Skip to main content
Journal of Virology logoLink to Journal of Virology
. 2018 Oct 12;92(21):e00830-18. doi: 10.1128/JVI.00830-18

Acute-Phase CD4+ T Cell Responses Targeting Invariant Viral Regions Are Associated with Control of Live Attenuated Simian Immunodeficiency Virus

Matthew S Sutton a, Amy Ellis-Connell a, Ryan V Moriarty b, Alexis J Balgeman a, Dane Gellerup b, Gabrielle Barry b, Andrea M Weiler b, Thomas C Friedrich b,c, Shelby L O'Connor a,b,
Editor: Guido Silvestrid
PMCID: PMC6189504  PMID: 30111562

Studies defining effective cellular immune responses to human immunodeficiency virus (HIV) and SIV have largely focused on a rare population that express specific MHC class I alleles and control virus replication in the absence of antiretroviral treatment. This leaves in question whether similar effective immune responses can be achieved in the larger population. The majority of HIV-infected individuals mount CD8+ T cell responses that target variable viral regions that accumulate high-frequency escape mutations. Limiting T cell responses to these variable regions and targeting invariant viral regions, similar to observations in rare “elite controllers,” may provide an ideal strategy for the development of effective T cell responses in individuals with diverse MHC genetics. Therefore, it is of paramount importance to determine whether T cell responses can be redirected toward invariant viral regions in individuals without protective MHC alleles and if these responses improve control of virus replication.

KEYWORDS: CD4+ T cells, CD8+ T cells, Mauritian cynomolgus macaque, invariant epitope, live attenuated SIV, variable epitope

ABSTRACT

We manipulated SIVmac239Δnef, a model of major histocompatibility complex (MHC)-independent viral control, to evaluate characteristics of effective cellular responses mounted by Mauritian cynomolgus macaques (MCMs) that express the M3 MHC haplotype, which has been associated with poor control of pathogenic simian immunodeficiency virus (SIV). We created SIVΔnef-8x to test the hypothesis that effective SIV-specific T cell responses targeting invariant viral regions can emerge in the absence of immunodominant CD8+ T cell responses targeting variable epitopes and that control is achievable in individuals lacking known “protective” MHC alleles. Full-proteome gamma interferon (IFN-γ) enzyme-linked immunospot (ELISPOT) assays identified six newly targeted immunogenic regions following SIVΔnef-8x infection of M3/M3 MCMs. We deep sequenced circulating virus and found that four of the six newly targeted regions rarely accumulated mutations. Six animals infected with SIVΔnef-8x had T cell responses that targeted at least one of the four invariant regions and had a lower set point viral load than two animals that did not have T cell responses that targeted any invariant regions. We found that MHC class II molecules restricted all four of the invariant peptide regions, while the two variable regions were restricted by MHC class I molecules. Therefore, in the absence of immunodominant CD8+ T cell responses that target variable regions during SIVmac239Δnef infection, individuals without protective MHC alleles developed predominantly CD4+ T cell responses specific for invariant regions that may improve control of virus replication. Our results provide some evidence that antiviral CD4+ T cells during acute SIV infection can contribute to effective viral control and should be considered in strategies to combat HIV infection.

IMPORTANCE Studies defining effective cellular immune responses to human immunodeficiency virus (HIV) and SIV have largely focused on a rare population that express specific MHC class I alleles and control virus replication in the absence of antiretroviral treatment. This leaves in question whether similar effective immune responses can be achieved in the larger population. The majority of HIV-infected individuals mount CD8+ T cell responses that target variable viral regions that accumulate high-frequency escape mutations. Limiting T cell responses to these variable regions and targeting invariant viral regions, similar to observations in rare “elite controllers,” may provide an ideal strategy for the development of effective T cell responses in individuals with diverse MHC genetics. Therefore, it is of paramount importance to determine whether T cell responses can be redirected toward invariant viral regions in individuals without protective MHC alleles and if these responses improve control of virus replication.

INTRODUCTION

During human immunodeficiency virus (HIV) infection, virus-specific CD8+ T cell responses are associated with resolution of peak viremia. These responses exert substantial immune pressure that often results in rapid selection for viral escape variants, suggesting that limiting viral escape is beneficial and may prove critical in the design of immunotherapies for HIV (14). There is evidence that the control of viremia associated with individuals expressing specific “protective” major histocompatibility complex (MHC) class I alleles may be attributed to CD8+ T cells that target specific peptide epitopes within highly invariant regions where mutations are likely to impose a significant fitness cost (58). However, this rare population of “elite controllers” is estimated at less than 1% of the infected population, while the majority of HIV-infected individuals do not express protective MHC alleles and more frequently mount CD8+ T cell responses that target viral regions that tolerate escape mutations easily (9, 10). Recently, multiple lines of evidence also indicate a nontraditional cytolytic role of HIV-specific CD4+ T cells that cooperate with HIV-specific CD8+ T cells to mediate suppression of virus replication and may be predictive of disease outcome (1113). It is essential for the design of vaccines and therapeutics to determine if virus-specific CD8+ and/or CD4+ T cell responses can be mounted by individuals not expressing protective MHC alleles and if these responses are effective at controlling viremia. For an intervention to be truly effective, a universal approach that can contend with the extraordinary sequence diversity of HIV in people with and without protective HLA alleles is needed.

Using nonhuman primates (NHPs), we can determine if acute-phase T cell responses targeting invariant viral regions can control primary viremia. Similar to humans, some macaques express protective MHC class I alleles associated with control of simian immunodeficiency virus (SIV) replication (1416). However, these studies have yet to define why control of virus replication in individuals expressing protective MHC alleles is incompletely penetrant, and they do not address how to induce viral control in animals without protective MHC alleles (17). Far less is known about the specificity of SIV-specific CD4+ T cell responses and whether they may also directly suppress virus replication. Only recently has there been interest in developing immunogens to elicit antiviral T cells targeting conserved viral regions across individuals with diverse MHC alleles in vivo (1820). Mauritian cynomolgus macaques (MCMs) are ideal for studying pathogen-specific T cells because they have extremely restricted MHC class I and II genetics, so that nearly all of their MHC alleles can be accounted for by 7 common haplotypes, termed M1 to M7 (21). As a result, animals with identical MHC alleles with the potential to present identical T cell peptide epitopes can be selected for studies (21, 22).

Our group and others have reported that M3/M3 MCMs poorly control infection with pathogenic SIVmac239, making them a good example of individuals with “nonprotective” MHC alleles in which to characterize favorable immune responses that could be elicited in a greater proportion of the population (23, 24). Unlike pathogenic SIVmac239, replication of live-attenuated SIVmac239Δnef is controlled in nearly every infected animal, regardless of host MHC genetics (25, 26). Control of SIVmac239Δnef replication in a host with nonprotective MHC alleles may provide a more favorable environment in which to find the characteristics of effective immune responses that control pathogenic virus replication in the broader population. Therefore, this unique model of MHC-independent control in M3/M3 MCMs may allow the characterization of effective T cell responses in animals without protective MHC alleles.

Previously, our group reported data suggesting that control of SIVmac239Δnef relied on immunodominant CD8+ T cell responses that select for escape mutations (25). However, at the time of our previous study, the CD8+ T cell responses restricted by MCMs expressing the M3 haplotype were incompletely known, and no SIV-specific M3-restricted CD4+ T cell responses had been identified. Additionally, the m3KOΔnef virus used in that study included additional mutations outside known M3-restricted epitopes with unknown impacts on virus replication (25). We wanted to improve upon the m3KOΔnef virus by creating a virus in which only known epitopes were disturbed and mutations in other regions of the virus were avoided. Since that time, we have improved our understanding of M3-restricted CD8+ T cell epitopes and now know of 10 epitopes in SIVmac239 that select for high-frequency mutations (22, 25, 27, 28).

In the current study, we used this new information to create a variant of SIVmac239Δnef, termed SIVΔnef-8x, that ablated the eight M3 MHC class I-restricted epitopes that accumulate mutations during infection with SIVmac239Δnef. We hypothesized that limiting the development of CD8+ T cell responses targeting highly variable epitopes might promote the development of alternate T cell responses that target invariant regions to suppress SIVmac239Δnef replication in animals with nonprotective MHC class I alleles. We identified six immunogenic regions in SIVΔnef-8x whose immunogenicity had not previously been defined in SIV-infected M3/M3 MCMs. Four of these regions did not accumulate mutations, despite eliciting detectable responses. Interestingly, all four invariant regions were restricted by M3 MHC class II molecules and were made exclusively by animals that controlled replication of SIVΔnef-8x. These data suggest that viral control is achievable in animals with nonprotective MHC alleles even when immunodominant CD8+ T cell responses that are normally elicited during SIVmac239Δnef infection are absent. Our findings provide support for the inclusion of immunogens able to elicit CD4+ T cell responses that target invariant viral antigens in the design of an effective HIV vaccine, as this approach may be applied for widespread use across individuals with a diverse array of MHC genetics.

(This article was submitted to an online preprint archive [29].)

RESULTS

Construction of SIVΔnef-8x.

