1
|
Pérot JB, Brouillet E, Flament J. The contribution of preclinical magnetic resonance imaging and spectroscopy to Huntington's disease. Front Aging Neurosci 2024; 16:1306312. [PMID: 38414634 PMCID: PMC10896846 DOI: 10.3389/fnagi.2024.1306312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 01/24/2024] [Indexed: 02/29/2024] Open
Abstract
Huntington's disease is an inherited disorder characterized by psychiatric, cognitive, and motor symptoms due to degeneration of medium spiny neurons in the striatum. A prodromal phase precedes the onset, lasting decades. Current biomarkers include clinical score and striatal atrophy using Magnetic Resonance Imaging (MRI). These markers lack sensitivity for subtle cellular changes during the prodromal phase. MRI and MR spectroscopy offer different contrasts for assessing metabolic, microstructural, functional, or vascular alterations in the disease. They have been used in patients and mouse models. Mouse models can be of great interest to study a specific mechanism of the degenerative process, allow better understanding of the pathogenesis from the prodromal to the symptomatic phase, and to evaluate therapeutic efficacy. Mouse models can be divided into three different constructions: transgenic mice expressing exon-1 of human huntingtin (HTT), mice with an artificial chromosome expressing full-length human HTT, and knock-in mouse models with CAG expansion inserted in the murine htt gene. Several studies have used MRI/S to characterized these models. However, the multiplicity of modalities and mouse models available complicates the understanding of this rich corpus. The present review aims at giving an overview of results obtained using MRI/S for each mouse model of HD, to provide a useful resource for the conception of neuroimaging studies using mouse models of HD. Finally, despite difficulties in translating preclinical protocols to clinical applications, many biomarkers identified in preclinical models have already been evaluated in patients. This review also aims to cover this aspect to demonstrate the importance of MRI/S for studying HD.
Collapse
Affiliation(s)
- Jean-Baptiste Pérot
- Laboratoire des Maladies Neurodégénératives, Molecular Imaging Research Center, Commissariat à l’Energie Atomique et aux Energies Alternatives, Centre National de la Recherche Scientifique, Université Paris-Saclay, Fontenay-aux-Roses, France
- Institut du Cerveau – Paris Brain Institute – ICM, Sorbonne Université, Paris, France
| | - Emmanuel Brouillet
- Laboratoire des Maladies Neurodégénératives, Molecular Imaging Research Center, Commissariat à l’Energie Atomique et aux Energies Alternatives, Centre National de la Recherche Scientifique, Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Julien Flament
- Laboratoire des Maladies Neurodégénératives, Molecular Imaging Research Center, Commissariat à l’Energie Atomique et aux Energies Alternatives, Centre National de la Recherche Scientifique, Université Paris-Saclay, Fontenay-aux-Roses, France
| |
Collapse
|
2
|
Hanrahan J, Locke DP, Cahill LS. Magnetic Resonance Imaging to Detect Structural Brain Changes in Huntington's Disease: A Review of Data from Mouse Models. J Huntingtons Dis 2024; 13:279-299. [PMID: 39213087 PMCID: PMC11494634 DOI: 10.3233/jhd-240045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/14/2024] [Indexed: 09/04/2024]
Abstract
Structural magnetic resonance imaging (MRI) is a powerful tool to visualize 3D neuroanatomy and assess pathology and disease progression in neurodegenerative disorders such as Huntington's disease (HD). The development of mouse models of HD that reproduce many of the psychiatric, motor and cognitive impairments observed in human HD has improved our understanding of the disease and provided opportunities for testing novel therapies. Similar to the clinical scenario, MRI of mouse models of HD demonstrates onset and progression of brain pathology. Here, we provided an overview of the articles that used structural MRI in mouse models of HD to date, highlighting the differences between studies and models and describing gaps in the current state of knowledge and recommendations for future studies.
Collapse
Affiliation(s)
- Jenna Hanrahan
- Department of Chemistry, Memorial University of Newfoundland, St. John’s, Newfoundland and Labrador, Canada
| | - Drew P. Locke
- Department of Chemistry, Memorial University of Newfoundland, St. John’s, Newfoundland and Labrador, Canada
| | - Lindsay S. Cahill
- Department of Chemistry, Memorial University of Newfoundland, St. John’s, Newfoundland and Labrador, Canada
- Discipline of Radiology, Memorial University of Newfoundland, St. John’s, Newfoundland and Labrador, Canada
| |
Collapse
|
3
|
Morelli KH, Wu Q, Gosztyla ML, Liu H, Yao M, Zhang C, Chen J, Marina RJ, Lee K, Jones KL, Huang MY, Li A, Smith-Geater C, Thompson LM, Duan W, Yeo GW. An RNA-targeting CRISPR-Cas13d system alleviates disease-related phenotypes in Huntington's disease models. Nat Neurosci 2023; 26:27-38. [PMID: 36510111 PMCID: PMC9829537 DOI: 10.1038/s41593-022-01207-1] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 10/18/2022] [Indexed: 12/14/2022]
Abstract
Huntington's disease (HD) is a fatal, dominantly inherited neurodegenerative disorder caused by CAG trinucleotide expansion in exon 1 of the huntingtin (HTT) gene. Since the reduction of pathogenic mutant HTT messenger RNA is therapeutic, we developed a mutant allele-sensitive CAGEX RNA-targeting CRISPR-Cas13d system (Cas13d-CAGEX) that eliminates toxic CAGEX RNA in fibroblasts derived from patients with HD and induced pluripotent stem cell-derived neurons. We show that intrastriatal delivery of Cas13d-CAGEX via an adeno-associated viral vector selectively reduces mutant HTT mRNA and protein levels in the striatum of heterozygous zQ175 mice, a model of HD. This also led to improved motor coordination, attenuated striatal atrophy and reduction of mutant HTT protein aggregates. These phenotypic improvements lasted for at least eight months without adverse effects and with minimal off-target transcriptomic effects. Taken together, we demonstrate proof of principle of an RNA-targeting CRISPR-Cas13d system as a therapeutic approach for HD, a strategy with implications for the treatment of other dominantly inherited disorders.
Collapse
Affiliation(s)
- Kathryn H Morelli
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Qian Wu
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Maya L Gosztyla
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Hongshuai Liu
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Minmin Yao
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Chuangchuang Zhang
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jiaxu Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ryan J Marina
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Kari Lee
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Krysten L Jones
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Megan Y Huang
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Allison Li
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Charlene Smith-Geater
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, CA, USA
| | - Leslie M Thompson
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, CA, USA
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA
| | - Wenzhen Duan
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- The Solomon H Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Gene W Yeo
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA.
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA.
- Stem Cell Program, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
4
|
Temporal Characterization of Behavioral and Hippocampal Dysfunction in the YAC128 Mouse Model of Huntington’s Disease. Biomedicines 2022; 10:biomedicines10061433. [PMID: 35740454 PMCID: PMC9219853 DOI: 10.3390/biomedicines10061433] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/09/2022] [Accepted: 06/14/2022] [Indexed: 11/17/2022] Open
Abstract
Huntington’s disease (HD) is a genetic neurodegenerative disease characterized by motor, psychiatric, and cognitive symptoms. Emerging evidence suggests that emotional and cognitive deficits seen in HD may be related to hippocampal dysfunction. We used the YAC128 HD mouse model to perform a temporal characterization of the behavioral and hippocampal dysfunctions. Early and late symptomatic YAC128 mice exhibited depressive-like behavior, as demonstrated by increased immobility times in the Tail Suspension Test. In addition, YAC128 mice exhibited cognitive deficits in the Swimming T-maze Test during the late symptomatic stage. Except for a reduction in basal mitochondrial respiration, no significant deficits in the mitochondrial respiratory rates were observed in the hippocampus of late symptomatic YAC128 mice. In agreement, YAC128 animals did not present robust alterations in mitochondrial ultrastructural morphology. However, light and electron microscopy analysis revealed the presence of dark neurons characterized by the intense staining of granule cell bodies and shrunken nuclei and cytoplasm in the hippocampal dentate gyrus (DG) of late symptomatic YAC128 mice. Furthermore, structural alterations in the rough endoplasmic reticulum and Golgi apparatus were detected in the hippocampal DG of YAC128 mice by electron microscopy. These results clearly show a degenerative process in the hippocampal DG in late symptomatic YAC128 animals.
Collapse
|
5
|
Shenoy SA, Zheng S, Liu W, Dai Y, Liu Y, Hou Z, Mori S, Tang Y, Cheng J, Duan W, Li C. A novel and accurate full-length HTT mouse model for Huntington's disease. eLife 2022; 11:e70217. [PMID: 35023827 PMCID: PMC8758142 DOI: 10.7554/elife.70217] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 12/31/2021] [Indexed: 01/29/2023] Open
Abstract
Here, we report the generation and characterization of a novel Huntington's disease (HD) mouse model BAC226Q by using a bacterial artificial chromosome (BAC) system, expressing full-length human HTT with ~226 CAG-CAA repeats and containing endogenous human HTT promoter and regulatory elements. BAC226Q recapitulated a full-spectrum of age-dependent and progressive HD-like phenotypes without unwanted and erroneous phenotypes. BAC226Q mice developed normally, and gradually exhibited HD-like psychiatric and cognitive phenotypes at 2 months. From 3 to 4 months, BAC226Q mice showed robust progressive motor deficits. At 11 months, BAC226Q mice showed significant reduced life span, gradual weight loss and exhibited neuropathology including significant brain atrophy specific to striatum and cortex, striatal neuronal death, widespread huntingtin inclusions, and reactive pathology. Therefore, the novel BAC226Q mouse accurately recapitulating robust, age-dependent, progressive HD-like phenotypes will be a valuable tool for studying disease mechanisms, identifying biomarkers, and testing gene-targeting therapeutic approaches for HD.
