1
|
Yi T, Liu Y, Wei W, He S, Jin K. Microstructural abnormalities of the right hemisphere in preschool autism spectrum disorders. J Psychiatr Res 2024; 180:258-264. [PMID: 39454493 DOI: 10.1016/j.jpsychires.2024.10.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 10/14/2024] [Accepted: 10/19/2024] [Indexed: 10/28/2024]
Abstract
BACKGROUND This study aims to investigate microstructural abnormalities within and between hemispheres in preschool children with autism spectrum disorders (ASD) using diffusion basis spectrum imaging (DBSI). METHODS A total of 35 ASD patients and 32 healthy controls (HC), matched for sex and age, underwent DBSI at 3T. We analyzed DBSI-derived indices of brain white matter using tract-based spatial statistics (TBSS) to compare ASD and HC groups. Support vector machine (SVM) classification was employed to evaluate the potential of positive DBSI parameters in distinguishing ASD patients. Additionally, correlation analyses were conducted to explore relationships between positive DBSI parameters and clinical scales. RESULTS Patients in the ASD group exhibited significantly higher fiber ratios in the right brainstem tracts, increased radial diffusivity in the left superior longitudinal fasciculus, and reduced fractional anisotropy (FA) in various fiber tracts, including projection, commissural, and association fibers, compared to HC. Notably, the FA of the right cingulum correlated positively with the Gesell scale (r = 0.439, p = 0.008) and achieved a specificity of 90% in identifying ASD. CONCLUSION The DBSI findings suggest asynchronous myelination in the right hemisphere and cerebellum in preschool ASD, with the FA value of the right cingulate gyrus appearing to be a reliable marker for ASD and may serve as a potential diagnostic parameter for preschool ASD.
Collapse
Affiliation(s)
- Ting Yi
- Department of Radiology, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan Children's Hospital) , 410007, Changsha, China
| | - Yuqing Liu
- Department of Radiology, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan Children's Hospital) , 410007, Changsha, China
| | - Weian Wei
- Department of Radiology, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan Children's Hospital) , 410007, Changsha, China
| | - Siping He
- Department of Radiology, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan Children's Hospital) , 410007, Changsha, China
| | - Ke Jin
- Department of Radiology, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University (Hunan Children's Hospital) , 410007, Changsha, China.
| |
Collapse
|
2
|
Mamah D, Patel A, Chen S, Wang Y, Wang Q. Diffusion Basis Spectrum Imaging of White Matter in Schizophrenia and Bipolar Disorder. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.07.602402. [PMID: 39005300 PMCID: PMC11245098 DOI: 10.1101/2024.07.07.602402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Background Multiple studies point to the role of neuroinflammation in the pathophysiology of schizophrenia (SCZ), however, there have been few in vivo tools for imaging brain inflammation. Diffusion basis spectrum imaging (DBSI) is an advanced diffusion-based MRI method developed to quantitatively assess microstructural alternations relating to neuroinflammation, axonal fiber, and other white matter (WM) pathologies. Methods We acquired one-hour-long high-directional diffusion MRI data from young control (CON, n = 27), schizophrenia (SCZ, n = 21), and bipolar disorder (BPD, n = 21) participants aged 18-30. We applied Tract-based Spatial Statistics (TBSS) to allow whole-brain WM analyses and compare DBSI-derived isotropic and anisotropic diffusion measures between groups. Clinical relationships of DBSI metrics with clinical symptoms were assessed across SCZ and control participants. Results In SCZ participants, we found a generalized increase in DBSI-derived cellularity (a putative marker of neuroinflammation), a decrease in restricted fiber fraction (a putative marker of apparent axonal density), and an increase in extra-axonal water (a putative marker of vasogenic edema) across several WM tracts. There were only minimal WM abnormalities noted in BPD, mainly in regions of the corpus callosum (increase in DTI-derived RD and extra-axonal water). DBSI metrics showed significant partial correlations with psychosis and mood symptoms across groups. Conclusion Our findings suggest that SCZ involves generalized white matter neuroinflammation, decreased fiber density, and demyelination, which is not seen in bipolar disorder. Larger studies are needed to identify medication-related effects. DBSI metrics could help identify high-risk groups requiring early interventions to prevent the onset of psychosis and improve outcomes.
Collapse
Affiliation(s)
- Daniel Mamah
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri
| | - Aakash Patel
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri
| | - ShingShiun Chen
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri
| | - Yong Wang
- Departments of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri
- Department of Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
- Department of Biomedical Engineering, Washington University School of Medicine, St. Louis, Missouri
- Mechanical Engineering and Materials Science, Washington University School of Medicine, St. Louis, Missouri
| | - Qing Wang
- Department of Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
- Department of Biomedical Engineering, Washington University School of Medicine, St. Louis, Missouri
- Mechanical Engineering and Materials Science, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
3
|
Jin C, Wang K, Ren Y, Li Y, Wang Z, Cheng L, Xie N. Role of durotomy on function outcome, tissue sparing, inflammation, and tissue stiffness after spinal cord injury in rats. MedComm (Beijing) 2024; 5:e530. [PMID: 38576458 PMCID: PMC10993870 DOI: 10.1002/mco2.530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 01/18/2024] [Accepted: 02/02/2024] [Indexed: 04/06/2024] Open
Abstract
Currently, there is a lack of effective treatments for spinal cord injury (SCI), a debilitating medical condition associated with enduring paralysis and irreversible neuronal damage. Extradural decompression of osseous as well as soft tissue components has historically been the principal objective of surgical procedures. Nevertheless, this particular surgical procedure fails to tackle the intradural compressive alterations that contribute to secondary SCI. Here, we propose an early intrathecal decompression strategy and evaluate its role on function outcome, tissue sparing, inflammation, and tissue stiffness after SCI. Durotomy surgery significantly promoted recovery of hindlimb locomotor function in an open-field test. Radiological analysis suggested that lesion size and tissue edema were significantly reduced in animals that received durotomy. Relative to the group with laminectomy alone, the animals treated with a durotomy had decreased cavitation, scar formation, and inflammatory responses at 4 weeks after SCI. An examination of the mechanical properties revealed that durotomy facilitated an expeditious restoration of the injured tissue's elastic rigidity. In general, early decompressive durotomy could serve as a significant strategy to mitigate the impairments caused by secondary injury and establish a more conducive microenvironment for prospective cellular or biomaterial transplantation.
