key: cord-0842913-mzgeqa79 authors: Lee, Abby C.; Castaneda, Grant; Li, Wei Tse; Chen, Chengyu; Shende, Neil; Chakladar, Jaideep; Taub, Pam R.; Chang, Eric Y.; Ongkeko, Weg M. title: COVID-19 Severity Potentially Modulated by Cardiovascular-Disease-Associated Immune Dysregulation date: 2021-05-28 journal: Viruses DOI: 10.3390/v13061018 sha: f5ebdee0f1f57f146c5c3e544d0341c157dad3cf doc_id: 842913 cord_uid: mzgeqa79 Patients with underlying cardiovascular conditions are particularly vulnerable to severe COVID-19. In this project, we aimed to characterize similarities in dysregulated immune pathways between COVID-19 patients and patients with cardiomyopathy, venous thromboembolism (VTE), or coronary artery disease (CAD). We hypothesized that these similarly dysregulated pathways may be critical to how cardiovascular diseases (CVDs) exacerbate COVID-19. To evaluate immune dysregulation in different diseases, we used four separate datasets, including RNA-sequencing data from human left ventricular cardiac muscle samples of patients with dilated or ischemic cardiomyopathy and healthy controls; RNA-sequencing data of whole blood samples from patients with single or recurrent event VTE and healthy controls; RNA-sequencing data of human peripheral blood mononuclear cells (PBMCs) from patients with and without obstructive CAD; and RNA-sequencing data of platelets from COVID-19 subjects and healthy controls. We found similar immune dysregulation profiles between patients with CVDs and COVID-19 patients. Interestingly, cardiomyopathy patients display the most similar immune landscape to COVID-19 patients. Additionally, COVID-19 patients experience greater upregulation of cytokine- and inflammasome-related genes than patients with CVDs. In all, patients with CVDs have a significant overlap of cytokine- and inflammasome-related gene expression profiles with that of COVID-19 patients, possibly explaining their greater vulnerability to severe COVID-19. In December 2019, widespread infection by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was reported in Wuhan, China [1] . Since then, SARS-CoV-2, which causes COVID-19, has spread rapidly, and COVID-19 was declared a pandemic by the World Health Organization (WHO) on 11 March 2020 [2] Current research suggests that patients with existing comorbidities, including hypertension, cardiovascular disease, diabetes, and obesity are more likely to develop severe COVID-19 [3] [4] [5] . COVID-19 has also been known to induce myocardial injury, arrhythmia, acute coronary syndrome, and venous thromboembolism (VTE) [6] [7] [8] . Such cardiovascular damage has been attributed to cytokine storms triggered by the SARS-CoV-2 infection that can cause multi-organ damage [9, 10] . Additionally, COVID-19 patients experience coagulation abnormalities, possibly leading to an increased risk of thromboembolic events [11] . In multiple autopsy studies, RNA-sequencing data were obtained from the following datasets: GSE116250 [40] , GSE19151 [41] , GSE90074 [42] , and SRP262885 [43] . GSE116250, provided by Sweet et al., consists of the RNA sequencing of human left ventricular samples from 14 patients with no major cardiac history (nonfailing), 37 patients with NIDCM, and 13 patients with ICM. GSE19151, provided by Lewis et al., consists of the high-throughput sequencing of whole blood samples from 63 healthy controls, 23 patients with single VTE, and 17 patients with recurrent VTE on warfarin. GSE90074 consists of the RNA-sequencing data of PBMCs from 93 patients with and 50 patients without CAD. Lastly, SRP262885 consists of the RNA-sequencing data of platelets from 10 COVID-19 subjects and 5 healthy controls. For the cardiomyopathy and VTE cohorts, the Kruskal-Wallis test (p < 0.05) was used to determine differentially expressed genes. CAD cohorts were analyzed using the GEO2R software, which employs the limma (linear models for microarray analysis) R package (p < 0.05). Differential expression was applied to the COVID-19 platelet data to determine the genes that were differentially expressed (p < 0.05). To correlate gene expression to immune-associated signatures, gene set enrichment analysis (GSEA) was utilized. Pathways were chosen from the C2: CP set of signatures from the Molecular Signatures Database [44] . Signatures that were significantly enriched were those with a nominal p-value < 0.05. The CIBERSORTx algorithm was used to deconvolute the RNA-sequencing data to estimate the infiltration levels of 22 immune cell types [45] . Gene dysregulation was determined by comparing COVID-19 and cardiomyopathy samples to healthy controls for each study. Cardiomyopathy samples were separated into patients with ischemic cardiomyopathy (ICM) or nonischemic