1
|
Hu N, Chen Z, Zhao X, Peng X, Wu Y, Yang K, Sun T. Endothelial Dysfunction in Huntington's Disease: Pathophysiology and Therapeutic Implications. Int J Mol Sci 2025; 26:1432. [PMID: 40003898 PMCID: PMC11855594 DOI: 10.3390/ijms26041432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/03/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Huntington's disease (HD) is a progressive neurodegenerative disorder characterized by motor, cognitive, and psychiatric symptoms. While traditionally viewed through the lens of neuronal dysfunction, emerging evidence highlights the critical role of endothelial dysfunction in HD pathogenesis. This review provides a comprehensive overview of endothelial dysfunction in HD, drawing on findings from both animal models and human studies. Key features of endothelial dysfunction in HD include impaired angiogenesis, altered cerebral blood flow, compromised neurovascular coupling and cerebrovascular reactivity, and increased blood-brain barrier permeability. Genetic factors such as the mutant huntingtin protein, peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), Brain-derived neurotrophic factor (BDNF), and the adenosine A2A receptor (ADORA2A) interact to influence endothelial function in complex ways. Various therapeutic approaches targeting endothelial dysfunction, including antioxidants, nitric oxide enhancers, calcium channel blockers, statins, and metformin, have shown promise in preclinical HD models but face translational challenges, particularly regarding optimal timing of intervention and patient stratification. The implications of these findings suggest that reconceptualizing HD as a neurovascular disorder, rather than purely neuronal, could lead to more effective treatment strategies. Future research priorities should include: (1) developing validated vascular biomarkers for disease progression, (2) advancing neuroimaging techniques to monitor endothelial dysfunction in real-time. These directions will be crucial for bridging the current gap between preclinical promise and clinical success in vascular-targeted HD therapeutics.
Collapse
Affiliation(s)
- Ning Hu
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China; (N.H.); (X.Z.); (X.P.); (Y.W.)
| | - Zihao Chen
- Institute of WUT-AMU, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China;
| | - Xinyue Zhao
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China; (N.H.); (X.Z.); (X.P.); (Y.W.)
| | - Xin Peng
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China; (N.H.); (X.Z.); (X.P.); (Y.W.)
| | - Yimeng Wu
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China; (N.H.); (X.Z.); (X.P.); (Y.W.)
| | - Kai Yang
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China; (N.H.); (X.Z.); (X.P.); (Y.W.)
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, Wuhan 430070, China
| | - Taolei Sun
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China; (N.H.); (X.Z.); (X.P.); (Y.W.)
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, Wuhan 430070, China
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| |
Collapse
|
2
|
Garcia FJ, Heiman M. Molecular and cellular characteristics of cerebrovascular cell types and their contribution to neurodegenerative diseases. Mol Neurodegener 2025; 20:13. [PMID: 39881338 PMCID: PMC11780804 DOI: 10.1186/s13024-025-00799-z] [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: 11/10/2024] [Accepted: 01/07/2025] [Indexed: 01/31/2025] Open
Abstract
Many diseases and disorders of the nervous system suffer from a lack of adequate therapeutics to halt or slow disease progression, and to this day, no cure exists for any of the fatal neurodegenerative diseases. In part this is due to the incredible diversity of cell types that comprise the brain, knowledge gaps in understanding basic mechanisms of disease, as well as a lack of reliable strategies for delivering new therapeutic modalities to affected areas. With the advent of single cell genomics, it is now possible to interrogate the molecular characteristics of diverse cell populations and their alterations in diseased states. More recently, much attention has been devoted to cell populations that have historically been difficult to profile with bulk single cell technologies. In particular, cell types that comprise the cerebrovasculature have become increasingly better characterized in normal and neurodegenerative disease contexts. In this review, we describe the current understanding of cerebrovasculature structure, function, and cell type diversity and its role in the mechanisms underlying various neurodegenerative diseases. We focus on human and mouse cerebrovasculature studies and discuss both origins and consequences of cerebrovascular dysfunction, emphasizing known cell type-specific vulnerabilities in neuronal and cerebrovascular cell populations. Lastly, we highlight how novel insights into cerebrovascular biology have impacted the development of modern therapeutic approaches and discuss outstanding questions in the field.
Collapse
Affiliation(s)
- Francisco J Garcia
- The Picower Institute for Learning and Memory, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| | - Myriam Heiman
- The Picower Institute for Learning and Memory, Cambridge, MA, USA.
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA.
| |
Collapse
|
3
|
Cousineau JP, Dawe AM, Alpaugh M. Investigating the Interplay between Cardiovascular and Neurodegenerative Disease. BIOLOGY 2024; 13:764. [PMID: 39452073 PMCID: PMC11505144 DOI: 10.3390/biology13100764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/18/2024] [Accepted: 09/21/2024] [Indexed: 10/26/2024]
Abstract
Neurological diseases, including neurodegenerative diseases (NDDs), are the primary cause of disability worldwide and the second leading cause of death. The chronic nature of these conditions and the lack of disease-modifying therapies highlight the urgent need for developing effective therapies. To accomplish this, effective models of NDDs are required to increase our understanding of underlying pathophysiology and for evaluating treatment efficacy. Traditionally, models of NDDs have focused on the central nervous system (CNS). However, evidence points to a relationship between systemic factors and the development of NDDs. Cardiovascular disease and related risk factors have been shown to modify the cerebral vasculature and the risk of developing Alzheimer's disease. These findings, combined with reports of changes to vascular density and blood-brain barrier integrity in other NDDs, such as Huntington's disease and Parkinson's disease, suggest that cardiovascular health may be predictive of brain function. To evaluate this, we explore evidence for disruptions to the circulatory system in murine models of NDDs, evidence of disruptions to the CNS in cardiovascular disease models and summarize models combining cardiovascular disruption with models of NDDs. In this study, we aim to increase our understanding of cardiovascular disease and neurodegeneration interactions across multiple disease states and evaluate the utility of combining model systems.
Collapse
Affiliation(s)
| | | | - Melanie Alpaugh
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON N1G 2W1, Canada; (J.P.C.); (A.M.D.)
| |
Collapse
|
4
|
Burtscher J, Strasser B, Pepe G, Burtscher M, Kopp M, Di Pardo A, Maglione V, Khamoui AV. Brain-Periphery Interactions in Huntington's Disease: Mediators and Lifestyle Interventions. Int J Mol Sci 2024; 25:4696. [PMID: 38731912 PMCID: PMC11083237 DOI: 10.3390/ijms25094696] [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: 03/28/2024] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Prominent pathological features of Huntington's disease (HD) are aggregations of mutated Huntingtin protein (mHtt) in the brain and neurodegeneration, which causes characteristic motor (such as chorea and dystonia) and non-motor symptoms. However, the numerous systemic and peripheral deficits in HD have gained increasing attention recently, since those factors likely modulate disease progression, including brain pathology. While whole-body metabolic abnormalities and organ-specific pathologies in HD have been relatively well described, the potential mediators of compromised inter-organ communication in HD have been insufficiently characterized. Therefore, we applied an exploratory literature search to identify such mediators. Unsurprisingly, dysregulation of inflammatory factors, circulating mHtt, and many other messenger molecules (hormones, lipids, RNAs) were found that suggest impaired inter-organ communication, including of the gut-brain and muscle-brain axis. Based on these findings, we aimed to assess the risks and potentials of lifestyle interventions that are thought to improve communication across these axes: dietary strategies and exercise. We conclude that appropriate lifestyle interventions have great potential to reduce symptoms and potentially modify disease progression (possibly via improving inter-organ signaling) in HD. However, impaired systemic metabolism and peripheral symptoms warrant particular care in the design of dietary and exercise programs for people with HD.
Collapse
Affiliation(s)
- Johannes Burtscher
- Institute of Sport Sciences, University of Lausanne, 1015 Lausanne, Switzerland
| | - Barbara Strasser
- Ludwig Boltzmann Institute for Rehabilitation Research, 1100 Vienna, Austria;
- Faculty of Medicine, Sigmund Freud Private University, 1020 Vienna, Austria
| | - Giuseppe Pepe
- IRCCS Neuromed, 86077 Pozzilli, Italy; (G.P.); (A.D.P.); (V.M.)
| | - Martin Burtscher
- Department of Sport Science, University of Innsbruck, 6020 Innsbruck, Austria; (M.B.); (M.K.)
| | - Martin Kopp
- Department of Sport Science, University of Innsbruck, 6020 Innsbruck, Austria; (M.B.); (M.K.)
| | - Alba Di Pardo
- IRCCS Neuromed, 86077 Pozzilli, Italy; (G.P.); (A.D.P.); (V.M.)
| | | | - Andy V. Khamoui
- Department of Exercise Science and Health Promotion, Florida Atlantic University, Boca Raton, FL 33458, USA;
- Institute for Human Health and Disease Intervention, Florida Atlantic University, Jupiter, FL 33458, USA
| |
Collapse
|
5
|
Vasilkovska T, Salajeghe S, Vanreusel V, Van Audekerke J, Verschuuren M, Hirschler L, Warnking J, Pintelon I, Pustina D, Cachope R, Mrzljak L, Muñoz-Sanjuan I, Barbier EL, De Vos WH, Van der Linden A, Verhoye M. Longitudinal alterations in brain perfusion and vascular reactivity in the zQ175DN mouse model of Huntington's disease. J Biomed Sci 2024; 31:37. [PMID: 38627751 PMCID: PMC11022401 DOI: 10.1186/s12929-024-01028-3] [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: 01/05/2024] [Accepted: 04/08/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Huntington's disease (HD) is marked by a CAG-repeat expansion in the huntingtin gene that causes neuronal dysfunction and loss, affecting mainly the striatum and the cortex. Alterations in the neurovascular coupling system have been shown to lead to dysregulated energy supply to brain regions in several neurological diseases, including HD, which could potentially trigger the process of neurodegeneration. In particular, it has been observed in cross-sectional human HD studies that vascular alterations are associated to impaired cerebral blood flow (CBF). To assess whether whole-brain changes in CBF are present and follow a pattern of progression, we investigated both resting-state brain perfusion and vascular reactivity longitudinally in the zQ175DN mouse model of HD. METHODS Using pseudo-continuous arterial spin labelling (pCASL) MRI in the zQ175DN model of HD and age-matched wild-type (WT) mice, we assessed whole-brain, resting-state perfusion at 3, 6 and 9 and 13 months of age, and assessed hypercapnia-induced cerebrovascular reactivity (CVR), at 4.5, 6, 9 and 15 months of age. RESULTS We found increased perfusion in cortical regions of zQ175DN HET mice at 3 months of age, and a reduction of this anomaly at 6 and 9 months, ages at which behavioural deficits have been reported. On the other hand, under hypercapnia, CBF was reduced in zQ175DN HET mice as compared to the WT: for multiple brain regions at 6 months of age, for only somatosensory and retrosplenial cortices at 9 months of age, and brain-wide by 15 months. CVR impairments in cortical regions, the thalamus and globus pallidus were observed in zQ175DN HET mice at 9 months, with whole brain reactivity diminished at 15 months of age. Interestingly, blood vessel density was increased in the motor cortex at 3 months, while average vessel length was reduced in the lateral portion of the caudate putamen at 6 months of age. CONCLUSION Our findings reveal early cortical resting-state hyperperfusion and impaired CVR at ages that present motor anomalies in this HD model, suggesting that further characterization of brain perfusion alterations in animal models is warranted as a potential therapeutic target in HD.
Collapse
Affiliation(s)
- Tamara Vasilkovska
- Bio-Imaging Lab, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Antwerp, Belgium.
