Plasma gradient of soluble urokinase-type plasminogen activator receptor is linked to pathogenic plasma proteome and immune transcriptome and stratifies outcomes in severe COVID-19 ==================================================================================================================================================================================== * Jafar Sarif * Deblina Raychaudhuri * Ranit D’Rozario * Purbita Bandopadhyay * Praveen Singh * Priyanka Mehta * Md. Asmaul Hoque * Bishnu Prasad Sinha * Manoj Kushwaha * Shweta Sahni * Priti Devi * Partha Chattopadhyay * Shekhar Ranjan Paul * Yogiraj Ray * Kausik Chaudhuri * Sayantan Banerjee * Debajyoti Majumdar * Bibhuti Saha * Biswanath Sharma Sarkar * Prasun Bhattacharya * Shilpak Chatterjee * Sandip Paul * Pramit Ghosh * Rajesh Pandey * Shantanu Sengupta * Dipyaman Ganguly ## ABSTRACT Disease caused by SARS-CoV-2 coronavirus (COVID-19) has resulted in significant morbidity and mortality world-wide. A systemic hyper-inflammation characterizes the severe COVID-19 disease often associated with acute respiratory distress syndrome (ARDS). Blood biomarkers capable of risk stratification are of great importance in effective triage and critical care of severe COVID-19 patients. In the present study we report higher plasma abundance of soluble urokinase-type plasminogen activator receptor (sUPAR), expressed by an abnormally expanded circulating myeloid cell population, in severe COVID-19 patients with ARDS. Plasma sUPAR level was found to be linked to a characteristic proteomic signature of plasma, linked to coagulation disorders and complement activation. Receiver operator characteristics curve analysis identified a cut-off value of sUPAR at 1996.809 pg/ml that could predict survival in our cohort (Odds ratio: 2.9286, 95% confidence interval 1.0427-8.2257). Lower sUPAR level than this threshold concentration was associated with a differential expression of the immune transcriptome as well as favourable clinical outcomes, both in terms of survival benefit (Hazard ratio: 0.3615, 95% confidence interval 0.1433-0.912) and faster disease remission in our patient cohort. Thus we identified sUPAR as a key pathogenic circulating molecule linking systemic hyperinflammation to the hypercoagulable state and stratifying clinical outcomes in severe COVID-19 patients with ARDS. Key words * Soluble UPAR * COVID-19 * PLAUR * Prognosis * Myeloid cells * ARDS * Proteomics ## INTRODUCTION The ongoing pandemic caused by the severe acute respiratory syndrome causing coronavirus 2 (SARS-COV-2) has resulted in close to 175 million documented infections and more than 3.7 million deaths. The respiratory disease caused by SARS-COV-2 or COVID-19 at times progresses to acute respiratory distress syndrome (ARDS), often with fatal outcomes in some patients [1]. Severe COVID-19 is characterized by a hyper-inflammation, the key manifestations being a systemic cytokine deluge and an abnormal myeloid expansion among circulating immune cells [2-5]. In addition to the hyper-inflammation, a number of patients with severe COVID-19 also present with intravascular coagulation as well as abnormal complement activation [6-9]. Thus the systemic cytokine surge, a hyper-coagulable state and tissue damage mediated by complement activation are the three established pathogenic triads in these patients. These also offer the possibility of identifying circulating factors with predictive potential for disease outcomes. Risk-stratifying biomarkers, which can be probed early enough in severe COVID-19 patients, can be useful as pre-assessors for effective triage or intensive care in low-resource settings. Previous single cell RNA sequencing (scRNAseq) studies in severe COVID-19 revealed abnormally expanded circulating myeloid cell compartment as well as an enriched expression of urokinase type plasminogen activator receptor (PLAUR gene, expressing UPAR protein) [3,4]. UPAR (or CD87) is a glycosylphosphatidylinositol (GPI)-anchored receptor present on the surface of various cells, including immune cells including monocytes, macrophages and neutrophils. Cell surface UPAR binds UPA, and transforms plasminogen into plasmin, which in turn initiates a proteolytic cascade to degrade the components of the ECM [10]. UPAR also lies within the complex regulatory network of the coagulation cascade as well as complement activation and complement mediated pathogen or