Your browser doesn't support javascript.
loading
Single-Cell Transcriptomic Profiling Identifies Molecular Phenotypes of Newborn Human Lung Cells.
Bhattacharya, Soumyaroop; Myers, Jacquelyn A; Baker, Cameron; Guo, Minzhe; Danopoulos, Soula; Myers, Jason R; Bandyopadhyay, Gautam; Romas, Stephen T; Huyck, Heidie L; Misra, Ravi S; Dutra, Jennifer; Holden-Wiltse, Jeanne; McDavid, Andrew N; Ashton, John M; Al Alam, Denise; Potter, S Steven; Whitsett, Jeffrey A; Xu, Yan; Pryhuber, Gloria S; Mariani, Thomas J.
Afiliação
  • Bhattacharya S; Department of Pediatrics, University of Rochester Medical Center, Rochester, NY 14642, USA.
  • Myers JA; Genomic Research Center, University of Rochester Medical Center, Rochester, NY 14642, USA.
  • Baker C; Genomic Research Center, University of Rochester Medical Center, Rochester, NY 14642, USA.
  • Guo M; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45219, USA.
  • Danopoulos S; Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Center, University of California Los Angeles, Los Angeles, CA 90024, USA.
  • Myers JR; Genomic Research Center, University of Rochester Medical Center, Rochester, NY 14642, USA.
  • Bandyopadhyay G; Department of Pediatrics, University of Rochester Medical Center, Rochester, NY 14642, USA.
  • Romas ST; Department of Pediatrics, University of Rochester Medical Center, Rochester, NY 14642, USA.
  • Huyck HL; Department of Pediatrics, University of Rochester Medical Center, Rochester, NY 14642, USA.
  • Misra RS; Department of Pediatrics, University of Rochester Medical Center, Rochester, NY 14642, USA.
  • Dutra J; Clinical & Translational Science Institute, University of Rochester, Rochester, NY 14642, USA.
  • Holden-Wiltse J; Clinical & Translational Science Institute, University of Rochester, Rochester, NY 14642, USA.
  • McDavid AN; Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY 14642, USA.
  • Ashton JM; Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY 14642, USA.
  • Al Alam D; Genomic Research Center, University of Rochester Medical Center, Rochester, NY 14642, USA.
  • Potter SS; Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Center, University of California Los Angeles, Los Angeles, CA 90024, USA.
  • Whitsett JA; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45219, USA.
  • Xu Y; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45219, USA.
  • Pryhuber GS; Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45219, USA.
  • Mariani TJ; Department of Pediatrics, University of Rochester Medical Center, Rochester, NY 14642, USA.
Genes (Basel) ; 15(3)2024 02 26.
Article em En | MEDLINE | ID: mdl-38540357
ABSTRACT
While animal model studies have extensively defined the mechanisms controlling cell diversity in the developing mammalian lung, there exists a significant knowledge gap with regards to late-stage human lung development. The NHLBI Molecular Atlas of Lung Development Program (LungMAP) seeks to fill this gap by creating a structural, cellular and molecular atlas of the human and mouse lung. Transcriptomic profiling at the single-cell level created a cellular atlas of newborn human lungs. Frozen single-cell isolates obtained from two newborn human lungs from the LungMAP Human Tissue Core Biorepository, were captured, and library preparation was completed on the Chromium 10X system. Data was analyzed in Seurat, and cellular annotation was performed using the ToppGene functional analysis tool. Transcriptional interrogation of 5500 newborn human lung cells identified distinct clusters representing multiple populations of epithelial, endothelial, fibroblasts, pericytes, smooth muscle, immune cells and their gene signatures. Computational integration of data from newborn human cells and with 32,000 cells from postnatal days 1 through 10 mouse lungs generated by the LungMAP Cincinnati Research Center facilitated the identification of distinct cellular lineages among all the major cell types. Integration of the newborn human and mouse cellular transcriptomes also demonstrated cell type-specific differences in maturation states of newborn human lung cells. Specifically, newborn human lung matrix fibroblasts could be separated into those representative of younger cells (n = 393), or older cells (n = 158). Cells with each molecular profile were spatially resolved within newborn human lung tissue. This is the first comprehensive molecular map of the cellular landscape of neonatal human lung, including biomarkers for cells at distinct states of maturity.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Perfilação da Expressão Gênica / Pulmão Limite: Animals / Humans / Newborn Idioma: En Revista: Genes (Basel) Ano de publicação: 2024 Tipo de documento: Article País de afiliação: Estados Unidos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Perfilação da Expressão Gênica / Pulmão Limite: Animals / Humans / Newborn Idioma: En Revista: Genes (Basel) Ano de publicação: 2024 Tipo de documento: Article País de afiliação: Estados Unidos