Spectroscopy: Neuro
Cancer/Spectroscopy/Molecular Imaging/Pre-Clinical Tuesday, 18 May 2021
Oral
Digital Poster
2199 - 2218

Oral Session - Spectroscopy: Neuro
Cancer/Spectroscopy/Molecular Imaging/Pre-Clinical
Tuesday, 18 May 2021 16:00 - 18:00
  • High Resolution Volumetric Diffusion-Weighted MRSI Using A Subspace Approach
    Zepeng Wang1,2 and Fan Lam1,2
    1Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, United States, 2Beckman Institute for Advanced Science and Technology, Urbana, IL, United States
    We propose a novel method to achieve fast high-resolution 3D DW-MRSI. In vivo experimental results have been obtained, which demonstrated that the proposed method can provide high-SNR DW-MRSI of the brain and metabolite-specific ADC maps with the highest ever resolution (3.4×3.4×5.3 mm3).
    Figure 5. Estimated metabolite ADC maps from the 64x64x12 data at a nominal resolution of 3.4×3.4×5.3 mm3. Row 1 shows the T1-weighted images for several slices from the 3D imaging volume. Rows 2-4 display the ADC maps of NAA, Cr, and Cho (from Gdir1) for the corresponding slices, respectively. Patterns of white matter and gray matter diffusion property differences (white matter having larger metabolite ADCs than gray matter) can be visualized due to the high resolution. To the best of our knowledge, these are the highest resolution metabolite ADC maps ever produced.
    Figure 4. Reconstructed DW metabolite and MD maps from the 32x32x8 data. Columns 1-3 present the metabolite maps at 3 b-values from Gdir1. Column 4 shows the MD maps of the matched slice for NAA, Cr, and Cho respectively (Rows 1-3). The MD maps are generated by averaging the registered ADC maps fitted from 3 orthogonal diffusion encoding directions and then overlaid on aligned anatomical images. DW contrast can be observed and the ranges and values of the MD for each metabolite are consistent with reported values7-9,17.
  • 3D-CRT-FID-MRSI in the brain at 7T: Evaluation of regional concentration estimates
    Gilbert Hangel1,2, Benjamin Spurny3, Philipp Lazen2, Cornelius Cadrien1,2, Sukrit Sharma2, Zoe Käfer2, Nikolaus Doblinger2, Lukas Hingerl2, Eva Hečková2, Bernhard Strasser2, Stanislav Motyka2, Alexandra Lipka2, Stephan Gruber2, Christoph Brandner4, Rupert Lanzenberger3, Karl Rössler1, Siegfried Trattnig2,5, and Wolfgang Bogner2
    1Department of Neurosurgery, Medical University of Vienna, Vienna, Austria, 2High Field MR Centre, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria, 3Division of General Psychiatry, Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria, 4High Field MR Centre, Centre for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria, 5Christian Doppler Laboratory for Clinical Molecular MR Imaging, Vienna, Austria
    Using 7T 3D-CRT-FID-MRSI, we successfully determined metabolite concentration estimates and their coefficients of variation for 13 metabolites in 44 brain regions for healthy volunteers.
    Figure 2: Exemplary concentration estimate maps of all successfully quantified metabolites in one subject. For lower SNR-metabolites, more artifacts or regions without successful quantification are visible. Good visibility of GM/WM and regional variations can be seen especially for neurotransmitters.
    Figure 3: Mean concentration estimates per ROI in [mM/l] for successfully quantified metabolites in all ROIs qualified as defined in Fig.1.
  • Simultaneous Detection of Metabolite Concentration Changes, Water BOLD Signal and pH Changes during Visual Stimulation in the Human Brain at 9.4T
    Johanna Dorst1, Tamas Borbath1, and Anke Henning1,2
    1High-Field MR Center, Max Planck Institute for Biological Cybernetics, Tübingen, Germany, 2Advanced Imaging Research Center, UT Southwestern Medical Center, Dallas, TX, United States
    The metabolite-cycling technique was introduced for 1H fMRS in the activated human brain at 9.4T. The correlation between water BOLD and increase of Lac and Glu during activation could be confirmed; in addition, dynamics in the PCr buffer system and pH were determined.

    Figure 1: Top: Schematic diagram of the fMRS visual stimulation paradigm using a red-black checkerboard pattern. Numbers indicate the acquired number of averages per block and needed acquisition time. FLASH insets below show the BOLD activated region and the fMRS voxel placement (15x18x20 mm3).

    Bottom: Representative MR spectra from one volunteer acquired during STIM (red, S1.2/S2.2) and REST (blue, R2.2/R3.2) periods (64 averages each spectrum) and the difference spectrum (yellow). Small amplitude and linewidth changes of Cr+PCr and NAA indicate the BOLD effect.

    Figure 4: Left: Mean time courses of Lac and Glu concentration differences relative to a baseline concentration (R1.2) with a time resolution of 40 s. Error bars represent standard deviations of the mean over all volunteers.

    Right: Correlation plots between the metabolite concentrations (from spectra summed across all volunteers) and the MC water amplitude (blue crosses). Solid red lines represent the linear fit, dotted red lines 95% confidence intervals. R and p values indicate the Spearman’s Rank Correlation Coefficient and significance level of the correlation, respectively.

