Inducible Bronchus-Associated Lymphoid Tissues (iBALT) Serve … · 2019. 4. 16. · 229 230 231...

12
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 112 113 114 ORIGINAL RESEARCH published: xx March 2019 doi: 10.3389/fimmu.2019.00611 Frontiers in Immunology | www.frontiersin.org 1 March 2019 | Volume 10 | Article 611 Edited by: Aurelio Cafaro, Istituto Superiore di Sanità (ISS), Italy Reviewed by: Michelle Angela Linterman, Babraham Institute (BBSRC), United Kingdom Andreas Hutloff, Deutsches Rheuma- Q2 Forschungszentrum (DRFZ), Germany *Correspondence: Stephen J. Kent [email protected] Adam K. Wheatley [email protected] Specialty section: This article was submitted to Viral Immunology, a section of the journal Frontiers in Immunology Received: 21 January 2019 Accepted: 07 March 2019 Published: xx March 2019 Citation: Tan H-X, Esterbauer R, Vanderven HA, Juno JA, Kent SJ and Wheatley AK (2019) Inducible Bronchus-Associated Lymphoid Tissues (iBALT) Serve as Sites of B Cell Selection and Maturation Following Influenza Infection in Mice. Front. Immunol. 10:611. doi: 10.3389/fimmu.2019.00611 Inducible Bronchus-Associated Lymphoid Tissues (iBALT) Serve as Sites of B Cell Selection and Maturation Following Influenza Infection in Mice Q1 Hyon-Xhi Tan 1 , Robyn Esterbauer 1 , Hillary A. Vanderven 2 , Jennifer A. Juno 1 , Stephen J. Kent 1,3,4 * and Adam K. Wheatley 1 * 1 Department of Microbiology and Immunology, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia, 2 Biomedicine, College of Public Health, Medical and Veterinary Sciences, James Cook University, Douglas, QLD, Australia, 3 Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, VIC, Australia, 4 ARC Centre for Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Parkville, VIC, Australia Seasonally recurrent influenza virus infections are a significant cause of global morbidity and mortality. In murine models, primary influenza infection in the respiratory tract elicits potent humoral responses concentrated in the draining mediastinal lymph node and the spleen. In addition to immunity within secondary lymphoid organs (SLO), pulmonary infection is also associated with formation of ectopic inducible bronchus-associated tissues (iBALT) in the lung. These structures display a lymphoid organization, but their function and protective benefits remain unclear. Here we examined the phenotype, transcriptional profile and antigen specificity of B cell populations forming iBALT in influenza infected mice. We show that the cellular composition of iBALT was comparable to SLO, containing populations of follicular dendritic cells (FDC), T-follicular helper (Tfh) cells, and germinal center (GC)-like B cells with classical dark- and light-zone polarization. Transcriptional profiles of GC B cells in iBALT and SLO were conserved regardless of anatomical localization. The architecture of iBALT was pleiomorphic and less structurally defined than SLO. Nevertheless, we show that GC-like structures within iBALT serve as a distinct niche that independently support the maturation and selection of B cells primarily targeted against the influenza virus nucleoprotein. Our findings suggest that iBALT, which are positioned at the frontline of the lung mucosa, drive long-lived, and unique GC reactions that contribute to the diversity of the humoral response targeting influenza. Keywords: germinal center (GC), influenza, iBALT, B cell, humoral immunity INTRODUCTION The generation of adaptive immune responses to infection requires the complex and elegant Q5 coordination of T and B lymphocytes, concentrated within secondary lymphoid organs (SLO), such as the spleen and lymph nodes. SLO are highly organized and regulated structures, facilitating antigen concentration, capture and presentation to naïve lymphocytes, thereby facilitating the

Transcript of Inducible Bronchus-Associated Lymphoid Tissues (iBALT) Serve … · 2019. 4. 16. · 229 230 231...

  • 1

    2

    3

    4

    5

    6

    7

    8

    9

    10

    11

    12

    13

    14

    15

    16

    17

    18

    19

    20

    21

    22

    23

    24

    25

    26

    27

    28

    29

    30

    31

    32

    33

    34

    35

    36

    37

    38

    39

    40

    41

    42

    43

    44

    45

    46

    47

    48

    49

    50

    51

    52

    53

    54

    55

    56

    57

    58

    59

    60

    61

    62

    63

    64

    65

    66

    67

    68

    69

    70

    71

    72

    73

    74

    75

    76

    77

    78

    79

    80

    81

    82

    83

    84

    85

    86

    87

    88

    89

    90

    91

    92

    93

    94

    95

    96

    97

    98

    99

    100

    101

    102

    103

    104

    105

    106

    107

    108

    109

    110

    111

    112

    113

    114

    ORIGINAL RESEARCHpublished: xx March 2019

    doi: 10.3389/fimmu.2019.00611

    Frontiers in Immunology | www.frontiersin.org 1 March 2019 | Volume 10 | Article 611

    Edited by:

    Aurelio Cafaro,

    Istituto Superiore di Sanità (ISS), Italy

    Reviewed by:

    Michelle Angela Linterman,

    Babraham Institute (BBSRC),

    United Kingdom

    Andreas Hutloff,

    Deutsches Rheuma-

    Q2

    Forschungszentrum

    (DRFZ), Germany

    *Correspondence:

    Stephen J. Kent

    [email protected]

    Adam K. Wheatley

    [email protected]

    Specialty section:

    This article was submitted to

    Viral Immunology,

    a section of the journal

    Frontiers in Immunology

    Received: 21 January 2019

    Accepted: 07 March 2019

    Published: xx March 2019

    Citation:

    Tan H-X, Esterbauer R,

    Vanderven HA, Juno JA, Kent SJ and

    Wheatley AK (2019) Inducible

    Bronchus-Associated Lymphoid

    Tissues (iBALT) Serve as Sites of B

    Cell Selection and Maturation

    Following Influenza Infection in Mice.

    Front. Immunol. 10:611.

    doi: 10.3389/fimmu.2019.00611

    Inducible Bronchus-AssociatedLymphoid Tissues (iBALT) Serve asSites of B Cell Selection andMaturation Following InfluenzaInfection in Mice

    Q1Hyon-Xhi Tan1, Robyn Esterbauer 1, Hillary A. Vanderven2, Jennifer A. Juno1,Stephen J. Kent 1,3,4* and Adam K. Wheatley 1*

    1 Department of Microbiology and Immunology, University of Melbourne, The Peter Doherty Institute for Infection and

    Immunity, Melbourne, VIC, Australia, 2 Biomedicine, College of Public Health, Medical and Veterinary Sciences, James Cook

    University, Douglas, QLD, Australia, 3 Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Hospital

    and Central Clinical School, Monash University, Melbourne, VIC, Australia, 4 ARC Centre for Excellence in Convergent

    Bio-Nano Science and Technology, University of Melbourne, Parkville, VIC, Australia

    Seasonally recurrent influenza virus infections are a significant cause of global morbidity

    and mortality. In murine models, primary influenza infection in the respiratory tract elicits

    potent humoral responses concentrated in the draining mediastinal lymph node and the

    spleen. In addition to immunity within secondary lymphoid organs (SLO), pulmonary

    infection is also associated with formation of ectopic inducible bronchus-associated

    tissues (iBALT) in the lung. These structures display a lymphoid organization, but their

    function and protective benefits remain unclear. Here we examined the phenotype,

    transcriptional profile and antigen specificity of B cell populations forming iBALT in

    influenza infected mice. We show that the cellular composition of iBALT was comparable

    to SLO, containing populations of follicular dendritic cells (FDC), T-follicular helper (Tfh)

    cells, and germinal center (GC)-like B cells with classical dark- and light-zone polarization.

    Transcriptional profiles of GC B cells in iBALT and SLO were conserved regardless of

    anatomical localization. The architecture of iBALT was pleiomorphic and less structurally

    defined than SLO. Nevertheless, we show that GC-like structures within iBALT serve as a

    distinct niche that independently support the maturation and selection of B cells primarily

    targeted against the influenza virus nucleoprotein. Our findings suggest that iBALT, which

    are positioned at the frontline of the lung mucosa, drive long-lived, and unique GC

    reactions that contribute to the diversity of the humoral response targeting influenza.

