Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Letter
  • Published:

MHC class II proteins mediate cross-species entry of bat influenza viruses

Abstract

Zoonotic influenza A viruses of avian origin can cause severe disease in individuals, or even global pandemics, and thus pose a threat to human populations. Waterfowl and shorebirds are believed to be the reservoir for all influenza A viruses, but this has recently been challenged by the identification of novel influenza A viruses in bats1,2. The major bat influenza A virus envelope glycoprotein, haemagglutinin, does not bind the canonical influenza A virus receptor, sialic acid or any other glycan1,3,4, despite its high sequence and structural homology with conventional haemagglutinins. This functionally uncharacterized plasticity of the bat influenza A virus haemagglutinin means the tropism and zoonotic potential of these viruses has not been fully determined. Here we show, using transcriptomic profiling of susceptible versus non-susceptible cells in combination with genome-wide CRISPR–Cas9 screening, that the major histocompatibility complex class II (MHC-II) human leukocyte antigen DR isotype (HLA-DR) is an essential entry determinant for bat influenza A viruses. Genetic ablation of the HLA-DR α-chain rendered cells resistant to infection by bat influenza A virus, whereas ectopic expression of the HLA-DR complex in non-susceptible cells conferred susceptibility. Expression of MHC-II from different bat species, pigs, mice or chickens also conferred susceptibility to infection. Notably, the infection of mice with bat influenza A virus resulted in robust virus replication in the upper respiratory tract, whereas mice deficient for MHC-II were resistant. Collectively, our data identify MHC-II as a crucial entry mediator for bat influenza A viruses in multiple species, which permits a broad vertebrate tropism.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Identification of HLA-DR as an entry determinant for bat IAV.
Fig. 2: HLA-DR mediates the entry of bat IAV into U-87MG cells.
Fig. 3: Homologues of HLA-DR from different species confer susceptibility to bat IAV.
Fig. 4: MHC-II knockout mice are resistant to bat IAV infection.

Similar content being viewed by others

Data availability

The authors declare that the data supporting the findings of this study are available within the paper and its Supplementary Information files. The associated raw data for Fig. 1a are provided in Supplementary Table 1, and raw data for Fig. 1c in Supplementary Table 2. Any further relevant data are available from the corresponding authors upon reasonable request.

References

  1. Tong, S. et al. New World bats harbor diverse influenza A viruses. PLoS Pathog. 9, e1003657 (2013).

    PubMed  PubMed Central  Google Scholar 

  2. Tong, S. et al. A distinct lineage of influenza A virus from bats. Proc. Natl Acad. Sci. USA 109, 4269–4274 (2012).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  3. Zhu, X. et al. Hemagglutinin homologue from H17N10 bat influenza virus exhibits divergent receptor-binding and pH-dependent fusion activities. Proc. Natl Acad. Sci. USA 110, 1458–1463 (2013).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  4. Sun, X. et al. Bat-derived influenza hemagglutinin H17 does not bind canonical avian or human receptors and most likely uses a unique entry mechanism. Cell Rep. 3, 769–778 (2013).

    CAS  PubMed  Google Scholar 

  5. Weis, W. et al. Structure of the influenza virus haemagglutinin complexed with its receptor, sialic acid. Nature 333, 426–431 (1988).

    ADS  CAS  PubMed  Google Scholar 

  6. Gamblin, S. J. et al. The structure and receptor binding properties of the 1918 influenza hemagglutinin. Science 303, 1838–1842 (2004).

    ADS  CAS  PubMed  Google Scholar 

  7. Stevens, J. et al. Glycan microarray analysis of the hemagglutinins from modern and pandemic influenza viruses reveals different receptor specificities. J. Mol. Biol. 355, 1143–1155 (2006).

    CAS  PubMed  Google Scholar 

  8. Edinger, T. O., Pohl, M. O., Yángüez, E. & Stertz, S. Cathepsin W is required for escape of influenza A virus from late endosomes. MBio 6, e00297-15 (2015).

