Varicella zoster virus encodes a viral decoy RHIM to inhibit cell death

Herpesviruses are known to encode a number of inhibitors of host cell death, including Rip Homotypic Interaction Motif (RHIM)-containing proteins. Varicella zoster virus (VZV) is a member of the alphaherpesvirus subfamily and is responsible for causing chickenpox and shingles. We have identified a novel viral RHIM in the VZV capsid triplex protein open reading frame (ORF) 20 that acts as a host cell death inhibitor. Like the human cellular RHIMs in RIPK1 and RIPK3 that stabilise the necrosome in TNF-induced necroptosis, and the viral RHIM in M45 from murine cytomegalovirus that inhibits cell death, the ORF20 RHIM is capable of forming fibrillar functional amyloid complexes. Notably, the ORF20 RHIM forms hybrid amyloid complexes with human ZBP1, a cytoplasmic sensor of viral nucleic acid. Although VZV can inhibit TNF-induced necroptosis, the ORF20 RHIM does not appear to be responsible for this inhibition. In contrast, the ZBP1 pathway is identified as important for VZV infection. Mutation of the ORF20 RHIM renders the virus incapable of efficient spread in ZBP1-expressing HT-29 cells, an effect which can be reversed by the inhibition of caspases. Therefore we conclude that the VZV ORF20 RHIM is important for preventing ZBP1-driven apoptosis during VZV infection, and propose that it mediates this effect by sequestering ZBP1 into decoy amyloid assemblies. Author Summary Rip homotypic interaction motifs (RHIMs) are found in host proteins that can signal for programmed cell death and in viral proteins that can prevent it. Complexes stabilized by intermolecular interactions involving RHIMs have a fibrillar amyloid structure. We have identified a novel RHIM within the ORF20 protein expressed by Varicella zoster virus (VZV) that forms amyloid-based complexes with human cellular RHIMs. Whereas other herpesvirus RHIMs inhibit TNF-driven necroptosis, this new VZV RHIM targets the host RHIM-containing protein ZBP1 to inhibit apoptosis during infection. This is the first study to demonstrate the importance of the ZBP1 pathway in VZV infection and to identify the role of a viral RHIM in apoptosis inhibition. It broadens our understanding of host defense pathways and demonstrates how a decoy amyloid strategy is employed by pathogens to circumvent the host response.

[1]  M. Sunde,et al.  RHIM-based protein:protein interactions in microbial defence against programmed cell death by necroptosis. , 2020, Seminars in cell & developmental biology.

[2]  M. Pasparakis,et al.  Z-nucleic acid sensing triggers ZBP1-dependent necroptosis and inflammation , 2020, Nature.

[3]  A. Sickmann,et al.  Herpesviruses induce aggregation and selective autophagy of host signalling proteins NEMO and RIPK1 as an immune-evasion mechanism , 2019, Nature Microbiology.

[4]  Katharina Gaus,et al.  Single-molecule detection on a portable 3D-printed microscope , 2019, Nature Communications.

[5]  J. L. Bernal,et al.  Burden of varicella complications in secondary care, England, 2004 to 2017 , 2019, Euro surveillance : bulletin Europeen sur les maladies transmissibles = European communicable disease bulletin.

[6]  H. Marshall,et al.  Burden of varicella in the Asia-Pacific region: a systematic literature review , 2019, Expert review of vaccines.

[7]  M. Sunde,et al.  Viral M45 and necroptosis‐associated proteins form heteromeric amyloid assemblies , 2018, EMBO reports.

[8]  B. Slobedman,et al.  Infection and Functional Modulation of Human Monocytes and Macrophages by Varicella-Zoster Virus , 2018, Journal of Virology.

[9]  J. Carette,et al.  Species-independent contribution of ZBP1/DAI/DLM-1-triggered necroptosis in host defense against HSV1 , 2018, Cell Death & Disease.

[10]  Hao Wu,et al.  The Structure of the Necrosome RIPK1-RIPK3 Core, a Human Hetero-Amyloid Signaling Complex , 2018, Cell.

[11]  B. Slobedman,et al.  Varicella zoster virus productively infects human natural killer cells and manipulates phenotype , 2018, PLoS pathogens.

