24
1 Type I and III interferons disrupt lung epithelial repair during recovery from viral infection J. Major 1 , S. Crotta 1 , M. Llorian 2 , T. M. McCabe 1 †, H. H. Gad 3 , R. Hartmann 3 , A. Wack 1 * 5 Affiliations: 1 Immunoregulation Laboratory, The Francis Crick Institute, London, UK. 2 Bioinformatics and Biostatistics, The Francis Crick Institute, London, UK. 3 Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark. 10 †Present address: Adaptive Immunity Research Unit, Glaxosmithkline, Stevenage, UK. *Correspondence to: Andreas Wack, Immunoregulation Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK; [email protected]. 15 20 25 30 35 . CC-BY 4.0 International license was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprint (which this version posted May 5, 2020. . https://doi.org/10.1101/2020.05.05.078360 doi: bioRxiv preprint

Type I and III interferons disrupt lung epithelial …...2020/05/05  · 4 Temporal overlap of antiviral IFN responses and lung epithelial repair following influenza virus infection

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Type I and III interferons disrupt lung epithelial …...2020/05/05  · 4 Temporal overlap of antiviral IFN responses and lung epithelial repair following influenza virus infection

1

Type I and III interferons disrupt lung epithelial repair during recovery from viral infection

J. Major1, S. Crotta1, M. Llorian2, T. M. McCabe1†, H. H. Gad3, R. Hartmann3, A. Wack1*

5

Affiliations: 1Immunoregulation Laboratory, The Francis Crick Institute, London, UK. 2Bioinformatics and Biostatistics, The Francis Crick Institute, London, UK. 3Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark. 10

†Present address: Adaptive Immunity Research Unit, Glaxosmithkline, Stevenage, UK. *Correspondence to: Andreas Wack, Immunoregulation Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK; [email protected].

15

20

25

30

35

.CC-BY 4.0 International licensewas not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprint (whichthis version posted May 5, 2020. . https://doi.org/10.1101/2020.05.05.078360doi: bioRxiv preprint

Page 2: Type I and III interferons disrupt lung epithelial …...2020/05/05  · 4 Temporal overlap of antiviral IFN responses and lung epithelial repair following influenza virus infection

2

Abstract:

Excessive cytokine signalling frequently exacerbates lung tissue damage during respiratory viral infection. Type I and III interferons (IFN-α/β and IFN-λ) are host-produced antiviral cytokines and currently considered as COVID-19 therapy. Prolonged IFN-α/β responses can lead 5

to harmful proinflammatory effects, whereas IFN-λ mainly signals in epithelia, inducing localised antiviral immunity. Here we show that IFN signalling interferes with lung repair during influenza recovery, with IFN-λ driving these effects most potently. IFN-induced p53 directly reduces epithelial proliferation and differentiation, increasing disease severity and susceptibility to bacterial superinfections. Hence, excessive or prolonged IFN-production aggravates viral infection 10

by impairing lung epithelial regeneration. Therefore, timing and duration are critical parameters of endogenous IFN action, and should be considered carefully for IFN therapeutic strategies against viral infections like influenza and COVID-19. 15

One Sentence Summary: A novel IFN-mediated mechanism of immunopathology during respiratory virus infection

by interference with lung tissue repair. 20

25

30

35

.CC-BY 4.0 International licensewas not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprint (whichthis version posted May 5, 2020. . https://doi.org/10.1101/2020.05.05.078360doi: bioRxiv preprint

Page 3: Type I and III interferons disrupt lung epithelial …...2020/05/05  · 4 Temporal overlap of antiviral IFN responses and lung epithelial repair following influenza virus infection

3

Main Text:

During infection with respiratory viruses, disease severity is linked to the destruction of epithelia in the lung. This occurs as a result of both cytopathic viral effects in host cells, and damage generated by the ensuing inflammatory immune response. Loss of airway and lung 5

epithelia can result in clinical complications, including lung failure, acute respiratory distress syndrome (ARDS), pneumonia, and increased susceptibility to bacterial superinfections by pathogens like Streptococcus pneumoniae (S. pneumoniae) owing to a loss of epithelial barrier function.

Restoration of damaged epithelial tissues is therefore paramount in order to maintain lung 10

function and barrier protection. The lung epithelium is largely quiescent during steady state, yet has a remarkable capacity to replenish following damage. Several types of progenitor cells can contribute to the regeneration of the respiratory epithelium. Trp63+Krt5+ basal cells in the upper airways and Scgb1a1+ secretory cells in conducting airways repopulate epithelial cell populations following depletion in mice (1, 2). Surfactant producing type II alveolar cells (AT2) cells 15

proliferate to reform damaged alveoli, whilst also functioning as stem cells through the differentiation into type I alveolar cells (AT1) (3, 4). In situations of severe damage, including influenza virus-induced injury, alveolar repair is supported by the emergence and expansion of Krt5+ basal cells from the airways (5–8).

Interferons (IFNs) represent a crucial component of the innate immune defence against 20

viral infections. Type I and III IFNs (IFN-α/β and IFN-λ, respectively) are induced upon viral recognition, and function by triggering transcription of an overlapping set of interferon-stimulated genes (ISGs), conferring an antiviral state in infected and bystander cells (9, 10). Owing to widespread expression of the type I IFN receptor (IFNAR) in immune cells, IFN-α/β responses can result in immunopathology during viral infections, including influenza virus and severe acute 25

respiratory syndrome coronavirus (SARS-CoV) (11–14). In contrast, the receptor for IFN-λ (IFNLR) is restricted mainly to epithelia at barrier sites like the gut and lung, with expression in the immune system largely restricted to neutrophils in mice (15–17). IFN-λ responses are therefore often characterised by their ability to confer localised antiviral protection at the site of infection, without driving damaging proinflammatory responses associated with IFN-α/β. Both type I and III 30

IFNs are currently being tested in clinical trials as antiviral therapeutics against SARS-CoV-2 (18–20), despite some scepticism owing to IFN-α/β-associated pathogenic effects (21). In addition to their antiviral and proinflammatory activity, IFNs exert antiproliferative and proapoptotic functions, and have a long history in antitumour therapies (22). Despite a growing understanding of the causes of immunopathology during infection with respiratory viruses, little is known 35

concerning the direct effect that IFN responses have on lung epithelial repair.

.CC-BY 4.0 International licensewas not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprint (whichthis version posted May 5, 2020. . https://doi.org/10.1101/2020.05.05.078360doi: bioRxiv preprint

Page 4: Type I and III interferons disrupt lung epithelial …...2020/05/05  · 4 Temporal overlap of antiviral IFN responses and lung epithelial repair following influenza virus infection

4

Temporal overlap of antiviral IFN responses and lung epithelial repair following influenza virus infection

To induce acute lung damage, C57BL/6 (B6) wild type (WT) mice were infected with a sublethal dose of the H3N2 influenza virus strain X31. Influenza virus matrix gene expression 5

peaked on day 2 post infection following viral replication, before viral clearance was reached on day 12 post infection (fig. S1A). Infection resulted in increasing morbidity and weight loss (fig. S1B), accompanied by significant immune cell infiltration and lung damage as determined by histology (fig. S1C, left, and S1D). Recovery from infection (~day 8 post infection; fig. S1B) coincided with the onset of epithelial regeneration (fig. S1C, right). To further investigate the 10

dynamics of lung repair following influenza virus infection, epithelial cells were harvested for analysis of proliferation by flow cytometry, using the marker of recent cell proliferation Ki67 (gating strategy in fig. S2). During steady-state conditions, type II alveolar epithelial cells (AT2; EpCam+MHCII+CD49flow) (3, 4, 23) have a low rate of turnover, remaining largely quiescent (Fig. 1A). However, following influenza virus-induced lung damage, AT2 cells undergo rapid 15

proliferation starting at ~day 7 post infection, correlating with mouse recovery and weight gain of mice (Fig. 1A and fig. S1B).