Collapse
Affiliation(s)
- Chen Jin
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of EducationOrthopaedic Department of Tongji HospitalSchool of MedicineTongji UniversityShanghaiChina
- Department of OrthopedicsTongren HospitalShanghai Jiaotong University School of MedicineShanghaiChina
| | - Kaiwei Wang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of EducationOrthopaedic Department of Tongji HospitalSchool of MedicineTongji UniversityShanghaiChina
| | - Yilong Ren
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of EducationOrthopaedic Department of Tongji HospitalSchool of MedicineTongji UniversityShanghaiChina
- Department of OrthopedicsShanghai General HospitalShanghai Jiaotong University School of MedicineShanghaiChina
| | - Yi Li
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of EducationOrthopaedic Department of Tongji HospitalSchool of MedicineTongji UniversityShanghaiChina
| | - Zhanwei Wang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of EducationOrthopaedic Department of Tongji HospitalSchool of MedicineTongji UniversityShanghaiChina
| | - Liming Cheng
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of EducationOrthopaedic Department of Tongji HospitalSchool of MedicineTongji UniversityShanghaiChina
| | - Ning Xie
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of EducationOrthopaedic Department of Tongji HospitalSchool of MedicineTongji UniversityShanghaiChina
| |
Collapse
|
4
|
Yang HC, Lavadi RS, Sauerbeck AD, Wallendorf M, Kummer TT, Song SK, Lin TH. Diffusion basis spectrum imaging detects subclinical traumatic optic neuropathy in a closed-head impact mouse model of traumatic brain injury. Front Neurol 2023; 14:1269817. [PMID: 38152638 PMCID: PMC10752006 DOI: 10.3389/fneur.2023.1269817] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 10/12/2023] [Indexed: 12/29/2023] Open
Abstract
Introduction Traumatic optic neuropathy (TON) is the optic nerve injury secondary to brain trauma leading to visual impairment and vision loss. Current clinical visual function assessments often fail to detect TON due to slow disease progression and clinically silent lesions resulting in potentially delayed or missed treatment in patients with traumatic brain injury (TBI). Methods Diffusion basis spectrum imaging (DBSI) is a novel imaging modality that can potentially fill this diagnostic gap. Twenty-two, 16-week-old, male mice were equally divided into a sham or TBI (induced by moderate Closed-Head Impact Model of Engineered Rotational Acceleration device) group. Briefly, mice were anesthetized with isoflurane (5% for 2.5 min followed by 2.5% maintenance during injury induction), had a helmet placed over the head, and were placed in a holder prior to a 2.1-joule impact. Serial visual acuity (VA) assessments, using the Virtual Optometry System, and DBSI scans were performed in both groups of mice. Immunohistochemistry (IHC) and histological analysis of optic nerves was also performed after in vivo MRI. Results VA of the TBI mice showed unilateral or bilateral impairment. DBSI of the optic nerves exhibited bilateral involvement. IHC results of the optic nerves revealed axonal loss, myelin injury, axonal injury, and increased cellularity in the optic nerves of the TBI mice. Increased DBSI axon volume, decreased DBSI λ||, and elevated DBSI restricted fraction correlated with decreased SMI-312, decreased SMI-31, and increased DAPI density, respectively, suggesting that DBSI can detect coexisting pathologies in the optic nerves of TBI mice. Conclusion DBSI provides an imaging modality capable of detecting subclinical changes of indirect TON in TBI mice.
Collapse
Affiliation(s)
- Hsin-Chieh Yang
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Raj Swaroop Lavadi
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Andrew D. Sauerbeck
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, United States
| | - Michael Wallendorf
- Department of Biostatistics, Washington University School of Medicine, St. Louis, MO, United States
| | - Terrance T. Kummer
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, United States
- VA Medical Center, St. Louis, MO, United States
| | - Sheng-Kwei Song
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, United States
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, United States
| | - Tsen-Hsuan Lin
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
5
|
Larrea A, Elexpe A, Díez-Martín E, Torrecilla M, Astigarraga E, Barreda-Gómez G. Neuroinflammation in the Evolution of Motor Function in Stroke and Trauma Patients: Treatment and Potential Biomarkers. Curr Issues Mol Biol 2023; 45:8552-8585. [PMID: 37998716 PMCID: PMC10670324 DOI: 10.3390/cimb45110539] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/25/2023] Open
Abstract
Neuroinflammation has a significant impact on different pathologies, such as stroke or spinal cord injury, intervening in their pathophysiology: expansion, progression, and resolution. Neuroinflammation involves oxidative stress, damage, and cell death, playing an important role in neuroplasticity and motor dysfunction by affecting the neuronal connection responsible for motor control. The diagnosis of this pathology is performed using neuroimaging techniques and molecular diagnostics based on identifying and measuring signaling molecules or specific markers. In parallel, new therapeutic targets are being investigated via the use of bionanomaterials and electrostimulation to modulate the neuroinflammatory response. These novel diagnostic and therapeutic strategies have the potential to facilitate the development of anticipatory patterns and deliver the most beneficial treatment to improve patients' quality of life and directly impact their motor skills. However, important challenges remain to be solved. Hence, the goal of this study was to review the implication of neuroinflammation in the evolution of motor function in stroke and trauma patients, with a particular focus on novel methods and potential biomarkers to aid clinicians in diagnosis, treatment, and therapy. A specific analysis of the strengths, weaknesses, threats, and opportunities was conducted, highlighting the key challenges to be faced in the coming years.
Collapse
Affiliation(s)
- Ane Larrea
- Research and Development Division, IMG Pharma Biotech, 48170 Zamudio, Spain; (A.L.); (A.E.); (E.D.-M.); (E.A.)
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, 48940 Leioa, Spain;
| | - Ane Elexpe
- Research and Development Division, IMG Pharma Biotech, 48170 Zamudio, Spain; (A.L.); (A.E.); (E.D.-M.); (E.A.)
| | - Eguzkiñe Díez-Martín
- Research and Development Division, IMG Pharma Biotech, 48170 Zamudio, Spain; (A.L.); (A.E.); (E.D.-M.); (E.A.)
- Department of Immunology, Microbiology and Parasitology, Faculty of Science and Technology, University of the Basque Country UPV/EHU, 48940 Leioa, Spain
| | - María Torrecilla
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, 48940 Leioa, Spain;
| | - Egoitz Astigarraga
- Research and Development Division, IMG Pharma Biotech, 48170 Zamudio, Spain; (A.L.); (A.E.); (E.D.-M.); (E.A.)
| | - Gabriel Barreda-Gómez
- Research and Development Division, IMG Pharma Biotech, 48170 Zamudio, Spain; (A.L.); (A.E.); (E.D.-M.); (E.A.)
