An optimized HMGB1 expressed by recombinant rabies virus enhances immunogenicity through activation of dendritic cells in mice.

Rabies remains an important public health threat, killing approximately 59,000 people worldwide annually, most of which are from the developing countries of Africa and Asia where dog rabies are endemic. Therefore, developing an affordable and efficacious vaccine for dog-mediated rabies control is needful in these countries. Our previous studies indicated that over-expression of granulocyte-macrophage colony stimulating factor (GM-CSF) or macrophage inflammatory protein-1 (MIP-1α or CCL3) by recombinant rabies virus (rRABV) could enhance the immunogenicity by activating dendritic cells (DCs). In this study, to further characterize the role of activating DCs in RABV immunogenicity, High mobility group box 1 (HMGB1), a highly conserved and non-histone chromosomal protein that can promote DCs maturation and activation, were investigated. The wild-type HMGB1 (HMGB1wt) and an optimized HMGB1 (HMGB1mut) were individually inserted into the genome of the rRABV strain LBNSE (designated as LBNSE-HMGB1wt and LBNSE-HMGB1mut, respectively), and the effect of over-expression of HMGB1 on the immunogenicity of RABV was investigated. The results demonstrated that LBNSE-HMGB1mut could promote significantly more DCs activation, and the recruitment of follicular helper T, germinal center B and plasma cells in vaccinated mice than those immunized with LBNSE-HMGB1wt or parent virus LBNSE. Further investigations suggested that mice vaccinated with LBNSE-HMGB1mut produced significantly higher level of RABV-neutralizing antibodies and offered a better protection than those vaccinated with LBNSE or LBNSE-HMGB1wt. Taken together, these data provides a better understanding of the mechanism for HMGB1 as a potential adjuvant in enhancing the immunogenicity of RABV, which would contribute to developing more-efficacious rabies vaccines.

[1]  Shengdi Wu,et al.  HMGB1-induced autophagy: a new pathway to maintain Treg function during chronic hepatitis B virus infection. , 2017, Clinical science.

[2]  D. Calado,et al.  Germinal Centers , 2017, Methods in Molecular Biology.

[3]  Huanchun Chen,et al.  A Novel Rabies Vaccine Expressing CXCL13 Enhances Humoral Immunity by Recruiting both T Follicular Helper and Germinal Center B Cells , 2016, Journal of Virology.

[4]  Caiping Guo,et al.  Rabies Control and Treatment: From Prophylaxis to Strategies with Curative Potential , 2016, Viruses.

[5]  M. Prausnitz,et al.  Rabies vaccination in dogs using a dissolving microneedle patch. , 2016, Journal of controlled release : official journal of the Controlled Release Society.

[6]  Y. Liao,et al.  Porcine epidemic diarrhea virus nucleoprotein contributes to HMGB1 transcription and release by interacting with C/EBP-β , 2016, Oncotarget.

[7]  A. Sarnaik,et al.  Intralesional rose bengal in melanoma elicits tumor immunity via activation of dendritic cells by the release of high mobility group box 1 , 2016, Oncotarget.

[8]  A. Sharpe,et al.  T follicular regulatory cells , 2016, Immunological reviews.

[9]  N. Câmara,et al.  Follicular helper T cell in immunity and autoimmunity , 2016, Brazilian journal of medical and biological research = Revista brasileira de pesquisas medicas e biologicas.

[10]  Bernadette,et al.  Human rabies transmitted by dogs: current status of global data, 2015. , 2016, Releve epidemiologique hebdomadaire.

[11]  Wei R. Chen,et al.  Stimulation of dendritic cells by DAMPs in ALA-PDT treated SCC tumor cells , 2015, Oncotarget.

[12]  G. Rall,et al.  Everything You Always Wanted to Know About Rabies Virus (But Were Afraid to Ask). , 2015, Annual review of virology.

[13]  M. Zhou,et al.  Critical Role of K1685 and K1829 in the Large Protein of Rabies Virus in Viral Pathogenicity and Immune Evasion , 2015, Journal of Virology.

[14]  M. Zhou,et al.  Recombinant rabies virus expressing dog GM-CSF is an efficacious oral rabies vaccine for dogs , 2015, Oncotarget.

[15]  J. Dushoff,et al.  Estimating the Global Burden of Endemic Canine Rabies , 2015, PLoS neglected tropical diseases.

[16]  H. Ueno,et al.  Pathophysiology of T follicular helper cells in humans and mice , 2015, Nature Immunology.

[17]  S. Crotty T follicular helper cell differentiation, function, and roles in disease. , 2014, Immunity.

[18]  D. Hooper,et al.  Expression of Interferon Gamma by a Recombinant Rabies Virus Strongly Attenuates the Pathogenicity of the Virus via Induction of Type I Interferon , 2014, Journal of Virology.

[19]  S. Cleaveland,et al.  Implementing Pasteur's vision for rabies elimination , 2014, Science.

[20]  S. Esener,et al.  TLR4-dependent activation of dendritic cells by an HMGB1-derived peptide adjuvant , 2014, Journal of Translational Medicine.

[21]  N. Johnson,et al.  Current status of rabies and prospects for elimination , 2014, The Lancet.

[22]  R. Basaraba,et al.  High mobility group box 1 acts as an adjuvant for tuberculosis subunit vaccines , 2014, Immunology.