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium.
| | - Somaie Salajeghe
- Bio-Imaging Lab, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Antwerp, Belgium
| | - Verdi Vanreusel
- Bio-Imaging Lab, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Antwerp, Belgium
| | - Johan Van Audekerke
- Bio-Imaging Lab, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Antwerp, Belgium
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Marlies Verschuuren
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Laboratory of Cell Biology and Histology, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Antwerp, Belgium
- Antwerp Centre for Advanced Microscopy, Universiteitsplein 1, 2610 Wilrijk, Antwerp, Belgium
| | - Lydiane Hirschler
- C.J. Gorter MRI Center, Leiden University Medical Center, Leiden, the Netherlands
| | - Jan Warnking
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, Grenoble, France
| | - Isabel Pintelon
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Laboratory of Cell Biology and Histology, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Antwerp, Belgium
- Antwerp Centre for Advanced Microscopy, Universiteitsplein 1, 2610 Wilrijk, Antwerp, Belgium
| | - Dorian Pustina
- CHDI Management, Inc., the company that manages the scientific activities of CHDI Foundation, Inc, Princeton, NJ, USA
| | - Roger Cachope
- CHDI Management, Inc., the company that manages the scientific activities of CHDI Foundation, Inc, Princeton, NJ, USA
| | - Ladislav Mrzljak
- CHDI Management, Inc., the company that manages the scientific activities of CHDI Foundation, Inc, Princeton, NJ, USA
- Present Address: Takeda Pharmaceuticals, Cambridge, MA, USA
| | - Ignacio Muñoz-Sanjuan
- CHDI Management, Inc., the company that manages the scientific activities of CHDI Foundation, Inc, Princeton, NJ, USA
- Present Address: Cajal Neuroscience Inc, Seattle, WA, USA
| | - Emmanuel L Barbier
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, Grenoble, France
| | - Winnok H De Vos
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Laboratory of Cell Biology and Histology, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Antwerp, Belgium
- Antwerp Centre for Advanced Microscopy, Universiteitsplein 1, 2610 Wilrijk, Antwerp, Belgium
| | - Annemie Van der Linden
- Bio-Imaging Lab, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Antwerp, Belgium
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Marleen Verhoye
- Bio-Imaging Lab, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Antwerp, Belgium
- µNEURO Research Centre of Excellence, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
6
|
van Eimeren T, Giehl K, Reetz K, Sampaio C, Mestre TA. Neuroimaging biomarkers in Huntington's disease: Preparing for a new era of therapeutic development. Parkinsonism Relat Disord 2023; 114:105488. [PMID: 37407343 DOI: 10.1016/j.parkreldis.2023.105488] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 06/05/2023] [Accepted: 06/10/2023] [Indexed: 07/07/2023]
Abstract
BACKGROUND A critical challenge for Huntington's disease (HD) clinical trials in disease modification is the definition of endpoints that can capture change when clinical signs are subtle/non-existent. Reliable biomarkers are therefore urgently needed to facilitate drug development by allowing the enrichment of clinical trial populations and providing measures of benefit that can support the establishment of efficacy. METHODS By systematically examining the published literature on HD neuroimaging biomarker studies, we sought to advance knowledge to guide the validation of neuroimaging biomarkers. We started by reviewing both cross-sectional and longitudinal studies and then conducted an in-depth review to make quantitative comparisons between biomarkers using data only from longitudinal studies with samples sizes larger than ten participants in PET studies or 30 participants in MRI studies. RESULTS From a total of 2202 publications initially identified, we included 32 studies, 19 of which underwent in-depth comparative review. The majority of included studies used various MRI-based methods (manual to automatic) to longitudinally assess either the volume of the putamen or the caudate, which have been shown to undergo significant structural change during HD natural history. CONCLUSION Despite the impressively large number of neuroimaging biomarker studies, only a small number of adequately designed studies met our criteria. Among these various biomarkers, MRI-based volumetric analyses of the caudate and putamen are currently the best validated for use in the disease phase before clinical motor diagnosis. A biomarker that can be used to demonstrate a disease-modifying effect is still missing.
Collapse
Affiliation(s)
- Thilo van Eimeren
- University of Cologne, Faculty of Medicine, Department of Nuclear Medicine, Cologne, Germany; University of Cologne, Faculty of Medicine, Department of Neurology, Cologne, Germany.
| | - Kathrin Giehl
- University of Cologne, Faculty of Medicine, Department of Nuclear Medicine, Cologne, Germany; Research Center Jülich, Institute for Neuroscience and Medicine (INM-2), Jülich, Germany
| | - Kathrin Reetz
- University of Aachen, Department of Neurology, Aachen, Germany
| | | | - Tiago A Mestre
- University of Ottawa, Department of Medicine, Division of Neurology, The Ottawa Hospital Research Institute, Parkinson's Disease and Movement Disorders Center, Canada
| |
Collapse
|
7
|
Saro A, Gao Z, Kambey PA, Pielnaa P, Marcellin DFH, Luo A, Zheng R, Huang Z, Liao L, Zhao M, Suo L, Lu S, Li M, Cai D, Chen D, Yu H, Huang J. HIV-Proteins-Associated CNS Neurotoxicity, Their Mediators, and Alternative Treatments. Cell Mol Neurobiol 2022; 42:2553-2569. [PMID: 34562223 PMCID: PMC11421612 DOI: 10.1007/s10571-021-01151-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 09/19/2021] [Indexed: 02/08/2023]
Abstract
Human immunodeficiency virus (HIV)-infected people's livelihoods are gradually being prolonged with the use of combined antiretroviral therapy (ART). Conversely, despite viral suppression by ART, the symptoms of HIV-associated neurocognitive disorder (HAND) endure. HAND persists because ART cannot really permanently confiscate the virus from the body. HAND encompasses a variety of conditions based on clinical presentation and severity level, comprising asymptomatic neurocognitive impairment, moderate neurocognitive disorder, and HIV-associated dementia. During the early stages of HIV infection, inflammation compromises the blood-brain barrier, allowing toxic virus, infected monocytes, macrophages, T-lymphocytes, and cellular products from the bloodstream to enter the brain and eventually the entire central nervous system. Since there are no resident T-lymphocytes in the brain, the virus will live for decades in macrophages and astrocytes, establishing a reservoir of infection. The HIV proteins then inflame neurons both directly and indirectly. The purpose of this review is to provide a synopsis of the effects of these proteins on the central nervous system and conceptualize avenues to be considered in mitigating HAND. We used bioinformatics repositories extensively to simulate the transcription factors that bind to the promoter of the HIV-1 protein and possibly could be used as a target to circumvent HIV-associated neurocognitive disorders. In the same vein, a protein-protein interaction complex was also deduced from a Search Tool for the Retrieval of Interacting Genes. In conclusion, this provides an alternative strategy that could be used to avert HAND.
Collapse
Affiliation(s)
- Adonira Saro
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410013, China
| | - Zhaolin Gao
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410013, China
| | - Piniel Alphayo Kambey
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, China
| | - Paul Pielnaa
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410013, China
| | | | - Aixiang Luo
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410013, China
| | - Ruping Zheng
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410013, China
| | - Zhongjun Huang
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410013, China
| | - Lvshuang Liao
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410013, China
| | - Mingxuan Zhao
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410013, China
| | - Liangpeng Suo
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410013, China
| | - Shuang Lu
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410013, China
| | - Min Li
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410013, China
| | - Deyang Cai
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410013, China
| | - Dan Chen
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410013, China
| | - Haiyang Yu
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410013, China.
| | - Jufang Huang
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, 410013, China.
- School of Life Sciences, Central South University, Changsha, 410013, China.
| |
Collapse
|
8
|
Rocha NP, Charron O, Colpo GD, Latham LB, Patino JE, Stimming EF, Freeman L, Teixeira AL. Cerebral blood flow is associated with markers of neurodegeneration in Huntington’s disease. Parkinsonism Relat Disord 2022; 102:79-85. [DOI: 10.1016/j.parkreldis.2022.07.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 07/21/2022] [Accepted: 07/30/2022] [Indexed: 10/16/2022]
|
9
|
Garcia FJ, Sun N, Lee H, Godlewski B, Mathys H, Galani K, Zhou B, Jiang X, Ng AP, Mantero J, Tsai LH, Bennett DA, Sahin M, Kellis M, Heiman M. Single-cell dissection of the human brain vasculature. Nature 2022; 603:893-899. [PMID: 35158371 PMCID: PMC9680899 DOI: 10.1038/s41586-022-04521-7] [Citation(s) in RCA: 189] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 02/04/2022] [Indexed: 11/08/2022]
Abstract
Despite the importance of the cerebrovasculature in maintaining normal brain physiology and in understanding neurodegeneration and drug delivery to the central nervous system1, human cerebrovascular cells remain poorly characterized owing to their sparsity and dispersion. Here we perform single-cell characterization of the human cerebrovasculature using both ex vivo fresh tissue experimental enrichment and post mortem in silico sorting of human cortical tissue samples. We capture 16,681 cerebrovascular nuclei across 11 subtypes, including endothelial cells, mural cells and three distinct subtypes of perivascular fibroblast along the vasculature. We uncover human-specific expression patterns along the arteriovenous axis and determine previously uncharacterized cell-type-specific markers. We use these human-specific signatures to study changes in 3,945 cerebrovascular cells from patients with Huntington's disease, which reveal activation of innate immune signalling in vascular and glial cell types and a concomitant reduction in the levels of proteins critical for maintenance of blood-brain barrier integrity. Finally, our study provides a comprehensive molecular atlas of the human cerebrovasculature to guide future biological and therapeutic studies.
Collapse
Affiliation(s)
- Francisco J Garcia
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
- Picower Institute for Learning and Memory, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Na Sun
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Electrical Engineering and Computer Science, MIT, Cambridge, MA, USA
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, USA
| | - Hyeseung Lee
- Picower Institute for Learning and Memory, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Brianna Godlewski
- Rosamund Stone Zander Translational Neuroscience Center, F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Hansruedi Mathys
- Picower Institute for Learning and Memory, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kyriaki Galani
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Electrical Engineering and Computer Science, MIT, Cambridge, MA, USA
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, USA
| | - Blake Zhou
- Picower Institute for Learning and Memory, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Xueqiao Jiang
- Picower Institute for Learning and Memory, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ayesha P Ng
- Picower Institute for Learning and Memory, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Julio Mantero
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Electrical Engineering and Computer Science, MIT, Cambridge, MA, USA
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, USA
| | - Li-Huei Tsai
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
- Picower Institute for Learning and Memory, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Mustafa Sahin
- Rosamund Stone Zander Translational Neuroscience Center, F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Manolis Kellis
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Electrical Engineering and Computer Science, MIT, Cambridge, MA, USA.
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, USA.
| | - Myriam Heiman
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA.
- Picower Institute for Learning and Memory, Cambridge, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
10
|
Vidyadhara DJ, Yarreiphang H, Raju TR, Alladi PA. Differences in Neuronal Numbers, Morphology, and Developmental Apoptosis in Mice Nigra Provide Experimental Evidence of Ontogenic Origin of Vulnerability to Parkinson's Disease. Neurotox Res 2021; 39:1892-1907. [PMID: 34762290 DOI: 10.1007/s12640-021-00439-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/26/2021] [Accepted: 10/29/2021] [Indexed: 10/19/2022]
Abstract
Parkinson disease (PD) prevalence varies by ethnicity. In an earlier study, we replicated the reduced vulnerability to PD in an admixed population, using 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-susceptible C57BL/6 J, MPTP-resistant CD-1 and their F1 crossbreds. In the present study, we investigated if the differences have a developmental origin. Substantia nigra was evaluated at postnatal days 2 (P2), P6, P10, P14, P18, and P22. C57BL/6 J mice had smaller nigra and fewer dopaminergic neurons than the CD-1 and crossbreds at P2, which persisted through development. A significant increase in numbers and nigral volume was observed across strains until P14. A drastic decline thereafter was specific to C57BL/6 J. CD-1 and crossbreds retained their numbers from P14 to stabilize with supernumerary neurons at adulthood. The neuronal size increased gradually to attain adult morphology at P10 in the resistant strains, vis-à-vis at P22 in C57BL/6 J. Accordingly, in comparison to C57BL/6 J, the nigra of CD-1 and reciprocal crossbreds possessed cytomorphological features of resilience, since birth. The considerably lesser dopaminergic neuronal loss in the CD-1 and crossbreds was seen at P2 and P14 and thereafter was complemented by attenuated developmental cell death. The differences in programmed cell death were confirmed by reduced TUNEL labelling, AIF, and caspase-3 expression. GDNF expression aligned with the cell death pattern at P2 and P14 in both nigra and striatum. Earlier maturity of nigra and its neurons appears to be better features that reflect as MPTP resistance at adulthood. Thus, variable MPTP vulnerability in mice and also differential susceptibility to PD in humans may arise early during nigral development.
Collapse
Affiliation(s)
- D J Vidyadhara
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, 560029, India
- Departments of Neurology and Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| | - Haorei Yarreiphang
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, 560029, India
| | - Trichur R Raju
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, 560029, India
| | - Phalguni Anand Alladi
- Department of Clinical Psychopharmacology and Neurotoxicology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, 560029, India.
- Formerly at Department of Neurophysiology, National Institute of Mental Health and Neuro-Sciences, Hosur Road, Bangalore, India.
| |
Collapse
|
11
|
Ouellette J, Lacoste B. From Neurodevelopmental to Neurodegenerative Disorders: The Vascular Continuum. Front Aging Neurosci 2021; 13:749026. [PMID: 34744690 PMCID: PMC8570842 DOI: 10.3389/fnagi.2021.749026] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/13/2021] [Indexed: 12/12/2022] Open
Abstract
Structural and functional integrity of the cerebral vasculature ensures proper brain development and function, as well as healthy aging. The inability of the brain to store energy makes it exceptionally dependent on an adequate supply of oxygen and nutrients from the blood stream for matching colossal demands of neural and glial cells. Key vascular features including a dense vasculature, a tightly controlled environment, and the regulation of cerebral blood flow (CBF) all take part in brain health throughout life. As such, healthy brain development and aging are both ensured by the anatomical and functional interaction between the vascular and nervous systems that are established during brain development and maintained throughout the lifespan. During critical periods of brain development, vascular networks remodel until they can actively respond to increases in neural activity through neurovascular coupling, which makes the brain particularly vulnerable to neurovascular alterations. The brain vasculature has been strongly associated with the onset and/or progression of conditions associated with aging, and more recently with neurodevelopmental disorders. Our understanding of cerebrovascular contributions to neurological disorders is rapidly evolving, and increasing evidence shows that deficits in angiogenesis, CBF and the blood-brain barrier (BBB) are causally linked to cognitive impairment. Moreover, it is of utmost curiosity that although neurodevelopmental and neurodegenerative disorders express different clinical features at different stages of life, they share similar vascular abnormalities. In this review, we present an overview of vascular dysfunctions associated with neurodevelopmental (autism spectrum disorders, schizophrenia, Down Syndrome) and neurodegenerative (multiple sclerosis, Huntington's, Parkinson's, and Alzheimer's diseases) disorders, with a focus on impairments in angiogenesis, CBF and the BBB. Finally, we discuss the impact of early vascular impairments on the expression of neurodegenerative diseases.
