1
|
Wu Q, Yao M, Liu H, Kakazu A, Ouyang Y, Liu C, Li R, Yang F, Wang A, Surasinghe S, Gerochi D, Baldo B, Jahn S, Tang H, Lu H, Wei Z, Duan W. Progressively reduced cerebral oxygen metabolism and elevated plasma NfL levels in the zQ175DN mouse model of Huntington's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.16.643551. [PMID: 40161725 PMCID: PMC11952543 DOI: 10.1101/2025.03.16.643551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Huntington's disease (HD) is a progressive neurodegenerative disorder caused by a CAG-repeat expansion in exon-1 of the huntingtin gene. Currently, no disease-modifying therapies are available, with a significant challenge in evaluating therapeutic efficacy before clinical symptoms emerge. This highlights the need for early biomarkers and intervention strategies. Therefore, it is essential to develop and characterize accurate mouse models and identify early biomarkers for preclinical therapeutic development. In this study, we characterized the pathological progression of the heterozygous zQ175 neo-deleted knock in (zQ175DN) mouse model across four age groups: 3, 6, 10, and 16 months to identify human translatable outcome measures. T2-relaxation-under-spin-tagging (TRUST) MRI was used to assess global CMRO 2 , while T2-weighted MRI was used to analyze brain volumes. Significant brain volume loss was detected as early as 6 months of age, worsening progressively with age in the zQ175 DN mice, resembling HD brain volumetric changes. A decline in CMRO 2 was observed in 6-month-old zQ175 DN mice, with significant and progressive reductions in 10- and 16-months old HD mice. Additionally, PHP1-positive mutant huntingtin (mHTT) aggregates were present in the striatum of zQ175 DN mice at all four age groups, with intranuclear localization prior to 6 months, transitioning to both intranuclear and neuropil aggregates in older zQ175 DN mice, suggesting that the localization of mHTT aggregates may reflect the severity of HD pathogenesis. Interestingly, plasma neurofilament light chain (NfL) protein concentrations were significantly elevated at 6 months of age and older zQ175DN mice. These findings provide valuable insights for selecting outcome measures in preclinical evaluations of HD therapies using the zQ175 DN mouse model.
Collapse
|
2
|
Ortega-Robles E, de Celis Alonso B, Cantillo-Negrete J, Carino-Escobar RI, Arias-Carrión O. Advanced Magnetic Resonance Imaging for Early Diagnosis and Monitoring of Movement Disorders. Brain Sci 2025; 15:79. [PMID: 39851446 PMCID: PMC11763950 DOI: 10.3390/brainsci15010079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/09/2025] [Accepted: 01/14/2025] [Indexed: 01/26/2025] Open
Abstract
Advanced magnetic resonance imaging (MRI) techniques are transforming the study of movement disorders by providing valuable insights into disease mechanisms. This narrative review presents a comprehensive overview of their applications in this field, offering an updated perspective on their potential for early diagnosis, disease monitoring, and therapeutic evaluation. Emerging MRI modalities such as neuromelanin-sensitive imaging, diffusion-weighted imaging, magnetization transfer imaging, and relaxometry provide sensitive biomarkers that can detect early microstructural degeneration, iron deposition, and connectivity disruptions in key regions like the substantia nigra. These techniques enable earlier and more accurate differentiation of movement disorders, including Parkinson's disease, progressive supranuclear palsy, multiple system atrophy, corticobasal degeneration, Lewy body and frontotemporal dementia, Huntington's disease, and dystonia. Furthermore, MRI provides objective metrics for tracking disease progression and assessing therapeutic efficacy, making it an indispensable tool in clinical trials. Despite these advances, the absence of standardized protocols limits their integration into routine clinical practice. Addressing this gap and incorporating these techniques more systematically could bring the field closer to leveraging advanced MRI for personalized treatment strategies, ultimately improving outcomes for individuals with movement disorders.
Collapse
Affiliation(s)
- Emmanuel Ortega-Robles
- Unidad de Trastornos del Movimiento y Sueño, Hospital General Dr. Manuel Gea González, Calzada de Tlalpan 4800, Mexico City 14080, Mexico;
| | - Benito de Celis Alonso
- Facultad de Ciencias Físico Matemáticas, Benemérita Universidad Autónoma de Puebla, Puebla 72570, Mexico;
| | - Jessica Cantillo-Negrete
- Technological Research Subdirection, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City 14389, Mexico;
| | - Ruben I. Carino-Escobar
- Division of Research in Clinical Neuroscience, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City 14389, Mexico;
| | - Oscar Arias-Carrión
- Unidad de Trastornos del Movimiento y Sueño, Hospital General Dr. Manuel Gea González, Calzada de Tlalpan 4800, Mexico City 14080, Mexico;
| |
Collapse
|
3
|
Martínez Lozada PS, Duque Perez J, Celi Salinas R, Miranda Morales B, Pazmiño Mesías JF, García Ríos CA, Rodas JA, Leon-Rojas JE. Early Diagnosis of Huntington Disease: Insights from Magnetic Resonance Spectroscopy-A Systematic Review. J Clin Med 2024; 13:6390. [PMID: 39518529 PMCID: PMC11546511 DOI: 10.3390/jcm13216390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/10/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
Background/Objectives: Huntington's disease (HD) is a fully penetrant neurodegenerative disease with a profound effect on quality of life. In recent years, there has been rapid growth in the description of its pathogenesis and diagnosis. Magnetic resonance spectroscopy (MRS) measurements can aid in the discrimination between premanifest Huntington's disease (Pre-HD) and healthy control (HC) subjects to establish early supportive and symptomatic management. Our objective was to evaluate metabolic changes using MRS to shed light on its potential as a biomarker through a systematic review. Methods: We followed the PRISMA guidelines, extracting articles from PubMed, Scopus, and the Virtual Health Library. We included patients with pre-HD, HD, and HC subjected to MRS, reporting the concentration of metabolites in at least one brain region. Results: In the putamen, N-acetyl Aspartate (NAA) was significantly decreased in 77.9% and total NAA (tNAA) was decreased in 72.4% of cases; no significant difference was found in 27.5% (n = 19) of cases. Furthermore, when looking into HD vs. pre-HD in the putamen, tNAA and NAA were decreased in 100% of participants. In the caudate nucleus, NAA and creatine were significantly decreased in 100% of HD in comparison to pre-HD participants, whereas tNAA showed a significant decrease in only 50%. Conclusions: MRS can be a relevant tool for the early diagnosis of HD; potential objective biomarkers related to its onset and pathogenesis exist and show differences between controls, pre-HD and HD patients. However, an effort should be made to standardize MRS methodology and reporting in subsequent studies.
Collapse
Affiliation(s)
- Pablo S. Martínez Lozada
- NeurALL Research Group, Quito 170157, Ecuador; (P.S.M.L.); (J.D.P.); (R.C.S.); (B.M.M.); (J.F.P.M.)
| | - José Duque Perez
- NeurALL Research Group, Quito 170157, Ecuador; (P.S.M.L.); (J.D.P.); (R.C.S.); (B.M.M.); (J.F.P.M.)
- Medical School, Universidad de las Américas (UDLA), Quito 170124, Ecuador
| | - Ronney Celi Salinas
- NeurALL Research Group, Quito 170157, Ecuador; (P.S.M.L.); (J.D.P.); (R.C.S.); (B.M.M.); (J.F.P.M.)
- Medignosis, Research Department, Quito 170157, Ecuador
- Christus Muguerza Hospital Alta Especialidad, Monterrey 64060, Mexico
| | - Bryan Miranda Morales
- NeurALL Research Group, Quito 170157, Ecuador; (P.S.M.L.); (J.D.P.); (R.C.S.); (B.M.M.); (J.F.P.M.)
| | | | | | - Jose A. Rodas
- School of Psychology, University College Dublin, D04 V1W8 Dublin, Ireland
- Escuela de Psicología, Universidad Espíritu Santo, Samborondón 092301, Ecuador
| | - Jose E. Leon-Rojas
- Medical School, Universidad de las Américas (UDLA), Quito 170124, Ecuador
| |
Collapse
|
4
|
Jing Y, Dogan I, Reetz K, Romanzetti S. Neurochemical changes in the progression of Huntington's disease: A meta-analysis of in vivo 1H-MRS studies. Neurobiol Dis 2024; 199:106574. [PMID: 38914172 DOI: 10.1016/j.nbd.2024.106574] [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: 05/13/2024] [Revised: 06/17/2024] [Accepted: 06/21/2024] [Indexed: 06/26/2024] Open
Abstract
Proton magnetic resonance spectroscopy (1H-MRS) allows measuring specific brain metabolic alterations in Huntington's disease (HD), and these metabolite profiles may serve as non-invasive biomarkers associated with disease progression. Despite this potential, previous findings are inconsistent. Accordingly, we performed a meta-analysis on available in vivo1H-MRS studies in premanifest (Pre-HD) and symptomatic HD stages (Symp-HD), and quantified neurometabolic changes relative to controls in 9 Pre-HD studies (227 controls and 188 mutation carriers) and 14 Symp-HD studies (326 controls and 306 patients). Our results indicated decreased N-acetylaspartate and creatine in the basal ganglia in both Pre-HD and Symp-HD. The overall level of myo-inositol was decreased in Pre-HD while increased in Symp-HD. Besides, Symp-HD patients showed more severe metabolism disruption than Pre-HD patients. Taken together, 1H-MRS is important for elucidating progressive metabolite changes from Pre-HD to clinical conversion; N-acetylaspartate and creatine in the basal ganglia are already sensitive at the preclinical stage and are promising biomarkers for tracking disease progression; overall myo-inositol is a possible characteristic metabolite for distinguishing HD stages.
Collapse
Affiliation(s)
- Yinghua Jing
- Department of Neurology, RWTH Aachen University, Aachen, Germany; JARA-Brain Institute Molecular Neuroscience and Neuroimaging (INM-11), Research Centre Jülich and RWTH Aachen University, Aachen, Germany
| | - Imis Dogan
- Department of Neurology, RWTH Aachen University, Aachen, Germany; JARA-Brain Institute Molecular Neuroscience and Neuroimaging (INM-11), Research Centre Jülich and RWTH Aachen University, Aachen, Germany
| | - Kathrin Reetz
- Department of Neurology, RWTH Aachen University, Aachen, Germany; JARA-Brain Institute Molecular Neuroscience and Neuroimaging (INM-11), Research Centre Jülich and RWTH Aachen University, Aachen, Germany
| | - Sandro Romanzetti
- Department of Neurology, RWTH Aachen University, Aachen, Germany; JARA-Brain Institute Molecular Neuroscience and Neuroimaging (INM-11), Research Centre Jülich and RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
5
|
Leem YH, Park JS, Park JE, Kim DY, Kim HS. Creatine supplementation with exercise reduces α-synuclein oligomerization and necroptosis in Parkinson's disease mouse model. J Nutr Biochem 2024; 126:109586. [PMID: 38262563 DOI: 10.1016/j.jnutbio.2024.109586] [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: 09/21/2023] [Revised: 01/04/2024] [Accepted: 01/17/2024] [Indexed: 01/25/2024]
Abstract
Parkinson's disease (PD) is an incurable neurological disorder that causes typical motor deficits. In this study, we investigated the effects of creatine supplementation and exercise in the subacute 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD. We found that 2% creatine supplementation and/or exercise intervention for 4 weeks elicited neurobehavioral recovery and neuroprotective effects regarding dopaminergic cell loss in MPTP-treated mice; this effect implies functional preservation of dopaminergic cells in the substantia nigra, as reflected by tyrosine hydroxylase expression recovery. Creatine and exercise reduced necroptotic activity in dopaminergic cells by lowering mixed lineage kinase domain-like protein (MLKL) modification to active phenotypes (phosphorylation at Ser345 and oligomerization) and phosphorylated receptor-interacting protein kinase 1 (RIPK1) (Ser166-p) and RIPK3 (Ser232-p) levels. In addition, creatine and exercise reduced the MPTP-induced increase in pathogenic α-synuclein forms, such as Ser129 phosphorylation and oligomerization. Furthermore, creatine and exercise had anti-inflammatory and antioxidative effects in MPTP mice, as evidenced by a decrease in microglia activation, NF-κB-dependent pro-inflammatory molecule expression, and increase in antioxidant enzyme expression. These phenotypic changes were associated with the exercise/creatine-induced AMP-activated protein kinase (AMPK)/nuclear factor erythroid 2-related factor 2 (Nrf2) and sirtuin 3 (SIRT3)/forkhead box O3 (FoxO3a) signaling pathways. In all experiments, combining creatine with exercise resulted in considerable improvement over either treatment alone. Consequently, these findings suggest that creatine supplementation with exercise has anti-inflammatory, antioxidative, and anti-α-synucleinopathy effects, thereby reducing necroptotic cell death in a PD mouse model.