host cell clearance [11-13]. Increased plasma abundance of soluble UPAR has been documented widely in chronic inflammatory contexts [6], thus it is also proposed to be a potential pathogenic molecule involved in the acute systemic hyper-inflammation in COVID-19 [10, 14]. In a cohort of COVID-19 patients from India, which was originally recruited for a randomised control trial on convalescent plasma therapy, we found significantly high plasma levels of soluble UPAR in severe COVID-19 patients early in the course of severity, which correlated with an expanded myeloid cell compartment in circulation. A characteristic proteomics signature of activation of coagulation cascade as well as complement activation was found to be associated with higher plasma concentrations of UPAR, as were specific immune related pathways enriched in peripheral blood transcriptome analysis. Finally we found that patients, suffering from ARDS in severe COVID-19 but with plasma levels of sUPAR below a computed cut-off value, registered significantly more favourable disease outcomes. ## RESULTS ### Expansion of circulating CD11c + HLA-DR— myeloid cells expressing sUPAR in severe COVID-19 patients COVID-19 patients with mild symptoms (N=8, Age=41.5±18.95 years) or ARDS (N=77, Age =61±11.86 years) were recruited at the ID & BG Hospital, Kolkata, India (Supplemental table 1). Peripheral blood sampling was done on the day of enrolment with due ethical approval from the institutional review board. Frequency of circulating CD11c+HLA-DR-- myeloid cells was assessed by flow cytometry. On comparing relative abundance of circulating CD11c+HLA-DR\---| proinflammatory macrophages between the COVID-19 patients with mild diseases and patients who progressed to ARDS, we found a prominent expansion of these cells in patients suffering from ARDS (Figure 1A, B), as has been reported by a number of previous studies [4-6]. We found no correlation between the abundance of these cells in circulation in ARDS patients and the plasma levels of most of the cytokines (among the 36 selected based on detectable levels in at least 70% of the ARDS patients in the cohort, data not shown), except for Eotaxin (Pearson R=0.3725, P=0.0029). HGF (Pearson R=0.2503, P=0.0497) and IL-1α (Pearson R=0.2588, P=0.0423) (Figure 1C-E). ![Fig. 1.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/06/21/2021.06.19.21259125/F1.medium.gif) [Fig. 1.](http://medrxiv.org/content/early/2021/06/21/2021.06.19.21259125/F1) Fig. 1. Expansion of circulating CD11c+HLA-DR- myeloid cell subset in severe COVID-19. (A) Representative flow cytometry plots for gating of circulating CD11c+HLA-DR- myeloid cells from COVID-19 patients with either mild disease or ARDS. (B) Violin plots showing frequency of circulating CD11c+HLA-DR-myeloid cells compared between COVID-19 patients with either mild disease or ARDS. Mann Whitney test was performed. (C) - (E) Correlation between the frequency of circulating CD11c+HLA-DR-myeloid cells and plasma level of the cytokines Eotaxin (C), HGF (D) and IL-1α (E) is plotted. Spearman ρ values are shown, **P <0.005, *P<0.05. Interestingly, a recent scRNAseq study reported an enriched expression of PLAUR, the gene for UPAR, in the similarly expanded myeloid cell compartment in severe COVID-19 [3]. It intrigued us to explore if the circulating CD11c+HLA-DR-myeloid cells in severe COVID-19 patients are enriched for UPAR expression. To this end we analyzed two public datasets on scRNAseq, one done on whole blood (GSE163668, Figure 2A, B) and the other on cells in the bronchoalveolar lavage fluid (GSE145926, Figure 2C, D) from COVID-19 patients with different disease states [15, 16]. We found that the CD11chighHLA-DRlow myeloid cell subsets were highly abundant in patients with severe COVID-19 both in the circulation as well as in the airways on these analyses as well as noted a highly enriched expression of PLAUR in these CD11chighHLA-DRlow myeloid cells (Figure 2B, D). ![Fig. 2.