1
|
Yoon S, Lee S, Joo Y, Ha E, Hong H, Song Y, Lee H, Kim S, Suh C, Lee CJ, Lyoo IK. Variations in Brain Glutamate and Glutamine Levels Throughout the Sleep-Wake Cycle. Biol Psychiatry 2024:S0006-3223(24)01785-2. [PMID: 39643103 DOI: 10.1016/j.biopsych.2024.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 10/25/2024] [Accepted: 11/21/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Glutamatergic signaling is essential for modulating synaptic plasticity and cognition. However, the dynamics of glutamatergic activity over the 24-hour sleep-wake cycle, particularly in relation to sleep, remain poorly understood. In this study, we aimed to investigate diurnal variations in brain Glx levels-representing the combined concentrations of glutamate and glutamine-in humans and to explore their implications for cognitive performance and sleep pressure. METHODS We conducted 2 independent experiments to measure Glx levels across the sleep-wake cycle using proton magnetic resonance spectroscopy. In experiment 1, 14 participants underwent 13 hours of Glx measurements during a typical sleep-wake cycle. Experiment 2 extended these measurements to an around-the-clock observation over a 6-day period. This period included 2 days of normal sleep-wake cycles, 24 hours of enforced wakefulness, and a 3-day recovery phase. Seven participants took part in experiment 2. RESULTS The study observed that brain Glx levels increased during wakefulness and decreased during sleep. Notably, Glx levels were lower during enforced wakefulness than during normal wakefulness. Reduced Glx levels were associated with diminished cognitive performance, while greater Glx exposure over the preceding 24 hours correlated with increased sleep pressure. CONCLUSIONS These findings suggest that Glx accumulation may contribute to increased sleep pressure, while its reduction appears to support wakefulness. These observations, together with the diurnal variations in Glx levels, underscore the dynamic nature of glutamatergic activity across the daily cycle. Further research is warranted to explore the potential role of sleep in regulating glutamatergic homeostasis.
Collapse
Affiliation(s)
- Sujung Yoon
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea; Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, South Korea
| | - Suji Lee
- Division of Psychology and Cognitive Science, Seoul Women's University, Seoul, South Korea
| | - Yoonji Joo
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea
| | - Eunji Ha
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea
| | - Haejin Hong
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea
| | - Yumi Song
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea; Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, South Korea
| | - Hyangwon Lee
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea; Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, South Korea
| | - Shinhye Kim
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea; Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, South Korea
| | - Chaewon Suh
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, South Korea
| | - In Kyoon Lyoo
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea; Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, South Korea; Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, South Korea.
| |
Collapse
|
2
|
Ma J, Li M, Bao Y, Huang W, He X, Hong Y, Wei W, Liu Z, Gao X, Yang Y, Cui Z, Wang W, Wang J, Zhu W, Zheng N, Pan L, Wang D, Ke Z, Zhou B, Sheng L, Li H. Gut microbiota-brain bile acid axis orchestrates aging-related neuroinflammation and behavior impairment in mice. Pharmacol Res 2024; 208:107361. [PMID: 39159729 DOI: 10.1016/j.phrs.2024.107361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/16/2024] [Accepted: 08/16/2024] [Indexed: 08/21/2024]
Abstract
Emerging evidence shows that disrupted gut microbiota-bile acid (BA) axis is critically involved in the development of neurodegenerative diseases. However, the alterations in spatial distribution of BAs among different brain regions that command important functions during aging and their exact roles in aging-related neurodegenerative diseases are poorly understood. Here, we analyzed the BA profiles in cerebral cortex, hippocampus, and hypothalamus of young and natural aging mice of both sexes. The results showed that aging altered brain BA profiles sex- and region- dependently, in which TβMCA was consistently elevated in aging mice of both sexes, particularly in the hippocampus and hypothalamus. Furthermore, we found that aging accumulated-TβMCA stimulated microglia inflammation in vitro and shortened the lifespan of C. elegans, as well as behavioral impairment and neuroinflammation in mice. In addition, metagenomic analysis suggested that the accumulation of brain TβMCA during aging was partially attributed to reduction in BSH-carrying bacteria. Finally, rejuvenation of gut microbiota by co-housing aged mice with young mice restored brain BA homeostasis and improved neurological dysfunctions in natural aging mice. In conclusion, our current study highlighted the potential of improving aging-related neuro-impairment by targeting gut microbiota-brain BA axis.
Collapse
Affiliation(s)
- Junli Ma
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Mingxiao Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yiyang Bao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wenjin Huang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiaofang He
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ying Hong
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Wenjing Wei
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zekun Liu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xinxin Gao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yang Yang
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhengyu Cui
- Department of Traditional Chinese Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Wantao Wang
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jie Wang
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, China
| | - Weize Zhu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ningning Zheng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Lingyun Pan
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Deheng Wang
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zunji Ke
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ben Zhou
- Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Lili Sheng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Houkai Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
3
|
Chen AM, Gajdošík M, Ahmed W, Ahn S, Babb JS, Blessing EM, Boutajangout A, de Leon MJ, Debure L, Gaggi N, Gajdošík M, George A, Ghuman M, Glodzik L, Harvey P, Juchem C, Marsh K, Peralta R, Rusinek H, Sheriff S, Vedvyas A, Wisniewski T, Zheng H, Osorio R, Kirov II. Retrospective analysis of Braak stage- and APOE4 allele-dependent associations between MR spectroscopy and markers of tau and neurodegeneration in cognitively unimpaired elderly. Neuroimage 2024; 297:120742. [PMID: 39029606 PMCID: PMC11404707 DOI: 10.1016/j.neuroimage.2024.120742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/28/2024] [Accepted: 07/16/2024] [Indexed: 07/21/2024] Open
Abstract
PURPOSE The pathological hallmarks of Alzheimer's disease (AD), amyloid, tau, and associated neurodegeneration, are present in the cortical gray matter (GM) years before symptom onset, and at significantly greater levels in carriers of the apolipoprotein E4 (APOE4) allele. Their respective biomarkers, A/T/N, have been found to correlate with aspects of brain biochemistry, measured with magnetic resonance spectroscopy (MRS), indicating a potential for MRS to augment the A/T/N framework for staging and prediction of AD. Unfortunately, the relationships between MRS and A/T/N biomarkers are unclear, largely due to a lack of studies examining them in the context of the spatial and temporal model of T/N progression. Advanced MRS acquisition and post-processing approaches have enabled us to address this knowledge gap and test the hypotheses, that glutamate-plus-glutamine (Glx) and N-acetyl-aspartate (NAA), metabolites reflecting synaptic and neuronal health, respectively, measured from regions on the Braak stage continuum, correlate with: (i) cerebrospinal fluid (CSF) p-tau181 level (T), and (ii) hippocampal volume or cortical thickness of parietal lobe GM (N). We hypothesized that these correlations will be moderated by Braak stage and APOE4 genotype. METHODS We conducted a retrospective imaging study of 34 cognitively unimpaired elderly individuals who received APOE4 genotyping and lumbar puncture from pre-existing prospective studies at the NYU Grossman School of Medicine between October 2014 and January 2019. Subjects returned for their imaging exam between April 2018 and February 2020. Metabolites were measured from the left hippocampus (Braak II) using a single-voxel semi-adiabatic localization by adiabatic selective refocusing sequence; and from the bilateral posterior cingulate cortex (PCC; Braak IV), bilateral precuneus (Braak V), and bilateral precentral gyrus (Braak VI) using a multi-voxel echo-planar spectroscopic imaging sequence. Pearson and Spearman correlations were used to examine the relationships between absolute levels of choline, creatine, myo-inositol, Glx, and NAA and CSF p-tau181, and between these metabolites and hippocampal volume or parietal cortical thicknesses. Covariates included age, sex, years of education, Fazekas score, and months between CSF collection and MRI exam. RESULTS There was a direct correlation between hippocampal Glx and CSF p-tau181 in APOE4 carriers (Pearson's r = 0.76, p = 0.02), but not after adjusting for covariates. In the entire cohort, there was a direct correlation between hippocampal NAA and hippocampal volume (Spearman's r = 0.55, p = 0.001), even after adjusting for age and Fazekas score (Spearman's r = 0.48, p = 0.006). This relationship was observed only in APOE4 carriers (Pearson's r = 0.66, p = 0.017), and was also retained after adjustment (Pearson's r = 0.76, p = 0.008; metabolite-by-carrier interaction p = 0.03). There were no findings in the PCC, nor in the negative control (late Braak stage) regions of the precuneus and precentral gyrus. CONCLUSIONS Our findings are in line with the spatially- and temporally-resolved Braak staging model of pathological severity in which the hippocampus is affected earlier than the PCC. The correlations, between MRS markers of synaptic and neuronal health and, respectively, T and N pathology, were found exclusively within APOE4 carriers, suggesting a connection with AD pathological change, rather than with normal aging. We therefore conclude that MRS has the potential to augment early A/T/N staging, with the hippocampus serving as a more sensitive MRS target compared to the PCC.
Collapse
Affiliation(s)
- Anna M Chen
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA; Center for Advanced Imaging Innovation and Research (CAI(2)R), Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA; Vilcek Institute of Graduate Biomedical Sciences, NYU Grossman School of Medicine, New York, NY, USA
| | - Martin Gajdošík
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA; Center for Advanced Imaging Innovation and Research (CAI(2)R), Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA; Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Wajiha Ahmed
- Center for Cognitive Neurology, Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
| | - Sinyeob Ahn
- Siemens Medical Solutions USA Inc., Malvern, PA, USA
| | - James S Babb
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA; Center for Advanced Imaging Innovation and Research (CAI(2)R), Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA
| | - Esther M Blessing
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY, USA; Healthy Brain Aging and Sleep Center, NYU Langone Health, New York, NY, USA
| | - Allal Boutajangout
- Center for Cognitive Neurology, Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA; Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York, NY, USA
| | - Mony J de Leon
- Retired Director, Center for Brain Health, Department of Psychiatry, NYU Grossman School of Medicine, New York, NY, USA; Brain Health Imaging Institute, Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Ludovic Debure
- Center for Cognitive Neurology, Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
| | - Naomi Gaggi
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY, USA; Healthy Brain Aging and Sleep Center, NYU Langone Health, New York, NY, USA
| | - Mia Gajdošík
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA; Center for Advanced Imaging Innovation and Research (CAI(2)R), Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA
| | - Ajax George
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA; Center for Advanced Imaging Innovation and Research (CAI(2)R), Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA
| | - Mobeena Ghuman
- Center for Cognitive Neurology, Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
| | - Lidia Glodzik
- Brain Health Imaging Institute, Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Patrick Harvey
- Brain Health Imaging Institute, Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Christoph Juchem
- Department of Biomedical Engineering, Columbia University, New York, NY, USA; Department of Radiology, Columbia University, New York, NY, USA
| | - Karyn Marsh
- Center for Cognitive Neurology, Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
| | - Rosemary Peralta
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA; Center for Advanced Imaging Innovation and Research (CAI(2)R), Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA
| | - Henry Rusinek
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA; Center for Advanced Imaging Innovation and Research (CAI(2)R), Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA
| | - Sulaiman Sheriff
- Department of Radiology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Alok Vedvyas
- Center for Cognitive Neurology, Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
| | - Thomas Wisniewski
- Center for Cognitive Neurology, Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA; Department of Psychiatry, NYU Grossman School of Medicine, New York, NY, USA; Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
| | - Helena Zheng
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA; Center for Advanced Imaging Innovation and Research (CAI(2)R), Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA
| | - Ricardo Osorio
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY, USA; Healthy Brain Aging and Sleep Center, NYU Langone Health, New York, NY, USA.
| | - Ivan I Kirov
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA; Center for Advanced Imaging Innovation and Research (CAI(2)R), Department of Radiology, NYU Grossman School of Medicine, New York, NY, USA; Vilcek Institute of Graduate Biomedical Sciences, NYU Grossman School of Medicine, New York, NY, USA; Center for Cognitive Neurology, Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA; Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
4
|
Tully J, Pereira AC, Sethi A, Griem J, Cross B, Williams SC, Blair RJ, Murphy D, Blackwood N. Impaired striatal glutamate/GABA regulation in violent offenders with antisocial personality disorder and psychopathy. Mol Psychiatry 2024; 29:1824-1832. [PMID: 38326560 PMCID: PMC11371654 DOI: 10.1038/s41380-024-02437-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 01/09/2024] [Accepted: 01/16/2024] [Indexed: 02/09/2024]
Abstract
Men with antisocial personality disorder (ASPD) with or without psychopathy (+/-P) are responsible for most violent crime in society. Development of effective treatments is hindered by poor understanding of the neurochemical underpinnings of the condition. Men with ASPD with and without psychopathy demonstrate impulsive decision-making, associated with striatal abnormalities in functional neuroimaging studies. However, to date, no study has directly examined the potential neurochemical underpinnings of such abnormalities. We therefore investigated striatal glutamate: GABA ratio using Magnetic Resonance Spectroscopy in 30 violent offenders (16 ASPD-P, 14 ASPD + P) and 21 healthy non-offenders. Men with ASPD +/- P had a significant reduction in striatal glutamate : GABA ratio compared to non-offenders. We report, for the first time, striatal Glutamate/GABA dysregulation in ASPD +/- P, and discuss how this may be related to core behavioral abnormalities in the disorders.
Collapse
Affiliation(s)
- John Tully
- Academic Unit of Mental Health and Clinical Neurosciences, School of Medicine, University of Nottingham, Jubilee Campus, University of Nottingham, Wollaton Rd, Lenton, Nottingham, NG8 1BB, United Kingdom.
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, 16 De Crespigny Park, London, SE5 8AF, United Kingdom.
| | - Andreia C Pereira
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, 16 De Crespigny Park, London, SE5 8AF, United Kingdom
| | - Arjun Sethi
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, 16 De Crespigny Park, London, SE5 8AF, United Kingdom
| | - Julia Griem
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, 16 De Crespigny Park, London, SE5 8AF, United Kingdom
| | - Ben Cross
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, 16 De Crespigny Park, London, SE5 8AF, United Kingdom
| | - Steve Cr Williams
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, 16 De Crespigny Park, London, SE58AF, United Kingdom
| | - Robert James Blair
- Child and Adolescent Mental Health Centre, Mental Health Services, Capital Region of Denmark, Copenhagen, Denmark
| | - Declan Murphy
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, 16 De Crespigny Park, London, SE5 8AF, United Kingdom
| | - Nigel Blackwood
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, 16 De Crespigny Park, London, SE5 8AF, United Kingdom
| |
Collapse
|
5
|
Niess F, Strasser B, Hingerl L, Bader V, Frese S, Clarke WT, Duguid A, Niess E, Motyka S, Krššák M, Trattnig S, Scherer T, Lanzenberger R, Bogner W. Whole-brain deuterium metabolic imaging via concentric ring trajectory readout enables assessment of regional variations in neuronal glucose metabolism. Hum Brain Mapp 2024; 45:e26686. [PMID: 38647048 PMCID: PMC11034002 DOI: 10.1002/hbm.26686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 03/13/2024] [Accepted: 04/04/2024] [Indexed: 04/25/2024] Open
Abstract
Deuterium metabolic imaging (DMI) is an emerging magnetic resonance technique, for non-invasive mapping of human brain glucose metabolism following oral or intravenous administration of deuterium-labeled glucose. Regional differences in glucose metabolism can be observed in various brain pathologies, such as Alzheimer's disease, cancer, epilepsy or schizophrenia, but the achievable spatial resolution of conventional phase-encoded DMI methods is limited due to prolonged acquisition times rendering submilliliter isotropic spatial resolution for dynamic whole brain DMI not feasible. The purpose of this study was to implement non-Cartesian spatial-spectral sampling schemes for whole-brain 2H FID-MR Spectroscopic Imaging to assess time-resolved metabolic maps with sufficient spatial resolution to reliably detect metabolic differences between healthy gray and white matter regions. Results were compared with lower-resolution DMI maps, conventionally acquired within the same session. Six healthy volunteers (4 m/2 f) were scanned for ~90 min after administration of 0.8 g/kg oral [6,6']-2H glucose. Time-resolved whole brain 2H FID-DMI maps of glucose (Glc) and glutamate + glutamine (Glx) were acquired with 0.75 and 2 mL isotropic spatial resolution using density-weighted concentric ring trajectory (CRT) and conventional phase encoding (PE) readout, respectively, at 7 T. To minimize the effect of decreased signal-to-noise ratios associated with smaller voxels, low-rank denoising of the spatiotemporal data was performed during reconstruction. Sixty-three minutes after oral tracer uptake three-dimensional (3D) CRT-DMI maps featured 19% higher (p = .006) deuterium-labeled Glc concentrations in GM (1.98 ± 0.43 mM) compared with WM (1.66 ± 0.36 mM) dominated regions, across all volunteers. Similarly, 48% higher (p = .01) 2H-Glx concentrations were observed in GM (2.21 ± 0.44 mM) compared with WM (1.49 ± 0.20 mM). Low-resolution PE-DMI maps acquired 70 min after tracer uptake featured smaller regional differences between GM- and WM-dominated areas for 2H-Glc concentrations with 2.00 ± 0.35 mM and 1.71 ± 0.31 mM, respectively (+16%; p = .045), while no regional differences were observed for 2H-Glx concentrations. In this study, we successfully implemented 3D FID-MRSI with fast CRT encoding for dynamic whole-brain DMI at 7 T with 2.5-fold increased spatial resolution compared with conventional whole-brain phase encoded (PE) DMI to visualize regional metabolic differences. The faster metabolic activity represented by 48% higher Glx concentrations was observed in GM- compared with WM-dominated regions, which could not be reproduced using whole-brain DMI with the low spatial resolution protocol. Improved assessment of regional pathologic alterations using a fully non-invasive imaging method is of high clinical relevance and could push DMI one step toward clinical applications.
