TOPICS

HOT TOPICS IN EIA RESEARCH

Horse Immune Response

 

The following is excerpted from an article authored by Cook RF, Cook SJ, and Issel CJ to appear in the Equine Veterinary Journal special publication on Equine Infectious Diseases

Host Immune Responses

Antibodies against EIAV may be detected in a variety of laboratory tests as early as 14-28 days post infection (pi) although most reports indicate they are non-neutralizing. Strain specific neutralizing antibodies are not generally observed until 38-87 days pi and may not reach maximal levels until 90-148 days pi (O'Rourke et al. 1988; Kono et al. 1973; Rwambo et al. 1990; Ball et al. 1992; Hammond et al. 1997), generally long after the acute disease episode has been resolved. As cytotoxic lymphocytes (CTL) can be detected in experimental infections at 14 days pi (Mealey et al. 2005) it is currently believed that cell-mediated and not humoral immune responses are responsible for initial control of EIAV infections. Once acute viral replication has been controlled the animal will remain free of overt clinical signs until a variant virus emerges that can evade immunological surveillance.

If EIAV has seemingly limitless mutational capacity, what terminates the recurring disease cycles and how do animals attain inapparent carrier status?

At one time it was thought there might be selective pressure to limit pathogenicity in the host and to ensure the progressive attenuation of EIAV (Belshan et al. 1998). However, viruses transferred from inapparent carrier equids often produce disease in naive recipients. Furthermore, recrudescence of clinical signs can be induced in carriers by immunosuppression with corticosteroids (Kono et al. 1976; Tumas et al. 1994; Howe et al. 2005; Craigo et al. 2002). These observations suggest the immune system and not viral attenuation is primarily responsible for the eventual cessation of clinical signs in EIAV infected animals. To envisage how this might occur, it is necessary to consider evolution the mammalian immune system in response to lentiviral infections. Cell-mediated and humoral immune responses to new infections are restricted by the phenomenon of immunodominance to just a handful of the many potential epitopes within microbial pathogens (Kedl, Kappler, and Marrack 2003; Borysiewicz et al. 1988; Busch and Pamer 1998; Pamer, Harty, and Bevan 1991; Rodriguez et al. 2002; Yu et al. 2002). Furthermore, immunodominance does not always translate into an effective immune response. EIAV epitopes recognized by high-avidity immunodominant CTL are subject to rapid mutational changes with the original sequences disappearing from the viral population (Mealey et al. 2003). In contrast, low or even moderate avidity immunodominant CTL do not appear to have substantially detrimental effects on the survival of EIAV and epitopes bound by these less efficient T-cells can persist in the viral population (Mealey et al. 2003; Mealey et al. 2005). Therefore, the initial adaptive immune response to EIAV is likely to consist of the eventual appearance of strain specific neutralizing antibodies coupled with a very limited number of effective CTLs. While this is apparently sufficient to resolve acute disease, it is easy to envisage how a highly mutable virus like EIAV could evade these initial immune responses by producing escape mutants with the replicative capacity to induce additional clinical episodes. As such, it appears the immune system is locked into a constant cycle of "catch-up". However, this cycle may be broken by a gradual broadening of the immune responses leading to the recognition of a greater number of epitopes including some that may be conserved because of functional constraints. The result of these enhanced responses is that complete escape from immunological surveillance becomes extremely difficult unless there is suppression of the immune system. For example, the number of CTL epitopes recognized in some HIV positive people increases from 2 during symptomatic acute infection to 27 after 18 months (Yu et al. 2002). The fact that relatively large numbers of T-helper and CTL epitopes can be identified within just the viral envelope glycoproteins 7 months after infection with an attenuated EIAV strain (Tagmyer et al. 2007) suggests a similar broadening of the immune response occur in the horse. An important additional consideration is that unlike HIV, EIAV does not infect T-helper cells and does not produce immunodeficiency. Therefore, in the case of EIAV infected equids broader cell-mediated immune responses are likely to remain protective for very long periods consistent with inapparent carrier status. In addition, antibody responses in EIAV infected animals gradually evolve from low-avidity interactions with linear epitopes to high-avidity binding with predominantly conformational epitopes (Hammond et al. 1997). Neutralizing antibodies also evolve from being highly strain specific to more generally reactive. However, while more broadly reactive cell-mediated and humoral immune responses may eventually restrict EIAV replication and prevent disease they are not sufficient to completely eradicate the virus.

References

  • Ball JM, Rushlow KE, Issel CJ, Montelaro RC (1992) Detailed mapping of the antigenicity of the surface unit glycoprotein of equine infectious anemia virus by using synthetic peptide strategies. J Virol, 66: 732-42.

  • Belshan M, Harris ME, Shoemaker AE, Hope TJ, Carpenter S (1998) Biological characterization of Rev variation in equine infectious anemia virus. J Virol, 72: 4421-6.

  • Borysiewicz LK, Hickling JK, Graham S, Sinclair J, Cranage MP, Smith GL, Sissons JG (1988) Human cytomegalovirus-specific cytotoxic T cells. Relative frequency of stage-specific CTL recognizing the 72-kD immediate early protein and glycoprotein B expressed by recombinant vaccinia viruses. J Exp Med, 168: 919-31.

  • Busch DH, Pamer EG (1998) MHC class I/peptide stability: implications for immunodominance, in vitro proliferation, and diversity of responding CTL. J Immunol, 160: 4441-8

  • Craigo JK, Leroux C, Howe L, Steckbeck JD, Cook SJ, Issel CJ, Montelaro RC (2002) Transient immune suppression of inapparent carriers infected with a principal neutralizing domain-deficient equine infectious anaemia virus induces neutralizing antibodies and lowers steady-state virus replication. J Gen Virol, 83: 1353-9.

  • Craigo JK, Zhang B, Barnes S, Tagmyer TL, Cook SJ, Issel CJ, Montelaro RC (2007) Envelope variation as a primary determinant of lentiviral vaccine efficacy. Proc Natl Acad Sci U S A, 104: 15105-10.

  • Hammond SA, Cook SJ, Lichtenstein DL, Issel CJ, Montelaro RC (1997) Maturation of the cellular and humoral immune responses to persistent infection in horses by equine infectious anemia virus is a complex and lengthy process. J Virol, 71: 3840-52.

  • Howe L, Craigo JK, Issel CJ, Montelaro RC (2005) Specificity of serum neutralizing antibodies induced by transient immune suppression of inapparent carrier ponies infected with a neutralization-resistant equine infectious anemia virus envelope strain. J Gen Virol, 86: 139-49.

  • Kedl RM, Kappler JW, Marrack P (2003) Epitope dominance, competition and T cell affinity maturation. Curr Opin Immunol, 15: 120-7.

  • Kono Y, Kobayashi K, Fukunaga Y (1973) Antigenic drift of equine infectious anemia virus in chronically infected horses. Arch Gesamte Virusforsch, 41: 1-10.

  • Kono Y, Hirasawa K, Fukunaga Y, Taniguchi T (1976) Recrudescence of equine infectious anemia by treatment with immunosuppressive drugs. Natl Inst Anim Health Q (Tokyo), 16: 8-15.

  • Mealey RH, Zhang B, Leib SR, Littke MH, McGuire TC (2003) Epitope specificity is critical for high and moderate avidity cytotoxic T lymphocytes associated with control of viral load and clinical disease in horses with equine infectious anemia virus. Virology, 313: 537-52.

  • Mealey RH, Sharif A, Ellis SA, Littke MH, Leib SR, McGuire TC (2005) Early detection of dominant Env-specific and subdominant Gag-specific CD8+ lymphocytes in equine infectious anemia virus-infected horses using major histocompatibility complex class I/peptide tetrameric complexes. Virology, 339: 110-26.

  • O'Rourke K, Perryman LE, McGuire TC (1988) Antiviral, anti-glycoprotein and neutralizing antibodies in foals with equine infectious anaemia virus. J Gen Virol, 69 ( Pt 3): 667-74.

  • Pamer EG, Harty JT, Bevan MJ (1991) Precise prediction of a dominant class I MHC-restricted epitope of Listeria monocytogenes. Nature, 353: 852-5.

  • Rodriguez F, Harkins S, Slifka MK, Whitton JL (2002) Immunodominance in virus-induced CD8(+) T-cell responses is dramatically modified by DNA immunization and is regulated by gamma interferon. J Virol, 76: 4251-9.

  • Rwambo PM, Issel CJ, Adams WV Jr, Hussain KA, Miller M, Montelaro RC (1990) Equine infectious anemia virus (EIAV) humoral responses of recipient ponies and antigenic variation during persistent infection. Arch Virol, 111: 199-212.

  • Tumas DB, Hines MT, Perryman LE, Davis WC, McGuire TC (1994) Corticosteroid immunosuppression and monoclonal antibody-mediated CD5+ T lymphocyte depletion in normal and equine infectious anaemia virus-carrier horses. J Gen Virol, 75 ( Pt 5): 959-68.

  • Yu XG, Addo MM, Rosenberg ES, Rodriguez WR, Lee PK, Fitzpatrick CA, Johnston MN, Strick D, Goulder PJ, Walker BD, Altfeld M (2002) Consistent patterns in the development and immunodominance of human immunodeficiency virus type 1 (HIV-1)-specific CD8+ T-cell responses following acute HIV-1 infection. J Virol, 76: 8690-701.

  • Updated on: February 17, 2010