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/06/21/2021.06.19.21259125/F2.medium.gif) [Fig. 2.](http://medrxiv.org/content/early/2021/06/21/2021.06.19.21259125/F2) Fig. 2. Reanalysis of single cell sequencing data to confirm myeloid sourcing of sUPAR. (A) and (B) Analysis of scRNAseq data (GSE163668) to compare frequency of peripheral blood CD11chighHLA-DRlowcells and expression of PLAUR in them between patients with mild (left panel) or severe (right panel) COVID-19 disease. (C) and (D) Analysis of scRNAseq data (GSE GSE145926) to compare frequency of CD11chighHLA-DRlowcells in the bronchoalveolar lavage fluid and expression of PLAUR in them between patients with mild (left panel) or severe (right panel) COVID-19 disease. In both cases, three smaller plots on the left show distribution of expression of ITGAX (gene for CD11c), HLA-DRA (gene for HLA-DR) and PLAUR (gene for UPAR) among all cells. The bigger plot on the right shows expression of PLAUR among CD11chighHLA-DRlowcells. ### Higher plasma abundance of sUPAR in severe COVID-19 is linked to systemic cytokine surge and TNFα-activated myeloid cells Next we measured the level of sUPAR in plasma samples from the patients in our cohort and found a significant correlation between the abundance of the circulating CD11c+HLA-DR- myeloid cells and plasma sUPAR levels (Figure 3A). Moreover, a significantly higher abundance of sUPAR was noted in patients who have progressed to ARDS, compared to patients with milder symptoms (Figure 3B). The plasma level of sUPAR had no relationship with either the viral load of the ARDS patients at the time of plasma sampling (Figure 3C) or the neutralizing antibody content of their plasma (Figure 3D). Age or gender of the patients also did not show any relationship with the plasma levels of sUPAR (Figure 3 E, F). ![Fig. 3.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/06/21/2021.06.19.21259125/F3.medium.gif) [Fig. 3.](http://medrxiv.org/content/early/2021/06/21/2021.06.19.21259125/F3) Fig. 3. Increased plasma abundance of soluble UPAR and its cellular source in severe COVID-19. (A) Correlation between frequency of circulating CD11c+HLA-DR--myeloid cells and plasma levels of sUPAR in COVID-19 patients. (B) Plasma level of sUPAR compared between COVID-19 patients with either mild disease or ARDS. (C) No correlation between average CT values for two SARS-CoV-2 genes and the plasma concentration of sUPAR. (D) No correlation between percent neutralization function of patient plasma (derived from a surrogate assay measuring neutralization of receptor binding protein and ACE2 binding) and the plasma concentration of sUPAR. (E) No correlation between age of patients with ARDS and the plasma level of sUPAR. (F) Comparison between male and female patients with ARDS for the plasma level of sUPAR. Mann Whitney test was performed. (G) Corr plot showing mutual correlation between the plasma levels of 36 cytokines and soluble UPAR in COVID-19 patients with ARDS. The dots are color-coded and size-scaled for the Spearman ρ values for individual correlations, significance levels for correlation between plasma levels of sUPAR and individual cytokines are noted as superscripts on cytokine names, \**\*|\*P<0.0001, \***|99% peptide confidence threshold, 1% peptide FDR. XIC extraction window was set to 10 min with 75 ppm XIC Width. All information was exported in the form of MarkerView (.mrkw) files. In MarkerView 1.2.1 (SCIEX), data normalization was performed using total area sum normalization and exported to excel. Proteomics data are submitted to the PRIDE database [33]. ### Receiver operator characteristic curve The ‘cutpointr’ package in R was used to generate the receiver operating characteristic (ROC) curve and calculate the corresponding AUC value for determining the suitability and dependability of sUPAR content in the plasma of ARDS Covid patients as an indicator of their survival. The optimum threshold sUPAR value which can be used to classify survival outcome in these patients with maximum sensitivity plus specificity was also determined. ### RNA-Seq library preparation and sequencing The RNASeq library was prepared using Illumina TruSeq Stranded Total RNA Library Prep Kit with Ribo-Zero Gold as per TruSeq Stranded Total RNA Reference Guide. We started with 250ng of the total RNA. Briefly, the cytoplasmic and mitochondrial rRNA was depleted from the total RNA, the remaining RNA was purified, fragmented and primed for cDNA synthesis. Subsequently, double stranded cDNA (ds cDNA) was synthesised, the 3’ end of the ds cDNA was adenylated to provide an overhang for adapter ligation. The IDT for Illumina - TruSeq RNA UD Indexes was used for indexing the samples to allow multiplexing, and then finally amplified and purified to enrich the adapter ligated library. The final library was quantified using Qubit™ dsDNA High