1
|
Lin Y, Li C, Chen Y, Gao J, Li J, Huang C, Liu Z, Wang W, Zheng X, Song X, Wu J, Wu J, Luo OJ, Tu Z, Li S, Li XJ, Lai L, Yan S. RNA-Targeting CRISPR/CasRx system relieves disease symptoms in Huntington's disease models. Mol Neurodegener 2025; 20:4. [PMID: 39806441 PMCID: PMC11727607 DOI: 10.1186/s13024-024-00794-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 12/22/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND HD is a devastating neurodegenerative disorder caused by the expansion of CAG repeats in the HTT. Silencing the expression of mutated proteins is a therapeutic direction to rescue HD patients, and recent advances in gene editing technology such as CRISPR/CasRx have opened up new avenues for therapeutic intervention. METHODS The CRISPR/CasRx system was employed to target human HTT exon 1, resulting in an efficient knockdown of HTT mRNA. This therapeutic effect was substantiated in various models: HEK 293 T cell, the HD 140Q-KI mouse, and the HD-KI pig model. The efficiency of the knockdown was analyzed through Western blot and RT-qPCR. Additionally, neuropathological changes were examined using Western blot, immunostaining, and RNA sequencing. The impact on motor abilities was assessed via behavioral experiments, providing a comprehensive evaluation of the treatment's effectiveness. RESULTS CRISPR/CasRx system can significantly reduce HTT mRNA levels across various models, including HEK 293 T cells, HD 140Q-KI mice at various disease stages, and HD-KI pigs, and resulted in decreased expression of mHTT. Utilizing the CRISPR/CasRx system to knock down HTT RNA has shown to ameliorate gliosis in HD 140Q-KI mice and delay neurodegeneration in HD pigs. CONCLUSIONS These findings highlight the effectiveness of the RNA-targeting CRISPR/CasRx as a potential therapeutic strategy for HD. Furthermore, the success of this approach provides valuable insights and novel avenues for the treatment of other genetic disorders caused by gene mutations.
Collapse
Affiliation(s)
- Yingqi Lin
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), School of Medicine, GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Caijuan Li
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), School of Medicine, GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Yizhi Chen
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), School of Medicine, GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Jiale Gao
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), School of Medicine, GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Jiawei Li
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), School of Medicine, GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Chunhui Huang
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), School of Medicine, GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Zhaoming Liu
- China-New Zealand Joint Laboratory On Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou, Institutes of Biomedicine and Health , Chinese Academy of Sciences, Guangzhou, China
| | - Wei Wang
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), School of Medicine, GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Xiao Zheng
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), School of Medicine, GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Xichen Song
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), School of Medicine, GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Jianhao Wu
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), School of Medicine, GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Jiaxi Wu
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), School of Medicine, GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Oscar Junhong Luo
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Zhuchi Tu
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), School of Medicine, GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 510632, China
| | - Shihua Li
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), School of Medicine, GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 510632, China
| | - Xiao-Jiang Li
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), School of Medicine, GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 510632, China
| | - Liangxue Lai
- China-New Zealand Joint Laboratory On Biomedicine and Health, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou, Institutes of Biomedicine and Health , Chinese Academy of Sciences, Guangzhou, China
| | - Sen Yan
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), School of Medicine, GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China.
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 510632, China.
- Department of Neurology, Faculty of Medical Science, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China.
| |
Collapse
|
2
|
Huang HW, Wu S, Chowdhury EA, Shah DK. Expansion of platform physiologically-based pharmacokinetic model for monoclonal antibodies towards different preclinical species: cats, sheep, and dogs. J Pharmacokinet Pharmacodyn 2024; 51:621-638. [PMID: 37947924 DOI: 10.1007/s10928-023-09893-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 10/16/2023] [Indexed: 11/12/2023]
Abstract
Monoclonal antibodies (mAbs) are becoming an important therapeutic option in veterinary medicine, and understanding the pharmacokinetic (PK) of mAbs in higher-order animal species is also important for human drug development. To better understand the PK of mAbs in these animals, here we have expanded a platform physiological-based pharmacokinetic (PBPK) model to characterize the disposition of mAbs in three different preclinical species: cats, sheep, and dogs. We obtained PK data for mAbs and physiological parameters for the three different species from the literature. We were able to describe the PK of mAbs following intravenous (IV) or subcutaneous administration in cats, IV administration in sheep, and IV administration dogs reasonably well by fixing the physiological parameters and just estimating the parameters related to the binding of mAbs to the neonatal Fc receptor. The platform PBPK model presented here provides a quantitative tool to predict the plasma PK of mAbs in dogs, cats, and sheep. The model can also predict mAb PK in different tissues where the site of action might be located. As such, the mAb PBPK model presented here can facilitate the discovery, development, and preclinical-to-clinical translation of mAbs for veterinary and human medicine. The model can also be modified in the future to account for more detailed compartments for certain organs, different pathophysiology in the animals, and target-mediated drug disposition.
Collapse
Affiliation(s)
- Hsien-Wei Huang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Pharmacy Building, Buffalo, NY, 14214-8033, USA
| | - Shengjia Wu
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Pharmacy Building, Buffalo, NY, 14214-8033, USA
| | - Ekram A Chowdhury
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Pharmacy Building, Buffalo, NY, 14214-8033, USA
| | - Dhaval K Shah
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Pharmacy Building, Buffalo, NY, 14214-8033, USA.
