Abstract
Type I and III interferons (IFN-I/λ) are key antiviral mediators against SARS-CoV-2 infection. Here, we demonstrate that the plasmacytoid dendritic cells (pDCs) are predominant IFN-I/λ source by sensing SARS-CoV-2-infected cells. We show that sensing of viral RNA by pDCs requires sustained cell adhesion with infected cells. In turn, the pDCs restrict viral spread by a local IFN-I/λ response directed toward SARS-CoV-2-infected cells. This specialized function enables pDCs to efficiently turn-off viral replication, likely by a concentrated flux of antiviral effectors at the contact site with infected cells. Therefore, we propose that pDC activation is essential to locally control SARS-CoV-2-infection. By exploring the pDC response in patients, we further demonstrate that pDC responsiveness correlates with the severity of the disease and in particular that it is impaired in severe COVID-19 patients. Thus, the ability of pDCs to respond to SARS-CoV-2-infected cells could be a key to understand severe cases of COVID-19.
Highlights
pDCs are immune cells against SARS-CoV-2-infected cells
pDC-mediated IFN-I/λ response against SARS-CoV-2 infected cells control COVID- 19 progression
pDC response by SARS-CoV-2 is restricted to IRF7-prioritized signaling leading to antiviral control
pDC antiviral response directed toward contacting SARS-CoV-2-infected cells
Introduction
The innate immune system acts as the first line of defense for the sensing of viral infection. This involves rapid recognition of pathogen-associated molecular patterns (PAMPs), including viral nucleic acids, by pattern recognition receptors (PRRs). This recognition results in an antiviral response characterized by the production of type I and III (λ) interferons (IFN) and other pro-inflammatory cytokines, along with the expression of IFN- stimulated genes (ISGs). Whilst type I and III/λ IFNs interact with distinct receptors, they both induce similar signaling pathways and effector factors, thus referred herein to as the IFN-I/λ response (Kim and Shin, 2021; Lazear et al., 2019). This host response suppresses viral spread by blocking the viral life cycle at multiple levels, hereby promoting virus clearance. The IFN-I/λ response also mediates immunomodulatory effects in surrounding tissues and imparts the onset of the adaptive immune response (Crouse et al., 2015; Makris et al., 2017).
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) emerged in December 2019 and is responsible for the still-ongoing coronavirus disease 2019 pandemic (Chan et al., 2020). Although most SARS-CoV-2-infected individuals experience asymptomatic to mild disease, others develop respiratory distress syndrome that is lethal in the most severe cases. Importantly, the IFN-I/λ response is now thought to be a critical host response against SARS- CoV-2 infection and its pathogenesis (Grajales-Reyes and Colonna, 2020; Hadjadj et al., 2020; Kim and Shin, 2021; Sa Ribero et al., 2020; Saichi et al., 2021; Trouillet-Assant et al., 2020). Recent studies have shown that SARS-CoV-2 can evade from this initial control by the IFN-I/λ response via manifold inhibitory mechanisms interfering with both the sensing and the signaling pathways within infected cells (reviewed in (Kim and Shin, 2021; Min et al., 2021; Park and Iwasaki, 2020; Sa Ribero et al., 2020)), and e.g., illustrated in (Kimura et al., 2021; Lei et al., 2020; Miorin et al., 2020; Saichi et al., 2021 ; Wang et al., 2021; Xia et al., 2020)). This immune evasion might lead to an increased viral load, followed by widespread inflammations (Sa Ribero et al., 2020). Individual immune responses against viral infection can thus be extremely heterogenous, ranging from robust and fast IFN-I/λ production to impaired and fatal defect in IFN-I/λ-mediated immunity, thereby highlighting the importance of IFN-I/λ for host defense against respiratory infections. Timing of the IFN-I/λ production also plays a crucial role in related Coronaviruses (e.g., MERS-CoV and SARS-CoV-1) (Channappanavar et al., 2016). Delayed IFN-I/λ signaling associated to robust virus replication indeed promotes the accumulation of pathological monocyte-macrophages, resulting in lung immunopathology, vascular leakage and suboptimal T cell responses (Channappanavar et al., 2016). Early reports on SARS-CoV-2 also suggest an association between severe COVID-19 and low level of IFN-I/λ but overproduction of inflammatory cytokines (Hadjadj et al., 2020; Lucas et al., 2020; Yao et al., 2021a). Accordingly, deficiency, neutralization by anti-IFNα-receptor autoantibodies or viral-mediated inhibition of the IFN-I/λ response aggravates SARS-CoV-2 pathogenesis (Asano et al., 2021; Bastard et al., 2021a; Bastard et al., 2021b; Bastard et al., 2020; King and Sprent, 2021; Lopez et al., 2021; Zhang et al., 2020). It is therefore critical to understand the regulation of the optimal production and activity of IFN-I/λ.
The plasmacytoid dendritic cells (pDCs) are a unique immune cell type specialized for rapid and massive production of IFN-I (approx. 1000-fold more than other cell types) and IFNλ (Reizis, 2019). pDCs possess multiple adaptations to efficiently produce IFN-I/λ (Blasius et al., 2010; Esashi et al., 2012; Reizis, 2019; Sasai et al., 2010; Webster et al., 2016). As pDCs are refractory to most viral infections, their response is not directly repressed by viral proteins. Thus, this aspect also contributes to the exceptional magnitude of pDC IFN-I/λ production (Decembre et al., 2014; Reizis, 2019). The main viral-sensors responsible for pDC activation are Toll-like receptors (TLR)-7 and -9 that recognize viral RNA and DNA, respectively (Reizis, 2019). A recent report suggested that pDCs activated by SARS-CoV-2 differentiate into cytokine- and IFN-secreting effector cells, in a TLR-dependent manner (Onodi et al., 2021). Studies on related coronaviruses have demonstrated that pDCs migration into the lungs, and their rapid production of IFN-I is essential for the control of CoV lethal infections (Cervantes-Barragan et al., 2007; Chen et al., 2010; Lucas et al., 2020). Nonetheless, how pDCs respond to SARS-CoV-2-infected cells and how this response evolves in the course of viral infection are still open questions.
Here, we explored the cellular mechanisms underlying the IFN-I/λ response against SARS- CoV-2 infection. Our results uncovered that pDCs establish cell contact with SARS-CoV-2 infected cells via αLβ2integrin/ICAM-1 adhesion complex and regulators of actin network. This physical contact between pDCs and infected cells is required for the pDC-mediated antiviral response via TLR7 recognition. Capitalizing on the identification of modalities of this previously-unsuspected activation mechanism, we further studied how pDC IFN-I/λ response associates with COVID-19 severity. We demonstrated that the differentiation of pDC into different subsets with distinct phenotypes and functions is restricted when activated by contact with SARS-CoV-2-infected cells. Consistently, this pDC activation preferentially induces a local IFNI/λ response, leading to efficient antiviral control directed towards the infected cells.
Results
Robust activation of pDCs in response to SARS-CoV-2-infected cells
Respiratory epithelial cells represent the first infected tissue in the course of SARS-CoV-2 infection. To investigate which hematopoietic cell type is primarily responsible for the IFN- I/λ response against SARS-CoV-2 infection, human peripheral blood mononuclear cells (PBMCs) isolated from healthy donors were cocultured with SARS-CoV-2-infected human lung-derived cells or exposed to their supernatants (SN). Human alveolar basal epithelial cell lines (Calu-3 cells and A549 cells expressing the angiotensin-converting enzyme 2, A549- ACE2 cells) were infected for 48 hours prior to the collection of their SN or cells for incubation or coculture with PBMCs (including pDCs), both for 16-18 hours. We found that PBMCs respond by a robust secretion of IFNα when cocultured with SARS-CoV-2-infected cells (Figure 1A-B). In contrast, the SN of SARS-CoV-2-infected cells failed to trigger IFNα secretion by PBMCs (Figure 1A-B). As expected from previous publications, SARS-CoV-2- infected cells did not produce themselves detectable level of IFNα (Blanco-Melo et al., 2020; Li et al., 2021; Lucas et al., 2020; Vanderheiden et al., 2020; Yamada et al., 2021; Yao et al., 2021b; Zhou et al., 2020). Plasmacytoid dendritic cells (pDCs) are known to robustly produce IFN-I/λ, notably IFNα (Reizis, 2019). In line with this, antibody mediated- depletion of pDCs from PBMCs abolished IFNα secretion in response to co-cultured SARS- CoV-2-infected cells (Figure 1A-B; line PBMC+/pDC-). To further demonstrate that pDCs are the major source of IFNα upon incubation with SARS-CoV-2-infected cells, pDCs were immuno-isolated from PBMCs (over 92% purity as we have previously reported (Assil et al., 2019; Decembre et al., 2014)). Purified pDCs potently produced IFNα in response to co- culture with SARS-CoV-2-infected cells, but not in the presence of uninfected cells (Figure 1A-B).
