Your browser doesn't support javascript.
loading
Variant to function mapping at single-cell resolution through network propagation.
Yu, Fulong; Cato, Liam D; Weng, Chen; Liggett, L Alexander; Jeon, Soyoung; Xu, Keren; Chiang, Charleston W K; Wiemels, Joseph L; Weissman, Jonathan S; de Smith, Adam J; Sankaran, Vijay G.
Afiliação
  • Yu F; Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
  • Cato LD; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA.
  • Weng C; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
  • Liggett LA; Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
  • Jeon S; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA.
  • Xu K; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
  • Chiang CWK; Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
  • Wiemels JL; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA.
  • Weissman JS; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
  • de Smith AJ; Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA.
  • Sankaran VG; Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
bioRxiv ; 2022 Jan 24.
Article em En | MEDLINE | ID: mdl-35118467
ABSTRACT
With burgeoning human disease genetic associations and single-cell genomic atlases covering a range of tissues, there are unprecedented opportunities to systematically gain insights into the mechanisms of disease-causal variation. However, sparsity and noise, particularly in the context of single-cell epigenomic data, hamper the identification of disease- or trait-relevant cell types, states, and trajectories. To overcome these challenges, we have developed the SCAVENGE method, which maps causal variants to their relevant cellular context at single-cell resolution by employing the strategy of network propagation. We demonstrate how SCAVENGE can help identify key biological mechanisms underlying human genetic variation including enrichment of blood traits at distinct stages of human hematopoiesis, defining monocyte subsets that increase the risk for severe coronavirus disease 2019 (COVID-19), and identifying intermediate lymphocyte developmental states that are critical for predisposition to acute leukemia. Our approach not only provides a framework for enabling variant-to-function insights at single-cell resolution, but also suggests a more general strategy for maximizing the inferences that can be made using single-cell genomic data.

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Idioma: En Ano de publicação: 2022 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Idioma: En Ano de publicação: 2022 Tipo de documento: Article