Clinical categorization of infected infants and changes in PBMC compositions
We collected blood samples and processed serum and peripheral blood mononuclear cells (PBMCs) from 26 infants (median, IQR age: 1.63 [0.93–7.59] months) hospitalized with SARS-CoV-2 infection (pediatric COVID; pCoV) and 14 matched pediatric healthy controls (pHC; median IQR age: 2.01 [1.86–4.28] months) (Fig. 1a). Patient demographics, clinical and laboratory data and treatment are summarized in Supplementary Table 1a, b. We categorized pCoV patients into three groups (G1, G2 and G3; Fig. 1b) based on (i) the time (days) since exposure (DSE) to a known COVID-19 case, (ii) disease severity, and (iii) viral loads (VL) measured within a median of 17 [11–36] hours since hospitalization. Infants in G1 (n = 10) were identified as part of the routine diagnostic work-up during hospitalization and had both milder disease and lower SARS-CoV-2 viral loads (subacute group). Infants in G2 (n = 11) had moderate disease and high SARS-CoV-2 loads, but did not require significant medical interventions (i.e., no supplemental oxygen or intravenous fluids). G3 (n = 5) infants also had high SARS-CoV-2 viral loads but required a higher level of care, such as oxygen supplementation or intensive care unit (ICU) admission (Supplementary Table 1a-d).
To assess the immune response to SARS-CoV-2 in these infants and to determine how these responses correlated with their varying clinical phenotypes, we performed: (i) single cell RNA-seq (scRNA-seq) of PBMCs, (ii) serum cytokine concentration assessment using the 92-Olink platform with a focus on inflammatory pathways, (iii) quantification of antibodies against coronaviruses (anti-Spike and anti-RBD) and anti-IFN activity, and (iv) viral load measurement in nasopharyngeal (NP) swabs (Extended Data Fig. 1a).
For scRNA-seq data analysis, raw data from pHC PBMCs (38% of the pool) and pCoV PBMCs (62% of the pool) were integrated (Fig. 1a). Hybrid transcriptomes (multiplets) were identified using scrublet11 and excluded from the rest of the analysis. After filtering steps (see Methods for details), pHC and pCoV samples yielded, respectively, a mean (± S.D.) of 6,559 (± 2,016) cells and 5,981 (± 1,807) cells per individual, and a mean of 1,411 and 1,603 genes per cell (Extended Data Fig. 1b,c). scRNA-seq profiles that passed quality control (n = 203,402 cells) were then corrected for batch effect using BBKNN12 (Extended Data Fig. 1d,e, see Methods). Unsupervised clustering of the corrected data, followed by a two-dimensional uniform manifold approximation and projection (UMAP), yielded 25 clusters independently of 10x run (Extended Data Fig. 1e) and subject (Extended Data Fig. 1f) batch effects. Using marker genes, clusters were first mapped to major immune cell types, including CD4+ and CD8+ T cells, B cells, NK cells, CD14+ and CD16+ monocytes, conventional dendritic cells (cDCs), plasmacytoid dendritic cells (pDCs), plasma cells (PCs), hematopoietic stem cells (HSCs) and Erythroblasts/Erythrocytes (Eryth) (Fig. 1c,d, Extended Data Fig. 1g,h). To analyze the cellular distribution of Interferon Stimulated Genes (ISGs) within different immune subsets, we calculated an ISG score based on expression levels of genes in interferon modules13 (Supplementary Table 3). This revealed increased ISG scores in most cell types, including monocytes, T cells and B cells (Fig. 1e). Cell compositional analyses in pHC, G1, G2 and G3 pCoV groups revealed comparable distribution of pHC and G1 groups, contrasting with striking differences observed in G2 and G3 groups (Fig. 1f). Overall, these high-level analyses revealed significant alterations in the PBMC composition of infants upon SARS-CoV-2 infection and a robust ISG signature in most immune cell types in response to the infection. To refine these alterations, we further subclustered and analyzed immune cell subset.