We engineered a variant of SIVmac239Δnef with point mutations in eight CD8+ T cell epitopes restricted by MHC class I molecules expressed by the M3 haplotype (Fig. 1a). All eight epitopes are highly immunogenic and accumulate high-frequency escape mutations in response to immunodominant CD8+ T cell pressure elicited during infection with either SIVmac239 or SIVmac239Δnef (16, 27, 28). For each epitope, we used sequence data from nine M3/M3 MCMs chronically infected with SIVmac239 to identify common variants in the replicating virus population (27). We performed gamma interferon (IFN-γ) enzyme-linked immunospot (ELISPOT) assays with the variant peptides of each of the 8 epitopes. Using peripheral blood mononuclear cells (PBMC) collected from several SIV-infected M3/M3 MCMs, we identified variant peptides for each epitope that elicited an IFN-γ ELISPOT response in vitro significantly lower than responses against the corresponding wild-type peptide (data not shown). We then used either these identified variant peptide sequences or the variant peptide sequences we included in the m3KOΔnef virus in SIVmac239Δnef virus (25). We referred to the resulting virus as SIVΔnef-8x to indicate its origin as an SIVmac239Δnef derivative with point mutations aimed at disrupting the immunogenicity of the eight variable M3-restricted CD8+ T cell epitopes present in SIVmac239Δnef (Fig. 1a).

FIG 1.

FIG 1

Construction of SIVΔnef-8x. (a) Diagram of live-attenuated SIV with epitope locations (top) and amino acid sequence differences between SIVΔnef-8x and SIVmac239Δnef (bottom). Point mutations were engineered into 8 M3-restricted epitopes known to accumulate high-frequency mutations during SIV infection. (b) SIVΔnef-8x and SIVmac239Δnef in vitro coculture fitness assay. For each assay, we included BCVΔnef as a reference at a 1:9, 1:1, or 9:1 ratio of p27 content relative to the query virus, either wild-type SIVmac239Δnef or SIVΔnef-8x, in the inoculum. The numbers of copies of the query virus and the barcoded virus were determined by qRT-PCR at each time point. A ratio of the number of copies of query virus to that of barcoded virus was determined and compared to the ratio present in the inoculum. All the data represent means with standard deviation of triplicate values. Unpaired t tests were performed at each time point. *, P < 0.05.

To test if the epitope variants incorporated into SIVΔnef-8x affected viral fitness, we performed in vitro coculture competition assays with a barcoded SIVmac239Δnef (BCVΔnef) containing 10 synonymous changes in gag that can be detected by a separate quantitative-PCR (qPCR) assay (25, 30). Using different ratios of BCVΔnef relative to the query virus (SIVmac239Δnef or SIVΔnef-8x), our data indicated that the eight variant epitopes we incorporated into SIVΔnef-8x did not substantially alter viral fitness in vitro (Fig. 1b).

Engineered variant epitopes in SIVΔnef-8x prevent the development of responses to wild-type epitopes and are minimally immunogenic in vivo.

We infected eight M3/M3 MCMs with SIVΔnef-8x, six M3/M3 MCMs with SIVmac239Δnef, and three M4/M6 MCMs with SIVΔnef-8x (Table 1). The T cell responses present in M4/M6 MCMs infected with SIVΔnef-8x should mirror those that would develop in M4/M6 MCMs infected with SIVmac239Δnef, as these animals do not express any of the alleles of the M3 MHC haplotype (21). To determine if the mutations we incorporated into SIVΔnef-8x were sufficient to prevent the development of the expected responses targeting the eight wild-type epitopes, we performed IFN-γ ELISPOT assays using wild-type and variant peptides with PBMC collected throughout infection. At 3 weeks postinfection, PBMC from all six M3/M3 animals infected with SIVmac239Δnef recognized up to five of the eight M3-restricted wild-type epitopes (Fig. 2a, blue). In contrast, only one of eight M3/M3 animals infected with SIVΔnef-8x had positive IFN-γ ELISPOT responses for any of the wild-type epitopes (Fig. 2a, black). None of the M4/M6 MCMs infected with SIVΔnef-8x made responses specific for the eight wild-type M3-restricted epitopes (data not shown). Similar results were observed in all the animals at 8 weeks post-SIVΔnef-8x infection (Fig. 2b, black) and 12 weeks post-SIVmac239Δnef infection (Fig. 2b, blue). Therefore, M3/M3 MCMs infected with SIVΔnef-8x did not develop the previously reported CD8+ T cell responses that target variable epitopes during SIVmac239Δnef infection.

TABLE 1.

Animals used in this study

Animal ID Gender MHC haplotype Infecting virus
cy0684 Female M3/M3 SIVmac239Δnef
cy0686 Female M3/M3 SIVmac239Δnef
cy0687 Male M3/M3 SIVmac239Δnef
cy0689 Male M3/M3 SIVmac239Δnef
cy0749 Male M3/recM1M3a SIVmac239Δnef
cy0752 Female M3/recM2M3a SIVmac239Δnef
cy0748 Male M4/M6 SIVΔnef-8x
cy0751 Female M4/M6 SIVΔnef-8x
cy0754 Male M4/M6 SIVΔnef-8x
cy0685 Female M3/M3 SIVΔnef-8x
cy0688 Male M3/M3 SIVΔnef-8x
cy0690 Male M3/M3 SIVΔnef-8x
cy0750 Male M3/M3 SIVΔnef-8x
cy0753 Female M3/M3 SIVΔnef-8x
cy0755 Female M3/M3 SIVΔnef-8x
cy0756 Female M3/M3 SIVΔnef-8x
cy0757 Female M3/M3 SIVΔnef-8x
a

Expressed the major MHC class I A and B alleles present in the M3 MHC haplotype, but also expressed minor MHC class I A alleles of the M1 (cy0749) and M2 (cy0752) MHC haplotypes. Transcriptionally abundant (major) MHC-I transcripts are responsible for restricting SIV-specific CD8+ T cell responses (27), and both animals expressed the major MHC class I A allele present in the M3 haplotype (A1*063). Accordingly, we include these functional M3/M3 animals in our group of M3/M3 MCMs infected with SIVmac239Δnef.

FIG 2.

FIG 2

M3-restricted CD8+ T cell responses elicited during SIVmac239Δnef infection are silenced in M3/M3 MCMs infected with SIVΔnef-8x. (a and b) IFN-γ ELISPOT assays using peptides that matched the wild-type epitope sequences were performed at week 3 (a) and week 8 or 12 (b) postinfection with SIVmac239Δnef (blue) or SIVΔnef-8x (black). (c and d) IFN-γ ELISPOT assays using peptides that matched the variant epitope sequences present in SIVΔnef-8x were performed at week 3 (c) and week 8 (d) post-SIVΔnef-8x infection.

To determine if the variants incorporated in SIVΔnef-8x were immunogenic, we performed parallel IFN-γ ELISPOT assays using peptides that matched the variant epitope sequences engineered into SIVΔnef-8x. At 3 weeks post-SIVΔnef-8x infection, we observed positive IFN-γ ELISPOT responses to only two of eight variant epitopes (Fig. 2c). One variant epitope, Env338-346RF9 K3R/W8R, was immunogenic in all eight animals at 3 weeks postinfection. Another variant epitope, Tat42-49QA8 R4H, elicited a response in two animals. These responses were diminished 8 weeks postinfection, with only one animal detecting the variant epitope Env338-346RF9 K3R/W8R (Fig. 2d).

Similar in vivo pathogenicities of SIVΔnef-8x and SIVmac239Δnef in M3/M3 MCMs.

Following infection, we assessed the pathogenicity of SIVΔnef-8x compared to SIVmac239Δnef by measuring plasma viremia and memory CD4+ T cell subsets. The limit of detection for the viral load assay was 100 copies/ml. We did not observe the peak viral load to be significantly different (P = 0.158) in M3/M3 MCMs infected with SIVΔnef-8x and those infected with SIVmac239Δnef (Fig. 3a). While peak viremia of SIVΔnef-8x was slightly lower (P = 0.046) in M4/M6 MCMs (n = 3) than in M3/M3 MCMs (n = 8), it was not significantly different (P = 0.275) from that of SIVmac239Δnef in M3/M3 MCMs (n = 6). We also evaluated the set point viral load, calculated as the geometric mean of viral loads between 14 and 30 weeks postinfection with SIVmac239Δnef or SIVΔnef-8x (Fig. 3b). Five of six M3/M3 MCMs infected with SIVmac239Δnef established a set point viral load at or near an undetectable level (median = 104 vRNA copies/ml), while one animal (cy0687) had a set point viral load of 18,300 vRNA copies/ml. In contrast, virus levels by week 30 were more diverse in SIVΔnef-8x-infected M3/M3 MCMs and ranged from nearly undetectable to over 7,000 vRNA copies/ml (median = 468 vRNA copies/ml). Virus replication was controlled below 1,000 vRNA copies/ml in six animals, four of which maintained a set point viral load below 250 vRNA copies/ml. In contrast, two animals (cy0690 and cy0755) failed to control replication during the chronic phase of infection and had circulating virus levels that exceeded 3,500 vRNA copies/ml. A set point viral load was undetectable in two M4/M6 MCMs and fluctuated around 1,000 vRNA copies/ml in one M4/M6 MCM. Set point viral load comparisons also revealed no significant differences between groups.