    PubMed  PubMed Central  Google Scholar 

  9. Wu, Y., Wu, Y., Tefsen, B., Shi, Y. & Gao, G. F. Bat-derived influenza-like viruses H17N10 and H18N11. Trends Microbiol. 22, 183–191 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Pugach, P. et al. HIV-1 clones resistant to a small molecule CCR5 inhibitor use the inhibitor-bound form of CCR5 for entry. Virology 361, 212–228 (2007).

    CAS  PubMed  Google Scholar 

  11. Tscherne, D. M., Manicassamy, B. & García-Sastre, A. An enzymatic virus-like particle assay for sensitive detection of virus entry. J. Virol. Methods 163, 336–343 (2010).

    CAS  PubMed  Google Scholar 

  12. Schröder, B. The multifaceted roles of the invariant chain CD74—more than just a chaperone. Biochim. Biophys. Acta 1863, 1269–1281 (2016).

    PubMed  Google Scholar 

  13. Stockinger, B. et al. A role of Ia-associated invariant chains in antigen processing and presentation. Cell 56, 683–689 (1989).

    CAS  PubMed  Google Scholar 

  14. Aydillo, T. et al. Specific mutations in the PB2 protein of influenza A virus compensate for the lack of efficient interferon antagonism of the NS1 protein of bat influenza A-like viruses. J. Virol. 92, e02021-17 (2018).

    PubMed  PubMed Central  Google Scholar 

  15. Irla, M. et al. Autoantigen-specific interactions with CD4+ thymocytes control mature medullary thymic epithelial cell cellularity. Immunity 29, 451–463 (2008).

    CAS  PubMed  Google Scholar 

  16. Crameri, G. et al. Establishment, immortalisation and characterisation of pteropid bat cell lines. PLoS ONE 4, e8266 (2009).

    ADS  PubMed  PubMed Central  Google Scholar 

  17. Moreira, E. A. et al. Synthetically derived bat influenza A-like viruses reveal a cell type- but not species-specific tropism. Proc. Natl Acad. Sci. USA 113, 12797–12802 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Sanjana, N. E., Shalem, O. & Zhang, F. Improved vectors and genome-wide libraries for CRISPR screening. Nat. Methods 11, 783–784 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Shalem, O. et al. Genome-scale CRISPR-Cas9 knockout screening in human cells. Science 343, 84–87 (2014).

    Article  ADS  CAS  PubMed  Google Scholar 

  20. Enomoto, M., Bunge, M. B. & Tsoulfas, P. A multifunctional neurotrophin with reduced affinity to p75NTR enhances transplanted Schwann cell survival and axon growth after spinal cord injury. Exp. Neurol. 248, 170–182 (2013).

    CAS  PubMed  Google Scholar 

  21. Doench, J. G. et al. Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPR–Cas9. Nat. Biotechnol. 34, 184–191 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Dehairs, J., Talebi, A., Cherifi, Y. & Swinnen, J. V. CRISP-ID: decoding CRISPR mediated indels by Sanger sequencing. Sci. Rep. 6, 28973 (2016).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  23. Hayer, A. et al. Engulfed cadherin fingers are polarized junctional structures between collectively migrating endothelial cells. Nat. Cell Biol. 18, 1311–1323 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Juozapaitis, M. et al. An infectious bat-derived chimeric influenza virus harbouring the entry machinery of an influenza A virus. Nat. Commun. 5, 4448 (2014).

    CAS  PubMed  Google Scholar 

  25. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    CAS  PubMed  Google Scholar 

  26. Lawrence, M. et al. Software for computing and annotating genomic ranges. PLOS Comput. Biol. 9, e1003118 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    CAS  PubMed  Google Scholar 

  28. Martin, M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet.journal 17, 10–12 (2011).

    Google Scholar 

  29. Li, W. et al. MAGeCK enables robust identification of essential genes from genome-scale CRISPR/Cas9 knockout screens. Genome Biol. 15, 554 (2014).

