1
|
Altmann S, Grauhan NF, Mercado MAA, Steinmetz S, Kronfeld A, Paul R, Benkert T, Uphaus T, Groppa S, Winter Y, Brockmann MA, Othman AE. Deep Learning Accelerated Brain Diffusion-Weighted MRI with Super Resolution Processing. Acad Radiol 2024; 31:4171-4182. [PMID: 38521612 DOI: 10.1016/j.acra.2024.02.049] [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: 12/31/2023] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 03/25/2024]
Abstract
OBJECTIVES To investigate the clinical feasibility and image quality of accelerated brain diffusion-weighted imaging (DWI) with deep learning image reconstruction and super resolution. METHODS 85 consecutive patients with clinically indicated MRI at a 3 T scanner were prospectively included. Conventional diffusion-weighted data (c-DWI) with four averages were obtained. Reconstructions of one and two averages, as well as deep learning diffusion-weighted imaging (DL-DWI), were accomplished. Three experienced readers evaluated the acquired data using a 5-point Likert scale regarding overall image quality, overall contrast, diagnostic confidence, occurrence of artefacts and evaluation of the central region, basal ganglia, brainstem, and cerebellum. To assess interrater agreement, Fleiss' kappa (ϰ) was determined. Signal intensity (SI) levels for basal ganglia and the central region were estimated via automated segmentation, and SI values of detected pathologies were measured. RESULTS Intracranial pathologies were identified in 35 patients. DL-DWI was significantly superior for all defined parameters, independently from applied averages (p-value <0.001). Optimum image quality was achieved with DL-DWI by utilizing a single average (p-value <0.001), demonstrating very good (80.9%) to excellent image quality (14.5%) in nearly all cases, compared to 12.5% with very good and 0% with excellent image quality for c-MRI (p-value <0.001). Comparable results could be shown for diagnostic confidence. Inter-rater Fleiss' Kappa demonstrated moderate to substantial agreement for virtually all defined parameters, with good accordance, particularly for the assessment of pathologies (p = 0.74). Regarding SI values, no significant difference was found. CONCLUSION Ultra-fast diffusion-weighted imaging with super resolution is feasible, resulting in highly accelerated brain imaging while increasing diagnostic image quality.
Collapse
Affiliation(s)
- Sebastian Altmann
- Department of Neuroradiology, University Medical Center Mainz, Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany.
| | - Nils F Grauhan
- Department of Neuroradiology, University Medical Center Mainz, Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Mario Alberto Abello Mercado
- Department of Neuroradiology, University Medical Center Mainz, Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Sebastian Steinmetz
- Department of Neuroradiology, University Medical Center Mainz, Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Andrea Kronfeld
- Department of Neuroradiology, University Medical Center Mainz, Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Roman Paul
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center Mainz, Johannes Gutenberg University, Rhabanusstr. 3/Tower A, 55118 Mainz, Germany
| | | | - Timo Uphaus
- Department of Neurology, University Medical Center Mainz, Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Sergiu Groppa
- Department of Neurology, University Medical Center Mainz, Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Yaroslav Winter
- Department of Neurology, University Medical Center Mainz, Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany; Department of Neurology, Philipps-University Marburg, Baldingerstr, 35043 Marburg, Germany
| | - Marc A Brockmann
- Department of Neuroradiology, University Medical Center Mainz, Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Ahmed E Othman
- Department of Neuroradiology, University Medical Center Mainz, Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany
| |
Collapse
|
2
|
Thomas DC, Oros-Peusquens AM, Schöneck M, Willuweit A, Abbas Z, Zimmermann M, Felder J, Celik A, Shah NJ. In Vivo Measurement of Rat Brain Water Content at 9.4 T MR Using Super-Resolution Reconstruction: Validation With Ex Vivo Experiments. J Magn Reson Imaging 2024; 60:161-172. [PMID: 37855368 DOI: 10.1002/jmri.29061] [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: 05/25/2022] [Revised: 09/28/2023] [Accepted: 09/29/2023] [Indexed: 10/20/2023] Open
Abstract
BACKGROUND Given that changes in brain water content are often correlated with disease, investigating water content non-invasively and in vivo could lead to a better understanding of the pathogenesis of several neurologic diseases. PURPOSE To adapt a super-resolution-based technique, previously developed for humans, to the rat brain and report in vivo high-resolution (HR) water content maps in comparison with ex vivo wet/dry methods. STUDY TYPE Prospective. ANIMAL MODEL Eight healthy male Wistar rats. FIELD STRENGTH/SEQUENCE 9.4-T, multi-echo gradient-echo (mGRE) sequence. ASSESSMENT Using super-resolution reconstruction (SRR), a HR mGRE image (200 μm isotropic) was reconstructed from three low-resolution (LR) orthogonal whole-brain images in each animal, which was followed by water content mapping in vivo. The animals were subsequently sacrificed, the brains excised and divided into five regions (front left, front right, middle left, middle right, and cerebellum-brainstem regions), and the water content was measured ex vivo using wet/dry measurements as the reference standard. The water content values of the in vivo and ex vivo methods were then compared for the whole brain and also for the different regions separately. STATISTICAL TESTS Friedman's non-parametric test was used to test difference between the five regions, and Pearson's correlation coefficient was used for correlation between in vivo and ex vivo measurements. A P-value <0.05 was considered statistically significant. RESULTS Water content values derived from in vivo MR measurements showed strong correlations with water content measured ex vivo at a regional level (r = 0.902). Different brain regions showed significantly different water content values. Water content values were highest in the frontal brain, followed by the midbrain, and lowest in the cerebellum and brainstem regions. DATA CONCLUSION An in vivo technique to achieve HR isotropic water content maps in the rat brain using SRR was adopted in this study. The MRI-derived water content values obtained using the technique showed strong correlations with water content values obtained using ex vivo wet/dry methods. LEVEL OF EVIDENCE 1 TECHNICAL EFFICACY: Stage 1.
Collapse
Affiliation(s)
- Dennis C Thomas
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich, Jülich, Germany
- Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | | | - Michael Schöneck
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich, Jülich, Germany
| | - Antje Willuweit
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich, Jülich, Germany
| | - Zaheer Abbas
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich, Jülich, Germany
| | - Markus Zimmermann
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich, Jülich, Germany
| | - Jörg Felder
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich, Jülich, Germany
- RWTH Aachen University, Aachen, Germany
| | - Avdo Celik
- Institute of Neuroscience and Medicine 11, INM-11, JARA, Forschungszentrum Jülich, Jülich, Germany
| | - Nadim Joni Shah
- Institute of Neuroscience and Medicine 4, INM-4, Forschungszentrum Jülich, Jülich, Germany
- Institute of Neuroscience and Medicine 11, INM-11, JARA, Forschungszentrum Jülich, Jülich, Germany
- JARA-BRAIN-Translational Medicine, Aachen, Germany
- Department of Neurology, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
3
|
Ishizuka K, Saito M, Shibata N, Kitagawa K. Cytoskeletal protein breakdown and serum albumin extravasation in MRI DWI-T2WI mismatch area in acute murine cerebral ischemia. Neurosci Res 2023; 190:85-91. [PMID: 36375655 DOI: 10.1016/j.neures.2022.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 10/12/2022] [Accepted: 11/10/2022] [Indexed: 11/13/2022]
Abstract
MRI diffusion-weighted imaging (DWI)-FLAIR mismatch is known as predictive of symptom onset within 4.5 h. This study assessed the breakdown of cytoskeletal protein and blood-brain barrier (BBB) in DWI-T2 mismatch. We employed occlusion of middle cerebral artery (MCAO) in C57BL/6 mice. We serially measured MRI including DWI and T2WI. After MRI, we prepared brain sections or samples and examined microtubule-associated protein 2 (MAP2) expression, alpha-fodrin degradation, extravasation of albumin and claudin-5 expression. In permanent or transient MCAO for 45 min, DWI hyperintensities was already found at 60 min without change of T2, showing DWI-T2 mismatch. In permanent MCAO, MAP2 expressions were preserved, and no extravasation of albumin was observed. In transient MCAO, MAP2 immunoreaction was already lost in the lateral part of the striatum. In both models, alpha-fodrin degradation was already detected. At 180 min, T2 hyperintensities appeared, where MAP2 signal was lost and albumin extravasation was found. At 24 h, hyperintensities of DWI and T2WI was found in the whole MCA territory, where MAP2 signal was completely lost with marked albumin extravasation and alpha-fodrin degradation. Immunoreaction for claudin-5 was preserved up to 180 min. DWI-T2 mismatch area may not always indicate intactness of cytoskeletal protein but shows preservation of BBB.
Collapse
Affiliation(s)
- Kentaro Ishizuka
- Department of Neurology, Tokyo Women's Medical University, Tokyo, Japan
| | - Moeko Saito
- Department of Neurology, Tokyo Women's Medical University, Tokyo, Japan
| | - Noriyuki Shibata
- Department of Pathology, Tokyo Women's Medical University, Tokyo, Japan
| | - Kazuo Kitagawa
- Department of Neurology, Tokyo Women's Medical University, Tokyo, Japan.
| |
Collapse
|
4
|
Ji Y, Lu D, Sun PZ, Zhou IY. In vivo pH mapping with omega plot-based quantitative chemical exchange saturation transfer MRI. Magn Reson Med 2023; 89:299-307. [PMID: 36089834 PMCID: PMC9617761 DOI: 10.1002/mrm.29444] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/05/2022] [Accepted: 08/15/2022] [Indexed: 02/01/2023]
Abstract
PURPOSE Chemical exchange saturation transfer (CEST) MRI is promising for detecting dilute metabolites and microenvironment properties, which has been increasingly adopted in imaging disorders such as acute stroke and cancer. However, in vivo CEST MRI quantification remains challenging because routine asymmetry analysis (MTRasym ) or Lorentzian decoupling measures a combined effect of the labile proton concentration and its exchange rate. Therefore, our study aimed to quantify amide proton concentration and exchange rate independently in a cardiac arrest-induced global ischemia rat model. METHODS The amide proton CEST (APT) effect was decoupled from tissue water, macromolecular magnetization transfer, nuclear Overhauser enhancement, guanidinium, and amine protons using the image downsampling expedited adaptive least-squares (IDEAL) fitting algorithm on Z-spectra obtained under multiple RF saturation power levels, before and after global ischemia. Omega plot analysis was applied to determine amide proton concentration and exchange rate simultaneously. RESULTS Global ischemia induces a significant APT signal drop from intact tissue. Using the modified omega plot analysis, we found that the amide proton exchange rate decreased from 29.6 ± 5.6 to 12.1 ± 1.3 s-1 (P < 0.001), whereas the amide proton concentration showed little change (0.241 ± 0.035% vs. 0.202 ± 0.034%, P = 0.074) following global ischemia. CONCLUSION Our study determined the labile proton concentration and exchange rate underlying the in vivo APT MRI. The significant change in the exchange rate, but not the concentration of amide proton demonstrated that the pH effect dominates the APT contrast during tissue ischemia.
Collapse
Affiliation(s)
- Yang Ji
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
- Wellcome Centre for Integrative Neuroimaging, FMRIB Division, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Dongshuang Lu
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Phillip Zhe Sun
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
- Emory Primate Imaging Center, Emory Primate Research Center, Emory University, Atlanta, GA, USA
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Iris Y. Zhou
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
5
|
Annus Á, Gera FZ, Sztriha L, Klivényi P. DWI-FLAIR mismatch guided thrombolysis in patients without large-vessel occlusion: real-world data from a comprehensive stroke centre. Heliyon 2022; 8:e12069. [PMID: 36506404 PMCID: PMC9730128 DOI: 10.1016/j.heliyon.2022.e12069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 09/27/2022] [Accepted: 11/25/2022] [Indexed: 12/12/2022] Open
Abstract
Introduction A significant proportion of ischaemic stroke patients present with unknown symptom onset time. DWI-FLAIR mismatch on MRI can help to identify those eligible for thrombolysis. We set out to analyse the short-term efficacy and safety of thrombolysis in a real-world setting. Methods A retrospective single-centre observational study was conducted. We collected data between January 2017 and April 2020. Patients with a large vessel occlusion (LVO) were excluded. Outcomes were compared between thrombolysed patients and those who did not receive alteplase due to lack of DWI-FLAIR mismatch or other contraindications. We analysed baseline and discharge NIHSS scores for efficacy and defined good outcome as any neurological improvement (ANI) on the NIHSS. In terms of safety, the presence and severity of intracerebral haemorrhage on follow-up imaging was analysed, and mortality at 90 days assessed. Results Seventy-one patients were included in this study, of whom 29 received thrombolysis. Significantly more patients had ANI in the thrombolysed group (OR, 3.16; 95% CI, 1.178-8.479; p = 0.020). In a multivariable logistic regression analysis, only thrombolysis correlated with ANI (OR, 3.051; 95% CI, 1.135-8.206; p = 0.027). Two thrombolysed patients suffered intracerebral haemorrhage (6.90%), of whom one was symptomatic and eventually fatal. We did not find a significant difference in 90-day mortality between the two groups (OR, 0.81, 95% CI, 0.134-4.856; p = 1.000). Conclusions Our real-world data demonstrate that thrombolysis based on DWI-FLAIR mismatch in patients without LVO has an early beneficial effect. The rate of intracerebral haemorrhage was similar to this complication reported in large thrombolysis trials with known onset times.