Expansion of ISG IL-1B monocyte In mild to severe disease
Previous studies have reported alterations in the CD14+ compartment of adults with severe COVID-19, including decreased HLA-DR surface expression and the accumulation of immature circulating myeloid cells, suggesting dysregulated myelopoiesis/innate immune response14,15. To investigate this in our infant cohort, we further clustered CD14+ monocytes (n = 16,906) into four subclusters (SCs) (Fig. 2a). SC0 (n = 5,495) expressed antigen presentation-related transcripts (e.g., “HLA-DRA”, “CD74”) together with small and large ribosomal subunits (e.g., “RPS3A”, “RPL3”), while SC2 (n = 4,590) expressed high levels of alarmins “S100A8” and “S100A9”. SC1 (n = 5,017) displayed an ISG signature, including “ISG15”, “IFI6” and “MX1”. SC3 (n = 1,804) was characterized by the upregulation of “IL1R2”, “CD163”, “FKBP5” as well as ISGs (Fig. 2b,c). SC1 and SC3 were expanded in G2/G3 and G3 respectively, while SC0 and SC2 were predominantly present in pHC and G1 (Fig. 2d). SC3 was almost entirely contributed by 3/5 severe patients (G3), two of whom had received systemic steroids. In fact, the SC3 transcriptional profile (Fig. 2b,c) was reminiscent of what was previously described in adults with SARS-CoV-2 infection receiving steroid treatment16,17,18.
The ISGhi subcluster (SC1) was rare in pHC and significantly expanded in 2/10 of G1, 11/11 of G2, and 2/5 of G3 patients (Fig. 2e). As both inflammation and type I IFN pathways were previously found activated in monocytes from adults with SARS-CoV-2 infection5, we analyzed the co-expression of inflammatory transcripts and ISGs at the single cell level in these infants. Approximately 49% of SC1 monocytes co-expressed “IL1B” and “ISG15” (Extended Data Fig. 2a-c).
Thus, infants with moderate and severe COVID-19 were separated in two groups: the first included all G2 and 2/5 G3 infants and was characterized by expansion of monocytes co-expressing inflammatory molecules and ISGs with low HLA gene expression. The second group consisted of 3/5 G3 infants whose monocytes expressed ISGs and IL1R2 along with CD163, likely reflecting the influence of steroid treatment.
SARS-CoV-2 infected infants maintain CD16 + monocyte percentages with increased interferon expression.
Contraction of CD16+ monocytes has been previously described as a feature of severe COVID-19 in adults with COVID-1915. Subclustering of CD16+ monocytes in infants (n = 5,987) generated three SCs (Figure. 2f). SC0 (n = 3,009) expressed an ISG signature (e.g., “ISG15”, “IFI6” and “MX1”). SC1 (n = 2,217) expressed ribosomal-associated genes “RPL5”, “RPL10A”, or “RPL3” and SC2 (n = 761) was characterized by the expression of inflammatory molecules (e.g., “IL1B, “CCL3”) (Figure. 2g). SC0 (n = 3,009) was significantly expanded in G2 and G3, SC1 was enriched in pHC and G1, while SC2 showed a mild elevation in G2/G3 (Figure. 2h). In contrast to the CD14+ population, only about 13% of CD16+ SC0 monocytes co-expressed “IL1B” and “ISG15” (Extended Data Fig. 2d,e). The ISGhi subcluster SC0 was rare in pHC and significantly expanded in 9/11 of G2 and 4/5 of G3 infants (Extended Data Fig. 2f). Unlike severe COVID-19 in adults, which was characterized by CD16+ monocyte depletion19, infants with moderate to severe disease (G2/G3) exhibit normal frequencies of monocytes due to the expansion of ISGhi CD16+ monocytes.
To explore how our findings compare with previous studies, we integrated our scRNA-seq data with a recent study by Wimmers et al.20, which analyzed PBMCs from older infants with mild disease (average 9 months of age, not hospitalized and untreated) during both Omicron and non-Omicron variants of SARS-CoV-2 infection. This cohort included: (i) nine samples collected before (pre, n = 9), during (acute; n = 10) and after infection (convalescent or conv; n = 9) with non-Omicron variants, (ii) eight samples during acute Omicron infection (acute omicron) and (iii) seven matched controls. Our integrative analysis of CD14+ and CD16+ monocytes from 83 infants in total confirmed the presence of an ISG signature in infants with moderate to severe disease (10/10 G2 and 5/5 G3 patients in our cohort), which contrasted with only 5/10 acute non-Omicron and 1/10 acute Omicron patients in the previous cohort (Extended Data Fig. 2g&i). The number of CD14+ monocytes co-expressing “ISG15” and “IL1B” in the acute non-Omicron group in the Wimmers cohort was lower than in G2, G3 groups in our cohort (Extended Data Fig. 2h). These differences are likely attributable to variations in the time from exposure to the virus, the age of the infants, and, most importantly, the clinical disease severity between the two cohorts.
