Patients
We studied a total of 64 patients with severe COVID-19 pneumonia admitted into the Infectious Diseases Clinics or to Intensive Care Unit (ICU) of the University Hospital in Modena over the period of March 2020-February 2021. All patients displayed symptoms including fever, cough, fatigue, and were classified as severe on the basis of the WHO’s scale 16.
Patients were divided into two groups: those aged <60 years (a total of 33 individuals, named “COVID-under”, CUN) and those aged >70 years (a total of 31 patients, named “COVID-over”, COV), as in Supplementary Table 1. Immunological features of CUN and COV are compared as described below. The clinical characteristics of CUN and COV patients are described in Table 1. For some analyses, patients were compared to a total of 32 healthy adults with a mean age of 54.1±15.9 years.
Table 1
Demographic and clinical characteristics of COVID-19 patients.
Variable | COVID UNDER (n=33) | COVID OVER (n=31) | p-value |
Demographic characteristics | | | |
Age (mean years, range)1 | 49.8 (34 – 59) | 76.4 (69 – 88) | < 0.0001 |
Sex (M, %)2 | 67.0 | 90.3 | 0.03 |
Deceased, N (%)2 | 1 (3.0) | 16 (51.6) | <0.0001 |
Clinical characteristics | | | |
Respiratory rate (±SD)1 | 23.5 (± 6.6) | 25.6 (± 6.6) | ns |
Heart rate (±SD)1 | 82.2 (± 11.5) | 82.2 (± 22.0) | ns |
SOFA score, mean (range)1 | 2.32 (1 – 5) | 3.91 (1 – 6) | 0.0006 |
Coexisting conditions | | | |
Type 2 diabetes, N (%)2 | 6 (18.2) | 10 (32.2) | ns |
Cardiovascular Dis., N (%)2 | 7 (21.2) | 15 (48.4) | ns |
Chronic Kidney Dis., N (%)2 | 2 (6.1) | 7 (25.8) | 0.044 |
Cancer, N (%)2 | 1 (3.0) | 1 (3.2) | ns |
Arterial blood gas analysis | | | |
pO2, mmHg (± SD)1 | 73.0 (± 26.8) | 61.99 (± 10.1) | ns |
sO2, % (± SD)1 | 93.6 (± 3.9) | 91.8 (± 3.3) | ns |
pCO2, mmHg (± SD)1 | 37.6 (± 4.7) | 35.6 (± 5.9) | ns |
pO2/FiO2 (± SD)1 | 149.9 (± 80.2) | 151.8 (± 86.2) | ns |
Clinical Blood parameters | | | |
ALT, U/L (± SD)1 | 39.3 (± 20.5) | 34.41 (± 21.38) | ns |
Total bilirubin, mg/dL (± SD)1 | 0.6 (± 0.2) | 0.8 (± 0.4) | ns |
CK, U/L (±SD)1 | 125.2 (± 121.3) | 166.6 (± 138.3) | ns |
Creatinine, mg/dL (± SD)1 | 0.8 (± 0.2) | 1.68 (± 1.4) | <0.0001 |
D-dimer, ng/mL (± SD)1 | 1,221.8 (± 542.2) | 1,612.86 (± 1,012.48) | ns |
LDH, U/L (± SD)1 | 673.4 (± 203.1) | 677.3 (± 232.3) | ns |
CRP, mg/dL (± SD)1 | 12.2 (± 8.3) | 11.68 (± 7.58) | ns |
Blood cell count | | | |
White blood cells, N/uL (± SD)1 | 6,369.8 (± 2,419.5) | 6,422.8 (± 2,057.0) | ns |
Lymphocytes, N/uL (± SD)1 | 1028.7 (± 405.4) | 898.1 (± 347.84) | ns |
Neutrophils, N/uL (± SD)1 | 5,056.9 (± 3,488.7) | 4,898.97 (± 1888.9) | ns |
Platelets, N*109/L (± SD)1 | 261.8 (± 139.9) | 199.45 (± 106.24) | 0.008 |
SD, standard deviation; Dis, disease; ns, not significant; M, male; pCO2, partial pressure of carbon dioxide; pO2, partial pressure of oxygen; sO2, oxygen saturation; pO2/FiO2, fraction of inspired oxygen; ALT, alanine aminotransferase; CK, creatine kinase; LDH, lactate dehydrogenase; CRP, C-reactive protein. |
1 Unpaired t test or Mann-Whitney t test |
2 Fisher's exact test |
COV patients showed higher plasma level of IL-6 and lower plasmatic levels of IFN-γ and wound healing growth factors if compared to CUN.