Collapse
Affiliation(s)
- Julie Ouellette
- Ottawa Hospital Research Institute, Neuroscience Program, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Baptiste Lacoste
- Ottawa Hospital Research Institute, Neuroscience Program, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada
| |
Collapse
|
12
|
Plinta K, Plewka A, Wójcik-Pędziwiatr M, Zmarzły N, Rudziński M, Rudzińska-Bar M. Is TGF-β1 a Biomarker of Huntington's Disease Progression? J Clin Med 2021; 10:jcm10133001. [PMID: 34279486 PMCID: PMC8269288 DOI: 10.3390/jcm10133001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 06/25/2021] [Accepted: 07/01/2021] [Indexed: 12/30/2022] Open
Abstract
Huntington's disease (HD) is an autosomal dominant genetic disease that can be divided into preclinical and symptomatic stages. Due to the diverse HD phenotype, there is an urgent need to identify markers that would independently assess its severity. The aim of this study was to evaluate the use of plasma levels of TGF-β1 in the assessment of HD severity. One hundred HD patients and 40 healthy volunteers were included in the study. All HD patients underwent neurological and cognitive function assessment. TGF-β1 levels were determined in the plasma of all patients. The correlations between TGF-β1 levels and clinical profile and HD severity were also investigated. In symptomatic patients, cognitive decline was demonstrated, while in preclinical patients, no symptoms were found. Plasma levels of TGF-β1 in HD patients did not differ significantly from the control group and did not change with the progression of the disease. In addition, TGF-β1 levels also did not correlate with the severity of motor dysfunction. Positive correlations between plasma TGF-β1 concentration and intensity of cognitive impairment were found, but only in the early disease stage. There was no clear benefit in assessing plasma TGF-β1 levels in HD patients as a marker of disease severity.
Collapse
Affiliation(s)
- Klaudia Plinta
- Neurology and Stroke Department, Regional Hospital of Saint Hedwig, 45-221 Opole, Poland;
| | - Andrzej Plewka
- Institute of Health Sciences, University of Opole, 45-040 Opole, Poland;
| | - Magdalena Wójcik-Pędziwiatr
- Department of Neurology, Faculty of Medicine and Health Sciences, Andrzej Frycz Modrzewski Krakow University, 30-705 Krakow, Poland;
| | - Nikola Zmarzły
- Department of Histology, Cytophysiology and Embryology, Faculty of Medicine, University of Technology in Katowice, 41-800 Zabrze, Poland;
| | - Marcin Rudziński
- Department of Laryngology, Jagiellonian University Medical College, 30-688 Krakow, Poland;
| | - Monika Rudzińska-Bar
- Department of Neurology, Faculty of Medicine and Health Sciences, Andrzej Frycz Modrzewski Krakow University, 30-705 Krakow, Poland;
- Correspondence:
| |
Collapse
|
13
|
Klinkmueller P, Kronenbuerger M, Miao X, Bang J, Ultz KE, Paez A, Zhang X, Duan W, Margolis RL, van Zijl PCM, Ross CA, Hua J. Impaired response of cerebral oxygen metabolism to visual stimulation in Huntington's disease. J Cereb Blood Flow Metab 2021; 41:1119-1130. [PMID: 32807001 PMCID: PMC8054727 DOI: 10.1177/0271678x20949286] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 06/30/2020] [Accepted: 07/15/2020] [Indexed: 01/29/2023]
Abstract
Huntington's disease (HD) is a neurodegenerative disease caused by a CAG triplet repeat expansion in the Huntingtin gene. Metabolic and microvascular abnormalities in the brain may contribute to early physiological changes that subserve the functional impairments in HD. This study is intended to investigate potential abnormality in dynamic changes in cerebral blood volume (CBV) and cerebral blood flow (CBF), and cerebral metabolic rate of oxygen (CMRO2) in the brain in response to functional stimulation in premanifest and early manifest HD patients. A recently developed 3-D-TRiple-acquisition-after-Inversion-Preparation magnetic resonance imaging (MRI) approach was used to measure dynamic responses in CBV, CBF, and CMRO2 during visual stimulation in one single MRI scan. Experiments were conducted in 23 HD patients and 16 healthy controls. Decreased occipital cortex CMRO2 responses were observed in premanifest and early manifest HD patients compared to controls (P < 0.001), correlating with the CAG-Age Product scores in these patients (R2 = 0.4, P = 0.001). The results suggest the potential value of this reduced CMRO2 response during visual stimulation as a biomarker for HD and may illuminate the role of metabolic alterations in the pathophysiology of HD.
Collapse
Affiliation(s)
- Peter Klinkmueller
- Department of Electrical and Computer Engineering, Johns Hopkins University, Baltimore, MD, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
- Neurosection, Division of MRI Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Martin Kronenbuerger
- Division of Movement Disorders, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, University of Greifswald, Greifswald, Germany
| | - Xinyuan Miao
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
- Neurosection, Division of MRI Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jee Bang
- Division of Movement Disorders, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kia E Ultz
- Division of Movement Disorders, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Adrian Paez
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
- Neurosection, Division of MRI Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xiaoyu Zhang
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
- Neurosection, Division of MRI Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Wenzhen Duan
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Departments of Neuroscience and Pharmacology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Russell L Margolis
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter CM van Zijl
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
- Neurosection, Division of MRI Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christopher A Ross
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Departments of Neuroscience and Pharmacology, 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
- Neurosection, Division of MRI Research, Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
14
|
Tan B, Shishegar R, Poudel GR, Fornito A, Georgiou-Karistianis N. Cortical morphometry and neural dysfunction in Huntington's disease: a review. Eur J Neurol 2020; 28:1406-1419. [PMID: 33210786 DOI: 10.1111/ene.14648] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/22/2020] [Accepted: 11/12/2020] [Indexed: 01/09/2023]
Abstract
Numerous neuroimaging techniques have been used to identify biomarkers of disease progression in Huntington's disease (HD). To date, the earliest and most sensitive of these is caudate volume; however, it is becoming increasingly evident that numerous changes to cortical structures, and their interconnected networks, occur throughout the course of the disease. The mechanisms by which atrophy spreads from the caudate to these cortical regions remains unknown. In this review, the neuroimaging literature specific to T1-weighted and diffusion-weighted magnetic resonance imaging is summarized and new strategies for the investigation of cortical morphometry and the network spread of degeneration in HD are proposed. This new avenue of research may enable further characterization of disease pathology and could add to a suite of biomarker/s of disease progression for patient stratification that will help guide future clinical trials.
Collapse
Affiliation(s)
- Brendan Tan
- School of Psychological Sciences, Turner Institute for Brain and Mental Health, Monash University, Melbourne, VIC, Australia
| | - Rosita Shishegar
- School of Psychological Sciences, Turner Institute for Brain and Mental Health, Monash University, Melbourne, VIC, Australia.,Australian e-Health Research Centre, CSIRO, Melbourne, VIC, Australia.,Monash Biomedical Imaging, Melbourne, VIC, Australia
| | - Govinda R Poudel
- School of Psychological Sciences, Turner Institute for Brain and Mental Health, Monash University, Melbourne, VIC, Australia.,Sydney Imaging, Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia.,Australian Catholic University, Melbourne, VIC, Australia
| | - Alex Fornito
- School of Psychological Sciences, Turner Institute for Brain and Mental Health, Monash University, Melbourne, VIC, Australia.,Monash Biomedical Imaging, Melbourne, VIC, Australia
| | - Nellie Georgiou-Karistianis
- School of Psychological Sciences, Turner Institute for Brain and Mental Health, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
15
|
Palomar-Garcia A, Camara E. SeSBAT: Single Subject Brain Analysis Toolbox. Application to Huntington's Disease as a Preliminary Study. Front Syst Neurosci 2020; 14:488652. [PMID: 33117135 PMCID: PMC7550747 DOI: 10.3389/fnsys.2020.488652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 08/21/2020] [Indexed: 12/02/2022] Open
Abstract
Magnetic resonance imaging (MRI) biomarkers require complex processing routines that are time-consuming and labor-intensive for clinical users. The Single Subject Brain Analysis Toolbox (SeSBAT) is a fully automated MATLAB toolbox with a graphical user interface (GUI) that offers standardized and optimized protocols for the pre-processing and analysis of anatomical MRI data at the single-subject level. In this study, the two-fold strategy provided by SeSBAT is illustrated through its application on a cohort of 42 patients with Huntington’s disease (HD), in pre-manifest and early manifest stages, as a suitable model of neurodegenerative processes. On the one hand, hypothesis-driven analysis can be used to extract biomarkers of neurodegeneration in specific brain regions of interest (ROI-based analysis). On the other hand, an exploratory voxel-based morphometry (VBM) approach can detect volume changes due to neurodegeneration throughout the whole brain (whole-brain analysis). That illustration reveals the potential of SeSBAT in providing potential prognostic biomarkers in neurodegenerative processes in clinics, which could be critical to overcoming the limitations of current qualitative evaluation strategies, and thus improve the diagnosis and monitoring of neurodegenerative disorders. Furthermore, the importance of the availability of tools for characterization at the single-subject level has been emphasized, as there is high interindividual variability in the pattern of neurodegeneration. Thus, tools like SeSBAT could pave the way towards more effective and personalized medicine.
Collapse
Affiliation(s)
- Alicia Palomar-Garcia
- Cognition and Brain Plasticity Unit, IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), Barcelona, Spain
| | - Estela Camara
- Department of Cognition, Development and Educational Psychology, University of Barcelona, Barcelona, Spain
| |
Collapse
|
16
|
Di Maio LG, Montorio D, Peluso S, Dolce P, Salvatore E, De Michele G, Cennamo G. Optical coherence tomography angiography findings in Huntington’s disease. Neurol Sci 2020; 42:995-1001. [DOI: 10.1007/s10072-020-04611-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 07/18/2020] [Indexed: 12/15/2022]
|
17
|
Steventon JJ, Furby H, Ralph J, O'Callaghan P, Rosser AE, Wise RG, Busse M, Murphy K. Altered cerebrovascular response to acute exercise in patients with Huntington's disease. Brain Commun 2020; 2:fcaa044. [PMID: 32566927 PMCID: PMC7293798 DOI: 10.1093/braincomms/fcaa044] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/26/2020] [Accepted: 03/16/2020] [Indexed: 01/20/2023] Open
Abstract
The objective of this study was to determine whether a single session of exercise was sufficient to induce cerebral adaptations in individuals with Huntington’s disease and to explore the time dynamics of any acute cerebrovascular response. In this case–control study, we employed arterial-spin labelling MRI in 19 Huntington’s disease gene-positive participants (32–65 years, 13 males) and 19 controls (29–63 years, 10 males) matched for age, gender, body mass index and self-reported activity levels, to measure global and regional perfusion in response to 20 min of moderate-intensity cycling. Cerebral perfusion was measured at baseline and 15, 40 and 60 min after exercise cessation. Relative to baseline, we found that cerebral perfusion increased in patients with Huntington’s disease yet was unchanged in control participants in the precentral gyrus (P = 0.016), middle frontal gyrus (P = 0.046) and hippocampus (P = 0.048) 40 min after exercise cessation (+15 to +32.5% change in Huntington’s disease participants, −7.7 to 0.8% change in controls). The length of the disease‐causing trinucleotide repeat expansion in the huntingtin gene predicted the change in the precentral gyrus (P = 0.03) and the intensity of the exercise intervention predicted hippocampal perfusion change in Huntington’s disease participants (P < 0.001). In both groups, exercise increased hippocampal blood flow 60 min after exercise cessation (P = 0.039). These findings demonstrate the utility of acute exercise as a clinically sensitive experimental paradigm to modulate the cerebrovasculature. Twenty minutes of aerobic exercise induced transient cerebrovascular adaptations in the hippocampus and cortex selectively in Huntington’s disease participants and likely represents latent neuropathology not evident at rest.