| |
Collapse
|
3
|
Williams GK, Akkermans J, Lawson M, Syta P, Staelens S, Adhikari MH, Morton AJ, Nitzsche B, Boltze J, Christou C, Bertoglio D, Ahamed M. Imaging Glucose Metabolism and Dopaminergic Dysfunction in Sheep ( Ovis aries) Brain Using Positron Emission Tomography Imaging Reveals Abnormalities in OVT73 Huntington's Disease Sheep. ACS Chem Neurosci 2024; 15:4082-4091. [PMID: 39420554 DOI: 10.1021/acschemneuro.4c00561] [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] [Indexed: 10/19/2024] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disease that causes cognitive, movement, behavioral, and sleep disturbances, which over time result in progressive disability and eventually death. Clinical translation of novel therapeutics and imaging probes could be accelerated by additional testing in well-characterized large animal models of HD. The major goal of our preliminary cross-sectional study is to demonstrate the feasibility and utility of the unique transgenic sheep model of HD (OVT73) in positron emission tomography (PET) imaging. PET imaging studies were performed in healthy merino sheep (6 year old, n = 3) and OVT73 HD sheep (5.5 year old, n = 3, and 11 year old, n = 3). Region-of-interest and brain atlas labels were defined for regional analyses by using a sheep brain template. [18F]fluorodeoxyglucose ([18F]FDG) was employed to compare the regional brain glucose metabolism and variations in FDG uptake between control and HD sheep. We also used [18F]fluoro-3,4-dihydroxyphenylalanine ([18F]FDOPA) to compare the extent of striatal dysfunction and evaluated the binding potential (BPND) in key brain regions between the groups. Compared with healthy controls and 11 year old HD sheep, the 5.5 year old HD sheep exhibited significantly increased [18F]FDG uptake in several cortical and subcortical brain regions (P < 0.05-0.01). No difference in [18F]FDG uptake was observed between healthy controls and 11 year old HD sheep. Analysis of the [18F]FDOPA BPND parametric maps revealed clusters of reduced binding potential in the 5.5 year old and 11 year old HD sheep compared to the 6 year old control sheep. In this first-of-its-kind study, we showed the usefulness and validity of HD sheep model in imaging cerebral glucose metabolism and dopamine uptake using PET imaging. The identification of discrete patterns of metabolic abnormality using [18F]FDG and decline of [18F]FDOPA uptake may provide a useful means of quantifying early HD-related changes in these models, particularly in the transition from presymptomatic to early symptomatic phases of HD.
Collapse
Affiliation(s)
- Georgia K Williams
- Preclinical, Imaging, and Research Laboratories (PIRL), South Australian Health and Medical Research Institute (SAHMRI), Gilles Plains, Adelaide 5086, Australia
- National Imaging Facility, SAHMRI, Adelaide 5000, Australia
| | - Jordy Akkermans
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Antwerp 2160, Belgium
- μNeuro Center for Excellence, University of Antwerp, Antwerp 2160, Belgium
| | - Matt Lawson
- Molecular Imaging and Therapy Research Unit, South Australian Health and Medical Research Institute (SAHMRI), Adelaide 5000, South Australia
| | - Patryk Syta
- Molecular Imaging and Therapy Research Unit, South Australian Health and Medical Research Institute (SAHMRI), Adelaide 5000, South Australia
| | - Steven Staelens
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Antwerp 2160, Belgium
- μNeuro Center for Excellence, University of Antwerp, Antwerp 2160, Belgium
| | - Mohit H Adhikari
- μNeuro Center for Excellence, University of Antwerp, Antwerp 2160, Belgium
- Bio-Imaging Lab, University of Antwerp, Antwerp 2160, Belgium
| | - A Jennifer Morton
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, U.K
| | - Björn Nitzsche
- Department of Nuclear Medicine, University Hospital Leipzig, Stephanstr. 11, Leipzig 04103, Germany
- Faculty of Veterinary Medicine, Institute of Anatomy, Histology and Embryology, University of Leipzig, An den Tierkliniken 43, Leipzig 04103, Germany
| | - Johannes Boltze
- School of Life Sciences, University of Warwick, Coventry CV4 7AL, U.K
| | - Chris Christou
- Preclinical, Imaging, and Research Laboratories (PIRL), South Australian Health and Medical Research Institute (SAHMRI), Gilles Plains, Adelaide 5086, Australia
- National Imaging Facility, SAHMRI, Adelaide 5000, Australia
| | - Daniele Bertoglio
- μNeuro Center for Excellence, University of Antwerp, Antwerp 2160, Belgium
- Bio-Imaging Lab, University of Antwerp, Antwerp 2160, Belgium
| | - Muneer Ahamed
- Preclinical, Imaging, and Research Laboratories (PIRL), South Australian Health and Medical Research Institute (SAHMRI), Gilles Plains, Adelaide 5086, Australia
- National Imaging Facility, SAHMRI, Adelaide 5000, Australia
- Molecular Imaging and Therapy Research Unit, South Australian Health and Medical Research Institute (SAHMRI), Adelaide 5000, South Australia
- Lambert Initiative for Cannabinoid Therapeutics, Brain and Mind Centre, The University of Sydney, Sydney, New South Wales 2050, Australia
| |
Collapse
|
4
|
Li X, Tong H, Xu S, Zhou G, Yang T, Yin S, Yang S, Li X, Li S. Neuroinflammatory Proteins in Huntington's Disease: Insights into Mechanisms, Diagnosis, and Therapeutic Implications. Int J Mol Sci 2024; 25:11787. [PMID: 39519337 PMCID: PMC11546928 DOI: 10.3390/ijms252111787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Huntington's disease (HD) is a hereditary neurodegenerative disorder caused by a CAG tract expansion in the huntingtin gene (HTT). HD is characterized by involuntary movements, cognitive decline, and behavioral changes. Pathologically, patients with HD show selective striatal neuronal vulnerability at the early disease stage, although the mutant protein is ubiquitously expressed. Activation of the immune system and glial cell-mediated neuroinflammatory responses are early pathological features and have been found in all neurodegenerative diseases (NDDs), including HD. However, the role of inflammation in HD, as well as its therapeutic significance, has been less extensively studied compared to other NDDs. This review highlights the significantly elevated levels of inflammatory proteins and cellular markers observed in various HD animal models and HD patient tissues, emphasizing the critical roles of microglia, astrocytes, and oligodendrocytes in mediating neuroinflammation in HD. Moreover, it expands on recent discoveries related to the peripheral immune system's involvement in HD. Although current immunomodulatory treatments and inflammatory biomarkers for adjunctive diagnosis in HD are limited, targeting inflammation in combination with other therapies, along with comprehensive personalized treatment approaches, shows promising therapeutic potential.