  • In Vivo Measurement of 13C Labeling of Glutamate and Glutamine in the Human Brain Using 1H MRS
    Li An1, Shizhe Li1, Maria Ferraris Araneta1, Milalynn Victorino1, Christopher Johnson1, and Jun Shen1
    1National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
    A single-step spectral editing 1H MRS technique with TE = 56 ms was used to measure fractional enrichments of Glu and Gln in the human brain after oral administration of [U-13C]glucose.
    FIGURE 2 Time-course spectra and corresponding fitted spectra of Glu, Gln, and their 13C satellites acquired from the dACC of subject 1. No linebroadening was applied to the spectra. Voxel size = 3.5 × 1.8 × 2 cm3; TR = 2.2 s; TE = 56 ms; number of averages = 264 and total scan time = 10 min for the pre-13C spectra; number of averages = 132 and total scan time = 5 min for each individual post-13C spectrum.
    FIGURE 4 Plots of fractional enrichments of Glu H4 and Gln H4 vs. time after oral administration of [U-13C]glucose for all five healthy volunteers.
  • Fast High-Resolution 19F-MRSI of Perfluorocarbon Nanoemulsions for MRI Cell Tracking Using SPICE with Learned Subspaces
    Yibo Zhao1,2, T. Kevin Hitchens3,4, Michele Herneisey5, Jelena M. Janjic5, Rong Guo1,2, Yudu Li1,2, and Zhi-Pei Liang1,2
    1Department of Electrical and Computer Engineering, University of Illinois, Urbana-Champaign, Urbana, IL, United States, 2Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, Urbana, IL, United States, 3Animal Imaging Center, University of Pittsburgh, Pittsburgh, PA, United States, 4Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, United States, 5Graduate School of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, United States
    A novel method is proposed for accelerated high-resolution 19F-MRSI of multiple perfluorocarbon nanoemulsions, using EPSI trajectories and union-of-subspaces modeling with pre-learned subspaces. The proposed method has been validated using both simulation and experimental data.
    Figure 4. Results obtained from a fixed rat brain injected with PCE and PFOB NEs. PCE (red) and PFOB (green) maps are overlaid on the 1H anatomical image, and representative spectra are shown. As can be seen, the proposed method obtained PCE and PFOB maps and spectra as expected.
    Figure 3. Proton structural image, field map, and comparison of Fourier-based and proposed 19F-MRSI processing results obtained from a phantom. As can be seen, Fourier-based processing results suffered from spatial chemical shift artifacts and spectral aliasing artifacts, while the proposed method produced artifact-free spatiospectral functions for PCE and PFOB.
  • Diffusion-weighted magnetic resonance spectroscopy in the cerebellum of a rat model of hepatic encephalopathy at 14.1T
    Jessie Mosso1,2,3, Julien Valette4, Katarzyna Pierzchala1,2, Dunja Simicic1,2,3, Ileana Ozana Jelescu1,2, and Cristina Cudalbu1,2
    1CIBM Center for Biomedical Imaging, Lausanne, Switzerland, 2Animal Imaging and Technology, EPFL, Lausanne, Switzerland, 3LIFMET, EPFL, Lausanne, Switzerland, 4Commissariat à l'Energie Atomique (CEA), Institut d'Imagerie Biomédicale (I2BM), Molecular Imaging Research Center (MIRCen), Fontenay-aux-Roses, France
    A significant decrease in the diffusion coefficient of Gln and an increase for Tau and Glu in the cerebellum of a rat model of chronic hepatic encephalopathy was measured, reflecting changes in astrocytes morphology measured ex vivo by histology.  
    A. Representative neurometabolic profiles in BDL and Sham rats acquired at 14.1T in the cerebellum (STEAM sequence - TE=3ms, TM=10ms, LB=8Hz) B. Representative spectra at different b-values for BDL and SHAM rats acquired with the STE-LASER4 sequence (LB=8Hz) in the cerebellum at 14.1T. C. Individual metabolite decays versus b-values for both groups. Error bars: dispersion around the mean value. Statistics: 2-way ANOVA with variables disease and b-value, *: p<0.05.
    IHC staining in the cerebellum of Sham and BDL rats. Top: anti-GFAP(red)/DAPI(blue) staining of the cerebellar folium, scale bar: 100μm. Middle: GFAP/DAPI staining of the granular layer, scale bar: 25μm. Bottom: Colgi-Cox staining of Purkinje cells, scale bar: 25μm Right: corresponding quantifications, ** p<0.01, *** p<0.001, **** p<0.0001.
  • Validation of dynamic Deuterium Metabolic Imaging (DMI) for the measurement of cerebral metabolic rates of glucose in rat.
    Claudius Sebastian Mathy1,2,3, Monique A. Thomas1, Graeme F. Mason1,4,5, Robin A. de Graaf1,5, and Henk M. De Feyter1
    1Department of Radiology and Biomedical Imaging, Magnetic Resonance Research Center, Yale University, New Haven, CT, United States, 2Institute of Physical and Theoretical Chemistry, University of Bonn, Bonn, Germany, 3Department of Diagnostic and Interventional Radiology, RWTH Aachen, Aachen, Germany, 4Department of Psychiatry, Yale University, New Haven, CT, United States, 5Department of Biomedical Engineering, Yale University, New Haven, CT, United States
    Dynamic, spatially localized DMI data, and proton-observed carbon-edited (POCE) MRS data were acquired during infusion of [6,6’-2H2]-glucose and [1-13C]-glucose. A metabolic model incorporating 2H-label loss provided metabolic flux rates that agreed with those based on POCE MRS.
    Fig. 1: Metabolic model: Glucose is transported through the blood-brain barrier by GLUT-1 transporters and metabolized to pyruvate/lactate by CMRgl. Lactate also enters via monocarboxylic acid transporters, characterized by Vmax, KM and KD. Lactate/pyruvate is further metabolized in the TCA cycle (Vtca). Unlabeled substrate in- and outflows are Vdil,gly = Vlac,out,brain and Vdil,tca. The glutamate/glutamine cycle is represented by Vgln. Modifications for 2H are indicated in red. 13C and 2H label positions are shown.
    Fig. 3: Axial MR image for spatial assignment. Time course of 2H MR spectra during [6,6’-2H2]-glucose infusion, starting from baseline prior to infusion until the end of the experiment with n=64 averages each, acquired from combined cortex/subcortex (Cx/SCx) volume. Line broadening=2Hz. Labeled metabolites are marked as double labeled but the signals include both single and double 2H-labeled metabolites.
  • Combining 1H MRS and deuterium labeled glucose for mapping of neural metabolism in humans
    Abigail T.J. Cember1, Laurie J. Rich1, Puneet Bagga1, Neil E. Wilson2, Ravi Prakash Reddy Nanga1, Deepa Thakuri1, Mark Elliott1, Mitchell D. Schnall3, John A. Detre4, and Ravinder Reddy1
    1Center for Magnetic Resonance and Optical Imaging, Department of Radiology, University of Pennsylvania, Philadelphia, PA, United States, 2Siemens Medical Solutions, USA, Malvern, PA, United States, 3Department of Radiology, University of Pennsylvania, Philadelphia, PA, United States, 4Department of Neurology, University of Pennsylvania, Philadelphia, PA, United States
    Using only 1H MRS at 7T, we present dynamic mapping of neurometabolism downstream of glucose absorption. Specifically, we focus on the possibility of measuring glutamate turnover in a spatially resolved fashion using chemical shift imaging in which the signal is modulated by labeling with 2H. 
    Figure 1. Schematic of qMRS experiment. Subject drinks aqueous deuterated glucose solution. Prior to this, subject has undergone MR spectroscopy and chemical shift imaging (qCSI) at 7T. After glucose ingestion, subject is scanned again. In this post-glucose scan, 1H resonances of metabolites experiencing labeling downstream from glucose will decrease. qCSI measurements are made repeatedly and, as labelled glucose is absorbed from the bloodstream and metabolized, the fractional enrichment of detected metabolites increases and eventually plateaus.
    Figure 3. Timecourse qCSI data of [Glu] and [NAA] from all subjects, gray and white matter. A) Glu/NAA ratio map (MRSI) illustrating ROIs used for calculation of the plotted data designated as gray and white matter regions. B) Bar graphs showing the timecourse measurements of Glu and NAA by qCSI in gray and white matter. In this plot, individual dots correspond to a single voxel (out of 15) in each ROI, averaged across four subjects who underwent this part of the experiment.
  • The Macromolecular Background Spectrum Does Not Change with Age in Healthy Participants
    Steve C.N. Hui1,2, Tao Gong3, Helge J. Zöllner1,2, Yulu Song3, Yufan Chen3, Muhammad G. Saleh4, Mark Mikkelsen1,2, Georg Oeltzschner1,2, Sofie Tapper1,2, Weibo Chen5, Richard A.E. Edden1,2, and Guangbin Wang3
    1Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States, 2F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States, 3Department of Imaging and Nuclear Medicine, Shandong Medical Imaging Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, China, 4Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, United States, 5Philips Healthcare, Shanghai, China
    Metabolite-suppressed MM spectra were acquired in 100 subjects from 20 to 70 years of age. The MM spectra are strikingly similar between male and female subjects, and extremely stable as a function of age.
    Figure 1. Osprey analysis workflow to remove residual metabolite signals and generate ‘clean’ MM spectrum.
    Figure 2. MM spectra from centrum semiovale white matter. Spectra from 96 subjects are overlaid in panel A, and the metabolite model signals are labeled in panel B. The mean spectra for male and female subjects are compared in panel C. The mean spectra for each decade of age are compared in panel D.
  • Ultra high-field, high-resolution, multi-voxel MRS in pre-manifest and early-manifest Huntington's Disease
    Yan Li1, Huawei Liu1, Angela Jakary1, Sivakami Avadiappan1, Melanie Morrison1, Ralph Noeske2, Peder E.Z. Larson1, Alexandra Nelson3, Katherine Possin3, Michael Geschwind3, Christopher Hess1, and Janine M Lupo1
    1Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, United States, 2GE Healthcare, Munich, Germany, 3Department of Neurology, University of California San Francisco, San Francisco, CA, United States
    This study demonstrates the feasibility of using a fast and high-resolution multi-voxel MRSI at 7T to obtain brain metabolites from patients with PM and EM at the spatial resolution of 0.5x0.5x2.0cm (0.5cm3) within a clinical feasible acquisition of 7 minutes.
    Figure 1. Example of multivoxel MRS and ROIs from a patient with pre-manifest HD.
    Figure 3. Significant differences in metabolite ratios between either the PM or EM groups and all of controls combined (HCEM+HCPM). Metabolites from healthy controls were plotted for each subgroup (HCEM, HCPM). When compared to age-/gender-matched healthy controls, only glutathione/creatine within the insula (EM vs. HCEM, p=0.002) remained significant.
Back to Top
