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:

MX2-mediated innate immunity against HIV-1 is regulated by serine phosphorylation

An Author Correction to this article was published on 13 August 2021

This article has been updated

Abstract

The antiviral cytokine interferon activates expression of interferon-stimulated genes to establish an antiviral state. Myxovirus resistance 2 (MX2, also known as MxB) is an interferon-stimulated gene that inhibits the nuclear import of HIV-1 and interacts with the viral capsid and cellular nuclear transport machinery. Here, we identified the myosin light chain phosphatase (MLCP) subunits myosin phosphatase target subunit 1 (MYPT1) and protein phosphatase 1 catalytic subunit-β (PPP1CB) as positively-acting regulators of MX2, interacting with its amino-terminal domain. We demonstrated that serine phosphorylation of the N-terminal domain at positions 14, 17 and 18 suppresses MX2 antiviral function, prevents interactions with the HIV-1 capsid and nuclear transport factors, and is reversed by MLCP. Notably, serine phosphorylation of the N-terminal domain also impedes MX2-mediated inhibition of nuclear import of cellular karyophilic cargo. We also found that interferon treatment reduces levels of phosphorylation at these serine residues and outline a homeostatic regulatory mechanism in which repression of MX2 by phosphorylation, together with MLCP-mediated dephosphorylation, balances the deleterious effects of MX2 on normal cell function with innate immunity against HIV-1.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: The NTD of MX2 interacts with MYPT1 and PPP1CB.
Fig. 2: MX2 requires functional MLCP for antiviral activity.
Fig. 3: IFN-induced MX2-mediated inhibition of HIV-1 requires MLCP.
Fig. 4: MX2 antiviral activity is antagonized by phosphorylation.
Fig. 5: Phosphorylation of MX2 reduces HIV-1 capsid binding and nuclear envelope accumulation.
Fig. 6: MX2-phosphorylation and nuclear import of cellular cargo.

Similar content being viewed by others

Data availability

The plasmids generated in this study are available upon request without restriction. All datasets generated in this study are available as source data. The mass spectrometry proteomics data have been deposited to the ProteomeXchange Consortium via the PRIDE49 partner repository with the dataset identifier PXD026073 (SILAC data) and PXD026090 and 10.6019/PXD026090 (phosphorylation data). Source data are provided with this paper.

Change history

References

  1. Doyle, T., Goujon, C. & Malim, M. H. HIV-1 and interferons: who’s interfering with whom? Nat. Rev. Microbiol. 13, 403–403 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Bourke, N. M. et al. Control of HIV infection by IFN-α: implications for latency and a cure. Cell. Mol. Life Sci. 75, 775–783 (2018).

    Article  CAS  PubMed  Google Scholar 

  3. Goujon, C. et al. Human MX2 is an interferon-induced post-entry inhibitor of HIV-1 infection. Nature 502, 559–562 (2013).

    Article  CAS  PubMed  Google Scholar 

  4. Kane, M. et al. MX2 is an interferon-induced inhibitor of HIV-1 infection. Nature 502, 563–566 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Liu, Z. et al. The interferon-inducible MxB protein inhibits HIV-1 infection. Cell Host Microbe 14, 398–410 (2013).

    Article  CAS  PubMed  Google Scholar 

  6. Crameri, M. et al. MxB is an interferon-induced restriction factor of human herpesviruses. Nat. Commun. 9, 1980–1980 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Schilling, M. et al. Human MxB protein is a pan-herpesvirus restriction factor. J. Virol. 92, 17 (2018).

    Article  Google Scholar 

  8. Staeheli, P. & Haller, O. Human MX2/MxB: a potent interferon-induced postentry inhibitor of herpesviruses and HIV-1. J. Virol. 92, 24 (2018).

    Article  Google Scholar 

  9. Jimenez-Guardeño, J. M., Apolonia, L., Betancor, G. & Malim, M. H. Immunoproteasome activation enables human TRIM5α restriction of HIV-1. Nat. Microbiol. 4, 933–940 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Wang, Y. X. et al. Interferon-inducible MX2 is a host restriction factor of hepatitis B virus replication. J. Hepatol. 72, 865–876 (2019).

