51
|
Sklinda K, Górecki A, Dorobek M, Walecki J, Modrzyńska A, Mruk B. Ischaemic background of brain fog in long-haul COVID-19 - a nuclear magnetic resonance spectroscopy-based metabonomic analysis. Preliminary results. Pol J Radiol 2021; 86:e654-e660. [PMID: 35059058 PMCID: PMC8757010 DOI: 10.5114/pjr.2021.111100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 11/12/2021] [Indexed: 11/25/2022] Open
Abstract
PURPOSE Long-haul COVID-19 is a condition of unknown background occurring in COVID-19 survivors regardless of the severity of the SARS-CoV-2 infection itself. The aim of the study was to evaluate brain changes in patients suffering from variable symptoms of brain fog after COVID-19. MATERIAL AND METHODS Eleven patients hospitalized due to symptoms of severe brain fog, i.e. insomnia, sudden impairment of cognitive function, headache, and depression, and 14 healthy volunteers underwent brain imaging including MR spectroscopy. RESULTS Routine MR imaging revealed no specific macroscopic changes in keeping with brain fog. Considering that the clinical manifestation of brain fog is transient, the evaluation of the metabolic status of the brain remained the method of choice. The concentration of the major cerebral metabolites, i.e. NAA, Cho, and Cr, remained stable. However, changes in Glx and Lac concentration were observed in MR spectroscopy. CONCLUSIONS Following results along with clinical course of the brain for imply probable ischaemic background of symptoms.
Collapse
Affiliation(s)
- Katarzyna Sklinda
- Department od Radiology, Centre of Postgraduate Medical Education, Poland
| | - Andrzej Górecki
- Medical Diagnostic Centre “Voxel”, Medical Hospital, Lancut, Poland
| | - Małgorzata Dorobek
- Department of Neurology, Central Clinical Hospital of Interior and Administration, Poland
| | - Jerzy Walecki
- Department od Radiology, Centre of Postgraduate Medical Education, Poland
| | - Aneta Modrzyńska
- Department of Neurology, Central Clinical Hospital of Interior and Administration, Poland
| | - Bartosz Mruk
- Department od Radiology, Centre of Postgraduate Medical Education, Poland
| |
Collapse
|
52
|
Fairless R, Bading H, Diem R. Pathophysiological Ionotropic Glutamate Signalling in Neuroinflammatory Disease as a Therapeutic Target. Front Neurosci 2021; 15:741280. [PMID: 34744612 PMCID: PMC8567076 DOI: 10.3389/fnins.2021.741280] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/30/2021] [Indexed: 01/15/2023] Open
Abstract
Glutamate signalling is an essential aspect of neuronal communication involving many different glutamate receptors, and underlies the processes of memory, learning and synaptic plasticity. Despite neuroinflammatory diseases covering a range of maladies with very different biological causes and pathophysiologies, a central role for dysfunctional glutamate signalling is becoming apparent. This is not just restricted to the well-described role of glutamate in mediating neurodegeneration, but also includes a myriad of other influences that glutamate can exert on the vasculature, as well as immune cell and glial regulation, reflecting the ability of neurons to communicate with these compartments in order to couple their activity with neuronal requirements. Here, we discuss the role of pathophysiological glutamate signalling in neuroinflammatory disease, using both multiple sclerosis and Alzheimer's disease as examples, and how current steps are being made to harness our growing understanding of these processes in the development of neuroprotective strategies. This review focuses in particular on N-methyl-D-aspartate (NMDA) and 2-amino-3-(3-hydroxy-5-methylisooxazol-4-yl) propionate (AMPA) type ionotropic glutamate receptors, although metabotropic, G-protein-coupled glutamate receptors may also contribute to neuroinflammatory processes. Given the indispensable roles of glutamate-gated ion channels in synaptic communication, means of pharmacologically distinguishing between physiological and pathophysiological actions of glutamate will be discussed that allow deleterious signalling to be inhibited whilst minimising the disturbance of essential neuronal function.
Collapse
Affiliation(s)
- Richard Fairless
- Department of Neurology, University Clinic Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit (CCU) Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hilmar Bading
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Ricarda Diem
- Department of Neurology, University Clinic Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit (CCU) Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
53
|
Stampanoni Bassi M, Nuzzo T, Gilio L, Miroballo M, Casamassa A, Buttari F, Bellantonio P, Fantozzi R, Galifi G, Furlan R, Finardi A, De Rosa A, Di Maio A, Errico F, Centonze D, Usiello A. Cerebrospinal fluid levels of L-glutamate signal central inflammatory neurodegeneration in multiple sclerosis. J Neurochem 2021; 159:857-866. [PMID: 34547109 DOI: 10.1111/jnc.15518] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/15/2021] [Accepted: 09/17/2021] [Indexed: 11/28/2022]
Abstract
Excessive extracellular concentrations of L-glutamate (L-Glu) can be neurotoxic and contribute to neurodegenerative processes in multiple sclerosis (MS). The association between cerebrospinal fluid (CSF) L-Glu levels, clinical features, and inflammatory biomarkers in patients with MS remains unclear. In 179 MS patients (relapsing remitting, RR, N = 157; secondary progressive/primary progressive, SP/PP, N = 22), CSF levels of L-Glu at diagnosis were determined and compared with those obtained in a group of 40 patients with non-inflammatory/non-degenerative disorders. Disability at the time of diagnosis, and after 1 year follow-up, was assessed using the Expanded Disability Status Scale (EDSS). CSF concentrations of lactate and of a large set of pro-inflammatory and anti-inflammatory molecules were explored. CSF levels of L-Glu were slightly reduced in MS patients compared to controls. In RR-MS patients, L-Glu levels correlated with EDSS after 1 year follow-up. Moreover, in MS patients, significant correlations were found between L-Glu and both CSF levels of lactate and the inflammatory molecules interleukin (IL)-2, IL-6, and IL-1 receptor antagonist. Altered expression of L-Glu is associated with disability progression, oxidative stress, and inflammation. These findings identify CSF L-Glu as a candidate neurochemical marker of inflammatory neurodegeneration in MS.
Collapse
Affiliation(s)
| | - Tommaso Nuzzo
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Caserta, Italy.,CEINGE Biotecnologie Avanzate, Naples, Italy
| | - Luana Gilio
- Unit of Neurology, IRCCS Neuromed, Pozzilli (IS), Italy
| | - Mattia Miroballo
- IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | | | - Fabio Buttari
- Unit of Neurology, IRCCS Neuromed, Pozzilli (IS), Italy
| | | | | | | | - Roberto Furlan
- Clinical Neuroimmunology Unit, Division of Neuroscience, Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy
| | - Annamaria Finardi
- Clinical Neuroimmunology Unit, Division of Neuroscience, Institute of Experimental Neurology, San Raffaele Scientific Institute, Milan, Italy
| | - Arianna De Rosa
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Caserta, Italy.,CEINGE Biotecnologie Avanzate, Naples, Italy
| | - Anna Di Maio
- CEINGE Biotecnologie Avanzate, Naples, Italy.,Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Francesco Errico
- CEINGE Biotecnologie Avanzate, Naples, Italy.,Department of Agricultural Sciences, University of Naples "Federico II", Portici, Italy
| | - Diego Centonze
- Unit of Neurology, IRCCS Neuromed, Pozzilli (IS), Italy.,Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Alessandro Usiello
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Caserta, Italy.,CEINGE Biotecnologie Avanzate, Naples, Italy
| |
Collapse
|
54
|
Dahmani S, Kaliss N, VanMeter JW, Moore DJ, Ellis RJ, Jiang X. Alterations of Brain Metabolites in Adults With HIV: A Systematic Meta-analysis of Magnetic Resonance Spectroscopy Studies. Neurology 2021; 97:e1085-e1096. [PMID: 34253633 PMCID: PMC8456358 DOI: 10.1212/wnl.0000000000012394] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 05/20/2021] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE A meta-analysis of proton magnetic resonance spectroscopy studies to investigate alterations in brain metabolites in people with HIV (PWH), the relationship between metabolite alterations and combination antiretroviral therapy (cART), and the relationship between metabolite alterations and cognitive impairment. METHODS The PubMed database was searched for studies published from 1997 to 2020. Twenty-seven studies were identified, which included 1255 PWH and 633 controls. Four metabolites (N-acetyl aspartate [NAA], myo-inositol [mI], choline [Cho], and glutamatergic metabolites [Glx]) from 5 brain regions (basal ganglia [BG], frontal gray and white matter [FGM and FWM], and parietal gray and white matter [PGM and PWM]) were pooled separately using random-effects meta-analysis. RESULTS During early HIV infection, metabolite alterations were largely limited to the BG, including Cho elevation, a marker of inflammation. cART led to global mI and Cho normalization (i.e., less elevations), but improvement in NAA was negligible. In chronic PWH on cART, there were consistent NAA reductions across brain regions, along with Cho and mI elevations in the FWM and BG, and Glx elevations in the FWM. Cognitive impairment was associated with NAA reduction and to a lesser degree mI elevation. CONCLUSIONS The BG are the primary region affected during early infection. cART is successful in partially controlling neuroinflammation (global mI and Cho normalization). However, neuronal dysfunction (NAA reductions) and neuroinflammation (mI and Cho elevations) persist and contribute to cognitive impairment in chronic PWH. Novel compounds targeting NAA signal pathways, along with better neuroinflammation control, may help to reduce cognitive impairment in PWH.
Collapse
Affiliation(s)
- Sophia Dahmani
- From the Department of Neuroscience (S.D., N.K., X.J.) and Department of Neurology (J.W.V.), Georgetown University Medical Center, Washington, DC; Department of Psychiatry (D.J.M., R.J.E.) and Department of Neurosciences (R.J.E.), University of California, San Diego, La Jolla
| | - Nicholas Kaliss
- From the Department of Neuroscience (S.D., N.K., X.J.) and Department of Neurology (J.W.V.), Georgetown University Medical Center, Washington, DC; Department of Psychiatry (D.J.M., R.J.E.) and Department of Neurosciences (R.J.E.), University of California, San Diego, La Jolla
| | - John W VanMeter
- From the Department of Neuroscience (S.D., N.K., X.J.) and Department of Neurology (J.W.V.), Georgetown University Medical Center, Washington, DC; Department of Psychiatry (D.J.M., R.J.E.) and Department of Neurosciences (R.J.E.), University of California, San Diego, La Jolla
| | - David J Moore
- From the Department of Neuroscience (S.D., N.K., X.J.) and Department of Neurology (J.W.V.), Georgetown University Medical Center, Washington, DC; Department of Psychiatry (D.J.M., R.J.E.) and Department of Neurosciences (R.J.E.), University of California, San Diego, La Jolla
| | - Ronald J Ellis
- From the Department of Neuroscience (S.D., N.K., X.J.) and Department of Neurology (J.W.V.), Georgetown University Medical Center, Washington, DC; Department of Psychiatry (D.J.M., R.J.E.) and Department of Neurosciences (R.J.E.), University of California, San Diego, La Jolla
| | - Xiong Jiang
- From the Department of Neuroscience (S.D., N.K., X.J.) and Department of Neurology (J.W.V.), Georgetown University Medical Center, Washington, DC; Department of Psychiatry (D.J.M., R.J.E.) and Department of Neurosciences (R.J.E.), University of California, San Diego, La Jolla.
| |
Collapse
|
55
|
Alger JR, O'Neill J, O'Connor MJ, Kalender G, Ly R, Ng A, Dillon A, Narr KL, Loo SK, Levitt JG. Neuroimaging of Supraventricular Frontal White Matter in Children with Familial Attention-Deficit Hyperactivity Disorder and Attention-Deficit Hyperactivity Disorder Due to Prenatal Alcohol Exposure. Neurotox Res 2021; 39:1054-1075. [PMID: 33751467 PMCID: PMC8442735 DOI: 10.1007/s12640-021-00342-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 02/15/2021] [Accepted: 02/17/2021] [Indexed: 10/21/2022]
Abstract
Attention-deficit hyperactivity disorder (ADHD) is common in patients with (ADHD+PAE) and without (ADHD-PAE) prenatal alcohol exposure (PAE). Many patients diagnosed with idiopathic ADHD actually have covert PAE, a treatment-relevant distinction. To improve differential diagnosis, we sought to identify brain differences between ADHD+PAE and ADHD-PAE using neurobehavioral, magnetic resonance spectroscopy, and diffusion tensor imaging metrics that had shown promise in past research. Children 8-13 were recruited in three groups: 23 ADHD+PAE, 19 familial ADHD-PAE, and 28 typically developing controls (TD). Neurobehavioral instruments included the Conners 3 Parent Behavior Rating Scale and the Delis-Kaplan Executive Function System (D-KEFS). Two dimensional magnetic resonance spectroscopic imaging was acquired from supraventricular white matter to measure N-acetylaspartate compounds, glutamate, creatine + phosphocreatine (creatine), and choline-compounds (choline). Whole brain diffusion tensor imaging was acquired and used to to calculate fractional anisotropy, mean diffusivity, axial diffusivity, and radial diffusivity from the same superventricular white matter regions that produced magnetic resonance spectroscopy data. The Conners 3 Parent Hyperactivity/Impulsivity Score, glutamate, mean diffusivity, axial diffusivity, and radial diffusivity were all higher in ADHD+PAE than ADHD-PAE. Glutamate was lower in ADHD-PAE than TD. Within ADHD+PAE, inferior performance on the D-KEFS Tower Test correlated with higher neurometabolite levels. These findings suggest white matter differences between the PAE and familial etiologies of ADHD. Abnormalities detected by magnetic resonance spectroscopy and diffusion tensor imaging co-localize in supraventricular white matter and are relevant to executive function symptoms of ADHD.