Consistent with these results, we further showed that IFNα producer cells were markedly enriched in the cell population gated as pDC as compared to other hematopoietic cell types (non-pDCs) (Figure 1C and S1A). Furthermore, IFNα expression level per pDC was very elevated (i.e., 2-log increased of mean fluorescence intensity over background; sub-selected as IFNαhi) (Figure 1C, right panel). Most IFNα−positive pDCs concomitantly produced IFNλ in response to SARS-CoV-2-infected cells (Figure 1C). Of note, the IFNα response mounted by pDCs in response to SARS-CoV-2-infected cells largely exceeded the one elicited by stimulation with TLR agonists i.e., imidazoquinoline compound [R848] and synthetic analog of double-stranded RNA polyinosinic-polycytidylic acid [polyI:C] (Figure 1C).
In contrast, this synthetic agonist led to a more potent upregulation of surface expression of activation markers including the programmed cell death ligand-1 (PD-L1) and CD83 as compared to coculture with SARS-CoV-2-infected cells (Figure 1D). This suggested that the pDC response to SARS-CoV-2-infected cells is likely qualitatively distinct from stimulation by soluble agonists, likely because PD-L1 and CD83 are both NF-κB-induced activation markers, and thus involving signaling distinct from the IFN-I/λ response (Antonangeli et al., 2020; Berchtold et al., 2002). Next, we tested whether the sensing of SARS-CoV-2-infected cells required a productive infection of pDCs, using a recombinant SARS-CoV-2 infectious clone expressing mNeongreen reporter (icSARS-CoV-2-mNG (Xie et al., 2020)). No infection of pDCs (defined as CTV+-pDCs; Figure S1I) was detected when in contact with icSARS-CoV-2-mNG-infected A549-ACE2 cells (Figure S1I). However, in the same coculture set up, infection by icSARS-CoV-2-mNG was readily detected in initially uninfected/naive RFP+ A549-ACE2 cells, hence proving validation of the efficient viral transmission (Figure S1I). The absence of productive infection of pDCs is consistent with previous observations made with different viruses and might be explained by the specialized function of pDCs for rapid and potent antiviral response leading to abortive infection (Decembre et al., 2014; Onodi et al., 2021; Reizis, 2019; Webster et al., 2018).
Taken together, these results strongly suggest that IFNα is robustly produced only by pDCs via sensing of SARS-CoV-2-infected cells and that this sensing most likely induces a specific activation state in pDCs.
Short range sensing of infected cells via cell contact triggers the pDC IFNα response
We observed that cell-free SN from SARS-CoV-2-infected cell types failed to trigger IFNα production by PBMCs (Figure 1A-B) or by purified pDCs (Figure 1E-H), even when used at a multiplicity of infection (MOI) of 5 per pDC. Furthermore, SARS-CoV-2 does not productively infect pDCs (Figure S1I). Distinct from a previous report and to avoid possible misinterpretation due to contamination by cell debris and/or floating cells (Onodi et al., 2021), we selected MOI and incubation time for SARS-CoV-2 infection so that no cytolytic effect was detectable in infected human lung-derived cells when cells and SNs were collected for coculture with pDCs (see Experimental procedures section). To further determine if cell- to-cell contacts were required for the transmission of the immunostimulatory signal to pDCs, we used transwell chambers containing SARS-CoV-2-infected cells and pDCs separated by a 0.4 m permeable membrane. This physical cell separation fully prevented IFNα production by pDCs (Figure 1E-H). Similar results were obtained for pDC stimulated by SARS-CoV-2- infected lung-derived cell lines Calu-3, A549-ACE2, H358-ACE2 as well as Huh7.5.1 cells, confirming that this feature was not cell type restricted (Figure 1E-F and S1D-E). Again, consistently with results shown in Figure 1A-B and previous reports (Blanco-Melo et al., 2020; Li et al., 2021; Lucas et al., 2020; Vanderheiden et al., 2020; Yamada et al., 2021; Yao et al., 2021b; Zhou et al., 2020), none of the SARS-CoV-2-infected cell types released themselves detectable levels of IFNα (Figure 1E-F).
To further define the mechanisms underlying pDC activation upon contact with SARS-CoV- 2-infected cells, we assessed the implication of cell adhesion complexes in this process. We focused on αL integrin and its ligand Intercellular Adhesion Molecule (ICAM)-1 (also called CD54) (Rothlein et al., 1986), respectively highly expressed by pDCs and by various cell types, guided by previous studies on the regulation of pDCs in other infection contexts (Assil et al., 2019). Antibody-mediated blockade of both αL integrin and ICAM-1 greatly prevented pDC IFNα production (Figure 1G). The engagement of integrins by their ligands is known to induce local recruitment of the actin network. Notably Arp2/3 complex mediates actin nucleation by recruiting and branching actin filaments within network (DeMali et al., 2002; DeMali et al., 2014; Rotty et al., 2013; Vicente-Manzanares et al., 2009). Pharmacological inhibition of Arp2/3 complex impaired IFNα production by pDCs in coculture with SARS- CoV-2-infected cells, in a dose dependent-manner (Figure S1G). This suggested that cell adhesion-induced actin recruitment is likely involved in the structuration of cell contacts. Using specific TLR7 antagonist (i.e., IRS661), we showed that the endosome-localized TLR7 sensor mediates the sensing of SARS-CoV-2-infected cells by pDCs (Figure S1H). Remarkably, similar levels of IFNα secretion were reproducibly obtained for pDCs isolated from the blood of a cohort of healthy donors (Figure 1H; distinct healthy donors as n=11 and n=15 for A549-ACE2 and Calu-3 cells respectively).
Together our results demonstrated that physical contacts between pDCs and SARS-CoV-2- infected cells are required for pDC IFNα production. This contact presumably involves cell adhesion mediated by αLintegrin/ICAM-1 complexes and the actin network, and leads to a robust TLR7-dependent IFNα response by the pDCs.
The IFN-I/λ signature observed in patients at early time point of SARS-CoV-2 infection is associated with viral clearance
Based on the importance of pDCs as the main cell type producing IFNα in response to SARS-CoV-2-infected cells, we sought to explore how this herein-uncovered mechanism for potent IFN-I/λ response is modulated in the course of the infection in patients and how it could relate to COVID-19 severity. A longitudinal study of IFN-I/λ response was done with different subsets of patients: i) critically ill, herein referred to as Severe patients, who presented acute respiratory distress syndrome or severe pneumonia at hospital admission and required mechanical ventilation in intensive care units, and ii) patients with mild symptoms (i.e., low-grade fever, cough, malaise, rhinorrhea, sore throat), that group was sub-divided according to the days of sample collection post-symptom onset, i.e., Mild early for the first two weeks and Mild late at the later time points. Of note, most patients were SARS-CoV-2 positive in nasal swab samples by qPCR at sampling time in the Mild early group but not anymore or presenting low viral levels in the Mild late group. All patients and the analyzed time-points are listed in Table 1 and S1, which provides information on the clinic and viral loads in nasal swab samples.
We quantified the IFN-I/λ levels in blood samples of infected patients at the transcriptional level (IFN-I signature by by Nanostring technology) and at the protein level for secreted IFNα, IFNλ 1, IFNγ together with IL-6. Both approaches revealed an elevated IFN-I/λ response at early time points (within the first 10-11 days post-onset of symptoms) for all patients with mild symptoms, that seemed to vanish over time, mirroring the controlled decrease of the viral load (Figure S2A and Table S1). Of note, the level of the pro- inflammatory cytokine IL6 was elevated in Severe patients (Figure S2A and Table S1), in line with the previously reported production of pro-inflammatory cytokines in COVID-19 patients (McGonagle et al., 2020; Vanderbeke et al., 2021).
Overall, the patients experiencing mild infection demonstrated high IFN-I/λ blood levels at early time-points. Furthermore, the inflammatory response appeared to be correlated with disease severity, in agreement with previous reports (Galani et al., 2021; Hadjadj et al., 2020; McGonagle et al., 2020; Vanderbeke et al., 2021).
The responsiveness of pDCs to SARS-CoV-2-infected cells is impaired in infected patients
We then determined the ability of PBMCs isolated from the different groups of patients to respond to ex vivo stimulation by SARS-CoV-2-infected cells. As our results pointed out the importance of pDCs in the IFN-I/λ response mounted against SARS-CoV-2 infected cells, we primarily analyzed pDCs as well as other cell types selected for their potential to play pivotal first defense against viruses. Especially, the Th1-promoting myeloid/conventional dendritic cell subset (mDC1), which produces IFNλ via TLR3-mediated recognition of viral RNA (Lauterbach et al., 2010; Yoshio et al., 2013) and the Th2-promoting myeloid/conventional DC subset (mDC2) and monocytes i.e., Human Leukocyte Antigen – DR isotype (HLA- DR)+CD14+populations known to produce proinflammatory cytokines (Bosteels et al., 2020; Leal Rojas et al., 2017; Merad et al., 2013; Mildner and Jung, 2014; Wong et al., 2011). This response was compared to the one induced by agonists. Agonists were selected in accordance with the targeted cell types: TLR7/TLR3 agonist [polyI:C/R848] for pDC (TLR7) and the mDC1 (TLR3), and TLR-4 agonist [lipopolysaccharide; LPS] for mDC2 and HLA- DR+CD14+ populations. A group of healthy donors comprising similarly treated samples was used as reference. Post-stimulation, we performed a multiparametric flow cytometry analysis to define the profiles of cell surface expression of several activation markers along with intracellular cytokines. These responses were assigned to different pivotal innate immune cells by designing a combined panel of cell type markers, along with the exclusion of other cell types (see gating strategy in Figure S2B).