Digital Poster Session - Spectroscopy: Neuro I
Cancer/Spectroscopy/Molecular Imaging/Pre-Clinical
Tuesday, 18 May 2021 17:00 - 18:00
  • Evaluation of a spectroscopy sequence using polychromatic refocusing to suppress J-modulation and measure lactate diffusion in the rodent brain
    Sophie Malaquin1, Eloïse Mougel1, Melissa Vincent1, and Julien Valette1
    1Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Centre National de la Recherche Scientifique (CNRS), Molecular Imaging Research Center (MIRCen), Laboratoire des Maladies Neurodégénératives, Fontenay aux Roses, France
    In an effort to optimize lactate detection in diffusion-weighted NMR spectroscopy, we compare different sequences including a spin echo sequence using polychromatic pulse to suppress the J-coupling. This sequence results in stronger lactate signal and more precise diffusion measurements.
    Figure 2: a. Spectra without diffusion-weighting obtained with the STE-LASER sequence (blue dotted line), the broad pulse SE-LASER sequence (green line) and the polychromatic SE-LASER sequence (red line). b. The gain of lactate signal is spectacular when using the polychromatic SE-LASER sequence.
    Figure 3: STE-LASER and polychromatic SE-LASER data at high diffusion-weighting in WT mice. a. Spectra acquired up to high diffusion-weighting (from b = 0.02 ms/µm² to 20 ms/µm²) with the STE-LASER sequence (left) and the polychromatic SE-LASER sequence (right). b. LCModel analysis for metabolites of interest. Macromolecules contribution on 1.31 ppm lactate peak is noticeable. c. Signal attenuation as a function of b for three metabolites by using the two sequences (blue diamonds = polychromatic SE-LASER, red squares = STE-LASER). Data points and error bars stand for mean ± s.d.
  • In Vivo J-difference Editing of (Phosphoryl)ethanolamine
    Muhammad G Saleh1, Steve Hui2, Helge Zöllner2, and Richard A Edden2
    1University of Maryland School of Medicine, Baltimore, MD, United States, 2The Johns Hopkins University School of Medicine, Baltimore, MD, United States
    MEGA-PRESS editing of Phosphorylethanolamine/ethanolamine was successfully demonstrated at both 3.22 and 3.98 ppm. In vivo experiments show that the 3.22 ppm signal is better resolved and yield reproducible quantification.
    Figure 2: In vivo MEGA-PRESS experiments at TE 90 and 110 ms for editing the PE3.22 (left) and PE3.98 (right). NAA: N-acetylaspartate; Lac: lactate; Cho: choline; and Cr: creatine.
    Figure 3: Linear-combination modeling of PE3.22 and PE3.98 difference spectra with complete fit (yellow), spline baseline (black), and PE estimates at both TEs 90 ms (blue) and 110 ms (red). Asp: aspartate, Cho: choline, Cr: creatine, Gln: glutamine, Ins: myo-Inositol, Lac: lactate, MM: macromolecules, Tau: taurine.
  • Spectral quality of J-refocused spectroscopic imaging at 7T
    Jullie W Pan1, Victor W Yushmanov2, Chan H Moon3, Brian Soher4, Frank H Lieberman5, and Hoby P Hetherington2
    1Radiology, University of Pittsburgh, Pittsburgh, PA, United States, 2university of pittsburgh, pittsburgh, PA, United States, 3University of Pittsburgh, Pittsburgh, PA, United States, 4Radiiology, Duke University, Durham, NC, United States, 5Neurology, University of Pittsburgh, Pittsburgh, PA, United States
    Spectral quality of fast 7T MR spectroscopic imaging is evaluated for multiplet compounds including Glutamine, myo-Inositol, Choline and demonstrated in brain tumor patients
    FIg. 2. (A,B) Spectral data from the frontal-parietal (A) and temporal (B) regions. CRLB % are shown for several loci in the temporal study. (C1,C2) The GLU/tNAA regresses significantly with fraction GM from both studied regions. (D) The CRLB % are shown for GLU, INS and GLN for the frontal-parietal and temporal regions.
    Fig. 3. (A) Regression of CRLB % with ξk=√LW/SNRk from the frontal-parietal region. (B) Same as (A) but with superimposed data from the temporal region, and magnified in X and Y axes. (C) Plots of CRk = CRLBk% * Ampk, with ξ0 =√LW*σ, with different compounds. The regressions for CRE, INS and CHO are shown.
  • Associations of tau aggregates and oxidative stress to apathy levels in progressive supranuclear palsy
    Kiwamu Matsuoka1,2, Yuhei Takado1, Kenji Tagai1, Manabu Kubota3, Yasunori Sano1, Keisuke Takahata1, Maiko Ono1, Chie Seki1, Hideki Matsumoto1,4, Hironobu Endo1, Hitoshi Shinotoh1, Jamie Near5, Kazunori Kawamura1, Ming-Rong Zhang1, Hitoshi Shimada1, and Makoto Higuchi1
    1National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan, 2Department of Psychiatry, Nara Medical University, Kashihara, Japan, 3Department of Psychiatry, Kyoto University Graduate School of Medicine, Kyoto, Japan, 4Department of Oral and Maxillofacial Radiology, Tokyo Dental College, the city of Chiyoda-ku, Japan, 5Douglas Mental Health University Institute and Department of Psychiatry, McGill University, Montréal, QC, Canada
    This multi-modal imaging study of tau aggregates and antioxidant glutathione using tau PET and magnetic resonance spectroscopy revealed associations of apathy in progressive supranuclear palsy with tau aggregates and oxidative stress.
    Representative spectrum in the ACC and PCC. Volumes of interest of magnetic resonance spectrum were placed at the ACC and PCC. A representative MRS acquired with the SPECIAL sequence, the corresponding LCModel spectral fit, fit residual, macromolecules (MM), baseline and individual metabolite fits including GSH in the ACC and PCC.
    a) Sagittal, coronal, and transverse brain views showed voxels with positive associations between apathy scale scores and 18F-PM-PBB3 SUVRs in patients with PSP after controlling for the effects of age and PSPRS. b, c) Scatterplots showing the association between apathy scale scores and 18F-PM-PBB3 SUVRs in the detected areas (AG).
  • Sleep-Wake Lactate Dynamics in Human Brain
    Selda Yildiz1,2, Miranda M. Lim2,3,4,5,6, Manoj K. Sammi1,7, Katherine Powers1, Charles F. Murchison2,8, Jeffrey J. Iliff9,10,11,12,13, and William D. Rooney1,2,5,10
    1Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR, United States, 2Department of Neurology, Oregon Health & Science University, Portland, OR, United States, 3VA Portland Health Care System, Portland, OR, United States, 4Department of Medicine, Division of Pulmonary and Critical Care Medicine, Oregon Health & Science University, Portland, OR, United States, 5Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States, 6Oregon Institute of Occupational Health Sciences;, Oregon Health & Science University, Portland, OR, United States, 7BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States, 8Department of Biostatistics, University of Alabama, Birmingham, AL, United States, 9Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR, United States, 10Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, United States, 11VISN 20 Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, Seattle, WA, United States, 12Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, United States, 13Department of Neurology, University of Washington, Seattle, WA, United States
    We present the first in-vivo demonstration of reductions in lactate levels and diffusivity during sleep versus wake in human brain. Findings, in consistent with invasive small-animal studies, support an altered lactate metabolism and/or increased glymphatics in sleeping human brain. 
    Figure 2. a) Subject 1: mean lactate concentration across sleep-wake cycles decline during sleep relative to wakefulness: 4.6 % in N1 (2.5 min), 12.0 % in N2 (12 min), and 18.0 % in N3. b) Representative sleep stages were visually scored for 30 s epochs and classified into: Wake (W), Non-REM sleep stage 1 (N1), Non-REM sleep stage 2 (N2), and Non-REM sleep stage 3 (N3).
    Figure 1. a) Measured single voxel (22 cm3, orange square) on a sagittal anatomical MR image of a 27-year old healthy young female (subject 1). b) A 7.5 sec 3T single voxel 1H-MR spectroscopy showing metabolites including lactate, c) Lactate concentrations, calculated within [1.23-1.48 ppm], are averaged over each sleep stage including Wake (W), non-REM stage 2 (N1), non-REM stage 2 (N2) and non-REM stage 3 (N3).
  • Reduced fMRI activation in the fusiform face area is related to higher hallucination proneness and lower glutamate levels assessed by 1H MRS
    Ralf Mekle1, Jochen B. Fiebach1, and Heiner Stuke2
    1Center for Stroke Research Berlin, Charité – Universitätsmedizin Berlin, Berlin, Germany, 2Department of Psychiatry and Psychotherapy, Charité – Universitätsmedizin Berlin, Berlin, Germany
    Combining fMRI and 1H MRS at 3T revealed that reduced activation in the fusiform face area is related to higher hallucination proneness and lower glutamate levels, thus supporting theories of impaired glutamatergic transmission being involved in the formation of hallucinations.
    Fig. 2. Activations in the a priori defined ROI of the bilateral gyrus fusisormis for face-stimuli versus non-face stimuli (a, N = 18, MNI peak voxel = 38, -38, -18, puncorr < 0.001, pfwe < 0.001) and correlations of these activations with hallucination proneness in the face task (b, negative correlation, N = 18, MNI peak voxel = 38, -38, -18, puncorr < 0.001, pfwe = 0.040) and glutamate/NAA ratios obtained from MRS (c, N = 16, MNI peak voxel = 32, -40, -16, puncorr < 0.001, pfwe = 0.121). In the figure, p-values of 0.005 were used for illustration purposes.
    Fig. 1. 1H difference MR spectra from a VOI in the right visual cortex (bottom inset) acquired at 3T with the MEGA-PRESS sequence (TR/TE = 3000/68 ms) for a healthy volunteer together with LCModel fit, LCModel output for GABA, Glu, fit residuals, and background. Preprocessing of data included coil combination, removal of bad averages, and phase and frequency correction.
  • Simultaneous spectral and bi-exponential diffusion modeling of doubly motion-corrected human brain spectra with very high b-values
    Kadir Şimşek1, André Döring1,2, André Pampel3, Harald E. Möller3, and Roland Kreis1
    1Department of Radiology and Biomedical Research, University of Bern, Bern, Switzerland, 2Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, United Kingdom, 3Max-Planck Institution for Human Cognitive and Brain Sciences, Leipzig, Germany
    Diffusion-weighted MRS is used at short-TE in human brain with two motion compensation methods and a novel simultaneous spectral/diffusion-decay model to study non-Gaussian diffusivity of metabolites at high b-values and to determine the macromolecular background spectrum by diffusion.