Collapse
Affiliation(s)
- Jessica J Steventon
- Cardiff University Brain Research Imaging Centre, School of Physics and Astronomy, Cardiff University, Cardiff CF24 4HQ, UK.,Neuroscience and Mental Health Research Institute, School of Medicine, Cardiff CF24 4HQ, UK
| | - Hannah Furby
- Neuroscience and Mental Health Research Institute, School of Medicine, Cardiff CF24 4HQ, UK.,Cardiff University Brain Research Imaging Centre, School of Psychology, Cardiff University, Cardiff CF24 4HQ, UK
| | - James Ralph
- Cardiff University Brain Research Imaging Centre, School of Psychology, Cardiff University, Cardiff CF24 4HQ, UK
| | - Peter O'Callaghan
- Cardiology Department, University Hospital of Wales, Cardiff CF14 4XW, UK
| | - Anne E Rosser
- Neuroscience and Mental Health Research Institute, School of Medicine, Cardiff CF24 4HQ, UK.,Cardiff Brain Repair Group, School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
| | - Richard G Wise
- Cardiff University Brain Research Imaging Centre, School of Psychology, Cardiff University, Cardiff CF24 4HQ, UK
| | - Monica Busse
- Centre for Trials Research, Cardiff University, Cardiff CF14 4YS, UK
| | - Kevin Murphy
- Cardiff University Brain Research Imaging Centre, School of Physics and Astronomy, Cardiff University, Cardiff CF24 4HQ, UK
| |
Collapse
|
18
|
Kielar C, Morton AJ. Early Neurodegeneration in R6/2 Mice Carrying the Huntington's Disease Mutation with a Super-Expanded CAG Repeat, Despite Normal Lifespan. J Huntingtons Dis 2019; 7:61-76. [PMID: 29480204 DOI: 10.3233/jhd-170265] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The threshold of CAG repeat expansion in the HTT gene that causes HD is 36 CAG repeats, although 'superlong' expansions are found in individual neurons in postmortem brains. Previously, we showed that, compared to mice with <250 CAG repeats, onset of disease in R6/2 mice carrying superlong (>440) CAG repeat expansions was delayed, and disease progression was slower. Inclusion pathology also differed from 250 CAG repeat mice, being dominated by a novel kind of extranuclear neuronal inclusion (nENNI) that resembles a class of aggregate seen in patients with the adult onset form of HD. Here, we characterised neuropathology in R6/2 mice with >400 CAG repeats using light and electron microscopy. nENNIs were found with increased frequency and wider distribution with age. Some nENNIs appear to 'mature' as the disease develops, developing a multi-layered cored structure. Mice with superlong CAG repeats do not develop clinical signs until they are around 30-40 weeks of age, and they attain a normal life span (>2 years). Nevertheless, they show brain atrophy and unequivocal neuron loss from the striatum and cortex by 22 weeks of age, an age at which similar pathology is seen in 250 CAG repeat mice. Since this time-point is 'end stage' for a 250 CAG mouse, but very far (at least 18 months) from end stage for a > 440 CAG repeat mouse, our data confirm that the appearance of clinical signs, the formation of inclusions, and neurodegeneration are processes that progress independently. A better understanding of the relationship between CAG repeat length, neurodegenerative pathways, and clinical behavioural signs is essential, if we are to find strategies to delay or reverse the course of this disease.
Collapse
Affiliation(s)
- Catherine Kielar
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - A Jennifer Morton
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| |
Collapse
|
19
|
Ikram MA, Zonneveld HI, Roshchupkin G, Smith AV, Franco OH, Sigurdsson S, van Duijn C, Uitterlinden AG, Launer LJ, Vernooij MW, Gudnason V, Adams HH. Heritability and genome-wide associations studies of cerebral blood flow in the general population. J Cereb Blood Flow Metab 2018; 38. [PMID: 28627999 PMCID: PMC6120124 DOI: 10.1177/0271678x17715861] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Cerebral blood flow is an important process for brain functioning and its dysregulation is implicated in multiple neurological disorders. While environmental risk factors have been identified, it remains unclear to what extent the flow is regulated by genetics. Here we performed heritability and genome-wide association analyses of cerebral blood flow in a population-based cohort study. We included 4472 persons free of cortical infarcts who underwent genotyping and phase-contrast magnetic resonance flow imaging (mean age 64.8 ± 10.8 years). The flow rate, cross-sectional area of the vessel, and flow velocity through the vessel were measured in the basilar artery and bilateral carotids. We found that the flow rate of the basilar artery is most heritable (h2 (SE) = 24.1 (9.8), p-value = 0.0056), and this increased over age. The association studies revealed two significant loci for the right carotid artery area (rs12546630, p-value = 2.0 × 10-8) and velocity (rs2971609, p-value = 1.4 × 10-8), with the latter showing a concordant effect in an independent sample (N = 1350, p-value = 0.057, meta-analyzed p-value = 2.5 × 10-9). These loci were also associated with other cerebral blood flow parameters below genome-wide significance, and rs2971609 lies in a known migraine locus. These findings establish that cerebral blood flow is under genetic control with potential relevance for neurological diseases.
Collapse
Affiliation(s)
- M Arfan Ikram
- 1 Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands.,2 Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands.,3 Department of Neurology, Erasmus MC, Rotterdam, the Netherlands
| | - Hazel I Zonneveld
- 1 Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands.,2 Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - Gennady Roshchupkin
- 2 Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands.,4 Department of Medical Informatics, Erasmus MC, Rotterdam, the Netherlands
| | - Albert V Smith
- 5 Icelandic Heart Association, Kopavogur, Iceland.,6 Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Oscar H Franco
- 1 Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands
| | | | | | | | | | - Meike W Vernooij
- 1 Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands.,2 Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - Vilmundur Gudnason
- 5 Icelandic Heart Association, Kopavogur, Iceland.,6 Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Hieab Hh Adams
- 1 Department of Epidemiology, Erasmus MC, Rotterdam, the Netherlands.,2 Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands
| |
Collapse
|
20
|
Hong Y, O'Donnell LJ, Savadjiev P, Zhang F, Wassermann D, Pasternak O, Johnson H, Paulsen J, Vonsattel JP, Makris N, Westin CF, Rathi Y. Genetic load determines atrophy in hand cortico-striatal pathways in presymptomatic Huntington's disease. Hum Brain Mapp 2018; 39:3871-3883. [PMID: 29797744 DOI: 10.1002/hbm.24217] [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] [Received: 12/03/2017] [Revised: 04/30/2018] [Accepted: 05/06/2018] [Indexed: 12/31/2022] Open
Abstract
Huntington's disease (HD) is an inherited neurodegenerative disorder that causes progressive breakdown of striatal neurons. Standard white matter integrity measures like fractional anisotropy and mean diffusivity derived from diffusion tensor imaging were analyzed in prodromal-HD subjects; however, they studied either a whole brain or specific subcortical white matter structures with connections to cortical motor areas. In this work, we propose a novel analysis of a longitudinal cohort of 243 prodromal-HD individuals and 88 healthy controls who underwent two or more diffusion MRI scans as part of the PREDICT-HD study. We separately trace specific white matter fiber tracts connecting the striatum (caudate and putamen) with four cortical regions corresponding to the hand, face, trunk, and leg motor areas. A multi-tensor tractography algorithm with an isotropic volume fraction compartment allows estimating diffusion of fast-moving extra-cellular water in regions containing crossing fibers and provides quantification of a microstructural property related to tissue atrophy. The tissue atrophy rate is separately analyzed in eight cortico-striatal pathways as a function of CAG-repeats (genetic load) by statistically regressing out age effect from our cohort. The results demonstrate a statistically significant increase in isotropic volume fraction (atrophy) bilaterally in hand fiber connections to the putamen with increasing CAG-repeats, which connects the genetic abnormality (CAG-repeats) to an imaging-based microstructural marker of tissue integrity in specific white matter pathways in HD. Isotropic volume fraction measures in eight cortico-striatal pathways are also correlated significantly with total motor scores and diagnostic confidence levels, providing evidence of their relevance to HD clinical presentation.
Collapse
Affiliation(s)
- Yi Hong
- Department of Computer Science, University of Georgia, Athens, Georgia
| | - Lauren J O'Donnell
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Peter Savadjiev
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Fan Zhang
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Ofer Pasternak
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Hans Johnson
- Department of Psychiatry, University of Iowa, Iowa City, Iowa
| | - Jane Paulsen
- Department of Psychiatry, University of Iowa, Iowa City, Iowa
| | - Jean-Paul Vonsattel
- Department of Pathology and Cell Biology, Columbia University, New York, New York
| | - Nikos Makris
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Carl F Westin
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Yogesh Rathi
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
21
|
Garcia‐Gorro C, de Diego‐Balaguer R, Martínez‐Horta S, Pérez‐Pérez J, Kulisevsky J, Rodríguez‐Dechicha N, Vaquer I, Subira S, Calopa M, Muñoz E, Santacruz P, Ruiz‐Idiago J, Mareca C, Caballol N, Camara E. Reduced striato-cortical and inhibitory transcallosal connectivity in the motor circuit of Huntington's disease patients. Hum Brain Mapp 2018; 39:54-71. [PMID: 28990240 PMCID: PMC6866479 DOI: 10.1002/hbm.23813] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 08/25/2017] [Accepted: 09/05/2017] [Indexed: 12/13/2022] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disorder which is primarily associated with striatal degeneration. However, the alterations in connectivity of this structure in HD have been underinvestigated. In this study, we analyzed the functional and structural connectivity of the left putamen, while participants performed a finger-tapping task. Using fMRI and DW-MRI, 30 HD gene expansion carriers (HDGEC) and 29 healthy participants were scanned. Psychophysiological interaction analysis and DTI-based tractography were employed to examine functional and structural connectivity, respectively. Manifest HDGEC exhibited a reduced functional connectivity of the left putamen with the left and the right primary sensorimotor areas (SM1). Based on this result, the inhibitory functional connectivity between the left SM1 and the right SM1 was explored, appearing to be also decreased. In addition, the tract connecting these areas (motor corpus callosum), and the tract connecting the left putamen with the left SM1 appeared disrupted in HDGEC compared to controls. Significant correlations were found between measures of functional and structural connectivity of the motor corpus callosum, showing a coupling of both types of alterations in this tract. The observed reduction of functional and structural connectivity was associated with worse motor scores, which highlights the clinical relevance of these results. Hum Brain Mapp 39:54-71, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Clara Garcia‐Gorro
- Cognition and Brain Plasticity UnitIDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de LlobregatBarcelonaSpain
- Department of Cognition, Development and Educational PsychologyUniversity of BarcelonaBarcelonaSpain
| | - Ruth de Diego‐Balaguer
- Cognition and Brain Plasticity UnitIDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de LlobregatBarcelonaSpain
- Department of Cognition, Development and Educational PsychologyUniversity of BarcelonaBarcelonaSpain
- The Institute of Neurosciences of the University of BarcelonaBarcelonaSpain
- ICREA (Catalan Institute for Research and Advanced Studies)BarcelonaSpain
| | - Saul Martínez‐Horta
- Movement Disorders Unit, Department of NeurologyBiomedical Research Institute Sant Pau (IIB‐Sant Pau), Hospital de la Santa Creu i Sant PauBarcelonaSpain
- CIBERNED (Center for Networked Biomedical Research on Neurodegenerative Diseases), Carlos III InstituteMadridSpain
| | - Jesus Pérez‐Pérez
- Movement Disorders Unit, Department of NeurologyBiomedical Research Institute Sant Pau (IIB‐Sant Pau), Hospital de la Santa Creu i Sant PauBarcelonaSpain
- CIBERNED (Center for Networked Biomedical Research on Neurodegenerative Diseases), Carlos III InstituteMadridSpain
| | - Jaime Kulisevsky
- Movement Disorders Unit, Department of NeurologyBiomedical Research Institute Sant Pau (IIB‐Sant Pau), Hospital de la Santa Creu i Sant PauBarcelonaSpain
- CIBERNED (Center for Networked Biomedical Research on Neurodegenerative Diseases), Carlos III InstituteMadridSpain
- Universidad Autónoma de BarcelonaBarcelonaSpain
| | | | - Irene Vaquer
- Hestia Duran i Reynals, Hospital Duran i Reynals, Hospitalet de LlobregatBarcelonaSpain
| | - Susana Subira
- Hestia Duran i Reynals, Hospital Duran i Reynals, Hospitalet de LlobregatBarcelonaSpain
- Department of Clinical and Health PsychologyUniversitat Autònoma de BarcelonaBarcelonaSpain
| | - Matilde Calopa
- Movement Disorders Unit, Neurology Service, Hospital Universitari de Bellvitge, L'Hospitalet de LlobregatBarcelonaSpain
| | - Esteban Muñoz
- Movement Disorders Unit, Neurology Service, Hospital ClínicBarcelonaSpain
- IDIBAPS (Institut d'Investigacions Biomèdiques August Pi i Sunyer)BarcelonaSpain
- Facultat de medicina, University of BarcelonaBarcelonaSpain
| | - Pilar Santacruz
- Movement Disorders Unit, Neurology Service, Hospital ClínicBarcelonaSpain
| | | | | | - Nuria Caballol
- Hospital de Sant Joan Despí Moisès Broggi, Sant Joan DespíBarcelonaSpain
| | - Estela Camara
- Cognition and Brain Plasticity UnitIDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de LlobregatBarcelonaSpain
- Department of Cognition, Development and Educational PsychologyUniversity of BarcelonaBarcelonaSpain
| |
Collapse
|
22
|
Neuroimaging as a tool to study the sources of phenotypic heterogeneity in Huntington's disease. Curr Opin Neurol 2017; 30:398-404. [PMID: 28509681 DOI: 10.1097/wco.0000000000000461] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Huntington's disease is a neurodegenerative disorder characterized by a triad of motor, cognitive and psychiatric disturbances. There is great variability regarding the prominence and evolution of each type of clinical sign. One possible source of phenotypic heterogeneity could be the more prominent degeneration of specific brain circuits. The scope of this review is to highlight the most recent neuroimaging studies that have analysed the relationship between brain changes and motor, cognitive and psychiatric alterations in Huntington's disease. RECENT FINDINGS The results from recent neuroimaging studies are heterogeneous. Although there is a great overlap between the different regions associated with each symptomatic domain, there is some degree of differentiation. For example, the motor network is associated with motor impairment, whereas the ventral striatum is especially involved in emotional deficits related with psychiatric problems. SUMMARY Motor, cognitive and psychiatric impairments are associated with structural and functional brain biomarkers. However, the specificity of the regions involved remains unknown, because these studies focused on specific regions and symptoms. In order to tease apart the neural substrates that underlie the phenotypic heterogeneity in Huntington's disease, multivariate approaches combining brain and behavioural measures related to all symptomatic domains should be considered in the future.
