key: cord-0711149-4jyqzztm authors: Manivannan, Jeganathan; Sundaresan, Lakshmikirupa title: Systems level insights into the impact of airborne exposure on SARS-CoV-2 pathogenesis and COVID-19 outcome – A multi-omics big data study date: 2021-08-12 journal: Gene Rep DOI: 10.1016/j.genrep.2021.101312 sha: c949d4ef8318cc77de23cdb25fc56ec2ae4c7af1 doc_id: 711149 cord_uid: 4jyqzztm Coronavirus disease 2019 (COVID-19) is a viral pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that led to more than 800,00 deaths and continues to be a major threat worldwide. The scientific community has been studying the risk factors associated with SARS-CoV-2 infection and pathogenesis. Recent studies highlight the possible contribution of atmospheric air pollution, specifically particulate matter (PM) exposure as a co-factor in COVID-19 severity. Hence, meaningful translation of suitable omics datasets of SARS-CoV-2 infection and PM exposure is warranted to understand the possible involvement of airborne exposome on COVID-19 outcome. Publicly available transcriptomic data (microarray and RNA-Seq) related to COVID-19 lung biopsy, SARS-CoV-2 infection in epithelial cells and PM exposure (lung tissue, epithelial and endothelial cells) were obtained in addition with proteome and interactome datasets. System-wide pathway/network analysis was done through appropriate software tools and data resources. The primary findings are; 1. There is no robust difference in the expression of SARS-CoV-2 entry factors upon particulate exposure, 2. The upstream pathways associated with upregulated genes during SARS-CoV-2 infection considerably overlap with that of PM exposure, 3. Similar pathways were differentially expressed during SARS-CoV-2 infection and PM exposure, 4. SARS-CoV-2 interacting host factors were predicted to be associated with the molecular impact of PM exposure and 5. Differentially expressed pathways during PM exposure may increase COVID-19 severity. Based on the observed molecular mechanisms (direct and indirect effects) the current study suggests that airborne PM exposure has to be considered as an additional co-factor in the outcome of COVID-19. The coronavirus disease 2019 pandemic caused by infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has led to more than 500,000 deaths worldwide (" " n.d.) . SARS-CoV-2 infection caused clusters of severe respiratory illness similar to severe acute respiratory syndrome coronavirus (SARS) and was associated with high mortality Wang et al., 2020) . In the throes of the COVID-19 crisis, Conticini et al conclude that the high level of atmospheric pollution in northern Italy should be considered an additional co-factor of the high level of lethality recorded in that area (Conticini et al., 2020) . In China, based on the daily confirmed cases, air pollution concentration and meteorological variables obtained from 120 cities indicate that there was a significant relationship between air pollution and COVID-19 infection . Further, a study that investigated the geographical expansion of the infection and correlated it with the annual indexes of air quality observed from the Sentinel-5 satellite provide initial evidence of higher morbidity and mortality due to SARS-CoV-2 in regions with poor air quality (Pansini and Fornacca, 2020) . Moreover, a nationwide cross-sectional study in the USA also found that a small increase in long-term exposure to PM2.5 (diameters of 2.5 μm or less) leads to are major contributors to disease severity and death in infected patients (Merad and Martin, 2020; . SARS-CoV-2 entry factors are highly expressed in nasal epithelial cells along with innate immune genes (Sungnak et al., 2020) . Apart from epithelial cells, recent findings have shown the presence of viral elements within endothelial cells and an accumulation of inflammatory cells, with evidence of endothelial and inflammatory cell death (Varga et al., 2020) . A recent study shows that SARS-CoV-2 utilizes the SARS-CoV receptor ACE2 for its entry and TMPRSS2, a serine protease for S protein priming (Hoffmann et al., 2020) . Long-term exposure to ambient PM 2.5 is often connected to with faster deterioration of lung function along with an higher risk of the occurrence of lung diseases (Guo et al., 2018) . Notably, previous in vivo and in vitro studies highlight the up-regulation of ACE2 (SARS-CoV-2 entry factor) expression during airborne particulate matter exposure Miyashita et al., 2020) . The possible role of the airborne exposome in promoting resilience or susceptibility to SARS-CoV-2 infection has been recently highlighted (Naughton et al., 2020) . Therefore, a systems approach that aims to target a list of disease molecular features, such as gene expression signatures, can be applied to understand the possible involvement of airborne particulate exposure in COVID19 pathogenesis and severity. Hence, the current study attempts an integrative analysis of omics big data (transcriptome, proteome and interactome) available for COVID19 pathogenesis and PM exposure in order to understand the possible molecular links between them and highlight the need to consider the physiological effects of air pollution during COVID19 clinical management. J o u r n a l P r e -p r o o f Journal Pre-proof Publicly available microarray datasets of various in vivo (mouse and