key: cord-0739021-kcuf5x8k authors: Sharif-Askari, Narjes Saheb; Sharif-Askari, Fatemeh Saheb; Mdkhana, Bushra; Hussain Alsayed, Hawra Ali; Alsafar, Habiba; Alrais, Zeyad Faoor; Hamid, Qutayba; Halwani, Rabih title: Upregulation of oxidative stress gene markers during SARS-COV-2 viral infection date: 2021-06-26 journal: Free Radic Biol Med DOI: 10.1016/j.freeradbiomed.2021.06.018 sha: 2183ef4fc4d347824fd7a0d9d01035cb7e417aa5 doc_id: 739021 cord_uid: kcuf5x8k Severe viral infections, including SARS-COV-2, could trigger disruption of the balance between pro-oxidant and antioxidant mediators; the magnitude of which could reflect the severity of infection and lung injury. Using publicly available COVID-19 transcriptomic datasets, we conducted an in-silico analyses to evaluate the expression levels of 125 oxidative stress genes, including 37 pro-oxidant genes, 32 oxidative-responsive genes, and 56 antioxidant genes. Seven oxidative stress genes were found to be upregulated in whole blood and lung autopsies (MPO, S100A8, S100A9, SRXN1, GCLM, SESN2, and TXN); these genes were higher in severe versus non-severe COVID-19 leucocytes. Oxidative genes were upregulated in inflammatory cells comprising macrophages and CD8(+) T cells isolated from bronchioalveolar fluid (BALF), and neutrophils isolated from peripheral blood. MPO, S100A8, and S100A9 were t/opmost upregulated oxidative markers within COVID-19's lung autopsies, whole blood, leucocytes, BALF derived macrophages and circulating neutrophils. The calprotectin's, S100A8 and S100A9 were upregulated in SARS-COV-2 infected human lung epithelium. To validate our in-silico analysis, we conducted qRT-PCR to measure MPO and calprotectin's levels in blood and saliva samples. Relative to uninfected donor controls, MPO, S100A8 and S100A9 were significantly higher in blood and saliva of severe versus asymptomatic COVID-19 patients. Compared to other different viral respiratory infections, coronavirus infection showed a prominent upregulation in oxidative stress genes with MPO and calprotectin at the top of the list. In conclusion, SARS-COV-2 induce the expression of oxidative stress genes via both immune as well as lung structural cells. The observed correlation between oxidative stress genes dysregulation and COVID-19 disease severity deserve more attention. Mechanistical studies are required to confirm the correlation between oxidative stress gene dysregulation, COVID-19 severity, and the net oxidative stress balance. to AT1 receptors, which in turn activates NADPH oxidase, and augments production of 139 reactive oxygen species (19, 20) . Expected Cellular response to oxidative stress is 140 mediated by Nuclear factor-erythroid 2 related factor 2 (NRF2) which activate the host 141 antioxidant defense by encoding transcription of oxidative-responsive and antioxidant 142 genes which contain antioxidant response elements (AREs) including thioredoxins, 143 sestrins, and glutathione system (21-23). The protective NRF2 antioxidant signaling was 144 found to be suppressed in severe COVID-19 lung autopsies as well as SARS-COV-2 in-145 vitro infection model (24) . These findings could suggest that SARS-COV-2 target NRF2 146 as an evasion mechanism to enhance their viral survival and replication (24) . 147 Although the contribution of oxidative stress to disease pathogenesis had been explored 149 in several viral infection (28, 29) , its relevance to COVID-19 respiratory infection 150 deserves more attention (25, 26) . This is due to the fact that immune derangement 151 during SARS-CoV-2 infection could switch on a lethal cycle of oxidative stress, 152 inflammation and lung tissue injury. Therefore, the aim of the current study is to 153 evaluate the dysregulation of oxidative balance during SARS-COV-2 infection through 154 measuring the gene expression levels of 125 oxidative stress genes known to be 155 associated with proinflammatory, antimicrobial, oxidant-scavenging and apoptosis-156 inducing activities . 