Collapse
|
23
|
Shaffer JJ, Ghayoor A, Long JD, Kim RE, Lourens S, O'Donnell LJ, Westin C, Rathi Y, Magnotta V, Paulsen JS, Johnson HJ. Longitudinal diffusion changes in prodromal and early HD: Evidence of white-matter tract deterioration. Hum Brain Mapp 2017; 38:1460-1477. [PMID: 28045213 PMCID: PMC5400101 DOI: 10.1002/hbm.23465] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 10/11/2016] [Accepted: 11/03/2016] [Indexed: 01/08/2023] Open
Abstract
INTRODUCTION Huntington's disease (HD) is a genetic neurodegenerative disorder that primarily affects striatal neurons. Striatal volume loss is present years before clinical diagnosis; however, white matter degradation may also occur prior to diagnosis. Diffusion-weighted imaging (DWI) can measure microstructural changes associated with degeneration that precede macrostructural changes. DWI derived measures enhance understanding of degeneration in prodromal HD (pre-HD). METHODS As part of the PREDICT-HD study, N = 191 pre-HD individuals and 70 healthy controls underwent two or more (baseline and 1-5 year follow-up) DWI, with n = 649 total sessions. Images were processed using cutting-edge DWI analysis methods for large multicenter studies. Diffusion tensor imaging (DTI) metrics were computed in selected tracts connecting the primary motor, primary somato-sensory, and premotor areas of the cortex with the subcortical caudate and putamen. Pre-HD participants were divided into three CAG-Age Product (CAP) score groups reflecting clinical diagnosis probability (low, medium, or high probabilities). Baseline and longitudinal group differences were examined using linear mixed models. RESULTS Cross-sectional and longitudinal differences in DTI measures were present in all three CAP groups compared with controls. The high CAP group was most affected. CONCLUSIONS This is the largest longitudinal DWI study of pre-HD to date. Findings showed DTI differences, consistent with white matter degeneration, were present up to a decade before predicted HD diagnosis. Our findings indicate a unique role for disrupted connectivity between the premotor area and the putamen, which may be closely tied to the onset of motor symptoms in HD. Hum Brain Mapp 38:1460-1477, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - Ali Ghayoor
- Department of Electrical and Computer EngineeringUniversity of IowaIowa CityIowa
| | - Jeffrey D. Long
- Department of PsychiatryUniversity of IowaIowa CityIowa
- Department of BiostatisticsUniversity of IowaIowa CityIowa
| | | | - Spencer Lourens
- Department of BiostatisticsIndiana University School of MedicineIndianapolisIndiana
| | - Lauren J. O'Donnell
- Department of RadiologyBrigham and Women's Hospital Harvard Medical SchoolBostonMassachusetts
| | - Carl‐Fredrik Westin
- Department of RadiologyBrigham and Women's Hospital Harvard Medical SchoolBostonMassachusetts
| | - Yogesh Rathi
- Department of RadiologyBrigham and Women's Hospital Harvard Medical SchoolBostonMassachusetts
| | - Vincent Magnotta
- Department of RadiologyUniversity of IowaIowa CityIowa
- Department of PsychiatryUniversity of IowaIowa CityIowa
- Biomedical EngineeringUniversity of IowaIowa CityIowa
| | - Jane S. Paulsen
- Department of PsychiatryUniversity of IowaIowa CityIowa
- Department of Neurology, Carver College of MedicineUniversity of IowaIowa CityIowa
- Neuroscience, Carver College of MedicineUniversity of IowaIowa CityIowa
| | - Hans J. Johnson
- Department of Electrical and Computer EngineeringUniversity of IowaIowa CityIowa
- Department of PsychiatryUniversity of IowaIowa CityIowa
- Biomedical EngineeringUniversity of IowaIowa CityIowa
| |
Collapse
|
24
|
The Complexity of Clinical Huntington's Disease: Developments in Molecular Genetics, Neuropathology and Neuroimaging Biomarkers. ADVANCES IN NEUROBIOLOGY 2017; 15:129-161. [PMID: 28674980 DOI: 10.1007/978-3-319-57193-5_5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder characterised by extensive neuronal loss in the striatum and cerebral cortex, and a triad of clinical symptoms affecting motor, cognitive/behavioural and mood functioning. The mutation causing HD is an expansion of a CAG tract in exon 1 of the HTT gene. This chapter provides a multifaceted overview of the clinical complexity of HD. We explore recent directions in molecular genetics including the identification of loci that are genetic modifiers of HD that could potentially reveal therapeutic targets beyond the HTT gene transcript and protein. The variability of clinical symptomatology in HD is considered alongside recent findings of variability in cellular and neurochemical changes in the striatum and cerebral cortex in human brain. We review evidence from structural neuroimaging methods of progressive changes of striatum, cerebral cortex and white matter in pre-symptomatic and symptomatic HD, with a particular focus on the potential identification of neuroimaging biomarkers that could be used to test promising disease-specific and modifying treatments. Finally we provide an overview of completed clinical trials in HD and future therapeutic developments.
Collapse
|
25
|
Admixing of MPTP-Resistant and Susceptible Mice Strains Augments Nigrostriatal Neuronal Correlates to Resist MPTP-Induced Neurodegeneration. Mol Neurobiol 2016; 54:6148-6162. [PMID: 27704331 DOI: 10.1007/s12035-016-0158-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 09/22/2016] [Indexed: 12/31/2022]
Abstract
Disease genetics in admixed populations like Hispanic-Americans, African-Americans, etc. are gaining importance due to high disease burden in them. Furthermore, epidemiological studies conclusively prove ethnicity-based differential prevalence of Parkinson's disease (PD), since the American-Caucasians are more susceptible than Asian-Indians and Africans. Contradictorily, Anglo-Indians, an admixture of Europeans and Asian-Indians are five-times less susceptible than Indians. We evaluated the neural basis of this phenomenon using the cytomorphological features of susceptibility to nigrostriatal neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). The nigral dopaminergic neuronal numbers, their size and tyrosine hydroxylase (TH), PitX3 and Nurr1 expression were compared in MPTP-susceptible C57BL/6J mice, MPTP-resistant CD-1 mice and their crossbreds using stereology, morphometry and densitometry. Apoptotic index was evaluated by TUNEL-assay and caspase-3 expression. Striatal volume, TH and glial derived neurotrophic factor (GDNF) expression were studied. The normal CD-1 and crossbreds had significantly more, although smaller, nigral dopaminergic neurons than C57BL/6J, and a larger striatum. The crossbreds had higher TH, Nurr1 and PitX3 levels. MPTP administration caused loss of ~50-60 % nigral dopaminergic neurons in C57BL/6J and ~15 % in CD-1, but none in crossbreds. MPTP-induced cellular shrinkage in C57BL/6J was contrasted by nuclear enlargement without somal alterations in resistant strains. MPTP lowered the striatal TH and GDNF in C57BL/6J. Elevated striatal GDNF in CD-1 and crossbreds could be of compensatory nature and complemented the reduced nigral caspase-3 expression to attenuate and/or block apoptosis. Similar neural correlates of resilience are envisaged in the Anglo-Indian population. Thus, we present the core neuroanatomical features of resilience against PD and evidence for ethnicity-based differential prevalence.
Collapse
|
26
|
Mason SL, Barker RA. Novel targets for Huntington's disease: future prospects. Degener Neurol Neuromuscul Dis 2016; 6:25-36. [PMID: 30050366 PMCID: PMC6053088 DOI: 10.2147/dnnd.s83808] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Huntington's disease (HD) is an incurable, inherited, progressive, neurodegenerative disorder that is characterized by a triad of motor, cognitive, and psychiatric problems. Despite the noticeable increase in therapeutic trials in HD in the last 20 years, there have, to date, been very few significant advances. The main hope for new and emerging therapeutics for HD is to develop a neuroprotective compound capable of slowing down or even stopping the progression of the disease and ultimately prevent the subtle early signs from developing into manifest disease. Recently, there has been a noticeable shift away from symptomatic therapies in favor of more mechanistic-based interventions, a change driven by a better understanding of the pathogenesis of this disorder. In this review, we discuss the status of, and supporting evidence for, potential novel treatments of HD that are currently under development or have reached the level of early Phase I/II clinical trials.
Collapse
Affiliation(s)
| | - Roger A Barker
- John van Geest Centre for Brain Repair, .,Department of Clinical Neuroscience, University of Cambridge, Cambridge, UK
| |
Collapse
|
27
|
van Bergen JMG, Hua J, Unschuld PG, Lim IAL, Jones CK, Margolis RL, Ross CA, van Zijl PCM, Li X. Quantitative Susceptibility Mapping Suggests Altered Brain Iron in Premanifest Huntington Disease. AJNR Am J Neuroradiol 2016; 37:789-96. [PMID: 26680466 DOI: 10.3174/ajnr.a4617] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 09/22/2015] [Indexed: 12/27/2022]
Abstract
BACKGROUND AND PURPOSE In patients with premanifest (nonsymptomatic) and advanced Huntington disease, changes in brain iron levels in the basal ganglia have been previously reported, especially in the striatum. Quantitative susceptibility mapping by using MR phase imaging allows in vivo measurements of tissue magnetic susceptibility, which has been shown to correlate well with iron levels in brain gray matter and is believed to be more specific than other imaging-based iron measures. The purpose of this study was to investigate the use of magnetic susceptibility as a biomarker of disease progression. MATERIALS AND METHODS Fifteen subjects with premanifest Huntington disease and 16 age-matched healthy controls were scanned at 7T. Magnetic susceptibility, effective relaxation, and tissue volume in deep gray matter structures were quantified and compared with genetic and clinical measures. RESULTS Subjects with premanifest Huntington disease showed significantly higher susceptibility values in the caudate nucleus, putamen, and globus pallidus, indicating increased iron levels in these structures. Significant decreases in magnetic susceptibility were found in the substantia nigra and hippocampus. In addition, significant volume loss (atrophy) and an increase effective relaxation were observed in the caudate nucleus and putamen. Susceptibility values in the caudate nucleus and putamen were found to be inversely correlated with structure volumes and directly correlated with the genetic burdens, represented by cytosine-adenine-guanine repeat age-product-scaled scores. CONCLUSIONS The significant magnetic susceptibility differences between subjects with premanifest Huntington disease and controls and their correlation with genetic burden scores indicate the potential use of magnetic susceptibility as a biomarker of disease progression in premanifest Huntington disease.
Collapse
Affiliation(s)
- J M G van Bergen
- From the Departments of Radiology and Radiological Sciences (J.M.G.v.B., J.H., I.A.L.L., C.K.J., P.C.M.v.Z., X.L.) F.M. Kirby Research Center for Functional Brain Imaging (J.M.G.v.B., J.H., I.A.L.L., C.K.J., P.C.M.v.Z., X.L.), Kennedy Krieger Institute, Baltimore, Maryland
| | - J Hua
- From the Departments of Radiology and Radiological Sciences (J.M.G.v.B., J.H., I.A.L.L., C.K.J., P.C.M.v.Z., X.L.) F.M. Kirby Research Center for Functional Brain Imaging (J.M.G.v.B., J.H., I.A.L.L., C.K.J., P.C.M.v.Z., X.L.), Kennedy Krieger Institute, Baltimore, Maryland
| | - P G Unschuld
- Psychiatry and Behavioral Sciences (P.G.U., R.L.M., C.A.R.) Division of Psychiatry Research and Psychogeriatric Medicine (P.G.U.), University of Zurich, Zurich, Switzerland
| | - I A L Lim
- From the Departments of Radiology and Radiological Sciences (J.M.G.v.B., J.H., I.A.L.L., C.K.J., P.C.M.v.Z., X.L.) F.M. Kirby Research Center for Functional Brain Imaging (J.M.G.v.B., J.H., I.A.L.L., C.K.J., P.C.M.v.Z., X.L.), Kennedy Krieger Institute, Baltimore, Maryland
| | - C K Jones
- From the Departments of Radiology and Radiological Sciences (J.M.G.v.B., J.H., I.A.L.L., C.K.J., P.C.M.v.Z., X.L.) F.M. Kirby Research Center for Functional Brain Imaging (J.M.G.v.B., J.H., I.A.L.L., C.K.J., P.C.M.v.Z., X.L.), Kennedy Krieger Institute, Baltimore, Maryland
| | - R L Margolis
- Psychiatry and Behavioral Sciences (P.G.U., R.L.M., C.A.R.) Neurology (R.L.M., C.A.R.), Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - C A Ross
- Psychiatry and Behavioral Sciences (P.G.U., R.L.M., C.A.R.) Neurology (R.L.M., C.A.R.), Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - P C M van Zijl
- From the Departments of Radiology and Radiological Sciences (J.M.G.v.B., J.H., I.A.L.L., C.K.J., P.C.M.v.Z., X.L.) F.M. Kirby Research Center for Functional Brain Imaging (J.M.G.v.B., J.H., I.A.L.L., C.K.J., P.C.M.v.Z., X.L.), Kennedy Krieger Institute, Baltimore, Maryland
| | - X Li
- From the Departments of Radiology and Radiological Sciences (J.M.G.v.B., J.H., I.A.L.L., C.K.J., P.C.M.v.Z., X.L.) F.M. Kirby Research Center for Functional Brain Imaging (J.M.G.v.B., J.H., I.A.L.L., C.K.J., P.C.M.v.Z., X.L.), Kennedy Krieger Institute, Baltimore, Maryland
| |
Collapse
|
28
|
TAKAGI Y. History of Neural Stem Cell Research and Its Clinical Application. Neurol Med Chir (Tokyo) 2016; 56:110-24. [PMID: 26888043 PMCID: PMC4791305 DOI: 10.2176/nmc.ra.2015-0340] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 01/15/2016] [Indexed: 12/11/2022] Open
Abstract
"Once development was ended…in the adult centers, the nerve paths are something fixed and immutable. Everything may die, nothing may be regenerated," wrote Santiago Ramón y Cajal, a Spanish neuroanatomist and Nobel Prize winner and the father of modern neuroscience. This statement was the central dogma in neuroscience for a long time. However, in the 1960s, neural stem cells (NSCs) were discovered. Since then, our knowledge about NSCs has continued to grow. This review focuses on our current knowledge about NSCs and their surrounding microenvironment. In addition, the clinical application of NSCs for the treatment of various central nervous system diseases is also summarized.