To compare the dynamics of epithelial recovery with production of IFN in our infection model, we collected bronchoalveolar lavage fluid (BALF) at different timepoints throughout influenza virus infection (Fig. 1B). IFN subtypes (IFN-α, -β and -λ) were produced rapidly, 20

peaking on day 2 post infection (Fig. 1B). The magnitude of IFN-λ production was significantly greater than that of IFN-α/β, both in concentration and duration. Whilst IFN-α/β production declined following the day 2 peak, high levels of IFN-λ production were maintained. Importantly, only IFN-λ was detected on day 7/8 post infection, coinciding with the onset of epithelial recovery (Fig. 1A and B). Together, these data show that following influenza virus infection, IFN signalling 25

(in particular IFN-λ induced) is ongoing throughout the onset of epithelial proliferation and lung repair.

IFN signalling during recovery from influenza virus infection impairs lung epithelial cell proliferation 30

Next, to understand the effect of IFNs on lung epithelial repair, mice were treated with IFN during recovery from influenza virus infection (day 7 to 10 post infection) (fig. S3A). To compare the effects of type I and III IFN subtypes at equal biological potency, IFN-α, -β and -λ subtypes were titrated on murine airway epithelial cell (AEC) cultures and analysed for the induction of antiviral ISGs (fig. S4A). Dose-response curves were then generated to obtain an EC50 for each 35

subtype, in order to determine a conversion factor for equipotency between IFN-α, -β and -λ (fig.

.CC-BY 4.0 International licensewas not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprint (whichthis version posted May 5, 2020. . https://doi.org/10.1101/2020.05.05.078360doi: bioRxiv preprint

Page 5: Type I and III interferons disrupt lung epithelial …...2020/05/05  · 4 Temporal overlap of antiviral IFN responses and lung epithelial repair following influenza virus infection

5

S4B). In order to study the effects of IFN treatment specifically on epithelial cells, we generated irradiation bone marrow (BM) chimeras where WT recipient mice were given Ifnar1-/- bone marrow cells, thus restricting IFNAR expression to the stromal compartment.

In chimeric mice, both IFN-α and -β treatments significantly reduced the proliferation of AT2 cells on day 11 post influenza virus infection, compared to PBS treated control mice (Fig. 5

1C). This reduction was independent of changes in viral burden, as influenza virus matrix gene expression remained unchanged following IFN treatment (fig. S3B). We also observed a reduction in the proliferation of AT2 cells in WT mice following IFN-λ treatment (Fig. 1B), again independently from changes in viral load (fig. S3C). To exclude the possibility of IFN-λ signalling in neutrophils, we treated WT mice with a monoclonal antibody against Ly6G (clone 1A8) (fig. 10

S3D). Even after depletion of neutrophils, we observed a significant reduction in AT2 cell proliferation following IFN-λ treatment (fig. S3E). A caveat when using inbred mouse strains in the context of influenza virus infection is the lack of a functional Mx1 protein; a crucial IFN-inducible influenza virus restriction factor in both mice and humans (24). We therefore infected mice that express functional Mx1 alleles (B6-Mx1) with the highly virulent influenza virus strain 15

hvPR8-DNS1, for a more clinically relevant influenza model. IFN-λ treatment also significantly reduced lung epithelial proliferation in the presence of functional Mx1 (Fig. 1E).

We next sought to determine the role of endogenous IFN during lung repair, using Ifnar1-

/- and Ifnlr1-/- mice. AT2 cells were analysed on day 8 post influenza virus infection; the time of overlap between IFN signalling and epithelial cell proliferation (Fig. 1A and B). Both Ifnar1-/- and 20

Ifnlr1-/- mice had improved AT2 cell proliferation, compared to WT controls (Fig. 1F and G). This was dependent on IFN signalling specifically through the epithelium, as receptor deficiency in the stromal compartment alone was sufficient to increase lung epithelial cell proliferation (Fig. 1H). Improved proliferation was independent of major changes in viral burden, with only a slight increase in influenza virus matrix gene expression observed in Ifnar1-/- mice on day 2 post 25

infection, and no change in Ifnlr1-/- mice, compared to WT controls (fig. S5A). Viral control in individual IFN receptor knockout mice is likely unaffected due to redundancy between type I and III IFN antiviral responses in epithelial cells (25, 26).

Despite type I and III IFN redundancy in viral control (fig. S5A), the lack of redundancy in antiproliferative IFN responses, with both Ifnar1-/- and Ifnlr1-/- mice displaying enhanced 30

epithelial proliferation (Fig. 1F to H), led us to further interrogate the phenotype. As IFNAR signalling has previously been shown to be important for the production of IFN-λ during influenza virus infection (27, 28), we measured IFN production in the BALF of IFN receptor knockout mice. Consistent with previous findings (28), we observed a significant reduction in IFN-λ (and indeed IFN-α/β) production in Ifnar1-/- mice compared to WT, yet we saw little change in IFN-α/β levels 35

in Ifnlr1-/- mice (fig. S5B). These findings suggest that the improved epithelial proliferation

.CC-BY 4.0 International licensewas not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprint (whichthis version posted May 5, 2020. . https://doi.org/10.1101/2020.05.05.078360doi: bioRxiv preprint

Page 6: Type I and III interferons disrupt lung epithelial …...2020/05/05  · 4 Temporal overlap of antiviral IFN responses and lung epithelial repair following influenza virus infection

6

observed in Ifnar1-/- mice, could be attributed to a reduction in IFN-λ. Reduced IFN production in Ifnar1-/- mice is linked with steady state priming defects in these mice owing to the absence of tonic IFNAR activation in immune cells (29). To circumvent this, we used an αIFNAR monoclonal antibody during influenza virus infection (MAR1-5A3) administered only from the time of infection (day 0). αIFNAR treatment maintained steady state priming required for IFN-λ 5

production (fig. S5C), yet was effective in reducing production of proinflammatory cytokines induced by IFN-α/β (fig. S5D) (12). Importantly, αIFNAR treatment from day 0 or day 3 post infection, had no effect on lung epithelial cell proliferation (fig. S5D). We therefore conclude that in our model of influenza virus infection in B6 mice, IFN-λ responses are most effective in disrupting epithelial regeneration during influenza recovery, and these effects are mediated 10

directly in epithelial cells. Type I and III IFNs block AEC proliferation and differentiation

To understand mechanistically how IFNs exert the antiproliferative effects observed in vivo, we set up primary murine AEC cultures. Progenitor basal cells from distal airway sites are 15

involved in the regeneration of the lung epithelium following influenza virus-induced damage (6, 8, 30). AEC cultures undergo rapid proliferation, and also differentiation upon exposure to an air-liquid interface (ALI), recapitulating repair processes observed in the lung and airway epithelium following damage in vivo (31, 32). Similar to IFNs used in vivo, IFNs used for in vitro assays were titrated on AEC cultures, to compare IFN subtypes at equivalent biological potency (fig. S6A and 20

B). AEC cultures seeded at a low density in the presence of IFN-β or -λ were unable to reach confluence, whilst IFN-α treatment significantly reduced AEC number and the time taken for cultures to reach confluence (Fig. 2A left, and fig. S7A).