Collapse
Affiliation(s)
- Sushila A Shenoy
- Department of Neuroscience, Weill Cornell Graduate School of Medical SciencesNew YorkUnited States
| | - Sushuang Zheng
- The MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking UniversityBeijingChina
| | - Wencheng Liu
- Department of Neuroscience, Weill Cornell Graduate School of Medical SciencesNew YorkUnited States
| | - Yuanyi Dai
- The MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking UniversityBeijingChina
| | - Yuanxiu Liu
- The MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking UniversityBeijingChina
| | - Zhipeng Hou
- The Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Susumu Mori
- The Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Yi Tang
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital Capital Medical University, National Center for Neurological DisordersBeijingChina
| | - Jerry Cheng
- Department of Computer Science, New York Institute of TechnologyNew YorkUnited States
| | - Wenzhen Duan
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences; Solomon H.Snyder Department of Neuroscience, Johns Hopkins University School of medicineBaltimoreUnited States
| | - Chenjian Li
- The MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking UniversityBeijingChina
| |
Collapse
|
6
|
Liu H, Zhang C, Xu J, Jin J, Cheng L, Miao X, Wu Q, Wei Z, Liu P, Lu H, van Zijl PCM, Ross CA, Hua J, Duan W. Huntingtin silencing delays onset and slows progression of Huntington's disease: a biomarker study. Brain 2021; 144:3101-3113. [PMID: 34043007 PMCID: PMC8634120 DOI: 10.1093/brain/awab190] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 04/19/2021] [Accepted: 04/29/2021] [Indexed: 01/29/2023] Open
Abstract
Huntington's disease is a dominantly inherited, fatal neurodegenerative disorder caused by a CAG expansion in the huntingtin (HTT) gene, coding for pathological mutant HTT protein (mHTT). Because of its gain-of-function mechanism and monogenic aetiology, strategies to lower HTT are being actively investigated as disease-modifying therapies. Most approaches are currently targeted at the manifest stage, where clinical outcomes are used to evaluate the effectiveness of therapy. However, as almost 50% of striatal volume has been lost at the time of onset of clinical manifest, it would be preferable to begin therapy in the premanifest period. An unmet challenge is how to evaluate therapeutic efficacy before the presence of clinical symptoms as outcome measures. To address this, we aim to develop non-invasive sensitive biomarkers that provide insight into therapeutic efficacy in the premanifest stage of Huntington's disease. In this study, we mapped the temporal trajectories of arteriolar cerebral blood volumes (CBVa) using inflow-based vascular-space-occupancy (iVASO) MRI in the heterozygous zQ175 mice, a full-length mHTT expressing and slowly progressing model with a premanifest period as in human Huntington's disease. Significantly elevated CBVa was evident in premanifest zQ175 mice prior to motor deficits and striatal atrophy, recapitulating altered CBVa in human premanifest Huntington's disease. CRISPR/Cas9-mediated non-allele-specific HTT silencing in striatal neurons restored altered CBVa in premanifest zQ175 mice, delayed onset of striatal atrophy, and slowed the progression of motor phenotype and brain pathology. This study-for the first time-shows that a non-invasive functional MRI measure detects therapeutic efficacy in the premanifest stage and demonstrates long-term benefits of a non-allele-selective HTT silencing treatment introduced in the premanifest Huntington's disease.
Collapse
Affiliation(s)
- Hongshuai Liu
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Chuangchuang Zhang
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jiadi Xu
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
- The Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jing Jin
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Liam Cheng
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xinyuan Miao
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
- The Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Qian Wu
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zhiliang Wei
- The Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peiying Liu
- The Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hanzhang Lu
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
- The Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Peter C M van Zijl
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
- The Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christopher A Ross
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jun Hua
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
- The Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Wenzhen Duan
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
7
|
Kim A, Lalonde K, Truesdell A, Gomes Welter P, Brocardo PS, Rosenstock TR, Gil-Mohapel J. New Avenues for the Treatment of Huntington's Disease. Int J Mol Sci 2021; 22:ijms22168363. [PMID: 34445070 PMCID: PMC8394361 DOI: 10.3390/ijms22168363] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 12/11/2022] Open
Abstract
Huntington’s disease (HD) is a neurodegenerative disorder caused by a CAG expansion in the HD gene. The disease is characterized by neurodegeneration, particularly in the striatum and cortex. The first symptoms usually appear in mid-life and include cognitive deficits and motor disturbances that progress over time. Despite being a genetic disorder with a known cause, several mechanisms are thought to contribute to neurodegeneration in HD, and numerous pre-clinical and clinical studies have been conducted and are currently underway to test the efficacy of therapeutic approaches targeting some of these mechanisms with varying degrees of success. Although current clinical trials may lead to the identification or refinement of treatments that are likely to improve the quality of life of those living with HD, major efforts continue to be invested at the pre-clinical level, with numerous studies testing novel approaches that show promise as disease-modifying strategies. This review offers a detailed overview of the currently approved treatment options for HD and the clinical trials for this neurodegenerative disorder that are underway and concludes by discussing potential disease-modifying treatments that have shown promise in pre-clinical studies, including increasing neurotropic support, modulating autophagy, epigenetic and genetic manipulations, and the use of nanocarriers and stem cells.
Collapse
Affiliation(s)
- Amy Kim
- Island Medical Program and Faculty of Medicine, University of British Columbia, Victoria, BC V8P 5C2, Canada; (A.K.); (K.L.)
| | - Kathryn Lalonde
- Island Medical Program and Faculty of Medicine, University of British Columbia, Victoria, BC V8P 5C2, Canada; (A.K.); (K.L.)
| | - Aaron Truesdell
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada;
- Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada
| | - Priscilla Gomes Welter
- Neuroscience Graduate Program, Federal University of Santa Catarina, Florianópolis 88040-900, Brazil; (P.G.W.); (P.S.B.)
| | - Patricia S. Brocardo
- Neuroscience Graduate Program, Federal University of Santa Catarina, Florianópolis 88040-900, Brazil; (P.G.W.); (P.S.B.)
| | - Tatiana R. Rosenstock
- Institute of Cancer and Genomic Science, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK;
- Department of Pharmacology, University of São Paulo, São Paulo 05508-000, Brazil
| | - Joana Gil-Mohapel
- Island Medical Program and Faculty of Medicine, University of British Columbia, Victoria, BC V8P 5C2, Canada; (A.K.); (K.L.)
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada;
- Correspondence: ; Tel.: +1-250-472-4597; Fax: +1-250-472-5505
| |
Collapse
|
8
|
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: 27] [Impact Index Per Article: 6.8] [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
|
9
|
Simmons DA, Mills BD, Butler Iii RR, Kuan J, McHugh TLM, Akers C, Zhou J, Syriani W, Grouban M, Zeineh M, Longo FM. Neuroimaging, Urinary, and Plasma Biomarkers of Treatment Response in Huntington's Disease: Preclinical Evidence with the p75 NTR Ligand LM11A-31. Neurotherapeutics 2021; 18:1039-1063. [PMID: 33786806 PMCID: PMC8423954 DOI: 10.1007/s13311-021-01023-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/02/2021] [Indexed: 12/13/2022] Open
Abstract
Huntington's disease (HD) is caused by an expansion of the CAG repeat in the huntingtin gene leading to preferential neurodegeneration of the striatum. Disease-modifying treatments are not yet available to HD patients and their development would be facilitated by translatable pharmacodynamic biomarkers. Multi-modal magnetic resonance imaging (MRI) and plasma cytokines have been suggested as disease onset/progression biomarkers, but their ability to detect treatment efficacy is understudied. This study used the R6/2 mouse model of HD to assess if structural neuroimaging and biofluid assays can detect treatment response using as a prototype the small molecule p75NTR ligand LM11A-31, shown previously to reduce HD phenotypes in these mice. LM11A-31 alleviated volume reductions in multiple brain regions, including striatum, of vehicle-treated R6/2 mice relative to wild-types (WTs), as assessed with in vivo MRI. LM11A-31 also normalized changes in diffusion tensor imaging (DTI) metrics and diminished increases in certain plasma cytokine levels, including tumor necrosis factor-alpha and interleukin-6, in R6/2 mice. Finally, R6/2-vehicle mice had increased urinary levels of the p75NTR extracellular domain (ecd), a cleavage product released with pro-apoptotic ligand binding that detects the progression of other neurodegenerative diseases; LM11A-31 reduced this increase. These results are the first to show that urinary p75NTR-ecd levels are elevated in an HD mouse model and can be used to detect therapeutic effects. These data also indicate that multi-modal MRI and plasma cytokine levels may be effective pharmacodynamic biomarkers and that using combinations of these markers would be a viable and powerful option for clinical trials.
Collapse
Affiliation(s)
- Danielle A Simmons
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Brian D Mills
- Department of Radiology, Stanford University Medical Center, Stanford, CA, 94305, USA
| | - Robert R Butler Iii
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Jason Kuan
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Tyne L M McHugh
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Carolyn Akers
- Department of Radiology, Stanford University Medical Center, Stanford, CA, 94305, USA
| | - James Zhou
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Wassim Syriani
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Maged Grouban
- Department of Radiology, Stanford University Medical Center, Stanford, CA, 94305, USA
| | - Michael Zeineh
- Department of Radiology, Stanford University Medical Center, Stanford, CA, 94305, USA
| | - Frank M Longo
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, 94305, USA
| |
Collapse
|
10
|
Salamon A, Maszlag-Török R, Veres G, Boros FA, Vágvölgyi-Sümegi E, Somogyi A, Vécsei L, Klivényi P, Zádori D. Cerebellar Predominant Increase in mRNA Expression Levels of Sirt1 and Sirt3 Isoforms in a Transgenic Mouse Model of Huntington's Disease. Neurochem Res 2020; 45:2072-2081. [PMID: 32524313 PMCID: PMC7423862 DOI: 10.1007/s11064-020-03069-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 05/01/2020] [Accepted: 06/04/2020] [Indexed: 01/28/2023]
Abstract
The potential role of Sirt1 and Sirt2 subtypes of Sirtuins (class III NAD+-dependent deacetylases) in the pathogenesis of Huntington’s disease (HD) has been extensively studied yielding some controversial results. However, data regarding the involvement of Sirt3 and their variants in HD are considerably limited. The aim of this study was to assess the expression pattern of Sirt1 and three Sirt3 mRNA isoforms (Sirt3-M1/2/3) in the striatum, cortex and cerebellum in respect of the effect of gender, age and the presence of the transgene using the N171-82Q transgenic mouse model of HD. Striatal, cortical and cerebellar Sirt1-Fl and Sirt3-M1/2/3 mRNA levels were measured in 8, 12 and 16 weeks old N171-82Q transgenic mice and in their wild-type littermates. Regarding the striatum and cortex, the presence of the transgene resulted in a significant increase in Sirt3-M3 and Sirt1 mRNA levels, respectively, whereas in case of the cerebellum the transgene resulted in increased expression of all the assessed subtypes and isoforms. Aging exerted minor influence on Sirt mRNA expression levels, both in transgene carriers and in their wild-type littermates, and there was no interaction between the presence of the transgene and aging. Furthermore, there was no difference between genders. The unequivocal cerebellar Sirtuin activation with presumed compensatory role suggests that the cerebellum might be another key player in HD in addition to the most severely affected striatum. The mitochondrially acting Sirt3 may serve as an interesting novel therapeutic target in this deleterious condition.
