GET THE APP

ACE2-Expressing Endothelial Cells in Aging Mouse Brain
..

Journal of Brain Research

ISSN: 2684-4583

Open Access

Research Article - (2020) Volume 3, Issue 3

ACE2-Expressing Endothelial Cells in Aging Mouse Brain

Su Bin Lim1, Valina L. Dawson1,2,3,4*, Ted M. Dawson1,3,4,5* and Sung-Ung Kang1*
*Correspondence: Valina L. Dawson, Department of Neurology, Johns Hopkins University School of Medicine, USA, Email: Ted M. Dawson, Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA, Email: Sung-Ung Kang, Department of Neuroregeneration and Stem Cell Programs, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA, Email:
1Department of Neuroregeneration and Stem Cell Programs, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
2Department of Physiology, Davis, USA
3Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
4Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
5Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA

Received: 06-Nov-2020 Published: 21-Nov-2020 , DOI: 10.37421/2684-4583.2020.3.116
Citation: Lim,Su Bin, Valina Dawson L, Ted Dawson M and Sung-Ung Kang. "ACE2-Expressing Endothelial Cells in Aging Mouse Brain." J Brain Res 3 (2020):116
Copyright: © 2020 Bin Lim, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Abstract

Angiotensin-Converting Enzyme 2 (ACE2) is a key receptor mediating the entry of SARS-CoV-2 into the host cell. Through a systematic analysis of publicly available mouse brain sc/snRNA-seq data, we found that ACE2 is specifically expressed in small sub-populations of endothelial cells and mural cells, namely pericytes and vascular smooth muscle cells. Further, functional changes in viral mRNA transcription and replication, and impaired blood-brain barrier regulation were most prominently implicated in the aged, ACE2-expressing endothelial cells, when compared to the young adult mouse brains. Concordant EC transcriptomic changes were further found in normal aged human brains. Overall, this work reveals an outline of ACE2 distribution in the mouse brain and identifies putative brain host cells that may underlie the selective susceptibility of the aging brain to viral infection.

Keywords

Brain • Aging • ACE2-expressing endothelial cells • Transcriptomic

Introduction

In addition to the well-known respiratory symptoms, COVID-19 patients suffer from a loss of smell and taste, headache, impaired consciousness, and nerve pain [1]. Raising possibility of virus infiltration in the nervous system, including brain. Despite cases emerging of COVID-19 patients with neurologic manifestations, potential neurotropic mechanisms underlying SARS-CoV-2-mediated entry into the cells of the brain are largely unexplored.

Evidenced by transgenic mice models [2,3]. The evolutionarily-related coronaviruses, such as SARS-CoV and MERS-CoV, can invade the brain by replicating and spreading through the nasal cavity, and possibly olfactory bulbs located in close proximity to the frontal lobes of the brain [4]. Once inside the brain, viruses can harm the brain directly and indirectly by infecting the cells and myelin sheaths, and by activating microglia, which may in turn consume healthy neurons to induce neuroinflammation and neurodegeneration [5].

Many of the observed neurological symptoms observed may in part be explained by a primary vasculopathy and hypercoagulability [6]. As endothelial dysfunction and the resulting clotting are increasingly being observed in patients with severe COVID-19 infection [7-9]. Consistent with these findings, the first pathologic evidence of direct viral infection of the EC and lymphocytic endotheliitis has been found in multiple organs, including lung, heart, kidney and liver, in a series of COVID-19 patients [10].

In contrast, recent clinical findings, including an MRI study [11]. And immunohistochemistry and RT-qPCR analyses [12,13]. Did not observe any signs of encephalitis from postmortem brain examination of COVID-19 patients. Similarly, postmortem analysis of SARS-CoV-2-exposed mice transgenically expressing ACE2 via mouse ACE2 promoter failed to detect the virus in the brain [14]. In light of such controversy regarding neuropathological features, a more comprehensive assessment on the distribution of ACE2 in a cell type-specific manner is required to identify putative brain host cells.

Here, we analyzed publicly available and spatially rich brain RNA-seq datasets to assess ACE2 distribution in mouse brains at the single-cell and single-nuclei level. We found that ACE2 was consistently expressed in small subpopulations of Endothelial Cells (ECs) and mural cells in all the analyzed datasets, in which the impaired blood-brain barrier was further implicated in the aged brains. These findings altogether may hold potential to initiate new avenues of research on specific cells types (EC and vascular mural cells) that remain poorly understood particularly in relation to the aging and viral infection in the brain.

