Type 2 and interferon inflammation regulate SARS-CoV-2 entry factor expression in the airway epithelium

Satria P Sajuthi, Peter DeFord, Yingchun Li, Nathan D Jackson, Michael T Montgomery, Jamie L Everman, Cydney L Rios, Elmar Pruesse, James D Nolin, Elizabeth G Plender, Michael E Wechsler, Angel C Y Mak, Celeste Eng, Sandra Salazar, Vivian Medina, Eric M Wohlford, Scott Huntsman, Deborah A Nickerson, Soren Germer, Michael C Zody, Gonçalo Abecasis, Hyun Min Kang, Kenneth M Rice, Rajesh Kumar, Sam Oh, Jose Rodriguez-Santana, Esteban G Burchard, Max A Seibold, Satria P Sajuthi, Peter DeFord, Yingchun Li, Nathan D Jackson, Michael T Montgomery, Jamie L Everman, Cydney L Rios, Elmar Pruesse, James D Nolin, Elizabeth G Plender, Michael E Wechsler, Angel C Y Mak, Celeste Eng, Sandra Salazar, Vivian Medina, Eric M Wohlford, Scott Huntsman, Deborah A Nickerson, Soren Germer, Michael C Zody, Gonçalo Abecasis, Hyun Min Kang, Kenneth M Rice, Rajesh Kumar, Sam Oh, Jose Rodriguez-Santana, Esteban G Burchard, Max A Seibold

Abstract

Coronavirus disease 2019 (COVID-19) is caused by SARS-CoV-2, an emerging virus that utilizes host proteins ACE2 and TMPRSS2 as entry factors. Understanding the factors affecting the pattern and levels of expression of these genes is important for deeper understanding of SARS-CoV-2 tropism and pathogenesis. Here we explore the role of genetics and co-expression networks in regulating these genes in the airway, through the analysis of nasal airway transcriptome data from 695 children. We identify expression quantitative trait loci for both ACE2 and TMPRSS2, that vary in frequency across world populations. We find TMPRSS2 is part of a mucus secretory network, highly upregulated by type 2 (T2) inflammation through the action of interleukin-13, and that the interferon response to respiratory viruses highly upregulates ACE2 expression. IL-13 and virus infection mediated effects on ACE2 expression were also observed at the protein level in the airway epithelium. Finally, we define airway responses to common coronavirus infections in children, finding that these infections generate host responses similar to other viral species, including upregulation of IL6 and ACE2. Our results reveal possible mechanisms influencing SARS-CoV-2 infectivity and COVID-19 clinical outcomes.

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1. ACE2 and TMPRSS2 are expressed…
Fig. 1. ACE2 and TMPRSS2 are expressed by multiple nasal airway cell types.
a UMAP visualization of 8,291 cells derived from a human nasal airway epithelial brushing depicts multiple epithelial and immune cell types identified through unsupervised clustering. b Log Count Per Million (CPM) normalized expression of ACE2 in epithelial and immune cell types. c Log CPM normalized expression of TMPRSS2 in epithelial and immune cell types.
Fig. 2. TMPRSS2 is part of a…
Fig. 2. TMPRSS2 is part of a T2 inflammation-induced mucus secretory network.
