feline lentiviruses demonstrate differences in receptor repertoire and envelope structural elements

17
Feline lentiviruses demonstrate differences in receptor repertoire and envelope structural elements Natalia Smirnova a , Jennifer L. Troyer a , Jennifer Schissler a , Julie Terwee a , Mary Poss b , Sue VandeWoude a, * a Department of Microbiology, Immunology, and Pathology, Colorado State University, 1619 Campus Delivery, Fort Collins, CO 80523-1619, USA b Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA Received 20 April 2005; returned to author for revision 9 June 2005; accepted 20 July 2005 Available online 24 August 2005 Abstract Feline immunodeficiency virus (FIV) causes fatal disease in domestic cats via T cell depletion-mediated immunodeficiency. Pumas and lions are hosts for apparently apathogenic lentiviruses (PLV, LLV) distinct from FIV. We compared receptor use among these viruses by: (1) evaluating target cell susceptibility; (2) measuring viral replication following exposure to specific and non-specific receptor antagonists; and (3) comparing Env sequence and structural motifs. Most isolates of LLVand PLV productively infected domestic feline T cells, but differed from domestic cat FIV by infecting cells independent of CXCR4, demonstrating equivalent or enhanced replication following heparin exposure, and demonstrating substantial divergence in amino acid sequence and secondary structure in Env receptor binding domains. PLV infection was, however, inhibited by CD134/OX40 antibody. Thus, although PLVand LLV infection interfere with FIV superinfection, we conclude that LLVand PLV utilize novel, more promiscuous mechanisms for cell entry than FIV, underlying divergent tropism and biological properties of these viruses. D 2005 Elsevier Inc. All rights reserved. Keywords: Feline; Lentivirus; Receptor; CXCR4; Envelope Introduction Feline immunodeficiency virus (FIV), like HIV, infects lymphocytes, results in T cell activation and CD4/CD8 inversion, and ultimately leads to the death of the host due to immunological exhaustion (Ackley et al., 1990). In this respect, FIV is unique in its similarity to the pathogenic primate lentiviruses, as ungulate lentiviruses induce disease conditions that are more typical of a chronic inflammatory reaction (reviewed in Miller et al., 2000; Overbaugh et al., 1997). FIV infection of the domestic cat has been established as an animal model for AIDS, providing valuable insights into maternal – fetal and trans-mucosal transmission, design and testing of anti-retroviral agents, pediatric AIDS, vaccine design, and lentiviral pathogenesis (reviewed in Bendinelli et al., 1995; Elder et al., 1998; Willett et al., 1997b). Serologic surveys of 36 non-domestic feline species have revealed a minimum of 20 species with antibodies that cross-react with FIV antigens (Barr et al., 1995; Brown et al., 1993; Carpenter and O’Brien, 1995; Olmsted et al., 1992; Troyer et al., 2005). Genetic characterization of the lentiviruses associated with seroconversion in several of these species has determined that they are distinct from each other, related to domestic cat FIV, and have marked intra- and inter-strain genetic heterogeneity (Barr et al., 1997; Biek et al., 2003; Brown et al., 1994; Carpenter et al., 1996; Troyer et al., 2004, 2005). Additionally, sequence compar- ison of pol sequences from lion and puma lentiviruses (LLV, PLV; also referred to as FIV-Ple and FIV-Pco) demonstrated increased diversity, saturation of synonymous sites, and an increased proportion of substitutions at non-synonymous 0042-6822/$ - see front matter D 2005 Elsevier Inc. All rights reserved. doi:10.1016/j.virol.2005.07.024 * Corresponding author. Fax: +1 970 491 0523. E-mail address: [email protected] (S. VandeWoude). Virology 342 (2005) 60 – 76 www.elsevier.com/locate/yviro

Upload: natalia-smirnova

Post on 12-Sep-2016

212 views

Category:

Documents


0 download

TRANSCRIPT

  • ffe

    ctu

    era,

    e V

    tate U

    sity o

    for re

    ne 24

    valuable insights into maternalfetal and trans-mucosal

    transmission, design and testing of anti-retroviral agents,

    Biek et al., 2003; Brown et al., 1994; Carpenter et al., 1996;

    Troyer et al., 2004, 2005). Additionally, sequence compar-

    ison of pol sequences from lion and puma lentiviruses (LLV,

    -Pco) demonstrated

    Virology 342 (2005) 6Introduction

    Feline immunodeficiency virus (FIV), like HIV, infects

    lymphocytes, results in T cell activation and CD4/CD8

    inversion, and ultimately leads to the death of the host due

    to immunological exhaustion (Ackley et al., 1990). In this

    respect, FIV is unique in its similarity to the pathogenic

    primate lentiviruses, as ungulate lentiviruses induce disease

    conditions that are more typical of a chronic inflammatory

    reaction (reviewed in Miller et al., 2000; Overbaugh et al.,

    1997). FIV infection of the domestic cat has been

    established as an animal model for AIDS, providing

    pediatric AIDS, vaccine design, and lentiviral pathogenesis

    (reviewed in Bendinelli et al., 1995; Elder et al., 1998;

    Willett et al., 1997b).

    Serologic surveys of 36 non-domestic feline species have

    revealed a minimum of 20 species with antibodies that

    cross-react with FIV antigens (Barr et al., 1995; Brown et

    al., 1993; Carpenter and OBrien, 1995; Olmsted et al.,

    1992; Troyer et al., 2005). Genetic characterization of the

    lentiviruses associated with seroconversion in several of

    these species has determined that they are distinct from each

    other, related to domestic cat FIV, and have marked intra-

    and inter-strain genetic heterogeneity (Barr et al., 1997;lions are hosts for apparently apathogenic lentiviruses (PLV, LLV) distinct from FIV. We compared receptor use among these viruses by: (1)

    evaluating target cell susceptibility; (2) measuring viral replication following exposure to specific and non-specific receptor antagonists; and

    (3) comparing Env sequence and structural motifs. Most isolates of LLV and PLV productively infected domestic feline T cells, but differed

    from domestic cat FIV by infecting cells independent of CXCR4, demonstrating equivalent or enhanced replication following heparin

    exposure, and demonstrating substantial divergence in amino acid sequence and secondary structure in Env receptor binding domains. PLV

    infection was, however, inhibited by CD134/OX40 antibody. Thus, although PLV and LLV infection interfere with FIV superinfection, we

    conclude that LLVand PLV utilize novel, more promiscuous mechanisms for cell entry than FIV, underlying divergent tropism and biological

    properties of these viruses.

    D 2005 Elsevier Inc. All rights reserved.

    Keywords: Feline; Lentivirus; Receptor; CXCR4; EnvelopeFeline immunodeficiency virus (FIV) causes fatal disease in domAbstract

    estic cats via T cell depletion-mediated immunodeficiency. Pumas andFeline lentiviruses demonstrate di

    envelope stru

    Natalia Smirnovaa, Jennifer L. Troy

    Mary Possb, Su

    aDepartment of Microbiology, Immunology, and Pathology, Colorado SbDivision of Biological Sciences, Univer

    Received 20 April 2005; returned to author

    Available onli0042-6822/$ - see front matter D 2005 Elsevier Inc. All rights reserved.

    doi:10.1016/j.virol.2005.07.024

    * Corresponding author. Fax: +1 970 491 0523.

    E-mail address: [email protected] (S. VandeWoude).rences in receptor repertoire and

    ral elements

    Jennifer Schisslera, Julie Terweea,

    andeWoudea,*

    niversity, 1619 Campus Delivery, Fort Collins, CO 80523-1619, USA

    f Montana, Missoula, MT 59812, USA

    vision 9 June 2005; accepted 20 July 2005

    August 2005

    0 76

    www.elsevier.com/locate/yviroPLV; also referred to as FIV-Ple and FIVincreased diversity, saturation of synonymous sites, and an

    increased proportion of substitutions at non-synonymous

  • irologsites when compared to sequences derived from FIV-

    infected cats (Biek et al., 2003; Brown et al., 1994;

    Carpenter et al., 1996, 1998; Troyer et al., 2004). These

    observations suggest that LLV and PLV have been main-

    tained in their host lineages longer than FIV has been in

    domestic cat populations.

    The clinical effects of the FIVs indigenous to wild and

    captive non-domestic cat populations have not been well

    studied. However, given high seroprevalence rates in the

    absence of indications of decreased population fitness, it

    appears likely that these infections are not responsible for

    high levels of pathology in free-ranging populations (Biek et

    al., 2003; Brown et al., 1994; Carpenter et al., 1996).

    Additionally, several strains of PLV and LLV can produc-

    tively infect domestic cats without resulting in disease,

    suggesting that viral as well as host factors mediate

    pathogenicity and disease outcome (VandeWoude et al.,

    1997a, 2000, 2002, 2003). PLV-1695, an isolate originally

    obtained from a British Columbia cougar, is infectious for

    domestic cats both parenterally and oronasally, and mucosal

    infection was contained in three of four cats in the absence

    of specific markers of enhanced host immune response.

    Moreover, this isolate demonstrated a greater propensity to

    target tissues of the gastrointestinal tract than FIV (Terwee

    et al., 2005). These observations support the supposition

    that LLV and PLV are less pathogenic and, at least in a

    domestic cat model, have significantly different biological

    properties than FIV.

    The virusreceptor interaction is the primary event in the

    process of lentivirus infection, is a critical determinant of

    cell tropism, and significantly influences the pathogenesis of

    disease. For example, human immunodeficiency virus

    targets helper T cells preferentially as these cells express

    the viral receptor CD4 (Klatzmann et al., 1984; Maddon et

    al., 1986), and variation at the level of viral receptor

    specificity underlies the classification of HIV isolates into

    syncytium-inducing (SI, X4-adapted strains; Feng et al.,

    1996) and non-syncytium-inducing (NSI, R5 strains;

    Alkhatib et al., 1996; Choe et al., 1996; Deng et al., 1996;

    Doranz et al., 1996; Dragic et al., 1996). It is generally

    thought that R5 tropic strains are the initial infecting

    subtype, and as a result, gastrointestinal tissues expressing

    high levels of CCR5 are targeted during initial infection

    (Apetrei et al., 2004). As disease progresses, X4-adapted

    strains are increasingly isolated, and at end stage of disease

    become the primary virus in circulation (Apetrei et al.,

    2004). Pathogenicity has been linked to X4 and R5 usage in

    both SIV and SIV-HIV recombinant (S/HIV) systems

    (Nishimura et al., 2004), and a paucity of CD4+CCR5+

    target cells has been correlated with lack of pathogenicity in

    natural SIV infections of African Green Monkeys (Allan et

    al., 2004). Receptor tropism has been mapped to the Env

    region of HIV and SIV, particularly in the V3 loop (Carrillo

    and Ratner, 1996; Chesebro et al., 1996; Johnston and

    N. Smirnova et al. / VPower, 2002; Kato et al., 1999; Pohlmann et al., 2004;

    Speck et al., 1997). Similarly, pathogenicity and cell tropismof FIV have also been linked to env V3 sequence (Hohdatsu

    et al., 1996; Hosie et al., 2002; Johnston and Power, 2002;

    Lerner and Elder, 2000; Pedersen et al., 2001; Verschoor et

    al., 1995). As described above, PLV-1695 localizes in the

    gastrointestinal tract tissues and draining lymph nodes in

    cats infected parenterally or mucosally, unlike domestic cat

    FIV, which is more highly tropic for thymus and bone

    marrow (Terwee et al., 2005).

    The chemokine receptor CXCR4 has been shown to act

    as a co-receptor for FIV, suggesting conservation of

    receptor usage between the feline and primate lentiviruses

    (Willett et al., 1997a, 1997c, 1998). However, despite the

    fact that FIV pathogenicity closely mimics that of HIV, FIV

    infection has not been demonstrated to involve interaction

    with CD4 and there are no firm data to support a role for

    CCR5 in infection of feline cells (Johnston and Power,

    1999; Johnston et al., 2001; Lerner and Elder, 2000).

