<<

1 presentation kinetics control / 2 interactions and Tfh generation in vivo.

3 Robert A Benson1, Megan KL MacLeod1, Benjamin G Hale2, Agapitos Patakas1, 4 Paul Garside1 & James M Brewer1

5

6 1Institute of , and , 7 College of Medical, Veterinary & Life Sciences, Sir Graeme Davies 8 Building, University of Glasgow, 120 University Place, Glasgow, G12 9 8TA, United Kingdom

10 2Institute of Medical Virology, University of Zurich 11

12 Running title: Antigen size dictates Tfh differentiation

13 Corresponding author: 14 Professor James Brewer 15 Institute of Infection, Immunity and Inflammation, 16 College of Medical, Veterinary & Life Sciences, 17 Sir Graeme Davies Building, University of Glasgow, 18 120 University Place, Glasgow, 19 G12 8TA, United Kingdom 20 Email: [email protected] 21 Telephone: +44 141 330 8417 22

23 Classification: biological sciences; and inflammation.

24 Key words: Tfh; multiphoton; antigen presentation; ; 25 influenza vaccine 26

1 27 Abstract

28 The production of high affinity, class switched produced by B cells 29 hinges on the effective differentiation of T follicular helper (Tfh) cells. Here we 30 define conditions specifically enhancing Tfh differentiation and providing 31 protection in a model of influenza infection. Tfh responses were associated with 32 prolonged antigen presentation by Dendritic cells (DCs), which maintained T 33 cell/DC interactions into stage 3 (>72 hours) of activation. Blocking stage 3 34 interactions ablated Tfh generation, demonstrating a causal link between T cell– 35 DC behaviour and functional outcomes. The current data therefore explain how 36 duration of antigen presentation affects the dynamics of T cell-DC interactions 37 and consequently determine Tfh cell differentiation in the developing immune 38 response.

39

40

2 41 INTRODUCTION

42 Following stimulation via the TcR, selective differentiation of 43 subsets is dependent on transcription factors, with Bcl-6 being responsible for 44 Tfh cell differentiation (1, 2) and antagonism of other T cell phenotypes (Th1, 45 Th2, Th17). Gene knockout studies in mice have revealed the requirement for 46 CD28 mediated-costimulation in the upstream control of Tfh differentiation (3), 47 with subsequent signalling through inducible T cell costimulator (ICOS) involved 48 in consolidation of differentiation (4, 5). The mechanisms controlling the 49 nduction of this differentiation program are less clear, a role for the IL- 50 6 and IL-21 and subsequent signalling via STAT3 have been implicated (6), while 51 other studies indicate that the strength and duration of the TCR signal itself is 52 the major factor determining effector cell differentiation to Tfh (Fazilleau et al., 53 2009; Tubo 2013). Using well defined and controlled conditions that 54 preferentially induce Tfh responses in vivo, we have therefore adopted a 55 reductionist approach to determine which of these factors are important in the 56 differentiation of this subset in vivo.

57 58 A variety of particle formulations, including alum, emulsions and liposomes have 59 received sustained interest as vaccine adjuvants for over 80 years (7-9). Their 60 ability to affect the availability of antigen and antigen presentation has 61 frequently been linked to their impact on the development of T cell responses 62 (10), but causal evidence has been absent. More recently, studies have suggested 63 that the use of nanoparticles as adjuvants can selectively induce Tfh responses 64 (11). Clearly selective differentiation of Tfh effector cells has implications in 65 vaccine design and pathogen control as for the majority of vaccines the principal 66 effector mechanism induced is production of high affinity, class switched 67 antibodies by B . Here we employ defined antigen particles to 68 selectively induce Tfh cells and antibody responses and show that increased 69 duration of antigen presentation and subsequent T cell/DC interactions are 70 associated with Tfh induction. The functional significance of these late stage 71 (72h) interactions in T cell differentiation was demonstrated by blockade of 72 T/DC interactions in vivo. These studies demonstrate that by manipulating the

3 73 form of antigen delivered late stage antigen presentation by DC and their 74 interaction with T cells can be controlled to drive Tfh differentiation in vivo. 75

76 Results 77 Antigen size impacts on protective . 78 In the present study, we employed polystyrene nanoparticles due to their inert 79 nature, defined size, narrow polydispersity, and surface functionalisation 80 allowing covalent linkage of antigen to their surface. Polystyrene nanoparticles 81 do not induce ERK activity or subsequent activation of conventional 82 inflammatory pathways in DC (12). Our study focused on polystyrene 83 nanoparticle sizes able to drain freely to the LN (13). Carbodiimide-mediated 84 coupling of was highly controlled, such that only particle size and 85 physically dependent parameters (surface area, antigen molecules per particle) 86 varied between immunisation conditions; all other conditions, antigen dose, 87 mass of particles, and antigen density were consistent between 40 nm and 200 88 nm particle immunisations (Table 1). Consequently, immunisations with 89 equivalent quantities of protein were not associated with any confounding 90 difference in total antigenic mass and a single variable, particle size could be 91 tested for its impact on antigen specific immune responses. Initial experiments 92 investigating the effect of particle size on antibody responses revealed that 93 immunisation of mice with Ag covalently linked to 200 nm particles induced 94 significantly higher amounts of specific IgG1 and IgG2c than smaller particles or 95 soluble Ag (Figure 1A, B). Subsequent studies focussed on the significance and 96 processes by which the 200 nm particles were able to support this antibody 97 response, in comparison to smaller particles (40 nm) and soluble antigen that 98 did not. 99 100 The functional importance of the observed antigen specific antibody response 101 was evaluated in a murine influenza infection model where antibodies to 102 the hemagglutinin (HA) component of influenza virus are important to 103 protective immunity (14, 15), blocking viral attachment and fusion of the viral 104 and host (16). Three weeks after immunisation with HA linked to