dilated cardiomyopathy (NIDCM). The two groups were individually compared against samples from patients with no major cardiovascular disease (healthy controls). We found a significant overlap between COVID-19 patients and ICM and NIDCM patients' immune-associated (IA) gene expression. About half of the IA genes dysregulated in COVID-19 are dysregulated in either or both types of cardiomyopathy ( Figure 1A) . A complete list of dysregulated IA genes are found in Appendix A, Table A1 . Cytokine-related genes that are dysregulated in both cardiomyopathy patients and COVID-19 patients include chemokines (CCL3, CCL4, CXCL4, etc.), interleukins or interleukin receptors (IL15, IL20RA, etc.), and genes in the transforming growth factor beta (TGFB) family. The inflammasome-related genes include genes in the caspase family (CASP2, CASP9, etc.), mitogen-activated protein kinase (MAPK)-related genes, and nuclear factor-kB (NF-kB) regulators (IKBKG, NFKBIA, etc.). IA gene dysregulation was very similar between dilated and ischemic cardiomyopathies. We observed that a significant number of IA genes were dysregulated in either of the cardiomyopathies but not in COVID-19 ( Figure 1A) . Interestingly, we found that most of the genes dysregulated in both COVID-19 and cardiomyopathy were dysregulated to a greater degree in COVID-19 than in cardiomyopathy samples. This was observed for TGFB3, CCL4, IL15, and IL20RA in both ICM samples vs. COVID-19 samples and NIDCM samples vs. COVID-19 samples ( Figure 1B ). Furthermore, these dysregulated genes appeared to be similarly dysregulated in COVID-19 and corresponding healthy samples ( Figure 1C ). In contrast, these genes' expression in cardiomyopathy samples and corresponding healthy samples were only sometimes similar, without overwhelming differences in expression levels between the two cohorts ( Figure 1C ). Therefore, we believe that these dysregulated genes are dysregulated to a greater degree in COVID-19 than in cardiomyopathy. The inflammasome-associated genes dysregulated in both COVID-19 and cardiomyopathy were upregulated in both conditions ( Figure 1C ), suggesting that they may upregulate inflammation through similar pathways. Interestingly, we found that most of the genes dysregulated in both COVID-19 and cardiomyopathy were dysregulated to a greater degree in COVID-19 than in cardiomyopathy samples. This was observed for TGFB3, CCL4, IL15, and IL20RA in both ICM samples vs. COVID-19 samples and NIDCM samples vs. COVID-19 samples ( Figure 1B ). Furthermore, these dysregulated genes appeared to be similarly dysregulated in COVID-19 and corresponding healthy samples ( Figure 1C ). In contrast, these genes' expression in cardiomyopathy samples and corresponding healthy samples were only sometimes similar, without overwhelming differences in expression levels between the two cohorts (Figure 1C) . Therefore, we believe that these dysregulated genes are dysregulated to a greater degree in COVID-19 than in cardiomyopathy. The inflammasome-associated genes dysregulated in both COVID-19 and cardiomyopathy were upregulated in both conditions ( Figure 1C ), suggesting that they may upregulate inflammation through similar pathways. We discovered that the levels of T and B cells were unchanged in healthy vs. COVID-19 patients (See Appendix A, Figure A1A ). The most noticeable change in immune cell abundance occurred in macrophages for COVID-19 patients, where M0 macrophage levels were dramatically reduced and M1 and M2 macrophage levels were slightly increased (See Appendix A, Figure A1A ). Both cardiomyopathies elicited greater immune cell abundance changes than COVID-19, with the changes being more pronounced for ICM. The levels of M1 and M2 macrophages increased in ICM, similar to what was observed for COVID-19 (See Appendix A, Figure A1B ). The levels of T-and B-cell subtypes changed more dramatically in ICM and NIDCM than in COVID-19. In summary, the levels of inflammatory macrophages increased for both cardiomyopathy and COVID-19 patients, while the levels of other immune cell types did not correlate between the two conditions. We analyzed genes that are dysregulated in both COVID-19 and cardiomyopathy to assess if they dysregulate common pathways in the two conditions. Interleukin 1 receptorassociated kinase 2 (IRAK2), upregulated in both COVID-19 and ICM, was associated with the upregulation of the FCER1 and TP63 pathways, both of which are associated with inflammation and immune activation (See Appendix A, Figure A2A ) [46, 47] . IRAK2 is a promoter of NF-kB signaling [48] . Caspase 2 (CASP2), also upregulated in COVID-19 and cardiomyopathy, is associated with the downregulation of IFIH, which is capable of recognizing viruses and inducing