157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186 Method 192 193 For this study, we first established a list of 125 oxidative stress genes including: 37 pro-194 oxidant genes, 32 oxidative-responsive genes, and 56 antioxidant genes (Table 1 -3). 195 The oxidative stress genes were derived from Gene Ontology (GO) term: 0006979 196 (response to oxidative stress), WikiPathways oxidative stress database (27), and a 197 number of previous reports (28-31). The expression of these genes was evaluated 198 using publicly available transcriptomic COVID-19 whole transcriptomic and single-cell 199 datasets of samples obtained from bronchioalveolar fluid (BALF), lung autopsies, and 200 whole blood of COVID-19 patients with different disease severity. We also compared 201 between the blood oxidative stress gene expression levels of COVID-19 and three 202 respiratory infections: SARS-CoV-1, influenza (IVA), and respiratory syncytial virus 203 (RSV). These datasets were publicly available at National Center for Biotechnology 204 while burden of co-morbidity was obtained by measuring the Charlson Comorbidity 227 Index score (35). Logistic regression analysis was then used to determine the 228 independent association between expression of oxidative stress genes and COVID-19 229 disease severity. ICU admission factor was used as the dependent factor and oxidative 230 stress gene expression as independent factor. The model was adjusted for age, gender, 231 body mass index and Charlson Comorbidity Index score (35). Statistical analyses were 232 performed using R software (v 3.0.2), SPSS 25.00 (SPSS Inc., Chicago, IL, USA), and 233 Prism (v8; GraphPad Software). P-value of <0.05 considered statistically significant. 234 We also examined how SARS-COV-2 and IAV infection may regulate the expression of 235 oxidative stress genes in whole blood and lung autopsies. We, hence, reanalyzed the 236 data deposited by Daniel Blanco-Melo (GEO: GSE147507) (6) to compare the 237 expression of these genes in viral-infected lung epithelial cells compared to Mock-238 infected controls. For leucocyte datasets (GEO: GSE157103) and Daniel Blanco-Melo 239 (GEO: GSE147507)), we processed the RNAseq raw count using the Bioconductor 240 package limma-voom (36), and presented the results as log2 counts per million (log 241 CPM). Log-transformed normalized intensities were also used in Linear Models for 242 MicroArray data (LIMMA) analyses to identify differentially expressed genes between 243 diseased and control groups. Expression of oxidative stress genes is increased in blood and lung tissue during 311 SARS-CoV-2 infection relative to disease severity. 312 313 Using publicly available transcriptomic datasets, we have determined the expression 314 levels of 125 oxidative stress genes, including 37 pro-oxidant genes, 32 oxidative-315 responsive genes, and 56 antioxidant genes. The lists of these genes are presented in 316 Tables 1-3. The datasets used in this study are presented in Table 4 . Expression levels 317 of the oxidative stress genes were determined in lung autopsies and whole blood of 318 COVID-19 patients ( Figure 1 ). For whole blood, RNA-sequencing data was extracted 319 from 20 severe COVID-19 patients and 10 controls ( Figure 1A ). For lung, RNA-320 sequencing data was obtained from 9 deceased COVID-19 patients and 10 negative 321 controls (PRJNA646224) ( Figure 1B ). Twenty-six oxidative stress genes were 322 upregulated in whole blood, while only 10 genes in lung autopsies (Figure 1C ). Seven of 323 these genes were commonly upregulated in both whole blood and lung autopsies 324 ( Figure 1C ), including the following five pro-oxidants/oxidative responsive genes: 325 Myeloperoxidase (MPO), Calprotectin (S100A8/S100A9), Sulfiredoxin-1 (SRXN1), 326 Glutamate-cysteine ligase modifier (GCLM), and two antioxidant genes: Sestrin 2 327 (SESN2) and Thioredoxin (TXN) (Figure 1C) . A significant increase in