    Article  PubMed  Google Scholar 

  11. Matreyek, K. A. et al. Host and viral determinants for MxB restriction of HIV-1 infection. Retrovirology 11, 90 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Busnadiego, I. et al. Host and viral determinants of Mx2 antiretroviral activity. J. Virol. 88, 7738–7752 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Bulli, L. et al. Complex interplay between HIV-1 capsid and MX2-independent alpha interferon-induced antiviral factors. J. Virol. 90, 7469–7480 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Staeheli, P., Haller, O., Boll, W., Lindenmann, J. & Weissmann, C. Mx protein: constitutive expression in 3T3 cells transformed with cloned Mx cDNA confers selective resistance to influenza virus. Cell 44, 147–158 (1986).

    Article  CAS  PubMed  Google Scholar 

  15. Gordien, E. et al. Inhibition of hepatitis B virus replication by the interferon-inducible MxA protein. J. Virol. 75, 2684–2691 (2002).

    Article  Google Scholar 

  16. Haller, O. & Kochs, G. Human MxA protein: an interferon-induced dynamin-like GTPase with broad antiviral activity. J. Interferon Cytokine Res. 31, 79–87 (2010).

    Article  PubMed  Google Scholar 

  17. Turan, K. et al. Nuclear MxA proteins form a complex with influenza virus NP and inhibit the transcription of the engineered influenza virus genome. Nucleic Acids Res. 32, 643–652 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Mitchell, P. S. et al. Evolution-guided identification of antiviral specificity determinants in the broadly acting interferon-induced innate immunity factor MxA. Cell Host Microbe 12, 598–604 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Mänz, B. et al. Pandemic influenza A viruses escape from restriction by human MxA through adaptive mutations in the nucleoprotein. PLoS Pathog. 9, 3 (2013).

    Article  Google Scholar 

  20. Patzina, C., Haller, O. & Kochs, G. Structural requirements for the antiviral activity of the human MxA protein against Thogoto and influenza A virus. J. Biol. Chem. 289, 6020–6027 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Goujon, C. et al. Transfer of the amino-terminal nuclear envelope targeting domain of human MX2 converts MX1 into an HIV-1 resistance factor. J. Virol. 88, 9017–9026 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Goujon, C., Greenbury, R. A., Papaioannou, S., Doyle, T. & Malim, M. H. A triple-arginine motif in the amino-terminal domain and oligomerization are required for HIV-1 inhibition by human MX2. J. Virol. 89, 4676–4680 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Fricke, T. et al. MxB binds to the HIV-1 core and prevents the uncoating process of HIV-1. Retrovirology 11, 68–68 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Dicks, M. D. J. et al. Oligomerization requirements for MX2-mediated suppression of HIV-1 infection. J. Virol. 90, 22–32 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Dicks, M. D. J. et al. Multiple components of the nuclear pore complex interact with the amino-terminus of MX2 to facilitate HIV-1 restriction. PLoS Pathog. 14, 1–24 (2018).

    Article  Google Scholar 

  26. Kane, M. et al. Nuclear pore heterogeneity influences HIV-1 infection and the antiviral activity of MX2. eLife 7, 1–44 (2018).

    Article  Google Scholar 

  27. Schulte, B. et al. Restriction of HIV-1 requires the N-terminal region of MxB as a capsid-binding motif but not as a nuclear localization signal. J. Virol. 89, 8599–8610 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Betancor, G. et al. The GTPase domain of MX2 interacts with the HIV-1 capsid, enabling its short isoform to moderate antiviral restriction. Cell Rep. 29, 1923–1933 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Smaga, S. S. et al. MxB restricts HIV-1 by targeting the tri-hexamer interface of the viral capsid. Structure 27, 1234–1245 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Buffone, C., Schulte, B., Opp, S. & Diaz-Griffero, F. Contribution of MxB oligomerization to HIV-1 capsid binding and restriction. J. Virol. 89, 3285–3294 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Kiss, A., Erdődi, F. & Lontay, B. Myosin phosphatase: unexpected functions of a long-known enzyme. Biochim. Biophys. Acta 1866, 2–15 (2019).