    Fig.3: Fitted diffusion decays as obtained with the different approaches described in the text. Free-Fit data was modeled as mono- and biexponential decay. Biexp-Fit (blue) is consistent for some metabolites but shows diverging results for others. The non-Gaussian nature of diffusion is evident in both approaches for most metabolites. At the bottom, spectra with the fits from Free-Fit as well as Biexp-Fit are shown documenting how small the differences are even though the decay function seem to differ substantially for some metabolites. (blue: Biexp-Fit; red; Free-Fit)
    Fig. 1: Illustration to motivate the need for the MM peak-based signal correction. For multiple subjects, the peak areas of MM0.9 are plotted in relation to the monoexponential decay fit for the cohort data from subjects without apparent strong artifactual signal decay. Subject 2 shows systematic additional signal decay with higher b-values on top of the MM diffusion effect, while subjects 1 and 3 show strong random fluctuations.
  • Rapid MRSI of the Brain on 7T Using Subspace-Based Processing
    Fan Lam1,2, Hoby Hetherington3, and Jullie Pan3
    1Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, United States, 2Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, United States, 3Radiology, University of Pittsburgh, Pittsburgh, PA, United States
    We present a new method for rapid 7T MRSI of the brain that uses subspace imaging to improve rosette MRSI (RSI). In vivo results showed substantial improvement over the standard RSI reconstruction and highlight the potentials of integrating acquisition and processing  for ultrahigh-field MRSI.
    Spatially-localized spectra for a 4x4 voxel array from an anterior brain region are compared, with results produced by the standard reconstruction (see texts) and the proposed method (the blue curves). The location of the region-of-interest is indicated by the red box in the anatomical image on the left with overlaid voxel grids. Significant reduction in lipid leakage and SNR improvement can be observed for the spectra from the proposed method, especially for the edge voxels. Spectra are shown in real parts.
    Comparison of NAA/Cr (column 1), Cho/Cr (column 2), Glu/NAA (column 3) maps from the standard reconstruction and the proposed method for one subject. As can be seen, the metabolite ratio maps produced by the proposed method have less artifacts (examples indicated by the red arrows) and better gray/white matter contrast (e.g., the Glu/NAA maps). The color scales were the same for the same column. The metabolite levels were obtained using spectral integration for this preliminary study, and no relaxation correction was performed.
  • Echo-planar Spectroscopic Imaging with Readout-segmented COKE at 7T:  Artifact Analysis using a Purpose-Built Phantom and Simulation
    Graeme A. Keith1, Amir Seginer2, David A. Porter1, and Rita Schmidt3
    1Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, Scotland, 2Siemens Healthcare Ltd, Rosh Ha’ayin, Israel, 3Neurobiology Department, Weizmann Institute of Science, Rehovot, Israel
    Readout-segmented COKE achieves high spectral width EPSI spectra, with coherent phase evolution at 7T, in the human brain. Residual lipid contamination, due to the readout-segmented trajectory, is investigated in simulations and phantom scans.
    Figure 3: Human scan results. a) Reference image with the scan location, b) water image with the same parameters as the spectroscopic imaging scan. c) 1H spectra at three regions shown in b). The scan parameters are described in the main text.
    Figure 4: 3D-shaped phantom imaging a) RS-EPSI versus RS-COKE with 3 readout segments with the “brain only” phantom. Images show water and NAA maps and the spectrum for the central box (5x5 pixels). b) 3D-shaped phantom including brain-like compounds and a superficial compartment with a lipid-like compound. The slice location is shown by blue overlay. Images compare single and 3-segment water images, a 3-segment acquisition with water suppression, and a 3-segment acquisition with both water suppression and three saturation bands. Sequence parameters are given in the main text.
  • Whole Brain 3D-FIDESI-MRSI: Revisiting Free-Induction-Decay & Spin-Echo Readouts with Concentric Rings at 7 T
    Lukas Hingerl1, Wolfgang Bogner1, Bernhard Strasser1, Petr Bednarik1, Stanislav Motyka1, Eva Heckova1, Ivica Just1, Alexandra Lipka1, Ovidiu Andronesi2, Stephan Gruber1, Siegfried Trattnig1,3, and Gilbert Hangel1,4
    1Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, High Field MR Centre, Vienna, Austria, 2Department of Radiology, Massachusetts General Hospital, Martinos Center for Biomedical Imaging, Boston, MA, United States, 3Christian Doppler Laboratory for Clinical Molecular MR Imaging, Vienna, Austria, 4Department of Neurosurgery, Medical University of Vienna, Vienna, Austria
    3D-FIDESI-MRSI enables whole-brain lactate mapping without lipid contamination by extending an FID sequence with a spin-echo readout, which features a reduced but oversampled spherical k-space readout.
    Figure 1: FIDESI features two analog-to-digital-converter readouts: Large k-space coverages for the high SNR FID scan and a small k-space coverage for the low SNR echo scan at TE 288 ms. The crusher gradient superimposed on the localization gradient ramp-down caused spectral acoustic sideband artifacts. A special pair of gradients was played out between the GOIA pulses for nulling the zero-order magnetization caused by the initial CRT gradients which move the trajectory to the specific ring radii.
    Figure 4: Whole brain metabolic Maps of the healthy Volunteer. FID maps have an effective measured matrix size of 64x64x29, echo maps have an effective measured matrix size of 32x32x15, but are interpolated to 32x32x29 before quantification. All maps are additionally interpolated by a factor of 2 for visualization purposes.
  • 13C-Glucose Labeling Effects measured in the Occipital Lobe and the Frontal Cortex using short TE 1H MC-semiLASER SVS at 9.4T
    Theresia Ziegs1,2, Johanna Dorst1,2, and Anke Henning1,3
    1MRZ, MPI for Biological Cybernetics, Tuebingen, Germany, 2IMPRS for Cognitive and Systems Neuroscience, Tuebingen, Germany, 3Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, United States
    Labeling effects after oral intake of [13C-1]glucose are observed in humans using a 1H MC-semiLASER sequence at 9.4 T. Spectral time series acquired in the frontal cortex as well as the occipital lobe show labelling induced changes of  Glu and Gln spectral pattern directly detected with 1H MRS.
    Figure 2: Time series of the MC-semiLASER spectra from the occipital lobe (left) and frontal cortex (right) and the corresponding difference spectra underneath. The difference spectra in the spectral region of interest between 1.9 and 2.8 is zoomed in to make changes more visible. The colorbar indicates the time of the (difference) spectra after the glucose administration. Changes in the spectral region of NAA and Cr can occur due to subtraction errors.
    Figure 3: a) 1H MC-semiLaser spectrum from one volunteer of the occipital lobe before glucose administration (black line) and 127 minutes after the administration (red line) and the difference spectrum (blue line). Obvious peak changes are marked: Decreasing peaks are marked in purple and increasing peaks are marked in green. The spectra contain 64 averages. b) Simulated spectra for selected metabolites and labeled metabolites.
  • Repeatability assessment of GABA and GSH concentrations with HERMES: a comparison between traditional analysis and a novel approach
    Diana Rotaru1, Georg Oeltzschner2,3, Richard Edden2,3, and David Lythgoe1
    1Neuroimaging, King's College London, London, United Kingdom, 2Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States, 3F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States
    A novel analysis method is proposed, the simultaneous modelling of HERMES GABA and GSH difference and sum spectra. The repeatability of this approach yielded comparable results to those produced by traditional single-spectrum analysis. 
    Figure.1 Transformation of the Hadamard reconstructed spectra (SUM - green, diff_GABA - red, diff_GSH - yellow) into a single concatenated spectrum with 1.2 ppm gap separation
    Figure 3. Mean processed spectral data is shown for test (black) and retest (grey) acquisitions at the top; mean LCModel fits, residuals and baselines are presented for test (black) and retest (grey) at the bottom
  • Constrained Optimized Water Suppression (COWS) for 1H Magnetic Resonance Spectroscopy
    Karl Landheer1, Martin Gajdošík1, and Christoph Juchem1,2
    1Biomedical Engineering, Columbia University, New York City, NY, United States, 2Radiology, Columbia University, New York City, NY, United States
    A novel algorithm for water suppression was introduced and validated, which can accommodate an arbitrary number of pulses, minimum durations between pulses, total module duration and maximum flip angles.
    Figure 3: Metabolite spectra obtained from the occipital lobe (top row) and parietal lobe (bottom row) for the three water suppression schemes tested here, VAPOR, COWS(7;236) and COWS(12;626). COWS(7;236) has comparable performance to VAPOR, while COWS(12;626) has marked improvement over both. Note that WS schemes were applied as theoretically optimized and no experimental fine tuning was performed.
    Figure 4: Macromolecule spectra obtained from the parietal lobe for VAPOR (red) and COWS(7;236) (black). Marked reduction of residual water for COWS(7;236) was observed over VAPOR. COWS(12;626) was not used due to its interference with the double inversion preparation module.
  • Feasibility of 1H-[13C] MRS of hippocampal and hypothalamic metabolism in the very same mouse with dynamic shim update (DSU)
    Hikari Yoshihara1, Nicolas Kunz2, and Hongxia Lei2,3
    1EPFL-LIFMET, Lausanne, Switzerland, 2EPFL-CIBM, Lausanne, Switzerland, 3United-Imaging Co. Ltd, Wuhan, China
    13C MRS of the very same mouse hippocampus and hypothalamus at 14.1T is feasible using dynamic shimming update (DSU).
    Figure 1. Schematic layout of the experimental design for 1H-[13C] MRS acquisition of both hippocampus and hypothalamus with dynamic shim update (DSU) (a), and typical non-edited (b) and edited (c) MR spectra of hippocampus (blue) and hypothalamus (orange).
    Figure 3. Neurochemical profiles of hippocampus and hypothalamus
  • Utility of Residual Water in Proton MR Spectroscopy. The Measurement of Voxel Temperature and Hypoxia
    Ralph E. Hurd1, Meng Gu1, Phil Adamson2, Kirk Riemer3, Ryan Beckman3, Michael Ma3, Kenichi Okamura3, Frank Hanley3, and Daniel Spielman1
    1Radiology, Stanford, Stanford, CA, United States, 2Electrical Engineering, Stanford, Stanford, CA, United States, 3Cardiothoracic Surgery, Stanford, Stanford, CA, United States
