A conserved transcriptional response to intranasal Ebola virus exposure in nonhuman primates prior to onset of fever.

Emily Speranza, Sandra L Bixler, Louis A Altamura, Catherine E Arnold, William D Pratt, Cheryl Taylor-Howell, Christina Burrows, William Aguilar, Franco Rossi, Joshua D Shamblin, Suzanne E Wollen, Justine M Zelko, Timothy Minogue, Elyse Nagle, Gustavo Palacios, Arthur J Goff, John H Connor
Author Information
  1. Emily Speranza: Department of Microbiology, Bioinformatics Program, National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA 02118, USA. ORCID
  2. Sandra L Bixler: Molecular and Translational Sciences Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA. ORCID
  3. Louis A Altamura: Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA. ORCID
  4. Catherine E Arnold: Center for Genome Sciences, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA. ORCID
  5. William D Pratt: Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA.
  6. Cheryl Taylor-Howell: Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA. ORCID
  7. Christina Burrows: Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA. ORCID
  8. William Aguilar: Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA. ORCID
  9. Franco Rossi: Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA.
  10. Joshua D Shamblin: Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA.
  11. Suzanne E Wollen: Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA. ORCID
  12. Justine M Zelko: Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA. ORCID
  13. Timothy Minogue: Diagnostic Systems Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA.
  14. Elyse Nagle: Center for Genome Sciences, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA.
  15. Gustavo Palacios: Center for Genome Sciences, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA. gustavo.f.palacios.ctr@mail.mil arthur.j.goff.civ@mail.mil jhconnor@bu.edu. ORCID
  16. Arthur J Goff: Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA. gustavo.f.palacios.ctr@mail.mil arthur.j.goff.civ@mail.mil jhconnor@bu.edu. ORCID
  17. John H Connor: Department of Microbiology, Bioinformatics Program, National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA 02118, USA. gustavo.f.palacios.ctr@mail.mil arthur.j.goff.civ@mail.mil jhconnor@bu.edu. ORCID

Abstract

Ebola virus disease (EVD), caused by Ebola virus (EBOV), is a severe illness characterized by case fatality rates of up to 90%. The sporadic nature of outbreaks in resource-limited areas has hindered the ability to characterize the pathogenesis of EVD at all stages of infection but particularly early host responses. Pathogenesis is often studied in nonhuman primate (NHP) models of disease that replicate major aspects of human EVD. Typically, NHP models use a large infectious dose, are carried out through intramuscular or aerosol exposure, and have a fairly uniform disease course. By contrast, we report our analysis of the host response to EBOV after intranasal exposure. Twelve cynomolgus macaques were infected with 100 plaque-forming units of EBOV/Makona through intranasal exposure and presented with varying times to onset of EVD. We used RNA sequencing and a newly developed NanoString CodeSet to monitor the host response via changes in RNA transcripts over time. When individual animal gene expression data were phased based on the onset of sustained fever, the first clinical sign of severe disease, mathematical models indicated that interferon-stimulated genes appeared as early as 4 days before fever onset. This demonstrates that lethal EVD has a uniform and predictable response to infection regardless of time to onset. Furthermore, expression of a subset of genes could predict disease development before other host-based indications of infection such as fever.

References

  1. Bioinformatics. 2015 Jan 15;31(2):166-9 [PMID: 25260700]
  2. BMC Genomics. 2014 Nov 06;15:960 [PMID: 25377889]
  3. BMC Genomics. 2016 Sep 05;17:707 [PMID: 27595844]
  4. Nucleic Acids Res. 2013 Jan;41(Database issue):D1040-6 [PMID: 23203888]
  5. Sci Rep. 2017 Aug 29;7(1):9730 [PMID: 28852031]
  6. J Infect Dis. 2011 Nov;204 Suppl 3:S1043-52 [PMID: 21987740]
  7. J Virol. 2010 Jun;84(11):5670-7 [PMID: 20335250]
  8. Viruses. 2017 Oct 29;9(11): [PMID: 29109373]
  9. BMC Genomics. 2014 Oct 03;15:846 [PMID: 25277458]
  10. Viruses. 2014 Nov 24;6(11):4666-82 [PMID: 25421892]
  11. Cell Host Microbe. 2017 Dec 13;22(6):817-829.e8 [PMID: 29154144]
  12. Int J Exp Pathol. 1995 Aug;76(4):227-36 [PMID: 7547435]
  13. Nat Protoc. 2009;4(8):1184-91 [PMID: 19617889]
  14. Clin Vaccine Immunol. 2015 Mar;22(3):354-6 [PMID: 25589554]
  15. J Virol. 2011 Sep;85(17):9060-8 [PMID: 21734050]
  16. Cancer Res. 2004 Aug 1;64(15):5245-50 [PMID: 15289330]
  17. Vaccines (Basel). 2017 Sep 20;5(3): [PMID: 28930167]
  18. Sci Rep. 2017 Nov 7;7(1):14756 [PMID: 29116224]
  19. Arch Pathol Lab Med. 1996 Feb;120(2):140-55 [PMID: 8712894]
  20. Genome Biol. 2014;15(12):550 [PMID: 25516281]
  21. Sci Rep. 2017 Mar 03;7:43776 [PMID: 28256637]
  22. PLoS Negl Trop Dis. 2013 Apr 25;7(4):e2171 [PMID: 23638192]
  23. Am J Trop Med Hyg. 2010 May;82(5):954-60 [PMID: 20439981]
  24. Front Microbiol. 2013 Sep 05;4:267 [PMID: 24046765]
  25. Genome Biol. 2017 Jan 19;18(1):4 [PMID: 28100256]
  26. Int J Biochem Cell Biol. 2005 Aug;37(8):1560-6 [PMID: 15896665]
  27. Viruses. 2015 Oct 23;7(10):5489-507 [PMID: 26512687]
  28. J Virol. 2015 Oct;89(19):9865-74 [PMID: 26202234]
  29. Nat Rev Immunol. 2015 Feb;15(2):104-16 [PMID: 25614320]
  30. Sci Transl Med. 2017 Apr 12;9(385): [PMID: 28404864]
  31. Lancet. 2011 Mar 5;377(9768):849-62 [PMID: 21084112]
  32. Bioinformatics. 2009 May 1;25(9):1105-11 [PMID: 19289445]
  33. Curr Opin Genet Dev. 2008 Oct;18(5):461-7 [PMID: 18782619]
  34. Cell Rep. 2017 Jan 17;18(3):816-829 [PMID: 28099857]
  35. Nat Methods. 2012 Mar 04;9(4):357-9 [PMID: 22388286]
  36. Genome Biol. 2007;8(8):R174 [PMID: 17725815]
  37. Viruses. 2012 Dec 06;4(12):3511-30 [PMID: 23223188]
  38. Dis Model Mech. 2009 Jan-Feb;2(1-2):12-7 [PMID: 19132113]
  39. Genome Res. 2003 Sep;13(9):2129-41 [PMID: 12952881]
  40. Curr Opin Virol. 2017 Feb;22:51-58 [PMID: 28012412]
  41. Ann Glob Health. 2014 Nov-Dec;80(6):444-51 [PMID: 25960093]
  42. PLoS Negl Trop Dis. 2014 Jul 31;8(7):e3061 [PMID: 25079789]
  43. Can J Infect Dis Med Microbiol. 2014 May;25(3):128-9 [PMID: 25285105]

Grants

  1. EP-D-15-015/EPA
  2. R01 AI096159/NIAID NIH HHS
  3. R21 AI121933/NIAID NIH HHS
  4. R21 AI135517/NIAID NIH HHS

MeSH Term

Administration, Intranasal
Animals
Disease Models, Animal
Ebolavirus
Hemorrhagic Fever, Ebola
Macaca fascicularis