Collapse
Affiliation(s)
- Yea-Hyun Leem
- Department of Molecular Medicine, Inflammation-Cancer Microenvironment Research Center, School of Medicine, Ewha Womans University, Seoul, South Korea
| | - Jin-Sun Park
- Department of Molecular Medicine, Inflammation-Cancer Microenvironment Research Center, School of Medicine, Ewha Womans University, Seoul, South Korea
| | - Jung-Eun Park
- Department of Molecular Medicine, Inflammation-Cancer Microenvironment Research Center, School of Medicine, Ewha Womans University, Seoul, South Korea
| | - Do-Yeon Kim
- Department of Molecular Medicine, Inflammation-Cancer Microenvironment Research Center, School of Medicine, Ewha Womans University, Seoul, South Korea
| | - Hee-Sun Kim
- Department of Molecular Medicine, Inflammation-Cancer Microenvironment Research Center, School of Medicine, Ewha Womans University, Seoul, South Korea; Department of Brain & Cognitive Sciences, Ewha Womans University, Seoul, South Korea.
| |
Collapse
|
6
|
Pérot JB, Brouillet E, Flament J. The contribution of preclinical magnetic resonance imaging and spectroscopy to Huntington's disease. Front Aging Neurosci 2024; 16:1306312. [PMID: 38414634 PMCID: PMC10896846 DOI: 10.3389/fnagi.2024.1306312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 01/24/2024] [Indexed: 02/29/2024] Open
Abstract
Huntington's disease is an inherited disorder characterized by psychiatric, cognitive, and motor symptoms due to degeneration of medium spiny neurons in the striatum. A prodromal phase precedes the onset, lasting decades. Current biomarkers include clinical score and striatal atrophy using Magnetic Resonance Imaging (MRI). These markers lack sensitivity for subtle cellular changes during the prodromal phase. MRI and MR spectroscopy offer different contrasts for assessing metabolic, microstructural, functional, or vascular alterations in the disease. They have been used in patients and mouse models. Mouse models can be of great interest to study a specific mechanism of the degenerative process, allow better understanding of the pathogenesis from the prodromal to the symptomatic phase, and to evaluate therapeutic efficacy. Mouse models can be divided into three different constructions: transgenic mice expressing exon-1 of human huntingtin (HTT), mice with an artificial chromosome expressing full-length human HTT, and knock-in mouse models with CAG expansion inserted in the murine htt gene. Several studies have used MRI/S to characterized these models. However, the multiplicity of modalities and mouse models available complicates the understanding of this rich corpus. The present review aims at giving an overview of results obtained using MRI/S for each mouse model of HD, to provide a useful resource for the conception of neuroimaging studies using mouse models of HD. Finally, despite difficulties in translating preclinical protocols to clinical applications, many biomarkers identified in preclinical models have already been evaluated in patients. This review also aims to cover this aspect to demonstrate the importance of MRI/S for studying HD.
Collapse
Affiliation(s)
- Jean-Baptiste Pérot
- Laboratoire des Maladies Neurodégénératives, Molecular Imaging Research Center, Commissariat à l’Energie Atomique et aux Energies Alternatives, Centre National de la Recherche Scientifique, Université Paris-Saclay, Fontenay-aux-Roses, France
- Institut du Cerveau – Paris Brain Institute – ICM, Sorbonne Université, Paris, France
| | - Emmanuel Brouillet
- Laboratoire des Maladies Neurodégénératives, Molecular Imaging Research Center, Commissariat à l’Energie Atomique et aux Energies Alternatives, Centre National de la Recherche Scientifique, Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Julien Flament
- Laboratoire des Maladies Neurodégénératives, Molecular Imaging Research Center, Commissariat à l’Energie Atomique et aux Energies Alternatives, Centre National de la Recherche Scientifique, Université Paris-Saclay, Fontenay-aux-Roses, France
| |
Collapse
|
7
|
Veeraiah P, Jansen JFA. Multinuclear Magnetic Resonance Spectroscopy at Ultra-High-Field: Assessing Human Cerebral Metabolism in Healthy and Diseased States. Metabolites 2023; 13:metabo13040577. [PMID: 37110235 PMCID: PMC10143499 DOI: 10.3390/metabo13040577] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 04/06/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
The brain is a highly energetic organ. Although the brain can consume metabolic substrates, such as lactate, glycogen, and ketone bodies, the energy metabolism in a healthy adult brain mainly relies on glucose provided via blood. The cerebral metabolism of glucose produces energy and a wide variety of intermediate metabolites. Since cerebral metabolic alterations have been repeatedly implicated in several brain disorders, understanding changes in metabolite levels and corresponding cell-specific neurotransmitter fluxes through different substrate utilization may highlight the underlying mechanisms that can be exploited to diagnose or treat various brain disorders. Magnetic resonance spectroscopy (MRS) is a noninvasive tool to measure tissue metabolism in vivo. 1H-MRS is widely applied in research at clinical field strengths (≤3T) to measure mostly high abundant metabolites. In addition, X-nuclei MRS including, 13C, 2H, 17O, and 31P, are also very promising. Exploiting the higher sensitivity at ultra-high-field (>4T; UHF) strengths enables obtaining unique insights into different aspects of the substrate metabolism towards measuring cell-specific metabolic fluxes in vivo. This review provides an overview about the potential role of multinuclear MRS (1H, 13C, 2H, 17O, and 31P) at UHF to assess the cerebral metabolism and the metabolic insights obtained by applying these techniques in both healthy and diseased states.
Collapse
Affiliation(s)
- Pandichelvam Veeraiah
- Scannexus (Ultra-High-Field MRI Center), 6229 EV Maastricht, The Netherlands
- Faculty of Health Medicine and Life Sciences, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
| | - Jacobus F A Jansen
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
- School for Mental Health and Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of Electrical Engineering, Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands
| |
Collapse
|
8
|
Taghian T, Gallagher J, Batcho E, Pullan C, Kuchel T, Denney T, Perumal R, Moore S, Muirhead R, Herde P, Johns D, Christou C, Taylor A, Passler T, Pulaparthi S, Hall E, Chandra S, O’Neill CA, Gray-Edwards H. Brain Alterations in Aged OVT73 Sheep Model of Huntington's Disease: An MRI Based Approach. J Huntingtons Dis 2022; 11:391-406. [PMID: 36189602 PMCID: PMC9837686 DOI: 10.3233/jhd-220526] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Huntington's disease (HD) is a fatal neurodegenerative autosomal dominant disorder with prevalence of 1 : 20000 that has no effective treatment to date. Translatability of candidate therapeutics could be enhanced by additional testing in large animal models because of similarities in brain anatomy, size, and immunophysiology. These features enable realistic pre-clinical studies of biodistribution, efficacy, and toxicity. OBJECTIVE AND METHODS Here we non-invasively characterized alterations in brain white matter microstructure, neurochemistry, neurological status, and mutant Huntingtin protein (mHTT) levels in cerebrospinal fluid (CSF) of aged OVT73 HD sheep. RESULTS Similar to HD patients, CSF mHTT differentiates HD from normal sheep. Our results are indicative of a decline in neurological status, and alterations in brain white matter diffusion and spectroscopy metric that are more severe in aged female HD sheep. Longitudinal analysis of aged female HD sheep suggests that the decline is detectable over the course of a year. In line with reports of HD human studies, white matter alterations in corpus callosum correlates with a decline in gait of HD sheep. Moreover, alterations in the occipital cortex white matter correlates with a decline in clinical rating score. In addition, the marker of energy metabolism in striatum of aged HD sheep, shows a correlation with decline of clinical rating score and eye coordination. CONCLUSION This data suggests that OVT73 HD sheep can serve as a pre-manifest large animal model of HD providing a platform for pre-clinical testing of HD therapeutics and non-invasive tracking of the efficacy of the therapy.
Collapse
Affiliation(s)
- Toloo Taghian
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA,
Department of Radiology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Jillian Gallagher
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
| | - Erin Batcho
- Department of Electrical and Computer Engineering, Auburn University, Auburn, AL, USA
| | - Caitlin Pullan
- South Australian Health and Medical Research Institute, Gillies Plains, SA, Australia
| | - Tim Kuchel
- South Australian Health and Medical Research Institute, Gillies Plains, SA, Australia
| | - Thomas Denney
- Department of Electrical and Computer Engineering, Auburn University, Auburn, AL, USA
| | - Raj Perumal
- South Australian Health and Medical Research Institute, Gillies Plains, SA, Australia
| | - Shamika Moore
- South Australian Health and Medical Research Institute, Gillies Plains, SA, Australia
| | - Robb Muirhead
- South Australian Health and Medical Research Institute, Gillies Plains, SA, Australia
| | - Paul Herde
- South Australian Health and Medical Research Institute, Gillies Plains, SA, Australia
| | - Daniel Johns
- South Australian Health and Medical Research Institute, Gillies Plains, SA, Australia
| | - Chris Christou
- South Australian Health and Medical Research Institute, Gillies Plains, SA, Australia
| | - Amanda Taylor
- Department of Clinical Sciences, Auburn University, Auburn, AL, USA
| | - Thomas Passler
- Department of Electrical and Computer Engineering, Auburn University, Auburn, AL, USA
| | - Sanjana Pulaparthi
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
| | - Erin Hall
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
| | - Sundeep Chandra
- Sana Biotechnology, South San Francisco, CA, USA,Bio Marin Pharmaceutical Inc., San Rafael, CA, USA
| | | | - Heather Gray-Edwards
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA,
Department of Radiology, University of Massachusetts Medical School, Worcester, MA, USA,Correspondence to: Heather L. Gray-Edwards, DVM, PhD, University of Massachusetts Medical School, Department of Radiology and Horae Gene Therapy Center, 368 Plantation Street, ASC6-2055, Worcester, MA 01605, USA. Tel.: +1 508 856 4051; Fax: +1 508 856 1552; E-mail:
| |
Collapse
|
9
|
Pérot JB, Célestine M, Palombo M, Dhenain M, Humbert S, Brouillet E, Flament J. Longitudinal multimodal MRI characterization of a knock-in mouse model of Huntington's disease reveals early gray and white matter alterations. Hum Mol Genet 2022; 31:3581-3596. [PMID: 35147158 PMCID: PMC9616570 DOI: 10.1093/hmg/ddac036] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 01/19/2022] [Accepted: 01/31/2022] [Indexed: 11/24/2022] Open
Abstract
Pathogenesis of the inherited neurodegenerative disorder Huntington's disease (HD) is progressive with a long presymptomatic phase in which subtle changes occur up to 15 years before the onset of symptoms. Thus, there is a need for early, functional biomarker to better understand disease progression and to evaluate treatment efficacy far from onset. Recent studies have shown that white matter may be affected early in mutant HTT gene carriers. A previous study performed on 12 months old Ki140CAG mice showed reduced glutamate level measured by Chemical Exchange Saturation Transfer of glutamate (gluCEST), especially in the corpus callosum. In this study, we scanned longitudinally Ki140CAG mice with structural MRI, diffusion tensor imaging, gluCEST and magnetization transfer imaging, in order to assess white matter integrity over the life of this mouse model characterized by slow progression of symptoms. Our results show early defects of diffusion properties in the anterior part of the corpus callosum at 5 months of age, preceding gluCEST defects in the same region at 8 and 12 months that spread to adjacent regions. At 12 months, frontal and piriform cortices showed reduced gluCEST, as well as the pallidum. MT imaging showed reduced signal in the septum at 12 months. Cortical and striatal atrophy then appear at 18 months. Vulnerability of the striatum and motor cortex, combined with alterations of anterior corpus callosum, seems to point out the potential role of white matter in the brain dysfunction that characterizes HD and the pertinence of gluCEST and DTI as biomarkers in HD.