Collapse
Affiliation(s)
- Xinhui Li
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| | - Huichun Tong
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Shuying Xu
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| | - Gongke Zhou
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| | - Tianqi Yang
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| | - Shurui Yin
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| | - Sitong Yang
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| | - Xiaojiang Li
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| | - Shihua Li
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| |
Collapse
|
5
|
Chang CP, Wu CW, Chern Y. Metabolic dysregulation in Huntington's disease: Neuronal and glial perspectives. Neurobiol Dis 2024; 201:106672. [PMID: 39306013 DOI: 10.1016/j.nbd.2024.106672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 09/15/2024] [Accepted: 09/16/2024] [Indexed: 09/29/2024] Open
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder caused by a mutant huntingtin protein with an abnormal CAG/polyQ expansion in the N-terminus of HTT exon 1. HD is characterized by progressive neurodegeneration and metabolic abnormalities, particularly in the brain, which accounts for approximately 20 % of the body's resting metabolic rate. Dysregulation of energy homeostasis in HD includes impaired glucose transporters, abnormal functions of glycolytic enzymes, changes in tricarboxylic acid (TCA) cycle activity and enzyme expression in the basal ganglia and cortical regions of both HD mouse models and HD patients. However, current understanding of brain cell behavior during energy dysregulation and its impact on neuron-glia crosstalk in HD remains limited. This review provides a comprehensive summary of the current understanding of the differences in glucose metabolism between neurons and glial cells in HD and how these differences contribute to disease development compared with normal conditions. We also discuss the potential impact of metabolic shifts on neuron-glia communication in HD. A deeper understanding of these metabolic alterations may reveal potential therapeutic targets for future drug development.
Collapse
Affiliation(s)
- Ching-Pang Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan; Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan; Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ching-Wen Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan; Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan
| | - Yijuang Chern
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan; Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
6
|
Rana N, Kapil L, Singh C, Singh A. Modeling Huntington's disease: An insight on in-vitro and in-vivo models. Behav Brain Res 2024; 459:114757. [PMID: 37952684 DOI: 10.1016/j.bbr.2023.114757] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/09/2023] [Accepted: 11/09/2023] [Indexed: 11/14/2023]
Abstract
Huntington's disease is a neurodegenerative illness that causes neuronal death most extensively within the basal ganglia. There is a broad class of neurologic disorders associated with the expansion of polyglutamine (polyQ) repeats in numerous proteins. Several other molecular mechanisms have also been implicated in HD pathology, including brain-derived neurotrophic factor (BDNF), mitochondrial dysfunction, and altered synaptic plasticity in central spiny neurons. HD pathogenesis and the effectiveness of therapy approaches have been better understood through the use of animal models. The pathological manifestations of the disease were reproduced by early models of glutamate analog toxicity and mitochondrial respiration inhibition. Because the treatments available for HD are quite limited, it is important to have a definite preclinical model that mimics all the aspects of the disease. It can be used to study mechanisms and validate candidate therapies. Although there hasn't been much success in translating animal research into clinical practice, each model has something special to offer in the quest for a deeper comprehension of HD's neurobehavioral foundations. This review provides insight into various in-vitro-and in-vivo models of HD which may be useful in the screening of newer therapeutics for this incapacitating disorder.
Collapse
Affiliation(s)
- Nitasha Rana
- Department of Pharmacology, ISF College of Pharmacy, Moga 142001, Affiliated to I.K Gujral Punjab Technical University, Jalandhar, Punjab, India
| | - Lakshay Kapil
- Department of Pharmacology, ISF College of Pharmacy, Moga 142001, Affiliated to I.K Gujral Punjab Technical University, Jalandhar, Punjab, India
| | - Charan Singh
- Department of Pharmaceutical Sciences, HNB Garhwal University (A Central University), Chauras Campus, Distt. Tehri Garhwal, Uttarakhand 246174, India
| | - Arti Singh
- Department of Pharmacology, ISF College of Pharmacy, Moga 142001, Affiliated to I.K Gujral Punjab Technical University, Jalandhar, Punjab, India.
| |
Collapse
|
7
|
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
|
8
|
Sapp E, Boudi A, Reid SJ, Trombetta BA, Kivisäkk P, Taghian T, Arnold SE, Howland D, Gray-Edwards H, Kegel-Gleason KB, DiFiglia M. Levels of Synaptic Proteins in Brain and Neurofilament Light Chain in Cerebrospinal Fluid and Plasma of OVT73 Huntington's Disease Sheep Support a Prodromal Disease State. J Huntingtons Dis 2023; 12:201-213. [PMID: 37661892 DOI: 10.3233/jhd-230590] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
BACKGROUND Synaptic changes occur early in patients with Huntington's disease (HD) and in mouse models of HD. An analysis of synaptic changes in HD transgenic sheep (OVT73) is fitting since they have been shown to have some phenotypes. They also have larger brains, longer lifespan, and greater motor and cognitive capacities more aligned with humans, and can provide abundant biofluids for in vivo monitoring of therapeutic interventions. OBJECTIVE The objective of this study was to determine if there were differences between 5- and 10-year-old OVT73 and wild-type (WT) sheep in levels of synaptic proteins in brain and in neurofilament light chain (NfL) in cerebrospinal fluid (CSF) and plasma. METHODS Mutant huntingtin (mHTT) and other proteins were measured by western blot assay in synaptosomes prepared from caudate, motor, and piriform cortex in 5-year-old and caudate, putamen, motor; and piriform cortex in 10-year-old WT and OVT73 sheep. Levels of NfL, a biomarker for neuronal damage increased in many neurological disorders including HD, were examined in CSF and plasma samples from 10-year-old WT and OVT73 sheep using the Simoa NfL Advantage kit. RESULTS Western blot analysis showed mHTT protein expression in synaptosomes from OVT73 sheep was 23% of endogenous sheep HTT levels at both ages. Significant changes were detected in brain levels of PDE10A, SCN4B, DARPP32, calmodulin, SNAP25, PSD95, VGLUT 1, VAMP1, and Na+/K+-ATPase, which depended on age and brain region. There was no difference in NfL levels in CSF and plasma in OVT73 sheep compared to age-matched WT sheep. CONCLUSIONS These results show that synaptic changes occur in brain of 5- and 10-year-old OVT73 sheep, but levels of NfL in biofluids are unaffected. Altogether, the data support a prodromal disease state in OVT73 sheep that involves the caudate, putamen and cortex.