a WGCNA identified networks of co-regulated genes related to mucus secretory function (black), T2 inflammation-induced mucus secretory function (pink), and canonical T2 inflammation biomarkers (saddle brown). TMPRSS2 was within the pink network. Select pathway and cell-type enrichments for network genes are shown. Enrichment p-values were obtained from a one-sided hypergeometric test. Benjamini–Hochberg correction was used to control for false discovery rate. b Scatterplot revealing a strong positive correlation between TMPRSS2 expression and summary (eigengene) expression of the T2 inflammatory, mucus secretory network. p-values were obtained from the two-sided Pearson correlation test. c Scatterplot revealing a strong positive correlation between TMPRSS2 expression and summary (eigengene) expression of the canonical T2 inflammation biomarker network. p-values were obtained from the two-sided Pearson correlation test. d Box plots revealing strong upregulation of TMPRSS2 expression among T2-high (n = 364) compared to T2-low (n = 331) subjects. Differential expression testing between T2-high and T2-low groups was performed using a two-sided Wilcoxon test. e Scatterplot revealing a strong negative correlation between ACE2 expression and summary (eigengene) expression of the T2 inflammation mucus secretory network. p-values were obtained from the two-sided Pearson correlation test. f Scatterplot revealing a strong negative correlation between ACE2 expression and summary (eigengene) expression of the canonical T2 inflammation biomarker network. p-values were obtained from the two-sided Pearson correlation test. g Box plots revealing strong downregulation of ACE2 expression among T2-high (n = 364) compared to T2-low (n = 331) subjects. Differential expression testing between T2-high and T2-low groups was performed using a two-sided Wilcoxon test. Box centers give the median, upper and lower box bounds correspond to first and third quartiles and the upper/lower whiskers extend from the upper/lower bounds up to/down from the largest/smallest value, no further than 1.5 × IQR from the upper/lower bound (where IQR is the inter-quartile range). Data beyond the end of whiskers are plotted individually.
Fig. 3. ACE2 and TMPRSS2 are both…
Fig. 3. ACE2 and TMPRSS2 are both regulated by IL-13 in the airway epithelium.
a Experimental schematic detailing the timeline for differentiation of basal airway epithelial cells into a mucociliary airway epithelium and treatment with chronic (10 days) or acute (72 h) IL-13 (10 ng mL−1). b Box plots of count-normalized expression between paired (n = 5 pairs) nasal airway cultures (control/IL-13) revealing strong downregulation of bulk ACE2 expression with IL-13 treatment. Differential expression results are from DESeq2. Benjamini–Hochberg correction was used to control for false discovery rate. c Box plots of count-normalized expression between paired (n = 5 pairs) nasal airway cultures (control/IL-13) revealing strong upregulation of bulk TMPRSS2 expression with IL-13 treatment. Differential expression results are from DESeq2. Benjamini–Hochberg correction was used to control for false discovery rate. d UMAP visualization of 6,969 cells derived from control and IL-13 stimulated tracheal airway ALI cultures depict multiple epithelial cell types identified through unsupervised clustering. e Violin plots of normalized ACE2 expression across epithelial cell types from tracheal airway ALI cultures, stratified by treatment (gray = control, red = IL-13). Differential expression using a two-sided Wilcoxon test was performed between control and IL-13-stimulated cells with significant differences in expression for a cell type indicated by a * (p < 0.05). p-values (left to right) = 0.62; 0.18; 2.8e−4; 0.66; 4.6e−6; 0.08; NA; 0.77; NA; NA; 0.12. f Violin plots of normalized TMPRSS2 expression across epithelial cell types from tracheal airway ALI cultures, stratified by treatment (gray = control, red = IL-13). Differential expression using a two-sided Wilcoxon test was performed between control and IL-13-stimulated cells with significant differences in expression for a cell type indicated by a * (p < 0.05). p-values (left to right) = 9.4e−4; 1.4e−11; 0.51; 2.5e−14; 6.9e−117; 1.5e−38; 2.3e−4; 2.3e−10; 0.46; NA; 0.95. Box centers give the median, upper and lower box bounds correspond to first and third quartiles and the upper/lower whiskers extend from the upper/lower bounds up to/down from the largest/smallest value, no further than 1.5 × IQR from the upper/lower bound (where IQR is the inter-quartile range). Data beyond the end of whiskers are plotted individually.