Plasma level of 62 cytokines in 26 CUN and 23 COV patients have been grouped according to one of their main functions, as reported in Figure 1. COV patients showed increased plasma level of IL-6 and IL-11 (Figure 1a). These two pro-inflammatory cytokines are involved in acute response to viruses and in lung fibrosis, respectively 17, 18. In COVID-19 patients, high levels of IL-6 have been associated with death 2, while during SARS-CoV-2 infection until now no alterations have been described in the levels of IL-11, a molecule which is linked to the hyperreactivity of the airways during viral infection.
Despite higher levels of IL-6, COV patients displayed lower levels of IL-1β, IL-1α, IL-2, IFN-β, IFN-γ, IL-3, IL-13 and IL-33 if compared to CUN patients (Figure 1a). These cytokines are involved in the regulation of inflammation, T cell activation, antiviral response, dendritic cell (DC) recruitment, tissue remodeling and hematopoietic progenitor cell mobilization 19, 20. Moreover, lower plasma levels of IFN-γ have been associated with increased lung fibrosis 21.
Regarding chemokines, we observed higher levels of fractalkine/CX3CL1 and CXCL10 in COV if compared to CUN patients. These molecules perform several activities, such as, among others, chemoattraction for monocytes/macrophages, T cells, NK cells, and dendritic cells, promotion of T cell adhesion to endothelial cells. Accordingly, it has been shown that in severe SARS-CoV-2 patients they can promote thrombosis and recruitment to the lung of monocytes and T lymphocytes 22. COV patients showed also lower levels of molecules able to recruit neutrophils in the site of infection, such as CXCL2 and CCL5 (Figure 1b). Soluble molecules involved in lung tissue repairing and vascular remodeling such as epidermal growth factor (EGF), platelets derived growth factor (PDGF-AA) and PDGF-AA/BB, were lower in COV if compared to CUN patients (Figure 1c).
We measured other soluble molecules, such as FAS, FAS-L and PD-L1, involved in different apoptotic pathways 23. COV were characterized by higher levels of soluble FAS and PD-L1 and lower levels of FAS-L, as compared to CUN patients (Figure 1d).
CUN and COV patients displayed similar plasma levels of the remaining 42 cytokines that we analysed (Supplementary Figure 1). The hyper-inflamed status for both CUN and COV was also highlighted by the high levels of soluble GM-CSF, TNF, granzyme B and IL-18 as compared to healthy donors (HD) (Supplementary Figure 1).
Different distribution and proliferative capacity of T and B cell subsets in CUN and COV patients.
To deeply characterize the landscape of human PBMC of during SARS-CoV-2 infection, we interrogated a total of 17 severe COVID-19 patients (CUN=7, COV=10). Peripheral blood mononuclear cells (PBMC) were stained and analyzed using a 38-markers mass cytometry panel. Unsupervised analysis revealed 23 different clusters, representing myeloid and lymphoid compartments (Figure 2a, b).
Among CD4+ T cells, we identified three main cell populations, i.e., naïve (CD4+CCR7+CD45RA+CD45R0−CD27+CD28+), central memory [CD4+CCR7+CD45RA− CD45R0+CD27+CD28+ (CM)] and effector memory [CD4+CCR7−CD45RA− CD45R0+CD27−CD28− (EM)]. CD8+ T cells were classified in four clusters, i.e., naïve [CD8+CCR7+CD45RA+CD45R0−CD27+CD28+], activated effector memory [CD8+CCR7−CD45RA−CD45R0+CD27+CD28+HLA-DR+CD38+ (act EM)] and two clusters of effector memory cells re-expressing CD45RA [CD8+CCR7−CD45RA+CD45RO−CD27−CD28− (EMRA)] expressing or not CD57. We also found double positive T cells [CD3+CD4+CD8+ (DP)] and two clusters of unconventional T lymphocytes, i.e., mucosal associated invariant T cells [CD3+CD8+CD161+ (MAIT)] and gamma-delta T cells (CD3+TCRγδ+).
