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.

  • Article
  • Published:

Dual inhibition of innate immunity and apoptosis by human cytomegalovirus protein UL37x1 enables efficient virus replication

Abstract

Immune evasion and inhibition of apoptosis are required for successful virus infection. However, inhibition of apoptosis can increase antiviral immune responses, which can then clear viral infections. Here we show that human cytomegalovirus (HCMV)-encoded UL37 exon-1 protein (UL37x1) not only inhibits apoptosis but also suppresses the cGAS-STING immune pathway. Using co-immunoprecipitation assays, we found that UL37x1 binds to TBK1 to abrogate the TBK1-STING-IRF3 interaction. Although the anti-apoptosis function of UL37x1 increases immune signalling, the immunosuppressive role of UL37x1 counteracts this undesirable side-effect. Furthermore, we used mutational analyses to show that the loss of either immunosuppressive or anti-apoptotic function of UL37x1 significantly reduced HCMV replication in human primary foreskin fibroblasts and humanized mice by over twofold. Finally, loss of both functions resulted in over fourfold reduction of HCMV replication in the same cell type and mouse model, showing that both UL37x1 functions are crucial for HCMV infection. We conclude that this sophisticated mechanism enables HCMV to control innate immunity and apoptosis to ensure efficient infection.

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: UL37x1 inhibits DNA-triggered innate immune signalling.
Fig. 2: UL37x1 interacts with TBK1 to abrogate TBK1-STING interaction.
Fig. 3: Lys40 of UL37x1 is required for targeting TBK1.
Fig. 4: UL37x1 inhibits HCMV-mediated immune responses.
Fig. 5: Immunosuppression by UL37x1 counterbalances apoptotic inhibition-induced immunity.
Fig. 6: UL37x1 functions are required for HCMV replication in cells and mice.

Similar content being viewed by others

Data availability

All data supporting the findings of this study are available within the Article and its Supplementary Information. The nucleotide sequences of HCMV strains mentioned in this study are publicly available on NCBI GenBank AD169 (FJ527563.1) and Towne (GQ121041.1). Source data are provided with this paper.

References

  1. Cohen, Y. & Stern-Ginossar, N. Manipulation of host pathways by human cytomegalovirus: insights from genome-wide studies. Semin. Immunopathol. 36, 651–658 (2014).

    Article  CAS  PubMed  Google Scholar 

  2. Wang, Y. Q. & Zhao, X. Y. Human cytomegalovirus primary infection and reactivation: insights from virion-carried molecules. Front. Microbiol. 11, 1511 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Liu, Y. et al. A cytomegalovirus peptide-specific antibody alters natural killer cell homeostasis and is shared in several autoimmune diseases. Cell Host Microbe 19, 400–408 (2016).

    Article  CAS  PubMed  Google Scholar 

  4. Manicklal, S., Emery, V. C., Lazzarotto, T., Boppana, S. B. & Gupta, R. K. The “silent” global burden of congenital cytomegalovirus. Clin. Microbiol. Rev. 26, 86–102 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Sun, L., Wu, J., Du, F., Chen, X. & Chen, Z. J. Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway. Science 339, 786–791 (2013).

    Article  CAS  PubMed  Google Scholar 

  6. Zhong, B. et al. The adaptor protein MITA links virus-sensing receptors to IRF3 transcription factor activation. Immunity 29, 538–550 (2008).

    Article  CAS  PubMed  Google Scholar 

  7. Sun, W. et al. ERIS, an endoplasmic reticulum IFN stimulator, activates innate immune signaling through dimerization. Proc. Natl Acad. Sci. USA 106, 8653–8658 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Ishikawa, H. & Barber, G. N. STING is an endoplasmic reticulum adaptor that facilitates innate immune signalling. Nature 455, 674–678 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Hopfner, K. P. & Hornung, V. Molecular mechanisms and cellular functions of cGAS-STING signalling. Nat. Rev. Mol. Cell Biol. 21, 501–521 (2020).

    CAS  Google Scholar 

  10. Zhang, X., Bai, X. C. & Chen, Z. J. Structures and mechanisms in the cGAS-STING innate immunity pathway. Immunity 53, 43–53 (2020).

