Single cell ‘omic profiles of human aortic endothelial cells in vitro and human atherosclerotic lesions ex vivo reveals heterogeneity of endothelial subtype and response to activating perturbations

  1. The Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ 85721, USA
  2. The Clinical Translational Sciences Graduate Program, The University of Arizona, Tucson, AZ, 85721, USA
  3. Asthma and Airway Disease Research Center, The University of Arizona, Tucson, AZ, 85721, USA

Peer review process

Not revised: This Reviewed Preprint includes the authors’ original preprint (without revision), an eLife assessment, public reviews, and a response from the authors (if available).

Read more about eLife’s peer review process.

Editors

  • Reviewing Editor
    Marcus Seldin
    University of California, Irvine, Irvine, United States of America
  • Senior Editor
    Olujimi Ajijola
    University of California, Los Angeles, Los Angeles, United States of America

Reviewer #1 (Public Review):

Summary:
The manuscript by Adelus and colleagues investigates the snRNA sequencing of endothelial cells isolated from deceased heart donor aortic trimmings. From n=6 donors, the authors have identified 5 distinct endothelial cell (EC) populations. The expression levels of a set of genes are different among the different donors and different EC clusters. Furthermore, treatment with IL-1B, TGFB, or ERGsi decreased the proportion of some of these clusters and increased others, with some migratory and ECM-producing capacity. Another interesting observation in this study is that IL-1B alone induces a shift in the clusters and that is different from the TGFB-induced cells. However, ex vivo analyses showed most of the TGFB-induced population matched the in vitro observations. Another interesting finding of the work is that the authors detected SNPs linked to chromatin accessibility to the set of genes identified within these EC populations.

Strengths:
Overall, the work is intriguing and has some novel aspects to it, especially the link between EC-derived EndMT in culture and comparing that with ex vivo atherosclerotic samples.

Weaknesses:
The experiments are lacking in controls, the purity of the isolation, and the use of multiple donors (deceased hearts) to draw conclusions. The lack of validation of the work is a concern.

Reviewer #2 (Public Review):

This study by Adelus et al. profiled the transcriptome and chromatin accessibility in cultured human aortic endothelial cells (ECs) at single-cell resolution. They also stimulated these cells with EC-activating agents, such as IL1b, TGFB2, or si-EGR, to knock down this master transcription factor in ECs. The results show a subpopulation, EC3, with the highest plasticity and sensitivity to perturbations. The authors also reviewed and meta-analyzed three independent publicly available scRNA-seq datasets, identifying two distinct EC subpopulations. Additionally, they aligned CAD-related SNPs with open chromatin regions in EC subpopulations. This study provides fundamental evidence to enrich our understanding of vascular ECs and highlights potential subpopulations that may contribute to health and diseases. The work exhibits the potential impact in the field. While the manuscript is comprehensive, there are some concerns that should be addressed.

1. My major concern is whether EC4 is derived from ECs. It seems that EC4 showed a lesser reaction to those perturbations and had lower expression levels of EC marker genes. Did the authors evaluate the purity of their isolated HAECs? Please discuss the potential cell lineage mapping of EC4.

2. Although all the donors are de-identified, is there any information about the severity of their vascular impairment, particularly in the case of patient 5, who exhibits the unique EC5?

3. The meta-analysis of the published datasets is comprehensive. The identified EC heterogeneity corresponds to their in vitro data. I am wondering, in terms of transcriptome, is there any similarity between endo1 and EC1/EC2, and also endo2 and EC3/EC4?

4. The in vitro data indicates that EC3 shows the highest plasticity and sensitivity to perturbations, which may act as the major subtype of ECs responding to risk factors. It's very interesting that CAD-related SNPs do not seem to be enriched in EC3. Please discuss this discrepancy.

5. The last sentence in the legend of Figure 1 seems incomplete: 'Module scores are generated for each cell barcode with Seurat function AddModuleScore().'

  1. Howard Hughes Medical Institute
  2. Wellcome Trust
  3. Max-Planck-Gesellschaft
  4. Knut and Alice Wallenberg Foundation