Collapse
Affiliation(s)
- Ádám Annus
- University of Szeged, Albert Szent-Györgyi Health Centre, Department of Neurology, Semmelweis u. 6, H-6725 Szeged, Hungary
| | - Franciska Zita Gera
- University of Szeged, Albert Szent-Györgyi Health Centre, Department of Neurology, Semmelweis u. 6, H-6725 Szeged, Hungary
| | - László Sztriha
- Department of Neurology, King’s College Hospital, Denmark Hill, SE5 9RS London, UK
| | - Péter Klivényi
- University of Szeged, Albert Szent-Györgyi Health Centre, Department of Neurology, Semmelweis u. 6, H-6725 Szeged, Hungary,Corresponding author.
| |
Collapse
|
6
|
Khoonkari M, Liang D, Kamperman M, Kruyt FAE, van Rijn P. Physics of Brain Cancer: Multiscale Alterations of Glioblastoma Cells under Extracellular Matrix Stiffening. Pharmaceutics 2022; 14:pharmaceutics14051031. [PMID: 35631616 PMCID: PMC9145282 DOI: 10.3390/pharmaceutics14051031] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/27/2022] [Accepted: 05/06/2022] [Indexed: 12/12/2022] Open
Abstract
The biology and physics underlying glioblastoma is not yet completely understood, resulting in the limited efficacy of current clinical therapy. Recent studies have indicated the importance of mechanical stress on the development and malignancy of cancer. Various types of mechanical stress activate adaptive tumor cell responses that include alterations in the extracellular matrix (ECM) which have an impact on tumor malignancy. In this review, we describe and discuss the current knowledge of the effects of ECM alterations and mechanical stress on GBM aggressiveness. Gradual changes in the brain ECM have been connected to the biological and physical alterations of GBM cells. For example, increased expression of several ECM components such as glycosaminoglycans (GAGs), hyaluronic acid (HA), proteoglycans and fibrous proteins result in stiffening of the brain ECM, which alters inter- and intracellular signaling activity. Several mechanosensing signaling pathways have been identified that orchestrate adaptive responses, such as Hippo/YAP, CD44, and actin skeleton signaling, which remodel the cytoskeleton and affect cellular properties such as cell–cell/ECM interactions, growth, and migration/invasion of GBM cells. In vitro, hydrogels are used as a model to mimic the stiffening of the brain ECM and reconstruct its mechanics, which we also discuss. Overall, we provide an overview of the tumor microenvironmental landscape of GBM with a focus on ECM stiffening and its associated adaptive cellular signaling pathways and their possible therapeutic exploitation.
Collapse
Affiliation(s)
- Mohammad Khoonkari
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (M.K.); (D.L.)
- Polymer Science, Zernike Institute for Advanced Materials, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands;
| | - Dong Liang
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (M.K.); (D.L.)
| | - Marleen Kamperman
- Polymer Science, Zernike Institute for Advanced Materials, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands;
| | - Frank A. E. Kruyt
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (M.K.); (D.L.)
- Correspondence: (F.A.E.K.); (P.v.R.)
| | - Patrick van Rijn
- Department of Biomedical Engineering-FB40, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials Science-FB41, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
- Correspondence: (F.A.E.K.); (P.v.R.)
| |
Collapse
|
7
|
Khodanovich MY, Gubskiy IL, Kudabaeva MS, Namestnikova DD, Kisel AA, Anan’ina TV, Tumentceva YA, Mustafina LR, Yarnykh VL. Long-term monitoring of chronic demyelination and remyelination in a rat ischemic stroke model using macromolecular proton fraction mapping. J Cereb Blood Flow Metab 2021; 41:2856-2869. [PMID: 34107787 PMCID: PMC8756474 DOI: 10.1177/0271678x211020860] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 04/24/2021] [Accepted: 04/26/2021] [Indexed: 12/23/2022]
Abstract
Remyelination is a key process enabling post-stroke brain tissue recovery and plasticity. This study aimed to explore the feasibility of demyelination and remyelination monitoring in experimental stroke from the acute to chronic stage using an emerging myelin imaging biomarker, macromolecular proton fraction (MPF). After stroke induction by transient middle cerebral artery occlusion, rats underwent repeated MRI examinations during 85 days after surgery with histological endpoints for the animal subgroups on the 7th, 21st, 56th, and 85th days. MPF maps revealed two sub-regions within the infarct characterized by distinct temporal profiles exhibiting either a persistent decrease by 30%-40% or a transient decrease followed by return to nearly normal values after one month of observation. Myelin histology confirmed that these sub-regions had nearly similar extent of demyelination in the sub-acute phase and then demonstrated either chronic demyelination or remyelination. The remyelination zones also exhibited active axonal regrowth, reconstitution of compact fiber bundles, and proliferation of neuronal and oligodendroglial precursors. The demyelination zones showed more extensive astrogliosis from the 21st day endpoint. Both sub-regions had substantially depleted neuronal population over all endpoints. These results histologically validate MPF mapping as a novel approach for quantitative assessment of myelin damage and repair in ischemic stroke.
Collapse
Affiliation(s)
| | - Ilya L Gubskiy
- Research Institute of Cerebrovascular Pathology and Stroke, Pirogov Russian Medical University, Moscow, Russian Federation
| | - Marina S Kudabaeva
- Laboratory of Neurobiology, Tomsk State University, Tomsk, Russian Federation
| | - Darya D Namestnikova
- Research Institute of Cerebrovascular Pathology and Stroke, Pirogov Russian Medical University, Moscow, Russian Federation
| | - Alena A Kisel
- Laboratory of Neurobiology, Tomsk State University, Tomsk, Russian Federation
- Department of Radiology, University of Washington, Seattle, USA
| | - Tatyana V Anan’ina
- Laboratory of Neurobiology, Tomsk State University, Tomsk, Russian Federation
| | - Yana A Tumentceva
- Laboratory of Neurobiology, Tomsk State University, Tomsk, Russian Federation
| | - Lilia R Mustafina
- Department of histology, embriology, and cytology, Siberian State Medical University, Tomsk, Russian Federation
| | - Vasily L Yarnykh
- Laboratory of Neurobiology, Tomsk State University, Tomsk, Russian Federation
- Department of Radiology, University of Washington, Seattle, USA
| |
Collapse
|
8
|
Lie AL, Pan X, White TW, Vaghefi E, Donaldson PJ. Age-Dependent Changes in Total and Free Water Content of In Vivo Human Lenses Measured by Magnetic Resonance Imaging. Invest Ophthalmol Vis Sci 2021; 62:33. [PMID: 34293079 PMCID: PMC8300047 DOI: 10.1167/iovs.62.9.33] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Purpose To use magnetic resonance imaging (MRI) to measure age-dependent changes in total and free water in human lenses in vivo. Methods Sixty-four healthy adults aged 18 to 86 years were recruited, fitted with a 32-channel head receiver coil, and placed in a 3 Tesla clinical MR scanner. Scans of the crystalline lens were obtained using a volumetric interpolated breath-hold examination sequence with dual flip angles, which were corrected for field inhomogeneity post-acquisition using a B1-map obtained using a turbo-FLASH sequence. The spatial distribution and content of corrected total (ρlens) and free (T1) water along the lens optical axis were extracted using custom-written code. Results Lens total water distribution and content did not change with age (all P > 0.05). In contrast to total water, a gradient in free water content that was highest in the periphery relative to the center was present in lenses across all ages. However, this initially parabolic free water gradient gradually developed an enhanced central plateau, as indicated by increasing profile shape parameter values (anterior: 0.067/y, P = 0.004; posterior: 0.050/y, P = 0.020) and central free water content (1.932 ms/y, P = 0.022) with age. Conclusions MRI can obtain repeatable total and free water measurements of in vivo human lenses. The observation that the lens steady-state free, but not total, water gradient is abolished with age raises the possibility that alterations in protein-water interactions are an underlying cause of the degradation in lens optics and overall vision observed with aging.
Collapse
Affiliation(s)
- Alyssa L Lie
- School of Optometry and Vision Science, New Zealand National Eye Centre, University of Auckland, New Zealand
| | - Xingzheng Pan
- School of Optometry and Vision Science, New Zealand National Eye Centre, University of Auckland, New Zealand.,Department of Physiology, School of Medical Sciences, New Zealand National Eye Centre, University of Auckland, New Zealand
| | - Thomas W White
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, New York, United States
| | - Ehsan Vaghefi
- School of Optometry and Vision Science, New Zealand National Eye Centre, University of Auckland, New Zealand
| | - Paul J Donaldson
- Department of Physiology, School of Medical Sciences, New Zealand National Eye Centre, University of Auckland, New Zealand
| |
Collapse
|
9
|
Alrayashi R, Braun RD, Muca A, Kühl A, Hali M, Holt AG. Postmortem neuroimaging: Temporal and spatial sensitivity of manganese-enhanced magnetic resonance imaging (MEMRI) and impact of Mn 2+ uptake. Hear Res 2021; 407:108276. [PMID: 34107410 DOI: 10.1016/j.heares.2021.108276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 04/20/2021] [Accepted: 05/08/2021] [Indexed: 11/29/2022]
Abstract
Magnetic resonance imaging data collection and analysis have been challenges in the field of auditory neuroscience. Recent studies have addressed these concerns by using manganese-enhanced magnetic resonance imaging (MEMRI). Basic challenges for in vivo application of MEMRI in rodents includes how to set inclusion criteria for adequate Mn2+ uptake and whether valid data can be collected from brains postmortem. Since brain Mn2+ uptake is complete within 2-4 h and clearance can take 2-4 weeks, one assumption has been that Mn2+-enhanced R1 values continue to reliably reflect the degree of Mn2+-uptake for some indeterminate time after death. To address these issues, the impact of death on R1 values was determined in rats administered Mn2+ and rats that were not. Images of auditory nuclei were collected at fixed intervals from rats before and after death for up to 10 h postmortem. By taking a ratio of pituitary and muscle T1-W intensities (P/M), a reliable quantitative method for assessing adequate brain Mn2+ uptake was created and suggest that P/M ratios should be adopted to objectively measure the quality of the Mn2+ injection. Postmortem R1 values decreased in all brain regions in both the After Mn2+ and No Mn2+ groups. However, the time-course of postmortem changes in R1 was dependent on brain region and degree of Mn2+ uptake. Thus, postmortem R1 values not only differ after death, but vary with time and across brain regions. Postmortem R1 values in unfixed brain tissue, including the auditory nuclei, should be interpreted with caution.