Collapse
Affiliation(s)
- Fabian Niess
- High Field MR Center, Department of Biomedical Imaging and Image‐Guided TherapyMedical University of ViennaViennaAustria
| | - Bernhard Strasser
- High Field MR Center, Department of Biomedical Imaging and Image‐Guided TherapyMedical University of ViennaViennaAustria
| | - Lukas Hingerl
- High Field MR Center, Department of Biomedical Imaging and Image‐Guided TherapyMedical University of ViennaViennaAustria
| | - Viola Bader
- High Field MR Center, Department of Biomedical Imaging and Image‐Guided TherapyMedical University of ViennaViennaAustria
| | - Sabina Frese
- High Field MR Center, Department of Biomedical Imaging and Image‐Guided TherapyMedical University of ViennaViennaAustria
| | - William T. Clarke
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Anna Duguid
- High Field MR Center, Department of Biomedical Imaging and Image‐Guided TherapyMedical University of ViennaViennaAustria
| | - Eva Niess
- High Field MR Center, Department of Biomedical Imaging and Image‐Guided TherapyMedical University of ViennaViennaAustria
- Christian Doppler Laboratory for MR Imaging Biomarkers (BIOMAK), Department of Biomedical Imaging and Image‐guided TherapyMedical University of ViennaViennaAustria
| | - Stanislav Motyka
- High Field MR Center, Department of Biomedical Imaging and Image‐Guided TherapyMedical University of ViennaViennaAustria
- Christian Doppler Laboratory for MR Imaging Biomarkers (BIOMAK), Department of Biomedical Imaging and Image‐guided TherapyMedical University of ViennaViennaAustria
| | - Martin Krššák
- Department of Medicine III, Division of Endocrinology and MetabolismMedical University of ViennaViennaAustria
| | - Siegfried Trattnig
- High Field MR Center, Department of Biomedical Imaging and Image‐Guided TherapyMedical University of ViennaViennaAustria
- Institute for Clinical Molecular MRIKarl Landsteiner SocietySt. PöltenAustria
| | - Thomas Scherer
- Department of Medicine III, Division of Endocrinology and MetabolismMedical University of ViennaViennaAustria
| | - Rupert Lanzenberger
- Department of Psychiatry and Psychotherapy, Comprehensive Center for Clinical Neurosciences and Mental Health (C3NMH)Medical University of ViennaViennaAustria
| | - Wolfgang Bogner
- High Field MR Center, Department of Biomedical Imaging and Image‐Guided TherapyMedical University of ViennaViennaAustria
- Christian Doppler Laboratory for MR Imaging Biomarkers (BIOMAK), Department of Biomedical Imaging and Image‐guided TherapyMedical University of ViennaViennaAustria
| |
Collapse
|
6
|
Schreiner SJ, Van Bergen JMG, Gietl AF, Buck A, Hock C, Pruessmann KP, Henning A, Unschuld PG. Gray matter gamma-hydroxy-butyric acid and glutamate reflect beta-amyloid burden at old age. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e12587. [PMID: 38690510 PMCID: PMC11058481 DOI: 10.1002/dad2.12587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 03/05/2024] [Accepted: 03/10/2024] [Indexed: 05/02/2024]
Abstract
Gamma-hydroxy-butyric acid (GABA) and glutamate are neurotransmitters with essential importance for cognitive processing. Here, we investigate relationships between GABA, glutamate, and brain ß-amyloid (Aß) burden before clinical manifestation of Alzheimer's disease (AD). Thirty cognitively healthy adults (age 69.9 ± 6 years) received high-resolution atlas-based 1H-magnetic resonance spectroscopic imaging (MRSI) at ultra-high magnetic field strength of 7 Tesla for gray matter-specific assessment of GABA and glutamate. We assessed Aß burden with positron emission tomography and risk factors for AD. Higher gray matter GABA and glutamate related to higher Aß-burden (ß = 0.60, p < 0.05; ß = 0.64, p < 0.02), with positive effect modification by apolipoprotein-E-epsilon-4-allele (APOE4) (p = 0.01-0.03). GABA and glutamate negatively related to longitudinal change in verbal episodic memory performance (ß = -0.48; p = 0.02; ß = -0.50; p = 0.01). In vivo measures of GABA and glutamate reflect early AD pathology at old age, in an APOE4-dependent manner. GABA and glutamate may represent promising biomarkers and potential targets for early therapeutic intervention and prevention. Highlights Gray matter-specific metabolic imaging with high-resolution atlas-based MRSI at 7 Tesla.Higher GABA and glutamate relate to ß-amyloid burden, in an APOE4-dependent manner.Gray matter GABA and glutamate identify older adults with high risk of future AD.GABA and glutamate might reflect altered synaptic and neuronal activity at early AD.
Collapse
Affiliation(s)
- Simon J. Schreiner
- Institute for Regenerative MedicineUniversity of ZurichZurichSwitzerland
- Department of Psychogeriatric MedicinePsychiatric University Hospital Zurich (PUK)ZurichSwitzerland
- Department of NeurologyUniversity Hospital Zurich and University of ZurichZurichSwitzerland
| | | | - Anton F. Gietl
- Institute for Regenerative MedicineUniversity of ZurichZurichSwitzerland
- Department of Psychogeriatric MedicinePsychiatric University Hospital Zurich (PUK)ZurichSwitzerland
| | - Alfred Buck
- Department of Nuclear MedicineUniversity Hospital Zurich and University of ZurichZurichSwitzerland
| | - Christoph Hock
- Institute for Regenerative MedicineUniversity of ZurichZurichSwitzerland
- NeurimmuneSchlierenSwitzerland
| | - Klaas P. Pruessmann
- Institute for Biomedical EngineeringUniversity of Zurich and ETH ZurichZurichSwitzerland
| | - Anke Henning
- Institute for Biomedical EngineeringUniversity of Zurich and ETH ZurichZurichSwitzerland
- High‐Field MR CenterMax Planck Institute for Biological CyberneticsTübingenGermany
- Advanced Imaging Research CenterUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Paul G. Unschuld
- Geriatric Psychiatry ServiceUniversity Hospitals of Geneva (HUG)ThônexSwitzerland
- Department of PsychiatryUniversity of Geneva (UniGE)GenevaSwitzerland
| |
Collapse
|
7
|
Hristov M, Nankova A, Andreeva-Gateva P. Alterations of the glutamatergic system in diabetes mellitus. Metab Brain Dis 2024; 39:321-333. [PMID: 37747631 DOI: 10.1007/s11011-023-01299-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 09/17/2023] [Indexed: 09/26/2023]
Abstract
Diabetes mellitus (DM) is a chronic disease characterized by elevated blood glucose levels caused by a lack of insulin production (type 1 diabetes) or insulin resistance (type 2 diabetes). It is well known that DM is associated with cognitive deficits and metabolic and neurophysiological changes in the brain. Glutamate is the main excitatory neurotransmitter in the central nervous system that plays a key role in synaptic plasticity, learning, and memory processes. An increasing number of studies have suggested that abnormal activity of the glutamatergic system is implicated in the pathophysiology of DM. Dysfunction of glutamatergic neurotransmission in the central nervous system can provide an important neurobiological substrate for many disorders. Magnetic resonance spectroscopy (MRS) is a non-invasive technique that allows a better understanding of the central nervous system factors by measuring in vivo the concentrations of brain metabolites within the area of interest. Here, we briefly review the MRS studies that have examined glutamate levels in the brain of patients with DM. The present article also summarizes the available data on abnormalities in glutamatergic neurotransmission observed in different animal models of DM. In addition, the role of gut microbiota in the development of glutamatergic alterations in DM is addressed. We speculate that therapeutic strategies targeting the glutamatergic system may be beneficial in the treatment of central nervous system-related changes in diabetic patients.
Collapse
Affiliation(s)
- Milen Hristov
- Department of Pharmacology and Toxicology, Faculty of Medicine, Medical University of Sofia, 2 "Zdrave" St, Sofia, 1431, Bulgaria.
| | - Anelia Nankova
- Department of Endocrinology, Faculty of Medicine, Medical University of Sofia, Sofia, 1431, Bulgaria
| | - Pavlina Andreeva-Gateva
- Department of Pharmacology and Toxicology, Faculty of Medicine, Medical University of Sofia, 2 "Zdrave" St, Sofia, 1431, Bulgaria
| |
Collapse
|
8
|
Ernst T, Ryan MC, Liang HJ, Wang JP, Cunningham E, Saleh MG, Kottilil S, Chang L. Neuronal and Glial Metabolite Abnormalities in Participants With Persistent Neuropsychiatric Symptoms After COVID-19: A Brain Proton Magnetic Resonance Spectroscopy Study. J Infect Dis 2023; 228:1559-1570. [PMID: 37540098 PMCID: PMC10681871 DOI: 10.1093/infdis/jiad309] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 06/01/2023] [Accepted: 08/02/2023] [Indexed: 08/05/2023] Open
Abstract
BACKGROUND The aim of this study was to determine whether neurometabolite abnormalities indicating neuroinflammation and neuronal injury are detectable in individuals post-coronavirus disease 2019 (COVID-19) with persistent neuropsychiatric symptoms. METHODS All participants were studied with proton magnetic resonance spectroscopy at 3 T to assess neurometabolite concentrations (point-resolved spectroscopy, relaxation time/echo time = 3000/30 ms) in frontal white matter (FWM) and anterior cingulate cortex-gray matter (ACC-GM). Participants also completed the National Institutes of Health Toolbox cognition and motor batteries and selected modules from the Patient-Reported Outcomes Measurement Information System. RESULTS Fifty-four participants were evaluated: 29 post-COVID-19 (mean ± SD age, 42.4 ± 12.3 years; approximately 8 months from COVID-19 diagnosis; 19 women) and 25 controls (age, 44.1 ± 12.3 years; 14 women). When compared with controls, the post-COVID-19 group had lower total N-acetyl compounds (tNAA; ACC-GM: -5.0%, P = .015; FWM: -4.4%, P = .13), FWM glutamate + glutamine (-9.5%, P = .001), and ACC-GM myo-inositol (-6.2%, P = .024). Additionally, only hospitalized patients post-COVID-19 showed age-related increases in myo-inositol, choline compounds, and total creatine (interaction P = .029 to <.001). Across all participants, lower FWM tNAA and higher ACC-GM myo-inositol predicted poorer performance on several cognitive measures (P = .001-.009), while lower ACC-GM tNAA predicted lower endurance on the 2-minute walk (P = .005). CONCLUSIONS In participants post-COVID-19 with persistent neuropsychiatric symptoms, the lower-than-normal tNAA and glutamate + glutamine indicate neuronal injury, while the lower-than-normal myo-inositol reflects glial dysfunction, possibly related to mitochondrial dysfunction and oxidative stress in Post-COVID participants with persistent neuropsychiatric symptoms.
Collapse
Affiliation(s)
- Thomas Ernst
- Department of Diagnostic Radiology and Nuclear Medicine, School of Medicine, University of Maryland
- Department of Neurology, School of Medicine, Johns Hopkins University
| | - Meghann C Ryan
- Department of Diagnostic Radiology and Nuclear Medicine, School of Medicine, University of Maryland
- Program in Neuroscience, School of Medicine, University of Maryland
| | - Hua-Jun Liang
- Department of Diagnostic Radiology and Nuclear Medicine, School of Medicine, University of Maryland
| | - Justin P Wang
- Department of Diagnostic Radiology and Nuclear Medicine, School of Medicine, University of Maryland
| | - Eric Cunningham
- Department of Diagnostic Radiology and Nuclear Medicine, School of Medicine, University of Maryland
| | - Muhammad G Saleh
- Department of Diagnostic Radiology and Nuclear Medicine, School of Medicine, University of Maryland
| | - Shyamasundaran Kottilil
- Institute of Human Virology, Division of Infectious Disease, Department of Medicine, School of Medicine, University of Maryland
| | - Linda Chang
- Department of Diagnostic Radiology and Nuclear Medicine, School of Medicine, University of Maryland
- Department of Neurology, School of Medicine, Johns Hopkins University
- Department of Neurology, School of Medicine, University of Maryland, Baltimore
| |
Collapse
|
9
|
Wójcik J, Kochański B, Cieśla K, Lewandowska M, Karpiesz L, Niedziałek I, Raj-Koziak D, Skarżyński PH, Wolak T. An MR spectroscopy study of temporal areas excluding primary auditory cortex and frontal regions in subjective bilateral and unilateral tinnitus. Sci Rep 2023; 13:18417. [PMID: 37891242 PMCID: PMC10611771 DOI: 10.1038/s41598-023-45024-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 10/14/2023] [Indexed: 10/29/2023] Open
Abstract
Previous studies indicate changes in neurotransmission along the auditory pathway in subjective tinnitus. Most authors, however, investigated brain regions including the primary auditory cortex, whose physiology can be affected by concurrent hearing deficits. In the present MR spectroscopy study we assumed increased levels of glutamate and glutamine (Glx), and other Central Nervous System metabolites in the temporal lobe outside the primary auditory cortex, in a region involved in conscious auditory perception and memory. We studied 52 participants with unilateral (n = 24) and bilateral (n = 28) tinnitus, and a control group without tinnitus (n = 25), all with no severe hearing losses and a similar hearing profile. None of the metabolite levels in the temporal regions of interest were found related to tinnitus status or laterality. Unexpectedly, we found a tendency of increased concentration of Glx in the control left medial frontal region in bilateral vs unilateral tinnitus. Slightly elevated depressive and anxiety symptoms were also shown in participants with tinnitus, as compared to healthy individuals, with the bilateral tinnitus group marginally more affected. We discuss no apparent effect in the temporal lobes, as well as the role of frontal brain areas, with respect to hearing loss, attention and psychological well-being in chronic tinnitus. We furthermore elaborate on the design-related and technical obstacles of MR spectroscopy.
Collapse
Affiliation(s)
- Joanna Wójcik
- Bioimaging Research Center, World Hearing Center, Institute of Physiology and Pathology of Hearing, Mokra 17 Street, Kajetany, 05-830, Nadarzyn, Poland
| | - Bartosz Kochański
- Bioimaging Research Center, World Hearing Center, Institute of Physiology and Pathology of Hearing, Mokra 17 Street, Kajetany, 05-830, Nadarzyn, Poland
| | - Katarzyna Cieśla
- Bioimaging Research Center, World Hearing Center, Institute of Physiology and Pathology of Hearing, Mokra 17 Street, Kajetany, 05-830, Nadarzyn, Poland.
| | - Monika Lewandowska
- Faculty of Philosophy and Social Sciences, Institute of Psychology, Nicolaus Copernicus University, Fosa Staromiejska 1a Street, 87-100, Toruń, Poland
| | - Lucyna Karpiesz
- Tinnitus Department, World Hearing Center, Institute of Physiology and Pathology of Hearing, Mokra 17 Street, Kajetany, 05-830, Nadarzyn, Poland
| | - Iwona Niedziałek
- Tinnitus Department, World Hearing Center, Institute of Physiology and Pathology of Hearing, Mokra 17 Street, Kajetany, 05-830, Nadarzyn, Poland
| | - Danuta Raj-Koziak
- Tinnitus Department, World Hearing Center, Institute of Physiology and Pathology of Hearing, Mokra 17 Street, Kajetany, 05-830, Nadarzyn, Poland
| | - Piotr Henryk Skarżyński
- Department of Teleaudiology and Screening, World Hearing Center, Institute of Physiology and Pathology of Hearing, Mokra 17 Street, Kajetany, 05-830, Nadarzyn, Poland
- Institute of Sensory Organs, Mokra 1 Street, Kajetany, 05-830, Nadarzyn, Poland
- Heart Failure and Cardiac Rehabilitation Department, Faculty of Medicine, Medical University of Warsaw, Kondratowicza 8 Street, 03-242, Warsaw, Poland
| | - Tomasz Wolak
- Bioimaging Research Center, World Hearing Center, Institute of Physiology and Pathology of Hearing, Mokra 17 Street, Kajetany, 05-830, Nadarzyn, Poland
| |
Collapse
|
10
|
Klietz M, Mahmoudi N, Maudsley AA, Sheriff S, Bronzlik P, Almohammad M, Nösel P, Wegner F, Höglinger GU, Lanfermann H, Ding XQ. Whole-Brain Magnetic Resonance Spectroscopy Reveals Distinct Alterations in Neurometabolic Profile in Progressive Supranuclear Palsy. Mov Disord 2023; 38:1503-1514. [PMID: 37289057 DOI: 10.1002/mds.29456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 03/16/2023] [Accepted: 05/09/2023] [Indexed: 06/09/2023] Open
Abstract
BACKGROUND Progressive supranuclear palsy (PSP) is an atypical Parkinsonian syndrome characterized by supranuclear gaze palsy, early postural instability, and a frontal dysexecutive syndrome. Contrary to normal brain magnetic resonance imaging in Parkinson's disease (PD), PSP shows specific cerebral atrophy patterns and alterations, but these findings are not present in every patient, and it is still unclear if these signs are also detectable in early disease stages. OBJECTIVE The aim of the present study was to analyze the metabolic profile of patients with clinically diagnosed PSP in comparison with matched healthy volunteers and PD patients using whole-brain magnetic resonance spectroscopic imaging (wbMRSI). METHODS Thirty-nine healthy controls (HCs), 29 PD, and 22 PSP patients underwent wbMRSI. PSP and PD patients were matched for age and handedness with HCs. Clinical characterization was performed using the Movement Disorder Society Unified Parkinson's Disease Rating Scale, PSP rating scale, and DemTect (test for cognitive assessment). RESULTS In PSP patients a significant reduction in N-acetyl-aspartate (NAA) was detected in all brain lobes. Fractional volume of the cerebrospinal fluid significantly increased in PSP patients compared to PD and healthy volunteers. CONCLUSIONS In PSP much more neuronal degeneration and cerebral atrophy have been detected compared with PD. The most relevant alteration is the decrease in NAA in all lobes of the brain, which also showed a partial correlation with clinical symptoms. However, more studies are needed to confirm the additional value of wbMRSI in clinical practice. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Martin Klietz
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Nima Mahmoudi
- Department of Neuroradiology, Hannover Medical School, Hannover, Germany
| | - Andrew A Maudsley
- Department of Radiology, University of Miami School of Medicine, Miami, Florida, USA
| | - Sulaiman Sheriff
- Department of Radiology, University of Miami School of Medicine, Miami, Florida, USA
| | - Paul Bronzlik
- Department of Neuroradiology, Hannover Medical School, Hannover, Germany
| | | | - Patrick Nösel
- Department of Neuroradiology, Hannover Medical School, Hannover, Germany
| | - Florian Wegner
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | | | | | - Xiao-Qi Ding
- Department of Neuroradiology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
11
|
Trivedi AG, Ramesh KK, Huang V, Mellon EA, Barker PB, Kleinberg LR, Weinberg BD, Shu HKG, Shim H. Spectroscopic MRI-Based Biomarkers Predict Survival for Newly Diagnosed Glioblastoma in a Clinical Trial. Cancers (Basel) 2023; 15:3524. [PMID: 37444634 PMCID: PMC10340675 DOI: 10.3390/cancers15133524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/22/2023] [Accepted: 07/05/2023] [Indexed: 07/15/2023] Open
Abstract
Despite aggressive treatment, glioblastoma has a poor prognosis due to its infiltrative nature. Spectroscopic MRI-measured brain metabolites, particularly the choline to N-acetylaspartate ratio (Cho/NAA), better characterizes the extent of tumor infiltration. In a previous pilot trial (NCT03137888), brain regions with Cho/NAA ≥ 2x normal were treated with high-dose radiation for newly diagnosed glioblastoma patients. This report is a secondary analysis of that trial where spectroscopic MRI-based biomarkers are evaluated for how they correlate with progression-free and overall survival (PFS/OS). Subgroups were created within the cohort based on pre-radiation treatment (pre-RT) median cutoff volumes of residual enhancement (2.1 cc) and metabolically abnormal volumes used for treatment (19.2 cc). We generated Kaplan-Meier PFS/OS curves and compared these curves via the log-rank test between subgroups. For the subgroups stratified by metabolic abnormality, statistically significant differences were observed for PFS (p = 0.019) and OS (p = 0.020). Stratification by residual enhancement did not lead to observable differences in the OS (p = 0.373) or PFS (p = 0.286) curves. This retrospective analysis shows that patients with lower post-surgical Cho/NAA volumes had significantly superior survival outcomes, while residual enhancement, which guides high-dose radiation in standard treatment, had little significance in PFS/OS. This suggests that the infiltrating, non-enhancing component of glioblastoma is an important factor in patient outcomes and should be treated accordingly.