B lymphocytes were identified on the basis of the expression of CD19, and were classified in four clusters, i.e., naïve (CD19+CD20+IgD+IgM+CD21+CD24+CD40+CXCR5+), memory (CD19+CD20+IgD−IgM−CD21+CD24+CD40+CXCR5+), plasmablasts (CD19lowCD20− IgD−IgM−CD80+CD38+) and exhausted cells (CD19+CD20+IgD−IgM−CD21−CD24−CD38−). Three populations of natural killer (NK) cells were identified according to the expression of CD56 and CD16: those defined as early NK (CD16lowCD56+CD57−) and those mature (CD16highCD56+), expressing or not CD57. CD57+ NK cells exhibit both memory-like features and potent effector functions and are one of the hallmarks of ageing 24.
Regarding myeloid compartment, monocytes were identified as CD14+CD11b+CD11c+HLA-DR+ cells (Figure 2b). DC were identified according to the expression of CD123. Four populations of DC were recognized: two populations of plasmacytoid DC [CD123+CD11c− (pDC)] expressing or not CXCR3, and two of myeloid DC [CD123+CD11c+ (mDC)], one of which is activated expressing both CD38 and HLA-DR. Finally, we identified low density neutrophils [CD66b+CXCR1+CCR6+ (LDN)] as those cells present among mononuclear cells after isolation by using Ficoll Hypaque, as previously described 25 (Figure 2b).
COV patients displayed a strong reduction of CM CD4+ T cells and naïve CD8+ T cells (p=0.00036 and p=0.00036, respectively, Figure 2c). Moreover, COV showed higher percentages of mature NK CD57+ cells if compared to CUN patients.
Among CD19+ cells, percentages of exhausted B cells were similar in both COV and CUN patients, while the percentages of memory B cells were significantly lower in COV patients (p= 0.021; Figure 2c). The percentage of plasmablasts was higher in COV if compared to CUN patients (p = 0.044, Figure 2c). Similar percentages of all other subpopulations were found in CUN and COV patients. Reclustering of monocytes and of CD4+ and CD8+ T cells was performed to better describe the populations with a deeper resolution, and described below.
The proliferative capacity of T and B cells was assessed along with phenotypic analysis, and we found that CM CD4+ T cells from COV showed a reduction of both proliferation index (PI) and percentage of divided cells (PD) if compared to CUN patients (Figure 2d). This defective proliferative capacity could partially explain the reduction of this population in the circulation of COV patients. No differences were found within the CD8+ T cell subset (Supplementary Figure 2). B cells from COV displayed higher PI and PD if compared to CUN patients, which likely explains the higher percentages of circulating plasmablasts observed in COV patients.
The principal components analysis (PCA), related to data regarding the complete phenotype obtained by CyTOF, revealed that COV and CUN patients cluster in different positions of the two-dimensional PCA space (Figure 2e, upper panel). Immune features related to the amount of LDN, plasmablasts and activated EM CD8+ T cells (more abundant in COV patients) were the main drivers of the clusterization of patients in two different areas (Figure 2e, lower panel). The same panel indicates that CUN patients were characterized by elevated frequencies of CM CD4+ T cells, naïve CD4+ T cells, naïve CD8+ T cells, naïve B cells and early NK cells.
CM CD4 + T cells in COV are transcriptionally different from those of CUN patients.
Given that in COV the percentage of CM CD4+ T cells was lower and that such cells were characterized by lower proliferative potential than those from CUN patients, we isolated this cell subset and analyzed their transcriptome by RNA-sequencing. We could study 3 CUN and COV patients (median age 46.3±8.1 and 73.3±5.9 years, respectively), who have been matched with 3 young (HUN) and 3 aged (HOV) healthy subjects (median age 50.7±4.9 and 75.0±6.4 years, respectively).