Collapse
Affiliation(s)
- Rasheed Alrayashi
- Department of Ophthalmology, Visual, and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Rod D Braun
- Department of Ophthalmology, Visual, and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Antonela Muca
- Department of Ophthalmology, Visual, and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - André Kühl
- Department of Ophthalmology, Visual, and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Mirabela Hali
- Department of Ophthalmology, Visual, and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Avril Genene Holt
- Department of Ophthalmology, Visual, and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, USA; John D. Dingell VAMC, Detroit, MI, USA.
| |
Collapse
|
10
|
Mac Grory B, Saldanha IJ, Mistry EA, Stretz C, Poli S, Sykora M, Kellert L, Feil K, Shah S, McTaggart R, Riebau D, Yaghi S, Gaines K, Xian Y, Feng W, Schrag M. Thrombolytic therapy for wake-up stroke: A systematic review and meta-analysis. Eur J Neurol 2021; 28:2006-2016. [PMID: 33772987 DOI: 10.1111/ene.14839] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 03/21/2021] [Indexed: 12/01/2022]
Abstract
BACKGROUND AND PURPOSE According to evidence-based clinical practice guidelines, patients presenting with disabling stroke symptoms should be treated with intravenous tissue plasminogen activator (IV tPA) within 4.5 h of time last known well. However, 25% of strokes are detected upon awakening (i.e., wake-up stroke [WUS]), which renders patients ineligible for IV tPA administered via time-based treatment algorithms, because it is impossible to establish a reliable time of symptom onset. We performed a systematic review and meta-analysis of the efficacy and safety of IV tPA compared with normal saline, placebo, or no treatment in patients with WUS using imaging-based treatment algorithms. METHODS We searched MEDLINE, Web of Science, and Scopus between January 1, 2006 and April 30, 2020. We included controlled trials (randomized or nonrandomized), observational cohort studies (prospective or retrospective), and single-arm studies in which adults with WUS were administered IV tPA after magnetic resonance imaging (MRI)- or computed tomography (CT)-based imaging. Our primary outcome was recovery at 90 days (defined as a modified Rankin Scale [mRS] score of 0-2), and our secondary outcomes were symptomatic intracranial hemorrhage (sICH) within 36 h, mortality, and other adverse effects. RESULTS We included 16 studies that enrolled a total of 14,017 patients. Most studies were conducted in Europe (37.5%) or North America (37.5%), and 1757 patients (12.5%) received IV tPA. All studies used MRI-based (five studies) or CT-based (10 studies) imaging selection, and one study used a combination of modalities. Sixty-one percent of patients receiving IV tPA achieved an mRS score of 0 to 2 at 90 days (95% confidence interval [CI]: 51%-70%, 12 studies), with a relative risk (RR) of 1.21 compared with patients not receiving IV tPA (95% CI: 1.01-1.46, four studies). Three percent of patients receiving IV tPA experienced sICH within 36 h (95% CI: 2.5%-4.1%; 16 studies), which is an RR of 4.00 compared with patients not receiving IV tPA (95% CI: 2.85-5.61, seven studies). CONCLUSIONS This systematic review and meta-analysis suggests that IV tPA is associated with a better functional outcome at 90 days despite the increased but acceptable risk of sICH. Based on these results, IV tPA should be offered as a treatment for WUS patients with favorable neuroimaging findings.
Collapse
Affiliation(s)
- Brian Mac Grory
- Department of Neurology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Ian J Saldanha
- Center for Evidence Synthesis in Health, Department of Health Services, Policy, and Practice, Brown University School of Public Health, Providence, Rhode Island, USA.,Department of Epidemiology, Brown University School of Public Health, Providence, Rhode Island, USA
| | - Eva A Mistry
- Department of Neurology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Christoph Stretz
- Department of Neurology, Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Sven Poli
- Department of Neurology With Focus on Neurovascular Diseases, University Hospital Tübingen, Tübingen, Germany.,Hertie Institute for Clinical Brain Research, University Hospital Tübingen, Tübingen, Germany
| | - Marek Sykora
- Department of Neurology, St. John's Hospital, Medical Faculty, Sigmund Freud University, Vienna, Austria
| | - Lars Kellert
- Department of Neurology, Ludwig Maximilians University, Munich, Germany
| | - Katharina Feil
- Department of Neurology, Ludwig Maximilians University, Munich, Germany
| | - Shreyansh Shah
- Department of Neurology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Ryan McTaggart
- Department of Neurology, Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA.,Department of Neurosurgery, Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA.,Department of Radiology, Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Derek Riebau
- Department of Neurology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Shadi Yaghi
- Department of Neurology, New York University School of Medicine, New York, New York, USA
| | - Kenneth Gaines
- Department of Neurology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Ying Xian
- Department of Neurology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Wuwei Feng
- Department of Neurology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Matthew Schrag
- Department of Neurology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|
11
|
Lee H, Lee EJ, Ham S, Lee HB, Lee JS, Kwon SU, Kim JS, Kim N, Kang DW. Machine Learning Approach to Identify Stroke Within 4.5 Hours. Stroke 2020; 51:860-866. [PMID: 31987014 DOI: 10.1161/strokeaha.119.027611] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background and Purpose- We aimed to investigate the ability of machine learning (ML) techniques analyzing diffusion-weighted imaging (DWI) and fluid-attenuated inversion recovery (FLAIR) magnetic resonance imaging to identify patients within the recommended time window for thrombolysis. Methods- We analyzed DWI and FLAIR images of consecutive patients with acute ischemic stroke within 24 hours of clear symptom onset by applying automatic image processing approaches. These processes included infarct segmentation, DWI, and FLAIR imaging registration and image feature extraction. A total of 89 vector features from each image sequence were captured and used in the ML. Three ML models were developed to estimate stroke onset time for binary classification (≤4.5 hours): logistic regression, support vector machine, and random forest. To evaluate the performance of ML models, the sensitivity and specificity for identifying patients within 4.5 hours were compared with the sensitivity and specificity of human readings of DWI-FLAIR mismatch. Results- Data from a total of 355 patients were analyzed. DWI-FLAIR mismatch from human readings identified patients within 4.5 hours of symptom onset with 48.5% sensitivity and 91.3% specificity. ML algorithms had significantly greater sensitivities than human readers (75.8% for logistic regression, P=0.020; 72.7% for support vector machine, P=0.033; 75.8% for random forest, P=0.013) in detecting patients within 4.5 hours, but their specificities were comparable (82.6% for logistic regression, P=0.157; 82.6% for support vector machine, P=0.157; 82.6% for random forest, P=0.157). Conclusions- ML algorithms using multiple magnetic resonance imaging features were feasible even more sensitive than human readings in identifying patients with stroke within the time window for acute thrombolysis.
Collapse
Affiliation(s)
- Hyunna Lee
- From the Health Innovation Big Data Center, Asan Institute for Life Science, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea. (H.L.)
| | - Eun-Jae Lee
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea. (E.-J.L., H.-B.L., S.U.K., J.S.K., D.-W.K.)
| | - Sungwon Ham
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea. (S.H.)
| | - Han-Bin Lee
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea. (E.-J.L., H.-B.L., S.U.K., J.S.K., D.-W.K.)
| | - Ji Sung Lee
- Clinical Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea. (J.S.L.)
| | - Sun U Kwon
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea. (E.-J.L., H.-B.L., S.U.K., J.S.K., D.-W.K.)
| | - Jong S Kim
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea. (E.-J.L., H.-B.L., S.U.K., J.S.K., D.-W.K.)
| | - Namkug Kim
- Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea. (N.K.).,Department of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea. (N.K.)
| | - Dong-Wha Kang
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea. (E.-J.L., H.-B.L., S.U.K., J.S.K., D.-W.K.)
| |
Collapse
|
12
|
Sghedoni R, Coniglio A, Mazzoni LN, Busoni S, Belli G, Tarducci R, Nocetti L, Fedeli L, Esposito M, Ciccarone A, Altabella L, Bellini A, Binotto L, Caivano R, Carnì M, Ricci A, Cimolai S, D'Urso D, Gasperi C, Levrero F, Mangili P, Morzenti S, Nitrosi A, Oberhofer N, Parruccini N, Toncelli A, Valastro LM, Gori C, Gobbi G, Giannelli M. A straightforward multiparametric quality control protocol for proton magnetic resonance spectroscopy: Validation and comparison of various 1.5 T and 3 T clinical scanner systems. Phys Med 2018; 54:49-55. [PMID: 30337010 DOI: 10.1016/j.ejmp.2018.08.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 07/25/2018] [Accepted: 08/13/2018] [Indexed: 02/08/2023] Open
Abstract
PURPOSE The aim of this study was to propose and validate across various clinical scanner systems a straightforward multiparametric quality assurance procedure for proton magnetic resonance spectroscopy (MRS). METHODS Eighteen clinical 1.5 T and 3 T scanner systems for MRS, from 16 centres and 3 different manufacturers, were enrolled in the study. A standard spherical water phantom was employed by all centres. The acquisition protocol included 3 sets of single (isotropic) voxel (size 20 mm) PRESS acquisitions with unsuppressed water signal and acquisition voxel position at isocenter as well as off-center, repeated 4/5 times within approximately 2 months. Water peak linewidth (LW) and area under the water peak (AP) were estimated. RESULTS LW values [mean (standard deviation)] were 1.4 (1.0) Hz and 0.8 (0.3) Hz for 3 T and 1.5 T scanners, respectively. The mean (standard deviation) (across all scanners) coefficient of variation of LW and AP for different spatial positions of acquisition voxel were 43% (20%) and 11% (11%), respectively. The mean (standard deviation) phantom T2values were 1145 (50) ms and 1010 (95) ms for 1.5 T and 3 T scanners, respectively. The mean (standard deviation) (across all scanners) coefficients of variation for repeated measurements of LW, AP and T2 were 25% (20%), 10% (14%) and 5% (2%), respectively. CONCLUSIONS We proposed a straightforward multiparametric and not time consuming quality control protocol for MRS, which can be included in routine and periodic quality assurance procedures. The protocol has been validated and proven to be feasible in a multicentre comparison study of a fairly large number of clinical 1.5 T and 3 T scanner systems.
Collapse
Affiliation(s)
| | - Angela Coniglio
- Medical Physics Unit, Ospedale San Giovanni Calibita Fatebenefratelli, Roma, Italy.
| | | | | | | | - Roberto Tarducci
- Health Physics Unit, Azienda Ospedaliera di Perugia, Perugia, Italy
| | - Luca Nocetti
- Health Physics Unit, Azienda Ospedaliera di Modena, Modena, Italy
| | - Luca Fedeli
- Physics and Astronomy Department, University of Florence, Firenze, Italy
| | - Marco Esposito
- Health Physics Unit, Azienda USL Toscana Centro, Firenze, Italy
| | | | | | | | - Luca Binotto
- Medical Physics Unit, Azienda ULSS 3 Serenissima, Mestre, Italy
| | - Rocchina Caivano
- Radiotherapy and Health Physics Unit, IRCCS CROB, Rionero in Vulture - Potenza, Italy
| | - Marco Carnì
- Health Physics Unit, Policlinico Umberto I, Roma, Italy
| | | | - Sara Cimolai
- Health Physics Unit, Azienda ULSS 2 Marca Trevigiana, Treviso, Italy
| | - Davide D'Urso
- Health Physics Unit, Azienda ULSS 2 Marca Trevigiana, Treviso, Italy
| | - Chiara Gasperi
- Health Physics Unit, Azienda USL Toscana Sud Est, Arezzo, Italy
| | - Fabrizio Levrero
- Medical and Health Physics Unit, IRCCS AOU San Martino, Genova, Italy
| | - Paola Mangili
- Medical Physics Unit, IRCCS San Raffaele, Milano, Italy
| | | | - Andrea Nitrosi
- Medical Physics Unit, Arcispedale Santa Maria Nuova - IRCCS, Reggio Emilia, Italy
| | - Nadia Oberhofer
- Health Physics, Azienda Sanitaria della Provincia Autonoma di Bolzano, Bolzano, Italy
| | | | | | | | - Cesare Gori
- Health Physics Unit, AOU Careggi, Firenze, Italy
| | - Gianni Gobbi
- Health Physics Unit, Azienda Ospedaliera di Perugia, Perugia, Italy
| | - Marco Giannelli
- Unit of Medical Physics, Pisa University Hospital "Azienda Ospedaliero-Universitaria Pisana", Pisa, Italy
| | | |
Collapse
|
13
|
Clinical applications of diffusion weighted imaging in neuroradiology. Insights Imaging 2018; 9:535-547. [PMID: 29846907 PMCID: PMC6108979 DOI: 10.1007/s13244-018-0624-3] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 03/12/2018] [Accepted: 03/20/2018] [Indexed: 12/21/2022] Open
Abstract
Abstract Diffusion-weighted imaging (DWI) has revolutionised stroke imaging since its introduction in the mid-1980s, and it has also become a pillar of current neuroimaging. Diffusion abnormalities represent alterations in the random movement of water molecules in tissues, revealing their microarchitecture, and occur in many neurological conditions. DWI provides useful information, increasing the sensitivity of MRI as a diagnostic tool, narrowing the differential diagnosis, providing prognostic information, aiding in treatment planning and evaluating response to treatment. Recently, there have been several technical improvements in DWI, leading to reduced acquisition time and artefacts and enabling the development of diffusion tensor imaging (DTI) as a tool for assessing white matter. We aim to review the main clinical uses of DWI, focusing on the physiological mechanisms that lead to diffusion abnormalities. Common pitfalls will also be addressed. Teaching Points • DWI includes EPI, TSE, RESOLVE or EPI combined with reduced volume excitation. • DWI is the most sensitive sequence in stroke diagnosis and provides information about prognosis. • DWI helps in the detection of intramural haematomas (arterial dissection). • In diffusion imaging, ADC is inversely proportional to tumour cellularity. • DWI and DTI derived parameters can be used as biomarkers in different pathologies.
