1
|
Patterson G, Dufford AJ, Morrissey TW, Styner M, Kim SH, Kim P. Prenatal Household Income Instability and Infant Subcortical Brain Volumes. Dev Sci 2025; 28:e70022. [PMID: 40331480 PMCID: PMC12056889 DOI: 10.1111/desc.70022] [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: 08/16/2024] [Revised: 02/12/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025]
Abstract
Prenatal stress exposure may negatively influence the development of the amygdala and hippocampus. Although there is significant income instability during pregnancy, and it can increase stress among pregnant parents, the impact of income instability on brain development is not well understood. The present study examined the association between household income losses during pregnancy and hippocampus and amygdala volumes in early infancy. A total of 63 infants from a prospective longitudinal study of pregnant individuals and their infants completed an MRI during natural sleep. The total number of negative month-to-month earnings shocks (defined as an arc percent change [APC] of -25 or greater) was significantly associated with smaller right hippocampal and right amygdala volumes. This suggests that household income losses during the perinatal period are associated with infant brain structure after birth. These findings provide support for the development of public programs that prioritize financial consistency, especially during sensitive periods like pregnancy.
Collapse
Affiliation(s)
| | - Alexander J. Dufford
- Center for Mental Health InnovationOregon Health & Science UniversityPortlandOregonUSA
- Department of PsychiatryOregon Health & Science UniversityPortlandOregonUSA
| | - Taryn W. Morrissey
- School of Public AffairsAmerican UniversityWashingtonDistrict of ColumbiaUSA
| | - Martin Styner
- Department of PsychiatryUniversity of North Carolina Chapel HillChapel HillNorth CarolinaUSA
| | - Sun Hyung Kim
- Department of PsychiatryUniversity of North Carolina Chapel HillChapel HillNorth CarolinaUSA
| | - Pilyoung Kim
- Department of PsychologyUniversity of DenverDenverColoradoUSA
- Department of PsychologyEwha Womans UniversitySeoulSouth Korea
| |
Collapse
|
2
|
Berisha DE, Rizvi B, Chappel-Farley MG, Tustison N, Taylor L, Dave A, Sattari NS, Chen IY, Lui KK, Janecek JC, Keator DB, Neikrug AB, Benca RM, Yassa MA, Mander BA. Association of Hypoxemia Due to Obstructive Sleep Apnea With White Matter Hyperintensities and Temporal Lobe Changes in Older Adults. Neurology 2025; 104:e213639. [PMID: 40334140 PMCID: PMC12060793 DOI: 10.1212/wnl.0000000000213639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 03/10/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Cerebral small vessel disease (CSVD) is a leading cause of cognitive decline and functional loss in older adults. Obstructive sleep apnea (OSA) is common in older adults, can increase cerebrovascular disease risk, and is linked to medial temporal lobe (MTL) degeneration and cognitive impairment. However, the interaction between OSA features and CSVD burden and their combined effect on MTL structure and function are not well understood. This study tested the hypothesis that CSVD burden is a candidate mechanism linking OSA to MTL degeneration and impaired memory in older adults. METHODS Cognitively unimpaired older adults from the Biomarker Exploration in Aging, Cognition, and Neurodegeneration cohort were recruited for an observational, in-lab overnight polysomnography (PSG) study with emotional mnemonic discrimination ability assessed before and after sleep. Participants had no neurologic or psychiatric disorders and were not on sleep-affecting medications. PSG-derived OSA variables included apnea-hypopnea index (AHI), total arousal index, and minimum SpO2. MRI was used to assess global and lobar white matter hyperintensity (WMH) volumes and MTL structure (hippocampal volume; entorhinal cortex [ERC] thickness) at an earlier time point. Regressions were implemented while adjusting for age, sex, and concurrent use of antihyperlipidemic and/or antihypertensive medication. Minimum SpO2 was transformed into a Hypoxemia Severity Index for normality, in which lower SpO2 values indicated more severe hypoxemia. RESULTS Thirty-seven older adults were included in the study (age 72.5 ± 5.6 years, 23 women, AHI = 13.8 ± 18.0 [range 0-80]). Hypoxemia measures significantly predicted global WMH volume (bminSpO2 = 0.141 [0.001-0.282], bduration <90% = 0.008 [0.000-0.016]). This relationship was driven by hypoxemia severity during REM sleep (bREMminSpO2 = 0.143 [0.003-0.284]), which also predicted frontal (bREMminSpO2 = 0.101 [0.004-0.198]) and parietal (bREMminSpO2 = 0.121 [0.024-0.219]) WMH burden. Greater frontal WMH burden indirectly mediated the relationship between REM sleep hypoxemia and ERC thickness (indirect effect = -0.043, 95% CI -0.1174 to -0.00015). Reduced ERC thickness was, in turn, associated with worse overnight mnemonic discrimination ability (bleftERCthickness = 0.112 [0.014-0.211]). DISCUSSION These findings identify CSVD as a candidate mechanism linking OSA-related hypoxemia to MTL degeneration and impaired sleep-dependent memory in older adults, specifically implicating hypoxic events during REM sleep.
Collapse
Affiliation(s)
- Destiny E Berisha
- Department of Neurobiology and Behavior, University of California Irvine
- Center for the Neurobiology of Learning and Memory, University of California Irvine
| | - Batool Rizvi
- Department of Neurobiology and Behavior, University of California Irvine
- Center for the Neurobiology of Learning and Memory, University of California Irvine
| | - Miranda G Chappel-Farley
- Department of Neurobiology and Behavior, University of California Irvine
- Center for the Neurobiology of Learning and Memory, University of California Irvine
- Current affiliation: Department of Psychiatry, University of Pittsburgh, PA
| | - Nicholas Tustison
- Center for the Neurobiology of Learning and Memory, University of California Irvine
| | - Lisa Taylor
- Department of Neurobiology and Behavior, University of California Irvine
- Center for the Neurobiology of Learning and Memory, University of California Irvine
- Department of Psychiatry and Human Behavior, University of California Irvine
| | - Abhishek Dave
- Department of Cognitive Sciences, University of California Irvine
| | - Negin S Sattari
- Department of Psychiatry and Human Behavior, University of California Irvine
| | - Ivy Y Chen
- Department of Psychiatry and Human Behavior, University of California Irvine
| | - Kitty K Lui
- Joint Doctoral Program in Clinical Psychology, San Diego State University/University of California San Diego
| | - John C Janecek
- Center for the Neurobiology of Learning and Memory, University of California Irvine
| | - David B Keator
- Department of Psychiatry and Human Behavior, University of California Irvine
| | - Ariel B Neikrug
- Department of Psychiatry and Human Behavior, University of California Irvine
| | - Ruth M Benca
- Department of Neurobiology and Behavior, University of California Irvine
- Center for the Neurobiology of Learning and Memory, University of California Irvine
- Department of Psychiatry and Human Behavior, University of California Irvine
- Neuroscience Training Program, University of Wisconsin-Madison
- Department of Psychiatry and Behavioral Medicine, Wake Forest University, Winston-Salem, NC
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine
| | - Michael A Yassa
- Department of Neurobiology and Behavior, University of California Irvine
- Center for the Neurobiology of Learning and Memory, University of California Irvine
- Department of Psychiatry and Human Behavior, University of California Irvine
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine
- Department of Neurology, University of California Irvine; and
| | - Bryce A Mander
- Center for the Neurobiology of Learning and Memory, University of California Irvine
- Department of Psychiatry and Human Behavior, University of California Irvine
- Department of Cognitive Sciences, University of California Irvine
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine
- Department of Pathology and Laboratory Medicine, University of California Irvine
| |
Collapse
|
3
|
Ljungberg E, Padormo F, Poorman M, Clemensson P, Bourke N, Evans JC, Gholam J, Vavasour I, Kollind SH, Lafayette SL, Bennallick C, Donald KA, Bradford LE, Lena B, Vokhiwa M, Shama T, Siew J, Sekoli L, van Rensburg J, Pepper MS, Khan A, Madhwani A, Banda FA, Mwila ML, Cassidy AR, Moabi K, Sephi D, Boakye RA, Ae‐Ngibise KA, Asante KP, Hollander WJ, Karaulanov T, Williams SCR, Deoni S. Characterization of Portable Ultra-Low Field MRI Scanners for Multi-Center Structural Neuroimaging. Hum Brain Mapp 2025; 46:e70217. [PMID: 40405769 PMCID: PMC12099222 DOI: 10.1002/hbm.70217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 03/17/2025] [Accepted: 04/08/2025] [Indexed: 05/24/2025] Open
Abstract
The lower infrastructure requirements of portable ultra-low field MRI (ULF-MRI) systems have enabled their use in diverse settings such as intensive care units and remote medical facilities. The UNITY Project is an international neuroimaging network harnessing this technology, deploying portable ULF-MRI systems globally to expand access to MRI for studies into brain development. Given the wide range of environments where ULF-MRI systems may operate, there are external factors that might influence image quality. This work aims to introduce the quality control (QC) framework used by the UNITY Project to investigate how robust the systems are and how QC metrics compare between sites and over time. We present a QC framework using a commercially available phantom, scanned with 64 mT portable MRI systems at 17 sites across 12 countries on four continents. Using automated, open-source analysis tools, we quantify signal-to-noise, image contrast, and geometric distortions. Our results demonstrated that the image quality is robust to the varying operational environment, for example, electromagnetic noise interference and temperature. The Larmor frequency was significantly correlated to room temperature, as was image noise and contrast. Image distortions were less than 2.5 mm, with high robustness over time. Similar to studies at higher field, we found that changes in pulse sequence parameters from software updates had an impact on QC metrics. This study demonstrates that portable ULF-MRI systems can be deployed in a variety of environments for multi-center neuroimaging studies and produce robust results.
Collapse
Affiliation(s)
- Emil Ljungberg
- Department of Medical Radiation PhysicsLund UniversityLundSweden
- Department of Neuroimaging, Institute of Psychiatry, Psychology & NeuroscienceKing's College LondonLondonUK
| | | | | | - Petter Clemensson
- Department of Neuroimaging, Institute of Psychiatry, Psychology & NeuroscienceKing's College LondonLondonUK
| | - Niall Bourke
- Department of Neuroimaging, Institute of Psychiatry, Psychology & NeuroscienceKing's College LondonLondonUK
| | - John C. Evans
- CUBRIC, Cardiff School of PsychologyCardiff UniversityCardiffUK
| | - James Gholam
- CUBRIC, Cardiff School of PsychologyCardiff UniversityCardiffUK
| | - Irene Vavasour
- Department of RadiologyUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Shannon H. Kollind
- Department of Medicine (Neurology)University of British ColumbiaVancouverBritish ColumbiaCanada
| | | | - Carly Bennallick
- Department of Neuroimaging, Institute of Psychiatry, Psychology & NeuroscienceKing's College LondonLondonUK
| | - Kirsten A. Donald
- Division of Developmental Paediatrics, Department of Paediatrics and Child HealthRed Cross War Memorial Children's HospitalCape TownSouth Africa
- Neuroscience InstituteUniversity of Cape TownCape TownSouth Africa
| | - Layla E. Bradford
- Division of Developmental Paediatrics, Department of Paediatrics and Child HealthRed Cross War Memorial Children's HospitalCape TownSouth Africa
- Neuroscience InstituteUniversity of Cape TownCape TownSouth Africa
| | - Beatrice Lena
- C.J. Gorter MRI Center, Radiology DepartmentLeids Universitair Medisch CentrumLeidenthe Netherlands
| | | | - Talat Shama
- Infectious Diseases DivisionInternational Centre for Diarrheal Disease ResearchDhakaBangladesh
| | - Jasmine Siew
- Laboratories of Cognitive Neuroscience, Division of Developmental Medicine, Department of MedicineBoston Children's HospitalBostonMassachusettsUSA
| | - Lydia Sekoli
- Institute for Cellular and Molecular Medicine, Department of Medical Immunology, Faculty of Health SciencesUniversity of PretoriaPretoriaSouth Africa
| | - Jeanne van Rensburg
- Institute for Cellular and Molecular Medicine, Department of Medical Immunology, Faculty of Health SciencesUniversity of PretoriaPretoriaSouth Africa
| | - Michael S. Pepper
- Institute for Cellular and Molecular Medicine, Department of Medical Immunology, Faculty of Health SciencesUniversity of PretoriaPretoriaSouth Africa
| | - Amna Khan
- Department of Paediatrics & Child HealthThe Aga Khan UniversityKarachiPakistan
| | - Akber Madhwani
- Department of Paediatrics & Child HealthThe Aga Khan UniversityKarachiPakistan
| | - Frank A. Banda
- University of North Carolina Global ProjectsLusakaZambia
| | - Mwila L. Mwila
- University of North Carolina Global ProjectsLusakaZambia
| | - Adam R. Cassidy
- Botswana Harvard Health PartnershipGaboroneBotswana
- Department of Psychiatry & PsychologyMayo ClinicRochesterMinnesotaUSA
- Department of Pediatric & Adolescent MedicineMayo ClinicRochesterMinnesotaUSA
| | | | - Dolly Sephi
- Botswana Harvard Health PartnershipGaboroneBotswana
| | | | | | | | | | | | - Steven C. R. Williams
- Department of Neuroimaging, Institute of Psychiatry, Psychology & NeuroscienceKing's College LondonLondonUK
| | - Sean Deoni
- MNCH D&T, Bill & Melinda Gates FoundationSeattleWAUSA
| |
Collapse
|
4
|
Ottesen JA, Tong E, Emblem KE, Latysheva A, Zaharchuk G, Bjørnerud A, Grøvik E. Semi-Supervised Learning Allows for Improved Segmentation With Reduced Annotations of Brain Metastases Using Multicenter MRI Data. J Magn Reson Imaging 2025; 61:2469-2479. [PMID: 39792624 PMCID: PMC12063759 DOI: 10.1002/jmri.29686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Deep learning-based segmentation of brain metastases relies on large amounts of fully annotated data by domain experts. Semi-supervised learning offers potential efficient methods to improve model performance without excessive annotation burden. PURPOSE This work tests the viability of semi-supervision for brain metastases segmentation. STUDY TYPE Retrospective. SUBJECTS There were 156, 65, 324, and 200 labeled scans from four institutions and 519 unlabeled scans from a single institution. All subjects included in the study had diagnosed with brain metastases. FIELD STRENGTH/SEQUENCES 1.5 T and 3 T, 2D and 3D T1-weighted pre- and post-contrast, and fluid-attenuated inversion recovery (FLAIR). ASSESSMENT Three semi-supervision methods (mean teacher, cross-pseudo supervision, and interpolation consistency training) were adapted with the U-Net architecture. The three semi-supervised methods were compared to their respective supervised baseline on the full and half-sized training. STATISTICAL TESTS Evaluation was performed on a multinational test set from four different institutions using 5-fold cross-validation. Method performance was evaluated by the following: the number of false-positive predictions, the number of true positive predictions, the 95th Hausdorff distance, and the Dice similarity coefficient (DSC). Significance was tested using a paired samples t test for a single fold, and across all folds within a given cohort. RESULTS Semi-supervision outperformed the supervised baseline for all sites with the best-performing semi-supervised method achieved an on average DSC improvement of 6.3% ± 1.6%, 8.2% ± 3.8%, 8.6% ± 2.6%, and 15.4% ± 1.4%, when trained on half the dataset and 3.6% ± 0.7%, 2.0% ± 1.5%, 1.8% ± 5.7%, and 4.7% ± 1.7%, compared to the supervised baseline on four test cohorts. In addition, in three of four datasets, the semi-supervised training produced equal or better results than the supervised models trained on twice the labeled data. DATA CONCLUSION Semi-supervised learning allows for improved segmentation performance over the supervised baseline, and the improvement was particularly notable for independent external test sets when trained on small amounts of labeled data. PLAIN LANGUAGE SUMMARY Artificial intelligence requires extensive datasets with large amounts of annotated data from medical experts which can be difficult to acquire due to the large workload. To compensate for this, it is possible to utilize large amounts of un-annotated clinical data in addition to annotated data. However, this method has not been widely tested for the most common intracranial brain tumor, brain metastases. This study shows that this approach allows for data efficient deep learning models across multiple institutions with different clinical protocols and scanners. LEVEL OF EVIDENCE 3 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Jon André Ottesen
- Computational Radiology and Artificial Intelligence (CRAI) Research Group, Division of Radiology and Nuclear MedicineOslo University HospitalOsloNorway
- Department of Physics, Faculty of Mathematics and Natural SciencesUniversity of OsloOsloNorway
| | - Elizabeth Tong
- Department of RadiologyStanford UniversityStanfordCaliforniaUSA
| | - Kyrre Eeg Emblem
- Department of Physics and Computational Radiology, Division of Radiology and Nuclear MedicineOslo University HospitalOsloNorway
- Institute of Clinical Medicine, Faculty of MedicineUniversity of OsloOsloNorway
| | - Anna Latysheva
- Division of Radiology and Nuclear MedicineOslo University HospitalOsloNorway
| | - Greg Zaharchuk
- Department of RadiologyStanford UniversityStanfordCaliforniaUSA
| | - Atle Bjørnerud
- Computational Radiology and Artificial Intelligence (CRAI) Research Group, Division of Radiology and Nuclear MedicineOslo University HospitalOsloNorway
- Department of Physics, Faculty of Mathematics and Natural SciencesUniversity of OsloOsloNorway
| | - Endre Grøvik
- Department of RadiologyÅlesund Hospital, Møre og Romsdal Hospital TrustÅlesundNorway
- Department of PhysicsNorwegian University of Science and TechnologyTrondheimNorway
| |
Collapse
|
5
|
Tomaka AA, Pojda D, Tarnawski M, Luchowski L. Transformation trees - Documentation of multimodal image registration. Comput Biol Med 2025; 193:110311. [PMID: 40409031 DOI: 10.1016/j.compbiomed.2025.110311] [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: 01/31/2025] [Revised: 04/18/2025] [Accepted: 04/29/2025] [Indexed: 05/25/2025]
Abstract
Multimodal image registration plays a key role in creating digital patient models by combining data from different imaging techniques into a single coordinate system. This process often involves multiple sequential and interconnected transformations, which must be well-documented to ensure transparency and reproducibility. In this paper, we propose the use of transformation trees as a method for structured recording and management of these transformations. This approach has been implemented in the dpVision software and uses a dedicated .dpw file format to store hierarchical relationships between images, transformations, and motion data. Transformation trees allow precise tracking of all image processing steps, reduce the need to store multiple copies of the same data, and enable the indirect registration of images that do not share common reference points. This improves the reproducibility of the analyses and facilitates later processing and integration of images from different sources. The practical application of this method is demonstrated with examples from orthodontics, including the integration of 3D face scans, intraoral scans, and CBCT images, as well as the documentation of mandibular motion. Beyond orthodontics, this method can be applied in other fields that require systematic management of image registration processes, such as maxillofacial surgery, oncology, and biomechanical analysis. Maintaining long-term data consistency is essential for both scientific research and clinical practice. It enables easier comparison of results in longitudinal studies, improves retrospective analysis, and supports the development of artificial intelligence algorithms by providing standardized and well-documented datasets. The proposed approach enhances data organization, allows for efficient analysis, and facilitates the reuse of information in future studies and diagnostic procedures.
Collapse
Affiliation(s)
- Agnieszka Anna Tomaka
- Institute of Theoretical and Applied Informatics, Polish Academy of Sciences, Baltycka 5, Gliwice, 44-100, Poland.
| | - Dariusz Pojda
- Institute of Theoretical and Applied Informatics, Polish Academy of Sciences, Baltycka 5, Gliwice, 44-100, Poland
| | - Michał Tarnawski
- Institute of Theoretical and Applied Informatics, Polish Academy of Sciences, Baltycka 5, Gliwice, 44-100, Poland
| | - Leszek Luchowski
- Institute of Theoretical and Applied Informatics, Polish Academy of Sciences, Baltycka 5, Gliwice, 44-100, Poland
| |
Collapse
|
6
|
Gibson K, Walsh M, Hynd M, Eisenlohr-Moul T, Walsh E, Bondy E, Gray R, Brierley J, Bizzell J, Styner M, Dichter GS, Schiller CE. The effects of estradiol on subcortical brain volumes in perimenopausal-onset depression. J Affect Disord 2025; 377:45-52. [PMID: 39983774 PMCID: PMC11997973 DOI: 10.1016/j.jad.2025.02.068] [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: 12/02/2024] [Revised: 02/05/2025] [Accepted: 02/17/2025] [Indexed: 02/23/2025]
Abstract
BACKGROUND Perimenopause is associated with increases in depressive and vasomotor symptoms (VMS), which can be alleviated with transdermal estradiol (TE2) administration. Subcortical brain regions are commonly implicated in depression, are dense with E2 receptors and are susceptible to volumetric changes resulting from E2 regulation of synaptic density. No studies have examined linkages among TE2 administration, perimenopausal-onset major depression (PO-MDD) and subcortical brain volumes. METHODS This is an exploratory data analysis of change in subcortical brain volumes measured via 3 T MRI before and after three-weeks of TE2 administration in 14 women with PO-MDD and 17 euthymic controls. Regions of interest were the hippocampus, amygdala, putamen, thalamus, and caudate nucleus. Multilevel models examined relations between baseline volumes and volumetric changes with symptom trajectories in the PO-MDD group. RESULTS In the PO-MDD group, anhedonia (p < 0.004) and VMS (p < 0.001) significantly reduced following TE2 administration. There was a significant Group X Time interaction in the right hippocampus (p < 0.01), driven by volume increases in the control group (p < 0.001). In the PO-MDD group, change in right hippocampal volumes significantly predicted decreases in anhedonia trajectories from baseline to week 2 and week 3 (p's < 0.001) and decreases in VMS across all timepoints (p's < 0.001). DISCUSSION Women with PO-MDD, who presented with more severe baseline anhedonia and VMS, experienced greater reductions in anhedonia, VMS, and hippocampal volumes, demonstrating a greater response to E2. Hippocampal volume change may be a candidate for predicting treatment response to E2 for anhedonia and vasomotor symptoms in women with PO-MDD. These findings should be validated with a placebo-controlled trial.