| |
Collapse
|
6
|
Zhang JK, Javeed S, Greenberg JK, Dibble CF, Song SK, Ray WZ. Diffusion Basis Spectrum Imaging Identifies Clinically Relevant Disease Phenotypes of Cervical Spondylotic Myelopathy. Clin Spine Surg 2023; 36:134-142. [PMID: 36959182 PMCID: PMC10042585 DOI: 10.1097/bsd.0000000000001451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 01/29/2023] [Indexed: 03/25/2023]
Abstract
STUDY DESIGN Prospective cohort study. OBJECTIVE Apply a machine learning clustering algorithm to baseline imaging data to identify clinically relevant cervical spondylotic myelopathy (CSM) patient phenotypes. SUMMARY OF BACKGROUND DATA A major shortcoming in improving care for CSM patients is the lack of robust quantitative imaging tools to guide surgical decision-making. Advanced diffusion-weighted magnetic resonance imaging (MRI) techniques, such as diffusion basis spectrum imaging (DBSI), may help address this limitation by providing detailed evaluations of white matter injury in CSM. METHODS Fifty CSM patients underwent comprehensive clinical assessments and diffusion-weighted MRI, followed by DBSI modeling. DBSI metrics included fractional anisotropy, axial and radial diffusivity, fiber fraction, extra-axonal fraction, restricted fraction, and nonrestricted fraction. Neurofunctional status was assessed by the modified Japanese Orthopedic Association, myelopathic disability index, and disabilities of the arm, shoulder, and hand. Quality-of-life was measured by the 36-Item Short Form Survey physical component summary and mental component summary. The neck disability index was used to measure self-reported neck pain. K-means clustering was applied to baseline DBSI measures to identify 3 clinically relevant CSM disease phenotypes. Baseline demographic, clinical, radiographic, and patient-reported outcome measures were compared among clusters using one-way analysis of variance (ANOVA). RESULTS Twenty-three (55%) mild, 9 (21%) moderate, and 10 (24%) severe myelopathy patients were enrolled. Eight patients were excluded due to MRI data of insufficient quality. Of the remaining 42 patients, 3 groups were generated by k-means clustering. When compared with clusters 1 and 2, cluster 3 performed significantly worse on the modified Japanese Orthopedic Association and all patient-reported outcome measures (P<0.001), except the 36-Item Short Form Survey mental component summary (P>0.05). Cluster 3 also possessed the highest proportion of non-Caucasian patients (43%, P=0.04), the worst hand dynamometer measurements (P<0.05), and significantly higher intra-axonal axial diffusivity and extra-axonal fraction values (P<0.001). CONCLUSIONS Using baseline imaging data, we delineated a clinically meaningful CSM disease phenotype, characterized by worse neurofunctional status, quality-of-life, and pain, and more severe imaging markers of vasogenic edema. LEVEL OF EVIDENCE II.
Collapse
Affiliation(s)
- Justin K. Zhang
- Department of Neurological Surgery, Washington University School of Medicine, Saint Louis, Missouri, 63110, USA
| | - Saad Javeed
- Department of Neurological Surgery, Washington University School of Medicine, Saint Louis, Missouri, 63110, USA
| | - Jacob K. Greenberg
- Department of Neurological Surgery, Washington University School of Medicine, Saint Louis, Missouri, 63110, USA
| | - Christopher F. Dibble
- Department of Neurological Surgery, Washington University School of Medicine, Saint Louis, Missouri, 63110, USA
| | - Sheng-Kwei Song
- Department of Neurological Surgery, Washington University School of Medicine, Saint Louis, Missouri, 63110, USA
| | - Wilson Z. Ray
- Department of Neurological Surgery, Washington University School of Medicine, Saint Louis, Missouri, 63110, USA
| |
Collapse
|
7
|
Han RH, Johanns TM, Roberts KF, Tao Y, Luo J, Ye Z, Sun P, Blum J, Lin TH, Song SK, Kim AH. Diffusion basis spectrum imaging as an adjunct to conventional MRI leads to earlier diagnosis of high-grade glioma tumor progression versus treatment effect. Neurooncol Adv 2023; 5:vdad050. [PMID: 37215950 PMCID: PMC10195207 DOI: 10.1093/noajnl/vdad050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023] Open
Abstract
Background Following chemoradiotherapy for high-grade glioma (HGG), it is often challenging to distinguish treatment changes from true tumor progression using conventional MRI. The diffusion basis spectrum imaging (DBSI) hindered fraction is associated with tissue edema or necrosis, which are common treatment-related changes. We hypothesized that DBSI hindered fraction may augment conventional imaging for earlier diagnosis of progression versus treatment effect. Methods Adult patients were prospectively recruited if they had a known histologic diagnosis of HGG and completed standard-of-care chemoradiotherapy. DBSI and conventional MRI data were acquired longitudinally beginning 4 weeks post-radiation. Conventional MRI and DBSI metrics were compared with respect to their ability to diagnose progression versus treatment effect. Results Twelve HGG patients were enrolled between August 2019 and February 2020, and 9 were ultimately analyzed (5 progression, 4 treatment effect). Within new or enlarging contrast-enhancing regions, DBSI hindered fraction was significantly higher in the treatment effect group compared to progression group (P = .0004). Compared to serial conventional MRI alone, inclusion of DBSI would have led to earlier diagnosis of either progression or treatment effect in 6 (66.7%) patients by a median of 7.7 (interquartile range = 0-20.1) weeks. Conclusions In the first longitudinal prospective study of DBSI in adult HGG patients, we found that in new or enlarging contrast-enhancing regions following therapy, DBSI hindered fraction is elevated in cases of treatment effect compared to those with progression. Hindered fraction map may be a valuable adjunct to conventional MRI to distinguish tumor progression from treatment effect.
Collapse
Affiliation(s)
- Rowland H Han
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Tanner M Johanns
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- The Brain Tumor Center, Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Kaleigh F Roberts
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Yu Tao
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jingqin Luo
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Zezhong Ye
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Peng Sun
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jacob Blum
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Tsen-Hsuan Lin
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Sheng-Kwei Song
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Albert H Kim
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
- The Brain Tumor Center, Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
8
|
Malomo T, Allard Brown A, Bale K, Yung A, Kozlowski P, Heran M, Streijger F, Kwon BK. Quantifying Intraparenchymal Hemorrhage after Traumatic Spinal Cord Injury: A Review of Methodology. J Neurotrauma 2022; 39:1603-1635. [PMID: 35538847 DOI: 10.1089/neu.2021.0317] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Intraparenchymal hemorrhage (IPH) after a traumatic injury has been associated with poor neurological outcomes. Although IPH may result from the initial mechanical trauma, the blood and its breakdown products have potentially deleterious effects. Further, the degree of IPH has been correlated with injury severity and the extent of subsequent recovery. Therefore, accurate evaluation and quantification of IPH following traumatic spinal cord injury (SCI) is important to define treatments' effects on IPH progression and secondary neuronal injury. Imaging modalities, such as magnetic resonance imaging (MRI) and ultrasound (US), have been explored by researchers for the detection and quantification of IPH following SCI. Both quantitative and semiquantitative MRI and US measurements have been applied to objectively assess IPH following SCI, but the optimal methods for doing so are not well established. Studies in animal SCI models (rodent and porcine) have explored US and histological techniques in evaluating SCI and have demonstrated the potential to detect and quantify IPH. Newer techniques using machine learning algorithms (such as convolutional neural networks [CNN]) have also been studied to calculate IPH volume and have yielded promising results. Despite long-standing recognition of the potential pathological significance of IPH within the spinal cord, quantifying IPH with MRI or US is a relatively new area of research. Further studies are warranted to investigate their potential use. Here, we review the different and emerging quantitative MRI, US, and histological approaches used to detect and quantify IPH following SCI.