FIG 3.

FIG 3

Similar in vivo pathogenicities of SIVΔnef-8x and SIVmac239Δnef in M3/M3 MCMs. Plasma viral load and memory CD4+ T cell populations were measured for 30 weeks after infection: M3/M3 MCMs infected with SIVΔnef-8x (black), M3/M3 MCMs infected with SIVmac239Δnef (blue), and M4/M6 MCMs infected with SIVΔnef-8x (red). (a) Individual peak viral loads in animals infected with SIVΔnef-8x or SIVmac239Δnef displayed as geometric means with 95% confidence intervals. (b) Comparisons of set point viral loads (geometric mean of viral loads between 14 and 30 weeks postinfection). (c) Longitudinal assessment of changes in the absolute count of memory CD4+ T cell subsets over time in animals infected with SIVΔnef-8x and SIVmac239Δnef. Effector memory CD4+ T cells were defined as CD3+ CD4+ CD8 CD95+ CD28 CCR7, and central memory CD4+ T cells were defined as CD3+ CD4+ CD8 CD95+ CD28+ CCR7+. An unpaired Student t test was used to calculate significant differences for the peak viral load and set point viral load.

To determine if CD4+ T cells were depleted following infection with SIVΔnef-8x compared to infection with SIVmac239Δnef, we evaluated changes in the absolute counts of central memory CD4+ T cells and effector memory CD4+ T cells in the blood at select time points (Fig. 3c). Though transient changes to both memory CD4+ T cell populations existed, we did not observe substantial differences between groups during peak viremia or the establishment of set point viral loads. Taken together, these results suggest that animals infected with SIVΔnef-8x did not exhibit enhanced peripheral depletion of CD4+ T cells compared to animals infected with SIVmac239Δnef.

A majority of M3/M3 MCMs control SIVΔnef-8x.

Next, we compared viral load trajectories and the time to control virus replication in MCMs infected with SIVΔnef-8x or SIVmac239Δnef. Plasma viral loads were measured in M3/M3 MCMs infected with SIVΔnef-8x (Fig. 4a, black) or SIVmac239Δnef (blue) and in M4/M6 MCMs infected with SIVΔnef-8x (red) for 30 weeks following infection.

FIG 4.

FIG 4

Similar in vivo pathogenicities of SIVΔnef-8x and SIVmac239Δnef in M3/M3 MCMs. Plasma viral loads and memory CD4+ T cell populations were measured for 30 weeks after infection: M3/M3 MCMs infected with SIVΔnef-8x (black), M3/M3 MCMs infected with SIVmac239Δnef (blue), and M4/M6 MCMs infected with SIVΔnef-8x (red). (a) Individual peak viral loads in animals infected with SIVΔnef-8x or SIVmac239Δnef displayed as geometric means with 95% confidence intervals. (b) Comparisons of set point viral loads (geometric mean of viral loads between 14 and 30 weeks postinfection). (c) Longitudinal assessment of changes in the absolute count of memory CD4+ T cell subsets over time in animals infected with SIVΔnef-8x and SIVmac239Δnef. Effector memory CD4+ T cells were defined as CD3+ CD4+ CD8 CD95+ CD28 CCR7, and central memory CD4+ T cells were defined as CD3+ CD4+ CD8 CD95+ CD28+ CCR7+. An unpaired Student t test was used to calculate significant differences for the peak viral load and set point viral load.

We compared the initial times required to control virus replication below a specific threshold and modeled this with Kaplan-Meier survival curves. We included all three groups in this analysis, even though the M4/M6 group had only three animals. The M4/M6 animals were used only to ensure that the in vivo fitness of SIVΔnef-8x was similar to that of SIVmac239Δnef, but our interest lay in comparing viral control in the two M3/M3 groups. Using a threshold for control similar to that of elite controller macaques infected with SIVmac239 (<1,000 vRNA copies/ml), we observed no statistically significant differences in the times required to control virus replication among the three cohorts (Fig. 4b) (14, 15, 24, 30, 31). We then evaluated time to control using a threshold matching the limit of detection for the viral load assay (<100 vRNA copies/ml) because, in most animals, SIVmac239Δnef was controlled at or near the limit of detection (25, 26). Using this threshold, we observed a statistically significant delay in the time required to control SIVΔnef-8x compared to SIVmac239Δnef in M3/M3 MCMs (Fig. 4c). Thus, when either threshold was set for viral control, at least half of the M3/M3 MCMs controlled virus replication. This suggests that M3/M3 MCMs can control virus replication even when they do not develop acute CD8+ T cells that select for escape mutations.

Acute-phase T cell responses elicited in M3/M3 MCMs infected with SIVΔnef-8x target several viral regions that do not accumulate mutations.

To determine if there were T cell responses present in M3/M3 MCMs infected with SIVΔnef-8x that targeted previously undefined T cell epitopes, we performed full-proteome IFN-γ ELISPOT assays with overlapping peptide pools to scan the entire SIVmac239 proteome. Positive peptide pools were then deconvoluted to assess responses to individual peptides. During acute infection (week 3) and post-peak infection (weeks 7 to 9), we identified six immunogenic regions in the SIVΔnef-8x proteome that elicited T cell responses to peptide sequences in Gag (n = 5) and Env (n = 1) that were not previously characterized in SIV-infected M3/M3 MCMs. During acute infection, all six animals that controlled SIVΔnef-8x replication responded to at least one of these new regions. Four out of these six animals made responses against 3 or more of the new regions (Fig. 5a, left). We observed similar results at 8 weeks postinfection, when five out of the six animals controlling virus replication below 1,000 vRNA copies/ml made one or more new responses (median = 2) (Fig. 5a, right). No responses were detected to any of these new regions during acute or post-peak infection in the two animals (cy0690 and cy0755) that did not control SIVΔnef-8x replication below either threshold.

FIG 5.

FIG 5

A majority of newly targeted regions do not accumulate high-frequency mutations during SIV infection. (a) Full-proteome IFN-γ ELISPOT assays were performed at an acute time point (week 3) and a postpeak time point (weeks 7 to 9). Peptide pools were deconvoluted to assess responses to individual peptides, resulting in the identification of responses targeting six viral regions previously undefined in M3/M3 MCMs. Only positive responses are shown, with solid bars representing assays using freshly isolated PBMC and hatched bars representing assays using frozen PBMC. (b) Virus populations were deep sequenced from plasma from M3/M3 MCMs during acute infection and post-peak infection with SIVΔnef-8x. (c) The six regions were also analyzed in M3/M3 MCMs chronically infected with SIVmac239 (>52 weeks) from a previous study by our group. The heat maps represent the percent sequence identity in relation to the inoculum. Darker colors correspond to higher sequence identity. n.c, no sequence coverage.

We deep sequenced viral populations in parallel to determine if the six newly targeted regions accumulated point mutations. During acute infection, only one of the six new regions, Gag57–71CG15, accumulated high-frequency mutations in a majority of animals (Fig. 5b, left). When we examined the sequences from virus populations isolated during post-peak infection (weeks 7 to 9), four of the six new regions remained nearly identical to the inoculum, while Gag57–71CG15 and Env329–347VG19 had accumulated high-frequency mutations (Fig. 5b, right). To determine whether these 4 apparently invariant regions could accumulate mutations during pathogenic SIV infection, we examined variant accumulation in these 4 regions in virus populations isolated and sequenced from 9 M3/M3 MCMs that had been infected with SIVmac239 for ∼52 weeks for a previous study by our group (Fig. 5c) (27). In 3 of these regions, we found that 70 to 99% of the sequences (median = 97%) matched wild-type SIVmac239 in all nine animals. In eight animals, more than 80% of Gag25–39GN15 sequences matched wild-type SIVmac239, while one animal had only 30% of sequences that matched wild-type SIVmac239. Taken together, our data suggest that it is possible to exert viral control when animals develop acute-phase T cell responses targeting regions that do not readily accumulate mutations.

CD4+ T cells targeting regions that do not accumulate mutations are common during SIVΔnef-8x infection.

We wanted to determine whether CD4+ or CD8+ T cells were responsible for targeting the six immunogenic regions identified in the IFN-γ ELISPOT assays. We grew polyclonal T cell lines specific for each of the six peptides and mapped MHC restriction. Of the four invariant viral regions that elicited responses, Mafa-DRA*01:02:01/DRB1*10:02 restricted both Gag249–263WY15 and Gag297–315Y19, while Mafa-DPA1*13:01/DPB1*09:02 restricted both Gag25–39GN15 and Gag413–427GC15 (Fig. 6a). MHC restriction was also mapped for the two responses targeting viral regions that accumulated high-frequency mutations during SIVΔnef-8x infection. Using the optimal peptides contained within the regions, we found that Gag57–71CG15 was restricted by Mafa-B*011:01 while Env329–347VG19 was restricted by Mafa-A1*063:02 (Fig. 6b). A summary of the newly targeted regions and their restricting alleles is shown in Table 2. Thus, acute-phase CD4+ T cells targeting viral regions that do not accumulate mutations are common during acute infection in animals without favorable MHC genetics that ultimately control virus replication.

FIG 6.