Methods

Mouse brain sc/snRNA-seq data and analysis

Of all the independent studies retrieved with the term “ACE2” from the Single Cell Portal 11 sc/snRNA-seq datasets were derived from (1) adult (young or old) mouse brains, and had (2) author-defined cell type annotations including endothelial cells (EC, PC, or VSMC). A total of 801,658 cells were analyzed in this study, based on the organ (i.e., brain) and species of origin (i.e., mouse), and diseased status (i.e., normal). 2-D tSNE/UMAP plots (colored by cell type) and box plots for cell type-specific ACE2 expression presented in this study were generated by the Single Cell Portal. t-SNE visualization (colored by age) and 25 cell type-specific DE gene lists of the aging mouse brains (T1) were obtained from the advanced interactive data viewer Genes with logGER (Gene Expression Ratio) |>0.1 and FDR<0.05 were defined as DEGs, and were analyzed for functional pathway enrichment using GeneAnalyics.

Human brain bulk RNA-seq data and analysis

De-identified processed human brain bulk RNA-seq data and annotation files for sample attributes and subject phenotypes were obtained from the Genotype-Tissue Expression (GTEx) portal. Data from amygdala (n=88), anterior cingulate cortex (n=107), caudate (basal ganglia) (n=142), cerebellar hemisphere (n=121), cerebellum (n=138), hippocampus (n=123), frontal cortex (BA9) (n=124), cortex (n=137), hypothalamus (n=120), nucleus accumbens (basal ganglia) (n=140), putamen (basal ganglia) (n=121), substantia nigra (n=74) were analyzed in this study. A total of 1,435 samples were divided into two age groups: young (<60) and old (≥ 60 years old). Expression levels (in TPM) were compared between the two age groups for all genes (n=56,200) by t-test with multiple testing correction. The performance of Bonferroni correction and False Discovery Rate (FDR)-Benjamini-Hochberg (BH) procedure was assessed using the RStudio (Version 1.2.5019) base function adjust. The R qvalue package from Bioconductor was used to assess the performance of q-value approach [15]. The R biomaRt package from Bioconductor was used to convert mouse EC DEGs to human gene symbols (hgnc) using getLDS() function [16]. Gene Set Enrichment Analysis (GSEA v4.0.3; [17]. was used to assess EC DEGs (EC up- and down-regulated genes) in GTEx human brain bulk RNA-seq samples. Following parameters were set to run enrichment tests: (1) number of permutations=1,000, (2) collapse/remap to gene symbols=no_collapse, and (3) permutation type=gene_set.

Conclusion

We have compared expression levels (in TPM) of human orthologs of the aged EC DEGs matched in the GTEx data between the young (<60) and old (≥ 60 years) human samples. Of all genes evaluated (n=56,200), 8,215 (15%) showed the expression level significantly different (t-test q-value ≤ 0.05; see Methods) while the expression levels of 37% of EC DEGs were different in the old group from that of the young population, indicating concordant EC transcriptomic changes in normal aged human brains. This conclusion was further supported by Gene Set Enrichment Analysis (GSEA) results, displaying a significant enrichment of the aged (n=141) and young (n=133) mouse EC DEGs in the old (n=814) and young (n=621) human samples, respectively. In conclusion, we have identified specific EC signatures that are functionally important and related to the aging and viral infection in the brain.

Acknowledgements

This work was supported by grants from NIH/NIA AG059686. T.M.D. is the Leonard and Madlyn Abramson Professor in Neurodegenerative Diseases.

Author Contributions

V.L.D., T.M.D., and S.U.K. supervised the project; S.B.L., V.L.D., T.M.D. and S.U.K. formulated the hypothesis; S.B.L. and S.U.K. performed research; S.B.L., V.L.D., T.M.D., and S.U.K. wrote the paper.

Competing Interests

The authors declare no competing interest.

References

Google Scholar citation report
Citations: 2

Journal of Brain Research received 2 citations as per Google Scholar report

Journal of Brain Research peer review process verified at publons

Indexed In

 
arrow_upward arrow_upward