Principal component analysis (PCA) of the flow cytometry datasets was performed within each cell population and stimulation condition to test whether differences in activation and cytokine expression could be linked to disease severity (Figure 2). PCA allows to reduce high dimensional data into principal components that maximize the variance of the projected data and potentially identify the underlying factors associated with it. In the upper panels (Figure 2), each patient is placed with respect to its distribution in the first two principal components (PCs), which explain most of the data variance. In the lower panels, vectors corresponding to the eigen decomposition of the data covariance matrix are depicted to identify features that explain the observed variance. For pDCs analyzed in the unstimulated condition, the first two principal components (PCs) revealed a differential segregation of the patients: the Severe group (in red), and to some extent mild early group (in purple) were mainly spread across the first PC (which was explained by IFNα, CD80 and PD-L1), while healthy and Mild late individuals were tightly clustered in both PCs (Figure 2A, left panels). An opposite segregation of the patient groups was observed in pDCs when PBMCs were cocultured with SARS-CoV-2-infected cells. Especially, this stimulation induced a spread across the first PC (mainly driven by IFN /IFNα and CD83 expression) of patients from the Mild and healthy groups, in accordance with the pDC ability to respond to SARS-CoV-2- infected cells (Figure 2A, middle panels). Of note, the healthy individuals were spread across the two PCs, whereas the Mild early and Mild late groups dispersed primarily along the first PCs. On the contrary, individuals from the Severe group were spread across the second PC (driven by CD80 and PD-L1 expression). Regarding pDC response to synthetic agonist, all groups were similarly spread across the first two PCs, while the Severe group was tightly grouped within the second PC (driven by IFNλ expression) (Figure 2A, right panels). These results suggested that pDCs from Severe individuals display differences in activation markers and cytokine expression compared to individuals from all the other groups, especially when stimulated by SARS-CoV-2-infected cells. PCA analysis performed on mDC1 subset demonstrated less marked contrast among the patient groups upon stimulation by SARS- CoV-2-infected cells, although the Severe group appeared more tightly grouped across both PCs (Figure 2B; middle panels). Upon agonist stimulation, mDC1 from the Severe group were also tightly grouped within the second PC (driven by IFNλ1, CD83 and PD-L1; Figure 2B; right panels). PCA analyses of mDC2 and HLA-DR+CD14+ populations revealed no marked contrast across the patient groups in the unstimulated condition and SARS-CoV-2 stimulation, while the Severe individuals tended again to be grouped in agonist stimulation (Figure 2C-D).
Taken together, our results highlighted that the contrast between Severe patients and the other groups is more pronounced in pDCs than in other PBMC subsets and upon stimulation by SARS-CoV-2-infected cells.
COVID-19 severity primarily associated with a blunted pDC IFNα response to SARS-CoV-2-infected cells
We further analyzed the expression of activation markers and cytokines individually at the single-cell level in the defined cell types, focusing first on markers identified as driving forces in the PCA analyses. In keeping with results of Figure 1, the frequency of IFNα + pDCs greatly increased in response to SARS-CoV-2-infected cells in the healthy donor group (Figure 3A, green arrow). In sharp contrast, pDCs from Severe patients failed to be activated by SARS-CoV-2-infected cells as similarly revealed by the detection of IFNα+, IFNλ+ and CD83+ pDC (Figure 3A-C; red bars and arrows). Of note, a strong basal level of pDC IFNα expression was observed in the absence of stimulation for the Severe patients. The response to agonist stimulation was also greatly diminished in pDCs from Severe patients compared to healthy donors and patients with mild symptoms, as shown by the level of activation markers (CD83, CD80 and PD-L1; Figure 3C-E; red arrows).
In line with the results obtained with pDCs, expression of IFNλ1 was barely detectable in the mDC1 subset of Severe patients upon stimulation by SARS-CoV-2-infected cells and agonists, comparatively to the other groups (Figure 3F; red arrows). Further, our data showed a potent upregulation of IL6 by HLA-DR+CD14+ subset upon agonist treatment in Severe patients and Mild early (Figure 3G). HLA-DR+CD14+ population represents a highly frequent cell subset of non-mDC2 (e.g., among the gated live cells+ lin- HLA-DR+: 65.7% are HLA-DR+CD14+ population and the exclusion of mDC2 represent only 4.85%; Figure S2A). In accordance, a high frequency of IL6+ cells was also observed for the non-mDC2 populations (Figure 3G, right panel). Likewise, Severe patients presented an elevated level of blood IL6 (Figure S2A). These results are consistent with the previously reported production of pro-inflammatory cytokines by monocytic cells in patients with severe disease (Hadjadj et al., 2020; Vanderbeke et al., 2021), and provided further insights into the differential responses of other cells.
We thus sought to define the dynamics of the response in COVID-19 patients. First, kinetic analyses in Mild patients revealed an elevated detection of pDC IFNα+ levels in absence of stimulation that vanished over time while, relatively, their ability to respond to SARS-COV- 2-infected cells increased at late time-points (Figure 3H, left panel). Basal level of pDC IFNα+ (i.e., in absence of stimulation) was also observed for Severe patients and associated to a limited induction in response to SARS-COV-2-infected cells at all analysed time-points (Figure 3H, right panel). Kinetic analysis was performed for a Severe patient, who had anti- IFNα antibody detected in the blood (patient description included in Table 1; legend) (Asano et al., 2021; Bastard et al., 2021a; Bastard et al., 2021b; Bastard et al., 2020; King and Sprent, 2021; Lopez et al., 2021; Zhang et al., 2020). It showed that the ability of pDC to respond to stimulation was blunted in this patient (Figure 3H, right panel; represented by starts with yellow-center).
Together our results demonstrated that the monocytic subset likely contributes to an exacerbated pro-inflammatory response implying notably IL6 production, but that is presumably not triggered directly by the contact with SARS-CoV-2-infected cells. As opposed, impaired IFN-I/λ response following cell contact with pDCs from Severe patients, including those with anti-IFNα, suggested a ‘silencing/exhaustion’ of pDCs in this context. This might be due to the lack of an amplification-loop by ISG resulting in lower activated state and IFN production by pDCs and to some extent, similarly, IFNα 1 expression by mDC1.
Limited pDC differentiation and cytotoxic activity induced by the contact with SARS- CoV-2-infected cells compared to stimulation by cell-free agonists
As we found that pDC activation is a salient feature that correlates with severe COVID-19, we aimed to determine how the contact with SARS-CoV-2-infected cells impacts the varied downstream signaling and function of pDCs. First, we analyzed the expression of pDC surface molecules enabling the stimulation of adaptive responses, namely HLA-DR, an MHC class II cell surface receptor driving the activation of CD4+ T cells, and the B cell ligand CD70 expressed by pDCs and known to interact with CD27 and to induce proliferation and differentiation of B cells into plasmablast (Shaw et al., 2010). Our results showed that pDCs upregulated both surface molecules in response to contact with SARS-CoV-2-infected cells, although at a lower level compared to stimulation by cell-free virus (influenza virus; flu) and synthetic agonists of TLR7/3 [R848/polyI:C; R/p] and TLR7 [imiquimod; IMQ] (Figure 4A- B). Similarly, surface expression of CD83, an activation marker for antigen presenting cells (Gomez et al., 2004; Li et al., 2019), was induced upon contact with SARS-CoV-2-infected cell and jointly expressed with PD-L1 by the same pDC subset (Figure 4C). These results obtained with purified pDCs are in agreement with results obtained with pDCs present in PBMCs and cocultured with SARS-CoV-2-infected cells (Figure 1D). Of note, SN from SARS-CoV-2-infected cells induced HLA-DR, CD83 and PD-L1 expression, yet not as potently as other soluble agonists (Figure 4A and 4C).