FIG 6

Characterization of M3-restricted SIVΔnef-8x T cell responses. ICS assays were performed to determine MHC class II restriction for CD4+ T cell lines specific for the four identified invariant regions (a) and MHC class I restriction for CD8+ T cell lines specific for the two identified variable regions (b). The data represent the percentages of cells positive for IFN-γ and/or TNF-α for each T cell line. MHC-matched 721.221 cells or K562 cells expressing the indicated M3 MHC class I alleles or RM3 cells expressing the indicated M3 MHC class II alleles were used as antigen-presenting cells. Peptide-pulsed untransfected 721.221 cells, K562 cells, or RM3 cells were used as negative controls.

TABLE 2.

Characterization of M3-restricted SIVΔnef-8x T cell responses

Immunogenic region Invariant Epitope sequence Allele specificity
Gag25-39GN15 Yes GKKKYMLKHVVWAAN DPA1*13:01/DPB1*09:02
Gag57-71CG15 No CQKILSVLAPLVPTG B*011:01
Gag249-263WY15 Yes WMYRQQNPIPVGNIY DRA*01:02:01/DRB1*10:02
Gag297-315YK19 Yes YVDRFYKSLRAEQTDAAVK DRA*01:02:01/DRB1*10:02
Gag413-427GC15 Yes GCWKCGKMDHVMAKC DPA1*13:01/DPB1*09:02
Env329-347VG19 No VFHSQPINDRPKQAWCWFG A1*063:02

DISCUSSION

Rare individuals who express protective MHC alleles control HIV replication without antiretroviral treatment and often make CD8+ T cell responses that target highly invariant, possibly evolutionarily conserved, viral regions (7, 8, 32). In contrast, responses to invariant viral regions are frequently subdominant in HIV-infected individuals who do not express protective MHC alleles (10). It is hypothesized that an effective vaccine will need to limit T cell responses to highly immunogenic variable regions in order to maximize the likelihood of developing T cell responses against invariant viral regions (3335). Using SIVΔnef-8x, we directly tested this hypothesis in M3/M3 MCMs that did not express protective MHC alleles and observed that control of SIVmac239Δnef could still be achieved.

Cellular responses that target Gag during acute HIV infection have been associated with a low viral load and improved disease outcome (2, 8, 9). Out of the six regions that elicited T cell responses in M3/M3 MCMs infected with SIVΔnef-8x, five were located within Gag. All six animals that controlled SIVΔnef-8x replication made T cell responses during acute infection that recognized one or more of the following five regions that span the majority of Gag: Gag249–263WY15 and Gag297–315YK19 are located within the p27 capsid (CA), Gag57–71CG15 and Gag25–39GN15 are located within the p15 matrix (MA), and Gag413–427GC15 is located within p6. Of note, the first 9 amino acids of Gag297–315YK19 are present in an evolutionarily conserved motif known as the major homology region (MHR) and share three of the four residues identified as highly conserved within the MHR (36). The MHR is also contained within the C-terminal subdomain of CA (CA-CTD), one of only two regions of HIV-1 Gag that are absolutely required for assembly, suggesting the region may represent an ideal target for T cell-based vaccines (37). In all six animals that controlled SIVΔnef-8x, four of the five Gag regions that were targeted did not accumulate mutations. Interestingly, the same four regions did not accumulate high-frequency mutations in nearly all nine M3/M3 MCMs chronically infected with SIVmac239 (>52 weeks) that were sequenced by our group for a previous study (Fig. 5c) (27).

Besides the four invariant regions, we found two regions that accumulated mutations during acute SIVΔnef-8x infection. Gag57–71CG15 accumulated both Q58R and V63A mutations by 3 weeks postinfection that were present at a frequency of 7% to 37% and 4% to 88%, respectively. By 9 weeks postinfection, less than 1% of the circulating virus matched the inoculum in Gag57–71CG15 for all eight M3/M3 MCMs infected with SIVΔnef-8x (Fig. 5b, right). Even though the V63A mutation in Gag57–71CG15 was observed coincident with breakthrough viremia in an elite controller rhesus macaque infected with SIVmac239 (31), we found the V63A mutation was present in virus populations from both controllers and noncontrollers of SIVΔnef-8x (data not shown). Our results suggest that this mutation alone does not confer breakthrough replication, so perhaps targeting this region of Gag may still offer some immunological benefit.

We also observed high-frequency mutations in Env329–349VG19, a region that is located at the C terminus of the V3 loop (38). This region also contains the variant epitope Env338–346RF9 K3R/W8R that we engineered into SIVΔnef-8x and was immunogenic during acute infection in all eight animals (Fig. 2c). The most common mutation we observed within the newly targeted Env329–349VG19 was an R-to-W change at position 345 of Env that restored one of the two mutations in Env338–346RF9 to the original SIVmac239 sequence. It is possible that the R-to-W reversion was a consequence of the development of T cells targeting Env338–346RF9 K3R/W8R during acute infection. Alternatively, it is possible that tryptophan at position 345 may be more favorable than the arginine we engineered into the virus. Given the proximity of this arginine to the C311-C344 link that serves as the base of the V3 loop, it seems possible that the polarity of the amino acid at position 345 may impact the formation of the V3 loop. The relatively conserved nature of the V3 loop, as well as its role in determining coreceptor tropism, further supports this region as a site for effective T cell responses to exert their antiviral function during acute infection (3840).

There is growing evidence that HIV-specific CD4+ T cells may play a bigger cytolytic role in control of virus replication than previously thought (11, 32, 41, 42). SIV-specific CD4+ responses have been previously detected in macaques infected with pathogenic SIV, as well as SIVmac239Δnef (4345). While observed to be typically subdominant in comparison to many CD8+ T cell responses, immune pressure imparted by CD4+ T cells has been sufficient to select for escape variants in certain cases (30, 31, 46). We found four immunogenic regions that elicited acute-phase CD4+ T cells in M3/M3 animals infected with SIVΔnef-8x. All four of these were located in Gag and did not accumulate mutations by 9 weeks postinfection despite eliciting positive IFN-γ ELISPOT responses, suggesting they may represent invariant viral regions that are immunogenic. Notably, we deep sequenced virus populations in M3/M3 MCMs infected with SIVmac239 for >52 weeks as part of a previous study by our laboratory and found that the same four regions of Gag did not routinely accumulate mutations, though it is unknown whether these regions elicited virus-specific CD4+ T cells (Fig. 5c) (27). Although we were unable to dissect a mechanism of CD4+ T cell-mediated control of virus replication during acute SIVΔnef-8x infection, cytolytic CD4+ T cells have been previously implicated in control of viral infections (11, 41, 47). This argument is in contrast to virus-specific CD4+ T cells as preferential targets of infection and may be explained by distinct transcriptional and functional signatures of cytolytic CD4+ T cells that mirror CD8+ T cells more than Th1 CD4+ cells (13, 48, 49). Together, we provide evidence that acute-phase CD4+ T cells may improve control of SIV replication, and we provide a model through which to further explore this mechanism in future studies.

It is certainly possible that infection with SIVΔnef-8x did not deplete peripheral CD4+ T cells, in contrast to reports of macaques infected with SIVmac239 (5052). As a result, this could increase the likelihood that SIV-specific CD4+ T cell responses were preserved and elicited following infection with SIVΔnef-8x. While changes to the numbers of effector memory and central memory CD4+ T cells could not account for the differential disease progression observed in animals infected with SIVΔnef-8x, it is possible that the activation states of these populations may have been an influencing factor. Previously, macaques infected with SIVmac239 have been shown to have increased activation of memory CD4+ T cells (52). In contrast, macaques infected with SIVmac239Δnef typically do not generate activated memory CD4+ T cells, and disease progression rarely occurs (45, 52). Still, there is evidence that animals infected with SIVmac239Δnef can exhibit some limited T cell proliferation during early infection (51). Thus, future studies of animals infected with SIVΔnef-8x may warrant an examination of T cell activation in the periphery and mucosal tissues to assess if there is an association with viral control.

We found that two M3/M3 MCMs infected with SIVΔnef-8x (cy0690 and cy0755) and one M3/M3 MCM infected with SIVmac239Δnef (cy0687) were unable to control virus replication, similar to what was seen in the animals of the Harris et al. study (25). Both cy0690 and cy0755 did not have T cell responses detectable by IFN-γ ELISPOT assays. When we sequenced virus populations isolated from cy0690 and cy0687, we found an additional deletion in nef that had previously been shown restore the reading frame and to result in increased pathogenicity (data not shown) (53). This observation prompted us to reexamine the sequences of virus populations replicating during chronic infection from the M3/M3 MCMs of the Harris et al. study. We found similar sequence changes within the same region of nef in virus populations isolated from these animals (data not shown). The precise mechanism by which nef is restored in certain animals and whether functional advantages are conferred remains to be determined and requires further investigation.

Together, our study suggests that perhaps expanding subdominant virus-specific CD4+ T cells toward invariant viral regions during early infection may improve viral control. Even though not every animal in our study was able to mount these responses and elicit viral control, our data provide compelling evidence that CD4+ T cell responses targeting MHC class II-restricted epitopes have the potential to be effective and can be mounted by individuals without protective MHC alleles. To our knowledge, this is the first identification of MHC class II-restricted SIV-specific T cell responses in MCMs. Future nonhuman primate studies may consider focusing on effective SIV-specific CD4+ T cell responses and evaluating their direct contribution to controlling virus replication in vivo.