4 105 200 or 40 nm particles, emulsified in complete Freund’s adjuvant (CFA) or as 106 soluble protein, mice were challenged intranasally with influenza A virus (H1N1; 107 A/WSN/1933 [WSN]). Protection from infection was determined by weight loss 108 (Figure 1C). While mice immunised with 200 nm HA-particles were protected 109 from virus induced pathology, significant weight loss was observed in groups 110 immunised with 40 nm HA-particles, similar weight loss to that observed in 111 unimmunised or soluble HA treated mice. 112 113 Antigen size influences the development of Tfh and responses. 114 To delineate the mechanisms that allow particle size to control antibody 115 responses, we employed an established adoptive transfer model (17). Antigen 116 specific CD4+ T cells and B cells were co-transferred into congenic recipients 117 then immunised with 130μg of OVA-HEL conjugate alone, in CFA or bound to 40 118 nm or 200 nm particles. Analysis of serum samples collected 10 days post 119 immunisation revealed OVA-HEL conjugated to 200 nm particles induced 120 significant levels of serum anti-HEL-IgMa, whereas antigen conjugated to 40 nm 121 particles did not (Figure 2A). Similarly, antigen conjugated to 200 nm particles 122 induced greater expansion of antigen-specific IgMa+ MD4 B cells than challenge 123 with 40 nm particles 7 days post immunisation (Figure 2B). Enhanced 124 expression of germinal centre (GC) markers (GL7 and FAS) following 125 immunisation with 200 nm OVA-HEL particles compared with 40 nm OVA-HEL 126 particles was also evident (Figure 2C, D). Conventional GC responses are 127 dependent on activated, antigen-specific CD4+ T cells (18, 19), suggesting that 128 different particles could induce changes in the magnitude or phenotype of the 129 resulting T cell response. However, immunisation with OVA-HEL conjugated to 130 either 40 nm or 200 nm particles could equally increase the proportion and 131 number of antigen specific OT-II T cells in vivo (Figure 3A, B), consistent with a 132 previous report (20). This implied that particle size induced a qualitative rather 133 than quantitative difference in T cell response, favouring generation of the Tfh 134 subset of T cells that have been defined as supporting B cell responses and GC 135 formation (21). Analysis of Tfh markers CXCR5, PD-1, ICOS and SLAM by 136 adoptively transferred OT-II T cells demonstrated the percentage and number of 137 antigen specific cells differentiating into Tfh cells was very low at day 5 post

5 138 immunisation with soluble antigen or antigen conjugated to 40 nm particles 139 (Figure 3C-E). However, a significant proportion of OT-II T cells were CXCR5+PD- 140 1+ following challenge with OVA-HEL conjugated to 200 nm particles (Figure 3C- 141 E). Changing a single physical characteristic of the immunising antigen 142 selectively enhanced Tfh differentiation without impacting on the magnitude of T 143 cell response, providing an excellent platform to dissect how external stimuli can 144 contribute to Tfh development in vivo. 145 146 147 Antigen size can determine duration of peptide/MHCII presentation LN 148 APCs 149 Adjuvants are thought to influence the availability of antigen in vivo and the 150 kinetics of antigen presentation influence consequent CD4+ T cell proliferative, 151 effector and memory responses (22-24). By covalently linking a traceable 152 antigen, EαGFP (25), to our particles we were able investigate the impact of 153 particle size on peptide/MHCII presentation in vivo. Immunisation with 40 nm 154 particles resulted in rapid presentation by both DCs and B cells within 6 hours of 155 subcutaneous injection, consistent with published reports (25, 26). 156 Immunisation with antigen conjugated to 40 nm particles resulted in similar 157 rapid presentation by DCs (Figure 4A, B) and B cells (Figure 5A, B C) as 158 previously observed with soluble antigen (25, 26). An equivalent proportion of 159 DCs also presented Eα/MHCII following immunisation with 200 nm particles. 160 Percentages and numbers of Eα/MHCII positive DCs equivalent to soluble and 40 161 nm particle antigen formulations were detected at 24 and 48 hours. However, in 162 contrast to soluble and 40 nm particle formulations, 200 nm particle challenge 163 resulted in presentation of antigen beyond 48 hours, with 21±3% of 164 conventional DCs staining positive for Eα/MHCII at 72 hours post immunisation 165 (Figure 4C). Presentation of antigen conjugated to 200 nm particles by the B cell 166 compartment was slower (Figure 5A, B, C) but did reach levels equivalent to 167 soluble and 40 nm particle-bound antigen. The sustained presentation observed 168 by DCs was not evident in the B cell compartment (Figure 5D), with no 169 significant differences in B cell presentation being detected beyond 24 hours

6 170 (Figure 5). Thus, larger particles did not alter the magnitude of presentation but 171 did prolong the duration of peptide/MHCII presentation by DCs. 172 173 174 175 Sustained peptide/MHCII presentation supports late T cell/DC interactions. 176 The relevance of antigen presentation at 72 hours induced by antigen conjugated 177 to 200 nm particles was questionable given evidence from intravital imaging 178 studies. Such studies have demonstrated that T cell by DCs occurs in 179 three successive stages (27-30); stage 1 (0-8 hours) characterised by rapid T cell 180 motility and short T/DC interactions (2- 3 minutes); stage 2 (12- 24 hours) with 181 long-term T/DC interactions associated with the induction of priming and; stage 182 3 (48 – 72 hours) where T cells resume motility and short T/DC interactions. We 183 therefore examined the impact of particle size on stage 3 T cell/DC interactions 184 in vivo. CD11c-YFP recipients were adoptively transferred with OT-II DsRed T 185 cells and challenged with OVA conjugated 40 nm or 200 nm particles 72 hours 186 before imaging by MPLSM (Video 1 and 6A). Multiple short duration 187 T cell-DC interactions (2.149 ± 0.139 minutes) were observed with 40 nm OVA 188 particles (Figure 6B), comparable to that seen with naïve T cells and therefore 189 consistent with an absence of cognate peptide/MHC and classic stage 3 motility 190 (30). Notably, interactions longer than 10 minutes were seen following 200 nm 191 particle challenge (Figure 6B), implying that antigen driven cognate recognition 192 was still occurring. This was further supported by the reduced T cell velocity 193 observed in the 200 nm particle group (Figure 6C) and again in a lower T cell 194 displacement rate (Figure 6D). T cell migratory patterns within the LNs were not 195 significantly different between challenges as evidenced by their equivalent 196 meandering indices (Figure 6E). Thus, the antigen presentation by DCs at 72 197 hours post challenge induced by antigen-conjugated 200 nm particles changed 198 the dynamics of T cell/DC interactions, with stable, long-term interactions 199 extending into the stage 3 time period, conventionally associated with transient 200 interactions and rapid T cell motility (29-32). 201 202