Collapse
Affiliation(s)
- Jean-Baptiste Pérot
- Laboratoire des Maladies Neurodégénératives, Molecular Imaging Research Center (MIRCen), Université Paris-Saclay, Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Centre National de la Recherche Scientifique (CNRS), Fontenay -aux-Roses 92260, France
| | - Marina Célestine
- Laboratoire des Maladies Neurodégénératives, Molecular Imaging Research Center (MIRCen), Université Paris-Saclay, Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Centre National de la Recherche Scientifique (CNRS), Fontenay -aux-Roses 92260, France
| | - Marco Palombo
- Department of Computer Science, Centre for Medical Image Computing, University College London, London WC1E 6BT, UK
| | - Marc Dhenain
- Laboratoire des Maladies Neurodégénératives, Molecular Imaging Research Center (MIRCen), Université Paris-Saclay, Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Centre National de la Recherche Scientifique (CNRS), Fontenay -aux-Roses 92260, France
| | - Sandrine Humbert
- Université Grenoble Alpes, INSERM, U1216, Grenoble Institut Neurosciences, Grenoble 38000 , France
| | - Emmanuel Brouillet
- Laboratoire des Maladies Neurodégénératives, Molecular Imaging Research Center (MIRCen), Université Paris-Saclay, Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Centre National de la Recherche Scientifique (CNRS), Fontenay -aux-Roses 92260, France
| | - Julien Flament
- Laboratoire des Maladies Neurodégénératives, Molecular Imaging Research Center (MIRCen), Université Paris-Saclay, Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Centre National de la Recherche Scientifique (CNRS), Fontenay -aux-Roses 92260, France
| |
Collapse
|
10
|
Pradhan SS, Thota SM, Rajaratnam S, Bhagavatham SKS, Pulukool SK, Rathnakumar S, Phalguna KS, Dandamudi RB, Pargaonkar A, Joseph P, Joshy EV, Sivaramakrishnan V. Integrated multi-omics analysis of Huntington disease identifies pathways that modulate protein aggregation. Dis Model Mech 2022; 15:dmm049492. [PMID: 36052548 PMCID: PMC10655815 DOI: 10.1242/dmm.049492] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 08/15/2022] [Indexed: 11/20/2022] Open
Abstract
Huntington disease (HD) is a neurodegenerative disease associated with polyglutamine expansion in the protein huntingtin (HTT). Although the length of the polyglutamine repeat correlates with age at disease onset and severity, psychological, cognitive and behavioral complications point to the existence of disease modifiers. Mitochondrial dysfunction and metabolic deregulation are both associated with the HD but, despite multi-omics characterization of patients and model systems, their mechanisms have remained elusive. Systems analysis of multi-omics data and its validation by using a yeast model could help to elucidate pathways that modulate protein aggregation. Metabolomics analysis of HD patients and of a yeast model of HD was, therefore, carried out. Our analysis showed a considerable overlap of deregulated metabolic pathways. Further, the multi-omics analysis showed deregulated pathways common in human, mice and yeast model systems, and those that are unique to them. The deregulated pathways include metabolic pathways of various amino acids, glutathione metabolism, longevity, autophagy and mitophagy. The addition of certain metabolites as well as gene knockouts targeting the deregulated metabolic and autophagy pathways in the yeast model system showed that these pathways do modulate protein aggregation. Taken together, our results showed that the modulation of deregulated pathways influences protein aggregation in HD, and has implications for progression and prognosis. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Sai S. Pradhan
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Anantapur, Andhra Pradesh, India515134
| | - Sai M. Thota
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Anantapur, Andhra Pradesh, India515134
| | - Saiswaroop Rajaratnam
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Anantapur, Andhra Pradesh, India515134
| | - Sai K. S. Bhagavatham
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Anantapur, Andhra Pradesh, India515134
| | - Sujith K. Pulukool
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Anantapur, Andhra Pradesh, India515134
| | - Sriram Rathnakumar
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Anantapur, Andhra Pradesh, India515134
| | - Kanikaram S. Phalguna
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Anantapur, Andhra Pradesh, India515134
| | - Rajesh B. Dandamudi
- Department of Chemistry, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Anantapur, Andhra Pradesh 515 134, India
| | - Ashish Pargaonkar
- Application Division, Agilent Technologies Ltd., Bengaluru 560048, India
| | - Prasanth Joseph
- Application Division, Agilent Technologies Ltd., Bengaluru 560048, India
| | - E. V. Joshy
- Department of Neurology, Sri Sathya Sai Institute of Higher Medical Sciences, Whitefield, Bengaluru, Karnataka 560066, India
| | - Venketesh Sivaramakrishnan
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Anantapur, Andhra Pradesh, India515134
| |
Collapse
|
11
|
Bolo NR, Jacobson AM, Musen G, Simonson DC. Hyperglycemia and hyperinsulinemia effects on anterior cingulate cortex myoinositol-relation to brain network functional connectivity in healthy adults. J Neurophysiol 2022; 127:1426-1437. [PMID: 35417272 PMCID: PMC9109787 DOI: 10.1152/jn.00408.2021] [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: 09/13/2021] [Revised: 03/22/2022] [Accepted: 04/11/2022] [Indexed: 11/22/2022] Open
Abstract
Brain mechanisms underlying the association of diabetes metabolic disorders-hyperglycemia and insulin resistance-with cognitive impairment are unknown. Myoinositol is a brain metabolite involved in cell osmotic balance, membrane phospholipid turnover, and second messenger neurotransmission, which affect brain function. Increased brain myoinositol and altered functional connectivity have been found in diabetes, mild cognitive impairment, and Alzheimer's disease, but the independent effects of plasma glucose and insulin on brain myoinositol and function are not characterized. We measured myoinositol concentrations in the pregenual anterior cingulate cortex (ACC), a region involved in self-reflective awareness and decision making, using proton magnetic resonance spectroscopy, and whole brain resting-state functional connectivity using fMRI, during acute hyperglycemia (with attendant hyperinsulinemia) and euglycemic-hyperinsulinemia compared with basal fasting-euglycemia (EU) in 11 healthy nondiabetic participants (5 women/6 men, means ± SD, age: 27 ± 7 yr, fasting-glucose: 5.2 ± 0.4 mmol/L, fasting-insulin: 4.9 ± 4.4 μU/mL). Brain MR data were acquired during two separate visits: 1) EU followed by a 60-min hyperglycemic-clamp (glucose: 10.7 ± 0.2 mmol/L, insulin: 33 ± 6 μU/mL); 2) EU followed by a hyperinsulinemic-euglycemic-clamp (glucose: 5.3 ± 0.1 mmol/L, insulin: 27 ± 5 μU/mL) designed to match individual insulin levels achieved during the visit 1 hyperglycemic-clamp. Myoinositol decreased by 14% during the hyperglycemic-clamp (from 7.7 ± 1.5 mmol/kg to 6.6 ± 0.8 mmol/kg, P = 0.031), and by 9% during the hyperinsulinemic-euglycemic-clamp (from 7.1 ± 0.7 mmol/kg to 6.5 ± 0.7 mmol/kg, P = 0.014), with no significant difference between the two clamps. Lower myoinositol was associated with higher functional connectivity of the thalamus and precentral cortex with insula-ACC-related networks, suggesting myoinositol is involved in insulin modulation of cognitive/emotional network function in healthy adults. Regional brain myoinositol levels may be useful biomarkers for monitoring cognitive and mood-enhancing treatment responses.NEW & NOTEWORTHY Hyperinsulinemia-related decreases of brain anterior cingulate cortex (ACC) myoinositol independent of plasma glucose levels and the association of low ACC myoinositol with increased functional connectivity between sensorimotor regions and ACC/insula-related networks suggest involvement of myoinositol in insulin-modulated brain network function in healthy adults. In diabetes, elevated brain myoinositol may be due to reduced brain insulin levels or action, rather than hyperglycemia, and may be involved in brain network dysfunctions leading to cognitive or mood disorders.
Collapse
Affiliation(s)
- Nicolas R Bolo
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - Alan M Jacobson
- Research Institute, NYU Long Island School of Medicine, Mineola, New York
| | - Gail Musen
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
- Research Division, Joslin Diabetes Center, Boston, Massachusetts
| | - Donald C Simonson
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
12
|
Ostojic SM, Engeset D. Improving Brain Creatine Uptake by Klotho Protein Stimulation: Can Diet Hit the Big Time? Front Nutr 2022; 8:795599. [PMID: 35004821 PMCID: PMC8732999 DOI: 10.3389/fnut.2021.795599] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 12/08/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Sergej M Ostojic
- Department of Nutrition and Public Health, University of Agder, Kristiansand, Norway
| | - Dagrun Engeset
- Department of Nutrition and Public Health, University of Agder, Kristiansand, Norway
| |
Collapse
|
13
|
Platt T, Ladd ME, Paech D. 7 Tesla and Beyond: Advanced Methods and Clinical Applications in Magnetic Resonance Imaging. Invest Radiol 2021; 56:705-725. [PMID: 34510098 PMCID: PMC8505159 DOI: 10.1097/rli.0000000000000820] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 08/07/2021] [Accepted: 08/07/2021] [Indexed: 12/15/2022]
Abstract
ABSTRACT Ultrahigh magnetic fields offer significantly higher signal-to-noise ratio, and several magnetic resonance applications additionally benefit from a higher contrast-to-noise ratio, with static magnetic field strengths of B0 ≥ 7 T currently being referred to as ultrahigh fields (UHFs). The advantages of UHF can be used to resolve structures more precisely or to visualize physiological/pathophysiological effects that would be difficult or even impossible to detect at lower field strengths. However, with these advantages also come challenges, such as inhomogeneities applying standard radiofrequency excitation techniques, higher energy deposition in the human body, and enhanced B0 field inhomogeneities. The advantages but also the challenges of UHF as well as promising advanced methodological developments and clinical applications that particularly benefit from UHF are discussed in this review article.
Collapse
Affiliation(s)
- Tanja Platt
- From the Medical Physics in Radiology, German Cancer Research Center (DKFZ)
| | - Mark E. Ladd
- From the Medical Physics in Radiology, German Cancer Research Center (DKFZ)
- Faculty of Physics and Astronomy
- Faculty of Medicine, University of Heidelberg, Heidelberg
- Erwin L. Hahn Institute for MRI, University of Duisburg-Essen, Essen
| | - Daniel Paech
- Division of Radiology, German Cancer Research Center (DKFZ), Heidelberg
- Clinic for Neuroradiology, University of Bonn, Bonn, Germany
| |
Collapse
|
14
|
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
|
15
|
O'Connell AB, Kuchel TR, Perumal SR, Sherwood V, Neumann D, Finnie JW, Hemsley KM, Morton AJ. Longitudinal Magnetic Resonance Spectroscopy and Diffusion Tensor Imaging in Sheep (Ovis aries) With Quinolinic Acid Lesions of the Striatum: Time-Dependent Recovery of N-Acetylaspartate and Fractional Anisotropy. J Neuropathol Exp Neurol 2021; 79:1084-1092. [PMID: 32743645 DOI: 10.1093/jnen/nlaa053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Indexed: 12/19/2022] Open
Abstract
We created an excitotoxic striatal lesion model of Huntington disease (HD) in sheep, using the N-methyl-d-aspartate receptor agonist, quinolinic acid (QA). Sixteen sheep received a bolus infusion of QA (75 µL, 180 mM) or saline, first into the left and then (4 weeks later) into the right striatum. Magnetic resonance spectroscopy (MRS) and diffusion tensor imaging (DTI) of the striata were performed. Metabolite concentrations and fractional anisotropy (FA) were measured at baseline, acutely (1 week after each surgery) and chronically (5 weeks or greater after the surgeries). There was a significant decrease in the neuronal marker N-acetylaspartate (NAA) and in FA in acutely lesioned striata of the QA-lesioned sheep, followed by a recovery of NAA and FA in the chronically lesioned striata. NAA level changes indicate acute death and/or impairment of neurons immediately after surgery, with recovery of reversibly impaired neurons over time. The change in FA values of the QA-lesioned striata is consistent with acute structural disruption, followed by re-organization and glial cell infiltration with time. Our study demonstrates that MRS and DTI changes in QA-sheep are consistent with HD-like pathology shown in other model species and that the MR investigations can be performed in sheep using a clinically relevant human 3T MRI scanner.
Collapse
Affiliation(s)
- Adam B O'Connell
- Pre-Clinical, Imaging and Research Laboratories (PIRL), South Australia Health and Medical Research Institute (SAHMRI), Adelaide, South Australia.,School of Medical Specialties, University of Adelaide, Adelaide, South Australia
| | - Timothy R Kuchel
- Pre-Clinical, Imaging and Research Laboratories (PIRL), South Australia Health and Medical Research Institute (SAHMRI), Adelaide, South Australia
| | - Sunthara R Perumal
- Pre-Clinical, Imaging and Research Laboratories (PIRL), South Australia Health and Medical Research Institute (SAHMRI), Adelaide, South Australia
| | | | - Daniel Neumann
- Childhood Dementia Research Group, Hopwood Centre for Neurobiology, SAHMRI, Adelaide, Australia.,Childhood Dementia Research Group, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - John W Finnie
- Discipline of Anatomy and Pathology, Adelaide Medical School, University of Adelaide and SA Pathology, Adelaide, Australia
| | - Kim M Hemsley
- Childhood Dementia Research Group, Hopwood Centre for Neurobiology, SAHMRI, Adelaide, Australia.,Childhood Dementia Research Group, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - A Jennifer Morton
- Department of Physiology, Development and Neuroscience, Cambridge University, Cambridge, UK
| |
Collapse
|
16
|
Wang N, Zeng Z, Wang B, Qin D, Wang T, Wang C, Guo S. High serum creatinine is associated with reduction of psychiatric symptoms in female patients with anti-NMDAR encephalitis. Neurosci Lett 2021; 746:135650. [PMID: 33485991 DOI: 10.1016/j.neulet.2021.135650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/21/2020] [Accepted: 01/07/2021] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Serum creatinine (SCR) has been shown to be associated with many neurodegenerative diseases. In this study, we investigated the relationship between SCR levels and the incidence of psychiatric symptoms in patients with anti-N-methyl-d-aspartate receptor (anti-NMDAR) encephalitis. METHODS The SCR levels were tested in 69 patients with anti-NMDAR encephalitis at admission. Clinical characteristics and blood and CSF parameters were compared between the group of patients with psychiatric symptoms (P + group) and the group of those without psychiatric symptoms (P- group). The association between SCR and the incidence of psychiatric symptoms was determined by multivariate-adjusted linear regression analyses. RESULTS The SCR levels in the P + group were significantly lower than those in the P- group (P < 0.001). In the female subgroup, the SCR levels in the P + group were significantly lower compared to the P- group (P < 0.001), whereas in the male subgroup, the SCR levels did not differ between the two groups (P = 0.084). Furthermore, the highest SCR tercile overall had a significantly lower incidence of psychiatric symptoms than the lowest tercile (P < 0.001), and a significant negative correlation between the SCR levels and the occurrence of psychiatric symptoms was observed (r = -0.392, P < 0.001). Multivariate logistic regression analysis showed that the association was independent after adjusting for age, cystatin C and the modified Rankin Scale (mRS) score (P = 0.001). A similar result was found in the female subgroup (P = 0.010), but not in the male subgroup (P = 0.225). CONCLUSION Our study indicated that the SCR level was negatively correlated with incidence of psychiatric symptoms in female patients, and higher SCR level could be a protective factor for psychiatric symptoms in female patients with anti-NMDAR encephalitis.
Collapse
Affiliation(s)
- Ningning Wang
- Department of Neurology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jing Wu Road, Huaiyin District, Jinan, 250021, Shandong, China
| | - Ziling Zeng
- Department of Neurology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jing Wu Road, Huaiyin District, Jinan, 250021, Shandong, China
| | - Baojie Wang
- Department of Neurology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jing Wu Road, Huaiyin District, Jinan, 250021, Shandong, China
| | - Danqing Qin
- Department of Neurology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jing Wu Road, Huaiyin District, Jinan, 250021, Shandong, China
| | - Tingting Wang
- Department of Neurology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jing Wu Road, Huaiyin District, Jinan, 250021, Shandong, China
| | - Chunjuan Wang
- Department of Neurology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jing Wu Road, Huaiyin District, Jinan, 250021, Shandong, China
| | - Shougang Guo
- Department of Neurology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jing Wu Road, Huaiyin District, Jinan, 250021, Shandong, China.
| |
Collapse
|
17
|
van Wamelen DJ, Aziz NA. Hypothalamic pathology in Huntington disease. HANDBOOK OF CLINICAL NEUROLOGY 2021; 182:245-255. [PMID: 34266596 DOI: 10.1016/b978-0-12-819973-2.00017-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Huntington's disease (HD), an autosomal dominant hereditary disorder associated with the accumulation of mutant huntingtin, is classically associated with cognitive decline and motor symptoms, notably chorea. However, growing evidence suggests that nonmotor symptoms are equally prevalent and debilitating. Some of these symptoms may be linked to hypothalamic pathology, demonstrated by findings in HD animal models and HD patients showing specific changes in hypothalamic neuropeptidergic populations and their associated functions. At least some of these alterations are likely due to local mutant huntingtin expression and toxicity, while others are likely caused by disturbed hypothalamic circuitry. Common problems include circadian rhythm disorders, including desynchronization of daily hormone excretion patterns, which could be targeted by novel therapeutic interventions, such as timed circadian interventions with light therapy or melatonin. However, translation of these findings from bench-to-bedside is hampered by differences in murine HD models and HD patients, including mutant huntingtin trinucleotide repeat length, which is highly heterogeneous across the various models. In this chapter, we summarize the current knowledge regarding hypothalamic alterations in HD patients and animal models, and the potential for these findings to be translated into clinical practice and management.