Collapse
Affiliation(s)
- Andras Salamon
- Department of Neurology, Interdisciplinary Excellence Center, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis u. 6, Szeged, 6725, Hungary
| | - Rita Maszlag-Török
- Department of Neurology, Interdisciplinary Excellence Center, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis u. 6, Szeged, 6725, Hungary
| | - Gábor Veres
- Department of Neurology, Interdisciplinary Excellence Center, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis u. 6, Szeged, 6725, Hungary
- MTA-SZTE Neuroscience Research Group of the Hungarian Academy of Sciences, Budapest, Hungary
| | - Fanni Annamária Boros
- Department of Neurology, Interdisciplinary Excellence Center, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis u. 6, Szeged, 6725, Hungary
| | - Evelin Vágvölgyi-Sümegi
- Department of Neurology, Interdisciplinary Excellence Center, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis u. 6, Szeged, 6725, Hungary
| | - Anett Somogyi
- Department of Neurology, Interdisciplinary Excellence Center, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis u. 6, Szeged, 6725, Hungary
| | - László Vécsei
- Department of Neurology, Interdisciplinary Excellence Center, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis u. 6, Szeged, 6725, Hungary
- MTA-SZTE Neuroscience Research Group of the Hungarian Academy of Sciences, Budapest, Hungary
| | - Péter Klivényi
- Department of Neurology, Interdisciplinary Excellence Center, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis u. 6, Szeged, 6725, Hungary
| | - Dénes Zádori
- Department of Neurology, Interdisciplinary Excellence Center, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Semmelweis u. 6, Szeged, 6725, Hungary.
| |
Collapse
|
11
|
Jiang M, Zhang X, Liu H, LeBron J, Alexandris A, Peng Q, Gu H, Yang F, Li Y, Wang R, Hou Z, Arbez N, Ren Q, Dong JL, Whela E, Wang R, Ratovitski T, Troncoso JC, Mori S, Ross CA, Lim J, Duan W. Nemo-like kinase reduces mutant huntingtin levels and mitigates Huntington's disease. Hum Mol Genet 2020; 29:1340-1352. [PMID: 32242231 PMCID: PMC7254850 DOI: 10.1093/hmg/ddaa061] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/15/2020] [Accepted: 03/30/2020] [Indexed: 11/12/2022] Open
Abstract
Nemo-like kinase (NLK), an evolutionarily conserved serine/threonine kinase, is highly expressed in the brain, but its function in the adult brain remains not well understood. In this study, we identify NLK as an interactor of huntingtin protein (HTT). We report that NLK levels are significantly decreased in HD human brain and HD models. Importantly, overexpression of NLK in the striatum attenuates brain atrophy, preserves striatal DARPP32 levels and reduces mutant HTT (mHTT) aggregation in HD mice. In contrast, genetic reduction of NLK exacerbates brain atrophy and loss of DARPP32 in HD mice. Moreover, we demonstrate that NLK lowers mHTT levels in a kinase activity-dependent manner, while having no significant effect on normal HTT protein levels in mouse striatal cells, human cells and HD mouse models. The NLK-mediated lowering of mHTT is associated with enhanced phosphorylation of mHTT. Phosphorylation defective mutation of serine at amino acid 120 (S120) abolishes the mHTT-lowering effect of NLK, suggesting that S120 phosphorylation is an important step in the NLK-mediated lowering of mHTT. A further mechanistic study suggests that NLK promotes mHTT ubiquitination and degradation via the proteasome pathway. Taken together, our results indicate a protective role of NLK in HD and reveal a new molecular target to reduce mHTT levels.
Collapse
Affiliation(s)
- Mali Jiang
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xiaoyan Zhang
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hongshuai Liu
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jared LeBron
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Athanasios Alexandris
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Qi Peng
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hao Gu
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Fanghan Yang
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yuchen Li
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ruiling Wang
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zhipeng Hou
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nicolas Arbez
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Qianwei Ren
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jen-Li Dong
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Emma Whela
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ronald Wang
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tamara Ratovitski
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Juan C Troncoso
- Division of Neuropathology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Susumu Mori
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christopher A Ross
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Janghoo Lim
- Departments of Genetics and of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| | - Wenzhen Duan
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
12
|
Petrella LI, Castelhano JM, Ribeiro M, Sereno JV, Gonçalves SI, Laço MN, Hayden MR, Rego AC, Castelo-Branco M. A whole brain longitudinal study in the YAC128 mouse model of Huntington's disease shows distinct trajectories of neurochemical, structural connectivity and volumetric changes. Hum Mol Genet 2019; 27:2125-2137. [PMID: 29668904 DOI: 10.1093/hmg/ddy119] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 03/26/2018] [Indexed: 12/20/2022] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disorder causing cognitive and motor impairments, evolving to death within 15-20 years after symptom onset. We previously established a mouse model with the entire human HD gene containing 128 CAG repeats (YAC128) which accurately recapitulates the natural history of the human disease. Defined time points in this natural history enable the understanding of longitudinal trajectories from the neurochemical and structural points of view using non-invasive high-resolution multi-modal imaging. Accordingly, we designed a longitudinal structural imaging (MRI and DTI) and spectroscopy (1H-MRS) study in YAC128, at 3, 6, 9 and 12 months of age, at 9.4 T. Structural analysis (MRI/DTI), confirmed that the striatum is the earliest affected brain region, but other regions were also identified through connectivity analysis (pre-frontal cortex, hippocampus, globus pallidus and thalamus), suggesting a striking homology with the human disease. Importantly, we found for the first time, a negative correlation between striatal and hippocampal changes only in YAC128. In fact, the striatum showed accelerated volumetric decay in HD, as opposed to the hippocampus. Neurochemical analysis of the HD striatum suggested early neurometabolic alterations in neurotransmission and metabolism, with a significant increase in striatal GABA levels, and specifically anticorrelated levels of N-acetyl aspartate and taurine, suggesting that the later is homeostatically adjusted for neuroprotection, as neural loss, indicated by the former, is progressing. These results provide novel insights into the natural history of HD and prove a valuable role for longitudinal multi-modal panels of structural and metabolite/neurotransmission in the YAC128 model.
Collapse
Affiliation(s)
- Lorena I Petrella
- Institute of Nuclear Science Applied to Health, University of Coimbra, 3000-548 Coimbra, Portugal.,Center for Neuroscience and Cell Biology-Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, 3000-548 Coimbra, Portugal
| | - João M Castelhano
- Institute of Nuclear Science Applied to Health, University of Coimbra, 3000-548 Coimbra, Portugal.,Center for Neuroscience and Cell Biology-Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Mario Ribeiro
- Institute of Nuclear Science Applied to Health, University of Coimbra, 3000-548 Coimbra, Portugal.,Center for Neuroscience and Cell Biology-Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, 3000-548 Coimbra, Portugal
| | - José V Sereno
- Institute of Nuclear Science Applied to Health, University of Coimbra, 3000-548 Coimbra, Portugal.,Center for Neuroscience and Cell Biology-Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Sónia I Gonçalves
- Institute of Nuclear Science Applied to Health, University of Coimbra, 3000-548 Coimbra, Portugal.,Center for Neuroscience and Cell Biology-Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, 3000-548 Coimbra, Portugal.,Neuroplasticity and Neural Activity Laboratory, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal
| | - Mário N Laço
- Center for Neuroscience and Cell Biology-Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Michael R Hayden
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | - A Cristina Rego
- Center for Neuroscience and Cell Biology-Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, 3000-548 Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Miguel Castelo-Branco
- Institute of Nuclear Science Applied to Health, University of Coimbra, 3000-548 Coimbra, Portugal.,Center for Neuroscience and Cell Biology-Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, 3000-548 Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| |
Collapse
|
13
|
Zhou X, Li G, Kaplan A, Gaschler MM, Zhang X, Hou Z, Jiang M, Zott R, Cremers S, Stockwell BR, Duan W. Small molecule modulator of protein disulfide isomerase attenuates mutant huntingtin toxicity and inhibits endoplasmic reticulum stress in a mouse model of Huntington's disease. Hum Mol Genet 2019; 27:1545-1555. [PMID: 29462355 DOI: 10.1093/hmg/ddy061] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 02/12/2018] [Indexed: 11/14/2022] Open
Abstract
Huntington's disease (HD) is caused by a cytosine-adenine-guanine (CAG) trinucleotide repeat expansion in the huntingtin (HTT) gene encoding an elongated polyglutamine tract within the N-terminal of the huntingtin protein (Htt) and leads to Htt misfolding, aberrant protein aggregation, and progressive appearance of disease symptoms. Chronic activation of endoplasmic reticulum (ER) stress by mutant Htt (mHtt) results in cellular dysfunction and ultimately cell death. Protein disulfide isomerase (PDI) is a chaperone protein located in the ER. Our previous studies demonstrated that mHtt caused PDI to accumulate at mitochondria-associated ER membranes and triggered cell death, and that modulating PDI activity using small molecules protected cells again mHtt toxicity in cell and brain slice models of HD. In this study, we demonstrated that PDI is upregulated in the HD human brain, in cell and mouse models. Chronic administration of a reversible, brain penetrable small molecule PDI modulator, LOC14 (20 mg/kg/day), significantly improved motor function, attenuated brain atrophy and extended survival in the N171-82Q HD mice. Moreover, LOC14 preserved medium spiny neuronal marker dopamine- and cyclic-AMP-regulated phosphoprotein of molecular weight 32 000 (DARPP32) levels in the striatum of HD mice. Mechanistic study revealed that LOC14 suppressed mHtt-induced ER stress, indicated by repressing the abnormally upregulated ER stress proteins in HD models. These findings suggest that LOC14 is promising to be further optimized for clinical trials of HD, and modulation of signaling pathways coping with ER stress may constitute an attractive approach to reduce mHtt toxicity and identify new therapeutic targets for treatment of HD.