Contraction of cDCs in SARS-CoV-2 infected infants
Similar to classical monocytes, DCs from SARS-CoV-2 infected adults exhibit lower levels of HLA-DR compared to healthy controls21. Subclustering conventional DCs (cDCs) in infants (n = 440) generated seven SCs (Fig. 2i and Extended Data Fig. 3a), which were further classified into (i) cDC1 (SC5; expressing “CLEC9A” and “XCR1”), (ii) cDC2 (SC0/SC3; expressing “CD1C” and “CLEC10A”), (iii) monocyte-derived cDCs (Mo-DCs; SC1/SC4; expressing “CD14” and S100s) and (iv) AXL+ DCs (SC6; expressing “AXL” and ”DAB2”). The AXL + DCs (SC6) expressed the highest levels of ISGs (Fig. 2j,k and Extended Data Fig. 3b-d). G2/G3 groups showed a marked contraction of cDC1, cDC2 and Mo-DCs (Fig. 2l), while two G2 patients showed an expansion of AXL+ ISGhi SC (SC6) (Fig. 2l and Extended Data Fig. 3e). Overall, our data showed that profiles of G2 and G3 infants were characterized by a decrease in cDC1, cDC2 and Mo-DCs.
The frequency of pDCs in peripheral blood has been found to decrease in adults with COVID-1922, possibly due to pDC apoptosis16. Subclustering of pDCs in infants (n = 386) generated two SCs (Fig. 2m). SC0 (n = 226) was significantly contracted in G2/G3 patients, while SC1 (n = 160) was significantly expanded in these infants and characterized by the upregulation of ISGs (Fig. 2n-o). The depletion of ISGlow pDCs is consistent with reports in adults with COVID-19; however, a subset of ISGhi circulating pDCs was found to be expanded in G2/G3 infants.
Circulating megakaryocytes (MGKs) have previously been linked to COVID-19 outcomes in adults23. Two of the infant’s SCs (in C22, Extended Data Fig. 1g,h) were annotated as megakaryocyte (MGK; n = 408; expression of “PPBP”, “CLU” and “PF4”) and hematopoietic stem cells (HSCs; n = 305; expression of “CD34” and “ITM2C”) (Extended Data Fig. 3f-h). MGK were significantly expanded while HSC were significantly contracted in G1 and G3 groups (Extended Data Fig. 3i).
ISG naïve CD4 T cells are expanded in SARS-CoV-2 infected infants
Activation and proliferation of both CD4+ and CD8+ T cells have been associated with COVID-19 disease severity16,4, though no specific T cell signatures have been identified as primary correlates in this context. In our infant study, subclustering CD4+ T cells (n = 100,482) yielded four SCs (Fig. 3a). SC0 (n = 43,567), expressing ribosomal protein-associated genes such as “RPL5”, “RPL10A” and “RPL3”, was significantly decreased in G2 and G3 infants. SC1 (n = 22,847), which exhibited an ISG signature (e.g., “IFI44L”, “ISG15” and “XAF1”), was expanded in G2 and G3 infants. SC2 (n = 14,383), characterized by expression of “JUNB/JUND”, “CD69” and “CXCR4” did not differ between infants with COVID-19 and pHC. Finally, SC3 (n = 14,383), expressing the memory marker “S100A4”, was expanded in all three pCoV groups (Fig. 3b,c). The ISGhi SC1 was present in 2/10 G1, 8/11 G2 and 5/5 G3 infants. Notably, more than 90% of CD4+ T cells from a fraction of G2 and G3 patients (e.g., pCoV11 and pCoV26) switched to an ISGhi state (Fig. 3d-f). SC0 and SC1 exhibited naïve (“CCR7”, “LEF1” and “SELL”) markers, while SC3 displayed memory markers (Fig. 3e-g). Further analysis revealed that SC3 contained both cytotoxic-associated transcripts (GZMK and “CCL5”), and Treg-associated transcripts (”FOXP3” and “IL2RA”) (Fig. 3h) indicating the presence of both CD4+ populations within the SC3 memory compartment. However, no Tfh (”CXCR5”), Th1 (“TBX21”) or Th2 (“GATA3”)-related transcripts were not detected in any SC. Analysis of granzyme (GZM) family expression revealed a broad upregulation of “GZMM”, contrasting with an absence of “GZMB” and restriction of “GZMK” to SC3. Our analysis also revealed the expression of the transcription factor “SOX4” within the naïve compartment, which is in line with an immature/naive phenotype. The absence of CX3CR1 expression a lack of CD4+ TEMRA cells in this age group (Fig. 3e). Interestingly, while the majority of ISGhi T cells originated from infected patients, we also detected these cells in pHC, indicating constitutive expression of ISGs (Extended Data Fig. 4a,b).