Collapse
Affiliation(s)
- Jeffry R Alger
- Department of Neurology, University of California Los Angeles, MC 708522, Los Angeles, CA, 90024, USA.
- Neurospectroscopics, LLC, Sherman Oaks, CA, USA.
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Hura Imaging Inc, Calabas, CA, USA.
| | - Joseph O'Neill
- Division of Child & Adolescent Psychiatry, Jane & Terry Semel Instutute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Mary J O'Connor
- Division of Child & Adolescent Psychiatry, Jane & Terry Semel Instutute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Guldamla Kalender
- Division of Child & Adolescent Psychiatry, Jane & Terry Semel Instutute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Ronald Ly
- Division of Child & Adolescent Psychiatry, Jane & Terry Semel Instutute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Andrea Ng
- Division of Child & Adolescent Psychiatry, Jane & Terry Semel Instutute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Andrea Dillon
- Division of Child & Adolescent Psychiatry, Jane & Terry Semel Instutute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Katherine L Narr
- Department of Neurology, University of California Los Angeles, MC 708522, Los Angeles, CA, 90024, USA
- Division of Child & Adolescent Psychiatry, Jane & Terry Semel Instutute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Sandra K Loo
- Division of Child & Adolescent Psychiatry, Jane & Terry Semel Instutute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Jennifer G Levitt
- Division of Child & Adolescent Psychiatry, Jane & Terry Semel Instutute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| |
Collapse
|
56
|
S1P 2-Gα 12 Signaling Controls Astrocytic Glutamate Uptake and Mitochondrial Oxygen Consumption. eNeuro 2021; 8:ENEURO.0040-21.2021. [PMID: 33893167 PMCID: PMC8287876 DOI: 10.1523/eneuro.0040-21.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/06/2021] [Accepted: 04/12/2021] [Indexed: 11/21/2022] Open
Abstract
Glutamate is the principal excitatory neurotransmitter in the human brain. Following neurotransmission, astrocytes remove excess extracellular glutamate to prevent neurotoxicity. Glutamate neurotoxicity has been reported in multiple neurologic diseases including multiple sclerosis (MS), representing a shared neurodegenerative mechanism. A potential modulator of glutamate neurotoxicity is the bioactive lysophospholipid sphingosine 1-phosphate (S1P) that signals through five cognate G-protein-coupled receptors, S1P1-S1P5; however, a clear link between glutamate homeostasis and S1P signaling has not been established. Here, S1P receptor knock-out mice, primary astrocyte cultures, and receptor-selective chemical tools were used to examine the effects of S1P on glutamate uptake. S1P inhibited astrocytic glutamate uptake in a dose-dependent manner and increased mitochondrial oxygen consumption, primarily through S1P2 Primary cultures of wild-type mouse astrocytes expressed S1P1,2,3 transcripts, and selective deletion of S1P1 and/or S1P3 in cerebral cortical astrocytes, did not alter S1P-mediated, dose-dependent inhibition of glutamate uptake. Pharmacological antagonists, S1P2-null astrocytes, and Gα12 hemizygous-null astrocytes indicated that S1P2-Gα12-Rho/ROCK signaling was primarily responsible for the S1P-dependent inhibition of glutamate uptake. In addition, S1P exposure increased mitochondrial oxygen consumption rates (OCRs) in wild-type astrocytes and reduced OCRs in S1P2-null astrocytes, implicating receptor selective metabolic consequences of S1P-mediated glutamate uptake inhibition. Astrocytic S1P-S1P2 signaling increased extracellular glutamate, which could contribute to neurotoxicity. This effect was not observed with the FDA-approved S1P receptor modulators, siponimod and fingolimod. Development and use of S1P2-selective antagonists may provide a new approach to reduce glutamate neurotoxicity in neurologic diseases.
Collapse
|
57
|
Woo MS, Ufer F, Rothammer N, Di Liberto G, Binkle L, Haferkamp U, Sonner JK, Engler JB, Hornig S, Bauer S, Wagner I, Egervari K, Raber J, Duvoisin RM, Pless O, Merkler D, Friese MA. Neuronal metabotropic glutamate receptor 8 protects against neurodegeneration in CNS inflammation. J Exp Med 2021; 218:e20201290. [PMID: 33661276 PMCID: PMC7938362 DOI: 10.1084/jem.20201290] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 12/17/2020] [Accepted: 02/02/2021] [Indexed: 12/13/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system with continuous neuronal loss. Treatment of clinical progression remains challenging due to lack of insights into inflammation-induced neurodegenerative pathways. Here, we show that an imbalance in the neuronal receptor interactome is driving glutamate excitotoxicity in neurons of MS patients and identify the MS risk-associated metabotropic glutamate receptor 8 (GRM8) as a decisive modulator. Mechanistically, GRM8 activation counteracted neuronal cAMP accumulation, thereby directly desensitizing the inositol 1,4,5-trisphosphate receptor (IP3R). This profoundly limited glutamate-induced calcium release from the endoplasmic reticulum and subsequent cell death. Notably, we found Grm8-deficient neurons to be more prone to glutamate excitotoxicity, whereas pharmacological activation of GRM8 augmented neuroprotection in mouse and human neurons as well as in a preclinical mouse model of MS. Thus, we demonstrate that GRM8 conveys neuronal resilience to CNS inflammation and is a promising neuroprotective target with broad therapeutic implications.
Collapse
Affiliation(s)
- Marcel S. Woo
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Friederike Ufer
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Nicola Rothammer
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Giovanni Di Liberto
- Division of Clinical Pathology, Department of Pathology and Immunology, Geneva Faculty of Medicine, Geneva, Switzerland
| | - Lars Binkle
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Undine Haferkamp
- Fraunhofer Institute for Translational Medicine and Pharmacology, Hamburg, Germany
| | - Jana K. Sonner
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Broder Engler
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Sönke Hornig
- Experimentelle Neuropädiatrie, Klinik für Kinder und Jugendmedizin, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Simone Bauer
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Ingrid Wagner
- Division of Clinical Pathology, Department of Pathology and Immunology, Geneva Faculty of Medicine, Geneva, Switzerland
| | - Kristof Egervari
- Division of Clinical Pathology, Department of Pathology and Immunology, Geneva Faculty of Medicine, Geneva, Switzerland
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR
- Department of Neurology, Oregon Health & Science University, Portland, OR
- Department of Radiation Medicine, Oregon Health & Science University, Portland, OR
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR
| | - Robert M. Duvoisin
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR
| | - Ole Pless
- Fraunhofer Institute for Translational Medicine and Pharmacology, Hamburg, Germany
| | - Doron Merkler
- Division of Clinical Pathology, Department of Pathology and Immunology, Geneva Faculty of Medicine, Geneva, Switzerland
| | - Manuel A. Friese
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
58
|
Alba-Arbalat S, Andorra M, Sanchez-Dalmau B, Camos-Carreras A, Dotti-Boada M, Pulido-Valdeolivas I, Llufriu S, Blanco Y, Sepulveda M, Saiz A, Batet O, Bilbao I, Torre I, Amat-Roldan I, Martinez-Lapiscina EH, Villoslada P. In Vivo Molecular Changes in the Retina of Patients With Multiple Sclerosis. Invest Ophthalmol Vis Sci 2021; 62:11. [PMID: 33974046 PMCID: PMC8114005 DOI: 10.1167/iovs.62.6.11] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Purpose Raman spectroscopy allows molecular changes to be quantified in vivo from the tissues like the retina. Here we aimed to assess the metabolic changes in the retina of patients with multiple sclerosis (MS). Methods We built a Raman spectroscopy prototype by connecting a scanning laser ophthalmoscope to a spectrophotometer. We defined the spectra of 10 molecules participating on energy supply, axon biology, or synaptic damage, which have been shown to be altered in the brain of patients with MS: cytochrome C, flavin adenine dinucleotide (FAD), nicotinamide adenine dinucleotide (NADH), N-acetyl-aspartate (NAA), excitotoxicity, glutamate, amyloid β (Aβ), τ and α-synuclein (SNCA), phosphatidyl-ethanolamine, and phosphatidyl-choline. We studied these molecules in a prospective cohort of patients with MS, either in the chronic phase or during relapses of acute optic neuritis (AON). Results Significant changes to all these molecules were associated with age in healthy individuals. There was a significant decrease in NADH and a trend toward a decrease in NAA in patients with MS, as well as an increase in Aβ compared with healthy controls. Moreover, NADH and FAD increased over time in a longitudinal analysis of patients with MS, whereas Aβ diminished. In patients with acute retinal inflammation due to AON, there was a significant increase in FAD and a decrease in SNCA in the affected retina. Moreover, glutamate levels increased in the affected eyes after a 6-month follow-up. Conclusions Alterations of molecules related to axonal degeneration are observed during neuroinflammation and show dynamic changes over time, suggesting progressive neurodegeneration.
Collapse
Affiliation(s)
- Salut Alba-Arbalat
- Center of Neuroimmunology, Institut d'Investigacions Biomediques August Pi Sunyer, Barcelona, Spain.,Department of Ophthalmology, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Magi Andorra
- Center of Neuroimmunology, Institut d'Investigacions Biomediques August Pi Sunyer, Barcelona, Spain
| | - Bernardo Sanchez-Dalmau
- Center of Neuroimmunology, Institut d'Investigacions Biomediques August Pi Sunyer, Barcelona, Spain.,Department of Ophthalmology, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Anna Camos-Carreras
- Center of Neuroimmunology, Institut d'Investigacions Biomediques August Pi Sunyer, Barcelona, Spain.,Department of Ophthalmology, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Marina Dotti-Boada
- Center of Neuroimmunology, Institut d'Investigacions Biomediques August Pi Sunyer, Barcelona, Spain.,Department of Ophthalmology, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Irene Pulido-Valdeolivas
- Center of Neuroimmunology, Institut d'Investigacions Biomediques August Pi Sunyer, Barcelona, Spain.,Department of Neurology, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Sara Llufriu
- Center of Neuroimmunology, Institut d'Investigacions Biomediques August Pi Sunyer, Barcelona, Spain.,Department of Neurology, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Yolanda Blanco
- Center of Neuroimmunology, Institut d'Investigacions Biomediques August Pi Sunyer, Barcelona, Spain.,Department of Neurology, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Maria Sepulveda
- Center of Neuroimmunology, Institut d'Investigacions Biomediques August Pi Sunyer, Barcelona, Spain.,Department of Neurology, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Albert Saiz
- Center of Neuroimmunology, Institut d'Investigacions Biomediques August Pi Sunyer, Barcelona, Spain.,Department of Neurology, Hospital Clinic, University of Barcelona, Barcelona, Spain.,Institut de Neurociències, University of Barcelona, Barcelona, Spain
| | | | | | | | | | - Elena H Martinez-Lapiscina
- Center of Neuroimmunology, Institut d'Investigacions Biomediques August Pi Sunyer, Barcelona, Spain.,Department of Neurology, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Pablo Villoslada
- Center of Neuroimmunology, Institut d'Investigacions Biomediques August Pi Sunyer, Barcelona, Spain.,Stanford University, Stanford, California, United States
| |
Collapse
|
59
|
Kulpanovich A, Tal A. What is the optimal schedule for multiparametric MRS? A magnetic resonance fingerprinting perspective. NMR IN BIOMEDICINE 2021; 34:e4196. [PMID: 31814197 PMCID: PMC9244865 DOI: 10.1002/nbm.4196] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/09/2019] [Accepted: 09/09/2019] [Indexed: 05/09/2023]
Abstract
Clinical magnetic resonance spectroscopy (MRS) mainly concerns itself with the quantification of metabolite concentrations. Metabolite relaxation values, which reflect the microscopic state of specific cellular and sub-cellular environments, could potentially hold additional valuable information, but are rarely acquired within clinical scan times. By varying the flip angle, repetition time and echo time in a preset way (termed a schedule), and matching the resulting signals to a pre-generated dictionary - an approach dubbed magnetic resonance fingerprinting - it is possible to encode the spins' relaxation times into the acquired signal, simultaneously quantifying multiple tissue parameters for each metabolite. Herein, we optimized the schedule to minimize the averaged root mean square error (RMSE) across all estimated parameters: concentrations, longitudinal and transverse relaxation time, and transmitter inhomogeneity. The optimal schedules were validated in phantoms and, subsequently, in a cohort of healthy volunteers, in a 4.5 mL parietal white matter single voxel and an acquisition time under 5 minutes. The average intra-subject, inter-scan coefficients of variation (CVs) for metabolite concentrations, T1 and T2 relaxation times were found to be 3.4%, 4.6% and 4.7% in-vivo, respectively, averaged over all major singlets. Coupled metabolites were quantified using the short echo time schedule entries and spectral fitting, and reliable estimates of glutamate+glutamine, glutathione and myo-inositol were obtained.
Collapse
Affiliation(s)
- Alexey Kulpanovich
- Department of Chemical Physics, Weizmann Institute of Science, 234 Herzel St., Rehovot 7610001, Israel
| | - Assaf Tal
- Department of Chemical Physics, Weizmann Institute of Science, 234 Herzel St., Rehovot 7610001, Israel
| |
Collapse
|
60
|
Roles of astrocytes in response to aging, Alzheimer's disease and multiple sclerosis. Brain Res 2021; 1764:147464. [PMID: 33812850 DOI: 10.1016/j.brainres.2021.147464] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 03/11/2021] [Accepted: 03/29/2021] [Indexed: 01/08/2023]
Abstract
Astrocytes are traditionally recognized for their multiple roles in support of brain function. However, additional changes in these roles are evident in response to brain diseases. In this review, we highlight positive and negative effects of astrocytes in response to aging, Alzheimer's disease and Multiple Sclerosis. We summarize data suggesting that reactive astrocytes may perform critical functions that might be relevant to the etiology of these conditions. In particular, we relate astrocytes effects to actions on synaptic transmission, cognition, and myelination. We suggest that a better understanding of astrocyte functions and how these become altered in response to aging or disease will lead to the appreciation of these cells as useful therapeutic targets.