Collapse
Affiliation(s)
- Toluyemi Malomo
- International Collaboration on Repair Discoveries, Division of Neuroradiology, Vancouver General Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Aysha Allard Brown
- International Collaboration on Repair Discoveries, Division of Neuroradiology, Vancouver General Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kirsten Bale
- International Collaboration on Repair Discoveries, Division of Neuroradiology, Vancouver General Hospital, University of British Columbia, Vancouver, British Columbia, Canada.,UBC MRI Research Center, Division of Neuroradiology, Vancouver General Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Andrew Yung
- International Collaboration on Repair Discoveries, Division of Neuroradiology, Vancouver General Hospital, University of British Columbia, Vancouver, British Columbia, Canada.,UBC MRI Research Center, Division of Neuroradiology, Vancouver General Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Piotr Kozlowski
- International Collaboration on Repair Discoveries, Division of Neuroradiology, Vancouver General Hospital, University of British Columbia, Vancouver, British Columbia, Canada.,UBC MRI Research Center, Division of Neuroradiology, Vancouver General Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Manraj Heran
- Department of Radiology, Division of Neuroradiology, Vancouver General Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Femke Streijger
- International Collaboration on Repair Discoveries, Division of Neuroradiology, Vancouver General Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Brian K Kwon
- International Collaboration on Repair Discoveries, Division of Neuroradiology, Vancouver General Hospital, University of British Columbia, Vancouver, British Columbia, Canada.,Vancouver Spine Surgery Institute, Department of Orthopaedics, and Division of Neuroradiology, Vancouver General Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
9
|
Isaacs AM, Neil JJ, McAllister JP, Dahiya S, Castaneyra-Ruiz L, Merisaari H, Botteron HE, Alexopoulos D, George A, Sun P, Morales DM, Shimony JS, Strahle J, Yan Y, Song SK, Limbrick DD, Smyser CD. Microstructural Periventricular White Matter Injury in Post-hemorrhagic Ventricular Dilatation. Neurology 2022; 98:e364-e375. [PMID: 34799460 PMCID: PMC8793106 DOI: 10.1212/wnl.0000000000013080] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 09/15/2021] [Accepted: 11/12/2021] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND AND OBJECTIVES The neurologic deficits of neonatal post-hemorrhagic hydrocephalus (PHH) have been linked to periventricular white matter injury. To improve understanding of PHH-related injury, diffusion basis spectrum imaging (DBSI) was applied in neonates, modeling axonal and myelin integrity, fiber density, and extrafiber pathologies. Objectives included characterizing DBSI measures in periventricular tracts, associating measures with ventricular size, and examining MRI findings in the context of postmortem white matter histology from similar cases. METHODS A prospective cohort of infants born very preterm underwent term equivalent MRI, including infants with PHH, high-grade intraventricular hemorrhage without hydrocephalus (IVH), and controls (very preterm [VPT]). DBSI metrics extracted from the corpus callosum, corticospinal tracts, and optic radiations included fiber axial diffusivity, fiber radial diffusivity, fiber fractional anisotropy, fiber fraction (fiber density), restricted fractions (cellular infiltration), and nonrestricted fractions (vasogenic edema). Measures were compared across groups and correlated with ventricular size. Corpus callosum postmortem immunohistochemistry in infants with and without PHH assessed intra- and extrafiber pathologies. RESULTS Ninety-five infants born very preterm were assessed (68 VPT, 15 IVH, 12 PHH). Infants with PHH had the most severe white matter abnormalities and there were no consistent differences in measures between IVH and VPT groups. Key tract-specific white matter injury patterns in PHH included reduced fiber fraction in the setting of axonal or myelin injury, increased cellular infiltration, vasogenic edema, and inflammation. Specifically, measures of axonal injury were highest in the corpus callosum; both axonal and myelin injury were observed in the corticospinal tracts; and axonal and myelin integrity were preserved in the setting of increased extrafiber cellular infiltration and edema in the optic radiations. Increasing ventricular size correlated with worse DBSI metrics across groups. On histology, infants with PHH had high cellularity, variable cytoplasmic vacuolation, and low synaptophysin marker intensity. DISCUSSION PHH was associated with diffuse white matter injury, including tract-specific patterns of axonal and myelin injury, fiber loss, cellular infiltration, and inflammation. Larger ventricular size was associated with greater disruption. Postmortem immunohistochemistry confirmed MRI findings. These results demonstrate DBSI provides an innovative approach extending beyond conventional diffusion MRI for investigating neuropathologic effects of PHH on neonatal brain development.
Collapse
Affiliation(s)
- Albert M Isaacs
- From the Department of Neuroscience (A.M.I.), Washington University in St. Louis, MO; Department of Clinical Neurosciences (A.M.I.), University of Calgary, Canada; and Departments of Neurology (J.J.N., D.A., C.D.S.), Neurosurgery (J.P.A., L.C.-R., H.E.B., D.M.M., J.S., D.D.L.), Pathology (S.D.), Public Health Sciences (Y.Y.,), and Pediatrics (C.D.S.), and Mallinckrodt Institute of Radiology (H.M., A.G., P.S., J.S., S.-K.S., C.D.S.), Washington University School of Medicine, St. Louis, MO.
