15 68

Cited 0 times in

Multimodal mapping of regional brain vulnerability to focal cortical dysplasia

DC Field Value Language
dc.contributor.author조규호-
dc.date.accessioned2024-05-30T06:54:44Z-
dc.date.available2024-05-30T06:54:44Z-
dc.date.issued2023-08-
dc.identifier.issn0006-8950-
dc.identifier.urihttps://ir.ymlib.yonsei.ac.kr/handle/22282913/199439-
dc.description.abstractFocal cortical dysplasia (FCD) is particularly likely to affect the frontal lobe. By examining associations between FCD and cytoarchitecture, gene expression and axes of cortical organization, Lee et al. show that frontal lobe vulnerability may reflect relatively early termination of prenatal neurogenesis, as well as aberrant postnatal synaptogenesis. Focal cortical dysplasia (FCD) type II is a highly epileptogenic developmental malformation and a common cause of surgically treated drug-resistant epilepsy. While clinical observations suggest frequent occurrence in the frontal lobe, mechanisms for such propensity remain unexplored. Here, we hypothesized that cortex-wide spatial associations of FCD distribution with cortical cytoarchitecture, gene expression and organizational axes may offer complementary insights into processes that predispose given cortical regions to harbour FCD. We mapped the cortex-wide MRI distribution of FCDs in 337 patients collected from 13 sites worldwide. We then determined its associations with (i) cytoarchitectural features using histological atlases by Von Economo and Koskinas and BigBrain; (ii) whole-brain gene expression and spatiotemporal dynamics from prenatal to adulthood stages using the Allen Human Brain Atlas and PsychENCODE BrainSpan; and (iii) macroscale developmental axes of cortical organization. FCD lesions were preferentially located in the prefrontal and fronto-limbic cortices typified by low neuron density, large soma and thick grey matter. Transcriptomic associations with FCD distribution uncovered a prenatal component related to neuroglial proliferation and differentiation, likely accounting for the dysplastic makeup, and a postnatal component related to synaptogenesis and circuit organization, possibly contributing to circuit-level hyperexcitability. FCD distribution showed a strong association with the anterior region of the antero-posterior axis derived from heritability analysis of interregional structural covariance of cortical thickness, but not with structural and functional hierarchical axes. Reliability of all results was confirmed through resampling techniques. Multimodal associations with cytoarchitecture, gene expression and axes of cortical organization indicate that prenatal neurogenesis and postnatal synaptogenesis may be key points of developmental vulnerability of the frontal lobe to FCD. Concordant with a causal role of atypical neuroglial proliferation and growth, our results indicate that FCD-vulnerable cortices display properties indicative of earlier termination of neurogenesis and initiation of cell growth. They also suggest a potential contribution of aberrant postnatal synaptogenesis and circuit development to FCD epileptogenicity.-
dc.description.statementOfResponsibilityopen-
dc.languageEnglish-
dc.publisherOxford University Press-
dc.relation.isPartOfBRAIN-
dc.rightsCC BY-NC-ND 2.0 KR-
dc.subject.MESHBrain / pathology-
dc.subject.MESHFocal Cortical Dysplasia*-
dc.subject.MESHHumans-
dc.subject.MESHMagnetic Resonance Imaging / methods-
dc.subject.MESHMalformations of Cortical Development* / diagnostic imaging-
dc.subject.MESHMalformations of Cortical Development* / genetics-
dc.subject.MESHMalformations of Cortical Development* / pathology-
dc.subject.MESHReproducibility of Results-
dc.titleMultimodal mapping of regional brain vulnerability to focal cortical dysplasia-
dc.typeArticle-
dc.contributor.collegeCollege of Medicine (의과대학)-
dc.contributor.departmentDept. of Neurology (신경과학교실)-
dc.contributor.googleauthorHyo M Lee-
dc.contributor.googleauthorSeok-Jun Hong-
dc.contributor.googleauthorRavnoor Gill-
dc.contributor.googleauthorBenoit Caldairou-
dc.contributor.googleauthorIrene Wang-
dc.contributor.googleauthorJian-Guo Zhang-
dc.contributor.googleauthorFrancesco Deleo-
dc.contributor.googleauthorDewi Schrader-
dc.contributor.googleauthorFabrice Bartolomei-
dc.contributor.googleauthorMaxime Guye-
dc.contributor.googleauthorKyoo Ho Cho-
dc.contributor.googleauthorCarmen Barba-
dc.contributor.googleauthorSanjay Sisodiya-
dc.contributor.googleauthorGraeme Jackson-
dc.contributor.googleauthorR Edward Hogan-
dc.contributor.googleauthorLily Wong-Kisiel-
dc.contributor.googleauthorGregory D Cascino-
dc.contributor.googleauthorAndreas Schulze-Bonhage-
dc.contributor.googleauthorIscia Lopes-Cendes-
dc.contributor.googleauthorFernando Cendes-
dc.contributor.googleauthorRenzo Guerrini-
dc.contributor.googleauthorBoris Bernhardt-
dc.contributor.googleauthorNeda Bernasconi-
dc.contributor.googleauthorAndrea Bernasconi-
dc.identifier.doi10.1093/brain/awad060-
dc.contributor.localIdA03811-
dc.relation.journalcodeJ00385-
dc.identifier.eissn1460-2156-
dc.identifier.pmid36852571-
dc.subject.keywordMRI-
dc.subject.keywordepilepsy-
dc.subject.keywordfocal cortical dysplasia-
dc.subject.keywordimaging-genetics-
dc.subject.keywordneurodevelopment-
dc.contributor.alternativeNameCho, Kyoo Ho-
dc.contributor.affiliatedAuthor조규호-
dc.citation.volume146-
dc.citation.number8-
dc.citation.startPage3404-
dc.citation.endPage3415-
dc.identifier.bibliographicCitationBRAIN, Vol.146(8) : 3404-3415, 2023-08-
Appears in Collections:
1. College of Medicine (의과대학) > Dept. of Neurology (신경과학교실) > 1. Journal Papers

qrcode

Items in DSpace are protected by copyright, with all rights reserved, unless otherwise indicated.