Collapse
Affiliation(s)
- Yasushi TAKAGI
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Sakyo, Kyoto
| |
Collapse
|
29
|
Robins Wahlin TB, Luszcz MA, Wahlin Å, Byrne GJ. Non-Verbal and Verbal Fluency in Prodromal Huntington's Disease. Dement Geriatr Cogn Dis Extra 2015; 5:517-29. [PMID: 26955384 PMCID: PMC4777933 DOI: 10.1159/000441942] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND This study examines non-verbal (design) and verbal (phonemic and semantic) fluency in prodromal Huntington's disease (HD). An accumulating body of research indicates subtle deficits in cognitive functioning among prodromal mutation carriers for HD. METHODS Performance was compared between 32 mutation carriers and 38 non-carriers in order to examine the magnitude of impairment across fluency tasks. The predicted years to onset (PYTO) in mutation carriers was calculated by a regression equation and used to divide the group according to whether onset was predicted as less than 12.75 years (HD+CLOSE; n = 16) or greater than 12.75 years (HD+DISTANT; n = 16). RESULTS The results indicate that both non-verbal and verbal fluency is sensitive to subtle impairment in prodromal HD. HD+CLOSE group produced fewer items in all assessed fluency tasks compared to non-carriers. HD+DISTANT produced fewer drawings than non-carriers in the non-verbal task. PYTO correlated significantly with all measures of non-verbal and verbal fluency. CONCLUSION The pattern of results indicates that subtle cognitive deficits exist in prodromal HD, and that less structured tasks with high executive demands are the most sensitive in detecting divergence from the normal range of functioning. These selective impairments can be attributed to the early involvement of frontostriatal circuitry and frontal lobes.
Collapse
Affiliation(s)
- Tarja-Brita Robins Wahlin
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- School of Medicine, University of Queensland, Brisbane, Qld., Australia
| | - Mary A. Luszcz
- School of Psychology, Flinders University, Adelaide, S.A., Australia
| | - Åke Wahlin
- School of Health and Welfare, Jönköping University, Jönköping, Sweden
| | - Gerard J. Byrne
- School of Medicine, University of Queensland, Brisbane, Qld., Australia
| |
Collapse
|
30
|
Hsiao HY, Chen YC, Huang CH, Chen CC, Hsu YH, Chen HM, Chiu FL, Kuo HC, Chang C, Chern Y. Aberrant astrocytes impair vascular reactivity in Huntington disease. Ann Neurol 2015; 78:178-92. [DOI: 10.1002/ana.24428] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 03/02/2015] [Accepted: 04/07/2015] [Indexed: 01/08/2023]
Affiliation(s)
- Han-Yun Hsiao
- Institute of Biomedical Sciences, Academia Sinica; Taipei Taiwan
- Institute of Neuroscience, National Yang-Ming University; Taipei Taiwan
| | - Yu-Chen Chen
- Institute of Biomedical Sciences, Academia Sinica; Taipei Taiwan
- Institute of Neuroscience, National Yang-Ming University; Taipei Taiwan
| | - Chien-Hsiang Huang
- Institute of Biomedical Sciences, Academia Sinica; Taipei Taiwan
- Institute of Biomedical Engineering, National Taiwan University; Taipei Taiwan
| | - Chiao-Chi Chen
- Institute of Biomedical Sciences, Academia Sinica; Taipei Taiwan
| | - Yi-Hua Hsu
- Institute of Biomedical Sciences, Academia Sinica; Taipei Taiwan
| | - Hui-Mei Chen
- Institute of Biomedical Sciences, Academia Sinica; Taipei Taiwan
| | - Feng-Lan Chiu
- Institute of Cellular and Organismic Biology, Academia Sinica; Taipei Taiwan
| | - Hung-Chih Kuo
- Institute of Cellular and Organismic Biology, Academia Sinica; Taipei Taiwan
| | - Chen Chang
- Institute of Biomedical Sciences, Academia Sinica; Taipei Taiwan
| | - Yijuang Chern
- Institute of Biomedical Sciences, Academia Sinica; Taipei Taiwan
- Institute of Neuroscience, National Yang-Ming University; Taipei Taiwan
| |
Collapse
|
31
|
Drouin-Ouellet J, Sawiak SJ, Cisbani G, Lagacé M, Kuan WL, Saint-Pierre M, Dury RJ, Alata W, St-Amour I, Mason SL, Calon F, Lacroix S, Gowland PA, Francis ST, Barker RA, Cicchetti F. Cerebrovascular and blood-brain barrier impairments in Huntington's disease: Potential implications for its pathophysiology. Ann Neurol 2015; 78:160-77. [PMID: 25866151 DOI: 10.1002/ana.24406] [Citation(s) in RCA: 215] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Revised: 03/06/2015] [Accepted: 03/06/2015] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Although the underlying cause of Huntington's disease (HD) is well established, the actual pathophysiological processes involved remain to be fully elucidated. In other proteinopathies such as Alzheimer's and Parkinson's diseases, there is evidence for impairments of the cerebral vasculature as well as the blood-brain barrier (BBB), which have been suggested to contribute to their pathophysiology. We investigated whether similar changes are also present in HD. METHODS We used 3- and 7-Tesla magnetic resonance imaging as well as postmortem tissue analyses to assess blood vessel impairments in HD patients. Our findings were further investigated in the R6/2 mouse model using in situ cerebral perfusion, histological analysis, Western blotting, as well as transmission and scanning electron microscopy. RESULTS We found mutant huntingtin protein (mHtt) aggregates to be present in all major components of the neurovascular unit of both R6/2 mice and HD patients. This was accompanied by an increase in blood vessel density, a reduction in blood vessel diameter, as well as BBB leakage in the striatum of R6/2 mice, which correlated with a reduced expression of tight junction-associated proteins and increased numbers of transcytotic vesicles, which occasionally contained mHtt aggregates. We confirmed the existence of similar vascular and BBB changes in HD patients. INTERPRETATION Taken together, our results provide evidence for alterations in the cerebral vasculature in HD leading to BBB leakage, both in the R6/2 mouse model and in HD patients, a phenomenon that may, in turn, have important pathophysiological implications.
Collapse
Affiliation(s)
- Janelle Drouin-Ouellet
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, United Kingdom
| | - Stephen J Sawiak
- Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Giulia Cisbani
- Centre de recherche du CHU de Québec (CHUQ), Québec, Québec, Canada
| | - Marie Lagacé
- Centre de recherche du CHU de Québec (CHUQ), Québec, Québec, Canada
| | - Wei-Li Kuan
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, United Kingdom
| | | | - Richard J Dury
- Sir Peter Mansfield Magnetic Resonance Centre, School of Physics and Astronomy, University of Nottingham, Nottingham, United Kingdom
| | - Wael Alata
- Centre de recherche du CHU de Québec (CHUQ), Québec, Québec, Canada
| | | | - Sarah L Mason
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, United Kingdom
| | - Frédéric Calon
- Centre de recherche du CHU de Québec (CHUQ), Québec, Québec, Canada.,Faculté de Pharmacie, Université Laval, Québec, Québec, Canada
| | - Steve Lacroix
- Centre de recherche du CHU de Québec (CHUQ), Québec, Québec, Canada.,Département de Médecine Moléculaire, Université Laval, Québec, Québec, Canada
| | - Penny A Gowland
- Sir Peter Mansfield Magnetic Resonance Centre, School of Physics and Astronomy, University of Nottingham, Nottingham, United Kingdom
| | - Susan T Francis
- Sir Peter Mansfield Magnetic Resonance Centre, School of Physics and Astronomy, University of Nottingham, Nottingham, United Kingdom
| | - Roger A Barker
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, United Kingdom
| | - Francesca Cicchetti
- Centre de recherche du CHU de Québec (CHUQ), Québec, Québec, Canada.,Département de Psychiatrie & Neurosciences, Université Laval, Québec, Québec, Canada
| |
Collapse
|
32
|
Progress in Huntington’s disease: the search for markers of disease onset and progression. J Neurol 2015; 262:1990-5. [DOI: 10.1007/s00415-015-7700-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 03/03/2015] [Accepted: 03/04/2015] [Indexed: 11/26/2022]
|
33
|
Chang KH, Wu YR, Chen YC, Chen CM. Plasma inflammatory biomarkers for Huntington's disease patients and mouse model. Brain Behav Immun 2015; 44:121-7. [PMID: 25266150 DOI: 10.1016/j.bbi.2014.09.011] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 09/19/2014] [Accepted: 09/20/2014] [Indexed: 12/19/2022] Open
Abstract
Huntington's disease (HD), caused by expanded CAG repeats encoding a polyglutamine tract in the huntingtin (HTT) protein, presents with a predominant degeneration of neurons in the striatum and cortex. Lines of evidence have observed neuroinflammation, particularly microglial activation, is involved in the pathogenesis of HD. Given that HTT is also expressed in peripheral inflammatory cells, it is possible that inflammatory changes detected in peripheral plasma may be biologically relevant and parallel the neuroinflammatory process of HD patients. By examining the expression levels of 13 microglia-derived inflammatory markers in the plasma of 5 PreHD carriers, 15 HD patients and 16 healthy controls, we found plasma levels of IL-6, MMP-9, VEGF and TGF-β1 were significantly increased in HD patients when compared with the controls, while plasma level of IL-18 were significantly reduced in HD patients compared with controls. Plasma level of IL-6 was reversely correlated with the UHDRS independence scale and functional capacity. To understand the temporal correlation between these inflammatory markers and HD progression, their levels were further tested in plasma from R6/2 mouse HD model at different ages. In rotarod test, R6/2 HD mice started to manifest HD phenotype at 7.5 weeks of age. Higher plasma VEGF levels of R6/2 mice than those of age-matched wild-type (WT) littermates were noted from 7 (presymptomatic stage) to 13 weeks of age (late symptomatic stage). The plasma IL-6 levels of R6/2 mice were higher than those of the WT littermates from 9 (early symptomatic stage) to 13 weeks of age. R6/2 mice demonstrated higher MMP-9 and TGF-β1 levels than their WT littermates from 11 (middle symptomatic stage) to 13 weeks of age. In contrast, the plasma IL-18 level was lower than those in WT littermates since 11 weeks of age. These altered expressions of inflammatory markers may serve as the potential biomarkers for HD onset and progression. Specific inhibition/activation of these inflammatory markers may be the targets of HD drug development.
Collapse
Affiliation(s)
- Kuo-Hsuan Chang
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Yih-Ru Wu
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Yi-Chun Chen
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Chiung-Mei Chen
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| |
Collapse
|
34
|
Kargieman L, Herrera E, Baez S, García AM, Dottori M, Gelormini C, Manes F, Gershanik O, Ibáñez A. Motor-Language Coupling in Huntington's Disease Families. Front Aging Neurosci 2014; 6:122. [PMID: 24971062 PMCID: PMC4054328 DOI: 10.3389/fnagi.2014.00122] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 05/27/2014] [Indexed: 11/24/2022] Open
Abstract
Traditionally, Huntington’s disease (HD) has been known as a movement disorder, characterized by motor, psychiatric, and cognitive impairments. Recent studies have shown that motor and action–language processes are neurally associated. The cognitive mechanisms underlying this interaction have been investigated through the action compatibility effect (ACE) paradigm, which induces a contextual coupling of ongoing motor actions and verbal processing. The present study is the first to use the ACE paradigm to evaluate action–word processing in HD patients (HDP) and their families. Specifically, we tested three groups: HDP, healthy first-degree relatives (HDR), and non-relative healthy controls. The results showed that ACE was abolished in HDP as well as HDR, but not in controls. Furthermore, we found that the processing deficits were primarily linguistic, given that they did not correlate executive function measurements. Our overall results underscore the role of cortico-basal ganglia circuits in action–word processing and indicate that the ACE task is a sensitive and robust early biomarker of HD and familial vulnerability.