    Article  CAS  PubMed  Google Scholar 

  11. Ma, Z., Ni, G. & Damania, B. Innate sensing of DNA virus genomes. Annu. Rev. Virol. 5, 341–362 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Wills, M. R., Poole, E., Lau, B., Krishna, B. & Sinclair, J. H. The immunology of human cytomegalovirus latency: could latent infection be cleared by novel immunotherapeutic strategies? Cell. Mol. Immunol. 12, 128–138 (2015).

    Article  CAS  PubMed  Google Scholar 

  13. Crough, T. & Khanna, R. Immunobiology of human cytomegalovirus: from bench to bedside. Clin. Microbiol. Rev. 22, 76–98 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Paijo, J. et al. cGAS senses human cytomegalovirus and induces type I interferon responses in human monocyte-derived cells. PLoS Pathog. 12, e1005546 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Fu, Y. Z. et al. Human cytomegalovirus tegument protein UL82 Inhibits STING-mediated signaling to evade antiviral immunity. Cell Host Microbe 21, 231–243 (2017).

    Article  CAS  PubMed  Google Scholar 

  16. Huang, Z. F. et al. Human cytomegalovirus protein UL31 inhibits DNA sensing of cGAS to mediate immune evasion. Cell Host Microbe 24, 69–80.e4 (2018).

    Article  CAS  PubMed  Google Scholar 

  17. Biolatti, M. et al. Human Cytomegalovirus Tegument Protein pp65 (pUL83) Dampens Type I Interferon Production by Inactivating the DNA Sensor cGAS without Affecting STING. J. Virol. 92, e01774-17 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Fu, Y. Z. et al. Human cytomegalovirus DNA polymerase subunit UL44 antagonizes antiviral immune responses by suppressing IRF3- and NF-κB-mediated transcription. J. Virol. 93, e00181-19 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Zou, H. M. et al. Human cytomegalovirus protein UL94 targets MITA to evade the antiviral immune response. J. Virol. 94, e00022-20 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Fu, Y. Z. et al. Human cytomegalovirus protein UL42 antagonizes cGAS/MITA-mediated innate antiviral response. PLoS Pathog. 15, e1007691 (2019).

  21. Everett, H. & McFadden, G. Apoptosis: an innate immune response to virus infection. Trends Microbiol. 7, 160–165 (1999).

    Article  CAS  PubMed  Google Scholar 

  22. Orzalli, M. H. & Kagan, J. C. Apoptosis and necroptosis as host defense strategies to prevent viral infection. Trends Cell Biol. 27, 800–809 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Luo, B. et al. Erythropoeitin signaling in macrophages promotes dying cell clearance and immune tolerance. Immunity 44, 287–302 (2016).

    Article  CAS  PubMed  Google Scholar 

  24. McGaha, T. L. & Karlsson, M. C. Apoptotic cell responses in the splenic marginal zone: a paradigm for immunologic reactions to apoptotic antigens with implications for autoimmunity. Immunol. Rev. 269, 26–43 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. White, M. J. et al. Apoptotic caspases suppress mtDNA-induced STING-mediated type I IFN production. Cell 159, 1549–1562 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Ning, X. et al. Apoptotic caspases suppress type I interferon production via the cleavage of cGAS, MAVS, and IRF3. Mol. Cell 74, 19–31.e7 (2019).

    Article  CAS  PubMed  Google Scholar 

  27. Peppenelli, M. A., Arend, K. C., Cojohari, O., Moorman, N. J. & Chan, G. C. Human cytomegalovirus stimulates the synthesis of select Akt-dependent antiapoptotic proteins during viral entry to promote survival of infected monocytes. J. Virol. 90, 3138–3147 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Reeves, M. B., Breidenstein, A. & Compton, T. Human cytomegalovirus activation of ERK and myeloid cell leukemia-1 protein correlates with survival of latently infected cells. Proc. Natl Acad. Sci. USA 109, 588–593 (2012).