    However, CD134/OX40, a member of the tumor necrosis

    factor/nerve growth factor-receptor superfamily, has been

    identified as a high-affinity receptor for field strains of FIV

    (de Parseval et al., 2004a; Shimojima et al., 2004); and

    binding mechanics between FIV Env and CD134 mimic

    that of HIV Env and CD4 (de Parseval et al., 2005). CD134

    is expressed primarily on activated CD4+ T cells, plays a

    role in CD4+ expansion, and has been implicated in

    modulating cell adhesion in HTLV-1 infection (Uchiyama,

    1997; Weinberg et al., 2004). In contrast to FIVs isolated

    from cats during the asymptomatic stage of disease, tissue

    culture-adapted FIVs appear to utilize CXCR4 as a sole

    receptor, as do FIVs isolated from cats with end stage

    immunodeficiency disease (Haining et al., 2004). These

    findings illustrate a virusreceptor interaction that varies

    with pathogenicity as observed in the primate lentivirus

    system. For these reasons, we have compared viral

    receptor interactions and env sequences between patho-

    genic domestic cat FIV and the presumably less pathogenic

    non-domestic feline lentiviruses.

    Our initial experimental data suggested that FIV and

    PLV have overlapping receptor repertoires, as viral

    interference has been observed in vitro and in vivo, and

    lion and puma lentiviruses were able to infect domestic cat

    PBMC and cell lines (VandeWoude et al., 1997a, 1997b).

    The aim of this study was to further characterize receptor

    use by these apparently less pathogenic, more highly host-

    adapted viruses. We have investigated the use of CD134,

    CXCR4, CD4, and CD25 by FIV, PLV, and LLV. We have

    also evaluated non-specific binding interactions via heparin

    and cyanovirin-N (CVN-N) interference. The data pre-

    sented here demonstrate that the non-domestic feline

    lentiviruses use different receptors and binding modalities

    than FIV. Comparisons of FIV- and PLV-Env amino acid

    sequences reveal substantial differences in biochemical

    properties in the V3 loop, providing a rationale for

    variation in receptor use and suggesting a plausible

    y 342 (2005) 6076 61mechanism for the observed biological differences noted

    among the feline lentiviruses.

  • Results

    Non-domestic cat lentiviruses replicate in domestic cat cells

    We used a representative panel of primary feline

    lymphoid cells, lymphoid cell lines, and adherent cells to

    assess in vitro growth characteristics of several strains of

    LLV and PLV. As previously described, several isolates of

    PLV and LLV are able to grow on domestic cat cells in vitro

    (Table 1; VandeWoude et al., 2003, 1997a). The feline

    lymphoid cell lines 3201 and Mya-1 cells were more likely

    to support growth of non-domestic cat lentiviruses than

    adherent cell lines. LLV was more difficult to propagate in

    vitro than PLV, though several isolates were recovered

    following co-cultivation with Mya-1 cells. PLV-1695 and

    FIV-C-PG were also able to replicate when grown on puma-

    derived PBMC stocks, and conversely, FIV-C-PG was

    replication competent on puma-origin PBMC. Thus, domes-

    effect of AMD3100 on infection with LLV and PLV.

    Twenty-four hour exposure to high concentrations of

    AMD3100 efficiently and reproducibly inhibited FIV

    infection in a dose-dependent manner as previously reported

    (Figs. 1A and 3A, Tables 2 and 3). Figs. 1B and C

    demonstrate that comparatively, LLV and PLV are not

    inhibited by AMD3100 in a dose-dependent manner. In

    order to compare results across all three viruses despite

    differences in assay technique (ELISA vs. reverse-tran-

    scriptase) and wide variation in raw data ranges, viral

    growth in the presence of inhibitor is presented as a

    percentage of growth in the presence of no AMD control

    in Fig. 1D and Figs. 2, 4, 5, and 6. Values from the raw data

    for these figures, and subsequent statistical comparisons of

    percent inhibition, are presented in Table 3.

    Comparisons of time course data between FIV and PLV

    exposed to lower concentrations of AMD3100 demonstrated

    that although the duration of FIV growth inhibition persisted

    ivoa

    = 25 background, nd = not done. Results ba In vivo virulence, see (VandeWoude et al., 1997b; VandeWoude et al., Tb 3201feline lymphosarcoma.c CrFKCrandell feline kidney.d Mya-1IL-2-dependent feline T cells.for several weeks post-inoculation (Fig. 2A), PLV infection

    was only inhibited at doses above 0.25 Ag/ml, and then onlytransiently (Fig. 2B). In order to evaluate the possibility that

    this transient inhibition was attributable to puma virus

    adaptation to domestic cat cells, AMD3100 inhibition was

    tested on puma-derived PBMC alongside Mya-1 cells. As

    previously noted in Mya-1 cells, FIV-C-PG growth was

    completely inhibited, and PLV-1695 growth was transiently

    inhibited at the highest concentration. AMD3100 also

    blocked FIV infection of puma PBMC; however, in contrast,

    PLV-1695 growth was not inhibited by AMD3100 in puma

    cells at any concentration (Fig. 3).

    Taken collectively, these results reveal that: (1)

    AMD3100 potently blocks FIVCXCR4 interactions;

    dilute concentrations of this drug present for 1 h are

    capable of significant reduction in viral replication greater

    than 2 weeks post-exposure; (2) high concentrations of

    AMD3100 are capable of inducing transient reductions in

    In vitro tropism

    Lion PBMC Cat PBMC 3201b CrFKc Mya-1d

    nd +++ +++ ++++ +++

    nd +++ +++nd +++ ++ ++nd +++ ++++ ++ ++++ nd ++

    +++ ++ ++++ + ++ ++++ +++ + + ++++ nd ++

    2 background; + = 25 background, ++ = 510 background, +++ =on at least duplicate estimations monitored for 28 days after infection.e et al., 2005).

  • irologN. Smirnova et al. / VPLV (and possibly LLV) cellular entry on domestic cat-

    derived cells; and (3) FIV infection of puma PBMC-

    derived cells is CXCR4 dependent, while PLV infection is

    Fig. 1. High dose AMD3100 inhibits FIV more significantly than LLV- or

    PLV-growth. (AC) Triplicate wells containing 3201 Tcells were exposed to

    a range of high concentrations of AMD3100, then inoculated with 10 TCID50FIV-B-2542, LLV-458, or PLV-1695. Cells were washed at 24 h and re-

    exposed to the same AMD concentrations as on day 0, providing an extended

    period of inhibitor exposure to maximize inhibition of PLV and LLV.

    Supernatants were collected bi-weekly for 3 weeks and assayed as described

    in Materials and methods. FIV growth was inhibited at all concentrations as

    illustrated in panel A for day 10 post-infection (PI). Replication of LLVand

    PLV was transiently inhibited (LLVon day 14 and PLVon day 10 PI only),

    but the trend was not statistically significant (B, C). Because y-axis scale

    range did not allow single graph comparisons, data were converted to %-no-

    inhibitor control (as described in Materials and methods) in panel D and in

    Figs. 2, 4, 5, and 6. Bars represent average and standard deviation of triplicate

    assays. Indicates P < 0.001 (see Table 3).not, as evidenced by response to the presence to CXCR4

    antagonist AMD3100.

    In order to evaluate the possibility that LLV and PLV

    attached to CXCR4 in a manner not disrupted by

    AMD3100, we attempted to block viral entry with anti-

    CXCR4 antibody. In contrast to AMD3100 interference, this

    agent did not inhibit infection by any of the feline

    lentiviruses. Others have observed that FIV infection was

    not inhibited by anti-CXCR4 antibody, a phenomenon

    attributed to the disparate sites of antibody vs. FIV binding

    (de Parseval et al., 2004b). SDF1-a, the natural ligand forCXCR4, inhibited FIV growth as expected, albeit to a lesser

    extent than AMD3100. Others have reported weaker

    inhibition of FIV growth by SDF1-a than by AMD3100,which may be observed because of cellular CXCR4

    upregulation following exposure to its ligand (Hosie et al.,

    1998). PLV growth was not abrogated in the presence of

    SDF1-a in multiple trials. LLV was only transientlyinhibited by SDF1-a in 2/5 trials, again demonstrating thatinfection of domestic cat cells by non-domestic FIVs is less

    sensitive to disruption of CXCR4 binding than the domestic

    cat virus (Table 2).

    Blocking CD134 significantly inhibits FIV, transiently

    inhibits PLV, and does not inhibit LLV in vitro

    The recent identification of CD134 as a primary FIV

    receptor (de Parseval et al., 2004a; Shimojima et al., 2004)

    led us to investigate whether this molecule also served as a

    receptor for LLVor PLV cell infection. Several experiments

    were conducted to determine the cross reactivity of human

    anti-CD134 with domestic cat CD134. Mean fluorescence

    intensity (MFI) correlated with antibody concentration over

    a range of 1 to 20 Ag/ml on Mya-1 cells (Fig. 4A).Approximately 13% of Mya-1 cells were labeled using 5

    Ag/ml antibody, whereas less than 5% of cells were labeledwhen the concentration was decreased to 2 Ag/ml (data notshown).

    Because of a generalized suppression of cell growth

    following addition of antibody (which precluded analysis in

    concentrations greater than 5 Ag/ml), isotype matchedirrelevant antibody (rather than no antibody) was used in

    similar dilutions as non-specific immunoglobulin control.

    Growth of FIV-C-PG was consistently and significantly

    inhibited on Mya-1 cells by anti-CD134 at a concentration

    of 5 Ag/ml compared to irrelevant isotype control antibody(P < 0.001), but was not inhibited at 2 Ag/ml (Fig. 4B,Table 3). PLV-1695 was also significantly inhibited by

    CD134 antibody at 5 Ag/ml (P < 0.001, Fig. 4B, Table 3)but not 2 Ag/ml, though percent inhibition was less than forFIV. LLV-458 was not significantly inhibited by 5 Ag/ml ofanti-CD134 antibody (P = 0.058; Fig. 4B). These data are

    similar to observations of AMD3100 inhibition, i.e. strong

    support for FIV use of CD134 for cellular entry that cannot

    y 342 (2005) 6076 63be replicated in identical inhibition experiments with PLV

    or LLV.

  • irologTable 2

    Summary of inhibition studies

    Inhibitor Virus Cell type Trials Result

    AMD-3100 FIV-B-2542 3201, Mya 3/3 Dose-dependent

    inhibition

    LLV-458 3201, Mya 2/2 Transient inhibition

    PLV-1695 3201, Mya 5/5 Transient inhibition

    FIV-C-PG Puma 1/1 Complete inhibition

    PLV-1695 Puma 5/5 Transient inhibition

    SDF-1a FIV-B-2542 3201,FePBMC

    3/3 Dose-dependent

    inhibition

    FIV-C-PG Mya 1/1 Dose-dependent

    inhibition

    LLV-458 3201, 3/5 No inhibition

    N. Smirnova et al. / V64Saturation of CD4 and CD25 surface molecules does not

    inhibit FIV, LLV, or PLV infection

    Given that blocking of either CXCR4 or CD134 did not

    fully inhibit the non-domestic lentiviruses, other surface

    molecules were assayed for a possible role in viral binding.

    Mya-1 cells are the only cell type found thus far that are

    permissive for all three feline lentiviruses (FIV, PLV, and

    LLV). These cells are CD4+CD25+ (data not shown).

    Therefore, the possible role of these surface molecules in

    viral entry was determined. Incubation of Mya-1 cells with

    high, medium, or low concentrations of anti-CD4 and anti-

    CD25 either alone or concurrently failed to inhibit FIV, PLV,

    and LLV infection (Tables 2 and 3).

    FePBMC

    LLV-458 Mya,

    FePBMC

    2/5 Transient inhibition

    PLV-1695 3201, Mya,

    FePBMC

    5/5 No inhibition

    Anti-CD134 FIV-C-PG Mya 1/1 Dose-dependent

    inhibition

    PLV-1695 Mya 1/1 Dose-dependent

    inhibition

    LLV-458 Mya 1/1 Marginal

    inhibition

    Anti-CD4 FIV-C-PG Mya 1/1 No inhibition

    LLV-458 Mya 1/1 No inhibition

    PLV-1695 Mya, 3201 3/3 No inhibition

    Anti-CD25 FIV-C-PG Mya 1/1 No inhibition

    LLV-458 Mya 1/1 No inhibition

    PLV-1695 Mya 1/1 No inhibition

    Anti-X4 PLV-1695 Mya 2/2 No inhibition

    TAK 779 PLV-1695 3201 2/2 No inhibition

    Cyanovirin FIV-C-PG Mya 2/2 Dose-dependent

    inhibition

    PLV-1695 Mya 2/2 No inhibition

    LLV-458 Mya 1/1 Dose-dependent

    inhibition

    Heparin FIV-C-PG Mya 1/1 Dose-dependent

    inhibition

    LLV-458 Mya, 3201 4/4 Enhancement

    PLV Mya, 3201 3/3 Enhancement

    Each trial consisted of samples run in triplicate or in sets of five

    (cyanovirin, anti-CD134, anti-CD25) at each inhibitor concentration.