MATERIALS AND METHODS

Animal care and use.

Seventeen MCMs were purchased from Bioculture Ltd. and were housed and cared for by the Wisconsin National Primate Research Center (WNPRC) according to protocols approved by the University of Wisconsin Graduate School Animal Care and Use Committee. The animals were chosen based on expression of particular MHC alleles as previously described (5458). Eight M3/M3 MCMs and three M4/M6 MCMs were infected intravenously with 10 ng of p27 SIVΔnef-8x. Four M3/M3 MCMs and two functional M3/M3 MCMs were infected intravenously with 10 ng p27 from SIVmac239Δnef. Four of these MCMs were infected as part of a previous study (26), and two animals (cy0749 and cy0752) were functionally M3/M3 (Table 1).

Creation of virus stocks.

We created plasmids (pUC57) containing the 5′ and 3′ viral genomes of SIVmac239Δnef by custom gene synthesis (GenScript, Piscataway, NJ), as was done previously (25). For SIVΔnef-8x, 10 substitutions were then incorporated into SIVmac239Δnef by site-directed mutagenesis (GenScript, Piscataway, NJ). The plasmids containing the 5′ and 3′ halves of the corresponding genomes were digested with SphI, treated with Antarctic phosphatase, precipitated, and ligated together. Vero cells were transfected with the ligated products and cocultured with CEMx174 cells for 48 h. The infected CEMx174 cells were grown for ∼2 weeks to produce high-titer viruses and harvested daily during the last week. Plasma SIV loads were determined by quantitative reverse transcription (qRT)-PCR, and the p27 content of the virus stocks was determined by enzyme-linked immunosorbent assay (ELISA) (ZeptoMetrix Corp., Buffalo, NY) according to the manufacturer's protocol. The p27 content of SIVmac239Δnef was 327 ng/ml, and the viral load was 2.19e9 copies/ml. The p27 content of SIVΔnef-8x was 245 ng/ml, and the viral load was 1.39e9 copies/ml.

In vitro coculture competition fitness assays.

In triplicate, SIVmac239Δnef or SIVΔnef-8x was mixed with BCVΔnef at p27 content ratios of 1:1, 1:9, and 9:1. Each virus mixture was incubated with 1e6 CEMx174 cells at 37°C for 4 h. After washing, 5e5 cells were plated and grown for 1 week, with supernatant sampled at days 3, 5, and 7. A discriminating qPCR assay was used to quantify the copies of SIVmac239Δnef or SIVΔnef-8x and BCVΔnef, as was done previously (25, 30). Briefly, viral RNA (vRNA) was isolated from the inoculum and each supernatant and then quantified with a SuperScript III Platinum one-step quantitative-PCR kit (Invitrogen, Carlsbad, CA). In one reaction, primers and probes targeting an 84-bp region of gag were used to quantify SIVmac239Δnef and SIVΔnef-8x. A separate reaction with a distinct set of primers and probes was used to quantify BCVΔnef. The p27 content ratio of SIVΔnef-8x or SIVmac239Δnef to BCVΔnef in each supernatant was normalized to the ratio that was present in the inoculum, and replicative differences between viruses were assessed at each time point with unpaired Student t tests (GraphPad Prism, La Jolla, CA). All the data represent means with standard deviations of triplicate values and are plotted on a log2 scale.

Plasma viral load analysis.

Plasma was isolated from undiluted whole blood by Ficoll-based density centrifugation and cryopreserved at −80°C. SIV gag loads were determined as previously described (26). Briefly, vRNA was isolated from plasma, reverse transcribed, and amplified with the Superscript III Platinum one-step qRT-PCR system (Invitrogen). The detection limit of the assay was 100 vRNA copy equivalents per ml of plasma. The limit of detection (100 vRNA copies/ml) was reported when the viral load was at or below the limit of detection.

Peptides.

The NIH AIDS Research and Reference Reagent Program (Germantown, MD) provided 15-mer peptides overlapping by 11 amino acid positions spanning the full SIVmac239 proteome. Additional peptides used for mapping were created by custom synthesis (GenScript, Piscataway, NJ). All the peptide sequences were derived from the SIVmac239 proteome.

IFN-γ ELISPOT assays.

IFN-γ ELISPOT assays were performed using fresh and frozen PBMC, as previously described (26, 27). Each of the eight variant epitopes was selected using frozen PBMC collected from at least five M3/M3 MCMs during acute or chronic SIV infection. Fresh PBMC were isolated from EDTA-anticoagulated blood by Ficoll-based density centrifugation. A precoated monkey IFN-γ ELISPOTplus plate (Mabtech, Mariemont, OH) was blocked, and individual peptides were added to each well at a final concentration of 1 μM. Multiple peptide pools containing 15-mer peptides that spanned the full SIVmac239 proteome, each overlapping by 11 amino acids, were used to assess new responses that emerged during infection. The peptide pools totaled 10 μM (1 μM each peptide) and were added to cells at a final pool concentration of 1 μM. Each peptide or peptide pool was tested in duplicate, as was concanavalin A (10 μM), which was used as a positive control. Four to 10 wells did not receive any peptides and served as a negative control for calculating background reactivity. Assays were performed according to the manufacturer's protocol, and wells were imaged with an ELISPOT reader (AID Autoimmun Diagnostika GmbH). Positive responses were determined using a one-tailed t test and an α level of 0.05, where the null hypothesis was that the background level would be greater than or equal to the treatment level (59). Positive responses were considered valid only if each duplicate well had a value of at least 50 spot-forming cells (SFCs) per 106 PBMC. If statistically positive, the reported values represent the average of the test wells minus the average of all negative-control wells.

Generation of M3/M3 BLCLs.

MHC-matched B lymphoblastoid cell lines (BLCLs) were generated as previously described (28). Briefly, PBMC were isolated by density-based centrifugation from whole blood containing EDTA. B cells were then immortalized with medium from an S549 cell line containing herpesvirus papio. The cells were maintained in R-10 medium and sequenced to verify the presence of appropriate MHC alleles.

Generation of peptide-specific T cell lines.

Polyclonal CD8+ and CD4+ T cell lines were generated from whole-blood PBMC that were isolated by Ficoll-based density centrifugation. Based on the availability of blood, approximately 3 to 5 peptide-specific CD8+ T cell lines were generated from animals infected with SIVΔnef-8x, as previously described (28). Briefly, 5e6 freshly isolated PBMC were incubated with 2 μM peptide in complete medium (RPMI 1640 medium supplemented with 15% fetal calf serum, 1% antibiotic/antimycotic, and 1% l-glutamine) with 100 IU of interleukin-2 (NIH AIDS Research and Reference Reagent Program). After 1 week, the cell lines were restimulated weekly with peptide-pulsed irradiated (9,000 rads) BLCLs, as previously described (28, 60). Approximately 2 or 3 CD4+ T cell lines per peptide were generated and cultured as previously described (61). Briefly, freshly isolated PBMC were depleted of CD8+ cells using NHP-specific anti-CD8 microbeads (Miltenyi Biotech, San Diego, CA) via magnetic separation according to the manufacturer's protocol. After separation, 5e6 CD8-depleted cells were used to generate CD4+ T cell lines and cultured in complete medium similarly to CD8+ T cell lines, with the addition of interleukin-7 (BioLegend, San Diego, CA) at a final concentration of 50 ng/ml. In select cases, we attempted to generate multiple CD4+ T cell lines using whole PBMC but were unable to establish peptide-specific polyclonal T cell lines similar to those established following magnetic separation of CD8+ cells.

Intracellular-cytokine-staining (ICS) assay.

To determine T cell line peptide specificity and the restricting MHC class I or class II molecule, we measured intracellular expression of IFN-γ and tumor necrosis factor alpha (TNF-α) as previously described (28, 61). Briefly, 2e5 peptide-pulsed BLCLs or MHC class I or class II transferents were incubated for 1.5 h with peptide at 37°C, washed twice with RPMI medium containing 10% fetal bovine serum (FBS) (R10), and then combined with 2e5 cells from corresponding CD4+ or CD8+ T cell lines for an additional 4 h at 37°C in the presence of brefeldin A (Sigma-Aldrich, St. Louis, MO). The cells were incubated with LIVE/DEAD fixable near-infrared (IR) dead cell stain for 15 min before being surface stained with CD3-AF700 (clone SP34-2; BD Biosciences), CD4-allophycocyanin (APC) (clone M-T466; Miltenyi Biotec), and CD8-Pacific Blue (clone RPA-T8; BD Biosciences) for 30 min in the dark at room temperature. The cells were fixed with 2% paraformaldehyde (PFA) for at least 20 min and then permeabilized with 0.1% saponin and stained with IFN-γ (clone 4S.B3; BD Biosciences) and TNF-α–peridinin chlorophyll protein (PerCP)/Cy5.5 (clone MAb11; BD Biosciences) for 30 min in the dark at room temperature. Flow cytometry was then performed on an LSR II instrument (BD Biosciences) using 2% PFA-fixed cells, and data were analyzed using FlowJo version 9.9.6 (Treestar, Ashland, OR).

