is a complex, error-prone process that often results in an irreparable protein by-product. These by-products can be recognized by cellular quality control machineries and targeted for proteasome-dependent degradation. The folding of proteins in the secretory pathway adds another layer to the protein folding "problem," as the endoplasmic reticulum maintains a unique chemical environment within the cell. In fact, a growing number of diseases are attributed to defects in secretory protein folding, and many of these by-products are targeted for a process known as endoplasmic reticulum-associated degradation (ERAD). Since its discovery, research on the mechanisms underlying the ERAD pathway has provided new insights into how ERAD contributes to human health during both normal and diseases states. Links between ERAD and disease are evidenced from the loss of protein function as a result of degradation, chronic cellular stress when ERAD fails to keep up with misfolded protein production, and the ability of some pathogens to coopt the ERAD pathway. The growing number of ERAD substrates has also illuminated the differences in the machineries used to recognize and degrade a vast array of potential clients for this pathway. Despite all that is known about ERAD, many questions remain, and new paradigms will likely emerge. Clearly, the key to successful disease treatment lies within defining the molecular details of the ERAD pathway and in understanding how this conserved pathway selects and degrades an innumerable cast of substrates.
Collapse
Affiliation(s)
- Christopher J Guerriero
- Department of Biological Sciences, University of Pittsburgh, A320 Langley Hall, Pittsburgh, PA 15260, USA
| | | |
Collapse
|
91
|
Xiao H, Yu Z, Wu Y, Nan J, Merry DE, Sekiguchi JM, Ferguson DO, Lieberman AP, Dressler GR. A polyglutamine expansion disease protein sequesters PTIP to attenuate DNA repair and increase genomic instability. Hum Mol Genet 2012; 21:4225-36. [PMID: 22736030 DOI: 10.1093/hmg/dds246] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Glutamine (Q) expansion diseases are a family of degenerative disorders caused by the lengthening of CAG triplet repeats present in the coding sequences of seemingly unrelated genes whose mutant proteins drive pathogenesis. Despite all the molecular evidence for the genetic basis of these diseases, how mutant poly-Q proteins promote cell death and drive pathogenesis remains controversial. In this report, we show a specific interaction between the mutant androgen receptor (AR), a protein associated with spinal and bulbar muscular atrophy (SBMA), and the nuclear protein PTIP (Pax Transactivation-domain Interacting Protein), a protein with an unusually long Q-rich domain that functions in DNA repair. Upon exposure to ionizing radiation, PTIP localizes to nuclear foci that are sites of DNA damage and repair. However, the expression of poly-Q AR sequesters PTIP away from radiation-induced nuclear foci. This results in sensitivity to DNA-damaging agents and chromosomal instabilities. In a mouse model of SBMA, evidence for DNA damage is detected in muscle cell nuclei and muscular atrophy is accelerated when one copy of the gene encoding PTIP is removed. These data provide a new paradigm for understanding the mechanisms of cellular degeneration observed in poly-Q expansion diseases.