The integration analysis with the Wimmers et al. dataset further confirmed the expansion of an ISG signature in all G2 and G3 individuals in our cohort, contrasting with 4/10 acute non-Omicron and 1/10 acute Omicron individuals in the Wimmers cohort (Extended Data Fig. 5a). Overall, these data indicate that the blood CD4+ T cells of COVID-19 infected infants fall into two main compartments: (i) naïve, as expected the predominant population, that had switched to an ISGhi state, and (ii) memory, which encompassed a mixture of effector memory T cells, CD4+ CTLs and Tregs and was expanded in SARS-CoV-2 infected infants.
ISG CD8 T cells are expanded in infants with COVID-19
COVID-19 associated lymphopenia3 is reported to preferentially impact CD8+ T cells4, possibly due to T cell exhaustion24. However, the overall CD8+ T cell compartment (n = 34,366), however, was not decreased in infants with COVID-19 and was distributed into five SCs (Fig. 4a). CD8+T-SC0 (naïve, n = 23,768) expressed ribosomal-associated genes (e.g., “RPL34” and “RPL39), and was contracted in G2 and G3 infants; CD8+T-SC1 (ISGhi; n = 3,483) exhibited an ISG signature (e.g., “IFI44L”, “ISG15” and “XAF1”) and was expanded in G2/G3 infants. CD8+T-SC2 (n = 2,967) displayed similar frequencies in infected infants and pHC and expressed cytotoxic genes, including “PRF1”, “GZMA” and “GNLY”, inflammatory chemokines (“CCL4” and “CCL5”), “S100A4” and “CX3CR1” consistent with a TEMRA phenotype. CD8+T-SC3 (GzK; n = 2,201) expressed “GZMK” and “CCL5” and was mostly present in G1 but contracted in G2/G3 infants. Finally, CD8+T-SC4 (Prolif.; n = 1,947), which expressed proliferation markers such as “MKI67” and HMGBs, was expanded in G2 and G3 infants (Fig. 4b,c). ISGhi SC was rare in pHC but present in 2/10 G1, 8/11 G2 and 3/5 G3 infants. Notably, more than 50% of CD8+ T cells in some G2 (pCoV13, pCoV24, pCoV26) and G3 (pCoV2 and pCoV11) infants demonstrated an ISGhi state (Fig. 4d). The proliferative SC4 included a subset of cells expressing TEMRA markers, such as “CX3CR1”, “FCGR3A” and “GZMB” (Fig. 4e,f). This SC represented 15–20% of CD8+ T cells in 6/16 of the G2/G3 infants (Fig. 4d). Comparison with published data20 on infants with mild disease further confirmed the presence of a robust ISG signature in all G2 and G3 patients in our cohort, contrasting with 4/10 acute non-Omicron and 1/10 acute Omicron individuals in the Wimmers cohort (Extended Data Fig. 5b). In summary, infants with more severe disease exhibited an expansion of ISGhi CD8+ T cells and an induction of CX3CR1+ proliferating CD8+ T cells, a finding that contrasts with previous reports adults25.