Collapse
|
14
|
Ji Y, Zhou IY, Qiu B, Sun PZ. Progress toward quantitative in vivo chemical exchange saturation transfer (CEST) MRI. Isr J Chem 2017. [DOI: 10.1002/ijch.201700025] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Yang Ji
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital; Harvard Medical School; Rm 2301, 149 13 Street Charlestown MA 02129
- Center for Biomedical Engineering, Department of Electronic Science and Technology; University of Science and Technology of China; Hefei China
| | - Iris Yuwen Zhou
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital; Harvard Medical School; Rm 2301, 149 13 Street Charlestown MA 02129
| | - Bensheng Qiu
- Center for Biomedical Engineering, Department of Electronic Science and Technology; University of Science and Technology of China; Hefei China
| | - Phillip Zhe Sun
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital; Harvard Medical School; Rm 2301, 149 13 Street Charlestown MA 02129
| |
Collapse
|
15
|
Wei XE, Zhou J, Li WB, Zhao YW, Li MH, Li YH. MRI based thrombolysis for FLAIR-negative stroke patients within 4.5-6h after symptom onset. J Neurol Sci 2016; 372:421-427. [PMID: 27839719 DOI: 10.1016/j.jns.2016.11.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Revised: 11/06/2016] [Accepted: 11/07/2016] [Indexed: 10/20/2022]
Abstract
To investigate the feasibility of DWI-FLAIR mismatch in identifying patients who might benefit from thrombolytic therapy within 4.5-6h, we analyzed the data of 105 ischemic stroke patients with known time of symptom onset who underwent MRI within 6h of stroke and thrombolysis between December 2006 and December 2013. They were divided into three groups: symptom onset within 4.5h (n=66); 4.5-6h and FLAIR images negative (n=9); and 4.5-6h and FLAIR images positive (n=30). Outcome of thrombolysis was assessed for each group by recanalization rate, National Institutes of Health Stroke Scale (NIHSS) and modified Rankin Scale (mRS) scores. The results showed that mismatch between positive DWI and negative FLAIR images identified patients within 4.5h of symptom onset with sensitivity, specificity, positive predictive value and negative predictive value of 40.9%, 76.9%, and 75% and 43.5%. Recanalization rate, NIHSS score and mRS score were all better in both the 0-4.5h and 4.5-6h FLAIR-negative groups than in the 4.5-6h FLAIR-positive group (p<0.05). These data demonstrate that within 4.5-6h of symptom onset, patients with negative FLAIR images may benefit from thrombolysis therapy.
Collapse
Affiliation(s)
- Xiao-Er Wei
- Institute of Diagnostic and Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600, Yi Shan Road, Shanghai 200233, China
| | - Jia Zhou
- Institute of Diagnostic and Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600, Yi Shan Road, Shanghai 200233, China
| | - Wen-Bin Li
- Institute of Diagnostic and Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600, Yi Shan Road, Shanghai 200233, China
| | - Yu-Wu Zhao
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600, Yi Shan Road, Shanghai 200233, China
| | - Ming-Hua Li
- Institute of Diagnostic and Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600, Yi Shan Road, Shanghai 200233, China
| | - Yue-Hua Li
- Institute of Diagnostic and Interventional Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600, Yi Shan Road, Shanghai 200233, China.
| |
Collapse
|
16
|
Winter B, Brunecker P, Fiebach JB, Jungehulsing GJ, Kronenberg G, Endres M. Striatal Infarction Elicits Secondary Extrafocal MRI Changes in Ipsilateral Substantia Nigra. PLoS One 2015; 10:e0136483. [PMID: 26325192 PMCID: PMC4556671 DOI: 10.1371/journal.pone.0136483] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2014] [Accepted: 12/24/2014] [Indexed: 02/05/2023] Open
Abstract
Focal ischemia may induce pathological alterations in brain areas distant from the primary lesion. In animal models, exofocal neuron death in the ipsilateral midbrain has been described after occlusion of the middle cerebral artery (MCA). Using sequential magnetic resonance imaging (T2- and diffusion-weighted) at 3 Tesla, we investigated acute ischemic stroke patients on days 1, 2, 6, 8, and 10 after stroke onset. Sixteen consecutive patients who had suffered a stroke involving the caudate nucleus and/or putamen of either hemisphere were recruited into the study. Four additional patients with strokes sparing the caudate nucleus and putamen but encompassing at least one-third of the MCA territory served as controls. Ischemic lesions involving striatal structures resulted in hyperintense lesions in ipsilateral midbrain that emerged between days 6 and 10 after stroke and were not present on the initial scans. In contrast, none of the control stroke patients developed secondary midbrain lesions. Hyperintense lesions in the pyramidal tract or the brain stem caused by degeneration of the corticospinal tract could be clearly distinguished from these secondary midbrain gray matter lesions and were detectable from day 2 after ischemia. Co-registration of high-resolution images with a digitized anatomic atlas revealed localization of secondary lesions primarily in the substantia nigra pars compacta. Apparent diffusion coefficient (ADC) values in the secondary lesions showed a delayed sharp decline through day 10. Normalization of ADC values was observed at late measurements. Taken together, our study demonstrates that striatal infarction elicits delayed degenerative changes in ipsilateral substantia nigra pars compacta.
Collapse
Affiliation(s)
- Benjamin Winter
- Center for Stroke Research Berlin (CSB), Charité-Universitätsmedizin Berlin, Berlin, Charitéplatz 1,10117, Berlin, Germany
- Department of Neurology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Peter Brunecker
- Center for Stroke Research Berlin (CSB), Charité-Universitätsmedizin Berlin, Berlin, Charitéplatz 1,10117, Berlin, Germany
| | - Jochen B. Fiebach
- Center for Stroke Research Berlin (CSB), Charité-Universitätsmedizin Berlin, Berlin, Charitéplatz 1,10117, Berlin, Germany
| | - Gerhard Jan Jungehulsing
- Center for Stroke Research Berlin (CSB), Charité-Universitätsmedizin Berlin, Berlin, Charitéplatz 1,10117, Berlin, Germany
- Department of Neurology, Jüdisches Krankenhaus Berlin, Heinz-Galinski-Strasse 1, 13347, Berlin, Germany
| | - Golo Kronenberg
- Center for Stroke Research Berlin (CSB), Charité-Universitätsmedizin Berlin, Berlin, Charitéplatz 1,10117, Berlin, Germany
- Department of Neurology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Department of Psychiatry, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Max-Delbrück Center and Charité Medical Faculty, Experimental and Clinical Research Center, Lindenbergerweg 80, 13125, Berlin, Germany
| | - Matthias Endres
- Center for Stroke Research Berlin (CSB), Charité-Universitätsmedizin Berlin, Berlin, Charitéplatz 1,10117, Berlin, Germany
- Department of Neurology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Excellence Cluster Neurocure, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE), Ludwig-Erhard-Allee, 53175, Bonn, Germany
- German Centre for Cardiovascular Research (DZHK), Oudenarder Straße 16, 13347, Berlin, Germany
- * E-mail:
| |
Collapse
|
17
|
Abstract
Wake-up stroke, defined as the situation where a patient awakens with stroke symptoms that were not present prior to falling asleep, represents roughly 1 in 5 acute ischemic strokes and remains a therapeutic dilemma. Patients with wake-up stroke were excluded from most ischemic stroke treatment trials and are often not eligible for acute reperfusion therapy in clinical practice, leading to poor outcomes. Studies of neuroimaging with standard noncontrast computed tomography (CT), magnetic resonance imaging (MRI), and multimodal perfusion-based CT and MRI suggest wake-up stroke may occur shortly before awakening and may assist in selecting patients for acute reperfusion therapies. Pilot studies of wake-up stroke treatment based on these neuroimaging features are promising but have limited generalizability. Ongoing randomized treatment trials using neuroimaging-based patient selection may identify a subset of patients with wake-up stroke that can safely benefit from acute reperfusion therapies.
Collapse
Affiliation(s)
- Mark N Rubin
- Department of Neurology, Divisions of Hospital & Vascular Neurology, Mayo Clinic, Scottsdale, AZ, USA
| | - Kevin M Barrett
- Department of Neurology, Division of Vascular Neurology, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
18
|
Spatio-temporal assessment of the neuroprotective effects of neuregulin-1 on ischemic stroke lesions using MRI. J Neurol Sci 2015; 357:28-34. [PMID: 26183085 DOI: 10.1016/j.jns.2015.06.055] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 05/25/2015] [Accepted: 06/25/2015] [Indexed: 11/23/2022]
Abstract
The neuroprotective effects of neuregulin-1 (NRG-1) on stroke lesions were assessed longitudinally in rats with middle cerebral artery occlusion (MCAo) using MRI. Sprague-Dawley rats (n=16, 250±20g) underwent permanent MCAo surgery with cerebral blood flow (CBF) monitored by laser doppler flowmetry at ipsilateral side of bregma for 20min post-occlusion. A single 50μl bolus dose of NRG-1 or vehicle was administered into the left internal carotid artery immediately prior to MCAo. The expansion of the ischemic lesion into the cortex was attenuated by NRG-1 over a 48-hour (h) time span as measured by diffusion weighted imaging (DWI). The final infarct volumes of NRG-1 treated rats were significantly smaller than those of the vehicle treated rats at 48h (264.8±192.1 vs. 533.4±175.5mm(3), p<0.05). The NRG-1 treated rats were further subdivided into 2 subgroups according to their CBF reduction during stroke surgery: mild ischemia (<70% CBF reduction) or severe ischemia (>70% CBF reduction). In particular, ischemic infarction was not usually observed in the cortex of NRG-1 treated rats with mild ischemia at 3 and 48h post-occlusion. Histological results validated the imaging findings and demonstrated that NRG-1 treated rats had fewer injured neurons in peri-infarct areas 48h post-ischemia. In summary, the neuroprotective effect of NRG-1 in the pMCAo stroke model was demonstrated by prevention of ischemic lesion expansion, reduced infarct volume and protection of neurons from ischemic damage.
Collapse
|
19
|
McVicar N, Li AX, Gonçalves DF, Bellyou M, Meakin SO, Prado MAM, Bartha R. Quantitative tissue pH measurement during cerebral ischemia using amine and amide concentration-independent detection (AACID) with MRI. J Cereb Blood Flow Metab 2014; 34:690-8. [PMID: 24496171 PMCID: PMC3982091 DOI: 10.1038/jcbfm.2014.12] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2013] [Revised: 11/26/2013] [Accepted: 12/23/2013] [Indexed: 11/09/2022]
Abstract
Tissue pH is an indicator of altered cellular metabolism in diseases including stroke and cancer. Ischemic tissue often becomes acidic due to increased anaerobic respiration leading to irreversible cellular damage. Chemical exchange saturation transfer (CEST) effects can be used to generate pH-weighted magnetic resonance imaging (MRI) contrast, which has been used to delineate the ischemic penumbra after ischemic stroke. In the current study, a novel MRI ratiometric technique is presented to measure absolute pH using the ratio of CEST-mediated contrast from amine and amide protons: amine/amide concentration-independent detection (AACID). Effects of CEST were observed at 2.75 parts per million (p.p.m.) for amine protons and at 3.50 p.p.m. for amide protons downfield (i.e., higher frequency) from bulk water. Using numerical simulations and in vitro MRI experiments, we showed that pH measured using AACID was independent of tissue relaxation time constants, macromolecular magnetization transfer effects, protein concentration, and temperature within the physiologic range. After in vivo pH calibration using phosphorus ((31)P) magnetic resonance spectroscopy ((31)P-MRS), local acidosis is detected in mouse brain after focal permanent middle cerebral artery occlusion. In summary, our results suggest that AACID represents a noninvasive method to directly measure the spatial distribution of absolute pH in vivo using CEST MRI.
Collapse
Affiliation(s)
- Nevin McVicar
- Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada
| | - Alex X Li
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Daniela F Gonçalves
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
- Department of Physiology and Biophysics, Neuroscience Centre, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Miranda Bellyou
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | - Susan O Meakin
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
- Department of Biochemistry, University of Western Ontario, Western University, London, Ontario, Canada
| | - Marco AM Prado
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
- Department of Anatomy and Cell Biology, University of Western Ontario, London, Ontario, Canada
| | - Robert Bartha
- Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
20
|
Rimmele DL, Thomalla G. Wake-up stroke: clinical characteristics, imaging findings, and treatment option - an update. Front Neurol 2014; 5:35. [PMID: 24723908 PMCID: PMC3972483 DOI: 10.3389/fneur.2014.00035] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 03/11/2014] [Indexed: 12/22/2022] Open
Abstract
About 25% of all strokes occur during sleep, i.e., without knowledge of exact time of symptom onset. According to licensing criteria, this large group of patients is excluded from treatment with received tissue-plasminogen activator, the only specific stroke treatment proven effective in large randomized trials. This paper reviews clinical and imaging characteristics of wake-up stroke and gives an update on treatment options for these patients. From clinical and imaging studies, there is evidence suggesting that many wake-up strokes occur close to awakening and thus, patients might be within the approved time-window of thrombolysis when presenting to the emergency department. Several imaging approaches are suggested to identify wake-up stroke patients likely to benefit from thrombolysis, including non-contrast CT, CT-perfusion, penumbral MRI, and the recent concept of diffusion weighted imaging-fluid attenuated inversion recovery (DWI-FLAIR). A number of small case series and observational studies report results of thrombolysis in wake-up stroke, and no safety concerns have occurred, while conclusions on efficacy cannot be drawn from these studies. To this end, there are ongoing clinical trials enrolling wake-up stroke patients based on imaging findings, i.e., the DWI-FLAIR-mismatch (WAKE-UP) or penumbral imaging (EXTEND). The results of these trials will provide evidence to guide thrombolysis in wake-up stroke and thus, expand treatment options for this large group of stroke patients.
Collapse
Affiliation(s)
- D Leander Rimmele
- Klinik und Poliklinik für Neurologie, Kopf- und Neurozentrum, Universitätsklinikum Hamburg-Eppendorf , Hamburg , Germany
| | - Götz Thomalla
- Klinik und Poliklinik für Neurologie, Kopf- und Neurozentrum, Universitätsklinikum Hamburg-Eppendorf , Hamburg , Germany
| |
Collapse
|
21
|
Tarpley J, Franc D, Tansy AP, Liebeskind DS. Use of perfusion imaging and other imaging techniques to assess risks/benefits of acute stroke interventions. Curr Atheroscler Rep 2014; 15:336. [PMID: 23666875 DOI: 10.1007/s11883-013-0336-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The advent of multimodal neuroimaging has provided acute stroke care providers with an armamentarium of sophisticated imaging options to utilize for guidance in clinical decision-making and management of acute ischemic stroke patients. Here, we propose a framework and potential algorithm-based methodology for imaging modality selection and utilization for the purpose of achieving optimal stroke clinical care. We first review imaging options that may best inform decision-making regarding revascularization eligibility, with a focus on the imaging modalities that best identify critical inclusion and exclusion criteria. Next, we review imaging methods that may guide the successful achievement of revascularization once it has been deemed desirable and feasible. Further, we review imaging modalities that may best assist in both the noninterventional care of acute stroke as well as the identification of stroke-mimics. Finally, we review imaging techniques under current investigation that show promise to improve future acute stroke management.
Collapse
Affiliation(s)
- Jason Tarpley
- UCLA Stroke Center, 710 Westwood Plaza, Los Angeles, CA 90095, USA
| | | | | | | |
Collapse
|
22
|
Thomalla G, Fiehler J. Treating Wake-Up Stroke Patients. CURRENT RADIOLOGY REPORTS 2014. [DOI: 10.1007/s40134-013-0030-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
23
|
Kuribayashi H, Sekino M, Minowa T, Maitani Y, Ohsaki H, Tsushima S, Hirai S, Ueda M, Katayama Y. Accuracy of equilibrium magnetization mapping in sliced two-dimensional spoiled gradient-recalled echo pulse sequence with variable flip angle. J Magn Reson Imaging 2013; 38:1245-50. [DOI: 10.1002/jmri.24023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2012] [Accepted: 12/07/2012] [Indexed: 11/08/2022] Open
Affiliation(s)
| | - Masaki Sekino
- School of Engineering; University of Tokyo; Chiba Japan
| | - Takuya Minowa
- Institute of Medicinal Chemistry; Hoshi University; Tokyo Japan
| | - Yoshie Maitani
- Institute of Medicinal Chemistry; Hoshi University; Tokyo Japan
| | | | - Shohji Tsushima
- Research Center for Carbon Recycling and Energy; Tokyo Institute of Technology; Tokyo Japan
| | - Shuichiro Hirai
- Research Center for Carbon Recycling and Energy; Tokyo Institute of Technology; Tokyo Japan
| | - Masayuki Ueda
- Department of Internal Medicine; Nippon Medical School; Tokyo Japan
| | - Yasuo Katayama
- Department of Internal Medicine; Nippon Medical School; Tokyo Japan
| |
Collapse
|
24
|
Water-content calculation in growth plate and cartilage using MR T1-mapping design and validation of a new method in a porcine model. Skeletal Radiol 2013; 42:1413-9. [PMID: 23842573 DOI: 10.1007/s00256-013-1674-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Revised: 05/22/2013] [Accepted: 06/09/2013] [Indexed: 02/02/2023]
Abstract
OBJECTIVE There is a close relation between cartilage health and its hydration state. Current magnetic resonance methods allow visualizing this tissue. However, a quantitative analysis is more useful when studying disease. The purpose of this study was to quantify water content in cartilage using magnetic resonance without contrast agents. MATERIALS AND METHODS Water-content estimations using T1 magnetic resonance mapping were done first in eight gelatin samples where the water content was previously known. The same method was used in the physeal areas of eight skeletally immature 30-kg pigs. To calculate accuracy, T1 calculations were compared to dry-freeze, which is considered the gold standard because it can remove the total water content form a tissue. Four fresh cartilage and seven gelatin samples were dry-frozen. Water content obtained from dry-freeze was compared to the one calculated from T1 map values. A mathematical model and statistical analysis were used to calculate the predictive value of the method and its significance. RESULTS T1-map-based magnetic resonance method can calculate water content in cartilage with an accuracy of 97.3 %. We calculated a coefficient of variance for this method against dry-frozen sample of 3.68 (SD = 1.2) in gelatin samples, and 2.73 (SD = 1.3) in in vivo samples. Between two independent observers, the coefficient of variance was 0.053, which suggests it can be easily reproduced. CONCLUSIONS Magnetic resonance was able to calculate, with high accuracy, the cartilage water content using T1 mapping sequences.
Collapse
|
25
|
Thomalla G, Ebinger M, Fiehler J, Fiebach JB, Endres M, Gerloff C. [EU-funded treatment study: WAKE-UP: a randomized, placebo-controlled MRI-based trial of thrombolysis in wake-up stroke]. DER NERVENARZT 2013; 83:1241-51. [PMID: 23015193 DOI: 10.1007/s00115-012-3532-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Patients waking up with stroke symptoms are generally excluded from intravenous thrombolysis. It was shown that magnetic resonance imaging (MRI) can identify patients within the time window for thrombolysis (≤ 4.5 h from symptom onset) by a mismatch between the acute ischemic lesion visible on diffusion-weighted imaging (DWI) but not visible on fluid-attenuated inversion recovery (FLAIR) imaging. The WAKE-UP trial is an investigator initiated, European, randomized, double-blind, placebo-controlled trial designed to test efficacy and safety of MRI-based thrombolysis with alteplase (tPA) in stroke patients with unknown time of symptom onset, e.g. due to symptom recognition on awakening. A total of 800 patients showing MRI findings of a DWI-FLAIR-mismatch will be randomized to either tPA or placebo. The primary efficacy endpoint will be favourable outcome defined by a modified Rankin scale score 0-1 at day 90. The primary safety outcome measures will be mortality and death or dependency defined by modified Rankin scale score 4-6 at 90 days. If positive the WAKE-UP trial is expected to change clinical practice and to make effective and safe treatment available for a large group of acute stroke patients currently excluded from specific acute treatment.
Collapse
Affiliation(s)
- G Thomalla
- Klinik und Poliklinik für Neurologie, Kopf- und Neurozentrum, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Deutschland.
| | | | | | | | | | | |
Collapse
|
26
|
Thomalla G, Fiebach JB, Østergaard L, Pedraza S, Thijs V, Nighoghossian N, Roy P, Muir KW, Ebinger M, Cheng B, Galinovic I, Cho TH, Puig J, Boutitie F, Simonsen CZ, Endres M, Fiehler J, Gerloff C. A multicenter, randomized, double-blind, placebo-controlled trial to test efficacy and safety of magnetic resonance imaging-based thrombolysis in wake-up stroke (WAKE-UP). Int J Stroke 2013; 9:829-36. [PMID: 23490032 DOI: 10.1111/ijs.12011] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
RATIONALE In about 20% of acute ischemic stroke patients stroke occurs during sleep. These patients are generally excluded from intravenous thrombolysis. MRI can identify patients within the time-window for thrombolysis (≤4·5 h from symptom onset) by a mismatch between the acute ischemic lesion visible on diffusion weighted imaging (DWI) but not visible on fluid-attenuated inversion recovery (FLAIR) imaging. AIMS AND HYPOTHESIS The study aims to test the efficacy and safety of MRI-guided thrombolysis with tissue plasminogen activator (rtPA) in ischemic stroke patients with unknown time of symptom onset, e.g., waking up with stroke symptoms. We hypothesize that stroke patients with unknown time of symptom onset with a DWI-FLAIR-mismatch pattern on MRI will have improved outcome when treated with rtPA compared to placebo. DESIGN WAKE-UP is an investigator initiated, European, multicentre, randomized, double-blind, placebo-controlled clinical trial. Patients with unknown time of symptom onset who fulfil clinical inclusion criteria (disabling neurological deficit, no contraindications against thrombolysis) will be studied by MRI. Patients with MRI findings of a DWI-FLAIR-mismatch will be randomised to either treatment with rtPA or placebo. STUDY OUTCOME The primary efficacy endpoint will be favourable outcome defined by modified Rankin Scale 0-1 at day 90. The primary safety outcome measures will be mortality and death or dependency defined by modified Rankin Scale 4-6 at 90 days. DISCUSSION If positive, WAKE-UP is expected to change clinical practice making effective and safe treatment available for a large group of acute stroke patients currently excluded from specific acute therapy.
Collapse
Affiliation(s)
- Götz Thomalla
- Klinik und Poliklinik für Neurologie, Kopf- und Neurozentrum, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Quantitative measurements of relative fluid-attenuated inversion recovery (FLAIR) signal intensities in acute stroke for the prediction of time from symptom onset. J Cereb Blood Flow Metab 2013; 33:76-84. [PMID: 23047272 PMCID: PMC3965287 DOI: 10.1038/jcbfm.2012.129] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In acute stroke magnetic resonance imaging, a 'mismatch' between visibility of an ischemic lesion on diffusion-weighted imaging (DWI) and missing corresponding parenchymal hyperintensities on fluid-attenuated inversion recovery (FLAIR) data sets was shown to identify patients with time from symptom onset ≤4.5 hours with high specificity. However, moderate sensitivity and suboptimal interpreter agreement are limitations of a visual rating of FLAIR lesion visibility. We tested refined image analysis methods in patients included in the previously published PREFLAIR study using refined visual analysis and quantitative measurements of relative FLAIR signal intensity (rSI) from a three-dimensional, segmented stroke lesion volume. A total of 399 patients were included. The rSI of FLAIR lesions showed a moderate correlation with time from symptom onset (r=0.382, P<0.001). A FLAIR rSI threshold of <1.0721 predicted symptom onset ≤4.5 hours with slightly increased specificity (0.85 versus 0.78) but also slightly decreased sensitivity (0.47 versus 0.58) as compared with visual analysis. Refined visual analysis differentiating between 'subtle' and 'obvious' FLAIR hyperintensities and classification and regression tree algorithms combining information from visual and quantitative analysis also did not improve diagnostic accuracy. Our results raise doubts whether the prediction of stroke onset time by visual image judgment can be improved by quantitative rSI measurements.
Collapse
|
28
|
Siemonsen S, Löbel U, Sedlacik J, Forkert ND, Mouridsen K, Østergaard L, Thomalla G, Fiehler J. Elevated T2-values in MRI of stroke patients shortly after symptom onset do not predict irreversible tissue infarction. Brain 2012; 135:1981-9. [PMID: 22505333 DOI: 10.1093/brain/aws079] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Distinct from signal alterations in diffusion-weighted images, T(2)-values are also dependent on tissue water content and known to increase with time from symptom onset in acute ischaemic stroke. The purpose of this study was to evaluate whether there is a detectable increase of T(2)-values in different regions in acute ischaemic stroke in the acute and subacute situation and to study the effect of recanalization on the evaluation of T(2)-values in the subacute phase. In addition, we sought to evaluate whether this increase in T(2)-values is reversible. For this purpose, 22 patients with acute ischaemic stroke in the territory of the middle cerebral artery underwent magnetic resonance imaging including diffusion-weighted imaging, perfusion-weighted imaging, fluid-attenuated inversion recovery to determine final infarct size, time-of-flight-angiography (acute and on day 1 or 2) and a triple echo-T(2)-sequence (calculation of T(2) maps) within 6 h after symptom onset. Images were co-registered and regions of diffusion restriction and prolonged time-to-peak as well as surviving tissue (surviving tissue = time-to-peak - final infarct size) and lesion growth (lesion growth = final infarct size-diffusion restriction) were defined and superimposed onto the quantitative T(2) map. In addition, patients were dichotomized according to recanalization information. Mean quantitative T(2)-values were derived for each patient within each region of interest. Mean T(2)-values for patients with recanalization (n = 15) in surviving tissue region of interest were 115.8 ± 7.2 ms (mean ± SD) and in the lesion growth region of interest 114.6 ± 7.0 ms. T(2)-values for patients without recanalization (n = 7) were 117.7 ± 11.4 ms in surviving tissue region of interest and 117.3 ± 12.1 ms in lesion growth region of interest. There was no significant difference between T(2)-values measured in lesion growth and surviving tissue region of interest for patients with or without recanalization. Even though it has been shown that T(2)-values increase with time from symptom onset within the infarct core, increased T(2)-values in areas of perfusion impairment do not identify irreversible damaged brain tissue and high T(2)-values are even found in tissue that is not part of the final infarct lesion and can therefore normalize. In conclusion, this study suggests that T(2)-values are not a valid imaging biomarker in acute stroke to predict tissue outcome.
Collapse
Affiliation(s)
- Susanne Siemonsen
- Department of Diagnostic and Interventional Neuroradiology, University Medical Centre Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
Here we describe MRI and (1)H MRS protocols for the investigation of animal models (mainly mice and rats) of psychiatric disorders. The introduction provides general findings from brain imaging studies in patients with psychiatric diseases and refers to general rules regarding the use of animal models in research. The methods section includes a selection of basic 9.4 T MRI and MRS protocols applicable for the investigation of animal models of psychiatric disorders (T1W, T2W, FLAIR, (1)H MRS). The notes section discusses in detail a series of factors that can influence the outcome of the experiment: from animal handling, stress-triggering aspects, and experimental design-related factors to technical aspects that affect T (1) and T (2) measurements.
Collapse
Affiliation(s)
- Dana S Poole
- Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| | | | | |
Collapse
|
30
|
Thomalla G, Cheng B, Ebinger M, Hao Q, Tourdias T, Wu O, Kim JS, Breuer L, Singer OC, Warach S, Christensen S, Treszl A, Forkert ND, Galinovic I, Rosenkranz M, Engelhorn T, Köhrmann M, Endres M, Kang DW, Dousset V, Sorensen AG, Liebeskind DS, Fiebach JB, Fiehler J, Gerloff C. DWI-FLAIR mismatch for the identification of patients with acute ischaemic stroke within 4·5 h of symptom onset (PRE-FLAIR): a multicentre observational study. Lancet Neurol 2011; 10:978-86. [PMID: 21978972 DOI: 10.1016/s1474-4422(11)70192-2] [Citation(s) in RCA: 386] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Many patients with stroke are precluded from thrombolysis treatment because the time from onset of their symptoms is unknown. We aimed to test whether a mismatch in visibility of an acute ischaemic lesion between diffusion-weighted MRI (DWI) and fluid-attenuated inversion recovery (FLAIR) MRI (DWI-FLAIR mismatch) can be used to detect patients within the recommended time window for thrombolysis. METHODS In this multicentre observational study, we analysed clinical and MRI data from patients presenting between Jan 1, 2001, and May 31, 2009, with acute stroke for whom DWI and FLAIR were done within 12 h of observed symptom onset. Two neurologists masked to clinical data judged the visibility of acute ischaemic lesions on DWI and FLAIR imaging, and DWI-FLAIR mismatch was diagnosed by consensus. We calculated predictive values of DWI-FLAIR mismatch for the identification of patients with symptom onset within 4·5 h and within 6 h and did multivariate regression analysis to identify potential confounding covariates. This study is registered with ClinicalTrials.gov, number NCT01021319. FINDINGS The final analysis included 543 patients. Mean age was 66·0 years (95% CI 64·7-67·3) and median National Institutes of Health Stroke Scale score was 8 (IQR 4-15). Acute ischaemic lesions were identified on DWI in 516 patients (95%) and on FLAIR in 271 patients (50%). Interobserver agreement for acute ischaemic lesion visibility on FLAIR imaging was moderate (κ=0·569, 95% CI 0·504-0·634). DWI-FLAIR mismatch identified patients within 4·5 h of symptom onset with 62% (95% CI 57-67) sensitivity, 78% (72-84) specificity, 83% (79-88) positive predictive value, and 54% (48-60) negative predictive value. Multivariate regression analysis identified a longer time to MRI (p<0·0001), a lower age (p=0·0009), and a larger DWI lesion volume (p=0·0226) as independent predictors of lesion visibility on FLAIR imaging. INTERPRETATION Patients with an acute ischaemic lesion detected with DWI but not with FLAIR imaging are likely to be within a time window for which thrombolysis is safe and effective. These findings lend support to the use of DWI-FLAIR mismatch for selection of patients in a future randomised trial of thrombolysis in patients with unknown time of symptom onset. FUNDING Else Kröner-Fresenius-Stiftung, National Institutes of Health.
Collapse
Affiliation(s)
- Götz Thomalla
- Klinik und Poliklinik für Neurologie, Kopf- und Neurozentrum, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Del Bigio MR, Slobodian I, Schellenberg AE, Buist RJ, Kemp-Buors TL. Magnetic resonance imaging indicators of blood-brain barrier and brain water changes in young rats with kaolin-induced hydrocephalus. Fluids Barriers CNS 2011; 8:22. [PMID: 21834998 PMCID: PMC3162928 DOI: 10.1186/2045-8118-8-22] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Accepted: 08/11/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Hydrocephalus is associated with enlargement of cerebral ventricles. We hypothesized that magnetic resonance (MR) imaging parameters known to be influenced by tissue water content would change in parallel with ventricle size in young rats and that changes in blood-brain barrier (BBB) permeability would be detected. METHODS Hydrocephalus was induced by injection of kaolin into the cisterna magna of 4-week-old rats, which were studied 1 or 3 weeks later. MR was used to measure longitudinal and transverse relaxation times (T1 and T2) and apparent diffusion coefficients in several regions. Brain tissue water content was measured by the wet-dry weight method, and tissue density was measured in Percoll gradient columns. BBB permeability was measured by quantitative imaging of changes on T1-weighted images following injection of gadolinium diethylenetriamine penta-acetate (Gd-DTPA) tracer and microscopically by detection of fluorescent dextran conjugates. RESULTS In nonhydrocephalic rats, water content decreased progressively from age 3 to 7 weeks. T1 and T2 and apparent diffusion coefficients did not exhibit parallel changes and there was no evidence of BBB permeability to tracers. The cerebral ventricles enlarged progressively in the weeks following kaolin injection. In hydrocephalic rats, the dorsal cortex was more dense and the white matter less so, indicating that the increased water content was largely confined to white matter. Hydrocephalus was associated with transient elevation of T1 in gray and white matter and persistent elevation of T2 in white matter. Changes in the apparent diffusion coefficients were significant only in white matter. Ventricle size correlated significantly with dorsal water content, T1, T2, and apparent diffusion coefficients. MR imaging showed evidence of Gd-DTPA leakage in periventricular tissue foci but not diffusely. These correlated with microscopic leak of larger dextran tracers. CONCLUSIONS MR characteristics cannot be used as direct surrogates for water content in the immature rat model of hydrocephalus, probably because they are also influenced by other changes in tissue composition that occur during brain maturation. There is no evidence for widespread persistent opening of BBB as a consequence of hydrocephalus in young rats. However, increase in focal BBB permeability suggests that periventricular blood vessels may be disrupted.
Collapse
Affiliation(s)
- Marc R Del Bigio
- Department of Pathology, University of Manitoba; 401 Brodie Centre, 727 McDermot Avenue, Winnipeg MB R3E 3P5 Canada.
| | | | | | | | | |
Collapse
|
32
|
Quantitative proton MRI and MRS of the rat brain with a 3T clinical MR scanner. J Neuroradiol 2011; 38:90-7. [DOI: 10.1016/j.neurad.2009.11.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2009] [Revised: 11/03/2009] [Accepted: 11/13/2009] [Indexed: 11/21/2022]
|
33
|
Petkova M, Rodrigo S, Lamy C, Oppenheim G, Touzé E, Mas JL, Méder JF, Oppenheim C. MR Imaging Helps Predict Time from Symptom Onset in Patients with Acute Stroke: Implications for Patients with Unknown Onset Time. Radiology 2010; 257:782-92. [DOI: 10.1148/radiol.10100461] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
34
|
Leithner C, Müller S, Füchtemeier M, Lindauer U, Dirnagl U, Royl G. Determination of the brain-blood partition coefficient for water in mice using MRI. J Cereb Blood Flow Metab 2010; 30:1821-4. [PMID: 20842161 PMCID: PMC3023928 DOI: 10.1038/jcbfm.2010.160] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cerebral blood flow (CBF) quantification is a valuable tool in stroke research. Mice are of special interest because of the potential of genetic engineering. Magnetic resonance imaging (MRI) provides repetitive, noninvasive CBF quantification. Many MRI techniques require the knowledge of the brain-blood partition coefficient (BBPC) for water. Adopting an MRI protocol described by Roberts et al (1996) in humans, we determined the BBPC for water in 129S6/SvEv mice from proton density measurements of brain and blood, calibrated with deuterium oxide/water phantoms. The average BBPC for water was 0.89 ± 0.03 mL/g, with little regional variation within the mouse brain.
