DNA Vaccine Molecular Adjuvants SP-D-BAFF and SP-D-APRIL Enhance Anti-gp120 Immune Response and Increase HIV-1 Neutralizing Antibody Titers

DNA Vaccine Molecular Adjuvants SP-D-BAFF and SP-D-APRIL Enhance Anti-gp120 Immune Response and Increase HIV-1 Neutralizing Antibody Titers

Products Related to p24, gp41, gp120, gp160

 
 

Product# 1103 HIV-1 p24 Monoclonal Antibody

 

Product#1091 Recombinant HIV-1 MN gp41 Envelope Protein (E.coli)

 

Product#1001 Recombinant HIV-1 IIIB Envelope Glycoprotein gp120 (Baculovirus)

 
 

Product#1024 Recombinant HIV-1 MN Envelope Glycoprotein gp160 (Baculovirus)

INTRODUCTION

It is generally believed that broadly neutralizing antibodies (bNAbs) are essential for the prevention of HIV-1 infection. Although several bNAbs have been isolated from HIV-infected individuals (,), they are not commonly generated in most humans following HIV-1 infection. Not surprisingly, a vaccine design that can induce high-titer bNAbs and immunological memory remains a major challenge. HIV-1 has unique structural features, including high variability of protein sequences, inaccessibility of the conserved structures in the Env protein to bNAbs, and extensive glycosylation masking Env antigens (Ags). Further compounding the issue is the potential loss of Env-specific B cell clones that are autoreactive and are therefore deleted during the process of immunological self-tolerance (,). Although most B cells are variably autoreactive in both humans and mice, the majority of B cells join the immunocompetent mature B cell repertoire (). This suggests that only B cell clones with autoreactivity beyond a certain threshold are deleted by negative selection, while the rest continue their development into mature B cells. Thus, the mature B cell repertoire likely contains weakly HIV-1 reactive B cell clones, as well as some strongly reactive B cell clones that escape tolerance checkpoints or assume an anergic state. Recent evidence suggests that the mature B cell repertoire in both mice and humans contains clones that bind HIV-1 Env and are often polyreactive (, ). Thus, in principle, it should be possible to design vaccines targeting HIV envelope-specific B cells that can serve as precursors for anti-HIV-1 bNAbs (, ).

A successful HIV vaccine design not only has to overcome some unusual evasive properties of HIV-1 but also has to mimic native Env trimers, which are highly unstable (). In addition, a vaccine should promote sufficiently high titers of bNAbs that are not diluted by immune responses to nonprotective epitopes. Most antibody responses depend on peptide-antigen presentation by dendritic cells (DCs) to CD4 T cells, upregulating the CD40 ligand (CD40L), which interacts with CD40 on B cells, providing cognate help to B cells. B cells stimulated in this manner migrate to the germinal centers (GCs), where they undergo isotype class switch recombination (CSR) and affinity maturation and differentiate into antibody-secreting plasma cells (ASCs). However, vaccines that mobilize this pathway exploit limited neutralizing epitopes on HIV-1 Env. HIV antigens can also activate B cells directly. Direct engagement of B cells with antigen may increase the number and complexity of the antibody-inducing epitopes available, including glycan-linked peptides. In this mode of immune activation, B cells can present antigen and activate the conventional CD4 T helper pathway as well as T cell-independent mechanisms of antibody production. We and others have shown that T cell-independent mechanisms depend heavily on B cell-activating factor (BAFF) receptor and ligand (,).

The BAFF receptors, BAFF-R (BAFF receptor), TACI (transmembrane activator and calcium-modulating cyclophilin ligand interactor), and BCMA (B cell maturation antigen), play crucial roles in many key aspects of B cell biology, including selection and survival during peripheral B cell maturation and the survival and differentiation of antigen-activated B cells and ASCs (,). Both BAFF and APRIL (a proliferation-inducing ligand) can stimulate B cells via BCMA and TACI. However, BAFF activation of TACI requires aggregates of BAFF (,). Additionally, only BAFF binds BAFF-R. Peripheral, transitional, and mature B cells express BAFF-R on their surfaces, whereas TACI is detectable on all peripheral B cells. BCMA is found primarily in plasma cells. Significantly, the expression of both BAFF-R and TACI on the cell surface is rapidly induced upon the engagement of B cell receptor (BCR) with antigen. Heightened expression of TACI and BAFF-R sensitizes antigen-specific B cells to the growth and differentiation signals of BAFF and APRIL () during an immune response, including the increased life span of GCs, class switching to IgG, and affinity maturation (, ). In fact, BAFF−/− transgenic mice display spontaneous formation of GCs, promoting a Th1-skewed response with IgG2a and IgG3 production (). Likewise, high serum BAFF concentrations support the survival and class switching of autoreactive B cells (). In contrast, gene deletion or neutralization of BAFF causes early dissolution of GCs, whereas transgenic overexpression of BAFF results in spontaneous formation of GCs. Further, an antibacterial vaccine formulation expressing BAFF from a viral vector enhanced and extended a protective high-titer antibody response against Pseudomonas aeruginosa (). Like BAFF, APRIL has been shown to induce T cell-independent isotype class switching, affinity maturation, and antibody production (, , , ). It was thought that BCMA was primarily responsible for the survival of ASCs. However, recent evidence suggests that TACI and BAFF-R are also required (, ). Thus, BAFF and APRIL may promote various aspects of class-switched and high-affinity antibody production.

As with other tumor necrosis factor (TNF) family members, stimulation through BAFF-R, TACI, and BCMA depends on the trimerization of BAFF and APRIL or on interactions between B cells and BAFF/APRIL-expressing cells such as B and T cells, monocytes, neutrophils, and macrophages/myeloid cells (). A prior study using BAFF and APRIL trimers as adjuvants in rabbits showed improved antibody response; however, only tier 1 viruses could be neutralized (). These observations led us to hypothesize that soluble multitrimers of BAFF and APRIL, as opposed to single trimers, would induce optimal immune activation of B cells and enhance the overall immune response. We have shown previously that TNF superfamily ligands, including BAFF, APRIL, and CD40L, can be formed into soluble multitrimers by using the lung protein SP-D (surfactant protein D) as a scaffold (,). In these studies, DNA vaccination with an SP-D-BAFF adjuvant enhanced cellular immune responses against an HIV-1 Gag antigen. However, we failed to observe a significant increase in antigen-specific antibody titers (), possibly due to the requirement for additional costimulatory signals in the vaccine formulation.

In the present study, we evaluated multitrimers of BAFF and APRIL as molecular adjuvants together with interleukin-12 p70 (IL-12p70) in an HIV-1 Env gp140 DNA vaccine model. Using this vaccine design, we tested both DNA vaccination and a DNA prime–protein boost vaccination strategy using a recombinant gp120 protein boost. B cell and antibody responses were evaluated following vaccination. Our data show that soluble multitrimeric forms of BAFF or APRIL, in combination with IL-12p70 and membrane-bound Env protein gp140, enhanced the gp120-specific antibody response and titers of tier 1 and autologous vaccine strain tier 2 neutralizing antibodies.

MATERIALS AND METHODS

 

Construction and preparation of DNA plasmids.

The HIV-1 Env DNA vaccine plasmid (pgp140; clade C, strain 96ZM651), was obtained from the NIH AIDS Reagent Repository program (catalog no. 8660). The secreted HIV-1 Gag DNA plasmid (pGag; also called pScGag) has been described previously () and was provided by George N. Pavlakis. Plasmids coding for the 4-trimer soluble forms of murine SP-D-BAFF and SP-D-APRIL were generated in vectors pVAX1 and pcDNA3.1, respectively, as described previously (). Constructs were cloned such that mouse SP-D from the N terminus to amino acid sequence ALFPDG was fused directly to the mouse TNF superfamily ligand (TNFSFL) extracellular domain, starting from the N-terminal amino acid sequences QLAALQ (BAFF) and MPASSP (APRIL). Plasmid pIL-12p70, encoding mouse single-chain IL-12, was purchased from InvivoGen Inc. All plasmids were propagated in Escherichia coli strain TOP10. Endotoxin-free DNA plasmid preparations were prepared using an EndoFree Giga plasmid kit (Qiagen). Plasmids were then further purified to remove residual endotoxins with additional Triton X-114 extractions as described previously (). Plasmid endotoxin levels were <0.2 endotoxin unit (EU)/ml for all constructs as confirmed by a Limulus amebocyte lysate (LAL) endotoxin assay (Lonza Inc.).