Collapse
Affiliation(s)
- Ellen Sapp
- Department of Neurology, Mass General Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Adel Boudi
- Department of Neurology, Mass General Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Suzanne J Reid
- Centre for Brain Research, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Bianca A Trombetta
- Department of Neurology, Alzheimer's Clinical and Translational Research Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Pia Kivisäkk
- Department of Neurology, Alzheimer's Clinical and Translational Research Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Toloo Taghian
- Department of Radiology and Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Steven E Arnold
- Department of Neurology, Alzheimer's Clinical and Translational Research Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Heather Gray-Edwards
- Department of Radiology and Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Kimberly B Kegel-Gleason
- Department of Neurology, Mass General Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Marian DiFiglia
- Department of Neurology, Mass General Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
9
|
Weiss AR, Bertoglio D, Liguore WA, Brandon K, Templon J, Link J, McBride JL. Reduced D 2 /D 3 Receptor Binding and Glucose Metabolism in a Macaque Model of Huntington's Disease. Mov Disord 2023; 38:143-147. [PMID: 36544385 PMCID: PMC9948637 DOI: 10.1002/mds.29271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 10/04/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Dopamine system dysfunction and altered glucose metabolism are implicated in Huntington's disease (HD), a neurological disease caused by mutant huntingtin (mHTT) expression. OBJECTIVE The aim was to characterize alterations in cerebral dopamine D2 /D3 receptor density and glucose utilization in a newly developed AAV-mediated NHP model of HD that expresses mHTT throughout numerous brain regions. METHODS Positron emission tomography (PET) imaging was performed using [18 F]fallypride to quantify D2 /D3 receptor density and 2-[18 F]fluoro-2-deoxy-d-glucose ([18 F]FDG) to measure cerebral glucose utilization in these HD macaques. RESULTS Compared to controls, HD macaques showed significantly reduced dopamine D2 /D3 receptor densities in basal ganglia (P < 0.05). In addition, HD macaques displayed significant glucose hypometabolism throughout the cortico-basal ganglia network (P < 0.05). CONCLUSIONS [18 F]Fallypride and [18 F]FDG are PET imaging biomarkers of mHTT-mediated disease progression that can be used as noninvasive outcome measures in future therapeutic studies with this AAV-mediated HD macaque model. © 2022 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Alison R. Weiss
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR
| | - Daniele Bertoglio
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Belgium
| | - William A. Liguore
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR
| | - Kristin Brandon
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR
| | - John Templon
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, OR
| | - Jeanne Link
- Center for Radiochemistry Research, Oregon Health and Science University, Portland, OR
| | - Jodi L. McBride
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR
- Dept. of Behavioral Neuroscience, Oregon National Primate Research Center, Beaverton, OR
| |
Collapse
|
10
|
Taghian T, Gallagher J, Batcho E, Pullan C, Kuchel T, Denney T, Perumal R, Moore S, Muirhead R, Herde P, Johns D, Christou C, Taylor A, Passler T, Pulaparthi S, Hall E, Chandra S, O’Neill CA, Gray-Edwards H. Brain Alterations in Aged OVT73 Sheep Model of Huntington's Disease: An MRI Based Approach. J Huntingtons Dis 2022; 11:391-406. [PMID: 36189602 PMCID: PMC9837686 DOI: 10.3233/jhd-220526] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Huntington's disease (HD) is a fatal neurodegenerative autosomal dominant disorder with prevalence of 1 : 20000 that has no effective treatment to date. Translatability of candidate therapeutics could be enhanced by additional testing in large animal models because of similarities in brain anatomy, size, and immunophysiology. These features enable realistic pre-clinical studies of biodistribution, efficacy, and toxicity. OBJECTIVE AND METHODS Here we non-invasively characterized alterations in brain white matter microstructure, neurochemistry, neurological status, and mutant Huntingtin protein (mHTT) levels in cerebrospinal fluid (CSF) of aged OVT73 HD sheep. RESULTS Similar to HD patients, CSF mHTT differentiates HD from normal sheep. Our results are indicative of a decline in neurological status, and alterations in brain white matter diffusion and spectroscopy metric that are more severe in aged female HD sheep. Longitudinal analysis of aged female HD sheep suggests that the decline is detectable over the course of a year. In line with reports of HD human studies, white matter alterations in corpus callosum correlates with a decline in gait of HD sheep. Moreover, alterations in the occipital cortex white matter correlates with a decline in clinical rating score. In addition, the marker of energy metabolism in striatum of aged HD sheep, shows a correlation with decline of clinical rating score and eye coordination. CONCLUSION This data suggests that OVT73 HD sheep can serve as a pre-manifest large animal model of HD providing a platform for pre-clinical testing of HD therapeutics and non-invasive tracking of the efficacy of the therapy.