Deep sequencing of SIV.

Replicating virus populations were subjected to genome-wide deep sequencing, as previously described (27). Briefly, viral RNA was isolated from plasma with a MinElute virus spin kit (Qiagen), and amplification of cDNA was performed using a Superscript III one-step reverse transcription (RT)-PCR system with high-fidelity Platinum Taq (Invitrogen). This resulted in four overlapping amplicons spanning the entire SIV coding sequence, which were then purified with the MinElute gel extraction kit (Qiagen) and quantified using a Quant-IT double-stranded DNA (dsDNA) HS assay kit (Invitrogen). Pooled amplicons (1 ng) were used to generate uniquely tagged libraries using a Nextera XT kit (Illumina). In select cases, cDNA was generated using a SuperScript IV First Strand synthesis system (Invitrogen) and PCR amplified with Q5 high-fidelity DNA polymerase (NEB) in duplicate to generate 37 overlapping amplicons spanning the entire SIV coding sequence. The amplified products were quantified using the Quant-IT dsDNA HS assay kit (Invitrogen) and diluted to 3 ng/μl for library preparation with TruSeq Nano HT (Illumina). Tagged libraries were then quantified using the Quant-IT dsDNA HS assay kit, and the fragment size distribution was assessed using a high-sensitivity Agilent bioanalyzer chip. The libraries were then pooled and sequenced on an Illumina MiSeq.

All sequences were analyzed with Geneious (Biomatters, Ltd.). Paired reads were initially quality trimmed using the BBDuk (decontamination using kmers) plugin, a part of the BBTools package by Brian Bushnell (Joint Genome Institute, Walnut Creek, CA) and then mapped with high sensitivity to the SIVmac239 sequence; gaps up to 500 bp were allowed when mapping to the SIVmac239 reference sequence (GenBank accession number M33262) in order to identify any additional deletions or insertions. Variant nucleotides were called at a threshold of 1%. The variants detected in the analyzed virus populations were then compared to the mutant sites originally incorporated into SIVΔnef-8x.

Statistics.

Student's t test was used evaluate the significance of differences between the peak viral load and set point viral load (the geometric mean of viral loads from 15 to 30 weeks postinfection). All viral load measurements were log10 transformed. Kaplan-Meier survival analyses were used to model the time to control virus replication in M3/M3 MCMs infected with SIVΔnef-8x (n = 8) or SIVmac239Δnef (n = 6) and M4/M6 MCMs infected with SIVΔnef-8x (n = 3); a log rank (Mantel-Cox) test was then used to determine the significance of differences. All statistical analyses were performed using GraphPad (La Jolla, CA) Prism.

Accession number(s).

The genome-sequencing and assembly data determined in this study are available in GenBank under BioProject number PRJNA479845.

ACKNOWLEDGMENTS

Research reported in this publication was supported in part by the Office of The Director, National Institutes of Health, under award number P51OD011106 to the Wisconsin National Primate Research Center, University of Wisconsin—Madison. Research reported in this publication was supported in part by National Institutes of Health award number R01AI108415, as well as by the National Institute of General Medical Sciences of the National Institutes of Health under award number T32GM081061.

The content is solely our responsibility and does not necessarily represent the official views of the National Institutes of Health.

Special thanks are due to Jason Weinfurter, Matthew Reynolds, and Adam Ericsen for helpful discussions.