Collapse
|
61
|
Bilingualism is a long-term cognitively challenging experience that modulates metabolite concentrations in the healthy brain. Sci Rep 2021; 11:7090. [PMID: 33782462 PMCID: PMC8007713 DOI: 10.1038/s41598-021-86443-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 03/16/2021] [Indexed: 02/01/2023] Open
Abstract
Cognitively demanding experiences, including complex skill acquisition and processing, have been shown to induce brain adaptations, at least at the macroscopic level, e.g. on brain volume and/or functional connectivity. However, the neurobiological bases of these adaptations, including at the cellular level, are unclear and understudied. Here we use bilingualism as a case study to investigate the metabolic correlates of experience-based brain adaptations. We employ Magnetic Resonance Spectroscopy to measure metabolite concentrations in the basal ganglia, a region critical to language control which is reshaped by bilingualism. Our results show increased myo-Inositol and decreased N-acetyl aspartate concentrations in bilinguals compared to monolinguals. Both metabolites are linked to synaptic pruning, a process underlying experience-based brain restructuring. Interestingly, both concentrations correlate with relative amount of bilingual engagement. This suggests that degree of long-term cognitive experiences matters at the level of metabolic concentrations, which might accompany, if not drive, macroscopic brain adaptations.
Collapse
|
62
|
Petracca M, Pontillo G, Moccia M, Carotenuto A, Cocozza S, Lanzillo R, Brunetti A, Brescia Morra V. Neuroimaging Correlates of Cognitive Dysfunction in Adults with Multiple Sclerosis. Brain Sci 2021; 11:346. [PMID: 33803287 PMCID: PMC8000635 DOI: 10.3390/brainsci11030346] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/02/2021] [Accepted: 03/04/2021] [Indexed: 02/06/2023] Open
Abstract
Cognitive impairment is a frequent and meaningful symptom in multiple sclerosis (MS), caused by the accrual of brain structural damage only partially counteracted by effective functional reorganization. As both these aspects can be successfully investigated through the application of advanced neuroimaging, here, we offer an up-to-date overview of the latest findings on structural, functional and metabolic correlates of cognitive impairment in adults with MS, focusing on the mechanisms sustaining damage accrual and on the identification of useful imaging markers of cognitive decline.
Collapse
Affiliation(s)
- Maria Petracca
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (M.P.); (M.M.); (A.C.); (V.B.M.)
| | - Giuseppe Pontillo
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (G.P.); (S.C.); (A.B.)
- Department of Electrical Engineering and Information Technology, University of Naples “Federico II”, 80125 Naples, Italy
| | - Marcello Moccia
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (M.P.); (M.M.); (A.C.); (V.B.M.)
| | - Antonio Carotenuto
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (M.P.); (M.M.); (A.C.); (V.B.M.)
| | - Sirio Cocozza
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (G.P.); (S.C.); (A.B.)
| | - Roberta Lanzillo
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (M.P.); (M.M.); (A.C.); (V.B.M.)
| | - Arturo Brunetti
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (G.P.); (S.C.); (A.B.)
| | - Vincenzo Brescia Morra
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (M.P.); (M.M.); (A.C.); (V.B.M.)
| |
Collapse
|
63
|
Altered in vivo brain GABA and glutamate levels are associated with multiple sclerosis central fatigue. Eur J Radiol 2021; 137:109610. [PMID: 33657474 DOI: 10.1016/j.ejrad.2021.109610] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 01/31/2021] [Accepted: 02/21/2021] [Indexed: 11/20/2022]
Abstract
PURPOSE Fatigue is a common symptom in patients with multiple sclerosis (MS) with unknown pathophysiology. Dysfunction of the GABAergic/glutamatergic pathways involving inhibitory and excitatory neurotransmitters such as γ-aminobutyric acid (GABA) and glutamine + glutamate pool (Glx) have been implicated in several neurological disorders. This study is aimed to evaluate the potential role of GABA and Glx in the origin of central fatigue in relapse remitting MS (RRMS) patients. METHODS 24 RRMS patients and 16 age- and sex-matched healthy controls (HC) were scanned using Mescher-Garwood point resolved spectroscopy (MEGA-PRESS) with a 3 T system to quantify GABA+ and Glx from prefrontal (PFC) and sensorimotor (SMC) cortices. Self-reported fatigue status was measured on all participants using the Modified Fatigue Impact Scale (MFIS). RESULTS RRMS patients had higher fatigue scores relative to HC (p ≤ 0.05). Compared to HC, Glx levels in RRMS patients were significantly decreased in SMC (p = 0.04). Significant correlations were found between fatigue scores and GABA+ (r = -0.531, p = 0.008) and Glx (r = 0.511, p = 0.018) in PFC. Physical fatigue was negatively correlated with GABA+ in SMC and PFC (r = -0.428 and -0.472 respectively, p ≤ 0.04) and positively with PFC Glx (r = 0.480, p = 0.028). CONCLUSION The associations between fatigue and GABA + and Glx suggest that there might be dysregulation of GABAergic/glutamatergic neurotransmission in the pathophysiological mechanism of central fatigue in MS.
Collapse
|
64
|
Bagnato F, Gauthier SA, Laule C, Moore GRW, Bove R, Cai Z, Cohen-Adad J, Harrison DM, Klawiter EC, Morrow SA, Öz G, Rooney WD, Smith SA, Calabresi PA, Henry RG, Oh J, Ontaneda D, Pelletier D, Reich DS, Shinohara RT, Sicotte NL. Imaging Mechanisms of Disease Progression in Multiple Sclerosis: Beyond Brain Atrophy. J Neuroimaging 2021; 30:251-266. [PMID: 32418324 DOI: 10.1111/jon.12700] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 02/04/2020] [Accepted: 02/18/2020] [Indexed: 12/11/2022] Open
Abstract
Clinicians involved with different aspects of the care of persons with multiple sclerosis (MS) and scientists with expertise on clinical and imaging techniques convened in Dallas, TX, USA on February 27, 2019 at a North American Imaging in Multiple Sclerosis Cooperative workshop meeting. The aim of the workshop was to discuss cardinal pathobiological mechanisms implicated in the progression of MS and novel imaging techniques, beyond brain atrophy, to unravel these pathologies. Indeed, although brain volume assessment demonstrates changes linked to disease progression, identifying the biological mechanisms leading up to that volume loss are key for understanding disease mechanisms. To this end, the workshop focused on the application of advanced magnetic resonance imaging (MRI) and positron emission tomography (PET) imaging techniques to assess and measure disease progression in both the brain and the spinal cord. Clinical translation of quantitative MRI was recognized as of vital importance, although the need to maintain a relatively short acquisition time mandated by most radiology departments remains the major obstacle toward this effort. Regarding PET, the panel agreed upon its utility to identify ongoing pathological processes. However, due to costs, required expertise, and the use of ionizing radiation, PET was not considered to be a viable option for ongoing care of persons with MS. Collaborative efforts fostering robust study designs and imaging technique standardization across scanners and centers are needed to unravel disease mechanisms leading to progression and discovering medications halting neurodegeneration and/or promoting repair.
Collapse
Affiliation(s)
- Francesca Bagnato
- Neuroimaging Unit, Neuroimmunology Division, Department of Neurology, Vanderbilt University Medical Center, Nashville, TN
| | - Susan A Gauthier
- Judith Jaffe Multiple Sclerosis Center, Department of Neurology, Feil Family Brain and Mind Institute, and Department of Radiology, Weill Cornell Medicine, New York, NY
| | - Cornelia Laule
- Department of Radiology, Pathology, and Laboratory Medicine, Department of Physics and Astronomy, and International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, British Columbia, Canada
| | - George R Wayne Moore
- Department of Pathology and Laboratory Medicine, and International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, British Columbia, Canada
| | - Riley Bove
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA
| | - Zhengxin Cai
- Department of Radiology and Biomedical Imaging, PET Center, Yale University, New Haven, CT
| | - Julien Cohen-Adad
- NeuroPoly Lab, Institute of Biomedical Engineering, Polytechnique Montreal and Functional Neuroimaging Unit, CRIUGM, University of Montreal, Montreal, Quebec, Canada
| | - Daniel M Harrison
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD
| | - Eric C Klawiter
- Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Sarah A Morrow
- Department of Clinical Neurological Sciences, University of Western Ontario, London, Ontario, Canada
| | - Gülin Öz
- Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN
| | - William D Rooney
- Advanced Imaging Research Center, Departments of Biomedical Engineering, Neurology, and Behavioral Neuroscience, Oregon Health & Science University, Portland, OR
| | - Seth A Smith
- Radiology and Radiological Sciences and Vanderbilt University Imaging Institute, Vanderbilt University Medical Center, and Biomedical Engineering, Vanderbilt University, Nashville, TN
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Roland G Henry
- Departments of Neurology, Radiology and Biomedical Imaging, and the UC San Francisco & Berkeley Bioengineering Graduate Group, University of California San Francisco, San Francisco, CA
| | - Jiwon Oh
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD.,Division of Neurology, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Daniel Ontaneda
- Mellen Center for Multiple Sclerosis, Neurological Institute, Cleveland Clinic, Cleveland, OH
| | - Daniel Pelletier
- Department of Neurology, University of Southern California Keck School of Medicine, Los Angeles, CA
| | - Daniel S Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, Bethesda, MD
| | - Russell T Shinohara
- Department of Biostatistics, Epidemiology, and Informatics, Penn Statistics in Imaging and Visualization Center, University of Pennsylvania, Philadelphia, PA
| | - Nancy L Sicotte
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA
| | -
- Neuroimaging Unit, Neuroimmunology Division, Department of Neurology, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
65
|
Lee DW, Kwon JI, Woo CW, Heo H, Kim KW, Woo DC, Kim JK, Lee DH. In Vivo Measurement of Neurochemical Abnormalities in the Hippocampus in a Rat Model of Cuprizone-Induced Demyelination. Diagnostics (Basel) 2020; 11:diagnostics11010045. [PMID: 33396601 PMCID: PMC7823778 DOI: 10.3390/diagnostics11010045] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/25/2020] [Accepted: 12/27/2020] [Indexed: 12/30/2022] Open
Abstract
This study quantitatively measured the changes in metabolites in the hippocampal lesions of a rat model of cuprizone-induced demyelination as detected using in vivo 7 T proton magnetic resonance spectroscopy. Nineteen Sprague Dawley rats were randomly divided into two groups and fed a normal chow diet or cuprizone (0.2%, w/w) for 7 weeks. Demyelinated hippocampal lesions were quantitatively measured using a 7 T magnetic resonance imaging scanner. All proton spectra were quantified for metabolite concentrations and relative ratios. Compared to those in the controls, the cuprizone-induced rats had significantly higher concentrations of glutamate (p = 0.001), gamma-aminobutyric acid (p = 0.019), and glutamate + glutamine (p = 0.001); however, creatine + phosphocreatine (p = 0.006) and myo-inositol (p = 0.001) concentrations were lower. In addition, we found that the glutamine and glutamate complex/total creatine (p < 0.001), glutamate/total creatine (p < 0.001), and GABA/total creatine (p = 0.002) ratios were significantly higher in cuprizone-treated rats than in control rats. Our results showed that cuprizone-induced neuronal demyelination may influence the severe abnormal metabolism in hippocampal lesions, and these responses could be caused by microglial activation, mitochondrial dysfunction, and astrocytic necrosis.
Collapse
Affiliation(s)
- Do-Wan Lee
- Department of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (K.W.K.); (J.K.K.)
- Correspondence: (D.-W.L.); (D.-H.L.)
| | - Jae-Im Kwon
- Convergence Medicine Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea; (J.-I.K.); (C.-W.W.); (D.-C.W.)
| | - Chul-Woong Woo
- Convergence Medicine Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea; (J.-I.K.); (C.-W.W.); (D.-C.W.)
| | - Hwon Heo
- Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea;
| | - Kyung Won Kim
- Department of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (K.W.K.); (J.K.K.)
| | - Dong-Cheol Woo
- Convergence Medicine Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea; (J.-I.K.); (C.-W.W.); (D.-C.W.)
- Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea;
| | - Jeong Kon Kim
- Department of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (K.W.K.); (J.K.K.)
| | - Dong-Hoon Lee
- Department of Radiation Convergence Engineering, Yonsei University, Wonju 26493, Korea
- Correspondence: (D.-W.L.); (D.-H.L.)
| |
Collapse
|
66
|
Gallego-Delgado P, James R, Browne E, Meng J, Umashankar S, Tan L, Picon C, Mazarakis ND, Faisal AA, Howell OW, Reynolds R. Neuroinflammation in the normal-appearing white matter (NAWM) of the multiple sclerosis brain causes abnormalities at the nodes of Ranvier. PLoS Biol 2020; 18:e3001008. [PMID: 33315860 PMCID: PMC7769608 DOI: 10.1371/journal.pbio.3001008] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 12/28/2020] [Accepted: 11/20/2020] [Indexed: 01/02/2023] Open
Abstract
Changes to the structure of nodes of Ranvier in the normal-appearing white matter (NAWM) of multiple sclerosis (MS) brains are associated with chronic inflammation. We show that the paranodal domains in MS NAWM are longer on average than control, with Kv1.2 channels dislocated into the paranode. These pathological features are reproduced in a model of chronic meningeal inflammation generated by the injection of lentiviral vectors for the lymphotoxin-α (LTα) and interferon-γ (IFNγ) genes. We show that tumour necrosis factor (TNF), IFNγ, and glutamate can provoke paranodal elongation in cerebellar slice cultures, which could be reversed by an N-methyl-D-aspartate (NMDA) receptor blocker. When these changes were inserted into a computational model to simulate axonal conduction, a rapid decrease in velocity was observed, reaching conduction failure in small diameter axons. We suggest that glial cells activated by pro-inflammatory cytokines can produce high levels of glutamate, which triggers paranodal pathology, contributing to axonal damage and conduction deficits. Current thinking on the mechanisms by which multiple sclerosis gives rise to cumulative neurological disability revolves largely around focal lesions of inflammation and demyelination. However, some of the debilitating symptoms, such as severe fatigue, might be better explained by a more diffuse pathology. This study shows that paranodes in the white matter become abnormal as a result of neuroinflammation, which may be the result of the action of cytokines that cause glia to release glutamate.
Collapse
Affiliation(s)
- Patricia Gallego-Delgado
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Rachel James
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Eleanor Browne
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Joanna Meng
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Swetha Umashankar
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Li Tan
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Carmen Picon
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Nicholas D. Mazarakis
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - A. Aldo Faisal
- Department of Bioengineering, Faculty of Engineering, Imperial College London, London, United Kingdom
- Department of Computing, Faculty of Engineering, Imperial College London, London, United Kingdom
- Data Science Institute, Imperial College London, London, United Kingdom
| | - Owain W. Howell
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
- Institute of Life Sciences, Swansea University Medical School, Swansea University, Swansea, Wales
| | - Richard Reynolds
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
- Centre for Molecular Neuropathology, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- * E-mail:
| |
Collapse
|
67
|
Porter L, Shoushtarizadeh A, Jelinek GA, Brown CR, Lim CK, de Livera AM, Jacobs KR, Weiland TJ. Metabolomic Biomarkers of Multiple Sclerosis: A Systematic Review. Front Mol Biosci 2020; 7:574133. [PMID: 33381517 PMCID: PMC7768024 DOI: 10.3389/fmolb.2020.574133] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 10/27/2020] [Indexed: 01/07/2023] Open
Abstract
Background Magnetic resonance imaging (MRI), cerebrospinal fluid (CSF) analysis, and the McDonald’s clinical criteria are currently utilized tools in diagnosing multiple sclerosis. However, a more conclusive, consistent, and efficient way of diagnosing multiple sclerosis (MS) is yet to be discovered. A potential biomarker, discovered using advances in high-throughput sequencing such as nuclear magnetic resonance (NMR) spectroscopy and other “Omics”-based techniques, may make diagnosis and prognosis more reliable resulting in a more personalized and targeted treatment regime and improved outcomes. The aim of this review was to systematically search the literature for potential biomarkers from any bodily fluid that could consistently and accurately diagnose MS and/or indicate disease progression. Methods A systematic literature review of EMBASE, PubMed (MEDLINE), The Cochrane Library, and CINAHL databases produced over a thousand potential studies. Inclusion criteria stated studies with potential biomarker outcomes for people with MS were to be included in the review. Studies were limited to those with human participants who had a clinically defined diagnosis of MS and published in English, with no limit placed on date of publication or the type of bodily fluid sampled. Results A total of 1,805 studies were recorded from the literature search. A total of 1,760 studies were removed based on their abstract, with a further 18 removed after considering the full text. A total of 30 studies were considered relevant and had their data retrieved and analyzed. Due to the heterogeneity of focus and results from the refined studies, a narrative synthesis was favored. Conclusion Several promising candidate biomarkers suitable for clinical application in MS have been studied. It is recommended follow-up studies with larger sample sizes be completed on several potential biomarkers.
Collapse
Affiliation(s)
- Lachlan Porter
- Neuroepidemiology Unit, Melbourne School of Population and Global Health, The University of Melbourne, Carlton, VIC, Australia
| | - Alireza Shoushtarizadeh
- Neuroepidemiology Unit, Melbourne School of Population and Global Health, The University of Melbourne, Carlton, VIC, Australia
| | - George A Jelinek
- Neuroepidemiology Unit, Melbourne School of Population and Global Health, The University of Melbourne, Carlton, VIC, Australia
| | - Chelsea R Brown
- The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Chai K Lim
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Macquarie Park, NSW, Australia
| | - Alysha M de Livera
- Neuroepidemiology Unit, Melbourne School of Population and Global Health, The University of Melbourne, Carlton, VIC, Australia
| | - Kelly R Jacobs
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Macquarie Park, NSW, Australia
| | - Tracey J Weiland
- Neuroepidemiology Unit, Melbourne School of Population and Global Health, The University of Melbourne, Carlton, VIC, Australia
| |
Collapse
|
68
|
Temporal Changes in In Vivo Glutamate Signal during Demyelination and Remyelination in the Corpus Callosum: A Glutamate-Weighted Chemical Exchange Saturation Transfer Imaging Study. Int J Mol Sci 2020; 21:ijms21249468. [PMID: 33322784 PMCID: PMC7764201 DOI: 10.3390/ijms21249468] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 12/04/2020] [Accepted: 12/11/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Glutamate-weighted chemical exchange saturation transfer (GluCEST) is a useful imaging tool that can be used to detect changes in glutamate levels in vivo and could also be helpful in the diagnosis of brain myelin changes. We investigated glutamate level changes in the cerebral white matter of a rat model of cuprizone-administered demyelination and remyelination using GluCEST. METHOD We used a 7 T pre-clinical magnetic resonance imaging (MRI) system. The rats were divided into the normal control (CTRL), cuprizone-administered demyelination (CPZDM), and remyelination (CPZRM) groups. GluCEST data were analyzed using the conventional magnetization transfer ratio asymmetry in the corpus callosum. Immunohistochemistry and transmission electron microscopy analyses were also performed to investigate the myelinated axon changes in each group. RESULTS The quantified GluCEST signals differed significantly between the CPZDM and CTRL groups (-7.25 ± 1.42% vs. -2.84 ± 1.30%; p = 0.001). The increased GluCEST signals in the CPZDM group decreased after remyelination (-6.52 ± 1.95% in CPZRM) to levels that did not differ significantly from those in the CTRL group (p = 0.734). CONCLUSION The apparent temporal signal changes in GluCEST imaging during demyelination and remyelination demonstrated the potential usefulness of GluCEST imaging as a tool to monitor the myelination process.
Collapse
|
69
|
Ye H, Shaghaghi M, Chen Q, Zhang Y, Lutz SE, Chen W, Cai K. In Vivo Proton Exchange Rate (k ex ) MRI for the Characterization of Multiple Sclerosis Lesions in Patients. J Magn Reson Imaging 2020; 53:408-415. [PMID: 32975008 DOI: 10.1002/jmri.27363] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 08/24/2020] [Accepted: 08/26/2020] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Currently available radiological methods do not completely capture the diversity of multiple sclerosis (MS) lesion subtypes. This lack of information hampers the understanding of disease progression and potential treatment stratification. For example, inflammation persists in some lesions after gadolinium (Gd) enhancement resolves. Novel metabolic and molecular imaging methods may improve the current assessments of MS pathophysiology. PURPOSE To compare the in vivo proton exchange rate (kex ) MRI with Gd-enhanced MRI for characterizing MS lesions. STUDY TYPE Retrospective. SUBJECTS Sixteen consecutively diagnosed relapsing-remitting multiple sclerosis (RRMS) patients. FIELD STRENGTH/SEQUENCE 3.0T MRI with T2 -weighted imaging, postcontrast T1 -weighted imaging, and single-slice chemical exchange saturation transfer imaging. ASSESSMENT MS lesions in white matter were assessed for Gd enhancement and kex elevation compared to normal-appearing white matter (NAWM). STATISTICAL TESTS Student's t-test was used for analyzing the difference of kex values between lesions and NAWM, with statistical significance set at 0.05. RESULTS Of all 153 MS lesions, 78 (51%) lesions were Gd-enhancing and 75 (49%) were Gd-negative. Without exception, all 78 Gd-enhancing lesions showed significantly elevated kex values compared to NAWM (924 ± 130 s-1 vs. 735 ± 61 s-1 , P < 0.05). Of 75 Gd-negative lesions, 18 lesions (24%) showed no kex elevation (762 ± 29 s-1 vs. 755 ± 28 s-1 , P = 0.47) and 57 (76%) showed significant kex elevation (950 ± 124 s-1 vs. 759 ± 48 s-1 , P < 0.05) compared to NAWM. MS lesions with kex elevation appeared nodular (118, 87.4%), ring-like (15, 11.1%), or irregular-shaped (2, 1.5%). DATA CONCLUSION For Gd-enhancing lesions, kex MRI is highly consistent with Gd-enhanced images by showing 100% of elevated kex . For all Gd-negative lesions, the discrepancy on kex MRI may further differentiate active slowly expanding lesions or chronic inactive lesions, supporting kex as an imaging biomarker for tissue oxidative stress and inflammation. Level of Evidence 2 Technical Efficacy Stage 3 J. MAGN. RESON. IMAGING 2021;53:408-415.
Collapse
Affiliation(s)
- Haiqi Ye
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mehran Shaghaghi
- Department of Radiology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Qianlan Chen
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Zhang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sarah E Lutz
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Weiwei Chen
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kejia Cai
- Department of Radiology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA.,Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois, USA.,Center for MR Research, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
70
|
Signal alterations of glutamate-weighted chemical exchange saturation transfer MRI in lysophosphatidylcholine-induced demyelination in the rat brain. Brain Res Bull 2020; 164:334-338. [PMID: 32926951 DOI: 10.1016/j.brainresbull.2020.09.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 08/10/2020] [Accepted: 09/05/2020] [Indexed: 01/07/2023]
Abstract
PURPOSE To compare in vivo glutamate-weighted chemical exchange saturation transfer (GluCEST-weighted) signal changes between in a rat model of demyelinated multiple sclerosis and control groups. PROCEDURES Using a pre-clinical 7 T magnetic resonance imaging (MRI) system, CEST imaging was applied to a toxin (lysophosphatidylcholine; LPC) induced rat (MSLPC) and control (CTRL) groups to compare in vivo glutamate signal changes. The GluCEST-weighted signals were analyzed based on the magnetization transfer ratio asymmetry approach at 3.0 ppm on the region-of-interests (ROIs) in the corpus callosum and hippocampus at each hemispheric region. RESULTS GluCEST-weighted signals were significantly changed between the CTRL and MSLPC groups, while higher glutamate signals were indicated in the MSLPC than the CTRL group ([MSLPC / CTRL]; hippocampus: [6.159 ± 0.790 / 4.336 ± 0.446] and corpus callosum: [-3.545 ± 0.945 / -6.038 ± 0.620], all p = 0.001). CONCLUSIONS Our results show increased GluCEST-weighted signals in the LPC-induced demyelination rat brain compared with control. GluCEST-weighted imaging could be a useful tool for defining a biomarker to estimate the glutamate-related metabolism in MS.
Collapse
|
71
|
Maramai S, Brindisi M. Targeting Endocannabinoid Metabolism: an Arrow with Multiple Tips Against Multiple Sclerosis. ChemMedChem 2020; 15:1985-2003. [PMID: 32762071 DOI: 10.1002/cmdc.202000310] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 07/24/2020] [Indexed: 12/19/2022]
Abstract
Multiple sclerosis (MS) is a chronic, immune-mediated disease of the central nervous system. At present, there is no definitive cure, and the few available disease-modifying options display either poor efficacy or life-threatening side effects. There is clear evidence that relapsing-remitting clinical attacks in MS are driven by inflammatory demyelination and that the subsequent disease steps, being irresponsive to immunotherapy, result from neurodegeneration. The endocannabinoid system (ECS) stands halfway between three key pathomechanisms underlying MS, namely inflammation, neurodegeneration and oxidative stress, thus representing a kingpin for the identification of novel therapeutic targets in MS. This review summarizes the current state of the art in the field of endocannabinoid metabolism modulators and their in vivo effects on relevant animal models. We also highlight key molecular underpinnings of their therapeutic efficacy as well as the potential to turn them into promising clinical candidates.
Collapse
Affiliation(s)
- Samuele Maramai
- Department of Excellence of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro, 2, 53100, Siena, Italy
| | - Margherita Brindisi
- Department of Excellence of Pharmacy, University of Naples Federico II, Via D. Montesano, 49, 80131, Naples, Italy
| |
Collapse
|
72
|
Al-Iedani O, Ribbons K, Gholizadeh N, Lechner-Scott J, Quadrelli S, Lea R, Andronesi O, Ramadan S. Spiral MRSI and tissue segmentation of normal-appearing white matter and white matter lesions in relapsing remitting multiple sclerosis patients ☆. Magn Reson Imaging 2020; 74:21-30. [PMID: 32898652 DOI: 10.1016/j.mri.2020.09.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 09/02/2020] [Accepted: 09/02/2020] [Indexed: 12/17/2022]
Abstract
PURPOSE To evaluate the performance of novel spiral MRSI and tissue segmentation pipeline of the brain, to investigate neurometabolic changes in normal-appearing white matter (NAWM) and white matter lesions (WML) of stable relapsing remitting multiple sclerosis (RRMS) compared to healthy controls (HCs). METHODS Spiral 3D MRSI using LASER-GOIA-W [16,4] was undertaken on 16 RRMS patients and 9 HCs, to acquire MRSI data from a large volume of interest (VOI) 320 cm3 and analyzed using LCModel. MRSI data and voxel tissue segmentation were compared between the two cohorts using t-tests. Support vector machine (SVM) was used to classify tissue types and assessed by accuracy, sensitivity and specificity. RESULTS Compared to HCs, RRMS demonstrated a statistically significant reduction in all mean brain tissues and increase in CSF volume. Within VOI, WM decreased (-10%) and CSF increased (41%) in RRMS compared to HCs (p < 0.001). MRSI revealed that total creatine (tCr) ratios of N-acetylaspartate and glutamate+glutamine in WML were significantly lower than NAWM-MS (-9%, -8%) and HCs (-14%, -10%), respectively. Myo-inositol/tCr in WML was significantly higher than NAWM-MS (14%) and HCs (10%). SVM of MRSI yielded accuracy, sensitivity and specificity of 86%, 95%, and 70%, respectively for HCs vs WML, which were higher than HC vs NAWM and WML vs NAWM models. CONCLUSION This study demonstrates the benefit of MRSI in evaluating MS neurometabolic changes in NAWM. SVM of MRSI data in the MS brain may be suited for clinical monitoring and progression of MS patients. Longitudinal MRSI studies are warranted.