Collapse
Affiliation(s)
- Daniel J van Wamelen
- Department of Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom; Parkinson's Foundation Centre of Excellence, King's College Hospital, London, United Kingdom; Department of Neurology, Radboud University Medical Centre, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands.
| | - N Ahmad Aziz
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany; Department of Neurology, Faculty of Medicine, University of Bonn, Bonn, Germany
| |
Collapse
|
18
|
van Diemen MPJ, Hart EP, Abbruscato A, Mead L, van Beelen I, Bergheanu SC, Hameeteman PW, Coppen E, Winder JY, Moerland M, Kan H, van der Grond J, Webb A, Roos RAC, Groeneveld GJ. Safety, pharmacokinetics and pharmacodynamics of SBT-020 in patients with early stage Huntington's disease, a 2-part study. Br J Clin Pharmacol 2020; 87:2290-2302. [PMID: 33197078 PMCID: PMC8247328 DOI: 10.1111/bcp.14656] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 10/29/2020] [Accepted: 10/30/2020] [Indexed: 12/19/2022] Open
Abstract
Aims Huntington's disease (HD) is a neurodegenerative disease with cognitive, motor and psychiatric symptoms. Toxic accumulation of misfolded mutant huntingtin protein induces mitochondrial dysfunction, leading to a bioenergetic insufficiency in neuronal and muscle cells. We evaluated the safety, pharmacokinetics and pharmacodynamics of SBT‐020, a novel compound to improve mitochondrial function, in a 2‐part study in early stage HD patients. Methods Part 1 consisted of 7‐day multiple ascending dose study to select the highest tolerable dose for Part 2, a 28‐day multiple dose study. Mitochondrial function was measured in the visual cortex and calf muscle, using phosphorous magnetic resonance spectroscopy, and in circulating peripheral blood mononuclear cells. Results Treatment‐emergent adverse events were mild and more present in the SBT‐020 group. Injection site reactions occurred in 91% in Part 1 and 97% in Part 2. Mitochondrial function in calf muscle, peripheral blood mononuclear cells or visual cortex was not changed overall due to treatment with SBT‐020. In a posthoc analysis, patients with a higher degree of mitochondrial dysfunction (below the median [∆Ψm < 3412 and τPCr > 42.5 s]) showed more improvement than patients with a relatively lower level of mitochondrial dysfunction. Conclusion SBT‐020 was safe at all doses, but no significant differences in any of the pharmacodynamic measurements between the treatment groups and placebo group could be demonstrated. The data suggest that the better than expected mitochondrial function in our patient population at baseline might explain the lack of effect of SBT‐020.
Collapse
Affiliation(s)
| | - Ellen P Hart
- Centre for Human Drug Research, Leiden, The Netherlands
| | | | - Liz Mead
- Stealth BioTherapeutics Inc, Newton, Massachusetts, USA
| | | | | | | | - Emma Coppen
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jessica Y Winder
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Hermien Kan
- Gorter Centre for high-field MRI, Leiden University Medical Center, Leiden, The Netherlands
| | - Jeroen van der Grond
- Radiology Research Center, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Andrew Webb
- Gorter Centre for high-field MRI, Leiden University Medical Center, Leiden, The Netherlands
| | - Raymund A C Roos
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
19
|
van Diemen MPJ, Hart EP, Hameeteman PW, Coppen EM, Winder JY, den Heijer J, Moerland M, Kan H, van der Grond J, Webb A, Roos RAC, Groeneveld GJ. Brain Bio-Energetic State Does Not Correlate to Muscle Mitochondrial Function in Huntington's Disease. J Huntingtons Dis 2020; 9:335-344. [PMID: 33325391 DOI: 10.3233/jhd-200413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Huntington's disease (HD) is a neurodegenerative disease with cognitive, motor and psychiatric symptoms. A toxic accumulation of misfolded mutant huntingtin protein (Htt) induces mitochondrial dysfunction, leading to a bioenergetic insufficiency in neuronal and muscle cells. Improving mitochondrial function has been proposed as an opportunity to treat HD, but it is not known how mitochondrial function in different tissues relates. OBJECTIVE We explored associations between central and peripheral mitochondrial function in a group of mild to moderate staged HD patients. METHODS We used phosphorous magnetic resonance spectroscopy (31P-MRS) to measure mitochondrial function in vivo in the calf muscle (peripheral) and the bio-energetic state in the visual cortex (central). Mitochondrial function was also assessed ex vivo in circulating peripheral blood mononuclear cells (PBMCs). Clinical function was determined by the Unified Huntington's Disease Rating Scale (UHDRS) total motor score. Pearson correlation coefficients were computed to assess the correlation between the different variables. RESULTS We included 23 manifest HD patients for analysis. There was no significant correlation between central bio-energetics and peripheral mitochondrial function. Central mitochondrial function at rest correlated significantly to the UHDRS total motor score (R = -0.45 and -0.48), which increased in a subgroup with the largest number of CAG repeats. DISCUSSION We did not observe a correlation between peripheral and central mitochondrial function. Central, but not peripheral, mitochondrial function correlated to clinical function. Muscle mitochondrial function is a promising biomarker to evaluate disease-modifying compounds that improve mitochondrial function, but Huntington researchers should use central mitochondrial function to demonstrate proof-of-pharmacology of disease-modifying compounds.
Collapse
Affiliation(s)
| | - Ellen P Hart
- Centre for Human Drug Research, Leiden, The Netherlands
| | | | - Emma M Coppen
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jessica Y Winder
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | - Hermien Kan
- Gorter Centre for High-field MRI, Leiden University Medical Center, Leiden, The Netherlands
| | - Jeroen van der Grond
- Radiology Research Center, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Andrew Webb
- Gorter Centre for High-field MRI, Leiden University Medical Center, Leiden, The Netherlands
| | - Raymund A C Roos
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Geert Jan Groeneveld
- Centre for Human Drug Research, Leiden, The Netherlands.,Department of Anesthesiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
20
|
Bertrand M, Decoville M, Meudal H, Birman S, Landon C. Metabolomic Nuclear Magnetic Resonance Studies at Presymptomatic and Symptomatic Stages of Huntington’s Disease on a Drosophila Model. J Proteome Res 2020; 19:4034-4045. [DOI: 10.1021/acs.jproteome.0c00335] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Marylène Bertrand
- Center for Molecular Biophysics, CBM, UPR 4301, CNRS, Rue Charles SADRON, F-45071 Orléans Cedex 02, France
| | - Martine Decoville
- Center for Molecular Biophysics, CBM, UPR 4301, CNRS, Rue Charles SADRON, F-45071 Orléans Cedex 02, France
- University of Orléans, 6 Avenue du Parc Floral, F-45100 Orléans, France
| | - Hervé Meudal
- Center for Molecular Biophysics, CBM, UPR 4301, CNRS, Rue Charles SADRON, F-45071 Orléans Cedex 02, France
| | - Serge Birman
- GCRN Team, Brain Plasticity Unit, UMR 8249, CNRS, ESPCI Paris, PSL Research University, F-75005 Paris, France
| | - Céline Landon
- Center for Molecular Biophysics, CBM, UPR 4301, CNRS, Rue Charles SADRON, F-45071 Orléans Cedex 02, France
| |
Collapse
|
21
|
Bolo NR, Jacobson AM, Musen G, Keshavan MS, Simonson DC. Acute Hyperglycemia Increases Brain Pregenual Anterior Cingulate Cortex Glutamate Concentrations in Type 1 Diabetes. Diabetes 2020; 69:1528-1539. [PMID: 32295804 PMCID: PMC7306132 DOI: 10.2337/db19-0936] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 04/09/2020] [Indexed: 12/15/2022]
Abstract
The brain mechanisms underlying the association of hyperglycemia with depressive symptoms are unknown. We hypothesized that disrupted glutamate metabolism in pregenual anterior cingulate cortex (ACC) in type 1 diabetes (T1D) without depression affects emotional processing. Using proton MRS, we measured glutamate concentrations in ACC and occipital lobe cortex (OCC) in 13 subjects with T1D without major depression (HbA1c 7.1 ± 0.7% [54 ± 7 mmol/mol]) and 11 healthy control subjects without diabetes (HbA1c 5.5 ± 0.2% [37 ± 3 mmol/mol]) during fasting euglycemia followed by a 60-min +5.5 mmol/L hyperglycemic clamp (HG). Intrinsic neuronal activity was assessed using resting-state blood oxygen level-dependent functional MRI to measure the fractional amplitude of low-frequency fluctuations in slow-4 band (fALFF4). Emotional processing and depressive symptoms were assessed using emotional tasks (emotional Stroop task, self-referent encoding task [SRET]) and clinical ratings (Hamilton Depression Rating Scale [HAM-D], Symptom Checklist-90 Revised [SCL-90-R]), respectively. During HG, ACC glutamate increased (1.2 mmol/kg, 10% P = 0.014) while ACC fALFF4 was unchanged (-0.007, -2%, P = 0.449) in the T1D group; in contrast, glutamate was unchanged (-0.2 mmol/kg, -2%, P = 0.578) while fALFF4 decreased (-0.05, -13%, P = 0.002) in the control group. OCC glutamate and fALFF4 were unchanged in both groups. T1D had longer SRET negative word response times (P = 0.017) and higher depression rating scores (HAM-D P = 0.020, SCL-90-R depression P = 0.008). Higher glutamate change tended to associate with longer emotional Stroop response times in T1D only. Brain glutamate must be tightly controlled during hyperglycemia because of the risk for neurotoxicity with excessive levels. Results suggest that ACC glutamate control mechanisms are disrupted in T1D, which affects glutamatergic neurotransmission related to emotional or cognitive processing. Increased prefrontal glutamate during acute hyperglycemic episodes could explain our previous findings of associations among chronic hyperglycemia, cortical thinning, and depressive symptoms in T1D.
Collapse
Affiliation(s)
- Nicolas R Bolo
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA
- Department of Psychiatry, Harvard Medical School, Boston, MA
| | - Alan M Jacobson
- Research Institute, NYU Long Island School of Medicine, Mineola, NY
| | - Gail Musen
- Department of Psychiatry, Harvard Medical School, Boston, MA
- Research Division, Joslin Diabetes Center, Boston, MA
| | - Matcheri S Keshavan
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA
- Department of Psychiatry, Harvard Medical School, Boston, MA
| | - Donald C Simonson
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
22
|
McColgan P, Joubert J, Tabrizi SJ, Rees G. The human motor cortex microcircuit: insights for neurodegenerative disease. Nat Rev Neurosci 2020; 21:401-415. [PMID: 32555340 DOI: 10.1038/s41583-020-0315-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2020] [Indexed: 12/22/2022]
Abstract
The human motor cortex comprises a microcircuit of five interconnected layers with different cell types. In this Review, we use a layer-specific and cell-specific approach to integrate physiological accounts of this motor cortex microcircuit with the pathophysiology of neurodegenerative diseases affecting motor functions. In doing so we can begin to link motor microcircuit pathology to specific disease stages and clinical phenotypes. Based on microcircuit physiology, we can make future predictions of axonal loss and microcircuit dysfunction. With recent advances in high-resolution neuroimaging we can then test these predictions in humans in vivo, providing mechanistic insights into neurodegenerative disease.
Collapse
Affiliation(s)
- Peter McColgan
- Huntington's Disease Research Centre, UCL Institute of Neurology, University College London, London, UK.
| | - Julie Joubert
- Huntington's Disease Research Centre, UCL Institute of Neurology, University College London, London, UK
| | - Sarah J Tabrizi
- Huntington's Disease Research Centre, UCL Institute of Neurology, University College London, London, UK.,Dementia Research Institute at UCL, London, UK
| | - Geraint Rees
- Wellcome Centre for Human Neuroimaging, UCL Institute of Neurology, University College London, London, UK.,UCL Institute of Cognitive Neuroscience, University College London, London, UK
| |
Collapse
|
23
|
Colzato L, Beste C. A literature review on the neurophysiological underpinnings and cognitive effects of transcutaneous vagus nerve stimulation: challenges and future directions. J Neurophysiol 2020; 123:1739-1755. [PMID: 32208895 DOI: 10.1152/jn.00057.2020] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Brain stimulation approaches are important to gain causal mechanistic insights into the relevance of functional brain regions and/or neurophysiological systems for human cognitive functions. In recent years, transcutaneous vagus nerve stimulation (tVNS) has attracted considerable popularity. It is a noninvasive brain stimulation technique based on the stimulation of the vagus nerve. The stimulation of this nerve activates subcortical nuclei, such as the locus coeruleus and the nucleus of the solitary tract, and from there, the activation propagates to the cortex. Since tVNS is a novel stimulation technique, this literature review outlines a brief historical background of tVNS, before detailing underlying neurophysiological mechanisms of action, stimulation parameters, cognitive effects of tVNS on healthy humans, and, lastly, current challenges and future directions of tVNS research in cognitive functions. Although more research is needed, we conclude that tVNS, by increasing norepineprine (NE) and gamma-aminobutyric acid (GABA) levels, affects NE- and GABA-related cognitive performance. The review provides detailed background information how to use tVNS as a neuromodulatory tool in cognitive neuroscience and outlines important future leads of research on tVNS.