    Transient inhibition is defined as dose-dependent inhibition noted only at

    the earliest timepoint that no-inhibitor control supernatants were RT

    positive. Enhancement is defined as inhibitor RT values significantly

    greater than no inhibitor control at multiple timepoints post-inoculation.Heparin inhibits FIV, but enhances LLV and PLV infection

    In addition to specific receptor binding, non-specific

    interactions between lentiviral envelope and target cell

    membranes are involved in HIV, SIV, and FIV infection.

    For example, heparin binding has been shown to play a role in

    both HIV- and FIV-cellular entry, purportedly through

    interactions between basic gp120 (gp95) with anionic cellular

    heparin sulfates (de Parseval and Elder, 2001; Ibrahim et al.,

    1999; Roderiquez et al., 1995; Tanabe-Tochikura et al.,

    1992). To better define non-specific interactions that might

    assist infection with LLV and PLV, we infected cells in the

    presence of heparin. FIV-C-PG infection was inhibited in a

    dose-dependent manner (P < 0.05) by heparin as previously

    reported for FIV-34TF10 and FIV-Pet (de Parseval and Elder,

    2001). In striking contrast, LLV and PLV infection was not

    inhibited, and in fact, viral growth in the face of heparin

    exceeded no-heparin controls in 4 of 4 (LLV) or 3 of 3 (PLV)

    trials (Fig. 5, Tables 2 and 3), suggesting substantial

    differences in non-specific binding interactions between

    domestic and non-domestic FIVs.

    High mannose glycosylation contributes to FIVand LLV, but

    not PLV, Envreceptor interactions

    CVN-N, an antibiotic which blocks high-mannose

    glycosylation sites on viral Env, interferes with both HIV

    and FIV receptor binding and subsequent infection in a non-

    specific manner (Barrientos et al., 2003; Bolmstedt et al.,

    2001; Dey et al., 2000; Mori and Boyd, 2001; OKeefe et

    al., 2000, 2003), and has recently been shown to interfere

    with vaginal and rectal transmission of SIV (Tsai et al.,

    2003, 2004). This activity is based upon binding of the

    molecule to envelope glycoproteins (specifically high-

    mannose oligosaccharides) which interferes with Env

    receptor binding and fusion. Fig. 6 demonstrates the ability

    of CVN-N to inhibit infection of LLV on Mya-1 cells,

    though results were not strictly dose dependent. FIV and

    PLV infection was only inhibited at one dose and timepoint

    (Fig. 6, Table 3). However, growth of all three viruses was

    blunted in the presence of any concentration of CVN-N.

    These results demonstrate that CVN-N stringency and

    efficiency of in vitro FIV-, LLV-, or PLV-growth inhibition

    are highly variable compared to the specific receptor

    antagonists AMD3100 and anti-CD134.

    FIV Env sequence and biochemical properties differ

    substantially from non-domestic cat lentiviral Env

    Given that Env has been shown to be the principal

    lentiviral receptor-binding domain, we compared Env

    among 2 PLV strains (PLV-1695 and PLV-14), and 2 FIV

    strains (Petaluma and C-PG). Addition of lentiviral-Env

    sequence obtained from a Pallas cat FIV-like virus

    y 342 (2005) 6076(Otocolobus manul, FIV-Oma) assisted in bridging align-

    ments of highly divergent regions of FIV- and PLV-Env.

  • Table 3

    Statistical analysis of inhibition studies represented in figures

    Virus AMD (Ag/ml) Viral growth Dose vs. control 2-tailed Students t Sequential dose 2-tailed Students t

    FIV 0 0.781 T 0.084

    0.3 0.098 T 0.006 P < 0.001 P < 0.05

    1.25 0.076 T 0.008 P < 0.001 P < 0.015 0.034 T 0.004 P < 0.001

    PLV 0 13,061 T 4626

    0.3 7533 T 3377 nsd nsd

    1.25 9215 T 3261 nsd nsd5 8901 T 4135 nsd

    LLV 0 1138 T 1044

    0.3 435 T 296 nsd nsd

    1.25 362 T 195 nsd nsd5 310 T 36 nsd

    Virus Antibody Ag/ml Viral growth Dose vs. control 2-tailed Students t Sequential dose 2-tailed Students t

    FIV Control 2 1.48 T 0.41

    Anti-CD134 1.20 T 0.27 nsdControl 5 1.17 T 0.11

    Anti-CD134 0.34 T 0.17 P < 0.001 P < 0.001

    PLV Control 2 335 T 82Anti-CD134 352 T 51 nsd

    Control 5 394 T 66

    Anti-CD134 218 T 40 P < 0.01 P < 0.05

    LLV Control 2 677 T 325Anti-CD134 698 T 263 nsd

    Control 5 567 T 120

    Anti-CD134 403 T 113 nsd nsd

    Virus Treatment Viral growth Dose vs. control 2-tailed Students t Sequential dose 2-tailed Students t

    FIV Control 1.845 T 0.055

    Anti-CD4+anti-CD25 1.900 T 0.133 nsd nsd

    PLV Control 816 T 152Anti-CD4+anti-CD25 1013 T 418 nsd nsd

    LLV Control 768 T 118

    Anti-CD4+anti-CD25 585 T 184 nsd nsd

    Virus Heparin (Ag/ml) Viral growth Dose vs. control 2-tailed Students t Sequential dose 2-tailed Students t

    FIV 0 0.374 T 0.260

    2.5 0.165 T 0.159 P < 0.05* nsd

    5 0.029 T 0.018 P < 0.05* nsd

    20 0.022 T 0.055 nsdPLV 0 511 T 550

    2.5 Not done

    5 1020 T 1150 nsd nsd

    20 646 T 809 nsd nsdLLV 0 331 T 52

    2.5 773 T 491 P < 0.10* nsd

    5 508 T 227 nsd20 508 T 105 P < 0.05* nsd

    Virus CV-N (Ag/ml) Viral growth Dose vs. control 2-tailed Students t Sequential dose 2-tailed Students t

    FIV 0 2.19 T 0.25

    1 1.72 T 0.70 nsd nsd10 0.38 T 0.39 P < 0.001 P < 0.01

    PLV 0 1683 T 304

    1 776 T 599 P < 0.05 nsd

    10 1316 T 772 nsd nsdLLV 0 1473 T 444

    1 1226 T 684 nsd nsd

    10 497 T 601 P < 0.05 nsd

    Statistical comparisons are made using 2-tailed two-sample homoscedastic Students t test except where indicated. Values for FIV represent OD450nm ELISA

    absorbance values. Values for PLV and LLV represent cpm (counts per minute) in reverse-transcriptase assays.

    * Indicates 1-tailed two-sample homoscedastic Students t test.

    N. Smirnova et al. / Virology 342 (2005) 6076 65

  • positive charge noted in this region among domestic cat

    FIVs (+8 to +12). Three to 6 additional N-glycosylation

    sites were predicted in non-domestic lentiviruses in com-

    parison with FIV, and 56 fewer cysteine residues were

    present in PLV vs. FIV. A similar trend was noted in V3, the

    region implicated as receptor binding domain for FIV

    (Hohdatsu et al., 1996; Sodora et al., 1994; Verschoor

    et al., 1995; Willett and Hosie, 1999). In contrast, the more

    conserved regions flanking V3V5 were more positively

    charged in non-domestic lentiviruses than FIV, and con-

    tained similar numbers of cysteine residues and putative

    N-glycosylation sites (Fig. 7, Table 4). Such differences

    indicate that substantial divergence occurs in secondary and

    tertiary structures at the receptor-binding region of PLV- vs.

    FIV-Env, providing an explanation for the differential

    susceptibilities to receptor inhibitors, and supporting the

    hypothesis that PLV uses a different receptor to gain cellular

    entry than FIV.

    Discussion

    irology 342 (2005) 6076Fig. 2. Low dose AMD3100 inhibits FIV-, but only transiently inhibits

    PLV-growth. Triplicate wells containing 3201 T cells were exposed to a

    N. Smirnova et al. / V66Sequences were aligned to compare structural regions,

    particularly those known to be important for receptor

    binding. Domestic cat FIV Env differed from non-domestic

    cat lentiviruses at approximately 77% of the amino acid

    sites. However, the sequence divergence was not uniform in

    that regions of relative homology (3050%) were inter-

    rupted by long tracts of unalignable sequence (12%homology). Despite substantial sequence disparity, three

    hydrophobic regions were conserved in similar positions

    across all feline lentiviruses, allowing more probable align-

    ments within the non-homologous regions (Fig. 7). The

    V3V5 region of PLV shared least homology with FIV Env

    (corresponding to aa 365 to 627 in the env alignment and

    defined in Pancino et al., 1993). In FIV, this region has been

    shown to contain the CXCR4 binding site (Willett et al.,

    1997a), neutralizing antibody binding sites (Lombardi et al.,

    1994, 1995; Tozzini et al., 1993) and several epitopes

    important for cell tropism and cell line adaptation (Hohdatsu

    et al., 1996; Vahlenkamp et al., 1997; Verschoor et al.,

    1995). Within the V3V5 region, several biochemical

    differences are notable that suggest corresponding differ-

    ences in secondary structure and function (Table 4). Total

    charge was negative in all three non-domestic lentivirus

    isolates (1 to 4); this differs substantially from the highly

    The data presented here strongly support the conclusion

    that lion and puma lentiviruses have different mechanismsrange of mid-to-low range concentrations of AMD3100, then inoculated

    with 10 TCID50 FIV-B-2542 (A) and PLV-1695 (B). Cells were washed at 1

    h and fed with AMD-free media. Supernatants were collected and processed

    as described in Materials and methods. As previously described, FIV was

    inhibited in a dose-dependent manner at all concentrations over a 2 week

    growth period (A). PLV replication was initially inhibited in an

    approximately does-dependent manner (days 810 PI, B) but the trend

    did not persist after day 15 PI (growth exceeds no AMD control, B). Bars

    represent the average % inhibition of triplicate assays. *Indicates P < 0.05

    (see Table 3).Fig. 3. AMD 3100 inhibits FIV-, but not PLV-, growth in domestic cat cells

    and puma-derived cells. Mya-1 (domestic cat) cells or puma PBMC-derived

    cells (2 105) were exposed to AMD 3100 at concentrations 5 Ag/ml, 1.25Ag/ml, and 0.3 Ag/ml, then inoculated with 10 TCID50 FIV-C-PG (A) andPLV-1695 (B). Experiments were performed in quintiplicate. Cells were

    incubated overnight (18 h), washed, and re-exposed to the same

    concentrations of AMD 3100 as on day 0. Supernatants were collected

    bi-weekly and subjected to reverse transcriptase assay. RT values shown are

    from day 10 (Mya-1 cell) or day 14 (puma cells) PI. (A) FIV growth was

    significantly inhibited at all concentrations in both cell types. (B) PLV

    growth was slightly inhibited at the highest AMD dose in Mya-1 cells, andnot for any AMD concentrations on puma cells. Indicates P < 0.01;

    *indicates P < 0.05 (see Table 3).

  • Fig. 5. Heparin inhibits FIV- but enhances LLV- and PLV-growth. Nave

    Mya-1 cells were incubated with decreasing dilutions of heparin for 1 h.

    FIV-C-PG, LLV-458, and PLV-1695 supernatant stocks were then plated on

    Mya-1 cells and processed as described in Materials and methods. Data are

    shown for day 7 PI for LLV and FIV, day 14 PI for PLV. Bars represent

    average of % inhibition in three assays. Whereas FIV replication was

    irology 342 (2005) 6076 67N. Smirnova et al. / Vfor cell entry than those of domestic cat FIV as manifested

    by the following findings: (a) AMD3100 and SDF-1a onlytransiently inhibited PLV and LLV replication relative to

    FIV in domestic cat cells, and, in contrast to FIV-C-PG,

    PLV-1695 replication was not impaired by AMD3100 in

    puma-derived PBMC; (b) replication of LLV and PLV on

    Mya-1 cells was not inhibited by heparin; and (c) V3V5

    Env differs substantially between FIV and two strains of

    PLV in amino acid sequence, charge, potential post-trans-

    lational modification sites, and cysteine content. The role of

    CD134 binding in LLV and PLV infection is less clear and

    discussed further below.