    Article  CAS  PubMed  Google Scholar 

  29. Goldmacher, V. S. vMIA, a viral inhibitor of apoptosis targeting mitochondria. Biochimie 84, 177–185 (2002).

    Article  CAS  PubMed  Google Scholar 

  30. McCormick, A. L., Meiering, C. D., Smith, G. B. & Mocarski, E. S. Mitochondrial cell death suppressors carried by human and murine cytomegalovirus confer resistance to proteasome inhibitor-induced apoptosis. J. Virol. 79, 12205–12217 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Zhang, A., Hildreth, R. L. & Colberg-Poley, A. M. Human cytomegalovirus inhibits apoptosis by proteasome-mediated degradation of Bax at endoplasmic reticulum-mitochondrion contacts. J. Virol. 87, 5657–5668 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Skaletskaya, A. et al. A cytomegalovirus-encoded inhibitor of apoptosis that suppresses caspase-8 activation. Proc. Natl Acad. Sci. USA 98, 7829–7834 (2001).

    CAS  Google Scholar 

  33. Hayajneh, W. A. et al. The sequence and antiapoptotic functional domains of the human cytomegalovirus UL37 exon 1 immediate early protein are conserved in multiple primary strains. Virology 279, 233–240 (2001).

    Article  CAS  PubMed  Google Scholar 

  34. Zhao, B. et al. A conserved PLPLRT/SD motif of STING mediates the recruitment and activation of TBK1. Nature 569, 718–722 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Zhang, C. et al. Structural basis of STING binding with and phosphorylation by TBK1. Nature 567, 394–398 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Mavinakere, M. S. & Colberg-Poley, A. M. Dual targeting of the human cytomegalovirus UL37 exon 1 protein during permissive infection. J. Gen. Virol. 85, 323–329 (2004).

    Article  CAS  PubMed  Google Scholar 

  37. Griffante, G. et al. Human cytomegalovirus-induced host protein citrullination is crucial for viral replication. Nat. Commun. 12, 3910 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Mocarski, E. S. Jr. Biology and replication of cytomegalovirus. Transfus. Med. Rev. 2, 229–234 (1988).

    Article  PubMed  Google Scholar 

  39. Smith, M. S. et al. Granulocyte-colony stimulating factor reactivates human cytomegalovirus in a latently infected humanized mouse model. Cell Host Microbe 8, 284–291 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Bravo, F. J., Cardin, R. D. & Bernstein, D. I. A model of human cytomegalovirus infection in severe combined immunodeficient mice. Antivir. Res. 76, 104–110 (2007).

    Article  CAS  PubMed  Google Scholar 

  41. Biolatti, M. et al. Regulatory interaction between the cellular restriction factor IFI16 and viral pp65 (pUL83) modulates viral gene expression and IFI16 protein stability. J. Virol. 90, 8238–8250 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. You, Y. et al. The suppression of apoptosis by alpha-herpesvirus. Cell Death Dis. 8, e2749 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Wang, Z. et al. A picorna-like virus suppresses the N-end rule pathway to inhibit apoptosis. eLife 6, e30590 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Pan, Y., Cheng, A., Wang, M., Yin, Z. & Jia, R. The dual regulation of apoptosis by flavivirus. Front. Microbiol. 12, 654494 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Lai, Y. et al. Regulation of apoptosis by enteroviruses. Front. Microbiol. 11, 1145 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Wasilenko, S. T., Meyers, A. F., Vander Helm, K. & Barry, M. Vaccinia virus infection disarms the mitochondrion-mediated pathway of the apoptotic cascade by modulating the permeability transition pore. J. Virol. 75, 11437–11448 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Shu, T. et al. SARS-Coronavirus-2 Nsp13 possesses NTPase and RNA helicase activities that can be inhibited by bismuth salts. Virol. Sin. 35, 321–329 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Lu, B. et al. Induction of INKIT by viral infection negatively regulates antiviral responses through inhibiting phosphorylation of p65 and IRF3. Cell Host Microbe 22, 86–98.e4 (2017).

    Article  CAS  PubMed  Google Scholar 

  49. Yu, D., Smith, G. A., Enquist, L. W. & Shenk, T. Construction of a self-excisable bacterial artificial chromosome containing the human cytomegalovirus genome and mutagenesis of the diploid TRL/IRL13 gene. J. Virol. 76, 2316–2328 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Tischer, B. K., Smith, G. A. & Osterrieder, N. En passant mutagenesis: a two step markerless red recombination system. Methods Mol. Biol. 634, 421–430 (2010).

    Article  CAS  PubMed  Google Scholar 

  51. Han, Y. et al. Feasibility study of mixing throat swab samples for severe acute respiratory syndrome coronavirus-2 screening. Virol. Sin. 35, 830–832 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Ren, Y. et al. The type I interferon-IRF7 axis mediates transcriptional expression of Usp25 gene. J. Biol. Chem. 291, 13206–13215 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Ren, Y. et al. The ORF3a protein of SARS-CoV-2 induces apoptosis in cells. Cell. Mol. Immunol. 17, 881–883 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank Y. Fu, Y. Wang and B. Zhong (Wuhan, China) for reagents.

This work was supported by the National Natural Science Foundation of China (NSFC) (31970169 to X.Z. and 32100106 to Y.R.), Strategic Priority Research Program of CAS (XDB29010300 to X.Z.), National Key R&D Program of China (2021YFC2300700 to X.Z.) and NSFC (U21A20423 to X.Z. and 81873964 to Y.Q.); a grant from the CAS Youth Innovation Promotion Association (2020332 to Y.Q.), the Hubei Province Natural Science Funds for Distinguished Young Scholars (2021CFA047 to Y.Q.) and the Young Top-notch Talent Cultivation Program of Hubei Province (Y.Q.).

Author information

Authors and Affiliations

Authors

Contributions

Y.R. performed most of the experiments. A.W., D.W., C.W., M.H., X.X. and L.J. performed specific experiments. Y.R. and X.Z. conceived the hypothesis. Y.R., W.Z., Y.Q. and X.Z. designed the experiments and analysed the data. Y.R., Y.Q. and X.Z. wrote the manuscript with inputs from all authors. X.Z. supervised the study.

Corresponding authors

Correspondence to Wei Zhou, Yang Qiu or Xi Zhou.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Microbiology thanks the anonymous reviewers for their contribution to the peer review of this work. Peer reviewer reports are available.

Additional information

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

Extended data

Extended Data Fig. 1 Sequence comparison of UL37x1 proteins.

The amino acid sequence alignment of UL37x1 proteins of different HCMV strains.

Extended Data Fig. 2 UL37x1 inhibits the DNA-triggered innate immune signaling in 293 T cells.

(a) Luciferase reporter assay analyzing IFNβ, ISRE, or NF-κB promoter activity was conducted in 293 T cells transfected with the plasmid of FLAG-cGAS plus FLAG-STING or an empty vector together with the indicated amounts of FLAG-UL37x1 expression plasmid for 24 h. The lower blots showed the expression levels of these transfected proteins. (b) 293 T cells were transfected with the plasmid of FLAG-cGAS plus FLAG-STING or an empty vector together with the indicated amounts of FLAG-UL37x1 expression plasmid for 24 h, followed by qPCR of the indicated antiviral genes. The lower blots showed the expression levels of these transfected proteins. (c) 293 T cells stably expressing STING were transfected with the plasmid of FLAG-UL37x1 or an empty vector. After 24 h, cells were untreated (mock) or transfected with indicated DNA ligands (HSV60, DNA90, or HSV120). At 3 and 6 h post transfection (h.p.t.), total RNA was extracted and subject to qPCR of indicated antiviral genes. Graphs show mean ± SD (n = 3, biologically independent experiments). Statistical significance was determined by two-way ANOVA. Immunoblots are representative of three independent experiments.

Source data

Extended Data Fig. 3 UL37x1 inhibits TBK1-mediated immune signaling.

(a) 293 T cells were transfected with the indicated plasmid in the presence or absence of the plasmid of FLAG-UL37x1. At 24 h.p.t., qRT-PCR was performed to measure the transcription of indicated antiviral genes. (b) HFFs were infected with HCMV for indicated time, followed by qPCR analysis of UL37x1. (c-d) HFFs were transfected with siRNA specifically for UL37x1 (UL37x1-siRNA#1, UL37x1-siRNA#2, or UL37x1-siRNA#3) or a control siRNA (Control). After 24 h, cells were infected with HCMV. At 6 h.p.i., qPCR was performed to measure the transcription of UL37x1 (c). At 12 h.p.i., cell lysates were subject to immunoblots with indicated antibodies (d). Immunoblots are representative of three independent experiments. Graphs show mean ± SD (n = 3, biologically independent experiments). Statistical significance was determined by two-way ANOVA in a, or one-way ANOVA in c.

Source data

Extended Data Fig. 4 Identifying the key residues for UL37x1 inhibiting cGAS-STING axis.

(a-b) Upper, the schematic illustration of the UL37x1 truncations. Middle, 293 T cells were transfected with the plasmid of FLAG-cGAS plus FLAG-STING together with the plasmid of FLAG-UL37x1 or indicated truncation for 24 h, followed by qPCR analysis of IFNB1. The lower blots showed the expression levels of the indicated proteins. (c) 293 T cells were transfected with the plasmid of FLAG-UL37x1, FLAG-UL37x140-44, or FLAG-UL37x1K40R together with the plasmid of FLAG-TBK1, FLAG-IRF3-5D, or an empty vector. At 24 h.p.t., qPCR was performed to measure the transcription of indicated antiviral genes. (d) 293 T cells were transfected with the plasmid of HA-TBK1 together with the plasmid of FLAG-UL37x1 or FLAG-UL37x140-44. At 24 h.p.t., cell lysates were subject to immunoprecipitation with anti-FLAG antibody or IgG, followed by immunoblots with indicated antibodies. (e) HFFs stably expressing UL37x1, UL37x1K40R, or empty vector were transfected with HA-MAVS, Myc-TBK1. At 24 h.p.i., cell lysates were subject to immunoprecipitation with anti-HA antibody or IgG, followed by immunoblots with indicated antibodies. Immunoblots are representative of three independent experiments. (f) Luciferase reporter assay analyzing IFNβ, ISRE, or NF-κB promoter activity was conducted in 293 T cells transfected with or without the plasmid of FLAG-RIG-I CARD together with the plasmid of FLAG-UL37x1 or FLAG-UL37x1K40R for 24 h. (g) 293 T cells were transfected with or without the plasmid of FLAG-RIG-I CARD together with the plasmids of FLAG-UL37x1 or FLAG-UL37x1K40R for 24 h. Total RNAs were extracted and subject to qPCR of the indicated antiviral genes. Graphs show mean ± SD (n = 3, biologically independent experiments). Statistical significance was determined by two-way ANOVA.

Source data

Extended Data Fig. 5 The effects of UL37x1 on immune signaling and apoptosis.