inflammation [49, 50] . Finally, CYLD lysine 63 deubiquitinase (CYLD) was correlated with multiple identical pathways for both COVID-19 samples and ICM samples. CYLD is upregulated in both COVID-19 and cardiomyopathy and was found to correlate with the activation of FGFR2, an important promoter of inflammation [51] , and TXA2, a gene that is upregulated in platelets (See Appendix A, Figure A2A ) [52] . CYLD is an inhibitor of inflammation [53] . Since the majority of correlations were between IA genes and pro-inflammatory pathways and signatures, the dysregulation of CYLD represents an exception, and we hypothesize that CYLD may be expressed as a response to attenuate excessive inflammation. We found that the overwhelming majority of pathways that correlated with dysregulated genes in both COVID-19 and NIDCM are associated with CYLD, and these pathways are primarily pro-plotting, pro-cell aggregation, and pro-inflammation (See Appendix A, Figure A2A ), supporting the possibility that CYLD is released in response to inflammation. We compared the immune landscape between COVID-19 samples and blood samples from VTE patients to find similarities in IA gene and pathway expression. VTE patients were classified into single occurrence VTE (single VTE) and recurrent VTE. Compared to the similarities in IA genes dysregulated between COVID-19 and cardiomyopathy, the similarities between COVID-19 and VTE are less pronounced. Two cytokine-associated genes (CCL4 and CD40) were dysregulated in COVID-19 and single VTE, and one cytokine-associated gene (CCL4) was dysregulated in COVID-19 and recurrent VTE (Figure 2A) . A complete list of dysregulated cytokine-and inflammasomeassociated genes are found in Appendix A, Table A2 . CCL4 recruits immune cells, including macrophages, monocytes, and T cells [54] , suggesting that COVID-19 and VTE may both exhibit the increased recruitment of inflammatory immune cells. The upregulation of CCL4 was much greater in COVID-19 than in VTE, however ( Figure 2B ). Two inflammasomerelated genes were found to be dysregulated in VTE and COVID-19. BCL2L1 is known to be highly upregulated in inflamed tissue [55] , and it was found to be upregulated in COVID-19 and both single and recurrent VTE ( Figure 2C ), while CASP4 directs the noncanonical upregulation of inflammasomes [56] . Interestingly, CASP4 was found to be upregulated in both COVID-19 and recurrent VTE but downregulated in single VTE ( Figure 2C ), which suggests that the gene could contribute to the development of recurrent VTE. found to be upregulated in both COVID-19 and recurrent VTE but downregulated in single VTE ( Figure 2C ), which suggests that the gene could contribute to the development of recurrent VTE. We found that naive B cells were dramatically reduced in abundance in VTE patients, which may indicate adaptive immune activation (See Appendix A, Figure A1C ). This was the only significant immune cell population change in VTE patients that was observed and does not correlate to changes in COVID-19. BCL2L1 and CASP4 were the only genes found to be dysregulated in both COVID-19 and VTE and also correlated with similar pathways in both patient cohorts ( Figure 3A ,B). BCL2L1 was found to be upregulated in COVID-19 and both VTE cohorts ( Figure 3C ). However, the direction of correlation to pathways was the complete opposite between COVID-19 and recurrent VTE (See Appendix A, Figure A2C ). The high correlation We found that naive B cells were dramatically reduced in abundance in VTE patients, which may indicate adaptive immune activation (See Appendix A, Figure A1C ). This was the only significant immune cell population change in VTE patients that was observed and does not correlate to changes in COVID-19. BCL2L1 and CASP4 were the only genes found to be dysregulated in both COVID-19 and VTE and also correlated with similar pathways in both patient cohorts ( Figure 3A ,B). BCL2L1 was found to be upregulated in COVID-19 and both VTE cohorts ( Figure 3C ). However, the direction of correlation to pathways was the complete opposite between COVID-19 and recurrent VTE (See Appendix A, Figure A2C ). The high correlation strength for each cohort suggests BCL2L1 is involved in both COVID-19 and recurrent VTE but functions in opposite ways. On the other hand, CASP4 expression was correlated with over 10 pathways in the same direction for both COVID-19 and recurrent VTE (See Appendix A, Figure A2C ). It was also found to be upregulated in both COVID-19 and recurrent VTE ( Figure 3C ). The pathways correlated with CASP4 were immune related (antigen process- ing and cross presentation) and general metabolism related (ABC transporter, oxidative phosphorylation). Therefore, while CASP4 likely