pDCs are now recognized to be an heterogeneous population composed of subsets endowed with diversified functions (Abbas et al., 2020; Alculumbre et al., 2018; Bryant et al., 2016; Matsui et al., 2009; Onodi et al., 2021; Reizis, 2019; Villani et al., 2017). As stimulation of pDCs can impact the frequency and phenotype of these subsets, we assessed the expression of a set of surface molecules previously assigned to define specific subpopulations of pDCs i.e., CD2, CD5, AXL, CD80 and PD-L1 (Alculumbre et al., 2018; Bryant et al., 2016; Matsui et al., 2009; Onodi et al., 2021; Reizis, 2019; Villani et al., 2017). CD2hi pDCs have a survival advantage and are able to efficiently trigger proliferation of naive allogenic T cells (Bryant et al., 2016; Matsui et al., 2009; Zhang et al., 2017). Stimulation of pDCs by contact with SARS-CoV-2-infected cells did not impact the frequency of CD2hi pDCs, and this CD2hi subset displayed an activation profile similar to the one of CD2low subset (Figure 4D and S4B). CD2hi CD5+ AXL+ pDCs were recently proposed as a defined subset that display limited IFN-I production capacity but can potently activate T cells (Villani et al., 2017). The CD2hi CD5+ AXL+ pDCs, which represent a very scarce subpopulation of pDCs (Villani et al., 2017), modestly decreased upon stimulation by SARS-CoV-2-infected cells, their SN or TLR-agonists, yet whether this intriguing decrease is relative to the activated state of pDCs requires further investigation (Figure S4C). We then addressed the diversification of pDCs into functionally distinct populations defined by PD-L1/CD80 expression. In agreement with previous reports, the stimulation by cell-free agonists and influenza virus triggered the differentiation into both PD-L1+CD80+ and PD-L1-CD80+ pDC subsets (Alculumbre et al., 2018; Onodi et al., 2021). In sharp contrast, direct contact with SARS- CoV-2-infected cells restricted the differentiation of pDCs towards PD-L1+ CD80- subset (Figure 4E).
Collectively, these results revealed that, unlike activation by cell-free viruses and agonists, direct activation of pDCs by SARS-CoV-2-infected cells restricted their differentiation into specific functional subsets.
We observed that pDCs expressed PD-L1 upon stimulation by SARS-CoV-2-infected cells. Therefore, we further examined the induction of cytotoxic/immune-suppressive activity of pDCs. In keeping with the previously-reported induction of membrane-bound TNF-related apoptosis-inducing ligand (mTRAIL) expression by pDCs upon viral stimulation (e.g., HIV) (Achard et al., 2017; Colisson et al., 2010; Gandini et al., 2013), we found that mTRAIL was readily co-expressed along with PD-L1 and CD83 upon stimulation by influenza virus and synthetic agonists. Nonetheless, mTRAIL upregulation was more limited upon pDC stimulation by SARS-CoV-2 compared to stimulation by cell free agonist (Figure 4F, total % of the bars). Moreover, the frequencies of Annexin V+/7-ADD+ apoptotic Calu-3 and A549 cells in cocultures with pDCs were similar between infected (i.e., pDC activation) and uninfected (i.e., no pDC activation) conditions, hence suggesting that mTRAIL expression on activated pDCs did not endow these cells with cytotoxic activity. (Figure 4G). In line with this, the contact with activated pDCs did not markedly impact the viability of the cocultured SARS-CoV-2-infected cells (i.e., when comparing condition with or without the anti-αL integrin, which inhibits contact and pDC activation), nor the viability of activated pDCs themselves, even when analyzed after 48 hours of coculture (Figure 4H).
Overall, these results showed that activation of pDCs by SARS-CoV-2-infected cells did not induce cytotoxic/immune-suppressive activity, mostly leading to a restricted diversification into functional pDC subsets and likely toward to their ability to robustly produce IFN-I/λ.
pDC activation by SARS-CoV-2-infected cells primarily leads to the antiviral state viaIFN-I/λ production
We then sought to examine deeper the signaling pathways at play in pDCs upon activation by coculture with SARS-CoV-2-infected cells. TLR7-dependent activation of pDCs can induce a ‘bifurcated’ signaling leading to (1) IFN-I/λ production mostly via IRF7-related signaling and other cytokines and (2) activation molecules via NF-κB-pathway (Reizis, 2019; Webster et al., 2018). We noticed that expression of the activation marker HLA-DR, CD70, CD83, mTRAIL by pDCs was weaker when induced by contact with SARS-CoV-2-infected cells compared to soluble agonist stimulation (Figure 4).
These markers/proteins are primarily regulated by NF-κB-mediated signaling (Table S3) (Antonangeli et al., 2020; Berchtold et al., 2002). To further define the signaling active in pDCs, we quantified transcripts levels of representative key effectors regulated by IRF/IFN-I signaling (i.e., mxA, isg15 and ifnλ) versus those primarily regulated by NF-κB-pathway (i.e., il-6 and tnfα) (Table S3) (Liu et al., 2017; Mesev et al., 2019). Cocultures of pDCs and SARS-CoV-2-infected cells induced more the IRF7/IFN-I-regulated molecules than the representatives of the NF-κB-pathway (Figure 5A). This was again confirmed by the quantification of secreted cytokines in the cocultures, demonstrating higher levels of IRF7/IFN-I-regulated IFNλ1/2/3 compared to TNFα (Figure 5B compared to 5C). This observation was also in agreement with the high level of secreted IFNα in similar experiments (Figure 1E-H). Of note, no detectable cytokine and low level of transcript upregulation were detected in SARS-CoV-2-infected cells in the absence of pDCs, or when pDCs were stimulated by SARS-CoV-2 SNs. In these assays, the low activation level of the NF-κB-pathway and/or its variability among different infected cell types can be explained by a contribution of the infected cells, as a feedback loop of the response to activated pDCs. Hence, we assessed cytokine expression at single-cell level by flow cytometry in the gated pDCs (Figure S4A). Stimulation by SARS-CoV-2-infected cells elicited higher frequency of IFNα+ pDCs compared to TNFα+ pDCs (Figure 5D). Remarkably, almost all TNFα+ pDCs were also IFNα+, possibly implying an activation of the NF-κB-pathway subsequent to IRF7/IFN-I response. Again, this result contrasted with the limited frequency of IFNα+ pDCs in response to cell-free agonist (Figure 5D). Similar analysis performed for IFNα+ combined with IFNλ1+ also revealed that virtually all IFNλ1+ pDCs were IFNα+ (Figure 5E).
Collectively, our data provided evidence that, in contrast to stimulation with cell-free viruses and agonists, the pDC response to contact with SARS-CoV-2-infected cells is biased towards IRF7-mediated signaling that leads to a robust IFN-I/λ production.
The pDC antiviral response is directed toward the contacting SARS-CoV-2 infected cells
As the response to SARS-CoV-2-infected cell primarily induced IFN-I/λ antiviral signaling, we next aimed to define how the pDC response inhibits viral propagation. pDCs were cocultured for 48 hours with icSARS-CoV-2-mNG-infected A549-ACE2 cells (mNG+) and uninfected A549-ACE2 RFP+ cells, and viral spread was quantified by flow cytometry. The results demonstrated that the pDC response readily prevented viral spread from mNG+ infected cells to initially uninfected RFP+ cells, whilst also slightly diminishing the replication in mNG+ infected cells during the coculture incubation time (Figure 5F-G). The inhibition of contact between pDCs and infected cells via blockage of αL integrin restored viral propagation to levels comparable to those measured in the absence of pDC (Figure 5F- G). This indicated that the establishment of cell-contact via adhesion molecules is required for pDC-mediated antiviral response.
Interestingly, the impact of pDC antiviral response on cells infected prior to coculture was quite modest as compared to the spread to uninfected cells, likely owing to the inhibition of IFN-I/λ signaling by SARS-CoV-2 (Chen et al., 2021; Yang et al., 2020). Therefore, we hypothesized that the reduction could be more potent if infected cells were directly in contact with pDCs as such contact could allow a concentrated antiviral response toward the infected cell. To test this hypothesis, we established an assay of 24 hour-long live-imaging of the coculture of pDCs (stained with CM-Dil; red) and icSARS-CoV-2-mNG infected cells (mNG+; green) using spinning-disk confocal microscopy. As depicted by the example of time-sequence imaging (Figure 6A), pDC contact with mNG+ infected cells lead to a control of viral replication in the targeted infected cells, reflected by the decreased mNG fluorescent reporter signal. We controlled that these infected cells, although not mNG+ anymore, were still physically present by using enhanced fluorescent signal analysis (Figure 6A; lower panels). Viral control by pDCs was seen for pDC/infected cell contacts starting at different time points in the course of the coculture (i.e., up to 14 hours after record onset). Similar analysis performed for several infected cells, either in contact with pDC versus not in contact, revealed that the decrease in viral replication (i.e., mNG fluorescence intensity) was observed only for infected cells directly in contact with pDCs (Figure 6B, 6D-E). Of note, live- tracking of mNG fluorescence intensity performed in infected cells cultured without pDCs, provided the basal level of the mNG fluorescence intensity (done simultaneously with record of coculture with infected cells). This basal level was comparable to the one measured in infected cells cocultured with pDCs that were not in direct contact with the latter (Figure 6C- D). pDCs established long-lasting contact with infected cells i.e., mostly above 10 hours, and accordingly, the diminution of the mNG fluorescence intensity was detected several hours after the onset of contact, likely owing to the time-window needed to decrease viral replication (Figure 6E-F).