rat) and human in vitro (epithelial and endothelial cells) studies related to particulate matter exposure were obtained from Gene Expression Omnibus (GEO; https://www.ncbi.nlm.nih.gov/geo/), a public functional genomics data repository (Clough and Barrett, 2016) . Table 1 demonstrates the details of microarray datasets with appropriate experimental comparison used in the current study. Differential gene expression analysis across experimental conditions was performed using GEO2R (https://www.ncbi.nlm.nih.gov/geo/geo2r/) by comparing two or more experimental conditions. GEO2R employs the GEOquery and limma R packages from the Bioconductor project (https://www.bioconductor.org/) and the results have been presented as a table of genes ordered by statistical significance. Expression of SARS-CoV-2 entry factors as mentioned in (www.covid19cellatlas.org) such as Angiotensin-Converting Enzyme 2 (ACE2), Transmembrane Serine Protease 2 (TMPRSS2), Cathepsin B (CTSB) and Cathepsin L (CTSL) were queried against the list of differentially expressed genes (DEGs) of the mouse, rat and human microarray datasets (control vs particulate exposure). The expression is considered as regulated genes derived from the above datasets to SPEED2 (https://speed2.sys-bio.net/). SPEED2 is a web server that utilizes pathway signatures derived from a large number of manually curated pathway perturbation experiments across many different cell types in human cells (Rydenfelt et al., 2020) . It allows inferring signaling pathways that likely caused these genes to be deregulated. In this analysis, the bates test was selected as test statistics of enrichment. In order to link the expression signature to upstream cell signaling kinase networks, the current study employed the eXpression2Kinases (https://amp.pharm.mssm.edu/X2K/) web server which computationally predicts involvement of upstream kinase signaling pathways. Current analysis utilized the list of significantly up-regulated genes derived from RNA-Seq datasets of COVID19 lung biopsy and SARS-CoV-2 infected Calu-3 cells. Additionally, significantly upregulated genes (FDR< 0.1 and logFC >1) obtained from PM exposed microarray datasets (GS2E4567 and GSE38172-4 th day) were also included. This study did not consider the down regulated genes since analysis with less than 20 genes may produce inaccurate results in the X2K analysis. The X2K tool first computes enrichment of transcription factors that are likely to regulate the expression of the differentially expressed genes followed by connecting the transcription factors through known protein-protein interactions (PPIs) to construct a subnetwork. Finally, it performs kinase enrichment analysis on the members of the sub-network and the complete upstream pathway is constructed via the eXpression2Kinases (X2K) algorithm (Clarke et al., 2018) . J o u r n a l P r e -p r o o f In this study, among the list of human microarray datasets, two of the real-world particulate matter exposed experimental designs were preferred to represent the pulmonary and vascular system (GSE38172-4 th day and GSE4567). The list of DEGs was obtained between appropriate experimental conditions using moderate cut-off values of FDR <0.1 and logFC >0.8 for pathway enrichment analysis. In addition, the list of DEGs derived from RNA-Seq transcriptome profile of SARS-CoV-2 infected Calu-3 cells also utilized for pathway membership analysis. The up and down regulated genes were submitted to EnrichR (http://amp.pharm.mssm.edu/Enrichr), a comprehensive gene set enrichment analysis web server and the analysis was done through Kyoto Encyclopedia of Genes and Genomes (KEGG) human pathways module (Kuleshov et al., 2016) . In this case, only the top 10 KEGG pathways based on the score in which terms with adjusted p value of < 0.05 were prioritized. This analysis anticipated to explore the overlapping pathological mechanisms between particulate matter exposed bronchial epithelial cells and endothelial cells and COVID19 affected lungs. The differentially expressed genes (with FDR< 0.05 and logFC >1) from RNA-Seq (GSE147507-COVID19) and microarray datasets (GSE4567, GSE38172-Day 1 and 4) were submitted to ToppCluster (http://toppcluster.cchmc.org) tool whose functionalities enables the identification of specialized biological functions and regulatory networks and systems biologybased dissection of biological states (Kaimal et al., 2010) . Cytoscape (https://cytoscape.org/), a software environment for integrated models of biomolecular interaction networks (Shannon et al., 2003) was utilized for visualizing the network with the enriched terms (FDR<0.05) between experimental conditions. J o u r n a l P r e -p r o o f Journal Pre-proof Gene Set Enrichment Analysis (GSEA) designed to analyze ranked lists of all available genes and do not require a threshold and whole-genome ranked list is suitable for input into pathway enrichment analysis (Reimand et al., 2019; Subramanian et al., 2005) . In this study, the genes from transcriptome experiments (GSE147507-COVID19 lung biopsy, GSE4567, GSE38172 -Day 1 and 4) were ranked by using sign (fold change) multiplied by (-log 10 (pvalue). The pre-ranked genes were subjected to GSEA with C2 pathways and C5 gene ontology biological processes from the MSigDb (v6.0) collection. Gene sets with a size between 