By analysing the differentially expressed genes (DEGs), we observed that a set of 21 immune-related genes clearly separates infected patients from healthy subjects (Figure 3a). Genes like DUSP4, NR4A1, TBX21(T-bet), ZEB2, CEBPA, SIGLEC5 and CIITA, involved in Th1 priming and T-cell receptor (TCR) response 26, were expressed at higher level in HUN and HOV if compared to CUN and COV patients. On the contrary, CM T cells from COV and CUN patients were clearly distinct from those of healthy donors for higher levels of CLTA4, LAG3, MCM6, MKI67 and IFI27 genes, that are associated with T cell exhaustion, proliferation, and antiviral activity. Both CUN and COV groups, compared with their respective control group, expressed higher level of genes involved in the antiviral response and proliferation, like IFIT5, IFI27 and PIM1, suggesting that those cells could have been activated by SARS-CoV-2 infection 27, 28 (Supplementary Figure 3b-c). When compared to their relative controls, COV, but not CUN patients expressed long non-coding RNAs like SNTG2−AS1, RGMB−AS1, ZNF32−AS2, SH3BP5−AS1, and LAG3, well-known markers of age-related dysfunction and exhaustion, respectively 29, 30 (Supplementary Figure 3c). Several other DEGs were identified and are reported in the supplementary Table 2.
Moreover, comparing CUN and COV patients, we found 7 differentially expressed genes (FDR <0.05, Figure 3b); 6 out 7 (CTLA4, LAG-3, DUSP4, CXCR3, CCR5 and LRRC32) were upregulated in COV patients; 3 out 6 (CTLA4, LAG-3, DUSP4) were associated with T cells exhaustion and defective TCR response 30–32. A different expression of these genes was not observed comparing HUN and HOV samples (Supplementary Figure 3a).
To further investigate the putative exhaustion of central memory compartment of COV patients, we quantified by flow cytometry the expression of PD-1 in CD4+ CM T cells, and found that the amount of this molecule per cell was significantly higher in COV patients (Figure 3c).
Reclustering of CD4 + T cells identifies different subpopulations of CM T cells, including circulating follicular T lymphocytes.
CM T cell subset is a heterogeneous population composed by elements that can migrate into the lymph node and towards the inflamed tissue thanks to the presence of different chemokine receptors, such as CXCR3 33. To gain more insights into CM compartment, we reclustered CD4+ T cells and identified 22 clusters. Besides naïve T cells, defined as CD45RA+CD27+CD28+CCR7+, we found 7 clusters of CM T cells defined as follow: CM (CD45RA−CD27+CD28+CCR7+), activated CM [CD45RA−CD27+CD28+CCR7+CD38+ (act CM)], CM Th1/Th2 [CD45RA−CD27+CD28+CCR7+CXCR3+CCR4lowCCR6low (CM Th1_2)], CM Th2 [CD45RA−CD27−CD28+CCR7+CXCR3−CCR4+ (CM Th2)], CM PD-1 [CD45RA−CD27+CD28+CCR7+CD38+PD-1+ (CM PD-1)], CM CXCR5 [CD45RA−CD27+CD28+CCR7+CXCR3−CXCR5+PD-1− (CM CXCR5)], and circulating follicular T cells [CD45RA−CD27+CD28+CCR7+CXCR3+CXCR5+PD-1+ (cTfh)]. Tfh cells that circulate in the blood have been identified as counterparts of germinal center Tfh. In particular, cTfh cells expressing CXCR3 are important in the response to influenza vaccine, inducing a strong antigen-specific antibody response 34.
Among TM, we found these 6 clusters, defined classical TM [CD45RA−CD27−CD28+CCR7−], TM Th1 expressing PD-1 [CD45RA−CD27+CD28+CCR7−PD-1+CXCR3+ (TM PD-1 Th1)], activated TM Th1 expressing PD-1[CD45RA−CD27+CD28+CCR7−PD-1+CXCR3+CD38+HLA-DR+ (TM PD-1 Th1_act)], TM Th2 [CD45RA−CD27−CD28+CCR7−CCR4+], TM Th2 expressing PD-1[CD45RA−CD27−CD28+CCR7−CCR4+ (TM PD-1 Th2)] and TM expressing both CD57 and PD-1 [CD45RA−CD27+CD28+CCR7−CD57+PD-1+CXCR3+ (TM CD57 PD-1)]. We also identified two populations of EM, i.e., those CD45RA−CD27−CD28−CCR7− and those expressing CD57 and PD-1 [CD45RA−CD27−CD28−CCR7−CD57+PD-1+ (EM CD57 PD-1)].