    PubMed  PubMed Central  Google Scholar 

  30. Reuther, P. et al. Generation of a variety of stable influenza A reporter viruses by genetic engineering of the NS gene segment. Sci. Rep. 5, 11346 (2015).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  31. Zimmer, G., Locher, S., Berger Rentsch, M. & Halbherr, S. J. Pseudotyping of vesicular stomatitis virus with the envelope glycoproteins of highly pathogenic avian influenza viruses. J. Gen. Virol. 95, 1634–1639 (2014).

    CAS  PubMed  Google Scholar 

  32. Krammer, F. et al. A carboxy-terminal trimerization domain stabilizes conformational epitopes on the stalk domain of soluble recombinant hemagglutinin substrates. PLoS ONE 7, e43603 (2012).

    ADS  CAS  PubMed  PubMed Central  Google Scholar 

  33. Bussmann, B. M., Reiche, S., Jacob, L. H., Braun, J. M. & Jassoy, C. Antigenic and cellular localisation analysis of the severe acute respiratory syndrome coronavirus nucleocapsid protein using monoclonal antibodies. Virus Res. 122, 119–126 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Monaghan, S. J. et al. Expression of immunogenic structural proteins of cyprinid herpesvirus 3 in vitro assessed using immunofluorescence. Vet. Res. 47, 8 (2016).

    PubMed  PubMed Central  Google Scholar 

  35. Edelstein, A., Amodaj, N., Hoover, K., Vale, R. & Stuurman, N. Computer control of microscopes using μManager. Curr. Protoc. Mol. Biol. 92, 14.20.1–14.20.17 (2010).

    Google Scholar 

  36. Ridler, T. W. & Calvard, S. Picture thresholding using an iterative selection method. IEEE Trans. Syst. Man Cybern. 8, 630–632 (1978).

    Google Scholar 

  37. Rueden, C. T. et al. ImageJ2: ImageJ for the next generation of scientific image data. BMC Bioinformatics 18, 529 (2017).

    PubMed  PubMed Central  Google Scholar 

  38. Papenfuss, A. T. et al. The immune gene repertoire of an important viral reservoir, the Australian black flying fox. BMC Genomics 13, 261 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Ng, J. H. J., Tachedjian, M., Wang, L. F. & Baker, M. L. Insights into the ancestral organisation of the mammalian MHC class II region from the genome of the pteropid bat, Pteropus alecto. BMC Genomics 18, 388 (2017).

    PubMed  PubMed Central  Google Scholar 

  40. Kittel, B. et al. Revised guides for organ sampling and trimming in rats and mice–part 2. A joint publication of the RITA and NACAD groups. Exp. Toxicol. Pathol. 55, 413–431 (2004).

    PubMed  Google Scholar 

  41. Yewdell, J. W., Frank, E. & Gerhard, W. Expression of influenza A virus internal antigens on the surface of infected P815 cells. J. Immunol. 126, 1814–1819 (1981).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank C. Kastenholz, K. Flämig, J. Brandel, S. Schuparis, S. Sander and G. Czerwinski for assistance; A. Dudek, P. Staeheli, D. Schnepf and A. Trkola for discussions; and R. Zengerle, D. Szabó, P. Koltay, A. Karsai and S. Zimmermann for their support. This work was supported by grants from the Swiss National Science Foundation to S.S. (310030E-164065) and B.G.H. (31003A_182464), a grant from the German Research Foundation to M.S. (SCHW 632/17-1) and M.B. (BE 5187/4-1) and the Excellence Initiative of the German Research Foundation (GSC-4, Spemann Graduate School) to T.T. This work was also partly supported by CRIP (Center for Research on Influenza Pathogenesis) and NIAID funded Center of Excellence in Influenza Research and Surveillance (CEIRS) to A.G.-S. (HHSN272201400008C). M.W.C. and C.B. were supported by NIAID grant U19AI135972.

Reviewer information

Nature thanks Michael Farzan, David Steinhauer and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Author information

Authors and Affiliations

Authors

Contributions

K.C. and W.R. contributed equally to this work. U.K., T.T., K.C., W.R., S.C.G., M.O.P., D.E., C.J., D.H., S.R., J.S., R.U., J.W., M.G.B.H., M.W.C., A. Hunziker, E.Y. and A. Halenius performed experiments; U.K., T.T., K.C., M.S. and S.S. designed experiments; T.A., F.K., J.O., A.O. and G.Z. contributed reagents and mice; M.M., C.B., B.G.H., A.G.-S., M.B., M.S. and S.S. supervised work and acquired funding; U.K., M.S. and S.S. wrote the manuscript.