    A dominant, but manageable residual water signal in MR spectroscopy provides substantial value. A residual water-by-design strategy should be considered in sequence and protocol design.
    Figure 2. Baseline 37°C spectrum after PWS to 1% residual water. Good fidelity observed for nearest neighbors, the C1α of glucose and the anomeric proton of NAA. Spectral baseline for normal fit region above 4.2 ppm is unperturbed.
    Figure 4. 37C Circulatory arrest. Powder-pattern develops during circulatory arrest and resolves after 5 minutes of recovery. The residual water reveals low level powder pattern which provides a measure of deoxy-Hb and hypoxia. Glucose recovers but lactate persists after hypoxia reversed.
  • Mapping of differential metabolic regions of RRMS patients in multi-slices dimensional using Spiral-MRSI technique
    Oun Al-iedani1,2, Jeannette Lechner-Scott2,3,4, Rodney Lea2, Ovidiu Andronesi5, and Saadallah Ramadan2,6
    1School of Health Sciences, University of Newcastle, Newcastle, Australia, 2Hunter Medical Research Institute, Newcastle, Australia, 3Department of Neurology, John Hunter Hospital, Newcastle, Australia, 4School of Medicine and Public Health, University of Newcastle, Newcastle, Australia, 5Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States, 6Faculty of Health and Medicine, University of Newcastle, Newcastle, Australia
    Novel pipeline of Multi-slice spiral-MRSI coupled with DMRs revealed NAA mapping  in WM-lesions was significantly lower than NAWM-MS and HCs, and is sensitive in diagnosing NAWM in RRMS.
    Pipeline of volumetric brain tissue segmentation from MRI and MRSI data. A. A binary mask of a MRSI single slice VOI (8x10 cm2) was created using the SPM toolbox. B. partial volume masks for each tissue type were created using FSL FAST. C. Partial volume segmentation of the lesion filled T1-MPRAGE structural images. D. Tissue segmentation of MRSI VOI (CSF, GM and WM) and lesion segmentation overlaid on the T2-FLAIR image.
    A. Box plot of significant differences in the neurometabolic ratios (NAA/tCr)and (m-Ins/tCr) for WML voxels, NAWM-MS and HC voxels in four slices of one DMR, located within deep cortical white matter in posterior parietal lobes (left). B. Mapping of NAA/tCr ratios in single slice and multi-slice of 3 DMRs located in one axial brain slice of an MS patient, using spiral-MRSI based representation of data, overlayed with structural image.
  • Metabolic Impact of Spontaneous Trigeminal Allodynia in Sprague-Dawley Rats: Implications for Migraine & other Neurological Disorders
    Samuel W. Holder1,2, Michael Graham Harrington3, and Samuel Colles Grant1,2
    1National High Magnetic Field Laboratory, Florida State University, Tallahassee, FL, United States, 2Chemical & Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, FL, United States, 3Molecular Neurology Program, Huntington Medical Research Institutes, Pasadena, CA, United States
    Relaxation-enhanced MRS at 21.1 T evaluated spontaneous trigeminal allodynia (STA) in the female rat thalamus with and without nitroglycerin-induced sensitization. STA impacts the buildup of naïve Glx signal but is reversed with nitroglycerin. 
    Figure 3: Temporal Glx increases are observed in saline-injected STA animals, showing both between group and baseline significance. Baseline significance begins 75 min post-injection, slightly before behavioral migraine symptoms present in the nitroglycerin (NTG) group. NTG (10 mg/mg) was injected at t=0 min. Brackets indicate significance between groups (p<0.05); * is significance to baseline (p<0.05).
    Figure 2: Periorbital Von Frey mechanical sensitivity thresholds (force based on filament stiffness) for both (a) Naturally Resilient (NR) and (b) Spontaneous Trigeminal Allodynic (STA) rats
  • 1H MRS biomarkers in spinocerebellar ataxia type 1
    Kirsten Kapteijns1, Teije van Prooije1, Jack JA van Asten2, Bart van de Warrenburg1, and Tom WJ Scheenen2
    1Dept of Neurology, Radboudumc, Nijmegen, Netherlands, 2Dept of Medical Imaging, Radboudumc, Nijmegen, Netherlands
    Altered relative metabolite levels in tNAA, glutamate, and myo-Inositol could be replicated in SCA1 and correlated with clinical scores. This indicates consistency of these MRS biomarkers, which can be considered for future trials.
    Figure 2 Visualization of the significant differences in relative metaboliteslevels between patients with SCA1 and controls in the three VOIs
    Figure 3 Correlation between relative metabolite levels in the brain and clinical SARA scores. tNAA is significantly correlated in all three VOIs, glutamate is correlated with clinical scores in the cerebellar white matter.
  • Examining the relationship between glutathione and post-traumatic headache in patients with persistent post-concussive symptoms
    Julie M Joyce1,2,3,4, Leah J Mercier2,4,5, Parker L La1,2,3,4, Tiffany Bell1,2,3, Julia M Batycky2,4,5, Chantel T Debert2,3,4,5, and Ashley D Harris1,2,3,4
    1Department of Radiology, University of Calgary, Calgary, AB, Canada, 2Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada, 3Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada, 4Integrated Concussion Research Program, University of Calgary, Calgary, AB, Canada, 5Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
    Using edited spectroscopy, we found increased glutathione in the anterior cingulate is associated with greater functional impact of headache in patients with persistent post-concussive symptoms suggesting oxidative stress is related to post-traumatic headache.
    Figure 2. Raincloud plot depicting the GSH concentration distribution (mmol/l) in the anterior cingulate cortex of PPCS participants with intermittent (mean = 2.31 ± 0.41; n = 6) and constant headache (mean = 2.86 ± 0.61; n = 10) and matched, injury-free controls (mean = 2.36 ± 0.37; n = 13).
    Figure 3. Association between functional impact of headache and GSH concentration (mmol/l) in the anterior cingulate cortex. GSH in the anterior cingulate cortex was positively correlated with HIT-6 scores; (higher HIT-6 scores indicates greater functional impact of headache) r(16) = 0.507, p = 0.045*. HIT-6 score classifications: (1) little-to-no impact (score: 36-39), (2) moderate impact (score: 50-55), (3) substantial impact (score: 56-59), (4) severe impact (score 60-78).
  • 1H MR spectroscopic imaging for identifying diffuse abnormalities in mild traumatic brain injury: initial results from a reproducibility study
    Anna M Chen1, Teresa Gerhalter1, Seena Dehkharghani1,2, Rosemary Peralta1, Fatemeh Adlparvar1, Martin Gajdošík3, Mickael Tordjman1,4, Julia Zabludovsky1, Sulaiman Sheriff5, James S Babb1, Tamara Bushnik6, Jonathan M Silver7, Brian S Im6, Stephen P Wall8, Guillaume Madelin1, and Ivan I Kirov1,2,9
    1Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York, NY, United States, 2Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States, 3Department of Biomedical Engineering, Columbia University School of Engineering and Applied Science, New York, NY, United States, 4Department of Radiology, Cochin Hospital, Paris, France, 5Department of Radiology, University of Miami Miller School of Medicine, Miami, FL, United States, 6Department of Rehabilitation Medicine, New York University Grossman School of Medicine, New York, NY, United States, 7Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, United States, 8Ronald O. Perelman Department of Emergency Medicine, New York University Grossman School of Medicine, New York, NY, United States, 9Center for Advanced Imaging Innovation and Research, Department of Radiology, New York University Grossman School of Medicine, New York, NY, United States
    Lobar linear regression analysis yielded higher Cho, Cr and mI in WM in mTBI patients compared to controls. No group differences for NAA were found in WM, nor for any metabolites in GM. Associations were detected between metabolites and symptoms in WM, and between metabolites and cognition in GM.
    Figure 1: Representative spectroscopic map images for a single control subject. Presented is every fifth slice from the volumetric data for (A) creatine (Cr), (B) choline (Cho), (C) myo-inositol (mI), (D) glutamate plus glutamine (Glx), (E) N-acetyl aspartate (NAA), (F) grey and (G) white matter segmentation (GM, WM), (H) lobar parcellation, and (I) water (used as quantification reference) at the spectroscopic image spatial resolution.
    Figure 3: Boxplots of lobar metabolite distributions within mTBI and control (CTL) cohorts. Occipital WM Cho and Cr, parietal WM Cr, and frontal WM mI are higher in mTBI patients compared to controls (MW, ♦: p 0.05). Note that in all lobes WM Cr and Cho medians are higher in mTBI patients compared to controls. Cho: choline, Cr: creatine, mI: myo-inositol, Glx: glutamine plus glutamate; NAA: N-acetyl aspartate; WM: white matter.
Back to Top
Digital Poster Session - Spectroscopy: Neuro II
Cancer/Spectroscopy/Molecular Imaging/Pre-Clinical
Tuesday, 18 May 2021 17:00 - 18:00
  • Assessing the agreement between 3T and 7T MRS measures of in-vivo neurochemistry.
    Dana Goerzen1,2, Tiffany Bell3,4,5, Jamie Near1,2, and Ashley D Harris3,4,5
    1McGill University, Montreal, QC, Canada, 2Centre d'Imagerie Cérébrale, Douglas Mental Health University Institute, Montreal, QC, Canada, 3Radiology, University of Calgary, Calgary, AB, Canada, 4Hotchkiss Brain Institution, Calgary, AB, Canada, 5Alberta Children's Hospital Research Institute, Calgary, AB, Canada
    We present an analysis of MRS reliability in the human brain between 3T and 7T. We show that scyllo-inositol is reliably detected at 3T and some 7T. Further, we show that myo-inositol (mI) and phosphocholine (PCh) may be better quantified individually, rather than as mI+gly and tCho.
    Figure 1. On left: Representative 2.5 x 2.5 x 2.5 cm3 voxel in occipital-parietal sulcus overlaid onto 7T MPRAGE anatomical image indicating region of MRS acquisition. Image and voxel reconstruction was performed prior to the follow-up scan, to ensure consistent placement of MRS voxel within participants. On right: fits from participant S01 for the two 7T and six 3T spectra spectra acquired in the voxel of interest.
    Figure 3. Pearson’s correlation coefficient (r) computed for each metabolite between 3T sequences shown in the x-axis and 7T sequences in legend. These were then averaged across all metabolites within a 3T-7T pairing to obtain the sequence-wide average correlation between 3T measures and 7T measures of metabolites.
  • Quantification of GABA in ultra-short TE 7-T human MR spectra
    Guglielmo Genovese1, Dinesh K. Deelchand1, Melissa Terpstra1, and Malgorzata Marjanska1
    1Center for Magnetic Resonance Research (CMRR), Department of Radiology, University of Minnesota, Minneapolis, MN, United States