7 203 Antigen size influences Tfh differentiation by sustaining peptide/MHCII 204 presentation. 205 Previous studies have demonstrated that differences in stage 2 interactions 206 underlie the outcome of the developing immune response, with long-term (>10 207 min) and short-term (<3 min) interactions being associated with induction or 208 priming and tolerance respectively (30-32). Significantly, the causal link between 209 T cell behaviour and function was subsequently revealed through disrupting 210 TcR/pMHCII interactions with a (Y3P), which resulted in of 211 loss long-term T/DC contacts, a return to rapid T cell motility, and a loss of T cell 212 activation (22). We hypothesised that the long-term T/DC interactions we 213 observed at 72 hours post immunisation with 200 nm particles were responsible 214 for selective Tfh differentiation. To test this hypothesis, we employed the 215 approach described by Celli et al (2007), using the Y3P monoclonal antibody 216 against MHCII (22) administered at 72 hours post challenge. Disruption of 217 TcR/pMHCII blocked the stable interactions between T cells and DCs (Video1 218 and 2, Figure 7A, and 7B) and increased T cell motility (data not shown). Unlike 219 blocking stage 2 T/DC interactions (22), robust T cell proliferative responses 220 were observed with Y3P treatment at 72 hours (Figure 7C). However, blocking 221 MHCII at this time point did significantly reduce the proportion of antigen 222 specific cells co-expressing CXCR5 and PD-1 (Figure 7D, E). Therefore, we 223 conclude that while the sustained T/DC interactions at 72 hours induced by 200 224 nm particles do not increase the magnitude of the resulting immune response, 225 these interactions specifically function to alter the quality of immune response 226 by promoting Tfh differentiation. 227 228 Analysis of antigen presentation kinetics following immunisation with 200 nm 229 particles revealed that at the time of MHCII blocking (72h) the major population 230 of APCs were DCs. Our multiphoton studies further confirmed that T cells were 231 continuing to interact with DCs at this time. However, interactions with antigen 232 presenting B cells could not be ruled out as a contributing factor, especially given 233 their critical role in affirming and maintaining the Tfh phenotype (1, 5, 33). To 234 establish whether the Tfh phenotype observed following immunisation with 200 235 nm particles was attributable purely to DC presentation at 72 hours we assessed

8 236 Tfh differentiation in the absence of cognate pMHCII B cell presentation. 237 Immunisation of MD4 mice with 200 nm OVA particles induced equivalent 238 proportional increases in adoptively transferred OT-II T cells as seen in C57BL/6 239 mice (Figure 8A). Interestingly, the Tfh phenotype induced following 240 immunisation with 200 nm particles was not significantly altered in the absence 241 of cognate T-B cell interactions (Figure 8B). Inclusion of pMHCII blockade at 72 242 hours post immunisation in MD4 mice did not influence expansion of the OT-II 243 population (Figure 8A) but did significantly reduce the proportion of cells co- 244 expressing CXCR5 and PD-1 (Figure 8B), reaffirming the importance of pMHCII 245 recognition at this time. Taken together, the data implicates DCs rather than B 246 cells as the APC population mediating this effect.

247 248 Discussion 249 We have provided, in unprecedented detail, the spatiotemporal sequence of 250 events that underlie enhanced differentiation of Tfh cells, GC response and 251 protective Ab production by antigen formulations in vivo. By combining highly 252 defined antigen delivery systems, with trackable antigen, antigen-receptor 253 transgenics and state of the art imaging techniques, we revealed that antigen size 254 impacts on the duration of peptide/MHCII presentation and the maintenance 255 beyond 48 hours of functional DC and T cell interactions in the draining LN. The 256 functional relevance of longer DC-T cell interactions, associated with antigen 257 conjugated to 200 nm particles, was dissected by specifically blocking later 258 interactions, resulting in reduced Tfh induction, while the overall magnitude of 259 the T cell response was unaffected. Thus, the temporal characteristics of T cell 260 stimulation can determine their functional differentiation towards a Tfh 261 phenotype, and this can be determined by the size of the particle upon which an 262 antigen is delivered. 263 264 Previous studies have investigated the impact of particle size on the immune 265 response to antigen using a variety of formulations, for example lipid vesicles 266 entrapping (11, 34) antigens or antigens non-specifically adsorbed to the surface 267 of inert particles (35). The inert nature, defined size and surface

9 268 functionalisation of particles employed in the present study, allowed a single 269 variable, size, to be tested for its impact on antigen immunogenicity. Initial 270 studies revealed that simply increasing particle size from 40 to 200 nm, could 271 induce antibody production following a single immunisation. The functional 272 importance of this observation was startlingly clear, with only the 200 nm 273 particles able to impart protective anti-HA humoral immunity to influenza 274 infection. Starting with a functional outcome relevant to vaccine design, we 275 sought to dissect the processes by which increasing particle size impacts on the 276 humoral response. 277 278 GC formation is central to development of high affinity antibody. GC structures 279 support , selection of high affinity B cells and their 280 differentiation into plasma and memory cells (for a comprehensive review see 281 (36)). Immunisation with 200 nm particles enhanced this process, explaining our 282 initial observation of increased antibody responses. Essential in this process is 283 the cognate interaction between Ag-specific B and T cells. The nature of this 284 interaction has been the focus of intense research in recent years, culminating in 285 the identification of Tfh cells and the molecules (surface and soluble) involved in 286 their differentiation and function (21). While both sizes of particle could equally 287 increase antigen specific T cell responses in vivo, we found that larger particles 288 (200 nm) induced greater Tfh differentiation than small (40 nm) particles, 289 consistent with their role in supporting GC responses. Even though the 290 endogenous molecular cues governing the development of Tfh cells are 291 multifactorial (5, 21), understanding how external stimuli can influence T cell 292 differentiation towards this phenotype is less well understood, yet has clear 293 implications in vaccine design. In this case we have demonstrated that simply 294 changing the size of the Ag can clearly impact on Tfh differentiation. 295 296 A key event in the initiation of any CD4+ T cell response is the recognition of 297 peptide/MHC class II complexes on dendritic cells. The kinetics of presentation 298 are known to influence the consequent CD4+ T cell proliferative, effector and 299 memory responses induced (22-24). By covalently linking a trackable antigen to 300 the particles we were able to map temporally in vivo peptide/MHC class II

10 301 presentation and its relation to particle size. Although 200 nm particle 302 immunisation prolonged the duration of peptide/MHC class II presentation by 303 DCs, significantly this did not impact upon the magnitude of the T cell response 304 compared with the smaller 40 nm particle, consistent with a previous report 305 (20). However, prolonged presentation was associated with the enhanced 306 differentiation of Tfh cells. Antigen availability can dictate the magnitude of a Tfh 307 response, with sustained presentation by B cells maintaining the Tfh phenotype 308 (33). Notably, immunisation with 200 nm particles resulted in sustained antigen 309 presentation by DCs with cognate interactions with B cells being dispensable for 310 maintenance of a Tfh phenotype. Indeed, sustained antigen presentation 311 associated with Tfh induction has been reported in the absence of cognate 312 interactions with B cells (37). In the latter study, CD4+ T cells deficient for SAP, 313 and therefore limited in their ability to interact with B cells, still adopted a Tfh 314 phenotype when a second dose of soluble antigen was given 72 hours later. 315 Presentation at this second challenge was attributed to DCs (37), consistent with 316 our own findings. Indeed, DCs can induce Tfh development in the absence of 317 cognate interactions with B cells (2) and likely represent the first step initiating 318 Tfh differentiation (38). Imaging draining LNs 72 hours post challenge revealed 319 that cognate T cell/DC interactions continued at this time. Importantly the 320 current study adds mechanistic detail to the observational data above; temporal 321 disruption of 72 hour (stage 3) interactions, while not affecting clonal expansion, 322 did inhibit 200 nm particle induced Tfh differentiation. Thus, increasing particle 323 size, and sustaining presentation by DCs and their cognate interactions with T 324 cells underpins the success of the larger particle in driving Tfh differentiation. It 325 is unclear as to how particle size influences temporal availability of pMHCII on 326 DCs. Particulates can impact on the mechanism of antigen uptake and therefore 327 consequent endosomal targeting, processing and dynamics of presentation (9, 328 39, 40). Thus the processing of antigen and loading of MHCII and/or its turnover 329 at the cell surface may be contributing factors. In either case, increasing 330 availability of TcR ligand and consequent TcR signal quantity may function in a 331 similar way as increasing TcR dwell time in determining Tfh differentiation (41, 332 42). 333

11 334 New delivery systems are required to generate safe and effective vaccines. This 335 has resulted in increased focus on particle delivery systems. Our findings provide 336 mechanistic insight as to why some formulations promote humoral immunity 337 while others do not (11, 35). We have linked T cell behaviour to function, 338 demonstrating the importance of stage 3 T/DC interactions in determining the 339 selective differentiation of T cells towards a Tfh lineage. These late stage 340 interactions could be selectively induced using immunisation with antigen 341 conjugated to 200 nm but not 40 nm particles, rationalising approaches to 342 vaccine design using nanoparticles for induction of Tfh, germinal centre 343 formation and high affinity, class switched protective antibody. Previous studies 344 have demonstrated that in the absence of microbial factors, parameters such as 345 the dose and persistence of antigen can affect T cell differentiation (10). The 346 current data therefore explain why parameters such as the magnitude and 347 duration of antigen presentation affect subsequent T/DC interactions and 348 consequently determine T cell differentiation in the developing immune 349 response. 350 351 352 Methods 353 Animals 354 hCD2-DsRed mice (gifted by D. Kioussis and A. Patel, National Institute for 355 Medical Research, London) were crossed with ovalbumin specific OT-II (43) T 356 cell receptor (TCR) transgenic (Tg) mice. MD4 BcR Tg mice (44) on C57BL/6 357 backgrounds were used as a source of hen egg lysozyme (HEL) specific B cells. 6- 358 8 week old C57BL/6J mice were used as recipients (Harlan, Bicester, UK). CD11c- 359 YFP mice (45) were used a recipients in multiphoton imaging experiments. All 360 animals were specified pathogen free and maintained under standard animal 361 house conditions at the University of Glasgow in accordance with UK Home 362 Office Regulations. 363 364 Antigen and bead preparations 365 The recombinant haemagglutinin (HA) protein used was expressed from DNA 366 encoding the extracellular domain of the influenza A virus subtype H1N1

12 367 (A/WSN/1933) HA (Life Technologies). OVA-HEL conjugates were prepared 368 using gluteraldehyde to couple the chicken ovalbumin (OVA; Worthington 369 Biochemical, Lakewood, NJ, USA) and hen egg lysozyme (HEL; BBI Enzymes, 370 Gwent, UK) as published (17). EαGFP was produced in house as detailed 371 previously (46). HA, OVA, OVA-HEL or EαGFP were covalently bound to 372 Polybead® Carboxylate Microspheres (Park Scientific, Northampton, UK). 373 Briefly, carboxylate-modified nanospheres were incubated with 2.5mg/ml 374 protein (EαGFP, HA, OVA or OVA-HEL) with 1mM EDAC (1-ethyl-3-(3- 375 dimethylaminopropyl) carbodiimide; Sigma, Dorset, UK) and 25mM MES (2-N- 376 morpholino ethane sulfonic acid; Sigma) overnight, in line with manufacturers 377 instructions. Conjugation mixes were then spun in an ultracentrifuge at 4°C for 378 40 minutes. Protein concentration in the supernatant was measured to allow the 379 amount bound to the microspheres to be calculated. Preparations were stored at 380 4°C and used within 3 days of preparation. 381 382 Infection with influenza virus 383 Mice were immunized subcutaneously (s.c.) in the scruff. with 384 influenza virus subtype H1N1 A/WSN/1933 (WSN) were carried out in mice 385 anesthetized with isofluorane and 20 µl of PBS containing 300 PFU of WSN virus 386 administered intranasally (5 mice per group). This dose was used as previous 387 experiments showed that this dose led to approximately 20% weight loss in 388 unvaccinated animals. Viral stocks were prepared and titred in MDCK cells. 389 390 Adoptive transfer of antigen specific lymphocytes and in vivo challenge 391 Adoptive transfers were performed as described previously (47). Briefly, single 392 cell suspensions of LNs and spleens were prepared from OT-II and MD4 mice. 393 The proportion of transgenic B and T cells was determined by flow cytometry 394 and 2-3x106 transgenic B cells and 2-3x106 transgenic T cells adoptively 395 transferred to age-matched C57BL/6J recipients via tail vein injection. For 396 imaging studies, DsRed expressing CD4+ OT-II T cells were magnetically sorted 397 from OT-II x hCD2-DsRed animals using mouse CD4+ T cell isolation kits 398 (Miltenyi Biotec, Surrey, UK). Mice were immunized via the footpad with 130µg 399 of OVA-HEL conjugate alone, with complete Freund’s adjuvant (CFA; Sigma,

