Zika virus (ZIKV) is a mosquito-borne pathogen that recently caused a series of increasingly severe outbreaks. We previously demonstrated that, compared with a pre-epidemic isolate (ZIKVCDN), a Brazilian ZIKV isolate (ZIKVBR) possesses a novel capacity to suppress host immunity, resulting in delayed viral clearance. However, whether ZIKVBR modulates CD4 T cell responses remains unknown. In this study, we show that, in comparison with ZIKVCDN infection, CD4 T cells are less polarized to the Th1 subtype following ZIKVBR challenge in mice. In contrast, we observed an enhanced accumulation of T follicular helper cells 10, 14, and 21 d postinfection with ZIKVBR. This response correlated with an enhanced germinal center B cell response and robust production of higher avidity-neutralizing Abs following ZIKVBR infection. Taken together, our data suggest that contemporary ZIKV strains have evolved to differentially induce CD4 T cell, B cell, and Ab responses and this could provide a model to further define the signals required for T follicular helper cell development.

Zika virus (ZIKV) was first isolated in the Zika Forest region of Uganda in 1947 (1, 2). Very little was known about the virus during the 20th century and it was linked to mostly innocuous symptoms. Improving our understanding of the immune response to ZIKV infection has become a major research priority following a series of increasingly severe outbreaks in the Federated States of Micronesia (2007), French Polynesia (2013), and South and Central America (with other outbreaks worldwide; 2015) (36). These outbreaks, together with smaller outbreaks that have occurred since 2015, have demonstrated the novel capacity of ZIKV to cause widespread disease and severe symptoms following infection. In particular, ZIKV infection has been associated with Guillain–Barré syndrome (an autoimmune paralysis) in adults, and fetal microcephaly along with a number of other developmental defects when infection occurs during pregnancy (7, 8). Together, these outbreaks and the symptoms that have become associated with infection raise the question of whether recent, epidemic ZIKV isolates have a distinct capacity to modulate host immunity.

CD4 T cells represent an important part of the host immune response against viral infections with multiple roles during primary infection. Following activation, CD4 T cells can differentiate into various Th cell subsets, which serve key functions in supporting the immune response. In the context of viral infection, IL-12 and IFN-γ lead to the generation of Th1 CD4 T cells (9). These cells express the T-box transcription factor T-bet and support the antiviral immune response by secreting effector cytokines such as IFN-γ, TNF-α, and IL-2 and by promoting CD8 T cell activation via dendritic cell (DC) licensing (9, 10). T follicular helper (Tfh) cells are an additional CD4 T cell subset that plays an important role in promoting Ab responses during viral infections. Tfh cells are defined by expression of CXCR5 and PD-1 on the cell surface, as well as the transcription factor B cell lymphoma-6 (Bcl-6) (11). Canonically, pre-Tfh cells must interact with both DCs and B cells for differentiation, and cytokine-dependent and -independent signals contribute to their development (11). Once differentiated, Tfh cells are able to form or enter germinal centers (GCs), distinct structures in secondary lymphoid tissues within which Tfh cells mediate GC B cell affinity maturation and proliferation (1114).

Several studies have explored CD4 T cell responses during ZIKV infection, in a variety of mouse models (15). In immunocompetent C57BL/6 mice, our group has demonstrated that infection with a pre-epidemic Canadian ZIKV isolate (ZIKVCDN, PLCal_ZV, GenBank accession no. KF993678, isolated in 2013 from a Canadian traveler returning from Thailand and exhibiting mild symptoms, https://www.ncbi.nlm.nih.gov/nuccore/KF993678) leads to a robust Th1 CD4 T cell response 7 d postinfection (dpi), characterized by upregulation of T-bet, and production of IFN-γ, TNF-α, and IL-2 (16). Experiments using a Cambodian ZIKV isolate (FSS13025) in LysMCre+Ifnarfl/fl mice, which lack IFN-α/β receptor (IFNAR) expression primarily in mature macrophages and granulocytes, revealed a similar Th1 polarization of CD4 T cells, and also identified formation of a Tfh cell response (17). Although CD4 T cells were required for generation of an IgG response, their depletion had no impact on viral burden or CD8 T cell responses (17). In contrast, depleting CD4 T cells from highly susceptible IFNAR knockout (KO) mice caused higher viral loads, more severe paralysis, and reduced survival in 10- to 12-wk-old mice infected with a Puerto Rican isolate (PRVABC59), and lethal infection in 3- to 4-wk-old mice infected with a Brazilian isolate (PE243) (18, 19). The Tfh cell response to ZIKV was more closely examined in a recent paper by Liang et al. (20) who described a robust Tfh cell response to a Chinese isolate (SZ-WIV01) characterized by IFN-γ production, IgG2c production, and long-lasting Ab responses in C57BL/6 mice treated with an IFNAR blocking Ab (20). Although these studies collectively suggest an important role for CD4 T cells during ZIKV infection, most were carried out in immunodeficient mouse models. Furthermore, none of these studies has compared whether pre-epidemic and epidemic ZIKV isolates differ in their capacity to induce Th1 or Tfh cell responses.

Previously, we have demonstrated that an epidemic ZIKV isolate from Brazil (ZIKVBR, HS-2015-BA-01, GenBank accession no. KX520666, isolated in 2015 from a symptomatic patient in Salvador, Bahia, Brazil, https://www.ncbi.nlm.nih.gov/nuccore/KX520666) actively suppresses the CD8 T cell response to infection, which correlates with delayed virus clearance, when compared with rapid control of infection with ZIKVCDN (21). Although virus is detectable by plaque assay in the spleen and kidney at 7 dpi in mice infected with ZIKVBR, it is cleared from these organs by 14 dpi, indicating that although ZIKVBR establishes a more sustained infection, it does not become chronic in mice (21). In this study, we sought to determine whether ZIKVBR modulates CD4 T cell responses, and to determine potential mechanisms for viral clearance following ZIKVBR infection. Our data demonstrate that ZIKVCDN induces a more robust Ag-experienced, Th1-polarized CD4 T cell response than does ZIKVBR at 7 dpi. In contrast, ZIKVBR-infected mice were more efficient at inducing Tfh and GC B cell responses 10 dpi, which persisted through 14 and 21 dpi. These responses correlated with enhanced formation of GCs in the spleen, as well as production of higher avidity Abs with greater neutralization potential, likely contributing to the control of ZIKVBR infection. Conversely, ZIKVCDN infection favored the induction of extrafollicular B cell responses that correlated with the production of lower quality virus-specific Abs with reduced neutralization capacity. Taken together, these data suggest that ZIKVBR may have evolved to dampen T cell responses that prevent rapid viral clearance, while simultaneously evoking more potent T-dependent humoral immunity that drives a contained, yet more sustained infection.

Vero cells (African green monkey kidney epithelial cells, provided by Steven Varga, The University of Iowa) were cultured in DMEM (Wisent) supplemented with 10% heat-inactivated FBS (Wisent), 1% l-glutamine (Wisent), 1% penicillin-streptomycin (Wisent), and 1% non-essential amino acids (Sigma-Aldrich) at 37°C and 5% CO2.