| | - Jeffrey J Neil
- From the Department of Neuroscience (A.M.I.), Washington University in St. Louis, MO; Department of Clinical Neurosciences (A.M.I.), University of Calgary, Canada; and Departments of Neurology (J.J.N., D.A., C.D.S.), Neurosurgery (J.P.A., L.C.-R., H.E.B., D.M.M., J.S., D.D.L.), Pathology (S.D.), Public Health Sciences (Y.Y.,), and Pediatrics (C.D.S.), and Mallinckrodt Institute of Radiology (H.M., A.G., P.S., J.S., S.-K.S., C.D.S.), Washington University School of Medicine, St. Louis, MO
| | - James P McAllister
- From the Department of Neuroscience (A.M.I.), Washington University in St. Louis, MO; Department of Clinical Neurosciences (A.M.I.), University of Calgary, Canada; and Departments of Neurology (J.J.N., D.A., C.D.S.), Neurosurgery (J.P.A., L.C.-R., H.E.B., D.M.M., J.S., D.D.L.), Pathology (S.D.), Public Health Sciences (Y.Y.,), and Pediatrics (C.D.S.), and Mallinckrodt Institute of Radiology (H.M., A.G., P.S., J.S., S.-K.S., C.D.S.), Washington University School of Medicine, St. Louis, MO
| | - Sonika Dahiya
- From the Department of Neuroscience (A.M.I.), Washington University in St. Louis, MO; Department of Clinical Neurosciences (A.M.I.), University of Calgary, Canada; and Departments of Neurology (J.J.N., D.A., C.D.S.), Neurosurgery (J.P.A., L.C.-R., H.E.B., D.M.M., J.S., D.D.L.), Pathology (S.D.), Public Health Sciences (Y.Y.,), and Pediatrics (C.D.S.), and Mallinckrodt Institute of Radiology (H.M., A.G., P.S., J.S., S.-K.S., C.D.S.), Washington University School of Medicine, St. Louis, MO
| | - Leandro Castaneyra-Ruiz
- From the Department of Neuroscience (A.M.I.), Washington University in St. Louis, MO; Department of Clinical Neurosciences (A.M.I.), University of Calgary, Canada; and Departments of Neurology (J.J.N., D.A., C.D.S.), Neurosurgery (J.P.A., L.C.-R., H.E.B., D.M.M., J.S., D.D.L.), Pathology (S.D.), Public Health Sciences (Y.Y.,), and Pediatrics (C.D.S.), and Mallinckrodt Institute of Radiology (H.M., A.G., P.S., J.S., S.-K.S., C.D.S.), Washington University School of Medicine, St. Louis, MO
| | - Harri Merisaari
- From the Department of Neuroscience (A.M.I.), Washington University in St. Louis, MO; Department of Clinical Neurosciences (A.M.I.), University of Calgary, Canada; and Departments of Neurology (J.J.N., D.A., C.D.S.), Neurosurgery (J.P.A., L.C.-R., H.E.B., D.M.M., J.S., D.D.L.), Pathology (S.D.), Public Health Sciences (Y.Y.,), and Pediatrics (C.D.S.), and Mallinckrodt Institute of Radiology (H.M., A.G., P.S., J.S., S.-K.S., C.D.S.), Washington University School of Medicine, St. Louis, MO
| | - Haley E Botteron
- From the Department of Neuroscience (A.M.I.), Washington University in St. Louis, MO; Department of Clinical Neurosciences (A.M.I.), University of Calgary, Canada; and Departments of Neurology (J.J.N., D.A., C.D.S.), Neurosurgery (J.P.A., L.C.-R., H.E.B., D.M.M., J.S., D.D.L.), Pathology (S.D.), Public Health Sciences (Y.Y.,), and Pediatrics (C.D.S.), and Mallinckrodt Institute of Radiology (H.M., A.G., P.S., J.S., S.-K.S., C.D.S.), Washington University School of Medicine, St. Louis, MO
| | - Dimitrios Alexopoulos
- From the Department of Neuroscience (A.M.I.), Washington University in St. Louis, MO; Department of Clinical Neurosciences (A.M.I.), University of Calgary, Canada; and Departments of Neurology (J.J.N., D.A., C.D.S.), Neurosurgery (J.P.A., L.C.-R., H.E.B., D.M.M., J.S., D.D.L.), Pathology (S.D.), Public Health Sciences (Y.Y.,), and Pediatrics (C.D.S.), and Mallinckrodt Institute of Radiology (H.M., A.G., P.S., J.S., S.-K.S., C.D.S.), Washington University School of Medicine, St. Louis, MO
| | - Ajit George
- From the Department of Neuroscience (A.M.I.), Washington University in St. Louis, MO; Department of Clinical Neurosciences (A.M.I.), University of Calgary, Canada; and Departments of Neurology (J.J.N., D.A., C.D.S.), Neurosurgery (J.P.A., L.C.-R., H.E.B., D.M.M., J.S., D.D.L.), Pathology (S.D.), Public Health Sciences (Y.Y.,), and Pediatrics (C.D.S.), and Mallinckrodt Institute of Radiology (H.M., A.G., P.S., J.S., S.-K.S., C.D.S.), Washington University School of Medicine, St. Louis, MO
| | - Peng Sun
- From the Department of Neuroscience (A.M.I.), Washington University in St. Louis, MO; Department of Clinical Neurosciences (A.M.I.), University of Calgary, Canada; and Departments of Neurology (J.J.N., D.A., C.D.S.), Neurosurgery (J.P.A., L.C.-R., H.E.B., D.M.M., J.S., D.D.L.), Pathology (S.D.), Public Health Sciences (Y.Y.,), and Pediatrics (C.D.S.), and Mallinckrodt Institute of Radiology (H.M., A.G., P.S., J.S., S.-K.S., C.D.S.), Washington University School of Medicine, St. Louis, MO
| | - Diego M Morales
- From the Department of Neuroscience (A.M.I.), Washington University in St. Louis, MO; Department of Clinical Neurosciences (A.M.I.), University of Calgary, Canada; and Departments of Neurology (J.J.N., D.A., C.D.S.), Neurosurgery (J.P.A., L.C.-R., H.E.B., D.M.M., J.S., D.D.L.), Pathology (S.D.), Public Health Sciences (Y.Y.,), and Pediatrics (C.D.S.), and Mallinckrodt Institute of Radiology (H.M., A.G., P.S., J.S., S.-K.S., C.D.S.), Washington University School of Medicine, St. Louis, MO
| | - Joshua S Shimony
- From the Department of Neuroscience (A.M.I.), Washington University in St. Louis, MO; Department of Clinical Neurosciences (A.M.I.), University of Calgary, Canada; and Departments of Neurology (J.J.N., D.A., C.D.S.), Neurosurgery (J.P.A., L.C.-R., H.E.B., D.M.M., J.S., D.D.L.), Pathology (S.D.), Public Health Sciences (Y.Y.,), and Pediatrics (C.D.S.), and Mallinckrodt Institute of Radiology (H.M., A.G., P.S., J.S., S.-K.S., C.D.S.), Washington University School of Medicine, St. Louis, MO
| | - Jennifer Strahle
- From the Department of Neuroscience (A.M.I.), Washington University in St. Louis, MO; Department of Clinical Neurosciences (A.M.I.), University of Calgary, Canada; and Departments of Neurology (J.J.N., D.A., C.D.S.), Neurosurgery (J.P.A., L.C.-R., H.E.B., D.M.M., J.S., D.D.L.), Pathology (S.D.), Public Health Sciences (Y.Y.,), and Pediatrics (C.D.S.), and Mallinckrodt Institute of Radiology (H.M., A.G., P.S., J.S., S.-K.S., C.D.S.), Washington University School of Medicine, St. Louis, MO
| | - Yan Yan
- From the Department of Neuroscience (A.M.I.), Washington University in St. Louis, MO; Department of Clinical Neurosciences (A.M.I.), University of Calgary, Canada; and Departments of Neurology (J.J.N., D.A., C.D.S.), Neurosurgery (J.P.A., L.C.-R., H.E.B., D.M.M., J.S., D.D.L.), Pathology (S.D.), Public Health Sciences (Y.Y.,), and Pediatrics (C.D.S.), and Mallinckrodt Institute of Radiology (H.M., A.G., P.S., J.S., S.-K.S., C.D.S.), Washington University School of Medicine, St. Louis, MO
| | - Sheng-Kwei Song
- From the Department of Neuroscience (A.M.I.), Washington University in St. Louis, MO; Department of Clinical Neurosciences (A.M.I.), University of Calgary, Canada; and Departments of Neurology (J.J.N., D.A., C.D.S.), Neurosurgery (J.P.A., L.C.-R., H.E.B., D.M.M., J.S., D.D.L.), Pathology (S.D.), Public Health Sciences (Y.Y.,), and Pediatrics (C.D.S.), and Mallinckrodt Institute of Radiology (H.M., A.G., P.S., J.S., S.-K.S., C.D.S.), Washington University School of Medicine, St. Louis, MO
| | - David D Limbrick