15 and 300 were included and the network was constructed with gene sets having FDR<0.05 using EnrichmentMap application (Merico et al., 2010) in Cytoscape. We first checked the expression of SARS-CoV-2 viral entry receptors upon particulate matter exposure. Among the 15 particulate exposure experimental conditions compared, none of the four SARS-CoV-2 viral entry receptors/factors (ACE2, TMPRSS2, CTSB and CTSL) passed the significant expression criteria (Table 1) . In order to obtain insights into the pathways modulated upon SARS-CoV-2 infection and PM exposure, we performed enrichment analysis of the DEG from different datasets. There were 1179 upregulated and 581 down-regulated genes in Calu-3 cells upon COVID-19 exposure. Various KEGG pathways were significantly (FDR <0.05) enriched among the DEGs of SARS-CoV-2 infected Calu-3 cells and PM exposed cells. Table. 2 represents the top 10 pathways deregulated in SARS-CoV-2 infected Calu-3 cells. Among that, notably TNF signaling pathway, IL-17 signaling pathway and NF-kappa B signaling pathways were enriched. Table 3 illustrates the top 10 pathways enriched from differentially expressed endothelial genes upon PM exposure which including TNF signaling pathway, IL-17 signaling pathway and NF-kappa B signaling pathway. Moreover, pathways enriched with DEGs of PM exposed bronchial epithelial cells indicate that PPAR signaling, ferroptosis and ovarian steroidogenesis as significantly enriched (FDR<0.05) among other pathways as presented in Table 4 . We predicted the possible upstream signaling pathways using SPEED. Upstream causal signaling pathway and kinase network associated with upregulated genes in SARS-CoV-2 infected Calu-3 cells and PM exposed bronchial epithelial cells have been illustrated in Fig.1 and Fig.2 respectively. In this observation, the top 3 causal signaling pathways in COVID-19 affected lung (JAK-STAT, IL-1 and TLR signaling) and SARS-CoV-2 infected Calu-3 cells (IL-1, TLR and TNF-α signaling) were considered. In the case of PM exposed models as well, top 3 causal pathways in PM exposed endothelial cells (IL-1, VEGF and TNF-α signaling) and bronchial epithelial cells (Insulin, TNF-α and VEGF signaling) were prioritized. Based on the X2K derived The list of differentially expressed proteins during SARS-CoV-2 infection and the results of reactome pathway analysis (list of genes increased in protein level corresponding to major metabolic pathways) obtained from a previous study (Bojkova et al., 2020) were adapted for proteomic and metabolic pathway analysis. Among the differentially expressed proteins none of them overlapped with PM induced differentially expressed genes except for F3 (upregulated). Altogether 22 genes namely, ENO1, IDH2, GOT2, CS, HK2, IDH3B, PKM, HKDC1, GAPDH, PDHB, MDH2, IDH3G, NDUFA10, PDK1, SCO1, VDAC1, COX11, NDUFAF2, NDUFB9, NDUFA7, NUP93, POM121C were identified from enriched terminologies such as carbon metabolism, citric acid cycle and respiratory electron transport, citrate cycle (TCA cycle), 2-oxocarboxylic acid metabolism, and glucose metabolism. Among the up-regulated metabolic enzymes none of them were differentially (both up and down) regulated in PM exposed datasets of bronchial epithelial cells and endothelial cells considered. We studied PTM using the protein-protein interaction network. The protein-protein interaction network suggests that the possibility of interconnections among the various host proteins (Fig.3) . Among the SARS-CoV-2 hijacked host factors, the current study focusses on proteins involved in PTM events, especially if their possible interaction partner also exists in the predicted network. In the context of molecular action, 7 post-translational modification partners namely MARK3 -PKP2, MARK2 -RNF41, TBK1 -MIB1, PRKACA -GNB1, PRKACA-GNG5, RHOA-PRKACA and PRKACA-ATP1B1 were predicted apart from the general J o u r n a l P r e -p r o o f interactions. While comparing the host genes against the PM exposed DEGs, totally 3 genes were found to be up-regulated in both the PM exposed conditions (Fig. 4) . Bronchial epithelial cells showed upregulation of HMOX1 while in endothelial cells, HMOX1, NPTX1 and RDX were found to be upregulated among the host protein interactors. No overlap between host factors and downregulated genes was observed. We used Toppcluster tool to perform clustering of COVID-19 and PM associated pathways. An overview of the Gene Ontology (GO) functional classification network constructed with the DEGs is illustrated in Fig.5 . Specifically, functional categories were enriched under biological process (BP), molecular function (MF) and pathways. The network illustrates that various biological processes and molecular pathways enriched in COVID19 infected lungs overlap with the enrichment observed in PM exposed endothelial cells and bronchial epithelial cells. Remarkably, many of them were associated with immune and inflammatory response associated molecular signaling pathways. In an attempt to connect the DEGs with a priori defined set of genes through GSEA for generating an added/improved result towards the system level understanding, we performed GSEA. GSEA identified several pathways that were highly enriched between genes of COVID-19 lung and PM exposed bronchial epithelial cells and endothelial cells (Fig.6) . Notably, it included