lung tissue 328 expression of S100A8 (4.20.3 log-fold vs 2.70.3 log-fold; p-value=0.001) and SRXN1 329 (2.10.2 log-fold vs 1.040.4 log-fold; p-value=0.03) compared to whole blood was 330 observed ( Figure 1C ). We then determined the expression of these seven shared genes 331 in NHBE infected with IAV or SARS-COV-2 using data deposited by Daniel Blanco-Melo 332 (GEO: GSE147507) (6). Of these seven genes, SRXN1 and SESN2 were slightly 333 increased in IAV infected NHBE (n=4 IAV NHBE vs n=4 mock infected NHBE), while 334 calprotectin genes of S100A8 (log-fold of 1.90.15; p-value < 0.0001) and S100A9 (log-335 fold of 1.10.07; p-value < 0.0001) were noticeably increased in SARS-COV-2 infected 336 The in-vitro infected results are displayed in the Figure 1D . 338 To confirm this increase in expression of oxidative genes during COVID-19 infection, we 340 also determined the expression of these seven genes in a transcriptome dataset of 341 leucocyte isolated from 37 severe and 51 non-severe COVID-19 patients (GSE157103). 342 Confirming the previous results, this analysis showed a significant upregulation of the 343 selected seven oxidative stress genes in severe versus non-severe COVID-19 (Figure 344 2). To further characterize the association between expression of these genes and 345 COVID-19 severity we carried a logistic regression analysis. After adjustment with age, 346 gender, body mass index, and Charlson Comorbidity Index score, the expression of 347 these seven genes were significantly associated with COVID-19 severity and ICU 348 admission. (Table S2) . increase in antioxidants genes. Among oxidative genes, the expression of S100A8 (1.8 367 log-fold; p-value <0.0001) and S100A9 (1.5 log-fold; p-value <0.0001) was significantly 368 increased ( Figure 4A ). Further, a distinct upregulation of oxidative stress genes in BALF 369 CD8+ T cells isolated from the same severe COVID-19 patients, while these markers 370 were not changed in BALF CD8+ T cells isolated from non-severe COVID-19 patients 371 ( Figure 4B ). All fold changes presented in Figure 4A and 4B were significant with a p 372 In this study overall neutrophil counts were increased in severe COVID-19, while 379 presence of low-density neutrophils was associated with severe COVID-19 phenotype 380 and development of ARDS (40). Low-density neutrophils showed upregulation of MPO 381 (1.6 log-fold; p-value <0.0001), CYBB (1.1 log-fold; p-value <0.0001), S100A8 (0.8 log-382 fold; p-value <0.0001) and S100A9 (0.34 log-fold; p-value <0.0001) calprotectin genes. patients relative to disease severity. We next examined whether the observed increase in these oxidative stress genes can 394 be detected in saliva of COVID-19 patients. This may hence suggest the usage of these 395 genes as non-invasive biomarkers for disease severity. To do that, we first validated the 396 increase of these markers in blood of asymptomatic and severe COVID-19 patients 397 using qRT-PCR. A significant increase in blood levels of MPO and calprotectin in severe 398 versus asymptomatic COVID-19 patients was observed ( Figure 4A ). MPO was 399 increased one log-fold more in severe versus asymptomatic (p-value= 0.0033), while 400 log-fold difference in S100A8 and S100A9 were 0.87 log-fold (p-value 0.004) and log-401 fold 0.9 (p-value=0.006), respectively. We then determined the level of these genes in 402 saliva from the same COVID19 patients ( Figure 4B ). This increase was comparable in 403 saliva compared to blood samples which may suggest that saliva level of expression of 404 these genes could reflect the level of COVID-19 severity. In severe versus 405 asymptomatic saliva, the log-fold difference was one log-fold (p-value=0.0001) for MPO, 406 2.7 log-fold (p-value<0.0001) for S100A8, and 0.3 log-fold (p-value=0001) for S100A9. We next compared the profile of enhanced oxidative stress gene expression observed 415 during SARS-CoV-2 to that detected during other respiratory viral