    Puma and lion lentiviruses infected domestic cat PBMC

    and T cell lines, particularly Mya-1 cells, demonstrating the

    capacity of LLV and PLV to adapt to use domestic cat cell

    surface moieties to gain cellular entry. The Mya-1 cell line

    was derived from feline blood mononuclear cells, and is

    readily infected with primary isolates of FIV (Miyazawa et

    Fig. 4. Anti-CD134 inhibits FIV- and PLV- but not LLV-growth. (A) Mya-1

    cells were incubated with dilutions of anti-human CD134 (OX40, Ancell

    Corporation: clone Ber Act 35) or Isotype control (Mouse IgG1, Ancell

    Corporation: clone MOPC 31C) for 30 min at 4 -C, washed, and stained

    using Anti-Mouse IgG FITC, and subjected to flow cytometry as described

    in Materials and methods. Mean fluorescence intensity decreased with anti-

    CD134 concentration. Twelve fifteen percent of Mya-1 cells were labeled

    at 5 Ag/ml compared to

  • intracellular enzymes responsible for interruption of trans-

    species viral replication (Gaddis et al., 2004; Mariani et al.,

    2003; Schrofelbauer et al., 2004).

    Primary strains of FIV are dependent upon both CD134

    and CXCR4 for cell entry (de Parseval et al., 2004a,

    2004b; Egberink et al., 1999; Hosie et al., 1998;

    Richardson et al., 1999; Shimojima et al., 2004), restricting

    their growth to CXCR4+ CD134+ target cells. Tissue

    culture adaptation can occur, rendering strains CD134

    independent, explaining the variation in cell susceptibilities

    secondary structure, provides plausible explanation for the

    results of inhibition experiments presented here. Our obser-

    vation of enhanced growth may occur because neutralization

    of anionic heparan sulfates by heparin facilitates binding of a

    neutral or anionically charged V3 loop found on PLV, and

    possibly LLV, isolates.

    Glycosylation of HIV Env is considered a major compo-

    nent of cell tropism and receptor targeting, and CVN-N has

    been shown to inhibit in vitro FIV infection, inferring a

    similar mechanistic property (Dey et al., 2000). Despite the

    show

    use o

    from

    sites a

    boxed

    grey

    >25%

    d C1

    ). Epit

    fined

    N. Smirnova et al. / Virology 342 (2005) 607668noted in Table 1. We found that anti-CD134 and the

    CXCR4 inhibitor, AMD3100 markedly disrupted in vitro

    infection by FIV-C-PG. Though CXCR4 blocking tran-

    siently inhibited LLV and PLV replication, in neither case

    was the degree of blocking as consistent and marked as for

    FIV. The fact that AMD3100 was ineffective at blocking

    PLV-1695 infection in puma-derived cells suggests that

    adaptation of PLV in domestic cat cells may confer some

    low-level degree of CXCR4 dependency that is rapidly

    reversed under pressure of an antagonist. We noted dose-

    dependent inhibition of PLV growth in the presence of

    anti-CD134 antibody. In light of recent reports that the

    binding site for anti-CD134 monoclonal antibody clone

    Ber Act 35 does not overlap with FIV-Env binding sites on

    CD134 for other FIV strains (de Parseval et al., 2005), and

    that two strains of PLV (PLV-14, CoLV) were able to

    infect X4CD134 AH927 cells (B. Willett, personalcommunication), further investigations using other strat-

    egies will be required to clarify the precise association

    between LLV and PLV usage of CXCR4 and CD134.

    Heparin binding did not interfere with LLV and PLV

    infection, in contrast to its ability to block FIV infection in

    vitro. The HIV/FIV blocking activity of heparin is thought to

    be dependent upon electrostatic interactions between the

    basic FIV V3 loop and acidic cell-bound heparin sulfates

    (Roderiquez et al., 1995). The V3 binding domain amino acid

    sequences of two highly divergent PLVs (PLV-14 and PLV-

    1695) have net charges substantially more acidic than FIV. de

    Parseval and Elder found that FIV strains with net charges of

    +6 or +8 in V3 were more significantly inhibited by heparin

    than strains with charges of +4 (de Parseval and Elder, 2001).

    Assuming that the V3 loop of PLV functions as a potential

    cellular binding domain, the neutral to anionic charge of this

    region, combined with the highly divergent amino acid and

    Fig. 7. Predicted amino acid alignment of domestic and non-domestic FIVs

    correlative sites from FIV-Pet Env. These numbers differ between strains beca

    672 (610 and 611 in the FIV-pet reference sequence) indicated the transition

    Cysteine residues, FC_, are indicated by pink, and predicted N-glycosylationcontain white letters. Fully conserved amino acid residues are dark grey and

    across taxa (>0.5 in the Gonnet PAM250 matrix; Yang, 1997) are in medium

    the Gonnet PAM250 matrix; Yang, 1997) are in light grey. Regions that have

    are indicated by white boxes above the amino acid sequence rows and labele

    sequences (light green, entire variable region; dark green, V3 binding region

    rows; stronger binding sites are darker orange. Other structural features de(PID), alpha-helix, Trp-rich motif, transmembrane region) have also been indicated

    region (residues 365627), where non-domestic feline lentiviruses have lower chfact that PLV-1695 Env sequence predicts a significant

    number of potential glycosylation sites, CVN-N inhibition

    experiments reported were unable to conclusively demon-

    strate dose-dependent inhibition of either this virus or LLV.

    These ambivalent results are likely attributable to variation in

    glycosylation patterns of the virus stocks used, since this

    variable is dependent upon the cell in which viral propagation

    occurs (Levy, 1998). It is also possible that the selectivity of

    CV-N for terminal Man(12)Man moieties on high-mannose

    Man8 or Man9 glycans precludes inhibition of glycosylation

    signatures of the virus stocks used in this study (Bewley and

    Otero-Quintero, 2001).

    Because the LLV and PLV strains we used for blocking

    studies readily infect domestic cat cells, it is difficult to

    ascribe our observations to variation between surface

    molecules on domestic vs. non-domestic T cells. It is

    possible that adaptations in viral genomic sequence and

    structure occur when PLV or LLV infect domestic cat cells,

    suggested by our experiments in puma-derived PBMC.

    Observations by Biek et al. (2003) have demonstrated that

    sequence diversity within PLV is very low, and preliminary

    studies analyzing a series of PLV-1695 isolates across pol

    has not detected sequence selection in tissue culture relative

    to wild type virus (Poss et al., data not shown), so such

    adaptation events appear to be rare. SIV receptor use has

    been assessed based upon infection of cells displaying

    human receptor molecules, even though these viruses

    originated in African monkeys, and are often been main-

    tained in, or adapted to, macaque cells (Chen et al., 1997;

    Liu et al., 2000; Vodros et al., 2001, 2003). While lion and

    puma candidate receptor proteins require testing to validate

    observations made using domestic cat cells, analogy to

    primate lentivirus receptor usage would support the validity

    of the data presented here.

    s variable regions of structural homology. Numbers across the top are for

    f amino acid mismatches/insertions. A vertical line between residues 671 and

    the surface protein (gp120;SU) to the transmembrane protein (gp41:TM).

    re shown in yellow. Hydrophobic regions are indicated by black boxes that

    with bold letters. Sites that contain strongly conserved amino acid changes

    , and those with weakly conserved amino acid changes across taxa (

  • N. Smirnova et al. / Virology 342 (2005) 6076 69

  • domestic cat FIV. Further studies comparing amino acid

    structure of LLV-Env to PLV and FIV-Oma will allow an

    8

    0

    4

    4

    3

    ddy-G

    ley et

    r each

    amino

    presen

    irologWe have previously observed viral interference between

    PLV and FIV (VandeWoude et al., 2002), and have here

    noted similar in vitro tropisms, leading us to initially

    hypothesize that these viruses used similar receptors. The

    work presented here does not support this hypothesis. Other

    mechanisms underlying viral interference could include: (a)

    suppression of superinfection by innate immune mecha-

    nisms, similar to observations that HIV-2 prevents super-

    infection by R5 strains of HIV-1 by inducing h-chemokineproduction, or by actions of intracellular nucleases (Gaddis

    et al., 2004; Mariani et al., 2003; Schrofelbauer et al., 2004);

    (b) selection of PLV laboratory-cultured isolates with altered

    mechanisms for cell entry relative to primary strains

    (discussed above); (c) different receptor binding sites on

    CD134 or CXCR4 for PLV vs. FIV, attributable to

    variations in puma vs. domestic cat molecules; or (d)

    disruption of primary PLV viral receptor expression

    following initial infection with FIV. Viral glycosylation

    sites and point mutations in Env substantially influence

    receptor usage among HIV strains (McCaffrey et al., 2004;

    Nabatov et al., 2004). It is also possible that the biological

    viral isolates used here contained viral quasi-species with

    Table 4

    Comparison of Env amino acid sequence properties

    V3 binding V3 V3V5

    376416 354419 354567

    T N C T N C T N

    FIVA-PET 6 0 2 7 1 3 12

    FIVC-PG 5 0 2 5 1 3 8 1

    OMA 3 0 1 5 4 1 2 1PLV-14 3 0 1 4 4 1 4 1PLV-1695 4 0 1 4 5 1 1 1FIVA-Pet = FIV Petaluma strain (Olmsted et al., 1989); FIV-C-PG = FIVC-Pa

    et al., 1997) FIV sequence; PLV-14 = Florida Panther FIV sequence (Lang

    Amino acid residues corresponding to FIV-Pet sequence are indicated unde

    homology among all depicted strains, FPC3 = region demonstrating high

    between amino terminus of V5 loop and beginning of conserved region, re

    N. Smirnova et al. / V70varying receptor affinities due to slight variation in sequence

    and N-glycosylation, which do not reflect all subtypes of

    PLV and LLV.

    Perhaps the most intriguing finding presented here is

    the marked differences in FIV, PLV, and FIV-Oma Env

    amino acid sequences and alignments. Alterations in the

    V3 loop of FIV and HIV have been strongly correlated

    with changes in host cell susceptibility, receptor affinity,

    neutralizing antibody binding sites, and pathogenicity

    (Bjorndal et al., 1997; Carrillo and Ratner, 1996; Chesebro

    et al., 1996; Nielsen and Yang, 1998; Nishimura et al.,

    1996; Pohlmann et al., 2004; Siebelink et al., 1995; Speck

    et al., 1997; Verschoor et al., 1995). Therefore, the high

    degree of nucleotide and amino acid divergence noted here

    provides an attractive explanation for differential receptor

    use by non-domestic cat lentiviruses vs. FIV. It is

    interesting that FIV-Oma demonstrates an intermediate

    alignment between FIV and PLV, despite the fact that theopportunity to further study this trend using an African-

    origin FIV. The lack of inhibition of LLV and PLV by

    agents that reproducibly inhibit FIV or HIV, together with

    substantial differences noted in predicted Env structure will

    provide models to relate changes in receptor use and Env

    structure to lentiviral pathogenicity.

    Materials and methods

    Cells and viruses

    Heparinized blood samples were obtained from cats in a

    specific pathogen free breeding colony housed in anPallas cat is an Asian cat species vs. the North American

    dwelling puma. That the V3V5 region of PLV more

    closely aligns with that of the Pallas cat suggests these

    older and more host-adapted viruses might have evolved

    convergently to use similar receptor mechanisms which

    vary from the more recently emerged and virulent

    FPC2 V5-FPC3 FPC3

    283331 568658 659687

    C T N C T N C T N C

    14 2 1 3 0 0 0 1 0 0

    13 1 1 3 0 0 0 1 0 0

    8 1 2 3 8 0 0 6 0 0

    8 1 1 3 9 0 0 4 0 08 1 1 3 5 0 0 3 0 0ammer strain (de Rozieres et al., 2004); OMA= Pallas cat (O. manul; Barr

    al., 1994); PLV-1695 = British Columbia cougar FIV sequence (see text).

    column heading; FPC2 = region demonstrating high amino acid sequence

    acid sequence homology among all depicted strains; V5-FPC3 = region

    ting an area of intermediate homology among depicted strains.

    y 342 (2005) 6076AAALAC International accredited animal facility. Periph-

    eral blood mononuclear cells (PBMC) were isolated from

    heparinized blood by ficoll gradient centrifugation (Quack-

    enbush et al., 1990). Lion and puma nave PBMC were

    provided by Dr. Stephen OBrien as viably frozen field

    samples; Drs. Don Hunter and Caroline Krumm provided

    anti-coagulated blood collected from a free-ranging puma.