Collapse
Affiliation(s)
- Hong Xiao
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Brooks S, Higgs G, Janghra N, Jones L, Dunnett SB. Longitudinal analysis of the behavioural phenotype in YAC128 (C57BL/6J) Huntington's disease transgenic mice. Brain Res Bull 2012; 88:113-20. [PMID: 20460143 DOI: 10.1016/j.brainresbull.2010.05.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2010] [Revised: 04/27/2010] [Accepted: 05/02/2010] [Indexed: 10/19/2022]
Abstract
To determine the suitability of mouse models of disease for therapeutic trials, the models must be characterised to determine their similarity to the human condition, and utility for specific therapeutic approaches. The YAC128 mouse model of HD has been bred on to C57BL/6J background in order to provide a mouse model of the disease better suited to behavioural testing, than the visually impaired original line on the FVB background. In the present study, the C57BL/6J YAC128 mice were assessed on several behavioural tasks bi-monthly between 4 and 24 months of age. On the rotarod early and stable deficits were demonstrated in the YAC128 mice from 4 months of age indicating an early abnormality in motor coordination. Early and stable deficits were also found on the balance beam measures of latency to orientate towards the beam and time to traverse it. Measures of fore and hind limb footslips on the balance beam demonstrated early and progressive limb use deficits in the YAC128 mice. On a 3-stage Morris water maze protocol, the YAC128 mice took longer and travelled further to find the hidden platform in each of the 3 locations, indicative of a spatial learning deficit. The YAC128 mice were also less reactive to the primary startle stimuli and the effects of the prepulse which may suggest striatal dysfunction. As a measure of general well being, the body weights of the mice were recorded and demonstrated increased weight in the YAC128 mice until 14 months of age, when they became comparable to that of their wildtype littermates. The YAC128 mouse on the C57BL/6J background has an early, robust and severe behavioural phenotype that shares some similarity to human HD symptomatology.
Collapse
Affiliation(s)
- Simon Brooks
- Brain Repair Group, School of Biosciences, Cardiff University, Wales, UK.
| | | | | | | | | |
Collapse
|
93
|
Bowles KR, Brooks SP, Dunnett SB, Jones L. Gene expression and behaviour in mouse models of HD. Brain Res Bull 2012; 88:276-84. [PMID: 21854837 DOI: 10.1016/j.brainresbull.2011.07.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Revised: 07/27/2011] [Accepted: 07/31/2011] [Indexed: 01/09/2023]
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disease, resulting in expansion of the CAG repeat in exon 1 of the HTT gene. The resulting mutant huntingtin protein has been implicated in the disruption of a variety of cellular functions, including transcription. Mouse models of HD have been central to the development of our understanding of gene expression changes in this disease, and are now beginning to elucidate the relationship between gene expression and behaviour. Here, we review current mouse models of HD and their characterisation in terms of gene expression. In addition, we look at how this can inform behaviours observed in mouse models of disease. The relationship between gene expression and behaviour in mouse models of HD is important, as this will further our knowledge of disease progression and its underlying molecular events, highlight new treatment targets, and potentially provide new biomarkers for therapeutic trials.
Collapse
Affiliation(s)
- K R Bowles
- Department of Psychological Medicine, MRC centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Wales, UK
| | | | | | | |
Collapse
|
94
|
Brooks SP, Janghra N, Higgs GV, Bayram-Weston Z, Heuer A, Jones L, Dunnett SB. Selective cognitive impairment in the YAC128 Huntington's disease mouse. Brain Res Bull 2012; 88:121-9. [PMID: 21624441 DOI: 10.1016/j.brainresbull.2011.05.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Revised: 05/11/2011] [Accepted: 05/13/2011] [Indexed: 01/01/2023]
Abstract
People with HD have a demonstrated early extra-dimensional set-shifting deficit. In the present study, we use a novel water T-maze set-shifting procedure and demonstrate its validity as a set-shifting task in a mouse model of Huntington's disease. Three groups of YAC128 mice of different ages (27, 69 and 117 weeks) were run on the task, which incorporated six distinct stages in which the mice must learn a rule and then switch to a different rule. The six stages were: directional learning, directional learning reversal, light discrimination, light discrimination reversal, return to place learning and a maze rotation spatial learning test. Rule changes from place learning to light discrimination and back constitute extra-dimensional shifts. The results of the study demonstrate robust light/dark discrimination reversal learning deficits in transgenic mice from 27 weeks of age, and a directional learning to light discrimination extra-dimensional set-shifting deficit from 69 weeks of age. The extra-dimensional shift deficit was confirmed with control trials demonstrating the validity of the deficit and the task. The onset of reversal learning and extra-dimensional shift deficits corresponded with the development of mutant huntingtin N-terminal fragment aggregates in neurons of relevant forebrain regions.