infections. To do that, 416 we used transcriptomic microarrays and RNA-sequencing data of PBMCs isolated from 417 SARS-CoV-1, IAV, and RSV infected patients at the peak of disease. For each 418 condition, differential gene expression was obtained by comparing the normalized gene 419 expression of the infected group to those of healthy donors ( Figure 5A ). For IAV and 420 RSV infections, none of the oxidative stress genes were increased more than one log 421 fold-change (FC), while 7 genes for SARS-CoV-1 and 27 genes for SARS-CoV-2 422 infections were upregulated to a than one log FC ( Figure 5A ). TXN, QSOX1, MAPK14, 423 MPO, S100A9, and S100A8 were the top shared oxidative stress genes appearing in 424 both coronavirus respiratory infections, with an increase in expression of more than 1.5 425 folds following infection ( Figure 5B) . Herein, the dysregulation in the expression levels of 125 oxidative stress genes during 432 severe COVID-19 viral infection was explored using bioinformatic analysis of publicly 433 available transcriptomic datasets of lung autopsies, bronchioalveolar fluid, and blood 434 from SARS-CoV-2 infected individuals. Seven oxidative stress genes were found to be 435 upregulated in whole blood and lung autopsies of severe COVID-19 (MPO, S100A8, 436 S100A9, SRXN1, GCLM, SESN2, and TXN) ( Figure 1C ). Of these genes, calprotectin 437 genes, S100A8 and S100A9, were distinctly elevated in NHBE infected with SARS-438 COV-2 as compared to cells infected with IAV ( Figure 1D ). We then examined if the 439 increase in these genes was relative to disease severity. These genes were significantly 440 increased in blood leucocytes of severe compared to non-severe COVID-19 (Figure 441 S1). Using logistic regression, this association remained significant even after 442 adjustment with cofounding factors of age, gender, body mass index, and Charlson 443 Comorbidity Index score (Table S1) . Here we observed an overall dysregulation of oxidative stress genes expression in 472 circulating blood and lung tissue during severe COVID-19 disease. MPO and S100A8/9 473 calprotectin's were the top upregulated oxidative markers in lung autopsies, whole 474 blood, leucocytes, BALF derived macrophages and PBMC derived neutrophils. The 475 three top upregulated oxidative genes were validated in blood of severe COVID-19 476 patients using qRT-PCR. Relative to uninfected donor controls, MPO, S100A8 and 477 S100A9 were significantly higher in blood of severe versus asymptomatic COVID-19 478 patients. Interestingly, these three oxidative genes were also significantly upregulated in 479 saliva of severe relative to asymptomatic COVID-19 patients (Figure 4 ). This suggest 480 that the saliva level of these oxidative genes can be used as non-invasive markers for 481 COVID-19 disease severity. 482 483 S100A8 and S100A9 genes encode calcium binding proteins also known as Migration 484 Inhibitory Factor-Related Protein 8 and 14 (MPR8 and MRP14), respectively. They form 485 heterodimers known as calprotectin that binds to toll-like receptor 4 (TLR4) and function 486 as alarmin to stimulate the innate immune system pathways, namely MAP-kinase and 487 -kappa-B signaling (49, 50) . Given that, S100A8 and S100A9 have extensive effects 488 on the net inflammation, redox balance, and cell death via autophagy and apoptosis 489 (51, 52). They are also expressed abundantly in cells of myeloid origin such as 490 neutrophils and monocytes. These genes are not expressed in normal tissue resident 491 macrophages, however, S100A9 (MRP14) was found to be expressed in macrophage 492 during acute inflammation, while macrophage infiltrate during chronic inflammation 493 expressed both S100A9 and S100A8 (53). Interestingly, we found both S100A8 and 494 S100A9 to be upregulated in BALF derived macrophages of severe COVID-19 in 495 contrast to mild COVID-19 ( Figure 3A) . Recently, Silvin et al showed that elevated 496 calprotectin levels in peripheral