Elsevier

Virus Research

Volume 153, Issue 1, October 2010, Pages 82-91
Virus Research

Mapping of amino acid residues responsible for adhesion of cell culture-adapted foot-and-mouth disease SAT type viruses

https://doi.org/10.1016/j.virusres.2010.07.010Get rights and content

Abstract

Foot-and-mouth disease virus (FMDV) infects host cells by adhering to the αV subgroup of the integrin family of cellular receptors in a Arg–Gly–Asp (RGD) dependent manner. FMD viruses, propagated in non-host cell cultures are reported to acquire the ability to enter cells via alternative cell surface molecules. Sequencing analysis of SAT1 and SAT2 cell culture-adapted variants showed acquisition of positively charged amino acid residues within surface-exposed loops of the outer capsid structural proteins. The fixation of positively charged residues at position 110–112 in the βF–βG loop of VP1 of SAT1 isolates is thought to correlate with the acquisition of the ability to utilise alternative glycosaminoglycan (GAG) molecules for cell entry. Similarly, two SAT2 viruses that adapted readily to BHK-21 cells accumulated positively charged residues at positions 83 and 85 of the βD–βE loop of VP1. Both regions surround the fivefold axis of the virion. Recombinant viruses containing positively charged residues at position 110 and 112 of VP1 were able to infect CHO-K1 cells (that expresses GAG) and demonstrated increased infectivity in BHK-21 cells. Therefore, recombinant SAT viruses engineered to express substitutions that induce GAG-binding could be exploited in the rational design of vaccine seed stocks with improved growth properties in cell cultures.

Introduction

Foot-and-mouth disease (FMD) is a highly contagious vesicular disease of cloven-hoofed animals causing significant distress and suffering in animals. Although mortality is usually low (<5%) (Thomson, 1995), morbidity can reach 100% and the impact can be catastrophical when an outbreak occurs in a FMD-free region with immunologically naïve population of livestock. Consequently FMD is classified by the OIE as one of the most important infectious diseases of livestock (Office International des Épizooties Terrestrial Manual, 2009). The economically critical effects to livestock farming due to the high cost of disease control and international trade restrictions (Sellers and Daggupaty, 1990) was evidenced during the 2000–2001 outbreaks in Europe and the virus escape that occurred more recently, in 2007 in the United Kingdom (Samuel and Knowles, 2001, Cottam et al., 2008). In endemic regions, FMD is controlled by restricting animal movement, the implementation of vaccination programmes and biosecurity measures. In disease-free countries where vaccination is normally not applied, ring-vaccination is only used in an emerging outbreak with subsequent slaughtering of vaccinated animals (Müller et al., 2001, Tomassen et al., 2002).

In South Africa, other regions of the African continent, as well as in some Asian and South American countries, regular immunisation is essential for disease control, and in maintaining FMD-free status. Current FMD vaccines are chemically inactivated preparations of concentrated, virus-infected cell culture supernatants (Office International des Épizooties Terrestrial Manual, 2009). Therefore, large-scale vaccine production utilize a suitable cell line, like BHK-21 cells, and requires that the vaccine strain is adapted and propagated in cell culture (Amadori et al., 1994, Amadori et al., 1997). However, these cell lines have limited (monolayers) or no (in suspension) expression of the required primary receptor for infection by FMDV (Amadori et al., 1994). In the early 1980s it was noted that viruses of the three SAT serotypes, endemic in Africa, are notorious for their difficulty to adapt to BHK-21 cells (Pay et al., 1978, Preston et al., 1982). It is thought that the cell surface molecules, which may act as virus receptors, exert an important selective pressure on viral RNA quasi-species, thereby enabling adaptation. Studies have shown that repeated passaging of FMDV in cultured cells rapidly gave rise to mutant viruses within the population (Rieder et al., 1994, Herrera et al., 2007). Adaptation of wild-type SAT viruses in cell culture to produce high yields of stable antigen is an intricate and time-consuming process that is often associated with a low success rate.