Corresponding authors

Correspondence to Martin Schwemmle or Silke Stertz.

Ethics declarations

Competing interests

C.J. is the patentee of the patent covering the technology of emulsion coupling (WO2016083793) and a shareholder in Actome GmbH (Germany), which holds the patent.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data figures and tables

Extended Data Fig. 1 Analysis of transcriptional profiles from H18N11-susceptible and non-susceptible cell lines.

a, MDCKII clone no. 1 (here labelled MDCKII #1), MDCKII clone no. 2 (here labelled MDCKII #2), U-87MG, U-118MG, Calu-3 and A549 cells were infected for 90 min at 37 °C with luciferase-encoding VLPs pseudotyped with H18N11 or H1N1, or control VLPs that lacked any surface glycoprotein (empty vector). At 48 h p.i., luciferase signals were measured and normalized to control samples. Fold increase in luciferase signals relative to samples infected with empty vector (EV) is plotted. b, MDCKII clone no. 1 (here labelled MDCKII #1), MDCKII clone no. 2 (here labelled MDCKII #2), U-87MG, U-118MG and Calu-3 cells were infected with BlaM1 VLPs pseudotyped with H1N1 or H18, for 4 h at 37 °C. Entry-positive cells were measured with the fluorogenic β-lactamase substrate CCF2-AM, and quantified by flow cytometry. Relative numbers of entry-positive cells are shown. **, signals from A549 were below the detection limit, owing to inefficient loading with the fluorogenic β-lactamase substrate. a, b, Data are mean ± s.d. from n = 3 independent experiments. Values below background levels of 1 are displayed on the x axes. ce, Transcriptional profiles of the indicated H18N11-susceptible versus non-susceptible cell lines are shown as volcano plots. The two-group comparison—each of which consists of n = 4 independent samples—was performed using the gene-wise negative binomial generalized linear model with quasi-likelihood tests, implemented in EdgeR. Adjustments for multiple comparisons were made by applying the Benjamini–Hochberg method. Significantly (P ≤ 0.01) upregulated (fold change (expressed as log2(transcript level susceptible cell line/non-susceptible cell line) ≥ 1) and downregulated (fold change (expressed as log2(transcript level susceptible cell line/non-susceptible cell line)≤ −1) transcripts are shown in red and blue, respectively. Transcripts expressed at similar levels (−1 ≤ fold change (expressed as log2(transcript level susceptible cell line/non-susceptible cell line) ≤ 1) are shown in grey. Black dots indicate significantly upregulated transcripts, filtered according to the Gene Ontology annotations GO:0016020 (membrane), GO:0016021 (integral component of membrane) and GO:0005886 (plasma membrane), that overlap in the H18N11-susceptible cell lines. f, HLA-DR surface staining of U-87MG, U-118MG, Calu-3 and A549 cells, and DLA surface staining of MDCKII clone no. 1 (here labelled MDCKII #1) and MDCKII clone no. 2 (here labelled MDCKII #2) cells, is shown in contour plots. Representative plots of n = 3 independent experiments are shown.

Extended Data Fig. 2 Genome-wide CRISPR–Cas9 screen on U-87MG cells to identify potential receptor candidates for bat influenza virus.

a, Schematic of the genome-wide CRISPR–Cas9 screen. U-87MG cells stably expressing Cas9 (U-87MG–Cas9) were transduced with lentiviruses encoding a human CRISPR-knockout pooled sgRNA library at an MOI of 0.3. Two days after transduction, cells (U-87MG–Cas9–sgRNA) were selected with puromycin (1 μg/ml) for ten days, and subsequently infected with VSV-H18 at an MOI of 10. Survivors were re-infected with VSV-H18 at an MOI of 10 for 4 additional rounds, and collected on day 27 p.i. Genomic DNA from cell pellets was isolated and analysed using next-generation sequencing. b, Susceptibility of the survivors and the wild-type U-87MG cells was monitored by flow cytometry measuring GFP signals from cells infected with H18-VSV reporter virus at different MOIs. Relative numbers of GFP-positive cells are shown.