ISGhi NK cells during severe SARS-CoV-2 infection in infants
A recent study reported an increase of “adaptive” NK cells in the blood of adults with severe COVID-1926. However, little is known about the role of these cells in infants. Herein, NK cell (n = 13,403) subclustering yielded four SCs (Fig. 4g). NK-SC0 (n = 5,013), contracted in G2 and G3 infants, expressed ribosomal-associated genes. NK-SC1 (n = 3,393), contracted in G2 infants, expressed “CD160” and “KLRB1”. NK-SC2 (n = 2,721), expanded in G2/G3 infants, exhibited an ISG signature. NK-SC3 (n = 2,276) displayed similar frequencies in pHC and infected infants and upregulated transcripts such as “XCL1”, “XCL2” and “GZMK” (Fig. 4h-k). While very rare in pHC, the ISGhi SC2 was present in 2/10 G1, 11/11 G2 and 5/5 G3 infants. Notably, more than 70% of NK cells from 9 G2 and all G3 infants switched to an ISGhi state (Fig. 4l).
The comparison with previously reported cohorts confirmed the presence of a strong ISG signature in all G2 and G3 patients in our cohort, contrasting with 5/10 acute non-Omicron and 1/10 acute Omicron patients in the Wimmers cohort (Extended Data Fig. 5c).
SARS-CoV-2 infection in infants is associated with increased ISG transitional and naïve B cells
In adults with COVID-19, activated B cells and plasmablasts are expanded27,4, and increased frequency of extrafollicular (Tbet+CD11c+CXCR5neg) B cells correlates with disease severity28. In our study, further clustering of infant’s B cells (n = 30,119) generated five SCs (Fig. 5a). In G2/G3 infants, B-SC1 (n = 5,228) and B-SC2 (n = 4,985) were expanded, while B-SC0 (n = 13,820), B-SC3 (n = 4857) and B-SC4 (n = 1,229) were contracted (Fig. 5b). Differential marker expression distinguished three compartments: (i) Naïve B cells (NBC; “IGHD”, “CCR7” and “SELL”), (ii) Transitional B cells (TrBC; “MME (CD10)”, “CD9”, and “CD24”) and (iii) Memory B cells (MBCs; “CD27”, “IGHA1”, “IGHG1” and “S100A4”). An ISG signature (e.g., “IFI44L”, “ISG15” and “IRF7”) was detected in both the NBC and TrBC compartments. Thus, NBCs included B-SC0 (ISGlow; expanded in pHC/G1) and B-SC2 (ISGhi; expanded in G2/G3); TrBCs encompassed B-SC3 (ISGlow; expanded in pHC/G1) and SC1 (ISGhi; expanded in G2/G3). SC4 included transcripts characteristic of double negative (DN)2 memory cells (e.g. “ITGAX”, “TFEC” and ”FGR”). Interestingly, the ISGhi naive B-SC2 expressed “TLR7”, a marker of both extrafollicular activated naïve and DN2 cells29 (Fig. 5c,d). In addition to memory markers, B-SC4 also expressed B cell survival and plasma cell differentiation markers such as “TNFRSF13B” (encoding TACI) and “TNFRSF17” (encoding BCMA), respectively. Although very rare in pHC, the ISGhi SC was present in 2/10 G1, 11/11 G2 and 5/5 G3 infants. Overall, > 70% of naïve (B-SC2) and transitional (B-SC1) B cell compartments from 13/16 of the G2/G3 infants switched to an ISGhi state (Fig. 5e). While the majority of the ISGhi B cells originated from infected patients, we also identified an ISG signature in both naïve and transitional B cells from healthy controls(Extended Data Fig. 4c,d).
The comparison between cohorts demonstrated the presence of a strong ISG signature in all G2 and G3 patients in our cohort, contrasting with 10% and 40% of acute Omicron and non-Omicron individuals, respectively, in the Wimmers cohort (Extended Data Fig. 5d).
Finally, PC (n = 544) subclustering generated three SCs (Fig. 5f), which were distinguishable based on IGH expression (Fig. 5g). Although the low number of cells is a limitation, cell compositional analysis did not reveal significant differences between COVID-19 infant groups and pHC (Fig. 5h), which contrasts with what was reported in adults with COVID-1930.
Patient stratification using cell frequencies.