13 400 Dorset, UK), or covalently bound to carboxylate-modified microspheres. 401 Injection with 50μl of PBS was used as a vehicle control. Popliteal LNs were 402 excised for ex vivo study. In MHCII blocking experiments, 500μg Y3P (BioXcell, 403 New Hampshire, USA) or control (mouse IgG2a; BioXcell) was given via 404 tail vein injection. In T cell/DC interaction studies, LNs were imaged 2 hours 405 after antibody treatment. 406 407 Flow cytometry 408 Single cell suspensions were stained using combinations of anti-CD11c-PE 409 (N418, BD Biosciences), anti-B220-PerCP (RA3-6B2, BD Biosciences), anti-I-

b 410 A /Ea52-68-biotin (YAe, eBioscience), anti-CD4-eFluor450 (RM4-5, eBioscience), 411 anti-ICOS-alexa-fluor-488 (C398.4A, BioLegend), anti-PD-1-PE-Cy7 (J43, 412 eBiosciences), anti-SLAM-APC (TC15-12F12.2, BioLegend), biotinylated anti- 413 CXCR5 (2G8, BD Biosciences), anti-B220-eFluor450 (RA3-6B2, eBioscience), 414 anti-GL-7-FITC (BD Biosciences), anti-FAS-PE (BD Biosciences) and biotinylated 415 anti-IgMa (DM-1, BD Biosciences). Biotinylated antibodies were detected by 416 incubation with fluorochrome-conjugated streptavidin (BD Biosciences). 417 Appropriate isotype controls were used throughout. Samples were acquired 418 using a MACSQuant analyzer (Miltenyi Biotec, Surrey, UK) and analysed using 419 FlowJo software (Tree Star Inc, Ashland, OR, USA). 420 421 Multiphoton Imaging 422 Multiphoton imaging was carried out using a Zeiss LSM7 MP system equipped 423 with a 20X/1.0NA water-immersion objective lens (Zeiss UK) and a tunable 424 Titanium: sapphire solid-state two-photon excitation source (Chameleon Ultra II; 425 Coherent Laser Group, Glasgow, UK) and optical parametric oscillator (OPO; 426 Coherent Laser Group). Excised LNs were continuously bathed in warmed

427 (37°C), oxygenated CO2 independent medium. A laser output of 820nm and OPO 428 signal at 1060 nm provided excitation of YFP CD11c+ and DsRed OT-II cells. 429 Movies were acquired for 20 to 30 minutes with an X-Y pixel resolution of 512 x 430 512 in 2μm Z increments. 3D tracking was performed using Volocity 6.1.1 431 (Perkin Elmer, Cambridge, UK). Values representing the mean velocity, 432 displacement, and meandering index were calculated for each object. The

14 433 intersection of DsRed and YFP objects was used to determine interaction 434 between T cells and DCs respectively.

435 436 Statistics 437 Results are expressed as mean ± standard deviation. Gaussian distribution was 438 confirmed by D'Agostino & Pearson omnibus normality test prior to significance 439 being determined by one-way ANOVA. Significance in DC-T cell interaction 440 experiments was confirmed by Mann Whitney. Values of p < 0.05 were regarded 441 as significant. 442 443 Acknowledgements

444 This work was supported by the Wellcome Trust (grant no. WT085589MA). BGH

445 is a Sir Henry Dale Fellow jointly funded by the Wellcome Trust and the Royal

446 Society (Grant Number 100034/Z/12/Z). The authors declare no competing

447 financial interests.

448

15 449 References 450 1. Johnston RJ, et al. (2009) Bcl6 and Blimp-1 are reciprocal and antagonistic 451 regulators of T follicular helper cell differentiation. Science 452 325(5943):1006-1010. 453 2. Choi YS, et al. (2011) ICOS receptor instructs T follicular helper cell versus 454 effector cell differentiation via induction of the transcriptional repressor 455 Bcl6. Immunity 34(6):932-946. 456 3. Linterman MA, et al. (2009) Roquin differentiates the specialized 457 functions of duplicated T cell costimulatory receptor genes CD28 and 458 ICOS. Immunity 30(2):228-241. 459 4. Xu H, et al. (2013) Follicular T-helper cell recruitment governed by 460 bystander B cells and ICOS-driven motility. Nature 496(7446):523-527. 461 5. Crotty S (2011) Follicular helper CD4 T cells (TFH). Annu Rev Immunol 462 29:621-663. 463 6. Nurieva RI, et al. (2008) Generation of T follicular helper cells is mediated 464 by interleukin-21 but independent of T helper 1, 2, or 17 cell lineages. 465 Immunity 29(1):138-149. 466 7. Brewer JM (2006) (How) do aluminium adjuvants work? Immunol Lett 467 102(1):10-15. 468 8. Xiang SD, et al. (2006) Pathogen recognition and development of 469 particulate vaccines: does size matter? Methods 40(1):1-9. 470 9. De Temmerman ML, et al. (2011) Particulate vaccines: on the quest for 471 optimal delivery and immune response. Drug Discov Today 16(13- 472 14):569-582. 473 10. Constant SL & Bottomly K (1997) Induction of Th1 and Th2 CD4+ T cell 474 responses: the alternative approaches. Annu Rev Immunol 15:297-322. 475 11. Moon JJ, et al. (2012) Enhancing humoral responses to a malaria antigen 476 with nanoparticle vaccines that expand Tfh cells and promote germinal 477 center induction. Proc Natl Acad Sci U S A 109(4):1080-1085. 478 12. Karlson Tde L, Kong YY, Hardy CL, Xiang SD, & Plebanski M (2013) The 479 signalling imprints of nanoparticle uptake by bone marrow derived 480 dendritic cells. Methods 60(3):275-283. 481 13. Manolova V, et al. (2008) Nanoparticles target distinct dendritic cell 482 populations according to their size. Eur J Immunol 38(5):1404-1413. 483 14. Gerhard W (2001) The role of the antibody response in influenza virus 484 infection. Curr Top Microbiol Immunol 260:171-190. 485 15. Murphy BR, et al. (1982) Secretory and systemic immunological response 486 in children infected with live attenuated influenza A virus vaccines. Infect 487 Immun 36(3):1102-1108. 488 16. Knossow M & Skehel JJ (2006) Variation and infectivity neutralization in 489 influenza. Immunology 119(1):1-7. 490 17. Garside P, et al. (1998) Visualization of specific B and T 491 interactions in the lymph node. Science 281(5373):96-99. 492 18. Owens T & Zeine R (1989) The cell biology of T-dependent B cell 493 activation. Biochem Cell Biol 67(9):481-489. 494 19. Nutt SL & Tarlinton DM (2011) Germinal center B and follicular helper T 495 cells: siblings, cousins or just good friends? Nat Immunol 12(6):472-477.