functions similarly in COVID-19 and recurrent VTE, its precise role requires further investigation. strength for each cohort suggests BCL2L1 is involved in both COVID-19 and recurrent VTE but functions in opposite ways. On the other hand, CASP4 expression was correlated with over 10 pathways in the same direction for both COVID-19 and recurrent VTE (See Appendix A, Figure A2C ). It was also found to be upregulated in both COVID-19 and recurrent VTE ( Figure 3C ). The pathways correlated with CASP4 were immune related (antigen processing and cross presentation) and general metabolism related (ABC transporter, oxidative phosphorylation). Therefore, while CASP4 likely functions similarly in COVID-19 and recurrent VTE, its precise role requires further investigation. We found a significant overlap in IA gene expression in COVID-19 and CAD. About a third of the IA genes dysregulated in CAD were also found to be dysregulated in COVID-19 ( Figure 4A ). The complete list of dysregulated cytokine and inflammasomeassociated genes are found in Appendix A, Table A3 . We found a significant overlap in IA gene expression in COVID-19 and CAD. About a third of the IA genes dysregulated in CAD were also found to be dysregulated in COVID-19 ( Figure 4A ). The complete list of dysregulated cytokine and inflammasome-associated genes are found in Appendix A, Table A3 . Cytokine-related genes that were found to be dysregulated in both CAD patients and COVID-19 patients include chemokines (CCL3 and CCL4), chemokine receptor CXCR1, and a TNFSF gene, LTB. The inflammasome-related genes that were found include NF-kB regulators (NFKBIA and CHUK), an alpha arrestin (TXNIP), and an F-BAR domaincontaining protein (PSTPIP1). Similar to cardiomyopathy, we found that most genes dysregulated in both COVID-19 and CAD were dysregulated to a greater degree in COVID-19 samples than in CAD samples. This was observed for CCL3, CCL4, CXCR1, and LTB ( Figure 4B ). Similar to COVID-19 patients, the memory B cells in CAD patients were more abundant (See Appendix A, Figure A1D ). We analyzed genes that are dysregulated in both COVID-19 and CAD to assess if they are associated with similar pathways in the two conditions. Notably, we discovered that CHUK, PSTPIP1, and CCL3, upregulated in both COVID-19 and CAD, were associated with the upregulation of many inflammatory pathways in both conditions, including the IL12, IL10, IL23, and P53 regulation pathways (See Appendix A, Figure A2C ). In this project, we characterized the immune landscape of cardiomyopathy, VTE, CAD, and COVID-19 patients, drawing important parallels between COVID-19 and cardiovascular-disease-mediated immune dysregulation. Of the four genes that were more severely dysregulated in COVID-19 compared to cardiomyopathy, two were reported to be dysregulated in COVID-19 patients: pro-inflammatory CCL4 was highly expressed in the bronchoalveolar lavage fluid of COVID-19 patients [57] , and IL15 modulates inflammation and functions in viral clearance [58, 59] . In fact, IL15 is part of an immune-based biomarker signature associated with mortality in COVID-19 patients, and CCL4 has been shown to be elevated in COVID-19 patients who eventually died due to the disease [60] . As we report that these cytokines are also upregulated in patients with cardiomyopathy, it is possible that such pre-existing immune dysregulation could explain the higher COVID-19 mortality rates of patients with cardiomyopathy and COVID-19. Our findings on immune cell abundance in COVID-19 and cardiomyopathy patients also point to a more robust innate immune response in COVID-19 patients, which is plausible as research has shown that a hyperinflammatory innate immune response coupled with an impaired adaptive immune response may lead to tissue damage in COVID-19 patients [59] [60] [61] . Conversely, the elevated levels of T and B cells in cardiomyopathy patients indicate a stronger adaptive immune response, which is now considered an increasingly important factor in Cytokine-related genes that were found to be dysregulated in both CAD patients and COVID-19 patients include chemokines (CCL3 and CCL4), chemokine receptor CXCR1, and a TNFSF gene, LTB. The inflammasome-related genes that were found include NF-kB regulators (NFKBIA and CHUK), an alpha arrestin (TXNIP), and an F-BAR domaincontaining protein (PSTPIP1). Similar to cardiomyopathy, we found that most genes dysregulated in both COVID-19 and CAD were dysregulated to a greater degree in COVID-19 samples than in CAD samples. This was observed for CCL3, CCL4, CXCR1, and LTB ( Figure 4B ). Similar to COVID-19 patients, the memory B cells in CAD patients were more abundant (See Appendix A, Figure A1D ). We analyzed genes that are dysregulated