Your browser doesn't support javascript.
loading
Sex specific molecular networks and key drivers of Alzheimer's disease.
Guo, Lei; Cao, Jiqing; Hou, Jianwei; Li, Yonghe; Huang, Min; Zhu, Li; Zhang, Larry; Lee, Yeji; Duarte, Mariana Lemos; Zhou, Xianxiao; Wang, Minghui; Liu, Chia-Chen; Martens, Yuka; Chao, Michael; Goate, Alison; Bu, Guojun; Haroutunian, Vahram; Cai, Dongming; Zhang, Bin.
Afiliación
  • Guo L; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
  • Cao J; Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
  • Hou J; Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
  • Li Y; James J Peters VA Medical Center, Research & Development, Bronx, NY, 10468, USA.
  • Huang M; Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
  • Zhu L; James J Peters VA Medical Center, Research & Development, Bronx, NY, 10468, USA.
  • Zhang L; Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA.
  • Lee Y; Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
  • Duarte ML; James J Peters VA Medical Center, Research & Development, Bronx, NY, 10468, USA.
  • Zhou X; Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
  • Wang M; James J Peters VA Medical Center, Research & Development, Bronx, NY, 10468, USA.
  • Liu CC; Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
  • Martens Y; James J Peters VA Medical Center, Research & Development, Bronx, NY, 10468, USA.
  • Chao M; James J Peters VA Medical Center, Research & Development, Bronx, NY, 10468, USA.
  • Goate A; Department of Neuroscience, Yale University, New Haven, CT, 06510, USA.
  • Bu G; Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
  • Haroutunian V; James J Peters VA Medical Center, Research & Development, Bronx, NY, 10468, USA.
  • Cai D; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
  • Zhang B; Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
Mol Neurodegener ; 18(1): 39, 2023 06 20.
Article en En | MEDLINE | ID: mdl-37340466
ABSTRACT

BACKGROUND:

Alzheimer's disease (AD) is a progressive and age-associated neurodegenerative disorder that affects women disproportionally. However, the underlying mechanisms are poorly characterized. Moreover, while the interplay between sex and ApoE genotype in AD has been investigated, multi-omics studies to understand this interaction are limited. Therefore, we applied systems biology approaches to investigate sex-specific molecular networks of AD.

METHODS:

We integrated large-scale human postmortem brain transcriptomic data of AD from two cohorts (MSBB and ROSMAP) via multiscale network analysis and identified key drivers with sexually dimorphic expression patterns and/or different responses to APOE genotypes between sexes. The expression patterns and functional relevance of the top sex-specific network driver of AD were further investigated using postmortem human brain samples and gene perturbation experiments in AD mouse models.

RESULTS:

Gene expression changes in AD versus control were identified for each sex. Gene co-expression networks were constructed for each sex to identify AD-associated co-expressed gene modules shared by males and females or specific to each sex. Key network regulators were further identified as potential drivers of sex differences in AD development. LRP10 was identified as a top driver of the sex differences in AD pathogenesis and manifestation. Changes of LRP10 expression at the mRNA and protein levels were further validated in human AD brain samples. Gene perturbation experiments in EFAD mouse models demonstrated that LRP10 differentially affected cognitive function and AD pathology in sex- and APOE genotype-specific manners. A comprehensive mapping of brain cells in LRP10 over-expressed (OE) female E4FAD mice suggested neurons and microglia as the most affected cell populations. The female-specific targets of LRP10 identified from the single cell RNA-sequencing (scRNA-seq) data of the LRP10 OE E4FAD mouse brains were significantly enriched in the LRP10-centered subnetworks in female AD subjects, validating LRP10 as a key network regulator of AD in females. Eight LRP10 binding partners were identified by the yeast two-hybrid system screening, and LRP10 over-expression reduced the association of LRP10 with one binding partner CD34.

CONCLUSIONS:

These findings provide insights into key mechanisms mediating sex differences in AD pathogenesis and will facilitate the development of sex- and APOE genotype-specific therapies for AD.
Asunto(s)
Palabras clave

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Enfermedad de Alzheimer Tipo de estudio: Prognostic_studies Límite: Animals / Female / Humans / Male Idioma: En Revista: Mol Neurodegener Año: 2023 Tipo del documento: Article País de afiliación: Estados Unidos

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Enfermedad de Alzheimer Tipo de estudio: Prognostic_studies Límite: Animals / Female / Humans / Male Idioma: En Revista: Mol Neurodegener Año: 2023 Tipo del documento: Article País de afiliación: Estados Unidos