Collapse
Affiliation(s)
- Lorenza Colzato
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Dresden, Germany.,Department of Cognitive Psychology, Institute of Cognitive Neuroscience, Faculty of Psychology, Ruhr University Bochum, Bochum, Germany.,Cognitive Psychology, Faculty of Psychology, Shandong Normal University, Jinan, China
| | - Christian Beste
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Dresden, Germany.,Cognitive Psychology, Faculty of Psychology, Shandong Normal University, Jinan, China
| |
Collapse
|
24
|
Altered hypothalamic metabolism in early multiple sclerosis – MR spectroscopy study. J Neurol Sci 2019; 407:116458. [DOI: 10.1016/j.jns.2019.116458] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 08/19/2019] [Accepted: 09/10/2019] [Indexed: 12/31/2022]
|
25
|
Marques EP, Ferreira FS, Santos TM, Prezzi CA, Martins LAM, Bobermin LD, Quincozes-Santos A, Wyse ATS. Cross-talk between guanidinoacetate neurotoxicity, memory and possible neuroprotective role of creatine. Biochim Biophys Acta Mol Basis Dis 2019; 1865:165529. [PMID: 31398469 DOI: 10.1016/j.bbadis.2019.08.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 07/21/2019] [Accepted: 08/05/2019] [Indexed: 12/18/2022]
Abstract
Guanidinoacetate Methyltransferase deficiency is an inborn error of metabolism that results in decreased creatine and increased guanidinoacetate (GAA) levels. Patients present neurological symptoms whose mechanisms are unclear. We investigated the effects of an intrastriatal administration of 10 μM of GAA (0.02 nmol/striatum) on energy metabolism, redox state, inflammation, glutamate homeostasis, and activities/immunocontents of acetylcholinesterase and Na+,K+-ATPase, as well as on memory acquisition. The neuroprotective role of creatine was also investigated. Male Wistar rats were pretreated with creatine (50 mg/kg) or saline for 7 days underwenting stereotactic surgery. Forty-eight hours after surgery, the animals (then sixty-days-old) were divided into groups: Control, GAA, GAA + Creatine, and Creatine. Experiments were performed 30 min after intrastriatal infusion. GAA decreased SDH, complexes II and IV activities, and ATP levels, but had no effect on mitochondrial mass/membrane potential. Creatine totally prevented SDH and complex II, and partially prevented COX and ATP alterations. GAA increased dichlorofluorescein levels and decreased superoxide dismutase and catalase activities. Creatine only prevented catalase and dichlorofluorescein alterations. GAA increased cytokines, nitrites levels and acetylcholinesterase activity, but not its immunocontent. Creatine prevented such effects, except nitrite levels. GAA decreased glutamate uptake, but had no effect on the immunocontent of its transporters. GAA decreased Na+,K+-ATPase activity and increased the immunocontent of its α3 subunit. The performance on the novel object recognition task was also impaired. Creatine partially prevented the changes in glutamate uptake and Na+,K+-ATPase activity, and completely prevented the memory impairment. This study helps to elucidate the protective effects of creatine against the damage caused by GAA.
Collapse
Affiliation(s)
- Eduardo Peil Marques
- Laboratory of Neuroprotection and Neurometabolic Diseases, Biochemistry Department, ICBS, Universidade Federal do Rio Grande do Sul, Street Ramiro Barcelos, 2600-Annex, CEP 90035-003 Porto Alegre, RS, Brazil; Program of Post-graduation in Biological Sciences-Biochemistry, Biochemistry Department, ICBS, Universidade Federal do Rio Grande do Sul, Street Ramiro Barcelos, 2600-Annex, CEP 90035-003 Porto Alegre, RS, Brazil
| | - Fernanda Silva Ferreira
- Laboratory of Neuroprotection and Neurometabolic Diseases, Biochemistry Department, ICBS, Universidade Federal do Rio Grande do Sul, Street Ramiro Barcelos, 2600-Annex, CEP 90035-003 Porto Alegre, RS, Brazil; Program of Post-graduation in Biological Sciences-Biochemistry, Biochemistry Department, ICBS, Universidade Federal do Rio Grande do Sul, Street Ramiro Barcelos, 2600-Annex, CEP 90035-003 Porto Alegre, RS, Brazil
| | - Tiago Marcon Santos
- Laboratory of Neuroprotection and Neurometabolic Diseases, Biochemistry Department, ICBS, Universidade Federal do Rio Grande do Sul, Street Ramiro Barcelos, 2600-Annex, CEP 90035-003 Porto Alegre, RS, Brazil; Program of Post-graduation in Biological Sciences-Biochemistry, Biochemistry Department, ICBS, Universidade Federal do Rio Grande do Sul, Street Ramiro Barcelos, 2600-Annex, CEP 90035-003 Porto Alegre, RS, Brazil
| | - Caroline Acauan Prezzi
- Laboratory of Neuroprotection and Neurometabolic Diseases, Biochemistry Department, ICBS, Universidade Federal do Rio Grande do Sul, Street Ramiro Barcelos, 2600-Annex, CEP 90035-003 Porto Alegre, RS, Brazil; Program of Post-graduation in Biological Sciences-Biochemistry, Biochemistry Department, ICBS, Universidade Federal do Rio Grande do Sul, Street Ramiro Barcelos, 2600-Annex, CEP 90035-003 Porto Alegre, RS, Brazil
| | - Leo A M Martins
- Program of Post-graduation in Biological Sciences-Biochemistry, Biochemistry Department, ICBS, Universidade Federal do Rio Grande do Sul, Street Ramiro Barcelos, 2600-Annex, CEP 90035-003 Porto Alegre, RS, Brazil
| | - Larissa Daniele Bobermin
- Program of Post-graduation in Biological Sciences-Biochemistry, Biochemistry Department, ICBS, Universidade Federal do Rio Grande do Sul, Street Ramiro Barcelos, 2600-Annex, CEP 90035-003 Porto Alegre, RS, Brazil
| | - André Quincozes-Santos
- Program of Post-graduation in Biological Sciences-Biochemistry, Biochemistry Department, ICBS, Universidade Federal do Rio Grande do Sul, Street Ramiro Barcelos, 2600-Annex, CEP 90035-003 Porto Alegre, RS, Brazil
| | - Angela T S Wyse
- Laboratory of Neuroprotection and Neurometabolic Diseases, Biochemistry Department, ICBS, Universidade Federal do Rio Grande do Sul, Street Ramiro Barcelos, 2600-Annex, CEP 90035-003 Porto Alegre, RS, Brazil; Program of Post-graduation in Biological Sciences-Biochemistry, Biochemistry Department, ICBS, Universidade Federal do Rio Grande do Sul, Street Ramiro Barcelos, 2600-Annex, CEP 90035-003 Porto Alegre, RS, Brazil.
| |
Collapse
|
26
|
Marques EP, Wyse ATS. Creatine as a Neuroprotector: an Actor that Can Play Many Parts. Neurotox Res 2019; 36:411-423. [PMID: 31069754 DOI: 10.1007/s12640-019-00053-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 04/12/2019] [Accepted: 04/23/2019] [Indexed: 12/11/2022]
Abstract
Creatine is a nitrogenous organic acid that plays a central role as an energy buffer in high energy demanding systems, including the muscular and the central nervous system. It can be acquired from diet or synthesized endogenously, and its main destination is the system creatine/phosphocreatine that strengthens cellular energetics via a temporal and spatial energy buffer that can restore cellular ATP without a reliance on oxygen. This compound has been proposed to possess secondary roles, such as direct and indirect antioxidant, immunomodulatory agent, and possible neuromodulator. However, these effects may be associated with its bioenergetic role in the mitochondria. Given the fundamental roles that creatine plays in the CNS, several preclinical and clinical studies have tested the potential that creatine has to treat degenerative disorders. However, although in vitro and in vivo animal models are highly encouraging, most clinical trials fail to reproduce positive results suggesting that the prophylactic use for neuroprotection in at-risk populations or patients is the most promising field. Nonetheless, the only clearly positive data of the creatine supplementation in human beings are related to the (rare) creatine deficiency syndromes. It seems critical that future studies must establish the best dosage regime to increase brain creatine in a way that can relate to animal studies, provide new ways for creatine to reach the brain, and seek larger experimental groups with biomarkers for prediction of efficacy.
Collapse
Affiliation(s)
- Eduardo Peil Marques
- Laboratory of Neuroprotection and Metabolic Disease, Biochemistry Department, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, RS, 90035-003, Brazil
- Post graduate program in Biological Science - Biochemistry, Biochemistry Department, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, RS, 90035-003, Brazil
| | - Angela T S Wyse
- Laboratory of Neuroprotection and Metabolic Disease, Biochemistry Department, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, RS, 90035-003, Brazil.
- Post graduate program in Biological Science - Biochemistry, Biochemistry Department, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre, RS, 90035-003, Brazil.
| |
Collapse
|
27
|
Petrella LI, Castelhano JM, Ribeiro M, Sereno JV, Gonçalves SI, Laço MN, Hayden MR, Rego AC, Castelo-Branco M. A whole brain longitudinal study in the YAC128 mouse model of Huntington's disease shows distinct trajectories of neurochemical, structural connectivity and volumetric changes. Hum Mol Genet 2019; 27:2125-2137. [PMID: 29668904 DOI: 10.1093/hmg/ddy119] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 03/26/2018] [Indexed: 12/20/2022] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disorder causing cognitive and motor impairments, evolving to death within 15-20 years after symptom onset. We previously established a mouse model with the entire human HD gene containing 128 CAG repeats (YAC128) which accurately recapitulates the natural history of the human disease. Defined time points in this natural history enable the understanding of longitudinal trajectories from the neurochemical and structural points of view using non-invasive high-resolution multi-modal imaging. Accordingly, we designed a longitudinal structural imaging (MRI and DTI) and spectroscopy (1H-MRS) study in YAC128, at 3, 6, 9 and 12 months of age, at 9.4 T. Structural analysis (MRI/DTI), confirmed that the striatum is the earliest affected brain region, but other regions were also identified through connectivity analysis (pre-frontal cortex, hippocampus, globus pallidus and thalamus), suggesting a striking homology with the human disease. Importantly, we found for the first time, a negative correlation between striatal and hippocampal changes only in YAC128. In fact, the striatum showed accelerated volumetric decay in HD, as opposed to the hippocampus. Neurochemical analysis of the HD striatum suggested early neurometabolic alterations in neurotransmission and metabolism, with a significant increase in striatal GABA levels, and specifically anticorrelated levels of N-acetyl aspartate and taurine, suggesting that the later is homeostatically adjusted for neuroprotection, as neural loss, indicated by the former, is progressing. These results provide novel insights into the natural history of HD and prove a valuable role for longitudinal multi-modal panels of structural and metabolite/neurotransmission in the YAC128 model.
Collapse
Affiliation(s)
- Lorena I Petrella
- Institute of Nuclear Science Applied to Health, University of Coimbra, 3000-548 Coimbra, Portugal.,Center for Neuroscience and Cell Biology-Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, 3000-548 Coimbra, Portugal
| | - João M Castelhano
- Institute of Nuclear Science Applied to Health, University of Coimbra, 3000-548 Coimbra, Portugal.,Center for Neuroscience and Cell Biology-Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Mario Ribeiro
- Institute of Nuclear Science Applied to Health, University of Coimbra, 3000-548 Coimbra, Portugal.,Center for Neuroscience and Cell Biology-Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, 3000-548 Coimbra, Portugal
| | - José V Sereno
- Institute of Nuclear Science Applied to Health, University of Coimbra, 3000-548 Coimbra, Portugal.,Center for Neuroscience and Cell Biology-Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Sónia I Gonçalves
- Institute of Nuclear Science Applied to Health, University of Coimbra, 3000-548 Coimbra, Portugal.,Center for Neuroscience and Cell Biology-Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, 3000-548 Coimbra, Portugal.,Neuroplasticity and Neural Activity Laboratory, Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal
| | - Mário N Laço
- Center for Neuroscience and Cell Biology-Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Michael R Hayden
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | - A Cristina Rego
- Center for Neuroscience and Cell Biology-Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, 3000-548 Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Miguel Castelo-Branco
- Institute of Nuclear Science Applied to Health, University of Coimbra, 3000-548 Coimbra, Portugal.,Center for Neuroscience and Cell Biology-Institute of Biomedical Imaging and Life Science (CNC.IBILI), University of Coimbra, 3000-548 Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| |
Collapse
|
28
|
Abstract
This review systematically examines the evidence for shifts in flux through energy generating biochemical pathways in Huntington’s disease (HD) brains from humans and model systems. Compromise of the electron transport chain (ETC) appears not to be the primary or earliest metabolic change in HD pathogenesis. Rather, compromise of glucose uptake facilitates glucose flux through glycolysis and may possibly decrease flux through the pentose phosphate pathway (PPP), limiting subsequent NADPH and GSH production needed for antioxidant protection. As a result, oxidative damage to key glycolytic and tricarboxylic acid (TCA) cycle enzymes further restricts energy production so that while basal needs may be met through oxidative phosphorylation, those of excessive stimulation cannot. Energy production may also be compromised by deficits in mitochondrial biogenesis, dynamics or trafficking. Restrictions on energy production may be compensated for by glutamate oxidation and/or stimulation of fatty acid oxidation. Transcriptional dysregulation generated by mutant huntingtin also contributes to energetic disruption at specific enzymatic steps. Many of the alterations in metabolic substrates and enzymes may derive from normal regulatory feedback mechanisms and appear oscillatory. Fine temporal sequencing of the shifts in metabolic flux and transcriptional and expression changes associated with mutant huntingtin expression remain largely unexplored and may be model dependent. Differences in disease progression among HD model systems at the time of experimentation and their varying states of metabolic compensation may explain conflicting reports in the literature. Progressive shifts in metabolic flux represent homeostatic compensatory mechanisms that maintain the model organism through presymptomatic and symptomatic stages.
Collapse
Affiliation(s)
- Janet M Dubinsky
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
29
|
Keuken MC, Isaacs BR, Trampel R, van der Zwaag W, Forstmann BU. Visualizing the Human Subcortex Using Ultra-high Field Magnetic Resonance Imaging. Brain Topogr 2018; 31:513-545. [PMID: 29497874 PMCID: PMC5999196 DOI: 10.1007/s10548-018-0638-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 01/28/2018] [Indexed: 12/15/2022]
Abstract
With the recent increased availability of ultra-high field (UHF) magnetic resonance imaging (MRI), substantial progress has been made in visualizing the human brain, which can now be done in extraordinary detail. This review provides an extensive overview of the use of UHF MRI in visualizing the human subcortex for both healthy and patient populations. The high inter-subject variability in size and location of subcortical structures limits the usability of atlases in the midbrain. Fortunately, the combined results of this review indicate that a large number of subcortical areas can be visualized in individual space using UHF MRI. Current limitations and potential solutions of UHF MRI for visualizing the subcortex are also discussed.
Collapse
Affiliation(s)
- M C Keuken
- Integrative Model-Based Cognitive Neuroscience Research Unit, University of Amsterdam, Postbus 15926, 1001NK, Amsterdam, The Netherlands.