Collapse
Affiliation(s)
- Anuradha G. Trivedi
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Karthik K. Ramesh
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Vicki Huang
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Eric A. Mellon
- Department of Radiation Oncology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 45056, USA
| | - Peter B. Barker
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Lawrence R. Kleinberg
- Department of Radiation Oncology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Brent D. Weinberg
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Hui-Kuo G. Shu
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Hyunsuk Shim
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA 30332, USA
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
12
|
Trivedi AG, Kim SH, Ramesh KK, Giuffrida AS, Weinberg BD, Mellon EA, Kleinberg LR, Barker PB, Han H, Shu HKG, Shim H, Schreibmann E. Applying a Radiation Therapy Volume Analysis Pipeline to Determine the Utility of Spectroscopic MRI-Guided Adaptive Radiation Therapy for Glioblastoma. Tomography 2023; 9:1052-1061. [PMID: 37218946 PMCID: PMC10204497 DOI: 10.3390/tomography9030086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/12/2023] [Accepted: 05/19/2023] [Indexed: 05/24/2023] Open
Abstract
Accurate radiation therapy (RT) targeting is crucial for glioblastoma treatment but may be challenging using clinical imaging alone due to the infiltrative nature of glioblastomas. Precise targeting by whole-brain spectroscopic MRI, which maps tumor metabolites including choline (Cho) and N-acetylaspartate (NAA), can quantify early treatment-induced molecular changes that other traditional modalities cannot measure. We developed a pipeline to determine how spectroscopic MRI changes during early RT are associated with patient outcomes to provide insight into the utility of adaptive RT planning. Data were obtained from a study (NCT03137888) where glioblastoma patients received high-dose RT guided by the pre-RT Cho/NAA twice normal (Cho/NAA ≥ 2x) volume, and received spectroscopic MRI scans pre- and mid-RT. Overlap statistics between pre- and mid-RT scans were used to quantify metabolic activity changes after two weeks of RT. Log-rank tests were used to quantify the relationship between imaging metrics and patient overall and progression-free survival (OS/PFS). Patients with lower Jaccard/Dice coefficients had longer PFS (p = 0.045 for both), and patients with lower Jaccard/Dice coefficients had higher OS trending towards significance (p = 0.060 for both). Cho/NAA ≥ 2x volumes changed significantly during early RT, putting healthy tissue at risk of irradiation, and warranting further study into using adaptive RT planning.
Collapse
Affiliation(s)
- Anuradha G. Trivedi
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Su Hyun Kim
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Karthik K. Ramesh
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Alexander S. Giuffrida
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Brent D. Weinberg
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Eric A. Mellon
- Department of Radiation Oncology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 45056, USA
| | - Lawrence R. Kleinberg
- Department of Radiation Oncology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Peter B. Barker
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Hui Han
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Hui-Kuo G. Shu
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Hyunsuk Shim
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Eduard Schreibmann
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
13
|
La PL, Joyce JM, Bell TK, Mauthner M, Craig W, Doan Q, Beauchamp MH, Zemek R, Yeates KO, Harris AD. Brain metabolites measured with magnetic resonance spectroscopy in pediatric concussion and orthopedic injury: An Advancing Concussion Assessment in Pediatrics (A-CAP) study. Hum Brain Mapp 2023; 44:2493-2508. [PMID: 36763547 PMCID: PMC10028643 DOI: 10.1002/hbm.26226] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/18/2022] [Accepted: 01/25/2023] [Indexed: 02/11/2023] Open
Abstract
Millions of children sustain a concussion annually. Concussion disrupts cellular signaling and neural pathways within the brain but the resulting metabolic disruptions are not well characterized. Magnetic resonance spectroscopy (MRS) can examine key brain metabolites (e.g., N-acetyl Aspartate (tNAA), glutamate (Glx), creatine (tCr), choline (tCho), and myo-Inositol (mI)) to better understand these disruptions. In this study, we used MRS to examine differences in brain metabolites between children and adolescents with concussion versus orthopedic injury. Children and adolescents with concussion (n = 361) or orthopedic injury (OI) (n = 184) aged 8 to 17 years were recruited from five emergency departments across Canada. MRS data were collected from the left dorsolateral prefrontal cortex (L-DLPFC) using point resolved spectroscopy (PRESS) at 3 T at a mean of 12 days post-injury (median 10 days post-injury, range 2-33 days). Univariate analyses for each metabolite found no statistically significant metabolite differences between groups. Within each analysis, several covariates were statistically significant. Follow-up analyses designed to account for possible confounding factors including age, site, scanner, vendor, time since injury, and tissue type (and interactions as appropriate) did not find any metabolite group differences. In the largest sample of pediatric concussion studied with MRS to date, we found no metabolite differences between concussion and OI groups in the L-DLPFC. We suggest that at 2 weeks post-injury in a general pediatric concussion population, brain metabolites in the L-DLPFC are not specifically affected by brain injury.
Collapse
Affiliation(s)
- Parker L La
- Department of Radiology, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Julie M Joyce
- Department of Radiology, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Tiffany K Bell
- Department of Radiology, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Micaela Mauthner
- Department of Radiology, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - William Craig
- Department of Pediatrics, University of Alberta and Stollery Children's Hospital, Edmonton, Alberta, Canada
| | - Quynh Doan
- Department of Pediatrics, University of British Columbia and BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Miriam H Beauchamp
- Department of Psychology, University of Montreal and Ste Justine Hospital Research Center, Montreal, Quebec, Canada
| | - Roger Zemek
- Department of Pediatrics and Emergency Medicine, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, Ontario, Canada
- Childrens' Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Keith Owen Yeates
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Ashley D Harris
- Department of Radiology, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
14
|
Huang V, Rejimon A, Reddy K, Trivedi AG, Ramesh KK, Giuffrida AS, Muiruri R, Shim H, Eaton BR. Spectroscopic MRI-Guided Proton Therapy in Non-Enhancing Pediatric High-Grade Glioma. Tomography 2023; 9:633-646. [PMID: 36961010 PMCID: PMC10037577 DOI: 10.3390/tomography9020051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/24/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023] Open
Abstract
Radiation therapy (RT) is a critical part of definitive therapy for pediatric high-grade glioma (pHGG). RT is designed to treat residual tumor defined on conventional MRI (cMRI), though pHGG lesions may be ill-characterized on standard imaging. Spectroscopic MRI (sMRI) measures endogenous metabolite concentrations in the brain, and Choline (Cho)/N-acetylaspartate (NAA) ratio is a highly sensitive biomarker for metabolically active tumor. We provide a preliminary report of our study introducing a novel treatment approach of whole brain sMRI-guided proton therapy for pHGG. An observational cohort (c1 = 10 patients) receives standard of care RT; a therapeutic cohort (c2 = 15 patients) receives sMRI-guided proton RT. All patients undergo cMRI and sMRI, a high-resolution 3D whole-brain echo-planar spectroscopic imaging (EPSI) sequence (interpolated resolution of 12 µL) prior to RT and at several follow-up timepoints integrated into diagnostic scans. Treatment volumes are defined by cMRI for c1 and by cMRI and Cho/NAA ≥ 2x for c2. A longitudinal imaging database is used to quantify changes in lesion and metabolite volumes. Four subjects have been enrolled (c1 = 1/c2 = 3) with sMRI imaging follow-up of 4-18 months. Preliminary data suggest sMRI improves identification of pHGG infiltration based on abnormal metabolic activity, and using proton therapy to target sMRI-defined high-risk regions is safe and feasible.
Collapse
Affiliation(s)
- Vicki Huang
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Abinand Rejimon
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Kartik Reddy
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Radiology, Children’s Healthcare of Atlanta, Atlanta, GA 30342, USA
| | - Anuradha G. Trivedi
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Karthik K. Ramesh
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Alexander S. Giuffrida
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Robert Muiruri
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Hyunsuk Shim
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA 30332, USA
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Bree R. Eaton
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Radiology, Children’s Healthcare of Atlanta, Atlanta, GA 30342, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
15
|
Consistency of frontal cortex metabolites quantified by magnetic resonance spectroscopy within overlapping small and large voxels. Sci Rep 2023; 13:2246. [PMID: 36755048 PMCID: PMC9908968 DOI: 10.1038/s41598-023-29190-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 01/31/2023] [Indexed: 02/10/2023] Open
Abstract
Single voxel magnetic resonance spectroscopy (MRS) quantifies metabolites within a specified volume of interest. MRS voxels are constrained to rectangular prism shapes. Therefore, they must define a small voxel contained within the anatomy of interest or include not of interest neighbouring tissue. When studying cortical regions without clearly demarcated boundaries, e.g. the dorsolateral prefrontal cortex (DLPFC), it is unclear how representative a larger voxel is of a smaller volume within it. To determine if a large voxel is representative of a small voxel placed within it, this study quantified total N-Acetylaspartate (tNAA), choline, glutamate, Glx (glutamate and glutamine combined), myo-inositol, and creatine in two overlapping MRS voxels in the DLPFC, a large (30×30x30 mm) and small (15×15x15 mm) voxel. Signal-to-noise ratio (SNR) and tissue type factors were specifically investigated. With water-referencing, only myo-inositol was significantly correlated between the two voxels, while all metabolites showed significant correlations with creatine-referencing. SNR had a minimal effect on the correspondence between voxels, while tissue type showed substantial influence. This study demonstrates substantial variability of metabolite estimates within the DLPFC. It suggests that when small anatomical structures are of interest, it may be valuable to spend additional acquisition time to obtain specific, localized data.
Collapse
|
16
|
Chen AM, Gerhalter T, Dehkharghani S, Peralta R, Gajdošík M, Gajdošík M, Tordjman M, Zabludovsky J, Sheriff S, Ahn S, Babb JS, Bushnik T, Zarate A, Silver JM, Im BS, Wall SP, Madelin G, Kirov II. Replicability of proton MR spectroscopic imaging findings in mild traumatic brain injury: Implications for clinical applications. Neuroimage Clin 2023; 37:103325. [PMID: 36724732 PMCID: PMC9898311 DOI: 10.1016/j.nicl.2023.103325] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 11/06/2022] [Accepted: 01/16/2023] [Indexed: 01/20/2023]
Abstract
PURPOSE Proton magnetic resonance spectroscopy (1H MRS) offers biomarkers of metabolic damage after mild traumatic brain injury (mTBI), but a lack of replicability studies hampers clinical translation. In a conceptual replication study design, the results reported in four previous publications were used as the hypotheses (H1-H7), specifically: abnormalities in patients are diffuse (H1), confined to white matter (WM) (H2), comprise low N-acetyl-aspartate (NAA) levels and normal choline (Cho), creatine (Cr) and myo-inositol (mI) (H3), and correlate with clinical outcome (H4); additionally, a lack of findings in regional subcortical WM (H5) and deep gray matter (GM) structures (H6), except for higher mI in patients' putamen (H7). METHODS 26 mTBI patients (20 female, age 36.5 ± 12.5 [mean ± standard deviation] years), within two months from injury and 21 age-, sex-, and education-matched healthy controls were scanned at 3 Tesla with 3D echo-planar spectroscopic imaging. To test H1-H3, global analysis using linear regression was used to obtain metabolite levels of GM and WM in each brain lobe. For H4, patients were stratified into non-recovered and recovered subgroups using the Glasgow Outcome Scale Extended. To test H5-H7, regional analysis using spectral averaging estimated metabolite levels in four GM and six WM structures segmented from T1-weighted MRI. The Mann-Whitney U test and weighted least squares analysis of covariance were used to examine mean group differences in metabolite levels between all patients and all controls (H1-H3, H5-H7), and between recovered and non-recovered patients and their respectively matched controls (H4). Replicability was defined as the support or failure to support the null hypotheses in accordance with the content of H1-H7, and was further evaluated using percent differences, coefficients of variation, and effect size (Cohen's d). RESULTS Patients' occipital lobe WM Cho and Cr levels were 6.0% and 4.6% higher than controls', respectively (Cho, d = 0.37, p = 0.04; Cr, d = 0.63, p = 0.03). The same findings, i.e., higher patients' occipital lobe WM Cho and Cr (both p = 0.01), but with larger percent differences (Cho, 8.6%; Cr, 6.3%) and effect sizes (Cho, d = 0.52; Cr, d = 0.88) were found in the comparison of non-recovered patients to their matched controls. For the lobar WM Cho and Cr comparisons without statistical significance (frontal, parietal, temporal), unidirectional effect sizes were observed (Cho, d = 0.07 - 0.37; Cr, d = 0.27 - 0.63). No differences were found in any metabolite in any lobe in the comparison between recovered patients and their matched controls. In the regional analyses, no differences in metabolite levels were found in any GM or WM region, but all WM regions (posterior, frontal, corona radiata, and the genu, body, and splenium of the corpus callosum) exhibited unidirectional effect sizes for Cho and Cr (Cho, d = 0.03 - 0.34; Cr, d = 0.16 - 0.51). CONCLUSIONS We replicated findings of diffuse WM injury, which correlated with clinical outcome (supporting H1-H2, H4). These findings, however, were among the glial markers Cho and Cr, not the neuronal marker NAA (not supporting H3). No differences were found in regional GM and WM metabolite levels (supporting H5-H6), nor in putaminal mI (not supporting H7). Unidirectional effect sizes of higher patients' Cho and Cr within all WM analyses suggest widespread injury, and are in line with the conclusion from the previous publications, i.e., that detection of WM injury may be more dependent upon sensitivity of the 1H MRS technique than on the selection of specific regions. The findings lend further support to the corollary that clinic-ready 1H MRS biomarkers for mTBI may best be achieved by using high signal-to-noise-ratio single-voxels placed anywhere within WM. The biochemical signature of the injury, however, may differ and therefore absolute levels, rather than ratios may be preferred. Future replication efforts should further test the generalizability of these findings.
Collapse
Affiliation(s)
- Anna M Chen
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Teresa Gerhalter
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Seena Dehkharghani
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York, NY, USA; Department of Neurology, New York University Grossman School of Medicine, New York, NY, USA
| | - Rosemary Peralta
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Mia Gajdošík
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Martin Gajdošík
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Mickael Tordjman
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York, NY, USA; Department of Radiology, Hôpital Cochin, Paris, France
| | - Julia Zabludovsky
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Sulaiman Sheriff
- Department of Radiology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Sinyeob Ahn
- Siemens Medical Solutions USA Inc., Malvern, PA, USA
| | - James S Babb
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Tamara Bushnik
- Department of Rehabilitation Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Alejandro Zarate
- Department of Rehabilitation Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Jonathan M Silver
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA
| | - Brian S Im
- Department of Rehabilitation Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Stephen P Wall
- Ronald O. Perelman Department of Emergency Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Guillaume Madelin
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York, NY, USA
| | - Ivan I Kirov
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York, NY, USA; Department of Neurology, New York University Grossman School of Medicine, New York, NY, USA; Center for Advanced Imaging Innovation and Research, Department of Radiology, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
17
|
Harris AD, Amiri H, Bento M, Cohen R, Ching CRK, Cudalbu C, Dennis EL, Doose A, Ehrlich S, Kirov II, Mekle R, Oeltzschner G, Porges E, Souza R, Tam FI, Taylor B, Thompson PM, Quidé Y, Wilde EA, Williamson J, Lin AP, Bartnik-Olson B. Harmonization of multi-scanner in vivo magnetic resonance spectroscopy: ENIGMA consortium task group considerations. Front Neurol 2023; 13:1045678. [PMID: 36686533 PMCID: PMC9845632 DOI: 10.3389/fneur.2022.1045678] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 12/06/2022] [Indexed: 01/06/2023] Open
Abstract
Magnetic resonance spectroscopy is a powerful, non-invasive, quantitative imaging technique that allows for the measurement of brain metabolites that has demonstrated utility in diagnosing and characterizing a broad range of neurological diseases. Its impact, however, has been limited due to small sample sizes and methodological variability in addition to intrinsic limitations of the method itself such as its sensitivity to motion. The lack of standardization from a data acquisition and data processing perspective makes it difficult to pool multiple studies and/or conduct multisite studies that are necessary for supporting clinically relevant findings. Based on the experience of the ENIGMA MRS work group and a review of the literature, this manuscript provides an overview of the current state of MRS data harmonization. Key factors that need to be taken into consideration when conducting both retrospective and prospective studies are described. These include (1) MRS acquisition issues such as pulse sequence, RF and B0 calibrations, echo time, and SNR; (2) data processing issues such as pre-processing steps, modeling, and quantitation; and (3) biological factors such as voxel location, age, sex, and pathology. Various approaches to MRS data harmonization are then described including meta-analysis, mega-analysis, linear modeling, ComBat and artificial intelligence approaches. The goal is to provide both novice and experienced readers with the necessary knowledge for conducting MRS data harmonization studies.