[12]  B. McSharry,et al.  Varicella-Zoster Virus ORF63 Protects Human Neuronal and Keratinocyte Cell Lines from Apoptosis and Changes Its Localization upon Apoptosis Induction , 2018, Journal of Virology.

[13]  Andrea Scrima,et al.  The invasin D protein from Yersinia pseudotuberculosis selectively binds the Fab region of host antibodies and affects colonization of the intestine , 2018, The Journal of Biological Chemistry.

[14]  J. Kagan,et al.  Apoptosis and Necroptosis as Host Defense Strategies to Prevent Viral Infection. , 2017, Trends in cell biology.

[15]  J. Rehwinkel,et al.  Sensing of viral and endogenous RNA by ZBP1/DAI induces necroptosis , 2017, The EMBO journal.

[16]  D. Green,et al.  ESCRT-III Acts Downstream of MLKL to Regulate Necroptotic Cell Death and Its Consequences , 2017, Cell.

[17]  W. Kaiser,et al.  Mouse cytomegalovirus M36 and M45 death suppressors cooperate to prevent inflammation resulting from antiviral programmed cell death pathways , 2017, Proceedings of the National Academy of Sciences.

[18]  David F. Boyd,et al.  DAI Senses Influenza A Virus Genomic RNA and Activates RIPK3-Dependent Cell Death. , 2016, Cell host & microbe.

[19]  A. Strasser,et al.  RIPK1 inhibits ZBP1-driven necroptosis during development , 2016, Nature.

[20]  M. Pasparakis,et al.  RIPK1 counteracts ZBP1-mediated necroptosis to inhibit inflammation , 2016, Nature.

[21]  Si Ming Man,et al.  ZBP1/DAI is an innate sensor of influenza virus triggering the NLRP3 inflammasome and programmed cell death pathways , 2016, Science Immunology.

[22]  Xiaoliang Yu,et al.  The interplay between human herpes simplex virus infection and the apoptosis and necroptosis cell death pathways , 2016, Virology Journal.

[23]  Akshay Bhumkar,et al.  Confocal Spectroscopy to Study Dimerization, Oligomerization and Aggregation of Proteins: A Practical Guide , 2016, International journal of molecular sciences.

[24]  M. Bertrand,et al.  Poly-ubiquitination in TNFR1-mediated necroptosis , 2016, Cellular and Molecular Life Sciences.

[25]  R. Cohrs,et al.  Varicella Zoster Virus in the Nervous System , 2015, F1000Research.

[26]  B. Slobedman,et al.  Varicella-Zoster Virus and Herpes Simplex Virus 1 Differentially Modulate NKG2D Ligand Expression during Productive Infection , 2015, Journal of Virology.

[27]  W. Kaiser,et al.  Necroptosis: The Trojan horse in cell autonomous antiviral host defense. , 2015, Virology.

[28]  W. Kaiser,et al.  Suppression of RIP3-dependent Necroptosis by Human Cytomegalovirus , 2015, The Journal of Biological Chemistry.

[29]  P. A. Harris,et al.  Herpes simplex virus suppresses necroptosis in human cells. , 2015, Cell host & microbe.

[30]  Chuan-Qi Zhong,et al.  RIP1/RIP3 binding to HSV-1 ICP6 initiates necroptosis to restrict virus propagation in mice. , 2015, Cell host & microbe.

[31]  Qin Chen,et al.  Direct activation of RIP3/MLKL-dependent necrosis by herpes simplex virus 1 (HSV-1) protein ICP6 triggers host antiviral defense , 2014, Proceedings of the National Academy of Sciences.

[32]  W. Alexander,et al.  Activation of the pseudokinase MLKL unleashes the four-helix bundle domain to induce membrane localization and necroptotic cell death , 2014, Proceedings of the National Academy of Sciences.

[33]  D. Gilden,et al.  Neurological complications of varicella zoster virus reactivation. , 2014, Current opinion in neurology.

[34]  A. Arvin,et al.  Molecular mechanisms of varicella zoster virus pathogenesis , 2014, Nature Reviews Microbiology.

[35]  W. Kaiser,et al.  Viral modulation of programmed necrosis. , 2013, Current opinion in virology.