Collapse
Affiliation(s)
- Christoph Leithner
- Department of Experimental Neurology, Charité-Universitätsmedizin, Center for Stroke Research Berlin, Berlin, Germany.
| | | | | | | | | | | |
Collapse
|
35
|
Overgaard-Steensen C, Stødkilde-Jørgensen H, Larsson A, Broch-Lips M, Tønnesen E, Frøkiaer J, Ring T. Regional differences in osmotic behavior in brain during acute hyponatremia: an in vivo MRI-study of brain and skeletal muscle in pigs. Am J Physiol Regul Integr Comp Physiol 2010; 299:R521-32. [PMID: 20445159 DOI: 10.1152/ajpregu.00139.2010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Brain edema is suggested to be the principal mechanism underlying the symptoms in acute hyponatremia. Identification of the mechanisms responsible for global and regional cerebral water homeostasis during hyponatremia is, therefore, of utmost importance. To examine the osmotic behavior of different brain regions and muscles, in vivo-determined water content (WC) was related to plasma sodium concentration ([Na(+)]) and brain/muscle electrolyte content. Acute hyponatremia was induced with desmopressin acetate and infusion of a 2.5% glucose solution in anesthetized pigs. WC in different brain regions and skeletal muscle was estimated in vivo from T(1) maps determined by magnetic resonance imaging (MRI). WC, expressed in gram water per 100 g dry weight, increased significantly in slices of the whole brain [342(SD = 14) to 363(SD = 21)] (6%), thalamus [277(SD = 13) to 311(SD = 24)] (12%) and white matter [219(SD = 7) to 225(SD = 5)] (3%). However, the WC increase in the whole brain and white mater WC was less than expected from perfect osmotic behavior, whereas in the thalamus, the water increase was as expected. Brain sodium content was significantly reduced. Muscle WC changed passively with plasma [Na(+)]. WC determined with deuterium dilution and tissue lyophilzation correlated well with MRI-determined WC. In conclusion, acute hyponatremia induces brain and muscle edema. In the brain as a whole and in the thalamus, regulatory volume decrease (RVD) is unlikely to occur. However, RVD may, in part, explain the observed lower WC in white matter. This may play a potential role in osmotic demyelination.
Collapse
|
36
|
Heinzer-Schweizer S, De Zanche N, Pavan M, Mens G, Sturzenegger U, Henning A, Boesiger P. In-vivo assessment of tissue metabolite levels using 1H MRS and the Electric REference To access In vivo Concentrations (ERETIC) method. NMR IN BIOMEDICINE 2010; 23:406-413. [PMID: 20101606 DOI: 10.1002/nbm.1476] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2008] [Revised: 10/19/2009] [Accepted: 10/19/2009] [Indexed: 05/28/2023]
Abstract
Quantitative values of metabolite concentrations in (1)H magnetic resonance spectroscopy have been obtained using the Electric REference To access In vivo Concentrations (ERETIC) method, whereby a synthetic reference signal is injected during the acquisition of spectra. The method has been improved to enable quantification of metabolite concentrations in vivo. Optical signal transmission was used to eliminate random fluctuations in ERETIC signal coupling to the receiver coil due to changes in position of cables and highly dielectric human tissue. Stability and reliability of the signal were tested in vitro, achieving stability with a mean error of 2.83%. Scaling of the signal in variable loading conditions was demonstrated and in-vivo measurements of brain were acquired on a 3T Philips system using a transmit/receive coil. The quantitative brain water and metabolite concentration values are in good agreement with those in the literature.
Collapse
Affiliation(s)
- S Heinzer-Schweizer
- Institute for Biomedical Engineering, University and ETH Zürich, Zürich, Switzerland.
| | | | | | | | | | | | | |
Collapse
|
37
|
Chuang KH, Koretsky AP, Sotak CH. Temporal changes in the T1 and T2 relaxation rates (DeltaR1 and DeltaR2) in the rat brain are consistent with the tissue-clearance rates of elemental manganese. Magn Reson Med 2009; 61:1528-32. [PMID: 19353652 DOI: 10.1002/mrm.21962] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Temporal changes in the T(1) and T(2) relaxation rates (DeltaR(1) and DeltaR(2)) in rat olfactory bulb (OB) and cortex were compared with the absolute manganese (Mn) concentrations from the corresponding excised tissue samples. In vivo T(1) and T(2) relaxation times were measured before, and at 1, 7, 28, and 35 d after intravenous infusion of 176 mg/kg MnCl(2). The values of DeltaR(1), DeltaR(2), and absolute Mn concentration peaked at day 1 and then declined to near control levels after 28 to 35 d. The Mn bioelimination rate from the rat brain was significantly faster than that reported using radioisotope techniques. The R(1) and R(2) relaxation rates were linearly proportional to the underlying tissue Mn concentration and reflect the total absolute amount of Mn present in the tissue. The in vivo Mn r(1) and r(2) tissue relaxivities were comparable to the in vitro values for aqueous Mn(2+). These results demonstrate that loss of manganese-enhanced MRI (MEMRI) contrast after systemic Mn(2+) administration is due to elimination of Mn(2+) from the brain.
Collapse
Affiliation(s)
- Kai-Hsiang Chuang
- Laboratory of Functional and Molecular Imaging, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | | | | |
Collapse
|
38
|
Thomalla G, Rossbach P, Rosenkranz M, Siemonsen S, Krützelmann A, Fiehler J, Gerloff C. Negative fluid-attenuated inversion recovery imaging identifies acute ischemic stroke at 3 hours or less. Ann Neurol 2009; 65:724-32. [PMID: 19557859 DOI: 10.1002/ana.21651] [Citation(s) in RCA: 174] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE To evaluate the use of fluid-attenuated inversion recovery (FLAIR) imaging as surrogate marker of lesion age within the first 6 hours of ischemic stroke. METHODS e analyzed FLAIR and diffusion-weighted imaging (DWI) sequences performed within 6 hours of symptom onset in 120 consecutive patients with ischemic stroke with known symptom onset. The visibility of acute ischemic lesions on FLAIR images was judged in two steps (on FLAIR alone and with knowledge of DWI) and compared with DWI. RESULTS egative FLAIR in the case of positive DWI allocated ischemic lesions to a time window 3 hours or less with a high specificity (0.93) and a high positive predictive value (0.94), whereas sensitivity (0.48) and negative predictive value (0.43) were low. Lesion visibility on FLAIR images alone (35.6%) and with knowledge of DWI (62.5%) was lower than on DWI (97.1%). The sensitivity of FLAIR increased with increasing time from symptom onset from 27.0/50.0% <or= 3 hours to 56.7/93.3% after 3 to 6 hours (FLAIR alone/with knowledge of DWI). Multivariate regression analysis spotted longer time from symptom onset and larger size of the ischemic lesion as independent predictors of lesion visibility on FLAIR images. INTERPRETATION "mismatch" between positive DWI and negative FLAIR allows the identification of patients that are highly likely to be within the 3-hour time window. Within the first 6 hours of stroke, the sensitivity of FLAIR sequences for acute ischemic lesions increases with time from symptom onset elapsing, approximating 100% after 3 to 6 hours.
Collapse
Affiliation(s)
- Götz Thomalla
- Department of Neurology, Center for Clinical Neurosciences, University Medical Center Hamburg, Eppendorf, Hamburg, Germany.
| | | | | | | | | | | | | |
Collapse
|
39
|
Siemonsen S, Mouridsen K, Holst B, Ries T, Finsterbusch J, Thomalla G, Ostergaard L, Fiehler J. Quantitative T2 Values Predict Time From Symptom Onset in Acute Stroke Patients. Stroke 2009; 40:1612-6. [PMID: 19325153 DOI: 10.1161/strokeaha.108.542548] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background and Purpose—
We hypothesize that in comparison to diffusion-weighted imaging, quantitative T2 values (qT2) are more directly related to water uptake in ischemic tissue, depending on time from symptom onset. We measured the increase of qT2 in the infarct core to quantify the correlation between time from symptom onset and change in qT2.
Methods—
Thirty-six patients with acute ischemic stroke in the territory of the proximal middle cerebral artery underwent MRI including diffusion-weighted imaging, fluid-attenuated inversion recovery, and a triple-echo T2 sequence (calculation of T2 maps) within 6 hours after symptom onset. Regions of decreased apparent diffusion coefficient <550×10
−9
mm
2
/sec were defined and superimposed onto the corresponding T2 map and the unaffected side in the horizontally flipped maps. Differences of T2/apparent diffusion coefficient values between affected and unaffected side were calculated (differences of T2/differences of apparent diffusion coefficient). Fluid-attenuated inversion recovery images were rated for lesion visibility.
Results—
Differences of T2 showed a significant correlation with time from symptom onset (
R
=0.580;
P
<0.001). T2 values measured in patients with visible fluid-attenuated inversion recovery lesions were significantly higher than in those without visible hyperintensity (
P
<0.001). The accuracy of qT2 to predict a time from symptom onset <3 hours was 0.794, whereas the corresponding accuracy for visual assessment of fluid-attenuated inversion recovery images was 0.676.
Conclusions—
T2 values demonstrated a strong correlation with time from onset, suggesting different pathophysiologic mechanisms than diffusion restriction. Whereas fluid-attenuated inversion recovery only provides binary information on lesion visibility, T2 values correlate well with time from symptom onset, and are free from operator bias, increasing reproducibility to determine time from symptom onset.
Collapse
Affiliation(s)
- Susanne Siemonsen
- From the Department of Neuroradiology (S.S., B.H., T.R., J. Fiehler), Systems Neuroscience (J. Finsterbusch), and Department of Neurology (G.T.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Center for Functionally Integrative Neuroscience (CFIN) (K.M., L.O.), Department of Neuroradiology, Aarhus University Hospital, Denmark
| | - Kim Mouridsen
- From the Department of Neuroradiology (S.S., B.H., T.R., J. Fiehler), Systems Neuroscience (J. Finsterbusch), and Department of Neurology (G.T.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Center for Functionally Integrative Neuroscience (CFIN) (K.M., L.O.), Department of Neuroradiology, Aarhus University Hospital, Denmark
| | - Brigitte Holst
- From the Department of Neuroradiology (S.S., B.H., T.R., J. Fiehler), Systems Neuroscience (J. Finsterbusch), and Department of Neurology (G.T.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Center for Functionally Integrative Neuroscience (CFIN) (K.M., L.O.), Department of Neuroradiology, Aarhus University Hospital, Denmark
| | - Thorsten Ries
- From the Department of Neuroradiology (S.S., B.H., T.R., J. Fiehler), Systems Neuroscience (J. Finsterbusch), and Department of Neurology (G.T.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Center for Functionally Integrative Neuroscience (CFIN) (K.M., L.O.), Department of Neuroradiology, Aarhus University Hospital, Denmark
| | - Jürgen Finsterbusch
- From the Department of Neuroradiology (S.S., B.H., T.R., J. Fiehler), Systems Neuroscience (J. Finsterbusch), and Department of Neurology (G.T.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Center for Functionally Integrative Neuroscience (CFIN) (K.M., L.O.), Department of Neuroradiology, Aarhus University Hospital, Denmark
| | - Götz Thomalla
- From the Department of Neuroradiology (S.S., B.H., T.R., J. Fiehler), Systems Neuroscience (J. Finsterbusch), and Department of Neurology (G.T.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Center for Functionally Integrative Neuroscience (CFIN) (K.M., L.O.), Department of Neuroradiology, Aarhus University Hospital, Denmark
| | - Leif Ostergaard
- From the Department of Neuroradiology (S.S., B.H., T.R., J. Fiehler), Systems Neuroscience (J. Finsterbusch), and Department of Neurology (G.T.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Center for Functionally Integrative Neuroscience (CFIN) (K.M., L.O.), Department of Neuroradiology, Aarhus University Hospital, Denmark
| | - Jens Fiehler
- From the Department of Neuroradiology (S.S., B.H., T.R., J. Fiehler), Systems Neuroscience (J. Finsterbusch), and Department of Neurology (G.T.), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Center for Functionally Integrative Neuroscience (CFIN) (K.M., L.O.), Department of Neuroradiology, Aarhus University Hospital, Denmark
| |
Collapse
|
40
|
Pedersen M, Vajda Z, Stødkilde-Jørgensen H, Nielsen S, Frøkiaer J. Furosemide increases water content in renal tissue. Am J Physiol Renal Physiol 2007; 292:F1645-51. [PMID: 17264309 DOI: 10.1152/ajprenal.00060.2006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The present study was designed to evaluate the short-term effects of intravenous administration of furosemide on key functions in the kidney cortex and the outer and inner medulla of rats by using magnetic resonance imaging (MRI). Renal tissue water content, renal tissue oxygenation (in relation to the magnetic resonance spin-spin relaxation rate), the apparent diffusion coefficient (ADC) of water, and volume of renal blood flow were measured. Furosemide administration resulted in an increased water content in all regions of the kidney. In parallel with this, we found a significant reduction in ADC in the cortex (2.7 +/- 0.1 x 10(-3) to 2.3 +/- 0.1 x 10(-3) mm(2)/s; P < 0.01) and in the outer medulla (2.3 +/- 0.1 x 10(-3) to 2.0 +/- 0.1 x 10(-3) mm(2)/s; P < 0.01), indicating that the intra- to extracellular volume fraction of water increased in response to furosemide administration. Furosemide also decreased the blood oxygenation in the cortex (49.1 +/- 2.9 to 40.9 +/- 2.0 s(-1); P < 0.01), outer medulla (41.9 +/- 2.8 to 33.2 +/- 1.6 s(-1); P < 0.01) and in the inner medulla (37.1 +/- 2.9 to 26.7 +/- 1.8 s(-1); P < 0.01), indicating an increased amount of oxygenated Hb in the renal tissue. Moreover, renal blood flow decreased in response to furosemide (6.9 +/- 0.2 to 4.4 +/- 0.2 ml/min; P < 0.001). In conclusion, furosemide administration was associated with increased renal water content, an increase in the intra- to extracellular volume fraction of water, an increased oxygen tension, and a decrease in the renal blood flow. Thus MRI provides an integrated evaluation of changes in renal function, leading to decreased renal water and solute reabsorption in response to furosemide, and, in addition, MRI provides an alternative tool to monitor noninvasively changes at the cellular level.