16 496 20. Gengoux C & Leclerc C (1995) In vivo induction of CD4+ T cell responses 497 by antigens covalently linked to synthetic microspheres does not require 498 adjuvant. Int Immunol 7(1):45-53. 499 21. Ma CS, Deenick EK, Batten M, & Tangye SG (2012) The origins, function, 500 and regulation of T follicular helper cells. J Exp Med 209(7):1241-1253. 501 22. Celli S, Lemaitre F, & Bousso P (2007) Real-time manipulation of T cell- 502 dendritic cell interactions in vivo reveals the importance of prolonged 503 contacts for CD4+ T cell activation. Immunity 27(4):625-634. 504 23. Obst R, van Santen HM, Mathis D, & Benoist C (2005) Antigen persistence 505 is required throughout the expansion phase of a CD4(+) T cell response. J 506 Exp Med 201(10):1555-1565. 507 24. Obst R, et al. (2007) Sustained antigen presentation can promote an 508 immunogenic T cell response, like dendritic cell activation. Proc Natl Acad 509 Sci U S A 104(39):15460-15465. 510 25. Pape KA, Catron DM, Itano AA, & Jenkins MK (2007) The humoral immune 511 response is initiated in lymph nodes by B cells that acquire soluble 512 antigen directly in the follicles. Immunity 26(4):491-502. 513 26. Itano AA, et al. (2003) Distinct dendritic cell populations sequentially 514 present antigen to CD4 T cells and stimulate different aspects of cell- 515 mediated immunity. Immunity 19(1):47-57. 516 27. Stoll S, Delon J, Brotz TM, & Germain RN (2002) Dynamic imaging of T 517 cell-dendritic cell interactions in lymph nodes. Science 296(5574):1873- 518 1876. 519 28. Bousso P & Robey E (2003) Dynamics of CD8+ T cell priming by dendritic 520 cells in intact lymph nodes. Nat Immunol 4(6):579-585. 521 29. Miller MJ, Safrina O, Parker I, & Cahalan MD (2004) Imaging the single cell 522 dynamics of CD4+ T cell activation by dendritic cells in lymph nodes. J Exp 523 Med 200(7):847-856. 524 30. Mempel TR, Henrickson SE, & Von Andrian UH (2004) T-cell priming by 525 dendritic cells in lymph nodes occurs in three distinct phases. Nature 526 427(6970):154-159. 527 31. Zinselmeyer BH, et al. (2005) In situ characterization of CD4+ T cell 528 behavior in mucosal and systemic lymphoid tissues during the induction 529 of oral priming and tolerance. J Exp Med 201(11):1815-1823. 530 32. Hugues S, et al. (2004) Distinct T cell dynamics in lymph nodes during the 531 induction of tolerance and immunity. Nat Immunol 5(12):1235-1242. 532 33. Baumjohann D, et al. (2013) Persistent antigen and germinal center B 533 cells sustain T follicular helper cell responses and phenotype. Immunity 534 38(3):596-605. 535 34. Brewer JM, Pollock KG, Tetley L, & Russell DG (2004) Vesicle size 536 influences the trafficking, processing, and presentation of antigens in lipid 537 vesicles. J Immunol 173(10):6143-6150. 538 35. Mottram PL, et al. (2007) Type 1 and 2 immunity following vaccination is 539 influenced by nanoparticle size: formulation of a model vaccine for 540 respiratory syncytial virus. Mol Pharm 4(1):73-84. 541 36. Victora GD & Nussenzweig MC (2012) Germinal centers. Annu Rev 542 Immunol 30:429-457.

17 543 37. Deenick EK, et al. (2010) Follicular helper T cell differentiation requires 544 continuous antigen presentation that is independent of unique B cell 545 signaling. Immunity 33(2):241-253. 546 38. Goenka R, et al. (2011) Cutting edge: dendritic cell-restricted antigen 547 presentation initiates the follicular helper T cell program but cannot 548 complete ultimate effector differentiation. J Immunol 187(3):1091-1095. 549 39. Zhao F, et al. (2011) Cellular uptake, intracellular trafficking, and 550 of nanomaterials. Small (Weinheim an der Bergstrasse, 551 Germany) 7(10):1322-1337. 552 40. Ghimire TR, Benson RA, Garside P, & Brewer JM (2012) Alum increases 553 antigen uptake, reduces antigen degradation and sustains antigen 554 presentation by DCs in vitro. Immunol Lett 147(1-2):55-62. 555 41. Tubo NJ, et al. (2013) Single naive CD4+ T cells from a diverse repertoire 556 produce different effector cell types during infection. Cell 153(4):785-796. 557 42. Tubo NJ & Jenkins MK (2014) TCR signal quantity and quality in CD4 T 558 cell differentiation. Trends in immunology 35(12):591-596. 559 43. Barnden MJ, Allison J, Heath WR, & Carbone FR (1998) Defective TCR 560 expression in transgenic mice constructed using cDNA-based alpha- and 561 beta-chain genes under the control of heterologous regulatory elements. 562 Immunol Cell Biol 76(1):34-40. 563 44. Mason DY, Jones M, & Goodnow CC (1992) Development and follicular 564 localization of tolerant B lymphocytes in lysozyme/anti-lysozyme 565 IgM/IgD transgenic mice. Int Immunol 4(2):163-175. 566 45. Lindquist RL, et al. (2004) Visualizing dendritic cell networks in vivo. Nat 567 Immunol 5(12):1243-1250. 568 46. Rush CM & Brewer JM (2010) Tracking dendritic cells in vivo. Methods 569 Mol Biol 626:169-185. 570 47. Smith KM, et al. (2000) Th1 and Th2 CD4+ T cells provide help for B cell 571 clonal expansion and antibody synthesis in a similar manner in vivo. J 572 Immunol 165(6):3136-3144.

573 574

18 575

Particle size 40 nm 200 nm

antigen dose/mouse 100 μg 100 μg

antigen molecules/mouse 1.3 x 1015 1.3 x 1015

latex dose/mouse 100 μg 130 μg

number of particles/mouse 1.4 x 1012 2.9 x 1010

antigen molecules/particle 1000 20,000

surface area/particle 5024 nm2 125,600 nm2

antigen density 1 per 5 nm2 1 per 6 nm2

576

577 Table 1: Comparison of particle conjugates. Data shows an example of a 578 typical carbodiimide-mediated conjugation of ovalbumin to either 40 or 200 nm. 579 Various parameters relating to a typical s.c. immunisation with 100μg of protein 580 are shown.

581

19 582 Figure Legends

583 Figure 1: Immunisation with antigen loaded 200 nm particles induces 584 elevated IgG and can provide protective immunity to influenza virus 585 infection. Mice were immunised s.c. in the scruff with 100μg of OVA alone or 586 conjugated to 20 nm, 40 nm, 100 nm, 200 nm or 1000 nm particles. 14 days later, 587 serum was tested by ELISA for anti-OVA-IgG2c (A) and anti-OVA-IgG1 (B). Data 588 show the mean (−) absorbance (405 nm) at a dilution of 1 in 200 with the 5 589 experimental animals per group ()*** p <0.001 comparing to the PBS group. To 590 assess functional responses, C57BL/6 mice were immunised s.c. with 50μg of HA 591 alone, conjugated to 40 or 200 nm particles, or emulsified with CFA. Weight 592 changes were then monitored following intranasal infection with 300 PFU of 593 influenza A virus (WSN) (C). Data shows the mean of 5 experimental animals per 594 group ± standard deviation *** p <0.001 comparing 200 nm group to 40 nm 595 group.