in both COVID-19 and CAD to assess if they are associated with similar pathways in the two conditions. Notably, we discovered that CHUK, PSTPIP1, and CCL3, upregulated in both COVID-19 and CAD, were associated with the upregulation of many inflammatory pathways in both conditions, including the IL12, IL10, IL23, and P53 regulation pathways (See Appendix A, Figure A2C ). In this project, we characterized the immune landscape of cardiomyopathy, VTE, CAD, and COVID-19 patients, drawing important parallels between COVID-19 and cardiovascular-disease-mediated immune dysregulation. Of the four genes that were more severely dysregulated in COVID-19 compared to cardiomyopathy, two were reported to be dysregulated in COVID-19 patients: pro-inflammatory CCL4 was highly expressed in the bronchoalveolar lavage fluid of COVID-19 patients [57] , and IL15 modulates inflammation and functions in viral clearance [58, 59] . In fact, IL15 is part of an immune-based biomarker signature associated with mortality in COVID-19 patients, and CCL4 has been shown to be elevated in COVID-19 patients who eventually died due to the disease [60] . As we report that these cytokines are also upregulated in patients with cardiomyopathy, it is possible that such pre-existing immune dysregulation could explain the higher COVID-19 mortality rates of patients with cardiomyopathy and COVID-19. Our findings on immune cell abundance in COVID-19 and cardiomyopathy patients also point to a more robust innate immune response in COVID-19 patients, which is plausible as research has shown that a hyperinflammatory innate immune response coupled with an impaired adaptive immune response may lead to tissue damage in COVID-19 patients [59] [60] [61] . Conversely, the elevated levels of T and B cells in cardiomyopathy patients indicate a stronger adaptive immune response, which is now considered an increasingly important factor in cardiovascular disease pathogenesis [62] [63] [64] . Comparing COVID-19 and cardiomyopathy patients, we found elevated levels of inflammatory macrophages in both groups of patients. This could suggest that cardiomyopathy patients are more susceptible to hyperinflammation in COVID-19 and are thus more likely to progress to severe COVID-19. We then analyzed overlapping gene expression pathway dysregulation between cardiomyopathy and COVID-19 patients. Upregulated CASP2 and IRAK2 are of particular interest due to their inflammatory roles. CASP2 is a pro-inflammatory gene and IRAK2 promotes NF-kB, which is a central activator of inflammation. Overall, genes dysregulated in both cardiomyopathy and COVID-19 appear to promote inflammation, which may indicate why cardiovascular disease patients experience poorer clinical outcomes, as greater inflammation correlates with severity and death in COVID-19 [56] . Exploring immune-associated (IA) gene dysregulation in VTE and COVID-19 revealed several IA genes dysregulated in both conditions. Cytokine CCL4 has been shown to be upregulated in COVID-19 patients [65] and in patients who develop cardiovascular diseases [66] . Inflammasome-related genes BCL2L1 and CASP4 are tied to inflammatory caspases. CASP4 is an inflammatory caspase and promotes pro-inflammatory cytokine secretion [67] . Conversely, BCL2L1 inhibits caspase release. With both genes being upregulated in VTE and COVID-19, future analysis must be carried out to examine how these genes function differently in VTE and COVID-19. Interestingly, in both COVID-19 and single VTE, BCL2L1 expression is negatively correlated to canonical pathway expression, but in recurring VTE they are positively correlated. CASP4, on the other hand, only has overlapping significant canonical pathways with COVID-19 in recurrent VTE. From these pathways, we observe that CASP4 functions similarly in recurrent VTE and COVID-19. Together, these results show that VTE and COVID-19 patients share similar upregulation of inflammation-associated genes, which could explain why rates of venous thromboembolism events are higher in COVID-19 patients, as well as why venous thromboembolism events are associated with higher risk of death in COVID-19 patients [68, 69] . Lastly, we compared the immune landscape and canonical pathways of CAD and COVID-19 patients. Of the significantly dysregulated cytokines in both COVID-19 and CAD, pro-inflammatory cytokines CCL3 and CCL4 have been associated with COVID-19 severity [59] . Of the inflammasome-related genes, CHUK is of particular interest. CHUK forms part of the IκB kinase (IKK) complex that is involved in the phosphorylation and degradation of IκBα, allowing for the transcription of NF-kB-dependent genes. Following coronavirus infection, the NF-kB pathway is activated via the MyD88 