 

Preparation of Env protein.

For the preparation of monomeric Env protein, plasmid pTpa-gp120-His was constructed by fusing HIV-1 Env gp120 (clade C, strain 96ZM651) to the first 21 amino acids of human tissue plasminogen activator (t-PA) at the N terminus and to a His tag at the C terminus. For the preparation of Env protein, 293T cells were transiently transfected with the pTpa-gp120-His plasmid by using the GenJet Plus transfection reagent (SignaGen Laboratories) according to the manufacturer's instructions. Supernatants were harvested 48 h after transfection and were purified on a Ni-nitrilotriacetic acid (NTA) affinity column using standard procedures. Briefly, after the separation of cell debris by high-speed centrifugation, the cell culture supernatant was loaded onto a Ni-NTA column, and the column was washed extensively with 20 mM imidazole to remove contaminating proteins. The Env protein was eluted with 200 mM imidazole and was dialyzed to remove excess imidazole. The purified Env protein was then concentrated using an Amicon Ultra-15 centrifugal filter unit with a 100,000-molecular-weight cutoff filter (Millipore Inc.) according to the manufacturer's instructions. The protein concentration was determined by a fluorescence-based Quant-iT assay (Invitrogen) and by spectrophotometry using an extinction coefficient of 37,930/M/cm, calculated using ProtParam online software at the ExPASy server (Invitrogen) ().

 

Mice and immunization schedule.

Female BALB/c mice (7 to 8 weeks old) were used in all vaccination experiments. Animals were housed at the University of Miami under the guidelines of the National Institutes of Health (NIH; Bethesda, MD). All animal experiments were performed in accordance with national and institutional guidance for animal care and were approved by the IACUC of the University of Miami. The pGag and pgp140 plasmids were diluted in phosphate-buffered saline (PBS) and were combined with either pcDNA3.1 or one of the SP-D-TNFSFL adjuvant plasmids as noted, with or without pIL-12. Plasmid mixtures were injected intramuscularly (i.m.) into the quadriceps muscles of both hind limbs. DNA vaccinations were given three times at 2-week intervals with 80 μg of the 96ZM651 membrane-bound gp140 plasmid mixed with 20 μg of the pcDNA3.1 or SP-D-TNFSFL adjuvant plasmid, with or without 20 μg of pIL-12. Doses were administered in a total volume of 100 μl PBS (50 μl per limb). For DNA vaccine studies, 2 weeks after the final DNA immunization, mice were anesthetized with ketamine-xylazine, and blood was drawn by cardiac puncture to obtain serum. For heterologous DNA prime–protein boost vaccinations, mice were injected intramuscularly with DNA three times at 2-week intervals, followed 3 weeks later by a single Env protein boost, given by intramuscular injection at the same site. Thirty micrograms of gp120 Env protein was formulated with an equal volume of Freund's incomplete adjuvant in a total volume of 100 μl. The vaccine was then injected intramuscularly at a 50-μl volume into each hind-leg quadriceps muscle. Two weeks following the Env protein boost, mice were anesthetized with ketamine-xylazine, and blood was drawn by cardiac puncture to obtain serum. Following euthanization, spleens were removed for B cell analysis. Half the spleen was quick-frozen in OCT compound for histology. Single-cell splenocyte preparations were obtained for the other half of each spleen by passage through a 40-μm nylon cell strainer (BD Falcon). Erythrocytes were depleted with lysis buffer (Sigma), and splenocytes were washed thoroughly with complete R10 medium (RPMI 1640 medium supplemented with 10% fetal bovine serum [FBS], 50 μM 2-mercaptomethanol, 100 U/ml of penicillin, 100 μg/ml streptomycin, and 10 mM HEPES).

 

ELISA for anti-Env IgG responses.

Antibody production was measured by an enzyme-linked immunosorbent assay (ELISA). Ninety-six-well ELISA plates were coated with HIV-1 IIIB p55 Gag protein (10 μg/ml; provided by the NIH AIDS Reagent Program) or HIV-1 Env 96ZM651 gp120 protein (10 μg/ml; provided by the NIH AIDS Reagent Program or prepared in the lab) overnight at 4°C. Mouse sera at varying dilutions were added to antigen-coated wells and were incubated at room temperature for 2 h with shaking. After plates were washed, antigen-specific IgG antibodies were detected using horseradish peroxidase-conjugated goat anti-mouse IgA, IgG, IgG1, IgG2b, or IgG2a (Jackson ImmunoResearch Inc.). The signal was developed using the SureBlue TMB microwell peroxidase substrate (Kirkegaard & Perry Laboratories, Inc.). Plates were analyzed using a 96-well plate absorbance reader at 650 nm. Endpoint titers were calculated as the highest dilutions with more than twice the background absorbance of control wells. For the quantification of total IgG, a standard curve using known amounts of purified mouse IgG was used.

 

HIV-1 neutralization assay.

Virus neutralization was measured using a luciferase-based assay on TZM.bl cells as described previously (). The TZM.bl cell assay was performed with HIV-1 tier 1 pseudovirus MW965.26 and tier 2 pseudovirus 96ZM651 (vaccine strain). The assay measures the reduction in luciferase reporter gene expression by TZM.bl cells following a 48-h incubation period with a single round of virus infection. The 50% inhibitory dose (ID50) was calculated as the serum dilution that caused a 50% reduction in relative luminescence units (RLU) from the level in the virus control wells after the subtraction of the RLU in cell control wells. Calculations were performed using a validated macro employing a point-based algorithm with linear interpolation between the two replicates on either side of 50% RLU reduction. The level of nonspecific neutralization for each mouse was determined by measuring the neutralization of murine leukemia virus (MLV) with SVA elements (SVA-MLV). Neutralization titers for each mouse were scored as positive only if they were >3-fold higher than SVA-MLV titers.

 

Avidity ELISA.

The avidity of antibodies against gp120 protein was determined by a particle disruption ELISA using the chaotropic agent sodium thiocyanate (NaSCN), as described previously (). The binding of gp120 protein and incubation with mouse serum were performed as described above. Plates were washed three times with 0.05% PBS–Tween 20, and the chaotropic agent NaSCN, diluted in PBS, was added to replicate wells (0, 1, 2, 3, 4, or 5 M). Plates were first incubated at room temperature for 30 min and then washed six times with PBS–Tween 20. Subsequent ELISA steps were performed as detailed above. The avidity index for each mouse sample was recorded as the mean molarity of NaSCN that reduced the optical density at 650 nm (OD650) by 50% from that for wells treated with PBS.

 

Flow cytometry.

Spleens from immunized mice were cut transversely; the anterior half was used for flow cytometry and an enzyme-linked immunosorbent spot (ELISPOT) assay, and the posterior half was used for tissue immunofluorescence (see below). Flow cytometry (with an LSR II system; BD Biosciences) was performed to determine the percentages of live splenic and GC B cells, the CD4/CD8 T cell ratios, and the level of purity of the B cell enrichment. Single-cell suspensions of splenocytes were stained with fluorescein isothiocyanate (FITC)-conjugated anti-B220 (RA3-6B2; eBioscience), phycoerythrin (PE)-Cy7-conjugated anti-CD19 (1D3; BD Biosciences), allophycocyanin (APC)-conjugated anti-CD4 (RM4-5; BD Biosciences), Pacific Blue (PB)-conjugated anti-CD8 (53-6.7; BD Biosciences), APC-conjugated anti-CD138 (anti-syndecan-1; BD Biosciences), and biotin-conjugated anti-GL7 (eBiosciences) and V500-conjugated streptavidin (BD Biosciences). Live/dead analysis was performed using LIVE/DEAD near-IR staining (Invitrogen). The values thus obtained were then used to calculate the numbers of ASCs, B cells, and GC B cells per half-spleen.

 

ELISPOT assay for anti-gp120 B cells.