Collapse
Affiliation(s)
- Xiao Zhou
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Gang Li
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Department of Pharmacology, Pharmacy School, Inner Mongolian Medical University, Hohhot, Inner Mongolian, 010110, China
| | - Anna Kaplan
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | | | - Xiaoyan Zhang
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Zhipeng Hou
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Mali Jiang
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Roseann Zott
- Irving Institute for Clinical and Translational Research, Columbia University Medical Center, New York, NY 10027, USA
| | - Serge Cremers
- Irving Institute for Clinical and Translational Research, Columbia University Medical Center, New York, NY 10027, USA.,Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10027, USA.,Department of Medicine, Columbia University Medical Center, New York, NY 10027, USA
| | - Brent R Stockwell
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA.,Department of Chemistry, Columbia University, New York, NY 10027, USA
| | - Wenzhen Duan
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
14
|
Antidepressant Effects of Probucol on Early-Symptomatic YAC128 Transgenic Mice for Huntington's Disease. Neural Plast 2018; 2018:4056383. [PMID: 30186318 PMCID: PMC6112232 DOI: 10.1155/2018/4056383] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 06/28/2018] [Accepted: 07/26/2018] [Indexed: 11/17/2022] Open
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder caused by a trinucleotide expansion in the HD gene, resulting in an extended polyglutamine tract in the protein huntingtin. HD is traditionally viewed as a movement disorder, but cognitive and neuropsychiatric symptoms also contribute to the clinical presentation. Depression is one of the most common psychiatric disturbances in HD, present even before manifestation of motor symptoms. Diagnosis and treatment of depression in HD-affected individuals are essential aspects of clinical management in this population, especially owing to the high risk of suicide. This study investigated whether chronic administration of the antioxidant probucol improved motor and affective symptoms as well as hippocampal neurogenic function in the YAC128 transgenic mouse model of HD during the early- to mild-symptomatic stages of disease progression. The motor performance and affective symptoms were monitored using well-validated behavioral tests in YAC128 mice and age-matched wild-type littermates at 2, 4, and 6 months of age, after 1, 3, or 5 months of treatment with probucol (30 mg/kg/day via water supplementation, starting on postnatal day 30). Endogenous markers were used to assess the effect of probucol on cell proliferation (Ki-67 and proliferation cell nuclear antigen (PCNA)) and neuronal differentiation (doublecortin (DCX)) in the hippocampal dentate gyrus (DG). Chronic treatment with probucol reduced the occurrence of depressive-like behaviors in early- and mild-symptomatic YAC128 mice. Functional improvements were not accompanied by increased progenitor cell proliferation and neuronal differentiation. Our findings provide evidence that administration of probucol may be of clinical benefit in the management of early- to mild-symptomatic HD.
Collapse
|
15
|
Kim H, Kim JH, Possin KL, Winer J, Geschwind MD, Xu D, Hess CP. Surface-based morphometry reveals caudate subnuclear structural damage in patients with premotor Huntington disease. Brain Imaging Behav 2018; 11:1365-1372. [PMID: 27730480 DOI: 10.1007/s11682-016-9616-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In patients with premotor Huntington's disease (pmHD), literature has reported decreases in caudate volume. However, the regional vulnerability of the caudate nucleus to pmHD remains to be clarified. We aimed to determine whether regional structural damage of the caudate nucleus was present in pmHD and was correlated with clinical profile using a surface-based morphometric technique applied to T1-weighted MRI. The study cohort consisted of 14 volunteers with genetically confirmed pmHD (6 males; 41.8 ± 13.2 years) and 11 age- and sex-matched controls (5 males; 46.2 ± 11.9 years, p > 0.3). On 3-T T1-weighted images, bilateral caudate volumes were manually delineated. The resulting labels were converted to a surface, triangulated with 1002 points equally distributed across subjects using SPHARM-PDM. Displacement vectors were then computed between each individual and a template surface representing the whole cohort. Computing point-wise Jacobian determinants (JD) from these vectors quantified local volumes. We found decreases in bilateral global caudate volumes in the pmHD group compared to controls (t = 3.4; p = 0.002). Point-wise analysis of local volumes mapped caudate atrophy in pmHD primarily onto medial surface (t > 2.7; FDR < 0.05), with most pronounced changes in anteromedial subdivision. In a combined group of patients and controls, volume within the area presenting significant group difference was positively correlated with scores of executive function (r = 0.7; p < 0.001) and working memory (r = 0.6; p = 0.002). In patients, the caudate atrophy was associated with increase in disease burden (r = 0.7; p = 0.005). Caudate subnuclear atrophy measured using our surface-based morphometric technique is evident in pmHD, correlates with clinical variables, and may provide a more sensitive biomarker than global volumes.
Collapse
Affiliation(s)
- Hosung Kim
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA.
| | - Ji-Hoon Kim
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA.,Department of Radiology, Seoul National Univeristy Hospital, Seoul, South Korea
| | - Katherine L Possin
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Joseph Winer
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Michael D Geschwind
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Duan Xu
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Christopher P Hess
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA.,Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
16
|
Mutant Huntingtin Causes a Selective Decrease in the Expression of Synaptic Vesicle Protein 2C. Neurosci Bull 2018; 34:747-758. [PMID: 29713895 DOI: 10.1007/s12264-018-0230-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 03/24/2018] [Indexed: 12/11/2022] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disease caused by a polyglutamine expansion in the huntingtin (Htt) protein. Mutant Htt causes synaptic transmission dysfunctions by interfering in the expression of synaptic proteins, leading to early HD symptoms. Synaptic vesicle proteins 2 (SV2s), a family of synaptic vesicle proteins including 3 members, SV2A, SV2B, and SV2C, plays important roles in synaptic physiology. Here, we investigated whether the expression of SV2s is affected by mutant Htt in the brains of HD transgenic (TG) mice and Neuro2a mouse neuroblastoma cells (N2a cells) expressing mutant Htt. Western blot analysis showed that the protein levels of SV2A and SV2B were not significantly changed in the brains of HD TG mice expressing mutant Htt with 82 glutamine repeats. However, in the TG mouse brain there was a dramatic decrease in the protein level of SV2C, which has a restricted distribution pattern in regions particularly vulnerable in HD. Immunostaining revealed that the immunoreactivity of SV2C was progressively weakened in the basal ganglia and hippocampus of TG mice. RT-PCR demonstrated that the mRNA level of SV2C progressively declined in the TG mouse brain without detectable changes in the mRNA levels of SV2A and SV2B, indicating that mutant Htt selectively inhibits the transcriptional expression of SV2C. Furthermore, we found that only SV2C expression was progressively inhibited in N2a cells expressing a mutant Htt containing 120 glutamine repeats. These findings suggest that the synaptic dysfunction in HD results from the mutant Htt-mediated inhibition of SV2C transcriptional expression. These data also imply that the restricted distribution and decreased expression of SV2C contribute to the brain region-selective pathology of HD.
Collapse
|
17
|
Miniarikova J, Evers MM, Konstantinova P. Translation of MicroRNA-Based Huntingtin-Lowering Therapies from Preclinical Studies to the Clinic. Mol Ther 2018; 26:947-962. [PMID: 29503201 DOI: 10.1016/j.ymthe.2018.02.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 01/30/2018] [Accepted: 02/05/2018] [Indexed: 12/21/2022] Open
Abstract
The single mutation underlying the fatal neuropathology of Huntington's disease (HD) is a CAG triplet expansion in exon 1 of the huntingtin (HTT) gene, which gives rise to a toxic mutant HTT protein. There have been a number of not yet successful therapeutic advances in the treatment of HD. The current excitement in the HD field is due to the recent development of therapies targeting the culprit of HD either at the DNA or RNA level to reduce the overall mutant HTT protein. In this review, we briefly describe short-term and long-term HTT-lowering strategies targeting HTT transcripts. One of the most advanced HTT-lowering strategies is a microRNA (miRNA)-based gene therapy delivered by a single administration of an adeno-associated viral (AAV) vector to the HD patient. We outline the outcome measures for the miRNA-based HTT-lowering therapy in the context of preclinical evaluation in HD animal and cell models. We highlight the strengths and ongoing queries of the HTT-lowering gene therapy as an HD intervention with a potential disease-modifying effect. This review provides a perspective on the fast-developing HTT-lowering therapies for HD and their translation to the clinic based on existing knowledge in preclinical models.
Collapse
Affiliation(s)
- Jana Miniarikova
- Department of Research and Development, uniQure, Amsterdam, the Netherlands; Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Melvin M Evers
- Department of Research and Development, uniQure, Amsterdam, the Netherlands
| | | |
Collapse
|
18
|
Resting-state functional MRI reveals altered brain connectivity and its correlation with motor dysfunction in a mouse model of Huntington's disease. Sci Rep 2017; 7:16742. [PMID: 29196686 PMCID: PMC5711837 DOI: 10.1038/s41598-017-17026-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 11/21/2017] [Indexed: 11/29/2022] Open
Abstract
Huntington’s disease (HD) is an autosomal dominant inherited neurodegenerative disorder, and no cure is available currently. Treatment of HD is likely to be most beneficial in the early, possibly pre-manifestation stage. The challenge is to determine the best time for intervention and evaluate putative efficacy in the absence of clinical symptoms. Resting-state functional MRI may represent a promising tool to develop biomarker reflecting early neuronal dysfunction in HD brain, because it can examine multiple brain networks without confounding effects of cognitive ability, which makes the resting-state fMRI promising as a translational bridge between preclinical study in animal models and clinical findings in HD patients. In this study, we examined brain regional connectivity and its correlation to brain atrophy, as well as motor function in the 18-week-old N171-82Q HD mice. HD mice exhibited significantly altered functional connectivity in multiple networks. Particularly, the weaker intra-striatum connectivity was positively correlated with striatal atrophy, while striatum-retrosplenial cortex connectivity is negatively correlated with striatal atrophy. The resting-state brain regional connectivity had no significant correlation with motor deficits in HD mice. Our results suggest that altered brain connectivity detected by resting-state fMRI might serve as an early disease biomarker in HD.
Collapse
|
19
|
Mouse models of neurodegenerative disease: preclinical imaging and neurovascular component. Brain Imaging Behav 2017; 12:1160-1196. [PMID: 29075922 DOI: 10.1007/s11682-017-9770-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Neurodegenerative diseases represent great challenges for basic science and clinical medicine because of their prevalence, pathologies, lack of mechanism-based treatments, and impacts on individuals. Translational research might contribute to the study of neurodegenerative diseases. The mouse has become a key model for studying disease mechanisms that might recapitulate in part some aspects of the corresponding human diseases. Neurodegenerative disorders are very complicated and multifactorial. This has to be taken in account when testing drugs. Most of the drugs screening in mice are very difficult to be interpretated and often useless. Mouse models could be condiderated a 'pathway models', rather than as models for the whole complicated construct that makes a human disease. Non-invasive in vivo imaging in mice has gained increasing interest in preclinical research in the last years thanks to the availability of high-resolution single-photon emission computed tomography (SPECT), positron emission tomography (PET), high field Magnetic resonance, Optical Imaging scanners and of highly specific contrast agents. Behavioral test are useful tool to characterize different animal models of neurodegenerative pathology. Furthermore, many authors have observed vascular pathological features associated to the different neurodegenerative disorders. Aim of this review is to focus on the different existing animal models of neurodegenerative disorders, describe behavioral tests and preclinical imaging techniques used for diagnose and describe the vascular pathological features associated to these diseases.
Collapse
|
20
|
Wu B, Jiang M, Peng Q, Li G, Hou Z, Milne GL, Mori S, Alonso R, Geisler JG, Duan W. 2,4 DNP improves motor function, preserves medium spiny neuronal identity, and reduces oxidative stress in a mouse model of Huntington's disease. Exp Neurol 2017; 293:83-90. [PMID: 28359739 PMCID: PMC9912814 DOI: 10.1016/j.expneurol.2017.03.020] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 03/17/2017] [Accepted: 03/26/2017] [Indexed: 12/18/2022]
Abstract
Huntington's disease (HD) is a neurodegenerative disorder caused by a CAG repeat expansion in the first exon of the gene huntingtin. There is no treatment to prevent or delay the disease course of HD currently. Oxidative stress and mitochondrial dysfunction have emerged as key determinants of the disease progression in HD. Therefore, counteracting mutant huntingtin (mHtt)-induced oxidative stress and mitochondrial dysfunction appears as a new approach to treat this devastating disease. Interestingly, mild mitochondrial uncoupling improves neuronal resistance to stress and facilitates neuronal survival. Mild mitochondrial uncoupling can be induced by the proper dose of 2,4-dinitrophenol (DNP), a proton ionophore that was previously used for weight loss. In this study, we evaluated the effects of chronic administration of DNP at three doses (0.5, 1, 5mg/kg/day) on mHtt-induced behavioral deficits and cellular abnormalities in the N171-82Q HD mouse model. DNP at a low dose (1mg/kg/day) significantly improved motor function and preserved medium spiny neuronal marker DARPP32 and postsynaptic protein PSD95 in the striatum of HD mice. Further mechanistic study suggests that DNP at this dose reduced oxidative stress in HD mice, which was indicated by reduced levels of F2-isoprostanes in the brain of HD mice treated with DNP. Our data indicated that DNP provided behavioral benefit and neuroprotective effect at a weight neutral dose in HD mice, suggesting that the potential value of repositioning DNP to HD treatment is warranted in well-controlled clinical trials in HD.
Collapse
Affiliation(s)
- Bin Wu
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States,Department of General Practice, The First hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Mali Jiang
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Qi Peng
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Gang Li
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States,Department of Pharmacology, Inner Mongolian Medical University School of Pharmacy, Hohhot, Inner Mongolian, China
| | - Zhipeng Hou
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Ginger L. Milne
- Eicosanoid Core Laboratory, Division of Clinical Pharmacology, Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Susumu Mori
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Robert Alonso
- Mitochon Pharmaceuticals Inc., Radnor, PA, United States
| | | | - Wenzhen Duan
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
21
|
Fang F, Peng T, Yang S, Wang W, Zhang Y, Li H. Lycium barbarum
polysaccharide attenuates the cytotoxicity of mutant huntingtin and increases the activity of AKT. Int J Dev Neurosci 2016; 52:66-74. [DOI: 10.1016/j.ijdevneu.2016.05.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 05/02/2016] [Accepted: 05/02/2016] [Indexed: 01/13/2023] Open
Affiliation(s)
- Fang Fang
- Division of Histology and EmbryologyDepartment of AnatomyTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030People's Republic of China
| | - Ting Peng
- Division of Histology and EmbryologyDepartment of AnatomyTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030People's Republic of China
- Institute for Brain SciencesTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030People's Republic of China
- Collaborative Innovation Center for Brain ScienceHuazhong University of Science and TechnologyWuhan430030People's Republic of China
| | - Shiming Yang
- Division of Histology and EmbryologyDepartment of AnatomyTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030People's Republic of China
| | - Weixi Wang
- Division of Histology and EmbryologyDepartment of AnatomyTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030People's Republic of China
| | - Yinong Zhang
- Division of Histology and EmbryologyDepartment of AnatomyTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030People's Republic of China
| | - He Li
- Division of Histology and EmbryologyDepartment of AnatomyTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030People's Republic of China
- Institute for Brain SciencesTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030People's Republic of China
- Collaborative Innovation Center for Brain ScienceHuazhong University of Science and TechnologyWuhan430030People's Republic of China
| |
Collapse
|
22
|
Fox J, Lu Z, Barrows L. Thiol-disulfide Oxidoreductases TRX1 and TMX3 Decrease Neuronal Atrophy in a Lentiviral Mouse Model of Huntington's Disease. PLOS CURRENTS 2015; 7. [PMID: 26664998 PMCID: PMC4650837 DOI: 10.1371/currents.hd.b966ec2eca8e2d89d2bb4d020be4351e] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Huntington’s disease (HD) is caused by a trinucleotide CAG repeat in the
huntingtin gene (HTT) that results in expression of a polyglutamine-expanded
mutant huntingtin protein (mHTT). N-terminal fragments of mHTT accumulate in
brain neurons and glia as soluble monomeric and oligomeric species as well as
insoluble protein aggregates and drive the disease process. Decreasing mHTT
levels in brain provides protection and reversal of disease signs in HD mice
making mHTT a prime target for disease modification. There is evidence for
aberrant thiol oxidation within mHTT and other proteins in HD models. Based on
this, we hypothesized that a specific thiol-disulfide oxidoreductase exists that
decreases mHTT levels in cells and provides protection in HD mice. We undertook
an in-vitro genetic screen of key thiol-disulfide oxidoreductases then completed
secondary screens to identify those with mHTT decreasing properties. Our
in-vitro experiments identified thioredoxin 1 and thioredoxin-related
transmembrane protein 3 as proteins that decrease soluble mHTT levels in
cultured cells. Using a lentiviral mouse model of HD we tested the effect of
these proteins in striatum. Both proteins decreased mHTT-induced striatal
neuronal atrophy. Findings provide evidence for a role of dysregulated
protein-thiol homeostasis in the pathogenesis of HD.
Collapse
Affiliation(s)
- Jonathan Fox
- Neuroscience Graduate Program, Department of Veterinary Sciences, University of Wyoming, Laramie, Wyoming, USA
| | - Zhen Lu
- Neuroscience Graduate Program, Department of Veterinary Sciences, University of Wyoming, Laramie, Wyoming, USA
| | | |
Collapse
|
23
|
Yoshimaru E, Totenhagen J, Alexander GE, Trouard TP. Design, manufacture, and analysis of customized phantoms for enhanced quality control in small animal MRI systems. Magn Reson Med 2015; 71:880-4. [PMID: 23440883 DOI: 10.1002/mrm.24678] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
PURPOSE Magnetic resonance imaging (MRI) is widely used in human brain research to evaluate the effects of healthy aging and development, as well as neurological disorders. Although standardized methods for quality assurance of human MRI instruments have been established, such approaches have typically not been translated to small animal imaging. We present a method for the generation and analysis of customized phantoms for small animal MRI systems that allows rapid and accurate system stability monitoring. METHODS Computer-aided design software was used to produce a customized phantom using a rapid prototyping printer. Automated registration algorithms were used on three-dimensional images of the phantom to allow system stability to be easily monitored over time. RESULTS The design of the custom phantom allowed reliable placement relative to the imaging coil. Automated registration showed superior ability to detect gradient changes reflected in the images than with manual measurements. Registering images acquired over time allowed monitoring of gradient drifts of less than one percent. CONCLUSION A low cost, MRI compatible phantom was successfully designed using computer-aided design software and a three-dimensional printer. Registering phantom images acquired over time allows monitoring of gradient stability of the MRI system.
Collapse
Affiliation(s)
- Eriko Yoshimaru
- Department of Biomedical Engineering, University of Arizona, Tucson, Arizona, USA
| | | | | | | |
Collapse
|
24
|
Swarnkar S, Chen Y, Pryor WM, Shahani N, Page DT, Subramaniam S. Ectopic expression of the striatal-enriched GTPase Rhes elicits cerebellar degeneration and an ataxia phenotype in Huntington's disease. Neurobiol Dis 2015; 82:66-77. [PMID: 26048156 DOI: 10.1016/j.nbd.2015.05.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 04/02/2015] [Accepted: 05/26/2015] [Indexed: 12/31/2022] Open
Abstract
Huntington's disease (HD) is caused by an expansion of glutamine repeats in the huntingtin protein (mHtt) that invokes early and prominent damage of the striatum, a region that controls motor behaviors. Despite its ubiquitous expression, why certain brain regions, such as the cerebellum, are relatively spared from neuronal loss by mHtt remains unclear. Previously, we implicated the striatal-enriched GTPase, Rhes (Ras homolog enriched in the striatum), which binds and SUMOylates mHtt and increases its solubility and cellular cytotoxicity, as the cause for striatal toxicity in HD. Here, we report that Rhes deletion in HD mice (N171-82Q), which express the N-terminal fragment of human Htt with 82 glutamines (Rhes(-/-)/N171-82Q), display markedly reduced HD-related behavioral deficits, and absence of lateral ventricle dilatation (secondary to striatal atrophy), compared to control HD mice (N171-82Q). To further validate the role of GTPase Rhes in HD, we tested whether ectopic Rhes expression would elicit a pathology in a brain region normally less affected in HD. Remarkably, ectopic expression of Rhes in the cerebellum of N171-82Q mice, during the asymptomatic period led to an exacerbation of motor deficits, including loss of balance and motor incoordination with ataxia-like features, not apparent in control-injected N171-82Q mice or Rhes injected wild-type mice. Pathological and biochemical analysis of Rhes-injected N171-82Q mice revealed a cerebellar lesion with marked loss of Purkinje neuron layer parvalbumin-immunoreactivity, induction of caspase 3 activation, and enhanced soluble forms of mHtt. Similarly reintroducing Rhes into the striatum of Rhes deleted Rhes(-/-)Hdh(150Q/150Q) knock-in mice, elicited a progressive HD-associated rotarod deficit. Overall, these studies establish that Rhes plays a pivotal role in vivo for the selective toxicity of mHtt in HD.
Collapse
Affiliation(s)
- Supriya Swarnkar
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - Youjun Chen
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - William M Pryor
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - Neelam Shahani
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - Damon T Page
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, USA
| | - Srinivasa Subramaniam
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida 33458, USA.
| |
Collapse
|
25
|
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
|
26
|
Reinstating aberrant mTORC1 activity in Huntington's disease mice improves disease phenotypes. Neuron 2014; 85:303-15. [PMID: 25556834 DOI: 10.1016/j.neuron.2014.12.019] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2014] [Indexed: 12/28/2022]
Abstract
Huntington's disease (HD) is caused by a polyglutamine tract expansion in huntingtin (HTT). Despite HTTs ubiquitous expression, there is early and robust vulnerability in striatum, the cause of which is poorly understood. Here, we provide evidence that impaired striatal mTORC1 activity underlies varied metabolic and degenerative phenotypes in HD brain and show that introducing the constitutively active form of the mTORC1 regulator, Rheb, into HD mouse brain, alleviates mitochondrial dysfunction, aberrant cholesterol homeostasis, striatal atrophy, impaired dopamine signaling, and increases autophagy. We also find that the expression of Rhes, a striatum-enriched mTOR activator, is reduced in HD patient and mouse brain and that exogenous addition of Rhes alleviates motor deficits and improves brain pathology in HD mice. Our combined work indicates that impaired Rhes/mTORC1 activity in HD brain may underlie the notable striatal susceptibility and thus presents a promising therapeutic target for HD therapy.
Collapse
|
27
|
Jiang M, Zheng J, Peng Q, Hou Z, Zhang J, Mori S, Ellis JL, Vlasuk GP, Fries H, Suri V, Duan W. Sirtuin 1 activator SRT2104 protects Huntington's disease mice. Ann Clin Transl Neurol 2014; 1:1047-52. [PMID: 25574479 PMCID: PMC4284130 DOI: 10.1002/acn3.135] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 09/06/2014] [Accepted: 10/07/2014] [Indexed: 02/01/2023] Open
Abstract
Sirtuin 1 is a nicotinamide adenine dinucleotide-dependent protein deacetylase which regulates longevity and improves metabolism. Activation of Sirtuin 1 confers beneficial effects in models of neurodegenerative diseases. We and others have provided convincing evidence that overexpression of Sirtuin 1 plays a neuroprotective role in mouse models of Huntington's disease. In this study, we report that SRT2104, a small molecule Sirtuin 1 activator, penetrated the blood–brain barrier, attenuated brain atrophy, improved motor function, and extended survival in a mouse model of Huntington's disease. These findings imply a novel therapeutic strategy for Huntington's disease by targeting Sirtuin 1.
Collapse
Affiliation(s)
- Mali Jiang
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, School of Medicine, Johns Hopkins University Baltimore, Maryland, 21287
| | - Jennifer Zheng
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, School of Medicine, Johns Hopkins University Baltimore, Maryland, 21287
| | - Qi Peng
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, School of Medicine, Johns Hopkins University Baltimore, Maryland, 21287
| | - Zhipeng Hou
- Department of Radiology, School of Medicine, Johns Hopkins University Baltimore, Maryland, 21287
| | - Jiangyang Zhang
- Department of Radiology, School of Medicine, Johns Hopkins University Baltimore, Maryland, 21287
| | - Susumu Mori
- Department of Radiology, School of Medicine, Johns Hopkins University Baltimore, Maryland, 21287
| | - James L Ellis
- Sirtris, a GSK company Collegeville, Pennsylvania, 19426
| | | | - Harvey Fries
- Sirtris, a GSK company Collegeville, Pennsylvania, 19426
| | - Vipin Suri
- Sirtris, a GSK company Collegeville, Pennsylvania, 19426
| | - Wenzhen Duan
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, School of Medicine, Johns Hopkins University Baltimore, Maryland, 21287 ; Department of Neuroscience, School of Medicine, Johns Hopkins University Baltimore, Maryland, 21287 ; Program in Cellular and Molecular Medicine, School of Medicine, Johns Hopkins University Baltimore, Maryland, 21287
| |
Collapse
|
28
|
Lee JH, Sowada MJ, Boudreau RL, Aerts AM, Thedens DR, Nopoulos P, Davidson BL. Rhes suppression enhances disease phenotypes in Huntington's disease mice. J Huntingtons Dis 2014; 3:65-71. [PMID: 25062765 DOI: 10.3233/jhd-140094] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
In Huntington's disease (HD) mutant HTT is ubiquitously expressed yet the striatum undergoes profound early degeneration. Cell culture studies suggest that a striatal-enriched protein, Rhes, may account for this vulnerability. We investigated the therapeutic potential of silencing Rhes in vivo using inhibitory RNAs (miRhes). While Rhes suppression was tolerated in wildtype mice, it failed to improve rotarod function in two distinct HD mouse models. Additionally, miRhes treated HD mice had increased anxiety-like behaviors and enhanced striatal atrophy as measured by longitudinal MRI when compared to control treated mice. These findings raise caution regarding the long-term implementation of inhibiting Rhes as a therapy for HD.
Collapse
Affiliation(s)
- John H Lee
- Medical Scientist Training Program, Roy J and Lucille A Carver College of Medicine, Iowa City, IA, USA Department of Molecular Physiology & Biophysics, Roy J and Lucille A Carver College of Medicine, Iowa City, IA, USA
| | - Matthew J Sowada
- Department of Internal Medicine, Roy J and Lucille A Carver College of Medicine, Iowa City, IA, USA
| | - Ryan L Boudreau
- Department of Internal Medicine, Roy J and Lucille A Carver College of Medicine, Iowa City, IA, USA
| | - Andrea M Aerts
- Department of Psychiatry, Roy J and Lucille A Carver College of Medicine, Iowa City, IA, USA
| | - Daniel R Thedens
- Department of Radiology, Roy J and Lucille A Carver College of Medicine, Iowa City, IA, USA
| | - Peg Nopoulos
- Department of Psychiatry, Roy J and Lucille A Carver College of Medicine, Iowa City, IA, USA
| | - Beverly L Davidson
- Department of Molecular Physiology & Biophysics, Roy J and Lucille A Carver College of Medicine, Iowa City, IA, USA Department of Internal Medicine, Roy J and Lucille A Carver College of Medicine, Iowa City, IA, USA Department of Neurology, Roy J and Lucille A Carver College of Medicine, Iowa City, IA, USA
| |
Collapse
|
29
|
Dada T, Rosenzweig JM, Shammary MA, Firdaus W, Rebh SA, Borbiev T, Tekes A, Zhang J, Alqahtani E, Mori S, Pletnikov MV, Johnston MV, Burd I. Mouse model of intrauterine inflammation: sex-specific differences in long-term neurologic and immune sequelae. Brain Behav Immun 2014; 38:142-50. [PMID: 24486323 PMCID: PMC3989501 DOI: 10.1016/j.bbi.2014.01.014] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 01/13/2014] [Accepted: 01/21/2014] [Indexed: 10/25/2022] Open
Abstract
Preterm infants, especially those that are exposed to prenatal intrauterine infection or inflammation, are at a major risk for adverse neurological outcomes, including cognitive, motor and behavioral disabilities. We have previously shown in a mouse model that there is an acute fetal brain insult associated with intrauterine inflammation. The objectives of this study were: (1) to elucidate long-term (into adolescence and adulthood) neurological outcomes by assessing neurobehavioral development, MRI, immunohistochemistry and flow cytometry of cells of immune origin and (2) to determine whether there are any sex-specific differences in brain development associated with intrauterine inflammation. Our results have shown that prenatal exposure appeared to lead to changes in MRI and behavior patterns throughout the neonatal period and during adulthood. Furthermore, we observed chronic brain inflammation in the offspring, with persistence of microglial activation and increased numbers of macrophages in the brain, ultimately resulting in neuronal loss. Moreover, our study highlights the sex-specific differences in long-term sequelae. This study, while extending the growing literature of adverse neurologic outcomes following exposure to inflammation during early development, presents novel findings in the context of intrauterine inflammation.
Collapse
Affiliation(s)
- Tahani Dada
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jason M. Rosenzweig
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Mofeedah Al Shammary
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Wance Firdaus
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Shorouq Al Rebh
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Talaibek Borbiev
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Aylin Tekes
- The Russell H. Morgan, Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jiangyang Zhang
- The Russell H. Morgan, Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Eman Alqahtani
- The Russell H. Morgan, Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Susumu Mori
- The Russell H. Morgan, Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Mikhail V. Pletnikov
- Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | - Irina Burd
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Neuroscience, Kennedy Krieger Institute, Baltimore, MD, United States; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
30
|
Intrajugular vein delivery of AAV9-RNAi prevents neuropathological changes and weight loss in Huntington's disease mice. Mol Ther 2014; 22:797-810. [PMID: 24390280 DOI: 10.1038/mt.2013.289] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 12/18/2013] [Indexed: 01/25/2023] Open
Abstract
Huntington's disease (HD) is a fatal neurological disorder caused by a CAG repeat expansion in the HTT gene, which encodes a mutant huntingtin protein (mHTT). The mutation confers a toxic gain of function on huntingtin, leading to widespread neurodegeneration and inclusion formation in many brain regions. Although the hallmark symptom of HD is hyperkinesia stemming from striatal degeneration, several other brain regions are affected which cause psychiatric, cognitive, and metabolic symptoms. Additionally, mHTT expression in peripheral tissue is associated with skeletal muscle atrophy, cardiac failure, weight loss, and diabetes. We, and others, have demonstrated a prevention of motor symptoms in HD mice following direct striatal injection of adeno-associated viral vector (AAV) serotype 1 encoding an RNA interference (RNAi) construct targeting mutant HTT mRNA (mHTT). Here, we expand these efforts and demonstrate that an intrajugular vein injection of AAV serotype 9 (AAV9) expressing a mutant HTT-specific RNAi construct significantly reduced mHTT expression in multiple brain regions and peripheral tissues affected in HD. Correspondingly, this approach prevented atrophy and inclusion formation in key brain regions as well as the severe weight loss germane to HD transgenic mice. These results demonstrate that systemic delivery of AAV9-RNAi may provide more widespread clinical benefit for patients suffering from HD.
Collapse
|
31
|
Lauterbach EC. Neuroprotective effects of psychotropic drugs in Huntington's disease. Int J Mol Sci 2013; 14:22558-603. [PMID: 24248060 PMCID: PMC3856079 DOI: 10.3390/ijms141122558] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 10/16/2013] [Accepted: 10/16/2013] [Indexed: 02/06/2023] Open
Abstract
Psychotropics (antipsychotics, mood stabilizers, antidepressants, anxiolytics, etc.) are commonly prescribed to treat Huntington’s disease (HD). In HD preclinical models, while no psychotropic has convincingly affected huntingtin gene, HD modifying gene, or huntingtin protein expression, psychotropic neuroprotective effects include upregulated huntingtin autophagy (lithium), histone acetylation (lithium, valproate, lamotrigine), miR-222 (lithium-plus-valproate), mitochondrial protection (haloperidol, trifluoperazine, imipramine, desipramine, nortriptyline, maprotiline, trazodone, sertraline, venlafaxine, melatonin), neurogenesis (lithium, valproate, fluoxetine, sertraline), and BDNF (lithium, valproate, sertraline) and downregulated AP-1 DNA binding (lithium), p53 (lithium), huntingtin aggregation (antipsychotics, lithium), and apoptosis (trifluoperazine, loxapine, lithium, desipramine, nortriptyline, maprotiline, cyproheptadine, melatonin). In HD live mouse models, delayed disease onset (nortriptyline, melatonin), striatal preservation (haloperidol, tetrabenazine, lithium, sertraline), memory preservation (imipramine, trazodone, fluoxetine, sertraline, venlafaxine), motor improvement (tetrabenazine, lithium, valproate, imipramine, nortriptyline, trazodone, sertraline, venlafaxine), and extended survival (lithium, valproate, sertraline, melatonin) have been documented. Upregulated CREB binding protein (CBP; valproate, dextromethorphan) and downregulated histone deacetylase (HDAC; valproate) await demonstration in HD models. Most preclinical findings await replication and their limitations are reviewed. The most promising findings involve replicated striatal neuroprotection and phenotypic disease modification in transgenic mice for tetrabenazine and for sertraline. Clinical data consist of an uncontrolled lithium case series (n = 3) suggesting non-progression and a primarily negative double-blind, placebo-controlled clinical trial of lamotrigine.
Collapse
Affiliation(s)
- Edward C Lauterbach
- Department of Psychiatry and Behavioral Sciences, Mercer University School of Medicine, 655 First Street, Macon, GA 31201, USA.
| |
Collapse
|
32
|
|
33
|
Li JY, Conforti L. Axonopathy in Huntington's disease. Exp Neurol 2013; 246:62-71. [DOI: 10.1016/j.expneurol.2012.08.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Revised: 06/27/2012] [Accepted: 08/11/2012] [Indexed: 02/02/2023]
|
34
|
Lin L, Wu S, Bin G, Yang C. Intensity Inhomogeneity Correction Using N3 on Mouse Brain Magnetic Resonance Microscopy. J Neuroimaging 2013; 23:502-7. [DOI: 10.1111/jon.12041] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2012] [Revised: 11/27/2012] [Accepted: 12/21/2012] [Indexed: 11/29/2022] Open
Affiliation(s)
- Lan Lin
- Biomedical Research Center, College of Life Science and Bioengineering; Beijing University of Technology; Beijing 100124 China
| | - Shuicai Wu
- Biomedical Research Center, College of Life Science and Bioengineering; Beijing University of Technology; Beijing 100124 China
| | - Guangyu Bin
- Biomedical Research Center, College of Life Science and Bioengineering; Beijing University of Technology; Beijing 100124 China
| | - Chunlan Yang
- Biomedical Research Center, College of Life Science and Bioengineering; Beijing University of Technology; Beijing 100124 China
| |
Collapse
|
35
|
Ullmann JFP, Watson C, Janke AL, Kurniawan ND, Paxinos G, Reutens DC. An MRI atlas of the mouse basal ganglia. Brain Struct Funct 2013; 219:1343-53. [PMID: 23689500 DOI: 10.1007/s00429-013-0572-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Accepted: 05/02/2013] [Indexed: 10/26/2022]
Abstract
The basal ganglia are a group of subpallial nuclei that play an important role in motor, emotional, and cognitive functions. Morphological changes and disrupted afferent/efferent connections in the basal ganglia have been associated with a variety of neurological disorders including psychiatric and movement disorders. While high-resolution magnetic resonance imaging has been used to characterize changes in brain structure in mouse models of these disorders, no systematic method for segmentation of the C57BL/6 J mouse basal ganglia exists. In this study we have used high-resolution MR images of ex vivo C57BL/6 J mouse brain to create a detailed protocol for segmenting the basal ganglia. We created a three-dimensional minimum deformation atlas, which includes the segmentation of 35 striatal, pallidal, and basal ganglia-related structures. In addition, we provide mean volumes, mean T2 contrast intensities and mean FA and ADC values for each structure. This MR atlas is available for download, and enables researchers to perform automated segmentation in genetic models of basal ganglia disorders.
Collapse
Affiliation(s)
- Jeremy F P Ullmann
- Center for Advanced Imaging, The University of Queensland, Brisbane, QLD, 4072, Australia,
| | | | | | | | | | | |
Collapse
|
36
|
Jiang M, Peng Q, Liu X, Jin J, Hou Z, Zhang J, Mori S, Ross CA, Ye K, Duan W. Small-molecule TrkB receptor agonists improve motor function and extend survival in a mouse model of Huntington's disease. Hum Mol Genet 2013; 22:2462-70. [PMID: 23446639 DOI: 10.1093/hmg/ddt098] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Huntington's disease (HD) is a fatal neurodegenerative disease characterized by abnormal motor coordination, cognitive decline and psychiatric disorders. This disease is caused by an expanded CAG trinucleotide repeat in the gene encoding the protein huntingtin. Reduced levels of brain-derived neurotrophic factor (BDNF) in the brain, which results from transcriptional inhibition and axonal transport deficits mediated by mutant huntingtin, have been suggested as critical factors underlying selective neurodegeneration in both HD patients and HD mouse models. BDNF activates its high-affinity receptor TrkB and promotes neuronal survival; restoring BDNF signaling is thus of particular therapeutic interest. In the present study, we evaluated the ability of a small-molecule TrkB agonist 7,8-dihydroxyflavone (7,8-DHF) and its synthetic derivative 4'-dimethylamino-7,8- dihydroxyflavone (4'-DMA-7,8-DHF) to protect neurons in the well-characterized N171-82Q HD mouse model. We found that chronic administration of 7, 8-DHF (5 mg/kg) or 4'-DMA-7,8-DHF (1 mg/kg) significantly improved motor deficits, ameliorated brain atrophy and extended survival in these N171-82Q HD mice. Moreover, 4'-DMA-7,8-DHF preserved DARPP32 levels in the striatum and rescued mutant huntingtin-induced impairment of neurogenesis in the N171-82Q HD mice. These data highlight consideration of TrkB as a therapeutic target in HD and suggest that small-molecule TrkB agonists that penetrate the brain have high potential to be further tested in clinical trials of HD.
Collapse
Affiliation(s)
- Mali Jiang
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Jin J, Cheng Y, Zhang Y, Wood W, Peng Q, Hutchison E, Mattson MP, Becker KG, Duan W. Interrogation of brain miRNA and mRNA expression profiles reveals a molecular regulatory network that is perturbed by mutant huntingtin. J Neurochem 2012; 123:477-90. [PMID: 22906125 PMCID: PMC3472040 DOI: 10.1111/j.1471-4159.2012.07925.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Revised: 08/13/2012] [Accepted: 08/14/2012] [Indexed: 11/27/2022]
Abstract
Emerging evidence indicates that microRNAs (miRNAs) may play an important role in the pathogenesis of Huntington's disease (HD). To identify the individual miRNAs that are altered in HD and may therefore regulate a gene network underlying mutant huntingtin-induced neuronal dysfunction in HD, we performed miRNA array analysis combined with mRNA profiling in the cerebral cortex from N171-82Q HD mice. Expression profiles of miRNAs as well as mRNAs in HD mouse cerebral cortex were analyzed and confirmed at different stages of disease progression; the most significant changes of miRNAs in the cerebral cortex were also detected in the striatum of HD mice. Our results revealed a significant alteration of miR-200 family members, miR-200a, and miR-200c in the cerebral cortex and the striatum, at the early stage of disease progression in N171-82Q HD mice. We used a coordinated approach to integrate miRNA and mRNA profiling, and applied bioinformatics to predict a target gene network potentially regulated by these significantly altered miRNAs that might be involved in HD disease progression. Interestingly, miR-200a and miR-200c are predicted to target genes regulating synaptic function, neurodevelopment, and neuronal survival. Our results suggest that altered expression of miR-200a and miR-200c may interrupt the production of proteins involved in neuronal plasticity and survival, and further investigation of the involvement of perturbed miRNA expression in HD pathogenesis is warranted, and may lead to reveal novel approaches for HD therapy.
Collapse
Affiliation(s)
- Jing Jin
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Yong Cheng
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Yongqing Zhang
- Gene Expression and Genomics Unit Research Resources Branch, National Institute on Aging Intramural Research Program, Baltimore, MD 21224
| | - William Wood
- Gene Expression and Genomics Unit Research Resources Branch, National Institute on Aging Intramural Research Program, Baltimore, MD 21224
| | - Qi Peng
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Emmette Hutchison
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD 21224
| | - Mark P. Mattson
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD 21224
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Kevin G. Becker
- Gene Expression and Genomics Unit Research Resources Branch, National Institute on Aging Intramural Research Program, Baltimore, MD 21224
| | - Wenzhen Duan
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| |
Collapse
|
38
|
Lin L, Wu S, Yang C. A template-based automatic skull-stripping approach for mouse brain MR microscopy. Microsc Res Tech 2012; 76:7-11. [PMID: 23034781 DOI: 10.1002/jemt.22128] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Accepted: 08/28/2012] [Indexed: 11/10/2022]
Abstract
Small animal magnetic resonance microscopy (MRM) has been widely used today in computational neuroanatomy. Accurate identification of brain tissue in a mouse MRM is a critical fundamental step in neuroimaging processing, which is given less attention. This study presents an automatic skull-stripping technique based on template models and histogram analysis. Results were evaluated by calculating the Jaccard similarity index (JSI) and boundary concordance ratio (BCR) between the automatically segmented and manually traced brain volumes. Results demonstrate that this technique accurately extracts the brain volume (mean JSI = 97.1%, BC = 94.4%). The brain extraction method presented in this study will greatly facilitate analysis of neuroimaging studies of rodent animals in neurodegenerative diseases.