Sensitivity Assay Kit (Catalog number: Q32851) and the library size was determined using Agilent High Sensitivity DNA Kit (Catalog number: 5067-4626) on Agilent Bioanalyzer 2100 platform. For sequencing, the individual library was diluted to 4nM and libraries were pooled. The pooled library was denatured using 0.2 N NaOH, and neutralized with 200mM Tris-HCl, pH 7.0. The libraries were sequenced on a Illumina NextSeq 550 platform, using high output Kit v2.5 (300 Cycles), at a final library concentration of 1.6 pM. (NextSeq 500 and NextSeq 550 SequencingSystems: Denature and Dilute Libraries Guide; Document # 15048776 v16). ### RNA-seq data processing Filtered fastq files were processed using Salmon v1.4.0 which provides fast and bias-aware quantification of transcript expression [34]. The mapping-based mode of Salmon was used for quantification [35]. The reference transcriptome (Ensembl GRCh38, release 103) was used for indexing and quantification of the individual genes.The quantification was performed on the full transcriptome and gene-level TPM-values(transcripts per million) were computed based on the effective length of the transcripts.The TPM values are normalised for the gene length and sequencing depth and used for further analysis of differentially expressed genes. ### RNA-seq data analysis TPM values were analysed using the online MeV software [36]. The Limma tool [37] was used to find out the differentially expressed genes (DEGs) between the 3 different groups having 3 patients each, categorised according to the concentration of sUPAR in their plasma as described in the figure legend. The list of DEGs (transcripts) (p<=0.05) provided by Limma was divided into two groups-(1) upregulated genes (having all differentially expressed transcripts upregulated) and (2) downregulated genes (having all differentially expressed transcripts downregulated) before being entered into the online NetworkAnalyst software [38] to obtain the list of enriched pathways (p<=0.05) from the Reactome database, separately for upregulated and downregulated genes. Moreover, the genes specifically implicated in each of the pathways was also obtained. ### Correlation matrix generation and visualisation A matrix of Spearman correlation coefficients indicating the degree and directionality of association between the concentrations of the indicated parameters in plasma of COVID-19 patients was generated using the ‘Hmisc’ package in R. For visualisation of the matrix, the ‘corrplot’ package in R was used. ### Statistics All statistical analyses, as depicted in the results as well in appropriate figures and their legends, were performed using Graphpad Prism 8 or in some cases using R. In all cases Spearman correlation analysis and Mann Whitney test was performed unless otherwise stated. Primary outcomes of survival and disease remission (in terms of time till discharge from hospital) were compared between the two arms using Kaplan Meier Curve analysis - Mantel-Haenszel Hazard Ratio was calculated and statistical significance was tested by Mantel-Cox log rank test. ## Supporting information Supplemental [[supplements/259125_file11.pdf]](pending:yes) ## Data Availability The mass spectrometry proteomics data have been deposited to the ProteomeXchange Consortium via the PRIDE partner repository with the dataset identifier PXD026510. Detailed pathway enrichment data derived from the transcriptome analysis are depicted in supplemental table 4. Codes for reanalysis of the single cell RNA sequencing data are provided in supplemental information. All other data pertaining to the manuscript are available with the corresponding authors. ## Author contributions J.S., R.D., P.B., M.A.H. and B.P.S. performed the ELISA, multiplex cytokine assay and flow cytometry; R.D., P.B. and S.C. did flow cytometric analysis; S.R.P., Y.R., K.C., S.B., D.M. B.S., B.S.S and P.B. recruited patients and participated in the clinical management of the patients; P.S., M.K. and S.S. performed the mass spectrometric proteomics; P.M., S.S, P.D., P.C. and R.P. performed the next generation sequencing and analysis; D.R.C. performed scRNAseq, RNAseq and ROC analysis; S.P. and P.G. helped with data analysis; D.G. conceptualized the study; D.G. and S.S. designed the experiments; D.G. wrote the manuscript. ## Funding The study was supported by grant no. MLP-129 from Council of Scientific and Industrial Research (CSIR), India, to