Collapse
Affiliation(s)
- Ashley D. Harris
- Department of Radiology, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Houshang Amiri
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mariana Bento
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
| | - Ronald Cohen
- Department of Clinical and Health Psychology, College of Public Health and Health Professions, University of Florida, Gainesville, FL, United States
| | - Christopher R. K. Ching
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, Los Angeles, CA, United States
| | - Christina Cudalbu
- CIBM Center for Biomedical Imaging, Lausanne, Switzerland
- Animal Imaging and Technology, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Emily L. Dennis
- TBI and Concussion Center, Department of Neurology, University of Utah, Salt Lake City, UT, United States
| | - Arne Doose
- Division of Psychological and Social Medicine and Developmental Neurosciences, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Stefan Ehrlich
- Division of Psychological and Social Medicine and Developmental Neurosciences, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Ivan I. Kirov
- Department of Radiology, Center for Advanced Imaging Innovation and Research, New York University Grossman School of Medicine, New York, NY, United States
| | - Ralf Mekle
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Georg Oeltzschner
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Eric Porges
- Department of Clinical and Health Psychology, College of Public Health and Health Professions, University of Florida, Gainesville, FL, United States
| | - Roberto Souza
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Electrical and Software Engineering, Schulich School of Engineering, University of Calgary, Calgary, AB, Canada
| | - Friederike I. Tam
- Division of Psychological and Social Medicine and Developmental Neurosciences, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Brian Taylor
- Division of Diagnostic Imaging, Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Paul M. Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, Los Angeles, CA, United States
| | - Yann Quidé
- School of Psychology, University of New South Wales (UNSW), Sydney, NSW, Australia
| | - Elisabeth A. Wilde
- TBI and Concussion Center, Department of Neurology, University of Utah, Salt Lake City, UT, United States
| | - John Williamson
- Department of Clinical and Health Psychology, College of Public Health and Health Professions, University of Florida, Gainesville, FL, United States
| | - Alexander P. Lin
- Center for Clinical Spectroscopy, Department of Radiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Brenda Bartnik-Olson
- Department of Radiology, Loma Linda University Medical Center, Loma Linda, CA, United States
| |
Collapse
|
18
|
Joyce JM, La PL, Walker R, Harris A. Magnetic resonance spectroscopy of traumatic brain injury and subconcussive hits: A systematic review and meta-analysis. J Neurotrauma 2022; 39:1455-1476. [PMID: 35838132 DOI: 10.1089/neu.2022.0125] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Magnetic resonance spectroscopy (MRS) is a non-invasive technique used to study metabolites in the brain. MRS findings in traumatic brain injury (TBI) and subconcussive hit literature have been mixed. The most common observation is a decrease in N-acetyl-aspartate (NAA), traditionally considered a marker of neuronal integrity. Other metabolites, however, such as creatine (Cr), choline (Cho), glutamate+glutamine (Glx) and myo-inositol (mI) have shown inconsistent changes in these populations. The objective of this systematic review and meta-analysis was to synthesize MRS literature in head injury and explore factors (brain region, injury severity, time since injury, demographic, technical imaging factors, etc.) that may contribute to differential findings. One hundred and thirty-eight studies met inclusion criteria for the systematic review and of those, 62 NAA, 24 Cr, 49 Cho, 18 Glx and 21 mI studies met inclusion criteria for meta-analysis. A random effects model was used for meta-analyses with brain region as a subgroup for each of the five metabolites studied. Meta-regression was used to examine the influence of potential moderators including injury severity, time since injury, age, sex, tissue composition and methodological factors. In this analysis of 1428 unique head-injured subjects and 1132 controls, the corpus callosum was identified as a brain region highly susceptible to metabolite alteration. NAA was consistently decreased in TBI of all severity, but not in subconcussive hits. Cho and mI were found to be increased in moderate-to-severe TBI but not mild TBI. Glx and Cr were largely unaffected, however did show alterations in certain conditions.
Collapse
Affiliation(s)
- Julie Michele Joyce
- University of Calgary, 2129, Radiology, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, 157742, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, 157744, Calgary, Alberta, Canada.,Integrated Concussion Research Program, Calgary, Alberta, Canada;
| | - Parker L La
- University of Calgary, 2129, Radiology, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, 157742, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, 157744, Calgary, Alberta, Canada.,Integrated Concussion Research Program, Calgary, Alberta, Canada;
| | - Robyn Walker
- University of Calgary, 2129, Radiology, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, 157742, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, 157744, Calgary, Alberta, Canada.,Integrated Concussion Research Program, Calgary, Alberta, Canada;
| | - Ashley Harris
- University of Calgary, Radiology, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, 157742, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, 157744, Calgary, Alberta, Canada.,Integrated Concussion Research Program, Calgary, Alberta, Canada;
| |
Collapse
|
19
|
Li X, Abiko K, Sheriff S, Maudsley AA, Urushibata Y, Ahn S, Tha KK. The Distribution of Major Brain Metabolites in Normal Adults: Short Echo Time Whole-Brain MR Spectroscopic Imaging Findings. Metabolites 2022; 12:metabo12060543. [PMID: 35736476 PMCID: PMC9228869 DOI: 10.3390/metabo12060543] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 06/05/2022] [Accepted: 06/08/2022] [Indexed: 12/10/2022] Open
Abstract
This prospective study aimed to evaluate the variation in magnetic resonance spectroscopic imaging (MRSI)-observed brain metabolite concentrations according to anatomical location, sex, and age, and the relationships among regional metabolite distributions, using short echo time (TE) whole-brain MRSI (WB-MRSI). Thirty-eight healthy participants underwent short TE WB-MRSI. The major metabolite ratios, i.e., N-acetyl aspartate (NAA)/creatine (Cr), choline (Cho)/Cr, glutamate + glutamine (Glx)/Cr, and myoinositol (mI)/Cr, were calculated voxel-by-voxel. Their variations according to anatomical regions, sex, and age, and their relationship to each other were evaluated by using repeated-measures analysis of variance, t-tests, and Pearson’s product-moment correlation analyses. All four metabolite ratios exhibited widespread regional variation across the cerebral hemispheres (corrected p < 0.05). Laterality between the two sides and sex-related variation were also shown (p < 0.05). In several regions, NAA/Cr and Glx/Cr decreased and mI/Cr increased with age (corrected p < 0.05). There was a moderate positive correlation between NAA/Cr and mI/Cr in the insular lobe and thalamus and between Glx/Cr and mI/Cr in the parietal lobe (r ≥ 0.348, corrected p ≤ 0.025). These observations demand age- and sex- specific regional reference values in interpreting these metabolites, and they may facilitate the understanding of glial-neuronal interactions in maintaining homeostasis.
Collapse
Affiliation(s)
- Xinnan Li
- Laboratory for Biomarker Imaging Science, Hokkaido University Graduate School of Biomedical Science and Engineering, Sapporo 060-8638, Japan;
| | - Kagari Abiko
- Department of Rehabilitation, Hokkaido University Hospital, Sapporo 060-8648, Japan;
- Department of Rehabilitation, Sapporo Azabu Neurosurgical Hospital, Sapporo 065-0022, Japan
| | - Sulaiman Sheriff
- Department of Radiology, University of Miami School of Medicine, Miami, FL 33146, USA; (S.S.); (A.A.M.)
| | - Andrew A. Maudsley
- Department of Radiology, University of Miami School of Medicine, Miami, FL 33146, USA; (S.S.); (A.A.M.)
| | | | - Sinyeob Ahn
- Siemens Healthineers, San Francisco, CA 94553, USA;
| | - Khin Khin Tha
- Laboratory for Biomarker Imaging Science, Hokkaido University Graduate School of Biomedical Science and Engineering, Sapporo 060-8638, Japan;
- Global Center for Biomedical Science and Engineering, Hokkaido University Faculty of Medicine, Sapporo 060-8638, Japan
- Correspondence: ; Tel.: +81-11-706-8183
| |
Collapse
|
20
|
Yakovlev A, Manzhurtsev A, Menshchikov P, Ublinskiy M, Melnikov I, Kupriyanov D, Akhadov T, Semenova N. Functional Magnetic Resonance Spectroscopy Study of Total Glutamate and Glutamine in the Human Visual Cortex Activated by a Short Stimulus. Biophysics (Nagoya-shi) 2022. [DOI: 10.1134/s0006350922020245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
21
|
Ramesh K, Mellon EA, Gurbani SS, Weinberg BD, Schreibmann E, Sheriff SA, Goryawala M, de le Fuente M, Eaton BR, Zhong J, Voloschin AD, Sengupta S, Dunbar EM, Holdhoff M, Barker PB, Maudsley AA, Kleinberg LR, Shim H, Shu HKG. A multi-institutional pilot clinical trial of spectroscopic MRI-guided radiation dose escalation for newly diagnosed glioblastoma. Neurooncol Adv 2022; 4:vdac006. [PMID: 35382436 PMCID: PMC8976280 DOI: 10.1093/noajnl/vdac006] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Background Glioblastomas (GBMs) are aggressive brain tumors despite radiation therapy (RT) to 60 Gy and temozolomide (TMZ). Spectroscopic magnetic resonance imaging (sMRI), which measures levels of specific brain metabolites, can delineate regions at high risk for GBM recurrence not visualized on contrast-enhanced (CE) MRI. We conducted a clinical trial to assess the feasibility, safety, and efficacy of sMRI-guided RT dose escalation to 75 Gy for newly diagnosed GBMs. Methods Our pilot trial (NCT03137888) enrolled patients at 3 institutions (Emory University, University of Miami, Johns Hopkins University) from September 2017 to June 2019. For RT, standard tumor volumes based on T2-FLAIR and T1w-CE MRIs with margins were treated in 30 fractions to 50.1 and 60 Gy, respectively. An additional high-risk volume based on residual CE tumor and Cho/NAA (on sMRI) ≥2× normal was treated to 75 Gy. Survival curves were generated by the Kaplan-Meier method. Toxicities were assessed according to CTCAE v4.0. Results Thirty patients were treated in the study. The median age was 59 years. 30% were MGMT promoter hypermethylated; 7% harbored IDH1 mutation. With a median follow-up of 21.4 months for censored patients, median overall survival (OS) and progression-free survival were 23.0 and 16.6 months, respectively. This regimen appeared well-tolerated with 70% of grade 3 or greater toxicity ascribed to TMZ and 23% occurring at least 1 year after RT. Conclusion Dose-escalated RT to 75 Gy guided by sMRI appears feasible and safe for patients with newly diagnosed GBMs. OS outcome is promising and warrants additional testing. Based on these results, a randomized phase II trial is in development.
Collapse
Affiliation(s)
- Karthik Ramesh
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, Georgia, USA,Department of Biomedical Engineering, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Eric A Mellon
- Department of Radiation Oncology, University of Miami, Miami, Florida, USA
| | - Saumya S Gurbani
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, Georgia, USA,Department of Biomedical Engineering, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Brent D Weinberg
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia, USA,Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Eduard Schreibmann
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | | | | | - Bree R Eaton
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Jim Zhong
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Alfredo D Voloschin
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Soma Sengupta
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia, USA,Present affiliation: Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | | | - Matthias Holdhoff
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Peter B Barker
- Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Lawrence R Kleinberg
- Department of Radiation Oncology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Hyunsuk Shim
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, Georgia, USA,Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, Georgia, USA,Department of Biomedical Engineering, Emory University School of Medicine, Atlanta, Georgia, USA,Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, USA,Corresponding Authors: Hyunsuk Shim, PhD and Hui-Kuo G. Shu, MD, PhD, Department of Radiation Oncology, Winship Cancer Institute of Emory University, 1701 Uppergate Drive, Atlanta, GA 30322, USA (. )
| | - Hui-Kuo G Shu
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, Georgia, USA,Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
22
|
Klauser A, Klauser P, Grouiller F, Courvoisier S, Lazeyras F. Whole-brain high-resolution metabolite mapping with 3D compressed-sensing SENSE low-rank 1 H FID-MRSI. NMR IN BIOMEDICINE 2022; 35:e4615. [PMID: 34595791 PMCID: PMC9285075 DOI: 10.1002/nbm.4615] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 08/16/2021] [Accepted: 08/20/2021] [Indexed: 05/07/2023]
Abstract
There is a growing interest in the neuroscience community to map the distribution of brain metabolites in vivo. Magnetic resonance spectroscopic imaging (MRSI) is often limited by either a poor spatial resolution and/or a long acquisition time, which severely restricts its applications for clinical and research purposes. Building on a recently developed technique of acquisition-reconstruction for 2D MRSI, we combined a fast Cartesian 1 H-FID-MRSI acquisition sequence, compressed-sensing acceleration, and low-rank total-generalized-variation constrained reconstruction to produce 3D high-resolution whole-brain MRSI with a significant acquisition time reduction. We first evaluated the acceleration performance using retrospective undersampling of a fully sampled dataset. Second, a 20 min accelerated MRSI acquisition was performed on three healthy volunteers, resulting in metabolite maps with 5 mm isotropic resolution. The metabolite maps exhibited the detailed neurochemical composition of all brain regions and revealed parts of the underlying brain anatomy. The latter assessment used previous reported knowledge and a atlas-based analysis to show consistency of the concentration contrasts and ratio across all brain regions. These results acquired on a clinical 3 T MRI scanner successfully combined 3D 1 H-FID-MRSI with a constrained reconstruction to produce detailed mapping of metabolite concentrations at high resolution over the whole brain, with an acquisition time suitable for clinical or research settings.
Collapse
Affiliation(s)
- Antoine Klauser
- Department of Radiology and Medical InformaticsUniversity of GenevaSwitzerland
- Center for Biomedical Imaging (CIBM)GenevaSwitzerland
| | - Paul Klauser
- Center for Psychiatric Neuroscience, Department of PsychiatryLausanne University HospitalSwitzerland
- Service of Child and Adolescent Psychiatry, Department of PsychiatryLausanne University HospitalSwitzerland
| | - Frédéric Grouiller
- Swiss Center for Affective SciencesUniversity of GenevaSwitzerland
- Laboratory of Behavioral Neurology and Imaging of Cognition, Department of Fundamental NeuroscienceUniversity of GenevaSwitzerland
| | - Sébastien Courvoisier
- Department of Radiology and Medical InformaticsUniversity of GenevaSwitzerland
- Center for Biomedical Imaging (CIBM)GenevaSwitzerland
| | - François Lazeyras
- Department of Radiology and Medical InformaticsUniversity of GenevaSwitzerland
- Center for Biomedical Imaging (CIBM)GenevaSwitzerland
| |
Collapse
|
23
|
Neman J, Franklin M, Madaj Z, Deshpande K, Triche TJ, Sadlik G, Carmichael JD, Chang E, Yu C, Strickland BA, Zada G. Use of predictive spatial modeling to reveal that primary cancers have distinct central nervous system topography patterns of brain metastasis. J Neurosurg 2021; 136:88-96. [PMID: 34271545 DOI: 10.3171/2021.1.jns203536] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 01/22/2021] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Brain metastasis is the most common intracranial neoplasm. Although anatomical spatial distributions of brain metastasis may vary according to primary cancer subtype, these patterns are not understood and may have major implications for treatment. METHODS To test the hypothesis that the spatial distribution of brain metastasis varies according to cancer origin in nonrandom patterns, the authors leveraged spatial 3D coordinate data derived from stereotactic Gamma Knife radiosurgery procedures performed to treat 2106 brain metastases arising from 5 common cancer types (melanoma, lung, breast, renal, and colorectal). Two predictive topographic models (regional brain metastasis echelon model [RBMEM] and brain region susceptibility model [BRSM]) were developed and independently validated. RESULTS RBMEM assessed the hierarchical distribution of brain metastasis to specific brain regions relative to other primary cancers and showed that distinct regions were relatively susceptible to metastasis, as follows: bilateral temporal/parietal and left frontal lobes were susceptible to lung cancer; right frontal and occipital lobes to melanoma; cerebellum to breast cancer; and brainstem to renal cell carcinoma. BRSM provided probability estimates for each cancer subtype, independent of other subtypes, to metastasize to brain regions, as follows: lung cancer had a propensity to metastasize to bilateral temporal lobes; breast cancer to right cerebellar hemisphere; melanoma to left temporal lobe; renal cell carcinoma to brainstem; and colon cancer to right cerebellar hemisphere. Patient topographic data further revealed that brain metastasis demonstrated distinct spatial patterns when stratified by patient age and tumor volume. CONCLUSIONS These data support the hypothesis that there is a nonuniform spatial distribution of brain metastasis to preferential brain regions that varies according to cancer subtype in patients treated with Gamma Knife radiosurgery. These topographic patterns may be indicative of the abilities of various cancers to adapt to regional neural microenvironments, facilitate colonization, and establish metastasis. Although the brain microenvironment likely modulates selective seeding of metastasis, it remains unknown how the anatomical spatial distribution of brain metastasis varies according to primary cancer subtype and contributes to diagnosis. For the first time, the authors have presented two predictive models to show that brain metastasis, depending on its origin, in fact demonstrates distinct geographic spread within the central nervous system. These findings could be used as a predictive diagnostic tool and could also potentially result in future translational and therapeutic work to disrupt growth of brain metastasis on the basis of anatomical region.