blood cells could be used to discriminate severe from 497 mild COVID-19 infection (54). In this study, they used high-dimensional flow cytometry 498 and single-cell RNA sequencing of COVID-19 peripheral blood and suggested that high 499 calprotectin production is mediated by abnormal myeloid subsets (54). Calprotectin 500 genes are also expressed in lungs, particularly in lung epithelial and alveolar type II 501 pneumocytes (55). Likewise, the expression of these genes is increased with viral 502 infection (56) and lipopolysaccharide stimulation (57). In our study, through 503 bioinformatic analyses, we showed that S100A8 and S100A9 were expressed in HBE 504 and lung autopsies, and their expression was upregulated post SARS-COV-2 infection 505 ( Figure 1D ). The observed increase of these markers, especially in lung autopsies, 506 could be attributed to the increase in expression of these genes in both inflammatory as 507 well as structural lung cells. We then associated elevated calprotectin level to severe 508 COVID-19 (Figure 2, Figure S1 , Figure S2 , Figure 3 and Table S2 ) and validated this in 509 whole blood and saliva of COVID-19 patients (Figure 4) . 510 In both blood and lung autopsies, myeloperoxidase enzyme gene, MPO, was among 512 the top three oxidative stress genes. This gene is mainly expressed in neutrophil, and it 513 mediates catalysis of reactive oxygen intermediates such as hypohalous acids (58). 514 Oxidative stress stimulates neutrophil extracellular traps (NETs) formation by 515 neutrophils, NETosis, and lead to burst of neutrophil granules containing 516 myeloperoxidase enzyme and calprotectin which in turn boost the cellular oxidative 517 levels further. Similar to S100A8 and S100A9, myeloperoxidase expression level was 518 associated with disease severity (Figure 2 ). Supporting these findings, a recent 519 investigation by FP Veras et. al showed viable SARS-COV-2 ability to directly induce 520 the release of NETs by healthy neutrophils (2). This group observed that NETs 521 concentration was increased in plasma, tracheal aspirate and lung autopsies during 522 and COVID-19 severity (3, 4) . 524 In conclusion, SARS-COV-2 induce the expression of oxidative stress genes via both 577 immune as well as lung structural cells. Myeloperoxidase and calprotectin gene levels 578 are increased in blood, lung tissue, and inflammatory cells. In our study, these genes 579 were detected in salivary samples and hence they could potentially be used as a non-580 invasive severe COVID-19 marker. The observed correlation between oxidative stress 581 genes dysregulation and COVID-19 disease severity deserve more attention. These 582 changes in oxidative stress gene expression may or may not reflect alteration in the net 583 oxidative stress balance. It is warranted that further mechanistical studies are performed 584 to confirm this association. 585 The authors declare no competing interests. were shared between whole blood and lung autopsies. MPO, S100A8, and S100A9 were among 868 the top upregulated oxidative genes, while S100A8 and SRXN1 were higher in lung autopsies. 869 Results are presented as fold change of gene expression between cases and controls. Unpaired 870 student t-test was used to compare between fold changes in mild and severe COVID-19. * p < 871 0.05, * * p < 0.01, * * * p < 0.001, * * * * p < 0.0001. (D) The seven shared oxidative genes were 872 analyzed in SAR-COV-2 and influenza A virus infected human lung epithelial cells. Independent 873 biological replicates of primary human lung epithelium (NHBE) were mock treated or infected 874 with SARS-CoV-2 (USA-WA1/2020), or IAV (A/Puerto Rico/8/1934 (H1N1)). Of these seven 875 genes, SRXN1 and SESN2 were slightly increased in IAV infected NHBE (n=4 IAV NHBE vs 876 n=4 mock infected NHBE, GEO: GSE147507), while calprotectin genes of S100A8 and S100A9 877 were noticeably increased in SARS-COV-2 infected NHBE (n=3 SARS-CO-V-2 NHBE vs n=3 878 patients relative to disease severity. (A) Gene expression level of MPO, S100A8 and S100A9 902 was higher in blood from severe COVID-19 (n=7) as compared to asymptomatic COVID-19 903 (n=9). (B) Gene expression level of MPO, S100A8 and S100A9 was higher in saliva from severe 904 COVID-19 (n=10) as compared to asymptomatic . Results are presented as log2 905 fold change. Unpaired student t-test was used to compare between the independent groups. * p < 906 0.05, * * p < 0.01, * * * p < 0.001, * * * * p < 0.0001. 