Collapse
Affiliation(s)
- Kathryn Gibson
- Department of Psychiatry, University of North Carolina Chapel Hill, Chapel Hill, NC 27514, USA.
| | - Melissa Walsh
- Department of Psychiatry, University of North Carolina Chapel Hill, Chapel Hill, NC 27514, USA
| | - Megan Hynd
- Department of Psychology and Neuroscience, University of North Carolina-Chapel Hill, Chapel Hill, NC 27514, USA
| | - Tory Eisenlohr-Moul
- Department of Psychiatry, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Erin Walsh
- Department of Psychiatry, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Erin Bondy
- Department of Psychiatry, University of North Carolina Chapel Hill, Chapel Hill, NC 27514, USA
| | - Reese Gray
- Department of Psychology and Neuroscience, University of North Carolina-Chapel Hill, Chapel Hill, NC 27514, USA
| | - James Brierley
- Department of Psychology and Neuroscience, University of North Carolina-Chapel Hill, Chapel Hill, NC 27514, USA
| | - Joshua Bizzell
- Department of Psychiatry, University of North Carolina Chapel Hill, Chapel Hill, NC 27514, USA
| | - Martin Styner
- Department of Psychiatry, University of North Carolina Chapel Hill, Chapel Hill, NC 27514, USA
| | - Gabriel S Dichter
- Department of Psychiatry, University of North Carolina Chapel Hill, Chapel Hill, NC 27514, USA; Department of Psychology and Neuroscience, University of North Carolina-Chapel Hill, Chapel Hill, NC 27514, USA; Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, Chapel Hill, NC 27510, USA
| | - Crystal E Schiller
- Department of Psychiatry, University of North Carolina Chapel Hill, Chapel Hill, NC 27514, USA
| |
Collapse
|
7
|
Blini E, D'Imperio D, Romeo Z, De Filippo De Grazia M, Passarini L, Pilosio C, Meneghello F, Bonato M, Zorzi M. Susceptibility to multitasking in stroke is associated to multiple-demand system damage and leads to lateralized visuospatial deficits. Commun Biol 2025; 8:734. [PMID: 40355698 PMCID: PMC12069553 DOI: 10.1038/s42003-025-08074-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 04/10/2025] [Indexed: 05/14/2025] Open
Abstract
Cognitive impairment after stroke is heterogeneous: there is no strict correspondence between brain damage and magnitude of deficit or recovery. Protective factors such as cognitive or brain reserve have been invoked to explain the mismatch. Here, we consider the opposite point of view: the instances in which this protection is overturned. We leveraged on multitasking to stress the brain's processing limits and unveil deficits that may be missed by standard testing in a sample of 46 patients with unilateral subacute to chronic stroke and no sign of lateralized spatial-attentional disorders at neuropsychological paper-and-pencil tests. Multivariate analyses identified a phenotype of patients with high susceptibility to multitasking, showing stark contralesional spatial awareness deficit only when multitasking. Multivariate brain-behavior mapping based on lesions location and structural disconnections pointed to the Multiple-Demand System, a network of frontal and fronto-parietal areas subserving domain-general processes. Damage in this network may critically interact with domain-specific processes, resulting in subtle and yet invalidating deficits. Indeed, these patients (one-third of the sample) presented worse performance in tests evaluating activities of daily living and domain-general abilities. We conclude that the theoretical construct of susceptibility to multitasking helps understanding what marks the passage to clinically visible deficits after brain damage.
Collapse
Affiliation(s)
- Elvio Blini
- Department of General Psychology and Padova Neuroscience Center, University of Padova, Padua, Italy
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Firenze, Florence, Italy
| | | | - Zaira Romeo
- Department of General Psychology and Padova Neuroscience Center, University of Padova, Padua, Italy
- Neuroscience Institute, National Research Council, Padova, Italy
| | | | | | | | | | - Mario Bonato
- Department of General Psychology and Padova Neuroscience Center, University of Padova, Padua, Italy
| | - Marco Zorzi
- Department of General Psychology and Padova Neuroscience Center, University of Padova, Padua, Italy.
- IRCCS San Camillo Hospital, Venice, Italy.
| |
Collapse
|
8
|
Madelung CF, Løkkegaard A, Fuglsang SA, Marques MM, Boer VO, Madsen KH, Hejl AM, Meder D, Siebner HR. High-resolution mapping of substantia nigra in Parkinson's disease using 7 tesla magnetic resonance imaging. NPJ Parkinsons Dis 2025; 11:113. [PMID: 40328786 PMCID: PMC12056087 DOI: 10.1038/s41531-025-00972-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 04/19/2025] [Indexed: 05/08/2025] Open
Abstract
Parkinson's disease causes a progressive loss of dopaminergic neurons and iron accumulation in the substantia nigra pars compacta. Using ultra-high field magnetic resonance imaging (MRI) at 7 tesla in 43 Parkinson's patients and 24 healthy controls, we analyzed the voxel-wise pattern of structural disintegration of dopamine neurons with neuromelanin-sensitive MRI, along with assessing iron accumulation using R2* and quantitative susceptibility mapping (QSM). We also explored correlations between these measures and the severity of residual motor symptoms in the on-medication state and other clinical variables. Differences were most notable in the nigrosomes within the pars compacta, with patients showing reduced neuromelanin signals and increased QSM values. Severity and asymmetry of motor symptoms correlated with higher R2* and QSM values in nigrosome N1. Thus, ultra-high field MRI provides high-resolution maps of various aspects of the underlying neurodegenerative process which reflect individual motor impairment in Parkinson's disease.
Collapse
Affiliation(s)
- Christopher F Madelung
- Danish Research Centre for Magnetic Resonance, Center for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Amager and Hvidovre, Copenhagen, Denmark
- Department of Neurology, Copenhagen University Hospital Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Annemette Løkkegaard
- Department of Neurology, Copenhagen University Hospital Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Søren A Fuglsang
- Danish Research Centre for Magnetic Resonance, Center for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Amager and Hvidovre, Copenhagen, Denmark
| | - Marta M Marques
- Danish Research Centre for Magnetic Resonance, Center for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Amager and Hvidovre, Copenhagen, Denmark
| | - Vincent O Boer
- Danish Research Centre for Magnetic Resonance, Center for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Amager and Hvidovre, Copenhagen, Denmark
| | - Kristoffer H Madsen
- Danish Research Centre for Magnetic Resonance, Center for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Amager and Hvidovre, Copenhagen, Denmark
- Department of Applied Mathematics and Computer Science, Technical University of Denmark, Kgs., Lyngby, Denmark
| | - Anne-Mette Hejl
- Department of Neurology, Copenhagen University Hospital Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - David Meder
- Danish Research Centre for Magnetic Resonance, Center for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Amager and Hvidovre, Copenhagen, Denmark.
| | - Hartwig R Siebner
- Danish Research Centre for Magnetic Resonance, Center for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Amager and Hvidovre, Copenhagen, Denmark.
- Department of Neurology, Copenhagen University Hospital Bispebjerg and Frederiksberg, Copenhagen, Denmark.
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
9
|
Struck AF, Garcia‐Ramos C, Prabhakaran V, Nair V, Adluru N, Adluru A, Almane D, Jones JE, Hermann BP. Cognitive and brain health in juvenile myoclonic epilepsy: Role of social determinants of health. Epilepsia 2025; 66:1641-1651. [PMID: 39963015 PMCID: PMC12097475 DOI: 10.1111/epi.18296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 01/21/2025] [Accepted: 01/21/2025] [Indexed: 03/01/2025]
Abstract
OBJECTIVE Juvenile myoclonic epilepsy (JME) is a prevalent genetic generalized epilepsy with linked abnormalities in cognition, behavior, and brain structure. Well recognized is the potential for advancing understanding of the epigenetic contributions to the neurobehavioral complications of JME, but to date there has been no examination of the role of socioeconomic disadvantage in regard to the cognitive and brain health of JME, which is the focus of this investigation. METHODS Seventy-seven patients with JME and 44 unrelated controls underwent neuropsychological assessment, structural neuroimaging, and clinical interview to delineate epilepsy history and aspects of family status. The Area Deprivation Index characterized the presence and degree of neighborhood disadvantage, which was examined in relation to cognitive factor scores underlying a comprehensive neuropsychological test battery, academic metrics, integrity of brain structure, and family characteristics. RESULTS JME participants resided in neighborhoods associated with significantly more socioeconomic disadvantage, which was associated with significantly poorer performance across all three cognitive factor scores and reading fluency. JME was associated with significant reduction of total subcortical gray matter (GM) but not total cortical gray or white matter volumes. Among controls, participants residing in more advantaged areas exhibited increased volumes of total subcortical GM and diverse subcortical structures as well as areas of increased cortical thickness and volume in frontal/prefrontal regions, findings that were compromised or not evident in JME, raising the possibility of disease-related attenuation of socioeconomic advantage. SIGNIFICANCE Socioeconomic disadvantage in JME is associated with adverse effects on cognitive and academic status, whereas socioeconomic advantage in controls is associated with increased brain volumes and thickness, markers of brain health that were largely attenuated or absent in JME. The associations detected here argue for the need to better integrate the social determinants of health with genetic and epigenetic factors in advancing understanding of cognitive and brain health in JME.
Collapse
Affiliation(s)
- Aaron F. Struck
- Department of NeurologyUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Department of NeurologyWilliam S. Middleton Veterans Administration HospitalMadisonWisconsinUSA
| | - Camille Garcia‐Ramos
- Department of NeurologyUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Vivek Prabhakaran
- Department of RadiologyUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Veena Nair
- Department of RadiologyUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Nagesh Adluru
- Department of RadiologyUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Waisman CenterUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Anusha Adluru
- Department of RadiologyUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
- Waisman CenterUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Dace Almane
- Department of NeurologyUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Jana E. Jones
- Department of NeurologyUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Bruce P. Hermann
- Department of NeurologyUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| |
Collapse
|
10
|
Fan D, Zhao J, Li C, Wang X, Zhang R, Zhu Q, Wang M, Si H, Zhang D, Sun L. MA-SAM: A Multi-Atlas Guided SAM Using Pseudo Mask Prompts Without Manual Annotation for Spine Image Segmentation. IEEE TRANSACTIONS ON MEDICAL IMAGING 2025; 44:2157-2169. [PMID: 40030770 DOI: 10.1109/tmi.2024.3524570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Accurate spine segmentation is crucial in clinical diagnosis and treatment of spine diseases. However, due to the complexity of spine anatomical structure, it has remained a challenging task to accurately segment spine images. Recently, the segment anything model (SAM) has achieved superior performance for image segmentation. However, generating high-quality points and boxes is still laborious for high-dimensional medical images. Meanwhile, an accurate mask is difficult to obtain. To address these issues, in this paper, we propose a multi-atlas guided SAM using multiple pseudo mask prompts for spine image segmentation, called MA-SAM. Specifically, we first design a multi-atlas prompt generation sub-network to obtain the anatomical structure prompts. More specifically, we use a network to obtain coarse mask of the input image. Then atlas label maps are registered to the coarse mask. Subsequently, a SAM-based segmentation sub-network is used to segment images. Specifically, we first utilize adapters to fine-tune the image encoder. Meanwhile, we use a prompt encoder to learn the anatomical structure prior knowledge from the multi-atlas prompts. Finally, a mask decoder is used to fuse the image and prompt features to obtain the segmentation results. Moreover, to boost the segmentation performance, different scale features from the prompt encoder are concatenated to the Upsample Block in the mask decoder. We validate our MA-SAM on the two spine segmentation tasks, including spine anatomical structure segmentation with CT images and lumbosacral plexus segmentation with MR images. Experiment results suggest that our method achieves better segmentation performance than SAM with points, boxes, and mask prompts.
Collapse
|
11
|
Ruohola A, Haapamäki V, Salli E, Kaseva T, Kangasniemi M, Savolainen S. Bone-wise rigid registration of femur, tibia, and fibula for the tracking of temporal changes. J Appl Clin Med Phys 2025; 26:e70053. [PMID: 40066783 PMCID: PMC12059268 DOI: 10.1002/acm2.70053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/27/2024] [Accepted: 12/25/2024] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND Multiple myeloma (MM) induces temporal alterations in bone structure, such as osteolytic bone lesions, which are challenging to identify through manual image interpretation. The large variation in radiologists' assessments, even at expert centers, further complicates diagnosis. Automatic image analysis methods, including segmentation and registration, can expedite detecting and tracking these bone changes. PURPOSE This study presents an automated pipeline for accurately tracking temporal changes in the femurs, tibiae, and fibulae of MM patients using 3D whole-body CT images. The pipeline leverages image segmentation, rigid registration, and temporal subtraction to accelerate disease monitoring and support clinical decision-making. METHODS A convolutional neural network (CNN) was trained to segment bones in 3D CT images of 30 MM patients. Nine patients with pre- and post-diagnosis CT scans were used to validate the segmentation and registration process. A two-phase bone-wise rigid registration was applied, followed by temporal subtraction to generate difference images. Segmentation and registration accuracy were assessed using the Dice similarity coefficient (DSC) and mean surface distance (MSD). The proposed method was compared to a non-rigid registration method. RESULTS The neural network segmentation resulted in a mean DSC of 0.93 across all bone types and all test data. The registration accuracy measured by the mean DSC across the test data was at least 0.94 for all bone types. The second phase of rigid registration improved the registration fibulae. Metric-wise, the nonrigid method performed better but diminished lesion visibility in difference images. CONCLUSIONS An automated pipeline for the longitudinal tracking of bone alterations was presented. Both segmentation and registration demonstrated high accuracy as measured by DSC and MSD. In the difference images produced by temporal subtraction, osteolytic lesions were clearly visible in the femurs. The methodology lays a solid foundation for future improvements, such as inclusion of the axial spine.
Collapse
Affiliation(s)
- Arttu Ruohola
- HUS Diagnostic CenterDepartment of RadiologyUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
| | - Ville Haapamäki
- HUS Diagnostic CenterDepartment of RadiologyUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
| | - Eero Salli
- HUS Diagnostic CenterDepartment of RadiologyUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
| | - Tuomas Kaseva
- HUS Diagnostic CenterDepartment of RadiologyUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
| | - Marko Kangasniemi
- HUS Diagnostic CenterDepartment of RadiologyUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
| | - Sauli Savolainen
- HUS Diagnostic CenterDepartment of RadiologyUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
- Department of PhysicsUniversity of HelsinkiHelsinkiFinland
| |
Collapse
|
12
|
Aglinskas A, Bergeron A, Anzellotti S. Understanding heterogeneity in psychiatric disorders: A method for identifying subtypes and parsing comorbidity. Psychiatry Clin Neurosci 2025. [PMID: 40304074 DOI: 10.1111/pcn.13829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 03/17/2025] [Accepted: 04/07/2025] [Indexed: 05/02/2025]
Abstract
AIM Most psychiatric and neurodevelopmental disorders are heterogeneous. Neural abnormalities in patients might differ in magnitude and kind, giving rise to distinct subtypes that can be partly overlapping (comorbidity). Identifying disorder-related individual differences is challenging due to the overwhelming presence of disorder-unrelated variation shared with healthy controls. Recently, Contrastive Variational Autoencoders (CVAEs) have been shown to separate disorder-related individual variation from disorder-unrelated variation. However, it is not known if CVAEs can also satisfy the other key desiderata for psychiatric research: capturing disease subtypes and disentangling comorbidity. In this paper, we compare CVAEs to other methods as a function of hyperparameters, such as model size and training data availability. We also introduce a new architecture for modeling comorbid disorders and test a novel training procedure for CVAEs that improves their reproducibility. METHODS We use synthetic neuroanatomical MRI data with known ground truth for shared and disorder-specific effects and study the performance of the CVAE and non-contrastive baseline models at detecting disorder-subtypes and disentangling comorbidity in brain images varying along shared and disorder-specific dimensions. RESULTS CVAE models consistently outperformed non-contrastive alternatives as measured by correlation with disorder-specific ground truth effects and accuracy of subtype discovery. The CVAE also successfully disentangled neuroanatomical loci of comorbid disorders, due to its novel architecture. Improved training procedure reduced variability in the results by up to 5.5×. CONCLUSION The results showcase how the CVAE can be used as an overall framework in precision psychiatry studies, enabling reliable detection of interpretable neuromarkers, discovering disorder subtypes and disentangling comorbidity.
Collapse
Affiliation(s)
- Aidas Aglinskas
- Department of Psychology and Neuroscience, Boston College, Chestnut Hill, Massachusetts, USA
| | - Alicia Bergeron
- Department of Psychology and Neuroscience, Boston College, Chestnut Hill, Massachusetts, USA
| | - Stefano Anzellotti
- Department of Psychology and Neuroscience, Boston College, Chestnut Hill, Massachusetts, USA
| |
Collapse
|
13
|
Im J, Xiang B, Levasseur VA, Sukstanskii AL, Quirk JD, Kothapalli SVVN, Cross AH, Yablonskiy DA. Unraveling the major role of vascular (R2') contributions to R2* signal relaxation at ultra-high-field MRI: A comprehensive analysis with quantitative gradient recalled echo in mouse brain. Magn Reson Med 2025. [PMID: 40294081 DOI: 10.1002/mrm.30529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 03/08/2025] [Accepted: 03/25/2025] [Indexed: 04/30/2025]
Abstract
PURPOSE Ultra-high-field (UHF) R2* relaxometry is often used for in vivo analysis of biological tissue microstructure without accounting for vascular contributions to R2* signal, that is, the BOLD signal component, and magnetic field inhomogeneities. These effects are especially important at UHF as their contribution to R2* scales linearly with magnetic field. Our study aims to report on the results of separate contributions of R2t* (tissue-specific sub-component) and R2' (vascular BOLD sub-component), corrected for the adverse effects of magnetic field inhomogeneities, to the total R2* signal at in vivo UHF MRI of mouse brain. METHODS Four healthy, 8-week-old C57BL/6J mice were imaged in vivo with multi-gradient echo MRI at 9.4 T and analyzed using the quantitative gradient recalled echo (qGRE) approach. A segmentation protocol was established using the Dorr Mouse Brain Atlas and ANTs Syn registration to warp template brain region labels to subject qGRE maps. RESULTS By separating R2' contribution from R2* signal, we have established normative R2t* data in mouse brain. Our findings revealed significant contributions of R2' to R2*, with approximately 42% of the R2* signal arising from vascular contributions, thus suggesting the R2t* as a more accurate metric for quantifying tissue microstructural information and its changes in neurodegenerative diseases. CONCLUSION qGRE approach allows efficient separation of tissue microstructure-specific (R2t*), vascular BOLD (R2'), and background gradients contributions to the total R2* relaxation at UHF MRI. Due to low concentration of non-heme iron in mouse brain, major contribution to R2t* results from tissue cellular components.
Collapse
Affiliation(s)
- Joanna Im
- Department of Biology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Biao Xiang
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Victoria A Levasseur
- Department of Neurology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Alexander L Sukstanskii
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - James D Quirk
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Satya V V N Kothapalli
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Anne H Cross
- Department of Neurology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Dmitriy A Yablonskiy
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
14
|
Tustison NJ, Chen M, Kronman FN, Duda JT, Gamlin C, Tustison MG, Kunst M, Dalley R, Sorenson S, Wang Q, Ng L, Kim Y, Gee JC. Modular strategies for spatial mapping of diverse cell type data of the mouse brain. RESEARCH SQUARE 2025:rs.3.rs-6289741. [PMID: 40297692 PMCID: PMC12036443 DOI: 10.21203/rs.3.rs-6289741/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Large-scale, international collaborative efforts by members of the BRAIN Initiative Cell Census Network (BICCN) consortium are aggregating the most comprehensive reference database to date for diverse cell type profiling of the mouse brain, which encompasses over 40 different multi-modal profiling techniques from more than 30 research groups. One central challenge for this integrative effort has been the need to map these unique datasets into common reference spaces such that the spatial, structural, and functional information from different cell types can be jointly analyzed. However, significant variation in the acquisition, tissue processing, and imaging techniques across data types makes mapping such diverse data a multifarious problem. Different data types exhibit unique tissue distortion and signal characteristics that precludes a single mapping strategy from being generally applicable across all cell type data. Tailored mapping approaches are often needed to address the unique barriers present in each modality. This work highlights modular atlas mapping strategies developed across separate BICCN studies using the Advanced Normalization Tools Ecosystem (ANTsX) to map spatial transcriptomic (MERFISH) and high-resolution morphology (fMOST) mouse brain data into the Allen Common Coordinate Framework (AllenCCFv3), and developmental (MRI and LSFM) data into the Developmental Common Coordinate Framework (DevCCF). We discuss common mapping strategies that can be shared across modalities and driven by specific challenges from each data type. These mapping strategies include novel open-source contributions that are made publicly available through ANTSX. These include 1) a velocity flow-based approach for continuously mapping developmental trajectories such as that characterizing the DevCCF and 2) an automated framework for determining structural morphology solely through the leveraging of publicly resources. Finally, we provide general guidance to aid investigators to tailor these strategies to address unique data challenges without the need to develop additional specialized software.