A single-cell spleen suspension was made as described above. CD43 (total conventional) B cells were enriched using the BD IMag B cell enrichment kit (BD Biosciences). A dilution series of B cells (1 × 106 to 0.5 × 105) was plated in duplicate wells of a Millipore ELISPOT plate with a polyvinylidene difluoride (PVDF) membrane that had been coated 24 h previously with 5 μg/ml gp120 protein. Cells were incubated overnight at 37°C, and then total IgG-producing gp120-specific ASCs were revealed using the mouse IgG ELISpot Plus kit (Mabtech) according to the manufacturer's instructions. The plates were dried and were read on an ImmunoSpot ELISPOT plate reader (Cellular Technology Ltd.). Flow cytometry (on an LSR II system; BD Biosciences) was used to determine the percentage of splenic B cells and the level of purity of the B cell enrichment (with FITC-conjugated anti-B220 [RA3-6B2; eBioscience] and PE-Cy7-conjugated anti-CD19 [1D3; BD Biosciences]). These values were then used to calculate the number of ASCs per spleen.

 

Tissue histology.

The remaining half of the spleen (see above) was embedded in OCT compound and frozen in liquid nitrogen, and 5-μm sagittal sections were cut. For staining, slides were incubated for 10 min in −20°C acetone, washed in Tris-buffered saline (TBS), and blocked in 3% bovine serum albumin (BSA) in TBS with 0.1% Tween 20 for 1 h at room temperature. Slides were then incubated with goat anti-mouse CD3 (M20; Santa Cruz Biotechnology), biotin-conjugated anti-CD19 (1D3; BD Biosciences), and tetramethyl rhodamine isocyanate (TRITC)-conjugated peanut agglutinin (PNA) (Sigma) in blocking buffer at 4°C overnight. Slides were first washed with TBS and then incubated for 1 h with an Alexa Fluor 647-conjugated anti-goat antibody (Life Technologies) and Alexa Fluor 488-conjugated streptavidin. Slides were washed with TBS and were then mounted using ProLong Gold Antifade reagent with 4′,6-diamidino-2-phenylindole (DAPI). Entire slides were digitally scanned using the VS120 virtual slide scanning system (Olympus America, Inc.) at ×40 magnification. The B cell areas in germinal centers were measured using ImageJ.

 

Statistical analysis.

GraphPad Prism software, version 6.0, was used to calculate significance by one-way analysis of variance (ANOVA) with multiple comparisons using Tukey's post hoc analysis. Correlation analysis was performed by linear regression using GraphPad Prism software, version 6.0.

RESULTS

 

Combining DNA vaccine adjuvant pSP-D-BAFF with pIL-12 increased antibody titers.

Previous studies with DNA vaccines containing the pSP-D-BAFF adjuvant plasmid have shown only low levels of antibody titers relative to those with an antigen-only control vaccine (). To explore methods of enhancing antibody titers from DNA vaccines, we performed a DNA vaccination experiment on mice with HIV-1 Gag DNA (as a model antigen) combined with plasmid pSP-D-BAFF or pIL-12, a single-chain IL-12p70 construct expressing both IL-12p35 and IL-12p40 joined by a flexible linker (). As shown in Fig. 1B, pSP-D-BAFF failed to increase anti-Gag antibody titers, while the pIL-12 adjuvant significantly increased antibody titers as measured by ELISA. Next, we moved to an HIV-1 gp140 DNA vaccine model to test the ability of combinations of IL-12p70 and SP-D-BAFF adjuvants to enhance antibody titers. Plasmid pgp140 (encoding membrane-bound gp140 of HIV-1 clade C strain 96ZM651) was combined with either the empty vector pcDNA3.1, pIL-12 plus pcDNA3.1, or pIL-12 plus pSP-D-BAFF. Again, addition of the IL-12p70 adjuvant significantly increased mean antibody titers over those with pgp140 alone (Fig. 1C). Mean antibody titers were also increased with the combination of pgp140, pIL-12, and pSP-D-BAFF over those with gp140 antigen alone. Although we observed a trend toward higher antibody titers with the combination of the pIL-12 and pSP-D-BAFF adjuvants than with pIL-12 alone, the difference was not statistically significant, a result consistent with those of previous DNA vaccination studies (, ).

IL-12p70 is necessary for the induction of antibodies by DNA vaccines encoding SP-D-BAFF. Mice were vaccinated i.m. with 80 μg of an antigen plasmid mixed with 20 μg of either an empty vector, the IL-12p70 single-chain dimer, SP-D-BAFF DNA, or a combination of the latter two (120 μg total DNA per mouse). (A) Animals were vaccinated every 2 weeks for a total of 3 vaccinations. Serum was obtained 2 weeks following the final vaccination and was assayed for binding to the antigen by ELISA. (B) The adjuvant plasmid pIL-12 induced higher antibody titers than did Gag alone or Gag plus SP-D-BAFF. (C) When HIV-1 clade C 96ZM651 gp140 was used as the antigen, either pIL-12 alone or pIL-12 plus pSP-D-BAFF induced higher titers of antibody than the antigen alone.

 

HIV-1 gp140 DNA vaccines expressing IL-12p70 plus SP-D-BAFF increased tier 1 and tier 2 neutralization antibody titers.

We next assessed the antibody titers and neutralization activity of vaccines incorporating SP-D-BAFF or the related construct SP-D-APRIL. Experiments again used an HIV-1 gp140 DNA vaccine mouse model. Vaccine groups included pgp140 HIV-1 Env antigen and the adjuvant plasmid pIL-12. As shown in Fig. 2B, all vaccine groups induced anti-Env antibody responses as measured by IgG ELISAs. The mean antibody titer with the SP-D-APRIL adjuvant was lower than that with SP-D-BAFF, but the difference did not reach statistical significance. Vaccines induced both IgG2a and IgG1 antibodies, suggesting a balanced Th1 and Th2 response. Next, sera from vaccinated mice were assayed for HIV-1 neutralization by a TZM.bl assay. As shown in Fig. 2C, pIL-12 plus pSP-D-BAFF significantly enhanced serum neutralization titers over those with the pIL-12 adjuvant alone. Vaccination with the BAFF adjuvant enhanced the neutralization of both tier 1 (MW965.26) and tier 2 (vaccine strain 96ZM651) viruses. Mice vaccinated with both the pIL-12 and pSP-D-APRIL adjuvants showed higher mean neutralization titers against tier 1 virus than those given the pIL-12 adjuvant alone, but the difference did not reach statistical significance (P = 0.076).

SP-D-BAFF enhanced neutralization titers but not IgG levels in a DNA vaccine model. Mice were vaccinated with the HIV-1 clade C 96ZM651 gp140 DNA vaccine plus pIL-12 and either pSP-D-BAFF or SP-D-APRIL. (A) Mice were vaccinated i.m. every 2 weeks for a total of 3 vaccinations and were sacrificed 2 weeks later. (B) Anti-gp120 ELISA. All groups generated similar IgG, IgG2a, and IgG1 titers against HIV-1 Env gp120 as measured by ELISA. (C) The addition of SP-D-BAFF significantly increased tier 1 (MW965.26) and autologous tier 2 (96ZM651) virus neutralization over that for the group receiving gp140 plus IL-12. Neutralization titers were considered positive if values were >3 times the titer against a murine leukemia virus control.

 

Neutralization correlated with IgG2a antibody titers.

To determine the role of the antibody subtype in the neutralization response, we compared IgG, IgG1, and IgG2a antibody titers with tier 1 neutralization titers. Mice from all vaccine groups with detectable neutralization titers were included. As shown in Fig. 3A, we observed a significant positive correlation between the anti-gp120 IgG2a titer and the neutralization titer. In contrast, anti-gp120 IgG and IgG1 titers did not correlate significantly with neutralization (Fig. 3B and data not shown).

An external file that holds a picture, illustration, etc.
Object name is zjv9990902310003.jpg

HIV-1 neutralization correlated with IgG2a antibody titers. Neutralization titers (both tier 1 and tier 2) were correlated with anti-gp120 IgG2a (A) or IgG (B) ELISA values (OD650) at a 1:480 serum dilution. Mice from all vaccine groups with a measurable neutralization titer were included in the analysis.

 

Evaluation of SP-D-BAFF and SP-D-APRIL by DNA prime–protein boost vaccination.

