* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
Download Targeting CD45RB alters T cell migration and delays viral clearance
Survey
Document related concepts
Transcript
International Immunology, Vol. 18, No. 2, pp. 291–300 doi:10.1093/intimm/dxh367 ª The Japanese Society for Immunology. 2005. All rights reserved. For permissions, please e-mail: [email protected] Targeting CD45RB alters T cell migration and delays viral clearance Bock Lim1*, Robyn M. Sutherland2*, Yifan Zhan2, Georgia Deliyannis1, Lorena E. Brown1 and Andrew M. Lew2 1 Department of Microbiology and Immunology and Cooperative Research Centre for Vaccine Technology, University of Melbourne, Royal Parade, Melbourne 3010, Australia 2 Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Melbourne 3050, Australia Keywords: CD45, CTL, homing, influenza, trafficking Abstract CD45 is a receptor tyrosine phosphatase essential for TCR signaling. One isoform, CD45RB, is down-regulated in memory cells and targeting CD45RB with a specific antibody has been shown to inhibit graft rejection. Its role in immunity to infection, however, has not been tested. Here, we report the effect of anti-CD45RB antibody treatment on the induction of anti-influenza CD81 T cells and viral clearance. Anti-CD45RB-treated mice had delayed pulmonary viral clearance compared with untreated mice whose infection was completely cleared by day 8 post-infection. In anti-CD45RBtreated mice, the total CD41 and CD81 T cell numbers in both the lungs and mediastinal nodes were substantially reduced at days 5 and 8; this effect was less marked for the spleen. CD81 T cells specific for influenza virus were also reduced compared with the control group in all three organs at day 8. By day 11, when both treated and control groups showed no virus remaining in the lungs, specific CD81 T cell numbers were at similar low levels. Homing to lymph nodes and lung of dye-labeled T cells was greatly inhibited (by >80%) by anti-CD45RB treatment. This reduced homing corresponded with reduced CD62L and b1-integrin expression in both uninfected and infected mice. Since CD62L plays a critical role in homing lymphocytes to lymph nodes, and high levels of CD62L and a4b1-integrin are expressed by lymphocytes that home to bronchus-associated lymphoid tissue, we suggest that reduced expression of these molecules is a key explanation for the delay in immune responses. Introduction Cell-mediated adaptive immune responses are critical for the recovery of primary influenza infection (1). The virus-specific CD8+ CTLs are thought to be the key effectors in the clearance of the virus especially in the first 8 days (2) and potentially provide heterologous immunity by recognizing epitopes that are highly conserved between different strains of influenza virus (3). CD4+ T-cell help is relatively dispensable in the induction of virus clearing CD8+ T cells during a primary influenza infection (4), although it may be critical for an optimal CD8+ T cell recall response during secondary infection (5–8). As we have focused on the primary infection, we have concentrated our investigations on CD8+ T cells. CD45 is expressed on all nucleated hematopoietic cells and is a receptor tyrosine phosphatase critical for antigen-specific B and T cell responses (9, 10). Due to alternative splicing, *These authors contributed equally. CD45 can exist in multiple isoforms (CD45RA, RB, RC and RO) that can be differentiated by their sizes. Although the functional relevance of these isoforms is not determined, they are highly regulated and T cell activation is associated with a shift from high- to low-molecular weight isoforms. In the mouse, naive peripheral T cells express abundant CD45RB and low-level expression is seen on primed/memory T cells, on cells that show increased secretion of Th2 cytokines, such as IL-4 and IL-10, and on a population of T cells with regulatory function (11, 12). These assignments are useful but may be a slight over-simplification as the CD45 isoform expression is not necessarily fixed (13, 14). Studies in transplantation indicate that treatment with the anti-CD45RB mAb, MB23G2, induces long-term tolerance to allogeneic grafts in mice (15–18). T cell responses are Received 4 March 2005, accepted 8 November 2005 Correspondence to: A. M. Lew; E-mail: [email protected]. L. E. Brown; E-mail: [email protected] Transmitting editor: E. Simpson Advance Access publication 16 December 2005 292 Anti-CD45RB alters T cell homing and viral clearance perturbed by this antibody and the cells remaining after antiCD45RB treatment have little or no CD45RB. One interpretation of these studies could be that the action of the antibody was to deplete CD45RB cells. However, the possibility of altered trafficking or sequestration of CD45RB+ cells could not be discounted. Although anti-CD45RB treatment is not associated with activation (CD69, CD25 and CD44 are not affected), it is associated with increased production of both Th2 cytokines and increased CTLA4 surface expression on CD4+ Tcells (17). This increase in CTLA4 may be critical in mediating tolerance (17) and/or reflect a common splice pathway between CD45 and CTLA4 (19). General immunosuppression by drugs (e.g. cyclosporin, FK506, rapamycin and mycophenolate mofetil) has been used clinically to inhibit transplantation rejection successfully; however, one of the adverse effects of such drugs is increased susceptibility to serious infections (20, 21). Anti-CD45RB appears to be less severe than most immunosuppressive regimens. For example, it does not deplete T cell numbers in the spleen (17, 22). Surprisingly, the effect of anti-CD45RB treatment on viral or bacterial immunity has not been reported. Therefore, we wanted to investigate how such a regimen would affect the host’s responses to adventitious infections, e.g. influenza. Several