To study growth inhibitory effects of IFNs without blocking the establishment of AEC cultures, we seeded cells at a higher density before treating with IFN for 5 days. All 3 IFN subtypes 25

delayed the time taken for cultures to reach confluency, and impaired cell growth, with IFN-β and IFN-λ having the most significant effects (Fig. 2A right, and 2B). Growth inhibitory effects are dependent on the presence of the respective IFN receptor (fig. S7B). Similar effects were observed when primary human AEC cultures were treated with equivalent doses of IFN types (fig. S6C and D), with human IFN-β and -λ treatment resulting in the most significant decrease in cell number 30

(Fig. 2C). To gain mechanistic insight into IFN-mediated disruption of AEC growth, we analysed

IFN treated AEC cultures by flow cytometry to study effects on apoptosis and proliferation. Consistent with the observed reduction in cell number, IFN-β or -λ treatment resulted in a significant increase in the frequency of apoptotic or necrotic cells (Annexin V+/TO-PRO-3+) (fig. 35

S7C and D). We also observed a more than 50% reduction in the number of actively proliferating

.CC-BY 4.0 International licensewas not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprint (whichthis version posted May 5, 2020. . https://doi.org/10.1101/2020.05.05.078360doi: bioRxiv preprint

Page 7: Type I and III interferons disrupt lung epithelial …...2020/05/05  · 4 Temporal overlap of antiviral IFN responses and lung epithelial repair following influenza virus infection

7

cells, as measured by incorporation of the thymidine analogue EdU, while IFN-α treatment had only a modest effect (Fig. 2D and E). These findings were confirmed by measuring CFSE dilution (fig. S7E and F, left). Interestingly, IFN-mediated antiproliferative effects were only observed in highly proliferative AEC cultures, as treating established cultures had no effect (fig. S7E to G, right). These data suggest that IFNs actively disrupt cell cycle progression without killing 5

quiescent cells. Therefore, the increase in apoptosis observed (fig. S7C and D) may occur as a result of failed progression through the cell cycle following IFN treatment, as seen previously (33).

We next sought to examine the effects of IFNs on AEC differentiation. Following acute damage, populations of basal cells and Scgb1a1+ secretory cells give rise to secretory and multiciliated cell subtypes (34). Timely repopulation of airway epithelial cell subsets following 10

damage is crucial in maintaining barrier function. Indeed, influenza virus infection has previously been shown to decrease mucociliary clearance in mice, resulting in an increased susceptibility to superinfection with S. pneumoniae (35). To study specific effects of IFNs on AEC differentiation, we initiated IFN treatment later during the course of AEC growth throughout exposure to an ALI, the time in which AEC differentiation is induced (fig. S8A). Similar to our findings for 15

proliferation, IFN-β and -λ treatment significantly reduced the expression of genes pertaining to multiciliated (Mcidas/Ccno) and secretory cell (Muc5AC, Scb1a1) differentiation, with IFN-λ having the strongest effect (Fig. 2F). Expression of the basal cell marker Krt5 remained unchanged, or increased by IFN-λ treatment, suggesting maintenance of stemness (fig. S8B). We also observed a significant reduction in the number of ciliated cells in AEC cultures (acetylated α-tubulin+) 20

following IFN-λ treatment, but not with IFN-α or -β (Fig. 2G and fig. S8C). To confirm these findings in vivo, WT and Ifnlr1-/- mice were infected with influenza virus, and lung sections were analysed by immunofluorescence. Ifnlr1-/- mice had increased acetylated α-tubulin+ ciliated cells in repairing conducting airways on day 10 post infection (Fig. 2H). We quantified these findings by flow cytometry, and observed an increase in the frequency of differentiated AECs 25

(EpCamhighCD49fhighCD24+) composed of ciliated, goblet and club cells (Fig. 2I and fig. S2)(5). From these results, we conclude that IFN-λ signalling reduces the capacity for basal cell differentiation during recovery from influenza virus infection. IFNs disrupt AEC growth via induction of p53-dependent downstream pathways 30

To understand mechanistically how IFNs mediate antiproliferative effects, we performed RNA-sequencing (RNA-seq) on IFN-treated AEC cultures (Fig. 3A). Principal component analysis (PCA) showed that untreated AEC control samples clustered depending on length of culture growth (including ALI exposure) along the principal component 2 (PC2), indicating that this axis represents proliferation and differentiation (Fig. 3B). AECs treated with IFN subtypes for 35

4 hours of culture clustered away from their respective mock on PC1, suggesting this principal

.CC-BY 4.0 International licensewas not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprint (whichthis version posted May 5, 2020. . https://doi.org/10.1101/2020.05.05.078360doi: bioRxiv preprint

Page 8: Type I and III interferons disrupt lung epithelial …...2020/05/05  · 4 Temporal overlap of antiviral IFN responses and lung epithelial repair following influenza virus infection

8

component of variance (46%) is attributable to induction of antiviral ISGs (Fig. 3B). Additionally, 4-hour IFN treatment clustered samples together regardless of subtype (Fig. 3B), confirming the equal subtype dosage based on previous titrations (fig. S6A, B, and S9A).

AECs were also treated for 2, 3, or 5 days to understand the effects of prolonged IFN treatment in growing AEC cultures. 4-hour and 5-day IFN treated cultures shared the same 5

experiment endpoint, and therefore share the same untreated controls (Fig. 3A). 5 days of IFN-β or -λ treatment, resulted in the clustering of samples away from corresponding untreated controls on both PC1 and PC2, indicating maintained ISG induction (PC1) and a block of AEC proliferation and development (PC2) (Fig. 3B). Conversely, the response induced by IFN-α was significantly reduced after just 2 days of treatment, consistent with previous findings (36). Ingenuity pathway 10

analysis (IPA) of differentially expressed genes following prolonged IFN treatment (2, 3 and 5 days) revealed the top cluster pertaining to biological pathways regulating cell cycle and cell death, most significantly induced by IFN-λ treatment across all timepoints (Fig. 3C). Predicted upstream transcriptional regulators identified typical regulators of IFN function, including interferon regulatory factors (IRFs) and signal transducer and activator of transcription (STAT) proteins, in 15

addition to regulators of cell cycle (Fig. 3D). We identified the tumour suppressor protein p53 as a top candidate regulating IFN-inducible antiproliferative effects, owing to its significant predicted activity following prolonged IFN-β and -λ treatment in AECs (Fig. 3D). Additionally, gene set enrichment analysis (GSEA) revealed IFN-mediated induction of the p53 downstream pathway and p53-regulated downstream targets (fig. S9B and C). 20

To confirm the role of p53, we utilised p53-/- AEC cultures. IFN treatment had no effect on p53-/- AEC number or proliferation following IFN treatment (Fig. 3E and F). IFN-mediated induction of antiproliferative downstream p53 target genes Gadd45g and Dusp5 (37, 38) was p53 dependent, as IFNs had no such effect in p53-/- AEC cultures (Fig. 3G). Additionally, IFN-λ had a much-reduced negative effect on multiciliogenesis in differentiating p53-/- cultures, compared to 25

its effect on WT and p53+/- cultures (fig. S9D). We next sought to examine whether IFNs regulate p53 activity in epithelial cells during lung repair in vivo. To study IFN effects specifically in the lung epithelium, we once again generated Ifnar1-/-> WT bone marrow chimeric mice for IFN-α or –β treatment, or depleted neutrophils in WT mice with αLy6G for IFN-λ treatment (fig. S3A). In agreement with in vitro observations, IFN-β and -λ, but not IFN-α, significantly upregulated p53 30

expression in repairing lung epithelial cells (Fig. 3H and I). Taken together, these results demonstrate that IFN-β and -λ mediate antiproliferative effects in AECs via induction of p53. Additionally, our RNA-seq analyses confirm the potent antiproliferative activity of IFN-λ, compared to type I IFN subtypes. 35

.CC-BY 4.0 International licensewas not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprint (whichthis version posted May 5, 2020. . https://doi.org/10.1101/2020.05.05.078360doi: bioRxiv preprint

Page 9: Type I and III interferons disrupt lung epithelial …...2020/05/05  · 4 Temporal overlap of antiviral IFN responses and lung epithelial repair following influenza virus infection