[23]  Todd G. Smith,et al.  Current and future tools for global canine rabies elimination. , 2013, Antiviral research.

[24]  S. Crotty,et al.  Modulation of SAP dependent T:B cell interactions as a strategy to improve vaccination. , 2013, Current opinion in virology.

[25]  Z. Fu,et al.  Recombinant Rabies Viruses Expressing GM-CSF or Flagellin Are Effective Vaccines for Both Intramuscular and Oral Immunizations , 2013, PloS one.

[26]  Laurence Zitvogel,et al.  Immunogenic cell death in cancer therapy. , 2013, Annual review of immunology.

[27]  M. Lotze,et al.  PAMPs and DAMPs: signal 0s that spur autophagy and immunity , 2012, Immunological reviews.

[28]  C. Qian,et al.  Regulatory dendritic cells program B cells to differentiate into CD19hiFcγIIbhi regulatory B cells through IFN-β and CD40L. , 2012, Blood.

[29]  M. Nussenzweig,et al.  Dopamine in germinal centers , 2017, Nature Immunology.

[30]  D. Weiner,et al.  Molecular adjuvant HMGB1 enhances anti-influenza immunity during DNA vaccination , 2011, Gene Therapy.

[31]  S. Crotty,et al.  Follicular helper CD4 T cells (TFH). , 2011, Annual review of immunology.

[32]  R. Ruigrok,et al.  Rabies virus transcription and replication. , 2011, Advances in virus research.

[33]  Hualei Wang,et al.  Rabies Virus Expressing Dendritic Cell-Activating Molecules Enhances the Innate and Adaptive Immune Response to Vaccination , 2010, Journal of Virology.

[34]  Hualei Wang,et al.  Expression of MIP-1α (CCL3) by a Recombinant Rabies Virus Enhances Its Immunogenicity by Inducing Innate Immunity and Recruiting Dendritic Cells and B Cells , 2010, Journal of Virology.

[35]  Andreas Radbruch,et al.  Organization of immunological memory by bone marrow stroma , 2010, Nature Reviews Immunology.

[36]  Huanchun Chen,et al.  The Roles of Chemokines in Rabies Virus Infection: Overexpression May Not Always Be Beneficial , 2009, Journal of Virology.

[37]  D. Weiner,et al.  Co‐immunization with an optimized plasmid‐encoded immune stimulatory interleukin, high‐mobility group box 1 protein, results in enhanced interferon‐γ secretion by antigen‐specific CD8 T cells , 2009, Immunology.

[38]  M. Gougeon,et al.  HMGB1-Dependent Triggering of HIV-1 Replication and Persistence in Dendritic Cells as a Consequence of NK-DC Cross-Talk , 2008, PloS one.

[39]  A. Izzo,et al.  Mycobacterial infection induces the secretion of high‐mobility group box 1 protein , 2008, Cellular microbiology.

[40]  T. Strowig,et al.  NK cells interactions with dendritic cells shape innate and adaptive immunity. , 2008, Frontiers in bioscience : a journal and virtual library.

[41]  C. Doglioni,et al.  Maturing Dendritic Cells Depend on RAGE for In Vivo Homing to Lymph Nodes1 , 2008, The Journal of Immunology.

[42]  Jeon-Soo Shin,et al.  Nucleocytoplasmic Shuttling of HMGB1 Is Regulated by Phosphorylation That Redirects It toward Secretion1 , 2006, The Journal of Immunology.

[43]  Giovanna Angelini,et al.  NK/iDC interaction results in IL-18 secretion by DCs at the synaptic cleft followed by NK cell activation and release of the DC maturation factor HMGB1. , 2005, Blood.

[44]  Pamini Rasalingam,et al.  Comparative pathogenesis of the SAD-L16 strain of rabies virus and a mutant modifying the dynein light chain binding site of the rabies virus phosphoprotein in young mice. , 2005, Virus research.

[45]  K. Calame,et al.  Regulation of plasma-cell development , 2005, Nature Reviews Immunology.

[46]  Andreas Radbruch,et al.  Maintenance of serum antibody levels. , 2005, Annual review of immunology.

[47]  S. Müller,et al.  HMGB1 is an endogenous immune adjuvant released by necrotic cells , 2004, EMBO reports.

[48]  Tiziana Bonaldi,et al.  Monocytic cells hyperacetylate chromatin protein HMGB1 to redirect it towards secretion , 2003, The EMBO journal.

[49]  J. Cyster,et al.  A Coordinated Change in Chemokine Responsiveness Guides Plasma Cell Movements , 2001, The Journal of experimental medicine.

[50]  G. Schuler,et al.  An advanced culture method for generating large quantities of highly pure dendritic cells from mouse bone marrow. , 1999, Journal of immunological methods.

[51]  R. Steinman,et al.  Dendritic cells and the control of immunity , 1998, Nature.

[52]  R. Ahmed,et al.  Humoral immunity due to long-lived plasma cells. , 1998, Immunity.

[53]  K. Conzelmann,et al.  Molecular cloning and complete nucleotide sequence of the attenuated rabies virus SAD B19. , 1990, Virology.

[54]  G. Goodwin,et al.  The isolation of the high mobility group non‐histone chromosomal protein HMG 14 , 1977, FEBS letters.

[55]  G. Goodwin,et al.  A new group of chromatin-associated proteins with a high content of acidic and basic amino acids. , 1973, European journal of biochemistry.