(a) HFFs stably expressing UL37x1 or empty vector were infected with HCMV (MOI = 1). At 0, 6, and 12 h.p.i., qPCR was performed to measure the transcription of indicated antiviral genes. (b) HFFs stably expressing UL37x1 or empty vector were uninfected (mock) or infected with HCMV in the absence or presence of z-VAD-FMK. At 12 h.p.i., cells were collected and stained with Annexin V-FITC/PI for flow cytometry analysis (left, early apoptosis: Annexin V-FITC single positive, Q3; late apoptosis: Annexin V-FITC and PI double positive, Q2), and the percentage of apoptotic cells (Q2 + Q3) was measured (right). This gating strategy applies to all flow cytometry analyses in this study. (c) 293 T cells were transfected with the plasmid of FLAG-cGAS plus FLAG-STING or an empty vector together with or without the plasmid of FLAG-UL37x1. After 24 h, cells were collected and stained with Annexin V-FITC/PI for flow cytometry analysis, and the percentage of apoptotic cells was measured. (d) HFFs were transfected with HSV60, DNA90, or HSV120. After 12 h, cells were collected and stained with Annexin V-FITC/ PI for flow cytometry analysis, and the percentage of apoptotic cells was measured. (e-f) HFFs were transfected with UL37x1-siRNA#1, UL37x1-siRNA#2, UL37x1-siRNA#3 or control siRNA for 24 h, followed by infection with HCMV (MOI = 1). At 12 h.p.i., the mRNA levels of the indicated HCMV genes were measured by qPCR (e) and the protein level of HCMV-expressed reporter GFP was measured by immunoblot (f). (g) HFFs were transfected as in (b) for 24 h, followed by infection with HCMVTowne (MOI = 1). At 12 h.p.i., cell lysate was subject to immunoblot with anti-UL37x1 and anti-β-Actin. (h) HFFs were transfected with UL37x1-siRNA#1, UL37x1-siRNA#3 or control siRNA and infected with HCMVTowne (MOI = 1) in the absence or presence of z-VAD-FMK. At 12 h.p.i., qPCR was performed to measure the transcription of indicated antiviral genes. Immunoblots are representative of three independent experiments. Graphs show mean ± SD (n = 3 for a, e, and h, or n = 2, for b, c, and d biologically independent experiments). Statistical significance was determined by two-way ANOVA in a, b, c, d, and h, or one-way ANOVA in e.

Source data

Extended Data Fig. 6 The immunosuppressive and anti-apoptotic activities of UL37x1 are functionally separated.

(a) 293 T cells were transfected with the plasmid of FLAG-UL37x1 or FLAG-UL37x12-23. At 24 h.p.t., cells were stained with Mito Tracker Red (mitochondria staining reagent) at 37 °C. After 15 min, cells were fixed and stained with anti-FLAG and Alexa-488 conjugated anti-mouse IgG antibodies, followed by immunofluorescence microscopy. Scale bar, 10 μM. (b) 293 T cells were transfected with the plasmid of FLAG-UL37x1 or FLAG-UL37x12-23. At 24 h.p.t., cells were collected and the mitochondria was separated via gradient centrifugation. The indicated cell lysate fractions were subject to immunoblots with anti-FLAG, anti-β-Actin, and anti-GDH (glutamate dehydrogenase, used to mark mitochondria) antibodies. Immunoblots are representative of three independent experiments. (c) HFFs stably expressing UL37x1, UL37x12-23, or empty vector were uninfected or infected with HCMV (MOI = 1). At 12 h.p.i., cells were collected and stained with Annexin V-FITC/ PI for flow cytometry analysis, and the percentage of apoptotic cells was measured. (d) HFFs stably expressing FLAG-UL37x1, FLAG-UL37x140-44, FLAG-UL37x1K40R, or empty vector were uninfected or infected with HCMV (MOI = 1). At 12 h.p.i., cells were collected and stained with Annexin V-FITC/ PI for flow cytometry analysis, and the percentage of apoptotic cells was measured. Graphs show mean ± SD (n = 2, biologically independent experiments). Statistical significance was determined by two-way ANOVA.

Source data

Extended Data Fig. 7 A scheme for homologous recombination mediated genome editing of the UL37x1 gene locus.

(a, c and e) A scheme for homologous recombination mediated genome editing of the UL37x1 gene locus. (b, d and f) DNA sequence and reading frame of wild-type and edited UL37x1 alleles. (g) HFFs were infected with HCMVWT, HCMVUL37x1, HCMV2-23, or HCMVK40R. At 12 h.p.i., cell lysates were subject to immunoblots with anti-UL37x1 and anti-UL36 antibodies. Immunoblots are representative of two independent experiments.

Source data

Extended Data Fig. 8 Endogenous UL37x1 inhibits innate immunity and apoptosis.