    Article  CAS  Google Scholar 

  32. Heroes, E. et al. The PP1 binding code: a molecular-lego strategy that governs specificity. FEBS J. 280, 584–595 (2013).

    Article  CAS  PubMed  Google Scholar 

  33. Wies, E. et al. Dephosphorylation of the RNA sensors RIG-I and MDA5 by the phosphatase PP1 is essential for innate immune signaling. Immunity 38, 437–449 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Ishihara, H. et al. Calyculin A and okadaic acid: inhibitors of protein phosphatase activity. Biochem. Biophys. Res. Commun. 159, 871–877 (1989).

    Article  CAS  PubMed  Google Scholar 

  35. Takai, A. et al. Inhibitory effect of okadaic acid derivatives on protein phosphatases. A study on structure-affinity relationship. Biochem. J. 284, 539–544 (2015).

    Article  Google Scholar 

  36. Ammosova, T. et al. Nuclear targeting of protein phosphatase-1 by HIV-1 Tat protein. J. Biol. Chem. 280, 36364–36371 (2005).

    Article  CAS  PubMed  Google Scholar 

  37. Wada, S. I. et al. Rubratoxin A specifically and potently inhibits protein phosphatase 2A and suppresses cancer metastasis. Cancer Sci. 101, 743–750 (2010).

    Article  CAS  PubMed  Google Scholar 

  38. Takai, A. et al. Protein phosphatases 1 and 2A and their naturally occurring inhibitors: current topics in smooth muscle physiology and chemical biology. J. Physiol. Sci. 68, 1–17 (2018).

    Article  CAS  PubMed  Google Scholar 

  39. Fribourgh, J. L. et al. Structural insight into HIV-1 restriction by MxB. Cell Host Microbe 16, 627–638 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Melén, K. et al. Human MxB protein, an interferon-α-inducible GTPase, contains a nuclear targeting signal and is localized in the heterochromatin region beneath the nuclear envelope. J. Biol. Chem. 271, 23478–23486 (1996).

    Article  PubMed  Google Scholar 

  41. Roy, J. & Cyert, M. S. Cracking the phosphatase code—docking interactions determine substrate specificity. Sci. Signal. 2, 100 (2009).

    Article  Google Scholar 

  42. Grassie, M. E., Moffat, L. D., Walsh, M. P. & MacDonald, J. A. A regulated mechanism for achieving substrate specificity of the catalytic subunit of protein phosphatase type 1δ. Arch. Biochem. Biophys. 510, 147–159 (2011).

    Article  CAS  PubMed  Google Scholar 

  43. Matsumura, F., Yamakita, Y. & Yamashiro, S. Myosin light chain kinases and phosphatase in mitosis and cytokinesis. Arch. Biochem. Biophys. 510, 76–82 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  46. Bolte, S. & Cordelières, F. P. A guided tour into subcellular colocalization analysis in light microscopy. J. Microsc. 224, 213–232 (2006).

    Article  CAS  PubMed  Google Scholar 

  47. Schaller, T. et al. Effects of inner nuclear membrane proteins SUN1/UNC-84A and SUN2/UNC-84B on the early steps of HIV-1 infection. J. Virol. 91, 19 (2017).

    Article  Google Scholar 

  48. Fouchier, R. A., Meyer, B. E., Simon, J. H., Fischer, U. & Malim, M. H. HIV-1 infection of non-dividing cells: evidence that the amino-terminal basic region of the viral matrix protein is important for Gag processing but not for post-entry nuclear import. EMBO J. 16, 4531–4539 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Perez-Riverol, Y. et al. The PRIDE database and related tools and resources in 2019: improving support for quantification data. Nucleic Acids Res. 47, D442–D450 (2019).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank D. Pollpeter, L. Ventimiglia, S. Papaioannou, P. Lehner, J. Cason, C. Mant, M. Kane and P. Bieniasz for the provision of reagents and helpful discussions. The work was supported by the Wellcome Trust (106223/Z/14/Z), the Medical Research Council (MR/M001199/1), the National Institutes of Health (U54 GM103368, AI150472) and the Department of Health via a National Institute for Health Research comprehensive Biomedical Research Centre award to Guy’s and St Thomas’ NHS Foundation Trust in partnership with King’s College London and King’s College Hospital NHS Foundation Trust. J.M.J.-G. is a long-term fellow of the European Molecular Biology Organization (ALTF 663-2016).

