Respiratory syncytial virus (RSV) infection can cause severe disease in the lower respiratory tract of infants and older people. Vaccination with a formalin-inactivated RSV vaccine (FI-RSV) and subsequent RSV infection has led to mild to severe pneumonia with two deaths among vaccinees. The vaccine-enhanced disease (VED) was recently demonstrated to be due to an elevated level of Th2 cell responses following loss of regulatory T (Treg) cells from the lungs. To induce high levels of neutralizing Abs and minimize pathogenic T cell responses, we developed a novel strategy of immunizing animals with a recombinant RSV G protein together with cyclosporine A. This novel vaccine induced not only a higher level of neutralizing Abs against RSV infection, but, most importantly, also significantly higher levels of Treg cells that suppressed VED in the lung after RSV infection. The induced responses provided protection against RSV challenge with no sign of pneumonia or bronchitis. Treg cell production of IL-10 was one of the key factors to suppress VED. These finding indicate that G protein plus cyclosporine A could be a promising vaccine against RSV infection in children and older people.

Respiratory syncytial virus (RSV) infection is a major cause of respiratory tract disease in children <5 years old. It leads to 64 million cases of bronchiolitis and viral pneumonia (15) and causes ∼200,000 deaths annually (4, 6). Moreover, there are direct correlations between RSV infection in childhood and development of asthma in adulthood (7). The infections have become a heavy medical and economic burden around the world, particularly in developing countries. During the infection, RSV modulates the host immune system through several mechanisms. These include arresting the cell cycle by downregulating p53 (8), inhibiting TNF-α via small hydrophobic protein (9), and suppressing maturation of dendritic cells (DCs) and some subtypes of T cells via NS1 and NS2 proteins (1012). Although the virus was identified >60 years ago, no vaccine is available currently. This problem is largely a legacy from the severity of the pathologic responses that were induced by vaccination with formalin-inactivated RSV (FI-RSV).

In the 1960s, the FI-RSV vaccine caused recipient children severe lung injuries, and two infants died in a phenomenon that is now called vaccine-enhanced disease (VED) (13, 14). Pathological analysis showed that the dead infants had an extended peribronchiolitis and alveolitis, with infiltration by neutrophils and a few eosinophils (1517). This problem can be reproduced in animal models, including mice and monkeys; when FI-RSV–immunized animals were challenged with RSV, there was extensive lung infiltration by neutrophils, lymphocytes, and some eosinophils, causing severe damage (1821). Increased complement-fixing Ab titers and lymphoproliferative responses were also observed during viral clearance (2225). These human cases and animal models suggested that the FI-RSV vaccine primes an overreactive and harmful inflammation that can be elicited when primed individuals encounter RSV infection.

Further reports showed that the harmful inflammation was due to the induction of a Th2-type response in lungs and overproduction of Th2 cytokines in a so-called cytokine storm. The Th2-type response was mainly a consequence of recruitment of infiltrating granulocytes, especially eosinophils (20, 2528). The eosinophils could not only promote naive CD4+ T cells to differentiate into Th2 cells by secreting cytokines such as IL-4 and IL-13, but they also could present Ags to naive CD4+ T cells directly to promote Th2 polarization. Moreover, chemokines such as eotaxin 1 and CXCL1 secreted by the eosinophils could recruit Th2 effector cells to the infected tissue (29). Based on the understanding that a correct Th1 versus Th2 balance is needed, the various prototype vaccines that have been developed are mainly aimed at inducing a strong Th1-type response and avoiding Th2. Krause et al. (30) found that an adenovirus-based RSV vaccine could increase the presence of plasmacytoid DCs (pDCs) in the lung during the RSV infection and these increased pDCs could promote the Th1 and regulatory T (Treg) cell responses against the RSV infection. Accordingly, an imbalanced pDC to conventional DC ratio was related to the Th2-type response.

The vaccines tested included attenuated RSV vaccines and subunit vaccines based on F or G glycoprotein and N protein. However, most only induced low levels of anti-RSV Abs, and a strong Ab response might be absolutely required to prevent RSV infections, because two commercially available mAbs, palivizumab and motavizumab, have been used to successfully treat children with RSV infections (31, 32).

Recently, a study demonstrated that within the harmful Th2 response elicited in the lung by infection after FI-RSV vaccination there was a low content of the Treg cells that are needed to attenuate such inflammation and tissue damage (33). The Treg subtype of CD4+ Th cells (34) uses a number of mechanisms for homeostatic control of overexuberant or incorrect immune responses. These include secretion of anti-inflammatory cytokines such as IL-10 and TGF-β (35, 36), disturbance of T cell metabolism by consuming IL-2 (37), or suppression of T cells by cell contact involving CTLA4 (38). Animals and human beings without Treg cells develop severe inflammation-related diseases, including allergy, asthma, and autoimmune disease. Because the lack of Treg cells can lead to severe tissue damage, it follows that induction of Treg cells might be used to treat such inflammation-related diseases (3941). Indeed, during some virus infections, Treg cells migrate into inflamed locations and minimize tissue damage by suppressing the excessive immune responses (42). The functional Treg cells express CCR4 and interact with chemokines, including CCL17 (TARC) and CCL22 (MDC), which are important for recruitment of the cells to skin and lung (43, 44). These important findings have been recently reported showing that both CCL17 and CCL22 are critical mediators in recruiting CCR4 Treg cells to suppress VED during RSV infection after the FI-RSV prime (45). Based on these findings, strategies for developing a safer RSV vaccine that circumvents FI-RSV–induced VED should include the induction of both Treg cells in lung and high levels of neutralizing Abs (20, 45, 46).

Among all of the 11 viral proteins of RSV, only G and F glycoproteins contain neutralizing epitopes and induce long-term neutralizing Abs (4749). For this reason, both have been considered as ideal candidates for RSV vaccine development. As the F protein is considered to induce high affinity of neutralizing Ab and only mild VED, several research groups have focused on this protein (50, 51). However, it is technically difficult to obtain the recombinant F glycoprotein in a stable prefusion form, and the misfolded or postfusion forms of F protein have shown less ability to induce sufficient neutralizing Abs (50, 51). A novel approach to make the prefusion form has been recently achieved (51). The G protein has been selected as an alternative promising candidate, because the immunogenicity is independent of the recombinant protein structure. It functions as an attachment protein during RSV infection by interacting with the CX3C motif of chemokine receptor CX3CR1 at the 182–186 aa position (52). Such interaction alerts CX3CL1 ligand–dependent T cell responses and impairs airway immune responses (53, 54). Another advantage of the G protein as a vaccine candidate is the presence of neutralizing epitopes that are comparatively independent of protein structure. Several approaches, including subunit-, nanoparticle-, peptide-, and bacterial-based vaccines have been tried, and an mAb against G protein has been demonstrated to inhibit the virus infection in animal models (5560). However, the association of the G protein with VED indicates the need for caution during its development as an RSV vaccine (26).

Cyclosporine A (CSA) is a widely used immunosuppressant in organ transplantation (6163), or in treatment of children with aplastic anemia (64, 65) and other autoimmune diseases (6668). CSA binds to the cytosolic protein cyclophilin and inhibits calcineurin of T cells, consequently suppressing transcription of IL-2 (6973). It also interferes with DC to T cell interaction (74) and can be used to induce Treg cells (7577). Moreover, the concept of immunizing with Ag together with an immunosuppressant to induce Treg cells and effectively treat autoimmune diseases has previously been proven in animal models (78, 79). Following this lead, we evaluated the ability of various immunosuppressants, including CSA, dexamethasone, rapamycin, and FK506, to induce tolerogenic reactions with human PBMCs in vitro. CSA showed the best ability to promote such reactions (data not shown). Although we found no report that a combination of CSA with RSV Ag could be useful for RSV vaccine development, in this study, we combined an RSV G truncated protein with CSA (G+CSA) and formulated the combination as a novel vaccine. We then evaluated its protective effect against RSV infection and its ability to inhibit lung inflammation. We demonstrated that G protein combined with CSA not only induced a high level of neutralizing anti-RSV Abs that led to reduced viral titers, but it also suppressed the lung inflammation that causes the severe lung injury often seen with FI-RSV vaccination. The suppression was due to induction of Treg cells. Therefore, G+CSA vaccination presents a novel approach to develop an effective and safe RSV vaccine for children.

Female 6- to 10-wk-old BALB/c mice were used for all experiments and purchased from the Shanghai SLAC Laboratory Animal Co. Ltd. (Shanghai, China). Foxp3-DTR/EGFP mice on a BALB/c background were from The Jackson Laboratory (Sacramento, CA). All mice were kept under specific pathogen-free conditions at Fudan University and handled according to the animal welfare guidelines for experimental animals.

Plaque-purified human RSV (type A2 strain from the American Type Culture Collection, Rockville, MD) was grown in HEp-2 cells and concentrated by ultracentrifugation (50,000 × g for 1 h). Mice were infected intranasally (i.n.) with 5 × 107 PFU RSV in 100 μl under anesthesia on day 14 after the second immunization.

FI-RSV vaccine was prepared as described by Kim et al. (14). In brief, 100 ml clarified RSV was incubated with formalin (37–40%) at 4000:1 (v/v) for 72 h at 37°C and then centrifuged at 50,000 × g for 1 h at 4°C. The pellet was diluted in 4 ml MEM and subsequently mixed with Imject alum adjuvant (Thermo Scientific, Rockford, IL) as 10:1 (v/v), suspended in 1 ml serum-free MEM, and stored at 4°C.

Nonglycosylated G protein was expressed in Escherichia coli BL21 (DE3), purified, and lyophilized. CSA (Santai, Taishan, Chain) was dissolved in propylene glycol/PBS (1:1) solution to 100 mg/ml. Lyophilized G protein was dissolved with the CSA solution or propylene glycol/PBS (1:1) solution to 100 mg/ml before injection.

The mice were randomly divided into groups and immunized s.c. with 10 μg G protein, CSA alone, or G+CSA on days 0 and 14. FI-RSV (50 μl) was used to immunize animals i.m. on days 0 and 14.

Mice were sacrificed by pentobarbital i.p. injection. Single-cell suspensions from the spleens and lung draining lymph nodes (dLNs) were collected with pestle and filter. Bronchoalveolar lavage (BAL) was collected by using 1 ml PBS to flush lungs three times before sacrificing animals.