    Keywords: germinal center (GC), influenza, iBALT, B cell, humoral immunity

    INTRODUCTION

    The generation of adaptive immune responses to infection requires the complex and elegant Q5

    coordination of T and B lymphocytes, concentrated within secondary lymphoid organs (SLO), suchas the spleen and lymph nodes. SLO are highly organized and regulated structures, facilitatingantigen concentration, capture and presentation to naïve lymphocytes, thereby facilitating the

    https://www.frontiersin.org/journals/immunologyhttps://www.frontiersin.org/journals/immunology#editorial-boardhttps://www.frontiersin.org/journals/immunology#editorial-boardhttps://www.frontiersin.org/journals/immunology#editorial-boardhttps://www.frontiersin.org/journals/immunology#editorial-boardhttps://doi.org/10.3389/fimmu.2019.00611http://crossmark.crossref.org/dialog/?doi=10.3389/fimmu.2019.00611&domain=pdf&date_stamp=2019-03-xxhttps://www.frontiersin.org/journals/immunologyhttps://www.frontiersin.orghttps://www.frontiersin.org/journals/immunology#articleshttps://creativecommons.org/licenses/by/4.0/mailto:[email protected]:[email protected]://doi.org/10.3389/fimmu.2019.00611https://www.frontiersin.org/articles/10.3389/fimmu.2019.00611/fullhttp://loop.frontiersin.org/people/655047/overviewhttp://loop.frontiersin.org/people/659268/overviewhttp://loop.frontiersin.org/people/660782/overviewhttp://loop.frontiersin.org/people/392226/overviewhttp://loop.frontiersin.org/people/43341/overviewhttp://loop.frontiersin.org/people/559990/overview

  • 115

    116

    117

    118

    119

    120

    121

    122

    123

    124

    125

    126

    127

    128

    129

    130

    131

    132

    133

    134

    135

    136

    137

    138

    139

    140

    141

    142

    143

    144

    145

    146

    147

    148

    149

    150

    151

    152

    153

    154

    155

    156

    157

    158

    159

    160

    161

    162

    163

    164

    165

    166

    167

    168

    169

    170

    171

    172

    173

    174

    175

    176

    177

    178

    179

    180

    181

    182

    183

    184

    185

    186

    187

    188

    189

    190

    191

    192

    193

    194

    195

    196

    197

    198

    199

    200

    201

    202

    203

    204

    205

    206

    207

    208

    209

    210

    211

    212

    213

    214

    215

    216

    217

    218

    219

    220

    221

    222

    223

    224

    225

    226

    227

    228

    Tan et al. B Cell Characterization in iBALT

    activation, and differentiation of T and B cells into effectorpopulations. Further specialized zones of germinal centers (GC)drive maturation of the humoral response via the selection ofhigh affinity antibodies, providing an effective source of long-lived protection in the form of serum antibody and antigen-experienced memory B and T cell populations.

    While SLO develop during embryogenesis at discreteanatomical locations (1), the induction of immunologicalstructures resembling SLO has been reported at peripheral sitesin response to inflammatory stimuli or infection with pathogenicmicroorganisms (2–5). Variably described as ectoptic lymphoidtissue or tertiary lymphoid organs, such lymphocyte aggregationsalso tend to form proximal to the bronchi within the lung and aretermed inducible bronchus-associated lymphoid tissues (iBALT).Induction of iBALT has been extensively reported followingpulmonary infections by bacterial (6, 7) or viral agents (8–10).iBALT can also form in response to exposure to particulates (11,12) or pro-inflammatory mediators (13, 14), and chronic allergicor inflammatory diseases such as COPD and asthma (15). Despiteconservation across numerous species and a wide number ofcontexts, the contribution of iBALT to humoral immunity againstinfluenza is not fully understood.

    While it is unlikely that a single pathway for iBALTgenesis exists given the wide range of originating stimuli, thedevelopmental models proposed for ectoptic lymphoid tissueshave commonalities (16). Based on these models, cytokine andchemokine production from innate cells such as ILCs andγδT cells recruit pro-inflammatory granulocyte populations,which in turn drive the CXCL13-dependent recruitment ofmature B cells into the lung (14, 17–19). Similar to themaintenance of conventional lymphoid tissues, T and B cellssupport the differentiation of stromal cells into mature folliculardendritic cells (FDC) and fibroblastic reticular cells (FRC),with homeostatic maintenance allowing these lymphoid-likestructures to persist for months in the absence of ongoinginflammation or infection (20–22). While iBALT is decidedlypleiomorphic (16), several groups have reported the presenceof B-cell clusters, networks of FDC, T cell areas, and zonesresembling germinal centers, with iBALT supporting theproliferation of both B and T cells (8–10, 13, 23). Notably,iBALT can initiate protective humoral and T cell responsesfollowing influenza infection in mice lacking SLO (8, 9),although the relative importance of lung-generated responses inimmunocompetent animals is unclear.

    Using the influenza infection model in mice, we sought toaddress two questions. First, to what extent do the GC-likestructures in the lung resemble GC in SLO? In particular, doiBALT GC-like structures facilitate B cell selection and affinitymaturation? Second, to what extent is the lung a primingsite for humoral immunity to pulmonary influenza infectionin immunocompetent animals? We show GC responses ininfluenza infection-induced iBALT were comparable to GCin SLO in terms of cellular composition, phenotype, andtranscriptional profile. However, the architecture of iBALTGC was non-classical, lacking discrete light and dark zonepolarization and T cell compartmentalization to the light zone.Nevertheless, using labeled recombinant influenza antigens

    to probe B cell specificities, we find that influenza-specificGC responses within iBALT predominantly targeted the viralnucleoprotein, constituted a unique clonal repertoire relative toSLO, and demonstrated a comparable capacity to drive somaticmutagenesis and B cell selection requisite for affinity maturation.Our work has implications for the development of improvedhumoral immunity to influenza by establishing long-lived andunique GC reactions at the frontline of the lung mucosa.

    MATERIALS AND METHODS

    Animal InfectionAll animal procedures were approved by the University Q7

    of Melbourne Animal Ethics Committee. C57BL/6 mice at6–10 weeks of age were used. Mice were anesthetizedby isoflurane inhalation prior to infection. For intranasalinfections, mice were instilled with 50 µL volume of 50TCID50 of A/Puerto Rico/8/34 (PR8).

    Confocal MicroscopyFresh tissues were snap-frozen in O.C.T. compound (SakuraFinetek USA) and stored at −80◦C. Tissues were sectioned at7µm thickness (Leica). Prior to staining, sectioned tissues werefixed in cold acetone solution (Sigma) for 10min then rehydratedwith PBS for 10min and blocked with 5% (w/v) bovine serumalbumin (Sigma) and 2% (v/v) normal goat serum (NGS).Cell staining was performed using the following antibodies:IgD (11-26c.2a; Biolegend), B220 (RA3-6B2; BD), GL7 (GL7;Biolegend), CD35 (8C12; BD), CD3 (17A2; Biolegend), CD169(3D6.112; BD), CD86 (GL1, Biolegend), CXCR4 (2B11; BD),CD4 (GK1.5; Biolegend), Ki67 (11F6, Biolegend), BCL6 (K112-91; BD). To amplify the CXCR4-PE signal, tissues were stainedsequentially with rabbit anti-PE antibody (polyclonal; NovusBiologicals) and a secondary goat anti-rabbit IgG Alexa Fluor555 antibody (polyclonal; Life Technologies). Slides were sealedwith ProLong Diamond Antifade Mountant (Life Technologies).Tiled Z-stack images covering were captured on a Zeiss LSM710microscope. Post-processing of confocal images was performedwith ImageJ v2.0.0.

    HA ProteinsRecombinant HA protein for use as flow cytometry probeswas derived for A/Puerto Rico/08/1934 as previously described(24). Briefly, expression constructs were synthesized (GeneArt)and cloned into mammalian expression vectors. Proteinswere expressed by transient transfection of Expi293 (LifeTechnologies) suspension cultures and purified by polyhistidine-tag affinity chromatography and gel filtration. Proteins werebiotinylated using BirA (Avidity) and stored at −80◦C.Prior to use, biotinylated HA proteins were labeled by thesequential addition of streptavidin (SA) conjugated to BV421,phycoerythrin (PE), or allophycocyanin (APC), and stored at4◦C. Recombinant influenza A H1N1 NP protein (11675-V08B;Sino Biological) was labeled with PE or APC fluorochromes usingcommercial conjugation kits, as per manufacturer’s protocol(AB102918, AB 201807; Abcam).

    Frontiers in Immunology | www.frontiersin.org 2 March 2019 | Volume 10 | Article 611

    https://www.frontiersin.org/journals/immunologyhttps://www.frontiersin.orghttps://www.frontiersin.org/journals/immunology#articles

  • 229

    230

    231

    232

    233

    234

    235

    236

    237

    238

    239

    240

    241

    242

    243

    244

    245

    246

    247

    248

    249

    250

    251

    252

    253

    254

    255

    256

    257

    258

    259

    260

    261

    262

    263

    264

    265

    266

    267

    268

    269

    270

    271

    272

    273

    274

    275

    276

    277

    278

    279

    280

    281

    282

    283

    284

    285

    286

    287

    288

    289

    290

    291

    292

    293

    294

    295

    296

    297

    298

    299

    300

    301

    302

    303

    304

    305

    306

    307

    308

    309

    310

    311

    312

    313

    314

    315

    316

    317

    318

    319

    320

    321

    322

    323

    324

    325

    326

    327

    328

    329

    330

    331

    332

    333

    334

    335

    336

    337

    338

    339

    340

    341

    342

    Tan et al. B Cell Characterization in iBALT

    Flow CytometryFor experiments requiring discrimination of blood and tissuepopulations, mice were intravenously labeled with 3 µg ofCD45.2 antibody (104; eBioscience) in a 200 µL volume priorto tissue collection. Tissues were mechanically homogenizedinto single cell suspensions in RF10 media (RPMI 1640, 10%FCS, 1 × penicillin-streptomycin-glutamine; Life Technologies).Except for MLN, red blood cell lysis was performed withPharm LyseTM (BD). Isolated cells were Fc-blocked with aCD16/32 antibody (93; Biolegend). For all B cell experiments,cells were surface stained with Live/dead Aqua viability dye(Thermofisher) and B220 (RA3-6B2; BD), IgD (11-26c.2a;BD), CD45 (30-F11, BD), GL7 (GL7; Biolegend), CD38 (90;Biolegend), Streptavidin (BD), CD3 (145-2C11; Biolegend),and F4/80 (BM8; Biolegend). For LZ/DZ experiments cellswere also stained with CD86 (GL1, Biolegend) and CXCR4(2B11; BD). Antigen-specific B cells were detected basedupon dual staining with HA or NP probes conjugated toAPC and PE. For Tfh cell characterization, cells were stainedin Transcription Factor Buffer Set (BD) with Live/deadRed viability dye (Life Technologies), BCL6 (IG191E/A8;Biolegend), CD3 (145-2C11; Biolegend), PD-1 (29F.1A12;Biolegend), CXCR5 (L138D7; Biolegend), CD4 (RM4-5; BD),B220 (RA3-6B2; BD), and F4/80 (T45-2342; BD). Stainedcells were washed twice and fixed with 1% formaldehyde(Polysciences) for acquisition on a BD LSR Fortessa, orresuspended in Optimem (Life Technologies) for sorting on aBD FACSAria III.

    RNAseq and Data AnalysisSingle cell suspensions were processed and stained as describedabove. Memory B cells (B220+ IgD- CD38hi GL7-), GCB cells (B220+ IgD- CD38lo GL7+), and T cells (CD3+)were sorted into RLT buffer (Qiagen) containing 0.14Mβ-mercaptoethanol (Sigma). Post-sort, cells were suspendedin a volume of RLT buffer representing 3.5x the sortingsolution volume. Genomic DNA was removed using a gDNAeliminator (Qiagen), according the manufacturer’s protocol.RNA was extracted by addition of 100% ethanol in a 5:7volume ratio to flow-through solution and processed withthe RNeasy Plus Micro Kit (Qiagen), according to themanufacturer’s instructions. The Australian Genome ResearchFacility (Melbourne, Australia) performed the RNAseq withan Illumina HiSeq 2500 and obtained 100bp single reads.The library was prepared with a TruSeq Stranded mRNALibrary Prep Kit (Illumina). RNAseq analysis was performedusing the web-based Galaxy platform maintained by MelbourneBioinformatics (25). Reads were mapped to the Mus musculusreference genome (mm10) using HISAT2 (26) and reads werequantified using HTSeq (27). Count matrices were generatedand inputted into Degust (http://degust.erc.monash.edu) fordata analysis and visualization with the Voom/Limma methodselected for data processing. Heat maps were generated usingMorpheus (The Broad Institute; https://software.broadinstitute.org/morpheus/). Raw sequence reads can be accessed with GEOcode: (pending approval).

    Sequencing and Analysis of Murine B CellReceptor GenesSequencing of murine heavy chain immunoglobulin sequenceswas performed as previously described (28). Briefly, single Bcells stained with the panel above and NP-PE or HA-BV421probes were sorted into 96-well plates and cDNA preparedusing SuperScript III Reverse Transcriptase (Life Technologies)and random hexamer primers (Life Technologies). Heavy chainimmunoglobulin sequences were amplified by nested PCR usingHotStar Taq polymerase (Qiagen) and multiplex primers bindingV-gene leader sequences or the immunoglobulin constantregions. PCR products were Sanger sequenced (Macrogen) andVDJ recombination analyzed using High V-QUEST on IMGT(29). Clonality was determined based upon shared gene usage andCDR-H3 length and sequence similarity. Circular layout graphicswere generated using the Circlize package in R (30).

    StatisticsData are generally presented as the median ± interquartilerange or the mean ± SD. Flow data was analyzed in FlowJo v9(FlowJo) and all statistical analyses were performed using Prismv7 (GraphPad).

    RESULTS

    Dynamics of iBALT Induction FollowingIntranasal Influenza InfectionTo study lung B cell responses to influenza, we first performedintranasal infection of mice with A/Puerto Rico/08/1934 (PR8)virus. Consistent with previous reports (8, 10, 31, 32),intranasal infection resulted in a pronounced infiltration ofB cells into the lung. Lung-infiltrating B cells, distinguishedfrom blood-circulating populations by intravenous CD45.2labeling, displayed peak numbers 14 days (d14) post-infectionand persisted up to d112 (Figure 1A; gating Figure S1). Asubpopulation of B cells expressing a GC-like phenotype (B220+IgD- GL7+ CD38lo) were evident in lungs at d14 post-infection and detectable up to d112, albeit waning at thelatter timepoint (Figure 1B). In comparison, mediastinal LN(MLN) displayed the largest proportion of GC B cells withcontinued maintenance at high frequencies up to d112 post-infection, in line with our previous observations (33). SplenicGC B cells frequencies peaked at d14, rapidly waned andwas proportionally lowest amongst the tissues analyzed fromd28 onward.

    When visualized using confocal microscopy, lung tissuesfrom infected mice similarly showed B cell infiltration andclustering (Figure 1C). Although B cell infiltration was observedat d7, cells were not structurally organized and were mostlydispersed around the bronchi regions. GC-like structures,demarcated by GL7 staining, were apparent at d14, peakingin size between d28 and d56, and subsequently waning insize by d112. When analyzed by flow cytometry, lung Bcells gradually acquired a canonical GC phenotype in latertimepoints, with full differentiation into GL7+ CD38lo and peakabsolute numbers of GC B cells observed by d35 (Figure S2).

    Frontiers in Immunology | www.frontiersin.org 3 March 2019 | Volume 10 | Article 611

    http://degust.erc.monash.eduhttps://software.broadinstitute.org/morpheus/https://software.broadinstitute.org/morpheus/https://www.frontiersin.org/journals/immunologyhttps://www.frontiersin.orghttps://www.frontiersin.org/journals/immunology#articles

  • 343

    344

    345

    346

    347

    348

    349

    350

    351

    352

    353

    354

    355

    356

    357

    358

    359

    360

    361

    362

    363

    364

    365

    366

    367

    368

    369

    370

    371

    372

    373

    374

    375

    376

    377

    378

    379

    380

    381

    382

    383

    384

    385

    386

    387

    388

    389

    390

    391

    392

    393

    394

    395

    396

    397

    398

    399

    400

    401

    402

    403

    404

    405

    406

    407

    408

    409

    410

    411

    412

    413

    414

    415

    416

    417

    418

    419

    420

    421

    422

    423

    424

    425

    426

    427

    428

    429

    430

    431

    432

    433

    434

    435

    436

    437

    438

    439

    440

    441

    442

    443

    444

    445

    446

    447

    448

    449

    450

    451

    452

    453

    454

    455

    456

    Tan et al. B Cell Characterization in iBALT

    FIGURE 1 | iBALT formation and characterization following intranasal influenza infection in mice. (A) Lung B cell infiltration (B220+ intravenous (IV) CD45.2-) and (B)Q4

    Q3 frequency of GC B cells (B220+ IgD- CD38lo GL7+) across various tissues were measured in mice infected intranasally with A/Puerto Rico/08/1934. Data representtwo independent experiment (n = 6). Error bars represent mean ± SD. (C) Induction and maturation of iBALT across various time-points visualized by composite

    images comprising B220 (orange), IgD (gray), GL7 (green), and CD35 (cyan); scale bar−100µM. (D) GC cellular composition of lungs and spleen visualized by

    immunofluorescence staining at d35 post-infection; scale bar−50µM. (E) Frequency and (F) visualization of light- and dark-zone GC B cells in lungs and SLO at d35

    post-infection. Data represent a single experiment (n = 6). Error bars represent mean ± SD. Scale bar−50 µM.

    Frontiers in Immunology | www.frontiersin.org 4 March 2019 | Volume 10 | Article 611

    https://www.frontiersin.org/journals/immunologyhttps://www.frontiersin.orghttps://www.frontiersin.org/journals/immunology#articles

  • 457

    458

    459

    460

    461

    462

    463

    464

    465

    466

    467

    468

    469

    470

    471

    472

    473

    474

    475

    476

    477

    478

    479

    480

    481

    482

    483

    484

    485

    486

    487

    488

    489

    490

    491

    492

    493

    494

    495

    496

    497

    498

    499

    500

    501

    502

    503

    504

    505

    506

    507

    508

    509

    510

    511

    512

    513

    514

    515

    516

    517

    518

    519

    520

    521

    522

    523

    524

    525

    526

    527

    528

    529

    530

    531

    532

    533

    534

    535

    536

    537

    538

    539

    540

    541

    542

    543

    544

    545

    546

    547

    548

    549

    550

    551

    552

    553

    554

    555

    556

    557

    558

    559

    560

    561

    562

    563

    564

    565

    566

    567

    568

    569

    570

    Tan et al. B Cell Characterization in iBALT

    As mature persistent GC responses are well-established inboth iBALT and the draining MLN by d35, we selectedthis as a suitable timepoint for subsequent GC analysis. Wecontrasted the structural characteristics of putative lung GC withcanonical splenic GC from infected mice at d35 post-infection(Figure 1D). Despite a pleiomorphic appearance (Figure S3),both FDC (CD35+) and highly clustered B cells (B220+)expressing GL7 were detected in iBALT analogous to thespleen. In contrast, T cells (CD3+) were dispersed throughoutthe lung tissues and CD169 staining, which demarcatessubcapsular sinus macrophages in the spleen, was negligible iniBALT structures.