596

597 Figure 2: Immunisation with antigen loaded 200 nm particles enhances B 598 cell expansion and germinal centre B cell responses. To dissect the cellular 599 responses in detail, MD4 BcR transgenic B cells and OT-II TcR transgenic T cells 600 and were adoptively transferred into C57BL/6 recipients. Mice were then 601 challenged with OVA-HEL conjugated to 40 nm or 200 nm particles. Challenges 602 with PBS, OVA-HEL alone or in CFA were included as negative and positive 603 controls. Day 10 serum samples were assayed by ELISA to determine anti-HEL- 604 IgMa levels (A). Expansion of B220+IgMa+ MD4 B cells in popliteal LNs was 605 assessed by flow cytometry 7 days post challenge (B). Development of germinal 606 centre B cell responses was determined by examining GL7 and FAS expressing 607 by MD4 B220+ cells (C). Representative staining of germinal centre markers GL7 608 and FAS are also shown (D). Panels show mean value and 5 individual animals in 609 a representative experiment of 3.

610

611

20 612 Figure 3: 200 nm particles induce Tfh differentiation. MD4 BcR transgenic B 613 cells and OT-II TcR transgenic T cells and were adoptively transferred into 614 C57BL/6 recipients. Mice were then challenged with OVA-HEL conjugated to 40 615 nm or 200 nm particles. Challenges with PBS, OVA-HEL alone or in CFA were 616 included as negative and positive controls. CD4+DsRed+ OT-II cell expansion in 617 LNs 5 days post challenge (A, B). These were defined as Tfh cells when being PD- 618 1+ and CXCR5+ (C, D). Cells were also confirmed as being both ICOS+ and SLAM+ 619 (E). Panels show mean value and 5 individual animals in a representative 620 experiment of 3. 621 622 Figure 4: 200 nm particulate antigen challenge promotes sustained 623 presentation of peptide/MHCII by DCs. C57BL/6 mice were immunized s.c. in 624 the footpad with 100μg of the model antigen EαGFP alone or covalently 625 conjugated to 40 nm or 200 nm particles. Challenge with PBS was included as a 626 negative control. The draining popliteal LNs were harvested after 1, 6, 24, 48 and 627 72 hours post challenge. Flow cytometry was used to determine levels of 628 presentation of the Eαpeptide/MHC class II complex using the monoclonal 629 antibody YAe on CD11c+B220- conventional DCs. The percentage and number of 630 conventional DCs cells staining YAe+ is shown in (A) and (B) respectively. 631 Representative flow cytometry plots are shown for 72 hours post challenge (C). 632 Data shows the mean of 3 animals per time point ± standard deviation and are 633 representative of ≥ 2 independent experiments. * 40 nm versus 200 nm p=0.012. 634 635 Figure 5: peptide/MHCII by B cells. C57BL/6 mice were immunized s.c. in the 636 footpad with 100μg of the model antigen EαGFP alone or covalently conjugated 637 to 40 nm or 200 nm particles. Challenge with PBS was included as a negative 638 control. The draining popliteal LNs were harvested after 1, 6, 24, 48 and 72 639 hours post challenge. Flow cytometry was used to determine levels of 640 presentation of the Eαpeptide/MHC class II complex using the monoclonal 641 antibody YAe on B220+CD11c- B cells. The percentage and number of 642 conventional DCs cells staining YAe+ is shown in (A) and (B) respectively. 643 Representative flow cytometry plots are shown for 6 and 72 hours post

21 644 challenge (C, D respectively). Data shows the mean of 3 animals per time point ± 645 standard deviation and are representative of ≥ 2 independent experiments. 646 647 Figure 6: DC and T cell interactions persist after challenge with 200 nm 648 particulate antigen. DsRed OT-II T cells were adoptively transferred into 649 CD11cYFP recipients and footpad challenged with 100µg of OVA alone, 650 conjugated to 40 nm or 200 nm particles. Popliteal LNs were imaged at 72 hours. 651 Representative images following challenge (A) correspond with Video 1 movie 652 file. The duration of OT-II T cell interaction with DCs was determined as 653 intersection of DsRed and YFP signals (B). The velocity (C), displacement rate (D) 654 and meandering index (E) for T cells were calculated. Data is representative of 655 ≥3 individual animals and shows pooled tracks with mean values of three 656 separate areas of a lymph node. Scale bars represent 50μm. 657 658 Figure 7: Blockade of stage 3 DC/T cell interactions following 200 nm 659 particulate challenge prevents Tfh differentiation. Disruption of stage 3 660 DC/T cell interaction was demonstrated by imaging DsRed OT-II T cells 661 adoptively transferred into CD11cYFP recipients. Mice were footpad challenged 662 with 100µg of OVA conjugated to 200 nm particles and popliteal LNs imaged at 663 72 hours. 500µg isotype (mIgG2a) or Y3P (anti-mouse I-A) was given i.v. 2 hours 664 prior to imaging. Representative movie frames show areas of intersection in grey 665 and highlighted by white arrows (A, see Video 2 for movie). Scale bars represent 666 25μm. The duration of OT-II T cell interaction with DCs was determined as 667 intersection of DsRed and YFP signals (B). Next, CFSE OT-II TcR transgenic T 668 cells were adoptively transferred into C57BL/6 recipients. Mice were then 669 challenged with PBS, OVA with CFA or OVA conjugated to 200 nm particles. Mice 670 were treated with a single dose of Y3P or isotype antibody i.v. 72 hours post 671 challenge with particles. Draining LNs were harvested on day 5 post challenge. 672 (C) Representative plots showing CFSE division of CD45.1+ OT-II TcR transgenic 673 T cells (gated on CD4+ population). Tfh cells were defined as being PD-1+CXCR5+ 674 (D) and confirmed as ICOS+ (data not shown) for plotting Tfh proportion as a 675 percentage of the OT-II population (E). Data is representative of 2 independent 676 experiments with 5 animals per group.