innate immune response, cytokine signaling and stress response. Currently, the effect of COVID-19 in people with other comorbidities remains inconclusive which necessitates suitable prospective studies. At the molecular level, genes/pathways closely linked with SARS-CoV-2 infection and/or associated pathological changes in lung and vascular cells need to be investigated. In this study, the plausible impact of particulate air pollution exposure on the progression of COVID-19 and associated pathophysiology of the pulmonary system has been discussed. While focusing on the expression of SARS-CoV-2 entry factors (ACE2, TMPRSS2, CTSB and CTSL), the current study did not observe any changes (especially up-regulation) in the existing microarray datasets related to particulate exposure. Whereas, a recent observation showed that traffic-related particulate matter upregulated the ACE2 expression in A549 cells (Miyashita et al., 2020) and another recent study suggests PM2.5 exposure may up-regulate ACE-2 a protein level (Borro et al., 2020) . In largescale gene expression experiments, such as transcriptomic studies, the false discover rate (FDR) correction and stringent fold change cut-off allow us to select only the genes showing a high confident expression change. Hence, a range of expression studies performed on various models clearly indicates that exposure to airborne PM pollution may not robustly deregulate the SARS-CoV-2 entry factors. Studying the upstream signaling (infer the mechanistic origin of transcriptomic dysregulation of genes) and downstream (consequent effects) pathways of SARS-CoV-2 infected cells is key to understanding the molecular mechanism of SARS-CoV-2 viral infection. This is in turn essential for correlating the existing knowledge of particulate exposure associated pathogenic signaling with COVID-19. The prediction of upstream pathways in SARS-CoV-2 infected conditions prioritized the association of TLR, TNF-α, IL-1 and JAK-STAT signaling pathways with the up-regulated genes where some of them are shared with PM exposed J o u r n a l P r e -p r o o f conditions. While considering the pathway membership of deregulated genes, many of the upregulated genes fall under pathways associated with viral pathogenesis and inflammatory signaling including TNF, IL-17 and NF-κB signaling pathways where overlapping of pathway terms between SARS-CoV-2 infected and PM exposure conditions are also observed. The abovementioned pathways are known to be associated with chronic inflammation of the airways (Athari, 2019) and also involved in the activation of proinflammatory responses in cells of the airway mucosa by particulate matter (Øvrevik et al., 2015) . Precisely, a previous study suggest that urban PM2.5 may aggravate allergic inflammation in the lungs via TLR2/TLR4/MyD88signaling pathway in a mouse model (He et al., 2017) . Exposure of human bronchial epithelial cells to airborne PM2.5 consistently up-regulated a number of genes including TNF-α implicated in the inflammatory response (Zhou et al., 2015) . While focusing on IL-17 signaling, an earlier study indicates that PM2.5 treatment changed the gene expression associated with IL-17 signaling pathway in the lung (Jeong et al., 2019) . In addition, cytokine expression profiling in bronchoalveolar lavage fluid (BALF) of animals exposed to traffic related PM10 also indicates increased expression of IL-17A and TNF-α . IL-17 is a proinflammatory cytokine which can induce A549 alveolar epithelial cells to undergo EMT via the TGF-β1 mediated Smad2/3 and ERK1/2 activation which is in turn associated with pulmonary fibrosis (T. , while the role of IL-17 in COVID-19-related pathogenesis has been already highlighted (Pacha et al., 2020) . In a precise perspective, the upstream kinase signaling network plausibly associated with the up-regulated genes during SARS-CoV-2 infection and PM exposure was found to be overlap. (Hou et al., 2020) . An earlier study explored that PM exposure enhanced the airway inflammatory response significantly through ROS-mediated activation of MAPK (ERK, JNK and p38 MAPK) and downstream NF-κB signaling pathways (J. . In addition, a study on human bronchial epithelial cells (HBECs) exposed to particulate matter observed an increased phosphorylation of ERK, JNK and PI3K which activating the associated signaling pathways in a time-dependent manner (C. . Further, PM exposure increase ICAM-1 expression in pulmonary epithelial cells in vitro and in vivo through the IL-6/AKT/STAT3/NF-κB signaling pathway . Studies on A549 lung cells showed that NRF2 regulates the steady-state levels of HIPK2 and their crosstalk shapes the cytoprotective responses (Torrente et al., 2017) , where the inhaled particulate matter toxicity is associated with redox imbalance and NRF2 activation (Deng et al., 2013; Pardo et al., 2020) . Further, PM2.5 rapidly induces severe cell cycle alterations including elevation of cyclin B1 level, which associates with CDK1 and drives the progression of cells through mitosis (Longhin et al., 2013) . From the above findings, we suggest that host cell signaling pathways that might be modulated by SARS-CoV-2 to facilitate their own replication cycle and signaling may be synergistically impacted during particulate exposure in the pulmonary system. The study also attempts to find clues