Collapse
Affiliation(s)
- Toloo Taghian
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA,
Department of Radiology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Jillian Gallagher
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
| | - Erin Batcho
- Department of Electrical and Computer Engineering, Auburn University, Auburn, AL, USA
| | - Caitlin Pullan
- South Australian Health and Medical Research Institute, Gillies Plains, SA, Australia
| | - Tim Kuchel
- South Australian Health and Medical Research Institute, Gillies Plains, SA, Australia
| | - Thomas Denney
- Department of Electrical and Computer Engineering, Auburn University, Auburn, AL, USA
| | - Raj Perumal
- South Australian Health and Medical Research Institute, Gillies Plains, SA, Australia
| | - Shamika Moore
- South Australian Health and Medical Research Institute, Gillies Plains, SA, Australia
| | - Robb Muirhead
- South Australian Health and Medical Research Institute, Gillies Plains, SA, Australia
| | - Paul Herde
- South Australian Health and Medical Research Institute, Gillies Plains, SA, Australia
| | - Daniel Johns
- South Australian Health and Medical Research Institute, Gillies Plains, SA, Australia
| | - Chris Christou
- South Australian Health and Medical Research Institute, Gillies Plains, SA, Australia
| | - Amanda Taylor
- Department of Clinical Sciences, Auburn University, Auburn, AL, USA
| | - Thomas Passler
- Department of Electrical and Computer Engineering, Auburn University, Auburn, AL, USA
| | - Sanjana Pulaparthi
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
| | - Erin Hall
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
| | - Sundeep Chandra
- Sana Biotechnology, South San Francisco, CA, USA,Bio Marin Pharmaceutical Inc., San Rafael, CA, USA
| | | | - Heather Gray-Edwards
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA,
Department of Radiology, University of Massachusetts Medical School, Worcester, MA, USA,Correspondence to: Heather L. Gray-Edwards, DVM, PhD, University of Massachusetts Medical School, Department of Radiology and Horae Gene Therapy Center, 368 Plantation Street, ASC6-2055, Worcester, MA 01605, USA. Tel.: +1 508 856 4051; Fax: +1 508 856 1552; E-mail:
| |
Collapse
|
11
|
Weiss AR, Liguore WA, Brandon K, Wang X, Liu Z, Domire JS, Button D, Srinivasan S, Kroenke CD, McBride JL. A novel rhesus macaque model of Huntington's disease recapitulates key neuropathological changes along with motor and cognitive decline. eLife 2022; 11:e77568. [PMID: 36205397 PMCID: PMC9545527 DOI: 10.7554/elife.77568] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 09/06/2022] [Indexed: 11/25/2022] Open
Abstract
We created a new nonhuman primate model of the genetic neurodegenerative disorder Huntington's disease (HD) by injecting a mixture of recombinant adeno-associated viral vectors, serotypes AAV2 and AAV2.retro, each expressing a fragment of human mutant HTT (mHTT) into the caudate and putamen of adult rhesus macaques. This modeling strategy results in expression of mutant huntingtin protein (mHTT) and aggregate formation in the injected brain regions, as well as dozens of other cortical and subcortical brain regions affected in human HD patients. We queried the disruption of cortico-basal ganglia circuitry for 30 months post-surgery using a variety of behavioral and imaging readouts. Compared to controls, mHTT-treated macaques developed working memory decline and progressive motor impairment. Multimodal imaging revealed circuit-wide white and gray matter degenerative processes in several key brain regions affected in HD. Taken together, we have developed a novel macaque model of HD that may be used to develop disease biomarkers and screen promising therapeutics.
Collapse
Affiliation(s)
- Alison R Weiss
- Division of Neuroscience, Oregon National Primate Research CenterBeavertonUnited States
| | - William A Liguore
- Division of Neuroscience, Oregon National Primate Research CenterBeavertonUnited States
| | - Kristin Brandon
- Division of Neuroscience, Oregon National Primate Research CenterBeavertonUnited States
| | - Xiaojie Wang
- Division of Neuroscience, Oregon National Primate Research CenterBeavertonUnited States
- Advanced Imaging Research Center, Oregon Health and Science UniversityPortlandUnited States
| | - Zheng Liu
- Division of Neuroscience, Oregon National Primate Research CenterBeavertonUnited States
- Advanced Imaging Research Center, Oregon Health and Science UniversityPortlandUnited States
| | - Jacqueline S Domire
- Division of Neuroscience, Oregon National Primate Research CenterBeavertonUnited States
| | - Dana Button
- Division of Neuroscience, Oregon National Primate Research CenterBeavertonUnited States
| | - Sathya Srinivasan
- Imaging and Morphology Support Core, Oregon National Primate Research CenterBeavertonUnited States
| | - Christopher D Kroenke
- Division of Neuroscience, Oregon National Primate Research CenterBeavertonUnited States
- Advanced Imaging Research Center, Oregon Health and Science UniversityPortlandUnited States
- Department of Behavioral Neuroscience, Oregon Health and Science UniversityPortlandUnited States
| | - Jodi L McBride
- Division of Neuroscience, Oregon National Primate Research CenterBeavertonUnited States
- Department of Behavioral Neuroscience, Oregon Health and Science UniversityPortlandUnited States
| |
Collapse
|
12
|
Jastrzębska-Więsek M, Wesołowska A, Kołaczkowski M, Varney MA, Newman-Tancredi A, Depoortere R. The selective 5-HT 1A receptor agonist, NLX-112, overcomes tetrabenazine-induced catalepsy and depression-like behavior in the rat. Behav Pharmacol 2022; 33:333-341. [PMID: 35695543 DOI: 10.1097/fbp.0000000000000681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Tetrabenazine, a preferential inhibitor of the vesicular monoamine transporter type 2, depletes the brain monoamines dopamine, serotonin and norepinephrine. Tetrabenazine and deutetrabenazine (Austedo ®) are used to treat chorea associated with Huntington's disease. However, both compounds are known to aggravate Parkinsonism and depression observed in Huntington's disease patients. NLX-112 (a.k.a. befiradol/F13640) is a highly selective, potent and efficacious serotonin 5-HT 1A agonist. In animal models, it has robust efficacy in combating other iatrogenic motor disorders such as L-DOPA-induced dyskinesia and has marked antidepressant-like activity in rodent tests. In the present study, we investigated, in rats, the efficacy of NLX-112 to counteract tetrabenazine-induced catalepsy (a model of Parkinsonism) and tetrabenazine-induced potentiation of immobility in the forced swim test (FST, a model to detect antidepressant-like activity). The prototypical 5-HT 1A agonist, (±)8-OH-DPAT, and the 5-HT 1A partial agonist/dopamine D2 receptor blocker, buspirone, were used as comparators. Both NLX-112 and (±)8-OH-DPAT (0.16-2.5 mg/kg p.o. or s.c., respectively) abolished catalepsy induced by tetrabenazine (2 mg/kg i.p.). In comparison, buspirone (0.63-5.0 mg/kg p.o.) was ineffective and even tended to potentiate tetrabenazine-induced catalepsy at 0.63 mg/kg. In the FST, NLX-112 and (±)8-OH-DPAT (0.63 mg/kg) strongly reduced immobility when administered alone but also significantly opposed potentiation of immobility induced by tetrabenazine (1.5 mg/kg i.p.). Buspirone (0.63 and 2.5 mg/kg p.o.) had no effect by itself or against tetrabenazine. These results strongly suggest that selective and highly efficacious 5-HT 1A agonists, such as NLX-112, may be useful in combating tetrabenazine-induced Parkinsonism and/or depression in Huntington's disease patients.