Extended Data Fig. 3 HLA-DR is required for bat IAV infection in U-87MG and Calu-3 cells.

a) U-87MG control, U-87MG KO no. 1 (here labelled KO #1) and U-87MG KO no. 2 (here labelled KO #2) cells were infected with recombinant VSV-H18 (red), VSV (light grey) and H7N7 (dark grey) reporter viruses expressing GFP at an MOI of 1, 0.1 and 0.1, respectively, for at least 24 h. The frequency of GFP-positive cells was quantified by flow cytometry. b, U-87MG control, U-87MG KO no. 1 and U-87MG KO no. 2 (labelled ‘KO #1’ and ‘KO #2’) were infected with recombinant VSV-H17 expressing GFP at an MOI of 1 for 72 h. Representative images of n = 3 independent experiments are shown. Scale bar, 100 μm. c, HLA-DR surface staining of U-87MG control, U-87MG KO no. 1 and U-87MG KO no. 2 (labelled ‘KO #1’ and ‘KO #2’), stably transduced with either a control lentivirus (labelled ‘LV-ctrl’) or a lentivirus encoding HLA-DRA (labelled ‘LV-HLA-DRA’) from a representative of n = 3 independent experiments is shown. d, e, Indicated U-87MG cell lines were infected with VSV-H18 reporter virus expressing GFP at an MOI of 5 for 48 h. Representative images are shown in d. Frequency of GFP-positive cells was quantified by flow cytometry and is depicted in e. a, e, Data are mean ± s.d. from n = 3 independent experiments. Values below background levels of 1 are displayed on the x axes. f, HLA-DR surface staining of Calu-3 HLA-DRA knockout cells (labelled ‘KO #1’ and ‘KO #2’) and Calu-3 cells (labelled ‘Ctrl’) is shown from a representative of n = 2 independent experiments. g, Calu-3 control, Calu-3 KO no. 1 and Calu-3 KO no. 2 cells (labelled ‘KO #1’ and ‘KO #2’) were infected with BlaM1 VLPs pseudotyped with H1N1 or H18 for 4 h at 37 °C. Entry-positive cells were measured with the fluorogenic β-lactamase substrate CCF2-AM, and quantified by flow cytometry. Relative numbers of entry-positive cells are shown. Data are mean from n = 2 independent experiments. Values below background levels of 1 are displayed on the x axis.

Extended Data Fig. 4 Alignment of protein sequences of haemagglutinins from different subtypes of IAV.

Residues conserved across all subtypes are highlighted in red. Residues known to be part of the receptor-binding site in conventional IAV are highlighted by light purple boxes, and the fusion peptide is marked by a yellow box.

Extended Data Fig. 5 Ectopic expression of HLA-DR renders non-susceptible cell lines susceptible.

a, HLA-DR surface staining of A549 LV control (LV-ctrl) and A549 LV HLA-DR (LV-HLA-DR) (left), MDCKII clone no. 2 LV control and MDCKII clone no. 2 LV HLA-DR (middle), U-118MG LV control and U-118MG LV HLA-DR (right) from a representative of one experiment is shown. b, Indicated cell lines were infected with luciferase-encoding VLPs pseudotyped with H18 or H1N1, or control VLPs that lack any surface glycoprotein (empty vector). Fold increase in luciferase signals relative to samples infected with empty vector is plotted. c, Indicated cell lines were infected with VSV-H18 at an MOI of 10 and GFP-positive cells were quantified by flow cytometry at 24 h p.i. for A549 LV control and A549 LV HLA-DR (left), at 24 h p.i. for MDCKII clone no. 2 LV control and MDCKII clone no. 2 LV HLA-DR (middle), and at 72 h p.i. for U-118MG LV control and U-118MG LV HLA-DR (right). b, c, Data are mean ± s.d. from n = 3 independent experiments. Values below background levels of 1 are displayed on the x axes. d, MDCKII clone no. 2 LV control and MDCKII clone no. 2 LV HLA-DR were infected with IAV H18N11 at MOI = 0.001. At indicated time points p.i., supernatants were collected and viral titres were determined by immunofluorescence on subconfluent MDCKII clone no. 1 cells, using rabbit polyclonal anti-H18. Data are mean ± s.d. from n = 3 independent experiments.