Collapse
Affiliation(s)
- Oun Al-Iedani
- School of Health Sciences, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, Newcastle, Australia
| | - Karen Ribbons
- Hunter Medical Research Institute, Newcastle, Australia
| | - Neda Gholizadeh
- School of Health Sciences, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Jeannette Lechner-Scott
- Hunter Medical Research Institute, Newcastle, Australia; Department of Neurology, John Hunter Hospital, New Lambton Heights, NSW 2305, Australia; School of Medicine and Public Health, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2305, Australia
| | - Scott Quadrelli
- Princess Alexandra Hospital, Radiology Department, Woolloongabba. QLD 4102, Australia; Faculty of Medicine, University of Queensland, Herston, QLD 4006, Australia
| | - Rodney Lea
- Hunter Medical Research Institute, Newcastle, Australia
| | - Ovidiu Andronesi
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Saadallah Ramadan
- School of Health Sciences, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, Newcastle, Australia.
| |
Collapse
|
73
|
Hnilicová P, Štrbák O, Kolisek M, Kurča E, Zeleňák K, Sivák Š, Kantorová E. Current Methods of Magnetic Resonance for Noninvasive Assessment of Molecular Aspects of Pathoetiology in Multiple Sclerosis. Int J Mol Sci 2020; 21:E6117. [PMID: 32854318 PMCID: PMC7504207 DOI: 10.3390/ijms21176117] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/13/2020] [Accepted: 08/21/2020] [Indexed: 12/29/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease with expanding axonal and neuronal degeneration in the central nervous system leading to motoric dysfunctions, psychical disability, and cognitive impairment during MS progression. The exact cascade of pathological processes (inflammation, demyelination, excitotoxicity, diffuse neuro-axonal degeneration, oxidative and metabolic stress, etc.) causing MS onset is still not fully understood, although several accompanying biomarkers are particularly suitable for the detection of early subclinical changes. Magnetic resonance (MR) methods are generally considered to be the most sensitive diagnostic tools. Their advantages include their noninvasive nature and their ability to image tissue in vivo. In particular, MR spectroscopy (proton 1H and phosphorus 31P MRS) is a powerful analytical tool for the detection and analysis of biomedically relevant metabolites, amino acids, and bioelements, and thus for providing information about neuro-axonal degradation, demyelination, reactive gliosis, mitochondrial and neurotransmitter failure, cellular energetic and membrane alternation, and the imbalance of magnesium homeostasis in specific tissues. Furthermore, the MR relaxometry-based detection of accumulated biogenic iron in the brain tissue is useful in disease evaluation. The early description and understanding of the developing pathological process might be critical for establishing clinically effective MS-modifying therapies.
Collapse
Affiliation(s)
- Petra Hnilicová
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia; (O.Š.); (M.K.)
| | - Oliver Štrbák
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia; (O.Š.); (M.K.)
| | - Martin Kolisek
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia; (O.Š.); (M.K.)
| | - Egon Kurča
- Clinic of Neurology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia; (E.K.); (Š.S.); (E.K.)
| | - Kamil Zeleňák
- Clinic of Radiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia;
| | - Štefan Sivák
- Clinic of Neurology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia; (E.K.); (Š.S.); (E.K.)
| | - Ema Kantorová
- Clinic of Neurology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia; (E.K.); (Š.S.); (E.K.)
| |
Collapse
|
74
|
Approved and Emerging Disease Modifying Therapies on Neurodegeneration in Multiple Sclerosis. Int J Mol Sci 2020; 21:ijms21124312. [PMID: 32560364 PMCID: PMC7348940 DOI: 10.3390/ijms21124312] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 06/15/2020] [Accepted: 06/15/2020] [Indexed: 12/16/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune, chronic, progressive disease leading to a combination of inflammation, demyelination, and neurodegeneration throughout the central nervous system (CNS). The outcome of these processes can be visualized in magnetic resonance imaging (MRI) scans as brain atrophy, or brain volume loss (BVL), as well as lesions, “black holes” and spinal cord atrophy. MRI outcomes such as BVL have been used as biomarkers of neurodegeneration and other measures of MS disease progression in clinical research settings. Several FDA-approved medications seek to alleviate disease progression by reducing the impact of such factors as demyelination and neurodegeneration, but there are still many shortcomings that current clinical research aims to mitigate. This review attempts to provide an overview of the FDA-approved medications available for treating multiple sclerosis and their effect on neurodegeneration, measured by BVL.
Collapse
|
75
|
Zhang Y, Shen J. Effects of noise and linewidth on in vivo analysis of glutamate at 3 T. JOURNAL OF MAGNETIC RESONANCE (SAN DIEGO, CALIF. : 1997) 2020; 314:106732. [PMID: 32361510 PMCID: PMC8485252 DOI: 10.1016/j.jmr.2020.106732] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/24/2020] [Accepted: 04/11/2020] [Indexed: 05/17/2023]
Abstract
Magnetic resonance spectroscopy (MRS) can noninvasively detect metabolites in vivo, including glutamate (Glu). However, quantification is known to be affected by the overlaps among metabolite resonance lines and background macromolecule signals. We found that adding a moderate amount of noise or line broadening (2 Hz) caused large variations in concentration of Glu and other metabolites, when determined by LCModel analysis of in vivo short-echo time (TE) spectra. Theses variations were largely attributed to strong spectral baselines in short TE spectra, especially near 2.35 ppm, as well as overlapping metabolite resonance lines. To address this issue, we acquired in vivo data at 3 T using both short-TE and the multiple echo time J-resolved point-resolved spectroscopy (JPRESS) MRS techniques. We found that one-dimensional (1D) JPRESS, by simultaneously fitting the two cross-sections of JPRESS at J = 0 and J = 7.5 Hz, was highly resistant to variations in noise levels and spectral linewidths. Our results demonstrate that LCModel analysis of short-TE data is highly sensitive to variations in noise levels and spectral linewidths and this sensitivity is greatly reduced by 1D JPRESS given its substantially reduced baselines and enhanced spectral resolution.
Collapse
Affiliation(s)
- Yan Zhang
- MR Spectroscopy Core Facility, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Jun Shen
- MR Spectroscopy Core Facility, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA; Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
76
|
Ding S, Gu Y, Cai Y, Cai M, Yang T, Bao S, Shen W, Ni X, Chen G, Xing L. Integrative systems and functional analyses reveal a role of dopaminergic signaling in myelin pathogenesis. J Transl Med 2020; 18:109. [PMID: 32122379 PMCID: PMC7053059 DOI: 10.1186/s12967-020-02276-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 02/18/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Myelin sheaths surrounding axons are critical for electrical signal transmission in the central nervous system (CNS). Diseases with myelin defects such as multiple sclerosis (MS) are devastating neurological conditions for which few effective treatments are available. Dysfunction of the dopaminergic system has been observed in multiple neurological disorders. Its role in myelin pathogenesis, however, is unclear. METHODS This work used a combination of literature curation, bioinformatics, pharmacological and genetic manipulation, as well as confocal imaging techniques. Literature search was used to establish a complete set of genes which is associated with MS in humans. Bioinformatics analyses include pathway enrichment and crosstalk analyses with human genetic association studies as well as gene set enrichment and causal relationship analyses with transcriptome data. Pharmacological and CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9) genetic manipulation were applied to inhibit the dopaminergic signaling in zebrafish. Imaging techniques were used to visualize myelin formation in vivo. RESULTS Systematic analysis of human genetic association studies revealed that the dopaminergic synapse signaling pathway is enriched in candidate gene sets. Transcriptome analysis confirmed that expression of multiple dopaminergic gene sets was significantly altered in patients with MS. Pathway crosstalk analysis and gene set causal relationship analysis reveal that the dopaminergic synapse signaling pathway interacts with or is associated with other critical pathways involved in MS. We also found that disruption of the dopaminergic system leads to myelin deficiency in zebrafish. CONCLUSIONS Dopaminergic signaling may be involved in myelin pathogenesis. This study may offer a novel molecular mechanism of demyelination in the nervous system.
Collapse
Affiliation(s)
- Sujun Ding
- School of Medicine, Nantong University, Nantong, China
- Department of Ultrasound, Affiliated Hospital of Nantong University, Nantong, China
| | - Yun Gu
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Yunyun Cai
- Department of Physiology, School of medicine, Nantong University, Nantong, China
| | - Meijuan Cai
- Department of Clinical Laboratory, Qilu Hospital of Shandong university, Shandong, China
| | - Tuo Yang
- Department of Hand Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Shuangxi Bao
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Weixing Shen
- Department of Physiology, School of medicine, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Xuejun Ni
- Department of Ultrasound, Affiliated Hospital of Nantong University, Nantong, China
| | - Gang Chen
- School of Medicine, Nantong University, Nantong, China
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, China
| | - Lingyan Xing
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
77
|
Bojcevski J, Stojic A, Hoffmann DB, Williams SK, Müller A, Diem R, Fairless R. Influence of retinal NMDA receptor activity during autoimmune optic neuritis. J Neurochem 2020; 153:693-709. [PMID: 32031240 DOI: 10.1111/jnc.14980] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 01/14/2020] [Accepted: 01/27/2020] [Indexed: 12/20/2022]
Abstract
Autoimmune optic neuritis (AON), a model of multiple sclerosis-associated optic neuritis, is accompanied by degeneration of retinal ganglion cells (RGCs) and optic nerve demyelination and axonal loss. In order to investigate the role of N-methyl-d-aspartate (NMDA) receptors in mediating RGC degeneration, upstream changes in the optic nerve actin cytoskeleton and associated deterioration in visual function, we induced AON in Brown Norway rats by immunization with myelin oligodendrocyte glycoprotein. Subsequently, visual acuity was assessed by recording visual evoked potentials and electroretinograms prior to extraction of optic nerves for western blot analysis and retinas for quantification of RGCs. As previously reported, in Brown Norway rats RGC degeneration is observed prior to onset of immune cell infiltration and demyelination of the optic nerves. However, within the optic nerve, destabilization of the actin cytoskeleton could be seen as indicated by an increase in the globular to filamentous actin ratio. Interestingly, these changes could be mimicked by intravitreal injection of glutamate, and similarly blocked by application of the NMDA receptor blocker MK-801, leading us to propose that prior to optic nerve lesion formation, NMDA receptor activation within the retina leads to retinal calcium accumulation, actin destabilization within the optic nerve as well as a deterioration of visual acuity during AON.
Collapse
Affiliation(s)
- Jovana Bojcevski
- Department of Neurology, University Clinic Heidelberg, Heidelberg, Germany.,CCU Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Aleksandar Stojic
- Department of Neurology, University Clinic Heidelberg, Heidelberg, Germany.,CCU Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Dorit B Hoffmann
- Department of Neurology, Saarland University, Homburg, Germany.,A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Sarah K Williams
- Department of Neurology, University Clinic Heidelberg, Heidelberg, Germany.,CCU Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Andreas Müller
- Department of Diagnostic and Interventional Radiology, Saarland University, Homburg, Germany
| | - Ricarda Diem
- Department of Neurology, University Clinic Heidelberg, Heidelberg, Germany.,CCU Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Richard Fairless
- Department of Neurology, University Clinic Heidelberg, Heidelberg, Germany.,CCU Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Centre (DKFZ), Heidelberg, Germany
| |
Collapse
|
78
|
Glutamate and Nitric Oxide as biomarkers for disease activity in patients with multiple sclerosis. Mult Scler Relat Disord 2020; 38:101873. [DOI: 10.1016/j.msard.2019.101873] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 10/24/2019] [Accepted: 11/24/2019] [Indexed: 11/23/2022]
|
79
|
Abstract
Multiple sclerosis (MS) affects approximately 1 million persons in the United States, and is the leading cause of neurological disability in young adults. The concept of precision medicine is now being applied to MS and has the promise of improved care. MS patients experience a variety of neurological symptoms, and disease severity ranges from mild to severe, and the biological underpinnings of these phenotypes are now starting to be elucidated. Precision medicine involves the classification of disease subtypes based on the underlying biology, rather than clinical phenotypes alone, and may govern disease course and treatment response. Over 18 disease-modifying drugs have been approved for the treatment of MS, and several biomarkers of treatment response are emerging. This article provides an overview of the concepts of precision medicine and emerging biological markers and their evolving role in decision-making in MS management.