On the other side, three populations of effector memory cells re-expressing CD45RA (EMRA) were detected, i.e., [CD45RA+CD27−CD28−CCR7− (EMRA)], EMRA expressing CD57 [CD45RA+CD27−CD28−CCR7−CD57+ (EMRA CD57)] and EMRA Th1/Th2 expressing CD57 [CD45RA+CD27−CD28−CCR7−CD57+CXCR3+CCR4+CCR6+ (EMRA CD57 Th1_2)]. Finally, we identified also putative effector T regulatory cells (eTreg) expressing CD45RA−CD127−CD25+CCR7−CCR4+ (Figure 4a-b).
Besides different percentages of activated and effector memory subset of both CUN and COV patients compared to HDs (Figure 4c), we found a higher percentage of activated CM in COV, but a lower percentage of cTfh cells if compared to CUN patients (Figure 4c, and Supplementary Figure 4a-b). Moreover, we observed a negative correlation among cTfh and plasmablasts in COV patients, in contrast with that observed in CUN patients (Supplementary Figure 4c).
To explore more in depth this correlation, we investigated in silico the plasma level of anti-spike immunoglobulin (Ig) A1, A2, G1, G3, Fcγ receptor binding and Fc effector activity in both CUN and COV patients using publicly available data 35. We selected 21 CUN and 18 COV with a median age of 49.0±6.1 and 83.0±5.9 years, respectively. We observed a reduced level of anti-S and anti-RBD IgG3 isotype during the first week of infection in COV patients if compared to CUN (Supplementary Figure 5). COV recovered to the level of CUN at the end of the second week. Similar level of anti-S and anti-RBD IgA1, IgA2 and IgG1 were found. Given the differences in IgG class switching, we also examined the ability of SARS-CoV-2-specific antibodies to bind to the low-affinity Fcγ receptors. We observed that the IgG of COV patients were less able to bound both activating receptor FcγR2A (also known as CD32A) and FcγR3A (also known as CD16) during the first week of onset but not during the second or third week (Supplementary Figure 5).
Finally, we reclustered CD8+ T cells, and found significant differences between patients and healthy donors. On the contrary, we could not identify any relevant difference between CUN and COV patients (Supplementary Figure 6).
SARS-CoV-2-reactive CD4 + cTfh cells from COV patients are more activated than those from CUN patients.
We performed an in silico analysis of published data containing single-cell transcriptomic and T cell receptor (TCR) analysis of >100,000 SARS-CoV-2-reactive CD4+ T cells 36. From the entire dataset, we selected 8 severe patients that we could classified as CUN or COV on the basis of their age. Among 8,959 SARS-CoV-2 specific CD4+ T cells, we found five clusters (Figure 5a). Clusters 0 and 3 were characterized by high levels of PRF1, GZMB, GZMH, GNLY, and NKG7 gene expression, and they were defined as cytotoxic CD4+ T cells (CTL) (Figure 5b). However, given the different expression of IFNG, TNF, CCL3 and CCL4, within these clusters we found CTL that were IFNhigh and CTL IFNlow. Cluster 1 expressed high levels of CD200, BTLA and POU2AF1 and was defined as formed by circulating follicular T helper (cTfh) 37. Cluster 2 expressed high level of STAT1, IL7R, SELL, TNFSF4 (also known as OX40L) characteristic of memory T cells that have recently engaged the Ag, and was defined as formed by activated-STAT1+ (act-STAT1) cells. Finally, cluster 4 displayed a transcriptional profile of proliferating cells expressing MKI67, TOP2A, HMGB1-2 and STMN1 (Figure 5b).
Although no differences were found in the proportion of clusters between CUN and COV patients (Figure 5c), the detailed analysis of their transcriptional level revealed that CTL-IFNhigh, CTL-IFNlow and act-STAT1 clusters from COV patients showed higher levels of TIGIT, CTLA4, ICOS, HAVCR2 (TIM-3) and ZBED2 genes (Figure 5d). In addition, the clusters from CUN patients displayed a transcriptional profile consistent with an increased antiviral activity, as they were expressing higher levels of genes related to IFN-response (IFI27, IFI6, ISG15), TNF-response (TNF, TNFSF10), cytotoxicity (GNLY), chemotaxis (CXCR3) and activation (CD69, CD28) (Figure 5d). cTfh cluster of COV patients expresses fewer genes involved in T cells activation and monocytes maturation, such as CD28, IFI27, CD74, CSF2 (also known as GM-CSF).