Extended Data Fig. 6 Polykaryon formation assay and PLAs detect interaction of H18 with HLA-DR complexes.

a, HEK293T cells were co-transfected with plasmids encoding mCherry and H1. Transfected HEK293T cells were treated with TPCK–trypsin to cleave haemagglutinin and seeded on top of control MDCKII clone no. 2 cells (labelled as LV-ctrl) or MDCKII clone no. 2 cells that stably express HLA-DR (labelled as LV-HLA-DR), in the presence or absence of the indicated antibodies. Polykaryon formation was examined after exposure of cells to low pH. Representative images of n = 3 independent experiments are shown. Cell nuclei were stained by DAPI (blue). Scale bar, 25 μm. b, Visualization of H18 and HLA-DR complexes by PLA. MDCKII clone no. 2 cells (LV-ctrl) or MDCKII clone no. 2 cells that express HLA-DR (LV-HLA-DR) were infected with VSV-H18 at an MOI of 100 at 37 °C. After 1 h, cells were fixed and subjected to PLA using mouse anti-H18 and rabbit anti-MHC-II antibodies. Specific PLA dots obtained after amplification of the PLA fluorescence probe are indicated by red dots. Cell nuclei were stained by DAPI (blue colour). Representative images of three independent experiments are shown. Scale bars, 20 μm. c, Scatter plot of the PLA dot count per cell obtained from PLA images taken under the conditions given in c is shown from a representative of three independent experiments. The cell number, counted at 37 °C, for LV-ctrl cells infected with VSV-H18, LV-HLA-DR mock-infected cells or LV-HLA-DR cells infected with VSV-H18 was 9,254, 10,747 and 14,966, respectively. Statistical significance was assessed by Mann–Whitney U test (two-sided). ***P ≤ 2.2 × 10−16. d, MDCKII clone no. 2 cells (LV-ctrl) or MDCKII clone no. 2 cells that express HLA-DR (LV-HLA-DR–HA-tag) were infected with VSV-H18 at an MOI of 10. GFP-positive cells were quantified by flow cytometry at 24 h p.i. Data are mean ± s.d. from n = 3 independent experiments. Values below background levels of 1 are displayed on the x axes. e, Cartoon highlighting the principal steps of the emulsion coupling assay. Emulsion coupling is a digital assay concept based on the detection of double-labelled, individual molecular complexes in emulsion, which are identified by ddPCR. In a first step, MDCKII clone no. 2 cells that express HLA-DR that contains a haemagglutinin tag (HLA-DR–HA tag) were incubated with VSV-H18 at an MOI of 100 for 15 min at room temperature (A). To stabilize and prevent the dissociation of complexes in the following steps, the interaction was fixed with a membrane-impermeable cross-linker. Cells were lysed and the cell lysate was incubated with single-stranded-DNA-labelled antibodies (B) that specifically recognize HLA-DR–HA or H18, to achieve equilibrium binding (C). Before emulsification, the reaction was highly diluted (~100,000 times) to achieve single-complex separation. ddPCR was carried out using the standard ddPCR protocol, and the reactions were measured using a QX200 Droplet Digital PCR instrument (D). The evaluation of the reaction was based on the partitioning of the labels in the ddPCR reactions using fluorescently tagged PCR products (FAM- or VIC-labelled), and indicated as RFU (see scatter plot). According to the ddPCR standard evaluation, the number of labelled antibodies was determined in each reaction (counting all label-positive droplets for a given label, and using the same definition of clusters for all reactions). Without complexes (interacting proteins with two antibodies bound), the partitioning of the labelled antibodies follows a Poisson distribution, and results in a calculable number of double-coloured droplets. In the case of complexes present in the reaction, the number of the detected double-coloured droplets is larger than would be expected by Poisson distribution. We developed a Python code to calculate the number of complexes, which explains this difference on the basis of Poisson-based modelling of the partitioning of the molecular species of the reaction; this results in the absolute counts for the detected complexes.