D.G., who is also supported by a Swarnajayanti Fellowship grant from Department of Science & Technology, Government of India. The RNA-Seq was funded by the grants from CSIR (MLP-2005), Fondation Botnar (CLP-0031), IUSSTF (CLP-0033) and Intel (CLP-0034). J.S., R.D. and M.A.H. are supported by research fellowships from University Grants Commission, India. P.B. and P.S. are supported by Senior Research Fellowship from CSIR. P.M. and S.S. are supported by MLP-2005. P.D. and P.C. acknowledge CSIR for their Junior Research Fellowship. ## Conflicts of Interest Authors declare no conflict of interest. ## Data availability The mass spectrometry proteomics data have been deposited to the ProteomeXchange Consortium via the PRIDE partner repository with the dataset identifier PXD026510. Detailed pathway enrichment data derived from the transcriptome analysis are depicted in supplemental table 4. Codes for reanalysis of the single cell RNA sequencing data are provided in supplemental information. * Received June 19, 2021. * Revision received June 19, 2021. * Accepted June 21, 2021. * © 2021, Posted by Cold Spring Harbor Laboratory The copyright holder for this pre-print is the author. All rights reserved. The material may not be redistributed, re-used or adapted without the author's permission. ## References 1. 1.WHO Working Group on the Clinical Characterisation and Management of COVID-19 infection. A minimal common outcome measure set for COVID-19 clinical research. Lancet Infect. Dis. 20, e192–e197 (2020) [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S1473-3099(20)30483-7&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32539990&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F06%2F21%2F2021.06.19.21259125.atom) 2. 2.Bandopadhyay P et al. Nature and dimensions of the systemic hyper-inflammation and its attenuation by convalescent plasma in severe COVID-19. J Infect Dis. 2021 Jan 12:jiab010. 3. 3.Schulte-Schrepping J et al. Severe COVID-19 Is Marked by a Dysregulated Myeloid Cell Compartment. Cell. 2020 Sep 17;182(6):1419–1440.e23. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F06%2F21%2F2021.06.19.21259125.atom) 4. 4.Silvin A et al. Elevated Calprotectin and Abnormal Myeloid Cell Subsets Discriminate Severe from Mild COVID-19. Cell. 2020 Sep 17;182(6):1401–1418.e18. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F06%2F21%2F2021.06.19.21259125.atom) 5. 5.Combes AJ et al. Global absence and targeting of protective immune states in severe COVID-19. Nature. 2021 Mar;591(7848):124–130. 6. 6.Zhang Y et al. Coagulopathy and Antiphospholipid Antibodies in Patients with Covid-19. N Engl J Med. 2020 Apr 23;382(17):e38. doi: 10.1056/NEJMc2007575. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMc2007575&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32268022&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F06%2F21%2F2021.06.19.21259125.atom) 7. 7.Sinkovits G et al. Complement Overactivation and Consumption Predicts In-Hospital Mortality in SARS-CoV-2 Infection. Front Immunol. 2021 Mar 25;12:663187. doi: 10.3389/fimmu.2021.663187. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3389/fimmu.2021.663187&link_type=DOI) 8. 8.Ma L et al. Increased complement activation is a distinctive feature of severe SARS-CoV-2 infection. bioRxiv. 2021 Feb 23;2021.02.22.432177. doi: 10.1101/2021.02.22.432177. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiYmlvcnhpdiI7czo1OiJyZXNpZCI7czoxOToiMjAyMS4wMi4yMi40MzIxNzd2MSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIxLzA2LzIxLzIwMjEuMDYuMTkuMjEyNTkxMjUuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 9. 9.Yan B et al. SARS-CoV-2 drives JAK1/2-dependent local complement hyperactivation. Sci Immunol. 2021 Apr 7;6(58):eabg0833. doi: 10.1126/sciimmunol.abg0833. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6ImltbXVub2xvZ3kiO3M6NToicmVzaWQiO3M6MTM6IjYvNTgvZWFiZzA4MzMiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMS8wNi8yMS8yMDIxLjA2LjE5LjIxMjU5MTI1LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 10. 10.D’Alonzo D, De Fenza M, Pavone V. COVID-19 and pneumonia: a role for the uPA/uPAR system. Drug Discov Today. 2020 Aug;25(8):1528–1534. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.drudis.2020.06.013&link_type=DOI) 11. 11.Del Rosso M et al. The plasminogen activation system in inflammation. Front Biosci. 2008 May 1;13:4667–86. doi: 10.2741/3032. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.2741/3032&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=18508538&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F06%2F21%2F2021.06.19.21259125.atom) 12. 12.Del Rosso M et al. The urokinase receptor system, a key regulator at the intersection between inflammation, immunity, and coagulation. Curr Pharm Des. 2011;17(19):1924–43. doi: 10.2174/138161211796718189. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.2174/138161211796718189&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21711238&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F06%2F21%2F2021.06.19.21259125.atom) 13. 13.Pliyev BK, Arefieva TI, Menshikov MY. Urokinase receptor (uPAR) regulates complement receptor 3 (CR3)-mediated neutrophil phagocytosis. Biochem Biophys Res Commun. 2010 Jun 25;397(2):277–82. doi: 10.1016/j.bbrc.2010.05.100. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.bbrc.2010.05.100&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20580686&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F06%2F21%2F2021.06.19.21259125.atom) 14. 14.Rovina N et al. Soluble urokinase plasminogen activator receptor (suPAR) as an early predictor of severe respiratory failure in patients with COVID-19 pneumonia. Crit Care. 2020 Apr 30;24(1):187. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/sl3054-020-02897-4&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32354367&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F06%2F21%2F2021.06.19.21259125.atom) 15. 15.Combes AJ et al. Global absence and targeting of protective immune states in severe COVID-19. Nature. 2021 Mar;591(7848):124–130. doi: 10.1038/s41586-021-03234-7. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41586-021-03234-7&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=33494096&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F06%2F21%2F2021.06.19.21259125.atom) 16. 16.Liao M et al. Single-cell landscape of bronchoalveolar immune cells in patients with COVID-19. Nat Med. 2020 Jun;26(6):842–844. doi: 10.1038/s41591-020-0901-9. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41591-020-0901-9&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32398875&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F06%2F21%2F2021.06.19.21259125.atom) 17. 17.Zhang F et al. IFN-γ and TNF-α drive a CXCL10+ CCL2+ macrophage phenotype expanded in severe COVID-19 lungs and inflammatory diseases with tissue inflammation. Genome Med. 2021 Apr 20;13(1):64. doi: 10.1186/s13073-021-00881-3. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/s13073-021-00881-3&link_type=DOI) 18. 18.Meiners, S., Eickelberg, O., Königshoff, M. Hallmarks of the ageing lung. Eur. Respir. J. 2015; 45, 807–827. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiZXJqIjtzOjU6InJlc2lkIjtzOjg6IjQ1LzMvODA3IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjEvMDYvMjEvMjAyMS4wNi4xOS4yMTI1OTEyNS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 19. 19.Sharma, G., Goodwin, J. Effect of aging on respiratory system physiology and immunology. Clin. Interv. Aging. 2006; 1, 253–260. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.2147/ciia.2006.1.3.253&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=18046878&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F06%2F21%2F2021.06.19.21259125.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000208238100008&link_type=ISI) 20. 20.Ray Y et al. Clinical and immunological benefits of convalescent plasma therapy in severe COVID-19: insights from a single center open label randomised control trial. medRxiv. 2020 Nov 29; 2020.11.25.20237883v1. 21. 21.Yoon HA et al. Treatment of Severe COVID-19 with Convalescent Plasma in the Bronx, NYC. medRxiv. 2020 Dec 4;2020.12.02.20242909. 22. 22.Pyrkov TV et al. Longitudinal analysis of blood markers reveals progressive loss of resilience and predicts human lifespan limit. Nat Commun. 2021 May 25;12(1):2765. doi: 10.1038/s41467-021-23014-1. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41467-021-23014-1&link_type=DOI) 23. 23.Matsumoto H et al. Decreased antithrombin activity in the early phase of trauma is strongly associated with extravascular leakage, but not with antithrombin consumption: a prospective observational study. Thromb J. 2018 Aug 1;16:17. doi: 10.1186/s12959-018-0171-7. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/s12959-018-0171-7&link_type=DOI) 24. 