Collapse
Affiliation(s)
- Josh Neman
- Departments of1Neurological Surgery.,2Physiology and Neuroscience.,3Norris Comprehensive Cancer Center.,6Keck School of Medicine, and.,8USC Brain Tumor Center, University of Southern California, Los Angeles, California; and
| | | | - Zachary Madaj
- 7Department of Bioinformatics, Van Andel Institute, Grand Rapids, Michigan
| | | | - Timothy J Triche
- 7Department of Bioinformatics, Van Andel Institute, Grand Rapids, Michigan
| | - Gal Sadlik
- Departments of1Neurological Surgery.,6Keck School of Medicine, and
| | - John D Carmichael
- Departments of1Neurological Surgery.,6Keck School of Medicine, and.,8USC Brain Tumor Center, University of Southern California, Los Angeles, California; and
| | - Eric Chang
- 3Norris Comprehensive Cancer Center.,5Radiation Oncology.,6Keck School of Medicine, and.,8USC Brain Tumor Center, University of Southern California, Los Angeles, California; and
| | - Cheng Yu
- Departments of1Neurological Surgery.,6Keck School of Medicine, and.,8USC Brain Tumor Center, University of Southern California, Los Angeles, California; and
| | - Ben A Strickland
- Departments of1Neurological Surgery.,6Keck School of Medicine, and
| | - Gabriel Zada
- Departments of1Neurological Surgery.,3Norris Comprehensive Cancer Center.,6Keck School of Medicine, and.,8USC Brain Tumor Center, University of Southern California, Los Angeles, California; and
| |
Collapse
|
24
|
Alterations of Striato-Thalamic Metabolism in Normal Aging Human Brain-An MR Metabolic Imaging Study. Metabolites 2021; 11:metabo11060371. [PMID: 34207758 PMCID: PMC8228538 DOI: 10.3390/metabo11060371] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 06/03/2021] [Accepted: 06/07/2021] [Indexed: 12/11/2022] Open
Abstract
Aging effects on striato-thalamic metabolism in healthy human brains were studied in vivo using short-TE whole brain 1H-MR spectroscopic imaging (wbMRSI) on eighty healthy subjects aged evenly between 20 to 70 years at 3T. Relative concentrations of N-acetyl-aspartate (NAA), choline, total creatine (tCr), myo-inositol (mI), glutamate, and glutamine in bilateral caudate nucleus, putamen, pallidum, and thalamus were determined using signal normalization relative to brain tissue water. Linear regression analysis was used to analyze the age-dependence of the metabolite concentrations. The metabolite concentrations revealed spatial inhomogeneity across brain regions and metabolites. With age, NAA decreased significantly in bilateral caudate nucleus and putamen, left pallidum, and left thalamus, tCr decreased in left putamen and bilateral pallidum, mI increased in bilateral caudate nucleus and right thalamus, and spectral linewidth increased in left putamen and right thalamus. In conclusion, normal aging of striato-thalamic metabolism in healthy human is associated with regional specific decreases of NAA and tCr and increases of mI, which may reflect the individual role of each brain structure within brain functionality.
Collapse
|
25
|
Ma RE, Murdoch JB, Bogner W, Andronesi O, Dydak U. Atlas-based GABA mapping with 3D MEGA-MRSI: Cross-correlation to single-voxel MRS. NMR IN BIOMEDICINE 2021; 34:e4275. [PMID: 32078755 PMCID: PMC7438238 DOI: 10.1002/nbm.4275] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 01/11/2020] [Accepted: 01/23/2020] [Indexed: 06/10/2023]
Abstract
The purpose of this work is to develop and validate a new atlas-based metabolite quantification pipeline for edited magnetic resonance spectroscopic imaging (MEGA-MRSI) that enables group comparisons of brain structure-specific GABA levels. By using brain structure masks segmented from high-resolution MPRAGE images and coregistering these to MEGA-LASER 3D MRSI data, an automated regional quantification of neurochemical levels is demonstrated for the example of the thalamus. Thalamic gamma-aminobutyric acid + coedited macromolecules (GABA+) levels from 21 healthy subjects scanned at 3 T were cross-validated both against a single-voxel MEGA-PRESS acquisition in the same subjects and same scan sessions, as well as alternative MRSI processing techniques (ROI approach, four-voxel approach) using Pearson correlation analysis. In addition, reproducibility was compared across the MRSI processing techniques in test-retest data from 14 subjects. The atlas-based approach showed a significant correlation with SV MEGA-PRESS (correlation coefficient r [GABA+] = 0.63, P < 0.0001). However, the actual values for GABA+, NAA, tCr, GABA+/tCr and tNAA/tCr obtained from the atlas-based approach showed an offset to SV MEGA-PRESS levels, likely due to the fact that on average the thalamus mask used for the atlas-based approach only occupied 30% of the SVS volume, ie, somewhat different anatomies were sampled. Furthermore, the new atlas-based approach showed highly reproducible GABA+/tCr values with a low median coefficient of variance of 6.3%. In conclusion, the atlas-based metabolite quantification approach enables a more brain structure-specific comparison of GABA+ and other neurochemical levels across populations, even when using an MRSI technique with only cm-level resolution. This approach was successfully cross-validated against the typically used SVS technique as well as other different MRSI analysis methods, indicating the robustness of this quantification approach.
Collapse
Affiliation(s)
- Ruoyun E. Ma
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota, Minneapolis, MN, USA
- School of Health Sciences, Purdue University, West Lafayette, IN, USA
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Wolfgang Bogner
- High Field MR Center, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Ovidiu Andronesi
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ulrike Dydak
- School of Health Sciences, Purdue University, West Lafayette, IN, USA
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
26
|
Maudsley AA, Andronesi OC, Barker PB, Bizzi A, Bogner W, Henning A, Nelson SJ, Posse S, Shungu DC, Soher BJ. Advanced magnetic resonance spectroscopic neuroimaging: Experts' consensus recommendations. NMR IN BIOMEDICINE 2021; 34:e4309. [PMID: 32350978 PMCID: PMC7606742 DOI: 10.1002/nbm.4309] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 02/01/2020] [Accepted: 03/10/2020] [Indexed: 05/04/2023]
Abstract
Magnetic resonance spectroscopic imaging (MRSI) offers considerable promise for monitoring metabolic alterations associated with disease or injury; however, to date, these methods have not had a significant impact on clinical care, and their use remains largely confined to the research community and a limited number of clinical sites. The MRSI methods currently implemented on clinical MRI instruments have remained essentially unchanged for two decades, with only incremental improvements in sequence implementation. During this time, a number of technological developments have taken place that have already greatly benefited the quality of MRSI measurements within the research community and which promise to bring advanced MRSI studies to the point where the technique becomes a true imaging modality, while making the traditional review of individual spectra a secondary requirement. Furthermore, the increasing use of biomedical MR spectroscopy studies has indicated clinical areas where advanced MRSI methods can provide valuable information for clinical care. In light of this rapidly changing technological environment and growing understanding of the value of MRSI studies for biomedical studies, this article presents a consensus from a group of experts in the field that reviews the state-of-the-art for clinical proton MRSI studies of the human brain, recommends minimal standards for further development of vendor-provided MRSI implementations, and identifies areas which need further technical development.
Collapse
Affiliation(s)
- Andrew A Maudsley
- Department of Radiology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Ovidiu C Andronesi
- Department of Radiology, Massachusetts General Hospital, Athinoula A. Martinos Center for Biomedical Imaging, Harvard Medical School, Boston, Massachusetts
| | - Peter B Barker
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, and the Kennedy Krieger Institute, F.M. Kirby Center for Functional Brain Imaging, Baltimore, Maryland
| | - Alberto Bizzi
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Wolfgang Bogner
- High Field MR Center, Department of Biomedical Imaging and Image-guided Therapy, Medical University Vienna, Vienna, Austria
| | - Anke Henning
- Max Planck Institute for Biological Cybernetics, Tübingen, Germany
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Sarah J Nelson
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Stefan Posse
- Department of Neurology, University of New Mexico, Albuquerque, New Mexico
| | - Dikoma C Shungu
- Department of Neuroradiology, Weill Cornell Medical College, New York, New York
| | - Brian J Soher
- Department of Radiology, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
27
|
Determinants of Schizophrenia Endophenotypes Based on Neuroimaging and Biochemical Parameters. Biomedicines 2021; 9:biomedicines9040372. [PMID: 33916324 PMCID: PMC8066217 DOI: 10.3390/biomedicines9040372] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/18/2021] [Accepted: 03/30/2021] [Indexed: 12/17/2022] Open
Abstract
Despite extensive research, there is no convincing evidence of a reliable diagnostic biomarker for schizophrenia beyond clinical observation. Disorders of glutamatergic neurotransmission associated with N-methyl-D-aspartate (NMDA) receptor insufficiency, neuroinflammation, and redox dysregulation are the principal common mechanism linking changes in the periphery with the brain, ultimately contributing to the emergence of negative symptoms of schizophrenia that underlie differential diagnosis. The aim of the study was to evaluate the influence of these systems via peripheral and cerebral biochemical indices in relation to the patient's clinical condition. Using neuroimaging diagnostics, we were able to define endophenotypes of schizophrenia based on objective laboratory data that form the basis of a personalized approach to diagnosis and treatment. The two distinguished endophenotypes differed in terms of the quality of life, specific schizophrenia symptoms, and glutamatergic neurotransmission metabolites in the anterior cingulate gyrus. Our results, as well as further studies of the excitatory or inhibitory balance of microcircuits, relating the redox systems on the periphery with the distant regions of the brain might allow for predicting potential biomarkers of neuropsychiatric diseases, including schizophrenia. To the best of our knowledge, our study is the first to identify an objective molecular biomarker of schizophrenia outcome.
Collapse
|
28
|
Abstract
RATIONALE Proton magnetic resonance spectroscopy (1H-MRS) is a cross-species neuroimaging technique that can measure concentrations of several brain metabolites, including glutamate and GABA. This non-invasive method has promise in developing centrally acting drugs, as it can be performed repeatedly within-subjects and be used to translate findings from the preclinical to clinical laboratory using the same imaging biomarker. OBJECTIVES This review focuses on the utility of single-voxel 1H-MRS in developing novel glutamatergic or GABAergic drugs for the treatment of psychiatric disorders and includes research performed in rodent models, healthy volunteers and patient cohorts. RESULTS Overall, these studies indicate that 1H-MRS is able to detect the predicted pharmacological effects of glutamatergic or GABAergic drugs on voxel glutamate or GABA concentrations, although there is a shortage of studies examining dose-related effects. Clinical studies have applied 1H-MRS to better understand drug therapeutic mechanisms, including the glutamatergic effects of ketamine in depression and of acamprosate in alcohol dependence. There is an emerging interest in identifying patient subgroups with 'high' or 'low' brain regional 1H-MRS glutamate levels for more targeted drug development, which may require ancillary biomarkers to improve the accuracy of subgroup discrimination. CONCLUSIONS Considerations for future research include the sensitivity of single-voxel 1H-MRS in detecting drug effects, inter-site measurement reliability and the interpretation of drug-induced changes in 1H-MRS metabolites relative to the known pharmacological molecular mechanisms. On-going technological development, in single-voxel 1H-MRS and in related complementary techniques, will further support applications within CNS drug discovery.
Collapse
Affiliation(s)
- Alice Egerton
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, UK.
| |
Collapse
|
29
|
Motegi T, Narita K, Fujihara K, Kasagi M, Suzuki Y, Tagawa M, Ujita K, Near J, Fukuda M. Glutamine + glutamate level predicts the magnitude of microstructural organization in the gray matter in the healthy elderly. Int Psychogeriatr 2021; 33:21-29. [PMID: 31578159 PMCID: PMC8482373 DOI: 10.1017/s1041610219001418] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 07/22/2019] [Accepted: 08/19/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Diffusion tensor imaging (DTI), which is a technique for measuring the degree and direction of movement of water molecules in tissue, has been widely used to noninvasively assess white matter (WM) or gray matter (GM) microstructures in vivo. Mean diffusivity (MD), which is the average diffusion across all directions, has been considered as a marker of WM tract degeneration or extracellular space enlargement in GM. Recent lines of evidence suggest that cortical MD can better identify early-stage Alzheimer's disease than structural morphometric parameters in magnetic resonance imaging. However, knowledge of the relationships between cortical MD and other biological factors in the same cortical region, e.g. metabolites, is still limited. METHODS Thirty-three healthy elderly individuals [aged 50-77 years (mean, 63.8±7.4 years); 11 males and 22 females] were enrolled. We estimated the associations between cortical MD and neurotransmitter levels. Specifically, we measured levels of γ-aminobutyric acid (GABA) and glutamate + glutamine (Glx), which are inhibitory and excitatory neurotransmitters, respectively, in medial prefrontal cortex (mPFC) and posterior cingulate cortex (PCC) using MEGA-PRESS magnetic resonance spectroscopy, and we measured regional cortical MD using DTI. RESULTS Cortical MD was significantly negatively associated with Glx levels in both mPFC and PCC. No significant association was observed between cortical MD and GABA levels in either GM region. CONCLUSION Our findings suggest that degeneration of microstructural organization in GM, as determined on the basis of cortical MD measured by DTI, is accompanied by the decline of Glx metabolism within the same GM region.
Collapse
Affiliation(s)
- Tomokazu Motegi
- Department of Psychiatry and Neuroscience, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Kosuke Narita
- Department of Psychiatry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Kazuyuki Fujihara
- Department of Psychiatry and Neuroscience, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Masato Kasagi
- Department of Psychiatry and Neuroscience, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Yusuke Suzuki
- Department of Psychiatry and Neuroscience, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Minami Tagawa
- Department of Psychiatry and Neuroscience, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Koichi Ujita
- Department of Diagnostic Radiology and Nuclear Medicine, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Jamie Near
- Douglas Mental Health University Institute and Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Masato Fukuda
- Department of Psychiatry and Neuroscience, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| |
Collapse
|
30
|
Bhogal AA, Broeders TAA, Morsinkhof L, Edens M, Nassirpour S, Chang P, Klomp DWJ, Vinkers CH, Wijnen JP. Lipid-suppressed and tissue-fraction corrected metabolic distributions in human central brain structures using 2D 1 H magnetic resonance spectroscopic imaging at 7 T. Brain Behav 2020; 10:e01852. [PMID: 33216472 PMCID: PMC7749561 DOI: 10.1002/brb3.1852] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 09/01/2020] [Accepted: 09/02/2020] [Indexed: 01/04/2023] Open
Abstract
INTRODUCTION Magnetic resonance spectroscopic imaging (MRSI) has the potential to add a layer of understanding of the neurobiological mechanisms underlying brain diseases, disease progression, and treatment efficacy. Limitations related to metabolite fitting of low signal-to-noise ratios data, signal variations due to partial-volume effects, acquisition and extracranial lipid artifacts, along with clinically relevant aspects such as scan time constraints, are among the challenges associated with in vivo MRSI. METHODS The aim of this work was to address some of these factors and to develop an acquisition, reconstruction, and postprocessing pipeline to derive lipid-suppressed metabolite values of central brain structures based on free-induction decay measurements made using a 7 T MR scanner. Anatomical images were used to perform high-resolution (1 mm3 ) partial-volume correction to account for gray matter, white matter (WM), and cerebral-spinal fluid signal contributions. Implementation of automatic quality control thresholds and normalization of metabolic maps from 23 subjects to the Montreal Neurological Institute (MNI) standard atlas facilitated the creation of high-resolution average metabolite maps of several clinically relevant metabolites in central brain regions, while accounting for macromolecular distributions. Partial-volume correction improved the delineation of deep brain nuclei. We report average metabolite values including glutamate + glutamine (Glx), glycerophosphocholine, choline and phosphocholine (tCho), (phospo)creatine, myo-inositol and glycine (mI-Gly), glutathione, N-acetyl-aspartyl glutamate(and glutamine), and N-acetyl-aspartate in the basal ganglia, central WM (thalamic radiation, corpus callosum) as well as insular cortex and intracalcarine sulcus. CONCLUSION MNI-registered average metabolite maps facilitate group-based analysis, thus offering the possibility to mitigate uncertainty in variable MRSI data.
Collapse
Affiliation(s)
- Alex A Bhogal
- Department of Radiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Tommy A A Broeders
- Department of Radiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lisan Morsinkhof
- Technical Medicine, University of Twente, Enchede, The Netherlands
| | - Mirte Edens
- Department of Radiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | - Dennis W J Klomp
- Department of Radiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Christiaan H Vinkers
- Department of Psychiatry, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Anatomy & Neurosciences, Amsterdam UMC (location VU University Medical Center), Amsterdam, The Netherlands.,Department of Psychiatry, Amsterdam UMC (location VU University Medical Center)/GGZ inGeest, Amsterdam, The Netherlands
| | - Jannie P Wijnen
- Department of Radiology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
31
|
Glutamatergic hypo-function in the left superior and middle temporal gyri in early schizophrenia: a data-driven three-dimensional proton spectroscopic imaging study. Neuropsychopharmacology 2020; 45:1851-1859. [PMID: 32403117 PMCID: PMC7608301 DOI: 10.1038/s41386-020-0707-y] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/23/2020] [Accepted: 05/06/2020] [Indexed: 12/26/2022]
Abstract
Proton magnetic resonance spectroscopy (1H-MRS) studies have examined glutamatergic abnormalities in schizophrenia, mostly in single voxels. Though the critical brain nodes remain unknown, schizophrenia involves networks with broad abnormalities. Hence, glutamine plus glutamate (Glx) and other metabolites were examined with whole-brain 1H-MRS, in early schizophrenia. Three dimensional 1H-MRS was acquired in young schizophrenia subjects (N = 36, 19 antipsychotic-naïve and 17 antipsychotic-treated) and healthy controls (HC, N = 29). Glx (as well as N-acetylaspartate, choline, myo-inositol and creatine) group contrasts from all individual voxels that met spectral quality, were analyzed in common brain space, followed by cluster-corrected level alpha-value (CCLAV ≤ 0.05). Schizophrenia subjects had lower Glx in the left superior (STG) and middle temporal gyri (16 voxels, CCLAV = 0.04) and increased creatine in two clusters involving left temporal, parietal and occipital regions (32, and 18 voxels, CCLAV = 0.02 and 0.04, respectively). Antipsychotic-treated and naïve patients (vs HC) had similar Glx reductions (8/16 vs 10/16 voxels respectively, but CCLAV's > 0.05). However, creatine was higher in antipsychotic-treated vs HC's in a larger left hemisphere cluster (100 voxels, CCLAV = 0.01). Also in treated patients, choline was increased in left middle frontal gyrus (18 voxels, CCLAV = 0.04). Finally in antipsychotic-naive patients, NAA was reduced in right frontal gyri (19 voxels, CCLAV = 0.05) and myo-inositol was reduced in the left cerebellum (34 voxels, CCLAV = 0.02). We conclude that data-driven spectroscopic brain examination supports that reductions in Glx in the left STG may be critical to the pathophysiology of schizophrenia. Postmortem and neuromodulation schizophrenia studies focusing on left STG, may provide critical mechanistic and therapeutic advancements, respectively.