To integrate subcluster analysis data (Extended Data Fig. 6a-d) with clinical disease severity, we performed an unsupervised clustering based on the abundance of detected subclusters (n = 40) across infants with COVID-19 (n = 26) and pHC (n = 14). The study subjects were accordingly clustered into two main sets (Fig. 6a). The first set, which included 10 infants with COVID-19 (3/5 G3, 6/11 G2 and 1/10 G1 infants), was characterized by the expansion of ISGhi SCs. The second set, which included the remaining 16 COVID-19 infants together with the 14 pHC, displayed ISGlow SCs including naïve CD4+, CD8+ T cells and B cells (Fig. 6b). Interestingly, the two G3 patients who clustered with pHC/G1 (pCoV17 and pCoV21) had received systemic steroids, consistent with the modulatory effect of corticosteroids on type I IFN expression31. These data therefore support that clinically severe SARS-CoV-2 infection in infants is associated with a robust ISG response.
Increased serum concentrations of inflammatory cytokines in SARS-CoV-2 infected infants.
To complement our transcriptional profiling studies, we measured serum cytokines in 34 children with COVID-19 (7 G1, 20 G2 and 7 G3) and 20 age-matched HCs using the Olink inflammatory panel (n = 92 analytes). This cohort included 24 of the 26 infants profiled with scRNAseq (Extended Data Fig. 1a, Supplementary Table. 1e). Overall, 72 cytokines were consistently detected in serum samples (Extended Data Fig. 6e); and 33 of them showed significantly different concentrations between disease severity groups (Fig. 6c). IFNg concentrations were increased in infants with COVID-19 across all disease groups. Cytokines and inflammatory proteins such as IL6, IL8, IL17C, IL18R1 and CXCL10 were particularly increased in G3 infants (Fig. 6d), while soluble CD6 and TNFS12/TWEAK were markedly decreased in this severe group compared with HCs. Patients in the Wimmers cohort did not show significant increases in plasma inflammatory cytokines possibly due to differences in time since infection and/or clinical disease severity. In summary, SARS-CoV-2 infected infants displayed increased serum concentrations of inflammatory cytokines, especially in those with severe disease (G3).
Anti-SARS-CoV-2 and anti-IFN antibody responses
To assess the infants’ humoral response to SARS-CoV-2 and seasonal coronaviruses, we measured antibody levels against: (i) SARS-CoV-2 full-length spike, RBD and S2 subunit, (ii) seasonal coronaviruses full-length spike, (iii) S1 subunit of beta-coronaviruses OC43 and HKU1, and (iv) full-length spike of alpha-coronavirus 229E. Samples from 16 infants were obtained at the time of hospitalization, and 13 of those had paired follow-up samples four weeks later. The 13 infants with paired samples, who were also included in the scRNAseq cohort (Extended Data Fig. 1a, Supplementary Table. 1f), predominantly (12/13) showed a significant increase in IgG antibody titers against SARS-CoV-2 antigens at follow-up, including spike, RBD and S2 subunit (Extended Data Fig. 7a,b). While 10 infants increased their anti-S2 and anti RB levels by 20 and ~ 8 fold, respectively, no increase in antibody titers against any of the seasonal alpha- or beta- coronaviruses was observed, despite the presence of detectable IgG levels at baseline (T0); (Extended Data Fig. 7c). Given the young age of this cohort (median 3.2 months), we hypothesized that the detected antibodies against seasonal coronaviruses are likely of maternal origin.
Finally, although anti-IFN autoantibodies have been identified in adults with severe COVID-1932, none of the patients (n = 16) or HCs (n = 6) in this pediatric cohort demonstrated anti-IFN activity in either the acute or convalescent serum samples (Extended Data Fig. 7d).
Lymphocytes from infants express a broader ISG signature compared to adults
We next compared our infant data with a scRNA-seq PBMC dataset (GSE16191816) from a reported cohort of 28 adults with COVID-19 and controls. For this comparison, we included data from the adult patients’ first time-point (T0), as well as from 11 healthy matched controls (aHC)16. After concatenating adult (n = 39) and infant (n = 40) datasets, we applied our pipeline (see methods for details) on doublet-cleaned data, which included pre-processing, batch correction (using Harmony33), and unsupervised clustering. Based on the criteria we used for the infant cohort (excluding viral loads, which were not available for the adult dataset), we categorized adult patients into three clinical groups, from lower to higher severity (aG1, aG2 and aG3; Fig. 7a). The first round of clustering generated 28 clusters and seven cell-types (Fig. 7b-d) that were then analyzed separately.