Collapse
Affiliation(s)
- Tanuja Chitnis
- Tanuja Chitnis Partners Multiple Sclerosis Center, Department of Neurology, Brigham and Women’s Hospital, Boston, MA, USA/Harvard Medical School, Boston, MA, USA/Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Boston, MA, USA
| | - Alexandre Prat
- Alexandre Prat Department of Neurology, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
80
|
Kelly R, Joers V, Tansey MG, McKernan DP, Dowd E. Microglial Phenotypes and Their Relationship to the Cannabinoid System: Therapeutic Implications for Parkinson's Disease. Molecules 2020; 25:molecules25030453. [PMID: 31973235 PMCID: PMC7037317 DOI: 10.3390/molecules25030453] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/14/2020] [Accepted: 01/16/2020] [Indexed: 12/11/2022] Open
Abstract
Parkinson’s disease is a neurodegenerative disorder, the motor symptoms of which are associated classically with Lewy body formation and nigrostriatal degeneration. Neuroinflammation has been implicated in the progression of this disease, by which microglia become chronically activated in response to α-synuclein pathology and dying neurons, thereby acquiring dishomeostatic phenotypes that are cytotoxic and can cause further neuronal death. Microglia have a functional endocannabinoid signaling system, expressing the cannabinoid receptors in addition to being capable of synthesizing and degrading endocannabinoids. Alterations in the cannabinoid system—particularly an upregulation in the immunomodulatory CB2 receptor—have been demonstrated to be related to the microglial activation state and hence the microglial phenotype. This paper will review studies that examine the relationship between the cannabinoid system and microglial activation, and how this association could be manipulated for therapeutic benefit in Parkinson’s disease.
Collapse
Affiliation(s)
- Rachel Kelly
- Pharmacology & Therapeutics, National University of Ireland, H91 W5P7 Galway, Ireland; (R.K.); (D.P.M.)
| | - Valerie Joers
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL 32611, USA; (V.J.); (M.G.T.)
| | - Malú G. Tansey
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL 32611, USA; (V.J.); (M.G.T.)
- Center for Translation Research in Neurodegenerative Disease, University of Florida College of Medicine, Gainesville, FL 32611, USA
| | - Declan P. McKernan
- Pharmacology & Therapeutics, National University of Ireland, H91 W5P7 Galway, Ireland; (R.K.); (D.P.M.)
| | - Eilís Dowd
- Pharmacology & Therapeutics, National University of Ireland, H91 W5P7 Galway, Ireland; (R.K.); (D.P.M.)
- Correspondence:
| |
Collapse
|
81
|
Debnath A, Hariharan H, Nanga RPR, Reddy R, Singh A. Glutamate-Weighted CEST Contrast After Removal of Magnetization Transfer Effect in Human Brain and Rat Brain with Tumor. Mol Imaging Biol 2020; 22:1087-1101. [DOI: 10.1007/s11307-019-01465-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
82
|
Kirov II, Tal A. Potential clinical impact of multiparametric quantitative MR spectroscopy in neurological disorders: A review and analysis. Magn Reson Med 2020; 83:22-44. [PMID: 31393032 PMCID: PMC6814297 DOI: 10.1002/mrm.27912] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 06/06/2019] [Accepted: 06/29/2019] [Indexed: 12/12/2022]
Abstract
PURPOSE Unlike conventional MR spectroscopy (MRS), which only measures metabolite concentrations, multiparametric MRS also quantifies their longitudinal (T1 ) and transverse (T2 ) relaxation times, as well as the radiofrequency transmitter inhomogeneity (B1+ ). To test whether knowledge of these additional parameters can improve the clinical utility of brain MRS, we compare the conventional and multiparametric approaches in terms of expected classification accuracy in differentiating controls from patients with neurological disorders. THEORY AND METHODS A literature review was conducted to compile metabolic concentrations and relaxation times in a wide range of neuropathologies and regions of interest. Simulations were performed to construct receiver operating characteristic curves and compute the associated areas (area under the curve) to examine the sensitivity and specificity of MRS for detecting each pathology in each region. Classification accuracy was assessed using metabolite concentrations corrected using population-averages for T1 , T2 , and B1+ (conventional MRS); using metabolite concentrations corrected using per-subject values (multiparametric MRS); and using an optimal linear multiparametric estimator comprised of the metabolites' concentrations and relaxation constants (multiparametric MRS). Additional simulations were conducted to find the minimal intra-subject precision needed for each parameter. RESULTS Compared with conventional MRS, multiparametric approaches yielded area under the curve improvements for almost all neuropathologies and regions of interest. The median area under the curve increased by 0.14 over the entire dataset, and by 0.24 over the 10 instances with the largest individual increases. CONCLUSIONS Multiparametric MRS can substantially improve the clinical utility of MRS in diagnosing and assessing brain pathology, motivating the design and use of novel multiparametric sequences.
Collapse
Affiliation(s)
- Ivan I. Kirov
- Center for Advanced Imaging Innovation and Research (CAIR), Bernard and Irene Schwartz Center for Biomedical Imaging, New York University School of Medicine, Department of Radiology, 660 1 Avenue, New York, NY 10016, United States of America
| | - Assaf Tal
- Department of Chemical and Biological Physics, Weizmann Institute of Science, 234 Herzel St., Rehovot 7610001, Israel
| |
Collapse
|
83
|
Evonuk KS, Doyle RE, Moseley CE, Thornell IM, Adler K, Bingaman AM, Bevensee MO, Weaver CT, Min B, DeSilva TM. Reduction of AMPA receptor activity on mature oligodendrocytes attenuates loss of myelinated axons in autoimmune neuroinflammation. SCIENCE ADVANCES 2020; 6:eaax5936. [PMID: 31934627 PMCID: PMC6949032 DOI: 10.1126/sciadv.aax5936] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 11/08/2019] [Indexed: 06/10/2023]
Abstract
Glutamate dysregulation occurs in multiple sclerosis (MS), but whether excitotoxic mechanisms in mature oligodendrocytes contribute to demyelination and axonal injury is unexplored. Although current treatments modulate the immune system, long-term disability ensues, highlighting the need for neuroprotection. Glutamate is elevated before T2-visible white matter lesions appear in MS. We previously reported that myelin-reactive T cells provoke microglia to release glutamate from the system xc - transporter promoting myelin degradation in experimental autoimmune encephalomyelitis (EAE). Here, we explore the target for glutamate in mature oligodendrocytes. Most glutamate-stimulated calcium influx into oligodendrocyte somas is AMPA receptor (AMPAR)-mediated, and genetic deletion of AMPAR subunit GluA4 decreased intracellular calcium responses. Inducible deletion of GluA4 on mature oligodendrocytes attenuated EAE and loss of myelinated axons was selectively reduced compared to unmyelinated axons. These data link AMPAR signaling in mature oligodendrocytes to the pathophysiology of myelinated axons, demonstrating glutamate regulation as a potential neuroprotective strategy in MS.
Collapse
Affiliation(s)
- Kirsten S. Evonuk
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Ryan E. Doyle
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Carson E. Moseley
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
- University of California, San Francisco, CA, USA
| | - Ian M. Thornell
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Keith Adler
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Amanda M. Bingaman
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Mark O. Bevensee
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Casey T. Weaver
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Booki Min
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Tara M. DeSilva
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| |
Collapse
|
84
|
Shen J, Shenkar D, An L, Tomar JS. Local and Interregional Neurochemical Associations Measured by Magnetic Resonance Spectroscopy for Studying Brain Functions and Psychiatric Disorders. Front Psychiatry 2020; 11:802. [PMID: 32848957 PMCID: PMC7432119 DOI: 10.3389/fpsyt.2020.00802] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 07/27/2020] [Indexed: 12/11/2022] Open
Abstract
Magnetic resonance spectroscopy (MRS) studies have found significant correlations among neurometabolites (e.g., between glutamate and GABA) across individual subjects and altered correlations in neuropsychiatric disorders. In this article, we discuss neurochemical associations among several major neurometabolites which underpin these observations by MRS. We also illustrate the role of spectral editing in eliminating unwanted correlations caused by spectral overlapping. Finally, we describe the prospects of mapping macroscopic neurochemical associations across the brain and characterizing excitation-inhibition balance of neural networks using glutamate- and GABA-editing MRS imaging.
Collapse
Affiliation(s)
- Jun Shen
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, United States
| | - Dina Shenkar
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, United States
| | - Li An
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, United States
| | - Jyoti Singh Tomar
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, United States
| |
Collapse
|
85
|
Chen Y, Li Y, Xu Z. Improved Low-Rank Filtering of MR Spectroscopic Imaging Data With Pre-Learnt Subspace and Spatial Constraints. IEEE Trans Biomed Eng 2019; 67:2381-2388. [PMID: 31870975 DOI: 10.1109/tbme.2019.2961698] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVE To investigate the use of pre-learnt subspace and spatial constraints for denoising magnetic resonance spectroscopic imaging (MRSI) data. METHOD We exploit the partial separability or subspace structures of high-dimensional MRSI data for denoising. More specifically, we incorporate a subspace model with pre-learnt spectral basis into the low-rank approximation (LORA) method. Spectral basis is determined based on empirical prior distributions of the spectral parameters variations learnt from auxiliary training data; spatial priors are also incorporated as is done in LORA to further improve denoising performance. RESULTS The effects of the explicit subspace and spatial constraints in reducing estimation bias and variance have been analyzed using Cramér-Rao Lower bound analysis, Monte-Carlo study, and experimental study. CONCLUSION The denoising effectiveness of LORA can be significantly improved by incorporating pre-learnt spectral basis and spatial priors into LORA. SIGNIFICANCE This study provides an effective method for denoising MRSI data along with comprehensive analyses of its performance. The proposed method is expected to be useful for a wide range of studies using MRSI.
Collapse
|
86
|
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]
|
87
|
Andravizou A, Dardiotis E, Artemiadis A, Sokratous M, Siokas V, Tsouris Z, Aloizou AM, Nikolaidis I, Bakirtzis C, Tsivgoulis G, Deretzi G, Grigoriadis N, Bogdanos DP, Hadjigeorgiou GM. Brain atrophy in multiple sclerosis: mechanisms, clinical relevance and treatment options. AUTO- IMMUNITY HIGHLIGHTS 2019; 10:7. [PMID: 32257063 PMCID: PMC7065319 DOI: 10.1186/s13317-019-0117-5] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 06/28/2019] [Indexed: 12/23/2022]
Abstract
Multiple sclerosis (MS) is an immune-mediated disease of the central nervous system characterized by focal or diffuse inflammation, demyelination, axonal loss and neurodegeneration. Brain atrophy can be seen in the earliest stages of MS, progresses faster compared to healthy adults, and is a reliable predictor of future physical and cognitive disability. In addition, it is widely accepted to be a valid, sensitive and reproducible measure of neurodegeneration in MS. Reducing the rate of brain atrophy has only recently been incorporated as a critical endpoint into the clinical trials of new or emerging disease modifying drugs (DMDs) in MS. With the advent of easily accessible neuroimaging softwares along with the accumulating evidence, clinicians may be able to use brain atrophy measures in their everyday clinical practice to monitor disease course and response to DMDs. In this review, we will describe the different mechanisms contributing to brain atrophy, their clinical relevance on disease presentation and course and the effect of current or emergent DMDs on brain atrophy and neuroprotection.
Collapse
Affiliation(s)
- Athina Andravizou
- Department of Neurology, Laboratory of Neurogenetics, Faculty of Medicine, University of Thessaly, University Hospital of Larissa, Biopolis, Mezourlo Hill, 41100 Larissa, Greece
| | - Efthimios Dardiotis
- Department of Neurology, Laboratory of Neurogenetics, Faculty of Medicine, University of Thessaly, University Hospital of Larissa, Biopolis, Mezourlo Hill, 41100 Larissa, Greece
| | - Artemios Artemiadis
- Immunogenetics Laboratory, 1st Department of Neurology, Medical School, National and Kapodistrian University of Athens, Aeginition Hospital, Vas. Sophias Ave 72-74, 11528 Athens, Greece
| | - Maria Sokratous
- Department of Neurology, Laboratory of Neurogenetics, Faculty of Medicine, University of Thessaly, University Hospital of Larissa, Biopolis, Mezourlo Hill, 41100 Larissa, Greece
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University General Hospital of Larissa, University of Thessaly, Viopolis, 40500 Larissa, Greece
| | - Vasileios Siokas
- Department of Neurology, Laboratory of Neurogenetics, Faculty of Medicine, University of Thessaly, University Hospital of Larissa, Biopolis, Mezourlo Hill, 41100 Larissa, Greece
| | - Zisis Tsouris
- Department of Neurology, Laboratory of Neurogenetics, Faculty of Medicine, University of Thessaly, University Hospital of Larissa, Biopolis, Mezourlo Hill, 41100 Larissa, Greece
| | - Athina-Maria Aloizou
- Department of Neurology, Laboratory of Neurogenetics, Faculty of Medicine, University of Thessaly, University Hospital of Larissa, Biopolis, Mezourlo Hill, 41100 Larissa, Greece
| | - Ioannis Nikolaidis
- Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Christos Bakirtzis
- Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Georgios Tsivgoulis
- Second Department of Neurology, School of Medicine, University of Athens, “Attikon” University Hospital, Athens, Greece
| | - Georgia Deretzi
- Department of Neurology, Papageorgiou General Hospital, Thessaloniki, Greece
| | - Nikolaos Grigoriadis
- Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Dimitrios P. Bogdanos
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University General Hospital of Larissa, University of Thessaly, Viopolis, 40500 Larissa, Greece
| | - Georgios M. Hadjigeorgiou
- Department of Neurology, Laboratory of Neurogenetics, Faculty of Medicine, University of Thessaly, University Hospital of Larissa, Biopolis, Mezourlo Hill, 41100 Larissa, Greece
- Department of Neurology, Medical School, University of Cyprus, Nicosia, Cyprus
| |
Collapse
|
88
|
Swanberg KM, Landheer K, Pitt D, Juchem C. Quantifying the Metabolic Signature of Multiple Sclerosis by in vivo Proton Magnetic Resonance Spectroscopy: Current Challenges and Future Outlook in the Translation From Proton Signal to Diagnostic Biomarker. Front Neurol 2019; 10:1173. [PMID: 31803127 PMCID: PMC6876616 DOI: 10.3389/fneur.2019.01173] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 10/21/2019] [Indexed: 01/03/2023] Open
Abstract
Proton magnetic resonance spectroscopy (1H-MRS) offers a growing variety of methods for querying potential diagnostic biomarkers of multiple sclerosis in living central nervous system tissue. For the past three decades, 1H-MRS has enabled the acquisition of a rich dataset suggestive of numerous metabolic alterations in lesions, normal-appearing white matter, gray matter, and spinal cord of individuals with multiple sclerosis, but this body of information is not free of seeming internal contradiction. The use of 1H-MRS signals as diagnostic biomarkers depends on reproducible and generalizable sensitivity and specificity to disease state that can be confounded by a multitude of influences, including experiment group classification and demographics; acquisition sequence; spectral quality and quantifiability; the contribution of macromolecules and lipids to the spectroscopic baseline; spectral quantification pipeline; voxel tissue and lesion composition; T1 and T2 relaxation; B1 field characteristics; and other features of study design, spectral acquisition and processing, and metabolite quantification about which the experimenter may possess imperfect or incomplete information. The direct comparison of 1H-MRS data from individuals with and without multiple sclerosis poses a special challenge in this regard, as several lines of evidence suggest that experimental cohorts may differ significantly in some of these parameters. We review the existing findings of in vivo1H-MRS on central nervous system metabolic abnormalities in multiple sclerosis and its subtypes within the context of study design, spectral acquisition and processing, and metabolite quantification and offer an outlook on technical considerations, including the growing use of machine learning, by future investigations into diagnostic biomarkers of multiple sclerosis measurable by 1H-MRS.