from the microarray experiments to explore the possible effects of particulate pollution in the context of COVID-19 pathogenesis such as pulmonary dysfunction and endotheliitis. Pathway analysis showed that genes involved in PPAR signaling pathway, ferroptosis and steroidogenesis were upregulated where the source study indicates that genes involved in inflammation, cytokine production and cell metabolism were found to be deregulated in addition with the up-regulation of genes related to cholesterol and lipid biosynthesis in cells exposed to PM10 for 4 days (Sun et al., 2012) . Apart from the J o u r n a l P r e -p r o o f enrichment of phenomena such as immune response and inflammatory signaling observed in various PM exposure models, the upregulation of ferroptosis related genes (HMOX1, SLC7A11 and GCLM) highlights the possible association of iron-dependent cell death (Hirschhorn and Stockwell, 2019). This is experimentally supported in endothelial cells where PM2.5 enhances ferroptosis sensitivity which is dependent on iron overload and oxidative stress (Wang and Tang, 2019) . Moreover, a study on cigarette smoke treated lung epithelial cells also highlights the involvement of epithelial cell ferroptosis in chronic obstructive pulmonary disease pathogenesis (Yoshida et al., 2019) . In this regard, a recent study also described the potential of iron chelation as an alternative beneficial adjuvant in treating SARS-CoV-2 infection . In the setting of COVID-19, pulmonary and peripheral endothelial injury due to direct viral attack might be an important inducer of hypercoagulation . Previous histopathological studies from COVID-19 patients have shown direct viral infection of endothelial cells, endotheliitis with diffuse endothelial inflammation and thrombosis in blood vessels (Pons et al., 2020; Varga et al., 2020) . In vitro experiments highlights that SARS-CoV-2 has the ability to directly infect human blood vessel organoids (Monteil et al., 2020 ). An earlier study (source study of microarray data GSE4567) on human pulmonary artery endothelial cells (HPAEC) exposed to ultrafine particles (UFPs) indicates that PM may cause adverse cardiovascular health effects by activating the coagulation-inflammation circuitry (Karoly et al., 2007) . In this regard, the current observation also highlights the elevation of TNF-α, IL-17 and NF-κB pathway components during PM exposure in endothelial cells. Notably, the blood coagulation pathway component F3 which is up-regulated in endothelial cells during UFPs exposure was also found to be elevated during SARS-CoV-2 infection in Caco-2 cells as evident from the proteome analysis (Bojkova et al., 2020) . In the host PPI network, few kinases associated with PTM events were prioritized among possibly hijacked signaling pathways associated with SARS-CoV-2 infection. A recent study showed that PRKACA is one among proteins possessing the highest degree centrality in SARS-CoV-2-host PPI network where GNB1 and GNG5 possess high centrality within the PPI network related to the immune system (Andrés et al., 2020) . A recent observation identified that ARHGEF2 phosphorylation by MARK3 induced RHOA activation and a regulatory switch controlled by MARK3 couples the microtubule and actin cytoskeletons to establish polarity in epithelial cells (Sandi et al., 2017) . Further, RNF41, an E3 ubiquitin ligase has been indicated as a novel MARK2 effector in the cell-ECM pathway (Lewandowski and Piwnica-Worms, 2014) . In alveolar epithelial cells, TBK1-AKT-ERK signaling pathway modulates radiation-induced EMT, which suggests that TBK1 is a potential target in the prevention of pulmonary fibrosis (Qu et al., 2019) and increased levels of active TBK1 are implicated in inflammatory diseases (Yu et al., 2012) . In the case of PKA signaling, protein kinase A-mediated CREB phosphorylation is a survival pathway in alveolar type II cells (Barlow et al., 2008) . In the vasculature, PRKACA was identified to directly regulate 137 neighbouring genes being located at the center of the gene network in Angiotensin II-induced arterial injury . PM2.5 induced activation of the RhoA/ROCK pathway where ROCK inhibitor attenuated the PM2.5 induced inflammatory response, NF-кB activation and epithelial barrier dysfunction in human lung epithelial BEAS-2B cells (Yan et al., 2017) . In cultured lung epithelial cells, chronic oxidative stress damages mitochondrial DNA (mtDNA) which in turn triggering inflammation via the TBK1/interferon regulatory factor 3 signaling pathway (Szczesny et al., 2018) wherein oxidative stress and mitochondrial DNA damage are considered as a significant consequence of PM exposure (Grevendonk et al., 2016; Hou et al., 2010) . Moreover, among the 332 high-confidence SARS-CoV-2 human protein interactors, HMOX1, NPTX1 and RDX were found to be up-regulated during PM exposure. The heme oxygenase 1 (HMOX1) acts as a critical mediator in ferroptosis induction and plays a causative factor in the progression of several diseases (Chiang et al., 2018) . It is a major intracellular source of iron and an essential enzyme for iron-dependent lipid peroxidation during ferroptotic cell death (Kwon et al., 2015) . Studies demonstrated that NPTX1 inhibited growth and promoted apoptosis in hepatocellular carcinoma (Zhao et al., 2019) and enhanced NPTX1 secretion