Collapse
|
32
|
Kahl KG, Atalay S, Maudsley AA, Sheriff S, Cummings A, Frieling H, Schmitz B, Lanfermann H, Ding XQ. Altered neurometabolism in major depressive disorder: A whole brain 1H-magnetic resonance spectroscopic imaging study at 3T. Prog Neuropsychopharmacol Biol Psychiatry 2020; 101:109916. [PMID: 32169561 DOI: 10.1016/j.pnpbp.2020.109916] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 02/25/2020] [Accepted: 03/07/2020] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Major depressive disorder (MDD) is a severe mental disorder with a neurobiological basis that is poorly understood. Several studies demonstrated widespread, functional and neurometabolic alterations in MDD. However, little is known about whole brain neurometabolic alterations in MDD. METHOD Thirty-two patients with MDD and 32 paired on a one-to-one basis healthy controls (CTRL) underwent 1H-whole brain spectroscopic (1H-WBS) imaging. Lobar and cerebellar metabolite concentrations of brain N-acetylaspartate (NAA), total choline (tCho), total creatine (tCr), glutamine (Gln), glutamate (Glu), and myo-Inositol (mI) were assessed in patients and controls. RESULTS Decreased NAA, tCho, and tCr were found in the right frontal and right parietal lobe in MDD compared to CTRL, and to a lesser extent in the left frontal lobe. Furthermore, in MDD increased glutamine was observed in the right frontal lobe and bitemporal lobes, and increased glutamate in the cerebellum. CONCLUSION Altered global neurometabolism examined using 1H-WBS imaging in MDD may be interpreted as signs of neuronal dysfunction, altered energy metabolism, and oligodendrocyte dysfunction. In particular, the parallel decrease in NAA, tCr and tCho in the same brain regions may be indicative of neuronal dysfunction that may be counterbalanced by an increase of the neuroprotective metabolite glutamine. Future prospective investigations are warranted to study the functional importance of these findings.
Collapse
Affiliation(s)
- Kai G Kahl
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany.
| | - Sirin Atalay
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Andrew A Maudsley
- Department of Radiology, University of Miami School of Medicine, Miami, FL, USA
| | - Sulaiman Sheriff
- Department of Radiology, University of Miami School of Medicine, Miami, FL, USA
| | - Anna Cummings
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Helge Frieling
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Birte Schmitz
- Institute of Diagnostic and Interventional Neuroradiology, Hannover Medical School, Hannover, Germany
| | - Heinrich Lanfermann
- Institute of Diagnostic and Interventional Neuroradiology, Hannover Medical School, Hannover, Germany
| | - Xiao-Qi Ding
- Institute of Diagnostic and Interventional Neuroradiology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
33
|
Zhang Y, Taub E, Mueller C, Younger J, Uswatte G, DeRamus TP, Knight DC. Reproducibility of whole-brain temperature mapping and metabolite quantification using proton magnetic resonance spectroscopy. NMR IN BIOMEDICINE 2020; 33:e4313. [PMID: 32348017 DOI: 10.1002/nbm.4313] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 06/11/2023]
Abstract
Assessing brain temperature can provide important information about disease processes (e.g., stroke, trauma) and therapeutic effects (e.g., cerebral hypothermia treatment). Whole-brain magnetic resonance spectroscopic imaging (WB-MRSI) is increasingly used to quantify brain metabolites across the entire brain. However, its feasibility and reliability for estimating brain temperature needs further validation. Therefore, the present study evaluates the reproducibility of WB-MRSI for temperature mapping as well as metabolite quantification across the whole brain in healthy volunteers. Ten healthy adults were scanned on three occasions 1 week apart. Brain temperature, along with four commonly assessed brain metabolites-total N-acetyl-aspartate (tNAA), total creatine (tCr), total choline (tCho) and myo-inositol (mI)-were measured from WB-MRSI data. Reproducibility was evaluated using the coefficient of variation (CV). The measured mean (range) of the intra-subject CVs was 0.9% (0.6%-1.6%) for brain temperature mapping, and 4.7% (2.5%-15.7%), 6.4% (2.4%-18.9%) and 14.2% (4.4%-52.6%) for tNAA, tCho and mI, respectively, with reference to tCr. Consistently larger variability was found when using H2 O as the reference for metabolite quantifications: 7.8% (3.3%-17.8%), 7.8% (3.1%-18.0%), 9.8% (3.7%-31.0%) and 17.0% (5.9%-54.0%) for tNAA, tCr, tCho and mI, respectively. Further, the larger the brain region (indicated by a greater number of voxels within that region), the better the reproducibility for both temperature and metabolite estimates. Our results demonstrate good reproducibility of whole-brain temperature and metabolite measurements using the WB-MRSI technique.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Psychology, University of Alabama at Birmingham, Alabama, US
- Department of Neurosurgery and Core for Advanced MRI, Baylor College of Medicine, Houston, Texas, US
| | - Edward Taub
- Department of Psychology, University of Alabama at Birmingham, Alabama, US
| | - Christina Mueller
- Department of Psychology, University of Alabama at Birmingham, Alabama, US
| | - Jarred Younger
- Department of Psychology, University of Alabama at Birmingham, Alabama, US
| | - Gitendra Uswatte
- Department of Psychology, University of Alabama at Birmingham, Alabama, US
- Department of Physical Therapy, University of Alabama at Birmingham, Alabama, US
| | - Thomas Patrick DeRamus
- TReNDs Center for Translational Research in Neuroimaging and Data Science, Georgia, US
- Department of Psychology, Georgia State University, Georgia, US
| | - David C Knight
- Department of Psychology, University of Alabama at Birmingham, Alabama, US
| |
Collapse
|
34
|
Wu L, Niu Z, Hu X, Liu H, Li S, Chen L, Zheng D, Liu Z, Liu T, Xu F, Manyande A, Wang J, Xia H. Regional cerebral metabolic levels and turnover in awake rats after acute or chronic spinal cord injury. FASEB J 2020; 34:10547-10559. [PMID: 32592196 DOI: 10.1096/fj.202000447r] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 05/14/2020] [Accepted: 05/26/2020] [Indexed: 11/11/2022]
Affiliation(s)
- Liang Wu
- Department of Neurosurgery General Hospital of Ningxia Medical University Yinchuan P.R. China
- Key Laboratory of Magnetic Resonance in Biological Systems State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics Wuhan Institute of Physics and MathematicsChinese Academy of SciencesInnovation Academy for Precision Measurement Science and Technology Wuhan P.R. China
- Ningxia Key Laboratory of Cerebrocranial Diseases Yinchuan P.R. China
- School of Clinical Medicine Ningxia Medical University Yinchuan P.R. China
| | - Zhanfeng Niu
- Department of Neurosurgery General Hospital of Ningxia Medical University Yinchuan P.R. China
| | - Xulei Hu
- Department of Neurosurgery General Hospital of Ningxia Medical University Yinchuan P.R. China
- Ningxia Key Laboratory of Cerebrocranial Diseases Yinchuan P.R. China
- School of Clinical Medicine Ningxia Medical University Yinchuan P.R. China
| | - Huili Liu
- Key Laboratory of Magnetic Resonance in Biological Systems State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics Wuhan Institute of Physics and MathematicsChinese Academy of SciencesInnovation Academy for Precision Measurement Science and Technology Wuhan P.R. China
- University of Chinese Academy of Sciences Beijing P.R. China
| | - Shuang Li
- Key Laboratory of Magnetic Resonance in Biological Systems State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics Wuhan Institute of Physics and MathematicsChinese Academy of SciencesInnovation Academy for Precision Measurement Science and Technology Wuhan P.R. China
| | - Lei Chen
- Key Laboratory of Magnetic Resonance in Biological Systems State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics Wuhan Institute of Physics and MathematicsChinese Academy of SciencesInnovation Academy for Precision Measurement Science and Technology Wuhan P.R. China
| | - Danhao Zheng
- Key Laboratory of Magnetic Resonance in Biological Systems State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics Wuhan Institute of Physics and MathematicsChinese Academy of SciencesInnovation Academy for Precision Measurement Science and Technology Wuhan P.R. China
- University of Chinese Academy of Sciences Beijing P.R. China
| | - Zhuang Liu
- Key Laboratory of Magnetic Resonance in Biological Systems State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics Wuhan Institute of Physics and MathematicsChinese Academy of SciencesInnovation Academy for Precision Measurement Science and Technology Wuhan P.R. China
- University of Chinese Academy of Sciences Beijing P.R. China
| | - Taotao Liu
- Department of Anesthesiology Peking University Third Hospital Beijing P.R. China
| | - Fuqiang Xu
- Key Laboratory of Magnetic Resonance in Biological Systems State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics Wuhan Institute of Physics and MathematicsChinese Academy of SciencesInnovation Academy for Precision Measurement Science and Technology Wuhan P.R. China
- University of Chinese Academy of Sciences Beijing P.R. China
| | - Anne Manyande
- School of Human and Social Sciences University of West London London UK
| | - Jie Wang
- Key Laboratory of Magnetic Resonance in Biological Systems State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics Wuhan Institute of Physics and MathematicsChinese Academy of SciencesInnovation Academy for Precision Measurement Science and Technology Wuhan P.R. China
- University of Chinese Academy of Sciences Beijing P.R. China
- Hebei Provincial Key Laboratory of Basic Medicine for Diabetes 2nd Hospital of Shijiazhuang Shijiazhuang P.R. China
| | - Hechun Xia
- Department of Neurosurgery General Hospital of Ningxia Medical University Yinchuan P.R. China
- Ningxia Human Stem Cell Research Institute General Hospital of Ningxia Medical University Yinchuan P.R. China
| |
Collapse
|
35
|
Tapper S, Göransson N, Lundberg P, Tisell A, Zsigmond P. A pilot study of essential tremor: cerebellar GABA+/Glx ratio is correlated with tremor severity. CEREBELLUM & ATAXIAS 2020; 7:8. [PMID: 32607248 PMCID: PMC7318770 DOI: 10.1186/s40673-020-00116-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 05/29/2020] [Indexed: 12/16/2022]
Abstract
Objective Essential tremor is a common movement disorder with an unclear origin. Emerging evidence suggests the role of the cerebellum and the thalamus in tremor pathophysiology. We examined the two main neurotransmitters acting inhibitory (GABA+) and excitatory (Glx) respectively, in the thalamus and cerebellum, in patients diagnosed with severe essential tremor. Furthermore, we also investigated the relationship between determined neurotransmitter concentrations and tremor severity in the essential tremor patients. Methods Ten essential tremor patients (prior to deep brain stimulation surgery) and six healthy controls, were scanned using a 3 T MR system. GABA+ and Glx concentrations were measured using magnetic resonance spectroscopy (MRS) performed using single voxel MEGA-PRESS. For the purpose of assessing the tremor severity, the essential tremor rating scale (ETRS) was used in accordance with Fahn, Tolosa, and Marin. Results We demonstrated that the cerebellar GABA+/Glx ratio was positively correlated to the ETRS (r = 0.70, p = 0.03) in essential tremor. Cerebellar and thalamic GABA+ and Glx concentrations did not show any significant difference when comparing essential tremor patients with healthy controls, at the group level. Conclusion We demonstrated a positive correlation between increasing tremor disability and the ratio of GABA+/ Glx in the cerebellum of essential tremor patients. This highlights the impact of an altered balance of the excitatory and inhibitory neurotransmitters in tremor severity. Rather than a change in GABA+, which was constant, we attribute this finding to an overall decrease of Glx.
Collapse
Affiliation(s)
- Sofie Tapper
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden.,Department of Medical Radiation Physics and Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Nathanael Göransson
- Department of Biomedical Engineering, Linköping University, Linköping, Sweden
| | - Peter Lundberg
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden.,Department of Medical Radiation Physics and Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Anders Tisell
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden.,Department of Medical Radiation Physics and Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Peter Zsigmond
- Department of Neurosurgery and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
36
|
Menshchikov P, Ivantsova A, Manzhurtsev A, Ublinskiy M, Yakovlev A, Melnikov I, Kupriyanov D, Akhadov T, Semenova N. Separate N-acetyl aspartyl glutamate, N-acetyl aspartate, aspartate, and glutamate quantification after pediatric mild traumatic brain injury in the acute phase. Magn Reson Med 2020; 84:2918-2931. [PMID: 32544309 DOI: 10.1002/mrm.28332] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 04/27/2020] [Accepted: 05/01/2020] [Indexed: 12/19/2022]
Abstract
PURPOSE To separately measure N-acetyl aspartul glutamate (NAAG), N-acetyl aspartate (NAA), aspartate (Asp), and glutamate (Glu) concentrations in white matter (WM) using J-editing techniques in patients with mild traumatic brain injury (mTBI) in the acute phase. METHODS Twenty-four patients with closed concussive head injury and 29 healthy volunteers were enrolled in the current study. For extended 1 H MRS examination, patients and controls were equally divided into two subgroups. In subgroup 1 (12 patients/15 controls), NAAG and NAA concentrations were measured in WM separately with MEGA-PRESS (echo time/repetition time [TE/TR] = 140/2000 ms; δ ON NAA / δ OFF NAA = 4.84/4.38 ppm, δ ON NAAG / δ OFF NAAG = 4.61/4.15 ppm). In subgroup 2 (12 patients/14 controls), Asp and Glu concentrations were acquired with MEGA-PRESS (TE/TR = 90/2000 ms; δ ON Asp / δ OFF Asp = 3.89/5.21 ppm) and TE-averaged PRESS (TE from 35 ms to 185 ms with 2.5-ms increments; TR = 2000 ms) pulse sequences, respectively. RESULTS tNAA and NAAG concentrations were found to be reduced, while NAA concentrations were unchanged, after mild mTBI. Reduced Asp and elevated myo-inositol (mI) concentrations were also found. CONCLUSION The main finding of the study is that the tNAA signal reduction in WM after mTBI is associated with a decrease in the NAAG concentration rather than a decrease in the NAA concentration, as was thought previously. This finding highlights the importance of separating these signals, at least for WM studies, to avoid misinterpretation of the results. NAAG plays an important role in selectively activating mGluR3 receptors, thus providing neuroprotective and neuroreparative functions immediately after mTBI. NAAG shows potential for the development of new therapeutic strategies for patients with injuries of varying severity.
Collapse
Affiliation(s)
- Petr Menshchikov
- Semenov Institute of Chemical Physics, Russian Academy of Sciences, Moscow, Russian Federation.,Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, Russian Federation.,Clinical and Research Institute of Emergency Paediatric Surgery and Traumatology, Moscow, Russian Federation
| | - Anna Ivantsova
- Clinical and Research Institute of Emergency Paediatric Surgery and Traumatology, Moscow, Russian Federation
| | - Andrei Manzhurtsev
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, Russian Federation.,Clinical and Research Institute of Emergency Paediatric Surgery and Traumatology, Moscow, Russian Federation
| | - Maxim Ublinskiy
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, Russian Federation.,Clinical and Research Institute of Emergency Paediatric Surgery and Traumatology, Moscow, Russian Federation
| | - Alexey Yakovlev
- Semenov Institute of Chemical Physics, Russian Academy of Sciences, Moscow, Russian Federation.,Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, Russian Federation.,Clinical and Research Institute of Emergency Paediatric Surgery and Traumatology, Moscow, Russian Federation
| | - Ilya Melnikov
- Clinical and Research Institute of Emergency Paediatric Surgery and Traumatology, Moscow, Russian Federation
| | | | - Tolib Akhadov
- Clinical and Research Institute of Emergency Paediatric Surgery and Traumatology, Moscow, Russian Federation
| | - Natalia Semenova
- Semenov Institute of Chemical Physics, Russian Academy of Sciences, Moscow, Russian Federation.,Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, Russian Federation.,Clinical and Research Institute of Emergency Paediatric Surgery and Traumatology, Moscow, Russian Federation
| |
Collapse
|
37
|
Savic I. MRS Shows Regionally Increased Glutamate Levels among Patients with Exhaustion Syndrome Due to Occupational Stress. Cereb Cortex 2020; 30:3759-3770. [PMID: 32195540 DOI: 10.1093/cercor/bhz340] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 12/17/2019] [Indexed: 12/21/2022] Open
Abstract
Despite the rapid increase of reports of exhaustion syndrome (ES) due to daily occupational stress, the mechanisms underlying ES are unknown. We used voxel-based 1H-MR spectroscopy to examine the potential role of glutamate in this condition. The levels of glutamate were found to be elevated among ES patients (n = 30, 16 females) compared with controls (n = 31, 15 females). Notably, this increase was detected only in the anterior cingulate and mesial prefrontal cortex (ACC/mPFC), and the glutamate levels were linearly correlated with the degree of perceived stress. Furthermore, there was a sex by group interaction, as the glutamate elevation was present only in female patients. Female but not male ES patients also showed an increase in N-acetyl aspartate (NAA) levels in the amygdala. No group differences were detected in glutamine concentration (also measured). These data show the key role of glutamate in stress-related neuronal signaling and the specific roles of the amygdala and ACC/mPFC. The data extend previous reports about the neurochemical basis of stress and identify a potential neural marker and mediator of ES due to occupational stress. The observation of specific sex differences provides a tentative explanation to the well-known female predominance in stress-related psychopathology.