observations that suggest a link between CD45 expression and trafficking have been reported. Firstly, CD45 has some regulatory role in selectin expression including CD62L (23) and in integrin-mediated adhesion (24). Secondly, although anti-CD45RB results in an increase in splenic T cells, there is also a decrease in circulating lymphocytes (15, 16) and lymph node T cells (22, 25). Such imbalance in cell numbers among the various sites could be interpreted as due to a trafficking aberration. As specific T cells can be more readily quantified in the murine influenza system than in allogeneic systems, the model we have chosen also allows trafficking of antigen-specific T cells to be examined. In this study, the ability of mice following anti-CD45RB treatment to induce effective CD8+ T cell-mediated viral clearing and anti-viral antibody responses during a primary influenza infection is reported. We also relate this to how T cell trafficking is affected after anti-CD45RB treatment. Methods Mice and anti-CD45RB treatment C57BL/6 female mice were obtained from the Department of Microbiology and Immunology, The University of Melbourne (Parkville, Victoria, Australia). CD4-deficient GK transgenic mice (26) on a C57BL/6 background were obtained from the Walter and Eliza Hall Institute (Parkville, Victoria, Australia). The animals were bred and maintained throughout the experiment under specific pathogen-free conditions in the same department. The anti-CD45RB mAb, MB23G2, was purified on immobilized protein G from hybridoma supernatant. Mice were injected intraperitoneally with 0.2 mg of MB23G2, 1 day prior to infection and thereafter every 3–4 days. The control mice were injected with either isotype control (GL117 = rat IgG2a) or PBS. Virus infection The type A strain of influenza virus used in this study was a genetic reassortant of A/Memphis/1/71 (H3N2) 3 A/Bellamy/ 42 (H1N1) referred to as Mem71 (H3N1) virus. The split Mem71 virus for use in ELISA was prepared by rate zonal centrifugation to purify the virus before inactivation with (b-propiolactone and disruption with sodium taurodeoxycholate. Mice were infected intranasally with 104.5 plaque-forming units of Mem71 virus after being anesthetized with penthrane. Each mouse received 50 ll of the virus in the form of allantoic fluid diluted in RPMI-1640. ELISPOT (Enzyme linked immunospot) assay for IFN-c-secreting CD8+ T cells Influenza-specific IFN-c-secreting CD8+ T cells were enumerated by an ELISPOT assay as previously described (27) with 2 lg ml1 of a peptide representing the dominant H-2Dbrestricted epitope NP366–374, ASNENMETM (28). Flow cytometric analysis of lymphocytes Lungs were minced, digested in 2 mg ml1 collagenase A (Roche, Mannheim, Germany) for 30 min at 37C, and then dispersed through a metal sieve. The resultant cell suspensions were pelleted and washed. Liver lymphocytes were obtained as described (29). Briefly, livers were minced and pushed through a metal sieve. Hepatocytes were removed by centrifugation in a 36% solution of isotonic Percoll in PBS, and the pellet containing lymphomyeloid cells and RBC was recovered. RBCs were removed from suspensions of liver, lung and spleen cells by lysis in Tris-buffered ammonium chloride solution. In experiments where liver lymphocytes were analyzed, mice were perfused by injecting 20 ml of PBS into the heart prior to recovery of organs. CD8+ and CD4+ T cells were enumerated by flow cytometry using a FACScan (Becton Dickinson, San Jose, CA, USA) after staining the lymphocytes with FITC-conjugated rat anti-mouse CD8 antibody (clone 53-6.7, Pharmingen, San Diego, CA, USA) and PE-conjugated rat anti-mouse CD4 antibody (clone GK1.5, Pharmingen). Data were analyzed using FlowJo version 3.6.1 (Tree Star Inc., San Carlos, CA, USA). Expression of b7-integrin and L-selectin on CD8+ T cells was examined by co-staining with PE-conjugated anti-CD8 antibody (clone CT-CD8a, Caltag Laboratories, Burlingame, CA, USA) and either FITC-conjugated anti-b7-integrin antibody (Pharmingen, clone M293) or FITC-conjugated anti-L-selectin antibody (clone MEL-14, Pharmingen). Expression of b1-integrin on CD8+ T cells was examined by co-staining with PE-Cy5-conjugated anti-CD8 antibody (clone 53-6.7, Pharmingen) plus FITCconjugated anti-b1-integrin antibody (clone Ha2/5, Pharmingen). Expression of CD45RB was examined with a biotinylated mAb that did not compete with MB23G2 (clone 16A, Pharmingen). Propidium iodide staining was used to exclude dead cells. Cells were analyzed using a FACScan and CellQuest software. ELISA To determine the titer of influenza-specific total and isotype (IgM, IgG1, IgG2c and IgG2b) antibodies in the sera of influenza-infected mice, flat-bottom 96-well polyvinyl chloride Anti-CD45RB alters T cell homing and viral clearance 293 microtiter plates (Dynatech) were coated with 50 ll per well of split Mem71 virus at a concentration of 5 mg ml1 in PBSN3 (30). The secondary antibodies used were 1:400 dilution of HRPconjugated rabbit anti-mouse antibodies (DakoCytomation, Carpinteria, CA, USA) or 1:5000 dilution of HRP-conjugated goat anti-mouse isotyping antibodies (Southern Biotechnology Associates Inc., Birmingham, AL, USA). The substrate used was 0.2 mM of 2-29-azino-bis(3-ethylbenzthiazoline-6-sulfonic acid) (Sigma Chemicals Co.) in 50 mM citrate buffer, pH 4.0, containing 0.004% H2O2 (v/v). The reaction was stopped by adding 50 ll of 45 mM sodium fluoride (Sigma Chemicals Co.) to each well. Plates were read at the dual wavelength of 405 and 450 nm (Multiskan Multisoft Labsystems, Helsinki, Finland). The antibody titer was expressed as the reciprocal of dilution of serum giving an optical density five times higher than the background with normal mouse serum. Plaque assay Infectious virus was quantified by plaque assay conducted on confluent monolayers of Madin–Darby canine kidney cells as previously described (30, 31). Results Clearance of influenza virus in mice after anti-CD45RB treatment To investigate the effect of anti-CD45RB on anti-influenza immunity that leads to viral clearance in the lungs, mice were treated intraperitoneally with 0.2 mg of anti-CD45RB 1 day prior to intranasal infection of influenza virus, and every 3 days thereafter. Control mice were either injected with PBS or the control isotype GL117; both controls behaved similarly. The ability of these mice to clear the pulmonary virus was examined 5, 8 and 11 days post-infection by plaque assay on samples from the lungs. On day 5, prior to the recruitment of CD8+ effector T cells to the lungs, viral titers were high in both groups (Fig. 1). By day 8, control mice had completely cleared the virus in their lungs (Fig. 1). In contrast, mice treated with antiCD45RB showed a delayed viral clearance, with individual mice displaying a range of viral titers in their lungs. All the treated mice ultimately cleared the virus by day 11 postinfection. Thus, anti-CD45RB hindered but did not abrogate the ability of the mice to clear a pulmonary influenza infection. Homing of CD8+ T cells labeled with carboxyfluorescein diacetate succinimidyl ester after anti-CD45RB treatment Expansion of lymphocytes in the mice following influenza infection A single-cell suspension was made from inguinal, axillary, popliteal, renal, iliac and mediastinal lymph nodes (MLN) recovered from C57BL/6 mice. The cells were washed twice with 0.1% BSA (CSL Ltd) in PBS before labeling with 10 lM 5,6-carboxyfluorescein diacetate succinimidyl ester (CFSE, Molecular Probes Inc., Eugene, OR, USA). After 10 min at 37C in the presence of CFSE, the cells were washed twice with medium and twice with PBS. CFSE-labeled cells (1.3 3 107) in PBS were injected intravenously into each C57BL/6 mouse, followed immediately by either MB23G2 or PBS delivered intraperitoneally. The mice were infected on the following day with Mem71. On day 5 post-infection, spleen, liver, lung and MLN and inguinal lymph nodes (ILN) were harvested from mice. The total number of CFSE-labeled CD8+ T cells in the various organs was determined by flow cytometry after staining with PE-conjugated rat anti-mouse CD8 antibodies (clone 53-6.7, Pharmingen). During infection, the rapid proliferation of lymphocytes is important for an efficient recovery from pulmonary influenza. Here, in an attempt to explain the delayed viral clearance in mice treated with anti-CD45RB, the expansion of the lymphocytes in response to the infection was examined. The total numbers of CD4+ T cells and CD8+ T cells in the MLN, lungs and spleen of mice on days 5, 8 and 11 are shown in Fig. 2. On day 5 post-infection, in the MLN and lungs, the total numbers of lymphocytes were significantly higher for control mice than for anti-CD45RB-treated mice. This early expansion of lymphocytes in the control mice is consistent with their ability to completely clear virus within the next 3 days. As expected, on day 8 post-infection, control mice that had just cleared the virus had very high numbers of CD4+ and CD8+ lymphocytes in the lung and MLN and these levels had decreased by day 11 in the absence of further stimulus. The rise in T cell numbers on day 8 post-infection was particularly notable in the lungs In vitro treatment of CD8 T cells with anti-CD45RB Pooled lymph node cells from the ILN, axilliary, popliteal and mesenteric lymph nodes of CD4 T cell-deficient GK mice were enriched for CD8 T cells by passing twice over nylon wool columns. Cells (4 3 106) in 1 ml of RPMI supplemented with 10% (v/v) heat-inactivated FCS and 50 lM 2-mercaptoethanol were added to a 24-well tissue culture plate. Either MB23G2 anti-CD45RB or GL117 isotype control antibody was added at a final concentration of 0.2 lg ml1 and cultures were incubated for 22 h at 37C. At the conclusion of culture, cells were recovered for FACS analysis. Statistical analysis Statistical analysis was performed using the non-parametric Mann–Whitney U test on the Prism 4 software program (GraphPad Software Inc., San Diego, CA, USA). Fig. 1. Anti-CD45RB treatment delays virus clearance. Influenza virus load in the lungs of control and anti-CD45RB-treated mice was determined on days 5, 8 and 11 after intranasal infection. The geometric mean titer of a group is shown as a line, and each symbol represents an individual mouse. Log10 1.6 is the minimum detectable titer. 294 Anti-CD45RB alters T cell homing and viral clearance Fig. 2. The total number of T cells in lymph nodes and lungs can be reduced by anti-CD45RB treatment. Total numbers of CD4+ and CD8+ T cells recovered from the MLN, lung and spleen of control and anti-CD45RB-treated mice on days 5, 8 and 11 post-infection. The mean is shown as a line, and each symbol represents an individual mouse. where the viral clearing activity was taking place (CD4+ T cells, P = 0.008; CD8+ T cells, P = 0.008). When the mice were treated with anti-CD45RB, fewer T cells were present in both MLN and lung. This deficit was particularly dramatic in the MLN (day 8: CD4+ T cells, P = 0.008; CD8+ T cells, P = 0.016 and day 11: CD4+ T cells, P = 0.016; CD8+ T cells, P = 0.008). In contrast to the MLN and lung, the difference in T cell numbers in the spleens between anti-CD45RB-treated and untreated mice through most of the study was unremarkable. Only for the CD8+ T cells and only on day 8 post-infection (when there was also a maximum increase in antigen-specific CD8+ T cells; Fig. 3) was there a significant difference. The general lack of changes in total T cell numbers in the spleen is consistent with recent reports that anti-CD45RB treatment did not decrease the splenic T cell numbers (17, 22). This modest effect on spleen compared with lymph nodes was one of the early indications that there may be altered trafficking. Influenza-specific CD8+ T cell responses in the mice Early control of primary influenza infection involves CD8+ T cell-mediated immunity. Influenza-specific IFN-c-secreting CD8+ T cells were enumerated in the MLN, lungs and spleen of the mice on the various days after viral infection using an ELISPOTassay (Fig. 3). The numbers of antigen-specific CD8+ T cells increased considerably between day 5 and day 8 postinfection in all organs. On day 8, the control mice had an abundance of influenza-specific IFN-c-secreting CD8+ T cells in their MLN, lungs and spleen, correlating with their ability to clear the virus (Fig. 1). However, mice treated with antiCD45RB had significantly fewer influenza-specific IFN-csecreting CD8+ T cells in the MLN (P = 0.008), lungs (P = 0.032) and spleen (P = 0.008) as compared with the controls. These data parallel the reduced total CD8+ T cell numbers (Fig. 2) in the MLN and lungs of anti-CD45RB-treated mice. Nevertheless, influenza-specific IFN-c-secreting CD8+ T cells were present in the virus-infected lungs of all the mice. In order to have an estimate of what proportion of CD8+ T cells was activated to secrete IFN-c, we divided the ELISPOT numbers in Fig. 3 by the total CD8+ Tcell numbers in Fig. 2 (Table 1). The numbers are so low on day 5 that they essentially correspond to uninfected mice. There were no statistical differences in the proportion of specific T cells to total CD8+ T cells for treated Anti-CD45RB alters T cell homing and viral clearance 295 compared with control mice in the various organs on day 8. By day 11 in the MLN, this proportion was actually higher for antiCD45RB-treated mice (11% versus 1%; P < 0.01), even though the total numbers of influenza-specific cells in the organ were similar between treated and control mice (Fig. 3). The finding that total CD8+ T cell numbers were dramatically reduced by anti-CD45RB without a substantial decrease in the proportion of specific versus total CD8+ T cells argues against any major effect of anti-CD45RB on T cell activation, but would be consistent with a lack of recruitment. By day 11 post-infection, the numbers of influenza-specific IFN-c-secreting CD8+ T cells in the lungs in untreated mice were greatly reduced, as the immune response subsided after viral clearance (Fig. 1). In the anti-CD45RB-treated mice, there was a modest increase of anti-viral CD8+ T cells such that on day 11 there were more CD8+ T cells in the anti-CD45RB mice than the controls. This could be due in part to the delayed kinetics of the immune response in anti-CD45RB-treated mice, but could also be in part due to the infection lasting beyond day 8 in these mice. In conclusion, the observed diminished CD8+ T cell response after anti-CD45RB treatment on day 8 post-infection correlates with the delayed viral clearance in these mice. However, all anti-CD45RB-treated mice had ultimately cleared the pulmonary influenza by day 11 postinfection, despite the lack of a strong peak in CD8+ T cell number as observed in control mice on day 8 post-infection. Anti-influenza humoral response in the mice Fig. 3. Anti-CD45RB treatment reduces total influenza-specific CD8+ T cells. Influenza-specific IFN-c-secreting CD8+ T cells in the MLN, lung and spleen of control and anti-CD45RB-treated mice on days 5, 8 and 11 post-infection were enumerated by ELISPOT assay and expressed as numbers per organ. Error bars represent standard deviation of the mean. The developing antibody response is known to be important in aiding recovery at later time points after infection. Since CD45 is known to play a role in antigen-specific B cell responses, treatment with anti-CD45RB might affect the humoral response against the infection, which in turn could influence the level of viral clearance. Hence, the total anti-influenza antibody in the sera of these mice was determined using ELISA and the results are illustrated in Fig. 4. On day 5 post-infection, no influenza-specific antibody was detected in either group of mice. On day 8 post-infection, mice treated with anti-CD45RB had significantly less antibody against influenza in their sera compared with the untreated controls. However, on day 11 post-infection, the total anti-influenza antibody titers in the anti-CD45RB-treated mice had risen dramatically (10-fold increase; P = 0.008). The profiles of IgM, IgG1, IgG2b and IgG2c (32) isotypes within these sera were also examined but no significant differences between the anti-CD45RBtreated mice and PBS-treated controls were apparent (data Table 1. Number of ELISPOTs per 100 CD8+ T cells MLN, lungs and spleens of mice treated with anti-CD45RB as compared with controls (n ¼ 6) Post-infection Treatment MLN 5 days PBS Anti-CD45RB PBS Anti-CD45RB PBS Anti-CD45RB 0.2 0.1 1.1 0.6 0.7 11.3 8 days 11 days Lung 6 6 6 6 6 6 0.1 0.1 (P ¼ 0.02) 0.5 0.5 (P ¼ 0.10) 0.4 6.6 (P ¼ 0.01) 2.2 1.2 63.5 131.4 38.2 51.6 Spleen 6 6 6 6 6 6 2.8 1.7 (P ¼ 0.55) 33.1 102.7 (P ¼ 0.22) 38.5 27.6 (P ¼ 0.31) 0.02 0.03 1.1 0.7 0.4 0.3 6 6 6 6 6 6 0.01 0.02 (P ¼ 0.31) 0.6 0.3 (P ¼ 0.31) 0.2 0.2 (P ¼ 0.84) 296 Anti-CD45RB alters T cell homing and viral clearance Fig. 4. Anti-CD45RB treatment delays the humoral response. Total influenza-specific antibody was determined by ELISA. Sera from control and anti-CD45RB-treated mice were collected on days 5, 8 and 11 post-infection and compared with normal mouse serum (nms). The geometric mean titer and standard deviation are indicated. not shown). Taken together, these data propose that antiCD45RB treatment delays the induction of the humoral response against influenza virus and suggest a possible role of influenza-specific antibodies in the clearance of virus observed in the anti-CD45RB-treated mice on day 11 postinfection in the absence of a strong CD8+ T cell response. Altered T cell homing after anti-CD45RB treatment We have shown above that the numbers of lymphocytes (especially CD8+ T cells) that gather at the site of infection are dramatically reduced. The lymphocytes that home to the bronchus-associated lymphoid tissue have high levels of CD62L and a4b1-integrin on their surface (33), whereas guthoming lymphocytes bear high levels of a4b7-integrin (34). However, there is no such preference in the spleen, so we decided to investigate the effect of anti-CD45RB antibody treatment on the homing molecules CD62L (L-selectin), b7 and b1 on CD8+ T cells in this organ. On day 8 after infection, antiCD45RB treatment resulted in far fewer cells bearing high levels of these homing molecules: for CD62L from 66 6 5% to 39 6 4% and for b1-integrin from 35 6 3% to 22 6 7% (Fig. 5). In uninfected mice (n = 3), the reduction was, respectively, from 87 6 3% to 44 6 4% and from 12 6 1% to 9 6 1%. Interestingly, cells with high levels of b7-integrin are also reduced, indicating that perhaps homing to the gut might also have been altered. The effect of the anti-CD45RB treatment on homing molecules was not simply due to deletion of CD45RB cells. Firstly, there was little reduction of CD45RBhi cells in the spleen (data not shown). Even in the lymph nodes, there was a substantial proportion (31.6%) of CD45RB+ cells remaining after anti-CD45RB treatment versus the 67.8% in control mice (Fig. 6). In contrast, only 11.9% of cells remained CD62L+ (Fig. 6) and therefore most CD45RB+ cells must have downmodulated their CD62L. To determine the effect of anti-CD45RB antibody on homing more directly, we tracked dye-labeled CD8+ T cells in vivo. CD8+ T cells from naive mice were labeled with the dye CFSE and injected intravenously. The numbers of CFSElabeled cells that tracked to the spleen, lung, draining MLN, Fig. 5. Anti-CD45RB treatment reduces the percentage of CD8+ T cells bearing high levels of CD62L or b1- or b7-integrins. Spleen cells were recovered from control and anti-CD45RB-treated mice 8 days post-infection. Representative plots are shown for one of three mice in each treatment group, and are gated on viable CD8+ T cells. Fig. 6. Anti-CD45RB treatment reduces CD62L expression on lymphocytes expressing high levels of CD45RB. Lymph nodes were recovered 3 days after treatment with control isotype or anti-CD45RB antibody. CD45RB staining was performed using the 16A antiCD45RB antibody which does not compete with the MB23G2 antiCD45RB antibody used for in vivo treatment. Plots are gated on viable lymphocytes. Anti-CD45RB alters T cell homing and viral clearance 297 non-draining ILN and liver all decreased with anti-CD45RB treatment (Fig. 7). Although the untreated donor CD8 T cells appeared homogeneous with respect to expression of CD45RB and CD62L (Fig. 7a), treatment with anti-CD45RB antibody preferentially reduced their ability to traffic to lungs and lymph node compared with spleen. Thus, in infected mice, anti-CD45RB treatment reduced trafficking to the spleen by 53% compared with a reduction in excess of 80% in lungs and lymph node (lung, 84%; MLN, 81%; ILN, 90%; Fig. 7b). Similarly, in uninfected mice, there was a 69% reduction in spleen compared with a reduction of at least 87% in lungs and lymph node (lung, 87%; MLN, 95%; ILN, 97%; Fig. 7b). Anti-CD45RB treatment directly reduces expression of CD62L on CD8 T cells The overall reduction in CD8 T cell recovery in anti-CD45RBtreated mice (Fig. 7) indicated that this antibody can deplete CD8 T cells or results in their being sequestered in an as yet unidentified site. Thus, it was important to determine if antiCD45RB simply depletes/sequesters naive CD8+T cells expressing high levels of CD62L/CD45RB or is also able to directly affect expression of CD62L. CD62L levels were clearly decreased upon in vitro incubation of purified CD8 T cells with anti-CD45RB antibody (Fig. 8). Notably, a concurrent decrease in CD45RB expression was not observed (Fig. 8). Discussion The effect of the anti-CD45RB mAb, MB23G2, on primary influenza infection was studied here. Virus-specific CD8+ Fig. 7. Anti-CD45RB treatment dramatically reduces CD8+ T cell trafficking to lymph nodes and lung. Lymph node cells were harvested from naive syngeneic female donors and analyzed for CD62L and CD45RB expression on CD8 cells (Fig. 7a). The donor cells were then labeled with CFSE dye and adoptively transferred to mice immediately prior to either anti-CD45RB or isotype control antibody treatment. The mice were infected on the following day. The spleen, lungs, MLN, ILN and liver of recipient mice were recovered on day 6 post-transfer and analyzed by flow cytometry. The number of CFSE-labeled CD8+ T cells in anti-CD45RB mice was expressed as a percentage of the number in isotype control–treated mice. The means and standard deviations of absolute numbers in isotype control–treated mice were as follows—infected: spleen, 57 269 (19 324); lung, 1882 (747); MLN, 4740 (3128); ILN, 4667 (407), and liver, 472 (120); and uninfected: spleen, 65 091 (11 337); lung, 1032 (334); MLN, 90 (52); ILN, 19 096 (14 854), and liver, 743 (352). Fig. 8. In vitro treatment with anti-CD45RB antibody results in decreased expression of CD62L on CD8 T cells. Purified CD8 T cells were incubated with 0.2 lg ml1 of either MB23G2 anti-CD45RB (open histogram) or GL117 isotype control (closed histogram) antibody for 22 h. FACS analysis was performed at the conclusion of culture. Data shown for CD8+propidium iodide gated lymphocytes. 298 Anti-CD45RB alters T cell homing and viral clearance T cells are known to be critical for the recovery of the early phase of influenza infection (2). In the murine influenza pneumonia model at the time of virus clearance, CD8+ T cells dominate the cellular population in the bronchoalveolar lavage (35). Mice that were treated with anti-CD45RB demonstrated a delayed viral clearance response in which significant viral load could be detected in the lungs of these mice on day 8 postinfection, while the control mice had completely cleared the pulmonary influenza. This delayed clearance corresponded with the significant reduction in numbers of influenza-specific CD8+ T cells in the lungs and MLN of the anti-CD45RB-treated mice. Our findings suggest that the anti-influenza-specific CD8+ T cell response leading to viral clearance is altered. This is concordant with studies that reported the perturbation of T cell response after anti-CD45RB treatment in allograft models (15, 16). The most marked effect of anti-CD45RB treatment in our study was the reduction in lymphocyte numbers in the MLN. In contrast, in the spleen, numbers were either unaltered or modestly reduced (Fig. 2). This resulted also in reduced numbers of influenza-specific CD8+ T cells in the whole MLN, but as a percentage of CD8+ T cells, there was no significant reduction (Table 1). This hinted that cell activation per se was not markedly affected by anti-CD45RB treatment and that the effect may be due to reduced naive cell trafficking to the regional lymph node for priming. For confirmation, we showed that the numbers of lymphocytes with high levels of the adhesion molecules associated with homing to lymph nodes were reduced (Fig. 5). The homing molecules we considered, L-selectin, a4b1 and a4b7, are important for homing from the blood to the high endothelial vessels of MLN (33); the latter two are also important in trafficking to the lungs (36). Less is known about what adhesion molecules are required (if any) for lymphocytes from afferent lymph; however, they comprise only 10% of lymphocytes entering lymph nodes, whereas blood lymphocytes comprise >85% (37). We contend that the reduction in cells expressing these homing molecules is not merely due to depletion/sequestration of naive cells. Firstly, there is very little depletion in the spleen. Secondly, the moderate reduction in numbers of CD45RB+ cells in lymph nodes cannot explain the dramatic reduction in numbers of CD62L+ cells (Fig. 6). Thirdly, CD62L expression was reduced under conditions where neither preferential depletion nor sequestration of CD45RB-high cells occurred viz. in vitro incubation with anti-CD45RB antibody (Fig. 8). Fourthly, the anti-CD45RB antibody used is a rat IgG2a and this isotype is inefficient at depletion (38), probably because its hinge region does not contain the Leu-Leu-Gly-Gly-Pro motif (39). As well as the surface phenotype studies, functional studies of homing by CFSE-labeled lymphocytes (Fig. 7) showed that the greatest reduction of homing was to the lymph node after anti-CD45RB treatment. The impaired recruitment of naive cells entering the nodes thus explains the dramatic decrease in CD4+ and CD8+ T cell numbers in the MLN of the antiCD45RB-treated mice (Fig. 2). This in turn would lead to reduced numbers of specific T cells being primed and hence (at least in part) delayed viral clearance. In support of our findings, a single dose of anti-L-selectin antibody treatment has been shown to delay the response to Sendai virus (40). Interestingly, mice that transgenically expressed only CD45RO did not have any alteration in lymphocyte distribution and were able to clear influenza infection by day 9 (41). We assume that some compensation has happened during lymphocyte development that allowed normal homing. Lymphocyte trafficking as a modality for controlling immune responses has received much attention recently, especially with the uncovering of the molecular basis of the immunosuppressive drug FTY720 (42, 43). This drug blocks egress of lymphocytes from lymph nodes by its action on the sphingosine1-phosphate 1 receptor. In contrast to egress, we have shown that anti-CD45RB antibody treatment results in reduced ingress of lymphocytes to lymph nodes. We therefore speculate that the combination of FTY720 and anti-CD45RB would have an even more dramatic effect on immune responses. Interestingly, by day 11 post-infection, mice treated with anti-CD45RB ultimately cleared the pulmonary infection. Although the homing mechanism to the draining MLN of the lungs appears affected after anti-CD45RB treatment, the suboptimal CD8+ T cell response that was observed in these animals could have been mostly induced in the spleen (44). This splenic population of activated CD8+ T cells might be sufficient to generate CTL effectors needed for viral clearance eventually. This is because the optimal CD8+ T cell response seen in control mice is probably in excess and a more modest response might be able to ultimately control the pulmonary influenza (45). The increase of anti-influenza antibody in antiCD45RB-treated mice on day 11 post-infection could also aid in viral clearance, as the humoral response is also known to play a role in the recovery of influenza (46, 47). The effect of anti-CD45RB treatment on the humoral response against the influenza infection was also analyzed here. Mice treated with anti-CD45RB showed a delayed humoral response on day 8 post-infection but total antiinfluenza antibody titer increased dramatically by day 11 postinfection. Though CD45 is known to play a role in B cell activation by mediating signals via the B cell antigen receptor (9), the mechanism in which anti-CD45RB treatment affects the humoral response is not well studied. As B cells share many of the homing receptors of T cells (e.g. CD62L, a4b1), the treatment of anti-CD45RB could also have impaired the trafficking of B cells and resulted in the delayed humoral response seen. We did not find significant differences in the isotypes of the total anti-influenza antibody between the antiCD45RB-treated mice and the controls, even though antiCD45RB treatment has been shown to enhance the production of Th2 cytokines (15, 25). Interestingly, treatment of anti-CD45RB has been reported to show greater immunosuppression and enhanced graft tolerance when accompanied with the blockade of B7/CD28 and CD40/CD40L co-stimulatory pathways (22, 25). Moreover, anti-CD45RB treatment is shown to up-regulate CTLA4 expression on T cells that block B7/CD28 co-stimulation (17). To prevent allograft rejection in clinical transplantation, immunosuppression is mediated usually by a combination of drugs (e.g. FK506, mycophenolate mofetil) that are broad spectrum. As such, one of the adverse effects is increased susceptibility to infections (20, 21). Thus, one of the main aims in transplantation is to have more selective immunosuppression and anti-CD45RB may be a candidate. If anti-CD45RB were ever to be used clinically in transplant patients, it would Anti-CD45RB alters T cell homing and viral clearance 299 be useful to know whether the host could resist infections. We have shown here that although influenza clearance was slower (by up to 3 days), the host was still able to eliminate the infection. Thus, anti-CD45RB may be a more strategic form of immunosuppression than the current drugs employed. The treatment of anti-CD45RB has been demonstrated here to impede viral clearance of pulmonary influenza by affecting the induction of an optimal CD8+ T cell response and causing a delay in the humoral response. Although the pulmonary infection was eventually cured in the absence of an optimal induction of CD8+ T cell response, this should be seen in the light of primary influenza infection. Recent experiments have shown that optimal induction of memory CD8+ T cells requires the interplay of various immune components such as CD4+ T cells and appropriate co-stimulation (5, 6, 8, 48, 49). This is vital for vaccination schemes to provide long-term protection. Thus, it will be interesting to examine the effect of anti-CD45RB treatment on secondary viral infection, although long-term studies with the anti-CD45RB mAb would be difficult, as mice eventually make antibody against the rat mAb rendering it ineffective. The insights on the roles of CD45RB in the induction of immune response will define important parameters in both transplantation and vaccination. In summary, this study indicates that CD45RBhi cells are critical in the optimal induction of influenza-specific CD8+ T cells response but are not mandatory for viral clearance. Acknowledgements This work was supported by Juvenile Diabetes Research Foundation, National Health and Medical Research Council of Australia and the Australian Government via its Cooperative Research Centres Program. B.L. is a recipient of the Australian Postgraduate Award. Abbreviations CFSE ILN MLN 5,6-carboxyfluorescein diacetate succinimidyl ester inguinal lymph nodes mediastinal lymph nodes References 1 Doherty, P. C., Topham, D. J., Tripp, R. A., Cardin, R. D., Brooks, J. W. and Stevenson, P. G. 1997. Effector CD4+ and CD8+ T-cell mechanisms in the control of respiratory virus infections. Immunol. Rev. 159:105. 2 Zinkernagel, R. M. and Doherty, P. C. 1979. MHC-restricted cytotoxic T cells: studies on the biological role of polymorphic major transplantation antigens determining T-cell restrictionspecificity, function, and responsiveness. Adv. Immunol. 27:51. 3 Effros, R. B., Doherty, P. C., Gerhard, W. and Bennink, J. 1977. Generation of both cross-reactive and virus-specific T-cell populations after immunization with serologically distinct influenza A viruses. J. Exp. Med. 145:557. 4 Allan, W., Tabi, Z., Cleary, A. and Doherty, P. C. 1990. Cellular events in the lymph node and lung of mice with influenza. Consequences of depleting CD4+ T cells. J. Immunol. 144:3980. 5 Riberdy, J. M., Christensen, J. P., Branum, K. and Doherty, P. C. 2000. Diminished primary and secondary influenza virus-specific CD8(+) T-cell responses in CD4-depleted Ig(/) mice. J. Virol. 74:9762. 6 Belz, G. T., Wodarz, D., Diaz, G., Nowak, M. A. and Doherty, P. C. 2002. Compromised influenza virus-specific CD8(+)-T-cell memory in CD4(+)-T-cell-deficient mice. J. Virol. 76:12388. 7 Janssen, E. M., Lemmens, E. E., Wolfe, T., Christen, U., von Herrath, M. G. and Schoenberger, S. P. 2003. CD4+ T cells are required for secondary expansion and memory in CD8+ T lymphocytes. Nature 421:852. 8 Sun, J. C. and Bevan, M. J. 2003. Defective CD8 T cell memory following acute infection without CD4 T cell help. Science 300:339. 9 Penninger, J. M., Irie-Sasaki, J., Sasaki, T. and Oliveira-dosSantos, A. J. 2001. CD45: new jobs for an old acquaintance. Nat. Immunol. 2:389. 10 Hermiston, M. L., Xu, Z. and Weiss, A. 2003. CD45: a critical regulator of signaling thresholds in immune cells. Annu. Rev. Immunol. 21:107. 11 Bottomly, K., Luqman, M., Greenbaum, L. et al. 1989. A monoclonal antibody to murine CD45R distinguishes CD4 T cell populations that produce different cytokines. Eur. J. Immunol. 19:617. 12 Lee, W. T., Yin, X. M. and Vitetta, E. S. 1990. Functional and ontogenetic analysis of murine CD45Rhi and CD45Rlo CD4+ T cells. J. Immunol. 144:3288. 13 Michie, C. A., McLean, A., Alcock, C. and Beverley, P. C. 1992. Lifespan of human lymphocyte subsets defined by CD45 isoforms. Nature 360:264. 14 Sparshott, S. M. and Bell, E. B. 1994. Membrane CD45R isoform exchange on CD4 T cells is rapid, frequent and dynamic in vivo. Eur. J. Immunol. 24:2573. 15 Basadonna, G. P., Auersvald, L., Khuong, C. Q. et al. 1998. Antibody-mediated targeting of CD45 isoforms: a novel immunotherapeutic strategy. Proc. Natl Acad. Sci. USA 95:3821. 16 Lazarovits, A. I., Poppema, S., Zhang, Z. et al. 1996. Prevention and reversal of renal allograft rejection by antibody against CD45RB. Nature 380:717. 17 Fecteau, S., Basadonna, G. P., Freitas, A., Ariyan, C., Sayegh, M. H. and Rothstein, D. M. 2001. CTLA-4 up-regulation plays a role in tolerance mediated by CD45. Nat. Immunol. 2:58. 18 Ariyan, C., Salvalaggio, P., Fecteau, S. et al. 2003. Cutting edge: transplantation tolerance through enhanced CTLA-4 expression. J. Immunol. 171:5673. 19 Rothrock, C., Cannon, B., Hahm, B. and Lynch, K. W. 2003. A conserved signal-responsive sequence mediates activationinduced alternative splicing of CD45. Mol. Cell 12:1317. 20 Avery, R. K. 2004. Prophylactic strategies before solid-organ transplantation. Curr. Opin. Infect. Dis. 17:353. 21 Soldatou, A. and Davies, E. G. 2003. Respiratory virus infections in the immunocompromised host. Paediatr. Respir. Rev. 4:193. 22 Sutherland, R. M., McKenzie, B. S., Zhan, Y. et al. 2002. AntiCD45RB antibody deters xenograft rejection by modulating T cell priming and homing. Int. Immunol. 14:953. 23 Wroblewski, M. and Hamann, A. 1997. CD45-mediated signals can trigger shedding of lymphocyte L-selectin. Int. Immunol. 9:555. 24 Shenoi, H., Seavitt, J., Zheleznyak, A., Thomas, M. L. and Brown, E. J. 1999. Regulation of integrin-mediated T cell adhesion by the transmembrane protein tyrosine phosphatase CD45. J. Immunol. 162:7120. 25 Rothstein, D. M., Livak, M. F., Kishimoto, K. et al. 2001. Targeting signal 1 through CD45RB synergizes with CD40 ligand blockade and promotes long term engraftment and tolerance in stringent transplant models. J. Immunol. 166:322. 26 Zhan, Y., Corbett, A. J., Brady, J. L., Sutherland, R. M. and Lew, A. M. 2000. CD4 help-independent induction of cytotoxic CD8 cells to allogeneic P815 tumor cells is absolutely dependent on costimulation. J. Immunol. 165:3612. 27 Deliyannis, G., Jackson, D. C., Ede, N. J., Zeng, W., Hourdakis, I., Sakabetis, E. and Brown, L. E. 2002. Induction of long-term memory CD8(+) T cells for recall of viral clearing responses against influenza virus. J. Virol. 76:4212. 28 Flynn, K. J., Belz, G. T., Altman, J. D., Ahmed, R., Woodland, D. L. and Doherty, P. C. 1998. Virus-specific CD8+ T cells in primary and secondary influenza pneumonia. Immunity 8:683. 29 Bertolino, P., Bowen, D. G., McCaughan, G. W. and Fazekas de St Groth, B. 2001. Antigen-specific primary activation of CD8+ T cells within the liver. J. Immunol. 166:5430. 300 Anti-CD45RB alters T cell homing and viral clearance 30 Deliyannis, G., Boyle, J. S., Brady, J. L., Brown, L. E. and Lew, A. M. 2000. A fusion DNA vaccine that targets antigen-presenting cells increases protection from viral challenge. Proc. Natl Acad. Sci. USA 97:6676. 31 Tannock, G. A., Paul, J. A. and Barry, R. D. 1984. Relative immunogenicity of the cold-adapted influenza virus A/Ann Arbor/6/ 60 (A/AA/6/60-ca), recombinants of A/AA/6/60-ca, and parental strains with similar surface antigens. Infect. Immun. 43:457. 32 Martin, R. M., Brady, J. L. and Lew, A. M. 1998. The need for IgG2c specific antiserum when isotyping antibodies from C57BL/6 and NOD mice. J. Immunol. Methods 212:187. 33 Xu, B., Wagner, N., Pham, L. N. et al. 2003. Lymphocyte homing to bronchus-associated lymphoid tissue (BALT) is mediated by L-selectin/PNAd, alpha4beta1 integrin/VCAM-1, and LFA-1 adhesion pathways. J. Exp. Med. 197:1255. 34 Berlin, C., Berg, E. L., Briskin, M. J. et al. 1993. Alpha 4 beta 7 integrin mediates lymphocyte binding to the mucosal vascular addressin MAdCAM-1. Cell 74:185. 35 Doherty, P. C., Topham, D.J. and Tripp, R. A. 1996. Analysing the T cells response to influenza viruses in mouse model systems. In Brown, L. E., Hampson, A. W. and Webster R. G., eds, Options for the Control of Influenza III, p. 153. Elsevier Science B.V., Amsterdam. 36 Lobb, R. R., Abraham, W. M., Burkly, L. C. et al. 1996. Pathophysiologic role of alpha 4 integrins in the lung. Ann. N. Y. Acad. Sci. 796:113. 37 Hall, J. G. and Morris, B. 1965. The origin of the cells in the efferent lymph from a single lymph node. J. Exp. Med. 121:901. 38 Cobbold, S. P., Jayasuriya, A., Nash, A., Prospero, T. D. and Waldmann, H. 1984. Therapy with monoclonal antibodies by elimination of T-cell subsets in vivo. Nature 312:548. 39 Woof, J. M. 1990. Fc receptors and their interaction with antibodies. Biochem. Soc. Trans. 18:217. 40 Hou, S., Hyland, L., Bradley, L. M., Watson, S. R. and Doherty, P. C. 1995. Subverting lymph node trafficking by treatment with the Mel-14 monoclonal antibody to L-selectin does not prevent an effective host response to Sendai virus. J. Immunol. 155:252. 41 Tchilian, E. Z., Dawes, R., Hyland, L. et al. 2004. Altered CD45 isoform expression affects lymphocyte function in CD45 Tg mice. Int. Immunol. 16:1323. 42 Rosen, H., Sanna, G. and Alfonso, C. 2003. Egress: a receptorregulated step in lymphocyte trafficking. Immunol. Rev. 195:160. 43 Matloubian, M., Lo, C. G., Cinamon, G. et al. 2004. Lymphocyte egress from thymus and peripheral lymphoid organs is dependent on S1P receptor 1. Nature 427:355. 44 Tripp, R. A., Hou, S., McMickle, A., Houston, J. and Doherty, P. C. 1995. Recruitment and proliferation of CD8+ T cells in respiratory virus infections. J. Immunol. 154:6013. 45 Tripp, R. A., Sarawar, S. R. and Doherty, P. C. 1995. Characteristics of the influenza virus-specific CD8+ T cell response in mice homozygous for disruption of the H-2lAb gene. J. Immunol. 155:2955. 46 Gerhard, W., Mozdzanowska, K., Furchner, M., Washko, G. and Maiese, K. 1997. Role of the B-cell response in recovery of mice from primary influenza virus infection. Immunol. Rev. 159:95. 47 Topham, D. J. and Doherty, P. C. 1998. Clearance of an influenza A virus by CD4+ T cells is inefficient in the absence of B cells. J. Virol. 72:882. 48 Bourgeois, C., Veiga-Fernandes, H., Joret, A. M., Rocha, B. and Tanchot, C. 2002. CD8 lethargy in the absence of CD4 help. Eur. J. Immunol. 32:2199. 49 Sun, J. C., Williams, M. A. and Bevan, M. J. 2004. CD4+ T cells are required for the maintenance, not programming, of memory CD8+ T cells after acute infection. Nat. Immunol. 5:927.