[HTML][HTML] The landscape of human tissue and cell type specific expression and co-regulation of senescence genes

P Xu, M Wang, W Song, Q Wang, GC Yuan… - Molecular …, 2022 - Springer
Molecular Neurodegeneration, 2022Springer
Background Cellular senescence is a complex stress response that impacts cellular function
and organismal health. Multiple developmental and environmental factors, such as intrinsic
cellular cues, radiation, oxidative stress, oncogenes, and protein accumulation, activate
genes and pathways that can lead to senescence. Enormous efforts have been made to
identify and characterize senescence genes (SnGs) in stress and disease systems.
However, the prevalence of senescent cells in healthy human tissues and the global SnG …
Background
Cellular senescence is a complex stress response that impacts cellular function and organismal health. Multiple developmental and environmental factors, such as intrinsic cellular cues, radiation, oxidative stress, oncogenes, and protein accumulation, activate genes and pathways that can lead to senescence. Enormous efforts have been made to identify and characterize senescence genes (SnGs) in stress and disease systems. However, the prevalence of senescent cells in healthy human tissues and the global SnG expression signature in different cell types are poorly understood.
Methods
This study performed an integrative gene network analysis of bulk and single-cell RNA-seq data in non-diseased human tissues to investigate SnG co-expression signatures and their cell-type specificity.
Results
Through a comprehensive transcriptomic network analysis of 50 human tissues in the Genotype-Tissue Expression Project (GTEx) cohort, we identified SnG-enriched gene modules, characterized SnG co-expression patterns, and constructed aggregated SnG networks across primary tissues of the human body. Our network approaches identified 51 SnGs highly conserved across the human tissues, including CDKN1A (p21)-centered regulators that control cell cycle progression and the senescence-associated secretory phenotype (SASP). The SnG-enriched modules showed remarkable cell-type specificity, especially in fibroblasts, endothelial cells, and immune cells. Further analyses of single-cell RNA-seq and spatial transcriptomic data independently validated the cell-type specific SnG signatures predicted by the network analysis.
Conclusions
This study systematically revealed the co-regulated organizations and cell type specificity of SnGs in major human tissues, which can serve as a blueprint for future studies to map senescent cells and their cellular interactions in human tissues.
Springer