Author information

Authors and Affiliations

Authors

Contributions

G.B. and M.H.M. designed the study and wrote the manuscript with input from all co-authors; G.B. carried out the experiments and analysed the data, with contributions from A.S. in Fig. 1, M.D.J.D. in Fig. 2, J.M.J.-G. in Figs. 3 and 5 and Extended Data Fig. 2, H.K. in Fig. 4, S.L. in Fig. 1 and R.A. in Fig. 4; A.E.S., M.D.J.D. and J.M.J.-G. contributed to the execution of experiments and provided reagents; and M.H.M. supervised all aspects of the project.

Corresponding authors

Correspondence to Gilberto Betancor or Michael H. Malim.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

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

Extended data

Extended Data Fig. 1 Effect of MYPT1 and PPP1CB depletion on MX2-mediated inhibition of HIV-1.

a, Individual siRNAs targeting MYPT1 or PPP1CB, as well as a CTR siRNA were used to transfect U87-MG CD4+ CXCR4+ cells expressing MX2 or Luc. 48 h after challenging with HIV-1/GFP, infectivity was determined by flow cytometry (n = 4 biological replicates mean ± SD; two-tailed unpaired t-test). Immunoblot analyses to the right demonstrate the extents of reductions in protein levels. b, U87-MG CD4+ CXCR4+ cells were transduced with pEasiLV expressing Luc or MX2 and pretreated with doxycycline for 48 h before being doubly transfected with CTR siRNA or siRNAs targeting MYPT1 and/or PPP1CB. Levels of HIV-1/GFP infectivity were determined by flow cytometry at 48 h (n = 4 biological replicates, mean ± SD; two-tailed unpaired t-test). Immunoblot analyses to the right demonstrate the extents of reductions in protein levels.

Source data

Extended Data Fig. 2 Role of functional MLCP in viral infection.

a, U87-MG CD4+ CXCR4+ cells were doubly transduced with pEasiLV expressing Flag-tagged Luc or MX2 and also with pEasiLV-BFP expressing Flag-tagged Luc, MYPT1 or PPP1CB. Infectivity data with HIV-1/GFP are shown (above) and immunoblotting was used to determine the expression of each construct (below) (n = 6 biological replicates, mean ± SD; two-tailed unpaired t-test). b, 293T cells doubly transfected with pCAGGs plasmids expressing HA-tagged MX2 and either Flag-tagged MX1 or Flag-tagged MYPT1 were lysed, MX2 was immunoprecipitated using an anti-HA antibody and recovered proteins analyzed by immunoblot using anti-PPP1CB, anti-HA or anti-Flag antibodies. All experiments were done at least 4 times. c, Titration of calyculin A and okadaic acid in U87-MG CD4+ CXCR4+ expressing Luc or MX2 and challenged with MLV/GFP. The number of infected cells was determined by enumeration of GFP expressing cells by flow cytometry 48 h after challenging (n = 5 biological replicates for calyculin A; n = 3 biological replicates for okadaic acid, mean ± SD, (ns) non-significant; two-tailed unpaired t-test).

Source data

Extended Data Fig. 3 MX2, MYPT1 and PPP1CB do not participate in IFN-mediated suppression of MLV infection.

U87-MG CD4+ CXCR4+ cells depleted of MX2 using CRISPR-Cas9 genome editing (bulk population and clone MX2 CRISPR_2) and CTR CRISPR cells were transduced with pEasiLV expressing Luc or MX2, incubated in the presence or absence of 500 U/ml IFNα, and challenged with MLV/GFP. Infectivity was determined 48 h later by flow cytometry (n = 4 biological replicates, mean ± SD; two-tailed unpaired t-test).