FMD virus (FMDV), the type species of the Aphtovirus genus in the family Picornaviridae (Racaniello, 2006), infects epithelial cells by adhering to any of four members of the αV subgroup of the integrin family of cellular receptors, i.e. αVβ1, αVβ3, αVβ6 and αVβ8 (Berinstein et al., 1995, Neff et al., 1998, Neff et al., 2000, Jackson et al., 1997, Jackson et al., 2000, Jackson et al., 2002, Jackson et al., 2004, Duque and Baxt, 2003). Attachment to the receptors is mediated via a highly conserved Arg–Gly–Asp (RGD) motif (Fox et al., 1989, Baxt and Becker, 1990, Mason et al., 1994, Leippert et al., 1997) located within the structurally disordered βG–βH loop of VP1 (Acharya et al., 1989, Lea et al., 1995, Curry et al., 1997). Following FMDV–receptor interactions, the virus is internalised and the viral genome is released in the cytosol. Adaptation of FMD field isolates to enable efficient replication in cultured cells is accompanied by changes in viral properties, including the acquisition of the ability to bind to alternative cellular receptors such as cell surface glycosaminoglycans (GAGs) (Jackson et al., 1996, Jackson et al., 2001, Sa-Carvalho et al., 1997, Zhao et al., 2003). The interactions of a diverse group of ligands, such as growth factors, chemokines, herpes simplex virus (HSV), human immunodeficiency virus, respiratory syncytial virus, alphaviruses, dengue virus, adeno-associated virus and FMDV, to the highly sulfated GAGs (also known as heparan sulfate proteoglycans, HSPG) is typically via a positively charged domain on these proteins (Patel et al., 1993, Gromm et al., 1995, Jackson et al., 1996, Chen et al., 1997, Krusat and Streckert, 1997, Sa-Carvalho et al., 1997, Byrnes and Griffin, 1998, Klimstra et al., 1998, Summerford and Samulski, 1998, Fry et al., 1999, Fry et al., 2005, Zhao et al., 2003). The ability of a type O virus to enter cells following adhesion to HSPG is thought to be dependent on the presence of the positively charged Arg residue at position 56 of VP3 which results in a net gain of positive charge on the virion surface (Sa-Carvalho et al., 1997, Fry et al., 1999).

Replacement of the external capsid-coding sequence of an infectious cDNA clone with the corresponding region of an outbreak virus results in the transfer of surface-exposed epitopes from the aetiological agent to the recombinant virus (Zibert et al., 1990, Rieder et al., 1993, Almeida et al., 1998, Van Rensburg and Mason, 2002, Van Rensburg et al., 2004). The chimeric viruses, produced in this manner, induce a protective immune response in animals similar to that of the outbreak virus. However, co-transferral of undesirable traits, such as capsid instability and poor cell culture adaptation of the field virus may also occur. Therefore, application of reverse genetics technology in FMD vaccinology includes the identification of amino acid sequences associated with the acquisition of HSPG-binding during cell culture adaptation of SAT viruses and the introduction of these changes into chimeric constructs.

In this report we identify novel amino acid residues within the capsid proteins of SAT1 and SAT2 viruses that affect the virus’ ability to grow in different cell lines. We demonstrated that cell culture adaptation to BHK-21 cells is acquired following repeated passaging of the field viruses in these cells. Furthermore, we illustrated that this phenotype can be transferred to an infectious cDNA clone of a FMD field virus from which viable cell culture-adapted viruses were recovered.

Section snippets

Cells, viruses and plasmids

Baby hamster kidney (BHK) cells, strain 21, clone 13 (ATCC CCL-10) were maintained as described by Rieder et al. (1993). Chinese hamster ovary (CHO) cells strain K1 (ATCC CCL-61) were maintained in Ham's F-12 medium (Invitrogen), supplemented with 10% foetal calf serum (Delta Bioproducts). Primary pig kidney (PK) and Instituto Biologico renal suino (IB-RS-2) cells Plaque assays were performed using a tragacanth overlay method and 1% methylene blue staining (Rieder et al., 1993).

Viruses used in

Adaptation of SAT viruses in cell culture selects variants that gain a net positive charge on the virion surface

SAT viruses used for vaccine production are typically adapted to cultured cells following limited passages in BHK-21 cells. In this study, we have investigated the phenotypic and genetic changes associated with the transition of SAT viruses from wild-type to the cell culture-adapted phenotype. The morphology of virus plaques produced on BHK-21 cell monolayers following infection with SAT1 and SAT2 vaccine strains differed from the plaques produced by the parental strains which have not been

Discussion

Despite the success of conventional vaccines in the control of FMD in the developed world, inactivated vaccines are unable to cover the vast antigenic variability within the SAT types in southern Africa (Hunter, 1996). Recombinant inactivated SAT type vaccines, structurally designed to be effective for specific geographic regions, may overcome the limitation of antigenic variation (Van Rensburg et al., 2004). However, the transfer of antigenic determinants during the replacement of the outer

Acknowledgments

This work was supported by funding from Intervet–Schering-Plough and the U.S. Department of Agriculture, Agricultural Research Service. The cell culture-adapted SAT1 and SAT2 viruses were received from the vaccine unit at Transboundary Animal Diseases of the ARC-OVI. We thank Juanita van Heerden for her contributions with sequencing part of SAT2/KNP/19/89. We would also like to thank Dr. Otto Koekemoer, Ms. Sonja Maree and Erika Kirkbride for critical reading of the manuscript.

References (59)

  • R. Acharya et al.

    The three-dimensional structure of foot-and-mouth disease virus at 2.9 Å resolution

    Nature

    (1989)
  • A.D.S. Bastos

    Detection and characterization of foot-and-mouth disease virus in sub-Saharan Africa

    Onderstepoort J. Vet. Res.

    (1998)
  • A.D.S. Bastos et al.

    Genetic heterogeneity of SAT-1 type foot-an-d-mouth disease viruses in southern Africa

    Arch. Virol.

    (2001)
  • A.D.S. Bastos et al.

    The implications of virus diversity within the SAT 2 serotype for control of foot-and-mouth disease in sub-Saharan Africa

    J. Gen. Virol.

    (2003)
  • B. Baxt et al.

    The effect of peptides containing the arginine–glycine–aspartic acid sequence on the adsorption of foot-and-mouth disease virus to tissue culture cells

    Virus Genes

    (1990)
  • A. Berinstein et al.

    Antibodies to the victronectin receptor (Integrin αvβ3) inhibit binding and infection of foot-and-mouth disease virus to cultured cells

    J. Virol.

    (1995)
  • Böhmer, B., 2004. Engineering of a chimeric SAT2 foot-and-mouth disease virus for vaccine production. M.Sc. thesis,...
  • A.P. Byrnes et al.

    Binding of Sindbis virus to cell surface heparan sulfate

    J. Virol.

    (1998)
  • A.P. Byrnes et al.

    Large-plaque mutants of Sindbis virus show reduced binding to heparan sulfate, heightened viremia, and slower clearance from the circulation

    J. Virol.

    (2000)
  • Y. Chen et al.

    Dengue virus infectivity depends on envelope protein binding to target cell heparan sulfate

    Nat. Med.

    (1997)
  • E.M. Cottam et al.

    Integrating genetic and epidemiological data to determine transmission pathways of foot and mouth disease virus

    Proc. Biol. Sci.

    (2008)
  • S. Curry et al.

    Dissecting the roles of VP0 cleavage and RNA packaging in picornavirus capsid stabilization, the structure of empty capsids of foot-and-mouth disease virus

    J. Virol.

    (1997)
  • H. Duque et al.

    FMDV receptors: comparison of bovine av integrin utilization by type A and O viruses

    J. Virol.

    (2003)
  • G. Fox et al.

    The cell attachment site on foot-and-mouth disease virus includes the amino acid sequence RGD (arginine–glycine–aspartic acid)

    J. Gen. Virol.

    (1989)
  • E.E. Fry et al.

    The structure and function of a foot-and-mouth disease virus-oligosaccharide receptor complex

    EMBO J.

    (1999)
  • E.E. Fry et al.

    Structure of FMDV serotype A1O61 alone and complexed with oligosaccharide receptor, receptor conservation in the face of antigenic variation

    J. Gen. Virol.

    (2005)
  • J.R. Gromm et al.

    Differences in the interaction of heparin with arginine and lysine and the importance of these basic amino acids in the binding of heparin to acidic fibroblast growth factor

    Arch. Biochem. Biophys.

    (1995)
  • M. Herrera et al.

    Molecular basis for a lack of correlation between viral fitness and cell killing capacity

    PLoS Pathog.

    (2007)
  • F.P. Heydrick et al.

    Host influence on the characteristics of Venezuelan equine encephalomyelitis virus

    J. Bacteriol.

    (1966)
  • Cited by (38)

    • The endocytosis of foot-and mouth disease virus requires clathrin and caveolin and is dependent on the existence of Rab5 and Rab7 in CHO-677 cells

      2022, Veterinary Microbiology
      Citation Excerpt :

      FMDV has gained the ability to enter cells through the alternative molecules in non-host cells. It has been reported that the recombinant FMDV cell adaptive-strain was able to infect CHO-K1 cells expressing GAGs and had increased infectivity to BHK-21 cells when the VP1 protein contained the positive residues at 110–112 position (Maree et al., 2010). Although in terms of pathogenicity, the mutations of some FMDV strains cause their infection of the hosts without using receptors such as HS and integrin, the mechanisms of endocytosis independent of HS and integrin receptors remain unknown (Rieder et al., 2005; Zhao et al., 2003b).

    • The selection of naturally stable candidate foot-and-mouth disease virus vaccine strains for East Africa

      2021, Vaccine
      Citation Excerpt :

      Sequence analyses of the external capsid proteins (VP1-3) for each strain identified a small number of amino acid changes in the O, A and SAT1 vaccine candidates and none in the SAT2 candidate. Four of the six amino acid changes in VP1-3 that were identified in this study following cell culture adaptation have been reported for the respective serotype [17]; a VP2132 V to I cell culture adaptation (instead of VP2132M to I described herein) has been reported for serotype O [18], a VP282 E to K adaptation for serotype A [19], and VP1111 N to K and VP39 D to A adaptations for serotype SAT1 [20,21]. Importantly, the changes accrued during cell culture adaptation did not affect capsid stability (data not shown).

    • Vaccine strain of O/ME-SA/Ind-2001e of foot-and-mouth disease virus provides high immunogenicity and broad antigenic coverage

      2020, Antiviral Research
      Citation Excerpt :

      FMDV infection and culture were performed in a Bio-Safety Level 3 facility in APQA. CHO–K1 (ATCC CCL-61) cells were used to test the heparan sulfate dependence of FMDV, as CHO–K1 cells use heparan sulfate as the secondary FMDV receptor and do not possess integrins as receptors in the field FMDV (Baranowski et al., 2000; Maree et al., 2010). Serial passage was performed for virus titration in CHO–K1 cells as follows: (1) five times in LFBK cells and three times in adherent BHK-21 cells, (2) five times in LFBK cells, four times in adherent BHK-21 cells, and one time in suspension BHK-21 cells, (3) five times in LFBK cells, four times in adherent BHK-21 cells, and four times in suspension BHK-21 cells, (4) five times in LFBK cells, four times in adherent BHK-21 cells, and six times in suspension BHK-21 cells, or (5) five times in LFBK cells, four times in adherent BHK-21 cells, and seven times in suspension BHK-21 cells.

    • Evaluation of immune responses of stabilised SAT2 antigens of foot-and-mouth disease in cattle

      2017, Vaccine
      Citation Excerpt :

      Several studies have shown that chimeric vaccines successfully induce protective immune responses and protect FMD host species against live virus challenge [23–25]. Additionally, the SAT capsid can be engineered to encode antigens required for vaccines in specific geographic localities [25] and for improved cell adaptation properties [26,27]. In many remote areas of Africa, the reliable maintenance of an adequate cold-chain for FMD vaccines from the manufacturer to the field is not possible.

    View all citing articles on Scopus
    1

    Currently at: European Bioinformatics Institute, Wellcome Trust Campus, Hinxton, Cambridge, CB10 1SD, United Kingdom.

    View full text