9

Ifnlr1-/- mice have improved epithelial repair and barrier function following influenza virus infection

Our data supports a significant role for IFN signalling, in particular IFN-λ, in the reduction of epithelial proliferation and differentiation during lung repair. We therefore determined whether this alters the state of lung epithelia or affects barrier function of the respiratory epithelium. RNA-5

seq analysis of sorted influenza virus infected lung epithelial cells (EpCam+CD31-CD45-) from WT or Ifnlr1-/- mice confirmed an upregulation of pathways pertaining to proliferation and multiciliogenesis in Ifnlr1-/- mice (Fig. 4A). Improved repair correlated with reduced lung damage, with a reduction in both the total, and red blood cell number in the BALF of Ifnlr1-/- mice day 8 post infection (Fig. 4B and C). Additionally, Ifnlr1-/- mice had less inflammatory immune cell 10

infiltrate dispersed in lung tissues (Fig. 4D). Influenza virus infections in humans are often exacerbated by opportunistic bacterial pathogen, including S. pneumoniae (39). A contributing factor driving this synergy, is a compromised epithelial barrier as a result of influenza virus-induced damage. To measure the effects of IFN-λ signalling on lung barrier function, we challenged mice with S. pneumoniae 8 days post influenza virus infection. We used BM chimeric 15

mice to study specific effects of IFN-λ in epithelial cells. Mice lacking IFNLR in the stromal compartment (WT > Ifnlr1-/-) had improved survival following bacterial superinfection (Fig. 4E). Overall, our data indicate that IFN-λ signalling reduces the capacity for epithelial repair following influenza virus infection, which results in prolonged lung damage and compromised barrier function, consequently increasing susceptibility to bacterial superinfection. 20

Discussion

Here we describe a novel mechanism by which type I and III IFN signalling aggravates lung pathology during respiratory viral infection. By focusing on the importance of epithelial repair during the recovery phase of infection, we highlight the potential for both IFN-α/β and IFN-25

λ in exacerbating influenza disease. Our data indicates that IFN-λ is most effective in disrupting lung epithelial repair. Specific

blockade of type I IFN responses with αIFNAR-treatment had no effect on lung epithelial proliferation. Comparison of IFN subtypes at equal biological potency in AECs revealed IFN-β and -λ to most significantly disrupt growth and induce antiproliferative pathways in primary 30

human and murine epithelial cell cultures. Factors that determine the antiproliferative differential of IFN subtypes include: ligand-receptor binding affinity, length of stimulation, receptor density, and negative feedback control (40–45). Additionally, we found the magnitude of IFN-λ production in BALF was greater compared to IFN-α/β, extending into the crucial time window in which epithelial repair manifests post influenza virus infection. We therefore conclude that the reduced 35

antiproliferative potential of IFN-α, combined with low IFN-α/β levels in vivo, limits the capacity

.CC-BY 4.0 International licensewas not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprint (whichthis version posted May 5, 2020. . https://doi.org/10.1101/2020.05.05.078360doi: bioRxiv preprint

Page 10: Type I and III interferons disrupt lung epithelial …...2020/05/05  · 4 Temporal overlap of antiviral IFN responses and lung epithelial repair following influenza virus infection

10

for type I IFNs to block lung repair directly in B6 mice. However, we do not exclude a potential for host IFN-α/β production in directly blocking lung epithelial repair. This may be the case in situations of excessive type I IFN production and lung pathology, as seen in other mouse strains following influenza virus or SARS-CoV infection (12, 13).

Mechanistically, we identified the tumour suppressor protein p53 as the downstream 5

regulator of IFN-induced antiproliferative effects. Research defining the mechanisms by which IFNs induce growth inhibitory effects have focused on type I IFNs, using transformed cell lines to explore antitumour therapeutics, with little understanding for their roles in normal physiology or during infection (22). In primary AECs, we observed no difference between WT and p53-/- cultures in baseline growth or expression of downstream effector genes (including Gadd45g and Dusp5), 10

indicating that p53 is inactive in these cells under normal conditions. Once activated, p53 triggers cell cycle arrest and apoptosis through induction of its target genes (46), and has previously been shown to directly regulate IFN-αβ antitumour and antiviral responses (47).

We also show that IFN-λ disrupts AEC differentiation during lung repair. The differentiation of AEC cultures triggered by air exposure is accompanied by reduced cell 15

proliferation. As we treated cultures specifically during this period, the block in differentiation appears to be independent of antiproliferative IFN effects. Multiciliogenesis in AECs requires tightly coordinated use of the mitotic machinery for centriole amplification, required for the generation of cilia (48). We therefore speculate that IFN-induced p53 activity disrupts this coordination, thereby impairing AEC differentiation. Respiratory virus infections are associated 20

with chronic lung disease such as asthma and chronic obstructive pulmonary disease (49), and proinflammatory cytokines have been shown to induce epithelial remodelling in human AECs (50). Therefore, our data suggest a mechanistic link between antiviral immune responses and long-term changes in lung physiology.

Pathogenic respiratory viruses, including IAV and CoV, are a significant global health 25

concern owing to their potential to cause pandemics. The ongoing outbreak of CoV disease 2019 (COVID-19) presents the most severe global health crisis since the Spanish flu pandemic of 1918 (51–53). Similar to influenza, severe cases of COVID-19 are associated with massive alveolar damage and progressive respiratory failure (54), and SARS-CoV-2 has been shown to induce expression type I and III IFNs in primary human AEC cultures (55). In addition, histopathological 30

findings in post-mortem lung biopsies of COVID-19 victims reveal neutrophilic infiltration consistent with superimposed bacterial bronchopneumonia (56) . Causes of variation in host disease severity during respiratory virus infection are not well understood. Studies comparing influenza patient cohorts found little or no correlation between disease severity and viral load (57–60), suggesting a role for genetically determined host factors, including mechanisms amplifying 35

immunopathology.

.CC-BY 4.0 International licensewas not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprint (whichthis version posted May 5, 2020. . https://doi.org/10.1101/2020.05.05.078360doi: bioRxiv preprint

Page 11: Type I and III interferons disrupt lung epithelial …...2020/05/05  · 4 Temporal overlap of antiviral IFN responses and lung epithelial repair following influenza virus infection

11

Attempts to gain a better understanding of human influenza pathogenesis have largely been restricted to whole blood expression analysis (61–63), and therefore exclude localised immune and epithelial repair responses in lung tissues (64). However, macaques infected with the highly virulent 1918 H1N1 strain, revealed an elevated ISG signature late during infection bronchial tissue (65). Additionally, SARS-CoV patients displayed robust IFN responses, and aberrant ISG 5

expression in severe cases (66). Whether susceptible individuals display excessive IFN responses late during infection, and the potential impact this has on lung repair, remains to be determined. Our transcriptomic analyses of regenerating epithelial cells in vivo link host IFN-λ responses with a reduction in the capacity for lung repair. Analysis of lung tissue and BALF from respiratory virus infected patients experiencing severe disease will provide insight into the mechanisms regulating 10

disease pathogenesis, including potential interplay between IFNs and epithelial cells. Lack of an available vaccine during the emergence of novel zoonotic viruses, like SARS-

CoV-2, highlights the importance of generating and implementing effective host-directed antiviral and immunomodulatory treatments. Our data highlights the importance of a well-timed IFN response, with regards to both host immunity and exogenous treatment. IFN-α/β treatment 15

exacerbates inflammation and disease severity in mice if administered past the peak of viral replication during influenza virus or Middle East respiratory-syndrome-CoV (MERS-CoV) infection (67, 68). IFN-λ treatment early during influenza virus infection was shown to be protective, owing to its ability to confer antiviral protection without generating the proinflammatory responses associated with IFN-α/β (68, 69). For these reasons, IFN-λ is therefore 20

being implemented as a candidate therapy for COVID-19 (70). However, our findings here identify a mechanism by which IFN-λ exacerbates respiratory virus disease acting specifically through the respiratory epithelium, independent of immunomodulation.