and reactive oxygen species generation in human endometrial stromal cells impair the survival of endothelial cells resulting in the loss in vascular integrity (Guzeloglu-Kayisli et al., 2014) . TNF-α and modulates permeability increases in human pulmonary microvascular endothelial cells (Koss et al., 2006) . Thus, the study hints that SARS-CoV-2 hijacked human factors involved in cell death and vascular integrity may modulated by PM exposure. Evaluating disease similarity merely on the basis of shared genes can be misleading as variable combinations of genes may be associated with similar conditions. This holds true for complex diseases as well, it is ideal to look for common biological processes rather than comparing gene matches (Mathur and Dinakarpandian, 2012) . Therefore, grouping of similar pathways and major biological themes (between disease or conditions) is necessary for the understanding of overlapping mechanisms of pathogenesis. A great deal of practical importance exists for computing mechanism based disease-disease similarities based on the shared underlying molecular processes (Hamaneh and Yu, 2016) . In this study, a cluster of pathways and biological mechanisms indicate that both COVID-19 and PM exposure conditions share many similar terms including innate immunity associated processes, inflammatory signaling, J o u r n a l P r e -p r o o f viral replication and stress associated pathways. Previously, Wu et al reported the inflammatory health effects of PM exposure and their substantial association with cardiopulmonary disease burden (Wu et al., 2018) . In this regard, it was previously highlighted that, controlling the inflammatory response may be as important as targeting the SARS-CoV-2 virus where modulation of deregulated immune responses along with inhibiting viral infection may synergize to prevent COVID-19 pathologies at multiple steps (Tay et al., 2020) . In addition, a recent study using RNA virus (influenza virus) indicates that PM exposure promotes viral infection through modulation of innate immune responses causing overall pathogenic burden in the lung cells (Mishra et al., 2020) . Further, the interleukin-mediated inflammatory mechanisms stimulated by the inhalation of PM2.5 is sufficient to activate coagulation and inhibit fibrinolysis in the lung and systemically (Budinger et al., 2011) . Meanwhile, the progressive increase of inflammation and an unusual trend of hypercoagulation has been noticed in severe and critical patients with COVID-19 . In the aspect of stress response, the possible role of endoplasmic reticulum (ER) stress (Banerjee et al., 2020) and oxidative stress (Delgado-Roche and Mesta, 2020) in COVID-19 pathogenesis and the need of its mitigation is previously suggested (Csukasi et al., 2020) while the role of ER stress and oxidative stress in the lung during PM exposure is well demonstrated (Valavanidis et al., 2013; Watterson et al., 2009) . Hence, along with the possible direct impact on the viral pathogenesis, the progression of indirect pathophysiological consequences due to PM exposure have to be considered for understanding the etiology of COVID-19. Finally, the SARS-CoV-2 modulated metabolic pathways as mentioned in the proteomic study have been prioritized with the context of PM exposure. Although the genes of metabolic pathways did not overlap with DEG from bronchial epithelial and endothelial datasets, the J o u r n a l P r e -p r o o f possible links were obtained through a literature survey. In human lung epithelial cells (A549), the citrate cycle, amino acid biosynthesis and glutathione metabolism were the 3 major metabolic pathways perturbed by PM2.5 exposure (Huang et al., 2015) . Further, seasonal PM2.5 modulated the metabolic rewiring and caused cardiac dysfunction in which a low dose of PM2.5 exposure induced the elevation of tricarboxylic acid cycle (TCA cycle), whereas a high dose of PM2.5 exposure mitigated TCA cycle, while increasing the glycolysis levels . Specifically, metabolic remodeling from oxidative phosphorylation to glycolysis was also found in human lung bronchial epithelial (BEAS-2B) cells treated with PM2.5 (Y. . On the other hand, Bojkova et al found that blocking glycolysis with hexokinase inhibitor, 2-deoxy-D-glucose (2-DG), prevented SARS-CoV-2 replication in Caco-2 cells suggesting glycolysis inhibitors as potential therapeutic agents for COVID-19 (Bojkova et al., 2020) . Therefore, the modulation of glycolysis during air particulate exposure has to be taken into account due to its supportive role in SARS-CoV-2 replication. In the exposome context, current perspectives focus on the possible role of exposome (including air pollution) in promoting resilience or susceptibility after SARS-CoV-2 infection (Naughton et al., 2020) . Apart from this, variations in pathogenic potential of airborne PM may be originated from the seasonal and compositional variability (ions and organic compounds) and also by means of its potential to cause oxidative stress and inflammation (Al Hanai et al., 2019; Park et al., 2018) . The current study spotlights the possible role of PM exposure as an encouraging factor in SARS-CoV-2 pathogenesis through the evidence of predicted overlapping system-wide molecular mechanisms. Moreover, along with the possible direct impact on viral pathogenesis, J o u r