[36]  Kenta Moriwaki,et al.  The RIP1/RIP3 Necrosome Forms a Functional Amyloid Signaling Complex Required for Programmed Necrosis , 2012, Cell.

[37]  W. Kaiser,et al.  DAI/ZBP1/DLM-1 complexes with RIP3 to mediate virus-induced programmed necrosis that is targeted by murine cytomegalovirus vIRA. , 2012, Cell host & microbe.

[38]  Xiaodong Wang,et al.  Mixed Lineage Kinase Domain-like Protein Mediates Necrosis Signaling Downstream of RIP3 Kinase , 2012, Cell.

[39]  William J. Kaiser,et al.  Viral infection and the evolution of caspase 8-regulated apoptotic and necrotic death pathways , 2011, Nature Reviews Immunology.

[40]  P. Kinchington,et al.  Varicella-Zoster Virus Inhibition of the NF-κB Pathway during Infection of Human Dendritic Cells: Role for Open Reading Frame 61 as a Modulator of NF-κB Activity , 2011, Journal of Virology.

[41]  B. Slobedman,et al.  Differentiated Neuroblastoma Cells Provide a Highly Efficient Model for Studies of Productive Varicella-Zoster Virus Infection of Neuronal Cells , 2011, Journal of Virology.

[42]  A. Sasseville,et al.  The ribonucleotide reductase R1 subunits of herpes simplex virus types 1 and 2 protect cells against TNFα- and FasL-induced apoptosis by interacting with caspase-8 , 2011, Apoptosis.

[43]  W. Kaiser,et al.  Virus inhibition of RIP3-dependent necrosis. , 2010, Cell host & microbe.

[44]  J. Tschopp,et al.  DAI/ZBP1 recruits RIP1 and RIP3 through RIP homotypic interaction motifs to activate NF‐κB , 2009, EMBO reports.

[45]  Na Zhang,et al.  RIP3, an Energy Metabolism Regulator That Switches TNF-Induced Cell Death from Apoptosis to Necrosis , 2009, Science.

[46]  F. Chan,et al.  Phosphorylation-Driven Assembly of the RIP1-RIP3 Complex Regulates Programmed Necrosis and Virus-Induced Inflammation , 2009, Cell.

[47]  Xiaodong Wang,et al.  Receptor Interacting Protein Kinase-3 Determines Cellular Necrotic Response to TNF-α , 2009, Cell.

[48]  Jennifer J. Brady,et al.  The Replication Cycle of Varicella-Zoster Virus: Analysis of the Kinetics of Viral Protein Expression, Genome Synthesis, and Virion Assembly at the Single-Cell Level , 2009, Journal of Virology.

[49]  P. Tomasec,et al.  Re-engineering adenovirus vector systems to enable high-throughput analyses of gene function. , 2008, BioTechniques.

[50]  W. Kaiser,et al.  Receptor-Interacting Protein Homotypic Interaction Motif-Dependent Control of NF-κB Activation via the DNA-Dependent Activator of IFN Regulatory Factors1 , 2008, The Journal of Immunology.

[51]  M. Sommer,et al.  Functions of Varicella-Zoster Virus ORF23 Capsid Protein in Viral Replication and the Pathogenesis of Skin Infection , 2008, Journal of Virology.

[52]  W. Kaiser,et al.  Cytomegalovirus M45 Cell Death Suppression Requires Receptor-interacting Protein (RIP) Homotypic Interaction Motif (RHIM)-dependent Interaction with RIP1* , 2008, Journal of Biological Chemistry.

[53]  Xiaodong Wang,et al.  TNF-α Induces Two Distinct Caspase-8 Activation Pathways , 2008, Cell.

[54]  N. Osterrieder,et al.  A Self-Excisable Infectious Bacterial Artificial Chromosome Clone of Varicella-Zoster Virus Allows Analysis of the Essential Tegument Protein Encoded by ORF9 , 2007, Journal of Virology.

[55]  K. Honda,et al.  DAI (DLM-1/ZBP1) is a cytosolic DNA sensor and an activator of innate immune response , 2007, Nature.

[56]  A. Sasseville,et al.  The ribonucleotide reductase domain of the R1 subunit of herpes simplex virus type 2 ribonucleotide reductase is essential for R1 antiapoptotic function. , 2007, The Journal of general virology.