(a) HFFs were infected with HCMVWT, HCMVUL37x1, HCMV2-23, or HCMVK40R (MOI = 1) in the presence of z-VAD-FMK. At 12 h.p.i., cells were collected and stained with Annexin V-FITC/PI for flow cytometry analysis, and the percentage of apoptotic cells was measured. (b-c) HFFs were infected with HCMVWT, HCMVUL37x1, HCMV2-23, or HCMVK40R (MOI = 1) in the presence of z-VAD-FMK. At 0, 6, and 12 h.p.i., total RNA were extracted and cell lysates were prepared, followed by qPCR to measure the transcription of indicated antiviral genes (b), and immunoprecipitation with anti-STING antibody followed by immunoblots with indicated antibodies (c), respectively. Immunoblots are representative of three independent experiments. (d-e) HFFs stably expressing UL37x1, UL37x12-23, UL37x1K40R, or empty vector were infected with HCMVUL37x1 (MOI = 1). At 96 h.p.i., GFP signals were analyzed by fluorescence microscopy (d, left). The percentage of the infected cells (GFP positive) were measured and shown as the graph (d, right). The mRNA levels of the indicated HCMV genes in HFFs were measured by qPCR at 96 h.p.i (e). (f-g) STING-/- or cGAS-/- HFFs were infected with HCMVWT, HCMVUL37x1, HCMV2-23, or HCMVK40R. At 96 h.p.i., the mRNA levels of the indicated HCMV genes in HFFs were measured by qPCR. (h) HFFs were infected with HCMVWT, HCMVUL37x1, HCMV2-23, or HCMVK40R in the presence of z-VAD-FMK. At 96 h.p.i., the mRNA levels of the indicated HCMV genes in HFFs were measured by qPCR. Graphs show mean ± SD (n = 2 for a, or n = 3 for b and e-h, biologically independent experiments). Statistical significance was determined by one-way ANOVA in a and e-h, or two-way ANOVA in b.

Source data

Extended Data Fig. 9 Humanized mouse model for HCMV infection.

(a) The experimental workflow of in vivo HCMV infection experiment in humanized mice: NOD-scid IL2RGγcnull mice were engrafted with human CD34+ HPCs and then transfused with NHDFs infected with HCMVWT, HCMVUL37x1, HCMV2-23 or HCMVK40R (n = 6/group). Uninfected mice served as the negative control group (n = 6). At 28 days post infection, in each group, three mice were euthanized for tissue harvesting and three mice were treated with G-CSF and AMD-3100 for another 10 days and then euthanized for tissue harvesting and analysis. (b-c) NHDF cells were infected with wild-type or mutant HCMV. At 12 h.p.i., the mRNA levels of the indicated HCMV genes in NHDFs were measured by qPCR (b) and the protein level of HCMV-expressed reporter GFP was measured by immunoblot (c). Immunoblots are representative of three independent experiments. Graphs in b show mean ± SD (n = 3, biologically independent experiments). Statistical significance was determined by one-way ANOVA.

Source data

Extended Data Fig. 10 Model for the dual inhibition of innate immunity and apoptosis by HCMV UL37x1.

Left: Apoptotic caspases cleave key components of the cGAS-STING axis to inhibit innate immune signaling. Right: During HCMV infection, UL37x1 inhibits apoptosis, which enhances cGAS-STING immune signaling. On the other hand, UL37x1 can directly target to TBK1 to disrupt TBK1 interaction with STING as well as IRF3 recruitment to STING, thereby suppressing the cGAS-STING pathway. This immunosuppressing activity of UL37x1 counterbalances the immune enhancing “side-effect” of UL37x1’s anti-apoptotic activity.

Supplementary information

Source data

Source Data Fig. 1

Unprocessed western blots.

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Unprocessed western blots.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Unprocessed western blots.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Unprocessed western blots.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Unprocessed western blots.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Unprocessed western blots.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 2

Unprocessed western blots.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Unprocessed western blots.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Unprocessed western blots.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Unprocessed western blots.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Unprocessed western blots.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Unprocessed western blots.

Source Data Extended Data Fig. 8

Unprocessed western blots.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Unprocessed western blots.

Source Data Extended Data Fig. 9

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ren, Y., Wang, A., Wu, D. et al. Dual inhibition of innate immunity and apoptosis by human cytomegalovirus protein UL37x1 enables efficient virus replication. Nat Microbiol 7, 1041–1053 (2022). https://doi.org/10.1038/s41564-022-01136-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-022-01136-6

This article is cited by

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