1. Academic Validation
  2. Direct activation of RIP3/MLKL-dependent necrosis by herpes simplex virus 1 (HSV-1) protein ICP6 triggers host antiviral defense

Direct activation of RIP3/MLKL-dependent necrosis by herpes simplex virus 1 (HSV-1) protein ICP6 triggers host antiviral defense

  • Proc Natl Acad Sci U S A. 2014 Oct 28;111(43):15438-43. doi: 10.1073/pnas.1412767111.
Xing Wang 1 Yun Li 1 Shan Liu 1 Xiaoliang Yu 1 Lin Li 2 Cuilin Shi 1 Wenhui He 2 Jun Li 1 Lei Xu 1 Zhilin Hu 1 Lu Yu 3 Zhongxu Yang 1 Qin Chen 1 Lin Ge 1 Zili Zhang 1 Biqi Zhou 1 Xuejun Jiang 4 She Chen 2 Sudan He 5
Affiliations

Affiliations

  • 1 Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, First Affiliated Hospital and Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215123, China;
  • 2 National Institute of Biological Sciences, Beijing 102206, China;
  • 3 Department of Emergency Medicine, First Affiliated Hospital, Soochow University, Suzhou 215006, China; and.
  • 4 Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065.
  • 5 Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, First Affiliated Hospital and Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215123, China; [email protected].
Abstract

The receptor-interacting kinase-3 (RIP3) and its downstream substrate Mixed Lineage Kinase domain-like protein (MLKL) have emerged as the key cellular components in programmed necrotic cell death. Receptors for the cytokines of tumor necrosis factor (TNF) family and Toll-like receptors (TLR) 3 and 4 are able to activate RIP3 through receptor-interacting kinase-1 and Toll/IL-1 receptor domain-containing adapter inducing IFN-β, respectively. This form of cell death has been implicated in the host-defense system. However, the molecular mechanisms that drive the activation of RIP3 by a variety of pathogens, other than the above-mentioned receptors, are largely unknown. Here, we report that human herpes simplex virus 1 (HSV-1) Infection triggers RIP3-dependent necrosis. This process requires MLKL but is independent of TNF Receptor, TLR3, cylindromatosis, and host RIP homotypic interaction motif-containing protein DNA-dependent activator of IFN regulatory factor. After HSV-1 Infection, the viral ribonucleotide reductase large subunit (ICP6) interacts with RIP3. The formation of the ICP6-RIP3 complex requires the RHIM domains of both proteins. An HSV-1 ICP6 deletion mutant failed to cause effective necrosis of HSV-1-infected cells. Furthermore, ectopic expression of ICP6, but not RHIM mutant ICP6, directly activated RIP3/MLKL-mediated necrosis. Mice lacking RIP3 exhibited severely impaired control of HSV-1 replication and pathogenesis. Therefore, this study reveals a previously uncharacterized host antipathogen mechanism.

Keywords

HSV-1; ICP6; MLKL; RIP3; programmed necrosis.

Figures