Collapse
Affiliation(s)
- Nicholas J. Tustison
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA
| | - Min Chen
- Department of Radiology, University of Pennsylvania, Philadelphia, PA
| | - Fae N. Kronman
- Department of Neural and Behavioral Sciences, Penn State University, Hershey, PA
| | - Jeffrey T. Duda
- Department of Radiology, University of Pennsylvania, Philadelphia, PA
| | | | - Mia G. Tustison
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA
- Department of Neural and Behavioral Sciences, Penn State University, Hershey, PA
- Allen Institute for Brain Science, Seattle, WA
| | | | | | | | | | - Lydia Ng
- Allen Institute for Brain Science, Seattle, WA
| | - Yongsoo Kim
- Department of Neural and Behavioral Sciences, Penn State University, Hershey, PA
| | - James C. Gee
- Department of Radiology, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
15
|
Zhang RZ, Ezhov I, Balcerak M, Zhu A, Wiestler B, Menze B, Lowengrub JS. Personalized predictions of Glioblastoma infiltration: Mathematical models, Physics-Informed Neural Networks and multimodal scans. Med Image Anal 2025; 101:103423. [PMID: 39700844 DOI: 10.1016/j.media.2024.103423] [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: 01/09/2024] [Revised: 09/01/2024] [Accepted: 12/01/2024] [Indexed: 12/21/2024]
Abstract
Predicting the infiltration of Glioblastoma (GBM) from medical MRI scans is crucial for understanding tumor growth dynamics and designing personalized radiotherapy treatment plans. Mathematical models of GBM growth can complement the data in the prediction of spatial distributions of tumor cells. However, this requires estimating patient-specific parameters of the model from clinical data, which is a challenging inverse problem due to limited temporal data and the limited time between imaging and diagnosis. This work proposes a method that uses Physics-Informed Neural Networks (PINNs) to estimate patient-specific parameters of a reaction-diffusion partial differential equation (PDE) model of GBM growth from a single 3D structural MRI snapshot. PINNs embed both the data and the PDE into a loss function, thus integrating theory and data. Key innovations include the identification and estimation of characteristic non-dimensional parameters, a pre-training step that utilizes the non-dimensional parameters and a fine-tuning step to determine the patient specific parameters. Additionally, the diffuse-domain method is employed to handle the complex brain geometry within the PINN framework. The method is validated on both synthetic and patient datasets, showing promise for personalized GBM treatment through parametric inference within clinically relevant timeframes.
Collapse
Affiliation(s)
- Ray Zirui Zhang
- Department of Mathematics, University of California Irvine, USA.
| | | | | | | | | | | | - John S Lowengrub
- Department of Mathematics, University of California Irvine, USA; Department of Biomedical Engineering, University of California Irvine, USA.
| |
Collapse
|
16
|
Baller EB, Luo AC, Schindler MK, Cooper EC, Pecsok MK, Cieslak MC, Martin ML, Bar-Or A, Elahi A, Perrone CM, Spangler BC, Satterthwaite TD, Shinohara RT. Uncinate Fasciculus Lesion Burden and Anxiety in Multiple Sclerosis. JAMA Netw Open 2025; 8:e254751. [PMID: 40227683 PMCID: PMC11997724 DOI: 10.1001/jamanetworkopen.2025.4751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 02/02/2025] [Indexed: 04/15/2025] Open
Abstract
Importance Multiple sclerosis (MS) is an immune-mediated neurological disorder that affects 2.4 million people worldwide, and up to 60% experience anxiety. Objective To investigate whether anxiety in MS is associated with white matter lesion burden in the uncinate fasciculus (UF). Design, Setting, and Participants This was a retrospective case-control study of participants aged 18 years or older diagnosed with MS by an MS specialist and identified from the electronic medical record at a single-center academic medical specialty MS clinic in Pennsylvania. Participants received research-quality 3-Tesla magnetic resonance neuroimaging as part of MS clinical care from January 6, 2010, to February 14, 2018. After excluding participants with poor image quality, participants were stratified into 3 groups naturally balanced in age and sex: (1) MS without anxiety, (2) MS with mild anxiety, and (3) MS with severe anxiety. Analyses were performed from June 1 to September 30, 2024. Exposure Anxiety diagnosis and anxiolytic medication. Main Outcomes and Measures Main outcomes were whether patients with severe anxiety had greater lesion burden in the UF than those without anxiety and whether higher anxiety severity was associated with greater UF lesion burden. Generalized additive models were used, with the burden of lesions (eg, proportion of fascicle impacted) within the UF as the outcome measure and sex, spline of age, and total brain volume as covariates. Results Among 372 patients with MS (mean [SD] age, 47.7 [11.4] years; 296 [80%] female), after anxiety phenotype stratification, 99 (27%) had no anxiety (mean [SD] age, 49.4 [11.7] years; 74 [75%] female), 249 (67%) had mild anxiety (mean [SD] age, 47.1 [11.1] years; 203 [82%] female), and 24 (6%) had severe anxiety (mean [SD] age, 47.0 [12.2] years; 19 [79%] female). UF burden was higher in patients with severe anxiety compared with no anxiety (T = 2.01 [P = .047]; Cohen f2, 0.19 [95% CI, 0.08-0.52]). Additionally, higher mean UF burden was associated with higher severity of anxiety (T = 2.09 [P = .04]; Cohen f2, 0.10 [95% CI, 0.05-0.21]). Conclusions and Relevance In this case-control study of UF lesion burden and anxiety in MS, overall lesion burden in the UF was associated with the presence and severity of anxiety. Future studies linking white matter lesion burden in the UF with treatment prognosis are warranted.
Collapse
Affiliation(s)
- Erica B. Baller
- Department of Psychiatry, University of Pennsylvania, Philadelphia
- Penn Lifespan Informatics and Neuroimaging Center, Philadelphia, Pennsylvania
| | - Audrey C. Luo
- Department of Psychiatry, University of Pennsylvania, Philadelphia
- Penn Lifespan Informatics and Neuroimaging Center, Philadelphia, Pennsylvania
| | - Matthew K. Schindler
- Department of Neurology, University of Pennsylvania, Philadelphia
- Center for Neuroinflammation and Neurotherapeutics, University of Pennsylvania, Philadelphia
| | - Elena C. Cooper
- Department of Psychiatry, University of Pennsylvania, Philadelphia
- Penn Lifespan Informatics and Neuroimaging Center, Philadelphia, Pennsylvania
| | | | - Matthew C. Cieslak
- Department of Psychiatry, University of Pennsylvania, Philadelphia
- Penn Lifespan Informatics and Neuroimaging Center, Philadelphia, Pennsylvania
| | - Melissa Lynne Martin
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia
| | - Amit Bar-Or
- Department of Neurology, University of Pennsylvania, Philadelphia
- Center for Neuroinflammation and Neurotherapeutics, University of Pennsylvania, Philadelphia
| | - Ameena Elahi
- Department of Information Services, University of Pennsylvania, Philadelphia
| | - Christopher M. Perrone
- Department of Neurology, University of Pennsylvania, Philadelphia
- Center for Neuroinflammation and Neurotherapeutics, University of Pennsylvania, Philadelphia
| | - Bailey C. Spangler
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia
| | - Theodore D. Satterthwaite
- Department of Psychiatry, University of Pennsylvania, Philadelphia
- Penn Lifespan Informatics and Neuroimaging Center, Philadelphia, Pennsylvania
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
| | - Russell T. Shinohara
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
| |
Collapse
|
17
|
Stone HL, Mitchell JL, Fuentes-Jimenez M, Tran JE, Yeatman JD, Yablonski M. Anatomically distinct regions in the inferior frontal cortex are modulated by task and reading skill. J Neurosci 2025; 45:e1767242025. [PMID: 40127940 PMCID: PMC12060616 DOI: 10.1523/jneurosci.1767-24.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 01/25/2025] [Accepted: 03/14/2025] [Indexed: 03/26/2025] Open
Abstract
Inferior frontal cortex (IFC) is a critical region for reading and language. This part of the cortex is highly heterogeneous in its structural and functional organization and shows high variability across individuals. Despite decades of research, the relationship between specific IFC regions and reading skill remains unclear. To shed light on the function of IFC in reading, we aim to (1) characterize the functional landscape of text-selective responses in IFC, while accounting for interindividual variability; and (2) examine how text-selective regions in the IFC relate to reading proficiency. To this end, children with a wide range of reading ability (N=66; age 7-14 years, 34 female, 32 male) completed functional MRI scans while performing two tasks on text and non-text visual stimuli. Importantly, both tasks do not explicitly require reading, and can be performed on all visual stimuli. This design allows us to tease apart stimulus-driven responses from task-driven responses and examine where in IFC task and stimulus interact. We were able to identify three anatomically-distinct, text-selective clusters of activation in IFC, in the inferior frontal sulcus (IFS), and dorsal and ventral precentral gyrus (PrG). These three regions showed a strong task effect that was highly specific to text. Furthermore, text-selectivity in the IFS and dorsal PrG was associated with reading proficiency, such that better readers showed higher selectivity to text. These findings suggest that text-selective regions in the IFC are sensitive to both stimulus and task, and highlight the importance of this region for proficient reading.Significance statement The inferior frontal cortex (IFC) is a critical region for language processing, yet despite decades of research, its relationship with reading skill remains unclear. In a group of children with a wide range of reading skills, we were able to identify three anatomically distinct text-selective clusters of activation in the IFC. These regions showed a strong task effect that was highly selective to text. Text-selectivity was positively correlated with reading proficiency, such that better readers showed higher selectivity to text, even in tasks that did not require reading. These findings suggest that multiple text-selective regions within IFC are sensitive to both stimulus and task, and highlight the critical role of IFC for reading proficiency.
Collapse
Affiliation(s)
- Hannah L Stone
- Graduate School of Education, Stanford University, Stanford, CA, 94305, USA
| | - Jamie L Mitchell
- Graduate School of Education, Stanford University, Stanford, CA, 94305, USA
- Department of Psychology, Stanford University, Stanford, CA, 94305, USA
| | | | - Jasmine E Tran
- Graduate School of Education, Stanford University, Stanford, CA, 94305, USA
| | - Jason D Yeatman
- Graduate School of Education, Stanford University, Stanford, CA, 94305, USA
- Department of Psychology, Stanford University, Stanford, CA, 94305, USA
- Division of Developmental-Behavioral Pediatrics, Department of Pediatrics, Stanford University School of Medicine, Stanford, California, 94305, USA
| | - Maya Yablonski
- Graduate School of Education, Stanford University, Stanford, CA, 94305, USA,
- Division of Developmental-Behavioral Pediatrics, Department of Pediatrics, Stanford University School of Medicine, Stanford, California, 94305, USA
| |
Collapse
|
18
|
Yang X, Li D, Deng L, Huang S, Wang J. TCDE-Net: An unsupervised dual-encoder network for 3D brain medical image registration. Comput Med Imaging Graph 2025; 123:102527. [PMID: 40147215 DOI: 10.1016/j.compmedimag.2025.102527] [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: 09/19/2024] [Revised: 02/24/2025] [Accepted: 03/05/2025] [Indexed: 03/29/2025]
Abstract
Medical image registration is a critical task in aligning medical images from different time points, modalities, or individuals, essential for accurate diagnosis and treatment planning. Despite significant progress in deep learning-based registration methods, current approaches still face considerable challenges, such as insufficient capture of local details, difficulty in effectively modeling global contextual information, and limited robustness in handling complex deformations. These limitations hinder the precision of high-resolution registration, particularly when dealing with medical images with intricate structures. To address these issues, this paper presents a novel registration network (TCDE-Net), an unsupervised medical image registration method based on a dual-encoder architecture. The dual encoders complement each other in feature extraction, enabling the model to effectively handle large-scale nonlinear deformations and capture intricate local details, thereby enhancing registration accuracy. Additionally, the detail-enhancement attention module aids in restoring fine-grained features, improving the network's capability to address complex deformations such as those at gray-white matter boundaries. Experimental results on the OASIS, IXI, and Hammers-n30r95 3D brain MR dataset demonstrate that this method outperforms commonly used registration techniques across multiple evaluation metrics, achieving superior performance and robustness. Our code is available at https://github.com/muzidongxue/TCDE-Net.
Collapse
Affiliation(s)
- Xin Yang
- Heilongjiang Provincial Key Laboratory of Complex Intelligent System and Integration, School of Automation, Harbin University of Science and Technology, Harbin, Heilongjiang 150080, China; Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, Guangdong 510060, China; Collaborative Innovation Center for Cancer Medicine, China; State Key Laboratory of Oncology in South China, China; Sun Yat-sen University Cancer Center, China; Department of Radiation Oncology, Guangzhou, Guangdong 510060, China.
| | - Dongxue Li
- Heilongjiang Provincial Key Laboratory of Complex Intelligent System and Integration, School of Automation, Harbin University of Science and Technology, Harbin, Heilongjiang 150080, China.
| | - Liwei Deng
- Heilongjiang Provincial Key Laboratory of Complex Intelligent System and Integration, School of Automation, Harbin University of Science and Technology, Harbin, Heilongjiang 150080, China.
| | - Sijuan Huang
- Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, Guangdong 510060, China; Collaborative Innovation Center for Cancer Medicine, China; State Key Laboratory of Oncology in South China, China; Sun Yat-sen University Cancer Center, China; Department of Radiation Oncology, Guangzhou, Guangdong 510060, China.
| | - Jing Wang
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, Guangdong 510631, China.
| |
Collapse
|
19
|
Gopalakrishnan V, Dey N, Chlorogiannis DD, Abumoussa A, Larson AM, Orbach DB, Frisken S, Golland P. Rapid patient-specific neural networks for intraoperative X-ray to volume registration. ARXIV 2025:arXiv:2503.16309v1. [PMID: 40166742 PMCID: PMC11957231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The integration of artificial intelligence in image-guided interventions holds transformative potential, promising to extract 3D geometric and quantitative information from conventional 2D imaging modalities during complex procedures. Achieving this requires the rapid and precise alignment of 2D intraoperative images (e.g., X-ray) with 3D preoperative volumes (e.g., CT, MRI). However, current 2D/3D registration methods fail across the broad spectrum of procedures dependent on X-ray guidance: traditional optimization techniques require custom parameter tuning for each subject, whereas neural networks trained on small datasets do not generalize to new patients or require labor-intensive manual annotations, increasing clinical burden and precluding application to new anatomical targets. To address these challenges, we present xvr, a fully automated framework for training patient-specific neural networks for 2D/3D registration. xvr uses physics-based simulation to generate abundant high-quality training data from a patient's own preoperative volumetric imaging, thereby overcoming the inherently limited ability of supervised models to generalize to new patients and procedures. Furthermore, xvr requires only 5 min of training per patient, making it suitable for emergency interventions as well as planned procedures. We perform the largest evaluation of a 2D/3D registration algorithm on real X-ray data to date and find that xvr robustly generalizes across a diverse dataset comprising multiple anatomical structures, imaging modalities, and hospitals. Across surgical tasks, xvr achieves submillimeter-accurate registration at intraoperative speeds, improving upon existing methods by an order of magnitude. xvr is released as open-source software freely available at https://github.com/eigenvivek/xvr.
Collapse
Affiliation(s)
- Vivek Gopalakrishnan
- Harvard-MIT Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Radiology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Neel Dey
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Andrew Abumoussa
- Department of Neurosurgery, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Anna M. Larson
- Department of Interventional Neuroradiology, Boston Children’s Hospital, Boston, MA, USA
| | - Darren B. Orbach
- Department of Interventional Neuroradiology, Boston Children’s Hospital, Boston, MA, USA
| | - Sarah Frisken
- Department of Radiology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Polina Golland
- Harvard-MIT Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
20
|
Pelcat A, Le Berre A, Ben Hassen W, Debacker C, Charron S, Thirion B, Legrand L, Turc G, Oppenheim C, Benzakoun J. Generative T2*-weighted images as a substitute for true T2*-weighted images on brain MRI in patients with acute stroke. Diagn Interv Imaging 2025:S2211-5684(25)00048-8. [PMID: 40113490 DOI: 10.1016/j.diii.2025.03.004] [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: 12/10/2024] [Revised: 03/11/2025] [Accepted: 03/14/2025] [Indexed: 03/22/2025]
Abstract
PURPOSE The purpose of this study was to validate a deep learning algorithm that generates T2*-weighted images from diffusion-weighted (DW) images and to compare its performance with that of true T2*-weighted images for hemorrhage detection on MRI in patients with acute stroke. MATERIALS AND METHODS This single-center, retrospective study included DW- and T2*-weighted images obtained less than 48 hours after symptom onset in consecutive patients admitted for acute stroke. Datasets were divided into training (60 %), validation (20 %), and test (20 %) sets, with stratification by stroke type (hemorrhagic/ischemic). A generative adversarial network was trained to produce generative T2*-weighted images using DW images. Concordance between true T2*-weighted images and generative T2*-weighted images for hemorrhage detection was independently graded by two readers into three categories (parenchymal hematoma, hemorrhagic infarct or no hemorrhage), and discordances were resolved by consensus reading. Sensitivity, specificity and accuracy of generative T2*-weighted images were estimated using true T2*-weighted images as the standard of reference. RESULTS A total of 1491 MRI sets from 939 patients (487 women, 452 men) with a median age of 71 years (first quartile, 57; third quartile, 81; range: 21-101) were included. In the test set (n = 300), there were no differences between true T2*-weighted images and generative T2*-weighted images for intraobserver reproducibility (κ = 0.97 [95 % CI: 0.95-0.99] vs. 0.95 [95 % CI: 0.92-0.97]; P = 0.27) and interobserver reproducibility (κ = 0.93 [95 % CI: 0.90-0.97] vs. 0.92 [95 % CI: 0.88-0.96]; P = 0.64). After consensus reading, concordance between true T2*-weighted images and generative T2*-weighted images was excellent (κ = 0.92; 95 % CI: 0.91-0.96). Generative T2*-weighted images achieved 90 % sensitivity (73/81; 95 % CI: 81-96), 97 % specificity (213/219; 95 % CI: 94-99) and 95 % accuracy (286/300; 95 % CI: 92-97) for the diagnosis of any cerebral hemorrhage (hemorrhagic infarct or parenchymal hemorrhage). CONCLUSION Generative T2*-weighted images and true T2*-weighted images have non-different diagnostic performances for hemorrhage detection in patients with acute stroke and may be used to shorten MRI protocols.
Collapse
Affiliation(s)
- Antoine Pelcat
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, IMA-BRAIN, 75014 Paris, France
| | - Alice Le Berre
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, IMA-BRAIN, 75014 Paris, France; GHU Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, Department of Neuroradiology, 75014 Paris, France
| | - Wagih Ben Hassen
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, IMA-BRAIN, 75014 Paris, France; GHU Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, Department of Neuroradiology, 75014 Paris, France
| | - Clement Debacker
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, IMA-BRAIN, 75014 Paris, France; GHU Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, Department of Neuroradiology, 75014 Paris, France
| | - Sylvain Charron
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, IMA-BRAIN, 75014 Paris, France
| | - Bertrand Thirion
- INRIA, CEA, Université Paris-Saclay, MIND Team, 91400 Palaiseau, France
| | - Laurence Legrand
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, IMA-BRAIN, 75014 Paris, France; GHU Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, Department of Neuroradiology, 75014 Paris, France
| | - Guillaume Turc
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Stroke Team, 75014 Paris, France; GHU Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, Department of Neurology, 75014 Paris, France
| | - Catherine Oppenheim
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, IMA-BRAIN, 75014 Paris, France; GHU Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, Department of Neuroradiology, 75014 Paris, France
| | - Joseph Benzakoun
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, IMA-BRAIN, 75014 Paris, France; GHU Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, Department of Neuroradiology, 75014 Paris, France.
| |
Collapse
|
21
|
MacKinnon MJ, Song S, Chao THH, Hsu LM, Albert ST, Ma Y, Shnitko TA, Wang TWW, Nonneman RJ, Freeman CD, Ozarkar SS, Emir UE, Shen MD, Philpot BD, Hantman AW, Lee SH, Chang WT, Shih YYI. SORDINO for Silent, Sensitive, Specific, and Artifact-Resisting fMRI in awake behaving mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.10.642406. [PMID: 40161795 PMCID: PMC11952411 DOI: 10.1101/2025.03.10.642406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Blood-oxygenation-level-dependent (BOLD) functional magnetic resonance imaging (fMRI) has revolutionized our understanding of the brain activity landscape, bridging circuit neuroscience in animal models with noninvasive brain mapping in humans. This immensely utilized technique, however, faces challenges such as acoustic noise, electromagnetic interference, motion artifacts, magnetic-field inhomogeneity, and limitations in sensitivity and specificity. Here, we introduce Steady-state On-the-Ramp Detection of INduction-decay with Oversampling (SORDINO), a transformative fMRI technique that addresses these challenges by maintaining a constant total gradient amplitude while acquiring data during continuously changing gradient direction. When benchmarked against conventional fMRI on a 9.4T system, SORDINO is silent, sensitive, specific, and resistant to motion and susceptibility artifacts. SORDINO offers superior compatibility with multimodal experiments and carries novel contrast mechanisms distinct from BOLD. It also enables brain-wide activity and connectivity mapping in awake, behaving mice, overcoming stress- and motion-related confounds that are among the most challenging barriers in current animal fMRI studies.