DNA prime–protein boost vaccination is a promising approach for the generation of anti-HIV-1 Env neutralizing antibodies (, ). To extend our encouraging results with SP-D-BAFF and SP-D-APRIL, we investigated whether the performance of these adjuvants could be further enhanced by a protein boost. Vaccination was performed with three biweekly DNA prime injections, followed by a single protein boost 3 weeks after the final DNA vaccination (Fig. 4A). Antibody responses were measured 2 weeks following the protein boost. All groups generated high titers of anti-gp120 IgG antibody following the protein boost (Fig. 4B). No significant differences in IgG, IgG1, IgG2b, or IgA titers were observed between groups. However, the differences in IgG2b titers between the groups given SP-D-BAFF or SP-D-APRIL and the group receiving gp140 plus IL-12p70 did approach significance (P = 0.0744). Importantly, IgG2a antibody titers showed a significant difference at a 10−5 serum dilution between groups given SP-D-BAFF or SP-D-APRIL and groups vaccinated with gp140 plus IL-12p70 alone. Endpoint titers were also determined. As shown in Fig. 4D, there was no significant difference in endpoint titers between groups.

pSP-D-BAFF and pSP-D-APRIL enhanced IgG2a antibody responses following DNA prime–protein boost vaccination. (A) Mice were vaccinated with an HIV-1 clade C 96ZM651 gp140 DNA vaccine as described in the legend to Fig. 2. Three weeks following the final DNA vaccination, mice were boosted with 10 μg 96ZM651 gp120 protein formulated in incomplete Freund's adjuvant. Sera and spleens were obtained 2 weeks later. (B) Anti-gp120 IgG and IgA responses, determined by ELISA. Both SP-D-BAFF and SP-D-APRIL significantly increased IgG2a titers but did not significantly change IgA, IgG, IgG2b, or IgG1 titers. (C) Total serum IgG concentration for each vaccine group. Adjuvants did not significantly increase total IgG concentrations over those obtained with the antigen alone. (D) Calculated anti-gp120 IgG endpoint titers. Anti-gp120 IgG endpoint titers were equivalent for all groups. (E and F) Total B cell numbers and percentages for each vaccine group. Total B cell numbers and percentages were equivalent for all vaccine groups.

Given the role of TNFSF members in B cell development, activation, and expansion, we evaluated the effects of these adjuvants on total IgG concentrations as well as on the proportion and numbers of splenic B cells. As shown in Fig. 4C, there was no significant difference in total IgG concentrations between groups as measured by ELISA, although there was a trend toward an increased mean IgG concentration for the group receiving SP-D-APRIL. Likewise, the numbers and percentages of splenic B cells did not differ significantly between the vaccination groups (Fig. 4E and andFF).

 

DNA prime–protein boost vaccination further enhanced the neutralization of tier 2 vaccine strain 96ZM651 by SP-D-APRIL.

Next, we evaluated tier 1 and tier 2 neutralization titers following DNA prime–protein boost vaccination. As shown in Fig. 5A, we observed no significant difference in tier 1 (MW965.26) neutralization titers among the groups. However, with the addition of the SP-D-APRIL adjuvant, we observed significant increases in tier 2 (vaccine strain 96ZM651) neutralization titers over those with gp140 alone or gp140 plus IL-12 (Fig. 5B). The SP-D-BAFF adjuvant also increased the mean 96ZM651 tier 2 neutralization titer, but the difference did not reach statistical significance (P = 0.0604). SP-D-CD40L did not increase mean 96ZM651 tier 2 neutralization titers over those with either gp140 alone or gp140 plus IL-12.

SP-D-APRIL significantly increased vaccine strain tier 2 neutralization titers in a DNA prime–protein boost vaccine model. The same sera used for the experiment for which results are shown in Fig. 4 were assayed for neutralization of tier 1 (MW965.26) or tier 2 (vaccine strain 96ZM651) HIV-1. (A) Tier 1 neutralization activity. Protein boosting gave similar tier 1 neutralization titers for all groups. (B) Tier 2 neutralization titers were significantly enhanced with the addition of the SP-D-APRIL adjuvant, and the enhancement obtained with SP-D-BAFF approached significance (P = 0.0604). Neutralization titers were considered positive if values were >3 times the titer against a murine leukemia virus control.

 

SP-D-BAFF and SP-D-APRIL significantly increased antibody avidity and the number of gp120-specific antibody-secreting cells.

BAFF and APRIL are known to enhance the germinal center reaction and the expression of activation-induced cytidine deaminase (AID), an enzyme essential for somatic hypermutation in complementarity-determining region 3 (CDR3). AID activity enhances antigen binding as well as immunoglobulin isotype class switch recombination, which determines the localization and function of the antibody molecule (, ,). We hypothesized that a possible mechanism of action of the SP-D-BAFF and SP-D-APRIL adjuvants was to increase the germinal center reaction, enhancing the affinity maturation of Env-specific B cells and thereby leading to enhanced neutralization. To assess affinity maturation, sera from vaccinated animals were evaluated for anti-gp120 functional avidity by ELISA in the presence of increasing concentrations of sodium thiocyanate (NaSCN). As shown in Fig. 6A, addition of either pSP-D-BAFF or pSP-D-APRIL during the DNA prime significantly increased the IgG avidity index over those for all other groups. In contrast, SP-D-CD40L did not significantly increase avidity.

The SP-D-BAFF and SP-D-APRIL adjuvants enhanced avidity, increased the number of gp120-specific B cells per spleen, and increased the number and proportion of effector (Eff) memory CD4 T cells. (A) The avidity index was calculated based on the dissociation of antibody in the presence of NaSCN. Avidity increased significantly with the addition of the SP-D-BAFF or SP-D-APRIL adjuvant, but not with SP-D-CD40L. (B) Number of anti-gp120 ASCs per spleen. An ELISPOT assay was performed on gp120 protein-coated plates to determine the total number of B cells secreting anti-gp120 antibody. The number of antibody-secreting B cells per spleen increased significantly with the addition of the SP-D-BAFF or SP-D-APRIL adjuvant. (C to G) Single-cell suspensions were prepared from spleens from vaccinated animals, and CD4 and CD8 T cells were analyzed by fluorescence-activated cell sorting. (C and D) Number and proportion of effector memory CD4 T cells. The number and proportion of effector memory CD4 T cells increased significantly with the addition of the SP-D-BAFF or SP-D-APRIL adjuvant. (E) Number of total CD4 T cells. (F) Number of CD8 T cells. (G) CD4/CD8 ratio per half-spleen sample. The ratio of CD4/CD8 T cells increased significantly with the addition of SP-D-CD40L, SP-D-BAFF, or SP-D-APRIL.

Next, we determined the number of gp120-specific IgG antibody-secreting cells by a B cell ELISPOT assay using ELISPOT plates coated with gp120 protein. As shown in Fig. 6B, the addition of an SP-D-BAFF or SP-D-APRIL plasmid to the DNA prime vaccine significantly increased the number of IgG antibody-secreting cells per spleen over those for other groups. Again, SP-D-CD40L failed to significantly increase the number of antibody-secreting cells, suggesting that the mechanisms employed by TACI and BAFF-R stimulation, but not CD40 stimulation, were capable of promoting the differentiation, expansion, and/or survival of gp120-specific ASCs. We hypothesized that gp120-specific activated B cells generated by BAFF and APRIL would serve as efficient antigen-presenting cells (APCs), activating CD4 T cells and soliciting T cell help for IgG antibody production. Analysis of splenic T cells revealed increased numbers and proportions of CD4 effector memory cells, with no apparent increase in overall CD4 cell counts (Fig. 6C to toE).E). Further, a modest decrease in the CD8 T cell count and a modest increase in the CD4/CD8 ratio were observed in the BAFF and APRIL groups (Fig. 6F and andG).G). These data provide initial support for our hypothesis.

 

SP-D-BAFF and SP-D-APRIL enhanced the germinal center reaction and the number of germinal centers.

Fluorescence immunohistochemistry was performed to directly evaluate the germinal center reaction 2 weeks following vaccination. Sections were stained for CD3 (T cells), B220 (B cells), and PNA to distinguish germinal centers. Representative spleens are shown in Fig. 7A. For each vaccination group, the top panel shows the entire half-spleen, highlighting the increased number of germinal centers with SP-D-BAFF or SP-D-APRIL. The other panels show individual germinal centers and the colocalization of CD3 and PNA staining. As shown in Fig. 7B, SP-D-BAFF or SP-D-APRIL increased the germinal center area per spleen over that with either IL-12p70 alone or IL-12p70 plus SP-D-CD40L. The numbers of germinal centers in vaccine groups receiving SP-D-BAFF or SP-D-APRIL were also significantly increased over that for the group receiving gp140 alone (Fig. 7C). Again, there was no significant difference with the SP-D-CD40L adjuvant. Taken together, these results suggest that multitrimers of BAFF and APRIL promote the germinal center reaction and increase and/or maintain antibody-secreting cells and long-lived plasma cells. In contrast, these mechanisms are not employed by multitrimers of CD40L.