907 Clinical characteristics and 602 intrauterine vertical transmission potential of COVID-19 infection in nine pregnant women: a 603 retrospective review of medical records Clinical features of patients infected 605 with 2019 novel coronavirus in Wuhan Transmission 607 of 2019-nCoV Infection from an Asymptomatic Contact in Germany Novel Coronavirus in the United States The many faces of the anti-COVID immune response Imbalanced Host Response to SARS-CoV-2 Drives Development of COVID-19 Immunophenotyping of COVID-19 618 and influenza highlights the role of type I interferons in development of severe COVID-19 Single-cell landscape of bronchoalveolar 621 immune cells in patients with COVID-19 Prevalence of co-623 morbidities and their association with mortality in patients with COVID-19: A systematic review 624 and meta-analysis Global pandemics interconnected -obesity, 626 impaired metabolic health and COVID-19 COVID-19 prevalence 628 and mortality in patients with cancer and the effect of primary tumour subtype and patient 629 demographics: a prospective cohort study Is Associated with Altered Host Survival and T Cell Phenotypes after Influenza A Virus Infection 632 in Mice Correlates With Poor Prognosis of Severe Influenza A Infection Viral-mediated 636 Inhibition of Antioxidant Enzymes Contributes to the Pathogenesis of Severe Respiratory 637 Tissue distribution 640 of ACE2 protein, the functional receptor for SARS coronavirus. A first step in understanding 641 SARS pathogenesis ACE2 localizes to the 643 respiratory cilia and is not increased by ACE inhibitors or ARBs Modulation of TNF-α-converting enzyme by the spike protein of SARS-CoV and ACE2 induces 647 TNF-α production and facilitates viral entry CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease 651 Inhibitor Hydrolysis of Biological 653 Peptides by Human Angiotensin-converting Enzyme-related Carboxypeptidase * SARS-CoV-2 and Other 658 Respiratory Viruses: What Does Oxidative Stress Have to Do with It? Oxidative Medicine and 659 Cellular Longevity Nrf2:INrf2 (Keap1) signaling in oxidative stress. Free 661 radical biology & medicine Regulation of Nrf2-an update. Free Radical Biology and 663 Medicine SARS-CoV2-665 mediated suppression of NRF2-signaling reveals potent antiviral and anti-inflammatory activity 666 of 4-octyl-itaconate and dimethyl fumarate A useful and 668 sensitive marker in the prediction of COVID-19 and disease severity: Thiol. Free Radical Biology 669 and Medicine Heme oxygenase-1 modulation: A potential therapeutic 671 target for COVID-19 and associated complications. Free Radical Biology and Medicine Available 674 at www.wikipathways.org/index.php/Pathway:WP408 The cysteine proteome. Free Radical Biology and 678 Medicine Oxidative cross-680 linking of calprotectin occurs in vivo, altering its structure and susceptibility to proteolysis Cellular response to oxidative stress: Signaling for suicide 683 and survival* Transcriptional and proteomic insights 685 into the host response in fatal COVID-19 cases Disease severity-specific neutrophil signatures in blood transcriptomes stratify COVID-19 689 patients Gene expression 699 signatures diagnose influenza and other symptomatic respiratory viral infections in humans Expression profile of 702 immune response genes in patients with Severe Acute Respiratory Syndrome Robust meta-analysis of gene expression using the elastic net. 705 Nucleic acids research A single-707 