- Cognitive Psychology Unit, Institute of Psychology and Leiden Institute for Brain and Cognition, Leiden University, Leiden, The Netherlands.
| | - B R Isaacs
- Integrative Model-Based Cognitive Neuroscience Research Unit, University of Amsterdam, Postbus 15926, 1001NK, Amsterdam, The Netherlands
- Maastricht University Medical Center, Maastricht, The Netherlands
| | - R Trampel
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | | | - B U Forstmann
- Integrative Model-Based Cognitive Neuroscience Research Unit, University of Amsterdam, Postbus 15926, 1001NK, Amsterdam, The Netherlands
- Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| |
Collapse
|
30
|
Adanyeguh IM, Monin ML, Rinaldi D, Freeman L, Durr A, Lehéricy S, Henry PG, Mochel F. Expanded neurochemical profile in the early stage of Huntington disease using proton magnetic resonance spectroscopy. NMR IN BIOMEDICINE 2018; 31:10.1002/nbm.3880. [PMID: 29315899 PMCID: PMC5841244 DOI: 10.1002/nbm.3880] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 11/13/2017] [Accepted: 11/14/2017] [Indexed: 05/29/2023]
Abstract
The striatum is a well-known region affected in Huntington disease (HD). However, other regions, including the visual cortex, are implicated. We have identified previously an abnormal energy response in the visual cortex of patients at an early stage of HD using 31 P magnetic resonance spectroscopy (31 P MRS). We therefore sought to further characterize these metabolic alterations with 1 H MRS using a well-validated semi-localized by adiabatic selective refocusing (semi-LASER) sequence that allows the measurement of an expanded number of neurometabolites. Ten early affected patients [Unified Huntington Disease Rating Scale (UHDRS), total motor score = 13.6 ± 10.8] and 10 healthy volunteers of similar age and body mass index (BMI) were recruited for the study. We performed 1 H MRS in the striatum - the region that is primarily affected in HD - and the visual cortex. The protocol allowed a reliable quantification of 10 metabolites in the visual cortex and eight in the striatum, compared with three to five metabolites in previous 1 H MRS studies performed in HD. We identified higher total creatine (p < 0.05) in the visual cortex and lower glutamate (p < 0.001) and total creatine (p < 0.05) in the striatum of patients with HD compared with controls. Less abundant neurometabolites [glutamine, γ-aminobutyric acid (GABA), glutathione, aspartate] showed similar concentrations in both groups. The protocol allowed the measurement of several additional metabolites compared with standard vendor protocols. Our study points to early changes in metabolites involved in energy metabolism in the visual cortex and striatum of patients with HD. Decreased striatal glutamate could reflect early neuronal dysfunction or impaired glutamatergic neurotransmission.
Collapse
Affiliation(s)
- Isaac M. Adanyeguh
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, F-75013, Paris, France
| | - Marie-Lorraine Monin
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, F-75013, Paris, France
- AP-HP, Pitié-Salpêtrière University Hospital, Department of Genetics, Paris, France
| | - Daisy Rinaldi
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, F-75013, Paris, France
| | - Léorah Freeman
- Department of Neurology, McGovern Medical School at UTHealth, Houston, TX, Unites States
| | - Alexandra Durr
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, F-75013, Paris, France
- AP-HP, Pitié-Salpêtrière University Hospital, Department of Genetics, Paris, France
| | - Stéphane Lehéricy
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, F-75013, Paris, France
- Center for NeuroImaging Research (CENIR), Institut du Cerveau et de la Moelle épinière, Paris, France
| | - Pierre-Gilles Henry
- Center for Magnetic Resonance Research (CMRR), University of Minnesota, Minneapolis, MN, United States
| | - Fanny Mochel
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, F-75013, Paris, France
- AP-HP, Pitié-Salpêtrière University Hospital, Department of Genetics, Paris, France
- University Pierre and Marie Curie, Neurometabolic Research Group, Paris, France
| |
Collapse
|
31
|
Trattnig S, Springer E, Bogner W, Hangel G, Strasser B, Dymerska B, Cardoso PL, Robinson SD. Key clinical benefits of neuroimaging at 7T. Neuroimage 2018; 168:477-489. [PMID: 27851995 PMCID: PMC5832016 DOI: 10.1016/j.neuroimage.2016.11.031] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 10/06/2016] [Accepted: 11/12/2016] [Indexed: 01/15/2023] Open
Abstract
The growing interest in ultra-high field MRI, with more than 35.000 MR examinations already performed at 7T, is related to improved clinical results with regard to morphological as well as functional and metabolic capabilities. Since the signal-to-noise ratio increases with the field strength of the MR scanner, the most evident application at 7T is to gain higher spatial resolution in the brain compared to 3T. Of specific clinical interest for neuro applications is the cerebral cortex at 7T, for the detection of changes in cortical structure, like the visualization of cortical microinfarcts and cortical plaques in Multiple Sclerosis. In imaging of the hippocampus, even subfields of the internal hippocampal anatomy and pathology may be visualized with excellent spatial resolution. Using Susceptibility Weighted Imaging, the plaque-vessel relationship and iron accumulations in Multiple Sclerosis can be visualized, which may provide a prognostic factor of disease. Vascular imaging is a highly promising field for 7T which is dealt with in a separate dedicated article in this special issue. The static and dynamic blood oxygenation level-dependent contrast also increases with the field strength, which significantly improves the accuracy of pre-surgical evaluation of vital brain areas before tumor removal. Improvement in acquisition and hardware technology have also resulted in an increasing number of MR spectroscopic imaging studies in patients at 7T. More recent parallel imaging and short-TR acquisition approaches have overcome the limitations of scan time and spatial resolution, thereby allowing imaging matrix sizes of up to 128×128. The benefits of these acquisition approaches for investigation of brain tumors and Multiple Sclerosis have been shown recently. Together, these possibilities demonstrate the feasibility and advantages of conducting routine diagnostic imaging and clinical research at 7T.
Collapse
Affiliation(s)
- Siegfried Trattnig
- High Field MR Center, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Lazarettgasse 14, A-1090, Vienna, Austria; Christian Doppler Laboratory for Clinical Molecular MRI, Vienna, Austria.
| | - Elisabeth Springer
- High Field MR Center, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Lazarettgasse 14, A-1090, Vienna, Austria; Christian Doppler Laboratory for Clinical Molecular MRI, Vienna, Austria.
| | - Wolfgang Bogner
- High Field MR Center, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Lazarettgasse 14, A-1090, Vienna, Austria.
| | - Gilbert Hangel
- High Field MR Center, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Lazarettgasse 14, A-1090, Vienna, Austria.
| | - Bernhard Strasser
- High Field MR Center, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Lazarettgasse 14, A-1090, Vienna, Austria.
| | - Barbara Dymerska
- High Field MR Center, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Lazarettgasse 14, A-1090, Vienna, Austria.
| | - Pedro Lima Cardoso
- High Field MR Center, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Lazarettgasse 14, A-1090, Vienna, Austria.
| | - Simon Daniel Robinson
- High Field MR Center, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Lazarettgasse 14, A-1090, Vienna, Austria.
| |
Collapse
|
32
|
Sawiak SJ, Wood NI, Morton AJ. Similar Progression of Morphological and Metabolic Phenotype in R6/2 Mice with Different CAG Repeats Revealed by In Vivo Magnetic Resonance Imaging and Spectroscopy. J Huntingtons Dis 2017; 5:271-283. [PMID: 27662335 DOI: 10.3233/jhd-160208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Huntington's disease (HD) is caused by an unstable polyglutamine (CAG) repeat in the HD gene, whereby a CAG repeat length greater than ∼36 leads to the disease. In HD patients, longer repeats correlate with more severe disease and earlier death. This is also seen in R6/2 mice carrying repeat lengths up to ∼200. Paradoxically, R6/2 mice with repeat lengths >300 have a less aggressive phenotype and longer lifespan than those with shorter repeats. The mechanism underlying this phenomenon is unknown. OBJECTIVE To investigate the consequences of longer repeat lengths on structural changes in the brains of R6/2 mice, especially with regard to progressive atrophy. METHODS We used longitudinal in vivo magnetic resonance imaging (MRI) and spectroscopy (MRS) to compare pathological changes in two strains of R6/2 mice, one with a rapidly progressing disease (250 CAG repeats), and the other with a less aggressive phenotype (350 CAG repeats). RESULTS We found significant progressive brain atrophy in both 250 and 350 CAG repeat mice, as well as changes in metabolites (glutamine/glutamate, choline and aspartate). Although similar in magnitude, atrophy in the brains of 350 CAG R6/2 mice progressed more slowly than that seen in 250 CAG mice, in line with the milder phenotype and longer lifespan. Interestingly, significant atrophy was detectable in 350 CAG mice as early as 8-12 weeks of age, although behavioural abnormalities in these mice are not apparent before 25-30 weeks. This finding fits well with human data from the PREDICT-HD and TRACK-HD project, where reductions in brain volume were found 10 years in advance of the onset of symptoms. CONCLUSIONS The similar brain atrophy with a mismatch between onset of brain atrophy and behavioural phenotype in HD mice with 350 repeats will make this mouse particularly useful for modelling early stages of HD pathology.
Collapse
Affiliation(s)
- Stephen J Sawiak
- Wolfson Brain Imaging Centre, University of Cambridge, Box 65 Addenbrooke's Hospital, Cambridge, UK.,Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, UK
| | - Nigel I Wood
- Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - A Jennifer Morton
- Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| |
Collapse
|
33
|
Ehrlich DJ, Walker RH. Functional neuroimaging and chorea: a systematic review. JOURNAL OF CLINICAL MOVEMENT DISORDERS 2017. [PMID: 28649394 PMCID: PMC5479019 DOI: 10.1186/s40734-017-0056-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Chorea is a hyperkinetic movement disorder consisting of involuntary irregular, flowing movements of the trunk, neck or face. Although Huntington’s disease is the most common cause of chorea in adults, chorea can also result from many other neurodegenerative, metabolic, and autoimmune conditions. While the pathophysiology of these different conditions is quite variable, recent advances in functional imaging have enabled the development of new methods for analysis of brain activity and neuronal dysfunction. In this paper we review the growing body of functional imaging data that has been performed in chorea syndromes and identify particular trends, which can be used to better understand the underlying network changes within the basal ganglia. While it can be challenging to identify whether changes are primary, secondary, or compensatory, identification of these trends can ultimately be useful in diagnostic testing and treatment in many of the conditions that cause chorea.
Collapse
Affiliation(s)
- Debra J Ehrlich
- Department of Neurology, Icahn School of Medicine at Mount Sinai, 5 East 98th Street, 1st Floor, Box 1637, New York, NY 10029 USA
| | - Ruth H Walker
- Department of Neurology, Icahn School of Medicine at Mount Sinai, 5 East 98th Street, 1st Floor, Box 1637, New York, NY 10029 USA.,Department of Neurology, James J Peters Veterans Affairs Medical Center, 130 West Kingsbridge Road, Bronx, NY 10468 USA
| |
Collapse
|
34
|
Abstract
Huntington disease (HD) is an autosomal dominant, neurodegenerative disorder with a primary etiology of striatal pathology. The Huntingtin gene (HTT) has a unique feature of a DNA trinucleotide (triplet) repeat, with repeat length ranging from 10 to 35 in the normal population. Repeat lengths between 36 and 39 cause HD at reduced penetrance (some will get the disease, others won't) and when expanded to 40 or more repeats (mHTT), causes HD at full penetrance (every person with this length or beyond will definitely develop the disease). The symptoms of HD may be motor, cognitive, and psychiatric, and are consistent with the pathophysiology of frontostriatal circuitry malfunction. Expressed ubiquitously and throughout the entire life cycle (development through adulthood), mHTT causes initial dysfunction and eventual death of a specific cell population within the striatum. Although all areas of the brain are eventually affected, the primary pathology of the disease is regionally specific. As a single-gene disorder, HD has the distinction of having the potential of treatment that is aimed directly at the known pathogenic mechanism by gene silencing, providing hope for neuroprotection and ultimately, prevention.
Collapse
Affiliation(s)
- Peggy C Nopoulos
- Department of Psychiatry, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
35
|
Abstract
Metabolic imaging enhances understanding of disease metabolisms and holds great potential as a measurement tool for evaluating disease prognosis and treatment effectiveness. Advancement of techniques, such as magnetic resonance spectroscopy, positron emission tomography, and mass spectrometry, allows for improved accuracy for quantification of metabolites and present unique possibilities for use in clinic. This article reviews and discusses literature reports of metabolic imaging in humans published since 2010 according to disease type, including cancer, degenerative disorders, psychiatric disorders, and others, as well as the current application of the various related techniques.
Collapse
Affiliation(s)
- Taylor L. Fuss
- Departments of Radiology and Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, 02114 USA
| | - Leo L. Cheng
- Departments of Radiology and Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, 02114 USA
- Corresponding Author: Leo L. Cheng, PhD, 149 13 Street, CNY-6, Charlestown, MA 02129, Ph.617-724-6593, Fax.617-726-5684,
| |
Collapse
|
36
|
Patassini S, Begley P, Xu J, Church SJ, Reid SJ, Kim EH, Curtis MA, Dragunow M, Waldvogel HJ, Snell RG, Unwin RD, Faull RLM, Cooper GJS. Metabolite mapping reveals severe widespread perturbation of multiple metabolic processes in Huntington's disease human brain. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1862:1650-62. [PMID: 27267344 DOI: 10.1016/j.bbadis.2016.06.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 05/31/2016] [Accepted: 06/01/2016] [Indexed: 01/01/2023]
Abstract
Huntington's disease (HD) is a genetically-mediated neurodegenerative disorder wherein the aetiological defect is a mutation in the Huntington's gene (HTT), which alters the structure of the huntingtin protein (Htt) through lengthening of its polyglutamine tract, thus initiating a cascade that ultimately leads to premature death. However, neurodegeneration typically manifests in HD only in middle age, and mechanisms linking the causative mutation to brain disease are poorly understood. Brain metabolism is severely perturbed in HD, and some studies have indicated a potential role for mutant Htt as a driver of these metabolic aberrations. Here, our objective was to determine the effects of HD on brain metabolism by measuring levels of polar metabolites in regions known to undergo varying degrees of damage. We performed gas-chromatography/mass spectrometry-based metabolomic analyses in a case-control study of eleven brain regions in short post-mortem-delay human tissue from nine well-characterized HD patients and nine matched controls. In each patient, we measured metabolite content in representative tissue-samples from eleven brain regions that display varying degrees of damage in HD, thus identifying the presence and abundance of 63 different metabolites from several molecular classes, including carbohydrates, amino acids, nucleosides, and neurotransmitters. Robust alterations in regional brain-metabolite abundances were observed in HD patients: these included changes in levels of small molecules that play important roles as intermediates in the tricarboxylic-acid and urea cycles, and amino-acid metabolism. Our findings point to widespread disruption of brain metabolism and indicate a complex phenotype beyond the gradient of neuropathologic damage observed in HD brain.