Collapse
Affiliation(s)
| | - Anna Wesołowska
- Department of Clinical Pharmacy, Jagiellonian University Medical College, Faculty of Pharmacy, Kraków, Poland
| | - Marcin Kołaczkowski
- Department of Clinical Pharmacy, Jagiellonian University Medical College, Faculty of Pharmacy, Kraków, Poland
| | | | | | | |
Collapse
|
13
|
Tabrizi SJ, Estevez-Fraga C, van Roon-Mom WMC, Flower MD, Scahill RI, Wild EJ, Muñoz-Sanjuan I, Sampaio C, Rosser AE, Leavitt BR. Potential disease-modifying therapies for Huntington's disease: lessons learned and future opportunities. Lancet Neurol 2022; 21:645-658. [PMID: 35716694 PMCID: PMC7613206 DOI: 10.1016/s1474-4422(22)00121-1] [Citation(s) in RCA: 153] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 02/18/2022] [Accepted: 03/04/2022] [Indexed: 01/03/2023]
Abstract
Huntington's disease is the most frequent autosomal dominant neurodegenerative disorder; however, no disease-modifying interventions are available for patients with this disease. The molecular pathogenesis of Huntington's disease is complex, with toxicity that arises from full-length expanded huntingtin and N-terminal fragments of huntingtin, which are both prone to misfolding due to proteolysis; aberrant intron-1 splicing of the HTT gene; and somatic expansion of the CAG repeat in the HTT gene. Potential interventions for Huntington's disease include therapies targeting huntingtin DNA and RNA, clearance of huntingtin protein, DNA repair pathways, and other treatment strategies targeting inflammation and cell replacement. The early termination of trials of the antisense oligonucleotide tominersen suggest that it is time to reflect on lessons learned, where the field stands now, and the challenges and opportunities for the future.
Collapse
Affiliation(s)
- Sarah J Tabrizi
- Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, UK.
| | - Carlos Estevez-Fraga
- Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | | | - Michael D Flower
- Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Rachael I Scahill
- Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Edward J Wild
- Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | | | - Cristina Sampaio
- CHDI Management, CHDI Foundation Los Angeles, CA, USA; Laboratory of Clinical Pharmacology, Faculdade de Medicina de Lisboa, Lisbon, Portugal
| | - Anne E Rosser
- BRAIN unit, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Blair R Leavitt
- Centre for Huntington's disease, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
14
|
Lawlor M, Zigo M, Kerns K, Cho IK, Easley IV CA, Sutovsky P. Spermatozoan Metabolism as a Non-Traditional Model for the Study of Huntington’s Disease. Int J Mol Sci 2022; 23:ijms23137163. [PMID: 35806166 PMCID: PMC9266437 DOI: 10.3390/ijms23137163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/22/2022] [Accepted: 06/27/2022] [Indexed: 12/10/2022] Open
Abstract
Huntington’s Disease (HD) is a fatal autosomal dominant neurodegenerative disease manifested through motor dysfunction and cognitive deficits. Decreased fertility is also observed in HD animal models and HD male patients, due to altered spermatogenesis and sperm function, thus resulting in reduced fertilization potential. Although some pharmaceuticals are currently utilized to mitigate HD symptoms, an effective treatment that remedies the pathogenesis of the disease is yet to be approved by the FDA. Identification of genes and relevant diagnostic biomarkers and therapeutic target pathways including glycolysis and mitochondrial complex-I-dependent respiration may be advantageous for early diagnosis, management, and treatment of the disease. This review addresses the HD pathway in neuronal and sperm metabolism, including relevant gene and protein expression in both neurons and spermatozoa, indicated in the pathogenesis of HD. Furthermore, zinc-containing and zinc-interacting proteins regulate and/or are regulated by zinc ion homeostasis in both neurons and spermatozoa. Therefore, this review also aims to explore the comparative role of zinc in both neuronal and sperm function. Ongoing studies aim to characterize the products of genes implicated in HD pathogenesis that are expressed in both neurons and spermatozoa to facilitate studies of future treatment avenues in HD and HD-related male infertility. The emerging link between zinc homeostasis and the HD pathway could lead to new treatments and diagnostic methods linking genetic sperm defects with somatic comorbidities.
Collapse
Affiliation(s)
- Meghan Lawlor
- Division of Animal Science, University of Missouri, Columbia, MO 65211, USA; (M.L.); (M.Z.); (K.K.)
| | - Michal Zigo
- Division of Animal Science, University of Missouri, Columbia, MO 65211, USA; (M.L.); (M.Z.); (K.K.)
| | - Karl Kerns
- Division of Animal Science, University of Missouri, Columbia, MO 65211, USA; (M.L.); (M.Z.); (K.K.)
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA
| | - In Ki Cho
- Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, GA 30602, USA; (I.K.C.); (C.A.E.IV)
- Regenerative Bioscience Center, University of Georgia, Athens, GA 30602, USA
| | - Charles A. Easley IV
- Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, GA 30602, USA; (I.K.C.); (C.A.E.IV)
- Regenerative Bioscience Center, University of Georgia, Athens, GA 30602, USA
| | - Peter Sutovsky
- Division of Animal Science, University of Missouri, Columbia, MO 65211, USA; (M.L.); (M.Z.); (K.K.)