Overall, these results demonstrated that pDCs established sustained contact with SARS-CoV- 2 infected cells via αLβ2integrin/ICAM-1 adhesion complex leading to an efficient antiviral response directed toward the infected cells that shut down viral replication.
Discussion
Here we demonstrate that pDCs are the key mediators of the IFN-I/λ response against SARS-CoV-2-infected cells. Moreover, our study exploring the responsiveness of PBMCs from COVID-19 patients establishes that pDC functions are of pivotal importance against COVID-19 severity. The sensing of SARS-CoV-2-infected cells by pDCs is mediated by the establishment of sustained contacts via cell adhesion molecules. These cell-cell contacts are required to enable the sensing of viral RNA by endolysosome-localized TLR7 and to restrict viral spread. This scanning function of pDCs does not involve their productive infection for immune surveillance of infected cells. We further demonstrate that pDC activation by contact with SARS-CoV-2-infected cells is characterized by IRF7/IFN-I/λ-prioritized signaling, while passively excluding NF- B-mediated responses. The pDC-mediated IFN-I/κ response is targeted towards SARS-CoV-2 infected cells. This specialized function thus enables pDCs to efficiently turn-off viral replication, likely owing to a concentrated efflux of antiviral effectors at the contact site with infected cells.
pDCs critically control viral replication and the outcome of COVID-19 severity
Insights on several hallmarks of IFN-I/λ pathway as being critical in COVID-19 severity are emanating from recent publications. First, neutralizing autoantibodies against several cytokines and genetic defects affecting the IFN-I pathway have been identified in life- threatening COVID-19, as autosomal disorders of IFN-I immunity and autoantibodies underlie at least 10% of critical COVID-19 pneumonia cases (Asano et al., 2021; Bastard et al., 2021a; Bastard et al., 2021b; Bastard et al., 2020; King and Sprent, 2021; Lopez et al., 2021; Zhang et al., 2020). Second, patients with severe diseases exhibit reduced circulating pDCs and low plasma IFN-I/λ levels when compared to mild COVID-19 patients (Hadjadj et al., 2020; Lucas et al., 2020; Yao et al., 2021a). Third, IL-3, which increases innate immunity likely by promoting the recruitment of circulating pDCs into the airways, is reduced in the plasma of patients with high viral load and severity/mortality (Benard et al., 2021). This evidence highlights that several biomarkers of IFN-I/λ response are diminished in COVID-19 patients, and hereby points to pDCs as a primary candidate in the progression of COVID-19. Here, using an original approach to study innate immunity in ex vivo-stimulated PBMCs from COVID19 patients, we report that the functionality of pDCs is markedly blunted in severe patients, as observed upon their stimulation by SARS-CoV-2-infected cells as well as other TLR agonists.
pDC effector functions against SARS-CoV-2
pDCs comprise distinct subpopulations capable of varied functions and efficacy levels to mount an IFN-I/λ response (Abbas et al., 2020; Alculumbre et al., 2018; Bryant et al., 2016; Matsui et al., 2009; Onodi et al., 2021; Reizis, 2019; Rhodes et al., 2019; Villani et al., 2017). Viral infections and attendant inflammation potentially impact the frequency and functionality of the distinct pDC subpopulation including effect on pDC renewal (Abbas et al., 2020; Alculumbre et al., 2018; Macal et al., 2018). Such modulations could be imprinted either by the micro-environment via crosstalk with immune cells, or by pDC activation itself.
We now report that neither pDC subpopulation frequencies nor their distinct ability to respond to SARS-CoV-2 are impacted by stimulation and contact with SARS-CoV-2-infected cells. This is illustrated for CD2low/CD2hi pDCs that display similar activation profile upon stimulation with SARS-CoV-2-infected cells, and the frequency of CD2+CD5+AXL+pDC- like, although their low number limit this analysis.
Interestingly, when comparing stimulations by contact with SARS-CoV-2-infected cells versus cell-free activators (i.e., viruses and synthetic agonists), we found that the differentiation of pDCs defined by CD80 and/or PD-L1 expression are readily distinguishable depending on the TLR7 inducers. Whilst pDCs diversify towards all the different PD- L1/CD80 subsets upon cell-free stimulation (agonist and virus) in agreement with prior publications (Alculumbre et al., 2018; Onodi et al., 2021), the contact with infected cells restricts their evolution to PD-L1+CD80- subsets. Of note, previous reports suggested that PD-L1+CD80- pDCs are more efficient for IFN-I response compared to the other subsets (Abbas et al., 2020; Alculumbre et al., 2018; Onodi et al., 2021).
In accordance with this phenotype, the contact with SARS-CoV-2-infected cells induces a IRF7/IFN-I/κ prioritized signaling in pDCs, while passively excluding NF-κB-mediated responses. The ‘bifurcated’ signaling in TLR7-activated pDCs can occur independently toward either IFN-I/ production or NF- B activation leading to pro-inflammatory cytokines and activation markers. This bifurcation of signaling is expected to be dependent on the sub- cellular compartment in which the TLRs encounter activating signal: activation in early endosomes induces IFN-I while activation in endolysosomes triggers NF-κB mediated signaling (Combes et al., 2017; Reizis, 2019). We previously reported that, in the context of
other viral infections (i.e., Dengue, Chikungunya, Hepatitis C, Zika viruses), contact with infected cells similarly induced a IRF7/IFN-I/λ prioritized signaling in activated pDCs, as opposed to incubation of pDCs with cell-free viruses (Assil et al., 2019; Webster et al., 2018). Of note, we also show that stimulation of pDCs by cell-free SARS-CoV-2 supernatants induced the upregulation of some activation markers (e.g., HLA-DR, PD-L1), in accordance with other reports (Alculumbre et al., 2018; Onodi et al., 2021), but elicited virtually no IFN-I/λ response. We propose that the signaling downstream of TLR7, including the phosphorylation cascade might be impacted by cell polarity and physical contact with infected cells (Olayioye et al., 2013; Saitoh et al., 2017). Further studies will aim at addressing this question.
Cross-regulation of immune responses
Progression to severe COVID-19 predominates in elderly patients, advanced age being a factor suspected to aggravate disease progression and to weaken innate immunity, potentially including pDC responsiveness (Feng et al., 2021; Jing et al., 2009). This is in accordance with the demographic analysis of our COVID-19 cohort, as the age-ranges were 63.5-year- old [interquartile of 48.0-73.0] and 40.5-year-old [24-57] for Severe and Mild patients, respectively. The patient history and genetic factors can explain the differential ability of pDCs from patients to mount a response against SARS-CoV-2-infected cells, as illustrated by the heterogeneous immune responses of patients, with critical anomalies in functionality of pDC in severe COVID-19. Here, a kinetic analysis performed on a COVID-19 patient that presents auto-antibodies against IFNα (Bastard et al., 2021a; Bastard et al., 2021b; Bastard et al., 2020; Lopez et al., 2021) show that the responsiveness of pDCs to stimulation by SARS-CoV-2-infected cells was blunted. Future study is needed to expand this interesting preliminary observation, which already indicates that the IFN-I/λ response can reinforce via a positive feedforward regulation the pDC antiviral function.
Of note, comparison of responses across patients with different COVID-19 severities suggest that pDC response to SARS-CoV-2-infected cells inversely correlates with an exacerbated inflammatory response (as illustrated by IL6 production by monocytic cells) and a basal level IFN-I/λ and inflammation activity. It is tempting to speculate that the progressive enrichment of proinflammatory cytokines in the lung micro-environment can imprint pDC responsiveness. In this scenario, an excessive elicitation of pDC would lead to their functional ‘silencing’. Future study is needed to elucidate the underlining mechanism that could lead to such ‘exhausted’ pDCs, as reminiscent of the findings of impaired pDC function in a distinct infection context (Macal et al., 2018). In turn, the deficit of the antiviral control at the infected site owing to exhausted pDCs can then feedback as fuel for uncontrolled viral replication leading to more lung inflammation. Altogether our results thus highlighted possible cross-regulation between immune cells in the course of COVID-19.
Concluding remarks: importance for SARS-COV2 and possible insight for predictive markers
SARS-CoV-2 is a still-ongoing worldwide health threat, currently causing a significant human and economic burden and exacerbated by divergence into more severe variants. We provide compelling evidence that pDCs are a key cell type in the initiation of antiviral responses against SARS-CoV-2. Further, this study identified the failure of pDC response as critical in COVID-19 severity. Moving forward our finding shall provide guidelines for predictive biomarkers, as associated with pDC responsiveness to SARS-CoV-2 infections and background IFN-I/λ response at different stages of disease. Furthermore, strategies to boost the pDC response, and especially their recruitment to the lung, can lead to the development of potential therapeutics against pulmonary viral infections, including SARS-COV-2.