Collapse
Affiliation(s)
- Martin J. MacKinnon
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sheng Song
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Tzu-Hao Harry Chao
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Li-Ming Hsu
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Scott T. Albert
- Neuroscience Center and Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yuncong Ma
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Tatiana A. Shnitko
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Tzu-Wen Winnie Wang
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Randy J. Nonneman
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Corey D. Freeman
- Neuroscience Center and Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Siddhi S. Ozarkar
- Neuroscience Center and Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Uzay E. Emir
- Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mark D. Shen
- Neuroscience Center and Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Benjamin D. Philpot
- Neuroscience Center and Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Adam W. Hantman
- Neuroscience Center and Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sung-Ho Lee
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Wei-Tang Chang
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yen-Yu Ian Shih
- Center for Animal MRI, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Biomedical Research Imaging Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
22
|
Zheng X, Li L, Gao JM, Hu Y, Deng L, Kang YF, Zhang Y. Radiation-induced white matter dysfunction in patients with nasopharyngeal carcinoma. Front Neurosci 2025; 19:1548744. [PMID: 40129723 PMCID: PMC11931022 DOI: 10.3389/fnins.2025.1548744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 02/26/2025] [Indexed: 03/26/2025] Open
Abstract
Radiation-induced structural abnormalities in white matter (WM) have been reported in patients with nasopharyngeal carcinoma (NPC); however, the alterations in functional domain were insufficiently investigated. A total of 111 NPC patients were included and these patients, based on whether completed radiation therapy (RT) or not, were divided into pre-RT (n = 47) and post-RT (n = 64) groups. Functional connectivity strength (FCS) between WM regions (WW-FCS) and between WM and gray matter (GM) regions (GW-FCS) was used to investigate the radiation-induced changes in WM function. Compared with the pre-RT patients, post-RT NPC patients showed decreased WW-FCS in the left superior cerebellar peduncle, right anterior limb of internal capsule, bilateral posterior thalamic radiation, and left tapetum. Compared with the pre-RT patients, post-RT NPC patients showed decreased GW-FCS in the left caudate, bilateral visual cortex, and the right ventral prefrontal cortex. In the post-RT group, the GW-FCS in left visual cortex was negatively correlated with radiation dosage for the brain stem (r = -0.35, p = 0.039), and for the left temporal lobe (r = -0.46, p = 0.0058). The GW-FCS in right visual cortex was negatively correlated with radiation dosage for the left temporal lobe (r = -0.38, p = 0.025). Our findings of decreased WW-FCS and GW-FCS in such brain regions (such as visual cortex, posterior thalamic radiation, and anterior limb of internal capsule, as well as superior cerebellar peduncle) suggest potential functional impairments in visual and motor systems.
Collapse
Affiliation(s)
- Xingyou Zheng
- Department of Medical Imaging, The Fourth Hospital of Changsha (Integrated Traditional Chinese and Western Medicine Hospital of Changsha, Changsha Hospital of Hunan Normal University), Changsha, Hunan, China
| | - Li Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jian-ming Gao
- State Key Laboratory of Oncology in South China, Department of Radiation Oncology, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yang Hu
- Independent Researcher, Shanghai, China
| | - Limeng Deng
- Department of Medical Imaging, The Fourth Hospital of Changsha (Integrated Traditional Chinese and Western Medicine Hospital of Changsha, Changsha Hospital of Hunan Normal University), Changsha, Hunan, China
| | - Ya-fei Kang
- School of Information, Xi’an University of Finance and Economics, Xi’an, Shaanxi, China
| | - Youming Zhang
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
23
|
Pagani M, Zerbi V, Gini S, Alvino F, Banerjee A, Barberis A, Basson MA, Bozzi Y, Galbusera A, Ellegood J, Fagiolini M, Lerch J, Matteoli M, Montani C, Pozzi D, Provenzano G, Scattoni ML, Wenderoth N, Xu T, Lombardo M, Milham MP, Martino AD, Gozzi A. Biological subtyping of autism via cross-species fMRI. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.04.641400. [PMID: 40093106 PMCID: PMC11908180 DOI: 10.1101/2025.03.04.641400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
It is frequently assumed that the phenotypic heterogeneity in autism spectrum disorder reflects underlying pathobiological variation. However, direct evidence in support of this hypothesis is lacking. Here, we leverage cross-species functional neuroimaging to examine whether variability in brain functional connectivity reflects distinct biological mechanisms. We find that fMRI connectivity alterations in 20 distinct mouse models of autism (n=549 individual mice) can be clustered into two prominent hypo- and hyperconnectivity subtypes. We show that these connectivity profiles are linked to distinct signaling pathways, with hypoconnectivity being associated with synaptic dysfunction, and hyperconnectivity reflecting transcriptional and immune-related alterations. Extending these findings to humans, we identify analogous hypo- and hyperconnectivity subtypes in a large, multicenter resting state fMRI dataset of n=940 autistic and n=1036 neurotypical individuals. Remarkably, hypo- and hyperconnectivity autism subtypes are replicable across independent cohorts (accounting for 25.1% of all autism data), exhibit distinct functional network architecture, are behaviorally dissociable, and recapitulate synaptic and immune mechanisms identified in corresponding mouse subtypes. Our cross-species investigation, thus, decodes the heterogeneity of fMRI connectivity in autism into distinct pathway-specific etiologies, offering a new empirical framework for targeted subtyping of autism.
Collapse
Affiliation(s)
- Marco Pagani
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, CNCS@UNITN, Rovereto, Italy
- Autism Center, Child Mind Institute, New York, NY, USA
- IMT School for Advanced Studies, Lucca, Italy
| | - Valerio Zerbi
- Department of Psychiatry, University of Geneva, Switzerland
- Department of Basic Neurosciences, University of Geneva, Switzerland
| | - Silvia Gini
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, CNCS@UNITN, Rovereto, Italy
- Center for Mind and Brain Sciences (CIMeC), University of Trento, Rovereto, Italy
| | - Filomena Alvino
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, CNCS@UNITN, Rovereto, Italy
| | | | - Andrea Barberis
- Synaptic Plasticity of Inhibitory Networks, Istituto Italiano di Tecnologia, Genova, Italy
| | - M. Albert Basson
- Centre for Craniofacial and Regenerative Biology, King’s College London, London, UK
- Department of Clinical and Biomedical Sciences, University of Exeter, Exeter, UK
| | - Yuri Bozzi
- Center for Mind and Brain Sciences (CIMeC), University of Trento, Rovereto, Italy
| | - Alberto Galbusera
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, CNCS@UNITN, Rovereto, Italy
| | - Jacob Ellegood
- Bloorview Research Institute, Holland Bloorview Kids Rehabilitation Hospital, Toronto, ON, Canada
| | | | - Jason Lerch
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Michela Matteoli
- Humanitas University, Milan, Italy
- CNR Institute of Neuroscience c/o Humanitas Clinical and Research Center - IRCCS, Rozzano, Milan, Italy
| | - Caterina Montani
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, CNCS@UNITN, Rovereto, Italy
| | - Davide Pozzi
- CNR Institute of Neuroscience c/o Humanitas Clinical and Research Center - IRCCS, Rozzano, Milan, Italy
| | - Giovanni Provenzano
- Department of Cellular, Computational and Integrative Biology. University of Trento, Trento, Italy
| | - Maria Luisa Scattoni
- Research Coordination and Support Service, Istituto Superiore di Sanità, Rome, Italy
| | | | - Ting Xu
- Center for Integrative Developing Brain, Child Mind Institute, New York, NY, USA
| | - Michael Lombardo
- Laboratory for Autism and Neurodevelopmental Disorders, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, Rovereto, Italy
| | - Michael P Milham
- Center for the Integrative Developmental Neuroscience, Child Mind Institute, New York, NY, USA
| | | | - Alessandro Gozzi
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, CNCS@UNITN, Rovereto, Italy
| |
Collapse
|
24
|
Shafiei G, Esper NB, Hoffmann MS, Ai L, Chen AA, Cluce J, Covitz S, Giavasis S, Lane C, Mehta K, Moore TM, Salo T, Tapera TM, Calkins ME, Colcombe S, Davatzikos C, Gur RE, Gur RC, Pan PM, Jackowski AP, Rokem A, Rohde LA, Shinohara RT, Tottenham N, Zuo XN, Cieslak M, Franco AR, Kiar G, Salum GA, Milham MP, Satterthwaite TD. Reproducible Brain Charts: An open data resource for mapping brain development and its associations with mental health. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.24.639850. [PMID: 40060681 PMCID: PMC11888297 DOI: 10.1101/2025.02.24.639850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/18/2025]
Abstract
Major mental disorders are increasingly understood as disorders of brain development. Large and heterogeneous samples are required to define generalizable links between brain development and psychopathology. To this end, we introduce the Reproducible Brain Charts (RBC), an open data resource that integrates data from 5 large studies of brain development in youth from three continents (N=6,346; 45% Female). Confirmatory bifactor models were used to create harmonized psychiatric phenotypes that capture major dimensions of psychopathology. Following rigorous quality assurance, neuroimaging data were carefully curated and processed using consistent pipelines in a reproducible manner with DataLad, the Configurable Pipeline for the Analysis of Connectomes (C-PAC), and FreeSurfer. Initial analyses of RBC data emphasize the benefit of careful quality assurance and data harmonization in delineating developmental effects and associations with psychopathology. Critically, all RBC data - including harmonized psychiatric phenotypes, unprocessed images, and fully processed imaging derivatives - are openly shared without a data use agreement via the International Neuroimaging Data-sharing Initiative. Together, RBC facilitates large-scale, reproducible, and generalizable research in developmental and psychiatric neuroscience.
Collapse
Affiliation(s)
- G Shafiei
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Lifespan Brain Institute (LiBI) of Penn Medicine and CHOP, University of Pennsylvania, Philadelphia, PA, USA
| | - N B Esper
- Child Mind Institute, New York, NY, USA
| | - M S Hoffmann
- Department of Neuropsychiatry, Universidade Federal de Santa Maria (UFSM), Santa Maria, Brazil
- Graduate Program in Psychiatry and Behavioral Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- National Institute of Developmental Psychiatry & National Center for Innovation and Research in Mental Health, Brazil
- Care Policy and Evaluation Centre, London School of Economics and Political Science, London, UK
| | - L Ai
- Child Mind Institute, New York, NY, USA
| | - A A Chen
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - J Cluce
- Child Mind Institute, New York, NY, USA
| | - S Covitz
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | | | - C Lane
- Child Mind Institute, New York, NY, USA
| | - K Mehta
- Department of Neuroscience, Columbia University, New York, NY, USA
| | - T M Moore
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Lifespan Brain Institute (LiBI) of Penn Medicine and CHOP, University of Pennsylvania, Philadelphia, PA, USA
| | - T Salo
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Lifespan Brain Institute (LiBI) of Penn Medicine and CHOP, University of Pennsylvania, Philadelphia, PA, USA
| | - T M Tapera
- Khoury College of Computer Sciences, Northeastern University, Boston, MA, USA
| | - M E Calkins
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Lifespan Brain Institute (LiBI) of Penn Medicine and CHOP, University of Pennsylvania, Philadelphia, PA, USA
| | - S Colcombe
- Center for Biomedical Imaging and Neuromodulation, Nathan Kline Institute, Orangeburg, NY, USA
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY, USA
| | - C Davatzikos
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - R E Gur
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Lifespan Brain Institute (LiBI) of Penn Medicine and CHOP, University of Pennsylvania, Philadelphia, PA, USA
| | - R C Gur
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Lifespan Brain Institute (LiBI) of Penn Medicine and CHOP, University of Pennsylvania, Philadelphia, PA, USA
| | - P M Pan
- Department of Psychiatry, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - A P Jackowski
- Department of Psychiatry, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
| | - A Rokem
- Department of Psychology, University of Washington, Seattle, WA, USA
- eScience Institute, University of Washington, Seattle, WA
| | - L A Rohde
- Graduate Program in Psychiatry and Behavioral Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - R T Shinohara
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - N Tottenham
- Department of Psychology, Columbia University, New York, NY, USA
| | - X N Zuo
- Developmental Population Neuroscience Research Center, IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - M Cieslak
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Lifespan Brain Institute (LiBI) of Penn Medicine and CHOP, University of Pennsylvania, Philadelphia, PA, USA
| | - A R Franco
- Child Mind Institute, New York, NY, USA
- Center for Biomedical Imaging and Neuromodulation, Nathan Kline Institute, Orangeburg, NY, USA
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY, USA
| | - G Kiar
- Child Mind Institute, New York, NY, USA
| | - G A Salum
- Child Mind Institute, New York, NY, USA
- Graduate Program in Psychiatry and Behavioral Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- National Institute of Developmental Psychiatry & National Center for Innovation and Research in Mental Health, Brazil
- ADHD Outpatient Program & Developmental Psychiatry Program, Hospital de Clinicas de Porto Alegre, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Medical Council UNIFAJ & UNIMAX, Brazil
| | - M P Milham
- Child Mind Institute, New York, NY, USA
- Center for Biomedical Imaging and Neuromodulation, Nathan Kline Institute, Orangeburg, NY, USA
| | - T D Satterthwaite
- Penn Lifespan Informatics and Neuroimaging Center, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Lifespan Brain Institute (LiBI) of Penn Medicine and CHOP, University of Pennsylvania, Philadelphia, PA, USA
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
25
|
Ottesen JA, Storas T, Vatnehol SAS, Løvland G, Vik-Mo EO, Schellhorn T, Skogen K, Larsson C, Bjørnerud A, Groote-Eindbaas IR, Caan MWA. Deep learning-based Intraoperative MRI reconstruction. Eur Radiol Exp 2025; 9:29. [PMID: 39998750 PMCID: PMC11861787 DOI: 10.1186/s41747-024-00548-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 12/27/2024] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND We retrospectively evaluated the quality of deep learning (DL) reconstructions of on-scanner accelerated intraoperative MRI (iMRI) during respective brain tumor surgery. METHODS Accelerated iMRI was performed using dual surface coils positioned around the area of resection. A DL model was trained on the fastMRI neuro dataset to mimic the data from the iMRI protocol. The evaluation was performed on imaging material from 40 patients imaged from Nov 1, 2021, to June 1, 2023, who underwent iMRI during tumor resection surgery. A comparative analysis was conducted between the conventional compressed sense (CS) method and the trained DL reconstruction method. Blinded evaluation of multiple image quality metrics was performed by two neuroradiologists and one neurosurgeon using a 1-to-5 Likert scale (1, nondiagnostic; 2, poor; 3, acceptable; 4, good; and 5, excellent), and the favored reconstruction variant. RESULTS The DL reconstruction was strongly favored or favored over the CS reconstruction for 33/40, 39/40, and 8/40 of cases for readers 1, 2, and 3, respectively. For the evaluation metrics, the DL reconstructions had a higher score than their respective CS counterparts for 72%, 72%, and 14% of the cases for readers 1, 2, and 3, respectively. Still, the DL reconstructions exhibited shortcomings such as a striping artifact and reduced signal. CONCLUSION DL shows promise in allowing for high-quality reconstructions of iMRI. The neuroradiologists noted an improvement in the perceived spatial resolution, signal-to-noise ratio, diagnostic confidence, diagnostic conspicuity, and spatial resolution compared to CS, while the neurosurgeon preferred the CS reconstructions across all metrics. RELEVANCE STATEMENT DL shows promise to allow for high-quality reconstructions of iMRI, however, due to the challenging setting of iMRI, further optimization is needed. KEY POINTS iMRI is a surgical tool with a challenging image setting. DL allowed for high-quality reconstructions of iMRI. Additional optimization is needed due to the challenging intraoperative setting.
Collapse
Affiliation(s)
- Jon André Ottesen
- Computational Radiology & Artificial Intelligence (CRAI) Research Group, Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway.
- Department of Physics, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway.
- Division of Radiology and Nuclear Medicine, Department of Physics and Computational Radiology, Oslo University Hospital, Oslo, Norway.
| | - Tryggve Storas
- Division of Radiology and Nuclear Medicine, Department of Physics and Computational Radiology, Oslo University Hospital, Oslo, Norway
| | - Svein Are Sirirud Vatnehol
- The Intervention Centre, Oslo University Hospital, Oslo, Norway
- Department of Optometry, Radiography and Lighting Design, University of South-Eastern Norway, Drammen, Norway
- Department of Health Sciences Gjøvik, Faculty of Medicine and Health Sciences, NTNU, Gjøvik, Norway
| | - Grethe Løvland
- The Intervention Centre, Oslo University Hospital, Oslo, Norway
| | - Einar Osland Vik-Mo
- Vilhelm Magnus Laboratory, Department of Neurosurgery, Oslo University Hospital, Oslo, Norway
- Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Till Schellhorn
- Computational Radiology & Artificial Intelligence (CRAI) Research Group, Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Karoline Skogen
- Computational Radiology & Artificial Intelligence (CRAI) Research Group, Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Christopher Larsson
- Computational Radiology & Artificial Intelligence (CRAI) Research Group, Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
- Department of Neurosurgery, Oslo University Hospital, Oslo, Norway
| | - Atle Bjørnerud
- Computational Radiology & Artificial Intelligence (CRAI) Research Group, Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
- Department of Physics, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
- Division of Radiology and Nuclear Medicine, Department of Physics and Computational Radiology, Oslo University Hospital, Oslo, Norway
| | - Inge Rasmus Groote-Eindbaas
- Computational Radiology & Artificial Intelligence (CRAI) Research Group, Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
- Department of Radiology, Vestfold Hospital Trust, Tønsberg, Norway
| | - Matthan W A Caan
- Computational Radiology & Artificial Intelligence (CRAI) Research Group, Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
- Biomedical Engineering and Physics, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
26
|
Khoury N, Pizzo ME, Discenza CB, Joy D, Tatarakis D, Todorov MI, Negwer M, Ha C, De Melo GL, Sarrafha L, Simon MJ, Chan D, Chau R, Chew KS, Chow J, Clemens A, Robles-Colmenares Y, Dugas JC, Duque J, Kaltenecker D, Kane H, Leung A, Lozano E, Moshkforoush A, Roche E, Sandmann T, Tong M, Xa K, Zhou Y, Lewcock JW, Ertürk A, Thorne RG, Calvert MEK, Yu Zuchero YJ. Fc-engineered large molecules targeting blood-brain barrier transferrin receptor and CD98hc have distinct central nervous system and peripheral biodistribution. Nat Commun 2025; 16:1822. [PMID: 39979268 PMCID: PMC11842567 DOI: 10.1038/s41467-025-57108-x] [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: 07/10/2024] [Accepted: 02/10/2025] [Indexed: 02/22/2025] Open
Abstract
Blood brain barrier-crossing molecules targeting transferrin receptor (TfR) and CD98 heavy chain (CD98hc) are widely reported to promote enhanced brain delivery of therapeutics. Here, we provide a comprehensive and unbiased biodistribution characterization of TfR and CD98hc antibody transport vehicles (ATVTfR and ATVCD98hc) compared to control IgG. Mouse whole-body tissue clearing reveals distinct organ localization for each molecule. In the brain, ATVTfR and ATVCD98hc achieve enhanced exposure and parenchymal distribution even when brain exposures are matched between ATV and control IgG in bulk tissue. Using a combination of cell sorting and single-cell RNAseq, we reveal that control IgG is nearly absent from parenchymal cells and is distributed primarily to brain perivascular and leptomeningeal cells. In contrast, ATVTfR and ATVCD98hc exhibit broad and unique parenchymal cell-type distribution. Finally, we profile in detail brain region-specific biodistribution of ATVTfR in cynomolgus monkey brain and spinal cord. Taken together, this in-depth multiscale characterization will guide platform selection for therapeutic targets of interest.
Collapse
Affiliation(s)
- Nathalie Khoury
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Michelle E Pizzo
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Claire B Discenza
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - David Joy
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - David Tatarakis
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | | | | | - Connie Ha
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Gabrielly L De Melo
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Lily Sarrafha
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Matthew J Simon
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Darren Chan
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Roni Chau
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Kylie S Chew
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Johann Chow
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Allisa Clemens
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | | | - Jason C Dugas
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Joseph Duque
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | | | - Holly Kane
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Amy Leung
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Edwin Lozano
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Arash Moshkforoush
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Elysia Roche
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Thomas Sandmann
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Mabel Tong
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Kaitlin Xa
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Yinhan Zhou
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Joseph W Lewcock
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | | | - Robert G Thorne
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA
| | - Meredith E K Calvert
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA.
| | - Y Joy Yu Zuchero
- Denali Therapeutics, Inc., 161 Oyster Point Blvd., South San Francisco, CA, 94080, USA.
| |
Collapse
|
27
|
Haubold J, Pollok OB, Holtkamp M, Salhöfer L, Schmidt CS, Bojahr C, Straus J, Schaarschmidt BM, Borys K, Kohnke J, Wen Y, Opitz M, Umutlu L, Forsting M, Friedrich CM, Nensa F, Hosch R. Moving Beyond CT Body Composition Analysis: Using Style Transfer for Bringing CT-Based Fully-Automated Body Composition Analysis to T2-Weighted MRI Sequences. Invest Radiol 2025:00004424-990000000-00294. [PMID: 39961134 DOI: 10.1097/rli.0000000000001162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2025]
Abstract
OBJECTIVES Deep learning for body composition analysis (BCA) is gaining traction in clinical research, offering rapid and automated ways to measure body features like muscle or fat volume. However, most current methods prioritize computed tomography (CT) over magnetic resonance imaging (MRI). This study presents a deep learning approach for automatic BCA using MR T2-weighted sequences. METHODS Initial BCA segmentations (10 body regions and 4 body parts) were generated by mapping CT segmentations from body and organ analysis (BOA) model to synthetic MR images created using an in-house trained CycleGAN. In total, 30 synthetic data pairs were used to train an initial nnU-Net V2 in 3D, and this preliminary model was then applied to segment 120 real T2-weighted MRI sequences from 120 patients (46% female) with a median age of 56 (interquartile range, 17.75), generating early segmentation proposals. These proposals were refined by human annotators, and nnU-Net V2 2D and 3D models were trained using 5-fold cross-validation on this optimized dataset of real MR images. Performance was evaluated using Sørensen-Dice, Surface Dice, and Hausdorff Distance metrics including 95% confidence intervals for cross-validation and ensemble models. RESULTS The 3D ensemble segmentation model achieved the highest Dice scores for the body region classes: bone 0.926 (95% confidence interval [CI], 0.914-0.937), muscle 0.968 (95% CI, 0.961-0.975), subcutaneous fat 0.98 (95% CI, 0.971-0.986), nervous system 0.973 (95% CI, 0.965-0.98), thoracic cavity 0.978 (95% CI, 0.969-0.984), abdominal cavity 0.989 (95% CI, 0.986-0.991), mediastinum 0.92 (95% CI, 0.901-0.936), pericardium 0.945 (95% CI, 0.924-0.96), brain 0.966 (95% CI, 0.927-0.989), and glands 0.905 (95% CI, 0.886-0.921). Furthermore, body part 2D ensemble model reached the highest Dice scores for all labels: arms 0.952 (95% CI, 0.937-0.965), head + neck 0.965 (95% CI, 0.953-0.976), legs 0.978 (95% CI, 0.968-0.988), and torso 0.99 (95% CI, 0.988-0.991). The overall average Dice across body parts (2D = 0.971, 3D = 0.969, P = ns) and body regions (2D = 0.935, 3D = 0.955, P < 0.001) ensemble models indicates stable performance across all classes. CONCLUSIONS The presented approach facilitates efficient and automated extraction of BCA parameters from T2-weighted MRI sequences, providing precise and detailed body composition information across various regions and body parts.
Collapse
Affiliation(s)
- Johannes Haubold
- From the Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany (J.H., O.B.P., M.H., L.S., C.B., J.S., B.M.S., K.B., J.K., M.O., L.U., M.F., F.N., R.H.); Institute for Artificial Intelligence in Medicine, University Hospital Essen, Essen, Germany (J.H., O.B.P., M.H., L.S., C.S.S., C.B., J.S., K.B., J.K., Y.W., M.O., L.U., M.F., F.N., R.H.); Institute for Transfusion Medicine, University Hospital Essen, Essen, Germany (C.S.S.); Center of Sleep and Telemedicine, University Hospital Essen-Ruhrlandklinik, Essen, Germany (C.S.S.); Data Integration Center, Central IT Department, University Hospital Essen, Essen, Germany (Y.W.); Department of Computer Science, University of Applied Sciences and Arts Dortmund (FHDO), Dortmund, Germany (C.M.F.); and Institute for Medical Informatics, Biometry, and Epidemiology (IMIBE), University Hospital Essen, Essen, Germany (C.M.F.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Gan Y, Lan Q, Huang C, Su W, Huang Z. Dense convolution-based attention network for Alzheimer's disease classification. Sci Rep 2025; 15:5693. [PMID: 39962113 PMCID: PMC11832751 DOI: 10.1038/s41598-025-85802-9] [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: 11/09/2024] [Accepted: 01/06/2025] [Indexed: 02/20/2025] Open
Abstract
Recently, deep learning-based medical image classification models have made substantial advancements. However, many existing models prioritize performance at the cost of efficiency, limiting their practicality in clinical use. Traditional Convolutional Neural Network (CNN)-based methods, Transformer-based methods, and hybrid approaches combining these two struggle to balance performance and model complexity. To achieve efficient predictions with a low parameter count, we propose DenseAttentionNetwork (DANet), a lightweight model for Alzheimer's disease detection in 3D MRI images. DANet leverages dense connections and a linear attention mechanism to enhance feature extraction and capture long-range dependencies. Its architecture integrates convolutional layers for localized feature extraction with linear attention for global context, enabling efficient multi-scale feature reuse across the network. By replacing traditional self-attention with a parameter-efficient linear attention mechanism, DANet overcomes some limitations of standard self-attention. Extensive experiments across multi-institutional datasets demonstrate that DANet achieves the best performance in area under the receiver operating characteristic curve (AUC), which underscores the model's robustness and effectiveness in capturing relevant features for Alzheimer's disease detection while also attaining a strong accuracy structure with fewer parameters. Visualizations based on activation maps further verify the model's ability to highlight AD-relevant regions in 3D MRI images, providing clinically interpretable insights into disease progression.
Collapse
Affiliation(s)
- Yingtong Gan
- Key Laboratory of Multimedia Trusted Perception and Efficient Computing, Ministry of Education of China, Xiamen University, Xiamen, 361005, People's Republic of China
- Institute of Artifical Intelligence, Xiamen University, Xiamen, 361005, People's Republic of China
| | - Quan Lan
- Department of Neurology, First Affiliated Hospital of Xiamen University, Xiamen, 361003, People's Republic of China
| | - ChenXi Huang
- Key Laboratory of Multimedia Trusted Perception and Efficient Computing, Ministry of Education of China, Xiamen University, Xiamen, 361005, People's Republic of China.
| | - Weichao Su
- Xiamen Xianyue Hospital, Xianyue Hospital Affiliated with Xiamen Medical College, Fujian Psychiatric Center, Fujian Clinical Research Center for Mental Disorders, Xiamen, 361012, People's Republic of China.
| | - Zhiyuan Huang
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, 350122, People's Republic of China.
- Xiamen Xianyue Hospital, Xianyue Hospital Affiliated with Xiamen Medical College, Fujian Psychiatric Center, Fujian Clinical Research Center for Mental Disorders, Xiamen, 361012, People's Republic of China.
| |
Collapse
|
29
|
Silberfeld A, Roe JM, Ellegood J, Lerch JP, Qiu L, Kim Y, Lee JG, Hopkins WD, Grandjean J, Ou Y, Pourquié O. Left-Right Brain-Wide Asymmetry of Neuroanatomy in the Mouse Brain. Neuroimage 2025; 307:121017. [PMID: 39798830 DOI: 10.1016/j.neuroimage.2025.121017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/16/2024] [Accepted: 01/09/2025] [Indexed: 01/15/2025] Open
Abstract
Left-right asymmetry of the human brain is widespread through its anatomy and function. However, limited microscopic understanding of it exists, particularly for anatomical asymmetry where there are few well-established animal models. In humans, most brain regions show subtle, population-average regional asymmetries in thickness or surface area, alongside a macro-scale twisting called the cerebral petalia in which the right hemisphere protrudes past the left. Here, we ask whether neuroanatomical asymmetries can be observed in mice, leveraging 6 mouse neuroimaging cohorts from 5 different research groups (∼3,500 animals). We found an anterior-posterior pattern of volume asymmetry with anterior regions larger on the right and posterior regions larger on the left. This pattern appears driven by similar trends in surface area and positional asymmetries, with the results together indicating a small brain-wide twisting pattern, similar to the human cerebral petalia. Furthermore, the results show no apparent relationship to known functional asymmetries in mice, emphasizing the complexity of the structure-function relationship in brain asymmetry. Our results recapitulate and extend previous patterns of asymmetry from two published studies as well as capture well-established, bilateral male-female differences in the mouse brain as a positive control. By establishing a signature of anatomical brain asymmetry in mice, we aim to provide a foundation for future studies to probe the mechanistic underpinnings of brain asymmetry seen in humans - a feature of the brain with extremely limited understanding.
Collapse
Affiliation(s)
- Andrew Silberfeld
- Department of Genetics, Harvard Medical School, Boston, MA, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - James M Roe
- Center for Lifespan Changes in Brain and Cognition (LCBC), Department of Psychology, University of Oslo, Norway
| | - Jacob Ellegood
- Bloorview Research Institute, Holland Bloorview Kids Rehabilitation Hospital, Toronto, Ontario, Canada; Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jason P Lerch
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada; Department of Medical Biophysics, The University of Toronto, Toronto, ON, Canada; Department of Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada; Department of Preclinical Imaging, Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, UK
| | - Lily Qiu
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada; Department of Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
| | - Yongsoo Kim
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| | - Jong Gwan Lee
- Department of Genetics, Harvard Medical School, Boston, MA, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - William D Hopkins
- Department of Comparative Medicine & Michale E Keeling Center for Comparative Medicine and Research, The University of Texas MD Anderson Cancer Center, United States
| | - Joanes Grandjean
- Donders Institute for Brain, Behaviour, and Cognition, Nijmegen, Netherlands; Department for Medical Imaging, Radboud University Medical Center, PO Box 9101, Nijmegen, Netherlands
| | - Yangming Ou
- Boston Children's Hospital, Harvard Medical School, Harvard University, Boston, MA, USA
| | - Olivier Pourquié
- Department of Genetics, Harvard Medical School, Boston, MA, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
30
|
Oshima S, Kim A, Sun XR, Rifi Z, Cross KA, Fu KA, Salamon N, Ellingson BM, Bari AA, Yao J. Predicting Post-Operative Side Effects in VIM MRgFUS Based on THalamus Optimized Multi Atlas Segmentation (THOMAS) on White-Matter-Nulled MRI: A Retrospective Study. AJNR Am J Neuroradiol 2025; 46:330-340. [PMID: 39730158 PMCID: PMC11878955 DOI: 10.3174/ajnr.a8448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 08/01/2024] [Indexed: 12/29/2024]
Abstract
BACKGROUND AND PURPOSE Precise and individualized targeting of the ventral intermediate thalamic nucleus for the MR-guided focused ultrasound is crucial for enhancing treatment efficacy and avoiding undesirable side effects. In this study, we tested the hypothesis that the spatial relationships between Thalamus Optimized Multi Atlas Segmentation derived segmentations and the post-focused ultrasound lesion can predict post-operative side effects in patients treated with MR-guided focused ultrasound. MATERIALS AND METHODS We retrospectively analyzed 30 patients (essential tremor, n = 26; tremor-dominant Parkinson's disease, n = 4) who underwent unilateral ventral intermediate thalamic nucleus focused ultrasound treatment. We created ROIs of coordinate-based indirect treatment target, focused ultrasound-induced lesion, and thalamus and ventral intermediate thalamic nucleus segmentations. We extracted imaging features including 1) focused ultrasound-induced lesion volumes, 2) overlap between lesions and thalamus and ventral intermediate thalamic nucleus segmentations, 3) distance between lesions and ventral intermediate thalamic nucleus segmentation and 4) distance between lesions and the indirect standard target. These imaging features were compared between patients with and without post-operative gait/balance side effects using Wilcoxon rank-sum test. Multivariate prediction models of side effects based on the imaging features were evaluated using the receiver operating characteristic analyses. RESULTS Patients with self-reported gait/balance side effects had a significantly larger extent of focused ultrasound-induced edema, a smaller fraction of the lesion within the ventral intermediate thalamic nucleus segmentation, a larger fraction of the off-target lesion outside the thalamus segmentation, a more inferior centroid of the lesion from the ventral intermediate thalamic nucleus segmentation, and a larger distance between the centroid of the lesion and ventral intermediate thalamic nucleus segmentation (p < 0.05). Similar results were found for exam-based side effects. Multivariate regression models based on the imaging features achieved areas under the curve of 0.99 (95% CI: 0.88 to 1.00) and 0.96 (95% CI: 0.73 to 1.00) for predicting self-reported and exam-based side effects, respectively. CONCLUSIONS Thalamus Optimized Multi Atlas Segmentation-based patient-specific segmentation of the ventral intermediate thalamic nucleus can predict post-operative side effects, which has implications for aiding the direct targeting of MR-guided focused ultrasound and reducing side effects.
Collapse
Affiliation(s)
- Sonoko Oshima
- From the UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers (S.O., A.K., B.M.E., J.Y.), University of California, Los Angeles, Los Angeles, California
- Department of Radiological Sciences (S.O., N.S., B.M.E., J.Y.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Asher Kim
- From the UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers (S.O., A.K., B.M.E., J.Y.), University of California, Los Angeles, Los Angeles, California
- Department of Bioengineering (A.K., B.M.E., J.Y.), Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, Los Angeles, California
| | - Xiaonan R Sun
- Department of Neurosurgery (X.R.S., Z.R., B.M.E., A.A.B.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Ziad Rifi
- Department of Neurosurgery (X.R.S., Z.R., B.M.E., A.A.B.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Katy A Cross
- Department of Neurology (K.A.C., K.A.F.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Katherine A Fu
- Department of Neurology (K.A.C., K.A.F.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Noriko Salamon
- Department of Radiological Sciences (S.O., N.S., B.M.E., J.Y.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Benjamin M Ellingson
- From the UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers (S.O., A.K., B.M.E., J.Y.), University of California, Los Angeles, Los Angeles, California
- Department of Radiological Sciences (S.O., N.S., B.M.E., J.Y.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
- Department of Bioengineering (A.K., B.M.E., J.Y.), Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, Los Angeles, California
- Department of Neurosurgery (X.R.S., Z.R., B.M.E., A.A.B.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
- Department of Psychiatry and Biobehavioral Sciences (B.M.E.), David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA
| | - Ausaf A Bari
- Department of Neurosurgery (X.R.S., Z.R., B.M.E., A.A.B.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Jingwen Yao
- From the UCLA Brain Tumor Imaging Laboratory (BTIL), Center for Computer Vision and Imaging Biomarkers (S.O., A.K., B.M.E., J.Y.), University of California, Los Angeles, Los Angeles, California
- Department of Radiological Sciences (S.O., N.S., B.M.E., J.Y.), David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
- Department of Bioengineering (A.K., B.M.E., J.Y.), Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, Los Angeles, California
| |
Collapse
|
31
|
Völzke Y, Akbey S, Löwen D, Pracht ED, Stirnberg R, Gras V, Boulant N, Zaiss M, Stöcker T. Calibration-free whole-brain CEST imaging at 7T with parallel transmit pulse design for saturation homogeneity utilizing universal pulses (PUSHUP). Magn Reson Med 2025; 93:630-642. [PMID: 39301770 PMCID: PMC11604840 DOI: 10.1002/mrm.30305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/30/2024] [Accepted: 08/30/2024] [Indexed: 09/22/2024]
Abstract
PURPOSE Chemical exchange saturation transfer (CEST) measurements at ultra-high field (UHF) suffer from strong saturation inhomogeneity. Retrospective correction of this inhomogeneity is possible to some extent, but requires a time-consuming repetition of the measurement. Here, we propose a calibration-free parallel transmit (pTx)-based saturation scheme that homogenizes the saturation over the imaging volume, which we call PUlse design for Saturation Homogeneity utilizing Universal Pulses (PUSHUP). THEORY Magnetization transfer effects depend on the saturationB 1 rms $$ {\mathrm{B}}_1^{\mathrm{rms}} $$ . PUSHUP homogenizes the saturationB 1 rms $$ {\mathrm{B}}_1^{\mathrm{rms}} $$ by using multiple saturation pulses with alternatingB 1 $$ {\mathrm{B}}_1 $$ -shims. Using a database ofB 1 $$ {\mathrm{B}}_1 $$ maps, universal pulses are calculated that remove the necessity of time-consuming, subject-based pulse calculation during the measurement. METHODS PUSHUP was combined with a whole-brain three-dimensional-echo planar imaging (3D-EPI) readout. Two PUSHUP saturation modules were calculated by either applying whole-brain or cerebellum masks to the database maps. The saturation homogeneity and the group mean CEST amplitudes were calculated for differentB 1 $$ {\mathrm{B}}_1 $$ -correction methods and were compared to circular polarized (CP) saturation in five healthy volunteers using an eight-channel transmit coil at 7 Tesla. RESULTS In contrast to CP saturation, where accurate CEST maps were impossible to obtain in the cerebellum, even with extensiveB 1 $$ {\mathrm{B}}_1 $$ -correction, PUSHUP CEST maps were artifact-free throughout the whole brain. A 1-point retrospectiveB 1 $$ {\mathrm{B}}_1 $$ -correction, that does not need repeated measurements, sufficiently removed the effect of residual saturation inhomogeneity. CONCLUSION The presented method allows for homogeneous whole-brain CEST imaging at 7 Tesla without the need of a repetition-basedB 1 $$ {\mathrm{B}}_1 $$ -correction or online pulse calculation. With the fast 3D-EPI readout, whole-brain CEST imaging with 45 saturation offsets is possible at 1.6 mm resolution in under 4 min.
Collapse
Affiliation(s)
- Yannik Völzke
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
| | - Suzan Akbey
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
| | - Daniel Löwen
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
| | | | | | - Vincent Gras
- CNRS, NeuroSpin, BaobabUniversité Paris‐Saclay, Commissariat à' l'Energie AtomiqueGif sur YvetteFrance
| | - Nicolas Boulant
- CNRS, NeuroSpin, BaobabUniversité Paris‐Saclay, Commissariat à' l'Energie AtomiqueGif sur YvetteFrance
| | - Moritz Zaiss
- Institute of Neuroradiology, Institute of NeuroradiologyFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)ErlangenGermany
| | - Tony Stöcker
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
- Department of Physics and AstronomyUniversity BonnBonnGermany
| |
Collapse
|
32
|
Boa Sorte Silva NC, Balbim GM, Stein RG, Gu Y, Tam RC, Dao E, Alkeridy W, Lam K, Kramer AF, Liu‐Ambrose T. Physical activity may protect myelin via modulation of high-density lipoprotein. Alzheimers Dement 2025; 21:e14599. [PMID: 39989020 PMCID: PMC11848041 DOI: 10.1002/alz.14599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/24/2024] [Accepted: 01/13/2025] [Indexed: 02/25/2025]
Abstract
INTRODUCTION Physical activity is associated with greater myelin content in older individuals with cerebral small vessel disease (CSVD), a condition marked by demyelination. However, potential mechanisms underlying this relationship remain understudied. METHODS We assessed cross-sectionally whether serum high-density lipoprotein (HDL), low-density lipoprotein, and triglycerides moderated the association between physical activity and in vivo myelin in older individuals with CSVD and mild cognitive impairment. RESULTS We included 81 highly educated, community-dwelling older individuals (mean age 74.57 years), 64% of whom were female. Regression models revealed that HDL levels significantly moderated the relationship between physical activity and myelin in the sagittal stratum, wherein higher physical activity levels were linked to greater myelin levels for those with average or high HDL (standardized B [95% CI] = 0.289 [0.087 to 0.491], p = 0.006). DISCUSSION Physical activity may promote myelin health partly through HDL. Data from longitudinal studies are needed to confirm our findings. HIGHLIGHTS Myelin loss is common in individuals with cerebral small vessel disease (CSVD). Physical activity was positively associated with myelin in older adults with CSVD. High-density lipoproteins (HDL) levels were also positively related to myelin. Physical activity effects on myelin were moderated by HDL levels.
Collapse
Affiliation(s)
- Nárlon C. Boa Sorte Silva
- Department of HealthKinesiology, and Applied PhysiologyFaculty of Arts and ScienceConcordia UniversityMontréalQuébecCanada
| | - Guilherme M. Balbim
- Department of Physical TherapyFaculty of MedicineUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Ryan G. Stein
- Department of Physical TherapyFaculty of MedicineUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Yi Gu
- Department of Physical TherapyFaculty of MedicineUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Roger C. Tam
- School of Biomedical EngineeringFaculty of Applied Science and Faculty of MedicineUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Elizabeth Dao
- Department of Physical TherapyFaculty of MedicineUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Walid Alkeridy
- Department of MedicineKing Saud UniversityCollege of MedicineRiyadhSaudi Arabia
| | - Kevin Lam
- Department of MedicineDivision of NeurologyFaculty of MedicineUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Arthur F. Kramer
- Department of PsychologyNortheastern UniversityBostonMassachusettsUSA
| | - Teresa Liu‐Ambrose
- Department of Physical TherapyFaculty of MedicineUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| |
Collapse
|
33
|
Apte AP, LoCastro E, Iyer A, Elguindi S, Jiang J, Oh JH, Veeraraghavan H, Shukla-Dave A, Deasy JO. Artificial Intelligence Apps for Medical Image Analysis using pyCERR and Cancer Genomics Cloud. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.19.633756. [PMID: 39896472 PMCID: PMC11785098 DOI: 10.1101/2025.01.19.633756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
This work introduces a user-friendly, cloud-based software framework for conducting Artificial Intelligence (AI) analyses of medical images. The framework allows users to deploy AI-based workflows by customizing software and hardware dependencies. The components of our software framework include the Python-native Computational Environment for Radiological Research (pyCERR) platform for radiological image processing, Cancer Genomics Cloud (CGC) for accessing hardware resources and user management utilities for accessing images from data repositories and installing AI models and their dependencies. GNU-GPL copyright pyCERR was ported to Python from MATLAB-based CERR to enable researchers to organize, access, and transform metadata from high dimensional, multi-modal datasets to build cloud-compatible workflows for AI modeling in radiation therapy and medical image analysis. pyCERR provides an extensible data structure to accommodate metadata from commonly used medical imaging file formats and a viewer to allow for multi-modal visualization. Analysis modules are provided to facilitate cloud-compatible AI-based workflows for image segmentation, radiomics, DCE MRI analysis, radiotherapy dose-volume histogram-based features, and normal tissue complication and tumor control models for radiotherapy. Image processing utilities are provided to help train and infer convolutional neural network-based models for image segmentation, registration and transformation. The framework allows for round-trip analysis of imaging data, enabling users to apply AI models to their images on CGC and retrieve and review results on their local machine without requiring local installation of specialized software or GPU hardware. The deployed AI models can be accessed using APIs provided by CGC, enabling their use in a variety of programming languages. In summary, the presented framework facilitates end-to-end radiological image analysis and reproducible research, including pulling data from sources, training or inferring from an AI model, utilities for data management, visualization, and simplified access to image metadata.
Collapse
|
34
|
Mitchell JL, Yablonski M, Stone HL, Fuentes-Jimenez M, Takada ME, Tang KA, Tran JE, Chou C, Yeatman JD. Small or absent Visual Word Form Area is a trait of dyslexia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.14.632854. [PMID: 39868322 PMCID: PMC11761755 DOI: 10.1101/2025.01.14.632854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Understanding the balance between plastic and persistent traits in the dyslexic brain is critical for developing effective interventions. This longitudinal intervention study examines the Visual Word Form Area (VWFA) in dyslexic and typical readers, exploring how this key component of the brain's reading circuitry changes with learning. We found that dyslexic readers show significant differences in VWFA presence, size, and tuning properties compared to typical readers. While reading intervention improved reading skills and increased VWFA size, disparities persisted, suggesting that VWFA abnormalities are an enduring trait of dyslexia. Notably, we found that even with sufficient intervention to close the reading skill gap, dyslexic readers are still expected to have smaller VWFAs. Our results reveal intervention-driven long-term neural and behavioral changes, while also elucidating stable differences in the functional architecture of the dyslexic brain. This provides new insights into the potential and limitations of learning-induced plasticity in the human visual cortex.
Collapse
Affiliation(s)
- Jamie L Mitchell
- Graduate School of Education, Stanford University, Stanford, CA, USA
- Department of Psychology, Stanford University, Stanford, CA, USA
| | - Maya Yablonski
- Graduate School of Education, Stanford University, Stanford, CA, USA
- Division of Developmental-Behavioral Pediatrics, Department of Pediatrics, Stanford University School of Medicine, Stanford,California, USA
| | - Hannah L Stone
- Graduate School of Education, Stanford University, Stanford, CA, USA
- Department of Psychological & Brain Sciences, University of California, Santa Baraba, CA, USA
| | | | - Megumi E Takada
- Graduate School of Education, Stanford University, Stanford, CA, USA
| | - Kenny A Tang
- Department of Special Education, Peabody College of Education and Human Development, Vanderbilt University, Nashville, TN, USA
| | - Jasmine E Tran
- Graduate School of Education, Stanford University, Stanford, CA, USA
- School of Education, University of California, Irvine, CA, USA
| | - Clementine Chou
- Graduate School of Education, Stanford University, Stanford, CA, USA
| | - Jason D Yeatman
- Graduate School of Education, Stanford University, Stanford, CA, USA
- Department of Psychology, Stanford University, Stanford, CA, USA
- Division of Developmental-Behavioral Pediatrics, Department of Pediatrics, Stanford University School of Medicine, Stanford,California, USA
| |
Collapse
|
35
|
Grzadzinski R, Mata K, Bhatt AS, Jatkar A, Garic D, Shen MD, Girault JB, St John T, Pandey J, Zwaigenbaum L, Estes A, Shen AM, Dager S, Schultz R, Botteron K, Marrus N, Styner M, Evans A, Kim SH, McKinstry R, Gerig G, Piven J, Hazlett H. Brain volumes, cognitive, and adaptive skills in school-age children with Down syndrome. J Neurodev Disord 2024; 16:70. [PMID: 39701965 PMCID: PMC11660842 DOI: 10.1186/s11689-024-09581-6] [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: 06/03/2024] [Accepted: 11/06/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Down syndrome (DS) is the most common congenital neurodevelopmental disorder, present in about 1 in every 700 live births. Despite its prevalence, literature exploring the neurobiology underlying DS and how this neurobiology is related to behavior is limited. This study fills this gap by examining cortical volumes and behavioral correlates in school-age children with DS. METHODS School-age children (mean = 9.7 years ± 1.1) underwent comprehensive assessments, including cognitive and adaptive assessments, as well as an MRI scan without the use of sedation. Children with DS (n = 35) were compared to available samples of typically developing (TD; n = 80) and ASD children (n = 29). ANOVAs were conducted to compare groups on cognitive and adaptive assessments. ANCOVAs (covarying for age, sex, and total cerebral volume; TCV) compared cortical brain volumes between groups. Correlations between behavioral metrics and cortical and cerebellar volumes (separately for gray (GM) and white matter (WM)) were conducted separately by group. RESULTS As expected, children with DS had significantly lower cognitive skills compared to ASD and TD children. Daily Living adaptive skills were comparable between ASD children and children with DS, and both groups scored lower than TD children. Children with DS exhibited a smaller TCV compared to ASD and TD children. Additionally, when controlling for TCV, age, and sex, children with DS had significantly smaller total GM and tissue volumes. Cerebellum volumes were significantly correlated with Daily Living adaptive behaviors in the DS group only. CONCLUSIONS Despite children with DS exhibiting lower cognitive skills and smaller brain volume overall than children with ASD, their deficits in Socialization and Daily Living adaptive skills are comparable. Differences in lobar volumes (e.g., Right Frontal GM/WM, Left Frontal WM, and Left and Right Temporal WM) were observed above and beyond overall differences in total volume. The correlation between cerebellum volumes and Daily Living adaptive behaviors in the DS group provides a novel area to explore in future research.
Collapse
Affiliation(s)
- Rebecca Grzadzinski
- Carolina Institute for Developmental Disabilities (CIDD), University of North Carolina at Chapel Hill, 101, Renee Lynne Court, Carrboro, NC, 27510, USA.
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Kattia Mata
- Carolina Institute for Developmental Disabilities (CIDD), University of North Carolina at Chapel Hill, 101, Renee Lynne Court, Carrboro, NC, 27510, USA
| | - Ambika S Bhatt
- Carolina Institute for Developmental Disabilities (CIDD), University of North Carolina at Chapel Hill, 101, Renee Lynne Court, Carrboro, NC, 27510, USA
| | | | - Dea Garic
- Carolina Institute for Developmental Disabilities (CIDD), University of North Carolina at Chapel Hill, 101, Renee Lynne Court, Carrboro, NC, 27510, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mark D Shen
- Carolina Institute for Developmental Disabilities (CIDD), University of North Carolina at Chapel Hill, 101, Renee Lynne Court, Carrboro, NC, 27510, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jessica B Girault
- Carolina Institute for Developmental Disabilities (CIDD), University of North Carolina at Chapel Hill, 101, Renee Lynne Court, Carrboro, NC, 27510, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Tanya St John
- University of Washington Autism Research Center, Seattle, WA, USA
- Department of Speech and Hearing Sciences, University of Washington, Seattle, WA, USA
| | - Juhi Pandey
- Center for Autism Research at the Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Lonnie Zwaigenbaum
- Autism Research Centre, Department of Pediatrics, University of Alberta, Edmonton, Canada
| | - Annette Estes
- University of Washington Autism Research Center, Seattle, WA, USA
- Department of Speech and Hearing Sciences, University of Washington, Seattle, WA, USA
| | | | - Stephen Dager
- Center On Human Development and Disability, University of Washington, Seattle, WA, USA
| | - Robert Schultz
- Center for Autism Research at the Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kelly Botteron
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA
| | - Natasha Marrus
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA
| | - Martin Styner
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alan Evans
- Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Sun Hyung Kim
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Robert McKinstry
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Guido Gerig
- Department of Computer Science and Engineering, New York University, New York, NY, USA
| | - Joseph Piven
- Carolina Institute for Developmental Disabilities (CIDD), University of North Carolina at Chapel Hill, 101, Renee Lynne Court, Carrboro, NC, 27510, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Heather Hazlett
- Carolina Institute for Developmental Disabilities (CIDD), University of North Carolina at Chapel Hill, 101, Renee Lynne Court, Carrboro, NC, 27510, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
36
|
Jeon H, Kim J, Kim J, Choi YK, Ho CLA, Pifferi F, Huber D, Feng L, Kim J. eLemur: A cellular-resolution 3D atlas of the mouse lemur brain. Proc Natl Acad Sci U S A 2024; 121:e2413687121. [PMID: 39630862 DOI: 10.1073/pnas.2413687121] [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: 07/11/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024] Open
Abstract
The gray mouse lemur (Microcebus murinus), one of the smallest living primates, emerges as a promising model organism for neuroscience research. This is due to its genetic similarity to humans, its evolutionary position between rodents and humans, and its primate-like features encapsulated within a rodent-sized brain. Despite its potential, the absence of a comprehensive reference brain atlas impedes the progress of research endeavors in this species, particularly at the microscopic level. Existing references have largely been confined to the macroscopic scale, lacking detailed anatomical information. Here, we present eLemur, a unique resource, comprising a repository of high-resolution brain-wide images immunostained with multiple cell type and structural markers, elucidating the cyto- and chemoarchitecture of the mouse lemur brain. Additionally, it encompasses a segmented two-dimensional reference and 3D anatomical brain atlas delineated into cortical, subcortical, and other vital regions. Furthermore, eLemur includes a comprehensive 3D cell atlas, providing densities and spatial distributions of non-neuronal and neuronal cells across the mouse lemur brain. Accessible via a web-based viewer (https://eeum-brain.com/#/lemurdatasets), the eLemur resource streamlines data sharing and integration, fostering the exploration of different hypotheses and experimental designs using the mouse lemur as a model organism. Moreover, in conjunction with the growing 3D datasets for rodents, nonhuman primates, and humans, our eLemur 3D digital framework enhances the potential for comparative analysis and translation research, facilitating the integration of extensive rodent study data into human studies.
Collapse
Affiliation(s)
- Hyungju Jeon
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, South Korea
| | - Jiwon Kim
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, South Korea
- Division of Bio-Medical Science & Technology, Korea Institute of Science and Technology-School, University of Science and Technology, Seoul 02792, South Korea
| | - Jayoung Kim
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, South Korea
- Division of Bio-Medical Science & Technology, Korea Institute of Science and Technology-School, University of Science and Technology, Seoul 02792, South Korea
| | - Yoon Kyoung Choi
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, South Korea
- Department of Computer Science and Engineering, Korea University, Seoul 02841, South Korea
| | - Chun Lum Andy Ho
- Department of Basic Neurosciences, University of Geneva, Geneva 1205, Switzerland
| | - Fabien Pifferi
- Musée National d'Histoire Naturelle, Adaptive Mechanisms and Evolution, UMR7179-CNRS, Paris 75005, France
| | - Daniel Huber
- Department of Basic Neurosciences, University of Geneva, Geneva 1205, Switzerland
| | - Linqing Feng
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, South Korea
| | - Jinhyun Kim
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, South Korea
- Division of Bio-Medical Science & Technology, Korea Institute of Science and Technology-School, University of Science and Technology, Seoul 02792, South Korea
- Department of Computer Science and Engineering, Korea University, Seoul 02841, South Korea
- Korea Institute of Science and Technology-Sungkyunkwan University Brain Research Center, Sungkyunkwan University Institute for Convergence, Sungkyunkwan University, Suwon 16419, South Korea
| |
Collapse
|
37
|
Stirnberg R, Deistung A, Reichenbach JR, Breteler MMB, Stöcker T. Rapid submillimeter QSM and R 2* mapping using interleaved multishot 3D-EPI at 7 and 3 Tesla. Magn Reson Med 2024; 92:2294-2311. [PMID: 38988040 DOI: 10.1002/mrm.30216] [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: 12/28/2023] [Revised: 06/05/2024] [Accepted: 06/24/2024] [Indexed: 07/12/2024]
Abstract
PURPOSE To explore the high signal-to-noise ratio (SNR) efficiency of interleaved multishot 3D-EPI with standard image reconstruction for fast and robust high-resolution whole-brain quantitative susceptibility (QSM) andR 2 ∗ $$ {R}_2^{\ast } $$ mapping at 7 and 3T. METHODS Single- and multi-TE segmented 3D-EPI is combined with conventional CAIPIRINHA undersampling for up to 72-fold effective gradient echo (GRE) imaging acceleration. Across multiple averages, scan parameters are varied (e.g., dual-polarity frequency-encoding) to additionally correct forB 0 $$ {\mathrm{B}}_0 $$ -induced artifacts, geometric distortions and motion retrospectively. A comparison to established GRE protocols is made. Resolutions range from 1.4 mm isotropic (1 multi-TE average in 36 s) up to 0.4 mm isotropic (2 single-TE averages in approximately 6 min) with whole-head coverage. RESULTS Only 1-4 averages are needed for sufficient SNR with 3D-EPI, depending on resolution and field strength. Fast scanning and small voxels together with retrospective corrections result in substantially reduced image artifacts, which improves susceptibility andR 2 ∗ $$ {R}_2^{\ast } $$ mapping. Additionally, much finer details are obtained in susceptibility-weighted image projections through significantly reduced partial voluming. CONCLUSION Using interleaved multishot 3D-EPI, single-TE and multi-TE data can readily be acquired 10 times faster than with conventional, accelerated GRE imaging. Even 0.4 mm isotropic whole-head QSM within 6 min becomes feasible at 7T. At 3T, motion-robust 0.8 mm isotropic whole-brain QSM andR 2 ∗ $$ {R}_2^{\ast } $$ mapping with no apparent distortion in less than 7 min becomes clinically feasible. Stronger gradient systems may allow for even higher effective acceleration rates through larger EPI factors while maintaining optimal contrast.
Collapse
Affiliation(s)
- Rüdiger Stirnberg
- MR Physics, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Andreas Deistung
- Clinic and Outpatient Clinic for Radiology, University Hospital Halle (Saale), University Medicine Halle, Halle (Saale), Germany
| | - Jürgen R Reichenbach
- Medical Physics Group, Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Jena, Germany
| | - Monique M B Breteler
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Faculty of Medicine, Institute for Medical Biometry, Informatics and Epidemiology (IMBIE), University of Bonn, Bonn, Germany
| | - Tony Stöcker
- MR Physics, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Physics and Astronomy, University of Bonn, Bonn, Germany
| |
Collapse
|
38
|
Ball G, Oldham S, Kyriakopoulou V, Williams LZJ, Karolis V, Price A, Hutter J, Seal ML, Alexander-Bloch A, Hajnal JV, Edwards AD, Robinson EC, Seidlitz J. Molecular signatures of cortical expansion in the human foetal brain. Nat Commun 2024; 15:9685. [PMID: 39516464 PMCID: PMC11549424 DOI: 10.1038/s41467-024-54034-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
The third trimester of human gestation is characterised by rapid increases in brain volume and cortical surface area. Recent studies have revealed a remarkable molecular diversity across the prenatal cortex but little is known about how this diversity translates into the differential rates of cortical expansion observed during gestation. We present a digital resource, μBrain, to facilitate knowledge translation between molecular and anatomical descriptions of the prenatal brain. Using μBrain, we evaluate the molecular signatures of preferentially-expanded cortical regions, quantified in utero using magnetic resonance imaging. Our findings demonstrate a spatial coupling between areal differences in the timing of neurogenesis and rates of neocortical expansion during gestation. We identify genes, upregulated from mid-gestation, that are highly expressed in rapidly expanding neocortex and implicated in genetic disorders with cognitive sequelae. The μBrain atlas provides a tool to comprehensively map early brain development across domains, model systems and resolution scales.
Collapse
Affiliation(s)
- G Ball
- Developmental Imaging, Murdoch Children's Research Institute, Melbourne, Australia.
- Department of Paediatrics, University of Melbourne, Melbourne, Australia.
| | - S Oldham
- Developmental Imaging, Murdoch Children's Research Institute, Melbourne, Australia
| | - V Kyriakopoulou
- Centre for the Developing Brain, King's College London, London, UK
- School of Biomedical Engineering & Imaging Science, King's College London, London, UK
| | - L Z J Williams
- Centre for the Developing Brain, King's College London, London, UK
- School of Biomedical Engineering & Imaging Science, King's College London, London, UK
| | - V Karolis
- Centre for the Developing Brain, King's College London, London, UK
- Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - A Price
- Centre for the Developing Brain, King's College London, London, UK
- School of Biomedical Engineering & Imaging Science, King's College London, London, UK
| | - J Hutter
- Centre for the Developing Brain, King's College London, London, UK
- School of Biomedical Engineering & Imaging Science, King's College London, London, UK
| | - M L Seal
- Developmental Imaging, Murdoch Children's Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - A Alexander-Bloch
- Department of Child and Adolescent Psychiatry and Behavioral Sciences, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Lifespan Brain Institute, The Children's Hospital of Philadelphia and Penn Medicine, Philadelphia, PA, USA
- Institute of Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
| | - J V Hajnal
- Centre for the Developing Brain, King's College London, London, UK
- School of Biomedical Engineering & Imaging Science, King's College London, London, UK
| | - A D Edwards
- Centre for the Developing Brain, King's College London, London, UK
- School of Biomedical Engineering & Imaging Science, King's College London, London, UK
| | - E C Robinson
- Centre for the Developing Brain, King's College London, London, UK
- School of Biomedical Engineering & Imaging Science, King's College London, London, UK
| | - J Seidlitz
- Department of Child and Adolescent Psychiatry and Behavioral Sciences, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
- Lifespan Brain Institute, The Children's Hospital of Philadelphia and Penn Medicine, Philadelphia, PA, USA
- Institute of Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
39
|
Veldmann M, Edwards LJ, Pine KJ, Ehses P, Ferreira M, Weiskopf N, Stoecker T. Improving MR axon radius estimation in human white matter using spiral acquisition and field monitoring. Magn Reson Med 2024; 92:1898-1912. [PMID: 38817204 DOI: 10.1002/mrm.30180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/08/2024] [Accepted: 05/15/2024] [Indexed: 06/01/2024]
Abstract
PURPOSE To compare MR axon radius estimation in human white matter using a multiband spiral sequence combined with field monitoring to the current state-of-the-art echo-planar imaging (EPI)-based approach. METHODS A custom multiband spiral sequence was used for diffusion-weighted imaging at ultra-highb $$ b $$ -values. Field monitoring and higher order image reconstruction were employed to greatly reduce artifacts in spiral images. Diffusion weighting parameters were chosen to match a state-of-the art EPI-based axon radius mapping protocol. The spiral approach was compared to the EPI approach by comparing the image signal-to-noise ratio (SNR) and performing a test-retest study to assess the respective variability and repeatability of axon radius mapping. Effective axon radius estimates were compared over white matter voxels and along the left corticospinal tract. RESULTS Increased SNR and reduced artifacts in spiral images led to reduced variability in resulting axon radius maps, especially in low-SNR regions. Test-retest variability was reduced by a factor of approximately 1.5 using the spiral approach. Reduced repeatability due to significant bias was found for some subjects in both spiral and EPI approaches, and attributed to scanner instability, pointing to a previously unknown limitation of the state-of-the-art approach. CONCLUSION Combining spiral readouts with field monitoring improved mapping of the effective axon radius compared to the conventional EPI approach.
Collapse
Affiliation(s)
- Marten Veldmann
- MR Physics, German Center for Neurodegenerative Diseases (DZNE) e.V, Bonn, Germany
| | - Luke J Edwards
- Department of Neurophysics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Kerrin J Pine
- Department of Neurophysics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Philipp Ehses
- MR Physics, German Center for Neurodegenerative Diseases (DZNE) e.V, Bonn, Germany
| | - Mónica Ferreira
- Clinical Research, German Center for Neurodegenerative Diseases (DZNE) e.V, Bonn, Germany
- University of Bonn, Bonn, Germany
| | - Nikolaus Weiskopf
- Department of Neurophysics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Felix Bloch Institute for Solid State Physics, Faculty of Physics and Earth System Sciences, Leipzig University, Leipzig, Germany
- Wellcome Centre for Human Neuroimaging, Institute of Neurology, University College London, London, UK
| | - Tony Stoecker
- MR Physics, German Center for Neurodegenerative Diseases (DZNE) e.V, Bonn, Germany
- Department of Physics & Astronomy, University of Bonn, Bonn, Germany
| |
Collapse
|
40
|
Grødem EOS, Leonardsen E, MacIntosh BJ, Bjørnerud A, Schellhorn T, Sørensen Ø, Amlien I, Fjell AM. A minimalistic approach to classifying Alzheimer's disease using simple and extremely small convolutional neural networks. J Neurosci Methods 2024; 411:110253. [PMID: 39168252 DOI: 10.1016/j.jneumeth.2024.110253] [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: 04/03/2024] [Revised: 07/12/2024] [Accepted: 08/16/2024] [Indexed: 08/23/2024]
Abstract
BACKGROUND There is a broad interest in deploying deep learning-based classification algorithms to identify individuals with Alzheimer's disease (AD) from healthy controls (HC) based on neuroimaging data, such as T1-weighted Magnetic Resonance Imaging (MRI). The goal of the current study is to investigate whether modern, flexible architectures such as EfficientNet provide any performance boost over more standard architectures. METHODS MRI data was sourced from the Alzheimer's Disease Neuroimaging Initiative (ADNI) and processed with a minimal preprocessing pipeline. Among the various architectures tested, the minimal 3D convolutional neural network SFCN stood out, composed solely of 3x3x3 convolution, batch normalization, ReLU, and max-pooling. We also examined the influence of scale on performance, testing SFCN versions with trainable parameters ranging from 720 up to 2.9 million. RESULTS SFCN achieves a test ROC AUC of 96.0% while EfficientNet got an ROC AUC of 94.9 %. SFCN retained high performance down to 720 trainable parameters, achieving an ROC AUC of 91.4%. COMPARISON WITH EXISTING METHODS The SFCN is compared to DenseNet and EfficientNet as well as the results of other publications in the field. CONCLUSIONS The results indicate that using the minimal 3D convolutional neural network SFCN with a minimal preprocessing pipeline can achieve competitive performance in AD classification, challenging the necessity of employing more complex architectures with a larger number of parameters. This finding supports the efficiency of simpler deep learning models for neuroimaging-based AD diagnosis, potentially aiding in better understanding and diagnosing Alzheimer's disease.
Collapse
Affiliation(s)
- Edvard O S Grødem
- Computational Radiology & Artificial Intelligence unit, Division of Radiology and Nuclear Medicine, Oslo University Hospital, 0372, Oslo, Norway; Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, 0373, Oslo, Norway.
| | - Esten Leonardsen
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, 0373, Oslo, Norway; Norwegian Centre for Mental Disorders Research, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, 0373, Oslo, Norway
| | - Bradley J MacIntosh
- Computational Radiology & Artificial Intelligence unit, Division of Radiology and Nuclear Medicine, Oslo University Hospital, 0372, Oslo, Norway; Department of Medical Biophysics, Sunnybrook Research Institute, University of Toronto, M5G 1L7, Toronto, Canada
| | - Atle Bjørnerud
- Computational Radiology & Artificial Intelligence unit, Division of Radiology and Nuclear Medicine, Oslo University Hospital, 0372, Oslo, Norway
| | - Till Schellhorn
- Computational Radiology & Artificial Intelligence unit, Division of Radiology and Nuclear Medicine, Oslo University Hospital, 0372, Oslo, Norway
| | - Øystein Sørensen
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, 0373, Oslo, Norway
| | - Inge Amlien
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, 0373, Oslo, Norway
| | - Anders M Fjell
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, 0373, Oslo, Norway
| |
Collapse
|
41
|
Karkalousos D, Išgum I, Marquering HA, Caan MWA. ATOMMIC: An Advanced Toolbox for Multitask Medical Imaging Consistency to facilitate Artificial Intelligence applications from acquisition to analysis in Magnetic Resonance Imaging. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 256:108377. [PMID: 39180913 DOI: 10.1016/j.cmpb.2024.108377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 07/26/2024] [Accepted: 08/15/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND AND OBJECTIVES Artificial intelligence (AI) is revolutionizing Magnetic Resonance Imaging (MRI) along the acquisition and processing chain. Advanced AI frameworks have been applied in various successive tasks, such as image reconstruction, quantitative parameter map estimation, and image segmentation. However, existing frameworks are often designed to perform tasks independently of each other or are focused on specific models or single datasets, limiting generalization. This work introduces the Advanced Toolbox for Multitask Medical Imaging Consistency (ATOMMIC), a novel open-source toolbox that streamlines AI applications for accelerated MRI reconstruction and analysis. ATOMMIC implements several tasks using deep learning (DL) models and enables MultiTask Learning (MTL) to perform related tasks in an integrated manner, targeting generalization in the MRI domain. METHODS We conducted a comprehensive literature review and analyzed 12,479 GitHub repositories to assess the current landscape of AI frameworks for MRI. Subsequently, we demonstrate how ATOMMIC standardizes workflows and improves data interoperability, enabling effective benchmarking of various DL models across MRI tasks and datasets. To showcase ATOMMIC's capabilities, we evaluated twenty-five DL models on eight publicly available datasets, focusing on accelerated MRI reconstruction, segmentation, quantitative parameter map estimation, and joint accelerated MRI reconstruction and segmentation using MTL. RESULTS ATOMMIC's high-performance training and testing capabilities, utilizing multiple GPUs and mixed precision support, enable efficient benchmarking of multiple models across various tasks. The framework's modular architecture implements each task through a collection of data loaders, models, loss functions, evaluation metrics, and pre-processing transformations, facilitating seamless integration of new tasks, datasets, and models. Our findings demonstrate that ATOMMIC supports MTL for multiple MRI tasks with harmonized complex-valued and real-valued data support while maintaining active development and documentation. Task-specific evaluations demonstrate that physics-based models outperform other approaches in reconstructing highly accelerated acquisitions. These high-quality reconstruction models also show superior accuracy in estimating quantitative parameter maps. Furthermore, when combining high-performing reconstruction models with robust segmentation networks through MTL, performance is improved in both tasks. CONCLUSIONS ATOMMIC advances MRI reconstruction and analysis by leveraging MTL and ensuring consistency across tasks, models, and datasets. This comprehensive framework serves as a versatile platform for researchers to use existing AI methods and develop new approaches in medical imaging.
Collapse
Affiliation(s)
- Dimitrios Karkalousos
- Department of Biomedical Engineering & Physics, Amsterdam University Medical Center, Location University of Amsterdam, Amsterdam, The Netherlands; Department of Radiology & Nuclear Medicine, Amsterdam University Medical Center, Location University of Amsterdam, Amsterdam, The Netherlands; Amsterdam Neuroscience, Brain Imaging, Amsterdam, The Netherlands.
| | - Ivana Išgum
- Department of Biomedical Engineering & Physics, Amsterdam University Medical Center, Location University of Amsterdam, Amsterdam, The Netherlands; Department of Radiology & Nuclear Medicine, Amsterdam University Medical Center, Location University of Amsterdam, Amsterdam, The Netherlands; Informatics Institute, University of Amsterdam, Amsterdam, The Netherlands
| | - Henk A Marquering
- Department of Biomedical Engineering & Physics, Amsterdam University Medical Center, Location University of Amsterdam, Amsterdam, The Netherlands; Department of Radiology & Nuclear Medicine, Amsterdam University Medical Center, Location University of Amsterdam, Amsterdam, The Netherlands; Amsterdam Neuroscience, Brain Imaging, Amsterdam, The Netherlands
| | - Matthan W A Caan
- Department of Biomedical Engineering & Physics, Amsterdam University Medical Center, Location University of Amsterdam, Amsterdam, The Netherlands; Amsterdam Neuroscience, Brain Imaging, Amsterdam, The Netherlands
| |
Collapse
|
42
|
Dadu A, Ta M, Tustison NJ, Daneshmand A, Marek K, Singleton AB, Campbell RH, Nalls MA, Iwaki H, Avants B, Faghri F. Prediction, prognosis and monitoring of neurodegeneration at biobank-scale via machine learning and imaging. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.27.24316215. [PMID: 39574848 PMCID: PMC11581077 DOI: 10.1101/2024.10.27.24316215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
Background Alzheimer's disease and related dementias (ADRD) and Parkinson's disease (PD) are the most common neurodegenerative conditions. These central nervous system disorders impact both the structure and function of the brain and may lead to imaging changes that precede symptoms. Patients with ADRD or PD have long asymptomatic phases that exhibit significant heterogeneity. Hence, quantitative measures that can provide early disease indicators are necessary to improve patient stratification, clinical care, and clinical trial design. This work uses machine learning techniques to derive such a quantitative marker from T1-weighted (T1w) brain Magnetic resonance imaging (MRI). Methods In this retrospective study, we developed machine learning (ML) based disease-specific scores based on T1w brain MRI utilizing Parkinson's Disease Progression Marker Initiative (PPMI) and Alzheimer's Disease Neuroimaging Initiative (ADNI) cohorts. We evaluated the potential of ML-based scores for early diagnosis, prognosis, and monitoring of ADRD and PD in an independent large-scale population-based longitudinal cohort, UK Biobank. Findings 1,826 dementia images from 731 participants, 3,161 healthy control images from 925 participants from the ADNI cohort, 684 PD images from 319 participants, and 232 healthy control images from 145 participants from the PPMI cohort were used to train machine learning models. The classification performance is 0.94 [95% CI: 0.93-0.96] area under the ROC Curve (AUC) for ADRD detection and 0.63 [95% CI: 0.57-0.71] for PD detection using 790 extracted structural brain features. The most predictive regions include the hippocampus and temporal brain regions in ADRD and the substantia nigra in PD. The normalized ML model's probabilistic output (ADRD and PD imaging scores) was evaluated on 42,835 participants with imaging data from the UK Biobank. There are 66 cases for ADRD and 40 PD cases whose T1 brain MRI is available during pre-diagnostic phases. For diagnosis occurrence events within 5 years, the integrated survival model achieves a time-dependent AUC of 0.86 [95% CI: 0.80-0.92] for dementia and 0.89 [95% CI: 0.85-0.94] for PD. ADRD imaging score is strongly associated with dementia-free survival (hazard ratio (HR) 1.76 [95% CI: 1.50-2.05] per S.D. of imaging score), and PD imaging score shows association with PD-free survival (hazard ratio 2.33 [95% CI: 1.55-3.50]) in our integrated model. HR and prevalence increased stepwise over imaging score quartiles for PD, demonstrating heterogeneity. As a proxy for diagnosis, we validated AD/PD polygenic risk scores of 42,835 subjects against the imaging scores, showing a highly significant association after adjusting for covariates. In both the PPMI and ADNI cohorts, the scores are associated with clinical assessments, including the Mini-Mental State Examination (MMSE), Alzheimer's Disease Assessment Scale-cognitive subscale (ADAS-Cog), and pathological markers, which include amyloid and tau. Finally, imaging scores are associated with polygenic risk scores for multiple diseases. Our results suggest that we can use imaging scores to assess the genetic architecture of such disorders in the future. Interpretation Our study demonstrates the use of quantitative markers generated using machine learning techniques for ADRD and PD. We show that disease probability scores obtained from brain structural features are useful for early detection, prognosis prediction, and monitoring disease progression. To facilitate community engagement and external tests of model utility, an interactive app to explore summary level data from this study and dive into external data can be found here https://ndds-brainimaging-ml.streamlit.app. As far as we know, this is the first publicly available cloud-based MRI prediction application. Funding US National Institute on Aging, and US National Institutes of Health.
Collapse
Affiliation(s)
- Anant Dadu
- Department of Computer Science, University of Illinois at Urbana-Champaign, Champaign, IL, 61820, USA
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, 20892, USA
- DataTecnica, Washington, DC, 20812, USA
| | - Michael Ta
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, 20892, USA
- DataTecnica, Washington, DC, 20812, USA
| | - Nicholas J Tustison
- University of Virginia, Dept of Radiology and Medical Imaging, Charlottesville, VA, 22903, USA
| | - Ali Daneshmand
- Department of Neurology, Boston Medical Center, Boston University School of Medicine, Boston, MA, 02118, USA
| | | | - Andrew B Singleton
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, 20892, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Roy H Campbell
- Department of Computer Science, University of Illinois at Urbana-Champaign, Champaign, IL, 61820, USA
| | - Mike A Nalls
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, 20892, USA
- DataTecnica, Washington, DC, 20812, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Hirotaka Iwaki
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, 20892, USA
- DataTecnica, Washington, DC, 20812, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Brian Avants
- University of Virginia, Dept of Radiology and Medical Imaging, Charlottesville, VA, 22903, USA
- InviCRO LLC, Boston, Massachusetts
| | - Faraz Faghri
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, 20892, USA
- DataTecnica, Washington, DC, 20812, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
43
|
Kronman FN, Liwang JK, Betty R, Vanselow DJ, Wu YT, Tustison NJ, Bhandiwad A, Manjila SB, Minteer JA, Shin D, Lee CH, Patil R, Duda JT, Xue J, Lin Y, Cheng KC, Puelles L, Gee JC, Zhang J, Ng L, Kim Y. Developmental mouse brain common coordinate framework. Nat Commun 2024; 15:9072. [PMID: 39433760 PMCID: PMC11494176 DOI: 10.1038/s41467-024-53254-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 10/07/2024] [Indexed: 10/23/2024] Open
Abstract
3D brain atlases are key resources to understand the brain's spatial organization and promote interoperability across different studies. However, unlike the adult mouse brain, the lack of developing mouse brain 3D reference atlases hinders advancements in understanding brain development. Here, we present a 3D developmental common coordinate framework (DevCCF) spanning embryonic day (E)11.5, E13.5, E15.5, E18.5, and postnatal day (P)4, P14, and P56, featuring undistorted morphologically averaged atlas templates created from magnetic resonance imaging and co-registered high-resolution light sheet fluorescence microscopy templates. The DevCCF with 3D anatomical segmentations can be downloaded or explored via an interactive 3D web-visualizer. As a use case, we utilize the DevCCF to unveil GABAergic neuron emergence in embryonic brains. Moreover, we map the Allen CCFv3 and spatial transcriptome cell-type data to our stereotaxic P56 atlas. In summary, the DevCCF is an openly accessible resource for multi-study data integration to advance our understanding of brain development.
Collapse
Affiliation(s)
- Fae N Kronman
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| | - Josephine K Liwang
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| | - Rebecca Betty
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| | - Daniel J Vanselow
- Department of Pathology, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| | - Yuan-Ting Wu
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| | - Nicholas J Tustison
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, USA
| | | | - Steffy B Manjila
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| | - Jennifer A Minteer
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| | - Donghui Shin
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| | - Choong Heon Lee
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY, USA
| | - Rohan Patil
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| | - Jeffrey T Duda
- Department of Radiology, Penn Image Computing and Science Lab, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jian Xue
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Yingxi Lin
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Keith C Cheng
- Department of Pathology, College of Medicine, The Pennsylvania State University, Hershey, PA, USA
| | - Luis Puelles
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, Universidad de Murcia, and Murcia Arrixaca Institute for Biomedical Research (IMIB), Murcia, Spain
| | - James C Gee
- Department of Radiology, Penn Image Computing and Science Lab, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jiangyang Zhang
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY, USA
| | - Lydia Ng
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Yongsoo Kim
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA, USA.
| |
Collapse
|
44
|
Baller EB, Luo AC, Schindler MK, Cooper EC, Pecsok MK, Cieslak MC, Martin ML, Bar-Or A, Elahi A, Perrone CM, Reid D, Spangler BC, Satterthwaite TD, Shinohara RT. Association of Anxiety with Uncinate Fasciculus Lesion Burden in Multiple Sclerosis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.08.24315108. [PMID: 39417125 PMCID: PMC11482984 DOI: 10.1101/2024.10.08.24315108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Importance Multiple sclerosis (MS) is an immune-mediated neurological disorder that affects 2.4 million people world-wide, and up to 60% experience anxiety. Objective We investigated how anxiety in MS is associated with white matter lesion burden in the uncinate fasciculus (UF). Design Retrospective case-control study of participants who received research-quality 3-tesla (3T) neuroimaging as part of MS clinical care from 2010-2018. Analyses were performed from June 1st to September 30th, 2024. Setting Single-center academic medical specialty MS clinic. Participants Participants were identified from the electronic medical record. All participants were diagnosed by an MS specialist and completed research-quality MRI at 3T. After excluding participants with poor image quality, 372 were stratified into three groups which were balanced for age and sex: 1) MS without anxiety (MS+noA, n=99); 2) MS with mild anxiety (MS+mildA, n=249); and 3) MS with severe anxiety (MS+severeA, n=24). Exposure Anxiety diagnosis and anxiolytic medication. Main Outcome and Measure We first evaluated whether MS+severeA patients had greater lesion burden in the UF than MS+noA. Next, we examined whether increasing anxiety severity was associated with greater UF lesion burden. Generalized additive models were employed, with the burden of lesions (e.g. proportion of fascicle impacted) within the UF as the outcome measure and sex and spline of age as covariates. Results UF burden was higher in MS+severeA as compared to MS+noA (T=2.02, P=0.045, Cohen's f 2=0.19). A dose-response effect was also found, where higher mean UF burden was associated with higher anxiety severity (T=2.08, P=0.038, Cohen's f 2=0.10). Conclusions and Relevance We demonstrate that overall lesion burden in UF was associated with the presence and severity of anxiety in patients with MS. Future studies linking white matter lesion burden in UF with treatment prognosis are warranted.
Collapse
Affiliation(s)
- Erica B. Baller
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA USA
- Penn Lifespan Informatics and Neuroimaging Center (PennLINC), Philadelphia, PA USA
| | - Audrey C. Luo
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA USA
- Penn Lifespan Informatics and Neuroimaging Center (PennLINC), Philadelphia, PA USA
| | - Matthew K. Schindler
- Department of Neurology, University of Pennsylvania, Philadelphia, PA USA
- Center for Neuroinflammation and Neurotherapeutics, University of Pennsylvania, Philadelphia, PA USA
| | - Elena C. Cooper
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA USA
- Penn Lifespan Informatics and Neuroimaging Center (PennLINC), Philadelphia, PA USA
| | - Margaret K. Pecsok
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA USA
| | - Matthew C. Cieslak
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA USA
- Penn Lifespan Informatics and Neuroimaging Center (PennLINC), Philadelphia, PA USA
| | - Melissa Lynne Martin
- Penn Statistics in Imaging and Visualization Center (PennSIVE), Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA USA
| | - Amit Bar-Or
- Department of Neurology, University of Pennsylvania, Philadelphia, PA USA
- Center for Neuroinflammation and Neurotherapeutics, University of Pennsylvania, Philadelphia, PA USA
| | - Ameena Elahi
- Department of Information Services, University of Pennsylvania, Philadelphia, PA USA
| | - Christopher M. Perrone
- Department of Neurology, University of Pennsylvania, Philadelphia, PA USA
- Center for Neuroinflammation and Neurotherapeutics, University of Pennsylvania, Philadelphia, PA USA
| | - Donovan Reid
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA USA
| | - Bailey C. Spangler
- Penn Statistics in Imaging and Visualization Center (PennSIVE), Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA USA
| | - Theodore D. Satterthwaite
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA USA
- Penn Lifespan Informatics and Neuroimaging Center (PennLINC), Philadelphia, PA USA
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA USA
| | - Russell T. Shinohara
- Penn Statistics in Imaging and Visualization Center (PennSIVE), Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA USA
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA USA
| |
Collapse
|
45
|
Cooper EC, Schindler MK, Bar-Or A, Brandstadter RB, Calkins ME, Gur RC, Jacobs DA, Markowitz CE, Moore TM, Naydovich LR, Perrone CM, Ruparel K, Spangler BC, Troyan S, Shinohara RT, Satterthwaite TD, Baller EB. Investigating Mood and Cognition in People with Multiple Sclerosis: A Prospective Study Protocol. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.02.24314787. [PMID: 39464257 PMCID: PMC11507955 DOI: 10.1101/2024.10.02.24314787] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Multiple sclerosis (MS) is an immune-mediated neurological disorder that affects one million people in the United States. Up to 50% of people with MS experience depression, yet the mechanisms of depression in MS remain under-investigated. Studies of medically healthy participants with depression have described associations between white matter variability and depressive symptoms, but frequently exclude participants with medical comorbidities and thus cannot be extrapolated to people with intracranial diseases. White matter lesions are a key pathologic feature of MS and could disrupt pathways involved in depression symptoms. The purpose of this study is to investigate the impact of brain network disruption on depression using MS as a model. We will obtain structured clinical and cognitive assessments from two hundred fifty participants with MS and prospectively evaluate white matter lesion burden as a predictor of depressive symptoms. Ethics approval was obtained from The University of Pennsylvania Institutional Review Board (Protocol #853883). The results of this study will be presented at scientific meetings and conferences and published in peer-reviewed journals.
Collapse
Affiliation(s)
- Elena C. Cooper
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA USA
- Penn Lifespan Informatics and Neuroimaging Center (PennLINC), Philadelphia, PA USA
| | - Matthew K. Schindler
- Department of Neurology, University of Pennsylvania, Philadelphia, PA USA
- Center for Neuroinflammation and Neurotherapeutics, University of Pennsylvania, Philadelphia, PA USA
| | - Amit Bar-Or
- Department of Neurology, University of Pennsylvania, Philadelphia, PA USA
- Center for Neuroinflammation and Neurotherapeutics, University of Pennsylvania, Philadelphia, PA USA
| | - Rachel B. Brandstadter
- Department of Neurology, University of Pennsylvania, Philadelphia, PA USA
- Center for Neuroinflammation and Neurotherapeutics, University of Pennsylvania, Philadelphia, PA USA
| | - Monica E. Calkins
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA USA
- Lifespan Brain Institute, Penn Medicine and Children’s Hospital of Philadelphia, Philadelphia, PA USA
| | - Ruben C. Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA USA
- Lifespan Brain Institute, Penn Medicine and Children’s Hospital of Philadelphia, Philadelphia, PA USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Dina A. Jacobs
- Department of Neurology, University of Pennsylvania, Philadelphia, PA USA
- Center for Neuroinflammation and Neurotherapeutics, University of Pennsylvania, Philadelphia, PA USA
| | - Clyde E. Markowitz
- Department of Neurology, University of Pennsylvania, Philadelphia, PA USA
- Center for Neuroinflammation and Neurotherapeutics, University of Pennsylvania, Philadelphia, PA USA
| | - Tyler M. Moore
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA USA
- Lifespan Brain Institute, Penn Medicine and Children’s Hospital of Philadelphia, Philadelphia, PA USA
| | - Laura R. Naydovich
- Department of Neurology, University of Pennsylvania, Philadelphia, PA USA
- Center for Neuroinflammation and Neurotherapeutics, University of Pennsylvania, Philadelphia, PA USA
| | - Christopher M. Perrone
- Department of Neurology, University of Pennsylvania, Philadelphia, PA USA
- Center for Neuroinflammation and Neurotherapeutics, University of Pennsylvania, Philadelphia, PA USA
| | - Kosha Ruparel
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA USA
| | - Bailey C. Spangler
- Penn Statistics in Imaging and Visualization Center (PennSIVE), Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA USA
| | - Scott Troyan
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA USA
| | - Russell T. Shinohara
- Penn Statistics in Imaging and Visualization Center (PennSIVE), Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA USA
- Department of Information Services, University of Pennsylvania, Philadelphia, PA USA
| | - Theodore D. Satterthwaite
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA USA
- Penn Lifespan Informatics and Neuroimaging Center (PennLINC), Philadelphia, PA USA
- Department of Information Services, University of Pennsylvania, Philadelphia, PA USA
| | - Erica B. Baller
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA USA
- Penn Lifespan Informatics and Neuroimaging Center (PennLINC), Philadelphia, PA USA
| |
Collapse
|
46
|
Shi Z, Li X, Todaro DR, Cao W, Lynch KG, Detre JA, Loughead J, Langleben DD, Wiers CE. Medial prefrontal neuroplasticity during extended-release naltrexone treatment of opioid use disorder - a longitudinal structural magnetic resonance imaging study. Transl Psychiatry 2024; 14:360. [PMID: 39237534 PMCID: PMC11377591 DOI: 10.1038/s41398-024-03061-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/07/2024] Open
Abstract
Opioid use disorder (OUD) has been linked to macroscopic structural alterations in the brain. The monthly injectable, extended-release formulation of μ-opioid antagonist naltrexone (XR-NTX) is highly effective in reducing opioid craving and preventing opioid relapse. Here, we investigated the neuroanatomical effects of XR-NTX by examining changes in cortical thickness during treatment for OUD. Forty-seven OUD patients underwent structural magnetic resonance imaging and subjectively rated their opioid craving ≤1 day before (pre-treatment) and 11 ± 3 days after (on-treatment) the first XR-NTX injection. A sample of fifty-six non-OUD individuals completed a single imaging session and served as the comparison group. A publicly available [¹¹C]carfentanil positron emission tomography dataset was used to assess the relationship between changes in cortical thickness and μ-opioid receptor (MOR) binding potential across brain regions. We found that the thickness of the medial prefrontal and anterior cingulate cortices (mPFC/aCC; regions with high MOR binding potential) was comparable between the non-OUD individuals and the OUD patients at pre-treatment. However, among the OUD patients, mPFC/aCC thickness significantly decreased from pre-treatment to on-treatment. A greater reduction in mPFC/aCC thickness was associated with a greater reduction in opioid craving. Taken together, our study suggests XR-NTX-induced cortical thickness reduction in the mPFC/aCC regions in OUD patients. The reduction in thickness does not appear to indicate a restoration to the non-OUD level but rather reflects XR-NTX's distinct therapeutic impact on an MOR-rich brain structure. Our findings highlight the neuroplastic effects of XR-NTX that may inform the development of novel OUD interventions.
Collapse
Affiliation(s)
- Zhenhao Shi
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - Xinyi Li
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Dustin R Todaro
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Wen Cao
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kevin G Lynch
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - John A Detre
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - James Loughead
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Daniel D Langleben
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Corinde E Wiers
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
47
|
Chen Y, Tozer D, Li R, Li H, Tuladhar A, De Leeuw FE, Markus HS. Improved Dementia Prediction in Cerebral Small Vessel Disease Using Deep Learning-Derived Diffusion Scalar Maps From T1. Stroke 2024; 55:2254-2263. [PMID: 39145386 PMCID: PMC11346716 DOI: 10.1161/strokeaha.124.047449] [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: 04/10/2024] [Revised: 06/23/2024] [Accepted: 07/22/2024] [Indexed: 08/16/2024]
Abstract
BACKGROUND Cerebral small vessel disease is the most common pathology underlying vascular dementia. In small vessel disease, diffusion tensor imaging is more sensitive to white matter damage and better predicts dementia risk than conventional magnetic resonance imaging sequences, such as T1 and fluid attenuation inversion recovery, but diffusion tensor imaging takes longer to acquire and is not routinely available in clinical practice. As diffusion tensor imaging-derived scalar maps-fractional anisotropy (FA) and mean diffusivity (MD)-are frequently used in clinical settings, one solution is to synthesize FA/MD from T1 images. METHODS We developed a deep learning model to synthesize FA/MD from T1. The training data set consisted of 4998 participants with the highest white matter hyperintensity volumes in the UK Biobank. Four external validations data sets with small vessel disease were included: SCANS (St George's Cognition and Neuroimaging in Stroke; n=120), RUN DMC (Radboud University Nijmegen Diffusion Tensor and Magnetic Resonance Imaging Cohort; n=502), PRESERVE (Blood Pressure in Established Cerebral Small Vessel Disease; n=105), and NETWORKS (n=26), along with 1000 normal controls from the UK Biobank. RESULTS The synthetic maps resembled ground-truth maps (structural similarity index >0.89 for MD maps and >0.80 for FA maps across all external validation data sets except for SCANS). The prediction accuracy of dementia using whole-brain median MD from the synthetic maps is comparable to the ground truth (SCANS ground-truth c-index, 0.822 and synthetic, 0.821; RUN DMC ground truth, 0.816 and synthetic, 0.812) and better than white matter hyperintensity volume (SCANS, 0.534; RUN DMC, 0.710). CONCLUSIONS We have developed a fast and generalizable method to synthesize FA/MD maps from T1 to improve the prediction accuracy of dementia in small vessel disease when diffusion tensor imaging data have not been acquired.
Collapse
Affiliation(s)
- Yutong Chen
- Department of Clinical Neuroscience, Stroke Research Group, University of Cambridge, United Kingdom (Y.C., D.T., R.L., H.S.M.)
| | - Daniel Tozer
- Department of Clinical Neuroscience, Stroke Research Group, University of Cambridge, United Kingdom (Y.C., D.T., R.L., H.S.M.)
| | - Rui Li
- Department of Clinical Neuroscience, Stroke Research Group, University of Cambridge, United Kingdom (Y.C., D.T., R.L., H.S.M.)
| | - Hao Li
- Department of Clinical Neuroscience, Stroke Research Group, University of Cambridge, United Kingdom (Y.C., D.T., R.L., H.S.M.)
- Department of Neurology, Radboud University Medical Center, Donders Center for Medical Neurosciences, Nijmegen, the Netherlands (H.L., A.T., F.E.D.L.)
| | - Anil Tuladhar
- Department of Neurology, Radboud University Medical Center, Donders Center for Medical Neurosciences, Nijmegen, the Netherlands (H.L., A.T., F.E.D.L.)
| | - Frank Erik De Leeuw
- Department of Neurology, Radboud University Medical Center, Donders Center for Medical Neurosciences, Nijmegen, the Netherlands (H.L., A.T., F.E.D.L.)
| | - Hugh S. Markus
- Department of Clinical Neuroscience, Stroke Research Group, University of Cambridge, United Kingdom (Y.C., D.T., R.L., H.S.M.)
| |
Collapse
|
48
|
Podobnik G, Ibragimov B, Tappeiner E, Lee C, Kim JS, Mesbah Z, Modzelewski R, Ma Y, Yang F, Rudecki M, Wodziński M, Peterlin P, Strojan P, Vrtovec T. HaN-Seg: The head and neck organ-at-risk CT and MR segmentation challenge. Radiother Oncol 2024; 198:110410. [PMID: 38917883 DOI: 10.1016/j.radonc.2024.110410] [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: 04/30/2024] [Revised: 06/12/2024] [Accepted: 06/15/2024] [Indexed: 06/27/2024]
Abstract
BACKGROUND AND PURPOSE To promote the development of auto-segmentation methods for head and neck (HaN) radiation treatment (RT) planning that exploit the information of computed tomography (CT) and magnetic resonance (MR) imaging modalities, we organized HaN-Seg: The Head and Neck Organ-at-Risk CT and MR Segmentation Challenge. MATERIALS AND METHODS The challenge task was to automatically segment 30 organs-at-risk (OARs) of the HaN region in 14 withheld test cases given the availability of 42 publicly available training cases. Each case consisted of one contrast-enhanced CT and one T1-weighted MR image of the HaN region of the same patient, with up to 30 corresponding reference OAR delineation masks. The performance was evaluated in terms of the Dice similarity coefficient (DSC) and 95-percentile Hausdorff distance (HD95), and statistical ranking was applied for each metric by pairwise comparison of the submitted methods using the Wilcoxon signed-rank test. RESULTS While 23 teams registered for the challenge, only seven submitted their methods for the final phase. The top-performing team achieved a DSC of 76.9 % and a HD95 of 3.5 mm. All participating teams utilized architectures based on U-Net, with the winning team leveraging rigid MR to CT registration combined with network entry-level concatenation of both modalities. CONCLUSION This challenge simulated a real-world clinical scenario by providing non-registered MR and CT images with varying fields-of-view and voxel sizes. Remarkably, the top-performing teams achieved segmentation performance surpassing the inter-observer agreement on the same dataset. These results set a benchmark for future research on this publicly available dataset and on paired multi-modal image segmentation in general.
Collapse
Affiliation(s)
- Gašper Podobnik
- University of Ljubljana, Faculty Electrical Engineering, Tržaška cesta 25, Ljubljana 1000, Slovenia.
| | - Bulat Ibragimov
- University of Ljubljana, Faculty Electrical Engineering, Tržaška cesta 25, Ljubljana 1000, Slovenia; University of Copenhagen, Department of Computer Science, Universitetsparken 1, Copenhagen 2100, Denmark
| | - Elias Tappeiner
- UMIT Tirol - Private University for Health Sciences and Health Technology, Eduard-Wallnöfer-Zentrum 1, Hall in Tirol 6060, Austria
| | - Chanwoong Lee
- Yonsei University, College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, South Korea; Yonsei Cancer Center, Department of RadiationOncology, 50-1 Yonsei-Ro, Seodaemun-gu, Seoul 03722, South Korea
| | - Jin Sung Kim
- Yonsei University, College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, South Korea; Yonsei Cancer Center, Department of RadiationOncology, 50-1 Yonsei-Ro, Seodaemun-gu, Seoul 03722, South Korea; Oncosoft Inc, 37 Myeongmul-gil, Seodaemun-gu, Seoul 03722, South Korea
| | - Zacharia Mesbah
- Henri Becquerel Cancer Center, 1 Rue d'Amiens, Rouen 76000, France; Siemens Healthineers, 6 Rue du Général Audran, CS20146, Courbevoie 92412, France
| | - Romain Modzelewski
- Henri Becquerel Cancer Center, 1 Rue d'Amiens, Rouen 76000, France; Litis UR 4108, 684 Av. de l'Université, Saint- Étienne-du-Rouvray 76800, France
| | - Yihao Ma
- Guizhou Medical University, School of Biology & Engineering, 9FW8+2P3, Ankang Avenue, Gui'an New Area, Guiyang, Guizhou Province 561113, China
| | - Fan Yang
- Guizhou Medical University, School of Biology & Engineering, 9FW8+2P3, Ankang Avenue, Gui'an New Area, Guiyang, Guizhou Province 561113, China
| | - Mikołaj Rudecki
- AGH University of Kraków, Department of Measurement and Electronicsal, Mickiewicza 30, Kraków 30-059, Poland
| | - Marek Wodziński
- AGH University of Kraków, Department of Measurement and Electronicsal, Mickiewicza 30, Kraków 30-059, Poland; University of Applied Sciences Western Switzerland, Information Systems Institute, Rue de la Plaine 2, Sierre 3960, Switzerland
| | - Primož Peterlin
- Institute of Oncology, Ljubljana, Zaloška cesta 2, Ljubljana 1000, Slovenia
| | - Primož Strojan
- Institute of Oncology, Ljubljana, Zaloška cesta 2, Ljubljana 1000, Slovenia
| | - Tomaž Vrtovec
- University of Ljubljana, Faculty Electrical Engineering, Tržaška cesta 25, Ljubljana 1000, Slovenia
| |
Collapse
|
49
|
Kwiatkowski A, Weidler C, Habel U, Coverdale NS, Hirad AA, Manning KY, Rauscher A, Bazarian JJ, Cook DJ, Li DKB, Mahon BZ, Menon RS, Taunton J, Reetz K, Romanzetti S, Huppertz C. Uncovering the hidden effects of repetitive subconcussive head impact exposure: A mega-analytic approach characterizing seasonal brain microstructural changes in contact and collision sports athletes. Hum Brain Mapp 2024; 45:e26811. [PMID: 39185683 PMCID: PMC11345636 DOI: 10.1002/hbm.26811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 07/16/2024] [Accepted: 07/20/2024] [Indexed: 08/27/2024] Open
Abstract
Repetitive subconcussive head impacts (RSHI) are believed to induce sub-clinical brain injuries, potentially resulting in cumulative, long-term brain alterations. This study explores patterns of longitudinal brain white matter changes across sports with RSHI-exposure. A systematic literature search identified 22 datasets with longitudinal diffusion magnetic resonance imaging data. Four datasets were centrally pooled to perform uniform quality control and data preprocessing. A total of 131 non-concussed active athletes (American football, rugby, ice hockey; mean age: 20.06 ± 2.06 years) with baseline and post-season data were included. Nonparametric permutation inference (one-sample t tests, one-sided) was applied to analyze the difference maps of multiple diffusion parameters. The analyses revealed widespread lateralized patterns of sports-season-related increases and decreases in mean diffusivity (MD), radial diffusivity (RD), and axial diffusivity (AD) across spatially distinct white matter regions. Increases were shown across one MD-cluster (3195 voxels; mean change: 2.34%), one AD-cluster (5740 voxels; mean change: 1.75%), and three RD-clusters (817 total voxels; mean change: 3.11 to 4.70%). Decreases were shown across two MD-clusters (1637 total voxels; mean change: -1.43 to -1.48%), two RD-clusters (1240 total voxels; mean change: -1.92 to -1.93%), and one AD-cluster (724 voxels; mean change: -1.28%). The resulting pattern implies the presence of strain-induced injuries in central and brainstem regions, with comparatively milder physical exercise-induced effects across frontal and superior regions of the left hemisphere, which need further investigation. This article highlights key considerations that need to be addressed in future work to enhance our understanding of the nature of observed white matter changes, improve the comparability of findings across studies, and promote data pooling initiatives to allow more detailed investigations (e.g., exploring sex- and sport-specific effects).
Collapse
Affiliation(s)
- Anna Kwiatkowski
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical FacultyRWTH Aachen UniversityAachenGermany
| | - Carmen Weidler
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical FacultyRWTH Aachen UniversityAachenGermany
| | - Ute Habel
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical FacultyRWTH Aachen UniversityAachenGermany
- Institute of Neuroscience and Medicine 10, Research Centre JülichJülichGermany
- JARA‐BRAIN Institute Brain Structure Function Relationship, Research Center Jülich and RWTH Aachen UniversityAachenGermany
| | | | - Adnan A. Hirad
- Department of SurgeryUniversity of Rochester Medical CenterRochesterNew YorkUSA
- Department of NeuroscienceUniversity of Rochester Medical CenterRochesterNew YorkUSA
- Del Monte Neuroscience Institute, University of RochesterNew YorkUSA
| | - Kathryn Y. Manning
- Department of RadiologyUniversity of Calgary and Alberta Children's Hospital Research InstituteCalgaryAlbertaCanada
| | - Alexander Rauscher
- Department of Radiology, Faculty of MedicineUniversity of British ColumbiaVancouverBritish ColumbiaCanada
- Department of Pediatrics, Division of NeurologyUniversity of British ColumbiaVancouverBritish ColumbiaCanada
- Department of Physics and AstronomyUniversity of British ColumbiaVancouverBritish ColumbiaCanada
- UBC MRI Research Centre, University of British ColumbiaVancouverBritish ColumbiaCanada
| | - Jeffrey J. Bazarian
- Department of Emergency MedicineUniversity of Rochester School of Medicine and DentistryRochesterNew YorkUSA
| | - Douglas J. Cook
- Centre for Neuroscience Studies, Queen's UniversityKingstonOntarioCanada
- Division of Neurosurgery, Department of SurgeryQueen's UniversityKingstonOntarioCanada
| | - David K. B. Li
- Department of Radiology, Faculty of MedicineUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Bradford Z. Mahon
- Department of PsychologyCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
- Carnegie Mellon Neuroscience InstitutePittsburghPennsylvaniaUSA
- Department of NeurosurgeryUniversity of Rochester Medical CenterRochesterNew YorkUSA
| | - Ravi S. Menon
- Centre for Functional and Metabolic Mapping, Robarts Research Institute, University of Western OntarioLondonOntarioCanada
| | - Jack Taunton
- Allan McGavin Sports Medicine Centre, Faculty of MedicineUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Kathrin Reetz
- Department of Neurology, Medical FacultyRWTH Aachen UniversityAachenGermany
- JARA‐BRAIN Institute Molecular Neuroscience and Neuroimaging, Research Center Jülich and RWTH Aachen UniversityAachenGermany
| | - Sandro Romanzetti
- Department of Neurology, Medical FacultyRWTH Aachen UniversityAachenGermany
- JARA‐BRAIN Institute Molecular Neuroscience and Neuroimaging, Research Center Jülich and RWTH Aachen UniversityAachenGermany
| | - Charlotte Huppertz
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical FacultyRWTH Aachen UniversityAachenGermany
| |
Collapse
|
50
|
Roston RA, Whikehart SM, Rolfe SM, Maga M. Morphological simulation tests the limits on phenotype discovery in 3D image analysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.30.601430. [PMID: 39005442 PMCID: PMC11244899 DOI: 10.1101/2024.06.30.601430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
In the past few decades, advances in 3D imaging have created new opportunities for reverse genetic screens. Rapidly growing datasets of 3D images of genetic knockouts require high-throughput, automated computational approaches for identifying and characterizing new phenotypes. However, exploratory, discovery-oriented image analysis pipelines used to discover these phenotypes can be difficult to validate because, by their nature, the expected outcome is not known a priori . Introducing known morphological variation through simulation can help distinguish between real phenotypic differences and random variation; elucidate the effects of sample size; and test the sensitivity and reproducibility of morphometric analyses. Here we present a novel approach for 3D morphological simulation that uses open-source, open-access tools available in 3D Slicer, SlicerMorph, and Advanced Normalization Tools in R (ANTsR). While we focus on diffusible-iodine contrast-enhanced micro-CT (diceCT) images, this approach can be used on any volumetric image. We then use our simulated datasets to test whether tensor-based morphometry (TBM) can recover our introduced differences; to test how effect size and sample size affect detectability; and to determine the reproducibility of our results. In our approach to morphological simulation, we first generate a simulated deformation based on a reference image and then propagate this deformation to subjects using inverse transforms obtained from the registration of subjects to the reference. This produces a new dataset with a shifted population mean while retaining individual variability because each sample deforms more or less based on how different or similar it is from the reference. TBM is a widely-used technique that statistically compares local volume differences associated with local deformations. Our results showed that TBM recovered our introduced morphological differences, but that detectability was dependent on the effect size, the sample size, and the region of interest (ROI) included in the analysis. Detectability of subtle phenotypes can be improved both by increasing the sample size and by limiting analyses to specific body regions. However, it is not always feasible to increase sample sizes in screens of essential genes. Therefore, methodical use of ROIs is a promising way to increase the power of TBM to detect subtle phenotypes. Generating known morphological variation through simulation has broad applicability in developmental, evolutionary, and biomedical morphometrics and is a useful way to distinguish between a failure to detect morphological difference and a true lack of morphological difference. Morphological simulation can also be applied to AI-based supervised learning to augment datasets and overcome dataset limitations.
Collapse
|