    - LCModel accurately quantifies alterations in [GABA] using measured macromolecules, stiff baseline and no concentration ratio priors.

    - Reducing SNR increased standard deviation across subjects.

    - The 10% [GABA] group differences could be observed with reasonable sample sizes.

    Figure 1. Overview of the synthetically altered spectra and their LCModel outputs for one representative subject. From top to bottom: synthetically altered spectra, fitted baselines, fitted GABA signals and residuals. The overlap of GABA resonances with other metabolite is shown with different colors. The alterations of the spectra caused by changes in GABA concentration are visible at 1.90 and 2.29 ppm whereas almost invisible at 3.00 ppm. The LCModel residuals do not show any structured signals for any of the spectra
    Figure 3. [GABA] values corresponding to the synthetically altered spectra (A) for different LCModel approaches and (B) for different number of averages are plotted as a function of the imposed change in GABA signal. The unity line between imposed change and measured concentration is also plotted. The results showed that the approach #1 (measured MM, stiff baseline and no concentration ratio priors) allows to correctly quantify the imposed change in GABA concentration. Reducing number of averages does not influence the mean value of [GABA] in a cohort but increase SD.
  • Improving the B0 homogeneity in 7 T MRSI applications using a 24-channel local array of shim coils
    Philipp Lazen1, Sahar Nassirpour2, Paul Chang2, Lukas Hingerl1, Karl Rössler3, Siegfried Trattnig1,4, Wolfgang Bogner1, and Gilbert Hangel1,3
    1Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria, 2MR Shim GmbH, Reutlingen, Germany, 3Department of Neurosurgery, Medical University of Vienna, Vienna, Austria, 4Christian Doppler Laboratory for Clinical Molecular MR Imaging, Vienna, Austria
    A 24-channel local shim coil array was tested for 7 T 3D MRSI in healthy volunteers. The B0 field strength’s standard deviation decreased by around 20% overall and signal to noise ratios and full widths at half maximum were improved. Some preliminary MRSI data was gathered.
    Figure 2: Representative B0 maps for both shim settings (once only with scanner shim of up to second order spherical harmonics and once with the 24-channel local shim array) are shown in three orientations overlaid on anatomical images for a single volunteer. B0 maps are plotted between [-100Hz,100Hz]. The standard deviation of the frequency shifts are reported under each corresponding B0 map.
    Figure 4: Representative map of glutamate and glutamine (Glu+Gln) for the standard scanner shim and the 24-channel shim, along with the corresponding B0 maps. Regions that suffer from poor B0 shimming quality with the scanner shim are marked by red arrows. The B0 maps are plotted between -60 Hz (blue) and 60 Hz (red) and the corresponding standard deviations are noted below each B0 map.
  • Comparison of Semi-LASER and Short-TE STEAM Pulse Sequences for Cerebral Glucose Quantification via Detecting H1-α-Glucose Peak in 1H MRS at 7 T
    Hideto Kuribayashi1, Yuta Urushibata1, Thuy Ha Duy Dinh2, Hirohiko Imai3, Sinyeob Ahn4, Ravi Teja Seethamraju5, Tadashi Isa2, and Tomohisa Okada2
    1Siemens Healthcare K.K., Tokyo, Japan, 2Human Brain Research Center, Kyoto University Graduate School of Medicine, Kyoto, Japan, 3Kyoto University Graduate School of Informatics, Kyoto, Japan, 4Siemens Medical Solutions, Berkeley, CA, United States, 5Siemens Medical Solutions, Boston, MA, United States
    H1-α-glucose peak at posterior cingulate cortex (27-mL volume) was detected with higher SNR (9.1 vs 5.1) using semi-LASER sequence (32-ms TE) than short-TE STEAM sequence (5-ms TE) at 7T in shorter scan time (11 vs 12.5 minutes), respectively.
    FIGURE 1. MR spectra acquired from all subjects and voxel position (3.0×3.0×3.0 (cm)3 on 3D MPRAGE T1-weighted images (TR/TI = 2300/1050 ms)). The spectra baseline-corrected on LCModel are stacked with SNR of the H1-α-glucose peak at 5.23 ppm in semi-LASER spectra from low (bottom) to high (top). The downfield spectra (5.0-5.4 ppm in the right panel) are scaled with 5-fold. In blue short-TE STEAM spectra, the glucose peak was not identified. In red spectra acquired from a subject, the glucose peak could not be fitted due to strong nuisance signals between the peaks of glucose and water.
    TABLE 1. Results from the spectral analysis.
  • 1H MRS of the Pediatric Brain Metabolism During Cardiopulmonary Bypass (CPB) Surgery
    Daniel Spielman1, Phil Adamson2, Ralph Hurd1, Meng Gu1, Kirk Riemer3, Ryan Beckman3, Michael Ma3, Kenichi Okamura3, and Frank Hanley3
    1Radiology, Stanford University, Stanford, CA, United States, 2Electrical Engineering, Stanford University, Stanford, CA, United States, 3Cardiothoracic Surgery, Stanford University, Stanford, CA, United States
    Acute brain metabolic changes were observed via 3T 1H MRS in a neonatal piglet model of cardiopulmonary bypass surgery, with the goal of finding optimal surgical parameters for minimizing brain injury. Findings include robust measures of brain temperature, hypoxia, and energy metabolism.
    Figure 1. 3T MRI scanner setup for neonatal pig DHCA-CPB study. A coronal T1-weighted MRI with the selected MRS voxel and representative spectra at baseline and end circulatory arrest (CA) time points are shown.
    Figure 4. Observation of NMR “powder pattern” from residual water signal from a brain voxel in a neonatal pig model under circulatory arrest. Dynamic spectra are plotted as a function of time post-circulatory arrest. A powder pattern is theoretically predicted from the intravascular water signal, based on a model of randomly oriented veins/capillaries containing deoxyhemoglobin [5].
  • Macromolecule suppressed GABA levels show no relationship with age in children and adolescents
    Tiffany Bell1,2,3, Mehak Stokoe1,2,3, and Ashley Harris1,2,3
    1Department of Radiology, University of Calgary, Calgary, AB, Canada, 2Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada, 3Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
    There were no significant correlations between age (7-14 years) and macromolecule-suppressed GABA levels in the thalamus, sensorimotor and occipital cortices when using either water (i.e., molal units) or creatine referencing.
    Figure 2:Correlations between the three versions of macromolecule-suppressed GABA levels and age. a) Correlation between macromolecule-suppressed GABA and age. b) Correlation between α-corrected macromolecule-suppressed GABA and age. c) Correlation between macromolecule-suppressed GABA/Cr and age. TH: Thalamus (red); SM: Sensorimotor Cortex (green); OC: Occipital Cortex (blue); MM Supp : macromolecule-suppressed.
    Table 1: Correlations between macromolecule-suppressed GABA measures and age (months)
  • Exploring Brain Metabolites in Pediatric Concussion: An A-CAP Study
    Parker L La1, Robyn Walker1, Julie M Joyce1, Tiffany Bell1, William Craig2, Quynh Doan3, Miriam Beauchamp4, Roger Zemek5, Pediatric Emergency Research Canada (PERC)6, Keith O Yeates7, and Ashley D Harris1
    1Radiology, University of Calgary, Calgary, AB, Canada, 2Pediatrics, University of Alberta and Stollery Children's Hospital, Edmonton, AB, Canada, 3Pediatrics, University of British Columbia, Vancouver, BC, Canada, 4Psychology, University of Montreal and St Justine Hospital, Montreal, QC, Canada, 5Pediatrics and Emergency Medicine, University of Ottawa and Children's Hospital of Eastern Ontario, Ottawa, ON, Canada, 6Alberta Children's Hospital, Calgary, AB, Canada, 7Psychology, University of Calgary, Calgary, AB, Canada
    In the largest magnetic resonance spectroscopy dataset in pediatric concussion study to date, no differences in metabolites were found between concussion and control groups, and only Glx was related to cognitive symptoms.
    Figure 2: Boxplot showing the average concentration of tNAA, tCr, tCho, and Glx in both concussion and OI groups. There are no significant differences between concussion and OI groups in the aforementioned metabolites utilizing multiple ANCOVAs while controlling for age, sex, and site. Circles outside of the box plots are outside the 1.5 standard deviations interquartile range, and the stars are 3 standard deviations outside the interquartile range. All outliers meet all data quality criteria.
    Figure 1: Example spectrum of a pediatric participant with concussion. This 1H-MRS PRESS data is from a 2x2x2 cm3 voxel in the left dorsolateral prefrontal cortex, shown in the voxel placements on the anatomical brain scan, to measure tNAA, tCr, tCho, and Glx. The PRESS sequence had a TE of 30ms and a TR of 2000ms, with 96 averages.
  • Measurement of local values of cerebral pH in mild traumatic brain injury using 1H MR spectroscopy
    Peter Bulanov1, Andrei Manzhurtsev1,2, Petr Menshchikov3,4, Maxim Ublinskiy2,3, Ilya Melnikov2, Natalia Semenova1,2,3,5, and Tolib Akhadov2
    1Lomonosov Moscow State University, Moscow, Russian Federation, 2Clinical and Research Institute of Emergency Pediatric Surgery and Traumatology, Moscow, Russian Federation, 3Emanuel Institute of Biochemical Physics of RAS, Moscow, Russian Federation, 4PHilips Healthcare, Russia, Moscow, Russian Federation, 5Semenov Institute of Chemical Physics of the Russian Academy of Sciences, Moscow, Russian Federation
    The pH value decreases in posterior cingulate cortex after acute mTBI by ~1%. The detected change may cause biochemical disorders in brain after mTBI, associated with a change in the pH-dependent activity of enzymes.
    Fig.2. Overview of the signals averaged over groups (NORM and mTBI) at 7 ppm and approximation of the signals.
    Fig.1. Voxel location: posterior cingulate cortex. Size 50 x 25 x 25 mm.
  • The Correlation of Neurometabolites on Presentation of Post-Concussion Phenotypes
    Katherine Breedlove1, David R Howell2, Inga K Koerte3,4, Eduardo Coello4, Molly Charney4, Huijin Liao4, Corey Lanois5, and Alexander Lin4
    1Radiology, Brigham and Women's Hospital, Boston, MA, United States, 2Childrens Hospital Colorado, Aurora, CO, United States, 3Ludwig-Maximilians-Universitat, Munich, Germany, 4Brigham and Women's Hospital, Boston, MA, United States, 5Boston Children's Hospital, Boston, MA, United States
    PCSS composite scores were found to significantly correlate with neurometabolite levels in the PCG.  Specifically, cognitive, vestibuolocular, cognitive, and sleep related scores were significantly correlated with concentrations of Glu, Glu+Gln, and NAA+NAAG.   
    Fig. 5. Regression plots showing fit to PCSS composite score vs. metabolite concentration. Regression p-value and R2 are indicated.
    Fig. 3. Table of significant regressions between neurometabolites and PCSS composite scores. Empty cells indicate that no significant regression was found. The p-value of the overall regression F-statistic is reported along with R2. VO=Vestibular-ocular
  • Hypoxia alters posterior cingulate cortex metabolism during a memory task: a 1H fMRS study
    Matthew Rogan1,2,3, Joseph B R Smith1,2,3, Alexander Friend3,4, Jamie H Macdonald3,4, Sam J Oliver3,4, Gabriella M K Rossetti5, Mark Mikkelsen6,7, Richard A E Edden6,7, and Paul G Mullins1,2,3
    1School of Psychology, Bangor University, Bangor, United Kingdom, 2The Bangor Imaging Unit, Bangor University, Bangor, United Kingdom, 3The Extremes Research Group, Bangor University, Bangor, United Kingdom, 4School of Sport Health and Exercise Sciences, Bangor University, Bangor, United Kingdom, 5Centre for Integrative Neuroscience and Neurodynamics, University of Reading, Reading, United Kingdom, 6Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, United States, 7F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States
    Exposure to environmental hypoxia resulted in a loss of the task induced increase in glutamate within the posterior cingulate cortex (PCC) and reduction in glucose concentration. This suggests hypoxia disrupts PCC oxidative metabolism. 
    Figure 1. Mean voxel location across participants and conditions (top left). Example spectrum (bottom left). Graph representing glutamate ANOVA results. Bars represent the group mean for within condition rest (light blue) to recall (dark blue). Individual connected dots and lines represent within participant rest to recall change (right).
  • Photoperiodic Regulation of hypothalamic metabolism : a preliminary single voxel Magnetic Resonance Spectroscopy investigation at 3T
    Nathalie Just1, Pierre-Marie Chevillard1, and Martine Migaud1
    1NhyRVana, INRAE, Nouzilly, France
    Photoperiodism regulated the metabolism of the female sheep hypothalamus as demontrated by significant changes of Glu, Gln and NAA during short days compared to long days. The sheep brain is an interesting model to explore hypothalamic mechanisms of metabolic regulation.
    Comparison of Neurochemical profiles (µmol/g) acquired within the hypothalamus of sheep at 4 time points (P01, P02, P03 and P04) during long days (Long period, LP) and at 4 time points during short days (Short period, SP). * p< 0.05; ** p<= 0.01

    A. Neurochemical profiles (µmol/g) during Long days (LP) at 4 time points. * p<0.05

    B. Neurochemical Profile (µmol/g) during Short days (SP) at 4 time points. * p<0.05 Gln and Glu concentrations at P01 were significantly different compared to concentrations at later time points

  • Hippocampal Single-Voxel MR Spectroscopy with Long Echo Time at 3 Tesla
    Martin Gajdošík1,2, Karl Landheer1, Kelley M. Swanberg1, Fatemeh Adlparvar2, Guillaume Madelin2, Wolfgang Bogner3, Christoph Juchem1,4, and Ivan I. Kirov2,5,6
    1Department of Biomedical Engineering, Columbia University, New York City, NY, United States, 2Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University Grossman School of Medicine, New York City, NY, United States, 3High-Field MR Center, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria, 4Department of Radiology, Columbia University Medical Center, New York City, NY, United States, 5Department of Neurology, New York University Grossman School of Medicine, New York City, NY, United States, 6Center for Advanced Imaging Innovation and Research, Department of Radiology, New York University Grossman School of Medicine, New York City, NY, United States
    Hippocampal proton MR spectroscopy was investigated for the feasibility and variability of metabolite concentrations at long TE of 120 ms, using sLASER localization.
    Figure 3: MRS acquisition and spectral analysis. (A) Placement of the 3.4 mL voxel (26 x 10 x 13 mm3) in the left hippocampus (52 years, woman), containing mainly its body and tail. (B) Spectrum from the hippocampus fitted with linear combination modelling. Note, no macromolecular background in the residual signal. (C) Basis set used for quantification of hippocampal metabolites.
    Figure 1: Simplified simulations of T2 relaxation of normalized signals for macromolecules (MM) and metabolites. The average T2 relaxation times and ranges of macromolecules and metabolites were calculated from literature3,14,15. Note that at TE = 120 ms, most MM signal is gone, while on average 60% of the metabolite signal remains. This demonstrates that relatively little metabolite signal was lost while the specific choice of TE = 120 ms enabled easily quantifiable Glu+Gln (Glx)) and m-Ins resonances.
  • Protective role of Creatine in chronic hepatic encephalopathy developing brain: in vivo longitudinal 1H and 31P-MRS study
    Dunja Simicic1,2,3, Katarzyna Pierzchala1,2, Olivier Braissant4, Stefanita-Octavian Mitrea1,2, Dario Sessa5, Valerie McLin5, and Cristina Cudalbu1,2
    1CIBM Center for Biomedical Imaging, Lausanne, Switzerland, 2Animal Imaging and Technology, EPFL, Lausanne, Switzerland, 3Laboratory of Fuctional and Metabolic Imaging, EPFL, Lausanne, Switzerland, 4Service of Clinical Chemistry, University of Lausanne and University Hospital of Lausanne, Lausanne, Switzerland, 5Swiss Center for Liver Disease in Children, University Hospitals Geneva, Geneva, Switzerland
    Type-C hepatic encephalopathy is a complication of chronic liver disease. It is known that children are more affected by CLD than adult patients. Our aim was to test whether Cr-supplementation dampens the changes observed in CHE in a longitudinal model of CLD acquired in early childhood P15.
    In vivo 31P-MR spectra from whole brain (VOI=5x9x9mm3) acquired at week 6 after BDL or sham surgery, each spectrum is from one animal.
    In vivo 1H-MR spectra from hippocampus (VOI=2x2.8x2mm3) acquired at week 6 after BDL or sham surgery, each spectrum is from one animal. Spectra clearly show increased tCr in both BDL+Cr and Sham+Cr. The positive effect of Cr treatment on other osmolytes (Ins, Tau and tCho) is also visible on BDL+Cr spectrum.
  • Fast lipid removal with multi-scale auto-alignment in full-FOV MRSI.
    Peter Adany1, In-Young Choi1,2, and Phil Lee1,3
    1Hoglund Biomedical Imaging Center, University of Kansas Medical Center, Kansas City, KS, United States, 2Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States, 3Department of Radiology, University of Kansas Medical Center, Kansas City, KS, United States
    Alignment optimization at 10 iterations/sec improves lipid signal removal and better quantitation in full-field of view MRSI metabolic imaging of the brain.
    Figure 4A. MRSI spectra from a subject with some misalignment between MRI and MRSI. (A) The approximate location of the lipid mask indicated with green voxels. Lipid signal strength is indicated by an orange color map. Without lipid auto-alignment, greater and asymmetric residual lipid signal are clearly visible (4A left). After lipid auto-alignment, the lipid removal is highly symmetric and the lipid residuals are smaller (4A right). The estimated displacement was 4.9 mm and 2.5 mm in X and Y directions, respectively. Spectral range is [0, 4.2] ppm.
    Figure 2. Results of spatial auto-alignment in 141 scans. Auto-alignment was performed on multiple scans of 73 healthy subjects. Red circles indicate positions outside a 5mm boundary (defined as Large Displacement). Results showed that coronal head movement was greater than sagittal during scan sessions, consistent with positioning of the head within the RF coil in supine position. The alignment precision exceeded the MRSI nominal voxel size of 20x20x25 mm3. Multi-scale spatial alignment used 7x7 search grids (40.0mm, 13.5mm, 4.5mm, and 1.5mm) to efficiently locate the minimum.
  • On the repeatability and reproducibility of SPECIAL-based in-vivo spectroscopy with different adiabatic inversion pulses
    Layla Tabea Riemann1, Christoph Stefan Aigner1, Ralf Mekle2, Sebastian Schmitter1, Bernd Ittermann1, and Ariane Fillmer1
    1Physikalisch-Technische Bundesanstalt (PTB), Braunschweig und Berlin, Germany, 2Center for Stroke Research Berlin, Charité Universitätsmedizin, Berlin, Germany
    This work shows improved repeatability and smaller CRLBs employing gradient-modulated adiabatic inversion pulses compared to the hyperbolic secant pulse that was originally used in the SPECIAL sequence for in-vivo 1H brain MRS at 7T.
    Fig.2: a) Measured spectra between 0.0 and 4.2 ppm for all pulses from session 1_1 for subject 2 b) Bland-Altman plots for HS (blue), GOIA (orange), WURST (green) for R0 (top), R1,M (middle), and R1,W (bottom) to visualize repeatability (R0) and reproducibility (R1,M and R1,W). One point is generated by using the two equations in the main text. The SD indicates the measure of repeatability/reproducibility and is, averaged over all pulses: 0.016, 0.039, and 0.064 for R0, R1,M and R1,W, respectively.
    Fig.1: a) Pulse diagram of SPECIAL with the HS (blue), GOIA (orange), and WURST (green) pulse; b) Scan scheme (exemplary): 1. day 1. session SPECIAL with HS, GOIA, and WURST; After repositioning the volunteer (1_2), scans were repeated as in 1_1. 2. day scans (i.e., one week later) 1. session, HS, and GOIA twice without repositioning. 2. session 2. day, WURST-SPECIAL twice without repositioning. c) Exemplary voxel position in the posterior cingulate cortex (PCC) of a volunteer.
  • Polarity Dependent Modulation of the Motor Region Using tDCS: A Proton MR Spectroscopy Study
    Rajakumar Nagarajan1, Anant Shinde2,3, Muhammed Enes Gunduz2,3, and Gottfried Schlaug2,3
    1Human Magnetic Resonance Center, Institute for Applied Life Sciences, University of Massachusetts Amherst, Amherst, MA, United States, 2Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, United States, 3Baystate Medical Center, University of Massachusetts Medical School, Springfield, MA, United States
    We studied the effects of motor region tDCS on metabolite levels in a spectroscopic voxel with repeated  5min of constant current dose. This study provides insights into how tDCS leads to polarity dependent modulation of metabolites and might provide a model for future MRS stimulation studies. 
    Figure 1. Experimental setup: a) electrode placement and stimulator modes; b) setup for applying tDCS inside the MR scanner; and c) 45 minutes of tDCS-MRS scanning protocol timing diagram
    Figure 3. Short term and Cumulative effect of GABA and Glx with respect to Creatine changes of Anode and Cathode
  • A time course of the glutamate+glutamine level in response to a short visual stimulus
    Alexey Yakovlev1,2,3, Andrei Manzhurtsev1,3, Petr Menshchikov3,4, Maxim Ublinskiy3, Tolib Akhadov1, and Natalia Semenova1,2,3
    1Clinical and Research Institute of Emergency Pediatric Surgery and Trauma, Moscow, Russian Federation, 2N.N.Semenov Federal research center of Chemical Physics Russian Academy of Sciences, Moscow, Russian Federation, 3N.M. Emanuel Institute of Biochemical Physics RAS, Moscow, Russian Federation, 4Philips Healthcare, Moscow, Russian Federation

    We assessed a time for glutamate neurotransmitter release, reuptake, and refilling in vesicles.  Early growth of Glx doesn't correlate with the maximum of BOLD response. For the first time, we investigated the dynamics of the BOLD-effect using the metabolic signal.

    Figure 2. The paradigm of the visual stimulation: 3 s - a checkerboard flickering with a frequency of 4 Hz and 21 s - dark screen; scheme of fMRI and 1H MRS data acquisition during stimulus presentation.
    Figure 1.T1-weighted image slices showing the voxel position for the single voxel spectroscopy in the activated visual cortex
  • Ultra-high field MR spectroscopic imaging at 7 Tesla in Multiple Sclerosis: myo-Inositol as early biomarker for MS pathologies
    Alexandra Lipka1,2, Eva Heckova1, Assunta Dal-Bianco3, Gilbert Hangel1, Bernhard Strasser1, Stanislav Motyka1, Lukas Hingerl1, Paulus Rommer3, Fritz Leutmezer3, Petra Hnilicová4, Ema Kantorová4, Stephan Gruber1, Siegfried Trattnig1,2, and Wolfgang Bogner1,2
    1High Field MR Centre, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria, 2Christian Doppler Laboratory for Clinical Molecular MR Imaging, Vienna, Austria, 3Department of Neurology, Medical University of Vienna, Vienna, Austria, 4Jessenius Faculty of Medicine in Martin, Comenius University, Bratislava, Slovakia
    Free Induction Decay - Magnetic Resonance Spectroscopic Imaging (FID-MRSI) enables a comprehensive biochemical characterization of Multiple Sclerosis (MS) lesions and strengthens the role of mIns/tNAA as an imaging biomarker for MS pathologies.
    Figure 1: Boxplot diagram for mIns/tNAA, mIns/tCr, tNAA/tCr and tCho/tCr and the different lesion categories “NWM”, “NAWM”, “nonBH”, “BH”, and “MRSI hotspot”. No differences between NWM and NAWM for each of the metabolites. Highly significant differences between lesion categories especially for mIns/tNAA
    Figure 2: Conventional MRI with T1-weighted MP2RAGE and T2-weighted FLAIR. Yellow arrows point to “black hole” lesions which are clearly visible on mIns/tNAA, mIns/tCr and tNAA/tCr. The red arrow depicts a “non black hole” lesion, which is already apparent on mIns/tNAA and mIns/tCr, though not yet visible on FLAIR
  • Ultra-high field MR spectroscopic imaging at 7 Tesla in Multiple Sclerosis: metabolic fingerprinting of iron rim lesions
    Alexandra Lipka1,2, Wolfgang Bogner1,2, Assunta Dal-Bianco3, Gilbert Hangel1, Bernhard Strasser1, Stanislav Motyka1, Lukas Hingerl1, Paulus Rommer3, Fritz Leutmezer3, Stephan Gruber1, Siegfried Trattnig1,2, and Eva Heckova1
    1High Field MR Centre, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria, 2Christian Doppler Laboratory for Clinical Molecular MR Imaging, Vienna, Austria, 3Department of Neurology, Medical University of Vienna, Vienna, Austria
    Free Induction Decay(FID)-MRSI at 7T shows metabolic changes associated with different types of iron accumulation and a metabolic gradient spanning from within the iron rim lesion to its close proximity
    Figure 1: T2-FLAIR, SWI and overlaid FLAIR-SWI images of(a) lesions categorized as “transition” with yellow arrow pointing towards an example, (b) “area” iron deposition indicated by a red arrow and (c) “rim” lesion with an abundant iron rim indicated by a green arrow.
    Figure 2: Boxplot diagram for mIns/tNAA, mIns/tCr, tNAA/tCr and tCho/tCr and the different lesion categories “no iron”, “area”, “transition” and “rim”. Significant results were found between lesion categories for mIns/tNAA and tNAA/tCr, but mIns/tCr stays constantly elevated for all “black hole” lesions.
  • Compression of Multivoxel Spectroscopic Data via Visualization of Grey and White Matter Contributions to Metabolite Concentrations
    Wufan Zhao1, Eduardo Coello1, Marcia Sahaya Louis1, Katherine Breedlove1, Assaf Tal2, and Alexander Lin1
    1Radiology, Brigham and Women's Hospital, Boston, MA, United States, 2Chemical and Biological Physics, Weizmann Institute of Science, Rehovot, Israel
    This work introduces a novel way to analyze MR spectroscopic imaging scans by characterizing grey matter and white matter tissue types based on their contributions to metabolite concentrations. The method detected and visualized different metabolic profiles between grey and white matter.
    Fig. 1. Maps of GM, WM, and CSF partial volumes of a single subject. (A) High resolution reconstructed images obtained from 60x60 matrices of grey matter, white matter, and cerebrospinal fluid partial volume fractions. (B) Partial volume maps with 16x16 grid overlay composed of 10x10x10 mm voxels. (C) Lower resolution reconstructed images from 8x8 matrices of the three partial fractions. Central 8x8 grids are in bold. A color scale of blue to yellow is used, with blue being lower and yellow being greater partial volume fraction at each voxel. Partial fractions were obtained via VDI.
    Fig. 2. Scatterplots of metabolite ratios to creatine (y-axis) against partial volume fractions of GM or WM (x-axis). GM is on the top row and WM is on the bottom row. The plots include a linear fit and report R-squared values. The intercepts of the linear fit where partial fractions are equal to 0.0 and 1.0 are shown by red markers (GM0% and GM100% for GM, and WM0% and WM100% for WM). Metabolites include tCho, tNAA, Glx, and mI. High correlations are observed with metabolites that are known to have different concentrations in grey and white matter.