    The feline lymphoid cell lines 3201 (Hardy et al., 1980) and

    Mya-1 (Miyazawa et al., 1990, 1992) were grown under

    standard conditions (Quackenbush et al., 1990) using

    commercially obtained media (Invitrogen Life Sciences,

    Carlsbad, CA). Puma-derived PBMC infection experiments

    were performed on cells isolated from EDTA anti-coagu-

    lated blood using Histopaque (Sigma) and were cultured for

    2 months in RPMI-1640 supplemented with Glutamax I,

    non-essential amino acids, sodium pyruvate, glucose,

    sodium bicarbonate, l-glucose, antibiotics, concanavalin A

    at 5 Ag/ml, and recombinant human IL-2 at 100 units/ml.

  • irologConcanavalin A exposure was discontinued after 8 dou-

    blings (2 months). Cells used in experiments described had

    been in culture for approximately 4 months and replicated

    continuously when grown in IL-2.

    PLV-1695 (gift of Dr. Ken Langelier) was amplified in

    feline PBMC or Mya-1 cells as previously described

    (Terwee et al., 2005; VandeWoude et al., 1997b). LLV-

    458 and PLV-14 supernatants were obtained from Dr.

    Stephen J. OBrien, and stocks were amplified in domestic

    cat PBMC or Mya-1 cells in vitro (VandeWoude et al.,

    1997a, 1997b). Supernatants were harvested at peak RT

    activity (Willey et al., 1988). FIV-B-2542 and FIV-C-Paddy

    Gammer (FIVC-PG) stocks were generated in feline PBMC

    or Mya-1 cells using standard conditions (Quackenbush et

    al., 1990). These two field strains of FIV have been shown

    in previous studies to infect cats mucosally and trans-

    placentally and cause immunological and clinical decline

    (Allison and Hoover, 2003; Burkhard et al., 1997; Diehl et

    al., 1996; Obert and Hoover, 2000; Pedersen et al., 2001;

    Rogers and Hoover, 1998, 2002). FIV-GL8 and FIV-PET

    were prepared as previously described (Willett et al.,

    1997a). Stocks were titrated on the targeted cell type for

    each assay using standard techniques (Harper, 1993) and

    used in inhibition experiments at concentrations of 2040

    TCID50.

    In vitro tropism studies

    Supernatants grown in nave cat, lion, or puma PBMC

    were co-cultivated on feline PBMC, CrFK, 3201, or Mya-

    1 cells. Cells were plated in 24-well plates in the

    appropriate media at a concentration of 2 106/ml. Viralstocks were added to constitute 10% total media volume.

    Twenty-five to 50% of the supernatants were collected and

    cells re-fed with fresh media bi-weekly for 4 weeks.

    Supernatants were subjected to microtiter RT assay as

    previously described (VandeWoude et al., 1997a; Willey et

    al., 1988).

    Inhibitors

    AMD3100 was a gift from Dr. Lou Hegedus (Colorado

    State University, Fort Collins, CO, Department of Chem-

    istry). Recombinant human SDF-1a was obtained fromPeprotech (Rocky Hill, NJ). Mouse monoclonal antibodies

    were obtained as follows: anti-human CXCR4, catalogue #

    MAB-172, R&D Systems (Minneapolis, MN); anti-feline

    CD4 clone 3-4F4, Southern Biotech (Birmingham AL);

    anti-human CD134 (OX40) clone Ber Act 35; mouse IgG1

    (clone MOPC 31C, Ancell Corporation, Bayport, MN);

    and IgG2a (Unlb Clone HOPC-1 Southern Biotech,

    Birmingham AL) isotype control. Porcine heparin (sodium

    salt), Grade I-A was purchased from Sigma Aldrich (St

    Louis, MO). Cyanovirin-N (CVN, purified recombinant

    N. Smirnova et al. / Vexpressed in E. coli) was a gift from Dr. John Elder (The

    Scripps Research Institute, La Jolla, California, Departmentof Molecular Biology). Anti-feline CD25 monoclonal

    antibody was a gift of Dr. Wayne Tompkins (North

    Carolina State University, Raleigh, NC, Immunology

    Program).

    Inhibition assays

    Inhibition of virus-CXCR4 binding was attempted using

    the CXCR4 inhibitor AMD3100, monoclonal antibody

    anti-CXCR4 172, and SDF-1a. Virus was plated inconstant concentration against serial dilutions of inhibitor,

    or in one AMD3100 experiment, virus was titrated against

    a constant concentration of inhibitor. CD4, CD25 and

    CD134 inhibition studies were performed with anti-feline

    CD4 or CD25 monoclonal antibody or anti-human CD134.

    Serial-dilutions of heparin and CVN were used for non-

    specific blocking studies as outlined below. Anti-CD4,

    anti-CD25, and anti-CD134 binding limits for serial

    dilutions were determined by flow cytometric analysis as

    described below. Positive controls consisted of FIV when

    appropriate (i.e. CXCR4 and CD134 inhibition) or flow

    cytometric analysis of cells labeled with the concentrations

    of antibodies used (i.e. CD4, CD25, CD134). Negative

    controls consisted of virus without inhibitor, cells without

    virus, or isotype control antibodies (for CD4, CD25,

    CD134, CXCR4).

    The standard format for inhibition assays was as

    follows: (1) target cells (Mya-1, 3201, or PBMC) were

    plated at 2 105 cells/well in replicates of 35 inlymphocyte or 3201 media; (2) inhibitors were added to

    2 final concentration in ranges shown previously toinhibit FIV infection (AMD3100, SDF1a, heparin, CVN;Donzella et al., 1998; Egberink et al., 1999) or in

    concentrations determined using flow cytometry to eval-

    uate limiting dilution (anti-CD134, anti-CD4, anti-CD25);

    (3) 2040 TCID50 viral supernatant stocks were added to

    wells in an equal volume to each well, or nave

    supernatant was added as negative control; cells were

    incubated at 37 -C with 5% CO2 for 24 h, then washedand re-fed with media alone. In CVN, CD25, CD4, and

    CD134 inhibition studies, protocols were modified so that

    cells were exposed to inhibitor for 1 h at 37 -C followedby addition of virus and a 2-h, 4 -C incubation. Cellswere then again incubated at 37 -C for 4 h. We employedthis protocol to synchronize viral-cell binding in each

    culture and eliminate any potential viral binding kinetic

    differences in the experiments. Cells were then centri-

    fuged at 400 g, washed and re-plated with media; CVNwas re-added to cultures in the original exposure

    concentration.

    Supernatants were collected bi-weekly starting at day 3

    and assayed for virus as described below for most samples;

    in some cases FIV supernatants were collected daily.

    Samples were considered positive when OD450 nm read-

    y 342 (2005) 6076 71ings or RT values exceeded 2.5 media control in at leasttwo samples, and was confirmed at later timepoints during

  • GATTT 3V and 3PLV14R, 5VACAGATATCTTTGAAGG-GACACAT 3V. PCR products were gel isolated and cloned

    irologsampling. Ranges for ELISA and RT assays varied widely

    between viruses and assays, which precluded multi-viral

    comparisons on one graph. Therefore, percent inhibition

    was calculated as [((NRx Neg) (Rx Neg)) / (NRx Neg)] * 100, where NRx = cells with virus and no inhibitor;

    Neg = cells with no virus; Rx = cells with virus and

    inhibitor. Percent of no-inhibitor control was calculated as

    100% (percent inhibition). Percent inhibition wasdetermined starting at timepoints where positive controls

    were clearly above background as described until 34

    weeks post-inoculation. Multiple replications were per-

    formed for several inhibitors as listed in Table 2 to verify

    reproducibility of results using multiple viral strains or cell

    types. Statistics presented in Table 3 were calculated using

    raw data subjected to two-sample homoscedastic Students t

    test analysis (Microsoft Excel, Microsoft Corporation,

    Redmond, WA).

    Viral detection

    The presence of FIV in culture supernatants was detected

    using p24 antigen capture ELISA (Dreitz et al., 1995).

    Previous studies have demonstrated that PLV and LLV are

    not detected by this ELISA assay (VandeWoude et al.,

    1997a). LLV and PLV growth was detected using a reverse

    transcriptase microtiter assay as described with the excep-

    tion that 3 Al of RT reaction was spotted on Wallac DEAEnylon microbeta counter filter paper and read with a Wallac

    microbeta counter (Wallac/Perkin Elmer, Wellesley, MA;

    Willey et al., 1988).

    Flow cytometry

    2.0 105 Mya-1 cells were plated and centrifuged at800 g in a 96-well-v-bottom plate (Costar, Corning Inc,Corning NY). Mouse monoclonal anti-human CD134 (1

    mg/ml), anti-feline CD4 (0.5 mg/ml), or anti-feline CD25

    (cell culture supernatant) described above was diluted in

    flow buffer at concentrations from 1:50 to 1:1000 (anti-

    CD134) or 1:100 to 1:10000 (anti-CD4, anti-CD25) in a

    final volume of 0.2 ml with 2 105 cells. Reactions wereincubated at 4 -C for 30 min, then washed twice. Cellswere incubated with 1:100 dilution of fluorescein iso-

    thiocyanate (FITC)-conjugated sheep anti-mouse IgG

    (Sigma, St Louis, MO), for 30 min. After washing,

    samples were passed through an EPICS XL-MCL flow

    cytometer (Beckman Coulter, Miami, FL). Cells stained

    with isotype control antibodies were used to set analysis

    gates. Results following acquisition of 10,000 events were

    analyzed using FlowJo software (Tree Star, Inc., San

    Carlos, CA).

    PLV-1695 env sequencing

    N. Smirnova et al. / V72Genomic DNA was extracted with QIAamp DNA blood

    mini kit (Qiagen, Valencia CA) from Mya-1 cells persis-into pDrive (Qiagen) and used to transform TOP10 cells

    (Invitrogen Corp, Carlsbad CA). Plasmid DNA was recov-

    ered from transformed colonies and sequenced.

    Comparison of domestic and non-domestic feline lentiviral

    Env regions

    env nucleotide sequences for FIV-Petaluma (accession

    no. M25381; Olmsted et al., 1989), FIV-C36 (accession no.

    AY600517; de Rozieres et al., 2004), Pallas cat lentivirus,

    FIV-Oma (accession no. U56928; Barr et al., 1997), and

    PLV-14 (accession no. U03982; Langley et al., 1994) were

    obtained from GenBank. Predicted amino acid sequences

    were aligned with those of PLV-1695 (see above) in Clustal

    X (Thompson et al., 1997) using the PAM 350 protein

    weight matrix (Yang, 1997) and realigned manually. The

    predicted secondary structure of these amino acid sequences

    including N-glycosylation sites (Gupta et al., in preparation;

    Gupta and Brunak, 2002; Gupta et al., 2002), protein

    domains (Corpet et al., 1998), disulfide bonds (Rost et

    al., 2004), hydrophobic regions, and net charge was

    determined using the PredictProtein server; http://

    www.embl-heidelberg.de/predictprotein/, NetNGlyc 1.0

    server; http://www.cbs.dtu.dk/services/NetNGlyc/, and Iso-

    electric Plot; http://www-biol.univ-mrs.fr/d_abim/d_docs/

    doc-tab-pk.html.

    Acknowledgments

    We thank Drs. Brian Willett and Margaret Hosie for

    discussions regarding PLV receptor use, and for manuscript

    review. Drs. J. Elder, L. Hegedus, W. Tompkins, K.

    Langelier, D. Hunter, and C. Krumm and S.J. OBrien

    generously contributed reagents, cells, and virus stocks, and

    we thank Dr. E.A. Hoover and the Feline Retrovirus

    Research Laboratory at CSU for provision of laboratory

    infrastructure support, and for manuscript review. We thank

    Jennifer Yactor, Kerry Sondgeroth, and Joy Zartman for

    technical assistance.tently infected with PLV-1695. Nested PCR were employed

    to amplify a 4.5 kb fragment that encompasses the 3V codingregions of the PLV genome. Reactions employed ExTaq

    DNA polymerase (Takara Mirus Bio, Madison WI) accord-

    ing to manufacturers protocol. PCR conditions were 3 min

    at 94 -C, and then 35 cycles of 94 -C for 30 s, 30 s annealingat 54 -C for first round PCR and 52 -C for second rounds, a70 -C extension for 4 min 30 s, and a final 10 min extensionat 70 -C. First round PCR primers were 3PLV11F, 5VGCCGGAACCTACAGACCCCTTAT 3V and 3PLV12R, 5VTTTGTTCTGCCCATTCTCCTATTG 3V and second roundprimers were 3770F, 5VAGAGAAGAAGATGCTAGATAT-

    y 342 (2005) 6076This work was supported by Public Health Service grants

    R29 AI41871 and R01 AI49765 from NIAID, DAIDS, the

  • 73 (4), 25762586.

    irologBrown, E.W., Miththapala, S., O Brien, S.J., 1993. Prevalence of exposure

    to feline immunodeficiency virus in exotic Felid species. J. Zoo Wildl.

    Med. 24 (3), 357364.

    Brown, E.W., Yuhki, N., Packer, C., OBrien, S.J., 1994. A lion lentivirusColorado State University College of Veterinary Medicine

    and Biomedical Science Research Council, and the Merck-

    Meriel Summer Fellowship Program.

    References

    Ackley, C.D., Yamamoto, J.K., Levy, N., Pedersen, N.C., Cooper, M.D.,

    1990. Immunologic abnormalities in pathogen-free cats experimen-

    tally infected with feline immunodeficiency virus. J. Virol. 64 (11),

    56525655.

    Alkhatib, G., Combadiere, C., Broder, C.C., Feng, Y., Kennedy, P.E.,

    Murphy, P.M., Berger, E.A., 1996. CC CKR5: a RANTES, MIP-1alpha,

    MIP-1beta receptor as a fusion cofactor for macrophage-tropic HIV-1.

    Science 272 (5270), 19551958.

    Allan, J.S., Wilson, B., Potter, J., Hodara, V., Giavedoni, L., White, R.,

    2004. 22nd Annual Symposium on Nonhuman Primate Models for

    AIDS, San Antonio, TX.

    Allison, R.W., Hoover, E.A., 2003. Feline immunodeficiency virus is

    concentrated in milk early in lactation. AIDS Res. Hum. Retroviruses

    19 (3), 245253.

    Apetrei, C., Robertson, D.L., Marx, P.A., 2004. The history of SIVS and

    AIDS: epidemiology, phylogeny and biology of isolates from naturally

    SIV infected non-human primates (NHP) in Africa. Front. Biosci. 9,

    225254.

    Barr, M.C., Zou, L., Holzschu, D.L., Phillips, L., Scott, F.W., Casey, J.W.,

    Avery, R.J., 1995. Isolation of a highly cytopathic lentivirus from a

    nondomestic cat. J. Virol. 69 (11), 73717374.

    Barr, M.C., Zou, L., Long, F., Hoose, W.A., Avery, R.J., 1997. Proviral

    organization and sequence analysis of feline immunodeficiency virus

    isolated from a Pallas cat. Virology 228 (1), 8491.

    Barrientos, L.G., OKeefe, B.R., Bray, M., Sanchez, A., Gronenborn, A.M.,

    Boyd, M.R., 2003. Cyanovirin-N binds to the viral surface glycopro-

    tein, GP1,2 and inhibits infectivity of Ebola virus. Antiviral Res. 58 (1),

    4756.

    Bendinelli, M., Pistello, M., Lombardi, S., Poli, A., Garzelli, C., Matteucci,

    D., Ceccherini-Nelli, L., Malvaldi, G., Tozzini, F., 1995. Feline

    immunodeficiency virus: an interesting model for AIDS studies and

    an important cat pathogen. Clin. Microbiol. Rev. 8 (1), 87112.

    Bewley, C.A., Otero-Quintero, S., 2001. The potent anti-HIV protein

    cyanovirin-N contains two novel carbohydrate binding sites that

    selectively bind to Man (8) D1D3 and Man (9) with nanomolar affinity:

    implications for binding to the HIV envelope protein gp120. J. Am.

    Chem. Soc. 123 (17), 38923902.

    Biek, R., Rodrigo, A.G., Holley, D., Drummond, A., Anderson Jr., C.R.,

    Ross, H.A., Poss, M., 2003. Epidemiology, genetic diversity, and

    evolution of endemic feline immunodeficiency virus in a population of

    wild cougars. J. Virol. 77 (17), 95789589.

    Bjorndal, A., Deng, H., Jansson, M., Fiore, J.R., Colognesi, C., Karlsson, A.,

    Albert, J., Scarlatti, G., Littman, D.R., Fenyo, E.M., 1997. Coreceptor

    usage of primary human immunodeficiency virus type 1 isolates varies

    according to biological phenotype. J. Virol. 71 (10), 74787487.

    Bolmstedt, A.J., OKeefe, B.R., Shenoy, S.R., McMahon, J.B., Boyd, M.R.,

    2001. Cyanovirin-N defines a new class of antiviral agent targeting N-

    linked, high-mannose glycans in an oligosaccharide-specific manner.

    Mol. Pharmacol. 59 (5), 949954.

    Brelot, A., Heveker, N., Adema, K., Hosie, M.J., Willett, B., Alizon, M.,

    1999. Effect of mutations in the second extracellular loop of CXCR4 on

    its utilization by human and feline immunodeficiency viruses. J. Virol.

    N. Smirnova et al. / Vrelated to feline immunodeficiency virus: epidemiologic and phyloge-

    netic aspects. J. Virol. 68 (9), 59535968.Burkhard, M.J., Obert, L.A., ONeil, L.L., Diehl, L.J., Hoover, E.A., 1997.

    Mucosal transmission of cell-associated and cell-free feline immunode-

    ficiency virus. AIDS Res. Hum. Retroviruses 13 (4), 347355.

    Caney, S.M., Day, M.J., Gruffydd-Jones, T.J., Helps, C.R., Hirst, T.R.,

    Stokes, C.R., 2002. Expression of chemokine receptors in the

    feline reproductive tract and large intestine. J. Comp. Pathol. 126 (4),

    289302.

    Carpenter, M.A., OBrien, S.J., 1995. Coadaptation and immunodefi-

    ciency virus: lessons from the Felidae. Curr. Opin. Genet. Dev. 5 (6),

    739745.

    Carpenter, M.A., Brown, E.W., Culver, M., Johnson, W.E., Pecon-Slattery,

    J., Brousset, D., OBrien, S.J., 1996. Genetic and phylogenetic

    divergence of feline immunodeficiency virus in the puma (Puma

    concolor). J. Virol. 70 (10), 66826693.

    Carpenter, M.A., Brown, E.W., MacDonald, D.W., OBrien, S.J., 1998.

    Phylogeographic patterns of feline immunodeficiency virus genetic

    diversity in the domestic cat. Virology 251 (2), 234243.

    Carrillo, A., Ratner, L., 1996. Human immunodeficiency virus type 1

    tropism for T-lymphoid cell lines: role of the V3 loop and C4 envelope

    determinants. J. Virol. 70 (2), 13011309.

    Chakrabarti, L.A., 2004. The paradox of simian immunodeficiency virus

    infectino in sooty mangebys: active viral replication without disease

    progression. Front. Biosci. 9, 521539.

    Chen, Z., Zhou, P., Ho, D.D., Landau, N.R., Marx, P.A., 1997. Genetically

    divergent strains of simian immunodeficiency virus use CCR5 as a

    coreceptor for entry. J. Virol. 71 (4), 27052714.

    Chesebro, B., Wehrly, K., Nishio, J., Perryman, S., 1996. Mapping of

    independent V3 envelope determinants of human immunodeficiency

    virus type 1 macrophage tropism and syncytium formation in

    lymphocytes. J. Virol. 70 (12), 90559059.

    Choe, H., Farzan, M., Sun, Y., Sullivan, N., Rollins, B., Ponath, P.D., Wu,

    L., Mackay, C.R., LaRosa, G., Newman, W., Gerard, N., Gerard, C.,

    Sodroski, J., 1996. The b-chemokine receptors CCR3 and CCR5

    facilitate infection by primary HIV-1 isolates. Cell 85 (7), 11351148.

    Corpet, F., Gouzy, J., Kahn, D., 1998. The ProDom database of protein

    domain families. Nucleic Acids Res. 26 (1), 323326.

    de Parseval, A., Elder, J.H., 2001. Binding of recombinant feline

    immunodeficiency virus surface glycoprotein to feline cells: role of

    CXCR4, cell-surface heparans, and an unidentified non-CXCR4

    receptor. J. Virol. 75 (10), 45284539.

    de Parseval, A., Chatterji, U., Sun, P., Elder, J.H., 2004a. Feline

    immunodeficiency virus targets activated CD4+ T cells by using

    CD134 as a binding receptor. Proc. Natl. Acad. Sci. U.S.A. 101 (35),

    1304413049.

    de Parseval, A., Ngo, S., Sun, P., Elder, J.H., 2004b. Factors that increase

    the effective concentration of CXCR4 dictate feline immunodefi-

    ciency virus tropism and kinetics of replication. J. Virol. 78 (17),

    91329143.

    de Parseval, A., Chatterji, U., Morris, G., Sun, P., Olson, A.J., Elder, J.H.,

    2005. Structural mapping of CD134 residues critical for interaction with

    feline immunodeficiency virus. Nat. Struct. Mol. Biol. 12 (1), 6066.

    de Rozieres, S., Mathiason, C.K., Rolston, M.R., Chatterji, U., Hoover,

    E.A., Elder, J.H., 2004. Characterization of a highly pathogenic

    molecular clone of feline immunodeficiency virus clade C. J. Virol. 78

    (17), 89718982.

    Deng, H.K., Lui, R., Ellmeier, W., Choe, S., Unutmaz, D., Burkhart, M., Di

    Marzio, P., Marmon, S., Sutton, R.E., Hill, C.M., Davis, C.B., Peiper,

    S.C., Schall, T.J., Littman, D.R., Landau, N.R., 1996. Identification of a

    major co-receptor for primary isolates of HIV-1. Nature 381, 661666.

    Dey, B., Lerner, D.L., Lusso, P., Boyd, M.R., Elder, J.H., Berger, E.A.,

    2000. Multiple antiviral activities of cyanovirin-N: blocking of human

    immunodeficiency virus type 1 gp120 interaction with CD4 and

    coreceptor and inhibition of diverse enveloped viruses. J. Virol. 74

    (10), 45624569.

    Diehl, L.J., Mathiason-DuBard, C.K., ONeil, L.L., Hoover, E.A., 1996.

    y 342 (2005) 6076 73Plasma viral RNA load predicts disease progression in an accelerated

    feline immunodeficiency virus model. J. Virol. 70, 25032507.

  • irologDonzella, G.A., Schols, D., Lin, S.W., Este, J.A., Nagashima, K.A.,

    Maddon, P.J., Allaway, G.P., Sakmar, T.P., Henson, G., De Clercq, E.,

    Moore, J.P., 1998. AMD3100, a small molecule inhibitor of HIV-1 entry

    via the CXCR4 co-receptor. Nat. Med. 4 (1), 7277.

    Doranz, B.J., Rucker, J., Yi, Y., Smyth, R.J., Samson, M., Peiper, S.C.,

    Parmentier, M., Collman, R.B., Doms, R.W., 1996. A dual-tropic

    primary HIV-1 isolate that uses fusin and the beta-chemokine

    receptors CKR-5, CKR-3, and CKR-2b as fusion cofactors. Cell 85

    (7), 11491158.

    Dragic, T., Litwin, V., Allaway, G.P., Martin, S.R., Huang, Y., Nagashima,

    K.A., Cayanan, C., Maddon, P.J., Koup, R.A., Moore, J.P., Paxton,

    W.A., 1996. HIV-1 entry into CD4+ cells is mediated by the chemokine

    receptor CC-CKR-5. Nature 381 (6584), 667673.

    Dreitz, M.J., Dow, S.W., Fiscus, S.A., Hoover, E.A., 1995. Developmental

    of monoclonal antibodies and capture immunoassays for feline

    immunodeficiency virus. Am. J. Vet. Res. 56, 764768.

    Egberink, H.F., De Clercq, E., Van Vliet, A.L., Balzarini, J., Bridger, G.J.,

    Henson, G., Horzinek, M.C., Schols, D., 1999. Bicyclams, selective

    antagonists of the human chemokine receptor CXCR4, potently inhibit

    feline immunodeficiency virus replication. J. Virol. 73 (8), 63466352.

    Elder, J.H., Dean, G.A., Hoover, E.A., Hoxie, J.A., Malim, M.H., Mathes,

    L., Neil, J.C., North, T.W., Sparger, E., Tompkins, M.B., Tompkins,

    W.A., Yamamoto, J., Yuhki, N., Pedersen, N.C., Miller, R.H., 1998.

    Lessons from the cat: feline immunodeficiency virus as a tool to

    develop intervention strategies against human immunodeficiency virus

    type 1. AIDS Res. Hum. Retroviruses 14 (9), 797801.

    Feng, Y., Broder, C.C., Kennedy, P.E., Berger, E.A., 1996. HIV-1 entry

    cofactor: functional cDNA cloning of a seven-transmembrane, G

    protein-coupled receptor. Science 272 (5263), 872877.

    Gaddis, N.C., Sheehy, A.M., Ahmad, K.M., Swanson, C.M., Bishop, K.N.,

    Beer, B.E., Marx, P.A., Gao, F., Bibollet-Ruche, F., Hahn, B.H., Malim,

    M.H., 2004. Further investigation of simian immunodeficiency virus

    Vif function in human cells. J. Virol. 78 (21), 1204112046.

    Gupta, R., Brunak, S., 2002. Prediction of glycosylation across the

    human proteome and the correlation to protein function. Pac. Symp.

    Biocomput., 310322.

    Gupta, R., Jensen, L.J., Brunak, S., 2002. Orphan protein function and its

    relation to glycosylation. Ernst Schering Res. Found Workshop (38),

    276294.

    Gupta, R., Jung, E., Brunak, S., in preparation. Prediction of

    N-glycosylation Sites in Human Proteins.

    Haining, H., McMonagle, E.L., Knapp, E., Klein, D., Willett, B.J., Hosie,

    M.J., 2004. 7th International Feline Retrovirus Research Symposium,

    Pisa, Italy.

    Hardy Jr., W.D., McClelland, A.J., Zuckerman, E.E., Snyder Jr., H.W.,

    MacEwen, E.G., Francis, D., Essex, M., 1980. Development of virus

    non-producer lymphosarcomas in pet cats exposed to FeLv. Nature 288

    (5786), 9092.

    Harper, D.R. (Ed.), 1993. Virology LabFax 1st edR In: Hames, B.D.,

    Rickwood, D. (Eds.), The LabFax Series. Academic Press, London, UK.

    Hohdatsu, T., Hirabayashi, H., Motokawa, K., Koyama, H., 1996.

    Comparative study of the cell tropism of feline immunodeficiency virus

    isolates of subtypes A, B and D classified on the basis of the env gene

    V3V5 sequence. J. Gen. Virol. 77 (Pt. 1), 93100.

    Hosie, M.J., Broere, N., Hesselgesser, J., Turner, J.D., Hoxie, J.A.,

    Neil, J.C., Willett, B.J., 1998. Modulation of feline immunodefi-

    ciency virus infection by stromal cell-derived factor. J. Virol. 72 (3),

    20972104.

    Hosie, M.J., Willett, B.J., Klein, D., Dunsford, T.H., Cannon, C.,

    Shimojima, M., Neil, J.C., Jarrett, O., 2002. Evolution of

    replication efficiency following infection with a molecularly cloned

    feline immunodeficiency virus of low virulence. J. Virol. 76 (12),

    60626072.

    Ibrahim, J., Griffin, P., Coombe, D.R., Rider, C.C., James, W., 1999. Cell-

    surface heparan sulfate facilitates human immunodeficiency virus Type

    N. Smirnova et al. / V741 entry into some cell lines but not primary lymphocytes. Virus Res. 60

    (2), 159169.Johnston, J., Power, C., 1999. Productive infection of human peripheral

    blood mononuclear cells by feline immunodeficiency virus: implica-

    tions for vector development. J. Virol. 73 (3), 24912498.

    Johnston, J.B., Power, C., 2002. Feline immunodeficiency virus xenoin-

    fection: the role of chemokine receptors and envelope diversity. J. Virol.

    76 (8), 36263636.

    Johnston, J.B., Olson, M.E., Rud, E.W., Power, C., 2001. Xenoinfection of

    nonhuman primates by feline immunodeficiency virus. Curr. Biol. 11

    (14), 11091113.

    Joshi, A., Vahlenkamp, T.W., Garg, H., Tompkins, W.A., Tompkins, M.B.,

    2004. Preferential replication of FIV in activated CD4(+)CD25(+)T

    cells independent of cellular proliferation. Virology 321 (2), 307322.

    Kato, K., Sato, H., Takebe, Y., 1999. Role of naturally occurring basic

    amino acid substitutions in the human immunodeficiency virus type 1

    subtype E envelope V3 loop on viral coreceptor usage and cell tropism.

    J. Virol. 73 (7), 55205526.

    Klatzmann, D., Champagne, E., Chamaret, S., Gruest, J., Guetard, D.,

    Hercend, T., Gluckman, J.C., Montagnier, L., 1984. T-lymphocyte T4

    molecule behaves as the receptor for human retrovirus LAV. Nature 312

    (5996), 767768.

    Langley, R.J., Hirsch, V.M., OBrien, S.J., Adger-Johnson, D., Goeken,

    R.M., Olmsted, R.A., 1994. Nucleotide sequence analysis of puma

    lentivirus (PLV-14): genomic organization and relationship to other

    lentiviruses. Virology 202 (2), 853864.

    Lerner, D.L., Elder, J.H., 2000. Expanded host cell tropism and cytopathic

    properties of feline immunodeficiency virus strain PPR subsequent to

    passage through interleukin-2-independent T cells. J. Virol. 74 (4),

    18541863.

    Levy, J.A., 1998. HIV and the Pathogenesis of AIDS, 2nd edR ASM Press,Washington, DC.

    Linenberger, M.L., Deng, T., 1999. The effects of feline retroviruses

    on cytokine expression. Vet. Immunol. Immunopathol. 72 (34),

    343368.

    Liu, H.Y., Soda, Y., Shimizu, N., Haraguchi, Y., Jinno, A., Takeuchi, Y.,

    Hoshino, H., 2000. CD4-dependent and CD4-independent utilization of

    coreceptors by human immunodeficiency viruses type 2 and simian

    immunodeficiency viruses. Virology 278 (1), 276288.

    Lombardi, S., Garzelli, C., Pistello, M., Massi, C., Matteucci, D.,

    Baldinotti, F., Cammarota, G., da Prato, L., Bandecchi, P., Tozzini, F.,

    1994. A neutralizing antibody-inducing peptide of the V3 domain of

    feline immunodeficiency virus envelope glycoprotein does not induce

    protective immunity. J. Virol. 68 (12), 83748379.

    Lombardi, S., Massi, C., Tozzini, F., Zaccaro, L., Bazzichi, A., Bandecchi,

    P., La Rosa, C., Bendinelli, M., Garzelli, C., 1995. Epitope mapping of

    the V3 domain of feline immunodeficiency virus envelope glycoprotein

    by monoclonal antibodies. J. Gen. Virol. 76 (Pt. 8), 18931899.

    Maddon, P.J., Dalgleish, A.G., McDougal, J.S., Clapham, P.R., Weiss, R.A.,

    Axel, R., 1986. The T4 gene encodes the AIDS virus receptor and is

    expressed in the immune system and the brain. Cell 47 (3), 333348.

    Mariani, R., Chen, D., Schrofelbauer, B., Navarro, F., Konig, R., Bollman,

    B., Munk, C., Nymark-McMahon, H., Landau, N.R., 2003. Species-

    specific exclusion of APOBEC3G from HIV-1 virions by Vif. Cell 114

    (1), 2131.

    Masuoka, K., Toyosaki, T., Tohya, Y., Norimine, J., Kai, C., Mikami, T.,

    1992. Monoclonal antibodies to feline lymphocyte membranes recog-

    nize the leukocyte-common antigen (CD45R). J. Vet. Med. Sci. 54 (5),

    865870.

    McCaffrey, R.A., Saunders, C., Hensel, M., Stamatatos, L., 2004. N-linked

    glycosylation of the V3 loop and the immunologically silent face of

    gp120 protects human immunodeficiency virus type 1 SF162 from

    neutralization by anti-gp120 and anti-gp41 antibodies. J. Virol. 78 (7),

    32793295.

    Miller, R.J., Cairns, J.S., Bridges, S., Sarver, N., 2000. Human immuno-

    deficiency virus and AIDS: insights from animal lentiviruses. J. Virol.

    74 (16), 71877195.

    y 342 (2005) 6076Miyazawa, T., Furuya, T., Itagaki, S., Tohya, Y., Takahashi, E., Mikami, T.,

    1989. Establishment of a feline T-lymphoblastoid cell line highly

  • irologsensitive for replication of feline immunodeficiency virus. Arch. Virol.

    108 (12), 131135.

    Miyazawa, T., Kawaguchi, Y., Furuya, T., Itagaki, S., Takahashi, E.,

    Mikami, T., 1990. Continuous production of feline immunodeficiency

    virus in a feline T-lymphoblastoid cell line (MYA-1 cells). Nippon

    Juigaku Zasshi 52 (4), 887890.

    Miyazawa, T., Toyosaki, T., Tomonaga, K., Norimine, J., Ohno, K.,

    Hasegawa, A., Kai, C., Mikami, T., 1992. Further characterization of a

    feline T-lymphoblastoid cell line (MYA-1 cells) highly sensitive for

    feline immunodeficiency virus. J. Vet. Med. Sci. 54 (1), 173175.

    Mori, T., Boyd, M.R., 2001. Cyanovirin-N, a potent human immunodefi-

    ciency virus-inactivating protein, blocks both CD4-dependent and CD4-

    independent binding of soluble gp120 (sgp120) to target cells, inhibits

    sCD4-induced binding of sgp120 to cell-associated CXCR4, and

    dissociates bound sgp120 from target cells. Antimicrob. Agents

    Chemother. 45 (3), 664672.

    Muller, M.C., Barre-Sinoussi, F., 2003. SIVagm: genetic and biological

    features associated with replication. Front. Biosci. 8, D1170D1185.

    Nabatov, A.A., Pollakis, G., Linnemann, T., Kliphius, A., Chalaby, M.I.,

    Paxton, W.A., 2004. Intrapatient alterations in the human immunode-

    ficiency virus type 1 gp120 V1V2 and V3 regions differentially

    modulate coreceptor usage, virus inhibition by CC/CXC chemokines,

    soluble CD4, and the b12 and 2G12 monoclonal antibodies. J. Virol. 78

    (1), 524530.

    Nielsen, R., Yang, Z., 1998. Likelihood models for detecting positively

    selected amino acid sites and applications to the HIV-1 envelope gene.

    Genetics 148 (3), 929936.

    Nishimura, Y., Nakamura, S., Goto, N., Hasegawa, T., Pang, H., Goto, Y.,

    Kato, H., Youn, H.Y., Endo, Y., Mizuno, T., Momoi, Y., Ohno, K.,

    Watari, T., Tsujimoto, H., Hasegawa, A., 1996. Molecular character-

    ization of feline immunodeficiency virus genome obtained directly from

    organs of a naturally infected cat with marked neurological symptoms

    and encephalitis. Arch. Virol. 141 (10), 19331948.

    Nishimura, Y., Miyazawa, T., Ikeda, Y., Izumiya, Y., Nakamura, K., Cai,

    J.S., Sato, E., Kohmoto, M., Mikami, T., 1998. Molecular cloning and

    expression of feline CD3epsilon. Vet. Immunol. Immunopathol. 65 (1),

    4350.

    Nishimura, Y., Igarashi, T., Donau, O.K., Buckler-White, A., Buckler, C.,

    Lafont, B.A., Goeken, R.M., Goldstein, S., Hirsch, V.M., Martin, M.A.,

    2004. Highly pathogenic SHIVs and SIVs target different CD4+ T cell

    subsets in rhesus monkeys, explaining their divergent clinical courses.

    Proc. Natl. Acad. Sci. U.S.A. 101 (33), 1232412329.

    OKeefe, B.R., Shenoy, S.R., Xie, D., Zhang, W., Muschik, J.M., Currens,

    M.J., Chaiken, I., Boyd, M.R., 2000. Analysis of the interaction

    between the HIV-inactivating protein cyanovirin-N and soluble forms of

    the envelope glycoproteins gp120 and gp41. Mol. Pharmacol. 58 (5),

    982992.

    OKeefe, B.R., Smee, D.F., Turpin, J.A., Saucedo, C.J., Gustafson,

    K.R., Mori, T., Blakeslee, D., Buckheit, R., Boyd, M.R., 2003.

    Potent anti-influenza activity of cyanovirin-N and interactions

    with viral hemagglutinin. Antimicrob. Agents Chemother. 47 (8),

    25182525.

    Obert, L.A., Hoover, E.A., 2000. Feline immunodeficiency virus clade C

    mucosal transmission and disease courses. AIDS Res. Hum. Retro-

    viruses 16 (7), 677688.

    Olmsted, R.A., Hirsch, V.M., Purcell, R.H., Johnson, P.R., 1989. Nucleo-

    tide sequence analysis of feline immunodeficiency virus: genome

    organization and relationship to other lentiviruses. Proc. Natl. Acad.

    Sci. U.S.A. 86 (20), 80888092.

    Olmsted, R.A., Langley, R., Roelke, M.E., Goeken, R.M., Adgerjohnson,

    D., Goff, J.P., Albert, J.P., Packer, C., Laurenson, M.K., Caro, T.M.,

    Scheepers, L., Wildt, D.E., Bush, M., Martenson, J.S., Obrien, S.J.,

    1992. Worldwide prevalence of lentivirus infection in wild feline

    species-epidemiologic and phylogenetic aspects. J. Virol. 66 (10),

    60086018.

    N. Smirnova et al. / VOverbaugh, J., Luciw, P.A., Hoover, E.A., 1997. Models for AIDS

    pathogenesis: simian immunodeficiency virus, simian-human immuno-deficiency virus and feline immunodeficiency virus infections. Aids 11

    (Suppl. A), S47S54.

    Pancino, G., Fossati, I., Chappey, C., Castelot, S., Hurtrel, B., Moraillon,

    A., Klatzmann, D., Sonigo, P., 1993. Structure and variations of feline

    immunodeficiency virus envelope glycoproteins. Virology 192 (2),

    659662.

    Pedersen, N.C., Leutenegger, C.M., Woo, J., Higgins, J., 2001.

    Virulence differences between two field isolates of feline immuno-

    deficiency virus (FIV-APetaluma and FIV-CPGammar) in young

    adult specific pathogen free cats. Vet. Immunol. Immunopathol. 79

    (12), 5367.

    Pohlmann, S., Davis, C., Meister, S., Leslie, G.J., Otto, C., Reeves, J.D.,

    Puffer, B.A., Papkalla, A., Krumbiegel, M., Marzi, A., Lorenz, S.,

    Munch, J., Doms, R.W., Kirchhoff, F., 2004. Amino acid 324 in the

    simian immunodeficiency virus SIVmac V3 loop can confer CD4

    independence and modulate the interaction with CCR5 and alternative

    coreceptors. J. Virol. 78 (7), 32233232.

    Quackenbush, S.L., Donahue, P.R., Dean, G.A., Myles, M.H., Ackley,

    C.D., Cooper, M.D., Mullins, J.I., Hoover, E.A., 1990. Lymphocyte

    subset alterations and viral determinants of immunodeficiency disease

    induction by the feline leukemia virus FeLV-FAIDS. J. Virol. 64 (11),

    54655474.

    Richardson, J., Pancino, G., Merat, R., Leste-Lasserre, T., Moraillon, A.,

    Schneider-Mergener, J., Alizon, M., Sonigo, P., Heveker, N., 1999.

    Shared usage of the chemokine receptor CXCR4 by primary and

    laboratory-adapted strains of feline immunodeficiency virus. J. Virol. 73

    (5), 36613671.

    Roderiquez, G., Oravecz, T., Yanagishita, M., Bou-Habib, D.C., Mostow-

    ski, H., Norcross, M.A., 1995. Mediation of human immunodeficiency

    virus type 1 binding by interaction of cell surface heparan sulfate

    proteoglycans with the V3 region of envelope gp120-gp41. J. Virol. 69

    (4), 22332239.

    Rogers, A.B., Hoover, E.A., 1998. Maternal fetal feline immunodeficiency

    virus transmission: timing and tissue tropisms. J. Infect. Dis. 178 (4),

    960967.

    Rogers, A.B., Hoover, E.A., 2002. Fetal feline immunodeficiency virus is

    prevalent and occult. J. Infect. Dis. 186 (7), 895904.

    Rost, B., Yachdav, G., Liu, J., 2004. The predict protein server. Nucleic

    Acids Res. 32, W321W326 (Web Server issue).

    Schrofelbauer, B., Chen, D., Landau, N.R., 2004. A single amino

    acid of APOBEC3G controls its species-specific interaction with

    virion infectivity factor (Vif). Proc. Natl. Acad. Sci. U.S.A. 101 (11),

    39273932.

    Shimojima, M., Nishimura, Y., Miyazawa, T., Kato, K., Tohya, Y., Akashi,

    H., 2003. CD56 expression in feline lymphoid cells. J. Vet. Med. Sci. 65

    (7), 769773.

    Shimojima, M., Miyazawa, T., Ikeda, Y., McMonagle, E.L., Haining, H.,

    Akashi, H., Takeuchi, Y., Hosie, M.J., Willett, B.J., 2004. Use of

    CD134 as a primary receptor by the feline immunodeficiency virus.

    Science 303 (5661), 11921195.

    Siebelink, K.H., Karlas, J.A., Rimmelzwaan, G.F., Osterhaus, A.D., Bosch,

    M.L., 1995. A determinant of feline immunodeficiency virus involved

    in Crandell feline kidney cell tropism. Vet. Immunol. Immunopathol. 46

    (12), 6169.

    Sodora, D.L., Shpaer, E.G., Kitchell, B.E., Dow, S.W., Hoover, E.A.,

    Mullins, J.I., 1994. Identification of three feline immunodeficiency

    virus (FIV) env gene subtypes and comparison of the FIV and human

    immunodeficiency virus type 1 evolutionary patterns. J. Virol. 68 (4),

    22302238.

    Speck, R.F., Wehrly, K., Platt, E.J., Atchison, R.E., Charo, I.F., Kabat, D.,

    Chesebro, B., Goldsmith, M.A., 1997. Selective employment of

    chemokine receptors as human immunodeficiency virus type 1

    coreceptors determined by individual amino acids within the envelope

    V3 loop. J. Virol. 71 (9), 71367139.

    Tanabe-Tochikura, A., Tochikura, T.S., Blakeslee Jr., J.R., Olsen, R.G.,

    y 342 (2005) 6076 75Mathes, L.E., 1992. Anti-human immunodeficiency virus (HIV)

    agents are also potent and selective inhibitors of feline immunodefi-

  • ciency virus (FIV)-induced cytopathic effect: development of a new

    method for screening of anti-FIV substances in vitro. Antivir. Res. 19

    (2), 161172.

    Terwee, J.A., Yactor, J.K., Sondgeroth, K.S., Vandewoude, S., 2005. Puma

    lentivirus is controlled in domestic cats after mucosal exposure in the

    absence of conventional indicators of immunity. J. Virol. 79 (5),

    27972806.

    Thompson, J.D., Gibson, T.J., Plewniak, F., Jeanmougin, F., Higgins, D.G.,

    1997. The CLUSTAL_X windows interface: flexible strategies for

    multiple sequence alignment aided by quality analysis tools. Nucleic

    Acids Res. 25 (24), 48764882.

    Tozzini, F., Matteucci, D., Bandecchi, P., Baldinotti, F., Siebelink, K.,

    Osterhaus, A., Bendinelli, M., 1993. Neutralizing antibodies in cats

    infected with feline immunodeficiency virus. J. Clin. Microbiol. 31 (6),

    VandeWoude, S., Hageman, C.L., OBrien, S.J., Hoover, E.A., 2000.

    Proceedings, 5th Intl. Feline Retrovirus Research Symposium, Cannes,

    France.

    VandeWoude, S., Hageman, C.A., OBrien, S.J., Hoover, E.A., 2002.

    Nonpathogenic lion and puma lentiviruses impart resistance to super-

    infection by virulent feline immunodeficiency virus. J. Acquir. Immune

    Defic. Syndr. 29 (1), 110.

    VandeWoude, S., Hageman, C.L., Hoover, E.A., 2003. Domestic cats

    infected with lion or puma lentivirus develop anti-feline immunodefi-

    ciency virus immune responses. J. Acquir. Immune Defic. Syndr. 34 (1),

    2031.

    Verschoor, E.J., Boven, L.A., Blaak, H., van Vliet, A.L., Horzinek, M.C.,

    de Ronde, A., 1995. A single mutation within the V3 envelope

    neutralization domain of feline immunodeficiency virus determines its

    tropism for CRFK cells. J. Virol. 69 (8), 47524757.

    N. Smirnova et al. / Virology 342 (2005) 607676Troyer, J.L., Pecon-Slattery, J., Roelke, M.E., Black, L., Packer, C.,

    OBrien, S.J., 2004. Patterns of feline immunodeficiency virus multiple

    infection and genome divergence in a free-ranging population of

    African lions. J. Virol. 78 (7), 37773791.

    Troyer, J.L., Pecon-Slattery, J., Roelke, M.E., Johnson, W.E., VandeWoude,

    S., Vazquez-Salat, N., Brown, M., Frank, L., Woodroffe, R., Winter-

    bach, C., Winterbach, H., Hemson, G., Bush, M., Alexander, K.A.,

    OBrien, S.J., 2005. Seroprevalence and genomic divergence of

    circulating strains of feline immunodeficiency virus (FIV) among

    felidae and hyaenidae species. J. Virol. (in revision).

    Tsai, C.C., Emau, P., Jiang, Y., Tian, B., Morton, W.R., Gustafson, K.R.,

    Boyd, M.R., 2003. Cyanovirin-N gel as a topical microbicide prevents

    rectal transmission of SHIV89.6P in macaques. AIDS Res. Hum.

    Retroviruses 19 (7), 535541.

    Tsai, C.C., Emau, P., Jiang, Y., Agy, M.B., Shattock, R.J., Schmidt, A.,

    Morton, W.R., Gustafson, K.R., Boyd, M.R., 2004. Cyanovirin-N

    inhibits AIDS virus infections in vaginal transmission models. AIDS

    Res. Hum. Retroviruses 20 (1), 1118.

    Uchiyama, T., 1997. Human T cell leukemia virus type I (HTLV-I) and

    human diseases. Annu. Rev. Immunol. 15, 1537.

    Vahlenkamp, T.W., Verschoor, E.J., Schuurman, N.N., van Vliet, A.L.,

    Horzinek, M.C., Egberink, H.F., de Ronde, A., 1997. A single amino

    acid substitution in the transmembrane envelope glycoprotein of

    feline immunodeficiency virus alters cellular tropism. J. Virol. 71 (9),

    71327135.

    Vahlenkamp, T.W., Tompkins, M.B., Tompkins, W.A., 2004. Feline

    immunodeficiency virus infection phenotypically and functionally

    activates immunosuppressive CD4+CD25+ T regulatory cells. J.

    Immunol. 172 (8), 47524761.

    VandeWoude, S., OBrien, S.J., Hoover, E.A., 1997a. Infectivity of lion and

    puma lentiviruses for domestic cats. J. Gen. Virol. 78 (Pt. 4), 795800.

    VandeWoude, S., OBrien, S.J., Langelier, K., Hardy, W.D., Slattery, J.P.,

    Zuckerman, E.E., Hoover, E.A., 1997b. Growth of lion and puma

    lentiviruses in domestic cat cells and comparisons with FIV. Virology

    233 (1), 185192.Vodros, D., Thorstensson, R., Biberfeld, G., Schols, D., De Clercq, E.,

    Fenyo, E.M., 2001. Coreceptor usage of sequential isolates from