Collapse
Affiliation(s)
- Stefano Patassini
- School of Biological Sciences, Faculty of Science, University of Auckland, Auckland, New Zealand; Centre for Brain Research and Department of Anatomy with Radiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand; Centre for Advanced Discovery and Experimental Therapeutics (CADET), Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester, UK; Institute of Human Development, Faculty of Medical and Human Sciences, The University of Manchester, Manchester, UK.
| | - Paul Begley
- Centre for Advanced Discovery and Experimental Therapeutics (CADET), Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester, UK; Institute of Human Development, Faculty of Medical and Human Sciences, The University of Manchester, Manchester, UK
| | - Jingshu Xu
- School of Biological Sciences, Faculty of Science, University of Auckland, Auckland, New Zealand; Centre for Brain Research and Department of Anatomy with Radiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand; Centre for Advanced Discovery and Experimental Therapeutics (CADET), Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester, UK; Institute of Human Development, Faculty of Medical and Human Sciences, The University of Manchester, Manchester, UK; Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Stephanie J Church
- Centre for Advanced Discovery and Experimental Therapeutics (CADET), Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester, UK; Institute of Human Development, Faculty of Medical and Human Sciences, The University of Manchester, Manchester, UK
| | - Suzanne J Reid
- School of Biological Sciences, Faculty of Science, University of Auckland, Auckland, New Zealand
| | - Eric H Kim
- Centre for Brain Research and Department of Anatomy with Radiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand; Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, United States
| | - Maurice A Curtis
- Centre for Brain Research and Department of Anatomy with Radiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Mike Dragunow
- Centre for Brain Research and Department of Anatomy with Radiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Henry J Waldvogel
- Centre for Brain Research and Department of Anatomy with Radiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Russell G Snell
- School of Biological Sciences, Faculty of Science, University of Auckland, Auckland, New Zealand; Centre for Brain Research and Department of Anatomy with Radiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Richard D Unwin
- Centre for Advanced Discovery and Experimental Therapeutics (CADET), Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester, UK; Institute of Human Development, Faculty of Medical and Human Sciences, The University of Manchester, Manchester, UK
| | - Richard L M Faull
- Centre for Brain Research and Department of Anatomy with Radiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Garth J S Cooper
- School of Biological Sciences, Faculty of Science, University of Auckland, Auckland, New Zealand; Centre for Brain Research and Department of Anatomy with Radiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand; Centre for Advanced Discovery and Experimental Therapeutics (CADET), Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Sciences Centre, Manchester, UK; Institute of Human Development, Faculty of Medical and Human Sciences, The University of Manchester, Manchester, UK; Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
37
|
The effects of creatine supplementation on thermoregulation and physical (cognitive) performance: a review and future prospects. Amino Acids 2016; 48:1843-55. [PMID: 27085634 DOI: 10.1007/s00726-016-2237-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 04/11/2016] [Indexed: 12/12/2022]
Abstract
Creatine (Cr) is produced endogenously in the liver or obtained exogenously from foods, such as meat and fish. In the human body, 95 % of Cr is located in the cytoplasm of skeletal muscle either in a phosphorylated (PCr) or free form (Cr). PCr is essential for the immediate rephosphorylation of adenosine diphosphate to adenosine triphosphate. PCr is rapidly degraded at the onset of maximal exercise at a rate that results in muscle PCr reservoirs being substantially depleted. A well-established strategy followed to increase muscle total Cr content is to increase exogenous intake by supplementation with chemically pure synthetic Cr. Most Cr supplementation regimens typically follow a well-established loading protocol of 20 g day(-1) of Cr for approximately 5-7 days, followed by a maintenance dose at between 2 and 5 g day(-1) for the duration of interest, although more recent studies tend to utilize a 0.3-g kg(-1) day(-1) supplementation regimen. Some studies have also investigated long-term supplementation of up to 1 year. Uptake of Cr is enhanced when taken together with carbohydrate and protein and/or while undertaking exercise. Cr supplementation has been shown to augment muscle total Cr content and enhance anaerobic performance; however, there is also some evidence of indirect benefits to aerobic endurance exercise through enhanced thermoregulation. While there is an abundance of data supporting the ergogenic effects of Cr supplementation in a variety of different applications, some individuals do not respond, the efficacy of which is dependent on a number of factors, such as dose, age, muscle fiber type, and diet, although further work in this field is warranted. Cr is increasingly being used in the management of some clinical conditions to enhance muscle mass and strength. The application of Cr in studies of health and disease has widened recently with encouraging results in studies involving sleep deprivation and cognitive performance.
Collapse
|
38
|
Characterization of Behavioral, Neuropathological, Brain Metabolic and Key Molecular Changes in zQ175 Knock-In Mouse Model of Huntington's Disease. PLoS One 2016; 11:e0148839. [PMID: 26859386 PMCID: PMC4747517 DOI: 10.1371/journal.pone.0148839] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 01/25/2016] [Indexed: 11/19/2022] Open
Abstract
Huntington’s disease (HD) is caused by an expansion of the trinucleotide poly (CAG) tract located in exon 1 of the huntingtin (Htt) gene leading to progressive neurodegeneration in selected brain regions, and associated functional impairments in motor, cognitive, and psychiatric domains. Since the discovery of the gene mutation that causes the disease, mouse models have been developed by different strategies. Recently, a new model, the zQ175 knock-in (KI) line, was developed in an attempt to have the Htt gene in a context and causing a phenotype that more closely mimics HD in humans. The behavioral phenotype was characterized across the independent laboratories and important features reminiscent of human HD are observed in zQ175 mice. In the current study, we characterized the zQ175 model housed in an academic laboratory under reversed dark-light cycle, including motor function, in vivo longitudinal structural MRI imaging for brain volume, MRS for striatal metabolites, neuropathology, as well as a panel of key disease marker proteins in the striatum at different ages. Our results suggest that homozygous zQ175 mice exhibited significant brain atrophy before the motor deficits and brain metabolite changes. Altered striatal medium spiny neuronal marker, postsynaptic marker protein and complement component C1qC also characterized zQ175 mice. Our results confirmed that the zQ175 KI model is valuable in understanding of HD-like pathophysiology and evaluation of potential therapeutics. Our data also provide suggestions to select appropriate outcome measurements in preclinical studies using the zQ175 mice.
Collapse
|
39
|
Abstract
Movement disorders can be hypokinetic (e.g., parkinsonism), hyperkinetic, or dystonic in nature and commonly arise from altered function in nuclei of the basal ganglia or their connections. As obvious structural changes are often limited, standard imaging plays less of a role than in other neurologic disorders. However, structural imaging is indicated where clinical presentation is atypical, particularly if the disorder is abrupt in onset or remains strictly unilateral. More recent advances in magnetic resonance imaging (MRI) may allow for differentiation between Parkinson's disease and atypical forms of parkinsonism. Functional imaging can assess regional cerebral blood flow (functional MRI (fMRI), positron emission tomography (PET), or single-photon emission computed tomography (SPECT)), cerebral glucose metabolism (PET), neurochemical and neuroreceptor status (PET and SPECT), and pathologic processes such as inflammation or abnormal protein deposition (PET) (Table 49.1). Cerebral blood flow can be assessed at rest, during the performance of motor or cognitive tasks, or in response to a variety of stimuli. In appropriate situations, the correct imaging modality and/or combination of modalities can be used to detect early disease or even preclinical disease, and to monitor disease progression and the effects of disease-modifying interventions. Various approaches are reviewed here.
Collapse
Affiliation(s)
- A Jon Stoessl
- Pacific Parkinson's Research Centre and Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia and Vancouver Coastal Health, Vancouver, BC, Canada.
| | - Martin J Mckeown
- Pacific Parkinson's Research Centre and Division of Neurology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia and Vancouver Coastal Health, Vancouver, BC, Canada
| |
Collapse
|
40
|
In Vivo NMR Studies of the Brain with Hereditary or Acquired Metabolic Disorders. Neurochem Res 2015; 40:2647-85. [PMID: 26610379 DOI: 10.1007/s11064-015-1772-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Revised: 11/10/2015] [Accepted: 11/12/2015] [Indexed: 01/09/2023]
Abstract
Metabolic disorders, whether hereditary or acquired, affect the brain, and abnormalities of the brain are related to cellular integrity; particularly in regard to neurons and astrocytes as well as interactions between them. Metabolic disturbances lead to alterations in cellular function as well as microscopic and macroscopic structural changes in the brain with diabetes, the most typical example of metabolic disorders, and a number of hereditary metabolic disorders. Alternatively, cellular dysfunction and degeneration of the brain lead to metabolic disturbances in hereditary neurological disorders with neurodegeneration. Nuclear magnetic resonance (NMR) techniques allow us to assess a range of pathophysiological changes of the brain in vivo. For example, magnetic resonance spectroscopy detects alterations in brain metabolism and energetics. Physiological magnetic resonance imaging (MRI) detects accompanying changes in cerebral blood flow related to neurovascular coupling. Diffusion and T1/T2-weighted MRI detect microscopic and macroscopic changes of the brain structure. This review summarizes current NMR findings of functional, physiological and biochemical alterations within a number of hereditary and acquired metabolic disorders in both animal models and humans. The global view of the impact of these metabolic disorders on the brain may be useful in identifying the unique and/or general patterns of abnormalities in the living brain related to the pathophysiology of the diseases, and identifying future fields of inquiry.
Collapse
|
41
|
Jozefovicova M, Herynek V, Jiru F, Dezortova M, Juhasova J, Juhas S, Motlik J, Hajek M. Minipig model of Huntington's disease: ¹H magnetic resonance spectroscopy of the brain. Physiol Res 2015; 65:155-63. [PMID: 26596319 DOI: 10.33549/physiolres.932967] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Huntington's disease (HD) is an inherited autosomal neurodegenerative disorder affecting predominantly the brain, characterized by motor dysfunctions, behavioral and cognitive disturbances. The aim of this study was to determine changes in the brain of transgenic minipigs before HD onset using (1)H magnetic resonance (MR) spectroscopy. Measurements were performed on a 3 T MR scanner using a single voxel spectroscopy sequence for spectra acquisition in the white matter and chemical shift imaging sequence for measurement in the striatum, hippocampus and thalamus. A decrease of (phospho)creatine (tCr) concentration was found only in the thalamus (p=0.002) of transgenic minipigs, nevertheless we found significant changes in metabolite ratios. Increase of the ratio choline compounds (tCho)/tCr was found in all examined areas: striatum (p=0.010), thalamus (p=0.011) as well as hippocampus (p=0.027). The ratio N-acetylaspartate+N-acetylaspartylglutamate (tNAA)/tCr (p=0.043) and glutamate+glutamine (Glx)/tCr (p=0.039) was elevated in the thalamus, the ratio myo-inositol (Ins)/tCr (p=0.048) was significantly increased in the hippocampus. No significant differences were observed in the metabolite concentrations in the white matter, however we found significant increase of ratios tNAA/tCr (p=0.018) and tCho/tCr (p=0.003) ratios in transgenic boars. We suppose that the majority of the observed changes are predominantly related to changes in energy metabolism caused by decrease of tCr.
Collapse
Affiliation(s)
- M Jozefovicova
- Magnetic Resonance Unit, Department of Diagnostic and Interventional Radiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic, Department of NMR Spectroscopy and Mass Spectroscopy, Faculty of Chemical and Food Technology, Slovak University of Technology, Bratislava, Slovak Republic.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Creatine supplementation enhances corticomotor excitability and cognitive performance during oxygen deprivation. J Neurosci 2015; 35:1773-80. [PMID: 25632150 DOI: 10.1523/jneurosci.3113-14.2015] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Impairment or interruption of oxygen supply compromises brain function and plays a role in neurological and neurodegenerative conditions. Creatine is a naturally occurring compound involved in the buffering, transport, and regulation of cellular energy, with the potential to replenish cellular adenosine triphosphate without oxygen. Creatine is also neuroprotective in vitro against anoxic/hypoxic damage. Dietary creatine supplementation has been associated with improved symptoms in neurological disorders defined by impaired neural energy provision. Here we investigate, for the first time in humans, the utility of creatine as a dietary supplement to protect against energetic insult. The aim of this study was to assess the influence of oral creatine supplementation on the neurophysiological and neuropsychological function of healthy young adults during acute oxygen deprivation. Fifteen healthy adults were supplemented with creatine and placebo treatments for 7 d, which increased brain creatine on average by 9.2%. A hypoxic gas mixture (10% oxygen) was administered for 90 min, causing global oxygen deficit and impairing a range of neuropsychological processes. Hypoxia-induced decrements in cognitive performance, specifically attentional capacity, were restored when participants were creatine supplemented, and corticomotor excitability increased. A neuromodulatory effect of creatine via increased energy availability is presumed to be a contributing factor of the restoration, perhaps by supporting the maintenance of appropriate neuronal membrane potentials. Dietary creatine monohydrate supplementation augments neural creatine, increases corticomotor excitability, and prevents the decline in attention that occurs during severe oxygen deficit. This is the first demonstration of creatine's utility as a neuroprotective supplement when cellular energy provision is compromised.