    GL7 expression in SLO is tightly linked to GC localization(34). The distributed GL7 staining throughout iBALT ledus to question if the observed GC-like B cell structureswere functionally analogous to actual germinal centers. Wetherefore examined the differential expression CXCR4 andCD86,previously described for the delineation of dark- and light-zoneresident cells in both human and murine GC (35). GC B cellsfrom the lung displayed the characteristic differential expressionprofiles of CXCR4 and CD86, analogous to staining in MLNand spleen (Figure 1E). However, CXCR4 and CD86 stainingvisualized by immunofluorescence microscopy on lung residentB cell clusters was diffuse, lacking the discrete localization oflight- and dark-zone staining seen in splenic samples (Figure 1F).Light and dark zone polarization requires differential chemokineexpression by stromal cells, including FDC (36). Comparedto the expected focused and GC-polarized FDC staining inMLN and spleen, FDC staining within the lung was diffusewith little indication of light-zone localization (Figure S4). Thus,while GC-like structures within influenza-induced iBALT displaymany phenotypic characteristics consistent with conventionalGC within SLO, they appear morphologically unconventional.

    Transcriptional Profile of GC-Like B CellsFrom iBALTTo facilitate an in-depth comparison of GC B cells fromdifferent anatomical sites, memory, and germinal center B cellswere sorted from the lung, spleen, MLN and blood of miceat d35 post-infection (gating Figure S1), and differential geneexpression profiles were generated by RNASeq. Based uponmulti-dimensional scaling, iBALT GC-like B cells clusteredclosely with GC B cells from SLO and were distinct from bothmemory B cell populations and T cell controls (Figure 2A).This was similarly evident within a heatmap generated usingthe top 200 differentially expressed genes, where memory andGC-like populations displayed discrete expression profiles acrosslung-, MLN-, and spleen-localized cells (Figure 2B). Notably,iBALT GC-like B cells, but not memory B cells, expressed themaster transcription factor bcl6, and canonical GC B cell markersfas, aidca, gcsam, and s1pr2 in a manner comparable to GC Bcells from SLO (Figure 2C). Conversely, we observed limitedexpression of genes associated with the memory phenotype inGC B cell populations (ccr7, s1pr1, ly6d, cd38, sell). Consistentwith previous BrdU incorporation data (8), iBALTGC-like B cellsappeared to be proliferating (expressing ki67).

    iBALT-Resident Tfh Cells Express Bcl6and Ki67Follicular T helper cells (Tfh) are critical for the generation andmaintenance of germinal centers in SLO (36), but the presence ofTfh in lung iBALT induced by influenza infection is understudied.We found comparable frequencies of Tfh cells (CD3+ CD4+CXCR5++ PD1++) were evident within the lungs and MLNof mice at d35 and d56 post-infection, encompassing close to10% of the total CD4+ T cell population (Figure 3A; gatingFigure S5A). Tfh from both MLN and lungs expressed elevatedlevels of the lineage-defining transcription factor BCL6 (37,38) compared to conventional CD4+ T cells (CXCR5- PD1-)(Figure 3B), althoughwith higher comparative expression (basedupon fluorescence intensity) observed within MLN-derived Tfhcells. Intracellular Ki67 expression suggested that lung Tfh cellscontained a similar proportion of recently proliferating cellscompared to MLN at d35, but marginally higher proliferationthan MLN Tfh cells at d56 (Figure 3C). B220+ cells expressinghigh levels of Ki67 and BCL6 were also present in both lungs andMLN of infected mice, although with frequencies higher in in thelatter tissue (Figure S5B).

    Ki67 (Figure 3D) and BCL6 (Figure 3E) expression in spleenand lungs of mice at d35 post-infection was visualized byconfocal microscopy. As expected, the spleen showed distinctspatial segregation of T and B cell zones. In contrast, we foundCD4T cell and B cell clusters to be substantially smaller in thelungs and their spatial segregation was not clearly delineated.Mirroring expression measured by flow cytometry, Ki67, andBCL6 expression was detected in B220 and CD4 cells of bothspleen and lungs, although expression of these markers was moreevident in B220 cells. Overall, while we observed differences inanatomical localization, Tfh within iBALT-resident GC structuresappear phenotypically analogous to SLO-resident Tfh cells.

    Specificity of GC-Resident B Cells forMajor Influenza AntigensThe antigen specificity of lung- or SLO-localized GC B cellswas analyzed using recombinant influenza HA and NP probes(Figure 4A). NP-specific GC B cells were evident in all infectedmice at both d35 and d56 post-infection at all sites, whereasHA-specific GC B cells were observed almost exclusively in theMLN and spleen, with little to no HA-specific GC B cells in thelungs (Figure 4B). In terms of absolute numbers of HA- and NP-specific GC B cells at both timepoints, the majority of HA-specificGC B cells were found in the MLN, a minor population in thespleen, and little to no HA-specific GC B cells observed in thelungs (Figure 4C). In contrast, NP-specific GC B cell were foundat high frequencies and counts in all tissues, withMLN and spleenshowing similar total cell counts while less cells were detectedin lungs.

    GC Within iBALT Support Maturation ofInfluenza-Specific B CellsWe next examined if iBALT GC structures display a functionalcapacity to drive maturation of the influenza-specific B cellresponse. NP- and HA-specific B cell populations were sorted

    Frontiers in Immunology | www.frontiersin.org 5 March 2019 | Volume 10 | Article 611

    https://www.frontiersin.org/journals/immunologyhttps://www.frontiersin.orghttps://www.frontiersin.org/journals/immunology#articles

  • 571

    572

    573

    574

    575

    576

    577

    578

    579

    580

    581

    582

    583

    584

    585

    586

    587

    588

    589

    590

    591

    592

    593

    594

    595

    596

    597

    598

    599

    600

    601

    602

    603

    604

    605

    606

    607

    608

    609

    610

    611

    612

    613

    614

    615

    616

    617

    618

    619

    620

    621

    622

    623

    624

    625

    626

    627

    628

    629

    630

    631

    632

    633

    634

    635

    636

    637

    638

    639

    640

    641

    642

    643

    644

    645

    646

    647

    648

    649

    650

    651

    652

    653

    654

    655

    656

    657

    658

    659

    660

    661

    662

    663

    664

    665

    666

    667

    668

    669

    670

    671

    672

    673

    674

    675

    676

    677

    678

    679

    680

    681

    682

    683

    684

    Tan et al. B Cell Characterization in iBALT

    FIGURE 2 | Transcriptional profile of GC-like B cells in iBALT. GC (B220+ IgD- GL7+ CD38lo) and memory B cells (B220+ IgD- GL7- CD38hi) were sorted from SLO

    and iBALT in mice at d35 post-infection with PR8. mRNA expression profiles were compared using RNAseq data from 2 to 3 independent sequencing experiments.

    (A) Multi-dimensional scaling plot showing clustering of memory and GC populations relative to T cell controls. (B) Heatmap of the top 200 differentially expressed

    genes, with shading indicating expression relative to the average (red–overexpressed, blue–underexpressed). (C) Differential expression of selected genes previously

    described to demarcate memory and GC subsets. Error bars represent mean ± SD.

    Frontiers in Immunology | www.frontiersin.org 6 March 2019 | Volume 10 | Article 611

    https://www.frontiersin.org/journals/immunologyhttps://www.frontiersin.orghttps://www.frontiersin.org/journals/immunology#articles

  • 685

    686

    687

    688

    689

    690

    691

    692

    693

    694

    695

    696

    697

    698

    699

    700

    701

    702

    703

    704

    705

    706

    707

    708

    709

    710

    711

    712

    713

    714

    715

    716

    717

    718

    719

    720

    721

    722

    723

    724

    725

    726

    727

    728

    729

    730

    731

    732

    733

    734

    735

    736

    737

    738

    739

    740

    741

    742

    743

    744

    745

    746

    747

    748

    749

    750

    751

    752

    753

    754

    755

    756

    757

    758

    759

    760

    761

    762

    763

    764

    765

    766

    767

    768

    769

    770

    771

    772

    773

    774

    775

    776

    777

    778

    779

    780

    781

    782

    783

    784

    785

    786

    787

    788

    789

    790

    791

    792

    793

    794

    795

    796

    797

    798

    Tan et al. B Cell Characterization in iBALT

    FIGURE 3 | Characterization of Tfh cells in iBALT. (A) Frequency of Tfh cells (CD3+ CD4+ CXCR5++ PD1++) in MLN and lungs of mice at d35 and d56

    post-infection with PR8. (B) Intracellular BCL6 staining measured by mean fluorescence intensities in Tfh and non-Tfh CD4T cells (CD3+ CD4+ CXCR5- PD1-)

    isolated from MLN and lungs. (C) Frequency of proliferation marker Ki67 expressed in Tfh and non-Tfh CD4T cells isolated from MLN and lungs. Data represent a

    single experiment (n = 4–5). Error bars represent mean ± SD. Visualization by immunofluorescence microscopy of (D) Ki67 and (E) BCL6 expression in T and B cells

    of spleen and lung iBALT; scale bar−50µM.

    Frontiers in Immunology | www.frontiersin.org 7 March 2019 | Volume 10 | Article 611

    https://www.frontiersin.org/journals/immunologyhttps://www.frontiersin.orghttps://www.frontiersin.org/journals/immunology#articles

  • 799

    800

    801

    802

    803

    804

    805

    806

    807

    808

    809

    810

    811

    812

    813

    814

    815

    816

    817

    818

    819

    820

    821

    822

    823

    824

    825

    826

    827

    828

    829

    830

    831

    832

    833

    834

    835

    836

    837

    838

    839

    840

    841

    842

    843

    844

    845

    846

    847

    848

    849

    850

    851

    852

    853

    854

    855

    856

    857

    858

    859

    860

    861

    862

    863

    864

    865

    866

    867

    868

    869

    870

    871

    872

    873

    874

    875

    876

    877

    878

    879

    880

    881

    882

    883

    884

    885

    886

    887

    888

    889

    890

    891

    892

    893

    894

    895

    896

    897

    898

    899

    900

    901

    902

    903

    904

    905

    906

    907

    908

    909

    910

    911

    912

    Tan et al. B Cell Characterization in iBALT

    FIGURE 4 | Antigen specificity of GC B cells in lung iBALT and SLO. (A) Representative HA and NP probe staining of GC B cells (B220+ IgD- CD38lo GL7+) isolated

    from lung iBALT, MLN, and spleen of mice at d35 post-infection with PR8. (B) Frequency of HA-, NP-specific, and undefined GC B cells isolated from individual mice

    at d35 and d56 post-infection. (C) Absolute counts of HA-specific and NP-specific GC B cells in lung iBALT, MLN, and spleen of mice at d35 and d56 post-infection.

    Data represent two independent experiment (n = 4–5). Error bars represent mean ± SD.

    from GC in the lung and MLN at d14, d35, and d56 and heavychain B cell receptor sequences were recovered, aligned andgrouped into clonal families. Antigen-specific B cell populations

    in the lung (NP) and MLN (NP and HA) consisted of numerousclonally-expanded lineages (Figure 5A), indicating proliferationconsistent with the observed Ki67 expression. We observed

    Frontiers in Immunology | www.frontiersin.org 8 March 2019 | Volume 10 | Article 611

    https://www.frontiersin.org/journals/immunologyhttps://www.frontiersin.orghttps://www.frontiersin.org/journals/immunology#articles

  • 913

    914

    915

    916

    917

    918

    919

    920

    921

    922

    923

    924

    925

    926

    927

    928

    929

    930

    931

    932

    933

    934

    935

    936

    937

    938

    939

    940

    941

    942

    943

    944

    945

    946

    947

    948

    949

    950

    951

    952

    953

    954

    955

    956

    957

    958

    959

    960

    961

    962

    963

    964

    965

    966

    967

    968

    969

    970

    971

    972

    973

    974

    975

    976

    977

    978

    979

    980

    981

    982

    983

    984

    985

    986

    987

    988

    989

    990

    991

    992

    993

    994

    995

    996

    997

    998

    999

    1000

    1001

    1002

    1003

    1004

    1005

    1006

    1007

    1008

    1009

    1010

    1011

    1012

    1013

    1014

    1015

    1016

    1017

    1018

    1019

    1020

    1021

    1022

    1023

    1024

    1025

    1026

    Tan et al. B Cell Characterization in iBALT

    FIGURE 5 | BCR sequencing of GC B cells in the lung and mediastinal lymph node. Heavy chain immunoglobulin sequences were recovered from NP- and

    HA-specific GC B cells sorted from the lung and mediastinal lymph nodes of PR8-infected mice on d14, d25, and d56. Data represents a single experiment with 5

    mice per timepoint. (A) Circular plots detailing the clonal distribution of NP- and HA-specific populations from each site. Each segment represents a unique B cell

    clonotype, the width proportional to the number of clonal family members recovered. Shared clonotypes between sites are indicated by the linking arcs. Number of

    unique clonal families and BCR sequences are indicated below. (B) The degree of V-gene somatic mutation in recovered heavy chain immunoglobulin sequences from

    each site. NP- and HA-specific B cells are indicated in blue and red, respectively. Error bars represent median ± interquartile range.

    discrete compartmentalization of the humoral response, withminimal sharing of clonotypes and different clonal hierarchiesseen between the lung and MLN, indicating B cell recruitmentto each of these sites was highly segregated. This pattern wasconserved across all animals and timepoints analyzed, and isconsistent with previous reports that the lung hosts a uniqueB cell repertoire (39). The observed rates of V-gene somaticmutation in the MLN were comparable for both NP- and HA-specific GC B cells, increasing over time from a median 3.1% to5.2% to 5.3% from d14 to d35 to d56 respectively (Figure 5B).

    Similarly within the lung, while few HA-specific B cells wererecovered, somatic mutation in B cells that were mostly NP-specific increased from 2.4% to 4.5% to 4.6% across the same

    timepoints. Notably, the mutation load of influenza-specific GCB cells within iBALT remained consistently lower (∼0.7%) incomparison to theMLN at each timepoint sampled. However, therate at which somatic mutations accumulated was comparable tothe MLN suggesting an equivalent capacity to drive maturationof humoral responses exists at both sites. Differences observedbetween the tissues are likely due to the delayed biogenesis ofiBALT-localized GC.

    DISCUSSION

    There is considerable interest in the generation of local immunitywithin the lungs to protect from influenza and other respiratory

    Frontiers in Immunology | www.frontiersin.org 9 March 2019 | Volume 10 | Article 611

    https://www.frontiersin.org/journals/immunologyhttps://www.frontiersin.orghttps://www.frontiersin.org/journals/immunology#articles

  • 1027

    1028

    1029

    1030

    1031

    1032

    1033

    1034

    1035

    1036

    1037

    1038

    1039

    1040

    1041

    1042

    1043

    1044

    1045

    1046

    1047

    1048

    1049

    1050

    1051

    1052

    1053

    1054

    1055

    1056

    1057

    1058

    1059

    1060

    1061

    1062

    1063

    1064

    1065

    1066

    1067

    1068

    1069

    1070

    1071

    1072

    1073

    1074

    1075

    1076

    1077

    1078

    1079

    1080

    1081

    1082

    1083

    1084

    1085

    1086

    1087

    1088

    1089

    1090

    1091

    1092

    1093

    1094

    1095

    1096

    1097

    1098

    1099

    1100

    1101

    1102

    1103

    1104

    1105

    1106

    1107

    1108

    1109

    1110

    1111

    1112

    1113

    1114

    1115

    1116

    1117

    1118

    1119

    1120

    1121

    1122

    1123

    1124

    1125

    1126

    1127

    1128

    1129

    1130

    1131

    1132

    1133

    1134

    1135

    1136

    1137

    1138

    1139

    1140

    Tan et al. B Cell Characterization in iBALT

    pathogens. iBALT represent a focus of lymphoid tissues that hasthe potential to improve immunity within the lower respiratorytract. Despite the iBALT being pleiomorphic and less structurallydefined than SLO GC structures, we found GC B cells in iBALTand SLO were transcriptionally similar and show that iBALTserve as a distinct anatomical niche for the maturation andselection of B cells primarily targeted against the influenzavirus nucleoprotein.

    The cellular composition of iBALT can vary, dependingupon the stimuli used for induction (16). Consistent withprevious reports, we observed cellular infiltration concentrated atbronchial junctions that included FDC (8), Tfh and conventionalT cells (8, 9, 32), and germinal center B cell populations(10) following pulmonary influenza infection. In contrast, LPS-induced iBALT exhibits lung-infiltrating T cells with Tfh-likefunction but lacking classical CXCR5/BCL6 expression (13).Further, inoculation with LPS or whole Pseudomonas aeruginosabacteria results in ectopic lung GC structures that lack FDC,in contrast to the CXCL13-expressing FDC observed followinginfection with modified vaccinia virus Ankara (7, 13, 40).Nuances in iBALT biogenesis appear attributable to the extentand nature of inflammation induced by infectious diseasescompared to alternative pulmonary stimuli.

    Germinal centers are highly specialized foci facilitating Bcell proliferation, competition, and selective differentiationin order to drive high affinity serological antibody responses.We find that clusters of GC-like B cells observed withiniBALT appear analogous to GC B cells in SLO with respect tocellular composition and phenotype. This includes evidence ofproliferation, division into light- and dark-zone phenotypes,expression of canonical GC transcription factors, and thepresence of Tfh cells. Here we extend previous studies(8, 40) by transcriptional profiling lung GC-like B cells byRNAseq. We found a conserved transcriptional signaturewas shared by GC B cells independent of anatomicallocation, with both lung- and SLO-resident populationsexpressing canonical transcription factors (e.g., bcl6), mediatorsof affinity maturation (e.g., aidca), and key chemotacticfactors (e.g., s1pr2).

    One key area of difference between GC within SLO vs. iBALTwas the variant morphology, with a lack of highly distinct Tand B cell zones observed in iBALT consistent with previousreports (8, 9, 41), and a lack of spatial segregation of B cellsphenotypically characteristic of light- and dark-zone B cells. Theimpact of atypical GC architecture upon the development of Bcell immunity in the lung is unclear. Previous studies have shownlight- and dark-zone B cells exhibit largely overlapping geneexpression patterns (42). Experiments using CXCR4-deficientmice (43) have established that B cells can cycle between these twostates, and that discrete light- and dark-zone architecture is nota requisite for affinity maturation nor plasma cell differentiation(44). Based upon recovered BCR sequences from GC-residentB cells, we find GC in the lung maintain a capacity to drivematuration of the influenza-specific B cell response, with thecontinued accumulation of somatic mutations observed over56 days post-infection. While overall mutation frequencieswere consistently higher in the draining MLN, the rate of

    accumulation was comparable between the two sites suggestingno detectable defect in the functionality of iBALT-localized GC.

    Given the importance of anti-HA antibody for protectionagainst influenza (45), the contribution of iBALT to serologicalHA-specific antibody seems minimal. Certainly, non-neutralizing antibody responses, such as those targetingNP, can contribute to immunity via Fc-mediated functions orantibody-dependent cellular cytotoxicity, and further provideprotection against heterosubtypic influenza virus challengein mice (46–50). iBALT might conceivably act as a source ofmucosal-resident antibody-secreting cells (ASCs) that contributeto ongoing barrier protection in the lung. We observed strongcompartmentalization of B cell receptor repertoires withinanatomical sites, implying that alternative B cell clonotypes arerecruited to and maintained in lung GC compared to SLO. Thisadditional clonotypic diversity may be advantageous duringrecurrent exposure to antigenically diverse pathogens suchas influenza. The extended persistence of iBALT, reported forup to 3 months (9, 10, 31, 40), may also serve as a broaderniche to position B cells proximal to a site of recent mucosalbarrier damage. Interestingly, we find a significant proportion oflung-resident GL7+ B cells, comprising up to half of total cells,were of undefined specificity. This corroborates previous studiesdemonstrating that the iBALT can act as a general priming site,thus supporting the proliferation of GC B cells binding antigensunrelated to the primary inducing agent (23, 51). While these GCB cells may also simply represent B cells specific for alternativeantigenic targets of the influenza virus not captured by ourprobes (52), or non-specific bystander B cells adopting a GCphenotype, they may also be elicited by antigenic translocationof microorganisms as a result of lung damage during infection.Additional definition of the specificities of iBALT-resident GC Bcells is warranted.

    In summary we find GC-like structures in iBALT arephenotypically, transcriptionally and functionally comparableto those within SLO, yet recruit a distinct subset of Bcell clonotypes into the humoral immune response. WhileiBALT can drive B cell diversification and may facilitateresidence of B and T lymphocytes proximal to the mucosa,the protective value of iBALT within immunocompetenthosts remains unclear, and in the context of our influenzainfection model, seems largely restricted to targeting theviral nucleoprotein.

    DATA AVAILABILITY

    The datasets generated for this study can befound in Gene Expression Omnibus, NCBI (Accession Q10

    Q11number pending).

    AUTHOR CONTRIBUTIONS

    H-XT, SK, andAWdesigned the study. H-XT, RE, HV, JJ, andAW Q9

    performed the experiments. H-XT, RE, JJ, SK, and AW wrote themanuscript. All authors read and revised the manuscript.

    Frontiers in Immunology | www.frontiersin.org 10 March 2019 | Volume 10 | Article 611

    https://www.frontiersin.org/journals/immunologyhttps://www.frontiersin.orghttps://www.frontiersin.org/journals/immunology#articles

  • 1141

    1142

    1143

    1144

    1145

    1146

    1147

    1148

    1149

    1150

    1151

    1152

    1153

    1154

    1155

    1156

    1157

    1158

    1159

    1160

    1161

    1162

    1163

    1164

    1165

    1166

    1167

    1168

    1169

    1170

    1171

    1172

    1173

    1174

    1175

    1176

    1177

    1178

    1179

    1180

    1181

    1182

    1183

    1184

    1185

    1186

    1187

    1188

    1189

    1190

    1191

    1192

    1193

    1194

    1195

    1196

    1197

    1198

    1199

    1200

    1201

    1202

    1203

    1204

    1205

    1206

    1207

    1208

    1209

    1210

    1211

    1212

    1213

    1214

    1215

    1216

    1217

    1218

    1219

    1220

    1221

    1222

    1223

    1224

    1225

    1226

    1227

    1228

    1229

    1230

    1231

    1232

    1233

    1234

    1235

    1236

    1237

    1238

    1239

    1240

    1241

    1242

    1243

    1244

    1245

    1246

    1247

    1248

    1249

    1250

    1251

    1252

    1253

    1254

    Tan et al. B Cell Characterization in iBALT

    ACKNOWLEDGMENTS

    This study was supported by an NHMRC program grantQ6 ID1052979 (SK) and NHMRC project grant ID1129099 (AW).

    JJ and SK are supported by NHMRC fellowships.

    SUPPLEMENTARY MATERIAL

    The Supplementary Material for this article can be foundonline at: https://www.frontiersin.org/articles/10.3389/fimmu.2019.00611/full#supplementary-material Q12

    Q8

    REFERENCES

    1. Rennert PD, Browning JL, Mebius R, Mackay F, Hochman PS. Surfacelymphotoxin alpha/beta complex is required for the developmentof peripheral lymphoid organs. J Exp Med. (1996) 184:1999–2006.doi: 10.1084/jem.184.5.1999

    2. Kendall PL, Yu G, Woodward EJ, Thomas JW. Tertiary lymphoid structuresin the pancreas promote selection of B lymphocytes in autoimmune diabetes.J Immunol. (2007) 178:5643–51. doi: 10.4049/jimmunol.178.9.5643

    3. Shomer NH, Fox JG, Juedes AE, Ruddle NH. Helicobacter-induced chronicactive lymphoid aggregates have characteristics of tertiary lymphoid tissue.Infect Immun. (2003) 71:3572–7. doi: 10.1128/IAI.71.6.3572-3577.2003

    4. Astorri E, Scrivo R, Bombardieri M, Picarelli G, Pecorella I, Porzia A,et al. CX3CL1 and CX3CR1 expression in tertiary lymphoid structuresin salivary gland infiltrates: fractalkine contribution to lymphoidneogenesis in Sjögren’s syndrome. Rheumatology. (2014) 53:611–20.doi: 10.1093/rheumatology/ket401

    5. Marinkovic T, Garin A, Yokota Y, Fu YX, Ruddle NH, Furtado GC, et al.Interaction of mature CD3+CD4+ T cells with dendritic cells triggers thedevelopment of tertiary lymphoid structures in the thyroid. J Clin Invest.(2006) 116:2622–32. doi: 10.1172/JCI28993

    6. Kahnert A, Höpken UE, Stein M, Bandermann S, Lipp M, Kaufmann SHE.Mycobacterium tuberculosis triggers formation of lymphoid structure inmurine lungs. J Infect Dis. (2007) 195:46–54. doi: 10.1086/508894

    7. Fleige H, Ravens S, Moschovakis GL, Bölter J, Willenzon S, Sutter G, et al.IL-17–induced CXCL12 recruits B cells and induces follicle formation inBALT in the absence of differentiated FDCs. J Exp Med. (2014) 211:643–51.doi: 10.1084/jem.20131737

    8. Moyron-Quiroz JE, Rangel-Moreno J, Kusser K, Hartson L, SpragueF, Goodrich S, et al. Role of inducible bronchus associated lymphoidtissue (iBALT) in respiratory immunity. Nat Med. (2004) 10:927–34.doi: 10.1038/nm1091

    9. Moyron-Quiroz JE, Rangel-Moreno J, Hartson L, Kusser K, Tighe MP,Klonowski KD, et al. Persistence and responsiveness of immunologic memoryin the absence of secondary lymphoid organs. Immunity. (2006) 25:643–54.doi: 10.1016/j.immuni.2006.08.022

    10. Onodera T, Takahashi Y, Yokoi Y, Ato M, Kodama Y, Hachimura S, et al.Memory B cells in the lung participate in protective humoral immuneresponses to pulmonary influenza virus reinfection. Proc Natl Acad Sci USA.(2012) 109:2485–90. doi: 10.1073/pnas.1115369109

    11. Hiramatsu K, Azuma A, Kudoh S, Desaki M, Takizawa H, Sugawara I.Inhalation of diesel exhaust for threemonths affectsmajor cytokine expressionand induces bronchus-associated lymphoid tissue formation in murine lungs.Exp Lung Res. (2003) 29:607–22. doi: 10.1080/01902140390240140

    12. van der Strate BWA, Postma DS, Brandsma CA, Melgert BN, Luinge MA,Geerlings M, et al. Cigarette smoke–induced emphysema. Am J Respir CritCare Med. (2006) 173:751–8. doi: 10.1164/rccm.200504-594OC

    13. Van DV, Beier KC, Pietzke LJ, Baz MSA, Feist RK, Gurka S, et al. Local T/Bcooperation in inflamed tissues is supported by T follicular helper-like cells.Nat Commun. (2016) 7:10875. doi: 10.1038/ncomms10875

    14. Rangel-Moreno J, Carragher DM, de la Luz Garcia-Hernandez M, HwangJY, Kusser K, Hartson L, et al. The development of inducible bronchus-associated lymphoid tissue depends on IL-17.Nat Immunol. (2011) 12:639–46.doi: 10.1038/ni.2053

    15. Elliot JG, Jensen CM, Mutavdzic S, Lamb JP, Carroll NG, James AL.Aggregations of lymphoid cells in the airways of nonsmokers, smokers,and subjects with asthma. Am J Respir Crit Care Med. (2004) 169:712–8.doi: 10.1164/rccm.200308-1167OC

    16. Hwang JY, Randall TD, Silva-Sanchez A. Inducible bronchus-associatedlymphoid tissue: taming inflammation in the lung. Front Immunol. (2016)7:258. doi: 10.3389/fimmu.2016.00258

    17. Foo SY, Zhang V, Lalwani A, Lynch JP, Zhuang A, Lam CE,et al. Regulatory T cells prevent inducible BALT formation bydampening neutrophilic inflammation. J Immunol. (2015) 194:4567–76.doi: 10.4049/jimmunol.1400909

    18. Sutton CE, Lalor SJ, Sweeney CM, Brereton CF, Lavelle EC, Mills KHG.Interleukin-1 and IL-23 induce innate IL-17 production from γδ T cells,amplifying Th17 responses and autoimmunity. Immunity. (2009) 31:331–41.doi: 10.1016/j.immuni.2009.08.001

    19. Takatori H, Kanno Y, Watford WT, Tato CM, Weiss G, Ivanov II, et al.Lymphoid tissue inducer–like cells are an innate source of IL-17 and IL-22.J Exp Med. (2009) 206:35–41. doi: 10.1084/jem.20072713

    20. Onder L, Narang P, Scandella E, Chai Q, Iolyeva M, Hoorweg K, et al. IL-7-producing stromal cells are critical for lymph node remodeling. Blood. (2012)120:4675–83. doi: 10.1182/blood-2012-03-416859

    21. Ngo VN, Korner H, Gunn MD, Schmidt KN, Riminton DS, Cooper MD,et al. Lymphotoxin α/β and tumor necrosis factor are required for stromalcell expression of homing chemokines in B and T cell areas of the spleen. JExp Med. (1999) 189:403–12. doi: 10.1084/jem.189.2.403

    22. Gonzalez M, Mackay F, Browning JL, Kosco-Vilbois MH, Noelle RJ. Thesequential role of lymphotoxin and B cells in the development of splenicfollicles. J Exp Med. (1998) 187:997–1007. doi: 10.1084/jem.187.7.997

    23. Halle S, Dujardin HC, Bakocevic N, Fleige H, Danzer H, Willenzon S, et al.Induced bronchus-associated lymphoid tissue serves as a general primingsite for T cells and is maintained by dendritic cells. J Exp Med. (2009)206:2593–601. doi: 10.1084/jem.20091472

    24. Whittle JRR, Wheatley AK, Wu L, Lingwood D, Kanekiyo M, Ma SS, et al.Flow cytometry reveals that H5N1 vaccination elicits cross-reactive stem-directed antibodies from multiple Ig heavy-chain lineages. J Virol. (2014)88:4047–57. doi: 10.1128/JVI.03422-13

    25. Afgan E, Sloggett C, Goonasekera N, Makunin I, Benson D, Crowe M, et al.Genomics virtual laboratory: a practical bioinformatics workbench for thecloud. PLoS ONE. (2015) 10:e0140829. doi: 10.1371/journal.pone.0140829

    26. Kim D, Langmead B, Salzberg SL. HISAT: a fast spliced alignerwith low memory requirements. Nat Methods. (2015) 12:357–60.doi: 10.1038/nmeth.3317

    27. Anders S, Pyl PT, Huber W. HTSeq—a Python framework to workwith high-throughput sequencing data. Bioinformatics. (2015) 31:166–9.doi: 10.1093/bioinformatics/btu638

    28. von Boehmer L, Liu C, Ackerman S, Gitlin AD, Wang Q, Gazumyan A, et al.Sequencing and cloning of antigen-specific antibodies frommouse memory Bcells. Nat Protoc. (2016) 11:1908–23. doi: 10.1038/nprot.2016.102

    29. Alamyar E, Duroux P, Lefranc MP, Giudicelli V. “IMGT R⃝ tools forthe nucleotide analysis of immunoglobulin (IG) and T cell receptor(TR) V-(D)-J repertoires, polymorphisms, and IG mutations: IMGT/V-QUEST and IMGT/HighV-QUEST for NGS,” In: Christiansen FT, TaitBD, editors. Immunogenetics: Methods and Applications in Clinical PracticeMethods in Molecular Biology (Totowa, NJ: Humana Press). p. 569–604.doi: 10.1007/978-1-61779-842-9_32

    30. Gu Z, Gu L, Eils R, Schlesner M, Brors B. Circlize implements andenhances circular visualization in R. Bioinformatics. (2014) 30:2811–2.doi: 10.1093/bioinformatics/btu393

    31. Joo HM, He Y, Sangster MY. Broad dispersion and lung localizationof virus-specific memory B cells induced by influenza pneumonia.Proc Natl Acad Sci USA. (2008) 105:3485–90. doi: 10.1073/pnas.0800003105

    Frontiers in Immunology | www.frontiersin.org 11 March 2019 | Volume 10 | Article 611

    https://www.frontiersin.org/articles/10.3389/fimmu.2019.00611/full#supplementary-materialhttps://doi.org/10.1084/jem.184.5.1999https://doi.org/10.4049/jimmunol.178.9.5643https://doi.org/10.1128/IAI.71.6.3572-3577.2003https://doi.org/10.1093/rheumatology/ket401https://doi.org/10.1172/JCI28993https://doi.org/10.1086/508894https://doi.org/10.1084/jem.20131737https://doi.org/10.1038/nm1091https://doi.org/10.1016/j.immuni.2006.08.022https://doi.org/10.1073/pnas.1115369109https://doi.org/10.1080/01902140390240140https://doi.org/10.1164/rccm.200504-594OChttps://doi.org/10.1038/ncomms10875https://doi.org/10.1038/ni.2053https://doi.org/10.1164/rccm.200308-1167OChttps://doi.org/10.3389/fimmu.2016.00258https://doi.org/10.4049/jimmunol.1400909https://doi.org/10.1016/j.immuni.2009.08.001https://doi.org/10.1084/jem.20072713https://doi.org/10.1182/blood-2012-03-416859https://doi.org/10.1084/jem.189.2.403https://doi.org/10.1084/jem.187.7.997https://doi.org/10.1084/jem.20091472https://doi.org/10.1128/JVI.03422-13https://doi.org/10.1371/journal.pone.0140829https://doi.org/10.1038/nmeth.3317https://doi.org/10.1093/bioinformatics/btu638https://doi.org/10.1038/nprot.2016.102https://doi.org/10.1007/978-1-61779-842-9_32https://doi.org/10.1093/bioinformatics/btu393https://doi.org/10.1073/pnas.0800003105https://www.frontiersin.org/journals/immunologyhttps://www.frontiersin.orghttps://www.frontiersin.org/journals/immunology#articles

  • 1255

    1256

    1257

    1258

    1259

    1260

    1261

    1262

    1263

    1264

    1265

    1266

    1267

    1268

    1269

    1270

    1271

    1272

    1273

    1274

    1275

    1276

    1277

    1278

    1279

    1280

    1281

    1282

    1283

    1284

    1285

    1286

    1287

    1288

    1289

    1290

    1291

    1292

    1293

    1294

    1295

    1296

    1297

    1298

    1299

    1300

    1301

    1302

    1303

    1304

    1305

    1306

    1307

    1308

    1309

    1310

    1311

    1312

    1313

    1314

    1315

    1316

    1317

    1318

    1319

    1320

    1321

    1322

    1323

    1324

    1325

    1326

    1327

    1328

    1329

    1330

    1331

    1332

    1333

    1334

    1335

    1336

    1337

    1338

    1339

    1340

    1341

    1342

    1343

    1344

    1345

    1346

    1347

    1348

    1349

    1350

    1351

    1352

    1353

    1354

    1355

    1356

    1357

    1358

    1359

    1360

    1361

    1362

    1363

    1364

    1365

    1366

    1367

    1368

    Tan et al. B Cell Characterization in iBALT

    32. Boyden AW, Legge KL, Waldschmidt TJ. Pulmonary infection with influenzaA virus induces site-specific germinal center and T follicular helper cellresponses. PLoS ONE. (2012) 7:e40733. doi: 10.1371/journal.pone.0040733

    33. Tan HX, Jegaskanda S, Juno JA, Esterbauer R, Wong J, Kelly HG,et al. Subdominance and poor intrinsic immunogenicity limit humoralimmunity targeting influenza HA-stem. J Clin Invest. (2018) 129:850–62.doi: 10.1172/JCI123366

    34. Naito Y, Takematsu H, Koyama S, Miyake S, Yamamoto H, Fujinawa R,et al. Germinal center marker GL7 probes activation-dependent repressionof N-glycolylneuraminic acid, a sialic acid species involved in thenegative modulation of B-cell activation. Mol Cell Biol. (2007) 27:3008–22.doi: 10.1128/MCB.02047-06

    35. Victora GD, Dominguez-Sola D, Holmes AB, Deroubaix S, Dalla-Favera R,Nussenzweig MC. Identification of human germinal center light and darkzone cells and their relationship to human B-cell lymphomas. Blood. (2012)120:2240–8. doi: 10.1182/blood-2012-03-415380

    36. Victora GD, Nussenzweig MC. Germinal centers. Annu Rev Immunol. (2012)30:429–57. doi: 10.1146/annurev-immunol-020711-075032

    37. Yu D, Rao S, Tsai LM, Lee SK, He Y, Sutcliffe EL, et al. The transcriptionalrepressor Bcl-6 directs T follicular helper cell lineage commitment. Immunity.(2009) 31:457–68. doi: 10.1016/j.immuni.2009.07.002

    38. Johnston RJ, Poholek AC, DiToro D, Yusuf I, Eto D, Barnett B, et al. Bcl6 andBlimp-1 are reciprocal and antagonistic regulators of T follicular helper celldifferentiation. Science. (2009) 325:1006–10. doi: 10.1126/science.1175870

    39. Adachi Y, Onodera T, Yamada Y, Daio R, Tsuiji M, Inoue T, et al. Distinctgerminal center selection at local sites shapes memory B cell response to viralescape. J Exp Med. (2015) 212:1709–23. doi: 10.1084/jem.20142284

    40. Hutloff A. T follicular helper-like cells in inflamed non-lymphoid tissues.Front Immunol. (2018) 9:1707. doi: 10.3389/fimmu.2018.01707

    41. GeurtsvanKessel CH, Willart MAM, Bergen IM, van Rijt LS, Muskens F,Elewaut D, et al. Dendritic cells are crucial for maintenance of tertiarylymphoid structures in the lung of influenza virus–infected mice. J Exp Med.(2009) 206:2339–49. doi: 10.1084/jem.20090410

    42. Victora GD, Schwickert TA, Fooksman DR, Kamphorst AO, Meyer-Hermann M, Dustin ML, et al. Germinal center dynamics revealed bymultiphoton microscopy with a photoactivatable fluorescent reporter. Cell.(2010) 143:592–605. doi: 10.1016/j.cell.2010.10.032

    43. Allen CDC, Okada T, Cyster JG. Germinal-center organization and cellulardynamics. Immunity. (2007) 27:190–202. doi: 10.1016/j.immuni.2007.07.009

    44. Bannard O, Horton RM, Allen CDC, An J, Nagasawa T, Cyster JG. Germinalcenter centroblasts transition to a centrocyte phenotype according to a timedprogram and depend on the dark zone for effective selection. Immunity.(2013) 39:912–24. doi: 10.1016/j.immuni.2013.08.038

    45. Coudeville L, Bailleux F, Riche B, Megas F, Andre P, Ecochard R. Relationshipbetween haemagglutination-inhibiting antibody titres and clinical protectionagainst influenza: development and application of a bayesian random-effects model. BMC Med Res Methodol. (2010) 10:18. doi: 10.1186/1471-2288-10-18

    46. Vanderven HA, Jegaskanda S, Wheatley AK, Kent SJ. Antibody-dependentcellular cytotoxicity and influenza virus. Curr Opin Virol. (2017) 22:89–96.doi: 10.1016/j.coviro.2016.12.002

    47. LaMere MW, Moquin A, Lee FEH, Misra RS, Blair PJ, Haynes L,et al. Regulation of antinucleoprotein IgG by systemic vaccination andits effect on influenza virus clearance. J Virol. (2011) 85:5027–35.doi: 10.1128/JVI.00150-11

    48. LaMere MW, Lam HT, Moquin A, Haynes L, Lund FE, RandallTD, et al. Contributions of antinucleoprotein IgG to heterosubtypicimmunity against influenza virus. J Immunol. (2011) 186:4331–9.doi: 10.4049/jimmunol.1003057

    49. Fujimoto Y, Tomioka Y, Takakuwa H, Uechi GI, Yabuta T, Ozaki K,et al. Cross-protective potential of anti-nucleoprotein human monoclonalantibodies against lethal influenza A virus infection. J Gen Virol. (2016)97:2104–16. doi: 10.1099/jgv.0.000518

    50. Carragher DM, Kaminski DA, Moquin A, Hartson L, Randall TD.A novel role for non-neutralizing antibodies against nucleoprotein infacilitating resistance to influenza virus. J Immunol. (2008) 181:4168–76.doi: 10.4049/jimmunol.181.6.4168

    51. Kuraoka M, Schmidt AG, Nojima T, Feng F, Watanabe A, Kitamura D,et al. Complex antigens drive permissive clonal selection in germinalcenters. Immunity. (2016) 44:542–52. doi: 10.1016/j.immuni.2016.02.010

    52. Angeletti D, Gibbs JS, Angel M, Kosik I, Hickman HD, Frank GM,et al. Defining B cell immunodominance to viruses. Nat Immunol. (2017)18:456–63. doi: 10.1038/ni.3680

    Conflict of Interest Statement: The authors declare that the research wasconducted in the absence of any commercial or financial relationships that couldbe construed as a potential conflict of interest.

    Copyright © 2019 Tan, Esterbauer, Vanderven, Juno, Kent and Wheatley. This is anopen-access article distributed under the terms of the Creative Commons AttributionLicense (CC BY). The use, distribution or reproduction in other forums is permitted,provided the original author(s) and the copyright owner(s) are credited and that theoriginal publication in this journal is cited, in accordance with accepted academicpractice. No use, distribution or reproduction is permitted which does not complywith these terms.

    Frontiers in Immunology | www.frontiersin.org 12 March 2019 | Volume 10 | Article 611

    https://doi.org/10.1371/journal.pone.0040733https://doi.org/10.1172/JCI123366https://doi.org/10.1128/MCB.02047-06https://doi.org/10.1182/blood-2012-03-415380https://doi.org/10.1146/annurev-immunol-020711-075032https://doi.org/10.1016/j.immuni.2009.07.002https://doi.org/10.1126/science.1175870https://doi.org/10.1084/jem.20142284https://doi.org/10.3389/fimmu.2018.01707https://doi.org/10.1084/jem.20090410https://doi.org/10.1016/j.cell.2010.10.032https://doi.org/10.1016/j.immuni.2007.07.009https://doi.org/10.1016/j.immuni.2013.08.038https://doi.org/10.1186/1471-2288-10-18https://doi.org/10.1016/j.coviro.2016.12.002https://doi.org/10.1128/JVI.00150-11https://doi.org/10.4049/jimmunol.1003057https://doi.org/10.1099/jgv.0.000518https://doi.org/10.4049/jimmunol.181.6.4168https://doi.org/10.1016/j.immuni.2016.02.010https://doi.org/10.1038/ni.3680http://creativecommons.org/licenses/by/4.0/http://creativecommons.org/licenses/by/4.0/http://creativecommons.org/licenses/by/4.0/http://creativecommons.org/licenses/by/4.0/http://creativecommons.org/licenses/by/4.0/https://www.frontiersin.org/journals/immunologyhttps://www.frontiersin.orghttps://www.frontiersin.org/journals/immunology#articles

    Inducible Bronchus-Associated Lymphoid Tissues (iBALT) Serve as Sites of B Cell Selection and Maturation Following Influenza Infection in MiceIntroductionMaterials and MethodsAnimal InfectionConfocal MicroscopyHA ProteinsFlow CytometryRNAseq and Data AnalysisSequencing and Analysis of Murine B Cell Receptor GenesStatistics

    ResultsDynamics of iBALT Induction Following Intranasal Influenza InfectionTranscriptional Profile of GC-Like B Cells From iBALTiBALT-Resident Tfh Cells Express Bcl6 and Ki67Specificity of GC-Resident B Cells for Major Influenza AntigensGC Within iBALT Support Maturation of Influenza-Specific B Cells

    DiscussionData AvailabilityAuthor ContributionsAcknowledgmentsSupplementary MaterialReferences