- From the Department of Neuroscience (A.M.I.), Washington University in St. Louis, MO; Department of Clinical Neurosciences (A.M.I.), University of Calgary, Canada; and Departments of Neurology (J.J.N., D.A., C.D.S.), Neurosurgery (J.P.A., L.C.-R., H.E.B., D.M.M., J.S., D.D.L.), Pathology (S.D.), Public Health Sciences (Y.Y.,), and Pediatrics (C.D.S.), and Mallinckrodt Institute of Radiology (H.M., A.G., P.S., J.S., S.-K.S., C.D.S.), Washington University School of Medicine, St. Louis, MO
| | - Christopher D Smyser
- From the Department of Neuroscience (A.M.I.), Washington University in St. Louis, MO; Department of Clinical Neurosciences (A.M.I.), University of Calgary, Canada; and Departments of Neurology (J.J.N., D.A., C.D.S.), Neurosurgery (J.P.A., L.C.-R., H.E.B., D.M.M., J.S., D.D.L.), Pathology (S.D.), Public Health Sciences (Y.Y.,), and Pediatrics (C.D.S.), and Mallinckrodt Institute of Radiology (H.M., A.G., P.S., J.S., S.-K.S., C.D.S.), Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
10
|
Spinal cord injury in mice impacts central and peripheral pathology in a severity-dependent manner. Pain 2021; 163:1172-1185. [PMID: 34490852 DOI: 10.1097/j.pain.0000000000002471] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 08/25/2021] [Indexed: 11/26/2022]
Abstract
ABSTRACT Chronic pain is a common medical complication experienced by those living with spinal cord injury (SCI) and leads to worsened quality of life. The pathophysiology of SCI pain is poorly understood, hampering the development of safe and efficacious therapeutics. We therefore sought to develop a clinically relevant model of SCI with a strong pain phenotype and characterize the central and peripheral pathology after injury. A contusion (50 kdyn) injury, with and without sustained compression (60 seconds) of the spinal cord, was carried out on female C57BL/6J mice. Mice with compression of the spinal cord exhibited significantly greater heat and mechanical hypersensitivity starting at 7 days post-injury, concomitant with reduced locomotor function, compared to those without compression. Immunohistochemical analysis of spinal cord tissue revealed significantly less myelin sparing and increased macrophage activation in mice with compression compared to those without. As measured by flow cytometry, immune cell infiltration and activation were significantly greater in the spinal cord (phagocytic myeloid cells and microglia) and dorsal root ganglia (Ly6C+ monocytes) following compression injury. We also decided to investigate the gastrointestinal microbiome, as it has been shown to be altered in SCI patients and has recently been shown to play a role in immune system maturation and pain. We found increased dysbiosis of the gastrointestinal microbiome in an injury severity-dependent manner. The use of this contusion-compression model of SCI may help advance the preclinical assessment of acute and chronic SCI pain and lead to a better understanding of mechanisms contributing to this pain.
Collapse
|
11
|
Rink S, Pavlov S, Wöhler A, Bendella H, Manthou M, Papamitsou T, Dunlop SA, Angelov DN. Numbers of Axons in Spared Neural Tissue Bridges But Not Their Widths or Areas Correlate With Functional Recovery in Spinal Cord-Injured Rats. J Neuropathol Exp Neurol 2021; 79:1203-1217. [PMID: 32594136 DOI: 10.1093/jnen/nlaa050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 04/11/2020] [Accepted: 05/08/2020] [Indexed: 11/14/2022] Open
Abstract
The relationships between various parameters of tissue damage and subsequent functional recovery after spinal cord injury (SCI) are not well understood. Patients may regain micturition control and walking despite large postinjury medullar cavities. The objective of this study was to establish possible correlations between morphological findings and degree of functional recovery after spinal cord compression at vertebra Th8 in rats. Recovery of motor (Basso, Beattie, Bresnahan, foot-stepping angle, rump-height index, and ladder climbing), sensory (withdrawal latency), and bladder functions was analyzed at 1, 3, 6, 9, and 12 weeks post-SCI. Following perfusion fixation, spinal cord tissue encompassing the injury site was cut in longitudinal frontal sections. Lesion lengths, lesion volumes, and areas of perilesional neural tissue bridges were determined after staining with cresyl violet. The numbers of axons in these bridges were quantified after staining for class III β-tubulin. We found that it was not the area of the spared tissue bridges, which is routinely determined by magnetic resonance imaging (MRI), but the numbers of axons in them that correlated with functional recovery after SCI (Spearman's ρ > 0.8; p < 0.001). We conclude that prognostic statements based only on MRI measurements should be considered with caution.
Collapse
Affiliation(s)
- Svenja Rink
- Department of Prosthetic Dentistry, School of Dental and Oral Medicine, University of Cologne, Germany
| | - Stoyan Pavlov
- Department of Anatomy, Histology and Embryology, Medical University, Varna, Bulgaria
| | | | - Habib Bendella
- Department of Neurosurgery, University of Witten/Herdecke, Cologne Merheim Medical Center (CMMC), Cologne, Germany
| | - Marilena Manthou
- Department of Histology and Embryology, Aristotle University Thessaloniki, Greece
| | - Theodora Papamitsou
- Department of Histology and Embryology, Aristotle University Thessaloniki, Greece
| | - Sarah A Dunlop
- School of Biological Sciences, The University of Western Australia, Australia
| | | |
Collapse
|
12
|
Mash-1 modified neural stem cells transplantation promotes neural stem cells differentiation into neurons to further improve locomotor functional recovery in spinal cord injury rats. Gene 2021; 781:145528. [PMID: 33631250 DOI: 10.1016/j.gene.2021.145528] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 12/28/2020] [Accepted: 02/09/2021] [Indexed: 12/28/2022]
Abstract
BACKGROUND Spinal cord injury (SCI) leads to severe motor and sensory dysfunctions. Neural stem cells (NSCs) transplantation therapy plays a positive role in functional recovery after SCI, but the effectiveness of this therapy is limited by inadequate differentiation ability of transplanted NSCs. Mammalian achaete-scute homologue-1 (Mash-1) has been reported to improve differentiation of NSCs. Thus, this study modified NSCs with Mash-1 to repair SCI. METHODS NSCs isolated from rat embryo hippocampus were cultured and identified in vitro and further transfected with the lentiviral vectors (Lv-Mash-1). After establishing a SCI rat model, the rats were transplanted with Mash-1 modified NSCs, the histopathological changes of rat spinal cord were detected by hematoxylin-eosin (HE) staining, and the locomotor activity of rats was evaluated with the Basso, Beattie and Bresnahan (BBB) scale. The NSCs cultured in vitro or extracted from SCI rat spinal cord were identified by immunofluorescence (IF). Mash-1, β3-Tubulin, and NeuN expressions in those cells were determined by Western blotting and reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR). RESULTS NSCs isolated from rat embryo hippocampus were Nestin- and NeuN-positive. NSC transplantation modified by Mash-1 increased BBB score of SCI rats and promoted recovery in lesion site of SCI rats. Mash-1 overexpression also promoted β3-Tubulin and NeuN expressions in NSCs cultured in vitro or extracted from spinal cord of SCI rats. CONCLUSION Mash-1 overexpression promoted NSC differentiation into neurons, and further improved locomotor functional recovery of SCI rats.