24.Lazarian G et al. Autoimmune haemolytic anaemia associated with COVID-19 infection. Br J Haematol. 2020 Jul;190(1):29–31. doi: 10.1111/bjh.16794. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/bjh.16794&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F06%2F21%2F2021.06.19.21259125.atom) 25. 25.Jacobs J, Eichbaum Q. COVID-19 associated with severe autoimmune hemolytic anemia. Transfusion. 2021 Feb;61(2):635–640. doi: 10.1111/trf.16226. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/trf.16226&link_type=DOI) 26. 26.Jawed M, Hart E, Saeed M. Haemolytic anaemia: a consequence of COVID-19. BMJ Case Rep. 2020 Dec 10;13(12):e238118. doi: 10.1136/bcr-2020-238118. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1136/bcr-2020-238118&link_type=DOI) 27. 27.Gussen H et al. Neutrophils are a main source of circulating suPAR predicting outcome in critical illness. J Intensive Care. 2019 Apr 27;7:26. 28. 28.Gumus A et al. Soluble urokinase-type plasminogen activator receptor is a novel biomarker predicting acute exacerbation in COPD. Int J Chron Obstruct Pulmon Dis. 2015 Feb 13;10:357–65. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.2147/COPD.S77654&link_type=DOI) 29. 29.Tsai PK et al. Plasma Soluble Urokinase-Type Plasminogen Activator Receptor Level as a Predictor of the Severity of Community-Acquired Pneumonia. Int J Environ Res Public Health. 2019 Mar 21;16(6):1035. 30. 30.Riisbro R et al. Soluble urokinase plasminogen activator receptor measurements: influence of sample handling. Int J Biol Markers. 2001 Oct-Dec;16(4):233–9. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=11820717&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F06%2F21%2F2021.06.19.21259125.atom) 31. 31.Osuchowski MF et al. The COVID-19 puzzle: deciphering pathophysiology and phenotypes of a new disease entity. Lancet Respir Med. 2021 Jun;9(6):622–642. 32. 32.McGonagle D, Bridgewood C, Meaney JFM. A tricompartmental model of lung oxygenation disruption to explain pulmonary and systemic pathology in severe COVID-19. Lancet Respir Med. 2021 Jun;9(6):665–672. 33. 33.Perez-Riverol Y et al. The PRIDE database and related tools and resources in 2019: improving support for quantification data. Nucleic Acids Res. 2019; 47(D1):D442–D450 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/nar/gky1106&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=30395289&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F06%2F21%2F2021.06.19.21259125.atom) 34. 34.Hao Y et al. Integrated analysis of multimodal single-cell data. bioRxiv. 2020 October 12; doi: 10.1101/2020.10.12.335331. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiYmlvcnhpdiI7czo1OiJyZXNpZCI7czoxOToiMjAyMC4xMC4xMi4zMzUzMzF2MSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIxLzA2LzIxLzIwMjEuMDYuMTkuMjEyNTkxMjUuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 35. 35.Patro R et al. Kingsford C. Salmon provides fast and bias-aware quantification of transcript expression. Nat Methods. 2017 Apr;14(4):417–419. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nmeth.4197&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28263959&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F06%2F21%2F2021.06.19.21259125.atom) 36. 36.Howe EA, Sinha R, Schlauch D, Quackenbush J. RNA-Seq analysis in MeV. Bioinformatics. 2011 Nov 15;27(22):3209–10. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/bioinformatics/btr490&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21976420&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F06%2F21%2F2021.06.19.21259125.atom) 37. 37.Wettenhall JM, Smyth GK. Limma GUI: a graphical user interface for linear modeling of microarray data. Bioinformatics. 2004 Dec 12;20(18):3705–6. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/bioinformatics/bth449&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15297296&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F06%2F21%2F2021.06.19.21259125.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000225786600062&link_type=ISI) 38. 38.Zhou G et al. NetworkAnalyst 3.0: a visual analytics platform for comprehensive gene expression profiling and meta-analysis. Nucleic Acids Res. 2019 Jul 2;47(W1):W234–W241. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/nar/gkz240&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=30931480&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F06%2F21%2F2021.06.19.21259125.atom)