Data Availability
all data referred to in the manuscript will be available
START Methods
Detailed methods are provided in the online version of this paper and include the following:
KEY RESOURCES TABLE
CONTACT FOR REAGENT AND RESOURCE SHARING
EXPERIMENTAL MODEL AND SUBJECT DETAILS
Preparation of Viral Stocks and Infections Cell Lines and Primary Cell Cultures Cohort of SARS-CoV-2 infected patients. Reagents
METHOD DETAILS
Quantification of SARS-CoV-2 level in nasal swab samples of infected patients. Analysis of IFN-I/λ signatures in blood samples of SARS-CoV-2-infected patients.
Ex-vivo stimulation of PBMCs isolated from SARS-CoV-2-infected patients and healthy donors.
Coculture experiments using isolated pDCs Immunostaining and flow cytometry analysis Analysis of transcriptional levels by RT-qPCR Analysis of extracellular infectivity Viral Spread Assay Live imaging of coculture with spinning-disc confocal microscopy analysis
QUANTIFICATION AND STATISTICAL ANALYSIS
PCA analysis
Bioinformatic analysis of the cis-acting regulatory element in the promoter Statistical methods.
CONTACT FOR REAGENT AND RESOURCE SHARING
Further information and requests for resources and reagents should be directed to and will be fulfilled by the Lead Contact, Dr. Marlène Dreux (marlene.dreux@ens-lyon.fr)
EXPERIMENTAL MODEL AND SUBJECT DETAILS Preparation of Viral Stocks and Infections
The clinical isolate was obtained from patients referenced in the GISAID EpiCoVTM database: as BetaCoV/France/IDF0571/2020; accession ID: EPI_ISL_411218 (Pizzorno et al., 2020), kindly provided by Dr B. Lina. The infectious-clone-derived mNeonGreen SARS- CoV-2 (referred to as icSARS-CoV-2-mNG) was kindly provided by Dr Pei-Yong Shi and generated by introducing mNeonGreen into ORF7 of the viral genome (Xie et al., 2020). Viral stock of Influenza A Virus (Flu A/H1N1/New caledonia ; infectious titer of ≈ 10 plaque forming unit (PFU)/ml) (de Chassey et al., 2013) was produced as previously described and kindly provided by Dr V. Lotteau (CIRI, Lyon France).
Cell Lines and Primary Cell Cultures
SARS-CoV-2-infected cells included the human alveolar basal epithelial cell lines, Calu-3 cells (ATCC HTB-55), A549 cells (ATCC CCL-185) and NCI-H358 cells (ATCC CRL-5807), Huh7.5.1 cells (Dreux et al., 2012) and HEK-293 cells (ATCC CRL-1573). The A549 cells, NCI-H358 cells and HEK-293 cells were transduced to stably express the human angiotensin converting enzyme 2 (ACE2; accession number: NM_021804) using a lentiviral vector, as previously described (Dreux et al., 2012; Rebendenne et al., 2021). A549 cells were maintained in Roswell Park Memorial Institute (RPMI) 1640 Medium (Life Technologies) supplemented with 10% FBS, 100 units (U)/ml penicillin, 100mg/ml streptomycin and non-essential amino acids (Life Technologies) at 37°C/5% CO2. The NCI- H358 and Huh7.5.1 cells were maintained in Dulbecco’s modified Eagle medium (DMEM) (Life Technologies) with the same supplements and 2mM L-glutamine. Calu-3 cells were maintained in DMEM/Nutrient mixture F-12 Ham (1:1) (Life Technologies) supplemented with glutaMAX, 10% FBS, 100 units (U)/ml penicillin, and 100mg/ml streptomycin. All cell were maintained at 37°C/5% CO2.
pDCs were isolated from 450 ml of blood units obtained from adult human healthy donors and according to procedures approved by the “Etablissement Français du sang” (EFS) Committee. PBMCs were isolated using Ficoll-Hypaque density centrifugation. pDCs were positively selected from PBMCs using BDCA-4-magnetic beads (MACS Miltenyi Biotec) and cultured as previously described (Assil et al., 2019). PBMCs and pDCs were cultured in RPMI 1640 Medium (Life Technologies) supplemented with 10% FBS, 100 units (U)/ml penicillin, 100 mg/ml streptomycin, 2 mM L-glutamine, non-essential amino acids, 1 mM sodium pyruvate and 10 mM Hepes (Life Technologies) at 37°C/5% CO2.
Cohort of SARS-CoV-2 infected patients
The constitution of the cohort was done by the collaboration of the Hospices Civils de Lyon (HCL). This cohort consists of patient groups recognized by clinicians as: i) patients admitted in intensive care units for severe disease at hospital admission (i.e., acute respiratory distress syndrome or severe pneumonia requiring mechanical ventilation, sepsis and septic shock) are referred to as Severe group and ii) patients with mild symptoms (i.e., low-grade fever, cough, malaise, rhinorrhea, sore throat) are referred to as Mild early group when collected in the first two weeks and Mild late group for later time points. A detailed description of the patient information along with the levels of the IFN-I/III signature and viremia, as determined in blood and nasal swab samples, respectively, is provided in Table 1 and S1. As reference, blood samples from healthy donors were obtained from EFS and experimentally processed similarly. To limit the risk of inclusion of asymptotic healthy donor: i/ part of the blood samples was collected prior to the pandemic and ii/ for blood collected during the SARS-CoV-2 pandemic, systemic examination and questioning/interview of the donors were performed and included symptoms, prior contacts at risk and vaccination, Thus, blood samples were excluded from our study if ongoing and/or recent COVID-positivity was suspected.
Reagents
The antibodies used for immuno-staining are listed in the Key Resource table. Ficoll- Hypaque (GE Healthcare Life Sciences). Other reagents included LPS, TLR3 agonist (Poly(I:C); LMW) and TLR7 agonist (R848 and Imiquimod) (Invivogen); TLR7 antagonist (IRS661, 5’-TGCTT GCAAGCTTGCAAGCA-3’ synthesized on a phosphorothionate backbone; MWG Biotech); mouse ant αL integrin (clone 38; Antibodies Online); mouse anti-ICAM-1 (Clone LB-2 ; BD Bioscience): Arp2/3 complex inhibitor I (CK-666 ; Merck Millipore) ; Fc Blocking solution (MACS Miltenyi Biotec); Golgi-Plug, cytoperm/cytofix and permeabilization-wash solutions (BD Bioscience); IFNα and IFNλ1/2/3 ELISA kit (PBL Interferon Source); IL6 and TNFα ELISA kit (Affymetrix, eBioscience); 96-well format transwell chambers (Corning); IL6 and IFNλ by U-PLEX Custom Human Cytokine assay (Meso Scale Diagnostics, Rockville, MD) ; 96-Well Optical-Bottom Plates (Thermo Fisher Scientific); cell-labeling solution using CellTraceTM Violet Cell Proliferation Kit (Life Technologies ref # C34557, C34571), Live/Dead Fixable Dead Cell Stain Near-IR (Life Technologies ref #10119); Fixable Viability Dye eFluor 450 (Life Technologies); Zombie Aqua and Zombie Green Fixable Viability Kits (Biolegend); FITC Annexin V Apoptosis Detection Kit with 7-AAD (Biolegend); cDNA synthesis and qPCR kit (Life Technologies) ; poly-L-lysin (P6282, Sigma-Aldrich).
METHOD DETAILS
Quantification of SARS-CoV-2 level in nasal swab samples of infected patients
Nucleic acid extraction was performed from 0.2 mL naso-pharyngeal swabs using NUCLISENS easyMAG and amplification was performed using Biorad CFX96. Quantitative viral load was determined using four internally developed quantification standards (QS) targeting the SARS-CoV-2 N gene: QS1 to QS4 respectively at 2.5.106, 2.5.105, 2.5.104, 2.5.103 copies/mL of a SARS-CoV-2 DNA standard. These QS were controlled and quantified using the Nanodrop spectrophotometer (ThermoFisher) and Applied Biosystems QuantStudio 3D Digital PCR. In parallel, naso-pharyngeal swabs were tested using the CELL Control R-GENE kit (amplification of the HPRT1 housekeeping gene) that contains two quantification standards QS1 and QS2, at 104 copies/µL (50,000 cells/PCR i.e. 1.25.106 cells/mL in our conditions) and 103 copies/µL (5000 cells/PCR i.e. 1.25.105 cells/mL in our conditions) of DNA standard, respectively, to normalize the viral load according to the sampling quality.