Collapse
Affiliation(s)
- Lan Lin
- Biomedical Research Center, College of Life Science and Bioengineering, Beijing University of Technology, Beijing 100022, China.
| | | | | |
Collapse
|
39
|
Renoir T, Pang TYC, Zajac MS, Chan G, Du X, Leang L, Chevarin C, Lanfumey L, Hannan AJ. Treatment of depressive-like behaviour in Huntington's disease mice by chronic sertraline and exercise. Br J Pharmacol 2012; 165:1375-89. [PMID: 21718306 DOI: 10.1111/j.1476-5381.2011.01567.x] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND AND PURPOSE Depression is the most common psychiatric disorder in Huntington's disease (HD) patients. Women are more prone to develop depression and such susceptibility might be related to 5-hydroxytryptaminergic (serotonergic) dysregulation. EXPERIMENTAL APPROACH We performed tests of depression-related behaviours on female R6/1 HD mice that had been chronically treated with sertraline or provided with running-wheels. Functional assessments of 5-HT(1A) and 5-HT(2A) receptors were performed by measuring behavioural and physiological responses following administration of specific agonists, in combination with analysis of hippocampal gene expression. Finally we assessed the effect of exercise on hippocampal cell proliferation. KEY RESULTS Female HD mice recorded increased immobility time in the forced-swimming test, reduced saccharin preference and a hyperthermic response to stress compared with wild-type animals. These alterations were improved by chronic sertraline treatment. Wheel-running also resulted in similar improvements with the exception of saccharin preference but failed to correct the hippocampal cell proliferation deficits displayed by HD mice. The benefits of sertraline treatment and exercise involved altered 5-HT(1A) autoreceptor function, as demonstrated by modulation of the exaggerated 8-OH-DPAT-induced hypothermia exhibited by female HD mice. On the other hand, sertraline treatment was unable to restore the reduced 5-HT(1A) and 5-HT(2) heteroceptor function observed in HD animals. CONCLUSIONS AND IMPLICATIONS We report for the first time a crucial role for 5-HT(1A) autoreceptor function in mediating the sex-specific depressive-like phenotype of female R6/1 HD mice. Our data further support a differential effect of chronic sertraline treatment and exercise on hippocampal cell proliferation despite common behavioural benefits.
Collapse
Affiliation(s)
- Thibault Renoir
- Howard Florey Institute, Florey Neuroscience Institutes, University of Melbourne, Parkville, Victoria, Australia.
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
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
|
41
|
Switonski PM, Szlachcic WJ, Gabka A, Krzyzosiak WJ, Figiel M. Mouse models of polyglutamine diseases in therapeutic approaches: review and data table. Part II. Mol Neurobiol 2012; 46:430-66. [PMID: 22944909 PMCID: PMC3461214 DOI: 10.1007/s12035-012-8316-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2012] [Accepted: 07/29/2012] [Indexed: 12/13/2022]
Abstract
Mouse models of human diseases are created both to understand the pathogenesis of the disorders and to find successful therapies for them. This work is the second part in a series of reviews of mouse models of polyglutamine (polyQ) hereditary disorders and focuses on in vivo experimental therapeutic approaches. Like part I of the polyQ mouse model review, this work is supplemented with a table that contains data from experimental studies of therapeutic approaches in polyQ mouse models. The aim of this review was to characterize the benefits and outcomes of various therapeutic strategies in mouse models. We examine whether the therapeutic strategies are specific to a single disease or are applicable to more than one polyQ disorder in mouse models. In addition, we discuss the suitability of mouse models in therapeutic approaches. Although the majority of therapeutic studies were performed in mouse models of Huntington disease, similar strategies were also used in other disease models.
Collapse
Affiliation(s)
- Pawel M Switonski
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland
| | | | | | | | | |
Collapse
|
42
|
Badea A, Gewalt S, Avants BB, Cook JJ, Johnson GA. Quantitative mouse brain phenotyping based on single and multispectral MR protocols. Neuroimage 2012; 63:1633-45. [PMID: 22836174 DOI: 10.1016/j.neuroimage.2012.07.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Revised: 06/26/2012] [Accepted: 07/07/2012] [Indexed: 12/13/2022] Open
Abstract
Sophisticated image analysis methods have been developed for the human brain, but such tools still need to be adapted and optimized for quantitative small animal imaging. We propose a framework for quantitative anatomical phenotyping in mouse models of neurological and psychiatric conditions. The framework encompasses an atlas space, image acquisition protocols, and software tools to register images into this space. We show that a suite of segmentation tools (Avants, Epstein et al., 2008) designed for human neuroimaging can be incorporated into a pipeline for segmenting mouse brain images acquired with multispectral magnetic resonance imaging (MR) protocols. We present a flexible approach for segmenting such hyperimages, optimizing registration, and identifying optimal combinations of image channels for particular structures. Brain imaging with T1, T2* and T2 contrasts yielded accuracy in the range of 83% for hippocampus and caudate putamen (Hc and CPu), but only 54% in white matter tracts, and 44% for the ventricles. The addition of diffusion tensor parameter images improved accuracy for large gray matter structures (by >5%), white matter (10%), and ventricles (15%). The use of Markov random field segmentation further improved overall accuracy in the C57BL/6 strain by 6%; so Dice coefficients for Hc and CPu reached 93%, for white matter 79%, for ventricles 68%, and for substantia nigra 80%. We demonstrate the segmentation pipeline for the widely used C57BL/6 strain, and two test strains (BXD29, APP/TTA). This approach appears promising for characterizing temporal changes in mouse models of human neurological and psychiatric conditions, and may provide anatomical constraints for other preclinical imaging, e.g. fMRI and molecular imaging. This is the first demonstration that multiple MR imaging modalities combined with multivariate segmentation methods lead to significant improvements in anatomical segmentation in the mouse brain.
Collapse
Affiliation(s)
- Alexandra Badea
- Center for InVivo Microscopy, Box 3302, Duke University Medical Center, Durham, NC 27710, USA.
| | | | | | | | | |
Collapse
|
43
|
Aggarwal M, Duan W, Hou Z, Rakesh N, Peng Q, Ross CA, Miller MI, Mori S, Zhang J. Spatiotemporal mapping of brain atrophy in mouse models of Huntington's disease using longitudinal in vivo magnetic resonance imaging. Neuroimage 2012; 60:2086-95. [PMID: 22342677 DOI: 10.1016/j.neuroimage.2012.01.141] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Revised: 12/29/2011] [Accepted: 01/31/2012] [Indexed: 12/19/2022] Open
Abstract
Mouse models of Huntington's disease (HD) that recapitulate some of the phenotypic features of human HD, play a crucial role in investigating disease mechanisms and testing potential therapeutic approaches. Longitudinal studies of these models can yield valuable insights into the temporal course of disease progression and the effect of drug treatments on the progressive phenotypes. Atrophy of the brain, particularly the striatum, is a characteristic phenotype of human HD, is known to begin long before the onset of motor symptoms, and correlates strongly with clinical features. Elucidating the spatial and temporal patterns of atrophy in HD mouse models is important to characterize the phenotypes of these models, as well as evaluate the effects of neuroprotective treatments at specific time frames during disease progression. In this study, three dimensional in vivo magnetic resonance imaging (MRI) and automated longitudinal deformation-based morphological analysis was used to elucidate the spatial and temporal patterns of brain atrophy in the R6/2 and N171-82Q mouse models of HD. Using an established MRI-based brain atlas and mixed-effects modeling of deformation-based metrics, we report the rates of progression and region-specificity of brain atrophy in the two models. Further, the longitudinal analysis approach was used to evaluate the effects of sertraline and coenzyme Q(10) (CoQ(10)) treatments on progressive atrophy in the N171-82Q model. Sertraline treatment resulted in significant slowing of atrophy, especially in the striatum and frontal cortex regions, while no significant effects of CoQ(10) treatment were observed. Progressive cortical and striatal atrophy in the N171-82Q mice showed significant positive correlations with measured functional deficits. The findings of this report can be used for future testing and comparison of potential therapeutics in mouse models of HD.
Collapse
Affiliation(s)
- Manisha Aggarwal
- Division of NMR Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Xiang Z, Valenza M, Cui L, Leoni V, Jeong HK, Brilli E, Zhang J, Peng Q, Duan W, Reeves SA, Cattaneo E, Krainc D. Peroxisome-proliferator-activated receptor gamma coactivator 1 α contributes to dysmyelination in experimental models of Huntington's disease. J Neurosci 2011; 31:9544-53. [PMID: 21715619 PMCID: PMC3132810 DOI: 10.1523/jneurosci.1291-11.2011] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Revised: 05/01/2011] [Accepted: 05/09/2011] [Indexed: 11/21/2022] Open
Abstract
The peroxisome-proliferator-activated receptor gamma coactivator 1 α (PGC1α) has been implicated in the pathogenesis of several neurodegenerative disorders, including Huntington's disease (HD). Recent data demonstrating white matter abnormalities in PGC1α knock-out (KO) mice prompted us to examine the role of PGC1α in CNS myelination and its relevance to HD pathogenesis. We found deficient postnatal myelination in the striatum of PGC1α KO mice, accompanied by a decrease in myelin basic protein (MBP). In addition, brain cholesterol, its precursors, and the rate-limiting enzymes for cholesterol synthesis, HMG CoA synthase (HMGCS1) and HMG CoA reductase (HMGCR), were also reduced in PGC1α KO mice. Moreover, knockdown of PGC1α in oligodendrocytes by lentiviral shRNA led to a decrease in MBP, HMGCS1, and Hmgcr mRNAs. Chromatin immunoprecipitations revealed the recruitment of PGC1α to MBP promoter in mouse brain, and PGC1α over-expression increased MBP and SREBP-2 promoter activity, suggesting that PGC1α regulates MBP and cholesterol synthesis at the transcriptional level. Importantly, expression of mutant huntingtin (Htt) in primary oligodendrocytes resulted in decreased expression of PGC1α and its targets HmgcS1, Hmgcr, and MBP. Decreased expression of MBP and deficient myelination were found postnatally and in adult R6/2 mouse model of HD. Diffusion tensor imaging detected white matter abnormalities in the corpus callosum of R6/2 mice, and electron microscopy revealed thinner myelin sheaths and increased myelin periodicity in BACHD [bacterial artificial chromosome (BAC)-mediated transgenic model for Huntington's disease] mice expressing full-length mutant Htt. Together, these data suggest that PGC1α plays a role in postnatal myelination and that deficient PGC1α activity in oligodendrocytes may contribute to abnormal myelination in HD.
Collapse
Affiliation(s)
- Zhongmin Xiang
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129
| | - Marta Valenza
- Department of Pharmacological Sciences and Centre for Stem Cell Research, University of Milan, 20133 Milan, Italy
| | - Libin Cui
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129
| | - Valerio Leoni
- Laboratory of Clinical Pathology and Medical Genetics, Institute of Neurology IRCCS “C. Besta”, 20133 Milan, Italy and
| | - Hyun-Kyung Jeong
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129
| | - Elisa Brilli
- Department of Pharmacological Sciences and Centre for Stem Cell Research, University of Milan, 20133 Milan, Italy
| | | | - Qi Peng
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, Maryland 21287
| | - Wenzhen Duan
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, Maryland 21287
| | - Steven A. Reeves
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129
| | - Elena Cattaneo
- Department of Pharmacological Sciences and Centre for Stem Cell Research, University of Milan, 20133 Milan, Italy
| | - Dimitri Krainc
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129
| |
Collapse
|