CD14+ monocytes included four subclusters (SCs), which were categorized into: (i) ISGlow (SC0), (ii) ISGhi (SC1), (iii) ISGhi inflam+ (SC2) and (iv) CD163+ IL1R2+ (SC3) (Fig. 7e & Extended Data Fig. 8a,b). Cell composition analysis of CD14+ monocytes showed a decrease of SC0 (ISGlo) and a switch to SC1 (ISGhi) and/or SC2 (IL1B+ ISGhi) in pG2/pG3 and aG2/G3 groups compared to their respective controls. Interestingly, we reproduced the increase in SC3 (IL1R2+ CD163+) in patients receiving steroid therapy (Fig. 7f). The analysis of CD16+ monocytes showed a switch to an ISGhi state in both G2/G3 infants and G3 adults (Extended Data Fig. 8c-e).
Subclustering CD4+ T cells generated six SCs (Fig. 7g), annotated as: (i) naïve (SC0 and SC1; “SELL” and “CCR7”), with SC0 being ISGlow and SC1 ISGhi, (ii) memory (SC2 and SC4, “S100A4”), (iii) SOX4+CD4+ T cells (SC3; “SOX4”) and (iv) regulatory T cells (SC5, “FOXP3”, “IL2RA”). SC4 (CTLs) exhibited the cytotoxic cell-associated chemokine “CCL5” (Extended Data Fig. 8f). Cell composition analysis showed: (i) an expansion of naïve cells in pediatric healthy controls (pHC) compared to adult healthy controls (aHC), (ii) a contraction of naïve cells in pG2/pG3 compared to pHC/pG1, (iii) expansion of memory cells (SC2) in aHC compared to pHC, (iv) expansion of CTLs (SC4) in aHC, and (v) an expansion of SOX4+ CD4+ T cells in pHC. Tregs (SC5) did not significantly change upon infection in either infants or adults. The ISGhi CD4+ T cell SC (SC1) was present in both infected infants (2/10 pG1, 8/11 pG2 and 5/5 pG3) and adults (6/21 aG3 patients), although their proportion was higher in infants (Fig. 7h).
B cell subclustering generated seven SCs (Fig. 7i), annotated as: (i) naïve (SC0, “CCR7”), (ii) activated (SC1, “CD69”) (iii) memory (SC2, “S100A4”) and (iv) transitional (SC4; “CD9”, “MME (CD10)”). SC1 encompassed ISGhi B cells and SC5 upregulated “CD83” (Extended Data Fig. 8g). Cell composition analysis showed: (i) an expansion of naïve B cells in pHC compared to aHC, (ii) an expansion of memory B cells in aHC compared to pHC, and (iii) a subtle ISGhi expansion in infected adults (aG3) contrasting with a near-complete ISGhi status in the infected infants with severe disease (pG2/pG3) (Fig. 7j).
CD8+ T cell subclustering generated five SCs (Extended Data Fig. 9a), classified as: (i) naïve (SC0; “RPLs”), (ii) TEMRA (SC1; “GZMB”), (iii) Gzk (SC2, “GZMK”), (iv) MAIT (SC3, “KLRB1” and “ZBTB16”) and (v) proliferative CD8 T cells (SC4, “MKI67”) (Extended Data Fig. 9b). As expected, cell composition analysis showed expanded naïve CD8+ T cells in pHC and TEMRA/MAIT in aHC (Extended Data Fig. 9c). NK cell subclustering generated two SCs (Extended Data Fig. 9d). SC0 upregulated “FCGR3A” (CD16+ NK), and SC1 was XCL1+ and no disease severity-associated differences were detected (Extended Data Fig. 9e,f).
Overall, these comparative analyses (Extended Data Fig. 10a-d) showed that T and B cells from infants with severe disease almost entirely switch to an ISGhi state – while only a fraction of T and B cells in adults with severe disease become ISGhi.