n a l P r e -p r o o f the progression of indirect pathophysiological pulmonary and vascular consequences of PM exposure which may enhance the susceptibility leading to severe outcomes after SARS-CoV-2 infection need to be emphasized (Fig. 7) . Overall, the current study suggests that airborne PM exposure in the population need to be taken into account as a co-factor in determining resilience or susceptibility during COVID-19 management. The authors declare that there are none. None. JM devised the study. JM and LS performed data analysis. JM wrote the manuscript. LS reviewed and edited the manuscript. Ontology and pathways. The association between terms enriched by differentially expressed genes in COVID-19 lung tissue, PM exposed bronchial epithelial cells -day 1 and 4 (GSE38172) and endothelial cells (GSE4567) has been illustrated. Association of enriched biological processes, pathways and molecular function among expressed genes from COVID-19 lung tissue, PM exposed bronchial epithelial cells -day 1 and 4 (GSE38172) and endothelial cells (GSE4567) has been illustrated. J o u r n a l P r e -p r o o f J o u r n a l P r e -p r o o f Seasonal variations in the oxidative stress and inflammatory potential of PM2.5 in Tehran using an alveolar macrophage model; The role of chemical composition and sources Silico Analyses of Immune System Protein Interactome Network, Single-Cell RNA Sequencing of Human Tissues, and Artificial Neural Networks Reveal Potential Therapeutic Targets for Drug Repurposing Against COVID-19 Protein kinase A-mediated CREB phosphorylation is an oxidant-induced survival pathway in alveolar type II cells Imbalanced Host Response to SARS-CoV-2 Drives Development of COVID-19 Re-analysis of SARS-CoV-2-infected host cell proteomics time-course data by impact pathway analysis and network analysis: a potential link with inflammatory response Proteomics of SARS-CoV-2-infected host cells reveals therapy targets Evidence-based considerations exploring relations between sars-cov-2 pandemic and air pollution: Involvement of PM2.5-mediated up-regulation of the viral receptor ace-2 Particulate Matter-Induced Lung Inflammation Increases Systemic Levels of PAI-1 and Activates Coagulation Through Distinct Mechanisms COVID-19: towards understanding of pathogenesis A Dual Role of Heme Oxygenase-1 in Cancer Cells eXpression2Kinases (X2K) Web: linking expression signatures to upstream cell signaling networks The Gene Expression Omnibus Database Can atmospheric pollution be considered a co-factor in extremely high level of SARS-CoV-2 lethality in Northern Italy? Should we unstress SARS-CoV-2 infected cells? oxidative stress triggers autophagy in human lung epithelial A549 cells. Toxicol. Vitr Tripartite combination of candidate pandemic mitigation agents: Vitamin D, Quercetin, and Estradiol Manifest properties of medicinal agents for targeted mitigation of the COVID-19 pandemic defined by genomics-guided tracing of SARS-CoV-2 targets in human cells Mitochondrial oxidative DNA damage and exposure to particulate air pollution in mother-newborn pairs Effect of long-term exposure to fine particulate matter on lung function decline and risk of chronic obstructive pulmonary disease in Taiwan: a longitudinal, cohort study Long-acting progestin-only contraceptives enhance human endometrial stromal cell expressed neuronal pentraxin-1 and reactive oxygen species to promote endothelial cell apoptosis Mechanism-based disease similarity Urban PM2.5 exacerbates allergic inflammation in the murine lung via a TLR2 SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor Airborne particulate matter and mitochondrial damage: A cross-sectional study Serum protein profiling reveals a landscape of inflammation and immune signaling in early-stage COVID-19 infection Clinical features of patients infected with 2019 novel coronavirus in Wuhan The effect of size-segregated ambient particulate matter on Th1/Th2-like immune responses in mice Metabolomics reveals disturbed metabolic pathways in human lung epithelial cells exposed to airborne fine particulate matter Exposure in the Respiratory System Induces Distinct Inflammatory Signaling in the Lung and the Liver of Mice ToppCluster: a multiple gene list feature analyzer for comparative enrichment clustering and network-based dissection of biological systems Up-regulation of tissue factor in human pulmonary artery endothelial cells after ultrafine particle exposure Ezrin/Radixin/Moesin Proteins Are Phosphorylated by TNF-α and Modulate Permeability Increases in Human Pulmonary Microvascular Endothelial Cells Phosphorylation of the E3 ubiquitin ligase RNF41 by the kinase Par-1b is required for epithelial cell polarity Instillation of particulate matter 2.5 induced acute lung injury and attenuated the injury recovery in ACE2 knockout mice PM2.5-induced oxidative stress increases intercellular adhesion molecule-1 expression in lung epithelial cells through the IL-6/AKT/STAT3/NF-ΚBdependent pathway Depriving Iron Supply to the Virus Represents a Promising Adjuvant Therapeutic Against Viral Survival Cell cycle alterations induced by urban PM2.5 in bronchial epithelial cells: Characterization of the process and possible mechanisms involved Moderated estimation of fold change and dispersion for RNAseq data with DESeq2 Finding disease similarity based