The SP-D-BAFF and SP-D-APRIL adjuvants increased germinal center reactions. Spleens from vaccinated animals were cryopreserved in OCT compound and were sectioned for immunohistochemistry analysis. (A) Representative images of spleens from mice 2 weeks following protein boost vaccination. Row 1, full half-spleen image; row 2, B220, CD3, and PNA staining; row 3, B220 and PNA staining; row 4, B220 and CD3 staining. (B) Size of the germinal center area per spleen (micrometers squared). The total germinal center area per spleen was calculated. Both the SP-D-BAFF and SP-D-APRIL adjuvants increased the germinal center area over that with SP-D-CD40L. (C) Number of germinal centers per half-spleen. PNA-positive germinal center reactions were counted per half-spleen sample. Addition of the SP-D-BAFF or SP-D-APRIL adjuvant significantly increased the total number of germinal centers.

DISCUSSION

The inability of most humans to mount a protective immune response to HIV-1 suggests that conventional vaccine designs may not be successful in inducing prophylactic immune responses, including the production of bNAbs. There are several barriers that may impede the development of an effective vaccine. The extremely high variability of the viral envelope protein and structural constraints hindering antibody access to conserved viral sequences pose challenges for vaccine design. Further, some HIV-1 Env-specific B cell clones have been shown to react strongly to self-antigens and therefore may be lost to the mechanisms of immunological self-tolerance (,). Consistently, HIV-1 bNAbs are polyreactive and autoreactive. Although polyreactivity and autoreactivity may not contribute to the neutralization activities of these antibodies, the two characteristics coincide frequently (,). With these challenges in mind, we tested the abilities of soluble multitrimerized forms of TNFSF members BAFF and APRIL to promote HIV-1 neutralizing antibodies. BAFF and APRIL were selected because they act primarily on B cells and because multiple aspects of their biology and activation are related to antibody production. For example, BAFF and APRIL enhance the selection and survival of B cells as well as class switch recombination, affinity maturation, the longevity of the GC reaction, and the retention of B cells undergoing affinity maturation in the GC (). In addition, TACI, which can bind both BAFF and APRIL, has been shown to be necessary for long-lasting protection against influenza (). Therefore, we used a combination of multitrimeric BAFF and APRIL together with IL-12 and membrane-bound gp140 to approximate the native conformation. Vaccination with SPD-APRIL or SPD-BAFF in this formulation and a DNA prime–protein boost protocol resulted in neutralizing antibody responses to both tier 1 and tier 2 viruses. DNA vaccination in the absence of a protein boost was also able to induce neutralizing antibody. This encouraging result may reflect the ability of IL-12 plus the BAFF or APRIL adjuvant to induce neutralizing antibody, despite the typically low levels of antibody generated following DNA vaccination. Future studies will assess the electroporation of the DNA vaccines, which may further enhance antibody titers without requiring a protein boost. Overall, these results provide proof of the concept that in spite of the many barriers, it may be possible to design vaccines that can elicit antibodies capable of neutralizing tier 1 and tier 2 HIV-1 clades, and this possibility should be further explored in designing vaccines against HIV-1.

We showed that vaccination with the SP-D-APRIL or SP-D-BAFF adjuvant together with IL-12 and HIV-1 Env elicited a neutralizing antibody response, whereas the CD40L adjuvant did not. It is not clear why CD40L failed or why APRIL was at least modestly better than BAFF at inducing an anti-HIV-1 antibody response. One explanation may be that, as we have shown previously (; unpublished data), BCR engagement robustly upregulates the expression of TACI and BAFF-R on the cell surface, whereas CD40 levels do not change. Additionally, large aggregates (or multitrimers) of BAFF, but not single BAFF trimers, can activate both BAFF-R and TACI signaling. APRIL can also bind the receptors TACI and BCMA, which are important for plasma cell survival. Thus, the provision of multitrimers of BAFF and APRIL would activate B cells and drive their differentiation into IgG ASCs. However, this model raises the question of how BAFF and APRIL activate antigen-specific B cells. A possible explanation is that under conditions of limiting antigen concentration, B cell clones specific for that Ag are more likely to grab the antigen and serve as antigen-presenting cells (APCs) for T cell activation. In contrast, innate APCs would take up antigen nonspecifically. Thus, the gp120 in our vaccine would sensitize Ag-specific B cells by increasing TACI and BAFF-R expression on the cell surface. Increased TACI and BAFF-R expression would focus BAFF and APRIL activity onto Ag-specific B cell clones, leading to enhanced Ag-specific antibody responses and neutralizing activity. In this scenario, APRIL would engage TACI on Ag-activated B cells, promote antibody production, and induce CSR, as well as promoting plasma cell survival via BCMA. BAFF would interact with gp120-sensitized B cells and would signal through TACI and BCMA as well as BAFF-R, thereby maintaining B cell survival and metabolic fitness during activation and antibody production.

How BAFF and APRIL act as adjuvants to generate effective anti-HIV humoral immunity upon vaccination is not known. Some researchers have reported some success with BAFF, and particularly with APRIL (). When given in soluble form, the BAFF, APRIL, and CD40L adjuvants can enhance IL-4 and IL-5 secretion by CD4 T cells but are unable to induce a significant increase in humoral responses. Direct targeting of B cells with soluble single trimers of CD40L, BAFF, and APRIL fused to HIV-1 gp120 in DNA prime–protein boost vaccination of rabbits enhanced neutralizing antibody titers (). In contrast to that study and previous data from our lab (), we show here that the combination of IL-12 with APRIL or BAFF multitrimers, but not CD40L multitrimers, enhanced tier 1 and tier 2 neutralization. While the data are limited to a tier 1 virus and the autologous tier 2 viral strain, these studies suggest that APRIL and BAFF are likely better adjuvants in multitrimeric than in single-trimer form, enhancing gp120 immunogenicity and generating Th1-like antibody responses when used in combination with IL-12. These data further suggest that direct targeting of B cells or antigen fusion with APRIL and BAFF may not provide an advantage in eliciting neutralizing antibodies.

Neutralization was associated with increased IgG2a titers and a trend toward increased IgG2b titers, but not with increases in total IgG or IgG1 titers, suggesting that a Th1 response was driving the generation of neutralizing antibodies. This Th1 response is consistent with a previous report () showing that BAFF increased titers of the Th1 antibodies IgG2a and IgG3 relative to those for the Th2 antibody isotype IgG1. Similarly, in previous studies, including studies with a prime-boost immunization approach, the IgG2a isotype showed 3- to 4-fold higher anti-herpes simplex virus 2 neutralization activity and provided better protection against viral infection in mice (,). Overall, these data support the concept that SP-D-BAFF and SP-D-APRIL can drive the production of neutralizing antibodies through enhanced Th1-biased antibody responses that promote IgG2a production.

It is possible that the neutralizing activity elicited by our vaccine design is derived from autoreactive B cell clones. This may be of concern due to the potential for self-tissue destruction or neutralization of self-molecules. First, this will depend on whether tolerance checkpoints acting on B cells specific for HIV-1 Env proteins are central or peripheral. If such autoreactive B cells are centrally deleted, as has been reported so far (, , ,), our vaccine design is unlikely to rescue them, given that adjuvants in the vaccine formulation act primarily on peripheral B cells. In this regard, a recent report has demonstrated that most B cells in both mice and humans are variably autoreactive, and therefore, any harmful effects that may occur will depend in part on the affinity of HIV-1 neutralizing antibodies to self-antigen. Another likely possibility is that the neutralizing antibodies observed arise from broadly reactive marginal zone B cells through affinity maturation. Mouse marginal zone B cells have recently been shown to harbor specificities similar to those of human bNAbs ().