- Department of Obstetrics, Gynecology and Women’s Health, University of Missouri, Columbia, MO 65211, USA
- Correspondence: ; Tel.: +1-(573)-882-3329
| |
Collapse
|
15
|
Huntingtin Co-Isolates with Small Extracellular Vesicles from Blood Plasma of TgHD and KI-HD Pig Models of Huntington's Disease and Human Blood Plasma. Int J Mol Sci 2022; 23:ijms23105598. [PMID: 35628406 PMCID: PMC9147436 DOI: 10.3390/ijms23105598] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/12/2022] [Accepted: 05/15/2022] [Indexed: 12/16/2022] Open
Abstract
(1) Background: Huntington’s disease (HD) is rare incurable hereditary neurodegenerative disorder caused by CAG repeat expansion in the gene coding for the protein huntingtin (HTT). Mutated huntingtin (mHTT) undergoes fragmentation and accumulation, affecting cellular functions and leading to neuronal cell death. Porcine models of HD are used in preclinical testing of currently emerging disease modifying therapies. Such therapies are aimed at reducing mHTT expression, postpone the disease onset, slow down the progression, and point out the need of biomarkers to monitor disease development and therapy efficacy. Recently, extracellular vesicles (EVs), particularly exosomes, gained attention as possible carriers of disease biomarkers. We aimed to characterize HTT and mHTT forms/fragments in blood plasma derived EVs in transgenic (TgHD) and knock-in (KI-HD) porcine models, as well as in HD patients’ plasma. (2) Methods: Small EVs were isolated by ultracentrifugation and HTT forms were visualized by western blotting. (3) Results: The full length 360 kDa HTT co-isolated with EVs from both the pig model and HD patient plasma. In addition, a ~70 kDa mutant HTT fragment was specific for TgHD pigs. Elevated total huntingtin levels in EVs from plasma of HD groups compared to controls were observed in both pig models and HD patients, however only in TgHD were they significant (p = 0.02). (4) Conclusions: Our study represents a valuable initial step towards the characterization of EV content in the search for HD biomarkers.
Collapse
|
16
|
Genetically modified mice for research on human diseases: A triumph for Biotechnology or a work in progress? THE EUROBIOTECH JOURNAL 2022. [DOI: 10.2478/ebtj-2022-0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2022] Open
Abstract
Abstract
Genetically modified mice are engineered as models for human diseases. These mouse models include inbred strains, mutants, gene knockouts, gene knockins, and ‘humanized’ mice. Each mouse model is engineered to mimic a specific disease based on a theory of the genetic basis of that disease. For example, to test the amyloid theory of Alzheimer’s disease, mice with amyloid precursor protein genes are engineered, and to test the tau theory, mice with tau genes are engineered. This paper discusses the importance of mouse models in basic research, drug discovery, and translational research, and examines the question of how to define the “best” mouse model of a disease. The critiques of animal models and the caveats in translating the results from animal models to the treatment of human disease are discussed. Since many diseases are heritable, multigenic, age-related and experience-dependent, resulting from multiple gene-gene and gene-environment interactions, it will be essential to develop mouse models that reflect these genetic, epigenetic and environmental factors from a developmental perspective. Such models would provide further insight into disease emergence, progression and the ability to model two-hit and multi-hit theories of disease. The summary examines the biotechnology for creating genetically modified mice which reflect these factors and how they might be used to discover new treatments for complex human diseases such as cancers, neurodevelopmental and neurodegenerative diseases.
Collapse
|
17
|
Rosser AE, Jones L. Huntington's Disease Gene Hunters: An Expanding Tale. Mov Disord Clin Pract 2022; 9:330-333. [PMID: 35392298 PMCID: PMC8974877 DOI: 10.1002/mdc3.13375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/25/2021] [Accepted: 11/09/2021] [Indexed: 11/08/2022] Open
Affiliation(s)
- Anne E. Rosser
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences Cardiff University School of Medicine Cardiff United Kingdom
- Cardiff Brain Repair Group Cardiff University School Biosciences Cardiff United Kingdom
- Wales Brain Research And Intracranial Neurotherapeutics (BRAIN) Unit Cardiff United Kingdom
| | - Lesley Jones
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences Cardiff University School of Medicine Cardiff United Kingdom
| |
Collapse
|
18
|
Abstract
Nonhuman primates are critically important animal models in which to study complex human diseases, understand biological functions, and address the safety of new diagnostics and therapies proposed for human use. They have genetic, physiologic, immunologic, and developmental similarities when compared to humans and therefore provide important preclinical models of human health and disease. This review highlights select research areas that demonstrate the importance of nonhuman primates in translational research. These include pregnancy and developmental disorders, infectious diseases, gene therapy, somatic cell genome editing, and applications of in vivo imaging. The power of the immune system and our increasing understanding of the role it plays in acute and chronic illnesses are being leveraged to produce new treatments for a range of medical conditions. Given the importance of the human immune system in health and disease, detailed study of the immune system of nonhuman primates is essential to advance preclinical translational research. The need for nonhuman primates continues to remain a high priority, which has been acutely evident during the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) global pandemic. Nonhuman primates will continue to address key questions and provide predictive models to identify the safety and efficiency of new diagnostics and therapies for human use across the lifespan.
