1
|
Solem MA, Pelzel RG, Rozema NB, Brown TG, Reid E, Mansky RH, Gomez-Pastor R. Absence of hippocampal pathology persists in the Q175DN mouse model of Huntington's disease despite elevated HTT aggregation. J Huntingtons Dis 2025; 14:59-84. [PMID: 39973391 PMCID: PMC11974504 DOI: 10.1177/18796397251316762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
BackgroundHuntington's disease (HD) is a neurodegenerative disorder causing motor, cognitive, and psychiatric impairments, with the striatum being the most affected brain region. However, the role of other regions, such as the hippocampus, in HD remains less understood.ObjectiveHere, we study the comparative impact of enhanced mHTT aggregation and neuropathology in the striatum and hippocampus of two HD mouse models.MethodsWe utilized the zQ175 as a control HD mouse model and the Q175DN mice lacking the PGK-Neomycin cassette generated in house. We performed a comparative characterization of the neuropathology between zQ175 and Q175DN mice in the striatum and the hippocampus by assessing HTT aggregation, neuronal and glial pathology, chaperone expression, and synaptic density.ResultsWe showed that Q175DN mice presented enhanced mHTT aggregation in both striatum and hippocampus compared to zQ175. Striatal neurons showed a greater susceptibility to enhanced accumulation of mHTT in Q175DN. On the contrary, no signs of hippocampal pathology were found in zQ175 and absence of hippocampal pathology persisted in Q175DN mice despite higher levels of mHTT. In addition, Q175DN hippocampus presented increased synaptic density, decreased Iba1+ microglia density and enhanced HSF1 levels in specific subregions of the hippocampus compared to zQ175.ConclusionsQ175DN mice are a valuable tool to understand the fundamental susceptibility differences to mHTT toxicity between striatal neurons and other neuronal subtypes. Furthermore, our findings also suggest that cognitive deficits observed in HD animals might arise from either striatum dysfunction or other regions involved in cognitive processes but not from hippocampal degeneration.
Collapse
Affiliation(s)
- Melissa A Solem
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Ross G Pelzel
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Nicholas B Rozema
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Taylor G Brown
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Emma Reid
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Rachel H Mansky
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Rocio Gomez-Pastor
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
2
|
Li X, Tong H, Xu S, Zhou G, Yang T, Yin S, Yang S, Li X, Li S. Neuroinflammatory Proteins in Huntington's Disease: Insights into Mechanisms, Diagnosis, and Therapeutic Implications. Int J Mol Sci 2024; 25:11787. [PMID: 39519337 PMCID: PMC11546928 DOI: 10.3390/ijms252111787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Huntington's disease (HD) is a hereditary neurodegenerative disorder caused by a CAG tract expansion in the huntingtin gene (HTT). HD is characterized by involuntary movements, cognitive decline, and behavioral changes. Pathologically, patients with HD show selective striatal neuronal vulnerability at the early disease stage, although the mutant protein is ubiquitously expressed. Activation of the immune system and glial cell-mediated neuroinflammatory responses are early pathological features and have been found in all neurodegenerative diseases (NDDs), including HD. However, the role of inflammation in HD, as well as its therapeutic significance, has been less extensively studied compared to other NDDs. This review highlights the significantly elevated levels of inflammatory proteins and cellular markers observed in various HD animal models and HD patient tissues, emphasizing the critical roles of microglia, astrocytes, and oligodendrocytes in mediating neuroinflammation in HD. Moreover, it expands on recent discoveries related to the peripheral immune system's involvement in HD. Although current immunomodulatory treatments and inflammatory biomarkers for adjunctive diagnosis in HD are limited, targeting inflammation in combination with other therapies, along with comprehensive personalized treatment approaches, shows promising therapeutic potential.
Collapse
Affiliation(s)
- Xinhui Li
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| | - Huichun Tong
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Shuying Xu
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| | - Gongke Zhou
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| | - Tianqi Yang
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| | - Shurui Yin
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| | - Sitong Yang
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| | - Xiaojiang Li
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| | - Shihua Li
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| |
Collapse
|
3
|
Solem MA, Pelzel R, Rozema NB, Brown TG, Reid E, Mansky RH, Gomez-Pastor R. Enhanced Hippocampal Spare Capacity in Q175DN Mice Despite Elevated mHTT Aggregation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.14.618355. [PMID: 39464002 PMCID: PMC11507687 DOI: 10.1101/2024.10.14.618355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Background Huntington's disease (HD) is a neurodegenerative disease resulting in devastating motor, cognitive, and psychiatric deficits. The striatum is a brain region that controls movement and some forms of cognition and is most significantly impacted in HD. However, despite well-documented deficits in learning and memory in HD, knowledge of the potential implication of other brain regions such as the hippocampus remains limited. Objective Here, we study the comparative impact of enhanced mHTT aggregation and neuropathology in the striatum and hippocampus of two HD mouse models. Methods We utilized the zQ175 as a control HD mouse model and the Q175DN mice lacking the PGK-Neomycin cassette generated in house. We performed a comparative characterization of the neuropathology between zQ175 and Q175DN mice in the striatum and the hippocampus by assessing HTT aggregation, neuronal and glial pathology, chaperone expression, and synaptic density. Results We showed that Q175DN mice presented enhanced mHTT aggregation in both striatum and hippocampus compared to zQ175. Striatal neurons showed a greater susceptibility to enhanced accumulation of mHTT than hippocampal neurons in Q175DN despite high levels of mHTT in both regions. Contrary to the pathology seen in the striatum, Q175DN hippocampus presented enhanced spare capacity showing increased synaptic density, decreased Iba1+ microglia density and enhanced HSF1 levels in specific subregions of the hippocampus compared to zQ175. Conclusions Q175DN mice are a valuable tool to understand the fundamental susceptibility differences to mHTT toxicity between striatal neurons and other neuronal subtypes. Furthermore, our findings also suggest that cognitive deficits observed in HD animals might arise from either striatum dysfunction or other regions involved in cognitive processes but not from hippocampal degeneration.
Collapse
Affiliation(s)
- Melissa A Solem
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Ross Pelzel
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Nicholas B. Rozema
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Taylor G. Brown
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Emma Reid
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Rachel H. Mansky
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - R Gomez-Pastor
- Department of Neuroscience, School of Medicine, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
4
|
Jia Q, Li S, Li XJ, Yin P. Neuroinflammation in Huntington's disease: From animal models to clinical therapeutics. Front Immunol 2022; 13:1088124. [PMID: 36618375 PMCID: PMC9815700 DOI: 10.3389/fimmu.2022.1088124] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 11/29/2022] [Indexed: 12/24/2022] Open
Abstract
Huntington's disease (HD) is a progressive neurodegenerative disease characterized by preferential loss of neurons in the striatum in patients, which leads to motor and cognitive impairments and death that often occurs 10-15 years after the onset of symptoms. The expansion of a glutamine repeat (>36 glutamines) in the N-terminal region of huntingtin (HTT) has been defined as the cause of HD, but the mechanism underlying neuronal death remains unclear. Multiple mechanisms, including inflammation, may jointly contribute to HD pathogenesis. Altered inflammation response is evident even before the onset of classical symptoms of HD. In this review, we summarize the current evidence on immune and inflammatory changes, from HD animal models to clinical phenomenon of patients with HD. The understanding of the impact of inflammation on HD would help develop novel strategies to treat HD.