pathway [70] , and increased transcription of NF-kB-dependent genes has implications for cardiomyopathy, atherosclerosis, and COVID-19 severity. Specifically, NF-kB activation in endothelial cells triggers the expression of adhesion molecules that are responsible for the invasion and homing of macrophages [71] [72] [73] [74] [75] [76] , contributing to atherosclerosis pathogenesis [77] . In addition, TNFa and IL6 expressions have been shown to be triggered by SARS via the NF-kB pathway [78] . These cytokines have been implicated in macrophage activation syndrome and cytokine storms and are associated with COVID-19 severity [79] [80] [81] . Interestingly, IRAK2, another NF-kB pathway regulator, is upregulated in cardiomyopathy patients. IRAK2, when phosphorylated with IRAK1 and IRAK4, recruits Ub ligase and activates TRAF6. TRAF6 activates the NF-kB pathway via the IKK complex. In summary, COVID-19 upregulates both IRAK2 and CHUK, while atherosclerosis only upregulates CHUK, and cardiomyopathy upregulates IRAK2, suggesting that NF-kB activation may be critical in all three conditions. Given that hyperactivation of the NF-kB pathway in B cells has been implicated in cytokine storms and the pathogenesis of severe and critical COVID-19 [82] , our results suggest that the upregulation of this pathway in patients with pre-existing cardiovascular disease could be key to explaining their poorer COVID-19 prognoses. In conclusion, we found that cardiomyopathy, VTE, and CAD patients display significant similarities in inflammation-related gene expression to COVID-19 patients. Therefore, when a patient with the above cardiovascular conditions contracts COVID-19, COVID-19 could further dysregulate the expression of inflammatory genes already dysregulated, leading to more severe inflammation. This may explain why patients with cardiovascular disease are more likely to develop severe COVID-19 and tend to have poorer clinical outcomes [83, 84] . Furthermore, we found that patients with CAD display a similar dysregulated immune landscape to COVID-19 patients, possibly indicating why CAD patients are at higher risk of severe COVID-19. Interestingly, cardiomyopathy patients display more similar immune dysregulation to COVID-19 patients than VTE or CAD patients vs. COVID-19 patients. This observation could explain the fact that COVID-19 mortality is increased in congestive heart failure patients, as demonstrated in a study of 31,461 adults [85] . Our findings suggest that investigating the relationships between specific cardiovascular diseases and COVID-19 severity and mortality is meaningful and offers insight into COVID-19 immune dysregulation. However, our study has several limitations. We had limited COVID-19 platelet data, specifically normal patients. This may have impacted our differential expression analysis and thus reduced the statistical power of our analysis. However, the direction of dysregulation of many of the genes identified was consistent with existing literature. Additionally, we used platelet data instead of blood samples. To validate our results, in vitro and in vivo experiments can be done. Despite these limitations, we believe our study advances our understanding of the relationship between cardiovascular disease and COVID-19. Our study also encourages the examination of potential treatment strategies such as anti-inflammatory steroids and ACE2 inhibitors to downregulate inflammation in COVID-19 patients with CVDs. TNF CCL3 CCL2 MAPK3 CCL11 CXCR4 TNF CCL3 CD40 MAPK8 CCL21 IFNA14 TNFSF12 CCL4 CKLF MAPK8 CCL17 EPO TNFSF11 CCL4 CXCL5 NAIP CCL22 IL11 TNFSF13 CD40 CXCL5 MAPK9 CCL2 IFNA14 TNFSF12 CXCL3 CXCR1 NFKBIB CCL24 IL11RA TNFSF13B CXCL3 EPOR NAIP CCL22 IL10 TNFSF13B IL15 EPOR POLR2J4 CCL5 IL12A TNFSF8 CXCR1 IL15RA NFKBIA CCL24 IL11 TNFSF14 IL20RA IL1RN RELA CCL8 IL16 TNFSF9 IL15 IL1RN POLR2J4 CCL5 IL11RA BIRC2 IL20RB LTB SUGT1 CCR10 IL17B APP IL20RA IL20RB PSTPIP1 CCL8 IL12A BIRC3 TGFB3 LTBR TAB1 CCR3 IL17C BIRC2 LTB TGFB1 RELA CCR1 IL13 CARD8 CASP2 TGFB1 TAB3 CCR4 IL17D CARD8 LTBR TGFBR2 SUGT1 CCR10 IL15RA CCL11 CASP9 CCL2 TNFAIP3 CCR7 IL1A CCL5 TGFB3 BCL2L1 TAB1 CCR3 IL16 CCL2 CYLD CKLF TRAF6 CD27 IL1B CCL8 CASP2 BIRC3 TAB3 CCR4 IL17B CCL5 HSP90AB1 IL15RA UBE2N CD4 IL2 CXCL1 CASP9 CARD6 TMEM189 CCR7 IL17D CCL8 IKBKG TGFBR2 BIRC3 CKLF IL23A CXCL2 CYLD CASP4 TRAF6 CD27 IL18 CXCL2 IRAK2 BCL2L1 HSP90AA1 CX3CL1 IL25 HSP90AA1 HSP90AB1 CASP8 TXN CD4 IL20 HSP90AA1 MAPK12 CARD6 IRAK1 CXCL1 IL27 IL1B IKBKB CHUK TXNIP CKLF IL23A IL18 NFKBIA CASP4 MAPK1 CXCL10 IL33 IL6 IKBKG HSP90AA1 UBE2N CX3CL1 IL25 IRAK1 NOD1 CASP8 MAPK10 CXCL14 IL34 MAPK1 IRAK2 HSP90B1 CXCL14 IL33 MAP3K7 TRIP6 CHUK MAPK9 CXCL16 IL6 MAPK11 MAPK12 IRAK1 CXCL16 OSMR MAPK1 TXNIP HSP90B1 PSTPIP1 CXCL2 IL9R MAPK9 