[57]  B. Banfield,et al.  Postentry Events Are Responsible for Restriction of Productive Varicella-Zoster Virus Infection in Chinese Hamster Ovary Cells , 2006, Journal of Virology.

[58]  I. Jurak,et al.  Induction of apoptosis limits cytomegalovirus cross‐species infection , 2006, The EMBO journal.

[59]  F. Rixon,et al.  Mutational Analysis of the Herpes Simplex Virus Triplex Protein VP19C , 2006, Journal of Virology.

[60]  P. Kinchington,et al.  Varicella-Zoster Virus ORF63 Inhibits Apoptosis of Primary Human Neurons , 2006, Journal of Virology.

[61]  Shannon L. Taylor,et al.  Replication of Varicella-Zoster Virus in Human Skin Organ Culture , 2005, Journal of Virology.

[62]  Xiaodong Wang,et al.  A Small Molecule Smac Mimic Potentiates TRAIL- and TNFα-Mediated Cell Death , 2004, Science.

[63]  A. Cunningham,et al.  Varicella-Zoster Virus-Infected Human Sensory Neurons Are Resistant to Apoptosis, yet Human Foreskin Fibroblasts Are Susceptible: Evidence for a Cell-Type-Specific Apoptotic Response , 2003, Journal of Virology.

[64]  U. Koszinowski,et al.  Role of Murine Cytomegalovirus US22 Gene Family Members in Replication in Macrophages , 2003, Journal of Virology.

[65]  R. Cohrs,et al.  Characterization of Varicella-Zoster Virus Gene 21 and 29 Proteins in Infected Cells , 2002, Journal of Virology.

[66]  V. Dixit,et al.  Identification of a Novel Homotypic Interaction Motif Required for the Phosphorylation of Receptor-interacting Protein (RIP) by RIP3* , 2002, The Journal of Biological Chemistry.

[67]  Brian Seed,et al.  Fas triggers an alternative, caspase-8–independent cell death pathway using the kinase RIP as effector molecule , 2000, Nature Immunology.

[68]  S. Nagata,et al.  Caspase-independent Cell Killing by Fas-associated Protein with Death Domain , 1998, The Journal of cell biology.

[69]  W. Fiers,et al.  Inhibition of Caspases Increases the Sensitivity of L929 Cells to Necrosis Mediated by Tumor Necrosis Factor , 1998, The Journal of experimental medicine.

[70]  L. Serpell,et al.  Common core structure of amyloid fibrils by synchrotron X-ray diffraction. , 1997, Journal of molecular biology.

[71]  J. Cohen,et al.  Deletion of the varicella-zoster virus large subunit of ribonucleotide reductase impairs growth of virus in vitro , 1994, Journal of virology.

[72]  I. Leigh,et al.  Replication of varicella zoster virus in primary human keratinocytes , 1992, Journal of medical virology.

[73]  T. H. Weller Serial Propagation in vitro of Agents Producing Inclusion Bodies Derived from Varicella and Herpes Zoster.∗ , 1953, Proceedings of the Society for Experimental Biology and Medicine. Society for Experimental Biology and Medicine.

[74]  T. H. Weller,et al.  Intranuclear inclusion bodies in cultures of human tissue inoculated with varicella vesicle fluid. , 1952, Journal of immunology.

[75]  Eunhee Kim,et al.  Kinase-independent role of nuclear RIPK1 in regulating parthanatos through physical interaction with PARP1 upon oxidative stress. , 2018, Biochimica et biophysica acta. Molecular cell research.

[76]  Si Ming Man,et al.  ZBP1/DAI is an innate sensor of influenza virus triggering the NLRP3 inflammasome and programmed cell death pathways. , 2016, Science immunology.

[77]  M. Gershon,et al.  VZV infection of keratinocytes: production of cell-free infectious virions in vivo. , 2010, Current topics in microbiology and immunology.

[78]  D. Lembo,et al.  Tinkering with a viral ribonucleotide reductase. , 2009, Trends in biochemical sciences.

[79]  A. Arvin,et al.  Varicella-zoster virus: aspects of pathogenesis and host response to natural infection and varicella vaccine. , 1996, Advances in virus research.