Collapse
|
13
|
Lin TH, Zhan J, Song C, Wallendorf M, Sun P, Niu X, Yang R, Cross AH, Song SK. Diffusion Basis Spectrum Imaging Detects Axonal Loss After Transient Dexamethasone Treatment in Optic Neuritis Mice. Front Neurosci 2021; 14:592063. [PMID: 33551721 PMCID: PMC7862582 DOI: 10.3389/fnins.2020.592063] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 12/28/2020] [Indexed: 11/24/2022] Open
Abstract
Optic neuritis is a frequent first symptom of multiple sclerosis (MS) for which corticosteroids are a widely employed treatment option. The Optic Neuritis Treatment Trial (ONTT) reported that corticosteroid treatment does not improve long-term visual acuity, although the evolution of underlying pathologies is unclear. In this study, we employed non-invasive diffusion basis spectrum imaging (DBSI)-derived fiber volume to quantify 11% axonal loss 2 months after corticosteroid treatment (vs. baseline) in experimental autoimmune encephalomyelitis mouse optic nerves affected by optic neuritis. Longitudinal DBSI was performed at baseline (before immunization), after a 2-week corticosteroid treatment period, and 1 and 2 months after treatment, followed by histological validation of neuropathology. Pathological metrics employed to assess the optic nerve revealed axonal protection and anti-inflammatory effects of dexamethasone treatment that were transient. Two months after treatment, axonal injury and loss were indistinguishable between PBS- and dexamethasone-treated optic nerves, similar to results of the human ONTT. Our findings in mice further support that corticosteroid treatment alone is not sufficient to prevent eventual axonal loss in ON, and strongly support the potential of DBSI as an in vivo imaging outcome measure to assess optic nerve pathology.
Collapse
Affiliation(s)
- Tsen-Hsuan Lin
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Jie Zhan
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, United States.,Department of Radiology, The First Affiliated Hospital, Nanchang University, Jiangxi, China
| | - Chunyu Song
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, United States
| | - Michael Wallendorf
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, United States
| | - Peng Sun
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Xuan Niu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Ruimeng Yang
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, United States.,Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Anne H Cross
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, United States
| | - Sheng-Kwei Song
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, United States.,Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, United States.,Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
14
|
Ye Z, Gary SE, Sun P, Mustafi SM, Glenn GR, Yeh FC, Merisaari H, Song C, Yang R, Huang GS, Kao HW, Lin CY, Wu YC, Jensen JH, Song SK. The impact of edema and fiber crossing on diffusion MRI metrics assessed in an ex vivo nerve phantom: Multi-tensor model vs. diffusion orientation distribution function. NMR IN BIOMEDICINE 2021; 34:e4414. [PMID: 33015890 PMCID: PMC9743958 DOI: 10.1002/nbm.4414] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 08/23/2020] [Accepted: 09/06/2020] [Indexed: 05/30/2023]
Abstract
Diffusion tensor imaging (DTI) has been employed for over 2 decades to noninvasively quantify central nervous system diseases/injuries. However, DTI is an inadequate simplification of diffusion modeling in the presence of coexisting inflammation, edema and crossing nerve fibers. We employed a tissue phantom using fixed mouse trigeminal nerves coated with various amounts of agarose gel to mimic crossing fibers in the presence of vasogenic edema. Diffusivity measures derived by DTI and diffusion basis spectrum imaging (DBSI) were compared at increasing levels of simulated edema and degrees of fiber crossing. Furthermore, we assessed the ability of DBSI, diffusion kurtosis imaging (DKI), generalized q-sampling imaging (GQI), q-ball imaging (QBI) and neurite orientation dispersion and density imaging to resolve fiber crossing, in reference to the gold standard angles measured from structural images. DTI-computed diffusivities and fractional anisotropy were significantly confounded by gel-mimicked edema and crossing fibers. Conversely, DBSI calculated accurate diffusivities of individual fibers regardless of the extent of simulated edema and degrees of fiber crossing angles. Additionally, DBSI accurately and consistently estimated crossing angles in various conditions of gel-mimicked edema when compared with the gold standard (r2 = 0.92, P = 1.9 × 10-9 , bias = 3.9°). Small crossing angles and edema significantly impact the diffusion orientation distribution function, making DKI, GQI and QBI less accurate in detecting and estimating fiber crossing angles. Lastly, we used diffusion tensor ellipsoids to demonstrate that DBSI resolves the confounds of edema and crossing fibers in the peritumoral edema region from a patient with lung cancer metastasis, while DTI failed. In summary, DBSI is able to separate two crossing fibers and accurately recover their diffusivities in a complex environment characterized by increasing crossing angles and amounts of gel-mimicked edema. DBSI also indicated better angular resolution compared with DKI, QBI and GQI.
Collapse
Affiliation(s)
- Zezhong Ye
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Sam E. Gary
- Medical Scientist Training Program, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Peng Sun
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Sourajit Mitra Mustafi
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202
| | - George Russell Glenn
- Department of Radiology and Imaging Science, Emory University School of Medicine, Atlanta, GA 30322
| | - Fang-Cheng Yeh
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Harri Merisaari
- Department of Diagnostic Radiology, University of Turku, Turku, Finland 20014
| | - Chunyu Song
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63130
| | - Ruimeng Yang
- Department of Radiology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510180, China
| | - Guo-Shu Huang
- Department of Radiology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan 114
| | - Hung-Wen Kao
- Department of Radiology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan 114
| | | | - Yu-Chien Wu
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Jens H. Jensen
- Department of Radiology and Radiological Science, Medical University of South Carolina, Charleston, SC 29425
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425
- Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC 29425
| | - Sheng-Kwei Song
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
15
|
Ye Z, Price RL, Liu X, Lin J, Yang Q, Sun P, Wu AT, Wang L, Han RH, Song C, Yang R, Gary SE, Mao DD, Wallendorf M, Campian JL, Li JS, Dahiya S, Kim AH, Song SK. Diffusion Histology Imaging Combining Diffusion Basis Spectrum Imaging (DBSI) and Machine Learning Improves Detection and Classification of Glioblastoma Pathology. Clin Cancer Res 2020; 26:5388-5399. [PMID: 32694155 DOI: 10.1158/1078-0432.ccr-20-0736] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 06/01/2020] [Accepted: 07/15/2020] [Indexed: 01/10/2023]
Abstract
PURPOSE Glioblastoma (GBM) is one of the deadliest cancers with no cure. While conventional MRI has been widely adopted to examine GBM clinically, accurate neuroimaging assessment of tumor histopathology for improved diagnosis, surgical planning, and treatment evaluation remains an unmet need in the clinical management of GBMs. EXPERIMENTAL DESIGN We employ a novel diffusion histology imaging (DHI) approach, combining diffusion basis spectrum imaging (DBSI) and machine learning, to detect, differentiate, and quantify areas of high cellularity, tumor necrosis, and tumor infiltration in GBM. RESULTS Gadolinium-enhanced T1-weighted or hyperintense fluid-attenuated inversion recovery failed to reflect the morphologic complexity underlying tumor in patients with GBM. Contrary to the conventional wisdom that apparent diffusion coefficient (ADC) negatively correlates with increased tumor cellularity, we demonstrate disagreement between ADC and histologically confirmed tumor cellularity in GBM specimens, whereas DBSI-derived restricted isotropic diffusion fraction positively correlated with tumor cellularity in the same specimens. By incorporating DBSI metrics as classifiers for a supervised machine learning algorithm, we accurately predicted high tumor cellularity, tumor necrosis, and tumor infiltration with 87.5%, 89.0%, and 93.4% accuracy, respectively. CONCLUSIONS Our results suggest that DHI could serve as a favorable alternative to current neuroimaging techniques in guiding biopsy or surgery as well as monitoring therapeutic response in the treatment of GBM.
Collapse
Affiliation(s)
- Zezhong Ye
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Richard L Price
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Xiran Liu
- Department of Electrical & System Engineering, Washington University, St. Louis, Missouri
| | - Joshua Lin
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Qingsong Yang
- Department of Radiology, Changhai Hospital, Yangpu District, Shanghai, China
| | - Peng Sun
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Anthony T Wu
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri
| | - Liang Wang
- Department of Electrical & System Engineering, Washington University, St. Louis, Missouri
| | - Rowland H Han
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Chunyu Song
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri
| | - Ruimeng Yang
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Sam E Gary
- Medical Scientist Training Program, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Diane D Mao
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Michael Wallendorf
- Department of Biostatistics, Washington University School of Medicine, St. Louis, Missouri
| | - Jian L Campian
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Jr-Shin Li
- Department of Electrical & System Engineering, Washington University, St. Louis, Missouri
| | - Sonika Dahiya
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri.
| | - Albert H Kim
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, Missouri.
| | - Sheng-Kwei Song
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
16
|
Warnock A, Toomey LM, Wright AJ, Fisher K, Won Y, Anyaegbu C, Fitzgerald M. Damage Mechanisms to Oligodendrocytes and White Matter in Central Nervous System Injury: The Australian Context. J Neurotrauma 2020; 37:739-769. [DOI: 10.1089/neu.2019.6890] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Andrew Warnock
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Lillian M. Toomey
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, Western Australia, Australia
| | - Alexander J. Wright
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Katherine Fisher
- School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Yerim Won
- School of Human Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Chidozie Anyaegbu
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Melinda Fitzgerald
- Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, Western Australia, Australia
| |
Collapse
|
17
|
Samara A, Murphy T, Strain J, Rutlin J, Sun P, Neyman O, Sreevalsan N, Shimony JS, Ances BM, Song SK, Hershey T, Eisenstein SA. Neuroinflammation and White Matter Alterations in Obesity Assessed by Diffusion Basis Spectrum Imaging. Front Hum Neurosci 2020; 13:464. [PMID: 31992978 PMCID: PMC6971102 DOI: 10.3389/fnhum.2019.00464] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 12/18/2019] [Indexed: 01/06/2023] Open
Abstract
Human obesity is associated with low-grade chronic systemic inflammation, alterations in brain structure and function, and cognitive impairment. Rodent models of obesity show that high-calorie diets cause brain inflammation (neuroinflammation) in multiple regions, including the hippocampus, and impairments in hippocampal-dependent memory tasks. To determine if similar effects exist in humans with obesity, we applied Diffusion Basis Spectrum Imaging (DBSI) to evaluate neuroinflammation and axonal integrity. We examined diffusion-weighted magnetic resonance imaging (MRI) data in two independent cohorts of obese and non-obese individuals (Cohort 1: 25 obese/21 non-obese; Cohort 2: 18 obese/41 non-obese). We applied Tract-based Spatial Statistics (TBSS) to allow whole-brain white matter (WM) analyses and compare DBSI-derived isotropic and anisotropic diffusion measures between the obese and non-obese groups. In both cohorts, the obese group had significantly greater DBSI-derived restricted fraction (DBSI-RF; an indicator of neuroinflammation-related cellularity), and significantly lower DBSI-derived fiber fraction (DBSI-FF; an indicator of apparent axonal density) in several WM tracts (all corrected p < 0.05). Moreover, using region of interest analyses, average DBSI-RF and DBSI-FF values in the hippocampus were significantly greater and lower, respectively, in obese relative to non-obese individuals (Cohort 1: p = 0.045; Cohort 2: p = 0.008). Hippocampal DBSI-FF and DBSI-RF and amygdalar DBSI-FF metrics related to cognitive performance in Cohort 2. In conclusion, these findings suggest that greater neuroinflammation-related cellularity and lower apparent axonal density are associated with human obesity and cognitive performance. Future studies are warranted to determine a potential role for neuroinflammation in obesity-related cognitive impairment.
Collapse
Affiliation(s)
- Amjad Samara
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States
| | - Tatianna Murphy
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States
| | - Jeremy Strain
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
| | - Jerrel Rutlin
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States
| | - Peng Sun
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Olga Neyman
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States
| | - Nitya Sreevalsan
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States
| | - Joshua S Shimony
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Beau M Ances
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States
| | - Sheng-Kwei Song
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Tamara Hershey
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States.,Department of Neurology, Washington University School of Medicine, St. Louis, MO, United States.,Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, United States.,Department of Psychological & Brain Sciences, Washington University School of Medicine, St. Louis, MO, United States
| | - Sarah A Eisenstein
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States.,Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|