The analysis of TCR clonality was studied by detecting gene expression of SARS-CoV-2-specific CD4+ T cells, and revealed that COV patients were characterized by a lower quantity of small clonotypes within cTfh and activated (act)-STAT1 clusters (p<0.0001; Figure 5e). This suggests the possible existence of a depletion of clonal repertoire of the SARS-CoV-2-reactive CD4+ T cells in COV patients. In particular, despite the frequency of cTfh cell was similar in CUN and COV patients as reported above, cTfh of CUN were likely able to recognize a wider range of SARS-CoV-2 antigens. This observation suggests that CUN patients may develop a broader antibody response against several immunogenic regions of the SARS-CoV-2 virus.
Finally, we observed that a massive clonal expansion occurred within CD4-CTL IFNhigh and CTL IFNlow clusters, in both CUN and COV groups, suggesting a probable active role of these cells in controlling the infection (Figure 5e). Specifically, we observed that CTL IFNhigh cells from CUN had a stronger expansion if compared to those from COV patients, as revealed by the higher level of hyperexpanded clones.
Detailed monocyte’s landscape revealed higher percentage of PD-L1+ intermediate monocytes in COV patients.
Reclustering of monocytes using markers such as CD14, CD16, CD4, CD56, CD38, CCR4, CCR6, CXCR3, CD294, CD80, CD40, CD11c, CD11b and PD-L1 allowed us to better characterize their different subpopulations and to point out differences between CUN and COV patients. Unsupervised analysis revealed 8 different clusters (Figure 6a-b). Two of them were classified as classical (CL) monocytes, expressing or not CD56, two were intermediate (INT) monocytes expressing or not PD-L1, three were non-classical (NC) monocytes expressing or not CD38, CD40 or PD-L1, and one was classified as formed by immature cells (CD14low). This detailed clusterization revealed the presence of one cluster of CL expressing CD56, who had been described as dysregulated monocyte producing basal level of IL-6 without stimulation 38, and that was more represented in COV patients (Figure 6c).
Another peculiar cluster was that of NC monocytes expressing CD40 and PD-L1, observed predominantly in COV patients. These could be suppressive antigen-presenting cells involved in the control of the activation of the adaptive immune response. Here we observe that their frequency was higher in COV patients (Figure 6c). Finally, we report that both CUN and COV patients display a significant percentage of immature monocytes, whose metabolism a particular population has been characterized recently 13. Such cells express high level of CXCR3, a chemokine receptor that is able to recruit monocytes in the inflamed tissue, and thus could likely play a role in guiding the inflammation in the lungs. Furthermore, we showed that the expression of HLA-DR on total monocytes were lower in COV patients if compared to CUN patients (Figure 6d), and this difference was due to a lower HLA-DR expression on different clusters, i.e., CL CD56−, CL CD56+, INT PD-L1+ (Supplementary Figure 7a). In addition, all monocytes from CUN and COV patients showed an upregulation of PD-L1 (Figure 6d, Supplementary Figure 7b).
Inflammatory molecules, including IL-6, inhibit HLA-DR and induce PD-L1 expression, suggesting that the high level of plasmatic IL-6 observed in COV patients may exacerbate the immune dysfunction of these patients. The amount of PD-L1 and HLA-DR molecules present on monocytes’ surface was quantified by flow cytometry, and we found that both COV and CUN patients showed a positive correlation between plasma level of IL-6 and the number of PD-L1 molecules (Figure 6e). On the other hand, comparing IL-6 plasma level with HLA-DR revealed that COV patients, but not CUN patients, showed a negative correlation between these two parameters (Figure 6e).
COV patients are characterized by lung fibrosis and activated macrophages.
Given that COV patients are characterized by profound immune alterations detectable in the blood, we aimed in better characterizing the immune feature of lung microenvironment. We interrogated a public dataset containing Imaging Mass Cytometry (IMC) data performed on lung biopsies from patients who died because of severe SARS-CoV-2 infection, and we stratified patients as CUN and COV patients.
An increased deposition of collagen type I mediates lung fibrosis. We observed that COV had higher deposition of collagen type I if compared to CUN patients (Figure 7a-b). Moreover, COV displayed similar number of lung infiltrated macrophages if compared to CUN patients, as outlined by CD68 expression (Figure 7d), but higher expression of vimentin that co-localized with caspase 3. These observations could indicate that the activation of tissue macrophages is higher in lungs from COV than in CUN patients (Figure 7c-d, Supplementary Figure 8).