Serum samples were obtained on days 0, 7, 21, and 38 after the first immunization and stored for ELISA analysis. Briefly, 96-well plates were coated with 5 × 106 PFU/ml heat-inactivated RSV (50 mM carbonate-bicarbonate buffer [pH 9.6]) at 37°C and blocked with 5% BSA in PBST (0.05% Tween 20 in PBS) at 37°C. The plates were incubated with serial 2-fold dilutions of serum for 1 h at 37°C. Bound Abs were detected with HRP-conjugated goat anti-mouse IgG (SouthernBiotech, Birmingham, AL), the enzymatic reaction was developed, and ODs were read at 450/620 nm by an ELISA plate reader (Bio-Rad, Hercules, CA)

The neutralization assay was performed as described previously (80). Briefly, sera were serially diluted 5-fold in a total of 100 μl PBS, heat inactivated at 56°C for 30 min, and incubated with 3 × 103 50% tissue culture–infective dose virus for 2 h at 4°C. Approximately 5 × 103 HEp-2 cells in 100 μl DMEM supplemented with 2% FCS were added to each well of a 96-well microtiter plate. The virus/serum mixture was added to the appropriate wells and incubated for 3 d in a 5% CO2 incubator at 37°C. Plates were then washed three times with PBST and fixed with 80% cold acetone in PBS followed by blocking with 3% blocking buffer. Goat anti-RSV Ab (Meridian Life Science, Saco, ME) was added to the appropriate wells and incubated for 60 min at 37°C. After three washings, bovine anti-goat IgG-HRP (Santa Cruz Biotechnology, Santa Cruz, CA) was added, the enzymatic reaction was developed, and ODs were read at 450/620 nm by an ELISA plate reader. Neutralizing Ab titers were calculated by the method of Reed and Muench (81); the average OD of triplicate wells was extrapolated by the inverse of the serum dilution that resulted in 50% reduction of RSV activity.

The quantity of IgG Ab produced in coculture medium was quantified by ELISA; 96-well plates were coated with IgG capture Ab (SouthernBiotech), and a curve was generated with mouse IgG-BIOT (SouthernBiotech).

On day 14 after the second immunization, single-splenocyte suspensions were collected and labeled with eFluor 670 proliferation dye (eBioscience, San Diego, CA). Cells at 5 × 105 were added to each well in a 96-well flat-bottom culture plate in 100 μl in a 37°C/5% CO2 incubator. Proliferation of responder T cells was determined after incubation for 4 d by analysis of eFluor 670 proliferation dye dilution.

Cells were stained with the following surface Abs (all from eBioscience unless noted otherwise): CD3 FITC, CD4 FITC or eFluor 450, CD25 allophycocyanin–eFluor 780, CCR4 BV405 (BioLegend, San Diego, CA), CXCR3 PerCP-Cy5.5, CD103 allophycocyanin, IFN-γ PE, IL-17a allophycocyanin, and IL-4 PE. All intracellular staining was done with the Foxp3/transcription factor staining buffer set (eBioscience) for IL-10 PerCP-Cy5.5 (eBioscience), Foxp3 FITC (eBioscience), and TGF-β (BioLegend). The data from BD LSRFortessa flow cytometer (BD Biosciences, San Diego, CA) was analyzed with FlowJo software (Tree Star, Ashland, OR). IL-10 levels in the BAL and cocultured medium were measured by a cytometric bead array flex set (BD Biosciences) following the manufacturer’s recommendations.

Splenocytes (1 × 106) from RSV noninfected mice were labeled with eFluor 670 proliferation dye (eBioscience) and cocultured with 5 × 104 cells of BAL and dLNs obtained from mice at 4 d after RSV (5 × 107 PFU) infection. Mixed cells were stimulated with heat-inactivated RSV at 37°C/5% CO2 for 3 d, and eFluor 670+ spleen T cells were monitored to determine the proliferation. The level of IL-10 in medium was tested by a cytometric bead array flex set (BD Biosciences) following the manufacturer’s recommendations.

Foxp3-DTR/EGFP mice were injected i.p. with 1 μg diphtheria toxin (DT; Sigma-Aldrich, St. Louis, MO) on days 1 and 2 before RSV infection to deplete Foxp3+ Treg cells. For neutralization of CCL17 and CCL22, 20 μg anti-CCL17 and anti-CCL22 Abs or IgG isotype control was i.p. injected on day 1 after RSV infection. IL-10 blocking was performed by adding anti–IL-10 Ab to the cell culture medium to the final concentration of 5 μg/ml, and mice were injected with one dose i.p. of 20 μg anti–IL-10 or IgG isotype control on day 1 after RSV infection.

Four days after RSV infection, lung tissues were fixed with 4% neutral buffered paraformaldehyde, and transverse sections (5–7 μm) were stained with H&E for histopathological evaluations. The infiltrated nuclei were counted with Image-Pro Plus (Media Cybernetics).

Statistical analysis was done using GraphPad Prism 6.0 (GraphPad Software, La Jolla, CA) and presented as means ± SEM. An unpaired Student t test and ANOVA were used in one-way or two-way data analyses. A p value <0.05 was considered statistically significant.

The RSV attachment protein without its transmembrane domain (G protein, aa 67–298) was cloned into the pET28a plasmid and expressed in E. coli BL21 (Supplemental Fig. 1A, 1B). The purified recombinant G protein was characterized by SDS-PAGE and identified by Western blot analysis with anti-RSV sera (Supplemental Fig. 1C, 1D).

To test responses to the combination of G protein with CSA, mice were immunized i.m. or s.c. twice with 2-wk intervals. As shown in Fig. 1A, 1B, and Supplemental Fig. 2, the levels of anti-RSV Abs and neutralizing Abs were higher in the group immunized with G+CSA compared with protein G without CSA, and even higher than in the FI-RSV–immunized group. Subcutaneous injections gave rise to significantly higher Ab responses (p < 0.05) than did the i.m. route (Fig. 1A). The optimal ratio of G protein/CSA to reach maximal inhibition of T cell activation while retaining induction of a high level of anti-RSV Abs was at 10 μg G protein plus 10 μg CSA (Fig. 1C). This dose was used throughout this study except where specifically mentioned.

FIGURE 1.

Induction of serum Ab and enhancement of splenic T cell proliferative capacity by G+CSA vaccine. Mice were immunized on days 0 and 14 and responses were measured at the stated intervals after the first immunization. (A) Rising titers of anti–RSV-IgG. (B) Neutralizing Ab titers at 28 d, defined from assays of 1:5 to 1:625 serum dilutions (n = 6). (C) Ag-specific splenic T cell proliferation and neutralizing Ab content of serum at 28 d after various doses of G protein and CSA (n = 5). Data represent means ± SEM. *p < 0.05.

FIGURE 1.

Induction of serum Ab and enhancement of splenic T cell proliferative capacity by G+CSA vaccine. Mice were immunized on days 0 and 14 and responses were measured at the stated intervals after the first immunization. (A) Rising titers of anti–RSV-IgG. (B) Neutralizing Ab titers at 28 d, defined from assays of 1:5 to 1:625 serum dilutions (n = 6). (C) Ag-specific splenic T cell proliferation and neutralizing Ab content of serum at 28 d after various doses of G protein and CSA (n = 5). Data represent means ± SEM. *p < 0.05.

Close modal

To evaluate protective efficacy of the 10 μg G+CSA formulation, animals were immunized on days 0 and 14 and then challenged i.n. with 5 × 107 PFU RSV (A2 strain) on day 28 (Fig. 2A). The viral loads in the lung were analyzed 4 d after the challenge. As depicted in Fig. 2B, the G+CSA immunization reduced the virus load in lung significantly more than did immunization with G protein (p = 0.0053) or FI-RSV (p = 0.0013). Meanwhile, the changes of body weight were assessed as an indication for morbidity of disease for 10 d after RSV challenge (Fig. 2C). Change in body weight in the G+CSA-immunized group was not significantly different from the noninfected controls. In contrast, early loss of body weight was revealed in the FI-RSV–immunized mice, and late losses of body weight were associated with groups receiving PBS, CSA, or G protein. These results suggested that G+CSA vaccine provided protection not only against infection but also against morbidity.

FIGURE 2.

G+CSA vaccine provides protection from RSV infection and VED. (A) Schematic outline of experimental approach: mice were immunized at days 0 and 14 and were challenged with RSV i.n. at 28 d. (B) Virus load of lung was tested 4 d after RSV infection (n = 5). (C) Changes of body weight were observed until 10 dpi; each line indicates the changing weight of one mouse (n = 10). (D) Total cell counts from BAL were tested 4 d after RSV infection (n = 5). (E) Lung index, indicating the ratio of the lung weight to the body weight (n = 5). (F) Representative lung sections made at 4 dpi and stained with H&E. Data represent means ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.001.

FIGURE 2.

G+CSA vaccine provides protection from RSV infection and VED. (A) Schematic outline of experimental approach: mice were immunized at days 0 and 14 and were challenged with RSV i.n. at 28 d. (B) Virus load of lung was tested 4 d after RSV infection (n = 5). (C) Changes of body weight were observed until 10 dpi; each line indicates the changing weight of one mouse (n = 10). (D) Total cell counts from BAL were tested 4 d after RSV infection (n = 5). (E) Lung index, indicating the ratio of the lung weight to the body weight (n = 5). (F) Representative lung sections made at 4 dpi and stained with H&E. Data represent means ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.001.

Close modal

Because VED is the major obstacle against RSV vaccine development, we analyzed lung pathology including the tissue index and histochemistry of the challenged mice. The lung tissue index was significantly lower in animals that had received G+CSA than in the other groups (Fig. 2E). H&E staining revealed that immunization with G protein or FI-RSV vaccine followed by challenge with RSV resulted in severe pneumonia associated with drastic perivasculitis, peribronchiolitis, and alveolitis. In contrast, G+CSA recipients had mild lung pathology with some peribronchiolitis and nearly normal alveolar morphology (Fig. 2F). This was consistent with the total cell counts in BAL fluid from the corresponding groups (Fig. 2D). These results demonstrated that the G+CSA vaccine, in contrast to the other vaccines, could significantly reduce the lung inflammation and injury that occurred after RSV challenge.

Because Treg cells were reported to be one of the most important factors in limiting VED (45), we examined whether the G+CSA vaccine could induce a high level of Treg cells in the BAL and dLNs of the lungs and spleens in these animals after RSV challenge. We observed that the percentage of CD4+ T cells in spleens that were CD25+Foxp3+ Treg cells had fallen by 4 d after challenge from around the 10% seen in PBS control mice down to 7% when the mice had been vaccinated with FI-RSV, to 3% with G, or to 5% with CSA (Fig. 3A). The Treg cells were purged to an even lower level (<2%) in the dLNs in these groups (Fig. 3B). In contrast, animals that had received G+CSA before challenge with RSV had higher levels of Treg cells in spleens and dLNs when compared with naive animals (Fig. 3A, 3B). Furthermore, in BAL there was a striking increase in Treg cells after challenge in the G+CSA group, both in their absolute numbers and as a percentage of total CD4+ T cells (Fig. 3C, 3D), and the increases were much higher than seen in the other groups (p < 0.0001). This demonstration that G+CSA vaccination leads to robust Treg cell responses in challenged lungs is consistent with the notion that Treg cells might abrogate the lymphocyte activation and subsequent excessive inflammatory responses and lung damage during the RSV infection.

FIGURE 3.

Treg cells prevent lung injury during RSV infection after G+CSA immunization. Mice were immunized at days 0 and 14 and challenged with RSV on day 28. Four days later, percentages of CD4+CD25+Foxp3+ Treg cells from spleen (A), dLNs (B), and BAL (C) were measured by flow cytometry with specific Abs. Treg cell counts of BAL were tested at the same time with standard beads as control (D). Data represent mean ± SEM. *p < 0.05, ***p < 0.001, ****p < 0.00001, n = 8.

FIGURE 3.

Treg cells prevent lung injury during RSV infection after G+CSA immunization. Mice were immunized at days 0 and 14 and challenged with RSV on day 28. Four days later, percentages of CD4+CD25+Foxp3+ Treg cells from spleen (A), dLNs (B), and BAL (C) were measured by flow cytometry with specific Abs. Treg cell counts of BAL were tested at the same time with standard beads as control (D). Data represent mean ± SEM. *p < 0.05, ***p < 0.001, ****p < 0.00001, n = 8.

Close modal

To elucidate the biological importance of G+CSA-induced Treg cells, Treg cell depletion or adoptive transfer studies were performed. Foxp3-DTR mice were immunized twice with G+CSA on days 0 and 14, given two injections of DT on days 26 and 27 to delete Treg cells, and then were RSV challenged on day 28 (Fig. 4A). Foxp3+ cells remained depleted for a week after the DT injections (Supplemental Fig. 4C). The viral loads in the lung were analyzed 4 d after the challenge. As depicted in Fig. 4B, the virus load of the Treg cell–depleted G+CSA mice was between those of the G+CSA- and G protein–immunized groups. The changes of body weight were assessed as an indication of disease morbidity 10 d after RSV challenge. As shown in Fig. 4C, even when the mice had been immunized with G+CSA, weight loss was severe once Treg cells were depleted. Moreover, the BAL from Treg cell–depleted mice contained more cells than did the BAL from Treg cell–sufficient mice (Fig. 4D), and among these cells, most CD4+ T cells expressed IL-4 (Fig. 4E). Lungs were harvested on day 4 after the RSV challenge, fixed, sectioned, and stained with H&E (Fig. 4F). The lung morphologies were found to be unchanged when animals had been immunized with G+CSA without Treg cell depletion, and massive cell infiltration was exhibited in the G+CSA-immunized mice that had been depleted of Treg cells.

FIGURE 4.

Treg depletion or complementation influences the lung injury during RSV infection. (A) Schematic outline of depletion experiment. Foxp3-DTR mice were immunized on days 0 and 14, DT was injected at days 26 and 27, and mice were challenged with RSV at day 28. (B) Virus load of lung was tested 4 d after RSV infection (n = 5). (C) Changes of body weight were observed until 10 dpi; each line indicates the changing weight of one mouse (n = 6). (D) Total cell counts from BAL were tested 4 d after RSV infection (n = 5). (E) Proportions of BAL CD4+ T cells producing IFN-γ, IL-4, and IL-17a (n = 5). (F) Representative lung sections made at 4 dpi and stained with H&E (n = 5). (G) Schematic outline of complementation experiment. Mice were immunized with G+CSA or PBS on days −2 and 12 and infected with RSV on day 26. Four days later, BAL (from G+CSA and PBS groups) or splenic Treg cells (from G+CSA group) were i.n. transferred into G protein–immunized and RSV-infected mice (2 dpi) and pathology was assessed 2 d later. Lung sections were stained with H&E and the infiltrated cells were counted (n = 6) (H). Original magnification ×173. Data represent means ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.00001.

FIGURE 4.

Treg depletion or complementation influences the lung injury during RSV infection. (A) Schematic outline of depletion experiment. Foxp3-DTR mice were immunized on days 0 and 14, DT was injected at days 26 and 27, and mice were challenged with RSV at day 28. (B) Virus load of lung was tested 4 d after RSV infection (n = 5). (C) Changes of body weight were observed until 10 dpi; each line indicates the changing weight of one mouse (n = 6). (D) Total cell counts from BAL were tested 4 d after RSV infection (n = 5). (E) Proportions of BAL CD4+ T cells producing IFN-γ, IL-4, and IL-17a (n = 5). (F) Representative lung sections made at 4 dpi and stained with H&E (n = 5). (G) Schematic outline of complementation experiment. Mice were immunized with G+CSA or PBS on days −2 and 12 and infected with RSV on day 26. Four days later, BAL (from G+CSA and PBS groups) or splenic Treg cells (from G+CSA group) were i.n. transferred into G protein–immunized and RSV-infected mice (2 dpi) and pathology was assessed 2 d later. Lung sections were stained with H&E and the infiltrated cells were counted (n = 6) (H). Original magnification ×173. Data represent means ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.00001.

Close modal

We tested whether any functional activities could be transferred with Treg cells or BAL fluid from G+CSA- or PBS-immunized animals. Splenocytes were isolated from Foxp3-EGFP mice and the Treg cells were sorted by FACS based on GFP positivity. G protein–immunized animals received either the BAL fluid or the sorted Treg cells by i.n. transfer on the second day after the RSV challenge (Fig. 4G). The recipients were sacrificed 2 d after the transfer and their lungs were removed for histochemistry analysis and staining with H&E (Fig. 4H). The recipients of either BAL fluid or Treg cells from the G+CSA-immunized animals showed much less lymphocyte infiltration than did the recipients of BAL from PBS-treated controls or recipients of medium only. Because both donor Treg cells and BAL fluid could suppress inflammatory T cell infiltration into lung, they both may play an important role to protect lung from VED upon RSV infection.

Because the expression of CCR4 by Treg cells is required for their movement from the peripheral circulation into target tissues where the chemokines CCL17/22 are released (82, 83), we asked whether it had a role here. We isolated Treg cells from BAL and looked for surface expression of several chemokine receptors. We found no differences between the groups in the expression levels of CD103 and CXCR3 on the Treg cells (Supplemental Fig. 3). However, the expression of CCR4 on the G+CSA-induced Treg cells was significantly higher (p < 0.05) than for Treg cells from the other groups (Fig. 5A).

FIGURE 5.

CCR4 and CCL17/22 control the Treg cell migration from dLNs to the lung. Mice were immunized on days 0 and 14 and infected with RSV at 14 d after the second injection. (A) Expression of CCR4 on Treg cells from BAL was tested by a flow cytometer at 4 dpi. Alternatively, at 1 dpi, G+CSA-immunized mice were i.p. injected with anti-CCL17 and anti-CCL22 Abs and after 3 d the body weight changes (B), pathology in lung sections stained with H&E (C) (original magnification ×173), and numbers of Treg cells in BAL (D) and dLNs (E) were tested. Data represent means ± SEM. *p < 0.05, **p < 0.01, ****p < 0.00001, n = 6.

FIGURE 5.

CCR4 and CCL17/22 control the Treg cell migration from dLNs to the lung. Mice were immunized on days 0 and 14 and infected with RSV at 14 d after the second injection. (A) Expression of CCR4 on Treg cells from BAL was tested by a flow cytometer at 4 dpi. Alternatively, at 1 dpi, G+CSA-immunized mice were i.p. injected with anti-CCL17 and anti-CCL22 Abs and after 3 d the body weight changes (B), pathology in lung sections stained with H&E (C) (original magnification ×173), and numbers of Treg cells in BAL (D) and dLNs (E) were tested. Data represent means ± SEM. *p < 0.05, **p < 0.01, ****p < 0.00001, n = 6.

Close modal

To demonstrate the functional relevance of CCR4 on the migration of the G+CSA-induced Treg cells, we neutralized CCL17 and CCL22 by i.p. injection of anti-CCL17 and anti-CCL22 Abs into the G+CSA vaccine recipients on 1 d postinfection (dpi) with RSV. Weight changes were tested for 4 d and the anti-CCL17/22–treated animals showed a significant weight loss (p < 0.01) at 4 dpi compared with IgG isotype Ab controls (Fig. 5B). This result was consistent with the severe lung inflammation and lung injuries seen when lung sections were analyzed by histochemistry and H&E staining (Fig. 5C). When CCL17/22 was blocked, the level of the G+CSA-induced Treg cells in the BAL was significantly reduced (p < 0.05) compared with the IgG isotype controls (Fig. 5D). Furthermore, the level of G+CSA-induced Treg cells in dLNs was slightly higher after the blockade compared with prior to the blocking (Fig. 5E), although the difference was not significant (p = 0.083). These findings imply that G+CSA-induced Treg cells could not migrate from LNs into lung due to the Ab-induced low level of CCL17/22 in the tissue. This is consistent with the evidence that CCR4 expression on the G+CSA-induced Treg cells plays an important role in migration from peripheral or dLNs to the lung after RSV infection.

Because IL-10 and TGF-β are both key anti-inflammatory cytokines produced in Treg cells, we sought to determine whether these were responsible for suppressing inflammation during RSV infection. Treg cells were harvested from spleen, dLNs, and BAL fluid in the various immunization groups and analyzed by intracellular staining and FACS. There were no differences in the level of TGF-β among all of the groups (Fig. 6A). However, a significantly higher level of IL-10 in Treg cells of the spleen and dLNs was induced by immunizing animals with G+CSA compared with other groups (p < 0.001; Fig. 6B, 6C, Supplemental Fig. 4A, 4B). Strikingly, the level of IL-10 in the BAL Treg cells of the G+CSA-immunized group was 6-fold higher than in other immunized groups after the RSV infection (Fig. 6D). These data suggested that IL-10 could have the anti-inflammatory role. To further investigate the source of the IL-10, we depleted Treg cells by injecting DT into Foxp3-DTR mice. The mice were immunized with G+CSA at days 0 and 14, and PBS or DT was injected i.p. at days 26 and 27. The mice were then infected with RSV at day 28. The concentration of IL-10 in the BAL was decreased significantly (p < 0.001) as tested 4 d after Treg cell depletion (Fig. 6J).

FIGURE 6.

IL-10 of Treg cells is required for the protection against VED following RSV infection. (AD) Vaccine-induced changes in the proportion of Treg cells producing TGF-β or IL-10 after RSV infection. Mice were immunized at days 0 and 14 and challenged i.n. with RSV at day 28. Four days later, Treg cell production of TGF-β in spleen (A) or IL-10 in spleen (B), LNs (C), and BAL (D) was tested (n = 6). (E) Neutralizing IL-10 in vivo inhibits VED. G+CSA-immunized mouse were infected with RSV and then at 2 dpi IL-10 was blocked by i.n. injection of anti–IL-10 or matching isotype control. Two days after that, lung sections were stained with H&E (n = 5). Original magnification ×173. (FH) Treg from G+CSA-immunized mice inhibit T cell proliferation and IL-10 production in vitro. (F) Schematic outline of experimental design. BAL from mice that had been G+CSA- or protein G–immunized and then infected with RSV were cocultured with splenocytes from naive or from noninfected protein G–immunized mice together with inactivated RSV for 3 d and then T cell proliferation (G) and IL-10 in medium (H) were measured (n = 4). (I) Presence of anti–IL-10 Ab during a 3-d culture of splenocytes from RSV-infected mice with inactivated RSV inhibits T cell proliferation. (J) Foxp3-DTR mice were immunized with G+CSA at 0 and 14 d, and PBS or DT was injected i.p. at days 26 and 27, and then the mice were infected with RSV at day 28. The concentration of IL-10 in the BAL was tested 4 d later (n = 5). Data represent means ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001.

FIGURE 6.

IL-10 of Treg cells is required for the protection against VED following RSV infection. (AD) Vaccine-induced changes in the proportion of Treg cells producing TGF-β or IL-10 after RSV infection. Mice were immunized at days 0 and 14 and challenged i.n. with RSV at day 28. Four days later, Treg cell production of TGF-β in spleen (A) or IL-10 in spleen (B), LNs (C), and BAL (D) was tested (n = 6). (E) Neutralizing IL-10 in vivo inhibits VED. G+CSA-immunized mouse were infected with RSV and then at 2 dpi IL-10 was blocked by i.n. injection of anti–IL-10 or matching isotype control. Two days after that, lung sections were stained with H&E (n = 5). Original magnification ×173. (FH) Treg from G+CSA-immunized mice inhibit T cell proliferation and IL-10 production in vitro. (F) Schematic outline of experimental design. BAL from mice that had been G+CSA- or protein G–immunized and then infected with RSV were cocultured with splenocytes from naive or from noninfected protein G–immunized mice together with inactivated RSV for 3 d and then T cell proliferation (G) and IL-10 in medium (H) were measured (n = 4). (I) Presence of anti–IL-10 Ab during a 3-d culture of splenocytes from RSV-infected mice with inactivated RSV inhibits T cell proliferation. (J) Foxp3-DTR mice were immunized with G+CSA at 0 and 14 d, and PBS or DT was injected i.p. at days 26 and 27, and then the mice were infected with RSV at day 28. The concentration of IL-10 in the BAL was tested 4 d later (n = 5). Data represent means ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001.

Close modal

To assess the role of IL-10, we performed an in vitro coculture inhibition assay with Treg cells isolated after RSV infection from the BAL and dLNs of the animals that had been immunized with G+CSA or G protein. These isolated cells were cocultured with splenocytes from PBS- or G protein–immunized mice, where IL-10 could be secreted by the cells in the presence of inactivated RSV as a stimulant (Fig. 6F). Proliferation of the splenocytes and the quantity of IL-10 in medium were measured 3 d later. As depicted in Fig. 6G, only the cells from the G+CSA-immunized animals suppressed G protein–primed T cell proliferation, whereas the cells from the G protein–immunized animals did not. The suppression was apparently associated with the high level of IL-10 produced (Fig. 6H). To demonstrate the direct involvement of IL-10 in suppression during RSV infection, splenocytes of the infected mice were stimulated with inactivated RSV in vitro with and without the addition of anti–IL-10 Ab or matched isotype IgG. T cell proliferation was measured after 3 d. As shown in Fig. 6I, the inhibition of T cell proliferation could be effectively reversed by addition of the anti–IL-10 Abs, but much less by the isotope control. Furthermore, when anti–IL-10 Ab was injected i.n. into G+CSA-immunized animals at 2 dpi, lymphocyte infiltration into the lung was more severe than in the isotype control (Fig. 6E). Evidently, IL-10 is one of the functional anti-inflammatory cytokines that limits pathogenic T cell activation and proliferation provoked by the RSV infection.

The effects and associated safety issues of RSV vaccines have puzzled researchers for >50 y, various strategies have been tried (84), but there is still no vaccine licensed for clinical use. Our study provides a new concept for RSV vaccine development, that is, that of inducing Treg cells without impairing Ab production.

The critical role of Treg cells in controlling the pathology of VED in RSV infection has been known for many years (47, 85). Openshaw and colleagues (45) showed that FI-RSV vaccination expelled Treg cells from lung and made pulmonary tissue prone to inflammation so that severe lung injury occurred upon RSV infection. Subsequently, manipulating the expelled Treg cells back into the lung was found to eliminate RSV infection–provoked inflammation and lung injury (45). We confirmed this phenomenon by the use of a Treg cell knockout animal model, because lack of Treg cells can be achieved by deletion in Foxp3-DTR transgenic mouse; the Treg-deleted mice showed more severe pneumonia than did the nondeleted mice (Fig. 4). Accordingly, we explored an implied corollary, that induction of Treg cells might be an important requisite for a successful RSV vaccine. Because we had previously shown that vaccination with a protein Ag plus an immunosuppressive agent such as CSA could induce potent immunosuppressive Treg cells, we tested RSV G protein plus CSA. The demonstrated efficacy of this approach may be ascribed, at least in part, to the induction of Treg cells in the dLNs and BAL where they may be most needed to suppress lung inflammation and injury (Fig. 3). This did not occur with the other vaccines, and they caused VED (Fig. 2). Inductions of these Treg cells may be associated with vaccine-induced rebalance of pDCs and conventional DCs as previous reported (30), but this remains to be tested. Although some Treg cell types have been documented to produce TGF-β (36, 86), the G+CSA-induced Treg cells did not (Fig. 6A), but they produced large amounts of IL-10, 6-fold more than the controls. Furthermore, the IL-10 was produced locally in the lung in response to RSV infection (Fig. 6B–D, 6J) and was demonstrated to participate in suppressing Ag-primed T cell activation and proliferation in vitro (Fig. 6G–I). Although IL-10 plays an important anti-inflammatory role, there may be other molecules, such as IL-35, that also contribute to this suppression, and this should be further investigated. Migration of these Treg cells from LNs to lung appeared to be required because it was prevented by inhibition of the CCL17/22–CCR4 axis of chemotaxis, and the Treg cells expressed high levels of CCR4 (Fig. 5).

There are contradictory reports of the effects of CSA upon immune responses. For example, some groups found that CSA reduced the function and level of Treg cells (8789), but other evidence showed that CSA could induce Treg cells (76, 77, 90, 91). This discrepancy may be due to dose effects because we observed that a lower dose CSA was more effective in inducing Treg cells than was a higher dose (Fig. 1C). This has been also reported previously (75). Also, there have been some studies of the influence of CSA on B cells and the humoral response (9294). CSA did not suppress Ab production in autoimmune diseases and transplantations (9597), suggesting that the target mechanism in T cells is different from that in B cells; CSA targets calcium activation in T cells but may not affect that pathway in B cells (93, 98100). However, we speculate that the mechanism by which CSA enhanced Ab production here may be indirect and via induction of Treg cells. There are contradictory reports of Treg effects on B cell function; that is, there may be suppression of B cells in one circumstance (101103), but help for B cells in another (104106). Help for B cells might be a consequence of IL-10 production by induced Treg cells. Although IL-10 is considered to be an anti-inflammatory cytokine, its enhancement of B cell activation and Ab production has been known for decades (97, 107110). Induced Treg cells produce high levels of IL-10, and this has been suggested to lead to B cell maturation (108, 111, 112). Although it seems likely that the IL-10–producing Treg cell acted in this case as an alternative Th cell to promote B cell Ab production, the mechanism by which G protein+CSA induce high titers of anti-RSV IgG and neutralizing Ab (Fig. 1, Supplemental Fig. 2) needs further investigation.

We chose to work with the RSV G rather than the F glycoprotein because it has the advantages as a vaccine candidate of being easy to produce, having fewer requirements for a particular structure, and having several neutralizing epitopes within its sequence (113). However, the G glycoprotein may potently stimulate host immunity by interacting with CXC3R1, DC-SIGN, and L-SIGN of DCs to modulate the host immune system during the infection (57, 58, 114116). Such strong stimulation of the immune responses can present a double-edged sword. On the one hand, there can be rapid host response to fend off infection, but on the other hand, the host system can overreact, leading to severe pathology. It appears that by adding CSA to the G protein the potent stimulation of the protective immune response was preserved and the harmful aspect eliminated, and further investigation of this vaccine is warranted.

We thank Dr. Douglas Lowrie (Shanghai Medical College, Fudan University, Shanghai, China) for valuable discussions and suggestions. We thank Xianghua Shi, Yongfu Xu, and Changsong Xu of Beijing Advaccine Biotechnology Co. Ltd. for fermentation and downstream processing, formulation, and analysis.

This work was supported in part by National Projects for New Drug Discovery Grant 2013ZX09102041, Nature Science Foundation of China Grant 31430027, and by National High Technology 863 Projects Grant 2012AA02A406 (to B.W.).

The online version of this article contains supplemental material.

Abbreviations used in this article:

BAL

bronchoalveolar lavage

CSA

cyclosporine A

DC

dendritic cell

dLN

draining lymph node

dpi

day postinfection

DT

diphtheria toxin

FI-RSV

formalin-inactivated respiratory syncytial virus

G+CSA

RSV G truncated protein with CSA

i.n.

intranasal(ly)

LN

lymph node

pDC

plasmacytoid DC

RSV

respiratory syncytial virus

Treg

regulatory T

VED

vaccine-enhanced disease.

1
Hon
K. L.
,
Leung
T. F.
,
Cheng
W. Y.
,
Ko
N. M.
,
Tang
W. K.
,
Wong
W. W.
,
Yeung
W. H.
,
Chan
P. K.
.
2012
.
Respiratory syncytial virus morbidity, premorbid factors, seasonality, and implications for prophylaxis.
J. Crit. Care
27
:
464
468
.
2
García
C. G.
,
Bhore
R.
,
Soriano-Fallas
A.
,
Trost
M.
,
Chason
R.
,
Ramilo
O.
,
Mejias
A.
.
2010
.
Risk factors in children hospitalized with RSV bronchiolitis versus non-RSV bronchiolitis.
Pediatrics
126
:
e1453
e1460
.
3
Collins
P. L.
,
Melero
J. A.
.
2011
.
Progress in understanding and controlling respiratory syncytial virus: still crazy after all these years.
Virus Res.
162
:
80
99
.
4
Nair
H.
,
Nokes
D. J.
,
Gessner
B. D.
,
Dherani
M.
,
Madhi
S. A.
,
Singleton
R. J.
,
O’Brien
K. L.
,
Roca
A.
,
Wright
P. F.
,
Bruce
N.
, et al
.
2010
.
Global burden of acute lower respiratory infections due to respiratory syncytial virus in young children: a systematic review and meta-analysis.
Lancet
375
:
1545
1555
.
5
Lucion
M. F.
,
Juarez
Mdel. V.
,
Viegas
M.
,
Castellano
V.
,
Romanin
V. S.
,
Grobaporto
M.
,
Bakir
J.
,
Mistchenko
A. S.
,
Gentile
A.
.
2014
.
Respiratory syncytial virus: clinical and epidemiological pattern in pediatric patients admitted to a children’s hospital between 2000 and 2013.
Arch. Argent. Pediatr.
112
:
397
404
.
6
Welliver
R. C.
 Sr.
,
Checchia
P. A.
,
Bauman
J. H.
,
Fernandes
A. W.
,
Mahadevia
P. J.
,
Hall
C. B.
.
2010
.
Fatality rates in published reports of RSV hospitalizations among high-risk and otherwise healthy children.
Curr. Med. Res. Opin.
26
:
2175
2181
.
7
Krishnamoorthy
N.
,
Khare
A.
,
Oriss
T. B.
,
Raundhal
M.
,
Morse
C.
,
Yarlagadda
M.
,
Wenzel
S. E.
,
Moore
M. L.
,
Peebles
R. S.
 Jr.
,
Ray
A.
,
Ray
P.
.
2012
.
Early infection with respiratory syncytial virus impairs regulatory T cell function and increases susceptibility to allergic asthma.
Nat. Med.
18
:
1525
1530
.
8
Gibbs
J. D.
,
Ornoff
D. M.
,
Igo
H. A.
,
Zeng
J. Y.
,
Imani
F.
.
2009
.
Cell cycle arrest by transforming growth factor β1 enhances replication of respiratory syncytial virus in lung epithelial cells.
J. Virol.
83
:
12424
12431
.
9
Rixon
H. W.
,
Brown
G.
,
Aitken
J.
,
McDonald
T.
,
Graham
S.
,
Sugrue
R. J.
.
2004
.
The small hydrophobic (SH) protein accumulates within lipid-raft structures of the Golgi complex during respiratory syncytial virus infection.
J. Gen. Virol.
85
:
1153
1165
.
10
Munir
S.
,
Hillyer
P.
,
Le Nouën
C.
,
Buchholz
U. J.
,
Rabin
R. L.
,
Collins
P. L.
,
Bukreyev
A.
.
2011
.
Respiratory syncytial virus interferon antagonist NS1 protein suppresses and skews the human T lymphocyte response.
PLoS Pathog.
7
:
e1001336
.
11
Bitko
V.
,
Shulyayeva
O.
,
Mazumder
B.
,
Musiyenko
A.
,
Ramaswamy
M.
,
Look
D. C.
,
Barik
S.
.
2007
.
Nonstructural proteins of respiratory syncytial virus suppress premature apoptosis by an NF-κB-dependent, interferon-independent mechanism and facilitate virus growth.
J. Virol.
81
:
1786
1795
.
12
Munir
S.
,
Le Nouen
C.
,
Luongo
C.
,
Buchholz
U. J.
,
Collins
P. L.
,
Bukreyev
A.
.
2008
.
Nonstructural proteins 1 and 2 of respiratory syncytial virus suppress maturation of human dendritic cells.
J. Virol.
82
:
8780
8796
.
13
Kapikian
A. Z.
,
Mitchell
R. H.
,
Chanock
R. M.
,
Shvedoff
R. A.
,
Stewart
C. E.
.
1969
.
An epidemiologic study of altered clinical reactivity to respiratory syncytial (RS) virus infection in children previously vaccinated with an inactivated RS virus vaccine.
Am. J. Epidemiol.
89
:
405
421
.
14
Kim
H. W.
,
Canchola
J. G.
,
Brandt
C. D.
,
Pyles
G.
,
Chanock
R. M.
,
Jensen
K.
,
Parrott
R. H.
.
1969
.
Respiratory syncytial virus disease in infants despite prior administration of antigenic inactivated vaccine.
Am. J. Epidemiol.
89
:
422
434
.
15
Bataki
E. L.
,
Evans
G. S.
,
Everard
M. L.
.
2005
.
Respiratory syncytial virus and neutrophil activation.
Clin. Exp. Immunol.
140
:
470
477
.
16
Lindemans
C. A.
,
Kimpen
J. L.
,
Luijk
B.
,
Heidema
J.
,
Kanters
D.
,
van der Ent
C. K.
,
Koenderman
L.
.
2006
.
Systemic eosinophil response induced by respiratory syncytial virus.
Clin. Exp. Immunol.
144
:
409
417
.
17
Stokes
K. L.
,
Currier
M. G.
,
Sakamoto
K.
,
Lee
S.
,
Collins
P. L.
,
Plemper
R. K.
,
Moore
M. L.
.
2013
.
The respiratory syncytial virus fusion protein and neutrophils mediate the airway mucin response to pathogenic respiratory syncytial virus infection.
J. Virol.
87
:
10070
10082
.
18
Cannon
M. J.
,
Openshaw
P. J.
,
Askonas
B. A.
.
1988
.
Cytotoxic T cells clear virus but augment lung pathology in mice infected with respiratory syncytial virus.
J. Exp. Med.
168
:
1163
1168
.
19
Graham
B. S.
,
Bunton
L. A.
,
Wright
P. F.
,
Karzon
D. T.
.
1991
.
Role of T lymphocyte subsets in the pathogenesis of primary infection and rechallenge with respiratory syncytial virus in mice.
J. Clin. Invest.
88
:
1026
1033
.
20
Connors
M.
,
Giese
N. A.
,
Kulkarni
A. B.
,
Firestone
C. Y.
,
Morse
H. C.
 III
,
Murphy
B. R.
.
1994
.
Enhanced pulmonary histopathology induced by respiratory syncytial virus (RSV) challenge of formalin-inactivated RSV-immunized BALB/c mice is abrogated by depletion of interleukin-4 (IL-4) and IL-10.
J. Virol.
68
:
5321
5325
.
21
Tang
Y. W.
,
Graham
B. S.
.
1995
.
Interleukin-12 treatment during immunization elicits a T helper cell type 1-like immune response in mice challenged with respiratory syncytial virus and improves vaccine immunogenicity.
J. Infect. Dis.
172
:
734
738
.
22
van Drunen Littel-van den Hurk
S.
,
Mapletoft
J. W.
,
Arsic
N.
,
Kovacs-Nolan
J.
.
2007
.
Immunopathology of RSV infection: prospects for developing vaccines without this complication.
Rev. Med. Virol.
17
:
5
34
.
23
Polack
F. P.
,
Teng
M. N.
,
Collins
P. L.
,
Prince
G. A.
,
Exner
M.
,
Regele
H.
,
Lirman
D. D.
,
Rabold
R.
,
Hoffman
S. J.
,
Karp
C. L.
, et al
.
2002
.
A role for immune complexes in enhanced respiratory syncytial virus disease.
J. Exp. Med.
196
:
859
865
.
24
Openshaw
P. J. M.
,
Tregoning
J. S.
.
2005
.
Immune responses and disease enhancement during respiratory syncytial virus infection.
Clin. Microbiol. Rev.
18
:
541
555
.
25
Castilow
E. M.
,
Olson
M. R.
,
Varga
S. M.
.
2007
.
Understanding respiratory syncytial virus (RSV) vaccine-enhanced disease.
Immunol. Res.
39
:
225
239
.
26
Johnson
T. R.
,
Varga
S. M.
,
Braciale
T. J.
,
Graham
B. S.
.
2004
.
Vβ14+ T cells mediate the vaccine-enhanced disease induced by immunization with respiratory syncytial virus (RSV) G glycoprotein but not with formalin-inactivated RSV.
J. Virol.
78
:
8753
8760
.
27
Power
U. F.
,
Huss
T.
,
Michaud
V.
,
Plotnicky-Gilquin
H.
,
Bonnefoy
J. Y.
,
Nguyen
T. N.
.
2001
.
Differential histopathology and chemokine gene expression in lung tissues following respiratory syncytial virus (RSV) challenge of formalin-inactivated RSV- or BBG2Na-immunized mice.
J. Virol.
75
:
12421
12430
.
28
Graham
B. S.
,
Henderson
G. S.
,
Tang
Y. W.
,
Lu
X.
,
Neuzil
K. M.
,
Colley
D. G.
.
1993
.
Priming immunization determines T helper cytokine mRNA expression patterns in lungs of mice challenged with respiratory syncytial virus.
J. Immunol.
151
:
2032
2040
.
29
Spencer
L. A.
,
Weller
P. F.
.
2010
.
Eosinophils and Th2 immunity: contemporary insights.
Immunol. Cell Biol.
88
:
250
256
.
30
Krause
A.
,
Xu
Y.
,
Ross
S.
,
Wu
W.
,
Joh
J.
,
Worgall
S.
.
2011
.
Absence of vaccine-enhanced RSV disease and changes in pulmonary dendritic cells with adenovirus-based RSV vaccine.
Virol. J.
8
:
375
.
31
Ramilo
O.
,
Lagos
R.
,
Sáez-Llorens
X.
,
Suzich
J.
,
Wang
C. K.
,
Jensen
K. M.
,
Harris
B. S.
,
Losonsky
G. A.
,
Griffin
M. P.
Motavizumab Study Group
.
2014
.
Motavizumab treatment of infants hospitalized with respiratory syncytial virus infection does not decrease viral load or severity of illness.
Pediatr. Infect. Dis. J.
33
:
703
709
.
32
Halsey
N. A.
,
Abramson
J. S.
,
Chesney
P. J.
,
Fisher
M. C.
,
Gerber
M. A.
,
Marcy
S. M.
,
Murray
D. L.
,
Overturf
G. D.
,
Prober
C. G.
,
Saari
T. N.
, et al
American Academy of Pediatrics Committee on Infectious Diseases and Committee of Fetus and Newborn
.
1998
.
Prevention of respiratory syncytial virus infections: indications for the use of palivizumab and update on the use of RSV-IGIV.
Pediatrics
102
:
1211
1216
.
33
Durant
L. R.
,
Makris
S.
,
Voorburg
C. M.
,
Loebbermann
J.
,
Johansson
C.
,
Openshaw
P. J.
.
2013
.
Regulatory T cells prevent Th2 immune responses and pulmonary eosinophilia during respiratory syncytial virus infection in mice.
J. Virol.
87
:
10946
10954
.
34
Sakaguchi
S.
,
Miyara
M.
,
Costantino
C. M.
,
Hafler
D. A.
.
2010
.
FOXP3+ regulatory T cells in the human immune system.
Nat. Rev. Immunol.
10
:
490
500
.
35
Annacker
O.
,
Asseman
C.
,
Read
S.
,
Powrie
F.
.
2003
.
Interleukin-10 in the regulation of T cell-induced colitis.
J. Autoimmun.
20
:
277
279
.
36
Beiting
D. P.
,
Gagliardo
L. F.
,
Hesse
M.
,
Bliss
S. K.
,
Meskill
D.
,
Appleton
J. A.
.
2007
.
Coordinated control of immunity to muscle stage Trichinella spiralis by IL-10, regulatory T cells, and TGF-β.
J. Immunol.
178
:
1039
1047
.
37
de la Rosa
M.
,
Rutz
S.
,
Dorninger
H.
,
Scheffold
A.
.
2004
.
Interleukin-2 is essential for CD4+CD25+ regulatory T cell function.
Eur. J. Immunol.
34
:
2480
2488
.
38
Read
S.
,
Malmström
V.
,
Powrie
F.
.
2000
.
Cytotoxic T lymphocyte-associated antigen 4 plays an essential role in the function of CD25+CD4+ regulatory cells that control intestinal inflammation.
J. Exp. Med.
192
:
295
302
.
39
Barron
L.
,
Dooms
H.
,
Hoyer
K. K.
,
Kuswanto
W.
,
Hofmann
J.
,
O’Gorman
W. E.
,
Abbas
A. K.
.
2010
.
Cutting edge: mechanisms of IL-2-dependent maintenance of functional regulatory T cells.
J. Immunol.
185
:
6426
6430
.
40
Jin
H.
,
Kang
Y.
,
Zhao
L.
,
Xiao
C.
,
Hu
Y.
,
She
R.
,
Yu
Y.
,
Du
X.
,
Zhao
G.
,
Ng
T.
, et al
.
2008
.
Induction of adaptive T regulatory cells that suppress the allergic response by coimmunization of DNA and protein vaccines.
J. Immunol.
180
:
5360
5372
.
41
Kang
Y.
,
Sun
Y.
,
Zhang
J.
,
Gao
W.
,
Kang
J.
,
Wang
Y.
,
Wang
B.
,
Xia
G.
.
2012
.
Treg cell resistance to apoptosis in DNA vaccination for experimental autoimmune encephalomyelitis treatment.
PLoS One
7
:
e49994
.
42
Veiga-Parga
T.
,
Sehrawat
S.
,
Rouse
B. T.
.
2013
.
Role of regulatory T cells during virus infection.
Immunol. Rev.
255
:
182
196
.
43
Sather
B. D.
,
Treuting
P.
,
Perdue
N.
,
Miazgowicz
M.
,
Fontenot
J. D.
,
Rudensky
A. Y.
,
Campbell
D. J.
.
2007
.
Altering the distribution of Foxp3+ regulatory T cells results in tissue-specific inflammatory disease.
J. Exp. Med.
204
:
1335
1347
.
44
Liston
A.
,
Gray
D. H. D.
.
2014
.
Homeostatic control of regulatory T cell diversity.
Nat. Rev. Immunol.
14
:
154
165
.
45
Loebbermann
J.
,
Durant
L.
,
Thornton
H.
,
Johansson
C.
,
Openshaw
P. J.
.
2013
.
Defective immunoregulation in RSV vaccine-augmented viral lung disease restored by selective chemoattraction of regulatory T cells.
Proc. Natl. Acad. Sci. USA
110
:
2987
2992
.
46
Lee
D. C.
,
Harker
J. A.
,
Tregoning
J. S.
,
Atabani
S. F.
,
Johansson
C.
,
Schwarze
J.
,
Openshaw
P. J.
.
2010
.
CD25+ natural regulatory T cells are critical in limiting innate and adaptive immunity and resolving disease following respiratory syncytial virus infection.
J. Virol.
84
:
8790
8798
.
47
Connors
M.
,
Collins
P. L.
,
Firestone
C. Y.
,
Murphy
B. R.
.
1991
.
Respiratory syncytial virus (RSV) F, G, M2 (22K), and N proteins each induce resistance to RSV challenge, but resistance induced by M2 and N proteins is relatively short-lived.
J. Virol.
65
:
1634
1637
.
48
Nguyen
T. N.
,
Power
U. F.
,
Robert
A.
,
Haeuw
J. F.
,
Helffer
K.
,
Perez
A.
,
Asin
M. A.
,
Corvaia
N.
,
Libon
C.
.
2012
.
The respiratory syncytial virus G protein conserved domain induces a persistent and protective antibody response in rodents.
PLoS One
7
:
e34331
.
49
Stott
E. J.
,
Taylor
G.
,
Ball
L. A.
,
Anderson
K.
,
Young
K. K.
,
King
A. M.
,
Wertz
G. W.
.
1987
.
Immune and histopathological responses in animals vaccinated with recombinant vaccinia viruses that express individual genes of human respiratory syncytial virus.
J. Virol.
61
:
3855
3861
.
50
Magro
M.
,
Mas
V.
,
Chappell
K.
,
Vázquez
M.
,
Cano
O.
,
Luque
D.
,
Terrón
M. C.
,
Melero
J. A.
,
Palomo
C.
.
2012
.
Neutralizing antibodies against the preactive form of respiratory syncytial virus fusion protein offer unique possibilities for clinical intervention.
Proc. Natl. Acad. Sci. USA
109
:
3089
3094
.
51
McLellan
J. S.
,
Chen
M.
,
Leung
S.
,
Graepel
K. W.
,
Du
X.
,
Yang
Y.
,
Zhou
T.
,
Baxa
U.
,
Yasuda
E.
,
Beaumont
T.
, et al
.
2013
.
Structure of RSV fusion glycoprotein trimer bound to a prefusion-specific neutralizing antibody.
Science
340
:
1113
1117
.
52
Tripp
R. A.
,
Jones
L. P.
,
Haynes
L. M.
,
Zheng
H.
,
Murphy
P. M.
,
Anderson
L. J.
.
2001
.
CX3C chemokine mimicry by respiratory syncytial virus G glycoprotein.
Nat. Immunol.
2
:
732
738
.
53
Chirkova
T.
,
Boyoglu-Barnum
S.
,
Gaston
K. A.
,
Malik
F. M.
,
Trau
S. P.
,
Oomens
A. G.
,
Anderson
L. J.
.
2013
.
Respiratory syncytial virus G protein CX3C motif impairs human airway epithelial and immune cell responses.
J. Virol.
87
:
13466
13479
.
54
Harcourt
J.
,
Alvarez
R.
,
Jones
L. P.
,
Henderson
C.
,
Anderson
L. J.
,
Tripp
R. A.
.
2006
.
Respiratory syncytial virus G protein and G protein CX3C motif adversely affect CX3CR1+ T cell responses.
J. Immunol.
176
:
1600
1608
.
55
Cautivo
K. M.
,
Bueno
S. M.
,
Cortes
C. M.
,
Wozniak
A.
,
Riedel
C. A.
,
Kalergis
A. M.
.
2010
.
Efficient lung recruitment of respiratory syncytial virus-specific Th1 cells induced by recombinant bacillus Calmette-Guérin promotes virus clearance and protects from infection.
J. Immunol.
185
:
7633
7645
.
56
Choi
Y.
,
Mason
C. S.
,
Jones
L. P.
,
Crabtree
J.
,
Jorquera
P. A.
,
Tripp
R. A.
.
2012
.
Antibodies to the central conserved region of respiratory syncytial virus (RSV) G protein block RSV G protein CX3C-CX3CR1 binding and cross-neutralize RSV A and B strains.
Viral Immunol.
25
:
193
203
.
57
Haynes
L. M.
,
Caidi
H.
,
Radu
G. U.
,
Miao
C.
,
Harcourt
J. L.
,
Tripp
R. A.
,
Anderson
L. J.
.
2009
.
Therapeutic monoclonal antibody treatment targeting respiratory syncytial virus (RSV) G protein mediates viral clearance and reduces the pathogenesis of RSV infection in BALB/c mice.
J. Infect. Dis.
200
:
439
447
.
58
Jorquera
P. A.
,
Choi
Y.
,
Oakley
K. E.
,
Powell
T. J.
,
Boyd
J. G.
,
Palath
N.
,
Haynes
L. M.
,
Anderson
L. J.
,
Tripp
R. A.
.
2013
.
Nanoparticle vaccines encompassing the respiratory syncytial virus (RSV) G protein CX3C chemokine motif induce robust immunity protecting from challenge and disease.
PLoS One
8
:
e74905
.
59
Li
X.
,
Sambhara
S.
,
Li
C. X.
,
Ettorre
L.
,
Switzer
I.
,
Cates
G.
,
James
O.
,
Parrington
M.
,
Oomen
R.
,
Du
R. P.
,
Klein
M.
.
2000
.
Plasmid DNA encoding the respiratory syncytial virus G protein is a promising vaccine candidate.
Virology
269
:
54
65
.
60
Harcourt
J. L.
,
Karron
R. A.
,
Tripp
R. A.
.
2004
.
Anti-G protein antibody responses to respiratory syncytial virus infection or vaccination are associated with inhibition of G protein CX3C-CX3CR1 binding and leukocyte chemotaxis.
J. Infect. Dis.
190
:
1936
1940
.
61
Mathis
A. S.
,
Egloff
G.
,
Ghin
H. L.
.
2014
.
Calcineurin inhibitor sparing strategies in renal transplantation, part one: late sparing strategies.
World J. Transplant.
4
:
57
80
.
62
Naoumov
N. V.
2014
.
Cyclophilin inhibition as potential therapy for liver diseases.
J. Hepatol.
61
:
1166
1174
.
63
Scheffert
J. L.
,
Raza
K.
.
2014
.
Immunosuppression in lung transplantation.
J. Thorac. Dis.
6
:
1039
1053
.
64
Frickhofen
N.
,
Kaltwasser
J. P.
,
Schrezenmeier
H.
,
Raghavachar
A.
,
Vogt
H. G.
,
Herrmann
F.
,
Freund
M.
,
Meusers
P.
,
Salama
A.
,
Heimpel
H.
The German Aplastic Anemia Study Group
.
1991
.
Treatment of aplastic anemia with antilymphocyte globulin and methylprednisolone with or without cyclosporine.
N. Engl. J. Med.
324
:
1297
1304
.
65
Nakao
S.
,
Takamatsu
H.
,
Chuhjo
T.
,
Ueda
M.
,
Shiobara
S.
,
Matsuda
T.
,
Kaneshige
T.
,
Mizoguchi
H.
.
1994
.
Identification of a specific HLA class II haplotype strongly associated with susceptibility to cyclosporine-dependent aplastic anemia.
Blood
84
:
4257
4261
.
66
Schultz
C.
2014
.
Safety and efficacy of cyclosporine in the treatment of chronic dry eye.
Ophthalmol. Eye Dis.
6
:
37
42
.
67
Stiller
C. R.
,
Dupré
J.
,
Gent
M.
,
Jenner
M. R.
,
Keown
P. A.
,
Laupacis
A.
,
Martell
R.
,
Rodger
N. W.
,
von Graffenried
B.
,
Wolfe
B. M.
.
1984
.
Effects of cyclosporine immunosuppression in insulin-dependent diabetes mellitus of recent onset.
Science
223
:
1362
1367
.
68
Bougneres
P. F.
,
Carel
J. C.
,
Castano
L.
,
Boitard
C.
,
Gardin
J. P.
,
Landais
P.
,
Hors
J.
,
Mihatsch
M. J.
,
Paillard
M.
,
Chaussain
J. L.
,
Bach
J. F.
.
1988
.
Factors associated with early remission of type I diabetes in children treated with cyclosporine.
N. Engl. J. Med.
318
:
663
670
.
69
Zenke
G.
,
Strittmatter
U.
,
Fuchs
S.
,
Quesniaux
V. F. J.
,
Brinkmann
V.
,
Schuler
W.
,
Zurini
M.
,
Enz
A.
,
Billich
A.
,
Sanglier
J. J.
,
Fehr
T.
.
2001
.
Sanglifehrin A, a novel cyclophilin-binding compound showing immunosuppressive activity with a new mechanism of action.
J. Immunol.
166
:
7165
7171
.
70
Stein
C. M.
,
Murray
J. J.
,
Wood
A. J. J.
.
1999
.
Inhibition of stimulated interleukin-2 production in whole blood: a practical measure of cyclosporine effect.
Clin. Chem.
45
:
1477
1484
.
71
Ho
S.
,
Clipstone
N.
,
Timmermann
L.
,
Northrop
J.
,
Graef
I.
,
Fiorentino
D.
,
Nourse
J.
,
Crabtree
G. R.
.
1996
.
The mechanism of action of cyclosporin A and FK506.
Clin. Immunol. Immunopathol.
80
:
S40
S45
.
72
Huai
Q.
,
Kim
H. Y.
,
Liu
Y.
,
Zhao
Y.
,
Mondragon
A.
,
Liu
J. O.
,
Ke
H.
.
2002
.
Crystal structure of calcineurin-cyclophilin-cyclosporin shows common but distinct recognition of immunophilin-drug complexes.
Proc. Natl. Acad. Sci. USA
99
:
12037
12042
.
73
Almawi
W. Y.
,
Melemedjian
O. K.
.
2000
.
Clinical and mechanistic differences between FK506 (tacrolimus) and cyclosporin A.
Nephrol. Dial. Transplant.
15
:
1916
1918
.
74
Hiroyuki
M.
,
Chendong
Y.
,
Keiko
M.
,
Dale
E.
,
Mark
M.
,
Akira
T.
.
2002
.
Contrasting impacts of immunosuppressive agents (rapamycin, FK506, cyclosporin A, and dexamethasone) on bidirectional dendritic cell-T cell interaction during antigen presentation.
J. Immunol.
169
:
3555
3564
.
75
Kawai
M.
,
Kitade
H.
,
Mathieu
C.
,
Waer
M.
,
Pirenne
J.
.
2005
.
Inhibitory and stimulatory effects of cyclosporine A on the development of regulatory T cells in vivo.
Transplantation
79
:
1073
1077
.
76
Yamada
Y.
,
Sekine
Y.
,
Yoshida
S.
,
Yasufuku
K.
,
Petrache
I.
,
Benson
H. L.
,
Brand
D. D.
,
Yoshino
I.
,
Wilkes
D. S.
.
2009
.
Type V collagen-induced oral tolerance plus low-dose cyclosporine prevents rejection of MHC class I and II incompatible lung allografts.
J. Immunol.
183
:
237
245
.
77
De Serres
S. A.
,
Sayegh
M. H.
,
Najafian
N.
.
2009
.
Immunosuppressive drugs and Tregs: a critical evaluation!
Clin. J. Am. Soc. Nephrol.
4
:
1661
1669
.
78
Kang
Y.
,
Xu
L.
,
Wang
B.
,
Chen
A.
,
Zheng
G.
.
2008
.
Cutting edge: immunosuppressant as adjuvant for tolerogenic immunization.
J. Immunol.
180
:
5172
5176
.
79
Zheng
G.
,
Zhong
S.
,
Geng
Y.
,
Munirathinam
G.
,
Cha
I.
,
Reardon
C.
,
Getz
G. S.
,
van Rooijen
N.
,
Kang
Y.
,
Wang
B.
,
Chen
A.
.
2013
.
Dexamethasone promotes tolerance in vivo by enriching CD11cloCD40lo tolerogenic macrophages.
Eur. J. Immunol.
43
:
219
227
.
80
Singh
S. R.
,
Dennis
V. A.
,
Carter
C. L.
,
Pillai
S. R.
,
Jefferson
A.
,
Sahi
S. V.
,
Moore
E. G.
.
2007
.
Immunogenicity and efficacy of recombinant RSV-F vaccine in a mouse model.
Vaccine
25
:
6211
6223
.
81
Reed
L. J.
,
Muench
H.
.
1938
.
A simple method of estimating fifty percent endpoints.
Am. J. Hyg.
27
:
493
497
.
82
Olkhanud
P. B.
,
Baatar
D.
,
Bodogai
M.
,
Hakim
F.
,
Gress
R.
,
Anderson
R. L.
,
Deng
J.
,
Xu
M.
,
Briest
S.
,
Biragyn
A.
.
2009
.
Breast cancer lung metastasis requires expression of chemokine receptor CCR4 and regulatory T cells.
Cancer Res.
69
:
5996
6004
.
83
Biragyn
A.
,
Bodogai
M.
,
Olkhanud
P. B.
,
Denny-Brown
S. R.
,
Puri
N.
,
Ayukawa
K.
,
Kanegasaki
S.
,
Hogaboam
C. M.
,
Wejksza
K.
,
Lee-Chang
C.
.
2013
.
Inhibition of lung metastasis by chemokine CCL17-mediated in vivo silencing of genes in CCR4+ Tregs.
J. Immunother.
36
:
258
267
.
84
Gomez
R. S.
,
Guisle-Marsollier
I.
,
Bohmwald
K.
,
Bueno
S. M.
,
Kalergis
A. M.
.
2014
.
Respiratory syncytial virus: pathology, therapeutic drugs and prophylaxis.
Immunol. Lett.
162
(
1 Pt A
):
237
247
.
85
Ruckwardt
T. J.
,
Bonaparte
K. L.
,
Nason
M. C.
,
Graham
B. S.
.
2009
.
Regulatory T cells promote early influx of CD8+ T cells in the lungs of respiratory syncytial virus-infected mice and diminish immunodominance disparities.
J. Virol.
83
:
3019
3028
.
86
Joetham
A.
,
Takeda
K.
,
Taube
C.
,
Miyahara
N.
,
Matsubara
S.
,
Koya
T.
,
Rha
Y. H.
,
Dakhama
A.
,
Gelfand
E. W.
.
2007
.
Naturally occurring lung CD4+CD25+ T cell regulation of airway allergic responses depends on IL-10 induction of TGF-β.
J. Immunol.
178
:
1433
1442
.
87
Bocian
K.
,
Borysowski
J.
,
Wierzbicki
P.
,
Wyzgal
J.
,
Klosowska
D.
,
Bialoszewska
A.
,
Paczek
L.
,
Górski
A.
,
Korczak-Kowalska
G.
.
2010
.
Rapamycin, unlike cyclosporine A, enhances suppressive functions of in vitro-induced CD4+CD25+ Tregs.
Nephrol. Dial. Transplant.
25
:
710
717
.
88
Miroux
C.
,
Moralès
O.
,
Carpentier
A.
,
Dharancy
S.
,
Conti
F.
,
Boleslowski
E.
,
Podevin
P.
,
Auriault
C.
,
Pancré
V.
,
Delhem
N.
.
2009
.
Inhibitory effects of cyclosporine on human regulatory T cells in vitro.
Transplant. Proc.
41
:
3371
3374
.
89
Stanojlovic
S.
,
Schlickeiser
S.
,
Appelt
C.
,
Vogt
K.
,
Schmitt-Knosalla
I.
,
Haase
S.
,
Ritter
T.
,
Sawitzki
B.
,
Pleyer
U.
.
2010
.
Influence of combined treatment of low dose rapamycin and cyclosporin A on corneal allograft survival.
Graefes Arch. Clin. Exp. Ophthalmol.
248
:
1447
1456
.
90
Brandt
C.
,
Pavlovic
V.
,
Radbruch
A.
,
Worm
M.
,
Baumgrass
R.
.
2009
.
Low-dose cyclosporine A therapy increases the regulatory T cell population in patients with atopic dermatitis.
Allergy
64
:
1588
1596
.
91
Meloni
F.
,
Morosini
M.
,
Solari
N.
,
Bini
F.
,
Vitulo
P.
,
Arbustini
E.
,
Pellegrini
C.
,
Fietta
A. M.
.
2006
.
Peripheral CD4+ CD25+ Treg cell expansion in lung transplant recipients is not affected by calcineurin inhibitors.
Int. Immunopharmacol.
6
:
2002
2010
.
92
Bierer
B. E.
,
Mattila
P. S.
,
Standaert
R. F.
,
Herzenberg
L. A.
,
Burakoff
S. J.
,
Crabtree
G.
,
Schreiber
S. L.
.
1990
.
Two distinct signal transmission pathways in T lymphocytes are inhibited by complexes formed between an immunophilin and either FK506 or rapamycin.
Proc. Natl. Acad. Sci. USA
87
:
9231
9235
.
93
Ghosh
P.
,
Buchholz
M. A.
,
Yano
S.
,
Taub
D.
,
Longo
D. L.
.
2002
.
Effect of rapamycin on the cyclosporin A-resistant CD28-mediated costimulatory pathway.
Blood
99
:
4517
4524
.
94
Riccardo
B.
,
Tatiana
S.
,
Carmela
G.
,
Thomas
P.
,
Jeremy
L.
,
Maurizio
M.
.
2010
.
Cyclosporine A-sensitive, cyclophilin B-dependent endoplasmic reticulum-associated degradation.
PLoS One
5
:
pii: e13008
.
doi:10.1371/journal.pone.0013008
.
95
Cippà
P. E.
,
Kamarashev
J.
,
Chen
J.
,
Kraus
A. K.
,
Segerer
S.
,
Feldmeyer
L.
,
Fehr
T.
.
2013
.
Synergistic Bcl-2 inhibition by ABT-737 and cyclosporine A.
Apoptosis
18
:
315
323
.
96
Heidt
S.
,
Roelen
D. L.
,
Eijsink
C.
,
van Kooten
C.
,
Claas
F. H.
,
Mulder
A.
.
2008
.
Effects of immunosuppressive drugs on purified human B cells: evidence supporting the use of MMF and rapamycin.
Transplantation
86
:
1292
1300
.
97
Jiang
H.
,
Wynn
C.
,
Pan
F.
,
Ebbs
A.
,
Erickson
L. M.
,
Kobayashi
M.
.
2002
.
Tacrolimus and cyclosporine differ in their capacity to overcome ongoing allograft rejection as a result of their differential abilities to inhibit interleukin-10 production.
Transplantation
73
:
1808
1817
.
98
Jiang
A.
,
Craxton
A.
,
Kurosaki
T.
,
Clark
E. A.
.
1998
.
Different protein tyrosine kinases are required for B cell antigen receptor-mediated activation of extracellular signal-regulated kinase, c-Jun NH2-terminal kinase 1, and p38 mitogen-activated protein kinase.
J. Exp. Med.
188
:
1297
1306
.
99
John
C.
,
Mohammed
A.
,
Bin
Y.
,
Jeremy
L.
.
2005
.
Cyclophilin A-deficient mice are resistant to immunosuppression by cyclosporine.
J. Immunol.
174
:
6030
6038
.
100
Matsuda
S.
,
Shibasaki
F.
,
Takehana
K.
,
Mori
H.
,
Nishida
E.
,
Koyasu
S.
.
2000
.
Two distinct action mechanisms of immunophilin-ligand complexes for the blockade of T-cell activation.
EMBO Rep.
1
:
428
434
.
101
Gotot
J.
,
Gottschalk
C.
,
Leopold
S.
,
Knolle
P. A.
,
Yagita
H.
,
Kurts
C.
,
Ludwig-Portugall
I.
.
2012
.
Regulatory T cells use programmed death 1 ligands to directly suppress autoreactive B cells in vivo.
Proc. Natl. Acad. Sci. USA
109
:
10468
10473
.
102
Wang
P.
,
Zheng
S. G.
.
2013
.
Regulatory T cells and B cells: implication on autoimmune diseases.
Int. J. Clin. Exp. Pathol.
6
:
2668
2674
.
103
Zhao
D. M.
,
Thornton
A. M.
,
DiPaolo
R. J.
,
Shevach
E. M.
.
2006
.
Activated CD4+CD25+ T cells selectively kill B lymphocytes.
Blood
107
:
3925
3932
.
104
Fuchs
A.
,
Atkinson
J. P.
,
Fremeaux-Bacchi
V.
,
Kemper
C.
.
2009
.
CD46-induced human Treg enhance B-cell responses.
Eur. J. Immunol.
39
:
3097
3109
.
105
Rapetti
L.
,
Chavele
K. M.
,
Evans
C. M.
,
Ehrenstein
M. R.
.
2015
.
B cell resistance to Fas-mediated apoptosis contributes to their ineffective control by regulatory T cells in rheumatoid arthritis.
Ann. Rheum. Dis.
74
:
294
302
.
106
Zhang
Y.
,
Eliav
Y.
,
Shin
S. U.
,
Schreiber
T. H.
,
Podack
E. R.
,
Tadmor
T.
,
Rosenblatt
J. D.
.
2013
.
B lymphocyte inhibition of anti-tumor response depends on expansion of Treg but is independent of B-cell IL-10 secretion.
Cancer Immunol. Immunother.
62
:
87
99
.
107
Levy
Y.
,
Brouet
J. C.
.
1994
.
Interleukin-10 prevents spontaneous death of germinal center B cells by induction of the bcl-2 protein.
J. Clin. Invest.
93
:
424
428
.
108
Llorente
L.
,
Zou
W.
,
Levy
Y.
,
Richaud-Patin
Y.
,
Wijdenes
J.
,
Alcocer-Varela
J.
,
Morel-Fourrier
B.
,
Brouet
J. C.
,
Alarcon-Segovia
D.
,
Galanaud
P.
,
Emilie
D.
.
1995
.
Role of interleukin 10 in the B lymphocyte hyperactivity and autoantibody production of human systemic lupus erythematosus.
J. Exp. Med.
181
:
839
844
.
109
Rousset
F.
,
Peyrol
S.
,
Garcia
E.
,
Vezzio
N.
,
Andujar
M.
,
Grimaud
J. A.
,
Banchereau
J.
.
1995
.
Long-term cultured CD40-activated B lymphocytes differentiate into plasma cells in response to IL-10 but not IL-4.
Int. Immunol.
7
:
1243
1253
.
110
Yoon
S.-O. O.
,
Zhang
X.
,
Berner
P.
,
Choi
Y. S.
.
2009
.
IL-21 and IL-10 have redundant roles but differential capacities at different stages of plasma cell generation from human germinal center B cells.
J. Leukoc. Biol.
86
:
1311
1318
.
111
Armitage
R. J.
,
Macduff
B. M.
,
Spriggs
M. K.
,
Fanslow
W. C.
.
1993
.
Human B cell proliferation and Ig secretion induced by recombinant CD40 ligand are modulated by soluble cytokines.
J. Immunol.
150
:
3671
3680
.
112
Moens
L.
,
Tangye
S. G.
.
2014
.
Cytokine-mediated regulation of plasma cell generation: IL-21 takes center stage.
Front. Immunol.
5
:
65
.
113
Anderson
R.
,
Huang
Y.
,
Langley
J. M.
.
2010
.
Prospects for defined epitope vaccines for respiratory syncytial virus.
Future Microbiol.
5
:
585
602
.
114
Boyoglu-Barnum
S.
,
Chirkova
T.
,
Todd
S. O.
,
Barnum
T. R.
,
Gaston
K. A.
,
Jorquera
P.
,
Haynes
L. M.
,
Tripp
R. A.
,
Moore
M. L.
,
Anderson
L. J.
.
2014
.
Prophylaxis with a respiratory syncytial virus (RSV) anti-G protein monoclonal antibody shifts the adaptive immune response to RSV rA2-line19F infection from Th2 to Th1 in BALB/c mice.
J. Virol.
88
:
10569
10583
.
115
Johnson
C. H.
,
Miao
C.
,
Blanchard
E. G.
,
Caidi
H.
,
Radu
G. U.
,
Harcourt
J. L.
,
Haynes
L. M.
.
2012
.
Effect of chemokine receptor CX3CR1 deficiency in a murine model of respiratory syncytial virus infection.
Comp. Med.
62
:
14
20
.
116
Tripp
R. A.
,
Dakhama
A.
,
Jones
L. P.
,
Barskey
A.
,
Gelfand
E. W.
,
Anderson
L. J.
.
2003
.
The G glycoprotein of respiratory syncytial virus depresses respiratory rates through the CX3C motif and substance P.
J. Virol.
77
:
6580
6584
.

Chaofan Li, Y.Z., X.Z., Z.H., and B.W. were included as coinventors on the patent “A RSV vaccine preparation and methods of using the same” (application no. 201510082407.8). The other authors have no financial conflicts of interest.

Supplementary data