NFKBIB MAPK1 CXCL2 TGFB2 MAPK10 IKBKB TMEM189 CXCL9 OSM NFKB1 NOD1 MAPK10 CXCL9 TGFBR1 MAPK9 MAPK13 TXN CXCR2 TGFB2 NLRC4 TNFAIP3 MAPK13 CXCR2 TGFBR2 NLRC4 MAPK14 CXCR3 TGFBR1 NLRP3 TRIP6 MAPK14 CXCR3 TGFBR3 NLRP1 MAPK3 TAB2 NLRP3 TAB3 PSTPIP1 TNF PYCARD TRAF6 TAB2 XIAP TMEM189 --------TNF ---TXN Table A2 . Complete list of dysregulated cytokine and inflammasome-related genes. Single VTE and COVID-19 Recurrent VTE Only CCL1 CCL4 CCL2 MAPK8 IKBKB CD27 CCL4 CCL2 MAPK3 IKBKB CCL16 CD40 CCL3 NAIP IKBKG CXCL10 BCL2L1 CCL3 MAPK8 IKBKG CCL18 BCL2L1 CKLF NFKBIA IRAK1 CXCL8 CASP4 CD40 NAIP IRAK1 CCL7 CXCL3 NFKBIB IRAK2 IFNG CKLF NFKBIA IRAK2 CD27 CXCL5 NOD1 TAB1 IL13 CXCL3 NFKBIB TAB1 CXCL10 CXCR1 POLR2J4 TAB3 IL16 CXCL5 NOD1 TAB3 CXCL8 EPOR PSTPIP1 TMEM189 IL1A CXCR1 POLR2J4 TMEM189 IL13 IL15 SUGT1 TNFAIP3 IL3 EPOR PSTPIP1 TNFAIP3 IL16 IL15RA CASP4 TRAF6 TNFSF10 IL15 SUGT1 TRAF6 IL1A IL1RN MAPK1 TRIP6 MAPK1 IL15RA MAPK1 TRIP6 IL3 IL20RA MAPK9 TXN MAPK9 IL1RN MAPK9 TXN TNFSF10 IL20RB RELA TXNIP RELA IL20RA RELA TXNIP IFNA5 LTB BIRC3 UBE2N BIRC2 IL20RB BIRC3 UBE2N BCL2 LTBR CARD6 CARD8 LTB CARD6 CASP5 TGFB1 CASP2 CASP5 LTBR CASP2 IL18 TGFB3 CASP8 IL18 TGFB1 CASP8 NLRP3 TGFBR2 CASP9 IL1B TGFB3 CASP9 NOD2 MAPK10 CHUK NLRP3 TGFBR2 CHUK CASP4 MAPK12 CYLD MAPK10 CYLD MAPK1 MAPK13 HSP90AA1 MAPK12 HSP90AA1 MAPK9 MAPK14 HSP90AB1 MAPK13 HSP90AB1 RELA MAPK3 HSP90B1 MAPK14 HSP90B1 Table A3 . Complete list of dysregulated cytokine and inflammasome-related genes. CCR4 CCL3 CCL2 CASP4 MAPK9NAIP CD27 CCL4 CKLF CASP8 NFKBIB CD40 CXCR1 CXCL3 CASP9 NOD1 CXCR3 LTB CXCL5 CYLD POLR2J4 CXCR4 CHUK EPOR HSP90AA1 RELA IL17B NFKBIA IL15 HSP90AB1 SUGT1 IL21 PSTPIP1 IL15RA HSP90B1 TAB1 IL6 TXNIP IL1RN IKBKB TAB3 OSM IL20RA IKBKG TMEM189 TNFSF10 IL20RB IRAK1 TNFAIP3 TNFSF13B LTBR IRAK2 TRAF6 TNFSF14 TGFB1 MAPK1 TRIP6 CASP1 TGFB3 MAPK10 TXN CASP5 TGFBR2 MAPK12 UBE2N MAPK11 BCL2L1 MAPK13 NLRP3 BIRC3 MAPK14 PYCARD CARD6 MAPK3 TAB2 CASP2 MAPK8 A Novel Coronavirus from Patients with Pneumonia in China A. microRNAs and the immune response Prevalence of comorbidities and its effects in patients infected with SARS-CoV-2: A systematic review and meta-analysis Impact of cerebrovascular and cardiovascular diseases on mortality and severity of COVID-19-systematic review, meta-analysis, and meta-regression Predictors of mortality in hospitalized COVID-19 patients: A systematic review and meta-analysis Association of Cardiac Injury with Mortality in Hospitalized Patients with COVID-19 in Wuhan, China Cardiovascular Implications of Fatal Outcomes of Patients With Coronavirus Disease 2019 (COVID-19) Characteristics and clinical significance of myocardial injury in patients with severe coronavirus disease 2019 The trinity of COVID-19: Immunity, inflammation and intervention In the eye of the COVID-19 cytokine storm Thromboinflammation and the hypercoagulability of COVID-19 Autopsy Findings and Venous Thromboembolism in Patients With COVID-19: A Prospective Cohort Study Viral presence and immunopathology in patients with lethal COVID-19: A prospective autopsy cohort study Critical Cytokine Pathways to Cardiac Inflammation Regulatory Mechanisms of the NLRP3 Inflammasome, a Novel Immune-Inflammatory Marker in Cardiovascular Diseases NLRP3 inflammasome-mediated pyroptosis contributes to the pathogenesis of non-ischemic di-lated cardiomyopathy Endothelial Dysfunction and Venous Thrombosis SARS-CoV-2 infection pathogenesis is related to oxidative stress as a response to aggression Endothelial Dysfunction in COVID-19: Lessons Learned from Coronaviruses Deep Vein Thrombosis in Hospitalized Patients With COVID-19 in Wuhan Thrombosis in Hospitalized Patients With COVID-19 in a New York City Health System The management of venous thromboembolism in hospitalized patients with COVID-19 Monocytes, neutrophils, and platelets cooperate to initiate and propagate venous thrombosis in mice in vivo Neutrophil extracellular traps promote deep vein thrombosis in mice Neutrophil Extracellular Trap (NET) Impact on Deep Vein Thrombosis Pathogenic T cells and inflammatory monocytes incite inflammatory storm in severe COVID-19 patients COVID-19 infection induces readily detectable morphological and inflammation-related phenotypic changes in peripheral blood monocytes, the severity of which correlate with patient outcome Clinical Characteristics of 138 Hospitalized Patients With 2019 Novel Coronavirus-Infected Pneumonia in Accelerated atherosclerosis in patients with SLE-mechanisms and management Direct Proinflammatory Effect of C-Reactive Protein on Human Endothelial Cells Increased adhesiveness of white blood cells in patients with unstable angina: Additional evidence for an involvement of the immune-inflammatory system Serum Cytokine Profile in