Your browser doesn't support javascript.
loading
Brain Region-Dependent Alternative Splicing of Alzheimer Disease (AD)-Risk Genes Is Associated With Neuropathological Features in AD.
Kim, Sara; Han, Seonggyun; Cho, Soo-Ah; Nho, Kwangsik; Koh, Insong; Lee, Younghee.
Afiliação
  • Kim S; Department of Biomedical Informatics, Hanyang University, Seoul, Korea.
  • Han S; Department of Biomedical Informatics, University of Utah School of Medicine, Salt Lake City, UT, USA.
  • Cho SA; College of Veterinary Medicine, Seoul National University, Seoul, Korea.
  • Nho K; Center for Neuroimaging, Department of Radiology and Imaging Sciences and Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA.
  • Koh I; Department of Biomedical Informatics, Hanyang University, Seoul, Korea.
  • Lee Y; College of Veterinary Medicine, Seoul National University, Seoul, Korea.
Int Neurourol J ; 26(Suppl 2): S126-136, 2022 Nov.
Article em En | MEDLINE | ID: mdl-36503215
ABSTRACT

PURPOSE:

Alzheimer disease (AD) is one of the most complex diseases and is characterized by AD-related neuropathological features, including accumulation of amyloid-ß plaques and tau neurofibrillary tangles. Dysregulation of alternative splicing (AS) contributes to these features, and there is heterogeneity in features across brain regions between AD patients, leading to different severity and progression rates; however, brain region-specific AS mechanisms still remain unclear. Therefore, we aimed to systemically investigate AS in multiple brain regions of AD patients and how they affect clinical features.

METHODS:

We analyzed RNA sequencing (RNA-Seq) data obtained from brain regions (frontal and temporal) of AD patients. Reads were mapped to the hg19 reference genome using the STAR aligner, and exon skipping (ES) rates were estimated as percent spliced in (PSI) by rMATs. We focused on AD-risk genes discovered by genome-wide association studies, and accordingly evaluated associations between PSI of skipped exons in AD-risk genes and Braak stage and plaque density mean (PM) for each brain region. We also integrated whole-genome sequencing data of the ascertained samples with RNA-Seq data to identify genetic regulators of feature-associated ES.

RESULTS:

We identified 26 and 41 ES associated with Braak stage in frontal and temporal regions, respectively, and 10 and 50 ES associated with PM. Among those, 10 were frontal-specific (CLU and NTRK2), 65 temporal-specific (HIF1A and TRPC4AP), and 26 shared ES (APP) that accompanied functional Gene Ontology terms, including axonogenesis in shared-ES genes. We further identified genetic regulators that account for 44 ES (44% of the total). Finally, we present as a case study the systematic regulation of an ES in APP, which is important in AD pathogenesis.

CONCLUSION:

This study provides new insights into brain region-dependent AS regulation of the architecture of AD-risk genes that contributes to AD pathologies, ultimately allowing identification of a treatment target and region-specific biomarkers for AD.
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Tipo de estudo: Etiology_studies / Risk_factors_studies Idioma: En Revista: Int Neurourol J Ano de publicação: 2022 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Tipo de estudo: Etiology_studies / Risk_factors_studies Idioma: En Revista: Int Neurourol J Ano de publicação: 2022 Tipo de documento: Article