Source data

Extended Data Fig. 4 Phosphorylation of MX2.

a, Example of LC-MSMS data from LysC generated MX2 peptides spanning A4 to K20, acquired on an Orbitrap Lumos and processed in PEAKS 8. The spectrum on the left shows the unphosphorylated form of the peptide and the spectrum on the right shows the corresponding peptide with putative phosphorylation at S14. b, 293T cells expressing C-terminally Flag-tagged MX2 were treated with 3 nM calyculin-A. After 6 h, cells were lysed, MX2 immunoprecipitated and the resulting protein digested with LysC. LC-MSMS data were processed in PEAKS 8. Sum peak areas for peptides spanning from M1 to K20 and covering serines 14, 17 and 18 were calculated using the areas under the curves, where the x-axis represents time and the y-axis represents intensity.

Source data

Extended Data Fig. 5 Phosphorylation of serines 14, 17 and 18 antagonizes the antiviral activity of MX2.

a, Residues S14, S17 and S18 in the N-terminal domain of the chimeras MX1 (NTDMX2) or Fv1b (NTDMX2) were substituted for alanine or aspartic acid and the effects on HIV-1/GFP infectivity measured at 48 h by flow cytometry (n = 4 biological replicates for Luc, MX2, MX1 (NTDMX2) and MX1 (NTDMX2) S14, 17-18A; n = 5 biological replicates for MX1 (NTDMX2) S14, 17-18D, Fv1b (NTDMX2), Fv1b (NTDMX2) S14, 17-18A and Fv1b (NTDMX2) S14, 17-18D, mean ± SD). Protein expression was confirmed by immunoblot. b, Residues 14, 17 and 18 of Flag-tagged MX2 were substituted for aspartic acid individually, in pairs or in triplicate. U87-MG CD4+ CXCR4+ cells expressing Luc, wild type or mutant proteins were challenged with HIV-1/GFP and infectivity was determined at 48 h by flow cytometry (n = 5 biological replicates, mean ± SD). Below, immunoblot showing protein expression.

Source data

Extended Data Fig. 6 Gating strategy for flow cytometry analysis of GFP+ infected cells.

a, Cells were distinguished from debris using the forward scatter (FCS, related to the cell size) and side scatter (SSC, related to the cell granularity). b, Selected population was gated for GFP+ (FITC channel) using uninfected cells as negative control. c, HIV-1/GFP infected cells were then gated for GFP. A representative plot showing GFP+ cells is shown.

Supplementary information

Source data

Source Data Fig. 1

Numerical source data.

Source Data Fig. 2

Numerical source data.

Source Data Fig. 3

Numerical source data.

Source Data Fig. 4

Numerical source data.

Source Data Fig. 5

Numerical source data.

Source Data Fig. 6

Numerical source data.

Source Data Extended Data Fig. 1

Numerical source data.

Source Data Extended Data Fig. 2

Numerical source data.

Source Data Extended Data Fig. 3

Numerical source data.

Source Data Extended Data Fig. 4

Numerical source data.

Source Data Extended Data Fig. 5

Numerical source data.

Source Data Fig. 1

Uncropped immunoblots.

Source Data Fig. 2

Uncropped immunoblots.

Source Data Fig. 3

Uncropped immunoblots.

Source Data Fig. 4

Uncropped immunoblots.

Source Data Fig. 5

Uncropped immunoblots.

Source Data Fig. 6

Uncropped immunoblots.

Source Data Extended Data Fig. 1

Uncropped immunoblots.

Source Data Extended Data Fig. 2

Uncropped immunoblots.

Source Data Extended Data Fig. 5

Uncropped immunoblots.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Betancor, G., Jimenez-Guardeño, J.M., Lynham, S. et al. MX2-mediated innate immunity against HIV-1 is regulated by serine phosphorylation. Nat Microbiol 6, 1031–1042 (2021). https://doi.org/10.1038/s41564-021-00937-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-021-00937-5

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