In conclusion, a DNA vaccine consisting of membrane-associated gp140, an IL-12p70 single-chain fusion, and either SP-D-BAFF or SP-D-APRIL elicited antibody production with increased HIV-1 neutralization activity. Vaccines including SP-D-BAFF or SP-D-APRIL as an adjuvant also showed increased numbers of gp120-specific ASCs and antibodies with increased affinity, suggesting that SP-D-BAFF and SP-D-APRIL promote the GC reaction and affinity maturation and support the maintenance of memory and/or long-lived plasma cells.

ACKNOWLEDGMENTS

We thank Yaelis Rivas and Francesca Raffa for technical assistance. We gratefully acknowledge Richard S. Kornbluth and Multimeric Biotherapeutics Inc. for providing the SP-D-BAFF, SP-D-APRIL, and SP-D-CD40L constructs used in this study. We thank George N. Pavlakis for providing the HIV-1 Gag DNA vaccine construct. We thank the Miami Center for AIDS Research Laboratory Core D for laboratory service support and reagents. We also thank Oliver Umland at the University of Miami Diabetes Research Center flow cytometry core facility for flow cytometry services.

G.W.S. was supported by National Institutes of Health (NIH), National Institute of Allergy and Infectious Diseases (NIAID) supplemental grant SB15 from the Miami Center for AIDS Research (CFAR) at the University of Miami Miller School of Medicine (CFAR grant P30 AI073961;; principal investigator, Savita Pahwa) and by grant K22 AI068489. W.N.K. was supported by NIH grant R21 AI088511. D.C.M. was supported by NIAID-NIH contract HHSN27201100016C. E.S.C. was supported by a CFAR training grant from the Miami CFAR (grant P30 AI073961). The following reagents were obtained through the NIH AIDS Reagent Program, Division of AIDS (DAIDS), NIAID: HIV-196ZM651 Env plasmids p96ZM651gp140-CD5-opt and p96ZM651gp120-CD5-opt from Yingying Li, Feng Gao, and Beatrice H. Hahn; HIV-196ZM651 gp120 protein and HIV-1 IIIB p55 Gag protein from DAIDS, NIAID.