Low-passage (p.4) ZIKVCDN (PLCal_ZV, GenBank accession no. KF993678, https://www.ncbi.nlm.nih.gov/nuccore/KF993678) derived from a ZIKV-infected traveler returning to Canada who acquired the infection in Thailand in 2013 was provided by Gary Kobinger and David Safronetz (National Microbiology Laboratory and Public Health Agency of Canada) and confirmed via sequencing to be of the Asian lineage (22). Low-passage (p.4) ZIKVBR (HS-2015-BA-01, GenBank accession no. KX520666, https://www.ncbi.nlm.nih.gov/nuccore/KX520666) isolated in August 2015 in Salvador, Bahia, Brazil, was provided by Mauro Teixeira (Universidade Federal de Minas Gerais). The stock was originally passaged three times in C6/36 mosquito cells and once in Vero cells. To propagate ZIKV stocks, 6 × 106 Vero cells were seeded in 150- × 2-mm dishes (Sigma-Aldrich), and 24 h later infected at a multiplicity of infection of 1 for 2 h in unsupplemented Eagle’s MEM (EMEM). After 2 h, infection media were removed and replaced with 15 ml of DMEM supplemented with 2% heat-inactivated FBS, 1% l-glutamine, 1% non-essential amino acids, 1% penicillin-streptomycin, and 15 mM HEPES. Supernatants were harvested after 48–72 h postinfection (hpi), centrifuged for 10 min at 3000 × g, aliquoted, and stored at −80°C. Viral stocks were titered by a plaque assay on Vero cells. Briefly, viral stocks were serially diluted (10-fold) in EMEM (Wisent) and 100 μl of each dilution was used to infect confluent monolayers of Vero cells in 1 ml of unsupplemented EMEM. At 2 hpi, infection media were removed and cells were overlaid with 1.2% carboxymethyl cellulose (Sigma-Aldrich) and 2% heat-inactivated FBS in EMEM for 4 d prior to fixation for 1 h with an equal volume of 10% formaldehyde. Monolayers were rinsed gently with distilled water and stained for 30 min with 0.1% crystal violet prior to counting plaques. ZIKVCDN was UV-inactivated by transferring 1 ml of ZIKV into one well of a six-well plate and exposing it to 3 J/cm2 of UV irradiation in a UVC 500 Crosslinker (Hoefer) as previously described (16). UV inactivation was confirmed by plaque assay.

C57BL/6 mice (wild-type) were purchased from Charles River Laboratories, the In Vivo Therapeutics Core at the Indiana University School of Medicine, or bred at McGill University. Infected mice were housed in biocontainment level 2, and all animal procedures were carried out in accordance with the Canadian Council on Animal Care and were approved by the McGill University Animal Care Committee or the Indiana University Institutional Animal Care and Use Committee. Six- to 12-wk-old mice of both sexes were used for all experiments.

ZIKVCDN (p.8 or p.9) or ZIKVBR (p.5 or p.6) were injected i.v. at 1 × 106 PFU. Mice injected with UV-inactivated ZIKV received a dose of inactivated virus equivalent to 1 × 106 PFU. Mock-infected mice were i.v. injected with an identical volume of mock-infected Vero cell culture supernatant (prepared in parallel to ZIKV stocks).

The following Abs were used in an appropriate combination of fluorochromes: B220 (clone RA3-6B2, BioLegend), Bcl-6 (clone K112-91, BD Biosciences), CD4 (clone GK1.5, BioLegend), CD8α (clone 53-6.7, BioLegend), CD11a (clone M17/4, BioLegend), CD19 (clone 1D3/CD19, BioLegend), CD21 (clone 7E9, BioLegend), CD23 (clone B3B4, BioLegend), CD49d (clone R1-2, BioLegend), CD86 (clone GL-1, BioLegend), CXCR5 (clone L138D7, BioLegend), Foxp3 (clone FJK-16s, eBioscience), GL7 (clone GL7, BioLegend), IFN-γ (clone XMG1.2, BioLegend), IgD (clone 11-26c.2a, BioLegend), IL-2 (clone JES6-5H4, BioLegend), IL-5 (clone TRFK5 BioLegend), IL-17A (clone TC11-18H10.1, BioLegend), PD-1 (clone 29F.1A12, BioLegend), T-bet (clone 4B10, BioLegend), TNF-α (clone MP6-XT22, BioLegend), and appropriate isotype controls.

Spleens were isolated and mechanically disrupted to generate single-cell suspensions. Erythrocytes were lysed with ammonium-chloride-potassium (ACK) buffer (0.15 M NH4Cl, 1 mM KHCO3, and 0.1mM Na2EDTA in dH2O [pH 7.2]), cells were incubated with TruStain FcX (anti-mouse CD16/CD32, clone 93, BioLegend) and stained with the indicated Abs, followed by fixation using IC (intracellular) fixation buffer (eBioscience). Intracellular staining for transcription factors was performed using the Foxp3/Transcription Factor Staining Buffer Set (eBioscience), as per the manufacturer’s instructions. Samples were analyzed with a BD LSRFortessa flow cytometer (BD Biosciences) and FlowJo software (Tree Star).

For ex vivo restimulation, spleens were harvested and processed as above. Erythrocytes were lysed with ACK buffer and samples were simulated for 5.5 h at 37°C with 5% CO2 in the presence of 1 μg per well of plate-bound anti-CD3ε Ab (clone 145-2C11, BioLegend) or media alone and brefeldin A (BioLegend). Anti-CD3ε was bound to the plate by incubating overnight in PBS at 4°C, and wells were washed with PBS prior to adding cells. Cells were then stained with surface Abs and fixed with IC fixation buffer (eBioscience), followed by intracellular staining for cytokines in Perm/Wash buffer (eBioscience). Samples were analyzed with a BD LSRFortessa flow cytometer (BD Biosciences) and FlowJo software (Tree Star).

Spleens from uninfected, ZIKVCDN-infected, and ZIKVBR-infected mice were cut in half, submerged in optimal cutting temperature compound, flash-frozen with liquid nitrogen vapor, and stored at −20°C. The tissues were sectioned into 5-μm slices using a cryostat. Spleen sections were fixed in an ice-cold acetone solution (75% acetone and 25% ethanol) for 5 min. The sections were then placed in PBS and stained in a dark, humid chamber at room temperature. Sections were blocked for 1 h in 2% BSA with Fc Block (1:100, clone 2.4G2) and then stained in 2% BSA with CD4 (clone GK1.5, eBioscience), GL7 (clone 81E2, BD Pharmingen), and IgD (clone 11-26c, eBioscience) for 1 h. The slides were then washed in PBS for 7 min twice while agitated in the dark and mounted using ProLong diamond antifade (Invitrogen). Images were taken using a Zeiss LSM 780 laser scanning confocal microscope and analyzed using Fiji software. Using the Zeiss tiling software, 4-by-4 images were taken to count GCs. Five to 13 tiled images were taken for each spleen, and the average number of GL7+ GCs per image for each spleen was graphed.

To extract serum, blood was collected via cardiac puncture and centrifuged at 7000 rpm for 10 min at 4°C. Serum was diluted 25-, 50-, 100-, 250-, and 500-fold in EMEM, or serially diluted 10-fold in EMEM, and mixed with the corresponding ZIKV isolate. One hundred microliters of each serum/virus mixture was used to infect confluent monolayers of Vero cells in 1 ml of unsupplemented EMEM. Each six-well plate included one virus-only control well. At 2 hpi, infection media were removed, and cells were overlaid with 1.2% carboxymethyl cellulose (Sigma-Aldrich) and 2% heat-inactivated FBS in EMEM for 4 d prior to fixation for 1 h with an equal volume of 10% formaldehyde. Monolayers were rinsed gently with distilled water and stained for 30 min with 0.1% crystal violet prior to counting plaques. Percent neutralization was calculated as: % neutralization = [(no. plaques in no-serum well − no. plaques in indicated dilution well)/no. plaques in no-serum well] × 100%.

To measure virus-specific IgG levels, Thermo Scientific MaxiSorp plates (Thermo Fisher Scientific) were coated with 1 × 105 PFU of heat-inactivated (30 min at 56°C) ZIKVCDN or ZIKVBR overnight at 4°C. Following incubation, plates were washed twice with PBS–0.1% Tween 20 and blocked for 2 h in 5% milk diluted in PBS–0.1% Tween 20 at room temperature. Serum was collected from naive or infected mice as described above and diluted in 5% milk PBS–0.1% Tween 20 and incubated for 1 h at room temperature (serum from infected mice was incubated in wells coated with the matching virus isolate). Wells were washed twice with PBS–0.1% Tween 20. For avidity determination, various concentrations of ammonium thiocyanate (0.15–4 M range) were added to each well and incubated for 15 min prior to washing three times with PBS–0.1% Tween 20. Wells were then incubated with HRP-conjugated donkey anti-mouse IgG Ab (Jackson ImmunoResearch Laboratories, 1:20,000 dilution in 5% milk PBS–Tween 20 solution) for 1 h at room temperature followed by washing three times in PBS–0.1% Tween 20. Plates were reacted with tetramethylbenzidine substrate and the reaction was stopped with 4 N sulfuric acid prior to being read at 450 nm using an accuSkan FC plate reader.

Data were analyzed using GraphPad Prism 8 or 9 software. Specific tests for determining statistical significance are indicated in the figure legends. A p value <0.05 was considered statistically significant.

To begin our analysis of the CD4 T cell response to each ZIKV isolate, we compared changes in the total and Ag-experienced CD4 T cell populations in the spleen at 7 dpi. Ag-experienced CD4 T cells were tracked using cell surface expression of CD11a and CD49d, previously established surrogate markers of Ag experience in multiple infection models that we have validated for tracking the global CD4 T cell response to ZIKV infection (16, 21, 23, 24). Consistent with our previous results, the total number of cells in the spleen was significantly higher following infection with ZIKVCDN than ZIKVBR (Fig. 1A). Although ZIKVBR-infected mice had a significantly higher frequency of CD4 T cells, there was no significant difference in the number of CD4 T cells in the spleen between the two infected groups (Fig. 1B–D). This difference in frequency without a corresponding increase in numbers is likely due to the differential CD8 T cell responses induced by these two infections, as we described previously (21). Similar to our previous observations regarding the splenic Ag-experienced CD8 T cell response, we observed a significant reduction in both the frequency and number of CD11a+CD49d+ CD4 T cells responding to infection with ZIKVBR in the spleen (Fig. 1B, 1E, 1F), which contrasted to what we previously observed in the blood (21). In addition, the frequency and number of CD11a+CD49d+ CD4 T cells expressing the Th1 transcription factor T-bet and the expression of T-bet on a per cell basis (as measured by geometric mean fluorescence intensity [gMFI]) were significantly lower following ZIKVBR infection (Fig. 1G–J). Taken together, these data demonstrate that ZIKVCDN induces a stronger CD11a+CD49d+ Ag-experienced CD4 T cell response than does ZIKVBR, and that those CD4 T cells that are responding to ZIKVBR infection are less polarized toward the Th1 subset.

FIGURE 1.

The Ag-experienced and Th1 CD4 T cell responses to ZIKVBR are reduced compared with ZIKVCDN infection. (A) Overall spleen cellularity following ZIKVCDN or ZIKVBR infection. (B) Gating strategy and representative plots to identify CD4 T cells and Ag-experienced, CD11a+CD49d+ CD4 T cells at 7 dpi in the spleen. (C and D) Frequency (C) and number (D) of CD4 T cells in the spleen at 7 dpi with ZIKVCDN or ZIKVBR. (E and F) Frequency (E) and number (F) of Ag-experienced CD11a+CD49d+ CD4 T cells in the spleen at 7 dpi with ZIKVCDN or ZIKVBR. (GJ) Representative histogram (G), frequency (H), number (I), and geometric mean fluorescence intensity (gMFI) (J) of T-bet expression in Ag-experienced CD11a+CD49d+ CD4 T cells in the spleen at 7 dpi with ZIKVCDN or ZIKVBR. Shaded gray curve in (G) represents isotype control staining, and line on histogram indicates the gating strategy to identify T-bet+ cells. Data are representative of three independent experiments with n = 3–4 mice per group in each experiment and are shown as mean ± SEM. Data were analyzed with a two-tailed, unpaired Student t test. *p < 0.05, **p < 0.01.

FIGURE 1.

The Ag-experienced and Th1 CD4 T cell responses to ZIKVBR are reduced compared with ZIKVCDN infection. (A) Overall spleen cellularity following ZIKVCDN or ZIKVBR infection. (B) Gating strategy and representative plots to identify CD4 T cells and Ag-experienced, CD11a+CD49d+ CD4 T cells at 7 dpi in the spleen. (C and D) Frequency (C) and number (D) of CD4 T cells in the spleen at 7 dpi with ZIKVCDN or ZIKVBR. (E and F) Frequency (E) and number (F) of Ag-experienced CD11a+CD49d+ CD4 T cells in the spleen at 7 dpi with ZIKVCDN or ZIKVBR. (GJ) Representative histogram (G), frequency (H), number (I), and geometric mean fluorescence intensity (gMFI) (J) of T-bet expression in Ag-experienced CD11a+CD49d+ CD4 T cells in the spleen at 7 dpi with ZIKVCDN or ZIKVBR. Shaded gray curve in (G) represents isotype control staining, and line on histogram indicates the gating strategy to identify T-bet+ cells. Data are representative of three independent experiments with n = 3–4 mice per group in each experiment and are shown as mean ± SEM. Data were analyzed with a two-tailed, unpaired Student t test. *p < 0.05, **p < 0.01.

Close modal

An important function of Th1-polarized CD4 T cells is the production of effector cytokines such as IFN-γ, TNF-α, and IL-2 to support the antiviral immune response (9). Thus, to investigate the functionality of responding Th1 cells at 7 dpi with each ZIKV isolate, we analyzed cytokine expression following restimulation with plate-bound anti-CD3ε Ab. Similar to our previous observations (16), and consistent with their Th1 polarization, Ag-experienced CD4 T cells produced Th1 cytokines (IFN-γ, TNF-α, and IL-2) following restimulation (Fig. 2). The frequency and number of IFN-γ+ and TNF-α+ Ag-experienced CD4 T cells were significantly higher following infection with ZIKVCDN compared with infection with ZIKVBR, although among IFN-γ+ or TNF-α+ cells, their expression on a per cell basis was similar between the two groups (Fig. 2A–H). Although the frequency of IL-2+ Ag-experienced CD4 T cells and the gMFI of IL-2 staining were similar between the two groups, we observed a significant reduction of IL-2+ Ag-experienced CD4 T cells following ZIKVBR infection (Fig. 2I–L). These data are consistent with the reduced induction of T-bet expression during ZIKVBR infection (Fig. 1G–J) and confirm a more robust Th1 response following ZIKVCDN infection. To determine whether the reduced polarization to the Th1 subset following ZIKVBR infection was due to polarization toward other Th subsets, we also analyzed expression of IL-5 (Th2) and IL-17A (Th17) following restimulation. Although in some samples we detected small frequencies of IL-5– and IL-17A–producing cells (<1% of CD11a+CD49d+), we observed no difference between ZIKVCDN and ZIKVBR infection (Supplemental Fig. 1). Thus, ZIKVBR infection induces a CD4 T cell response that is less polarized to the Th1 subset and is less capable of producing Th1 cytokines without inducing polarization toward other Th subsets.

FIGURE 2.

Ag-experienced CD4 T cells produce less IFN-γ and TNF-α following ZIKVBR infection. Mice were infected with ZIKVCDN or ZIKVBR and spleens were harvested at 7 dpi. Splenocytes were restimulated with anti-CD3ε Ab or media alone prior to intracellular staining for cytokines. (AD) Representative plots (A), frequency (B), number (C), and gMFI (D) of IFN-γ production by Ag-experienced CD11a+CD49d+ CD4 T cells. (EH) Same as (A)–(D) but for TNF-α production. (IL) Same as (A)–(D) but for IL-2 production. Data are representative of three independent experiments with n = 3 mice per group in each experiment and are shown as mean ± SEM. Data were analyzed with a two-tailed, unpaired Student t test. *p < 0.05, **p < 0.01.

FIGURE 2.

Ag-experienced CD4 T cells produce less IFN-γ and TNF-α following ZIKVBR infection. Mice were infected with ZIKVCDN or ZIKVBR and spleens were harvested at 7 dpi. Splenocytes were restimulated with anti-CD3ε Ab or media alone prior to intracellular staining for cytokines. (AD) Representative plots (A), frequency (B), number (C), and gMFI (D) of IFN-γ production by Ag-experienced CD11a+CD49d+ CD4 T cells. (EH) Same as (A)–(D) but for TNF-α production. (IL) Same as (A)–(D) but for IL-2 production. Data are representative of three independent experiments with n = 3 mice per group in each experiment and are shown as mean ± SEM. Data were analyzed with a two-tailed, unpaired Student t test. *p < 0.05, **p < 0.01.

Close modal

CD4 T cell priming events involve a fate decision-making process directing CD4 T cells toward either effector (e.g., Th1) differentiation or Tfh cell differentiation (11). While the signals that influence this fate decision remain incompletely understood, which subset is favored may depend on the balance of signals that are present. For example, whereas IL-2 or IL-12 signaling and T-bet expression have been shown to favor Th1 differentiation, IL-6 stimulation and strong TCR signaling promote Tfh cell responses (11, 25, 26). To address whether differences exist between the Tfh cell response to ZIKVCDN and ZIKVBR, mice were infected with either isolate, or injected with UV-inactivated ZIKVCDN (UV-ZIKV) as a control, and the Tfh cell response was analyzed in the spleen at 10, 14, and 21 dpi. We observed a small population of PD-1hiCXCR5+ Tfh cells that remained comparable across all time points in mice injected with UV-ZIKV or ZIKVCDN-infected mice (Fig. 3A–I). However, following ZIKVBR infection we observed a significantly higher frequency and number of PD-1hiCXCR5+ Tfh cells across all time points tested (Fig. 3A–I). To confirm that these cells represented a bona fide Tfh cell response, we analyzed the PD-1hiCXCR5+ Tfh cells for expression of the transcription factor Bcl-6, a master regulator of Tfh function (11). As expected, Tfh cells from ZIKVBR-infected mice expressed significantly more Bcl-6 on a per cell basis than did non-Tfh CD4 T cells from the same mouse at all time points tested (Fig. 3J–L), confirming that ZIKVBR induces a prototypical Tfh cell response. We conducted the same analysis within ZIKVCDN-infected mice and found that the small frequency of Tfh cells present within these mice expressed significantly more Bcl-6 than did their non-Tfh CD4 T cell counterparts (Supplemental Fig. 2). Nonetheless, at 10 dpi the expression of Bcl-6 (as measured by gMFI) was higher among Tfh cells from ZIKVBR-infected mice than ZIKVCDN-infected mice (Fig. 3J, Supplemental Fig. 2). These data indicate that infection with ZIKVBR promotes a more robust Tfh response than infection with ZIKVCDN.

FIGURE 3.

ZIKVBR infection induces enhanced and sustained Tfh cell responses. Mice were injected with UV-ZIKV or infected with ZIKVCDN or ZIKVBR, and PD-1hiCXCR5+ Tfh cell responses were analyzed. (AC) Representative histograms (A), frequency (B), and number (C) of PD-1hiCXCR5+ Tfh cells in the spleen at 10 dpi. (DF) Same as (A)–(D) but at 14 dpi. (GI) Same as (A)–(D) but at 21 dpi. (J) Representative histogram and gMFI of Bcl-6 expression in PD-1hiCXCR5+ Tfh cells and CXCR5 non–Tfh cells at 10 dpi with ZIKVBR. (K and L) Same as (J) but for 14 and 21 dpi, respectively. Data are representative of two independent experiments with n = 3 mice per group in each experiment and are shown as mean ± SEM. Data in (B), (C), (E), (F), (H), and (I) were analyzed by one-way ANOVA with a Tukey posttest of multiple comparisons. Data in (J)–(L) were analyzed by a two-tailed, paired Student t test. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

FIGURE 3.

ZIKVBR infection induces enhanced and sustained Tfh cell responses. Mice were injected with UV-ZIKV or infected with ZIKVCDN or ZIKVBR, and PD-1hiCXCR5+ Tfh cell responses were analyzed. (AC) Representative histograms (A), frequency (B), and number (C) of PD-1hiCXCR5+ Tfh cells in the spleen at 10 dpi. (DF) Same as (A)–(D) but at 14 dpi. (GI) Same as (A)–(D) but at 21 dpi. (J) Representative histogram and gMFI of Bcl-6 expression in PD-1hiCXCR5+ Tfh cells and CXCR5 non–Tfh cells at 10 dpi with ZIKVBR. (K and L) Same as (J) but for 14 and 21 dpi, respectively. Data are representative of two independent experiments with n = 3 mice per group in each experiment and are shown as mean ± SEM. Data in (B), (C), (E), (F), (H), and (I) were analyzed by one-way ANOVA with a Tukey posttest of multiple comparisons. Data in (J)–(L) were analyzed by a two-tailed, paired Student t test. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

Close modal

Within the Tfh cell population, a subset of Foxp3+ T follicular regulatory (Tfr) cells, which develop from T regulatory cells, may also be induced following infection to regulate the Tfh and GC B cell responses (27). To determine whether each ZIKV isolate differentially induced Tfr cell responses, we analyzed Foxp3 expression among the Tfh subset at 10, 14, and 21 dpi with either ZIKV isolate. Although we observed a trend toward a higher number of Tfr cells at 10 and 21 dpi, and significantly more Tfr cells at 14 dpi with ZIKVBR, this population represented a much smaller proportion of the total Tfh cell population within the ZIKVBR-infected mice due to the larger increase in overall Tfh cell numbers in those mice (Supplemental Fig. 3). In contrast, the frequency of Tfr cells among CXCR5+PD-1+ cells in ZIKVCDN-infected mice showed a trending increase at 10 dpi and was significantly higher at 14 and 21 dpi (Supplemental Fig. 3). Thus, the enhanced Tfh cell response to ZIKVBR infection is not associated with a significant change in Tfr numbers at most of the time points examined, suggesting that changes in the Tfr population are unlikely to be responsible for the modulation of Tfh populations in this model.

One of the primary functions of Tfh cells is to promote GC reactions in secondary lymphoid organs, which function to mediate GC B cell isotype class switching and affinity maturation (13, 14). Together, these processes generate B cells with higher affinity for cognate Ag, which in turn leads to production of higher affinity Abs against the pathogen (13, 14). Because we observed a more robust Tfh cell response following ZIKVBR infection, we next analyzed the GC B cell response to UV-ZIKV, ZIKVCDN, or ZIKVBR at 10, 14, and 21 dpi. Similar to the Tfh cell response, we detected only small frequencies of IgDloGL7+ GC B cells at all time points following injection with UV-ZIKV or infection with ZIKVCDN (Fig. 4A–I). Both the frequency and number of IgDloGL7+ GC B cells were significantly increased following infection with ZIKVBR across all time points analyzed (Fig. 4A–I). In addition, we analyzed splenic tissue sections for the presence of GL7+ GCs by confocal microscopy. We observed similar numbers of GCs in uninfected and ZIKVCDN-infected mice, whereas the average number of GCs per spleen section was significantly higher in ZIKVBR-infected mice (Fig. 4J, 4K). Taken together, these data demonstrate that infection with ZIKVBR leads to enhanced generation of GCs in the spleen and a more robust GC B cell response than with ZIKVCDN.

FIGURE 4.

GC formation and GC B cell response are enhanced following ZIKVBR infection. Mice were injected with UV-ZIKV or infected with ZIKVCDN or ZIKVBR, and IgDloGL7+ GC B cell responses were analyzed. (AC) Representative histograms (A), frequency (B), and number (C) of IgDloGL7+ GC B cells in the spleen at 10 dpi. (DF) Same as (A)–(D) but at 14 dpi. (GI) Same as (A)–(D) but at 21 dpi. (J) Representative confocal microscopy images of spleen sections from uninfected, ZIKVCDN-infected, and ZIKVBR-infected mice 10 dpi. CD4 (CD4 T cells) is in red, IgD (naive B cells) is in green, and GL7 (GC B cells) is in blue. Scale bars, 100 μm. (K) Average number of GL7+ GCs counted per section. Data in (A)–(I) are representative of two independent experiments with n = 3 mice per group in each experiment and are shown as mean ± SEM. Data in (J) and (K) are pooled from three independent experiments with n = 1 uninfected mouse per experiment and n = 2 mice per infection group in each experiment, and are shown as mean ± SEM. Data in (B), (C), (E), (F), (H), (I), and (K) were analyzed by one-way ANOVA with a Tukey posttest of multiple comparisons. **p < 0.01, ***p < 0.001, ****p < 0.0001.

FIGURE 4.

GC formation and GC B cell response are enhanced following ZIKVBR infection. Mice were injected with UV-ZIKV or infected with ZIKVCDN or ZIKVBR, and IgDloGL7+ GC B cell responses were analyzed. (AC) Representative histograms (A), frequency (B), and number (C) of IgDloGL7+ GC B cells in the spleen at 10 dpi. (DF) Same as (A)–(D) but at 14 dpi. (GI) Same as (A)–(D) but at 21 dpi. (J) Representative confocal microscopy images of spleen sections from uninfected, ZIKVCDN-infected, and ZIKVBR-infected mice 10 dpi. CD4 (CD4 T cells) is in red, IgD (naive B cells) is in green, and GL7 (GC B cells) is in blue. Scale bars, 100 μm. (K) Average number of GL7+ GCs counted per section. Data in (A)–(I) are representative of two independent experiments with n = 3 mice per group in each experiment and are shown as mean ± SEM. Data in (J) and (K) are pooled from three independent experiments with n = 1 uninfected mouse per experiment and n = 2 mice per infection group in each experiment, and are shown as mean ± SEM. Data in (B), (C), (E), (F), (H), (I), and (K) were analyzed by one-way ANOVA with a Tukey posttest of multiple comparisons. **p < 0.01, ***p < 0.001, ****p < 0.0001.

Close modal

One of the primary functions of Abs is to neutralize virus particles, thereby preventing further spread to uninfected cells. Thus, we sought to determine whether the generation of neutralizing Abs differed between infection with each ZIKV isolate. To address this question, we conducted plaque reduction neutralization tests to analyze the capacity of serum from UV-ZIKV–, ZIKVCDN-, and ZIKVBR-infected mice at 10 and 21 dpi to neutralize virus and prevent infection of Vero cell monolayers. Serum was serially diluted and incubated with the same isolate with which the mice had been infected (i.e., serum from ZIKVCDN-infected mice was incubated with ZIKVCDN virus and serum from ZIKVBR-infected mice was incubated with ZIKVBR virus) prior to incubation with Vero cells for plaque assay. Each serum dilution was compared with a no serum control well to calculate the percentage neutralization. Serum from UV-ZIKV–injected mice was unable to neutralize ZIKVCDN at any serum dilution (data not shown), indicating that there was no non-specific neutralization within mouse serum. Serum from ZIKVCDN-infected mice neutralized virus only at low dilutions at 10 dpi, which was quickly lost at dilutions of 100-fold or higher (Fig. 5A). Strikingly, serum from ZIKVBR-infected mice at 10 dpi showed high neutralization across the same range of dilutions, and our data indicate that neutralization capacity was only lost after being diluted 10,000-fold (Fig. 5A, Supplemental Fig. 4). This pattern was maintained at 21 dpi, with serum from ZIKVCDN-infected mice showing only minor increases in neutralization capacity, and serum from ZIKVBR-infected mice retaining robust neutralization capacity (Fig. 5B). Thus, ZIKVBR induces an Ab response that potently neutralizes viral infection in cell culture.

FIGURE 5.

ZIKVBR infection induces a more potent neutralizing Ab response. (A and B) Plaque reduction neutralization tests were performed using serially diluted (25- to 500-fold) serum from ZIKVCDN- and ZIKVBR-infected mice (A) at 10 dpi and (B) at 21 dpi to test each sample’s capacity to neutralize infection of Vero cell monolayers by the same ZIKV isolate with which the mice had been infected. Data are expressed as the percent neutralization when compared with a no serum control well. Data were pooled from two independent experiments with n = 3 mice per group and are shown as mean ± SEM. (C) ELISA analysis of virus-specific IgG. Serum was isolated from ZIKVCDN or ZIKVBR at 21 dpi or from naive mice and reacted with plate-bound heat-killed corresponding virus isolate. Data are representative of two experiments with five infected mice per group in each experiment. Naive sera were pooled from more than five uninfected mice and are shown as mean ± SEM. (D) IC50 of ammonium thiocyanate needed to remove IgG binding. Diluted sera from ZIKV-infected mice were incubated with increasing concentrations of ammonium thiocyanate, and percent binding when compared with untreated samples was used to calculate IC50 for each individual mouse. Data are representative of two experiments with five infected mice per group in each experiment and are shown as mean ± SEM. Data in (A), (B), and (D) were analyzed by a two-tailed, unpaired Student t test. ***p < 0.001, ****p < 0.0001.

FIGURE 5.

ZIKVBR infection induces a more potent neutralizing Ab response. (A and B) Plaque reduction neutralization tests were performed using serially diluted (25- to 500-fold) serum from ZIKVCDN- and ZIKVBR-infected mice (A) at 10 dpi and (B) at 21 dpi to test each sample’s capacity to neutralize infection of Vero cell monolayers by the same ZIKV isolate with which the mice had been infected. Data are expressed as the percent neutralization when compared with a no serum control well. Data were pooled from two independent experiments with n = 3 mice per group and are shown as mean ± SEM. (C) ELISA analysis of virus-specific IgG. Serum was isolated from ZIKVCDN or ZIKVBR at 21 dpi or from naive mice and reacted with plate-bound heat-killed corresponding virus isolate. Data are representative of two experiments with five infected mice per group in each experiment. Naive sera were pooled from more than five uninfected mice and are shown as mean ± SEM. (D) IC50 of ammonium thiocyanate needed to remove IgG binding. Diluted sera from ZIKV-infected mice were incubated with increasing concentrations of ammonium thiocyanate, and percent binding when compared with untreated samples was used to calculate IC50 for each individual mouse. Data are representative of two experiments with five infected mice per group in each experiment and are shown as mean ± SEM. Data in (A), (B), and (D) were analyzed by a two-tailed, unpaired Student t test. ***p < 0.001, ****p < 0.0001.

Close modal

Increased neutralization by Abs from ZIKVBR-infected mice could be due to a difference in either Ab quantity, quality, or both. To address this question, we harvested serum from naive mice (pooled serum from naive mice was used for these experiments) or on 21 dpi with either ZIKVCDN or ZIKVBR and measured ZIKV-specific IgG levels via ELISA. We observed that despite a reduced neutralization capacity, ZIKVCDN infection induces a stronger virus-specific IgG response at 21 dpi (Fig. 5C). Next, we tested the avidity of the Ab response via ELISA and thiocyanate elution (28). We observed that the concentration of thiocyanate required to remove 50% of IgG binding (IC50, also referred to as affinity index) was significantly greater for ZIKVBR infection–induced Abs, establishing that these Abs are of higher avidity (Fig. 5D). Taken together, this suggests that the stronger Tfh and GC B cell responses induced following ZIKVBR infection leads to a higher avidity Ab response that is correlated with a greatly enhanced neutralization capacity despite lower overall production of virus-specific IgG.

The production of higher levels of low-avidity virus-specific Abs following ZIKVCDN infection in the absence of a robust Tfh response suggests that this viral isolate may favor the induction of an extrafollicular response. This response involves the early production of lower avidity Abs and the activation of non-GC B cell subsets such as marginal zone B (MZB) cells, which can rapidly produce IgM and IgG Abs in response to pathogens, including viruses (2933). At 7 dpi, we observed that infection with ZIKVCDN led to a significant increase in the activation of MZB cells, as measured by their surface expression of the costimulatory molecule CD86, compared with mock-infected or ZIKVBR-infected mice (Fig. 6A, 6B). In addition, we observed that ZIKVCDN infection, but not ZIKVBR infection, led to the induction of a virus-specific IgG response at this early time point (Fig. 6C). Taken together, this suggests that infection with ZIKVCDN primarily drives an extrafollicular B cell response that is associated with greater production of virus-specific IgG Abs with lower avidity and less capacity to neutralize infection.

FIGURE 6.

ZIKVCDN infection induces enhanced activation of MZB cells and early virus-specific Ab production. (A and B) Representative histogram (A) and gMFI (B) of CD86 expression on MZB cells (gated on B220+CD19+CD21+CD23lo cells) at 7 dpi with either ZIKVCDN, ZIKVBR, or mock infection. Data are representative of two experiments with five infected mice per group in each experiment and are shown as mean ± SEM. (C) ELISA analysis of virus-specific IgG. Serum was isolated from ZIKVCDN or ZIKVBR at 7 dpi or from naive mice and reacted with plate-bound heat-killed corresponding virus isolate. Data are representative of two experiments with five infected mice per group in each experiment. Naive sera were pooled from more than five uninfected mice and are shown as mean ± SEM. Data in (B) were analyzed by one-way ANOVA with a Tukey posttest of multiple comparisons. ****p < 0.0001.

FIGURE 6.

ZIKVCDN infection induces enhanced activation of MZB cells and early virus-specific Ab production. (A and B) Representative histogram (A) and gMFI (B) of CD86 expression on MZB cells (gated on B220+CD19+CD21+CD23lo cells) at 7 dpi with either ZIKVCDN, ZIKVBR, or mock infection. Data are representative of two experiments with five infected mice per group in each experiment and are shown as mean ± SEM. (C) ELISA analysis of virus-specific IgG. Serum was isolated from ZIKVCDN or ZIKVBR at 7 dpi or from naive mice and reacted with plate-bound heat-killed corresponding virus isolate. Data are representative of two experiments with five infected mice per group in each experiment. Naive sera were pooled from more than five uninfected mice and are shown as mean ± SEM. Data in (B) were analyzed by one-way ANOVA with a Tukey posttest of multiple comparisons. ****p < 0.0001.

Close modal

Since the South and Central American outbreak, many studies have sought to improve our understanding of the immune response to ZIKV infection. However, few of these studies have directly compared the immune response induced by distinct ZIKV isolates, an approach that may help further our understanding of how ZIKV has changed in its interactions with the host immune system as it acquired epidemic capacity. In this study, we have conducted an analysis of CD4 T cell differentiation following infection with a pre-epidemic isolate (ZIKVCDN) or an epidemic isolate (ZIKVBR). Our data demonstrate that although ZIKVCDN induces more robust Th1 and global Ag-experienced CD4 T cell responses than does ZIKVBR at 7 dpi, there is a stronger Tfh cell response to ZIKVBR at 10 dpi, which persists through to 21 dpi. This Tfh cell response correlates with increased GC formation and increased generation of GC B cells, which in turn leads to a more potent neutralizing Ab response in the serum and correlates with the induction of a higher avidity virus-specific Ab response. Conversely, ZIKVCDN induces the production of more virus-specific IgG at various time points postinfection, even as early as 7 dpi. However, these Abs are of lower avidity, have poor neutralization capacity, and correlate with the stronger activation of extrafollicular B cells such as MZB cells. Taken together, these data suggest that although ZIKVBR may have evolved to subvert antiviral Th1 CD4 T cell responses and CD8 T cell responses that develop earlier following infection (21), this ultimately results in stronger GC B cell–mediated immunity that develops later in the infection and likely prevents the establishment of chronic infection. One caveat that remains is that our study centered on investigating the total Ag-experienced CD4 T cells using surrogate markers rather than defined Ag-specific CD4 T cells using ZIKV-specific peptides or tetramers. Our findings regarding a change of polarization within the total pool of virus-activated CD4 T cells provide an overview of the virus-specific CD4 T cell response to these two ZIKV isolates. However, it will be of interest to determine whether these two isolates induce CD4 T cells that target different viral epitopes, as we have recently observed regarding the CD8 T cell response following infection with these ZIKV isolates, and this will be the focus of future investigation (21).

Impaired CD4 T cell immunity following ZIKVBR infection could have a broad impact on the host’s capacity to respond to infection. The cytokines produced by Th1 CD4 T cells play an important role in activating other immune cells and enhancing the antiviral immune response (9). If the reduced presence of Th1 CD4 T cells impacted DC licensing as well, it could in turn impact the generation of protective CD8 T cell immunity, which would be consistent with our previous observations of an impaired CD8 T cell response to ZIKVBR infection, which correlated with delayed viral clearance in some tissues (21). CD4 T cells clearly play an important role in protection against ZIKV infection in susceptible mouse strains, as their depletion leads to more severe paralysis, higher viral loads, and increased mortality (18, 19). In addition, transferring memory CD4 T cells from ZIKV-immune to naive IFNAR KO mice protected these mice from lethal ZIKV challenge (18, 19). The fact that this protection was lost in the absence of IFN-γ receptor signaling or B cells implies an important role for both Th1 and Tfh-dependent B cell responses in controlling ZIKV infection (19).

Only a few studies have directly analyzed Tfh and GC B cell responses to ZIKV infection. In LysMCre+Ifnarfl/fl mice, ZIKV infection induces Tfh and GC B cell responses that are important for IgG isotype class switching (17). Liang et al. (20) similarly detected a robust “Th1-like” (IFN-γ+) Tfh cell response following ZIKV infection in mice that had been treated with an IFNAR-blocking Ab prior to infection. Similar to our observations in the present study, this Tfh response was readily detectable at 10 and 14 dpi, and correlated with generation of GC B cells, IgG2a production, and neutralizing Ab responses (20). Our previous work has identified that type I IFN production is reduced following ZIKVBR infection, consistent with the role that Liang et al. (20) identified for IFNAR signaling in modulating the Tfh cell response to ZIKV (21). Whether IFNAR signaling directly or indirectly influences Tfh cell differentiation following ZIKV infection remains an intriguing question for future study. We also demonstrated previously that ZIKVBR remains detectable by plaque assay in the spleen and kidney at 7 dpi but is cleared by 14 dpi (21). Thus, the enhanced Tfh and GC B cell responses we observe, as well as the resulting increase in Ab avidity and neutralization capacity following ZIKVBR infection, could provide a potential mechanism for the eventual clearance of this virus.

Our observation of a balance between the induction of Th1- or Tfh-mediated immunity following infection with ZIKVCDN or ZIKVBR is similar to observations that have been made following Salmonella enterica and Toxoplasma gondii infection (25, 26). Following S. enterica infection, IL-12 enhances Th1 differentiation at the expense of the Tfh cell response, which is dependent on IL-12 receptor signaling and T-bet (26). Furthermore, this contributes to the inhibition of GC formation observed following S. enterica infection. During T. gondii infection, although IL-12 initially induces genes that may contribute to both Th1 and Tfh cell phenotypes, T-bet and IFN-γ antagonize Tfh cell responses and promote Th1 differentiation, which in turn limits GC formation and Ab responses (25). Thus, the robust Th1 response to ZIKVCDN could be limiting the development of Tfh cell and GC B cell responses, whereas the lack of a Th1 response and Th1 cytokines may have the opposite impact following ZIKVBR infection. However, Tfh cell differentiation does not represent a default differentiation pathway and requires more than an absence of conditions or cytokines that promote Th1 differentiation for their generation (11). For example, Tfh cell responses are promoted by strong TCR signals, such as those that occur following high-affinity TCR–peptide/MHC class II interactions, or by repeated stimulation (11). Fahey et al. (34) demonstrated that although both LCMV Armstrong and clone 13 initially drive comparable Th1 responses, over time as LCMV clone 13 persists, the CD4 T cell response becomes increasingly Tfh cell dominant. This suggests that delayed viral clearance (longer Ag exposure) following ZIKVBR infection could contribute to the more robust Tfh cell response we observe. Interestingly, infection with ZIKVCDN yielded the production of higher levels of virus-specific Abs, but these Abs had significantly lower avidity for ZIKV Ags and lower neutralization capacity. This was associated with stronger activation of MZB cells, suggesting that this virus isolate primarily activates extrafollicular B cell responses. The signals that determine the activation of extrafollicular and follicular B cell responses following ZIKV infection remain unknown and will be the focus of future investigation. In addition, future studies will seek to determine the cytokine-dependent and -independent signals that impact the Th1/Tfh cell balance following ZIKV infection. Nonetheless, the ability to compare the Th1 and Tfh cell responses between two isolates of the same virus suggests that ZIKV could provide a useful model for dissecting other signals that promote or restrain Tfh cell differentiation.

In conclusion, our data demonstrate that while ZIKVBR infection induced a weaker Th1 response than did infection with ZIKVCDN, this contrasts with more robust Tfh and GC B cell responses. As a result, ZIKVBR infection generates a potent neutralizing Ab response, which persists as late as 21 dpi. In contrast, infection with ZIKVCDN induces only a minor neutralizing Ab response despite producing high levels of virus-specific IgG Abs. Taken together, these data suggest that ZIKVBR may have evolved to subvert Th1 CD4 T cell responses, but at the cost of promoting B cell–mediated immunity, which develops later in the infection. However, the difference in timing of these responses could provide a crucial window of viremia within which ZIKVBR continues to circulate, which may have important implications during ZIKV infection, including potentially increasing the transmission window of the virus and increased risk of pathogenesis in the context of pregnancy.

We thank David Safronetz and Gary Kobinger at the National Microbiology Laboratory and Public Health Agency of Canada for providing ZIKVCDN, and Mauro Teixeira (Universidade Federal de Minas Gerais) for providing ZIKVBR. We also thank Steven Varga (University of Iowa) for providing Vero cells. Thank you to Noah Butler, Alexander Dent, Stefanie Valbon and Miranda Yu for discussion and feedback. We thank the members of the Indiana University Melvin and Bren Simon Cancer Center Flow Cytometry Resource Facility for outstanding technical support.

The Richer Lab was supported by Canadian Institutes of Health Research Grants PJT-152903 and PJT-162212; Natural Sciences and Engineering Research Council Grant RGPIN-2016-04713; Fonds de Recherche du Québec–Santé Grant 32807; the Canada Foundation for Innovation; and by funds from Indiana University. R.D.P is supported by the Frederick Banting and Charles Best Canada Graduate Scholarships–Doctoral Award from the Canadian Institutes of Health Research. The Indiana University Melvin and Bren Simon Comprehensive Cancer Center Flow Cytometry Resource Facility is funded in part by National Institutes of Health/National Cancer Institute Grant P30 CA082709 and National Institute of Diabetes and Digestive and Kidney Diseases Grant U54 DK106846. The Flow Cytometry Resource Facility is supported in part by National Institutes of Health Instrumentation Grant 1S10D012270.

The online version of this article contains supplemental material.

Abbreviations used in this article:

Bcl-6

B cell lymphoma-6

DC

dendritic cell

dpi

day postinfection

EMEM

Eagle’s MEM

GC

germinal center

gMFI

geometric mean fluorescence intensity

hpi

hour postinfection

IFNAR

IFN-α/β receptor

KO

knockout

MZB cell

marginal zone B cell

Tfh

T follicular helper

Tfr

T follicular regulatory

UV-ZIKV

UV-inactivated ZIKVCDN

ZIKV

Zika virus

1.
Dick
G. W.
1952
.
Zika virus. II. Pathogenicity and physical properties.
Trans. R. Soc. Trop. Med. Hyg.
46
:
521
534
.
2.
Dick
G. W.
,
S. F.
Kitchen
,
A. J.
Haddow
.
1952
.
Zika virus. I. Isolations and serological specificity.
Trans. R. Soc. Trop. Med. Hyg.
46
:
509
520
.
3.
Rajah
M. M.
,
R. D.
Pardy
,
S. A.
Condotta
,
M. J.
Richer
,
S. M.
Sagan
.
2016
.
Zika virus: emergence, phylogenetics, challenges, and opportunities.
ACS Infect. Dis.
2
:
763
772
.
4.
Duffy
M. R.
,
T. H.
Chen
,
W. T.
Hancock
,
A. M.
Powers
,
J. L.
Kool
,
R. S.
Lanciotti
,
M.
Pretrick
,
M.
Marfel
,
S.
Holzbauer
,
C.
Dubray
, et al
2009
.
Zika virus outbreak on Yap Island, Federated States of Micronesia.
N. Engl. J. Med.
360
:
2536
2543
.
5.
Oehler
E.
,
L.
Watrin
,
P.
Larre
,
I.
Leparc-Goffart
,
S.
Lastere
,
F.
Valour
,
L.
Baudouin
,
H.
Mallet
,
D.
Musso
,
F.
Ghawche
.
2014
.
Zika virus infection complicated by Guillain-Barre syndrome—case report, French Polynesia, December 2013.
Euro Surveill.
19
:
20720
.
6.
World Health Organization
.
2016
.
The history of Zika virus.
.
7.
Pardy
R. D.
,
M. J.
Richer
.
2019
.
Zika virus pathogenesis: from early case reports to epidemics.
Viruses
11
:
886
.
8.
Krauer
F.
,
M.
Riesen
,
L.
Reveiz
,
O. T.
Oladapo
,
R.
Martínez-Vega
,
T. V.
Porgo
,
A.
Haefliger
,
N. J.
Broutet
,
N.
Low
;
WHO Zika Causality Working Group
.
2017
.
Zika virus infection as a cause of congenital brain abnormalities and Guillain-Barré syndrome: systematic review.
PLoS Med.
14
:
e1002203
.
9.
Luckheeram
R. V.
,
R.
Zhou
,
A. D.
Verma
,
B.
Xia
.
2012
.
CD4+T cells: differentiation and functions.
Clin. Dev. Immunol.
2012
:
925135
.
10.
Smith
C. M.
,
N. S.
Wilson
,
J.
Waithman
,
J. A.
Villadangos
,
F. R.
Carbone
,
W. R.
Heath
,
G. T.
Belz
.
2004
.
Cognate CD4+ T cell licensing of dendritic cells in CD8+ T cell immunity.
Nat. Immunol.
5
:
1143
1148
.
11.
Crotty
S.
2014
.
T follicular helper cell differentiation, function, and roles in disease.
Immunity
41
:
529
542
.
12.
Krishnaswamy
J. K.
,
S.
Alsén
,
U.
Yrlid
,
S. C.
Eisenbarth
,
A.
Williams
.
2018
.
Determination of T follicular helper cell fate by dendritic cells.
Front. Immunol.
9
:
2169
.
13.
Vinuesa
C. G.
,
M. A.
Linterman
,
D.
Yu
,
I. C.
MacLennan
.
2016
.
Follicular helper T cells.
Annu. Rev. Immunol.
34
:
335
368
.
14.
Victora
G. D.
,
M. C.
Nussenzweig
.
2012
.
Germinal centers.
Annu. Rev. Immunol.
30
:
429
457
.
15.
Pardy
R. D.
,
M. J.
Richer
.
2019
.
Protective to a T: the role of T cells during Zika virus infection.
Cells
8
:
820
.
16.
Pardy
R. D.
,
M. M.
Rajah
,
S. A.
Condotta
,
N. G.
Taylor
,
S. M.
Sagan
,
M. J.
Richer
.
2017
.
Analysis of the T cell response to Zika virus and identification of a novel CD8+ T cell epitope in immunocompetent mice.
PLoS Pathog.
13
:
e1006184
.
17.
Elong Ngono
A.
,
M. P.
Young
,
M.
Bunz
,
Z.
Xu
,
S.
Hattakam
,
E.
Vizcarra
,
J. A.
Regla-Nava
,
W. W.
Tang
,
M.
Yamabhai
,
J.
Wen
,
S.
Shresta
.
2019
.
CD4+ T cells promote humoral immunity and viral control during Zika virus infection. [Published erratum appears in 2019 PLoS Pathog. 15: e1007821.]
PLoS Pathog.
15
:
e1007474
.
18.
Hassert
M.
,
K. J.
Wolf
,
K. E.
Schwetye
,
R. J.
DiPaolo
,
J. D.
Brien
,
A. K.
Pinto
.
2018
.
CD4+T cells mediate protection against Zika associated severe disease in a mouse model of infection.
PLoS Pathog.
14
:
e1007237
.
19.
Lucas
C. G. O.
,
J. Z.
Kitoko
,
F. M.
Ferreira
,
V. G.
Suzart
,
M. P.
Papa
,
S. V. A.
Coelho
,
C. B.
Cavazzoni
,
H. A.
Paula-Neto
,
P. C.
Olsen
,
A.
Iwasaki
, et al
2018
.
Critical role of CD4+ T cells and IFNγ signaling in antibody-mediated resistance to Zika virus infection.
Nat. Commun.
9
:
3136
.
20.
Liang
H.
,
J.
Tang
,
Z.
Liu
,
Y.
Liu
,
Y.
Huang
,
Y.
Xu
,
P.
Hao
,
Z.
Yin
,
J.
Zhong
,
L.
Ye
, et al
2019
.
ZIKV infection induces robust Th1-like Tfh cell and long-term protective antibody responses in immunocompetent mice.
Nat. Commun.
10
:
3859
.
21.
Pardy
R. D.
,
S. F.
Valbon
,
B.
Cordeiro
,
C. M.
Krawczyk
,
M. J.
Richer
.
2021
.
An epidemic Zika virus isolate suppresses antiviral immunity by disrupting antigen presentation pathways.
Nat. Commun.
12
:
4051
.
22.
Fonseca
K.
,
B.
Meatherall
,
D.
Zarra
,
M.
Drebot
,
J.
MacDonald
,
K.
Pabbaraju
,
S.
Wong
,
P.
Webster
,
R.
Lindsay
,
R.
Tellier
.
2014
.
First case of Zika virus infection in a returning Canadian traveler.
Am. J. Trop. Med. Hyg.
91
:
1035
1038
.
23.
McDermott
D. S.
,
S. M.
Varga
.
2011
.
Quantifying antigen-specific CD4 T cells during a viral infection: CD4 T cell responses are larger than we think.
J. Immunol.
187
:
5568
5576
.
24.
Butler
N. S.
,
J.
Moebius
,
L. L.
Pewe
,
B.
Traore
,
O. K.
Doumbo
,
L. T.
Tygrett
,
T. J.
Waldschmidt
,
P. D.
Crompton
,
J. T.
Harty
.
2011
.
Therapeutic blockade of PD-L1 and LAG-3 rapidly clears established blood-stage Plasmodium infection.
Nat. Immunol.
13
:
188
195
.
25.
Nakayamada
S.
,
Y.
Kanno
,
H.
Takahashi
,
D.
Jankovic
,
K. T.
Lu
,
T. A.
Johnson
,
H. W.
Sun
,
G.
Vahedi
,
O.
Hakim
,
R.
Handon
, et al
2011
.
Early Th1 cell differentiation is marked by a Tfh cell-like transition.
Immunity
35
:
919
931
.
26.
Elsner
R. A.
,
M. J.
Shlomchik
.
2019
.
IL-12 blocks Tfh cell differentiation during Salmonella infection, thereby contributing to germinal center suppression.
Cell Rep.
29
:
2796
2809.e5
.
27.
Xie
M. M.
,
A. L.
Dent
.
2018
.
Unexpected help: follicular regulatory T cells in the germinal center.
Front. Immunol.
9
:
1536
.
28.
Pullen
G. R.
,
M. G.
Fitzgerald
,
C. S.
Hosking
.
1986
.
Antibody avidity determination by ELISA using thiocyanate elution.
J. Immunol. Methods
86
:
83
87
.
29.
Cerutti
A.
,
M.
Cols
,
I.
Puga
.
2013
.
Marginal zone B cells: virtues of innate-like antibody-producing lymphocytes.
Nat. Rev. Immunol.
13
:
118
132
.
30.
Gatto
D.
,
C.
Ruedl
,
B.
Odermatt
,
M. F.
Bachmann
.
2004
.
Rapid response of marginal zone B cells to viral particles.
J. Immunol.
173
:
4308
4316
.
31.
Guay
H. M.
,
R.
Mishra
,
R. L.
Garcea
,
R. M.
Welsh
,
E.
Szomolanyi-Tsuda
.
2009
.
Generation of protective T cell-independent antiviral antibody responses in SCID mice reconstituted with follicular or marginal zone B cells.
J. Immunol.
183
:
518
523
.
32.
Szomolanyi-Tsuda
E.
,
J. D.
Brien
,
J. E.
Dorgan
,
R. L.
Garcea
,
R. T.
Woodland
,
R. M.
Welsh
.
2001
.
Antiviral T-cell-independent type 2 antibody responses induced in vivo in the absence of T and NK cells.
Virology
280
:
160
168
.
33.
Szomolanyi-Tsuda
E.
,
R. M.
Welsh
.
1996
.
T cell-independent antibody-mediated clearance of polyoma virus in T cell-deficient mice.
J. Exp. Med.
183
:
403
411
.
34.
Fahey
L. M.
,
E. B.
Wilson
,
H.
Elsaesser
,
C. D.
Fistonich
,
D. B.
McGavern
,
D. G.
Brooks
.
2011
.
Viral persistence redirects CD4 T cell differentiation toward T follicular helper cells.
J. Exp. Med.
208
:
987
999
.

The authors have no financial conflicts of interest.

Supplementary data