Collapse
Affiliation(s)
- Alice F Tarantal
- Departments of Pediatrics and Cell Biology and Human Anatomy, University of California, Davis, California, USA;
- California National Primate Research Center, University of California, Davis, California, USA
| | - Stephen C Noctor
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, California, USA;
| | - Dennis J Hartigan-O'Connor
- California National Primate Research Center, University of California, Davis, California, USA
- Medical Microbiology and Immunology, School of Medicine, University of California, Davis, California, USA;
| |
Collapse
|
19
|
Karwacka M, Olejniczak M. Advances in Modeling Polyglutamine Diseases Using Genome Editing Tools. Cells 2022; 11:cells11030517. [PMID: 35159326 PMCID: PMC8834129 DOI: 10.3390/cells11030517] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 01/29/2022] [Accepted: 02/01/2022] [Indexed: 11/18/2022] Open
Abstract
Polyglutamine (polyQ) diseases, including Huntington’s disease, are a group of late-onset progressive neurological disorders caused by CAG repeat expansions. Although recently, many studies have investigated the pathological features and development of polyQ diseases, many questions remain unanswered. The advancement of new gene-editing technologies, especially the CRISPR-Cas9 technique, has undeniable value for the generation of relevant polyQ models, which substantially support the research process. Here, we review how these tools have been used to correct disease-causing mutations or create isogenic cell lines with different numbers of CAG repeats. We characterize various cellular models such as HEK 293 cells, patient-derived fibroblasts, human embryonic stem cells (hESCs), induced pluripotent stem cells (iPSCs) and animal models generated with the use of genome-editing technology.
Collapse
|
20
|
Benatti HR, Gray-Edwards HL. Adeno-Associated Virus Delivery Limitations for Neurological Indications. Hum Gene Ther 2022; 33:1-7. [PMID: 35049369 DOI: 10.1089/hum.2022.29196.hrb] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Hector Ribeiro Benatti
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Heather L Gray-Edwards
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA.,Department of Radiology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
21
|
Sawant N, Morton H, Kshirsagar S, Reddy AP, Reddy PH. Mitochondrial Abnormalities and Synaptic Damage in Huntington's Disease: a Focus on Defective Mitophagy and Mitochondria-Targeted Therapeutics. Mol Neurobiol 2021; 58:6350-6377. [PMID: 34519969 DOI: 10.1007/s12035-021-02556-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 09/05/2021] [Indexed: 12/12/2022]
Abstract
Huntington's disease (HD) is a fatal and pure genetic disease with a progressive loss of medium spiny neurons (MSN). HD is caused by expanded polyglutamine repeats in the exon 1 of HD gene. Clinically, HD is characterized by chorea, seizures, involuntary movements, dystonia, cognitive decline, intellectual impairment, and emotional disturbances. Several years of intense research revealed that multiple cellular changes, including defective axonal transport, protein-protein interactions, defective bioenergetics, calcium dyshomeostasis, NMDAR activation, synaptic damage, mitochondrial abnormalities, and selective loss of medium spiny neurons are implicated in HD. Recent research on mutant huntingtin (mHtt) and mitochondria has found that mHtt interacts with the mitochondrial division protein, dynamin-related protein 1 (DRP1), enhances GTPase DRP1 enzymatic activity, and causes excessive mitochondrial fragmentation and abnormal distribution, leading to defective axonal transport of mitochondria and selective synaptic degeneration. Recent research also revealed that failure to remove dead and/or dying mitochondria is an early event in the disease progression. Currently, efforts are being made to reduce abnormal protein interactions and enhance synaptic mitophagy as therapeutic strategies for HD. The purpose of this article is to discuss recent research in HD progression. This article also discusses recent developments of cell and mouse models, cellular changes, mitochondrial abnormalities, DNA damage, bioenergetics, oxidative stress, mitophagy, and therapeutics strategies in HD.
Collapse
Affiliation(s)
- Neha Sawant
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Hallie Morton
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Sudhir Kshirsagar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Arubala P Reddy
- Nutritional Sciences Department, College of Human Sciences, Texas Tech University, 1301 Akron Ave, Lubbock, TX, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
- Neuroscience & Pharmacology, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
- Neurology, Department of School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
- Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
- Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
- Department of Internal Medicine, Cell Biology & Biochemistry, Public Health and School of Health Professions, Texas Tech University Health Sciences Center, Neuroscience & Pharmacology3601 4th Street, NeurologyLubbock, TX, 79430, USA.
| |
Collapse
|
22
|
Past, present and future role of retinal imaging in neurodegenerative disease. Prog Retin Eye Res 2021; 83:100938. [PMID: 33460813 PMCID: PMC8280255 DOI: 10.1016/j.preteyeres.2020.100938] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 12/11/2020] [Accepted: 12/17/2020] [Indexed: 02/08/2023]
Abstract
Retinal imaging technology is rapidly advancing and can provide ever-increasing amounts of information about the structure, function and molecular composition of retinal tissue in humans in vivo. Most importantly, this information can be obtained rapidly, non-invasively and in many cases using Food and Drug Administration-approved devices that are commercially available. Technologies such as optical coherence tomography have dramatically changed our understanding of retinal disease and in many cases have significantly improved their clinical management. Since the retina is an extension of the brain and shares a common embryological origin with the central nervous system, there has also been intense interest in leveraging the expanding armamentarium of retinal imaging technology to understand, diagnose and monitor neurological diseases. This is particularly appealing because of the high spatial resolution, relatively low-cost and wide availability of retinal imaging modalities such as fundus photography or OCT compared to brain imaging modalities such as magnetic resonance imaging or positron emission tomography. The purpose of this article is to review and synthesize current research about retinal imaging in neurodegenerative disease by providing examples from the literature and elaborating on limitations, challenges and future directions. We begin by providing a general background of the most relevant retinal imaging modalities to ensure that the reader has a foundation on which to understand the clinical studies that are subsequently discussed. We then review the application and results of retinal imaging methodologies to several prevalent neurodegenerative diseases where extensive work has been done including sporadic late onset Alzheimer's Disease, Parkinson's Disease and Huntington's Disease. We also discuss Autosomal Dominant Alzheimer's Disease and cerebrovascular small vessel disease, where the application of retinal imaging holds promise but data is currently scarce. Although cerebrovascular disease is not generally considered a neurodegenerative process, it is both a confounder and contributor to neurodegenerative disease processes that requires more attention. Finally, we discuss ongoing efforts to overcome the limitations in the field and unmet clinical and scientific needs.
Collapse
|