Collapse
Affiliation(s)
- Simon P Brooks
- Brain Repair Group, School of Biosciences, Cardiff University, Wales, UK.
| | | | | | | | | | | | | |
Collapse
|
95
|
Trueman RC, Jones L, Dunnett SB, Brooks SP. Early onset deficits on the delayed alternation task in the Hdh(Q92) knock-in mouse model of Huntington's disease. Brain Res Bull 2012; 88:156-62. [PMID: 21440047 DOI: 10.1016/j.brainresbull.2011.03.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2010] [Revised: 03/14/2011] [Accepted: 03/18/2011] [Indexed: 10/18/2022]
Abstract
A number of genetic mouse models of Huntington's disease have been created, in order to examine the pathogenesis of Huntington's disease and to test potential therapeutics. In the present study we demonstrate that the full-length knock-in homozygote Hdh(Q92) mice exhibit impairments at 5 months of age on the delayed alternation task, conducted in 9-hole operant chambers. This test is sensitive to cortico-striatal dysfunction and demonstrates again that although Hdh(Q92) mice do not display an overt motor phenotype, they do exhibit clear impairments that can be related to deficits seen in HD patients. This indicates that if appropriately sensitive tasks are used, the more subtle and specific Hdh(Q92) knock-in model could be of use for the examination of pathogenic mechanisms in Huntington's disease and to test potential therapeutics.
Collapse
Affiliation(s)
- R C Trueman
- Brain Repair Group, School of Biosciences, Cardiff University, Wales, UK.
| | | | | | | |
Collapse
|
96
|
Gil-Mohapel JM. Screening of therapeutic strategies for Huntington's disease in YAC128 transgenic mice. CNS Neurosci Ther 2012; 18:77-86. [PMID: 21501423 DOI: 10.1111/j.1755-5949.2011.00246.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Huntington’s disease (HD) is a hereditary neurodegenerative disorder caused by an unstable expansion of cytosine-adenine-guanine (CAG) repeats in the HD gene. The symptoms include cognitive dysfunction and severe motor impairment with loss of voluntary movement coordination that is later replaced by bradykinesia and rigidity. The neuropathology is characterized by neuronal loss mainly in the striatum and cortex, and the appearance of neuronal intranuclear inclusions of mutant huntingtin. The mechanisms responsible for neurodegeneration are still not fully understood although excitotoxicity and a consequent increase in intracellular calcium concentration as well as the activation of caspases and calapins are known to play a key role. There is currently no satisfactory treatment or cure for this disease. The YAC128 transgenic mice express the full-length human HD gene with 128 CAG repeats and constitute a unique model for the study of HD as they replicate the slow and biphasic progression of behavioral deficits characteristic of the human condition and show striatal neuronal loss. As such, these transgenic mice have been an invaluable model not only for the elucidation of the neurodegenerative pathways in HD, but also for the screening and development of new therapeutic approaches. Here, I will review the unique characteristics of this transgenic HD model and will provide a summary of the therapies that have been tested in these mice, namely: potentiation of the protective roles of wild-type huntingtin and mutant huntingtin aggregation, transglutaminase inhibition, inhibition of glutamate- and dopamine-induced toxicity, apoptosis inhibition, use of essential fatty acids, and the novel approach of intrabody gene therapy. The insights obtained from these and future studies will help identify potential candidates for clinical trials and will ultimately contribute to the discovery of a successful treatment for this devastating neurodegenerative disorder.
Collapse
Affiliation(s)
- Joana M Gil-Mohapel
- Division of Medical Sciences, Island Medical Program, University of Victoria, British Columbia, Canada.
| |
Collapse
|
97
|
Ehrlich ME. Huntington's disease and the striatal medium spiny neuron: cell-autonomous and non-cell-autonomous mechanisms of disease. Neurotherapeutics 2012; 9:270-84. [PMID: 22441874 PMCID: PMC3337013 DOI: 10.1007/s13311-012-0112-2] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Huntington's disease is an autosomal dominant disorder caused by a mutation in the gene encoding the protein huntingtin on chromosome 4. The mutation is an expanded CAG repeat in the first exon, encoding a polyglutamine tract. If the polyglutamine tract is > 40, penetrance is 100% and death is inevitable. Despite the widespread expression of huntingtin, HD has long been considered primarily as a disease of the striatum. It is characterized by selective vulnerability with dysfunction followed by death of the medium size spiny neuron. Considerable effort is being expended to determine whether striatal damage is cell-autonomous, non-cell-autonomous, requiring cell-cell and region to region communication, or both. We review data supporting both mechanisms. We also attempt to organize the data into common mechanisms that may arise outside the medium, spiny neuron, but ultimately have their greatest impact in the striatum.
Collapse
Affiliation(s)
- Michelle E Ehrlich
- Department of Pediatrics, Mount Sinai School of Medicine, Annenberg 14-44, 1 Gustave L. Levy Place, New York, NY 10019, USA.
| |
Collapse
|
98
|
Nucifora LG, Burke KA, Feng X, Arbez N, Zhu S, Miller J, Yang G, Ratovitski T, Delannoy M, Muchowski PJ, Finkbeiner S, Legleiter J, Ross CA, Poirier MA. Identification of novel potentially toxic oligomers formed in vitro from mammalian-derived expanded huntingtin exon-1 protein. J Biol Chem 2012; 287:16017-28. [PMID: 22433867 DOI: 10.1074/jbc.m111.252577] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Huntington disease is a genetic neurodegenerative disorder that arises from an expanded polyglutamine region in the N terminus of the HD gene product, huntingtin. Protein inclusions comprised of N-terminal fragments of mutant huntingtin are a characteristic feature of disease, though are likely to play a protective role rather than a causative one in neurodegeneration. Soluble oligomeric assemblies of huntingtin formed early in the aggregation process are candidate toxic species in HD. In the present study, we established an in vitro system to generate recombinant huntingtin in mammalian cells. Using both denaturing and native gel analysis, we have identified novel oligomeric forms of mammalian-derived expanded huntingtin exon-1 N-terminal fragment. These species are transient and were not previously detected using bacterially expressed exon-1 protein. Importantly, these species are recognized by 3B5H10, an antibody that recognizes a two-stranded hairpin conformation of expanded polyglutamine believed to be associated with a toxic form of huntingtin. Interestingly, comparable oligomeric species were not observed for expanded huntingtin shortstop, a 117-amino acid fragment of huntingtin shown previously in mammalian cell lines and transgenic mice, and here in primary cortical neurons, to be non-toxic. Further, we demonstrate that expanded huntingtin shortstop has a reduced ability to form amyloid-like fibrils characteristic of the aggregation pathway for toxic expanded polyglutamine proteins. Taken together, these data provide a possible candidate toxic species in HD. In addition, these studies demonstrate the fundamental differences in early aggregation events between mutant huntingtin exon-1 and shortstop proteins that may underlie the differences in toxicity.
Collapse
Affiliation(s)
- Leslie G Nucifora
- Division of Neurobiology, Department of Psychiatry, Children's Medical Surgical Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Sapp E, Valencia A, Li X, Aronin N, Kegel KB, Vonsattel JP, Young AB, Wexler N, DiFiglia M. Native mutant huntingtin in human brain: evidence for prevalence of full-length monomer. J Biol Chem 2012; 287:13487-99. [PMID: 22375012 DOI: 10.1074/jbc.m111.286609] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Huntington disease (HD) is caused by polyglutamine expansion in the N terminus of huntingtin (htt). Analysis of human postmortem brain lysates by SDS-PAGE and Western blot reveals htt as full-length and fragmented. Here we used Blue Native PAGE (BNP) and Western blots to study native htt in human postmortem brain. Antisera against htt detected a single band broadly migrating at 575-850 kDa in control brain and at 650-885 kDa in heterozygous and Venezuelan homozygous HD brains. Anti-polyglutamine antisera detected full-length mutant htt in HD brain. There was little htt cleavage even if lysates were pretreated with trypsin, indicating a property of native htt to resist protease cleavage. A soluble mutant htt fragment of about 180 kDa was detected with anti-htt antibody Ab1 (htt-(1-17)) and increased when lysates were treated with denaturants (SDS, 8 M urea, DTT, or trypsin) before BNP. Wild-type htt was more resistant to denaturants. Based on migration of in vitro translated htt fragments, the 180-kDa segment terminated ≈htt 670-880 amino acids. If second dimension SDS-PAGE followed BNP, the 180-kDa mutant htt was absent, and 43-50 kDa htt fragments appeared. Brain lysates from two HD mouse models expressed native full-length htt; a mutant fragment formed if lysates were pretreated with 8 M urea + DTT. Native full-length mutant htt in embryonic HD(140Q/140Q) mouse primary neurons was intact during cell death and when cell lysates were exposed to denaturants before BNP. Thus, native mutant htt occurs in brain and primary neurons as a soluble full-length monomer.
Collapse
Affiliation(s)
- Ellen Sapp
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Butler DC, Messer A. Bifunctional anti-huntingtin proteasome-directed intrabodies mediate efficient degradation of mutant huntingtin exon 1 protein fragments. PLoS One 2011; 6:e29199. [PMID: 22216210 PMCID: PMC3245261 DOI: 10.1371/journal.pone.0029199] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Accepted: 11/22/2011] [Indexed: 12/31/2022] Open
Abstract
Huntington's disease (HD) is a fatal autosomal dominant neurodegenerative disorder caused by a trinucleotide (CAG)(n) repeat expansion in the coding sequence of the huntingtin gene, and an expanded polyglutamine (>37Q) tract in the protein. This results in misfolding and accumulation of huntingtin protein (htt), formation of neuronal intranuclear and cytoplasmic inclusions, and neuronal dysfunction/degeneration. Single-chain Fv antibodies (scFvs), expressed as intrabodies that bind htt and prevent aggregation, show promise as immunotherapeutics for HD. Intrastriatal delivery of anti-N-terminal htt scFv-C4 using an adeno-associated virus vector (AAV2/1) significantly reduces the size and number of aggregates in HDR6/1 transgenic mice; however, this protective effect diminishes with age and time after injection. We therefore explored enhancing intrabody efficacy via fusions to heterologous functional domains. Proteins containing a PEST motif are often targeted for proteasomal degradation and generally have a short half life. In ST14A cells, fusion of the C-terminal PEST region of mouse ornithine decarboxylase (mODC) to scFv-C4 reduces htt exon 1 protein fragments with 72 glutamine repeats (httex1-72Q) by ~80-90% when compared to scFv-C4 alone. Proteasomal targeting was verified by either scrambling the mODC-PEST motif, or via proteasomal inhibition with epoxomicin. For these constructs, the proteasomal degradation of the scFv intrabody proteins themselves was reduced<25% by the addition of the mODC-PEST motif, with or without antigens. The remaining intrabody levels were amply sufficient to target N-terminal httex1-72Q protein fragment turnover. Critically, scFv-C4-PEST prevents aggregation and toxicity of httex1-72Q fragments at significantly lower doses than scFv-C4. Fusion of the mODC-PEST motif to intrabodies is a valuable general approach to specifically target toxic antigens to the proteasome for degradation.
Collapse
Affiliation(s)
- David C. Butler
- Wadsworth Center, New York State Department of Health, Albany, New York, United States of America
- Department of Biomedical Sciences, University at Albany, Albany, New York, United States of America
| | - Anne Messer
- Wadsworth Center, New York State Department of Health, Albany, New York, United States of America
- Department of Biomedical Sciences, University at Albany, Albany, New York, United States of America
- * E-mail:
| |
Collapse
|