Collapse
|
43
|
Jin J, Peng Q, Hou Z, Jiang M, Wang X, Langseth AJ, Tao M, Barker PB, Mori S, Bergles DE, Ross CA, Detloff PJ, Zhang J, Duan W. Early white matter abnormalities, progressive brain pathology and motor deficits in a novel knock-in mouse model of Huntington's disease. Hum Mol Genet 2015; 24:2508-27. [PMID: 25609071 DOI: 10.1093/hmg/ddv016] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 01/19/2015] [Indexed: 12/21/2022] Open
Abstract
White matter abnormalities have been reported in premanifest Huntington's disease (HD) subjects before overt striatal neuronal loss, but whether the white matter changes represent a necessary step towards further pathology and the underlying mechanism of these changes remains unknown. Here, we characterized a novel knock-in mouse model that expresses mouse HD gene homolog (Hdh) with extended CAG repeat- HdhQ250, which was derived from the selective breeding of HdhQ150 mice. HdhQ250 mice manifest an accelerated and robust phenotype compared with its parent line. HdhQ250 mice exhibit progressive motor deficits, reduction in striatal and cortical volume, accumulation of mutant huntingtin aggregation, decreased levels of DARPP32 and BDNF and altered striatal metabolites. The abnormalities detected in this mouse model are reminiscent of several aspects of human HD. In addition, disturbed myelination was evident in postnatal Day 14 HdhQ250 mouse brain, including reduced levels of myelin regulatory factor and myelin basic protein, and decreased numbers of myelinated axons in the corpus callosum. Thinner myelin sheaths, indicated by increased G-ratio of myelin, were also detected in the corpus callosum of adult HdhQ250 mice. Moreover, proliferation of oligodendrocyte precursor cells is altered by mutant huntingtin both in vitro and in vivo. Our data indicate that this model is suitable for understanding comprehensive pathogenesis of HD in white matter and gray matter as well as developing therapeutics for HD.
Collapse
Affiliation(s)
- Jing Jin
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences
| | - Qi Peng
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences
| | | | - Mali Jiang
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences
| | - Xin Wang
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY 10065, USA
| | | | - Michael Tao
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences
| | | | | | | | - Christopher A Ross
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA, Department of Neuroscience, Department of Neurology and Pharmacology and Molecular Sciences and
| | - Peter J Detloff
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35242, USA
| | | | - Wenzhen Duan
- Division of Neurobiology, Department of Psychiatry and Behavioral Sciences, Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA, Department of Neuroscience,
| |
Collapse
|
44
|
Aylward EH, Harrington DL, Mills JA, Nopoulos PC, Ross CA, Long JD, Liu D, Westervelt HK, Paulsen JS. Regional atrophy associated with cognitive and motor function in prodromal Huntington disease. J Huntingtons Dis 2014; 2:477-89. [PMID: 25062732 DOI: 10.3233/jhd-130076] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Neuroimaging studies suggest that volumetric MRI measures of specific brain structures may serve as excellent biomarkers in future clinical trials of Huntington disease (HD). OBJECTIVE Demonstration of the clinical significance of these measures is an important step in determining their appropriateness as potential outcome measures. METHODS Measures of gray- and white-matter lobular volumes and subcortical volumes (caudate, putamen, globus pallidus, thalamus, nucleus accumbens, hippocampus) were obtained from MRI scans of 516 individuals who tested positive for the HD gene expansion, but were not yet exhibiting signs or symptoms severe enough to warrant diagnosis ("pre-HD"). MRI volumes (corrected for intracranial volume) were correlated with cognitive, motor, psychiatric, and functional measures known to be sensitive to subtle changes in pre-HD. RESULTS Caudate, putamen, and globus pallidus volumes consistently correlated with cognitive and motor, but not psychiatric or functional measures in pre-HD. Volumes of white matter, nucleus accumbens, and thalamus, but not cortical gray matter, also correlated with some of the motor and cognitive measures. CONCLUSIONS Results of regression analyses suggest that volumes of basal ganglia structures contributed more highly to the prediction of most motor and cognitive variables than volumes of other brain regions. These results support the use of volumetric measures, especially of the basal ganglia, as outcome measures in future clinical trials in pre-HD. Results may also assist investigators in selecting the most appropriate measures for treatment trials that target specific clinical features or regions of neuropathology.
Collapse
Affiliation(s)
- Elizabeth H Aylward
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Deborah L Harrington
- Department of Radiology, University of California, San Diego, La Jolla, CA, USA VA San Diego Healthcare System, Research Service, San Diego, CA, USA
| | - James A Mills
- Department of Psychiatry, The University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Peggy C Nopoulos
- Department of Psychiatry, The University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Christopher A Ross
- Departments of Psychiatry, Neurology and Neuroscience, Johns Hopkins University, Baltimore, MD, USA
| | - Jeffrey D Long
- Department of Psychiatry, The University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Dawei Liu
- Department of Psychiatry, The University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Holly K Westervelt
- Division of Biology and Medicine, Department of Psychiatry and Human Behavior, Brown University, Providence, RI, USA
| | - Jane S Paulsen
- Departments of Psychiatry, Neurology, Psychology and Neuroscience, The University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | | |
Collapse
|
45
|
Abstract
A plethora of magnetic resonance (MR) techniques developed in the last two decades provide unique and noninvasive measurement capabilities for studies of basic brain function and brain diseases in humans. Animal model experiments have been an indispensible part of this development. MR imaging and spectroscopy measurements have been employed in animal models, either by themselves or in combination with complementary and often invasive techniques, to enlighten us about the information content of such MR methods and/or verify observations made in the human brain. They have also been employed, with or independently of human efforts, to examine mechanisms underlying pathological developments in the brain, exploiting the wealth of animal models available for such studies. In this endeavor, the desire to push for ever-higher spatial and/or spectral resolution, better signal-to-noise ratio, and unique image contrast has inevitably led to the introduction of increasingly higher magnetic fields. As a result, today, animal model studies are starting to be conducted at magnetic fields ranging from ~ 11 to 17 Tesla, significantly enhancing the armamentarium of tools available for the probing brain function and brain pathologies.
Collapse
Affiliation(s)
- Gülin Öz
- Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, Minnesota, USA
| | | | | |
Collapse
|
46
|
Andre R, Scahill RI, Haider S, Tabrizi SJ. Biomarker development for Huntington's disease. Drug Discov Today 2014; 19:972-9. [PMID: 24632006 DOI: 10.1016/j.drudis.2014.03.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 03/04/2014] [Indexed: 02/02/2023]
Abstract
Huntington's disease (HD) is a fatal inherited neurodegenerative disorder, treatment to slow the progression of which has not yet been found. Human clinical trials to test a number of therapeutic strategies are underway or imminent, facilitated in part by the recent development of biomarkers that might be used as surrogate endpoints in such trials. However, although much progress in developing HD biomarkers has been made, ongoing work seeks to improve the sensitivity and reliability of current measures, and to demonstrate that they correspond to clear meaningful benefit to patients. Of particular importance is the identification of state biomarkers that can be used in pre-manifest HD gene carriers to test therapies hoped to delay symptom onset in these individuals. Functional, neuroimaging and biochemical biomarkers continue to be investigated for use in the development of disease-modifying treatments of HD.
Collapse
Affiliation(s)
- Ralph Andre
- UCL Institute of Neurology, Department of Neurodegenerative Disease, Queen Square, London WC1N 3BG, UK
| | - Rachael I Scahill
- UCL Institute of Neurology, Department of Neurodegenerative Disease, Queen Square, London WC1N 3BG, UK
| | - Salman Haider
- UCL Institute of Neurology, Department of Neurodegenerative Disease, Queen Square, London WC1N 3BG, UK
| | - Sarah J Tabrizi
- UCL Institute of Neurology, Department of Neurodegenerative Disease, Queen Square, London WC1N 3BG, UK.
| |
Collapse
|
47
|
Padowski JM, Weaver KE, Richards TL, Laurino MY, Samii A, Aylward EH, Conley KE. Neurochemical correlates of caudate atrophy in Huntington's disease. Mov Disord 2014; 29:327-35. [PMID: 24442623 DOI: 10.1002/mds.25801] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 11/19/2013] [Accepted: 12/09/2013] [Indexed: 11/11/2022] Open
Abstract
The precise pathogenic mechanisms of Huntington's disease (HD) are unknown but can be tested in vivo using proton magnetic resonance spectroscopy ((1)H MRS) to measure neurochemical changes. The objective of this study was to evaluate neurochemical differences in HD gene mutation carriers (HGMCs) versus controls and to investigate relationships among function, brain structure, and neurochemistry in HD. Because previous (1)H MRS studies have yielded varied conclusions about HD neurochemical changes, an additional goal was to compare two (1)H MRS data analysis approaches. HGMCs with premanifest to early HD and controls underwent evaluation of motor function, magnetic resonance imaging, and localized (1)H MRS in the caudate and the frontal lobe. Analytical approaches that were tested included absolute quantitation (unsuppressed water signal as an internal reference) and relative quantification (calculating ratios of all neurochemical signals within a voxel). We identified a suite of neurochemicals that were reduced in concentration proportionally to loss of caudate volume in HGMCs. Caudate concentrations of N-acetylaspartate (NAA), creatine, choline, and caudate and frontal lobe concentrations of glutamate plus glutamine (Glx) and glutamate were correlated with caudate volume in HGMCs. The relative, but not the absolute, quantitation approach revealed disease-related differences; the Glx signal was decreased relative to other neurochemicals in the caudate of HGMCs versus controls. This is the first study to demonstrate a correlation among structure, function, and chemical measures in HD brain. Additionally, we demonstrate that a relative quantitation approach may enable the magnification of subtle differences between groups. Observation of decreased Glx suggests that glutamate signaling may be disrupted relatively early in HD, which has important implications for therapeutic approaches.
Collapse
Affiliation(s)
- Jeannie M Padowski
- Department of Radiology, University of Washington School of Medicine, Seattle, Washington, USA; Integrated Brain Imaging Center, University of Washington School of Medicine, Seattle, Washington, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Early deficits in glycolysis are specific to striatal neurons from a rat model of huntington disease. PLoS One 2013; 8:e81528. [PMID: 24303051 PMCID: PMC3841140 DOI: 10.1371/journal.pone.0081528] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 10/14/2013] [Indexed: 11/28/2022] Open
Abstract
In Huntington disease (HD), there is increasing evidence for a link between mutant huntingtin expression, mitochondrial dysfunction, energetic deficits and neurodegeneration but the precise nature, causes and order of these events remain to be determined. In this work, our objective was to evaluate mitochondrial respiratory function in intact, non-permeabilized, neurons derived from a transgenic rat model for HD compared to their wild type littermates by measuring oxygen consumption rates and extracellular acidification rates. Although HD striatal neurons had similar respiratory capacity as those from their wild-type littermates when they were incubated in rich medium containing a supra-physiological glucose concentration (25 mM), pyruvate and amino acids, respiratory defects emerged when cells were incubated in media containing only a physiological cerebral level of glucose (2.5 mM). According to the concept that glucose is not the sole substrate used by the brain for neuronal energy production, we provide evidence that primary neurons can use lactate as well as pyruvate to fuel the mitochondrial respiratory chain. In contrast to glucose, we found no major deficits in HD striatal neurons’ capacity to use pyruvate as a respiratory substrate compared to wild type littermates. Additionally, we used extracellular acidification rates to confirm a reduction in anaerobic glycolysis in the same cells. Interestingly, the metabolic disturbances observed in striatal neurons were not seen in primary cortical neurons, a brain region affected in later stages of HD. In conclusion, our results argue for a dysfunction in glycolysis, which might precede any defects in the respiratory chain itself, and these are early events in the onset of disease.
Collapse
|
49
|
Scahill R. Recent advances in imaging the onset and progression of Huntington’s disease. Neurodegener Dis Manag 2013. [DOI: 10.2217/nmt.13.19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
SUMMARY Huntington’s disease is a devastating autosomal-dominant neurodegenerative disorder resulting in progressive decline in motor and cognitive function, accompanied by neuropsychiatric disturbances. In vivo imaging can reveal the underlying neuropathological changes that contribute to symptom manifestation. Observational studies of individuals carrying the causative gene have demonstrated that structural and functional brain changes are apparent decades before clinical onset of the disease; imaging measures can predict those individuals who subsequently undergo clinical conversion. Such studies have improved our understanding of neurodegeneration across the disease spectrum and aided the identification of therapeutic targets. Clinical trials of potentially disease-modifying treatments are likely to be investigated in the near future and imaging provides a powerful tool to monitor disease progression and thereby assess therapeutic efficacy.
Collapse
Affiliation(s)
- Rachael Scahill
- Huntington’s Disease Research Group, Department of Neurodegeneration, UCL Institute of Neurology, University College London, London, WC1N 3BG, UK
| |
Collapse
|
50
|
Hall EL, Stephenson MC, Price D, Morris PG. Methodology for improved detection of low concentration metabolites in MRS: optimised combination of signals from multi-element coil arrays. Neuroimage 2013; 86:35-42. [PMID: 23639258 DOI: 10.1016/j.neuroimage.2013.04.077] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 04/16/2013] [Accepted: 04/17/2013] [Indexed: 01/08/2023] Open
Abstract
State of the art magnetic resonance imaging (MRI) scanners are generally equipped with multi-element receive coils; 16 or 32 channel coils are common. Their development has been predominant for parallel imaging to enable faster scanning. Less consideration has been given to localized magnetic resonance spectroscopy (MRS). Multinuclear studies, for example (31)P or (13)C MRS, are often conducted with a single element coil located over the region of interest. (1)H MRS studies have generally employed the same multi-element coils used for MRI, but little consideration has been given as to how the spectroscopic data from the different channels are combined. In many cases it is simply co-added with detrimental effect on the signal to noise ratio. In this study, we derive the optimum method for combining multi-coil data, namely weighting with the ratio of signal to the square of the noise. We show that provided that the noise is uncorrelated, this is the theoretical optimal combination. The method is demonstrated for in vivo proton MRS data acquired using a 32 channel receive coil at 7T in four different brain areas; left motor and right motor, occipital cortex and medial frontal cortex.
Collapse
Affiliation(s)
- Emma L Hall
- Sir Peter Mansfield Magnetic Resonance Centre, School of Physics and Astronomy, University of Nottingham, Nottingham, UK.
| | - Mary C Stephenson
- Sir Peter Mansfield Magnetic Resonance Centre, School of Physics and Astronomy, University of Nottingham, Nottingham, UK
| | - Darren Price
- Sir Peter Mansfield Magnetic Resonance Centre, School of Physics and Astronomy, University of Nottingham, Nottingham, UK
| | - Peter G Morris
- Sir Peter Mansfield Magnetic Resonance Centre, School of Physics and Astronomy, University of Nottingham, Nottingham, UK
| |
Collapse
|