Analysis of IFN-I/λ signatures in blood samples of SARS-CoV-2-infected patients
Transcript levels using Nanostring technology
Targeted transcripts of the IFN-I pathway included SIGLEC1, IFI27, IFI44L, IFIT1, ISG15, RSAD2, HPRT1, POLR2A and ACTB (Pescarmona et al., 2019). Transcript levels were determined using RNA extracted from the patient’s blood samples by Maxwell 16 LEV simply RNA Blood kits that comprised an individual DNAse treatment (Pescarmona et al., 2019). Next, total RNA was eluted in 40 µL RNAse-free water and concentration was quantified by spectrophotometry using a NanoVue (Biochrom). Only 200 ng of RNA were needed to achieve IFN signature with Nanostring technology. For the Elements system, capture (probe A) and tag (probe B) probe DNA oligos were designed by NanoString and synthesized by Integrated DNA Technologies (IDT). After hybridization at 67°C for 18-20 hours, samples were analyzed using the “High Sensitivity” protocol option on the nCounter Prep Station and counted on nCounter Digital Analyzer using maximal data resolution. Data were processed with nSolver software (NanoString Technologies, Seattle, WA), which included assessment of the quality of the runs, and combined, normalized, and analyzed in nSolver and Excel.
Normalization was performed by applying a scaling factor that normalizes the geometric mean of housekeeping genes (β-Actin; ACTB, hypoxanthine phosphoribosyltransferase 1; HPRT1 and RNA polymerase II subunit A ; POLR2A) for each sample and these normalized counts were used to calculate the scores, as previously reported (Pescarmona et al., 2019). The median of these ISG relative expression was used as an IFN score.
IFNα protein measurement by Simoa technology
All reagents were purchased from Quanterix (reference 100860) and loaded onto the Simoa HD-1 Analyzer (Quanterix) according to the manufacturer’s instructions and using three-step assay configurations. Briefly, the beads were pelleted with a magnet to remove supernatant (SN). Following several washes, 100 uL of detector antibody were added, according to the manufacturer’s instructions. The beads were then pelleted with a magnet, followed by washes and 100μL of ß–D–galactosidase (SβG) were added. The beads were washed, re-suspended in resorufin ß- D-galactopy-ranoside (RGP) solution, and loaded onto the array. The array was then sealed with oil and imaged. Images of the arrays were analyzed and AEB (average enzyme per bead) values were calculated by the software in the HD-1 Analyzer, as previously reported (Wilson et al., 2016). Human plasma samples along with calibration curves were measured using the Simoa HD-1 Analyzer. The calibration curves were fit using a 4PLfit with 1/y2 weighting factor and were used to determine the concentrations of the unknown human plasma samples. This analysis was done automatically using the software provided by Quanterix with the Simoa HD-1 Analyzer.
IL6, IFNλ1 and IFNγ protein measurements
Concentrations were determined in patient’s serum using U-PLEX Custom Human Cytokine assay (Meso Scale Diagnostics, Rockville, MD). The assays were performed according to the manufacturer’s instructions with overnight incubation of the diluted samples and standards at 4°C. The electrochemiluminescence signals (ECL) were detected by MESO QuickPlex SQ 120 plate reader (MSD) and analyzed with Discovery Workbench Software (v4.0, MSD).
Ex-vivo stimulation of PBMCs isolated from SARS-CoV-2-infected patients and healthy donors
Bloods of SARS-CoV-2-infected patients and healthy donors were collected in EDTA tubes. PBMCs were freshly isolated by Ficoll-Hypaque density centrifugation followed by washing in pDC/PMBC culture medium (i.e., RPMI 1640 Medium supplemented with 10% FBS, 100 U/ml penicillin, 100 mg/ml streptomycin, 2 mM L-glutamine, non-essential amino acids, 1 mM sodium pyruvate and 10 mM Hepes). PMBCs were frozen in 1mL freezing medium (10% DMSO, 90% FBS) and cryopreserved in vapor phase liquid nitrogen (>-135°C). Two hours prior to ex vivo stimulation, PBMCs were thawed out at 37°C rinsed with 10 mL of FCS, incubated in 40 mL of pDC/PMBC culture medium at 37C°/5% CO2 for 30 minutes and resuspended in culture medium. 2.5x10e5 PBMCs were cocultured with 1x10e5 SARS-CoV- 2-infected versus uninfected A549-ACE2 cells, as negative control, or were stimulated with TLR agonists [31.8 µM R848 and 42.22 µM polyI:C] for the FACS panel of mDC1/pDC analysis and LPS stimulation [2.04 µM] for the mDC2/non-mDC2/HLA-DR+ CD14+ panel in a final volume of 200µl in 96-well round-bottom plates incubated at 37C°/5% CO2 for 14 to 16 hours. Cell-culture SNs were collected for quantification of cytokine levels (IFNα by ELISA/Simoa; IL6, IFNγ and IFNλ 1 by U-PLEX Custom Human Cytokine assay) while cells were harvested for flow cytometry or for Nanostring analyses.
RNAs were isolated from these cells by phenol/chloroform extraction procedure as previously described (Assil et al., 2019). The subsequent steps of the procedure used for Nanostring analysis on ex-vivo stimulated PBMCs were performed as described above for the patient blood samples.
Coculture experiments using isolated pDCs
Unless indicated differently, 2x10e4 pDCs were cocultured with SARS-CoV-2-infected or uninfected cells as 5x10e4 or 1x10e5 cells for analysis by RT-qPCR or flow cytometry, respectively or were stimulated with 100µl of cell-free SN collected from SARS-CoV-2- infected cells. The cells were infected at MOI 0.01, 0.1, 0.02 and 0.5 for, respectively, NCI- H358-ACE2, Huh7.5.1, A549-ACE2 and Calu-3 cells for 48 hours maximum prior to collection of the cells and their SNs for coculture. As comparison pDCs were stimulated with TLR agonists [31.8 µM R848 and 42.22 µM polyI:C] in a final volume of 200µl in 96-well round-bottom plates incubated at 37C°/5% CO2. When indicated, cells were cocultured in 96- well format transwell chambers (Corning) with a 0.4 µm permeable membrane. At the indicated time, cell-culture supernatants were collected for quantification of cytokine levels:
IFNα, IFNλ 1/2/3 (IL29/28A/28B), TNFα and IL6 using specific ELISA kit (PBL Interferon Source, Affymetrix, respectively) following the manufacturer’s instructions. Cells were harvested at the indicated times for analysis by flow cytometry or RT-qPCR.
Immunostaining and flow cytometry analysis
At the indicated times, harvested cells were resuspended using 2 mM EDTA-PBS solution for the coculture with PBMCs and 0.48 mM EDTA-PBS solution for pDC cocultures. Cells were incubated with 1 μL/mL viability marker diluted in PBS for 20 minutes at RT. After a 10- minute incubation with Fc receptor blocking reagent (MACS Miltenyi Biotec) at 4°C followed by two PBS washes, cells were stained for surface markers for 30 minutes at 4°C with antibodies diluted in staining buffer (PBS without calcium and magnesium, with 2% FBS and 2mM EDTA), followed by two PBS washes. These markers included generic lineage markers (CD3, CD19, CD20, CD56 for exclusion, and CD11c, HLA-DR for selection of cell populations), and specific markers of pDCs (CD123, BDCA-2, CD2 and Axl), mDC2 (BDCA-1), mDC1 (BDCA-3), monocytes (CD14 and CD16) and/or cell differentiation markers (CD83, CD80 and PD-L1). The references and used concentrations for the antibodies are listed in the Key resource table. For the identification of apoptotic and necrotic cells, surface-stained cells were labelled using FITC Annexin V Apoptosis Detection Kit with 7- AAD according to the manufacturer’s instructions. Following one wash with Annexin V Binding Buffer (Biolegend), cells were fixed with 4% PFA for 30 minutes at 4°C. For intracellular-immunostaining, cells were treated with 1 µl/ml GolgiPlug solution (BD Bioscience) for 3 hours at 37°C/5% CO2 before collection. After surface staining and fixation with cytoperm/cytofix solution (BD Bioscience) for 20 minutes at 4°C, IFNα, IL6, IFNλ 1, and TNFα were stained by a 45-minute incubation at 4°C with antibodies diluted in permeabilization buffer (BD Bioscience ; antibodies are listed in the Key resource table). Cells were then washed with permeabilization buffer and resuspended in staining buffer. Flow cytometric analysis was performed using a BD LSR Fortessa 4L. Compensation beads were used as reference for the analysis. The data were analyzed using Flow Jo software (Tree Star).
Analysis of transcriptional levels by RT-qPCR
RNAs were isolated from cells harvested in guanidinium thiocyanate citrate buffer (GTC) by phenol/chloroform extraction procedure as described previously (Assil et al., 2019). The mRNA levels of human MXA, ISG15, IFNL, IL6, TNFA and glyceraldehyde-3-phosphate dehydrogenase (GADPH) were determined by RT-qPCR using iScript RT kit (Life Technologies) and PCR Master Mix kit (Life Technologies) for qPCR and analyzed using StepOnePlus Real-Time PCR system (Life Technologies). The sequences of the primers used for RT-qPCR are described in Table S1. The mRNA levels were normalized to GADPH mRNA levels.