Relation to the Severity of Coronary Artery Disease Elevated level of C-reactive protein may be an early marker to predict risk for severity of COVID-19 Clinical features in 52 patients with COVID-19 who have increased leukocyte count: A retrospective analysis Cytokine Storm in COVID-19: The Current Evidence and Treatment Strategies COVID-19 and vascular disease COVID-19 and cardiovascular disease: From basic mechanisms to clinical perspectives Comorbidity and its impact on 1590 patients with COVID-19 in China: A nationwide analysis Whole blood gene expression analyses in patients with single versus recurrent venous thromboembolism Clinical Evidence Supports a Protective Role for CXCL5 in Coronary Artery Disease RNA-seq of Platelets from SARS-CoV-2 Covid-19 Molecular signatures database (MSigDB) 3.0 Profiling Tumor Infiltrating Immune Cells with CIBERSORT The role of FcepsilonRI expressed in dendritic cells and monocytes Regulation of the interferon-gamma (IFN-gamma) pathway by p63 and Delta133p53 isoform in different breast cancer subtypes IRAK-2 participates in multiple toll-like receptor signaling pathways to NFkappaB via activation of TRAF6 ubiquitination Incoming RNA Virus Nucleocapsids Containing a 5 -Triphosphorylated Genome Activate RIG-I and Antiviral Signaling Intrinsic FGFR2 and Ectopic FGFR1 Signaling in the Prostate and Prostate Cancer Deubiquitination of NLRP6 inflammasome by Cyld critically regulates intestinal inflammation Chemokines enhance immunity by guiding naive CD8+ T cells to sites of CD4+ T cell-dendritic cell interaction Persistent Inflammatory Pathways Associated with Early Onset Myocardial Infarction in a Medicated Multiethnic Hawaiian Cohort Human caspase-4 mediates noncanonical inflammasome activation against gram-negative bacterial pathogens Transcriptomic characteristics of bronchoalveolar lavage fluid and peripheral blood mononucle-ar cells in COVID-19 patients Synergy of Interleukin (IL)-5 and IL-18 in eosinophil mediated pathogenesis of allergic diseases Functions of IL-15 in anti-viral immunity: Multiplicity and variety Clinical features of patients infected with 2019 novel coronavirus in An immune-based biomarker signature is associated with mortality in COVID-19 patients Pathological findings of COVID-19 associated with acute respiratory distress syndrome Dysfunctional and Proinflammatory Regulatory T-Lymphocytes Are Essential for Ad-verse Cardiac Remodeling in Ischemic Cardiomyopathy Activated T Lymphocytes are Essential Drivers of Pathological Remodeling in Ischemic Heart Failure T-cell recruitment to the heart: Friendly guests or unwelcome visitors? Pathological inflammation in patients with COVID-19: A key role for monocytes and macrophages Emerging role of chemokine CC motif ligand 4 related mechanisms in diabetes mellitus and cardiovas-cular disease: Friends or foes? CASP4 gene silencing in epithelial cancer cells leads to impairment of cell migration, cell-matrix adhesion and tissue invasion Thromboembolism risk of COVID-19 is high and associated with a higher risk of mortality: A systematic review and meta-analysis Arterial and venous thromboembolism in COVID-19: A study-level meta-analysis COVID-19 and Cardiovascular Disease Recent insights into emerging coronaviruses P-Selectin or intercellular adhesion molecule (ICAM)-1 deficiency substantially protects against atherosclerosis in apolipoprotein E-deficient mice The nuclear factor-kappaB-interleukin-6 signalling pathway mediating vascular inflammation Endothelial cell-specific NF-kappaB inhibition protects mice from atherosclerosis Interleukin-1 plays a major role in vascular inflammation and atherosclerosis in male apolipoprotein E-knockout mice Upregulation of VCAM-1 and ICAM-1 at atherosclerosis-prone sites on the endothelium in the ApoE-deficient mouse Reduced plaque formation induced by rosiglitazone in an STZ-diabetes mouse model of atherosclerosis is associated with downregulation of adhesion molecules The role of monocytes in atherosclerotic coronary artery disease Up-regulation of IL-6 and TNF-alpha induced by SARS-coronavirus spike protein in murine macrophages via NF-kappaB pathway The Role of Cytokines including Interleukin-6 in COVID-19 induced Pneumonia and Macrophage Activation Syndrome-Like Disease Articles Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: A retrospective cohort study COVID-19: A New Virus, but a Familiar Receptor and Cytokine Release Syndrome The pathogenesis and treatment of the 'Cytokine Storm' in COVID-19 Potential Effects of Coronaviruses on the Cardiovascular System: A Review Transcriptome analysis of human heart failure reveals dysregulated cell adhesion in dilated cardiomyopathy and activated immune pathways in ischemic heart failure