Taken together, these data indicate the necessity for effective regulation of host IFN responses, and the importance of timing and duration when considering IFNs as therapeutic 25

strategies to treat respiratory virus infections. Optimal protection would be achieved by strong induction of ISGs early during infection to curb viral replication, followed by timely downregulation of IFN responses, enabling efficient lung epithelial repair. These data change our understanding of immunopathology during viral infection, and have important implications for IFN usage as antiviral therapy. 30

.CC-BY 4.0 International licensewas not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprint (whichthis version posted May 5, 2020. . https://doi.org/10.1101/2020.05.05.078360doi: bioRxiv preprint

Page 12: Type I and III interferons disrupt lung epithelial …...2020/05/05  · 4 Temporal overlap of antiviral IFN responses and lung epithelial repair following influenza virus infection

12

References and Notes:

1. J. R. Rock et al., Basal cells as stem cells of the mouse trachea and human airway epithelium. Proc. Natl. Acad. Sci. U. S. A. 106, 12771–5 (2009).

2. P. R. Tata et al., Dedifferentiation of committed epithelial cells into stem cells in vivo. Nature 503, 218–23 (2013). 5

3. A. Nabhan et al., Single-cell Wnt signaling niches maintain stemness of alveolar type 2 cells. Science 1123, 1–12 (2018).

4. W. J. Zacharias et al., Regeneration of the lung alveolus by an evolutionarily conserved epithelial progenitor. Nature 555, 251–255 (2018).

5. J. Quantius et al., Influenza virus infects epithelial stem/progenitor cells of the distal lung: 10 impact on Fgfr2b-driven epithelial repair. PLoS Pathog. 12 (2016), doi:10.1371/journal.ppat.1005544.

6. P. A. Kumar et al., Distal airway stem cells yield alveoli in vitro and during lung regeneration following H1N1 influenza infection. Cell 147, 525–538 (2011).

7. A. E. Vaughan et al., Lineage-negative progenitors mobilize to regenerate lung epithelium 15 after major injury. Nature 517, 621–5 (2015).

8. W. Zuo et al., P63(+)Krt5(+) distal airway stem cells are essential for lung regeneration. Nature 517, 616–620 (2014).

9. F. McNab et al., Type I interferons in infectious disease. Nat Rev Immunol. 15, 87–103 (2015). 20

10. A. Wack, E. Terczyńska-Dyla, R. Hartmann, Guarding the frontiers: the biology of type III interferons. Nat. Immunol. 16, 802–809 (2015).

11. J. R. Teijaro et al., Persistent LCMV infection is controlled by blockade of type I interferon signaling. Science 340, 207–211 (2013).

12. S. Davidson et al., Pathogenic potential of interferon αβ in acute influenza infection. Nat. 25 Commun. 5 (2014), doi:10.1038/ncomms4864.

13. R. Channappanavar et al., Dysregulated type I interferon and inflammatory monocyte-macrophage responses cause lethal pneumonia in SARS-CoV-infected mice. Cell Host Microbe 19, 181–193 (2016).

14. E. B. Wilson et al., Blockade of chronic type I interferon signaling to control persistent 30 LCMV infection. Science 340, 202–207 (2013).

15. K. Blazek et al., IFN-λ resolves inflammation via suppression of neutrophil infiltration and IL-1β production. J. Exp. Med. 212, 845–853 (2015).

16. A. Broggi et al., IFN-λ suppresses intestinal inflammation by non-translational regulation of neutrophil function. Nat. Immunol. 18, 1084–1093 (2017). 35

17. V. Espinosa et al., Type III interferon is a critical regulator of innate antifungal immunity. 5357 (2017), doi:10.1126/sciimmunol.aan5357.

18. J. Phua et al., Intensive care management of coronavirus disease 2019 (COVID-19):

.CC-BY 4.0 International licensewas not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprint (whichthis version posted May 5, 2020. . https://doi.org/10.1101/2020.05.05.078360doi: bioRxiv preprint

Page 13: Type I and III interferons disrupt lung epithelial …...2020/05/05  · 4 Temporal overlap of antiviral IFN responses and lung epithelial repair following influenza virus infection

13

challenges and recommendations. Lancet. Respir. Med. 2019, 1–12 (2020). 19. Lopinavir/ritonavir, ribavirin and IFN-beta combination for nCoV treatment.

https://clinicaltrials.gov/ct2/show/NCT04276688 (2020). 20. Pegylated Interferon Lambda Treatment for COVID-19.

https://clinicaltrials.gov/ct2/show/NCT04343976 (2020). 5

21. P. J. Richardson, M. Corbellino, J. Stebbing, Baricitinib for COVID-19: a suitable treatment? Lancet Infect. Dis. 3099, 30270 (2020).

22. B. S. Parker, J. Rautela, P. J. Hertzog, Antitumour actions of interferons: implications for cancer therapy. Nat. Rev. Cancer. 16, 131–44 (2016).

23. K. Hasegawa et al., Fraction of MHCII and EpCAM expression characterizes distal lung 10 epithelial cells for alveolar type 2 cell isolation. Respir. Res. 18, 1–13 (2017).

24. O. Haller et al., Mx GTPases: Dynamin-like antiviral machines of innate immunity. Trends Microbiol. 23, 154–163 (2015).

25. M. Mordstein et al., Interferon-λ contributes to innate immunity of mice against influenza A virus but not against hepatotropic viruses. PLoS Pathog. 4, 1–7 (2008). 15

26. S. Crotta et al., Type I and type III interferons drive redundant amplification loops to induce a transcriptional signature in influenza-infected airway epithelia. PLoS Pathog. 9, e1003773 (2013).

27. P. Osterlund et al., Gene expression and antiviral activity of alpha/beta Interferons and interleukin-29 in virus-infected human myeloid dendritic cells. J. Virol. 79, 9608–9617 20 (2005).

28. N. A. Jewell et al., Lambda interferon is the predominant interferon induced by influenza A virus infection in vivo. J. Virol. 84, 11515–11522 (2010).

29. D. J. Gough et al., Constitutive type I interferon modulates homeostatic balance through tonic signaling. Immunity 36, 166–174 (2012). 25

30. A. E. Vaughan et al., Lineage-negative progenitors mobilize to regenerate lung epithelium after major injury. Nature 517, 621–5 (2015).

31. Y. You et al., Growth and differentiation of mouse tracheal epithelial cells: selection of a proliferative population. Am. J. Physiol. Lung Cell. Mol. Physiol. 283, L1315–L1321 (2002). 30

32. J. A. Zepp, E. E. Morrisey, Cellular crosstalk in the development and regeneration of the respiratory system. Nat. Rev. Mol. Cell Biol. (2019), doi:10.1038/s41580-019-0141-3.

33. P. S. Subramaniam et al., Type I interferon induction of the Cdk-inhibitor p21 is accompanied by ordered G1 arrest, differentiation and apoptosis of the Daudi B-cell line. Oncogene 16, 1885–90 (1998). 35

34. B. L. M. Hogan et al., Repair and regeneration of the respiratory system: Complexity, plasticity, and mechanisms of lung stem cell function. Cell Stem Cell 15, 123–138 (2014).

35. L. A. Pittet et al., Influenza virus infection decreases tracheal mucociliary velocity and clearance of Streptococcus pneumoniae. Am. J. Respir. Cell Mol. Biol. 42, 450–460

.CC-BY 4.0 International licensewas not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprint (whichthis version posted May 5, 2020. . https://doi.org/10.1101/2020.05.05.078360doi: bioRxiv preprint

Page 14: Type I and III interferons disrupt lung epithelial …...2020/05/05  · 4 Temporal overlap of antiviral IFN responses and lung epithelial repair following influenza virus infection

14

(2010). 36. J. Klinkhammer et al., IFN-λ prevents influenza virus spread from the upper airways to

the lungs and limits virus transmission. Elife 7, e33354 (2018). 37. M. L. Smith et al., Interaction of the p53-regulated protein GADD45 with proliferating

cell nuclear antigen. Science 266, 1376–1380 (1994). 5

38. K. Ueda, H. Arakawa, Y. Nakamura, Dual-specificity phosphatase 5 ( DUSP5 ) as a direct transcriptional target of tumor suppressor p53. Oncogene 22, 5586–5591 (2003).

39. J. A. McCullers, The co-pathogenesis of influenza viruses with bacteria in the lung. Nat. Rev. Microbiol. 12, 252–262 (2014).

40. D. A. Jaitin et al., Inquiring into the Differential Action of Interferons (IFNs): an IFN-α2 10 Mutant with Enhanced Affinity to IFNAR1 Is Functionally Similar to IFN-β. Mol. Cell. Biol. 26, 1888–1897 (2006).

41. I. Moraga et al., Receptor Density Is Key to the Alpha2/Beta Interferon Differential Activities. Mol. Cell. Biol. 29, 4778–4787 (2009).

42. G. Schreiber, The molecular basis for differential type i interferon signaling. J. Biol. 15 Chem. 292, 7285–7294 (2017).

43. V. François-Newton et al., USP18-based negative feedback control is induced by type I and type III interferons and specifically inactivates interferon α response. PLoS One 6 (2011), doi:10.1371/journal.pone.0022200.

44. V. Francois-Newton et al., USP18 establishes the transcriptional and anti-proliferative 20 interferon α/β differential. Biochem. J. 446, 509–516 (2012).

45. G. Schreiber, J. Piehler, The molecular basis for functional plasticity in type I interferon signaling. Trends Immunol. 36, 139–149 (2015).

46. K. H. Vousden, D. P. Lane, P53 in health and disease. Nat. Rev. Mol. Cell Biol. 8, 275–283 (2007). 25

47. A. Takaoka et al., Integration of interferon-α/β signalling to p53 responses in tumour suppression and antiviral defence. Nature 424, 516–523 (2003).

48. A. Al Jord et al., Calibrated mitotic oscillator drives motile ciliogenesis. Science 358, 803–806 (2017).

49. M. J. Holtzman, Asthma as a chronic disease of the innate and adaptive immune systems 30 responding to viruses and allergens. J. Clin. Invest. 122, 2741–2748 (2012).

50. H. Danahay et al., Notch2 is required for inflammatory cytokine-driven goblet cell metaplasia in the lung. Cell Rep. 10, 239–252 (2015).

51. C. Huang et al., Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 395, 497–506 (2020). 35

52. C. Wang et al., A novel coronavirus outbreak of global health concern. Lancet 395, 470–473 (2020).

53. N. Zhu et al., A novel coronavirus from patients with pneumonia in China, 2019. N. Engl. J. Med. 382, 727–733 (2020).

.CC-BY 4.0 International licensewas not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprint (whichthis version posted May 5, 2020. . https://doi.org/10.1101/2020.05.05.078360doi: bioRxiv preprint

Page 15: Type I and III interferons disrupt lung epithelial …...2020/05/05  · 4 Temporal overlap of antiviral IFN responses and lung epithelial repair following influenza virus infection

15

54. Z. Xu et al., Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir. Med. 2600, 19–21 (2020).

55. A. Pizzorno et al., Characterization and treatment of SARS-CoV-2 in nasal and bronchial human airway epithelia. bioRxiv, in press, doi:10.1101/2020.03.31.017889.

56. S. Tian et al., Pathological study of the 2019 novel coronavirus disease (COVID-19) 5 through postmortem core biopsies. Mod. Pathol., 10–12 (2020).

57. A. Granados et al., Influenza and rhinovirus viral load and disease severity in upper respiratory tract infections. J. Clin. Virol. 86, 14–19 (2017).

58. C. K. Lee et al., Comparison of pandemic (H1N1) 2009 and seasonal influenza viral loads, Singapore. Emerg. Infect. Dis. 17, 287–291 (2011). 10

59. C. Launes et al., Viral load at diagnosis and influenza A H1N1 (2009) disease severity in children. Influenza Other Respi. Viruses. 6, 89–92 (2012).

60. A. B. Gorini da Veiga, N. A. Kretzmann, L. T. Correa, Viral load and epidemiological profile of patients infected by pandemic influenza A (H1N1) 2009 and seasonal influenza A virus in southern brazil. J. Med. Virol. 84, 371–379 (2012). 15

61. M. D. De Jong et al., Fatal outcome of human influenza A (H5N1) is associated with high viral load and hypercytokinemia. Nat. Med. 12, 1203–1207 (2006).

62. Y. Zhai et al., Host transcriptional response to influenza and other acute respiratory viral infections – a prospective cohort study. PLoS Pathog. 11, 1–29 (2015).

63. J. Dunning et al., Progression of whole-blood transcriptional signatures from interferon-20 induced to neutrophil-associated patterns in severe influenza. Nat. Immunol. 19, 625–635 (2018).

64. Y. Nagaoka et al., Local and systemic immune responses to influenza A virus infection in pneumonia and encephalitis mouse models. Dis. Markers. 2017 (2017), doi:10.1155/2017/2594231. 25

65. D. Kobasa et al., Aberrant innate immune response in lethal infection of macaques with the 1918 influenza virus. Nature 445, 319–323 (2007).

66. M. J. Cameron et al., Interferon-mediated immunopathological events are associated with atypical innate and adaptive immune responses in patients with severe acute respiratory syndrome. J. Virol. 81, 8692–8706 (2007). 30

67. R. Channappanavar et al., IFN-I response timing relative to virus replication determines MERS coronavirus infection outcomes. J. Clin. Invest. 129, 3625–3639 (2019).

68. S. Davidson et al., IFNλ is a potent anti-influenza therapeutic without the inflammatory side effects of IFN a treatment. EMBO Mol. Med. 8, 1–14 (2016).

69. I. E. Galani et al., Interferon-λ mediates non-redundant front-line antiviral protection 35 against influenza virus infection without compromising host fitness. Immunity 46, 875-890.e6 (2017).

70. L. Prokunina-olsson et al., COVID-19 and emerging viral infections : The case for interferon lambda. J. Exp. Med. 217, 5–8 (2020).

.CC-BY 4.0 International licensewas not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprint (whichthis version posted May 5, 2020. . https://doi.org/10.1101/2020.05.05.078360doi: bioRxiv preprint

Page 16: Type I and III interferons disrupt lung epithelial …...2020/05/05  · 4 Temporal overlap of antiviral IFN responses and lung epithelial repair following influenza virus infection

16

71. P. Staeheli et al., Polyclonal and monoclonal antibodies to the interferon-inducible protein Mx of influenza virus-resistant mice. J. Biol. Chem. 260, 1821–1825 (1985).

72. G. Kochs et al., Strong interferon-inducing capacity of a highly virulent variant of influenza A virus strain PR8 with deletions in the NS1 gene. J. Gen. Virol. 90, 2990–2994 (2009). 5

73. C. Dellgren et al., Human interferon-λ3 is a potent member of the type III interferon family. Genes Immun. 10, 125–131 (2009).

74. A. Horani et al., Rho-associated protein kinase inhibition enhances airway epithelial basal-cell proliferation and lentivirus transduction. Am. J. Respir. Cell Mol. Biol. 49, 341–347 (2013). 10

75. C. L. Ward et al., Design and performance testing of quantitative real time PCR assays for influenza A and B viral load measurement. J. Clin. Virol. 29, 179–188 (2004).

15 20 25 30 35 40

.CC-BY 4.0 International licensewas not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprint (whichthis version posted May 5, 2020. . https://doi.org/10.1101/2020.05.05.078360doi: bioRxiv preprint

Page 17: Type I and III interferons disrupt lung epithelial …...2020/05/05  · 4 Temporal overlap of antiviral IFN responses and lung epithelial repair following influenza virus infection

17

Acknowledgments: We thank the Crick sequencing, histopathology, flow cytometry and animal facilities for excellent support. We are grateful to Simon Priestnall and Alejandro Suarez-Bonnet for histopathology scoring of lung H&E sections, and to Anne O’Garra, Gitta Stockinger, Peter Staeheli and Daniel Schnepf for critically reading the manuscript. 5

Author contributions: Conception: A.W., S.C. and J.M; research design and experimentation: J.M., S.C. and T.McC.; data analysis: J.M., S.C., T.McC. and M.L.; production and provision of key reagents: H.H.G. and R.H.; writing and editing of the manuscript: J.M., A.W., and S.C. All authors read and approved the final manuscript. 10

Funding: JM, SC, ML, TMcC and AW were supported by the Francis Crick Institute which receives its core funding from Cancer Research UK (FC001206), the UK Medical Research Council (FC001206), and the Wellcome Trust (FC001206). RH and HHG were supported by Independent Research Fund Denmark | Medical Sciences, Research grant agreement 11-107588; and by Novo Nordisk Foundation, Grant agreement NNF190C0058287. 15

Competing interests: The authors declare no competing interests. Data and materials availability: All data supporting the findings of this study are available within the paper or in the supplementary materials, or are available from the authors upon request. 20

Supplementary Materials: Materials and Methods Figures S1-S9 Tables S1 25

Data S1 References (31, 71–75)

.CC-BY 4.0 International licensewas not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprint (whichthis version posted May 5, 2020. . https://doi.org/10.1101/2020.05.05.078360doi: bioRxiv preprint

Page 18: Type I and III interferons disrupt lung epithelial …...2020/05/05  · 4 Temporal overlap of antiviral IFN responses and lung epithelial repair following influenza virus infection

18

Fig. 1. Type I and III IFNs reduce epithelial cell proliferation during lung repair. (A and B) Mice were infected with 104 TCID50 X31 (H3N2) influenza virus in 30 µl intranasally. (A) Proliferating (Ki67+) AT2 cells (EpCam+MHCII+CD49flow) were measured by flow cytometry (n 5

= 5), and (B) type I and III IFN levels were detected in BALF (n = 4), on indicated days post infection. (C and D) X31 infected mice were administered IFNs every 24 hours (on days 7 to 10 post infection), and proliferating (Ki67+) AT2 cells (EpCam+MHCII+CD49flow) were measured by flow cytometry on day 11 post infection. (C) Lethally irradiated WT mice were injected with Ifnar1-/- bone marrow cells. Following reconstitution, influenza virus-infected chimeric mice were 10

treated with PBS control (n = 8), IFN-α (n = 9), or IFN-β (n = 9). Naïve controls are uninfected, untreated bone marrow chimeric mice (n = 2). (D) Infected WT mice were treated with IFN-λ (n = 4) or PBS control (n = 4). Naïve controls are uninfected, untreated WT mice (n = 5). (E) B6-Mx1 mice were infected with 2.5 x 103 TCID50 hvPR8-DNS1 (H1N1) and treated with IFN-λ (n = 4) or PBS control (n = 4) (IFN treatment and lung analysis was performed as for C and D). (F to 15

H) Lungs from X31 infected WT (n = 4 to 7), (F) Ifnar1-/- (n = 4), (G) Ifnlr1-/- (n = 7), and (H) BM chimeric mice (n = 4 to 5) mice were harvested and proliferating (Ki67+) AT2 cells were measured by flow cytometry on day 8 post infection. All data are representative of at least two independent experiments. Data are shown as mean ± s.e.m. and statistical significance was

Naive

PBS α β0

10

20

30

40

Ki67

+ (%

Of E

pCam

+ M

HC

-II+) **

***

Naive

PBS λ0

20

40

60

Ki67

+ (%

Of E

pCam

+ M

HC

-II+) *

WT

Ifnar1-/-

0

5

10

15

Ki67

+ (%

Of E

pCam

+ M

HC

-II+)

***

WT

Ifnlr1-/-

0

10

20

30

Ki67

+ (%

Of E

pCam

+ M

HC

-II+ ) *

0

5

10

15

Ki67

+ (%

Of E

pCam

+ M

HC

-II+)

P = 0.07*

WT >

WT

WT > Ifnar1-/-

WT > Ifnlr1-/-

IAV (d11) IAV (d11)

C

PBS λ0

5

10

15

20Ki

67+

(% O

f EpC

am+

MH

C-II

+) *

Ifnar1-/- > WT D E F G H

A B

B6-Mx1 + hvPR8-ΔNS1

0 2 4 6 8 10 12 140

20

40

60

Days post infection

Ki67

+ (%

Of E

pCam

+ M

HC

-II+) Lung epithelial cell

proliferation

0 2 4 6 80

100

200

300

400

Days post infectionpg

/ml

IFN-α

0 2 4 6 80

20

40

60

Days post infection

pg/m

l

IFN-β

0 2 4 6 80

200

400

600

Days post infection

pg/m

l

IFN-λ

.CC-BY 4.0 International licensewas not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprint (whichthis version posted May 5, 2020. . https://doi.org/10.1101/2020.05.05.078360doi: bioRxiv preprint

Page 19: Type I and III interferons disrupt lung epithelial …...2020/05/05  · 4 Temporal overlap of antiviral IFN responses and lung epithelial repair following influenza virus infection

19

assessed by one-way ANOVA with Dunnett’s post-test (C, D and H), or unpaired two-tailed Student’s t-test (E to G). ns, not significant (P >0.05); *P ≤ 0.05, **P ≤ 0.01, ***P ≤ 0.001. 5

.CC-BY 4.0 International licensewas not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprint (whichthis version posted May 5, 2020. . https://doi.org/10.1101/2020.05.05.078360doi: bioRxiv preprint

Page 20: Type I and III interferons disrupt lung epithelial …...2020/05/05  · 4 Temporal overlap of antiviral IFN responses and lung epithelial repair following influenza virus infection

20

Fig. 2. IFN signalling blocks airway epithelial cell growth and differentiation. (A) AECs were seeded at a low density (500 cells/transwell) or high density (104 cells/transwell) in the presence of equivalent doses of IFN-α, -β, -λ, or media control, and grown for 12 days (n = 3 for all 5

- α β λ0

40

80

120

****ns

40K 60K 80K 100K 120K 140K 40K 60K 80K 100K 120K 140K40K 60K 80K 100K 120K 140K 40K 60K 80K 100K 120K 140K

- α β λ0

40

80

120

S-ph

ase

(% re

lativ

e to

moc

k)

***ns

**

Low cell density High cell densityA Mouse AECs Human AECs

DAPI

EdU

38.3 31.4 18.418.1

Mock IFN-α IFN-λIFN-β

4 6 8 10 120

500

1000

>1500

Days post plating

TEER

(Ω)

4 6 8 10 120

500

1000

>1500

Days post plating

Confluence

0

5

10

15

Cel

l num

ber (

x105 ) *

****

0

10

20

30

Cel

l num

ber (

x104 )

********

**

-αβλ

β

λ

% A

cety

late

d α

-tubu

lin

(rela

tive

to m

ock)

*

0 3 5 810-1

100

101

102

Days post ALI

Ccn

o m

RN

A (fo

ld)

***

ns

0 3 5 810-1

10 0

10 1

10 2

10 3

Days post ALI

Mci

das

mR

NA

(fold

)

***

****

ns

0 3 5 810-1

100

101

102

103

104

Days post ALI

Scb1

a1 m

RN

A (fo

ld)

*******

*****

0 3 5 810-1

100

101

102

Days post ALI

Muc

5ac

mR

NA

(fold

)

********

Naive IAV (d14)0

5

10

15

20

CD

24hi

gh C

D49

f+ (%

Of E

pCam

+)

ns

**

Ciliated cell markers Secretory cell markers

WT

Ifnlr1-/-

-

α

β

λ

ns

B C

D E

F

G H I

100 μmDAPI

Acetylated α-tubulin

WT Ifnlr1-/-

.CC-BY 4.0 International licensewas not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprint (whichthis version posted May 5, 2020. . https://doi.org/10.1101/2020.05.05.078360doi: bioRxiv preprint

Page 21: Type I and III interferons disrupt lung epithelial …...2020/05/05  · 4 Temporal overlap of antiviral IFN responses and lung epithelial repair following influenza virus infection

21

conditions). Confluence was determined by measuring transepithelial electrical resistance (TEER; >1000Ω = confluent cultures). (B, D and E) Proliferating mouse AEC cultures (2 days prior to exposure to an ALI) were treated for 5 days with IFNs (day -2 ALI to day 3 post ALI), and effects on growth were determined by cell number (B) (n = 9) and EdU incorporation to measure proliferation (D and E) (n = 9). (C) Primary human AEC cultures were treated with IFNs for 5 5

days, and cells were counted (n = 4 to 6). (F and G) Mouse AECs were grown to confluence, then exposed to an air-liquid interface (ALI) for 2 days. IFNs were then administrated for 6 days during ALI exposure (n = 6 for all conditions). Differentiation was determined by mRNA expression of the indicated genes (F) and the level of acetylated α-tubulin staining in cultures (G). (H and I) WT and Ifnlr1-/- mice were infected with influenza virus, and lungs were analysed by 10

immunofluorescence (DAPI/acetylated α-tubulin) on day 10 post infection (n = 4) (H), and flow cytometry (EpCam+CD49fhighCD24+) on day 14 post infection (n = 3) (I). All data are representative of at least three independent experiments. Data are shown as mean ± s.e.m. and statistical significance was assessed by one-way (B, C, E and G) or two-way (F and I) ANOVA with Dunnett’s post-test. ns, not significant (P >0.05); *P ≤ 0.05, **P ≤ 0.01, ***P ≤ 0.001, ****P 15

≤ 0.0001.

.CC-BY 4.0 International licensewas not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprint (whichthis version posted May 5, 2020. . https://doi.org/10.1101/2020.05.05.078360doi: bioRxiv preprint

Page 22: Type I and III interferons disrupt lung epithelial …...2020/05/05  · 4 Temporal overlap of antiviral IFN responses and lung epithelial repair following influenza virus infection

22

Fig. 3. Type I and III IFNs activate antiproliferative and cell death pathways in AECs via induction of p53. (A) Schematic diagram for IFN treatment of AECs for RNA-seq analysis. (B) PCA plot of RNA-seq data from AECs following IFN treatment, and untreated controls. (C) 5

.CC-BY 4.0 International licensewas not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprint (whichthis version posted May 5, 2020. . https://doi.org/10.1101/2020.05.05.078360doi: bioRxiv preprint

Page 23: Type I and III interferons disrupt lung epithelial …...2020/05/05  · 4 Temporal overlap of antiviral IFN responses and lung epithelial repair following influenza virus infection

23

Heatmap for significant differences in ‘Canonical Pathways’ for 9 pairwise comparisons between indicated IFN treatment and the respective mock, at each time point (fold change > 1.5, one-way ANOVA with Benjamini–Hochberg correction, P < 0.05), Gene expression was compared using IPA Comparison Analysis. (D) Predicted upstream transcriptional regulators of differentially expressed genes (IPA analysis). (E to G) WT and p53-/- AECs were treated with IFN subtypes for 5

5 days, and measured for growth by cell number (E), CFSE dilution (F), and mRNA expression of indicated genes (G) (n = 3 for all conditions). (H and I) Ifnar1-/- > WT BM chimeric mice (H) (n = 4 to 5) and α-Ly6G treated mice (I) (n = 4) infected with influenza virus (X31), and treated with IFN every 24 hours consecutively for 4 days (day 7 to 10 post infection), before EpCam+MHCII+CD49flow AT2 cells were analysed for p53 mean fluorescence intensity (MFI) on 10

day 11 post infection by flow cytometry. All data are representative of at least two independent experiments (E to I). Data are shown as mean ± s.e.m. and statistical significance was assessed by two-way (E to G) or one-way (H) ANOVA with Dunnett’s post-test, or unpaired two-tailed Student’s t-test (I). ns, not significant (P >0.05); *P ≤ 0.05, **P ≤ 0.01, ***P ≤ 0.001, ****P ≤ 0.0001. 15

.CC-BY 4.0 International licensewas not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprint (whichthis version posted May 5, 2020. . https://doi.org/10.1101/2020.05.05.078360doi: bioRxiv preprint

Page 24: Type I and III interferons disrupt lung epithelial …...2020/05/05  · 4 Temporal overlap of antiviral IFN responses and lung epithelial repair following influenza virus infection

24

Fig. 4. Ifnlr1-/- mice have improved lung repair, reduced damage, and improved epithelial barrier function. WT or Ifnlr1-/- mice were infected with 104 TCID50 X31 influenza virus (X31). (A) GSEA plots of RNA-seq datasets from WT or Ifnlr1-/- bulk lung epithelial cells (EpCam+) on day 8 post infection. (B and C) Total cell and red blood cell number in BALF on day 8 post 5

infection (n = 4 for both WT and Ifnlr1-/- mice). (D) Histopathological analysis of H&E lung sections on day 9 post infection (n = 4 for both WT and Ifnlr1-/- mice). (E) Lethally irradiated WT and Ifnlr1-/- mice were injected with WT bone marrow cells. Following reconstitution, chimeric mice were challenged with 2x105 colony-forming units (c.f.u.) TIGR4 in 30µl on day 8 post influenza virus infection (n = 8 WT, n = 9 Ifnlr1-/-). All data are representative of at least two 10

independent experiments (B to E). Data are shown as mean ± s.e.m. and statistical significance was assessed by unpaired two-tailed Student’s t-test (B to D) or log-rank (Mantel–Cox) test (E, survival). *P ≤ 0.05, **P ≤ 0.01.

A

Ifnlr1-/- WT

Canonical pathways: Cell cycle0.1

0.05

0

-0.05FDR q value: 0.043

NES: 2.27P value: 0

Ifnlr1-/- WT

GO biological processes: Cilium organisation0.40.30.20.1

0

FDR q value: 0NES: 6.08P value: 0

0

10

20

30

40

Cel

ls/m

L (x

105 )

**

0

10

20

30

Cel

ls/m

L (x

105 )

**WT

Ifnlr1-/-

Ifnlr1-/-

WT

0

1

2

3

His

topa

thol

ogy

scor

e

*

Inflammation

Total BALF cells RBCs in BALF

WT > WT

WT > Ifnlr1-/-

S. pneumoniae (d8)

0 8 10 12 140

50

100

Days post influenza infection

% S

urvi

val

**

B

C

D E

.CC-BY 4.0 International licensewas not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprint (whichthis version posted May 5, 2020. . https://doi.org/10.1101/2020.05.05.078360doi: bioRxiv preprint