22 677 678 Figure 8: Cognate interactions between B and T cells are not required for 679 early Tfh differentiation. OT-II TcR transgenic T cells and were adoptively 680 transferred into C57BL/6 or intact MD4 BcR transgenic recipients. Mice were 681 then challenged with OVA conjugated to 200 nm particles and 500µg isotype 682 (mIgG2a) or Y3P (anti-mouse I-A) given i.v. 72 hours later. Challenges with PBS, 683 OVA/CFA were included as negative and positive controls. Flow cytometry was 684 used to assess CD4+CD45.1+ OT-II cell expansion in LNs 5 days post challenge (A). 685 Tfh were defined by PD-1 and CXCR5 co-expression (B). Panels show mean value 686 and 5 individual animals, *** p <0.001. 687 688 689

690 Video 1: Imaging DC and T cell behaviour after challenge with 200 nm 691 particulate antigen. DsRed OT-II T cells were adoptively transferred into 692 CD11cYFP recipients and footpad challenged with 100µg of OVA conjugated to 693 40 nm or 200 nm particles. Popliteal LNs were imaged at 72 hours. 2 hours prior 694 to imaging, 200 nm challenged groups were given 500ug mIgG2a or Y3P (anti- 695 mouse I-A). Data is representative of ≥3 individual animals and shows one of 696 three separate areas imaged per lymph node. Scale bar represents 50μm.

697 Video 2: Disruption of DC and T cell interactions using Y3P. As for video 1, 698 DsRed OT-II T cells were adoptively transferred into CD11cYFP recipients and 699 footpad challenged with 100µg of OVA conjugated to 40 nm or 200 nm particles. 700 Popliteal LNs were imaged at 72 hours. 2 hours prior to imaging, 200 nm 701 challenged groups were given 500µg mIgG2a or Y3P (anti-mouse I-A). Movies 702 were acquired using a 20X/1.0NA water-immersion objective lens and 2X zoom. 703 Areas of interaction are shown in grey. Scale bars represent 25μm.

704

23     0.4  0.4 0.3 0.3    0.2 0.2  Absorbance Absorbance

0.1 IgG1 anti-OVA 0.1 anti-OVA IgG2c anti-OVA

0.0 0.0

PBS OVA PBS OVA 20 nm 40 nm 20 nm 40 nm 100 nm200 nm 100 nm200 nm 1000 nmCFA/OVA 1000 nmCFA/OVA

110     PBS 100 soluble HA 40nm HA 200nm HA 90 CFA/HA

80 0 1 2 3 4 5 6 7 8 9 10 11 12 day post infection  

1.00   4.0x105

0.75 3.0x105   0.50 ( 2.0x105  ""! 5 0.25 ( 1.0x10

0.00  0      #! "!!      #! "!!        50  '  ( 40

""! 30 (

 20 ( 10  $ 0      #! "!!   

        4 4 4  10 10 10 4 4 10 10             

3 3 3 3 10 10 10 103 10

2 2 2 10 10 10 102 102 FL2-H: FAS PE FL2-H: FAS PE FL2-H: FAS PE FL2-H: FAS PE FL2-H: FAS PE   1 1 1 1 10 10 10 10 101      0 0 0 0 10 10 10 10 100 0 1 2 3 4 0 1 2 3 4 0 1 2 3 4 0 1 2 3 4 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 100 101 102 103 104 FL1-H: GL7 FITC FL1-H: GL7 FITC FL1-H: GL7 FITC FL1-H: GL7 FITC FL1-H: GL7 FITC       14 %  7.5x105 %  12 '3( 4 10 5.0x105 '" ( 8 4 - 4

6 -

4 5 4 2.5x10   2 0   0  &  % -* ,**  &  % -* ,**  &   &  4 ) 12 ))) 7.5x10 10 8 5.0x104 6 4 2.5x104

2 "  & 

 ! &  0 0  &  % -* ,**  &  % -* ,** &  & 

 100 ,** 80 60

5 10 3# 40

4 20 104 &+ 0 4 2 3 4 5 0$*/ . 010 10 10 10 103   &+ 100 102 80 0 60

2 3 4 5

010 10 10 10 3# 40

20

0 . 2 3 4 5 010 10 10 10       PBS EGFP 25 )) 1.0x106 40nm 200nm 20 7.5x105 &   &   α 15 α 5.0x105 ) 10  # 2.5x105

  5 "  # 0 0 0 20 40 60 80 0 20 40 60 80 hours hours   α  .* ,**

*%.4*%,3 04-%,3 /4.%-3 ,+4-%+3 "

α &  '(    PBS

35 8×106 EGFP 30 40nm 6

'   6×10 25 '   α α 200nm 20 4×106 15 10 2×106 5  !$  #     !$ 0 0 0 20406080 0 20406080 hours hours  1#   α  /+ -++

,&.37+&06 -2&/706 ,.7,&/6 ,&07+&-6 #

α!'  ()  2-#   α  /+ -++

+&-.7+&,6 ,&07+&.6 +&-07+&,6 +&237+&/6 #

α!'  ()         

!!!  !!! "20 20

15 15

10 10   5 5          0 0 naive 40nm 200nm naive 40nm 200nm   12 !!! 1.00 10 0.75 8 6 0.50

  4 0.25      2      0 0.00 naive 40nm 200nm naive 40nm 200nm          &&& 20

15

    10

5

      0  isotype Y3P

    $ +)) +))1, 

105 105 105 105 105

104 104 104 104 104 C5 C5 CD45.1 A C5

103 103 103 103 103 VioBlue

-.#*

102 102 102 V110 A: 2 102

0 0 0 0 0

0102 103 104 105 0102 103 104 105 0102 103 104 105 0102 103 104 105 0102 103 104 105  CCS CCS CCS CCS CCS     $ +)) +))1, 

105 105 105 105 105 )#,00.38 -#*/4.16 *+#+12.2 *+ 12 ,#+ 3.2 A 4 4 4 - 4 4 1

10 10 10 10- 10 PD 1 A PD - 7 y y C 103 103 103 10- 3 103 PE Cy7 PE

: A %* - B4-A: PE-Cy7 -PD-1-A B4 A: B4 B4-A: PE-Cy7 -PD-1-A 102 102 102 102 102

0 0 0 0 0

0102 103 104 105 0102 103 104 105 0102 103 104 105 0102 103 104 105 0102 103 104 105 R1 A: APC CXCR5 A R1-A: APC -CXCR5-A R1-A: APC -CXCR5-A R1 A: APC CXCR5 A R1-A: APC -CXCR5-A  . &&&  14 12 10 8 6 4 2

 %  0    $ +)) +)) 1! 1,       10  14 8 12 10 6 8 4 6

% of OT-II cells 4 CXCR5+ PD-1+ CXCR5+ % of LN cells

DsRed+ CD4+ DsRed+ 2 2 0 0