Abstract

In this article we report that the M2 protein encoded by the vaccinia virus is secreted as a homo-oligomer by infected cells and binds two central costimulation molecules, CD80 (B7-1) and CD86 (B7-2). These interactions block the ligation of the two B7 proteins to both soluble CD28 and soluble cytotoxic T-lymphocyte associated protein 4 (CTLA4) but favor the binding of soluble PD-L1 to soluble CD80. M2L gene orthologues are found in several other poxviruses, and the B7-CD28/CTLA4 blocking activity has been identified for several culture supernatants of orthopoxvirus-infected cells and for a recombinant myxoma virus M2 protein homolog (i.e., Gp120-like protein, or Gp120LP). Overall, these data indicate that the M2 poxvirus family of proteins may be involved in immunosuppressive activities broader than the NF-κB inhibition already reported (R. Gedey, X. L. Jin, O. Hinthong, and J. L. Shisler, J Virol 80:8676-8685, 2006, https://doi.org/10.1128/JVI.00935-06). A Copenhagen vaccinia virus with a deletion of the nonessential M2L locus was generated and compared with its parental virus. This M2L-deleted vaccinia virus, unlike the parental virus, does not generate interference with the B7-CD28/CTLA4/PD-L1 interactions. Moreover, this deletion did not affect any key features of the virus (in vitro replication, oncolytic activities in vitro and in vivo, and intratumoral expression of a transgene in an immunocompetent murine model). Altogether, these first results suggest that the M2 protein has the potential to be used as a new immunosuppressive biotherapeutic and that the M2L-deleted vaccinia virus represents an attractive new oncolytic platform with an improved immunological profile.IMPORTANCE The vaccinia virus harbors in its genome several genes dedicated to the inhibition of the host immune response. Among them, M2L was reported to inhibit the intracellular NF-κB pathway. We report here several new putative immunosuppressive activities of M2 protein. M2 protein is secreted and binds cornerstone costimulatory molecules (CD80/CD86). M2 binding to CD80/CD86 blocks their interaction with soluble CD28/CTLA4 but also favors the soluble PD-L1-CD80 association. These findings open the way for new investigations deciphering the immune system effects of soluble M2 protein. Moreover, a vaccinia virus with a deletion of its M2L has been generated and characterized as a new oncolytic platform. The replication and oncolytic activities of the M2L-deleted vaccinia virus are indistinguishable from those of the parental virus. More investigations are needed to characterize in detail the immune response triggered against both the tumor and the virus by this M2-defective vaccinia virus.

Highlights

  • IMPORTANCE The vaccinia virus harbors in its genome several genes dedicated to the inhibition of the host immune response

  • In this article we show that the vaccinia virus M2 protein is secreted by infected cells as a homo-oligomer that interacts with CD80 and CD86 of at least human and murine origins

  • The M2 binding to CD80 and CD86 inhibits their interaction with both soluble CD28 and cytotoxic T-lymphocyte associated protein 4 (CTLA4), their cognate ligands

Read more

Summary

Introduction

IMPORTANCE The vaccinia virus harbors in its genome several genes dedicated to the inhibition of the host immune response. About 40 out of the 250 genes of the vaccinia virus genome are well known to encode immunosuppressors that contribute altogether to hamper the antiviral immune response [13] These immunosuppressors target a wide variety of pathways by interacting with both intracellular (several NF-␬B activation inhibitors) and extracellular (e.g., type I/II interferon decoy receptors) central immune mediators [13, 14]. Several of these immunosuppressor genes have been eliminated during the generation of the highly immunogenic, nononcolytic, modified vaccinia virus Ankara (MVA) that is widely used as a prophylactic or therapeutic vaccine platform to fight infectious diseases and cancers. A vaccinia virus with a triple deletion (TD) of thymidine kinase, ribonucleotide reductase, and M2L (TKϪ RRϪ M2LϪ vaccinia virus, or VV-TD) was generated and characterized for its in vitro and in vivo replication and oncolytic activities

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call