Fig. 4. ACE2 is an interferon network…
Fig. 4. ACE2 is an interferon network gene regulated by a viral infection.
a Scatterplot revealing a strong positive correlation between ACE2 expression and summary (eigengene) expression of the cytotoxic immune response network (purple). p-values were obtained from a two-sided Pearson correlation test. b Scatterplot revealing a strong positive correlation between ACE2 expression and summary (eigengene) expression of the interferon response network (tan). p-values were obtained from a two-sided Pearson correlation test. c WGCNA analysis identified networks of co-regulated genes related to cytotoxic immune response (purple) and interferon response (tan). ACE2 was within the purple network. Select pathway and cell-type enrichments for network genes are shown. Enrichment p-values were obtained from a one-sided hypergeometric test. Benjamini–Hochberg correction was used to control for false discovery rate. d Box plots of count-normalized expression from GALA II nasal epithelial samples reveal strong upregulation of ACE2 expression among interferon-high (n = 78) compared to interferon-low (n = 617) subjects. Differential expression results are from DESeq2. Benjamini–Hochberg correction was used to control for false discovery rate. e Pie graph depicting the percentage of each type of respiratory virus infection found among GALA II subjects in whom viral reads were found. f Experimental schematic detailing a timeline for differentiation of basal airway epithelial cells into a mucociliary airway epithelium and experimental infection with HRV-A16. g Box plots of count-normalized expression between paired (n = 5 pairs) nasal airway cultures (control/HRV-A16 infected) revealing strong upregulation of ACE2 expression with HRV-A16 infection. Differential expression results are from DESeq2. Benjamini–Hochberg correction was used to control for false discovery rate. h Box plots of count-normalized expression between paired (n = 5 pairs) nasal airway cultures (control/HRV-A16-infected) revealing no effect of HRVA-16 on TMPRSS2 expression. Differential expression results are from DESeq2. Benjamini–Hochberg correction was used to control for false discovery rate. Box centers give the median, upper and lower box bounds correspond to first and third quartiles and the upper/lower whiskers extend from the upper/lower bounds up to/down from the largest/smallest value, no further than 1.5 × IQR from the upper/lower bound (where IQR is the inter-quartile range). Data beyond the end of whiskers are plotted individually.
Fig. 5. Airway surface ACE2 protein is…
Fig. 5. Airway surface ACE2 protein is regulated by IL-13 and viral infection.
ac Immunofluorescence staining of in vitro nasal airway epithelial ALI cultures (derived from a single GALA II asthmatic child) derived from vehicle-treated (a), 5 days IL-13(10 ng mL−1) treated (b), and 24 h HRV-A infected epithelium (c). Representative images of ciliated cells (ACT; red) and ACE2-positive (white) cells. The nuclei were counterstained with DAPI (blue). ACE2 protein was located in the apical compartment and decreased with IL-13 treatment and increased in HRV-A infection. Scale bars: 60 μm. d Zoomed in image of c. e Quantification of apical ACE2-positive cells per 20x field for each condition in the IL-13/HRV mature culture stimulation experiment (n = 1 donor). p-values were obtained from a two-sided t-test. Data are presented as mean values ± SEM. fh Immunofluorescence staining of in vitro nasal airway epithelial ALI cultures from a single child non-asthmatic donor derived from vehicle-treated (f), 21 days IL-13 (10 ng mL−1) treated (g), and 21 days DAPT(5 μM) treated epithelium (h). Representative images of ciliated cells (ACT; red), basal cells (KRT5; green), and ACE2-positive (white) cells. The nuclei were counterstained with DAPI (blue). ACE2 protein located in the apical compartment and decreased in IL-13 treatment and increased in DAPT treatment. Scale bars: 60 μm. i Quantification of apical ACE2-positive cells per 20x field for each condition in the IL-13/DAPT differentiation experiment (n = 1 donor). p-values were obtained from a two-sided t-test. Data are presented as mean values ± SEM. j Immunofluorescence staining of in vivo trachea tissues from a single healthy donor. Representative images of ciliated cells (ACT; red), basal cells (KRT5; green), and ACE2-positive (white) cells. The nuclei were counterstained with DAPI (blue). ACE2 protein located in the apical compartment of ACT positive ciliated cells. Scale bars: 60 μm. All images were captured on the Echo Revolve R4 and images were cropped and resized using Affinity Designer. For each image, brightness and contrasts were uniformly adjusted relative to the brightest feature to balance exposure of each color channel.