Collapse
Affiliation(s)
- Kelley M Swanberg
- Department of Biomedical Engineering, Columbia University Fu Foundation School of Engineering and Applied Science, New York, NY, United States
| | - Karl Landheer
- Department of Biomedical Engineering, Columbia University Fu Foundation School of Engineering and Applied Science, New York, NY, United States
| | - David Pitt
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States
| | - Christoph Juchem
- Department of Biomedical Engineering, Columbia University Fu Foundation School of Engineering and Applied Science, New York, NY, United States.,Department of Radiology, Columbia University College of Physicians and Surgeons, New York, NY, United States
| |
Collapse
|
89
|
Tobore TO. Towards a comprehensive etiopathogenetic and pathophysiological theory of multiple sclerosis. Int J Neurosci 2019; 130:279-300. [PMID: 31588832 DOI: 10.1080/00207454.2019.1677648] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background: Multiple sclerosis (MS) is a neurodegenerative disease caused by dysfunction of the immune system that affects the central nervous system (CNS). It is characterized by demyelination, chronic inflammation, neuronal and oligodendrocyte loss and reactive astrogliosis. It can result in physical disability and acute neurological and cognitive problems. Despite the gains in knowledge of immunology, cell biology, and genetics in the last five decades, the ultimate etiology or specific elements that trigger MS remain unknown. The objective of this review is to propose a theoretical basis for MS etiopathogenesis.Methods: Search was done by accessing PubMed/Medline, EBSCO, and PsycINFO databases. The search string used was "(multiple sclerosis* OR EAE) AND (pathophysiology* OR etiopathogenesis)". The electronic databases were searched for titles or abstracts containing these terms in all published articles between January 1, 1960, and June 30, 2019. The search was filtered down to 362 articles which were included in this review.Results: A framework to better understand the etiopathogenesis and pathophysiology of MS can be derived from four essential factors; mitochondria dysfunction (MtD) & oxidative stress (OS), vitamin D (VD), sex hormones and thyroid hormones. These factors play a direct role in MS etiopathogenesis and have a modulatory effect on many other factors involved in the disease.Conclusions: For better MS prevention and treatment outcomes, efforts should be geared towards treating thyroid problems, sex hormone alterations, VD deficiency, sleep problems and melatonin alterations. MS patients should be encouraged to engage in activities that boost total antioxidant capacity (TAC) including diet and regular exercise and discouraged from activities that promote OS including smoking and alcohol consumption.
Collapse
|
90
|
Hollinger KR, Smith MD, Kirby LA, Prchalova E, Alt J, Rais R, Calabresi PA, Slusher BS. Glutamine antagonism attenuates physical and cognitive deficits in a model of MS. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2019; 6:e609. [PMID: 31467038 PMCID: PMC6745721 DOI: 10.1212/nxi.0000000000000609] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 07/09/2019] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To measure the impact of JHU-083, a novel prodrug of the glutamine antagonist 6-diazo-5-oxo-l-norleucine, on immune cell proliferation and activation, along with physical and cognitive impairments associated with the experimental autoimmune encephalomyelitis (EAE) mouse model of MS. METHODS Splenic-derived T cells and bone marrow-derived dendritic cells (DCs) were cultured, activated, and treated daily with vehicle or JHU-083. Proliferation and activation were measured via flow cytometry and IncuCyte live cell analysis. C57BL/6 mice were immunized for EAE. Vehicle or JHU-083 was administered orally every other day either from the time of immunization in the prevention paradigm or from the time of disease onset in the treatment paradigm. Disease scores and body weight were monitored. In the treatment paradigm, cognition was evaluated using the Barnes maze test. RESULTS JHU-083 selectively inhibits T-cell proliferation and decreases T-cell activation, with no effect on DCs. In vivo, orally administered JHU-083 significantly decreases EAE severity in both prevention and treatment paradigms and reverses EAE-induced cognitive impairment. CONCLUSIONS JHU-083, a well-tolerated, brain penetrable glutamine antagonist, is a promising novel treatment for both the physical and cognitive deficits of MS.
Collapse
Affiliation(s)
- Kristen R Hollinger
- From the Johns Hopkins Drug Discovery (K.R.H., E.P., J.A., R.R., B.S.S.), Johns Hopkins University; and Department of Psychiatry and Behavioral Sciences (K.R.H., B.S.S.), Department of Neurology (K.R.H., M.D.S., L.A.K., E.P., R.R., P.A.C., B.S.S.), Department of Neuroscience (B.S.S.), Department of Medicine (B.S.S.), Department of Oncology (B.S.S.), Johns Hopkins University, Baltimore, MD
| | - Matthew D Smith
- From the Johns Hopkins Drug Discovery (K.R.H., E.P., J.A., R.R., B.S.S.), Johns Hopkins University; and Department of Psychiatry and Behavioral Sciences (K.R.H., B.S.S.), Department of Neurology (K.R.H., M.D.S., L.A.K., E.P., R.R., P.A.C., B.S.S.), Department of Neuroscience (B.S.S.), Department of Medicine (B.S.S.), Department of Oncology (B.S.S.), Johns Hopkins University, Baltimore, MD
| | - Leslie A Kirby
- From the Johns Hopkins Drug Discovery (K.R.H., E.P., J.A., R.R., B.S.S.), Johns Hopkins University; and Department of Psychiatry and Behavioral Sciences (K.R.H., B.S.S.), Department of Neurology (K.R.H., M.D.S., L.A.K., E.P., R.R., P.A.C., B.S.S.), Department of Neuroscience (B.S.S.), Department of Medicine (B.S.S.), Department of Oncology (B.S.S.), Johns Hopkins University, Baltimore, MD
| | - Eva Prchalova
- From the Johns Hopkins Drug Discovery (K.R.H., E.P., J.A., R.R., B.S.S.), Johns Hopkins University; and Department of Psychiatry and Behavioral Sciences (K.R.H., B.S.S.), Department of Neurology (K.R.H., M.D.S., L.A.K., E.P., R.R., P.A.C., B.S.S.), Department of Neuroscience (B.S.S.), Department of Medicine (B.S.S.), Department of Oncology (B.S.S.), Johns Hopkins University, Baltimore, MD
| | - Jesse Alt
- From the Johns Hopkins Drug Discovery (K.R.H., E.P., J.A., R.R., B.S.S.), Johns Hopkins University; and Department of Psychiatry and Behavioral Sciences (K.R.H., B.S.S.), Department of Neurology (K.R.H., M.D.S., L.A.K., E.P., R.R., P.A.C., B.S.S.), Department of Neuroscience (B.S.S.), Department of Medicine (B.S.S.), Department of Oncology (B.S.S.), Johns Hopkins University, Baltimore, MD
| | - Rana Rais
- From the Johns Hopkins Drug Discovery (K.R.H., E.P., J.A., R.R., B.S.S.), Johns Hopkins University; and Department of Psychiatry and Behavioral Sciences (K.R.H., B.S.S.), Department of Neurology (K.R.H., M.D.S., L.A.K., E.P., R.R., P.A.C., B.S.S.), Department of Neuroscience (B.S.S.), Department of Medicine (B.S.S.), Department of Oncology (B.S.S.), Johns Hopkins University, Baltimore, MD
| | - Peter A Calabresi
- From the Johns Hopkins Drug Discovery (K.R.H., E.P., J.A., R.R., B.S.S.), Johns Hopkins University; and Department of Psychiatry and Behavioral Sciences (K.R.H., B.S.S.), Department of Neurology (K.R.H., M.D.S., L.A.K., E.P., R.R., P.A.C., B.S.S.), Department of Neuroscience (B.S.S.), Department of Medicine (B.S.S.), Department of Oncology (B.S.S.), Johns Hopkins University, Baltimore, MD.
| | - Barbara S Slusher
- From the Johns Hopkins Drug Discovery (K.R.H., E.P., J.A., R.R., B.S.S.), Johns Hopkins University; and Department of Psychiatry and Behavioral Sciences (K.R.H., B.S.S.), Department of Neurology (K.R.H., M.D.S., L.A.K., E.P., R.R., P.A.C., B.S.S.), Department of Neuroscience (B.S.S.), Department of Medicine (B.S.S.), Department of Oncology (B.S.S.), Johns Hopkins University, Baltimore, MD.
| |
Collapse
|
91
|
Tobore TO. On elucidation of the role of mitochondria dysfunction and oxidative stress in multiple sclerosis. ACTA ACUST UNITED AC 2019. [DOI: 10.1111/ncn3.12335] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
92
|
Rao VTS, Fuh SC, Karamchandani JR, Woulfe JMJ, Munoz DG, Ellezam B, Blain M, Ho MK, Bedell BJ, Antel JP, Ludwin SK. Astrocytes in the Pathogenesis of Multiple Sclerosis: An In Situ MicroRNA Study. J Neuropathol Exp Neurol 2019; 78:1130-1146. [DOI: 10.1093/jnen/nlz098] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Abstract
Astrocytes are increasingly recognized as active contributors to the disease process in multiple sclerosis (MS), rather than being merely reactive. We investigated the expression of a selected microRNA (miRNA) panel that could contribute both to the injury and to the recovery phases of the disease. Individual astrocytes were laser microdissected from brain sections. We then compared the miRNAs’ expressions in MS and control brain samples at different lesional stages in white versus grey matter regions. In active MS lesions, we found upregulation of ischemia-related miRNAs in white but not grey matter, often with reversion to the normal state in inactive lesions. In contrast to our previous findings on MS macrophages, expression of 2 classical inflammatory-related miRNAs, miRNA-155 and miRNA-146a, was reduced in astrocytes from active and chronic active MS lesions in white and grey matter, suggesting a lesser direct pathogenetic role for these miRNAs in astrocytes. miRNAs within the categories regulating aquaporin4 (-100, -145, -320) and glutamate transport/apoptosis/neuroprotection (-124a, -181a, and -29a) showed some contrasting responses. The regional and lesion-stage differences of expression of these miRNAs indicate the remarkable ability of astrocytes to show a wide range of selective responses in the face of differing insults and phases of resolution.