Collapse
Affiliation(s)
- Lucila Kargieman
- Laboratory of Experimental Psychology and Neuroscience (LPEN), Institute of Cognitive Neurology (INECO), Favaloro University , Buenos Aires , Argentina ; National Scientific and Technical Research Council (CONICET) , Buenos Aires , Argentina ; UDP-INECO Foundation Core on Neuroscience (UIFCoN), Diego Portales University , Santiago , Chile
| | - Eduar Herrera
- Laboratory of Experimental Psychology and Neuroscience (LPEN), Institute of Cognitive Neurology (INECO), Favaloro University , Buenos Aires , Argentina ; National Scientific and Technical Research Council (CONICET) , Buenos Aires , Argentina ; Universidad Autónoma del Caribe , Barranquilla , Colombia
| | - Sandra Baez
- Laboratory of Experimental Psychology and Neuroscience (LPEN), Institute of Cognitive Neurology (INECO), Favaloro University , Buenos Aires , Argentina ; National Scientific and Technical Research Council (CONICET) , Buenos Aires , Argentina ; UDP-INECO Foundation Core on Neuroscience (UIFCoN), Diego Portales University , Santiago , Chile
| | - Adolfo M García
- Laboratory of Experimental Psychology and Neuroscience (LPEN), Institute of Cognitive Neurology (INECO), Favaloro University , Buenos Aires , Argentina ; National Scientific and Technical Research Council (CONICET) , Buenos Aires , Argentina ; UDP-INECO Foundation Core on Neuroscience (UIFCoN), Diego Portales University , Santiago , Chile ; School of Languages, National University of Córdoba (UNC) , Córdoba , Argentina
| | - Martin Dottori
- Laboratory of Experimental Psychology and Neuroscience (LPEN), Institute of Cognitive Neurology (INECO), Favaloro University , Buenos Aires , Argentina
| | - Carlos Gelormini
- Laboratory of Experimental Psychology and Neuroscience (LPEN), Institute of Cognitive Neurology (INECO), Favaloro University , Buenos Aires , Argentina
| | - Facundo Manes
- Laboratory of Experimental Psychology and Neuroscience (LPEN), Institute of Cognitive Neurology (INECO), Favaloro University , Buenos Aires , Argentina ; National Scientific and Technical Research Council (CONICET) , Buenos Aires , Argentina ; Australian Research Council (ARC) Centre of Excellence in Cognition and its Disorders , Sydney, NSW , Australia
| | - Oscar Gershanik
- Laboratory of Experimental Psychology and Neuroscience (LPEN), Institute of Cognitive Neurology (INECO), Favaloro University , Buenos Aires , Argentina ; National Scientific and Technical Research Council (CONICET) , Buenos Aires , Argentina
| | - Agustín Ibáñez
- Laboratory of Experimental Psychology and Neuroscience (LPEN), Institute of Cognitive Neurology (INECO), Favaloro University , Buenos Aires , Argentina ; National Scientific and Technical Research Council (CONICET) , Buenos Aires , Argentina ; UDP-INECO Foundation Core on Neuroscience (UIFCoN), Diego Portales University , Santiago , Chile ; Universidad Autónoma del Caribe , Barranquilla , Colombia
| |
Collapse
|
35
|
Chandra A, Johri A, Beal MF. Prospects for neuroprotective therapies in prodromal Huntington's disease. Mov Disord 2014; 29:285-93. [PMID: 24573776 DOI: 10.1002/mds.25835] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 01/14/2014] [Accepted: 01/16/2014] [Indexed: 12/13/2022] Open
Abstract
Huntington's disease (HD) is a prototypical dominantly inherited neurodegenerative disorder characterized by progressive cognitive deterioration, psychiatric disturbances, and a movement disorder. The genetic cause of the illness is a CAG repeat expansion in the huntingtin gene, which leads to a polyglutamine expansion in the huntingtin protein. The exact mechanism by which mutant huntingtin causes HD is unknown, but it causes abnormalities in gene transcription as well as both mitochondrial dysfunction and oxidative damage. Because the penetrance of HD is complete with CAG repeats greater than 39, patients can be diagnosed well before disease onset with genetic testing. Longitudinal studies of HD patients before disease onset have shown that subtle cognitive and motor deficits occur as much as 10 years before onset, as do reductions in glucose utilization and striatal atrophy. An increase in inflammation, as shown by elevated interleukin-6, occurs approximately 15 years before onset. Detection of these abnormalities may be useful in defining an optimal time for disease intervention to try to slow or halt the degenerative process. Although reducing gene expression with small interfering RNA or short hairpin RNA is an attractive approach, other approaches targeting energy metabolism, inflammation, and oxidative damage may be more easily and rapidly moved into the clinic. The recent PREQUEL study of coenzyme Q10 in presymptomatic gene carriers showed the feasibility of carrying out clinical trials to slow or halt onset of HD. We review both the earliest detectable clinical and laboratory manifestations of HD, as well as potential neuroprotective therapies that could be utilized in presymptomatic HD.
Collapse
Affiliation(s)
- Abhishek Chandra
- Brain and Mind Research Institute, Weill Medical College of Cornell University, New York Presbyterian Hospital, New York, New York, USA
| | | | | |
Collapse
|
36
|
Kim SU, Lee HJ, Kim YB. Neural stem cell-based treatment for neurodegenerative diseases. Neuropathology 2013; 33:491-504. [PMID: 23384285 DOI: 10.1111/neup.12020] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 12/27/2012] [Accepted: 12/28/2012] [Indexed: 12/11/2022]
Abstract
Human neurodegenerative diseases such as Parkinson's disease (PD), Huntington's disease (HD), amyotrophic lateral sclerosis (ALS) and Alzheimer's disease (AD) are caused by a loss of neurons and glia in the brain or spinal cord. Neurons and glial cells have successfully been generated from stem cells such as embryonic stem cells (ESCs), mesenchymal stem cells (MSCs) and neural stem cells (NSCs), and stem cell-based cell therapies for neurodegenerative diseases have been developed. A recent advance in generation of a new class of pluripotent stem cells, induced pluripotent stem cells (iPSCs), derived from patients' own skin fibroblasts, opens doors for a totally new field of personalized medicine. Transplantation of NSCs, neurons or glia generated from stem cells in animal models of neurodegenerative diseases, including PD, HD, ALS and AD, demonstrates clinical improvement and also life extension of these animals. Additional therapeutic benefits in these animals can be provided by stem cell-mediated gene transfer of therapeutic genes such as neurotrophic factors and enzymes. Although further research is still needed, cell and gene therapy based on stem cells, particularly using neurons and glia derived from iPSCs, ESCs or NSCs, will become a routine treatment for patients suffering from neurodegenerative diseases and also stroke and spinal cord injury.
Collapse
Affiliation(s)
- Seung U Kim
- Medical Research Institute, Chung-Ang University College of Medicine, Seoul, Korea; Division of Neurology, Department of Medicine, UBC Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | |
Collapse
|
37
|
Lin CY, Hsu YH, Lin MH, Yang TH, Chen HM, Chen YC, Hsiao HY, Chen CC, Chern Y, Chang C. Neurovascular abnormalities in humans and mice with Huntington's disease. Exp Neurol 2013; 250:20-30. [PMID: 24036415 DOI: 10.1016/j.expneurol.2013.08.019] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2013] [Revised: 08/04/2013] [Accepted: 08/30/2013] [Indexed: 01/27/2023]
Abstract
Cerebral microvascular aberrations have recently become recognized as a source of pathologies in neurodegenerative disorders, but this concept has not been fully examined with respect to Huntington's disease (HD). A novel in vivo technique, three-dimensional microscopic magnetic resonance angiography (μMRA), allows visualization of the neurovascular system in exquisite detail and provides quantitative structural and functional information. This technique was applied in the present study, in parallel with immunohistological analysis and behavioral assessment, to a well-characterized mouse model of HD (R6/2). Dynamic contrast-enhanced magnetic resonance imaging was used to examine the integrity of the blood-brain barrier (BBB). The μMRA findings revealed an increase in vessel volume fraction and cerebral blood volume in the brains of R6/2 mice at the age of 7weeks when no apparent motor dysfunction was detected. Collagen IV immunostaining disclosed an enhancement in vessel density, but not in vessel size of the microvasculature in the mouse HD brain. This change in neurovasculature worsened with disease progression, with no apparent disruption in the BBB. Most importantly, immunohistological assays of human tissues revealed that the vessel densities in the cortex, caudate/putamen, and substantia nigra were higher in HD patients than in non-HD human subjects. The early onset of such vessel aberrations could be used as a biomarker for the early diagnosis of HD.
Collapse
Affiliation(s)
- Chien-Yuan Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan; Functional and Micro-Magnetic Resonance Imaging Core Facility, Academia Sinica, Taipei 11529, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Hua J, Unschuld PG, Margolis RL, van Zijl PCM, Ross CA. Elevated arteriolar cerebral blood volume in prodromal Huntington's disease. Mov Disord 2013; 29:396-401. [PMID: 23847161 DOI: 10.1002/mds.25591] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 05/24/2013] [Accepted: 05/29/2013] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Neurovascular alterations have been implicated in the pathophysiology of Huntington's disease (HD). Because arterioles are most responsive to metabolic alterations, arteriolar cerebral blood volume (CBVa) is an important indicator of cerebrovascular regulation. The objective of this pilot study was to investigate potential neurovascular (CBVa ) abnormality in prodromal-HD patients and compare it with the widely used imaging marker: brain atrophy. METHODS CBVa and brain volumes were measured with ultra-high-field (7.0-Telsa) magnetic resonance imaging in seven prodromal-HD patients and nine age-matched controls. RESULTS Cortical CBVa was elevated significantly in prodromal-HD patients compared with controls (relative difference, 38.5%; effect size, 1.48). Significant correlations were found between CBVa in the frontal cortex and genetic measures. By contrast, no significant brain atrophy was detected in the prodromal-HD patients. CONCLUSIONS CBVa may be abnormal in prodromal-HD, even before substantial brain atrophy occurs. Further investigation with a larger cohort and longitudinal follow-up is merited to determine whether CBVa could be used as a potential biomarker for clinical trials.
Collapse
Affiliation(s)
- Jun Hua
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Magnetic Resonance Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA
| | | | | | | | | |
Collapse
|
39
|
Matsui JT, Vaidya JG, Johnson HJ, Magnotta VA, Long JD, Mills JA, Lowe MJ, Sakaie KE, Rao SM, Smith MM, Paulsen JS. Diffusion weighted imaging of prefrontal cortex in prodromal Huntington's disease. Hum Brain Mapp 2013; 35:1562-73. [PMID: 23568433 DOI: 10.1002/hbm.22273] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Revised: 11/09/2012] [Accepted: 01/28/2013] [Indexed: 11/07/2022] Open
Abstract
Huntington's disease (HD) is a devastating neurodegenerative disease with no effective disease-modifying treatments. There is considerable interest in finding reliable indicators of disease progression to judge the efficacy of novel treatments that slow or stop disease onset before debilitating signs appear. Diffusion-weighted imaging (DWI) may provide a reliable marker of disease progression by characterizing diffusivity changes in white matter (WM) in individuals with prodromal HD. The prefrontal cortex (PFC) may play a role in HD progression due to its prominent striatal connections and documented role in executive function. This study uses DWI to characterize diffusivity in specific regions of PFC WM defined by FreeSurfer in 53 prodromal HD participants and 34 controls. Prodromal HD individuals were separated into three CAG-Age Product (CAP) groups (16 low, 22 medium, 15 high) that indexed baseline progression. Statistically significant increases in mean diffusivity (MD) and radial diffusivity (RD) among CAP groups relative to controls were seen in inferior and lateral PFC regions. For MD and RD, differences among controls and HD participants tracked with baseline disease progression. The smallest difference was for the low group and the largest for the high group. Significant correlations between Trail Making Test B (TMTB) and mean fractional anisotropy (FA) and/or RD paralleled group differences in mean MD and/or RD in several right hemisphere regions. The gradient of effects that tracked with CAP group suggests DWI may provide markers of disease progression in future longitudinal studies as increasing diffusivity abnormalities in the lateral PFC of prodromal HD individuals.
Collapse
Affiliation(s)
- Joy T Matsui
- Department of Psychiatry, The University of Iowa, Iowa City, Iowa; John A. Burns School of Medicine, The University of Hawaii, Honolulu, Hawaii
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Leoni V, Long JD, Mills JA, Di Donato S, Paulsen JS. Plasma 24S-hydroxycholesterol correlation with markers of Huntington disease progression. Neurobiol Dis 2013; 55:37-43. [PMID: 23557875 DOI: 10.1016/j.nbd.2013.03.013] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Revised: 02/15/2013] [Accepted: 03/14/2013] [Indexed: 10/27/2022] Open
Abstract
24S-hydroxycholesterol (24OHC) is involved in the conversion of excess cholesterol in the brain, and its level in plasma is related to the number of metabolically active neuronal cells. Previous research suggests that plasma 24OHC is substantially reduced in the presence of neurodegenerative disease. Huntington disease (HD) is an inherited autosomal dominant neurodegenerative disorder caused by a cytosine-adenine-guanine (CAG) triplet repeat expansion in the coding region of the huntingtin (HTT) gene. The current study focused on the relative importance of 24OHC as a marker of HD progression. Using mass spectrometry methods, we examined plasma 24OHC levels in three groups of gene-expanded individuals (Low, Medium, High) characterized by their progression at entry into the parent PREDICT-HD study, along with a group of non-gene-expanded controls (total N=150). In addition, the correlation of 24OHC with a number of motor, cognitive, and imagining markers was examined, and effect sizes for group differences among the markers were computed for comparison with 24OHC. Results show a progression gradient as 24OHC levels decreased as the progression group increased (Low to High). The effect size of group differences for 24OHC was larger than all the other variables, except striatal volume. 24OHC was significantly correlated with many of the other key variables. The results are interpreted in terms of cholesterol synthesis and neuronal degeneration. This study provides evidence that 24OHC is a relatively important marker of HD progression.