cell atlas of the peripheral immune response in patients with severe COVID-19 SARS-CoV-2 712 Infection-Induced Promoter Hypomethylation as an Epigenetic Modulator of Heat Shock 713 Protein A1L (HSPA1L) Gene SARS-CoVe reduction of nuclear DUSP1 716 and DUSP5 expression 18S rRNA is a 718 reliable normalisation gene for real time PCR based on influenza virus infected cells Reactive oxygen and nitrogen intermediates in the relationship 725 between mammalian hosts and microbial pathogens Oxidative Stress: 728 Harms and Benefits for Human Health. Oxidative medicine and cellular longevity Induction of nuclear factor-κB 731 responses by the S100A9 protein is Toll-like receptor-4-dependent Induction of neutrophil degranulation by S100A9 via a 734 MAPK-dependent mechanism S100A8/A9 736 induces autophagy and apoptosis via ROS-mediated cross-talk between mitochondria and 737 lysosomes that involves BNIP3 Proinflammatory activities of 739 S100: proteins S100A8, S100A9, and S100A8/A9 induce neutrophil chemotaxis and adhesion Elevated 744 Calprotectin and Abnormal Myeloid Cell Subsets Discriminate Severe from Mild COVID-19 Induction of alarmin S100A8/A9 mediates 749 activation of aberrant neutrophils in the pathogenesis of COVID-19 Up-regulation of S100A8 and 752 S100A9 protein in bronchial epithelial cells by lipopolysaccharide Its role for host defense, inflammation, and neutrophil 755 function Oxidative stress during viral infection: a review Biological effects of the oxygen molecule in critically ill patients SARS-CoV-2-triggered neutrophil extracellular traps mediate COVID-19 pathology Inhibitory effect of melatonin on lung 764 oxidative stress induced by respiratory syncytial virus infection in mice Oxygen free radical involvement 767 in acute lung injury induced by H5N1 virus in mice. Influenza and Other Respiratory Viruses Modulation of Antiviral Immunity by Heme 770 Therapeutic Impact 772 of Human Serum Albumin-Thioredoxin Fusion Protein on Influenza Virus-Induced Lung Injury 773 Mice Extracellular thioredoxin levels are increased in patients with acute lung injury Active Protein Thioredoxin-1 Administration Ameliorates Influenza A Virus (H1N1 Acute Lung Injury in Mice Can Activation of NRF2 Be a Strategy against COVID-19? Direct and indirect antioxidant properties of inducers of 784 cytoprotective proteins Impaired 786 efferocytosis and neutrophil extracellular trap clearance by macrophages in ARDS The expression of MPO, S100A8, and S100A9 were higher in severe compared to mild Results are presented as fold change of gene expression between cases and 925 controls The sample size presented in A-C belonged to EGAS00001004503 928 dataset and were as following Single-cell gene expression from PBMCs of severe COVID-19 patients Single-cell RNA sequencing was performed on PBMCs from 6 severe COVID-19 934 patients and 7 healthy controls (GEO: GSE150728). Low-density neutrophils were associated 935 with severe COVID-19 phenotype and development of acute respiratory distress syndrome Model-based analysis of single cell transcriptomics (MAST) algorithm in Seurat v3 was used by 937 authors to identify differentially expressed genes and to determine the fold change Prominent upregulation of S100A8 and S100A9 in M1 macrophage group. Macrophages were 891 clustered into four groups; M1 macrophages were presented with group 1 and 2, while M2 892 macrophages were presented with group 3. Both M1 and M2 like macrophages were enriched in 893 severe COVID-19. Group 4 macrophages were predominant in moderate and healthy controls 894 and presented the less severe COVID-19. Fold changes were generated for each group of 895 macrophages relative to total macrophages. (B) Specific upregulation of oxidative stress genes in 896 CD8+ T cells from severe COVID-19 patients, while none of the oxidative genes appeared in the 897 non-severe COVID-19 cluster. All fold changes presented in the figure were significant with a p 898 value <0.05. 899 900