Collapse
Affiliation(s)
| | | | | | - Peng Yin
- *Correspondence: Xiao-Jiang Li, ; Peng Yin,
| |
Collapse
|
5
|
Barry LA, Kay GW, Mitchell NL, Murray SJ, Jay NP, Palmer DN. Aggregation chimeras provide evidence of in vivo intercellular correction in ovine CLN6 neuronal ceroid lipofuscinosis (Batten disease). PLoS One 2022; 17:e0261544. [PMID: 35404973 PMCID: PMC9000108 DOI: 10.1371/journal.pone.0261544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 03/29/2022] [Indexed: 11/29/2022] Open
Abstract
The neuronal ceroid lipofuscinoses (NCLs; Batten disease) are fatal, mainly childhood, inherited neurodegenerative lysosomal storage diseases. Sheep affected with a CLN6 form display progressive regionally defined glial activation and subsequent neurodegeneration, indicating that neuroinflammation may be causative of pathogenesis. In this study, aggregation chimeras were generated from homozygous unaffected normal and CLN6 affected sheep embryos, resulting in seven chimeric animals with varied proportions of normal to affected cells. These sheep were classified as affected-like, recovering-like or normal-like, based on their cell-genotype ratios and their clinical and neuropathological profiles. Neuropathological examination of the affected-like animals revealed intense glial activation, prominent storage body accumulation and severe neurodegeneration within all cortical brain regions, along with vision loss and decreasing intracranial volumes and cortical thicknesses consistent with ovine CLN6 disease. In contrast, intercellular communication affecting pathology was evident at both the gross and histological level in the normal-like and recovering-like chimeras, resulting in a lack of glial activation and rare storage body accumulation in only a few cells. Initial intracranial volumes of the recovering-like chimeras were below normal but progressively recovered to about normal by two years of age. All had normal cortical thicknesses, and none went blind. Extended neurogenesis was evident in the brains of all the chimeras. This study indicates that although CLN6 is a membrane bound protein, the consequent defect is not cell intrinsic. The lack of glial activation and inflammatory responses in the normal-like and recovering-like chimeras indicate that newly generated cells are borne into a microenvironment conducive to maturation and survival.
Collapse
Affiliation(s)
- Lucy Anne Barry
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, Canterbury, New Zealand
| | - Graham William Kay
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, Canterbury, New Zealand
| | - Nadia Lesley Mitchell
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, Canterbury, New Zealand
- Department of Radiology, University of Otago, Christchurch, Canterbury, New Zealand
| | - Samantha Jane Murray
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, Canterbury, New Zealand
| | - Nigel P. Jay
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, Canterbury, New Zealand
| | - David Norris Palmer
- Faculty of Agriculture and Life Sciences, Lincoln University, Lincoln, Canterbury, New Zealand
- Department of Radiology, University of Otago, Christchurch, Canterbury, New Zealand
- * E-mail:
| |
Collapse
|
6
|
Oura S, Noda T, Morimura N, Hitoshi S, Nishimasu H, Nagai Y, Nureki O, Ikawa M. Precise CAG repeat contraction in a Huntington's Disease mouse model is enabled by gene editing with SpCas9-NG. Commun Biol 2021; 4:771. [PMID: 34163001 PMCID: PMC8222283 DOI: 10.1038/s42003-021-02304-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 06/03/2021] [Indexed: 12/22/2022] Open
Abstract
The clustered regularly interspaced palindromic repeats (CRISPR)/Cas9 system is a research hotspot in gene therapy. However, the widely used Streptococcus pyogenes Cas9 (WT-SpCas9) requires an NGG protospacer adjacent motif (PAM) for target recognition, thereby restricting targetable disease mutations. To address this issue, we recently reported an engineered SpCas9 nuclease variant (SpCas9-NG) recognizing NGN PAMs. Here, as a feasibility study, we report SpCas9-NG-mediated repair of the abnormally expanded CAG repeat tract in Huntington's disease (HD). By targeting the boundary of CAG repeats with SpCas9-NG, we precisely contracted the repeat tracts in HD-mouse-derived embryonic stem (ES) cells. Further, we confirmed the recovery of phenotypic abnormalities in differentiated neurons and animals produced from repaired ES cells. Our study shows that SpCas9-NG can be a powerful tool for repairing abnormally expanded CAG repeats as well as other disease mutations that are difficult to access with WT-SpCas9.
Collapse
Affiliation(s)
- Seiya Oura
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Taichi Noda
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Division of Reproductive Biology, Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan
| | - Naoko Morimura
- Department of Integrative Physiology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Seiji Hitoshi
- Department of Integrative Physiology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Hiroshi Nishimasu
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
- Department of Structural Biology, Research center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Yoshitaka Nagai
- Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
- Department of Neurotherapeutics, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Neurology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Osamu Nureki
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Masahito Ikawa
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.
- Laboratory of Reproductive Systems Biology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
7
|
Deng Y, Wang H, Joni M, Sekhri R, Reiner A. Progression of basal ganglia pathology in heterozygous Q175 knock-in Huntington's disease mice. J Comp Neurol 2021; 529:1327-1371. [PMID: 32869871 PMCID: PMC8049038 DOI: 10.1002/cne.25023] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 08/07/2020] [Accepted: 08/26/2020] [Indexed: 12/29/2022]
Abstract
We used behavioral testing and morphological methods to detail the progression of basal ganglia neuron type-specific pathology and the deficits stemming from them in male heterozygous Q175 mice, compared to age-matched WT males. A rotarod deficit was not present in Q175 mice until 18 months, but increased open field turn rate (reflecting hyperkinesia) and open field anxiety were evident at 6 months. No loss of striatal neurons was seen out to 18 months, but ENK+ and DARPP32+ striatal perikarya were fewer by 6 months, due to diminished expression, with further decline by 18 months. No reduction in SP+ striatal perikarya or striatal interneurons was seen in Q175 mice at 18 months, but cholinergic interneurons showed dendrite attenuation by 6 months. Despite reduced ENK expression in indirect pathway striatal perikarya, ENK-immunostained terminals in globus pallidus externus (GPe) were more abundant at 6 months and remained so out to 18 months. Similarly, SP-immunostained terminals from striatal direct pathway neurons were more abundant in globus pallidus internus and substantia nigra at 6 months and remained so at 18 months. FoxP2+ arkypallidal GPe neurons and subthalamic nucleus neurons were lost by 18 months but not prototypical PARV+ GPe neurons or dopaminergic nigral neurons. Our results show that striatal projection neuron abnormalities and behavioral abnormalities reflecting them develop between 2 and 6 months of age in Q175 male heterozygotes, indicating early effects of the HD mutation. The striatal pathologies resemble those in human HD, but are less severe at 18 months than even in premanifest HD.
Collapse
Affiliation(s)
- Yunping Deng
- Department of Anatomy and NeurobiologyThe University of Tennessee Health Science CenterMemphisTennesseeUSA
| | - Hongbing Wang
- Department of Anatomy and NeurobiologyThe University of Tennessee Health Science CenterMemphisTennesseeUSA
| | - Marion Joni
- Department of Anatomy and NeurobiologyThe University of Tennessee Health Science CenterMemphisTennesseeUSA
| | - Radhika Sekhri
- Department of PathologyThe University of Tennessee Health Science CenterMemphisTennesseeUSA
| | - Anton Reiner
- Department of Anatomy and NeurobiologyThe University of Tennessee Health Science CenterMemphisTennesseeUSA
- Department of OphthalmologyThe University of Tennessee Health Science CenterMemphisTennesseeUSA
| |
Collapse
|
8
|
Chivet M, Marchioretti C, Pirazzini M, Piol D, Scaramuzzino C, Polanco MJ, Romanello V, Zuccaro E, Parodi S, D’Antonio M, Rinaldi C, Sambataro F, Pegoraro E, Soraru G, Pandey UB, Sandri M, Basso M, Pennuto M. Polyglutamine-Expanded Androgen Receptor Alteration of Skeletal Muscle Homeostasis and Myonuclear Aggregation Are Affected by Sex, Age and Muscle Metabolism. Cells 2020; 9:cells9020325. [PMID: 32019272 PMCID: PMC7072234 DOI: 10.3390/cells9020325] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/23/2020] [Accepted: 01/24/2020] [Indexed: 12/18/2022] Open
Abstract
Polyglutamine (polyQ) expansions in the androgen receptor (AR) gene cause spinal and bulbar muscular atrophy (SBMA), a neuromuscular disease characterized by lower motor neuron (MN) loss and skeletal muscle atrophy, with an unknown mechanism. We generated new mouse models of SBMA for constitutive and inducible expression of mutant AR and performed biochemical, histological and functional analyses of phenotype. We show that polyQ-expanded AR causes motor dysfunction, premature death, IIb-to-IIa/IIx fiber-type change, glycolytic-to-oxidative fiber-type switching, upregulation of atrogenes and autophagy genes and mitochondrial dysfunction in skeletal muscle, together with signs of muscle denervation at late stage of disease. PolyQ expansions in the AR resulted in nuclear enrichment. Within the nucleus, mutant AR formed 2% sodium dodecyl sulfate (SDS)-resistant aggregates and inclusion bodies in myofibers, but not spinal cord and brainstem, in a process exacerbated by age and sex. Finally, we found that two-week induction of expression of polyQ-expanded AR in adult mice was sufficient to cause premature death, body weight loss and muscle atrophy, but not aggregation, metabolic alterations, motor coordination and fiber-type switch, indicating that expression of the disease protein in the adulthood is sufficient to recapitulate several, but not all SBMA manifestations in mice. These results imply that chronic expression of polyQ-expanded AR, i.e. during development and prepuberty, is key to induce the full SBMA muscle pathology observed in patients. Our data support a model whereby chronic expression of polyQ-expanded AR triggers muscle atrophy through toxic (neomorphic) gain of function mechanisms distinct from normal (hypermorphic) gain of function mechanisms.
Collapse
Affiliation(s)
- Mathilde Chivet
- Dulbecco Telethon Institute, Centre for Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy; (M.C.); (D.P.); (M.J.P.)
| | - Caterina Marchioretti
- Department of Biomedical Sciences (DBS), University of Padova, 35131 Padova, Italy; (C.M.); (M.P.); (V.R.); (E.Z.); (M.S.)
- Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Marco Pirazzini
- Department of Biomedical Sciences (DBS), University of Padova, 35131 Padova, Italy; (C.M.); (M.P.); (V.R.); (E.Z.); (M.S.)
- Myology Center (Cir-Myo), University of Padova, 35129 Padova, Italy; (E.P.); (G.S.)
| | - Diana Piol
- Dulbecco Telethon Institute, Centre for Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy; (M.C.); (D.P.); (M.J.P.)
- Department of Biomedical Sciences (DBS), University of Padova, 35131 Padova, Italy; (C.M.); (M.P.); (V.R.); (E.Z.); (M.S.)
| | - Chiara Scaramuzzino
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia (IIT), 16163 Genova, Italy; (C.S.); (S.P.)
| | - Maria Josè Polanco
- Dulbecco Telethon Institute, Centre for Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy; (M.C.); (D.P.); (M.J.P.)
| | - Vanina Romanello
- Department of Biomedical Sciences (DBS), University of Padova, 35131 Padova, Italy; (C.M.); (M.P.); (V.R.); (E.Z.); (M.S.)
- Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
- Myology Center (Cir-Myo), University of Padova, 35129 Padova, Italy; (E.P.); (G.S.)
| | - Emanuela Zuccaro
- Department of Biomedical Sciences (DBS), University of Padova, 35131 Padova, Italy; (C.M.); (M.P.); (V.R.); (E.Z.); (M.S.)
- Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
| | - Sara Parodi
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia (IIT), 16163 Genova, Italy; (C.S.); (S.P.)
| | - Maurizio D’Antonio
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, 20132 Milan, Italy;
| | - Carlo Rinaldi
- Department of Paediatrics, University of Oxford, OX1 3QX Oxford, UK;
| | - Fabio Sambataro
- Department of Neuroscience (DNS), University of Padova, 35128 Padova, Italy;
- Padova Neuroscience Center (PNC), 35100 Padova, Italy
| | - Elena Pegoraro
- Myology Center (Cir-Myo), University of Padova, 35129 Padova, Italy; (E.P.); (G.S.)
- Department of Neuroscience (DNS), University of Padova, 35128 Padova, Italy;
- Padova Neuroscience Center (PNC), 35100 Padova, Italy
| | - Gianni Soraru
- Myology Center (Cir-Myo), University of Padova, 35129 Padova, Italy; (E.P.); (G.S.)
- Department of Neuroscience (DNS), University of Padova, 35128 Padova, Italy;
- Padova Neuroscience Center (PNC), 35100 Padova, Italy
| | - Udai Bhan Pandey
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA 15261, USA;
- Division of Child Neurology, Department of Pediatrics, Children’s Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA 15224, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Marco Sandri
- Department of Biomedical Sciences (DBS), University of Padova, 35131 Padova, Italy; (C.M.); (M.P.); (V.R.); (E.Z.); (M.S.)
- Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
- Myology Center (Cir-Myo), University of Padova, 35129 Padova, Italy; (E.P.); (G.S.)
| | - Manuela Basso
- Centre for Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy;
| | - Maria Pennuto
- Dulbecco Telethon Institute, Centre for Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy; (M.C.); (D.P.); (M.J.P.)
- Department of Biomedical Sciences (DBS), University of Padova, 35131 Padova, Italy; (C.M.); (M.P.); (V.R.); (E.Z.); (M.S.)
- Veneto Institute of Molecular Medicine (VIMM), 35129 Padova, Italy
- Myology Center (Cir-Myo), University of Padova, 35129 Padova, Italy; (E.P.); (G.S.)
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia (IIT), 16163 Genova, Italy; (C.S.); (S.P.)
- Padova Neuroscience Center (PNC), 35100 Padova, Italy
- Correspondence: ; Tel.: +39 049 8276069
| |
Collapse
|
9
|
Dragatsis I, Dietrich P, Ren H, Deng YP, Del Mar N, Wang HB, Johnson IM, Jones KR, Reiner A. Effect of early embryonic deletion of huntingtin from pyramidal neurons on the development and long-term survival of neurons in cerebral cortex and striatum. Neurobiol Dis 2017; 111:102-117. [PMID: 29274742 PMCID: PMC5821111 DOI: 10.1016/j.nbd.2017.12.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 11/07/2017] [Accepted: 12/19/2017] [Indexed: 12/12/2022] Open
Abstract
We evaluated the impact of early embryonic deletion of huntingtin (htt) from pyramidal neurons on cortical development, cortical neuron survival and motor behavior, using a cre-loxP strategy to inactivate the mouse htt gene (Hdh) in emx1-expressing cell lineages. Western blot confirmed substantial htt reduction in cerebral cortex of these Emx-httKO mice, with residual cortical htt in all likelihood restricted to cortical interneurons of the subpallial lineage and/or vascular endothelial cells. Despite the loss of htt early in development, cortical lamination was normal, as revealed by layer-specific markers. Cortical volume and neuron abundance were, however, significantly less than normal, and cortical neurons showed reduced brain-derived neurotrophic factor (BDNF) expression and reduced activation of BDNF signaling pathways. Nonetheless, cortical volume and neuron abundance did not show progressive age-related decline in Emx-httKO mice out to 24 months. Although striatal neurochemistry was normal, reductions in striatal volume and neuron abundance were seen in Emx-httKO mice, which were again not progressive. Weight maintenance was normal in Emx-httKO mice, but a slight rotarod deficit and persistent hyperactivity were observed throughout the lifespan. Our results show that embryonic deletion of htt from developing pallium does not substantially alter migration of cortical neurons to their correct laminar destinations, but does yield reduced cortical and striatal size and neuron numbers. The Emx-httKO mice were persistently hyperactive, possibly due to defects in corticostriatal development. Importantly, deletion of htt from cortical pyramidal neurons did not yield age-related progressive cortical or striatal pathology.
Collapse
Affiliation(s)
- I Dragatsis
- Department of Physiology, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - P Dietrich
- Department of Physiology, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - H Ren
- Department of Anatomy & Neurobiology, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Y P Deng
- Department of Anatomy & Neurobiology, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - N Del Mar
- Department of Anatomy & Neurobiology, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - H B Wang
- Department of Anatomy & Neurobiology, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - I M Johnson
- Department of Physiology, The University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - K R Jones
- Department of Molecular, Cellular, & Developmental Biology, 347 UCB, University of Colorado, Boulder, CO 80309, United States
| | - A Reiner
- Department of Anatomy & Neurobiology, The University of Tennessee Health Science Center, Memphis, TN 38163, United States; Department of Ophthalmology, The University of Tennessee Health Science Center, Memphis, TN 38163, United States.
| |
Collapse
|
10
|
Guley NH, Rogers JT, Del Mar NA, Deng Y, Islam RM, D'Surney L, Ferrell J, Deng B, Hines-Beard J, Bu W, Ren H, Elberger AJ, Marchetta JG, Rex TS, Honig MG, Reiner A. A Novel Closed-Head Model of Mild Traumatic Brain Injury Using Focal Primary Overpressure Blast to the Cranium in Mice. J Neurotrauma 2016; 33:403-22. [PMID: 26414413 PMCID: PMC4761824 DOI: 10.1089/neu.2015.3886] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Mild traumatic brain injury (TBI) from focal head impact is the most common form of TBI in humans. Animal models, however, typically use direct impact to the exposed dura or skull, or blast to the entire head. We present a detailed characterization of a novel overpressure blast system to create focal closed-head mild TBI in mice. A high-pressure air pulse limited to a 7.5 mm diameter area on the left side of the head overlying the forebrain is delivered to anesthetized mice. The mouse eyes and ears are shielded, and its head and body are cushioned to minimize movement. This approach creates mild TBI by a pressure wave that acts on the brain, with minimal accompanying head acceleration-deceleration. A single 20-psi blast yields no functional deficits or brain injury, while a single 25-40 psi blast yields only slight motor deficits and brain damage. By contrast, a single 50-60 psi blast produces significant visual, motor, and neuropsychiatric impairments and axonal damage and microglial activation in major fiber tracts, but no contusive brain injury. This model thus reproduces the widespread axonal injury and functional impairments characteristic of closed-head mild TBI, without the complications of systemic or ocular blast effects or head acceleration that typically occur in other blast or impact models of closed-skull mild TBI. Accordingly, our model provides a simple way to examine the biomechanics, pathophysiology, and functional deficits that result from TBI and can serve as a reliable platform for testing therapies that reduce brain pathology and deficits.
Collapse
Affiliation(s)
- Natalie H. Guley
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Joshua T. Rogers
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Nobel A. Del Mar
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Yunping Deng
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Rafiqul M. Islam
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, Tennessee
- Department of Anatomy and Histology, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Lauren D'Surney
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, Tennessee
- Department of Ophthalmology, The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Jessica Ferrell
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Bowei Deng
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Jessica Hines-Beard
- Department of Ophthalmology, The University of Tennessee Health Science Center, Memphis, Tennessee
- Department of Ophthalmology and Visual Sciences, Vanderbilt University, Nashville, Tennessee
| | - Wei Bu
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Huiling Ren
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Andrea J. Elberger
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, Tennessee
| | | | - Tonia S. Rex
- Department of Ophthalmology, The University of Tennessee Health Science Center, Memphis, Tennessee
- Department of Ophthalmology and Visual Sciences, Vanderbilt University, Nashville, Tennessee
| | - Marcia G. Honig
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, Tennessee
| | - Anton Reiner
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, Tennessee
- Department of Ophthalmology, The University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
11
|
Ultrasensitive measurement of huntingtin protein in cerebrospinal fluid demonstrates increase with Huntington disease stage and decrease following brain huntingtin suppression. Sci Rep 2015; 5:12166. [PMID: 26174131 PMCID: PMC4502413 DOI: 10.1038/srep12166] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 06/18/2015] [Indexed: 11/26/2022] Open
Abstract
Quantitation of huntingtin protein in the brain is needed, both as a marker of Huntington disease (HD) progression and for use in clinical gene silencing trials. Measurement of huntingtin in cerebrospinal fluid could be a biomarker of brain huntingtin, but traditional protein quantitation methods have failed to detect huntingtin in cerebrospinal fluid. Using micro-bead based immunoprecipitation and flow cytometry (IP-FCM), we have developed a highly sensitive mutant huntingtin detection assay. The sensitivity of huntingtin IP-FCM enables accurate detection of mutant huntingtin protein in the cerebrospinal fluid of HD patients and model mice, demonstrating that mutant huntingtin levels in cerebrospinal fluid reflect brain levels, increasing with disease stage and decreasing following brain huntingtin suppression. This technique has potential applications as a research tool and as a clinical biomarker.
Collapse
|
12
|
Waldvogel HJ, Kim EH, Tippett LJ, Vonsattel JPG, Faull RLM. The Neuropathology of Huntington's Disease. Curr Top Behav Neurosci 2015; 22:33-80. [PMID: 25300927 DOI: 10.1007/7854_2014_354] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
The basal ganglia are a highly interconnected set of subcortical nuclei and major atrophy in one or more regions may have major effects on other regions of the brain. Therefore, the striatum which is preferentially degenerated and receives projections from the entire cortex also affects the regions to which it targets, especially the globus pallidus and substantia nigra pars reticulata. Additionally, the cerebral cortex is itself severely affected as are many other regions of the brain, especially in more advanced cases. The cell loss in the basal ganglia and the cerebral cortex is extensive. The most important new findings in Huntington's disease pathology is the highly variable nature of the degeneration in the brain. Most interestingly, this variable pattern of pathology appears to reflect the highly variable symptomatology of cases with Huntington's disease even among cases possessing the same number of CAG repeats.
Collapse
Affiliation(s)
- Henry J Waldvogel
- Centre for Brain Research, Department of Anatomy with Radiology, University of Auckland, Auckland, New Zealand,
| | | | | | | | | |
Collapse
|
13
|
Motor, visual and emotional deficits in mice after closed-head mild traumatic brain injury are alleviated by the novel CB2 inverse agonist SMM-189. Int J Mol Sci 2014; 16:758-87. [PMID: 25561230 PMCID: PMC4307274 DOI: 10.3390/ijms16010758] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 12/22/2014] [Indexed: 11/17/2022] Open
Abstract
We have developed a focal blast model of closed-head mild traumatic brain injury (TBI) in mice. As true for individuals that have experienced mild TBI, mice subjected to 50-60 psi blast show motor, visual and emotional deficits, diffuse axonal injury and microglial activation, but no overt neuron loss. Because microglial activation can worsen brain damage after a concussive event and because microglia can be modulated by their cannabinoid type 2 receptors (CB2), we evaluated the effectiveness of the novel CB2 receptor inverse agonist SMM-189 in altering microglial activation and mitigating deficits after mild TBI. In vitro analysis indicated that SMM-189 converted human microglia from the pro-inflammatory M1 phenotype to the pro-healing M2 phenotype. Studies in mice showed that daily administration of SMM-189 for two weeks beginning shortly after blast greatly reduced the motor, visual, and emotional deficits otherwise evident after 50-60 psi blasts, and prevented brain injury that may contribute to these deficits. Our results suggest that treatment with the CB2 inverse agonist SMM-189 after a mild TBI event can reduce its adverse consequences by beneficially modulating microglial activation. These findings recommend further evaluation of CB2 inverse agonists as a novel therapeutic approach for treating mild TBI.
Collapse
|
14
|
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
|
15
|
Heldt SA, Elberger AJ, Deng Y, Guley NH, Del Mar N, Rogers J, Choi GW, Ferrell J, Rex TS, Honig MG, Reiner A. A novel closed-head model of mild traumatic brain injury caused by primary overpressure blast to the cranium produces sustained emotional deficits in mice. Front Neurol 2014; 5:2. [PMID: 24478749 PMCID: PMC3898331 DOI: 10.3389/fneur.2014.00002] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 01/06/2014] [Indexed: 12/14/2022] Open
Abstract
Emotional disorders are a common outcome from mild traumatic brain injury (TBI) in humans, but their pathophysiological basis is poorly understood. We have developed a mouse model of closed-head blast injury using an air pressure wave delivered to a small area on one side of the cranium, to create mild TBI. We found that 20-psi blasts in 3-month-old C57BL/6 male mice yielded no obvious behavioral or histological evidence of brain injury, while 25-40 psi blasts produced transient anxiety in an open field arena but little histological evidence of brain damage. By contrast, 50-60 psi blasts resulted in anxiety-like behavior in an open field arena that became more evident with time after blast. In additional behavioral tests conducted 2-8 weeks after blast, 50-60 psi mice also demonstrated increased acoustic startle, perseverance of learned fear, and enhanced contextual fear, as well as depression-like behavior and diminished prepulse inhibition. We found no evident cerebral pathology, but did observe scattered axonal degeneration in brain sections from 50 to 60 psi mice 3-8 weeks after blast. Thus, the TBI caused by single 50-60 psi blasts in mice exhibits the minimal neuronal loss coupled to "diffuse" axonal injury characteristic of human mild TBI. A reduction in the abundance of a subpopulation of excitatory projection neurons in basolateral amygdala enriched in Thy1 was, however, observed. The reported link of this neuronal population to fear suppression suggests their damage by mild TBI may contribute to the heightened anxiety and fearfulness observed after blast in our mice. Our overpressure air blast model of concussion in mice will enable further studies of the mechanisms underlying the diverse emotional deficits seen after mild TBI.
Collapse
Affiliation(s)
- Scott A. Heldt
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Andrea J. Elberger
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Yunping Deng
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Natalie H. Guley
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Nobel Del Mar
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Joshua Rogers
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Gy Won Choi
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Jessica Ferrell
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Tonia S. Rex
- Department of Ophthalmology, The University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Ophthalmology and Visual Sciences, Vanderbilt University, Nashville, TN, USA
| | - Marcia G. Honig
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Anton Reiner
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Ophthalmology, The University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
16
|
Todd TW, Lim J. Aggregation formation in the polyglutamine diseases: protection at a cost? Mol Cells 2013; 36:185-94. [PMID: 23794019 PMCID: PMC3800151 DOI: 10.1007/s10059-013-0167-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 06/02/2013] [Indexed: 12/30/2022] Open
Abstract
Mutant protein aggregation is a hallmark of many neurodegenerative diseases, including the polyglutamine disorders. Although the correlation between aggregation formation and disease pathology originally suggested that the visible inclusions seen in patient tissue might directly contribute to pathology, additional studies failed to confirm this hypothesis. Current opinion in the field of polyglutamine disease research now favors a model in which large inclusions are cytoprotective and smaller oligomers or misfolded monomers underlie pathogenesis. Nonetheless, therapies aimed at reducing or preventing aggregation show promise. This review outlines the debate about the role of aggregation in the polyglutamine diseases as it has unfolded in the literature and concludes with a brief discussion on the manipulation of aggregation formation and clearance mechanisms as a means of therapeutic intervention.
Collapse
Affiliation(s)
- Tiffany W. Todd
- Department of Genetics, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT 06510, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Janghoo Lim
- Department of Genetics, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT 06510, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
17
|
Loss of corticostriatal and thalamostriatal synaptic terminals precedes striatal projection neuron pathology in heterozygous Q140 Huntington's disease mice. Neurobiol Dis 2013; 60:89-107. [PMID: 23969239 DOI: 10.1016/j.nbd.2013.08.009] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 07/22/2013] [Accepted: 08/07/2013] [Indexed: 01/18/2023] Open
Abstract
Motor slowing, forebrain white matter loss, and striatal shrinkage have been reported in premanifest Huntington's disease (HD) prior to overt striatal neuron loss. We carried out detailed LM and EM studies in a genetically precise HD mimic, heterozygous Q140 HD knock-in mice, to examine the possibility that loss of corticostriatal and thalamostriatal terminals prior to striatal neuron loss underlies these premanifest HD abnormalities. In our studies, we used VGLUT1 and VGLUT2 immunolabeling to detect corticostriatal and thalamostriatal (respectively) terminals in dorsolateral (motor) striatum over the first year of life, prior to striatal projection neuron pathology. VGLUT1+ axospinous corticostriatal terminals represented about 55% of all excitatory terminals in striatum, and VGLUT2+ axospinous thalamostriatal terminals represented about 35%, with VGLUT1+ and VGLUT2+ axodendritic terminals accounting for the remainder. In Q140 mice, a significant 40% shortfall in VGLUT2+ axodendritic thalamostriatal terminals and a 20% shortfall in axospinous thalamostriatal terminals were already observed at 1 month of age, but VGLUT1+ terminals were normal in abundance. The 20% deficiency in VGLUT2+ thalamostriatal axospinous terminals persisted at 4 and 12 months in Q140 mice, and an additional 30% loss of VGLUT1+ corticostriatal terminals was observed at 12 months. The early and persistent deficiency in thalamostriatal axospinous terminals in Q140 mice may reflect a development defect, and the impoverishment of this excitatory drive to striatum may help explain early motor defects in Q140 mice and in premanifest HD. The loss of corticostriatal terminals at 1 year in Q140 mice is consistent with prior evidence from other mouse models of corticostriatal disconnection early during progression, and can explain both the measurable bradykinesia and striatal white matter loss in late premanifest HD.
Collapse
|
18
|
Bayram-Weston Z, Jones L, Dunnett SB, Brooks SP. Light and electron microscopic characterization of the evolution of cellular pathology in HdhQ92 Huntington's disease knock-in mice. Brain Res Bull 2012; 88:171-81. [DOI: 10.1016/j.brainresbull.2011.03.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Revised: 03/15/2011] [Accepted: 03/18/2011] [Indexed: 01/01/2023]
|
19
|
Reiner A, Lafferty DC, Wang HB, Del Mar N, Deng YP. The group 2 metabotropic glutamate receptor agonist LY379268 rescues neuronal, neurochemical and motor abnormalities in R6/2 Huntington's disease mice. Neurobiol Dis 2012; 47:75-91. [PMID: 22472187 DOI: 10.1016/j.nbd.2012.03.025] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Revised: 03/14/2012] [Accepted: 03/17/2012] [Indexed: 12/11/2022] Open
Abstract
Excitotoxic injury to striatum by dysfunctional cortical input or aberrant glutamate uptake may contribute to Huntington's disease (HD) pathogenesis. Since corticostriatal terminals possess mGluR2/3 autoreceptors, whose activation dampens glutamate release, we tested the ability of the mGluR2/3 agonist LY379268 to improve the phenotype in R6/2 HD mice with 120-125 CAG repeats. Daily subcutaneous injection of a maximum tolerated dose (MTD) of LY379268 (20mg/kg) had no evident adverse effects in WT mice, and diverse benefits in R6/2 mice, both in a cohort of mice tested behaviorally until the end of R6/2 lifespan and in a cohort sacrificed at 10weeks of age for blinded histological analysis. MTD LY379268 yielded a significant 11% increase in R6/2 survival, an improvement on rotarod, normalization and/or improvement in locomotor parameters measured in open field (activity, speed, acceleration, endurance, and gait), a rescue of a 15-20% cortical and striatal neuron loss, normalization of SP striatal neuron neurochemistry, and to a lesser extent enkephalinergic striatal neuron neurochemistry. Deficits were greater in male than female R6/2 mice, and drug benefit tended to be greater in males. The improvements in SP striatal neurons, which facilitate movement, are consistent with the improved movement in LY379268-treated R6/2 mice. Our data indicate that mGluR2/3 agonists may be particularly useful for ameliorating the morphological, neurochemical and motor defects observed in HD.
Collapse
Affiliation(s)
- A Reiner
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | | | | | | | | |
Collapse
|
20
|
Bayram-Weston Z, Jones L, Dunnett SB, Brooks SP. Light and electron microscopic characterization of the evolution of cellular pathology in YAC128 Huntington's disease transgenic mice. Brain Res Bull 2011; 88:137-47. [PMID: 21620935 DOI: 10.1016/j.brainresbull.2011.05.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Revised: 04/21/2011] [Accepted: 05/08/2011] [Indexed: 11/19/2022]
Abstract
Huntington's disease (HD) is a progressive neurodegenerative disease caused by the insertion of an expanded polyglutamine sequence within the huntingtin protein. This mutation induces the formation of abnormal protein fragment aggregations and intra-nuclear neuronal inclusions in the brain. The present study aimed to produce a detailed longitudinal characterization of the neuronal pathology in the YAC128 transgenic mouse brain, to determine the similarity of this mouse model to other mouse models and the human condition in the spatial and temporal deposition pattern of the mutant protein fragments. Brain samples were taken from mice aged between 4 and 27 months of age, and assessed using S830 and GFAP immunohistochemistry, stereology and electron microscopy. Four month old mice did not exhibit intra-nuclear or extra-nuclear inclusions using the S830 antibody. Diffuse nuclear staining was present in the cortex, hippocampus and cerebellum from 6 months of age onwards. By 15 months of age, intra-nuclear inclusions were visible in most brain regions including nucleus accumbens, ventral striatum, lateral striatum, motor cortex, sensory cortex and cerebellum. The ventral striatum had a greater density of inclusions than the dorsal striatum. At 15 and 24 months of age, the mice showed increased reactive astrogliosis in the cortex, but no differences were found in the striatum. Necrotic cell death with vacuolation, uneven cell membrane and degenerated Golgi apparatus were detected ultrastructurally at 14 months of age, with some cells showing signs of apoptosis. By 26 months of age, most cells were degenerated in the transgenic animals, with lipofuscin granules being more abundant and larger in these mice than in their wildtype littermates. Our results demonstrate a progressive and widespread neuropathology in the YAC128 mice line that shares some similarity to the human condition. This article is part of a Special Issue entitled 'HD Transgenic Mouse'.
Collapse
|
21
|
Abstract
Mouse models for Huntington's Disease (HD) and HD patients demonstrate motor and behavioral dysfunctions, such as progressive loss of coordination and memory, and share similar transcriptional profiles and striatal neuron atrophy. Clear differences between the mouse and human diseases include almost complete striatal degeneration and rarity of intranuclear inclusions in HD, and the fact that mice expressing full-length mutant huntingtin do not demonstrate a shortened life span characteristic of HD. While no clinical interventions tested in mouse models to date have delayed disease progression, the mouse models provide an invaluable tool for both investigating the underlying pathogenic processes and developing new effective therapies. Inherent differences between humans and mice must be considered in the search for efficacious treatments for HD, but the striking similarities between human HD and mouse models support the view that these models are a biologically relevant system to support the identification and testing of potential clinical therapies.
Collapse
Affiliation(s)
- Zachary R Crook
- The David H. Koch Institute for Integrative Cancer Research at MIT, 500 Main Street, Building 76-553, Cambridge, MA 02139, USA
| | | |
Collapse
|
22
|
Bayram-Weston Z, Torres EM, Jones L, Dunnett SB, Brooks SP. Light and electron microscopic characterization of the evolution of cellular pathology in the Hdh(CAG)150 Huntington's disease knock-in mouse. Brain Res Bull 2011; 88:189-98. [PMID: 21511013 DOI: 10.1016/j.brainresbull.2011.03.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2010] [Revised: 03/15/2011] [Accepted: 03/18/2011] [Indexed: 12/30/2022]
Abstract
Huntington's disease is an autosomal dominant, progressive neurodegenerative disease in which a single mutation in the gene responsible for the protein huntingtin leads to a primarily striatal and cortical neuronal loss, resulting progressive motor, cognitive and psychiatric disability and ultimately death. The mutation induces an abnormal protein accumulation within cells, although the precise role of this accumulation in the disease process is unknown. Several animal models have been created to model the disease. In the present study, the pathology of the Hdh(CAG(150)) mouse model was analyzed longitudinally over 24 months. At 5 months of age, the mutant N-terminal antibody S830 found dense nuclear staining and nuclear inclusions in the olfactory tubercle and striatum of the Hdh(Q150/Q150) mice. Nuclear inclusions increased in number and size with age and disease progression, and spread in ventral to dorsal, and anterior to posterior pattern. Electron microscopy observations at 14 months of age revealed that the neurons showed a normal nucleus having a circular shape and regular membranes in a densely packed cytoplasm, whereas by 21 months the cytoplasm was vacuolated and contained swollen mitochondria with many degenerated cytoplasmic organelles. Immunogold labelling of the S830 antibody was found to be specifically localised to the inner area of the neuronal intra-nuclear inclusions. Our data demonstrate a marked and progressive cellular phenotype that begins at 5 months of age and progresses with time. The pathology the Hdh(Q150/Q150) line was focused on the striatum and cortex until the late stage of the disease, consistent with the human condition.
Collapse
|
23
|
Swanson DJ, Goldowitz D. Experimental Sey mouse chimeras reveal the developmental deficiencies of Pax6-null granule cells in the postnatal cerebellum. Dev Biol 2011; 351:1-12. [DOI: 10.1016/j.ydbio.2010.11.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Revised: 11/06/2010] [Accepted: 11/10/2010] [Indexed: 10/18/2022]
|
24
|
Silva-Fernandes A, Costa MDC, Duarte-Silva S, Oliveira P, Botelho CM, Martins L, Mariz JA, Ferreira T, Ribeiro F, Correia-Neves M, Costa C, Maciel P. Motor uncoordination and neuropathology in a transgenic mouse model of Machado–Joseph disease lacking intranuclear inclusions and ataxin-3 cleavage products. Neurobiol Dis 2010; 40:163-76. [DOI: 10.1016/j.nbd.2010.05.021] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2010] [Revised: 04/30/2010] [Accepted: 05/17/2010] [Indexed: 10/19/2022] Open
|
25
|
Giralt A, Friedman HC, Caneda-Ferrón B, Urbán N, Moreno E, Rubio N, Blanco J, Peterson A, Canals JM, Alberch J. BDNF regulation under GFAP promoter provides engineered astrocytes as a new approach for long-term protection in Huntington's disease. Gene Ther 2010; 17:1294-308. [PMID: 20463759 DOI: 10.1038/gt.2010.71] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is the main candidate for neuroprotective therapeutic strategies for Huntington's disease. However, the administration system and the control over the dosage are still important problems to be solved. Here we generated transgenic mice overexpressing BDNF under the promoter of the glial fibrillary acidic protein (GFAP) (pGFAP-BDNF mice). These mice are viable and have a normal phenotype. However, intrastriatal administration of quinolinate increased the number of reactive astrocytes and enhanced the release of BDNF in pGFAP-BDNF mice compared with wild-type mice. Coincidentally, pGFAP-BDNF mice are more resistant to quinolinate than wild-type mice, suggesting a protective effect of astrocyte-derived BDNF. To verify this, we next cultured astrocytes from pGFAP-BDNF and wild-type mice for grafting. Wild-type and pGFAP-BDNF-derived astrocytes behave similarly in nonlesioned mice. However, pGFAP-BDNF-derived astrocytes showed higher levels of BDNF and larger neuroprotective effects than the wild-type ones when quinolinate was injected 30 days after grafting. Interestingly, mice grafted with pGFAP-BDNF astrocytes showed important and sustained behavioral improvements over time after quinolinate administration as compared with mice grafted with wild-type astrocytes. These findings show that astrocytes engineered to release BDNF can constitute a therapeutic approach for Huntington's disease.
Collapse
Affiliation(s)
- A Giralt
- Facultat de Medicina, Departament de Biologia Cel·lular, Immunologia i Neurociències, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Universitat de Barcelona, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Genetic mouse models of Huntington's disease: focus on electrophysiological mechanisms. ASN Neuro 2010; 2:e00033. [PMID: 20396376 PMCID: PMC2850512 DOI: 10.1042/an20090058] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Revised: 03/11/2010] [Accepted: 03/16/2010] [Indexed: 11/21/2022] Open
Abstract
The discovery of the HD (Huntington’s disease) gene in 1993 led to the creation of genetic mouse models of the disease and opened the doors for mechanistic studies. In particular, the early changes and progression of the disease could be followed and examined systematically. The present review focuses on the contribution of these genetic mouse models to the understanding of functional changes in neurons as the HD phenotype progresses, and concentrates on two brain areas: the striatum, the site of most conspicuous pathology in HD, and the cortex, a site that is becoming increasingly important in understanding the widespread behavioural abnormalities. Mounting evidence points to synaptic abnormalities in communication between the cortex and striatum and cell–cell interactions as major determinants of HD symptoms, even in the absence of severe neuronal degeneration and death.
Collapse
Key Words
- AMPA, α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate
- BAC, bacterial artificial chromosome
- BDNF, brain-derived neurotrophic factor
- DA, dopamine
- EPSC, excitatory postsynaptic current
- GABA, γ-aminobutyric acid
- HD, Huntington’s disease
- Huntington’s disease
- IPSC, inhibitory postsynaptic current
- IR-DIC, infrared differential interference contrast
- MSSN, medium-sized spiny projection neuron
- NII, neuronal intranuclear inclusion
- NMDA, N-methyl-d-aspartate
- WT, wild-type
- YAC, yeast artificial chromosome
- enk, enkephalin
- excitatory amino acid
- htt, huntingtin
- mouse model
- neurodegeneration
- striatum
- synaptic activity
Collapse
|
27
|
Schwarcz R, Guidetti P, Sathyasaikumar KV, Muchowski PJ. Of mice, rats and men: Revisiting the quinolinic acid hypothesis of Huntington's disease. Prog Neurobiol 2010; 90:230-45. [PMID: 19394403 PMCID: PMC2829333 DOI: 10.1016/j.pneurobio.2009.04.005] [Citation(s) in RCA: 130] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2009] [Accepted: 04/17/2009] [Indexed: 12/31/2022]
Abstract
The neurodegenerative disease Huntington's disease (HD) is caused by an expanded polyglutamine (polyQ) tract in the protein huntingtin (htt). Although the gene encoding htt was identified and cloned more than 15 years ago, and in spite of impressive efforts to unravel the mechanism(s) by which mutant htt induces nerve cell death, these studies have so far not led to a good understanding of pathophysiology or an effective therapy. Set against a historical background, we review data supporting the idea that metabolites of the kynurenine pathway (KP) of tryptophan degradation provide a critical link between mutant htt and the pathophysiology of HD. New studies in HD brain and genetic model organisms suggest that the disease may in fact be causally related to early abnormalities in KP metabolism, favoring the formation of two neurotoxic metabolites, 3-hydroxykynurenine and quinolinic acid, over the related neuroprotective agent kynurenic acid. These findings not only link the excitotoxic hypothesis of HD pathology to an impairment of the KP but also define new drug targets and therefore have direct therapeutic implications. Thus, pharmacological normalization of the imbalance in brain KP metabolism may provide clinical benefits, which could be especially effective in early stages of the disease.
Collapse
Affiliation(s)
- Robert Schwarcz
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD, USA.
| | | | | | | |
Collapse
|
28
|
Kvam E, Nannenga BL, Wang MS, Jia Z, Sierks MR, Messer A. Conformational targeting of fibrillar polyglutamine proteins in live cells escalates aggregation and cytotoxicity. PLoS One 2009; 4:e5727. [PMID: 19492089 PMCID: PMC2683928 DOI: 10.1371/journal.pone.0005727] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2008] [Accepted: 04/29/2009] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Misfolding- and aggregation-prone proteins underlying Parkinson's, Huntington's and Machado-Joseph diseases, namely alpha-synuclein, huntingtin, and ataxin-3 respectively, adopt numerous intracellular conformations during pathogenesis, including globular intermediates and insoluble amyloid-like fibrils. Such conformational diversity has complicated research into amyloid-associated intracellular dysfunction and neurodegeneration. To this end, recombinant single-chain Fv antibodies (scFvs) are compelling molecular tools that can be selected against specific protein conformations, and expressed inside cells as intrabodies, for investigative and therapeutic purposes. METHODOLOGY/PRINCIPAL FINDINGS Using atomic force microscopy (AFM) and live-cell fluorescence microscopy, we report that a human scFv selected against the fibrillar form of alpha-synuclein targets isomorphic conformations of misfolded polyglutamine proteins. When expressed in the cytoplasm of striatal cells, this conformation-specific intrabody co-localizes with intracellular aggregates of misfolded ataxin-3 and a pathological fragment of huntingtin, and enhances the aggregation propensity of both disease-linked polyglutamine proteins. Using this intrabody as a tool for modulating the kinetics of amyloidogenesis, we show that escalating aggregate formation of a pathologic huntingtin fragment is not cytoprotective in striatal cells, but rather heightens oxidative stress and cell death as detected by flow cytometry. Instead, cellular protection is achieved by suppressing aggregation using a previously described intrabody that binds to the amyloidogenic N-terminus of huntingtin. Analogous cytotoxic results are observed following conformational targeting of normal or polyglutamine-expanded human ataxin-3, which partially aggregate through non-polyglutamine domains. CONCLUSIONS/SIGNIFICANCE These findings validate that the rate of aggregation modulates polyglutamine-mediated intracellular dysfunction, and caution that molecules designed to specifically hasten aggregation may be detrimental as therapies for polyglutamine disorders. Moreover, our findings introduce a novel antibody-based tool that, as a consequence of its general specificity for fibrillar conformations and its ability to function intracellularly, offers broad research potential for a variety of human amyloid diseases.
Collapse
Affiliation(s)
- Erik Kvam
- 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
| | - Brent L. Nannenga
- Department of Chemical Engineering, Arizona State University, Tempe, Arizona, United States of America
| | - Min S. Wang
- Department of Chemical Engineering, Arizona State University, Tempe, Arizona, United States of America
| | - Zongjian Jia
- Department of Chemical Engineering, Arizona State University, Tempe, Arizona, United States of America
| | - Michael R. Sierks
- Department of Chemical Engineering, Arizona State University, Tempe, Arizona, 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
| |
Collapse
|
29
|
Dragatsis I, Goldowitz D, Del Mar N, Deng YP, Meade CA, Liu L, Sun Z, Dietrich P, Yue J, Reiner A. CAG repeat lengths > or =335 attenuate the phenotype in the R6/2 Huntington's disease transgenic mouse. Neurobiol Dis 2008; 33:315-30. [PMID: 19027857 DOI: 10.1016/j.nbd.2008.10.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2008] [Revised: 10/15/2008] [Accepted: 10/17/2008] [Indexed: 11/17/2022] Open
Abstract
With spontaneous elongation of the CAG repeat in the R6/2 transgene to > or =335, resulting in a transgene protein too large for passive entry into nuclei via the nuclear pore, we observed an abrupt increase in lifespan to >20 weeks, compared to the 12 weeks common in R6/2 mice with 150 repeats. In the > or =335 CAG mice, large ubiquitinated aggregates of mutant protein were common in neuronal dendrites and perikaryal cytoplasm, but intranuclear aggregates were small and infrequent. Message and protein for the > or =335 CAG transgene were reduced to one-third that in 150 CAG R6/2 mice. Neurological and neurochemical abnormalities were delayed in onset and less severe than in 150 CAG R6/2 mice. These findings suggest that polyQ length and pathogenicity in Huntington's disease may not be linearly related, and pathogenicity may be less severe with extreme repeats. Both diminished mutant protein and reduced nuclear entry may contribute to phenotype attenuation.
Collapse
Affiliation(s)
- I Dragatsis
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|