Collapse
Affiliation(s)
- Valerio Leoni
- Laboratory of Clinical Pathology and Medical Genetics, Fondazione IRCCS Institute of Neurology Carlo Besta, Milan, Italy
| | | | | | | | | | | |
Collapse
|
41
|
Automated differentiation of pre-diagnosis Huntington's disease from healthy control individuals based on quadratic discriminant analysis of the basal ganglia: The IMAGE-HD study. Neurobiol Dis 2013; 51:82-92. [DOI: 10.1016/j.nbd.2012.10.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Revised: 08/31/2012] [Accepted: 10/03/2012] [Indexed: 01/18/2023] Open
|
42
|
Georgiou-Karistianis N, Scahill R, Tabrizi SJ, Squitieri F, Aylward E. Structural MRI in Huntington's disease and recommendations for its potential use in clinical trials. Neurosci Biobehav Rev 2013; 37:480-90. [PMID: 23376047 DOI: 10.1016/j.neubiorev.2013.01.022] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Revised: 01/02/2013] [Accepted: 01/22/2013] [Indexed: 01/18/2023]
Abstract
Huntington's disease (HD) results in progressive impairment of motor and cognitive function and neuropsychiatric disturbance. There are no disease-modifying treatments available, but HD research is entering a critical phase where promising disease-specific therapies are on the horizon. Thus, a pressing need exists for biomarkers capable of monitoring progression and ultimately determining drug efficacy. Neuroimaging provides a powerful tool for assessing disease progression. However, in order to be accepted as biomarkers for clinical trials, imaging measures must be reproducible, robust to scanner differences, sensitive to disease-related change and demonstrate a relationship to clinically meaningful measures. We provide a review of the current structural imaging literature in HD and highlight inconsistencies between studies. We make recommendations for the standardisation of reporting for future studies, such as appropriate cohort characterisation and documentation of methodologies to facilitate comparisons and inform trial design. We also argue for an intensified effort to consider issues highlighted here so that we have the best chance of assessing the efficacy of the therapeutic benefit in forestalling this devastating disease.
Collapse
|
43
|
Mickes L, Wixted JT, Peavy GM, Jacobson MW, Goldstein JL, Corey-Bloom J. Difficulty modifying a sustained motor response in prodromal Huntington's disease. J Clin Exp Neuropsychol 2013. [DOI: 10.1080/13803395.2012.742039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
44
|
Kim SU. Regenerative Medicine in the Central Nervous System: Stem Cell-Based Cell- and Gene-Therapy. Regen Med 2013. [DOI: 10.1007/978-94-007-5690-8_27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
45
|
Characterization of neurophysiological and behavioral changes, MRI brain volumetry and 1H MRS in zQ175 knock-in mouse model of Huntington's disease. PLoS One 2012; 7:e50717. [PMID: 23284644 PMCID: PMC3527436 DOI: 10.1371/journal.pone.0050717] [Citation(s) in RCA: 195] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Accepted: 10/23/2012] [Indexed: 11/22/2022] Open
Abstract
Huntington's disease (HD) is an autosomal neurodegenerative disorder, characterized by severe behavioral, cognitive, and motor deficits. Since the discovery of the huntingtin gene (HTT) mutation that causes the disease, several mouse lines have been developed using different gene constructs of Htt. Recently, a new model, the zQ175 knock-in (KI) mouse, was developed (see description by Menalled et al, [1]) in an attempt to have the Htt gene in a context and causing a phenotype that more closely mimics HD in humans. Here we confirm the behavioral phenotypes reported by Menalled et al [1], and extend the characterization to include brain volumetry, striatal metabolite concentration, and early neurophysiological changes. The overall reproducibility of the behavioral phenotype across the two independent laboratories demonstrates the utility of this new model. Further, important features reminiscent of human HD pathology are observed in zQ175 mice: compared to wild-type neurons, electrophysiological recordings from acute brain slices reveal that medium spiny neurons from zQ175 mice display a progressive hyperexcitability; glutamatergic transmission in the striatum is severely attenuated; decreased striatal and cortical volumes from 3 and 4 months of age in homo- and heterozygous mice, respectively, with whole brain volumes only decreased in homozygotes. MR spectroscopy reveals decreased concentrations of N-acetylaspartate and increased concentrations of glutamine, taurine and creatine + phosphocreatine in the striatum of 12-month old homozygotes, the latter also measured in 12-month-old heterozygotes. Motor, behavioral, and cognitive deficits in homozygotes occur concurrently with the structural and metabolic changes observed. In sum, the zQ175 KI model has robust behavioral, electrophysiological, and histopathological features that may be valuable in both furthering our understanding of HD-like pathophyisology and the evaluation of potential therapeutic strategies to slow the progression of disease.
Collapse
|
46
|
Increased brain tissue sodium concentration in Huntington's Disease — A sodium imaging study at 4T. Neuroimage 2012; 63:517-24. [DOI: 10.1016/j.neuroimage.2012.07.009] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Revised: 07/05/2012] [Accepted: 07/06/2012] [Indexed: 02/05/2023] Open
|
47
|
Guo Z, Rudow G, Pletnikova O, Codispoti KE, Orr BA, Crain BJ, Duan W, Margolis RL, Rosenblatt A, Ross CA, Troncoso JC. Striatal neuronal loss correlates with clinical motor impairment in Huntington's disease. Mov Disord 2012; 27:1379-86. [PMID: 22975850 DOI: 10.1002/mds.25159] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Revised: 07/22/2012] [Accepted: 07/27/2012] [Indexed: 11/06/2022] Open
Abstract
Huntington's disease (HD) is characterized clinically by chorea, motor impairment, psychiatric manifestations, and dementia. Atrophy of the striatum is the neuropathological hallmark of HD, and previous studies have suggested that striatal atrophy correlates more closely with motor impairment than with chorea. Motor impairment, as measured by motor impairment score, correlates with functional disability in HD patients, but chorea does not. In this study, we investigated the relation between neuronal loss and these motor features. We conducted neuropathological and stereologic assessments of neurons in putamen and subthalamic nuclei in HD patients and age-matched controls. In putamen, we estimated the total number and volume of medium spiny neurons labeled with dopamine- and cAMP-regulated phosphoprotein 32 kDa (DARPP-32). In subthalamic nuclei, we estimated the total number of neurons on hematoxylin & eosin/luxol fast blue stains. In putamen of HD, immunohistochemistry showed DARPP-32 neuronal atrophy with extensive disruption of neurites and neuropil; stereologic studies found significant decreases in both the number and size of DARPP-32 neurons; we also detected a significant reduction of overall putamen volume in HD patients, compared to controls. In subthalamic nuclei, there was a mild, but significant, neuronal loss in the HD group. The loss of neurons in putamen and subthalamic nuclei as well as putaminal atrophy were significantly correlated with severity of motor impairment, but not with chorea. Our findings suggest that neuronal loss and atrophy in striatum and neuronal loss in subthalamic nuclei contribute specifically to the motor impairment of HD, but not to chorea.
Collapse
Affiliation(s)
- Zhihong Guo
- Division of Neuropathology, Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Dogan I, Eickhoff SB, Schulz JB, Shah NJ, Laird AR, Fox PT, Reetz K. Consistent neurodegeneration and its association with clinical progression in Huntington's disease: a coordinate-based meta-analysis. NEURODEGENER DIS 2012; 12:23-35. [PMID: 22922585 DOI: 10.1159/000339528] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Accepted: 05/10/2012] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND The neuropathological hallmark of Huntington's disease (HD) is progressive striatal loss starting several years prior to clinical onset. In the past decade, whole-brain magnetic resonance imaging (MRI) studies have provided accumulating evidence for widely distributed cortical and subcortical atrophy in the early course of the disease. OBJECTIVE In order to synthesize current morphometric MRI findings and to investigate the impact of clinical and genetic features on structural changes, we performed a coordinate-based meta-analysis of voxel-based morphometry (VBM) studies in HD. METHODS Twenty HD samples derived from 17 studies were integrated in the analysis comparing a total of 685 HD mutation carriers [345 presymptomatic (pre-HD) and 340 symptomatic (symp-HD) subjects] and 507 controls. Convergent findings across studies were delineated using the anatomical likelihood estimation approach. Effects of genetic and clinical parameters on the likelihood of observing VBM findings were calculated by means of correlation analyses. RESULTS Pre-HD studies featured convergent evidence for neurodegeneration in the basal ganglia, amygdala, thalamus, insula and occipital regions. In symp-HD, cerebral atrophy was more pronounced and spread to cortical regions (i.e., inferior frontal, premotor, sensorimotor, midcingulate, frontoparietal and temporoparietal cortices). Higher cytosine-adenosine-guanosine repeats were associated with striatal degeneration, while parameters of disease progression and motor impairment additionally correlated with cortical atrophy, especially in sensorimotor areas. CONCLUSION This first quantitative meta-analysis in HD demonstrates the extent of striatal atrophy and further consistent extrastriatal degeneration before clinical conversion. Sensorimotor areas seem to be core regions affected in symp-HD and, along with widespread cortical atrophy, may account for the clinical heterogeneity in HD.
Collapse
Affiliation(s)
- Imis Dogan
- Department of Neurology, RWTH Aachen University, Aachen.,Institute of Neuroscience and Medicine, Research Center Jülich GmbH, Jülich.,Translational Brain Medicine, Jülich Aachen Research Alliance, Jülich
| | - Simon B Eickhoff
- Institute of Neuroscience and Medicine, Research Center Jülich GmbH, Jülich.,Institute of Clinical Neuroscience and Medical Psychology, Heinrich Heine University, Düsseldorf , Germany
| | - Jörg B Schulz
- Department of Neurology, RWTH Aachen University, Aachen.,Translational Brain Medicine, Jülich Aachen Research Alliance, Jülich
| | - N Jon Shah
- Department of Neurology, RWTH Aachen University, Aachen.,Institute of Neuroscience and Medicine, Research Center Jülich GmbH, Jülich.,Translational Brain Medicine, Jülich Aachen Research Alliance, Jülich
| | - Angela R Laird
- Research Imaging Center, University of Texas Health Science Center San Antonio, San Antonio, Tex. , USA
| | - Peter T Fox
- Research Imaging Center, University of Texas Health Science Center San Antonio, San Antonio, Tex. , USA
| | - Kathrin Reetz
- Department of Neurology, RWTH Aachen University, Aachen.,Institute of Neuroscience and Medicine, Research Center Jülich GmbH, Jülich.,Translational Brain Medicine, Jülich Aachen Research Alliance, Jülich
| |
Collapse
|
49
|
Abstract
Huntington's disease (HD) is a genetic neurodegenerative disorder affecting the basal ganglia. These subcortical structures are particularly important for motor functions, response selection and implicit learning. In the current study, we have assessed prodromal and symptomatic HD participants with an implicit contextual learning task that is not based on motor learning, but on a purely visual implicit learning mechanism. We used an implicit contextual learning task in which subjects need to locate a target among several distractors. In half of the trials, the positions of the distractors and target stimuli were repeated. By memorizing this contextual information, attention can be guided faster to the target stimulus. Nine symptomatic HD participants, 16 prodromal HD participants and 22 control subjects were included. We found that the responses of the control subjects were faster for the repeated trials than for the new trials, indicating that their visual search was facilitated when repeated contextual information was present. In contrast, no difference in response times between the repeated and new trials was found for the symptomatic and prodromal HD participants. The results of the current study indicate that both prodromal and symptomatic HD participants are impaired on an implicit contextual learning task.
Collapse
|
50
|
Wolf RC, Sambataro F, Vasic N, Wolf ND, Thomann PA, Saft C, Landwehrmeyer GB, Orth M. Default-mode network changes in preclinical Huntington's disease. Exp Neurol 2012; 237:191-8. [PMID: 22742947 DOI: 10.1016/j.expneurol.2012.06.014] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Accepted: 06/16/2012] [Indexed: 10/28/2022]
Abstract
The default-mode network (DMN) refers to as a set of brain regions which are active when the brain does not engage in a cognitive task and which are deactivated with task-related cognitive effort. Altered function of the DMN has been associated with a decline of cognition in several neurodegenerative diseases and related at-risk conditions. In Huntington's disease, an autosomal dominant inherited neurodegenerative disorder, several studies so far have shown abnormal task-related brain activation patterns even in preclinical carriers of the Huntington's disease gene mutation (preHD). To date, however, the functional integrity of the DMN has not been addressed in this population. The aim of this study was to study the functional connectivity of the DMN in 18 preHD and 18 healthy controls who underwent functional magnetic resonance imaging during an attention task. A group independent component analysis identified spatiotemporally distinct patterns of two DMN subsystems. The spatial distribution of these components in preHD was similar to controls. However, preHD showed lower subsystem-specific connectivity in the anterior medial prefrontal cortex, the left inferior parietal and the posterior cingulate cortex (p<0.05, cluster-corrected). Connectivity between the two DMN subsystems was increased in preHD compared to controls. In preHD individuals lower functional connectivity of the left inferior parietal cortex was associated with shorter reaction times in the attention task. This suggests that some functionally critical regions of the DMN may have to remain active to maintain or optimise cognitive performance in preHD.
Collapse
Affiliation(s)
- Robert Christian Wolf
- Center for Psychosocial Medicine, Department of General Psychiatry, University of Heidelberg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|