Extended Data Fig. 7 HLA-DR homologues from different species confer susceptibility to bat IAV.

a, HEK293T cells were transfected with plasmids encoding CIITA, HLA-DRA or HLA-DRB1, and co-transfected with HLA-DRA and HLA-DRB1 or empty vector. At 48 h after transfection, cells were infected with VSV-H18 at an MOI of 10. Fluorescent microscopy images were taken at 72 h p.i. Representative images of n = 3 independent experiments are shown. Scale bar, 100 μm. b, c, HEK293T cells were co-transfected with plasmids encoding the α- and β-chains of HLA-DR homologues from different species: S. scrofa (SLA-DR), G. gallus (B-L), E. fuscus (E. fuscus DR), M. lucifugus (M. lucifugus DR) and P. alecto (P. alecto DR). HLA-DR was included as a positive control; cells transfected with empty vector served as a negative control. At 48 h after transfection, cells were infected with GFP encoding VSV-H18 (b) or VSV-H17 (c) at an MOI of 1. At 48 h (b) or 72 h (c) p.i., the frequency of GFP-positive cells was quantified by flow cytometry. d, CIITA from P. alecto (PaCIITA) was stably expressed by lentiviral transduction in kidney-derived cell lines from bat species S. lilium and P. alecto. Cells were infected with VSV-H18 at an MOI of 10 for at least 24 h. The frequency of GFP-positive cells was quantified by flow cytometry. bd, Data are mean ± s.d. from n = 3 independent experiments. Values below background levels of 1 are displayed on the x axes.

Extended Data Fig. 8 Alignment of protein sequences of surface MHC-II α-chains from different species.

Sequences included are the α-chains of MHC-II from Homo sapiens (HLA-DRA, HLA-DPA1 and HLA-DQA1), S. scrofa (SLA-DRA), G. gallus (B-LA), E. fuscus (EfDRA), M. lucifugus (MlDRA) and P. alecto (PaDRA). Residues conserved across all species are highlighted in red.

Extended Data Fig. 9 Alignment of protein sequences of surface MHC-II β-chains from different species.

Sequences included are the β-chains of MHC-II from H. sapiens (HLA-DRB1, HLA-DPB1 and HLA-DQB1), S. scrofa (SLA-DRB1), G. gallus (B-LB2), E. fuscus (EfDRB5), M. lucifugus (MlDRB5) and P. alecto (PaDRB). Residues conserved across all species are highlighted in red.

Extended Data Fig. 10 MHC-II knockout mice are resistant to bat IAV infection.