Fig. 6. Nasal airway ACE2 and TMPRSS2…
Fig. 6. Nasal airway ACE2 and TMPRSS2 are regulated by eQTL variants.
a Locuszoom plot of ACE2 eQTL signals. The lead eQTL variant (rs181603331) is highlighted with a purple dot. The strength of linkage disequilibrium (LD) between rs181603331 and other variants is discretely divided into five quantiles and mapped into five colors (dark blue, sky blue, green, orange, and red) sequentially from low LD to high LD. b Locuszoom plot of TMPRSS2 eQTL signals. The three independent eQTL variants (rs1475908, rs2838057, rs74659079) and their LD with other variants (r2) are represented by red, blue, and green color gradient, respectively. c Box plots of normalized ACE2 expression among the three genotypes of the lead ACE2 eQTL variant (rs181603331). log2AFC = log2 of the allelic fold change associated with the variant. (n: GG = 654, GT = 8, TT = 3). eQTL p-values were obtained from testing the additive genotype effect on gene expression using the linear regression model implemented in FastQTL. Benjamini–Hochberg correction was used to control for false discovery rate. d Box plots of normalized TMPRSS2 expression among the three genotypes of the lead TMPRSS2 eQTL variant (rs1475908). log2AFC = log2 of the allelic fold change associated with the variant. (n: GG = 432, GA = 218, AA = 31). eQTL p-values were obtained from testing the additive genotype effect on gene expression using the linear regression model implemented in FastQTL. Benjamini–Hochberg correction was used to control for false discovery rate. e Bar plots depicting allele frequencies of the ACE2 eQTL variant rs181603331 and TMPRSS2 eQTL variants (rs1475908, rs2838057, rs74659079) across world populations. Allele frequency data were obtained from gnomAD v2.1.1. Box centers give the median, upper and lower box bounds correspond to first and third quartiles, and the upper/lower whiskers extend from the upper/lower bounds up to/down from the largest/smallest value, no further than 1.5× IQR from the upper/lower bound (where IQR is the inter-quartile range). Data beyond the end of whiskers are plotted individually.
Fig. 7. Host response to CoV strains…
Fig. 7. Host response to CoV strains are similar to other respiratory viruses.
a Box plots revealing a strong and equivalent upregulation of summary (eigengene [Eg]) expression for the interferon response network among ORV- and CoV-infected GALA II subjects (n = 22, 9 respectively), compared to uninfected subjects (n = 582). Differential expression analysis between infected and uninfected groups was performed with a two-sided t-test. b Box plots revealing a strong and equivalent upregulation of summary (eigengene [Eg]) expression for the cytotoxic immune response network among ORV- and CoV-infected GALA II subjects (n = 22, 9 respectively), compared to uninfected subjects (n = 582). Differential expression analysis between infected and uninfected groups was performed with a two-sided t-test. c Scatterplot showing the similarity in log2FC differential expression of genes in ORV (x axis) and CoV (y axis) infected individuals relative to uninfected subjects. The color of the points corresponds to the group in which each gene was identified as significant (FDR < 0.05, absolute log2FC > 0.5). The Pearson correlation coefficient of the significant genes (excluding the “Neither” category) is 0.95. Top enrichment terms for genes that were significantly differentially expressed in both virus infections (blue) are shown. d Top upstream regulators predicted by Ingenuity Pathway Analysis to be regulating the genes that were upregulated in CoV. Enrichment values for these CoV regulators, using the ORV upregulated genes are also shown. Enrichment p-values were obtained from IPA. No multiple comparison adjustment was performed. e Box plots revealing upregulation of IL6 expression in virus-infected individuals (n: CoV+ = 9, ORV+ = 22, non-infected = 582). Differential expression analysis was performed with limma. Benjamini–Hochberg correction was used to control for false discovery rate. f Box plots revealing upregulation of ACE2 expression in virus-infected individuals (n: CoV+ = 9, ORV+ = 22, non-infected=582). Differential expression analysis was performed with limma. Benjamini–Hochberg correction was used to control for false discovery rate. Box centers give the median, upper and lower box bounds correspond to first and third quartiles and the upper/lower whiskers extend from the upper/lower bounds up to/down from the largest/smallest value, no further than 1.5 × IQR from the upper/lower bound (where IQR is the inter-quartile range). Data beyond the end of whiskers are plotted individually.

References

    1. Wang C, Horby PW, Hayden FG, Gao GF. A novel coronavirus outbreak of global health concern. Lancet. 2020;395:470–473. doi: 10.1016/S0140-6736(20)30185-9.
    1. Zhu N, et al. A novel coronavirus from patients with pneumonia in China, 2019. N. Engl. J. Med. 2020;382:727–733. doi: 10.1056/NEJMoa2001017.
    1. Baud, D. et al. Real estimates of mortality following COVID-19 infection. Lancet Infect. Dis.20, 773 (2020).
    1. Du, Y. et al. Clinical features of 85 fatal cases of COVID-19 from Wuhan: a retrospective observational study. Am. J. Respir. Crit. Care Med.201, 1372–1379 (2020).
    1. Zhou F, et al. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. Lancet. 2020;395:1054–1062. doi: 10.1016/S0140-6736(20)30566-3.
    1. CDC. Coronavirus disease 2019 in children—United States, February 12-April 2, 2020. MMWR Morb. Mortal Wkly Rep.69, 422–426 (2020).
    1. Dong, Y. et al. Epidemiology of COVID-19 among children in China. Pediatrics 145, 10.1542/peds.2020-0702 (2020).
    1. Hoffmann, M. et al. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell181, 271–280.e8 (2020).
    1. Goldfarbmuren KC, et al. Dissecting the cellular specificity of smoking effects and reconstructing lineages in the human airway epithelium. Nat. Commun. 2020;11:2485. doi: 10.1038/s41467-020-16239-z.
    1. Montoro DT, et al. A revised airway epithelial hierarchy includes CFTR-expressing ionocytes. Nature. 2018;560:319–324. doi: 10.1038/s41586-018-0393-7.
    1. Plasschaert LW, et al. A single-cell atlas of the airway epithelium reveals the CFTR-rich pulmonary ionocyte. Nature. 2018;560:377–381. doi: 10.1038/s41586-018-0394-6.
    1. Woodruff PG, et al. T-helper Type 2-driven inflammation defines major subphenotypes of asthma. Am. J. Respir. Crit. Care Med. 2009;180:388–395. doi: 10.1164/rccm.200903-0392OC.
    1. Barcelo B, et al. Intracellular cytokine profile of T lymphocytes in patients with chronic obstructive pulmonary disease. Clin. Exp. Immunol. 2006;145:474–479. doi: 10.1111/j.1365-2249.2006.03167.x.
    1. George L, Brightling CE. Eosinophilic airway inflammation: role in asthma and chronic obstructive pulmonary disease. Therap. Adv. Chronic Dis. 2016;7:34–51. doi: 10.1177/2040622315609251.
    1. Chen G, et al. SPDEF is required for mouse pulmonary goblet cell differentiation and regulates a network of genes associated with mucus production. J. Clin. Investig. 2009;119:2914–2924. doi: 10.1172/JCI35314.
    1. Lachowicz-Scroggins ME, et al. Abnormalities in MUC5AC and MUC5B protein in airway mucus in asthma. Am. J. Respiratory Crit. Care Med. 2016;194:1296–1299. doi: 10.1164/rccm.201603-0526LE.
    1. de Lamballerie, C. N. et al. Characterization of cellular transcriptomic signatures induced by different respiratory viruses in human reconstituted airway epithelia. Sci. Rep.9, 11493 (2019).
    1. Steuerman, Y. et al. Dissection of influenza infection in vivo by single-cell RNA sequencing. Cell Syst.6, 679–691 (2018).
    1. Terrier, O. et al. Cellular transcriptional profiling in human lung epithelial cells infected by different subtypes of influenza A viruses reveals an overall down-regulation of the host p53 pathway. Virol. J.8, 285 (2011).
    1. Wesolowska-Andersen A, et al. Dual RNA-seq reveals viral infections in asthmatic children without respiratory illness which are associated with changes in the airway transcriptome. Genome Biol. 2017;18:12. doi: 10.1186/s13059-016-1140-8.
    1. Aguet F, et al. Genetic effects on gene expression across human tissues. Nature. 2017;550:204–217.
    1. Everman JL, et al. Functional genomics of CDHR3 confirms its role in HRV-C infection and childhood asthma exacerbations. J. Allergy Clin. Immunol. 2019;144:962–971. doi: 10.1016/j.jaci.2019.01.052.
    1. Caliskan M, et al. Rhinovirus wheezing illness and genetic risk of childhood-onset asthma. N. Engl. J. Med. 2013;368:1398–1407. doi: 10.1056/NEJMoa1211592.
    1. Poole A, et al. Dissecting childhood asthma with nasal transcriptomics distinguishes subphenotypes of disease. J. Allergy Clin. Immunol. 2014;133:670–678 e612. doi: 10.1016/j.jaci.2013.11.025.
    1. Voehringer D, Reese TA, Huang X, Shinkai K, Locksley RM. Type 2 immunity is controlled by IL-4/IL-13 expression in hematopoietic non-eosinophil cells of the innate immune system. J. Exp. Med. 2006;203:1435–1446. doi: 10.1084/jem.20052448.
    1. Robinson D, et al. Revisiting Type 2-high and type 2-low airway inflammation in asthma: current knowledge and therapeutic implications. Clin. Exp. Allergy. 2017;47:161–175. doi: 10.1111/cea.12880.
    1. Edwards KM, et al. Burden of human metapneumovirus infection in young children. N. Engl. J. Med. 2013;368:633–643. doi: 10.1056/NEJMoa1204630.
    1. Thompson WW, et al. Estimating influenza-associated deaths in the United States. Am. J. Public Health. 2009;99:S225–S230. doi: 10.2105/AJPH.2008.151944.
    1. Nair H, et al. Global burden of acute lower respiratory infections due to respiratory syncytial virus in young children: a systematic review and meta-analysis. Lancet. 2010;375:1545–1555. doi: 10.1016/S0140-6736(10)60206-1.
    1. Weinberg GA, et al. Parainfluenza virus infection of young children: estimates of the population-based burden of hospitalization. J. Pediatr. 2009;154:694–699. doi: 10.1016/j.jpeds.2008.11.034.
    1. Tran, D. et al. Hospitalization for influenza A versus B. Pediatrics138, e20154643 (2016).
    1. Schuster, J. E. et al. Severe enterovirus 68 respiratory illness in children requiring intensive care management. J. Clin. Virol. 70, 77–82 (2015).
    1. Lambert KA, et al. The role of human rhinovirus (HRV) species on asthma exacerbation severity in children and adolescents. J. Asthma. 2018;55:596–602. doi: 10.1080/02770903.2017.1362425.
    1. Mehta P, et al. COVID-19: consider cytokine storm syndromes and immunosuppression. Lancet. 2020;395:1033–1034.
    1. Kesic MJ, Meyer M, Bauer R, Jaspers I. Exposure to ozone modulates human airway protease/antiprotease balance contributing to increased influenza A infection. PLoS ONE. 2012;7:e35108. doi: 10.1371/journal.pone.0035108.
    1. Sungnak W, et al. SARS-CoV-2 entry factors are highly expressed in nasal epithelial cells together with innate immune genes. Nat. Med. 2020;26:681–687. doi: 10.1038/s41591-020-0868-6.
    1. Ziegler CGK, et al. SARS-CoV-2 receptor ACE2 is an interferon-stimulated gene in human airway epithelial cells and is detected in specific cell subsets across tissues. Cells. 2020;181:1016–1035.e1019. doi: 10.1016/j.cell.2020.04.035.
    1. Maughan, E. F. et al. Cell-intrinsic differences between human airway epithelial cells from children and adults. Preprint at (2020).
    1. Peters MC, et al. A transcriptomic method to determine airway immune dysfunction in T2-high and T2-low asthma. Am. J. Respir. Crit. Care Med. 2019;199:465–477. doi: 10.1164/rccm.201807-1291OC.
    1. Advani S, Sengupta A, Forman M, Valsamakis A, Milstone AM. Detecting respiratory viruses in asymptomatic children. Pediatr. Infect. Dis. J. 2012;31:1221–1226. doi: 10.1097/INF.0b013e318265a804.
    1. Jartti T, Jartti L, Peltola V, Waris M, Ruuskanen O. Identification of respiratory viruses in asymptomatic subjects: asymptomatic respiratory viral infections. Pediatr. Infect. Dis. J. 2008;27:1103–1107. doi: 10.1097/INF.0b013e31817e695d.
    1. Singleton RJ, et al. Viral respiratory infections in hospitalized and community control children in Alaska. J. Med. Virol. 2010;82:1282–1290. doi: 10.1002/jmv.21790.
    1. Stelzer-Braid S, et al. Absence of back to school peaks in human rhinovirus detections and respiratory symptoms in a cohort of children with asthma. J. Med. Virol. 2016;88:578–587. doi: 10.1002/jmv.24371.
    1. Lee, J. S. et al. Immunophenotyping of COVID-19 and influenza highlights the role of type I interferons in development of severe COVID-19. Sci. Immunol. 5, eabd1554 (2020).
    1. Neophytou AM, et al. Air pollution and lung function in minority youth with asthma in the GALA II (genes-environments and admixture in Latino Americans) and SAGE II (study of african americans, asthma, genes, and environments) Studies. Am. J. Respir. Crit. Care Med. 2016;193:1271–1280. doi: 10.1164/rccm.201508-1706OC.
    1. Nishimura KK, et al. Early-life air pollution and asthma risk in minority children. The GALA II and SAGE II studies. Am. J. Respir. Crit. Care Med. 2013;188:309–318. doi: 10.1164/rccm.201302-0264OC.
    1. Thakur N, et al. Socioeconomic status and childhood asthma in urban minority youths. The GALA II and SAGE II studies. Am. J. Respir. Crit. Care Med. 2013;188:1202–1209. doi: 10.1164/rccm.201306-1016OC.
    1. Everman JL, Rios C, Seibold MA. Utilization of air-liquid interface cultures as an in vitro model to assess primary airway epithelial cell responses to the type 2 cytokine interleukin-13. Methods Mol. Biol. 2018;1799:419–432. doi: 10.1007/978-1-4939-7896-0_30.
    1. Taliun, D. et al. Sequencing of 53,831 diverse genomes from the NHLBI TOPMed program. Preprint at (2019).
    1. Li, H. Aligning sequence reads, clone sequences and assembly contigs with BWA-MEM. Preprint at (2013).
    1. Jun G, Wing MK, Abecasis GR, Kang HM. An efficient and scalable analysis framework for variant extraction and refinement from population-scale DNA sequence data. Genome Res. 2015;25:918–925. doi: 10.1101/gr.176552.114.
    1. Loh PR, et al. Reference-based phasing using the Haplotype Reference Consortium panel. Nat. Genet. 2016;48:1443–1448. doi: 10.1038/ng.3679.
    1. Jiang H, Lei R, Ding SW, Zhu S. Skewer: a fast and accurate adapter trimmer for next-generation sequencing paired-end reads. BMC Bioinformatics. 2014;15:182. doi: 10.1186/1471-2105-15-182.
    1. Wu TD, Nacu S. Fast and SNP-tolerant detection of complex variants and splicing in short reads. Bioinformatics. 2010;26:873–881. doi: 10.1093/bioinformatics/btq057.
    1. Anders S, Pyl PT, Huber W. HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics. 2015;31:166–169. doi: 10.1093/bioinformatics/btu638.
    1. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15:550. doi: 10.1186/s13059-014-0550-8.
    1. Jackson ND, et al. Single-cell and population transcriptomics reveal pan-epithelial remodeling in type 2-high asthma. Cell Rep. 2020;32:107872. doi: 10.1016/j.celrep.2020.107872.
    1. Langfelder P, Horvath S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinformatics. 2008;9:559. doi: 10.1186/1471-2105-9-559.
    1. Consortium GT, et al. Genetic effects on gene expression across human tissues. Nature. 2017;550:204–213. doi: 10.1038/nature24277.
    1. Bray NL, Pimentel H, Melsted P, Pachter L. Near-optimal probabilistic RNA-seq quantification. Nat. Biotechnol. 2016;34:525–527. doi: 10.1038/nbt.3519.
    1. Robinson MD, McCarthy DJ, Smyth GK. edgeR: a bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26:139–140. doi: 10.1093/bioinformatics/btp616.
    1. Alexander DH, Lange K. Enhancements to the ADMIXTURE algorithm for individual ancestry estimation. BMC Bioinformatics. 2011;12:246. doi: 10.1186/1471-2105-12-246.
    1. Stegle O, Parts L, Durbin R, Winn J. A Bayesian framework to account for complex non-genetic factors in gene expression levels greatly increases power in eQTL studies. PLoS Comput Biol. 2010;6:e1000770. doi: 10.1371/journal.pcbi.1000770.
    1. Ongen H, Buil A, Brown AA, Dermitzakis ET, Delaneau O. Fast and efficient QTL mapper for thousands of molecular phenotypes. Bioinformatics. 2016;32:1479–1485. doi: 10.1093/bioinformatics/btv722.
    1. Delaneau O, et al. A complete tool set for molecular QTL discovery and analysis. Nat. Commun. 2017;8:15452. doi: 10.1038/ncomms15452.
    1. Mohammadi P, Castel SE, Brown AA, Lappalainen T. Quantifying the regulatory effect size of cis-acting genetic variation using allelic fold change. Genome Res. 2017;27:1872–1884. doi: 10.1101/gr.216747.116.
    1. Bushnell B, Rood J, Singer E. BBMerge—accurate paired shotgun read merging via overlap. PLoS ONE. 2017;12:e0185056. doi: 10.1371/journal.pone.0185056.
    1. Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nat. Methods. 2012;9:357–359.
    1. Nurk S, Meleshko D, Korobeynikov A, Pevzner PA. metaSPAdes: a new versatile metagenomic assembler. Genome Res. 2017;27:824–834. doi: 10.1101/gr.213959.116.
    1. Li H, et al. The sequence alignment/map format and SAMtools. Bioinformatics. 2009;25:2078–2079. doi: 10.1093/bioinformatics/btp352.
    1. Altschul SF, Gish W, Miller W, Myers EW, Lipman DJ. Basic local alignment search tool. J. Mol. Biol. 1990;215:403–410. doi: 10.1016/S0022-2836(05)80360-2.
    1. Ritchie ME, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43:e47. doi: 10.1093/nar/gkv007.
    1. Han B, Eskin E. Random-effects model aimed at discovering associations in meta-analysis of genome-wide association studies. Am. J. Hum. Genet. 2011;88:586–598. doi: 10.1016/j.ajhg.2011.04.014.
    1. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc. Ser. 1995;57:289–300.
    1. Chen EY, et al. Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinformatics. 2013;14:128. doi: 10.1186/1471-2105-14-128.
    1. Travaglini, K. J. et al. A molecular cell atlas of the human lung from single cell RNA sequencing. Preprint at (2020).
    1. Butler A, Hoffman P, Smibert P, Papalexi E, Satija R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 2018;36:411–420. doi: 10.1038/nbt.4096.
    1. Hafemeister C, Satija R. Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression. Genome Biol. 2019;20:296. doi: 10.1186/s13059-019-1874-1.
    1. Waltman L, Van Eck NJ. A smart local moving algorithm for large-scale modularity-based community detection. Eur. Phys. J. B. 2013;86:471. doi: 10.1140/epjb/e2013-40829-0.
    1. McInnes, L. & Healy, J. Uniform manifold approximation and projection for dimension reduction. Preprint at (2018).
    1. Kim D, Langmead B, Salzberg SL. HISAT: a fast spliced aligner with low memory requirements. Nat. Methods. 2015;12:357–360. doi: 10.1038/nmeth.3317.
    1. Stuart T, et al. Comprehensive integration of single-cell data. Cell. 2019;177:1888–1902. doi: 10.1016/j.cell.2019.05.031.

Source: PubMed

3
Tilaa