Analysis of extracellular infectivity
Infectivity titers in supernatants were determined by end-point dilution in plaque assay. Briefly, 10-fold serial dilutions of SARS-CoV-2-containing supernatants were added to 2x10e5 Vero cells seeded in 12-well plates for a 2 hour-incubation. The medium was then replaced by DMEM containing 2% FBS and 2% carboxymethylcellulose (CMC). The cytopathic effect was scored 96 hours post-infection : cells were fixed for 30 minutes with 4% PFA and colored by cristal violet solution.
For icSARS-CoV-2-mNG infection, foci were directly detected according to mNeongreen- positive cells. Briefly, 10-fold serial dilutions of icSARS-CoV-2-mNG-containing supernatants were added to 2x10e4 Vero cells seeded in 96-well plates and fixed 24 hours post-infection. GFP-expressing cells were quantified by foci counting using a Zeiss Axiovert 135 microscope.
Viral Spread Assay
A549-ACE2 cells were transduced with lentiviral-based vector pseudotyped with VSV glycoprotein to stably express RFP, as previously reported (Assil et al., 2019). After immuno-isolation, pDCs were stained with CellTrace Violet Cell Proliferation kit (Life Technologies) for 20 minutes at 37°C in the dark. Labeled pDCs were then spinned down and resuspended in pDC culture medium. 2.5x10e4 pDCs were cocultured with 2.5x10e4 icSARS-CoV-2-mNG-infected cells (infected for 24 hours prior to coculture) and with 2.5x10e4 RFP+ uninfected cells for 48 hours at 37°C/5% CO2. When indicated, the cocultures were treated with an anti-αL integrin blocking antibody at 10mg/mL. After coculture, harvested cells were stained with Live/Dead Fixable Dead Cell Stain Near-IR marker for 30 minutes at RT, washed with PBS and fixed with 4% PFA for 30 minutes at 4°C. The level of viral spread from icSARS-CoV-2-mNG-infected cells (mNG+) to uninfected cells (RFP+) during coculture was determined by flow cytometric analysis as the frequency of infected cells (mNG+/RFP+ population) among the RFP+ cell population and similarly in RFP- populations. Flow cytometric analysis was performed using a BD LSR Fortessa 4L and the data were analyzed using Flow Jo software (Tree Star).
Live imaging of coculture with spinning-disc confocal microscopy analysis
A549-ACE2 cells were infected with icSARS-CoV-2-mNG for 48 hours prior to coculture with pDCs. Infected cells were seeded (2x10e4cells per well) in a 96-Well Optical-Bottom Plate pre-coated with poly-L-lysine (1 hour incubation at 37°C/5% CO2 with 8mg/mL poly- L-lysine). Isolated pDCs were stained with 0.5µM Vybrant cell-labeling solution (CM-DiI, Life Technologies) by successive incubations for 10 and 15 minutes at 37°C and 4°C respectively. After addition of pDCs to seeded infected cells, the cocultures were imaged every 30 minutes during 24 hours at magnification x10 with a BSL3-based spinning-disc confocal microscope (AxioObserver Z1, Zeiss). The cocultures were maintained at 37°C/5% CO2 in an incubation chamber. Analyses of pDC motion were performed using projection of Z-stacks with maximal intensity (i.e., about 5-to-10 selected Z-stacks per fields out of 30 Z- stacks in total). The quantification of mNeonGreen fluorescence intensity of infected cells and the duration of contacts between pDCs and infected cells were performed using Image J software package (http://rsb.info.nih.gov/ij). The calculations of pDC positions were performed using Trackmate plug-in of Image J software.
QUANTIFICATION AND STATISTICAL ANALYSIS PCA Analysis
PCA analyses were performed using the R princomp function (R version 4.1.0) with the correlation matrix approach (Jolliffe and Cadima, 2016; Mardia et al., 1979; Sánchez, 1982; Venables and Ripley, 2002). All time points available for each patient with a complete dataset for any given cell type were used. The time points of the patients presenting a mild severity were discriminated between Mild early (≤ 15 days after estimated primo-infection) and Mild late (> 15 days). For each cell type, PCA analyses were performed independently for each stimulation condition with the parameters presented in the vectors plots.
Bioinformatic analysis of the cis-acting regulatory element in the promoter
For each candidate gene, we recovered the genomic nucleotide sequence spanning from 1500 nucleotides upstream, until 100∼200 nucleotides downstream of the annotated Refseq transcription start site. For genes with more than one annotated transcription start site, we collected the sequence from 1500 nucleotides upstream of the most 5’ transcription start site until 100∼200 nucleotides downstream of the most 3’ transcription start site. The bioinformatic analyses were performed using the FIMO and AME tools available on https://meme-suite.org/meme/ (Grant et al., 2011; McLeay and Bailey, 2010) and the consensus sequence as follows:
NF-KB GGGRNYYYCC
IRF7 MCGAAARYGAAAVT
IRF3 NSRRAAMGGAAACCGAAACYR
IRF1 NTTYASTTTCACTTTCDBTTT
IRF5 cCGAAACCGAAmCy
STAT1:STAT2 tyAGTTTCrkTTYCy
IRF9 AwCGAAACCGAAACy
Statistical methods
Statistical analysis was performed using R software environment for statistical computing and graphics, version 3.3.2.
For quantifications by ELISA, RT-qPCR and flux cytometry analyses of the levels of cytokines, ISG and cell surface markers, the statistical analyses were performed using one- way ANOVA on ranks (Kruskal−Wallis rank sum test). When the test was considered significant (p-values ≤ 0.05), we used the Tukey Kramer (Nemenyi) pairwise test as post hoc test for multiple comparisons of mean rank sums to determine which contrasts between individual experimental condition pairs were significant.
Of note, for each independent experiment preformed using PBMCs isolated from SARS- CoV-2 infected patients, the same procedures and analyses were done in parallel using different healthy donors, as references (Figure 2, S2, 3). Likewise, all independent experiments of cocultures with PBMCs or pDCs were performed using distinct healthy donors as reference (Figure 1, S1, 4, S4, 5, S5, 6). To test difference between the patients groups in the Flow cytometry analysis of the biomarker the different cell population (Figure 3), we used the Beta regression model with the logit link function from the R ’betareg" package, which is the suitable statistical approach for modeling continuous response Y variables that vary in the open standard unit interval (0, 1). Beta regression being unable to model in case Y contains exactly 0 or 1 values, Y data was transformed to Y’ as following : where n is the number of patients in all compared groups.
For the quantification by flow cytometry analysis of the viral spread of the icSARS-CoV-2- mNG SARS-CoV-2 molecular clones from RFP- cells to RFP+ cells (initially uninfected)
(Figure 5E-F) and for quantification of data from the live-imaging analysis using spinning- disk confocal microscopy analysis (Figure 6), the statistical analyses were performed using one-way ANOVA followed by Tukey multiple comparisons of means.
The set of Figures was prepared using PRISM software.
Acknowledgments
We thank Dr B. Lina for kindly provided the clinical isolate of SARS-CoV-2 (CIRI, Lyon, France); Dr Pei-Yong Shi for the infectious-clone-derived mNeonGreen SARS-CoV-2 (University of Texas Medical Branch, Galveston, US); Dr V. Lotteau (CIRI, Lyon, France) for viral stock of the influenza A virus; Dr C. Goujon for the ACE2-Lentiviral construct (Institut de Recherche en Infectiologie de Montpellier, IRIM, France) and Dr F.V. Chisari (Scripps Research Institute, La Jolla, CA) for the Huh7.5.1 cells. We are grateful to Drs Y. Jaillais, A Marçais, B. Webster, S Assil, P.Y Lozach and A. Bosseboeuf for critical reading of the manuscript and to our colleagues for their encouragement and help. We thank the technical assistance of the staffs of PLATIM and AniRA-cytometry facilities – SFR Biosciences Lyon, Gerland (UMS3444/US8) for imaging and FACS analyses. We acknowledge the contribution of the EFS Confluence-Lyon. This work was supported by grants from the Agence Nationale de la Recherche (ANRJCJC-iSYN); the Fondation pour le recherche médicale (FRM ; ANR Flash COVID-19) ; the Agence Nationale pour la Recherche contre le SIDA et les Hépatites Virales (ANRS – N21006CR and N19017CR) ; the UDL/ANR IA ELAN ERC (G19005CC); and fondation FINOVI (AO11 - Collaboration); from EU H2020 ZIKAlliance. PhD fellowships for S.R. and G.J. are respectively sponsored by ANRS and Université Lyon 1 ‘Contrats doctoraux Lyon 1 dédiés à l’International’.
Footnotes
↵* Egal contributions as first co-authors.