Collapse
Affiliation(s)
- Michael Pedersen
- MR Research Centre, Aarhus University Hospital, University of Aarhus, Aarhus, Denmark
| | | | | | | | | |
Collapse
|
41
|
Samson RS, Thornton JS, McLean MA, Williams SCR, Tofts PS. 1H-MRS internal thermometry in test-objects (phantoms) to within 0.1 K for quality assurance in long-term quantitative MR studies. NMR IN BIOMEDICINE 2006; 19:560-5. [PMID: 16612806 DOI: 10.1002/nbm.1033] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Many magnetic resonance test-object properties are temperature-dependent, with typical temperature coefficients of approximately 2-3% K(-1). Therefore, to achieve consistent quality assurance measurements to within 1%, test object temperatures should ideally be known to within 0.3 K. Proton magnetic resonance spectroscopy has previously been used to estimate accurately absolute tissue temperature in vivo, based on the linear temperature dependence of the chemical shift difference between water and temperature-stable reference metabolites such as N-acetylaspartate. In this study, this method of 'internal thermometry' in quality assurance test-objects was investigated, and in particular the value of sodium 3-(trimethylsilyl)propane-1-sulfonate (DSS) as a chemical shift reference was demonstrated. The relationship between the DSS-water chemical shift difference (sigma, expressed in ppm) and temperature tau (in K) was shown to be tau = 764.55 (+/-5.05) - 97.72 (+/-1.05) sigma (286 <or= tau <or= 309 K). Internal thermometry in MRI test-objects is feasible and straightforward, using readily available (1)H-MRS pulse sequences and standard spectroscopy evaluation packages, with a minimum detectable temperature difference of 100 (+/-20) mK.
Collapse
Affiliation(s)
- R S Samson
- Department of Neuroinflammation, NMR Research Unit, Institute of Neurology, University College London, Queen Square, London WC1N 3BG, UK.
| | | | | | | | | |
Collapse
|
42
|
Tailor DR, Baumgardner JE, Regatte RR, Leigh JS, Reddy R. Proton MRI of metabolically produced H2 17O using an efficient 17O2 delivery system. Neuroimage 2004; 22:611-8. [PMID: 15193589 DOI: 10.1016/j.neuroimage.2004.02.024] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2003] [Revised: 02/11/2004] [Accepted: 02/12/2004] [Indexed: 11/16/2022] Open
Abstract
In vivo detection of H(2)(17)O produced via metabolic reduction of inhaled (17)O-enriched gas is demonstrated using proton magnetic resonance imaging (MRI). Specifically, (1)H T(1rho)-weighted MRI, which may be readily implemented on any MRI scanner, is applied as an indirect (17)O imaging method to quantitatively monitor the distribution of metabolically produced (17)O water (mpH(2)(17)O) in the rat brain. The delivery of (17)O(2) to rats is conducted via a specially designed closed respiration circuit that conserves the expensive gas. Quantitative mapping of H(2)(17)O performed via (1)H T(1rho)-weighted MRI is validated by direct (17)O-magnetic resonance spectroscopy. The MRI data show that a steady-state H(2)(17)O concentration of 25.7 +/- 1.66 mM (n = 4) is achieved in the rat brain within approximately 30 min under the (17)O inhalation paradigm used. From the first minute of the mpH(2)(17)O time courses, cerebral metabolic rate of oxygen (CMRO(2)) is estimated to be 2.10 +/- 0.44 micromol g(-1) min(-1) (n = 4), a value that is consistent with the literature.
Collapse
Affiliation(s)
- Dharmesh R Tailor
- University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA.
| | | | | | | | | |
Collapse
|
43
|
Tailor DR, Roy A, Regatte RR, Charagundla SR, McLaughlin AC, Leigh JS, Reddy R. Indirect 17(O)-magnetic resonance imaging of cerebral blood flow in the rat. Magn Reson Med 2003; 49:479-87. [PMID: 12594750 DOI: 10.1002/mrm.10403] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Proton T(1rho)-dispersion MRI is demonstrated for indirect, in vivo detection of (17)O in the brain. This technique, which may be readily implemented on any clinical MRI scanner, is applied towards high-resolution, quantitative mapping of cerebral blood flow (CBF) in the rat by monitoring the clearance of (17)O-enriched water. Strategies are derived and employed for 1) quantitation of absolute H(2) (17)O tracer concentration from a ratio of high- and low-frequency spin-locked T(1rho) images, and 2) mapping CBF without having to transform the T(1rho) signal to H(2) (17)O tracer concentration. Absolute regional blood flow was mapped in a single 3-mm brain slice at an in-plane resolution of 0.4 x 0.8 mm within a 5-min tracer washout time; these data are consistent with the less localized CBF measurements reported in the literature. T(1rho)-weighted imaging yields excellent signal-to-noise ratios, spatiotemporal resolution, and anatomical contrast for mapping CBF.
Collapse
Affiliation(s)
- Dharmesh R Tailor
- Metabolic Magnetic Resonance Research and Computing Center, Department of Radiology, University of Pennsylvania, Philadelphia 19104, USA.
| | | | | | | | | | | | | |
Collapse
|
44
|
Benveniste H, Blackband S. MR microscopy and high resolution small animal MRI: applications in neuroscience research. Prog Neurobiol 2002; 67:393-420. [PMID: 12234501 DOI: 10.1016/s0301-0082(02)00020-5] [Citation(s) in RCA: 168] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The application of magnetic resonance (MR) imaging in the study of human disease using small animals has steadily evolved over the past two decades and strongly established the fields of "small animal MR imaging" and "MR microscopy." An increasing number of neuroscience related investigations now implement MR microscopy in their experiments. Research areas of growth pertaining to MR microscopy studies are focused on (1). phenotyping of genetically engineered mice models of human neurological diseases and (2). rodent brain atlases. MR microscopy can be performed in vitro on tissue specimens, ex vivo on brain slice preparations and in vivo (typically on rodents). Like most new imaging technologies, MR microscopy is technologically demanding and requires broad expertise. Uniform guidelines or "standards" of a given MR microscopy experiment are non-existent. The main focus therefore of this review will be on biological applications of MR microscopy and the experimental requirements. We also take a critical look at the biological information that small animal (rodent) MR imaging has provided in neuroscience research.
Collapse
Affiliation(s)
- Helene Benveniste
- Medical Department, Brookhaven National Laboratory, Building 490, 30 Bell Avenue, Upton, NY 11793, USA.
| | | |
Collapse
|
45
|
Kucinski T, Väterlein O, Glauche V, Fiehler J, Klotz E, Eckert B, Koch C, Röther J, Zeumer H. Correlation of apparent diffusion coefficient and computed tomography density in acute ischemic stroke. Stroke 2002; 33:1786-91. [PMID: 12105353 DOI: 10.1161/01.str.0000019125.80118.99] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Diffusion-weighted MR imaging is very sensitive for the detection of restricted molecular water diffusion in acute ischemic stroke. CT is sensitive to net water uptake in ischemic edema. We compared the decrease in the apparent diffusion coefficient (ADC) in diffusion-weighted MR imaging with CT density changes to study the correlation between diffusion restriction and water uptake in acute stroke patients. METHODS Twenty-five patients with acute ischemic stroke of the anterior cerebral circulation underwent MR and CT imaging 1.3 to 5.4 hours after symptom onset. ADC and CT data were transferred into a common 3-dimensional space, and regions of decreased ADC (dADC) were superimposed onto the corresponding CT. Mean values of ADC and Hounsfield units (HU) were determined in comparison with the nonaffected hemisphere. RESULTS Mean decrease in ADC (dADC) was 170+/-53x 10(-6) mm2/s and corresponded to a decrease (dCT) in CT density of 1.3+/-0.7 HU. dCT showed a continuous linear decrease of 0.4 HU/h (r=0.55, P<0.01), whereas the decrease is ADC was almost complete after 1.5 hours. A correlation between the decrease in ADC and dCT was found (r=0.41, P=0.04). CONCLUSIONS The severity of diffusion restriction correlates with net water uptake in acute ischemic stroke. However, the underlying pathophysiology and different time courses indicate a common reason rather than a direct causality for both phenomena. The time delay and low value of CT density changes provide a reasonable explanation for the higher sensitivity of MR imaging in ischemic stroke.
Collapse
Affiliation(s)
- Thomas Kucinski
- Department of Neuroradiology, University-Hospital Hamburg Eppendorf, Hamburg, Germany. kucinski@ uke.uni-hanburg.de
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Gutteridge S, Ramanathan C, Bowtell R. Mapping the absolute value of M0 using dipolar field effects. Magn Reson Med 2002; 47:871-9. [PMID: 11979565 DOI: 10.1002/mrm.10142] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The ability to map the spatial variation of the absolute, rather than the relative value of the equilibrium magnetization could be advantageous in many areas of NMR. However, direct measurement of M(0) is usually difficult because of the multiparametric dependence of the NMR signal. Here we propose a technique for mapping the spatial variation of the absolute value of M(0), independent of relaxation weighting and flip angle calibration. This method, which works best at high field strengths, is based on the effect of the dipolar field due to the nuclear magnetization that is normally neglected in liquid-state NMR. The experimental implementation of this sequence at 3.0 T is described, and its initial application to the measurement of the water content of brain tissue is outlined.
Collapse
Affiliation(s)
- S Gutteridge
- Magnetic Resonance Centre, School of Physics and Astronomy, University of Nottingham, Nottingham, UK
| | | | | |
Collapse
|
47
|
Schwarcz A, Berente Z, Osz E, Dóczi T. In vivo water quantification in mouse brain at 9.4 Tesla in a vasogenic edema model. Magn Reson Med 2001; 46:1246-9. [PMID: 11746594 DOI: 10.1002/mrm.1324] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The aim of our study was to establish a simple in vivo method for water quantification in vasogenic edema, and provide data on imaging of mouse brain at 9.4 Tesla. Apparent T1 and spin density values determined by MRI were found to strongly correlate with the gravimetric water content of mouse brain undergoing cold injury. Using a two-point calibration line between the spin density values for pure water and cortex of mouse brain, as well as the corresponding water contents in vivo, water could be quantified with satisfactory accuracy.
Collapse
Affiliation(s)
- A Schwarcz
- Department of Neurosurgery, University of Pécs, Pécs, Hungary
| | | | | | | |
Collapse
|