on implicit semantic similarity Pathological inflammation in patients with COVID-19: a key role for monocytes and macrophages Enrichment Map: A Network-Based Method for Gene-Set Enrichment Visualization and Interpretation Particulate matter (PM10) enhances RNA virus infection through modulation of innate immune responses Traffic-derived particulate matter and angiotensinconverting enzyme 2 expression in human airway epithelial cells Inhibition of SARS-CoV-2 Infections in Engineered Human Tissues Using Clinical-Grade Soluble Human ACE2 The role of the exposome in promoting resilience or susceptibility after SARS-CoV-2 infection Activation of proinflammatory responses in cells of the airway mucosa by particulate matter: Oxidant-and non-oxidant-mediated triggering mechanisms COVID-19: a case for inhibiting IL-17? COVID-19 higher induced mortality in Chinese regions with lower air quality. medRxiv 1-16 Particulate matter toxicity is nrf2 and mitochondria dependent: The roles of metals and polycyclic aromatic hydrocarbons Differential toxicities of fine particulate matters from various sources The vascular endothelium: The cornerstone of organ dysfunction in severe SARS-CoV-2 infection Blocking TBK1 alleviated radiation-induced pulmonary fibrosis and epithelial-mesenchymal transition through Akt-Erk inactivation Pathway enrichment analysis and visualization of omics data using g:Profiler, GSEA, Cytoscape and EnrichmentMap SPEED2: inferring upstream pathway activity from differential gene expression MARK3-mediated phosphorylation of ARHGEF2 couples microtubules to the actin cytoskeleton to establish cell polarity Cytoscape: A software Environment for integrated models of biomolecular interaction networks Evidence for the critical role of the PI3K signaling pathway in particulate matter-induced dysregulation of the inflammatory mediators COX-2/PGE2 and the associated epithelial barrier protein Filaggrin in the bronchial epithelium Mass spectrometry-based metabolomics reveals the mechanism of ambient fine particulate matter and its components on energy metabolic reprogramming in BEAS-2B cells Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles Gene expression profiling and pathway analysis of human bronchial epithelial cells exposed to airborne particulate matter collected from Saudi Arabia SARS-CoV-2 entry factors are highly expressed in nasal epithelial cells together with innate immune genes Mitochondrial DNA damage and subsequent activation of Z-DNA binding protein 1 links oxidative stress to inflammation in epithelial cells STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets The trinity of COVID-19: immunity, inflammation and intervention Crosstalk between NRF2 and HIPK2 shapes cytoprotective responses Pulmonary oxidative stress, inflammation and cancer: Respirable particulate matter, fibrous dusts and ozone as major causes of lung carcinogenesis through reactive oxygen species mechanisms Endothelial cell infection and endotheliitis in COVID-19 Clinical Characteristics of 138 Hospitalized Patients with 2019 Novel Coronavirus-Infected Pneumonia in Wuhan, China Urban particulate matter triggers lung inflammation via the ROS-MAPK-NF-κB signaling pathway Interleukin-17 induces human alveolar epithelial to mesenchymal cell transition via the TGF-β1 mediated Smad2/3 and ERK1/2 activation PM2.5 induces ferroptosis in human endothelial cells through iron overload and redox imbalance Urban Particulate Matter Causes ER Stress and the Unfolded Protein Response in Human Lung Cells WHO COVID-19 Statistics Risk Factors Associated With Acute Respiratory Distress Syndrome and Death in Patients With Coronavirus Disease Exposure to air pollution and COVID-19 mortality in the United States: A nationwide cross-sectional study Inflammatory health effects of indoor and outdoor particulate matter Industrial PM2.5 cause pulmonary adverse effect through RhoA/ROCK pathway Involvement of cigarette smoke-induced epithelial cell ferroptosis in COPD pathogenesis The Pivotal Role of TBK1 in Inflammatory Responses Mediated by Macrophages Cardiac dysfunction and metabolic remodeling due to seasonally ambient fine particles exposure Analysis of Genes Related to Angiotensin II-Induced Arterial Injury Using a Time Series Microarray Online Analysis Of Differential Gene Expression With Comprehensive Options Of Statistical Methods. bioRxiv 122747 As a downstream target of the AKT pathway, NPTX1 inhibits proliferation and promotes apoptosis in hepatocellular carcinoma Transcriptomic Analyses of the Biological Effects of Airborne PM2.5 Exposure on Human Bronchial Epithelial Cells Association between short-term exposure to air pollution and COVID-19 infection: Evidence from China Credit Author Statement Jeganathan Manivannan: Study design, Data analysis Considerable overlapping of upstream pathways associated with upregulated genes during SARS-CoV-2 infection with that of PM exposure 3. Differentially expressed pathways during PM exposure may increase COVID-19 severity 4. PM exposure has direct effects on COVID-19 infections as evident by overlapping molecular mechanisms ☒ The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.☐The authors declare the following financial interests/personal relationships which may be considered as potential competing interests:J o u r n a l P r e -p r o o f