REFERENCES

  • 1.Streeck H, Nixon DF. 2010. T cell immunity in acute HIV-1 infection. J Infect Dis 202(Suppl 2):S302–S308. doi: 10.1086/655652. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Koup RA, Safrit JT, Cao Y, Andrews CA, McLeod G, Borkowsky W, Farthing C, Ho DD. 1994. Temporal association of cellular immune responses with the initial control of viremia in primary human immunodeficiency virus type 1 syndrome. J Virol 68:4650–4655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Borrow P, Lewicki H, Hahn BH, Shaw GM, Oldstone MB. 1994. Virus-specific CD8+ cytotoxic T-lymphocyte activity associated with control of viremia in primary human immunodeficiency virus type 1 infection. J Virol 68:6103–6110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Ogg GS, Jin X, Bonhoeffer S, Dunbar PR, Nowak MA, Monard S, Segal JP, Cao Y, Rowland-Jones SL, Cerundolo V, Hurley A, Markowitz M, Ho DD, Nixon DF, McMichael AJ. 1998. Quantitation of HIV-1-specific cytotoxic T lymphocytes and plasma load of viral RNA. Science 279:2103–2106. doi: 10.1126/science.279.5359.2103. [DOI] [PubMed] [Google Scholar]
  • 5.International HIV Controllers Study, Pereyra F, Jia X, McLaren PJ, Telenti A, de Bakker PI, Walker BD, Ripke S, Brumme CJ, Pulit SL, Carrington M, Kadie CM, Carlson JM, Heckerman D, Graham RR, Plenge RM, Deeks SG, Gianniny L, Crawford G, Sullivan J, Gonzalez E, Davies L, Camargo A, Moore JM, Beattie N, Gupta S, Crenshaw A, Burtt NP, Guiducci C, Gupta N, Gao X, Qi Y, Yuki Y, Piechocka-Trocha A, Cutrell E, Rosenberg R, Moss KL, Lemay P, O'Leary J, Schaefer T, Verma P, Toth I, Block B, Baker B, Rothchild A, Lian J, Proudfoot J, Alvino DM, Vine S, Addo MM, Allen TM, Altfeld M, Henn MR, Le Gall S, et al. 2010. The major genetic determinants of HIV-1 control affect HLA class I peptide presentation. Science 330:1551–1557. doi: 10.1126/science.1195271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Altfeld M, Kalife ET, Qi Y, Streeck H, Lichterfeld M, Johnston MN, Burgett N, Swartz ME, Yang A, Alter G, Yu XG, Meier A, Rockstroh JK, Allen TM, Jessen H, Rosenberg ES, Carrington M, Walker BD. 2006. HLA Alleles associated with delayed progression to AIDS contribute strongly to the initial CD8(+) T cell response against HIV-1. PLoS Med 3:e403. doi: 10.1371/journal.pmed.0030403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Pereyra F, Heckerman D, Carlson JM, Kadie C, Soghoian DZ, Karel D, Goldenthal A, Davis OB, DeZiel CE, Lin T, Peng J, Piechocka A, Carrington M, Walker BD. 2014. HIV control is mediated in part by CD8+ T-cell targeting of specific epitopes. J Virol 88:12937–12948. doi: 10.1128/JVI.01004-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Wang YE, Li B, Carlson JM, Streeck H, Gladden AD, Goodman R, Schneidewind A, Power KA, Toth I, Frahm N, Alter G, Brander C, Carrington M, Walker BD, Altfeld M, Heckerman D, Allen TM. 2009. Protective HLA class I alleles that restrict acute-phase CD8+ T-cell responses are associated with viral escape mutations located in highly conserved regions of human immunodeficiency virus type 1. J Virol 83:1845–1855. doi: 10.1128/JVI.01061-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Deeks SG, Walker BD. 2007. Human immunodeficiency virus controllers: mechanisms of durable virus control in the absence of antiretroviral therapy. Immunity 27:406–416. doi: 10.1016/j.immuni.2007.08.010. [DOI] [PubMed] [Google Scholar]
  • 10.Liu Y, McNevin J, Rolland M, Zhao H, Deng W, Maenza J, Stevens CE, Collier AC, McElrath MJ, Mullins JI. 2009. Conserved HIV-1 epitopes continuously elicit subdominant cytotoxic T-lymphocyte responses. J Infect Dis 200:1825–1833. doi: 10.1086/648401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Soghoian DZ, Jessen H, Flanders M, Sierra-Davidson K, Cutler S, Pertel T, Ranasinghe S, Lindqvist M, Davis I, Lane K, Rychert J, Rosenberg ES, Piechocka-Trocha A, Brass AL, Brenchley JM, Walker BD, Streeck H. 2012. HIV-specific cytolytic CD4 T cell responses during acute HIV infection predict disease outcome. Sci Transl Med 4:123ra25. doi: 10.1126/scitranslmed.3003165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Ranasinghe S, Flanders M, Cutler S, Soghoian DZ, Ghebremichael M, Davis I, Lindqvist M, Pereyra F, Walker BD, Heckerman D, Streeck H. 2012. HIV-specific CD4 T cell responses to different viral proteins have discordant associations with viral load and clinical outcome. J Virol 86:277–283. doi: 10.1128/JVI.05577-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Johnson S, Eller M, Teigler JE, Maloveste SM, Schultz BT, Soghoian DZ, Lu R, Oster AF, Chenine AL, Alter G, Dittmer U, Marovich M, Robb ML, Michael NL, Bolton D, Streeck H. 2015. Cooperativity of HIV-specific cytolytic CD4 T cells and CD8 T cells in control of HIV viremia. J Virol 89:7494–7505. doi: 10.1128/JVI.00438-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Yant LJ, Friedrich TC, Johnson RC, May GE, Maness NJ, Enz AM, Lifson JD, O'Connor DH, Carrington M, Watkins DI. 2006. The high-frequency major histocompatibility complex class I allele Mamu-B*17 is associated with control of simian immunodeficiency virus SIVmac239 replication. J Virol 80:5074–5077. doi: 10.1128/JVI.80.10.5074-5077.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Loffredo JT, Maxwell J, Qi Y, Glidden CE, Borchardt GJ, Soma T, Bean AT, Beal DR, Wilson NA, Rehrauer WM, Lifson JD, Carrington M, Watkins DI. 2007. Mamu-B*08-positive macaques control simian immunodeficiency virus replication. J Virol 81:8827–8832. doi: 10.1128/JVI.00895-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Budde ML, Greene JM, Chin EN, Ericsen AJ, Scarlotta M, Cain BT, Pham NH, Becker EA, Harris M, Weinfurter JT, O'Connor SL, Piatak M, Lifson JD, Gostick E, Price DA, Friedrich TC, O'Connor DH. 2012. Specific CD8+ T cell responses correlate with control of simian immunodeficiency virus replication in Mauritian cynomolgus macaques. J Virol 86:7596–7604. doi: 10.1128/JVI.00716-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Maness NJ, Yant LJ, Chung C, Loffredo JT, Friedrich TC, Piaskowski SM, Furlott J, May GE, Soma T, León EJ, Wilson NA, Piontkivska H, Hughes AL, Sidney J, Sette A, Watkins DI. 2008. Comprehensive immunological evaluation reveals surprisingly few differences between elite controller and progressor Mamu-B*17-positive simian immunodeficiency virus-infected rhesus macaques. J Virol 82:5245–5254. doi: 10.1128/JVI.00292-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Hu X, Valentin A, Dayton F, Kulkarni V, Alicea C, Rosati M, Chowdhury B, Gautam R, Broderick KE, Sardesai NY, Martin MA, Mullins JI, Pavlakis GN, Felber BK. 2016. DNA prime-boost vaccine regimen to increase breadth, magnitude, and cytotoxicity of the cellular immune responses to subdominant Gag epitopes of simian immunodeficiency virus and HIV. J Immunol 197:3999–4013. doi: 10.4049/jimmunol.1600697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Mothe B, Hu X, Llano A, Rosati M, Olvera A, Kulkarni V, Valentin A, Alicea C, Pilkington GR, Sardesai NY, Rocafort M, Crespo M, Carrillo J, Marco A, Mullins JI, Dorrell L, Hanke T, Clotet B, Pavlakis GN, Felber BK, Brander C. 2015. A human immune data-informed vaccine concept elicits strong and broad T-cell specificities associated with HIV-1 control in mice and macaques. J Transl Med 13:60. doi: 10.1186/s12967-015-0392-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Hu X, Valentin A, Rosati M, Manocheewa S, Alicea C, Chowdhury B, Bear J, Broderick KE, Sardesai NY, Gall SL, Mullins JI, Pavlakis GN, Felber BK. 2017. HIV Env conserved element DNA vaccine alters immunodominance in macaques. Hum Vaccin Immunother 13:2859–2871. doi: 10.1080/21645515.2017.1339852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Budde ML, Wiseman RW, Karl JA, Hanczaruk B, Simen BB, O'Connor DH. 2010. Characterization of Mauritian cynomolgus macaque major histocompatibility complex class I haplotypes by high-resolution pyrosequencing. Immunogenetics 62:773–780. doi: 10.1007/s00251-010-0481-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.O'Connor SL, Becker EA, Weinfurter JT, Chin EN, Budde ML, Gostick E, Correll M, Gleicher M, Hughes AL, Price DA, Friedrich TC, O'Connor DH. 2012. Conditional CD8+ T cell escape during acute simian immunodeficiency virus infection. J Virol 86:605–609. doi: 10.1128/JVI.05511-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.O'Connor SL, Lhost JJ, Becker EA, Detmer AM, Johnson RC, Macnair CE, Wiseman RW, Karl JA, Greene JM, Burwitz BJ, Bimber BN, Lank SM, Tuscher JJ, Mee ET, Rose NJ, Desrosiers RC, Hughes AL, Friedrich TC, Carrington M, O'Connor DH. 2010. MHC heterozygote advantage in simian immunodeficiency virus-infected Mauritian cynomolgus macaques. Sci Transl Med 2:22ra18. doi: 10.1126/scitranslmed.3000524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Ericsen AJ, Starrett GJ, Greene JM, Lauck M, Raveendran M, Deiros DR, Mohns MS, Vince N, Cain BT, Pham NH, Weinfurter JT, Bailey AL, Budde ML, Wiseman RW, Gibbs R, Muzny D, Friedrich TC, Rogers J, O'Connor DH. 2014. Whole genome sequencing of SIV-infected macaques identifies candidate loci that may contribute to host control of virus replication. Genome Biol 15:478. doi: 10.1186/s13059-014-0478-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Harris M, Burns CM, Becker EA, Braasch AT, Gostick E, Johnson RC, Broman KW, Price DA, Friedrich TC, O'Connor SL. 2013. Acute-phase CD8 T cell responses that select for escape variants are needed to control live attenuated simian immunodeficiency virus. J Virol 87:9353–9364. doi: 10.1128/JVI.00909-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Sutton MS, Burns CM, Weiler AM, Balgeman AJ, Braasch A, Lehrer-Brey G, Friedrich TC, O'Connor SL. 2016. Vaccination with live attenuated simian immunodeficiency virus (SIV) protects from mucosal, but not necessarily intravenous, challenge with a minimally heterologous SIV. J Virol 90:5541–5548. doi: 10.1128/JVI.00192-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Gellerup DD, Balgeman AJ, Nelson CW, Ericsen AJ, Scarlotta M, Hughes AL, O'Connor SL. 2016. Conditional immune escape during chronic simian immunodeficiency virus infection. J Virol 90:545–552. doi: 10.1128/JVI.02587-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Budde ML, Lhost JJ, Burwitz BJ, Becker EA, Burns CM, O'Connor SL, Karl JA, Wiseman RW, Bimber BN, Zhang GL, Hildebrand W, Brusic V, O'Connor DH. 2011. Transcriptionally abundant major histocompatibility complex class I alleles are fundamental to nonhuman primate simian immunodeficiency virus-specific CD8+ T cell responses. J Virol 85:3250–3261. doi: 10.1128/JVI.02355-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Sutton MS, Ellis-Connell A, Moriarty RV, Balgeman AJ, Gellerup D, Barry G, Weiler AM, Friedrich TC, O'Connor SL. 2018. Acute-phase CD4+ T cell responses targeting invariant viral regions are associated with control of live attenuated simian immunodeficiency virus. bioRxiv doi: 10.1101/321000. [DOI] [PMC free article] [PubMed]