Collapse
Affiliation(s)
- Ivanka Savic
- Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, SE-171 76 Stockholm, Sweden.,Department of Neurology, Karolinska University Hospital, SE-171 76 Stockholm, Sweden.,Department of Neurology, UCLA, Los Angeles, CA 90095-1769, USA
| |
Collapse
|
38
|
Goryawala M, Sullivan M, Maudsley AA. Effects of apodization smoothing and denoising on spectral fitting. Magn Reson Imaging 2020; 70:108-114. [PMID: 32333950 DOI: 10.1016/j.mri.2020.04.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 03/13/2020] [Accepted: 04/19/2020] [Indexed: 11/20/2022]
Abstract
PURPOSE Visual review of individual spectra in magnetic resonance spectroscopic imaging (MRSI) data benefits from the application of spectral smoothing; however, if this processing step is applied prior to spectral analysis this can impact the accuracy of the quantitation. This study aims to analyze the effect of spectral denoising and apodization smoothing on the quantitation of whole-brain MRSI data obtained at short TE. METHODS Short-TE MRSI data obtained at 3 T were analyzed with no spectral smoothing, following (i) Gaussian apodization with values of 1, 2, 4, 6, and 8 Hz, and (ii) denoising using principal component analysis (dnPCA) with 3 different values for the number of retained principal components. The mean lobar white matter estimates for four metabolites, signal-to-noise ratio (SNR), spectral linewidth, and confidence intervals were compared to data reconstructed using no smoothing. Additionally, a voxel-wise comparison for N-acetylaspartate quantitation with different smoothing schemes was performed. RESULTS Significant pairwise differences were seen for all Gaussian smoothing methods as compared to no smoothing (p<0.001) in linewidth and metabolite estimates, whereas dnPCA methods showing no statistically significant differences in these measures. Confidence intervals decreased, and SNR increased with increasing levels of apodization smoothing or dnPCA denoising. CONCLUSION Mild Gaussian apodization (≤2 Hz at 3 T) can be applied with minimal (1%) errors in quantitation; however, smoothing values greater than that can significantly affect metabolite quantification. In contrast, mild to moderate dnPCA based denoising provides quantitative results that are consistent with the analysis of unsmoothed data and this method is recommended for spectral denoising.
Collapse
Affiliation(s)
| | - Molly Sullivan
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | | |
Collapse
|
39
|
Goryawala M, Saraf-Lavi E, Nagornaya N, Heros D, Komotar R, Maudsley AA. The Association between Whole-Brain MR Spectroscopy and IDH Mutation Status in Gliomas. J Neuroimaging 2019; 30:58-64. [PMID: 31868291 DOI: 10.1111/jon.12685] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 11/25/2019] [Accepted: 12/13/2019] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND PURPOSE Mutations in isocitrate dehydrogenase (IDH) have a direct effect on gliomagenesis. The purpose of this study is to quantify differences in brain metabolites due to IDH mutations. METHODS Magnetic Resonance Spectroscopic Imaging (MRSI) was performed in 35 patients with gliomas of different grade and varied IDH mutation status. Volumes of interest (VOIs) for active tumor (tVOI), peritumoral area (pVOI), and contralateral normal-appearing white matter (cVOI) were created. Metabolite ratios of Choline (Cho) to both N-acetylaspartate (NAA) and Creatine (Cr) were estimated. Ratios of Glutamate/Glutamine complex (Glx) and myoinositol (mIno) to Cr were also quantified. General linear models (GLMs) were used to estimate the effects of IDH mutation on metabolite measures, with age, gender, and tumor grade used as covariates. RESULTS GLM analysis showed that maximum Cho/NAA and Cho/Cr in the tVOI were significantly (P < .05) higher in IDH mutant lesions as compared to wild-type. In the pVOI, mean Cho/Cr was found to be significantly different among IDH mutant and wild-type gliomas. Mean Cho/NAA (P = .306) and Cho/Cr (P = .292) within the tVOI were not significantly different. Ratios of Glx/Cr and mIno/Cr in any region showed no significant differences between IDH mutant and wild-type gliomas. No significant differences in metabolite ratios were seen in the cVOI between IDH mutants and wild-types. CONCLUSION IDH mutation's effect in gliomas show an increase in Cho in the tumor and perilesional regions as compared to wild-type lesions but do not show widespread changes across the brain.
Collapse
Affiliation(s)
| | | | | | - Deborah Heros
- Department of Neurology, University of Miami, Miami, FL
| | - Ricardo Komotar
- Department of Neurological Surgery, University of Miami, Miami, FL
| | | |
Collapse
|
40
|
Cardiovascular risks impact human brain N-acetylaspartate in regionally specific patterns. Proc Natl Acad Sci U S A 2019; 116:25243-25249. [PMID: 31754041 DOI: 10.1073/pnas.1907730116] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Cardiovascular risk factors such as dyslipidemia and hypertension increase the risk for white matter pathology and cognitive decline. We hypothesize that white matter levels of N-acetylaspartate (NAA), a chemical involved in the metabolic pathway for myelin lipid synthesis, could serve as a biomarker that tracks the influence of cardiovascular risk factors on white matter prior to emergence of clinical changes. To test this, we measured levels of NAA across white matter and gray matter in the brain using echo planar spectroscopic imaging (EPSI) in 163 individuals and examined the relationship of regional NAA levels and cardiovascular risk factors as indexed by the Framingham Cardiovascular Risk Score (FCVRS). NAA was strongly and negatively correlated with FCVRS across the brain, but, after accounting for age and sex, the association was found primarily in white matter regions, with additional effects found in the thalamus, hippocampus, and cingulate gyrus. FCVRS was also negatively correlated with creatine levels, again primarily in white matter. The results suggest that cardiovascular risks are related to neurochemistry with a predominantly white matter pattern and some subcortical and cortical gray matter involvement. NAA mapping of the brain may provide early surveillance for the potential subclinical impact of cardiovascular and metabolic risk factors on the brain.
Collapse
|
41
|
Klietz M, Bronzlik P, Nösel P, Wegner F, Dressler DW, Dadak M, Maudsley AA, Sheriff S, Lanfermann H, Ding XQ. Altered Neurometabolic Profile in Early Parkinson's Disease: A Study With Short Echo-Time Whole Brain MR Spectroscopic Imaging. Front Neurol 2019; 10:777. [PMID: 31379726 PMCID: PMC6651356 DOI: 10.3389/fneur.2019.00777] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 07/03/2019] [Indexed: 12/11/2022] Open
Abstract
Objective: To estimate alterations in neurometabolic profile of patients with early stage Parkinson's disease (PD) by using a short echo-time whole brain magnetic resonance spectroscopic imaging (wbMRSI) as possible biomarker for early diagnosis and monitoring of PD. Methods: 20 PD patients in early stage (H&Y ≤ 2) without evidence of severe other diseases and 20 age and sex matched healthy controls underwent wbMRSI. In each subject brain regional concentrations of metabolites N-acetyl-aspartate (NAA), choline (Cho), total creatine (tCr), glutamine (Gln), glutamate (Glu), and myo-inositol (mIns) were obtained in atlas-defined lobar structures including subcortical basal ganglia structures (the left and right frontal lobes, temporal lobes, parietal lobes, occipital lobes, and the cerebellum) and compared between patients and matched healthy controls. Clinical characteristics of the PD patients were correlated with spectroscopic findings. Results: In comparison to controls the PD patients revealed altered lobar metabolite levels in all brain lobes contralateral to dominantly affected body side, i.e., decreases of temporal NAA, Cho, and tCr, parietal NAA and tCr, and frontal as well as occipital NAA. The frontal NAA correlated negatively with the MDS-UPDRS II (R = 22120.585, p = 0.008), MDS-UPDRS IV (R = −0.458, p = 0.048) and total MDS-UPDRS scores (R = −0.679, p = 0.001). Conclusion: In early PD stages metabolic alterations are evident in all contralateral brain lobes demonstrating that the neurodegenerative process affects not only local areas by dopaminergic denervation, but also the functional network within different brain regions. The wbMRSI-detectable brain metabolic alterations reveal the potential to serve as biomarkers for early PD.
Collapse
Affiliation(s)
- Martin Klietz
- Department of Neurology, Hannover Medical School, Hanover, Germany
| | - Paul Bronzlik
- Department of Neuroradiology, Hannover Medical School, Hanover, Germany
| | - Patrick Nösel
- Department of Neuroradiology, Hannover Medical School, Hanover, Germany
| | - Florian Wegner
- Department of Neurology, Hannover Medical School, Hanover, Germany
| | - Dirk W Dressler
- Department of Neurology, Hannover Medical School, Hanover, Germany
| | - Mete Dadak
- Department of Neuroradiology, Hannover Medical School, Hanover, Germany
| | - Andrew A Maudsley
- Department of Radiology, University of Miami School of Medicine, Miami, FL, United States
| | - Sulaiman Sheriff
- Department of Radiology, University of Miami School of Medicine, Miami, FL, United States
| | | | - Xiao-Qi Ding
- Department of Neuroradiology, Hannover Medical School, Hanover, Germany
| |
Collapse
|
42
|
Quevenco FC, Schreiner SJ, Preti MG, van Bergen JMG, Kirchner T, Wyss M, Steininger SC, Gietl A, Leh SE, Buck A, Pruessmann KP, Hock C, Nitsch RM, Henning A, Van De Ville D, Unschuld PG. GABA and glutamate moderate beta-amyloid related functional connectivity in cognitively unimpaired old-aged adults. NEUROIMAGE-CLINICAL 2019; 22:101776. [PMID: 30927605 PMCID: PMC6439267 DOI: 10.1016/j.nicl.2019.101776] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 02/03/2019] [Accepted: 03/10/2019] [Indexed: 02/08/2023]
Abstract
Background Effects of beta-amyloid accumulation on neuronal function precede the clinical manifestation of Alzheimer's disease (AD) by years and affect distinct cognitive brain networks. As previous studies suggest a link between beta-amyloid and dysregulation of excitatory and inhibitory neurotransmitters, we aimed to investigate the impact of GABA and glutamate on beta-amyloid related functional connectivity. Methods 29 cognitively unimpaired old-aged adults (age = 70.03 ± 5.77 years) were administered 11C-Pittsburgh Compound B (PiB) positron-emission tomography (PET), and MRI at 7 Tesla (7T) including blood oxygen level dependent (BOLD) functional MRI (fMRI) at rest for measuring static and dynamic functional connectivity. An advanced 7T MR spectroscopic imaging (MRSI) sequence based on the free induction decay acquisition localized by outer volume suppression’ (FIDLOVS) technology was used for gray matter specific measures of GABA and glutamate in the posterior cingulate and precuneus (PCP) region. Results GABA and glutamate MR-spectra indicated significantly higher levels in gray matter than in white matter. A global effect of beta-amyloid on functional connectivity in the frontal, occipital and inferior temporal lobes was observable. Interactive effects of beta-amyloid with gray matter GABA displayed positive PCP connectivity to the frontomedial regions, and the interaction of beta-amyloid with gray matter glutamate indicated positive PCP connectivity to frontal and cerebellar regions. Furthermore, decreased whole-brain but increased fronto-occipital and temporo-parietal dynamic connectivity was found, when GABA interacted with regional beta-amyloid deposits in the amygdala, frontal lobe, hippocampus, insula and striatum. Conclusions GABA, and less so glutamate, may moderate beta-amyloid related functional connectivity. Additional research is needed to better characterize their interaction and potential impact on AD. Combined ultra-high fieldstrength FIDLOVS MRSI at 7 Tesla with 11C-PIB PET. Assessment of gray matter specific levels of GABA and glutamate. Identification of interactive effects of GABA, glutamate and beta-Amyloid. GABA may moderate dysfunctional beta-Amyloid effects on pre-clinical brain pathology.
Collapse
Affiliation(s)
- F C Quevenco
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - S J Schreiner
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland; Hospital for Psychogeriatric Medicine, Psychiatric University Hospital Zurich (PUK), Zurich, Switzerland
| | - M G Preti
- Department of Radiology and Medical Informatics, Université de Genève, Switzerland; Institute of Bioengineering, École polytechnique fédérale de Lausanne, Switzerland
| | - J M G van Bergen
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - T Kirchner
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - M Wyss
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - S C Steininger
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland; Hospital for Psychogeriatric Medicine, Psychiatric University Hospital Zurich (PUK), Zurich, Switzerland
| | - A Gietl
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - S E Leh
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland; Hospital for Psychogeriatric Medicine, Psychiatric University Hospital Zurich (PUK), Zurich, Switzerland
| | - A Buck
- Division of Nuclear Medicine, University Hospital Zurich (USZ), Zurich, Switzerland
| | - K P Pruessmann
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland; Neuroscience Center Zurich (ZNZ), Zurich, Switzerland
| | - C Hock
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland; Hospital for Psychogeriatric Medicine, Psychiatric University Hospital Zurich (PUK), Zurich, Switzerland; Neuroscience Center Zurich (ZNZ), Zurich, Switzerland
| | - R M Nitsch
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland; Hospital for Psychogeriatric Medicine, Psychiatric University Hospital Zurich (PUK), Zurich, Switzerland; Neuroscience Center Zurich (ZNZ), Zurich, Switzerland
| | - A Henning
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland; Max Planck Institute for Biological Cybernetics, Tubingen, Germany
| | - D Van De Ville
- Department of Radiology and Medical Informatics, Université de Genève, Switzerland; Institute of Bioengineering, École polytechnique fédérale de Lausanne, Switzerland
| | - P G Unschuld
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland; Hospital for Psychogeriatric Medicine, Psychiatric University Hospital Zurich (PUK), Zurich, Switzerland; Neuroscience Center Zurich (ZNZ), Zurich, Switzerland.
| |
Collapse
|
43
|
Spurny B, Heckova E, Seiger R, Moser P, Klöbl M, Vanicek T, Spies M, Bogner W, Lanzenberger R. Automated ROI-Based Labeling for Multi-Voxel Magnetic Resonance Spectroscopy Data Using FreeSurfer. Front Mol Neurosci 2019; 12:28. [PMID: 30837839 PMCID: PMC6382749 DOI: 10.3389/fnmol.2019.00028] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 01/22/2019] [Indexed: 12/14/2022] Open
Abstract
Purpose: Advanced analysis methods for multi-voxel magnetic resonance spectroscopy (MRS) are crucial for neurotransmitter quantification, especially for neurotransmitters showing different distributions across tissue types. So far, only a handful of studies have used region of interest (ROI)-based labeling approaches for multi-voxel MRS data. Hence, this study aims to provide an automated ROI-based labeling tool for 3D-multi-voxel MRS data. Methods: MRS data, for automated ROI-based labeling, was acquired in two different spatial resolutions using a spiral-encoded, LASER-localized 3D-MRS imaging sequence with and without MEGA-editing. To calculate the mean metabolite distribution within selected ROIs, masks of individual brain regions were extracted from structural T1-weighted images using FreeSurfer. For reliability testing of automated labeling a comparison to manual labeling and single voxel selection approaches was performed for six different subcortical regions. Results: Automated ROI-based labeling showed high consistency [intra-class correlation coefficient (ICC) > 0.8] for all regions compared to manual labeling. Higher variation was shown when selected voxels, chosen from a multi-voxel grid, uncorrected for voxel composition, were compared to labeling methods using spatial averaging based on anatomical features within gray matter (GM) volumes. Conclusion: We provide an automated ROI-based analysis approach for various types of 3D-multi-voxel MRS data, which dramatically reduces hands-on time compared to manual labeling without any possible inter-rater bias.
Collapse
Affiliation(s)
- Benjamin Spurny
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Eva Heckova
- Department of Biomedical Imaging and Image-Guided Therapy, High Field MR Centre, Medical University of Vienna, Vienna, Austria
| | - Rene Seiger
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Philipp Moser
- Department of Biomedical Imaging and Image-Guided Therapy, High Field MR Centre, Medical University of Vienna, Vienna, Austria
| | - Manfred Klöbl
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Thomas Vanicek
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Marie Spies
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Bogner
- Department of Biomedical Imaging and Image-Guided Therapy, High Field MR Centre, Medical University of Vienna, Vienna, Austria
| | - Rupert Lanzenberger
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
44
|
Regional Metabolite Concentrations in Aging Human Brain: Comparison of Short-TE Whole Brain MR Spectroscopic Imaging and Single Voxel Spectroscopy at 3T. Clin Neuroradiol 2019; 30:251-261. [PMID: 30659340 DOI: 10.1007/s00062-018-00757-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 12/31/2018] [Indexed: 10/27/2022]
Abstract
PURPOSE The aim of this study was to compare a recently established whole brain MR spectroscopic imaging (wbMRSI) technique using spin-echo planar spectroscopic imaging (EPSI) acquisition and the Metabolic Imaging and Data Analysis System (MIDAS) software package with single voxel spectroscopy (SVS) technique and LCModel analysis for determination of relative metabolite concentrations in aging human brain. METHODS A total of 59 healthy subjects aged 20-70 years (n ≥ 5 per age decade for each gender) underwent a wbEPSI scan and 3 SVS scans of a 4 ml voxel volume located in the right basal ganglia, occipital grey matter and parietal white matter. Concentration ratios to total creatine (tCr) for N‑acetylaspartate (NAA/tCr), total choline (tCho/tCr), glutamine (Gln/tCr), glutamate (Glu/tCr) and myoinositol (mI/tCr) were obtained both from EPSI and SVS acquisitions with either LCModel or MIDAS. In addition, an aqueous phantom containing known metabolite concentrations was also measured. RESULTS Metabolite concentrations obtained with wbMRSI and SVS were comparable and consistent with those reported previously. Decreases of NAA/tCr and increases of line width with age were found with both techniques, while the results obtained from EPSI acquisition revealed generally narrower line widths and smaller Cramer-Rao lower bounds than those from SVS data. CONCLUSION The wbMRSI could be used to estimate metabolites in vivo and in vitro with the same reliability as using SVS, with the main advantage being the ability to determine metabolite concentrations in multiple brain structure simultaneously in vivo. It is expected to be widely used in clinical diagnostics and neuroscience.
Collapse
|
45
|
Oeltzschner G, Saleh MG, Rimbault D, Mikkelsen M, Chan KL, Puts NAJ, Edden RAE. Advanced Hadamard-encoded editing of seven low-concentration brain metabolites: Principles of HERCULES. Neuroimage 2019; 185:181-190. [PMID: 30296560 PMCID: PMC6289748 DOI: 10.1016/j.neuroimage.2018.10.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 09/17/2018] [Accepted: 10/01/2018] [Indexed: 11/25/2022] Open
Abstract
PURPOSE To demonstrate the framework of a novel Hadamard-encoded spectral editing approach for simultaneously detecting multiple low-concentration brain metabolites in vivo at 3T. METHODS HERCULES (Hadamard Editing Resolves Chemicals Using Linear-combination Estimation of Spectra) is a four-step Hadamard-encoded editing scheme. 20-ms editing pulses are applied at: (A) 4.58 and 1.9 ppm; (B) 4.18 and 1.9 ppm; (C) 4.58 ppm; and (D) 4.18 ppm. Edited signals from γ-aminobutyric acid (GABA), glutathione (GSH), ascorbate (Asc), N-acetylaspartate (NAA), N-acetylaspartylglutamate (NAAG), aspartate (Asp), lactate (Lac), and likely 2-hydroxyglutarate (2-HG) are separated with reduced signal overlap into distinct Hadamard combinations: (A+B+C+D); (A+B-C-D); and (A-B+C-D). HERCULES uses a novel multiplexed linear-combination modeling approach, fitting all three Hadamard combinations at the same time, maximizing the amount of information used for model parameter estimation, in order to quantify the levels of these compounds. Fitting also allows estimation of the levels of total choline (tCho), myo-inositol (Ins), glutamate (Glu), and glutamine (Gln). Quantitative HERCULES results were compared between two grey- and white-matter-rich brain regions (11 min acquisition time each) in 10 healthy volunteers. Coefficients of variation (CV) of quantified measurements from the HERCULES fitting approach were compared against those from a single-spectrum fitting approach, and against estimates from short-TE PRESS data. RESULTS HERCULES successfully segregates overlapping resonances into separate Hadamard combinations, allowing for the estimation of levels of seven coupled metabolites that would usually require a single 11-min editing experiment each. Metabolite levels and CVs agree well with published values. CVs of quantified measurements from the multiplexed HERCULES fitting approach outperform single-spectrum fitting and short-TE PRESS for most of the edited metabolites, performing only slightly to moderately worse than the fitting method that gives the lowest CVs for tCho, NAA, NAAG, and Asp. CONCLUSION HERCULES is a new experimental approach with the potential for simultaneous editing and multiplexed fitting of up to seven coupled low-concentration and six high-concentration metabolites within a single 11-min acquisition at 3T.
Collapse
Affiliation(s)
- Georg Oeltzschner
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States.
| | - Muhammad G Saleh
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States
| | - Daniel Rimbault
- Medical Imaging Research Unit, Division of Biomedical Engineering, Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - Mark Mikkelsen
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States
| | - Kimberly L Chan
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States; Department of Bioengineering, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Nicolaas A J Puts
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States
| | - Richard A E Edden
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States
| |
Collapse
|
46
|
Monnig MA, Woods AJ, Walsh E, Martone CM, Blumenthal J, Monti PM, Cohen RA. Cerebral Metabolites on the Descending Limb of Acute Alcohol: A Preliminary 1H MRS Study. Alcohol Alcohol 2019; 54:487-496. [PMID: 31322647 DOI: 10.1093/alcalc/agz062] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 05/28/2019] [Accepted: 06/28/2019] [Indexed: 12/19/2022] Open
Abstract
AIMS Chronic alcohol use is associated with cerebral metabolite abnormalities, yet alcohol's acute effects on neurometabolism are not well understood. This preliminary study investigated cerebral metabolite changes in vivo on the descending limb of blood alcohol in healthy moderate drinkers. METHODS In a pre/post design, participants (N = 13) completed magnetic resonance imaging (MRI) scans prior to and approximately 5 hours after consuming a moderate dose of alcohol (0.60 grams alcohol per kilogram of body weight). Magnetic resonance spectroscopy (1H MRS) was used to quantify cerebral metabolites related to glutamatergic transmission (Glx) and neuroimmune activity (Cho, GSH, myo-inositol) in the thalamus and frontal white matter. RESULTS Breath alcohol concentration (BrAC) peaked at 0.070±0.008% (mean ± standard deviation) and averaged 0.025±0.011% directly prior to the descending limb scan. In the thalamus, Glx/Cr and Cho/Cr were significantly elevated on the descending limb scan relative to baseline. BrAC area under the curve, an index of alcohol exposure during the session, was significantly, positively associated with levels of Glx/Cr, Cho/Cr and GSH/Cr in the thalamus. GSH/Cr on the descending limb was inversely correlated with subjective alcohol sedation. CONCLUSIONS This study offers preliminary evidence of alcohol-related increases in Glx/Cr, Cho/Cr and GSH/Cr on the descending limb of blood alcohol concentration. Findings add novel information to previous research on neurometabolic changes at peak blood alcohol in healthy individuals and during withdrawal in individuals with alcohol use disorder.
Collapse
Affiliation(s)
- Mollie A Monnig
- Center for Alcohol and Addiction Studies, Brown University, Providence, RI, USA
| | - Adam J Woods
- Department of Clinical and Health Psychology and Center for Cognitive Aging and Memory, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Edward Walsh
- Department of Neuroscience, Brown University, Providence, RI, USA
| | | | - Jonah Blumenthal
- Undergraduate Neuroscience Program, Brown University, Providence, RI, USA
| | - Peter M Monti
- Center for Alcohol and Addiction Studies, Brown University, Providence, RI, USA
| | - Ronald A Cohen
- Department of Clinical and Health Psychology and Center for Cognitive Aging and Memory, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| |
Collapse
|
47
|
Psychostimulant drug effects on glutamate, Glx, and creatine in the anterior cingulate cortex and subjective response in healthy humans. Neuropsychopharmacology 2018; 43:1498-1509. [PMID: 29511334 PMCID: PMC5983539 DOI: 10.1038/s41386-018-0027-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 01/19/2018] [Accepted: 02/01/2018] [Indexed: 12/11/2022]
Abstract
Prescription psychostimulants produce rapid changes in mood, energy, and attention. These drugs are widely used and abused. However, their effects in human neocortex on glutamate and glutamine (pooled as Glx), and key neurometabolites such as N-acetylaspartate (tNAA), creatine (tCr), choline (Cho), and myo-inositol (Ins) are poorly understood. Changes in these compounds could inform the mechanism of action of psychostimulant drugs and their abuse potential in humans. We investigated the acute impact of two FDA-approved psychostimulant drugs on neurometabolites using magnetic resonance spectroscopy (1H MRS). Single clinically relevant doses of d-amphetamine (AMP, 20 mg oral), methamphetamine (MA, 20 mg oral; Desoxyn®), or placebo were administered to healthy participants (n = 26) on three separate test days in a placebo-controlled, double-blinded, within-subjects crossover design. Each participant experienced all three conditions and thus served as his/her own control. 1H MRS was conducted in the dorsal anterior cingulate cortex (dACC), an integrative neocortical hub, during the peak period of drug responses (140-150 m post ingestion). D-amphetamine increased the level of Glu (p = .0001), Glx (p = .003), and tCr (p = .0067) in the dACC. Methamphetamine increased Glu in females, producing a significant crossover interaction pattern with gender (p = .02). Drug effects on Glu, tCr, and Glx were positively correlated with subjective drug responses, predicting both the duration of AMP liking (Glu: r = +.49, p = .02; tCr: r = +.41, p = .047) and the magnitude of peak drug high to MA (Glu: r = +.52, p = .016; Glx: r = +.42, p = .049). Neither drug affected the levels of tNAA, Cho, or Ins after correction for multiple comparisons. We conclude that d-amphetamine increased the concentration of glutamate, Glx, and tCr in the dACC in male and female volunteers 21/2 hours after drug consumption. There was evidence that methamphetamine differentially affects dACC Glu levels in women and men. These findings provide the first experimental evidence that specific psychostimulants increase the level of glutamatergic compounds in the human brain, and that glutamatergic changes predict the extent and magnitude of subjective responses to psychostimulants.
Collapse
|
48
|
Steel A, Chiew M, Jezzard P, Voets NL, Plaha P, Thomas MA, Stagg CJ, Emir UE. Metabolite-cycled density-weighted concentric rings k-space trajectory (DW-CRT) enables high-resolution 1 H magnetic resonance spectroscopic imaging at 3-Tesla. Sci Rep 2018; 8:7792. [PMID: 29773892 PMCID: PMC5958083 DOI: 10.1038/s41598-018-26096-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 05/02/2018] [Indexed: 11/10/2022] Open
Abstract
Magnetic resonance spectroscopic imaging (MRSI) is a promising technique in both experimental and clinical settings. However, to date, MRSI has been hampered by prohibitively long acquisition times and artifacts caused by subject motion and hardware-related frequency drift. In the present study, we demonstrate that density weighted concentric ring trajectory (DW-CRT) k-space sampling in combination with semi-LASER excitation and metabolite-cycling enables high-resolution MRSI data to be rapidly acquired at 3 Tesla. Single-slice full-intensity MRSI data (short echo time (TE) semi-LASER TE = 32 ms) were acquired from 6 healthy volunteers with an in-plane resolution of 5 × 5 mm in 13 min 30 sec using this approach. Using LCModel analysis, we found that the acquired spectra allowed for the mapping of total N-acetylaspartate (median Cramer-Rao Lower Bound [CRLB] = 3%), glutamate+glutamine (8%), and glutathione (13%). In addition, we demonstrate potential clinical utility of this technique by optimizing the TE to detect 2-hydroxyglutarate (long TE semi-LASER, TE = 110 ms), to produce relevant high-resolution metabolite maps of grade III IDH-mutant oligodendroglioma in a single patient. This study demonstrates the potential utility of MRSI in the clinical setting at 3 Tesla.
Collapse
Affiliation(s)
- Adam Steel
- Wellcome Centre for Integrative Neuroimaging, FMRIB Division, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX3 9DU, UK
- Laboratory of Brain and Cognition, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20814, USA
| | - Mark Chiew
- Wellcome Centre for Integrative Neuroimaging, FMRIB Division, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX3 9DU, UK
| | - Peter Jezzard
- Wellcome Centre for Integrative Neuroimaging, FMRIB Division, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX3 9DU, UK
| | - Natalie L Voets
- Wellcome Centre for Integrative Neuroimaging, FMRIB Division, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX3 9DU, UK
- Department of Neurosurgery, John Radcliffe Hospital, Oxford, United Kingdom
| | - Puneet Plaha
- Wellcome Centre for Integrative Neuroimaging, FMRIB Division, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX3 9DU, UK
- Department of Neurosurgery, John Radcliffe Hospital, Oxford, United Kingdom
| | - Michael Albert Thomas
- Department of Radiological Sciences, University of California, Los Angeles, California, USA
| | - Charlotte J Stagg
- Wellcome Centre for Integrative Neuroimaging, FMRIB Division, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX3 9DU, UK
- Oxford Centre for Human Brain Activity, Wellcome Centre for Integrative Neuroimaging, Department of Psychiatry, University of Oxford, Oxford, OX3 7JX, UK
| | - Uzay E Emir
- Wellcome Centre for Integrative Neuroimaging, FMRIB Division, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX3 9DU, UK.
- Purdue University, School of Health Sciences, West Lafayette, IN, 47907, USA.
| |
Collapse
|
49
|
Zhang Y, Taub E, Salibi N, Uswatte G, Maudsley AA, Sheriff S, Womble B, Mark VW, Knight DC. Comparison of reproducibility of single voxel spectroscopy and whole-brain magnetic resonance spectroscopy imaging at 3T. NMR IN BIOMEDICINE 2018; 31:e3898. [PMID: 29436038 PMCID: PMC6291009 DOI: 10.1002/nbm.3898] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 12/11/2017] [Accepted: 12/26/2017] [Indexed: 05/09/2023]
Abstract
To date, single voxel spectroscopy (SVS) is the most commonly used MRS technique. SVS is relatively easy to use and provides automated and immediate access to the resulting spectra. However, it is also limited in spatial coverage. A new and very promising MRS technique allows for whole-brain MR spectroscopic imaging (WB-MRSI) with much improved spatial resolution. Establishing the reproducibility of data obtained using SVS and WB-MRSI is an important first step for using these techniques to evaluate longitudinal changes in metabolite concentration. The purpose of this study was to assess and directly compare the reproducibility of metabolite quantification at 3T using SVS and WB-MRSI in 'hand-knob' areas of motor cortices and hippocampi in healthy volunteers. Ten healthy adults were scanned using both SVS and WB-MRSI on three occasions one week apart. N-acetyl aspartate (NAA), creatine (Cr), choline (Cho) and myo-inositol (mI) were quantified using SVS and WB-MRSI with reference to both Cr and H2 O. The reproducibility of each technique was evaluated using the coefficient of variation (CV), and the correspondence between the two techniques was assessed using Pearson correlation analysis. The measured mean (range) intra-subject CVs for SVS were 5.90 (2.65-10.66)% for metabolites (i.e. NAA, Cho, mI) relative to Cr, and 8.46 (4.21-21.07)% for metabolites (NAA, Cr, Cho, mI) relative to H2 O. The mean (range) CVs for WB-MRSI were 7.56 (2.78-11.41)% for metabolites relative to Cr, and 7.79 (4.57-14.11)% for metabolites relative to H2 O. Significant positive correlations were observed between metabolites quantified using SVS and WB-MRSI techniques when the Cr but not H2 O reference was used. The results demonstrate that reproducibilities of SVS and WB-MRSI are similar for quantifying the four major metabolites (NAA, Cr, Cho, mI); both SVS and WB-MRSI exhibited good reproducibility. Our findings add reference information for choosing the appropriate 1 H-MRS technique in future studies.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Psychology, University of Alabama at Birmingham, AL, USA
| | - Edward Taub
- Department of Psychology, University of Alabama at Birmingham, AL, USA
| | | | - Gitendra Uswatte
- Department of Psychology, University of Alabama at Birmingham, AL, USA
- Department of Physical Therapy, University of Alabama at Birmingham, AL, USA
| | | | | | - Brent Womble
- Department of Psychology, University of Alabama at Birmingham, AL, USA
| | - Victor W Mark
- Department of Psychology, University of Alabama at Birmingham, AL, USA
- Department of Physical Medicine and Rehabilitation, University of Alabama at Birmingham, AL, USA
- Department of Neurology, University of Alabama at Birmingham, AL, USA
| | - David C Knight
- Department of Psychology, University of Alabama at Birmingham, AL, USA
| |
Collapse
|
50
|
Raschke F, Noeske R, Dineen RA, Auer DP. Measuring Cerebral and Cerebellar Glutathione in Children Using 1H MEGA-PRESS MRS. AJNR Am J Neuroradiol 2017; 39:375-379. [PMID: 29242361 DOI: 10.3174/ajnr.a5457] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 09/13/2017] [Indexed: 12/29/2022]
Abstract
BACKGROUND AND PURPOSE Glutathione is an important antioxidant in the human brain and therefore of interest in neurodegenerative disorders. The purpose of this study was to investigate the feasibility of measuring glutathione in healthy nonsedated children by using the 1H Mescher-Garwood point-resolved spectroscopy (MEGA-PRESS) sequence at 3T and to compare glutathione levels between the medial parietal gray matter and the cerebellum. MATERIALS AND METHODS Glutathione was measured using MEGA-PRESS MRS (TR = 1.8 seconds, TE = 131 ms) in the parietal gray matter (35 × 25 × 20 mm3) of 6 healthy children (10.0 ± 2.4 years of age; range, 7-14 years; 3 males) and in the cerebellum of 11 healthy children (12.0 ± 2.7 years of age; range, 7-16 years; 6 males). A postprocessing pipeline was developed to account for frequency and phase variations in the edited ON and nonedited OFF spectra. Metabolites were quantified with LCModel and reported both as ratios and water-scaled values. Glutathione was quantified in the ON-OFF spectra, whereas total NAA, total Cho, total Cr, mIns, Glx, and taurine were quantified in the OFF spectra. RESULTS We found significantly higher glutathione, total Cho, total Cr, mIns, and taurine in the cerebellum (P < .01). Glx and total NAA were significantly higher in the parietal gray matter (P < .01). There was no significant difference in glutathione/total Cr (P = .93) between parietal gray matter and cerebellum. CONCLUSIONS We demonstrated that glutathione measurement in nonsedated children is feasible. We found significantly higher glutathione in the cerebellum compared with the parietal gray matter. Metabolite differences between the parietal gray matter and cerebellum agree with published MRS data in adults.
Collapse
Affiliation(s)
- F Raschke
- From the National Center for Tumor Diseases (F.R.), Partner Site Dresden, Dresden, Germany.,German Cancer Research Center (F.R.), Heidelberg, Germany.,Medical Faculty and University Hospital Carl Gustav Carus (F.R.), Technische Universität Dresden, Dresden, Germany.,Helmholtz Association/Helmholtz Zentrum Dresden Rossendorf (F.R.), Dresden, Germany
| | - R Noeske
- GE Healthcare (R.N.), Applied Science Lab Europe, Berlin, Germany
| | - R A Dineen
- Sir Peter Mansfield Imaging Centre (R.A.D., D.P.A.), University of Nottingham, Nottingham, UK .,Radiological Sciences, (R.A.D., D.P.A.), Division of Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham, UK.,National Institute for Health Research (R.A.D., D.P.A.), Nottingham Biomedical Research Centre, Nottingham, UK
| | - D P Auer
- Sir Peter Mansfield Imaging Centre (R.A.D., D.P.A.), University of Nottingham, Nottingham, UK.,Radiological Sciences, (R.A.D., D.P.A.), Division of Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham, UK.,National Institute for Health Research (R.A.D., D.P.A.), Nottingham Biomedical Research Centre, Nottingham, UK
| |
Collapse
|