a, HEK293T cells were co-transfected with plasmids encoding the α- and β-chains of mouse MHC-II H-2A or H-2E or transfected with control plasmid. At 48 h after transfection, cells were infected with BlaM1 VLPs pseudotyped with H1N1 or H18 for 4 h at 37 °C. Entry-positive cells were measured with the fluorogenic β-lactamase substrate CCF2-AM and quantified by flow cytometry. b, HEK293T cells were co-transfected with plasmids encoding the α- and β-chains of mouse MHC-II H-2A or H-2E, or transfected with control plasmid. At 48 h p.i., cells were infected with VSV-H18 expressing GFP at an MOI of 10 for 48 h. Representative images are shown on the left, and a quantification of GFP-positive cells by flow cytometry is depicted on the right. a, b, Data are mean ± s.d. from n = 3 independent experiments. Values below background levels of 1 are displayed on the x axes. c, Detection of viral antigen in paraffin-embedded tissue from B6 mice intranasally infected with H18N11 (n = 4, 1 × 105 ffu in 40 μl, panels A and B) or H3N2 (n = 3, 1 × 103 pfu in 20 μl, panels C and D) or mock-infected (n = 2, panels E and F) at 4 days p.i. IAV matrix protein M1 immunoreactivity was observed in an oligofocal and laminar pattern in the apical ciliary border of the epithelium (arrow), and in epithelial cells (arrowhead), which displayed equivocal faint cytoplasmic and nuclear immunoreactivity (panel A). The inset (B) provides an overview of the epithelium (arrow) highlighted in panel A, and shows unspecific immunoreactivity in the lumen of a blood vessel in the lamina propria (arrowhead). Panel C shows intense matrix protein immunoreactivity in a multifocal-to-coalescing cytoplasmic pattern, occasionally with an apically increased intensity (arrow). Some nuclei within the immunoreactive cells are also labelled (arrowhead). The inset (D) provides an overview of the localization of the immunoreactive respiratory epithelium (arrow) on the cranial nasal septum in the area of the transition from transitional (left side) to respiratory (right side) epithelium. In mock-infected animals (panel E), respiratory epithelium never displayed immunoreactivity. Only faint unspecific immunoreactivity was observed in the lumen of a blood vessel in the lamina propria (arrow) in panel E. The inset (F) provides an overview of the epithelium highlighted in panel E. Scale bars, 10 μm (panels A, C, E), 50 μm (panels B, D, F). d, Detection of viral antigen and viral RNA in paraffin-embedded tissue of B6 mice intranasally infected with H3N2 (n = 3) for four days as described in c. A moderate amount of oligo- to multifocally distributed IAV matrix protein (panel A) but no H18-haemagglutinin-specific RNA (panel B) was detectable in the respiratory epithelium in following sections of the nasal cavities by using immunohistochemistry and in situ hybridization, respectively.

Supplementary information

Reporting Summary

Supplementary Table 1

Differentially expressed genes in bat IAV susceptible versus non-susceptible cell lines. Transcriptional profiles of MDCKII #1, Calu-3 and U-87MG cells susceptible to H18-pseudotyped VLP infection were compared to those of non-susceptible MDCKII #2, A549 and U-118MG cells, respectively. The two-groups comparison, each group consisting of n = 4 independent samples, was performed using the genewise negative binomial generalized linear model with quasi-likelihood tests (glmQLfit) implemented in EdgeR. Adjustments for multiple comparison were made by applying the Benjamini-Hochberg method. The indicated numbers represent genes in the H18N11-susceptible cell lines significantly (P ≤ 0.01) upregulated (log2 (fold change) ≥1) and filtered according to gene ontology annotations GO:0016020 (membrane), GO:0016021 (integral component of membrane) and GO:0005886 (plasma membrane). The 10 overlapping genes from MDCK #1, Calu-3 and U-87MG cells are listed.

Supplementary Table 2

Results of CRISPR screen. Cas9 expressing U-87MG cells transduced with a lentiviral library encoding genome-wide pools of single-guide RNAs (sgRNA) were infected with VSV-H18. sgRNA sequences from surviving cells were sequenced. Data analysis was performed by MAGeCK to identify enriched sgRNAs and genes were rank-ordered by robust rank aggregation (RRA) scores. Tab 1 contains positive selection summaries at the gene level. Tab 2 contains the sgRNA score summary. Tab 3 contains raw sgRNA counts. Tab 4 contains normalized sgRNA counts. The statistical significance of selection at the gene level was assessed using modified robust ranking aggregation (RRA), a permutation test implemented as part of the MAGeCK software suite. Corrections for multiple comparisons were made using the Benjamini-Hochberg method. The screen was performed once (n = 1).

Supplementary Table 3

siRNA sequences used to screen potential receptor candidates.

Supplementary Table 4

PCR primers used for Illumina sequencing.

Supplementary Table 5

Oligos used in Emulsion Coupling.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Karakus, U., Thamamongood, T., Ciminski, K. et al. MHC class II proteins mediate cross-species entry of bat influenza viruses. Nature 567, 109–112 (2019). https://doi.org/10.1038/s41586-019-0955-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-019-0955-3

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing