학술논문

Sex-Stratified Single-Cell RNA-Seq Analysis Identifies Sex-Specific and Cell Type-Specific Transcriptional Responses in Alzheimer’s Disease Across Two Brain Regions
Document Type
article
Source
Molecular Neurobiology. 59(1)
Subject
Biological Sciences
Bioinformatics and Computational Biology
Alzheimer's Disease
Aging
Neurodegenerative
Genetics
Human Genome
Neurosciences
Brain Disorders
Dementia
Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD)
Acquired Cognitive Impairment
Aetiology
2.1 Biological and endogenous factors
1.1 Normal biological development and functioning
Underpinning research
Neurological
Aged
Aged
80 and over
Alzheimer Disease
Entorhinal Cortex
Female
Humans
Male
Neurons
Prefrontal Cortex
Sequence Analysis
RNA
Sex Factors
Single-Cell Analysis
Transcription
Genetic
Transcriptome
Alzheimer's disease
Sex differences
Single-cell
RNA sequencing
Alzheimer’s disease
Psychology
Cognitive Sciences
Neurology & Neurosurgery
Biochemistry and cell biology
Language
Abstract
Alzheimer's disease (AD) is a pervasive neurodegenerative disorder that disproportionately affects women. Since neural anatomy and disease pathophysiology differ by sex, investigating sex-specific mechanisms in AD pathophysiology can inform new therapeutic approaches for both sexes. Previous bulk human brain RNA sequencing studies have revealed sex differences in dysregulated molecular pathways related to energy production, neuronal function, and immune response; however, the sex differences in disease mechanisms are yet to be examined comprehensively on a single-cell level. We leveraged nearly 74,000 cells from human prefrontal and entorhinal cortex samples from the first two publicly available single-cell RNA sequencing AD datasets to perform a case versus control sex-stratified differential gene expression analysis and pathway network enrichment in a cell type-specific manner for each brain region. Our examination at the single-cell level revealed sex differences in AD prominently in glial cells of the prefrontal cortex. In the entorhinal cortex, we observed the same genes and networks to be perturbed in opposing directions between sexes in AD relative to healthy state. Our findings contribute to growing evidence of sex differences in AD-related transcriptomic changes, which can fuel the development of therapies that may prove more effective at reversing AD pathophysiology.