Collapse
Affiliation(s)
- Vijayaraghava T S Rao
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University
| | - Shih-Chieh Fuh
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | | | - John M J Woulfe
- Department of Pathology, The Ottawa Hospital, University of Ottawa
| | - David G Munoz
- Department of Pathology, St. Michaels Hospital, Toronto University, Toronto
| | | | - Manon Blain
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON, Canada
| | - Ming-Kai Ho
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University
| | - Barry J Bedell
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Jack P Antel
- Department of Neuropathology, Montreal Neurological Institute
| | - Samuel K Ludwin
- Department of Pathology, The Ottawa Hospital, University of Ottawa
| |
Collapse
|
93
|
Landheer K, Schulte RF, Treacy MS, Swanberg KM, Juchem C. Theoretical description of modern1H in Vivo magnetic resonance spectroscopic pulse sequences. J Magn Reson Imaging 2019; 51:1008-1029. [DOI: 10.1002/jmri.26846] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/14/2019] [Accepted: 06/17/2019] [Indexed: 01/20/2023] Open
Affiliation(s)
- Karl Landheer
- Biomedical Engineering, Columbia University Fu Foundation School of Engineering and Applied Science New York New York USA
| | | | - Michael S. Treacy
- Biomedical Engineering, Columbia University Fu Foundation School of Engineering and Applied Science New York New York USA
| | - Kelley M. Swanberg
- Biomedical Engineering, Columbia University Fu Foundation School of Engineering and Applied Science New York New York USA
| | - Christoph Juchem
- Biomedical Engineering, Columbia University Fu Foundation School of Engineering and Applied Science New York New York USA
- Radiology, Columbia University College of Physicians and Surgeons New York New York USA
| |
Collapse
|
94
|
Coe S, Cossington J, Collett J, Soundy A, Izadi H, Ovington M, Durkin L, Kirsten M, Clegg M, Cavey A, Wade DT, Palace J, DeLuca GC, Chapman K, Harrison JM, Buckingham E, Dawes H. A randomised double-blind placebo-controlled feasibility trial of flavonoid-rich cocoa for fatigue in people with relapsing and remitting multiple sclerosis. J Neurol Neurosurg Psychiatry 2019; 90:507-513. [PMID: 30833449 DOI: 10.1136/jnnp-2018-319496] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 12/21/2018] [Accepted: 12/26/2018] [Indexed: 02/02/2023]
Abstract
UNLABELLED The impact of flavonoids on fatigue has not been investigated in relapsing and remitting multiple sclerosis (RRMS). OBJECTIVE To determine the feasibility and estimate the potential effect of flavonoid-rich cocoa on fatigue and fatigability in RRMS. METHODS A randomised double-blind placebo-controlled feasibility study in people recently diagnosed with RRMS and fatigue, throughout the Thames Valley, UK (ISRCTN69897291). During a 6-week intervention participants consumed a high or low flavonoid cocoa beverage daily. Fatigue and fatigability were measured at three visits (weeks 0, 3 and 6). Feasibility and fidelity were assessed through recruitment and retention, adherence and a process evaluation. RESULTS 40 people with multiple sclerosis (10 men, 30 women, age 44±10 years) were randomised and allocated to high (n=19) or low (n=21) flavonoid groups and included in analysis. Missing data were <20% and adherence to intervention of allocated individuals was >75%. There was a small effect on fatigue (Neuro-QoL: effect size (ES) 0.04, 95% CI -0.40 to 0.48) and a moderate effect on fatigability (6 min walk test: ES 0.45, 95% CI -0.18 to 1.07). There were seven adverse events (four control, three intervention), only one of which was possibly related and it was resolved. CONCLUSION A flavonoid beverage demonstrates the potential to improve fatigue and fatigability in RRMS.
Collapse
Affiliation(s)
- Shelly Coe
- Department of Sport Health Sciences and Social Work, Centre for Movement Occupational and Rehabilitation Sciences, Oxford Brookes Centre for Nutrition and Health, Oxford Brookes Univeristy, Oxford, UK
| | - Jo Cossington
- Department of Sport Health Sciences and Social Work, Centre for Movement Occupational and Rehabilitation Sciences, Oxford Brookes Centre for Nutrition and Health, Oxford Brookes Univeristy, Oxford, UK
| | - Johnny Collett
- Department of Sport Health Sciences and Social Work, Centre for Movement Occupational and Rehabilitation Sciences, Oxford Brookes Centre for Nutrition and Health, Oxford Brookes Univeristy, Oxford, UK
| | - Andrew Soundy
- Department of Neurology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Hooshang Izadi
- Department of Sport Health Sciences and Social Work, Centre for Movement Occupational and Rehabilitation Sciences, Oxford Brookes Centre for Nutrition and Health, Oxford Brookes Univeristy, Oxford, UK
| | - Martin Ovington
- Department of Sport Health Sciences and Social Work, Centre for Movement Occupational and Rehabilitation Sciences, Oxford Brookes Centre for Nutrition and Health, Oxford Brookes Univeristy, Oxford, UK
| | - Luke Durkin
- Department of Sport Health Sciences and Social Work, Centre for Movement Occupational and Rehabilitation Sciences, Oxford Brookes Centre for Nutrition and Health, Oxford Brookes Univeristy, Oxford, UK
| | - Maja Kirsten
- Department of Sport Health Sciences and Social Work, Centre for Movement Occupational and Rehabilitation Sciences, Oxford Brookes Centre for Nutrition and Health, Oxford Brookes Univeristy, Oxford, UK
| | - Miriam Clegg
- Department of Food and Nutritional Sciences, University of Reading, Reading, UK
| | - Ana Cavey
- University of Birmingham, Birmingham, UK
| | - Derick T Wade
- Department of Sport Health Sciences and Social Work, Centre for Movement Occupational and Rehabilitation Sciences, Oxford Brookes Centre for Nutrition and Health, Oxford Brookes Univeristy, Oxford, UK
| | - Jacqueline Palace
- Department of Neurology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Gabriele C DeLuca
- Department of Neurology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Kim Chapman
- Department of Sport Health Sciences and Social Work, Centre for Movement Occupational and Rehabilitation Sciences, Oxford Brookes Centre for Nutrition and Health, Oxford Brookes Univeristy, Oxford, UK
| | - Jane-Marie Harrison
- Department of Sport Health Sciences and Social Work, Centre for Movement Occupational and Rehabilitation Sciences, Oxford Brookes Centre for Nutrition and Health, Oxford Brookes Univeristy, Oxford, UK
| | - Elizabeth Buckingham
- Department of Sport Health Sciences and Social Work, Centre for Movement Occupational and Rehabilitation Sciences, Oxford Brookes Centre for Nutrition and Health, Oxford Brookes Univeristy, Oxford, UK
| | - Helen Dawes
- Department of Sport Health Sciences and Social Work, Centre for Movement Occupational and Rehabilitation Sciences, Oxford Brookes Centre for Nutrition and Health, Oxford Brookes Univeristy, Oxford, UK
| |
Collapse
|
95
|
Aiello M, Cavaliere C, Fiorenza D, Duggento A, Passamonti L, Toschi N. Neuroinflammation in Neurodegenerative Diseases: Current Multi-modal Imaging Studies and Future Opportunities for Hybrid PET/MRI. Neuroscience 2019; 403:125-135. [DOI: 10.1016/j.neuroscience.2018.07.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 07/18/2018] [Accepted: 07/19/2018] [Indexed: 12/28/2022]
|
96
|
Fingolimod reduces neuropathic pain behaviors in a mouse model of multiple sclerosis by a sphingosine-1 phosphate receptor 1-dependent inhibition of central sensitization in the dorsal horn. Pain 2019; 159:224-238. [PMID: 29140922 DOI: 10.1097/j.pain.0000000000001106] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune-inflammatory neurodegenerative disease that is often accompanied by a debilitating neuropathic pain. Disease-modifying agents slow down the progression of multiple sclerosis and prevent relapses, yet it remains unclear if they yield analgesia. We explored the analgesic potential of fingolimod (FTY720), an agonist and/or functional antagonist at the sphingosine-1-phosphate receptor 1 (S1PR1), because it reduces hyperalgesia in models of peripheral inflammatory and neuropathic pain. We used a myelin oligodendrocyte glycoprotein 35 to 55 (MOG35-55) mouse model of experimental autoimmune encephalomyelitis, modified to avoid frank paralysis, and thus, allow for assessment of withdrawal behaviors to somatosensory stimuli. Daily intraperitoneal fingolimod reduced behavioral signs of central neuropathic pain (mechanical and cold hypersensitivity) in a dose-dependent and reversible manner. Both autoimmune encephalomyelitis and fingolimod changed hyperalgesia before modifying motor function, suggesting that pain-related effects and clinical neurological deficits were modulated independently. Fingolimod also reduced cellular markers of central sensitization of neurons in the dorsal horn of the spinal cord: glutamate-evoked Ca signaling and stimulus-evoked phospho-extracellular signal-related kinase ERK (pERK) expression, as well as upregulation of astrocytes (GFAP) and macrophage/microglia (Iba1) immunoreactivity. The antihyperalgesic effects of fingolimod were prevented or reversed by the S1PR1 antagonist W146 (1 mg/kg daily, i.p.) and could be mimicked by either repeated or single injection of the S1PR1-selective agonist SEW2871. Fingolimod did not change spinal membrane S1PR1 content, arguing against a functional antagonist mechanism. We conclude that fingolimod behaves as an S1PR1 agonist to reduce pain in multiple sclerosis by reversing central sensitization of spinal nociceptive neurons.
Collapse
|
97
|
Henstridge CM, Tzioras M, Paolicelli RC. Glial Contribution to Excitatory and Inhibitory Synapse Loss in Neurodegeneration. Front Cell Neurosci 2019; 13:63. [PMID: 30863284 PMCID: PMC6399113 DOI: 10.3389/fncel.2019.00063] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 02/08/2019] [Indexed: 12/12/2022] Open
Abstract
Synapse loss is an early feature shared by many neurodegenerative diseases, and it represents the major correlate of cognitive impairment. Recent studies reveal that microglia and astrocytes play a major role in synapse elimination, contributing to network dysfunction associated with neurodegeneration. Excitatory and inhibitory activity can be affected by glia-mediated synapse loss, resulting in imbalanced synaptic transmission and subsequent synaptic dysfunction. Here, we review the recent literature on the contribution of glia to excitatory/inhibitory imbalance, in the context of the most common neurodegenerative disorders. A better understanding of the mechanisms underlying pathological synapse loss will be instrumental to design targeted therapeutic interventions, taking in account the emerging roles of microglia and astrocytes in synapse remodeling.
Collapse
Affiliation(s)
- Christopher M Henstridge
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom.,Dementia Research Institute UK, The University of Edinburgh, Edinburgh, United Kingdom
| | - Makis Tzioras
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom.,Dementia Research Institute UK, The University of Edinburgh, Edinburgh, United Kingdom
| | - Rosa C Paolicelli
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
98
|
Kuzmina US, Zainullina LF, Vakhitov VA, Bakhtiyarova KZ, Vakhitova YV. The role of glutamate in the pathogenesis of multiple sclerosis. Zh Nevrol Psikhiatr Im S S Korsakova 2019; 119:160-167. [DOI: 10.17116/jnevro2019119081160] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
99
|
Fern R, Matute C. Glutamate receptors and white matter stroke. Neurosci Lett 2018; 694:86-92. [PMID: 30476568 DOI: 10.1016/j.neulet.2018.11.031] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 11/19/2018] [Accepted: 11/21/2018] [Indexed: 12/23/2022]
Abstract
White matter (WM) damage during ischemia occurs at multiple sites including myelin, oligodendrocytes, astrocytes and axons. A major driver of WM demise is excitoxicity as a consequence of excessive glutamate release by vesicular and non-vesicular mechanisms from axons and glial cells. This results in over-activation of ionotropic glutamate receptors (GluRs) profusely expressed by all cell compartments in WM. Thus, blocking excitotoxicity in WM with selective antagonists of those receptors has a potential therapeutic value. The significance of WM GluR expression for WM stroke injury is the focus of this review, and we will examine the role of GluRs in injury to myelin, oligodendrocytes, astrocytes and the axon cylinder.
Collapse
Affiliation(s)
- Robert Fern
- Faculty of Medicine and Dentistry, University of Plymouth, Plymouth, United Kingdom
| | - Carlos Matute
- Achucarro Basque Center for Neuroscience, CIBERNED and Department of Neuroscience, University of the Basque Country, Leioa, Spain.
| |
Collapse
|
100
|
O'Grady KP, Dula AN, Lyttle BD, Thompson LM, Conrad BN, Box BA, McKeithan LJ, Pawate S, Bagnato F, Landman BA, Newhouse P, Smith SA. Glutamate-sensitive imaging and evaluation of cognitive impairment in multiple sclerosis. Mult Scler 2018; 25:1580-1592. [PMID: 30230400 DOI: 10.1177/1352458518799583] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND Cognitive impairment (CI) profoundly impacts quality of life for patients with multiple sclerosis (MS). Dysfunctional regulation of glutamate in gray matter (GM) has been implicated in the pathogenesis of MS by post-mortem pathological studies and in CI by in vivo magnetic resonance spectroscopy, yet GM pathology is subtle and difficult to detect using conventional T1- and T2-weighted magnetic resonance imaging (MRI). There is a need for high-resolution, clinically accessible imaging techniques that probe molecular changes in GM. OBJECTIVE To study cortical GM pathology related to CI in MS using glutamate-sensitive chemical exchange saturation transfer (GluCEST) MRI at 7.0 Tesla (7T). METHODS A total of 20 patients with relapsing-remitting MS and 20 healthy controls underwent cognitive testing, anatomical imaging, and GluCEST imaging. Glutamate-sensitive image contrast was quantified for cortical GM, compared between cohorts, and correlated with clinical measures of CI. RESULTS AND CONCLUSION Glutamate-sensitive contrast was significantly increased in the prefrontal cortex of MS patients with accumulated disability (p < 0.05). In addition, glutamate-sensitive contrast in the prefrontal cortex was significantly correlated with symbol digit modality test (rS = -0.814) and choice reaction time (rS = 0.772) scores in patients (p < 0.05), suggesting that GluCEST MRI may have utility as a marker for GM pathology and CI.
Collapse
Affiliation(s)
- Kristin P O'Grady
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Adrienne N Dula
- Department of Neurology, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Bailey D Lyttle
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lindsey M Thompson
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Benjamin N Conrad
- Neuroscience Graduate Program, Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bailey A Box
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lydia J McKeithan
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Siddharama Pawate
- Vanderbilt Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Francesca Bagnato
- Neuroimaging Unit, Neuroimmunology Division, Vanderbilt Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bennett A Landman
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA/Department of Electrical Engineering and Computer Science, Vanderbilt University, Nashville, TN, USA/Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA/Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Paul Newhouse
- Department of Psychiatry and Behavioral Sciences, Center for Cognitive Medicine, Vanderbilt University Medical Center, Nashville, TN, USA/Veterans Affairs Tennessee Valley Healthcare System Geriatric Research, Education, and Clinical Center (VA TVHS GRECC), Nashville, TN, USA
| | - Seth A Smith
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA/Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA/Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|