REFERENCES

1. Burton DR, Pyati J, Koduri R, Sharp SJ, Thornton GB, Parren PW, Sawyer LS, Hendry RM, Dunlop N, Nara PL, Lamacchia M, Garratty E, Stiehm ER, Bryson YJ, Cao Y, Moore JP, Ho DD, Barbas CF. 1994. Efficient neutralization of primary isolates of HIV-1 by a recombinant human monoclonal antibody. Science 266:1024–1027. doi:10.1126/science.7973652. [PubMed] [CrossRef] []
2. Walker LM, Huber M, Doores KJ, Falkowska E, Pejchal R, Julien JP, Wang SK, Ramos A, Chan-Hui PY, Moyle M, Mitcham JL, Hammond PW, Olsen OA, Phung P, Fling S, Wong CH, Phogat S, Wrin T, Simek MD, Koff WC, Wilson IA, Burton DR, Poignard P. 2011. Broad neutralization coverage of HIV by multiple highly potent antibodies. Nature 477:466–470. doi:10.1038/nature10373. [PMC free article] [PubMed] [CrossRef] []
3. Walker LM, Phogat SK, Chan-Hui PY, Wagner D, Phung P, Goss JL, Wrin T, Simek MD, Fling S, Mitcham JL, Lehrman JK, Priddy FH, Olsen OA, Frey SM, Hammond PW, Kaminsky S, Zamb T, Moyle M, Koff WC, Poignard P, Burton DR. 2009. Broad and potent neutralizing antibodies from an African donor reveal a new HIV-1 vaccine target. Science 326:285–289. doi:10.1126/science.1178746. [PMC free article] [PubMed] [CrossRef] []
4. Wu X, Yang ZY, Li Y, Hogerkorp CM, Schief WR, Seaman MS, Zhou T, Schmidt SD, Wu L, Xu L, Longo NS, McKee K, O'Dell S, Louder MK, Wycuff DL, Feng Y, Nason M, Doria-Rose N, Connors M, Kwong PD, Roederer M, Wyatt RT, Nabel GJ, Mascola JR. 2010. Rational design of envelope identifies broadly neutralizing human monoclonal antibodies to HIV-1. Science 329:856–861. doi:10.1126/science.1187659. [PMC free article] [PubMed] [CrossRef] []
5. Finton KA, Larimore K, Larman HB, Friend D, Correnti C, Rupert PB, Elledge SJ, Greenberg PD, Strong RK. 2013. Autoreactivity and exceptional CDR plasticity (but not unusual polyspecificity) hinder elicitation of the anti-HIV antibody 4E10. PLoS Pathog 9:e1003639. doi:10.1371/journal.ppat.1003639. [PMC free article] [PubMed] [CrossRef] []
6. Haynes BF, Fleming J, St Clair EW, Katinger H, Stiegler G, Kunert R, Robinson J, Scearce RM, Plonk K, Staats HF, Ortel TL, Liao HX, Alam SM. 2005. Cardiolipin polyspecific autoreactivity in two broadly neutralizing HIV-1 antibodies. Science 308:1906–1908. doi:10.1126/science.1111781. [PubMed] [CrossRef] []
7. Verkoczy L, Diaz M, Holl TM, Ouyang YB, Bouton-Verville H, Alam SM, Liao HX, Kelsoe G, Haynes BF. 2010. Autoreactivity in an HIV-1 broadly reactive neutralizing antibody variable region heavy chain induces immunologic tolerance. Proc Natl Acad Sci U S A 107:181–186. doi:10.1073/pnas.0912914107. [PMC free article] [PubMed] [CrossRef] []
8. Zikherman J, Parameswaran R, Weiss A. 2012. Endogenous antigen tunes the responsiveness of naive B cells but not T cells. Nature 489:160–164. doi:10.1038/nature11311. [PMC free article] [PubMed] [CrossRef] []
9. McElrath MJ, Haynes BF. 2010. Induction of immunity to human immunodeficiency virus type-1 by vaccination. Immunity 33:542–554. doi:10.1016/j.immuni.2010.09.011. [PMC free article] [PubMed] [CrossRef] []
10. Pujanauski LM, Janoff EN, McCarter MD, Pelanda R, Torres RM. 2013. Mouse marginal zone B cells harbor specificities similar to human broadly neutralizing HIV antibodies. Proc Natl Acad Sci U S A 110:1422–1427. doi:10.1073/pnas.1213713110. [PMC free article] [PubMed] [CrossRef] []
11. Haynes BF, Moody MA, Liao HX, Verkoczy L, Tomaras GD. 2011. B cell responses to HIV-1 infection and vaccination: pathways to preventing infection. Trends Mol Med 17:108–116. doi:10.1016/j.molmed.2010.10.008. [PMC free article] [PubMed] [CrossRef] []
12. Pantophlet R. 2010. Antibody epitope exposure and neutralization of HIV-1. Curr Pharm Des 16:3729–3743. doi:10.2174/138161210794079182. [PubMed] [CrossRef] []
13. He B, Santamaria R, Xu W, Cols M, Chen K, Puga I, Shan M, Xiong H, Bussel JB, Chiu A, Puel A, Reichenbach J, Marodi L, Doffinger R, Vasconcelos J, Issekutz A, Krause J, Davies G, Li X, Grimbacher B, Plebani A, Meffre E, Picard C, Cunningham-Rundles C, Casanova JL, Cerutti A. 2010. The transmembrane activator TACI triggers immunoglobulin class switching by activating B cells through the adaptor MyD88. Nat Immunol 11:836–845. doi:10.1038/ni.1914. [PMC free article] [PubMed] [CrossRef] []
14. Khan WN. 2009. B cell receptor and BAFF receptor signaling regulation of B cell homeostasis. J Immunol 183:3561–3567. doi:10.4049/jimmunol.0800933. [PubMed] [CrossRef] []
15. Puga I, Cols M, Barra CM, He B, Cassis L, Gentile M, Comerma L, Chorny A, Shan M, Xu W, Magri G, Knowles DM, Tam W, Chiu A, Bussel JB, Serrano S, Lorente JA, Bellosillo B, Lloreta J, Juanpere N, Alameda F, Baro T, de Heredia CD, Toran N, Catala A, Torrebadell M, Fortuny C, Cusi V, Carreras C, Diaz GA, Blander JM, Farber CM, Silvestri G, Cunningham-Rundles C, Calvillo M, Dufour C, Notarangelo LD, Lougaris V, Plebani A, Casanova JL, Ganal SC, Diefenbach A, Arostegui JI, Juan M, Yague J, Mahlaoui N, Donadieu J, Chen K, Cerutti A. 2012. B cell-helper neutrophils stimulate the diversification and production of immunoglobulin in the marginal zone of the spleen. Nat Immunol 13:170–180. doi:10.1038/ni.2194. [PMC free article] [PubMed] [CrossRef] []
16. Treml LS, Carlesso G, Hoek KL, Stadanlick JE, Kambayashi T, Bram RJ, Cancro MP, Khan WN. 2007. TLR stimulation modifies BLyS receptor expression in follicular and marginal zone B cells. J Immunol 178:7531–7539. doi:10.4049/jimmunol.178.12.7531. [PubMed] [CrossRef] []
17. Uslu K, Coleman AS, Allman WR, Katsenelson N, Bram RJ, Alugupalli KR, Akkoyunlu M. 2014. Impaired B cell receptor signaling is responsible for reduced TACI expression and function in X-linked immunodeficient mice. J Immunol 192:3582–3595. doi:10.4049/jimmunol.1203468. [PubMed] [CrossRef] []
18. Lesley R, Xu Y, Kalled SL, Hess DM, Schwab SR, Shu HB, Cyster JG. 2004. Reduced competitiveness of autoantigen-engaged B cells due to increased dependence on BAFF. Immunity 20:441–453. doi:10.1016/S1074-7613(04)00079-2. [PubMed] [CrossRef] []
19. Liu Z, Davidson A. 2011. BAFF and selection of autoreactive B cells. Trends Immunol 32:388–394. doi:10.1016/j.it.2011.06.004. [PMC free article] [PubMed] [CrossRef] []
20. Ota M, Duong BH, Torkamani A, Doyle CM, Gavin AL, Ota T, Nemazee D. 2010. Regulation of the B cell receptor repertoire and self-reactivity by BAFF. J Immunol 185:4128–4136. doi:10.4049/jimmunol.1002176. [PMC free article] [PubMed] [CrossRef] []
21. Thien M, Phan TG, Gardam S, Amesbury M, Basten A, Mackay F, Brink R. 2004. Excess BAFF rescues self-reactive B cells from peripheral deletion and allows them to enter forbidden follicular and marginal zone niches. Immunity 20:785–798. doi:10.1016/j.immuni.2004.05.010. [PubMed] [CrossRef] []
22. Bossen C, Cachero TG, Tardivel A, Ingold K, Willen L, Dobles M, Scott ML, Maquelin A, Belnoue E, Siegrist CA, Chevrier S, Acha-Orbea H, Leung H, Mackay F, Tschopp J, Schneider P. 2008. TACI, unlike BAFF-R, is solely activated by oligomeric BAFF and APRIL to support survival of activated B cells and plasmablasts. Blood 111:1004–1012. doi:10.1182/blood-2007-09-110874. [PubMed] [CrossRef] []
23. Cerutti A, Puga I, Cols M. 2011. Innate control of B cell responses. Trends Immunol 32:202–211. doi:10.1016/j.it.2011.02.004. [PMC free article] [PubMed] [CrossRef] []
24. Mackay F, Schneider P. 2009. Cracking the BAFF code. Nat Rev Immunol 9:491–502. doi:10.1038/nri2572. [PubMed] [CrossRef] []
25. Scholz JL, Oropallo MA, Sindhava V, Goenka R, Cancro MP. 2013. The role of B lymphocyte stimulator in B cell biology: implications for the treatment of lupus. Lupus 22:350–360. doi:10.1177/0961203312469453. [PubMed] [CrossRef] []
26. Castigli E, Wilson SA, Scott S, Dedeoglu F, Xu S, Lam KP, Bram RJ, Jabara H, Geha RS. 2005. TACI and BAFF-R mediate isotype switching in B cells. J Exp Med 201:35–39. doi:10.1084/jem.20032000. [PMC free article] [PubMed] [CrossRef] []
27. Rickert RC, Jellusova J, Miletic AV. 2011. Signaling by the tumor necrosis factor receptor superfamily in B-cell biology and disease. Immunol Rev 244:115–133. doi:10.1111/j.1600-065X.2011.01067.x. [PMC free article] [PubMed] [CrossRef] []
28. Groom JR, Fletcher CA, Walters SN, Grey ST, Watt SV, Sweet MJ, Smyth MJ, Mackay CR, Mackay F. 2007. BAFF and MyD88 signals promote a lupuslike disease independent of T cells. J Exp Med 204:1959–1971. doi:10.1084/jem.20062567. [PMC free article] [PubMed] [CrossRef] []
29. Tertilt C, Joh J, Krause A, Chou P, Schneeweiss K, Crystal RG, Worgall S. 2009. Expression of B-cell activating factor enhances protective immunity of a vaccine against Pseudomonas aeruginosa. Infect Immun 77:3044–3055. doi:10.1128/IAI.00927-08. [PMC free article] [PubMed] [CrossRef] []
30. O'Connor BP, Raman VS, Erickson LD, Cook WJ, Weaver LK, Ahonen C, Lin LL, Mantchev GT, Bram RJ, Noelle RJ. 2004. BCMA is essential for the survival of long-lived bone marrow plasma cells. J Exp Med 199:91–98. doi:10.1084/jem.20031330. [PMC free article] [PubMed] [CrossRef] []
31. Benson MJ, Dillon SR, Castigli E, Geha RS, Xu S, Lam KP, Noelle RJ. 2008. The dependence of plasma cells and independence of memory B cells on BAFF and APRIL. J Immunol 180:3655–3659. doi:10.4049/jimmunol.180.6.3655. [PubMed] [CrossRef] []
32. Ou X, Xu S, Lam KP. 2012. Deficiency in TNFRSF13B (TACI) expands T-follicular helper and germinal center B cells via increased ICOS-ligand expression but impairs plasma cell survival. Proc Natl Acad Sci U S A 109:15401–15406. doi:10.1073/pnas.1200386109. [PMC free article] [PubMed] [CrossRef] []
33. Melchers M, Bontjer I, Tong T, Chung NP, Klasse PJ, Eggink D, Montefiori DC, Gentile M, Cerutti A, Olson WC, Berkhout B, Binley JM, Moore JP, Sanders RW. 2012. Targeting HIV-1 envelope glycoprotein trimers to B cells by using APRIL improves antibody responses. J Virol 86:2488–2500. doi:10.1128/JVI.06259-11. [PMC free article] [PubMed] [CrossRef] []
34. Kanagavelu SK, Snarsky V, Termini JM, Gupta S, Barzee S, Wright JA, Khan WN, Kornbluth RS, Stone GW. 2012. Soluble multi-trimeric TNF superfamily ligand adjuvants enhance immune responses to a HIV-1 Gag DNA vaccine. Vaccine 30:691–702. doi:10.1016/j.vaccine.2011.11.088. [PMC free article] [PubMed] [CrossRef] []
35. Stone GW, Barzee S, Snarsky V, Kee K, Spina CA, Yu XF, Kornbluth RS. 2006. Multimeric soluble CD40 ligand and GITR ligand as adjuvants for human immunodeficiency virus DNA vaccines. J Virol 80:1762–1772. doi:10.1128/JVI.80.4.1762-1772.2006. [PMC free article] [PubMed] [CrossRef] []
36. Stone GW, Barzee S, Snarsky V, Spina CA, Lifson JD, Pillai VK, Amara RR, Villinger F, Kornbluth RS. 2006. Macaque multimeric soluble CD40 ligand and GITR ligand constructs are immunostimulatory molecules in vitro. Clin Vaccine Immunol 13:1223–1230. doi:10.1128/CVI.00198-06. [PMC free article] [PubMed] [CrossRef] []
37. Qiu JT, Liu B, Tian C, Pavlakis GN, Yu XF. 2000. Enhancement of primary and secondary cellular immune responses against human immunodeficiency virus type 1 Gag by using DNA expression vectors that target Gag antigen to the secretory pathway. J Virol 74:5997–6005. doi:10.1128/JVI.74.13.5997-6005.2000. [PMC free article] [PubMed] [CrossRef] []
38. Stone GW, Barzee S, Snarsky V, Santucci C, Tran B, Kornbluth RS. 2009. Regression of established AB1 murine mesothelioma induced by peritumoral injections of CpG oligodeoxynucleotide either alone or in combination with poly(I:C) and CD40 ligand plasmid DNA. J Thorac Oncol 4:802–808. doi:10.1097/JTO.0b013e3181a8634d. [PubMed] [CrossRef] []
39. Gasteiger E, Gattiker A, Hoogland C, Ivanyi I, Appel RD, Bairoch A. 2003. ExPASy: The proteomics server for in-depth protein knowledge and analysis. Nucleic Acids Res 31:3784–3788. doi:10.1093/nar/gkg563. [PMC free article] [PubMed] [CrossRef] []
40. Seaman MS, Janes H, Hawkins N, Grandpre LE, Devoy C, Giri A, Coffey RT, Harris L, Wood B, Daniels MG, Bhattacharya T, Lapedes A, Polonis VR, McCutchan FE, Gilbert PB, Self SG, Korber BT, Montefiori DC, Mascola JR. 2010. Tiered categorization of a diverse panel of HIV-1 Env pseudoviruses for assessment of neutralizing antibodies. J Virol 84:1439–1452. doi:10.1128/JVI.02108-09. [PMC free article] [PubMed] [CrossRef] []
41. Ross TM, Xu Y, Green TD, Montefiori DC, Robinson HL. 2001. Enhanced avidity maturation of antibody to human immunodeficiency virus envelope: DNA vaccination with gp120-C3d fusion proteins. AIDS Res Hum Retroviruses 17:829–835. doi:10.1089/088922201750252025. [PMC free article] [PubMed] [CrossRef] []
42. Lieschke GJ, Rao PK, Gately MK, Mulligan RC. 1997. Bioactive murine and human interleukin-12 fusion proteins which retain antitumor activity in vivo. Nat Biotechnol 15:35–40. doi:10.1038/nbt0197-35. [PubMed] [CrossRef] []
43. Vaine M, Wang S, Crooks ET, Jiang P, Montefiori DC, Binley J, Lu S. 2008. Improved induction of antibodies against key neutralizing epitopes by human immunodeficiency virus type 1 gp120 DNA prime-protein boost vaccination compared to gp120 protein-only vaccination. J Virol 82:7369–7378. doi:10.1128/JVI.00562-08. [PMC free article] [PubMed] [CrossRef] []
44. Wang S, Arthos J, Lawrence JM, Van Ryk D, Mboudjeka I, Shen S, Chou TH, Montefiori DC, Lu S. 2005. Enhanced immunogenicity of gp120 protein when combined with recombinant DNA priming to generate antibodies that neutralize the JR-FL primary isolate of human immunodeficiency virus type 1. J Virol 79:7933–7937. doi:10.1128/JVI.79.12.7933-7937.2005. [PMC free article] [PubMed] [CrossRef] []
45. Rahman ZS, Manser T. 2004. B cells expressing Bcl-2 and a signaling-impaired BAFF-specific receptor fail to mature and are deficient in the formation of lymphoid follicles and germinal centers. J Immunol 173:6179–6188. doi:10.4049/jimmunol.173.10.6179. [PubMed] [CrossRef] []
46. Rahman ZS, Rao SP, Kalled SL, Manser T. 2003. Normal induction but attenuated progression of germinal center responses in BAFF and BAFF-R signaling-deficient mice. J Exp Med 198:1157–1169. doi:10.1084/jem.20030495. [PMC free article] [PubMed] [CrossRef] []
47. Yoon SO, Zhang X, Berner P, Blom B, Choi YS. 2009. Notch ligands expressed by follicular dendritic cells protect germinal center B cells from apoptosis. J Immunol 183:352–358. doi:10.4049/jimmunol.0803183. [PubMed] [CrossRef] []
48. Holl TM, Yang G, Kuraoka M, Verkoczy L, Alam SM, Moody MA, Haynes BF, Kelsoe G. 2014. Enhanced antibody responses to an HIV-1 membrane-proximal external region antigen in mice reconstituted with cultured lymphocytes. J Immunol 192:3269–3279. doi:10.4049/jimmunol.1302829. [PMC free article] [PubMed] [CrossRef] []
49. Liu M, Yang G, Wiehe K, Nicely NI, Vandergrift NA, Rountree W, Bonsignori M, Alam SM, Gao J, Haynes BF, Kelsoe G. 2015. Polyreactivity and autoreactivity among HIV-1 antibodies. J Virol 89:784–798. doi:10.1128/JVI.02378-14. [PMC free article] [PubMed] [CrossRef] []
50. Verkoczy L, Kelsoe G, Haynes BF. 2014. HIV-1 envelope gp41 broadly neutralizing antibodies: hurdles for vaccine development. PLoS Pathog 10:e1004073. doi:10.1371/journal.ppat.1004073. [PMC free article] [PubMed] [CrossRef] []
51. Sasaki Y, Casola S, Kutok JL, Rajewsky K, Schmidt-Supprian M. 2004. TNF family member B cell-activating factor (BAFF) receptor-dependent and -independent roles for BAFF in B cell physiology. J Immunol 173:2245–2252. doi:10.4049/jimmunol.173.4.2245. [PubMed] [CrossRef] []
52. Wolf AI, Mozdzanowska K, Quinn WJ III, Metzgar M, Williams KL, Caton AJ, Meffre E, Bram RJ, Erickson LD, Allman D, Cancro MP, Erikson J. 2011. Protective antiviral antibody responses in a mouse model of influenza virus infection require TACI. J Clin Invest 121:3954–3964. doi:10.1172/JCI57362. [PMC free article] [PubMed] [CrossRef] []
53. Ishizaka ST, Piacente P, Silva J, Mishkin EM. 1995. IgG subtype is correlated with efficiency of passive protection and effector function of anti-herpes simplex virus glycoprotein D monoclonal antibodies. J Infect Dis 172:1108–1111. doi:10.1093/infdis/172.4.1108. [PubMed] [CrossRef] []
54. Morello CS, Kraynyak KA, Levinson MS, Chen Z, Lee KF, Spector DH. 2012. Inactivated HSV-2 in MPL/alum adjuvant provides nearly complete protection against genital infection and shedding following long term challenge and rechallenge. Vaccine 30:6541–6550. doi:10.1016/j.vaccine.2012.08.049. [PMC free article] [PubMed] [CrossRef] []
55. Sin JI, Bagarazzi M, Pachuk C, Weiner DB. 1999. DNA priming-protein boosting enhances both antigen-specific antibody and Th1-type cellular immune responses in a murine herpes simplex virus-2 gD vaccine model. DNA Cell Biol 18:771–779. doi:10.1089/104454999314917. [PubMed] [CrossRef] []
56. Bonsignori M, Wiehe K, Grimm SK, Lynch R, Yang G, Kozink DM, Perrin F, Cooper AJ, Hwang KK, Chen X, Liu M, McKee K, Parks RJ, Eudailey J, Wang M, Clowse M, Criscione-Schreiber LG, Moody MA, Ackerman ME, Boyd SD, Gao F, Kelsoe G, Verkoczy L, Tomaras GD, Liao HX, Kepler TB, Montefiori DC, Mascola JR, Haynes BF. 2014. An autoreactive antibody from an SLE/HIV-1 individual broadly neutralizes HIV-1. J Clin Invest 124:1835–1843. doi:10.1172/JCI73441. [PMC free article] [PubMed] [CrossRef] []
57. Haynes BF, Verkoczy L. 2014. AIDS/HIV. Host controls of HIV neutralizing antibodies. Science 344:588–589. doi:10.1126/science.1254990. [PMC free article] [PubMed] [CrossRef] []
58. Verkoczy L, Chen Y, Zhang J, Bouton-Verville H, Newman A, Lockwood B, Scearce RM, Montefiori DC, Dennison SM, Xia SM, Hwang KK, Liao HX, Alam SM, Haynes BF. 2013. Induction of HIV-1 broad neutralizing antibodies in 2F5 knock-in mice: selection against membrane proximal external region-associated autoreactivity limits T-dependent responses. J Immunol 191:2538–2550. doi:10.4049/jimmunol.1300971. [PMC free article] [PubMed] [CrossRef] []

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