  • 30.Valentine LE, Loffredo JT, Bean AT, León EJ, MacNair CE, Beal DR, Piaskowski SM, Klimentidis YC, Lank SM, Wiseman RW, Weinfurter JT, May GE, Rakasz EG, Wilson NA, Friedrich TC, O'Connor DH, Allison DB, Watkins DI. 2009. Infection with “escaped” virus variants impairs control of simian immunodeficiency virus SIVmac239 replication in Mamu-B*08-positive macaques. J Virol 83:11514–11527. doi: 10.1128/JVI.01298-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Burwitz BJ, Giraldo-Vela JP, Reed J, Newman LP, Bean AT, Nimityongskul FA, Castrovinci PA, Maness NJ, Leon EJ, Rudersdorf R, Sacha JB. 2012. CD8+ and CD4+ cytotoxic T cell escape mutations precede breakthrough SIVmac239 viremia in an elite controller. Retrovirology 9:91. doi: 10.1186/1742-4690-9-91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Streeck H, Lichterfeld M, Alter G, Meier A, Teigen N, Yassine-Diab B, Sidhu HK, Little S, Kelleher A, Routy JP, Rosenberg ES, Sekaly RP, Walker BD, Altfeld M. 2007. Recognition of a defined region within p24 gag by CD8+ T cells during primary human immunodeficiency virus type 1 infection in individuals expressing protective HLA class I alleles. J Virol 81:7725–7731. doi: 10.1128/JVI.00708-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Létourneau S, Im EJ, Mashishi T, Brereton C, Bridgeman A, Yang H, Dorrell L, Dong T, Korber B, McMichael AJ, Hanke T. 2007. Design and pre-clinical evaluation of a universal HIV-1 vaccine. PLoS One 2:e984. doi: 10.1371/journal.pone.0000984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Rolland M, Nickle DC, Mullins JI. 2007. HIV-1 group M conserved elements vaccine. PLoS Pathog 3:e157. doi: 10.1371/journal.ppat.0030157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Kunwar P, Hawkins N, Dinges WL, Liu Y, Gabriel EE, Swan DA, Stevens CE, Maenza J, Collier AC, Mullins JI, Hertz T, Yu X, Horton H. 2013. Superior control of HIV-1 replication by CD8+ T cells targeting conserved epitopes: implications for HIV vaccine design. PLoS One 8:e64405. doi: 10.1371/journal.pone.0064405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Tanaka M, Robinson BA, Chutiraka K, Geary CD, Reed JC, Lingappa JR. 2016. Mutations of conserved residues in the major homology region arrest assembling HIV-1 Gag as a membrane-targeted intermediate containing genomic RNA and cellular proteins. J Virol 90:1944–1963. doi: 10.1128/JVI.02698-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Accola MA, Strack B, Göttlinger HG. 2000. Efficient particle production by minimal Gag constructs which retain the carboxy-terminal domain of human immunodeficiency virus type 1 capsid-p2 and a late assembly domain. J Virol 74:5395–5402. doi: 10.1128/JVI.74.12.5395-5402.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Kirchhoff F, Mori K, Desrosiers RC. 1994. The “V3” domain is a determinant of simian immunodeficiency virus cell tropism. J Virol 68:3682–3692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Hung CS, Vander Heyden N, Ratner L. 1999. Analysis of the critical domain in the V3 loop of human immunodeficiency virus type 1 gp120 involved in CCR5 utilization. J Virol 73:8216–8226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Chen B, Vogan EM, Gong H, Skehel JJ, Wiley DC, Harrison SC. 2005. Structure of an unliganded simian immunodeficiency virus gp120 core. Nature 433:834–841. doi: 10.1038/nature03327. [DOI] [PubMed] [Google Scholar]
  • 41.von Gegerfelt A, Valentin A, Alicea C, Van Rompay KK, Marthas ML, Montefiori DC, Pavlakis GN, Felber BK. 2010. Emergence of simian immunodeficiency virus-specific cytotoxic CD4+ T cells and increased humoral responses correlate with control of rebounding viremia in CD8-depleted macaques infected with Rev-independent live-attenuated simian immunodeficiency virus. J Immunol 185:3348–3358. doi: 10.4049/jimmunol.1000572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Leng J, Ho HP, Buzon MJ, Pereyra F, Walker BD, Yu XG, Chang EJ, Lichterfeld M. 2014. A cell-intrinsic inhibitor of HIV-1 reverse transcription in CD4(+) T cells from elite controllers. Cell Host Microbe 15:717–728. doi: 10.1016/j.chom.2014.05.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Wilson NA, Keele BF, Reed JS, Piaskowski SM, MacNair CE, Bett AJ, Liang X, Wang F, Thoryk E, Heidecker GJ, Citron MP, Huang L, Lin J, Vitelli S, Ahn CD, Kaizu M, Maness NJ, Reynolds MR, Friedrich TC, Loffredo JT, Rakasz EG, Erickson S, Allison DB, Piatak M, Lifson JD, Shiver JW, Casimiro DR, Shaw GM, Hahn BH, Watkins DI. 2009. Vaccine-induced cellular responses control simian immunodeficiency virus replication after heterologous challenge. J Virol 83:6508–6521. doi: 10.1128/JVI.00272-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Friedrich TC, Valentine LE, Yant LJ, Rakasz EG, Piaskowski SM, Furlott JR, Weisgrau KL, Burwitz B, May GE, León EJ, Soma T, Napoe G, Capuano SV, Wilson NA, Watkins DI. 2007. Subdominant CD8+ T-cell responses are involved in durable control of AIDS virus replication. J Virol 81:3465–3476. doi: 10.1128/JVI.02392-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Gauduin MC, Yu Y, Barabasz A, Carville A, Piatak M, Lifson JD, Desrosiers RC, Johnson RP. 2006. Induction of virus-specific effector-memory CD4+ T cell response by attenuated SIV infection. J Exp Med 203:2661–2672. doi: 10.1084/jem.20060134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Sacha JB, Reynolds MR, Buechler MB, Chung C, Jonas AK, Wallace LT, Weiler AM, Lee W, Piaskowski SM, Soma T, Friedrich TC, Wilson NA, Watkins DI. 2008. Differential antigen presentation kinetics of CD8+ T-cell epitopes derived from the same viral protein. J Virol 82:9293–9298. doi: 10.1128/JVI.00749-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Brown DM, Lampe AT, Workman AM. 2016. The differentiation and protective function of cytolytic CD4 T cells in influenza infection. Front Immunol 7:93. doi: 10.3389/fimmu.2016.00093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Douek DC, Brenchley JM, Betts MR, Ambrozak DR, Hill BJ, Okamoto Y, Casazza JP, Kuruppu J, Kunstman K, Wolinsky S, Grossman Z, Dybul M, Oxenius A, Price DA, Connors M, Koup RA. 2002. HIV preferentially infects HIV-specific CD4+ T cells. Nature 417:95–98. doi: 10.1038/417095a. [DOI] [PubMed] [Google Scholar]
  • 49.Takeuchi A, Saito T. 2017. CD4 CTL, a cytotoxic subset of CD4. Front Immunol 8:194. doi: 10.3389/fimmu.2017.00194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Veazey RS, DeMario M, Chalifoux LV, Shvetz DE, Pauley DR, Knight HL, Rosenzweig M, Johnson RP, Desrosiers RC, Lackner AA. 1998. Gastrointestinal tract a major site of CD4+ T cell depletion and viral replication in SIV infection. Science 280:427–431. doi: 10.1126/science.280.5362.427. [DOI] [PubMed] [Google Scholar]
  • 51.Reeves RK, Gillis J, Wong FE, Johnson RP. 2009. Vaccination with SIVΔnef activates CD4+ T cells in the absence of CD4+ T cell loss. J Med Primatol 38(Suppl 1):8–16. doi: 10.1111/j.1600-0684.2009.00370.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Picker LJ, Hagen SI, Lum R, Reed-Inderbitzin EF, Daly LM, Sylwester AW, Walker JM, Siess DC, Piatak M Jr, Wang C, Allison DB, Maino VC, Lifson JD, Kodama T, Axthelm MK. 2004. Insufficient production and tissue delivery of CD4+ memory T cells in rapidly progressive simian immunodeficiency virus infection. J Exp Med 200:1299–1314. doi: 10.1084/jem.20041049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Chakrabarti LA, Metzner KJ, Ivanovic T, Cheng H, Louis-Virelizier J, Connor RI, Cheng-Mayer C. 2003. A truncated form of Nef selected during pathogenic reversion of simian immunodeficiency virus SIVmac239Deltanef increases viral replication. J Virol 77:1245–1256. doi: 10.1128/JVI.77.2.1245-1256.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Wiseman RW, Wojcechowskyj JA, Greene JM, Blasky AJ, Gopon T, Soma T, Friedrich TC, O'Connor SL, O'Connor DH. 2007. Simian immunodeficiency virus SIVmac239 infection of major histocompatibility complex-identical cynomolgus macaques from Mauritius. J Virol 81:349–361. doi: 10.1128/JVI.01841-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Karl JA, Wiseman RW, Campbell KJ, Blasky AJ, Hughes AL, Ferguson B, Read DS, O'Connor DH. 2008. Identification of MHC class I sequences in Chinese-origin rhesus macaques. Immunogenetics 60:37–46. doi: 10.1007/s00251-007-0267-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Wiseman RW, Karl JA, Bohn PS, Nimityongskul FA, Starrett GJ, O'Connor DH. 2013. Haplessly hoping: macaque major histocompatibility complex made easy. ILAR J 54:196–210. doi: 10.1093/ilar/ilt036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Karl JA, Heimbruch KE, Vriezen CE, Mironczuk CJ, Dudley DM, Wiseman RW, O'Connor DH. 2014. Survey of major histocompatibility complex class II diversity in pig-tailed macaques. Immunogenetics 66:613–623. doi: 10.1007/s00251-014-0797-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Karl JA, Graham ME, Wiseman RW, Heimbruch KE, Gieger SM, Doxiadis GG, Bontrop RE, O'Connor DH. 2017. Major histocompatibility complex haplotyping and long-amplicon allele discovery in cynomolgus macaques from Chinese breeding facilities. Immunogenetics 69:211–229. doi: 10.1007/s00251-017-0969-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Reynolds MR, Weiler AM, Piaskowski SM, Piatak M, Robertson HT, Allison DB, Bett AJ, Casimiro DR, Shiver JW, Wilson NA, Lifson JD, Koff WC, Watkins DI. 2012. A trivalent recombinant Ad5 gag/pol/nef vaccine fails to protect rhesus macaques from infection or control virus replication after a limiting-dose heterologous SIV challenge. Vaccine 30:4465–4475. doi: 10.1016/j.vaccine.2012.04.082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Burwitz BJ, Pendley CJ, Greene JM, Detmer AM, Lhost JJ, Karl JA, Piaskowski SM, Rudersdorf RA, Wallace LT, Bimber BB, Loffredo JT, Cox DG, Bardet W, Hildebrand W, Wiseman RW, O'Connor SL, O'Connor DH. 2009. Mauritian cynomolgus macaques share two exceptionally common major histocompatibility complex class I alleles that restrict simian immunodeficiency virus-specific CD8+ T cells. J Virol 83:6011–6019. doi: 10.1128/JVI.00199-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Ellis A, Balgeman A, Rodgers M, Updike C, Tomko J, Maiello P, Scanga CA, O'Connor SL. 2017. Characterization of T cells specific for CFP-10 and ESAT-6 in Mycobacterium tuberculosis-infected Mauritian cynomolgus macaques. Infect Immun 85:e01009-16. doi: 10.1128/IAI.01009-16. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Journal of Virology are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES