38
1 Local blockade of epithelial PDL-1 in the airways enhances T cell function and 1 viral clearance during influenza virus infection 2 3 Beth McNally 1,* , Fang Ye 1 , Meredith Willette 1 , and Emilio Flaño 1,2 4 5 1 Center for Vaccines and Immunity, The Research Institute at Nationwide Children’s 6 Hospital, Columbus, OH 43205 7 2 The Ohio State University College of Medicine, Columbus, OH 43210 8 9 * Corresponding author. Mailing address: Center for Vaccines and Immunity, The 10 Research Institute at Nationwide Children’s Hospital, 700 Children’s Dr., Columbus, OH 11 43205. [email protected] 12 13 Running title: PD-1 blockade in the airways 14 15 Abbreviations: bronchoalveolar lavage (BAL), Human airway epithelial (HAE), 16 interferon-α receptor (IFNAR), mediastinal lymph node (MLN), programmed death 17 receptor-1 (PD-1), programmed death receptor 1-ligand (PDL-1), tracheal epithelial cell 18 (TEC) 19 Word count abstract: 205 20 Word count text: 4,538 21 22 JVI Accepts, published online ahead of print on 25 September 2013 J. Virol. doi:10.1128/JVI.02423-13 Copyright © 2013, American Society for Microbiology. All Rights Reserved. on April 4, 2019 by guest http://jvi.asm.org/ Downloaded from

Local blockade of epithelial PDL-1 in the airways enhances T cell

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

1

Local blockade of epithelial PDL-1 in the airways enhances T cell function and 1

viral clearance during influenza virus infection 2

3

Beth McNally1,*, Fang Ye1, Meredith Willette1, and Emilio Flaño1,2 4

5

1 Center for Vaccines and Immunity, The Research Institute at Nationwide Children’s 6

Hospital, Columbus, OH 43205 7

2 The Ohio State University College of Medicine, Columbus, OH 43210 8

9

* Corresponding author. Mailing address: Center for Vaccines and Immunity, The 10

Research Institute at Nationwide Children’s Hospital, 700 Children’s Dr., Columbus, OH 11

43205. [email protected] 12

13

Running title: PD-1 blockade in the airways 14

15

Abbreviations: bronchoalveolar lavage (BAL), Human airway epithelial (HAE), 16

interferon-α receptor (IFNAR), mediastinal lymph node (MLN), programmed death 17

receptor-1 (PD-1), programmed death receptor 1-ligand (PDL-1), tracheal epithelial cell 18

(TEC) 19

Word count abstract: 205 20

Word count text: 4,538 21

22

JVI Accepts, published online ahead of print on 25 September 2013J. Virol. doi:10.1128/JVI.02423-13Copyright © 2013, American Society for Microbiology. All Rights Reserved.

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from

2

ABSTRACT 23

In order to maintain the gas exchange function of the lung following influenza virus 24

infection, a delicate orchestration of positive and negative regulatory pathways must be 25

maintained to attain viral eradication while minimizing local inflammation. The 26

programmed death receptor 1-ligand /programmed death receptor-1 (PDL-1/PD-1) 27

pathway plays an important immunoregulatory role particularly in the context of T cell 28

function. Here, we show that influenza virus infection of primary airway epithelial cells 29

strongly enhances PDL-1 expression, and does so in an interferon-α receptor (IFNAR) 30

signaling dependent manner. PD-1 is primarily expressed on effector T cells in the lung 31

compared to effector memory and central memory cells, and shortly after influenza virus 32

infection an increased number of PD-1+ T cells are recruited to the airways. Using in 33

vitro co-cultures of airway epithelial cells and T cells, and in vivo models of influenza 34

virus infection, we demonstrate that blockade of airway epithelial PDL-1 improves CD8 35

T cell function defined by increased production of IFNγ, granzyme B, and expression of 36

CD107ab. Furthermore, PDL-1 blockade in the airways served to accelerate influenza 37

virus clearance and enhance infection recovery. Our findings suggest that local 38

manipulation of the PDL-1/PD-1 axis in the airways may represent a therapeutic 39

alternative during acute influenza virus infection. 40

41

42

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from

3

INTRODUCTION 43

The respiratory tract is constantly exposed to a broad range of foreign antigens due to 44

respiration. The airway epithelium lining the respiratory tract not only represents the first 45

barrier of defense against respiratory pathogens it is often the primary target of infection 46

(1). Consequently, apart from a critical role in gas exchange function, airway epithelial 47

cells additionally participate in host defense mechanisms by producing cytokines and 48

chemokines (2-8). As exacerbated responses by airway epithelial cells in response to 49

innocuous antigens or pathogens can prove detrimental to lung function, airway 50

epithelial cells have also evolved to regulate pulmonary homeostasis by multiple 51

mechanisms including, among others, expression of CD200 (9), MUC-1 (10), surfactant 52

proteins (11), and reduced expression of adaptor molecules (12-14). 53

Interaction of programmed cell death-ligand 1 (PDL-1) with its receptor, programmed 54

death 1 (PD-1), a member of the B7 family of signaling molecules, has been specifically 55

linked to negative regulation of T cell immune responses (15, 16). Upon activation, PD-1 56

inhibits T cell receptor signaling by blocking PI3k/Akt activation (17). During chronic viral 57

infection and cancer, PD-1 expression leads to T cell dysfunction and exhaustion (15, 58

16, 18). Conversely, blockade of the PD-1/PDL-1 axis has been shown to either 59

enhance T cell function or reduce viral burden in a number of chronic viral infections 60

(18-21). Our understanding of the role of the PD-1/PDL-1 pathway during acute viral 61

infection, however, is just beginning to unfold. PD-1 expression is rapidly down-62

regulated during acute LCMV infection (22) but is expressed on functional CD8 T cells 63

in other acute viral infections (23, 24). PD-1 has also been shown to regulate the 64

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from

4

development of T cell memory during vaccinia virus infection (25). Fairly recently, it has 65

been shown that a number of acute respiratory viral infections including human 66

metapneumovirus, respiratory syncytial virus and influenza virus impair CD8 T cell 67

function through PD-1 (26)(27). Although previous studies have shown that lack of PD-68

1 function due to genetic mutation or to systemic blockade prevented CD8 T cell 69

inhibition and enhanced viral control, the specific role that the airway epithelium plays 70

during PDL-1/PD-1 regulation of pulmonary T cell function has not been fully elucidated. 71

Our study builds on the observations by Erickson et al. (27) to further show that PDL-1 72

is constitutively expressed in airway epithelial cells, and its expression is up-regulated 73

by influenza virus and dependent on interferon-α receptor (IFNAR) signaling. Co-culture 74

studies with airway epithelial cells and T cells in the presence of PDL-1 blocking 75

antibody revealed that PDL-1 expression on airway epithelial cells repressed T cell 76

function. Additionally, intranasal blockade of PDL-1 in influenza virus infected mice 77

resulted in increased levels of cytotoxic proteins and cytokines in the airways; enhanced 78

CD8 T cell function; and reduced viral load. Our findings suggest that PDL-1 expression 79

by airway epithelial cells during the early phase of the recall response to influenza virus 80

infection represents a mechanism for regulating lung immune homeostasis, and reveal 81

a promising strategy to modulate local epithelial-T cell interaction in the airways. 82

83

MATERIALS AND METHODS 84

85

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from

5

Mice. C57BL/6J, Balb/c, and IFNAR−/− mice (Balb/c background) were obtained from 86

The Jackson Laboratory (Bar Harbor, ME) or Harlan (Indianapolis, IN), or were bred at 87

The Research Institute at Nationwide Children’s Hospital. Mice were housed in BL2 88

containment under pathogen-free conditions. The Institutional Animal Care and Use 89

Committee at The Research Institute at Nationwide Children’s Hospital approved all of 90

the animal studies described in this study. 91

Cell culture. Primary mouse tracheal epithelial cell (TEC) cultures were obtained from 92

tracheas resected from 3-10 week old wild-type Balb/c and IFNAR-/- mice. Tracheal cell 93

isolation and culture was performed as previously described (28). In short, tracheas 94

were collected and incubated with 1.5 mg/ml pronase and 0.5 mg/ml crude pancreatic 95

DNase I. Non-adherent cells were collected and seeded onto 24 mm coated 96

membranes with 50 µg/ml type I rat tail collagen solution. Cells were incubated for 10-14 97

days post-harvest then allowed to differentiate by incubating under air-liquid interface 98

for an additional 14 days before use. 99

Virus infection. Influenza A viruses Udorn, WSN, PR8 and X31 were grown in eggs, 100

and virus titers were determined by an immunofluorescence foci assay. Male mice (6-101

12wks of age) were anesthetized with 2,2,2,-tribromoethanol and intranasally inoculated 102

with 3000 EID50 influenza virus X31 and challenged with 1x102 PFU/mouse PR8, in a 30 103

µl volume. Organs were harvested on the indicated days post treatment. Groups of 104

three to six animals were used for each data point. For the analysis of PDL-1 transcript, 105

TEC cultures derived from Balb/c and IFNAR-/- mice were exposed to 2x105 WSN for 106

2h or mock inoculated and harvested 24h post infection. Human airway epithelial (HAE) 107

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from

6

cultures were exposed to 2x105 influenza A Udorn strain, or mock treated for 2 hours 108

and harvested after 24 hours of incubation as indicated (28). 109

Immunofluorescence. Influenza infected HAE cultures maintained on Snapwell inserts 110

were fixed in 10% paraformaldehyde then washed and stored in 70% ethanol until 111

paraffin embedded. Six-micrometer sections of HAE the cultures or human trachea 112

were deparaffinized for immunofluorescence. Following 30’ antigen retrieval in sodium 113

citrate (pH 6), tissues were incubated in 0.2% Triton X-100 for 15’ and blocked with 5% 114

goat serum for 1 hour. PDL-1 was visualized using rabbit polyclonal to CD274, (Abcam, 115

Cambridge, MA) and Texas-red goat anti rabbit (Vector Laboratories, Burlingame, CA). 116

Control sections were stained with rabbit immunoglobulin or with secondary antibody 117

only. Tissues were mounted with Prolong Gold anti-fade reagent containing DAPI 118

(Invitrogen/Molecular Probes). Immunofluorescent images were viewed on an Olympus 119

BX61 using a 40x objective and captured using a Hamamatsu ORCA-GR digital camera 120

in concert with Slidebook version 4.2 software (Intelligent Imaging Innovations, Inc., 121

Denver, CO). 122

RNA extraction, reverse transcription, and real-time PCR. RNA isolation from 123

infected HAE cultures and TEC cultures was achieved by direct lysis of cells with TRIzol 124

(Invitrogen) and application of the resulting aqueous phase to Qiagen RNeasy Mini 125

Columns. Total RNA harvested from triplicate (TEC) or quadruplicate (HAE) cultures 126

was processed and analyzed as described in (28). 1 たg RNA extracted from infected 127

lungs or cell cultures using a combination of Trizol (Invitrogen) and/or the RNeasy Mini 128

Kit, (Qiagen), was also used for cDNA synthesis (High capacity RT Kit, Applied 129

Biosytems) followed by quantitative PCR performed using Cyber Green (Applied 130

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from

7

Biosytems) and primers for human or mouse PDL-1, the Flu NP gene and human or 131

mouse GAPDH. Data were analyzed using Prism 7500 SDS software. Mock infected 132

groups served as the calibrator sample and all samples were normalized to the 133

endogenous control, GAPDH. 134

TEC/T lymphocyte co-cultures. TEC cultures derived from Balb/c and IFNAR-/- mice 135

were co-cultured with splenic T lymphocytes at a 1:1 ratio (TECs:T cells). T 136

lymphocytes were isolated from spleens of naïve Balb/c mice by negative selection. 137

TECs seeded in 24-well plates were exposed to 1 PFU/cell influenza virus WSN for 2 138

hours. Fresh media was added after the removal of inoculum, and panned T 139

lymphocytes (purity, >95%) were added 24h post infection. Blocking antibodies to PDL-140

1 (clone MIH5, eBioscience) or control antibody (ChromePure Rat IgG, Jackson 141

Immuno Research) were added 3 hrs before the addition of T lymphocytes such that 10 142

たg blocking antibody was added to 2.5�×�105 TECs. 143

In vivo antibody blockade. Mice initially infected with influenza virus X31 were 144

challenged 30-40 days later with influenza virus PR8 as described above, and either 145

intraperitoneally injected with 200 ug of functional grade anti-mouse PDL-1, 200 ug IgG 146

or HBSS; or were intranasally treated with 150 ug anti-PDL-1 or control antibody 147

administered successively over a three day time period in 50 ug (50 ul) doses. Sham 148

mice received the same volume of HBSS in lieu of antibody. 149

Plaque assay. Lung homogenates collected in 0.5 ml DMEM-0.1% were used for viral 150

titer determination. Plaque assays were performed by exposing 80-90% confluent 151

MDCK cells grown in 12-well plates to 100 µl quantities of 10-fold serial dilutions of lung 152

homogenates for 2 hours at 37°C. The inoculated monolayers were covered with DMEM 153

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from

8

containing 0.8% methylcellulose, L-glutamine, 0.1% BSA, antibiotics and 1 たg/ml TPCK 154

trypsin (Sigma, St. Louis, MO) and incubated for 3 days at 37°C. After incubation, the 155

monolayers were permeablized with 0.2% Triton X-100. Virus infected cells were 156

immuno-stained using a monoclonal antibody that reacts with the influenza A 157

nucleoprotein (Cal Bioreagents, San Mateo, CA) followed by a 1 hr incubation with anti-158

mouse IgG peroxidase-congjugate (Sigma, St. Louis, MO) and a 10-30 min incubation 159

with True Blue Substrate (KPL, Gathersburg, MD). Quantification was performed by 160

counting plaques using a dissecting microscope and counts were expressed as log10 161

PFU per milliliter. 162

Flow cytometry 163

Epithelial cells. TECs were trypsinized to obtain a cell suspension then stained and 164

fixed with paraformaldehyde. Analysis of cell surface expression of PDL-1 on TECs was 165

achieved using PE-Rat anti-mouse CD274 (BD Pharmingen, Franklin Lakes, NJ) and 166

the BD LSRII flow cytometer with FlowJo software (TreeStar Inc., Ashland, OR). Results 167

were expressed as median fluorescence intensity (MFI) after subtracting the MFI of 168

control cells stained with the appropriate isotype control antibodies (BD Pharmingen). 169

T lymphocytes. Single-cell suspensions were obtained from the bronchoalveolar lavage, 170

lung, MLN and spleen. RBCs were lysed, and the number of cells per organ was 171

determined. Cells were blocked with Fc-block (CD16/32) and washed and stained with a 172

combination of NP366–374/Db tetramer and antibodies against CD3, CD4, CD8, CD44, 173

CD62L, CD43-activation associated glycoform, CD107ab, and PD-1 or combined with 174

intracellular staining for cytokines/cytotoxic proteins: IFN-け, IL-2, perforin and granzyme 175

B (eBioscience). Flow cytometry data were acquired on a BD LSR II (BD Biosciences) 176

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from

9

and analyzed using FlowJo software. Gates were set using negative controls and 177

isotype controls. 178

ELISA for IFN-γ and granzyme B. Concentrations of gamma interferon (IFN-γ) and 179

Granzyme B, were measured in BAL sample supernatants and co-cultutres with 180

commercially available ELISA kits (Ready-SET-Go! Kit, eBioscience). All assays were 181

carried out according to the manufacturer's instructions, and 100 µl from each sample 182

was assayed in duplicate. The lower limits of detection for these assays were 15 pg/ml 183

for IFN-け, and 40 pg/mL (Granzyme B). For statistical analysis, samples with optical 184

density readings below the limit of the standard curve of the assay were assigned a 185

value half that of the detection level. 186

Statistical analysis. T tests were used to compare both the percentage of cells positive 187

for a given antigen of interest as well as the mean relative fluorescence intensity (MRFI) 188

determined by flow cytometry analysis. Two sample homoscedastic student t-tests were 189

used for single comparisons in which a P value less than 0.05 was considered 190

statistically significant. Results are representative of at least 2-3 separate experiments 191

using 3-6 mice per group and are expressed as means�±�SD. One-way analysis of 192

variance (ANOVA) was used to determine statistical significance when comparing three 193

or more independent groups (particularly in cases in which antibody treatment was 194

compared to mock, Flu and IgG control treatments). When ANOVA demonstrated that 195

the differences among means were statistically significant (P < 0.05), the Tukey post-196

test was used to correct for multiple comparisons, and to generate multiplicity adjusted 197

P values (Prism GraphPad, version 6). 198

199

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from

10

RESULTS 200

PDL-1 is constitutively expressed in airway epithelial cells, and influenza virus 201

infection and interferon- signaling regulate its expression 202

The epithelial surfaces of the lung play an important role in regulating local immunity 203

and homeostasis. In an initial attempt to analyze the role of regulatory factors critical to 204

infection of the respiratory tract, we established polarized, well-differentiated human 205

primary airway epithelial cell cultures (HAEs) and exposed them to influenza A virus. 206

HEA cultures differentiate to form a pseudostratified mucociliary cell layer and 207

effectively mimic the airway epithelial environment. A comparison of cluster of 208

differentiation molecules following gene expression microarray-based screening 209

revealed that one gene in particular, PDL-1, was drastically upregulated in HAE cultures 210

24 h post exposure to influenza virus (S1). Our analysis showed that PDL-1 expression 211

was enhanced 83.9-fold 24h post exposure to Influenza virus compared to mock-treated 212

HAEs (Figure 1A). qRT-PCR analysis corroborated the above findings showing a 54.3-213

fold increase in PDL-1 expression in HAEs following influenza virus exposure (Figure 214

1B). Additionally, immunoflourescent staining of human trachea sections showed that 215

PDL-1 was not only constitutively expressed in airway epithelial cells, but, perhaps more 216

importantly, that the staining was prominent on the apical surface (Figure 1C). To 217

determine if influenza virus infection affected PDL-1 expression at the protein level, we 218

performed immunofluorescent analysis of PDL-1 expression on HAE cultures infected or 219

not with influenza virus. As we observed in human trachea epithelium, PDL-1 was 220

constitutively expressed in control HAEs (Figure 1D, upper panel). However, when 221

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from

11

compared with mock treated cultures, PDL-1 staining increased in intensity in HAEs 24 222

h post exposure to influenza (Figure 1D, lower panel). Altogether, these data indicate 223

that PDL-1 is constitutively expressed by the human airway epithelium, and that 224

influenza virus infection increases the expression of PDL-1 in airway epithelial cells 225

within 24 hours of infection. 226

To further analyze regulation of PDL-1 expression on airway epithelial cells we 227

generated primary murine tracheal epithelial cell cultures (TECs). As we observed with 228

the HAEs, exposure of TECs to influenza virus resulted in a significant increase (27.6-229

fold) in PDL-1 mRNA expression by 24 h (Figure 2A). As interferon-α receptor (IFNAR) 230

signaling is critical for the anti-viral response of the airway epithelium (28), we also 231

analyzed the role of IFNAR on the regulation of PD-1L expression in airway epithelial 232

cells during the response to influenza virus. The data show that there was a 21.1-fold 233

difference in PDL-1 mRNA expression when comparing influenza virus infected IFNAR-234

/- TECs to wild-type TECs, indicating that PDL-1 mRNA expression is dependent on 235

signaling. We next corroborated these findings at the protein level by FACS analysis of 236

PDL-1 cell surface expression. The data show that influenza virus infection enhanced 237

the cell surface expression of PDL-1 on TECs, but that PDL-1 expression on IFNAR-238

deficient TEC cultures was not different from isotype control staining (Figure 2B-C). 239

Altogether, these data indicate that influenza virus-induced increase of PDL-1 240

expression on epithelial cells is IFNAR signaling dependent. 241

PD-1+ T cells are present in the lung early during influenza virus infection 242

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from

12

In order for PDL-1 to be functional on airway epithelial cells, contact with cells 243

expressing its receptor is necessary. Therefore, we investigated whether or not a 244

significant population of PD-1 expressing cells was available in the respiratory tract. In 245

order to do this, cohorts of mice were intranasally infected with influenza A virus, and 246

cell populations were analyzed in the bronchoalveolar lavage (BAL) and lung 247

parenchyma at different times points after infection. The data shows that PD-1 was 248

expressed in 10-40% of the CD4 and CD8 T cells present in the airways and lung 249

parenchyma of naïve mice, and that there were not significant changes in the frequency 250

of PD-1-expressing T cells during the first week after influenza virus infection (Figures 251

3A-B). The analysis of T cell numbers shows that a small population of PD-1-expressing 252

CD4 and CD8 T cells is present in the lung parenchyma (~1000 cells) while the number 253

of PD-1+ T cells in the airways was very low (10 CD8 T cells, 100 CD4 T cells, Figure 254

3C-D). However, we observed a significant increase in the absolute number of PD-1+ 255

CD4 and CD8 T cells present in the airways by day 3 after influenza virus infection 256

(Figure 3C). Additionally, the number of PD-1+ T cells in the lung parenchyma also 257

increased over time though the differences were not statistically significant (Figure 3D). 258

Although the number of PD1-expressing T cells returned to baseline levels in the lung 259

parenchyma by day 12 post-infection, greater numbers of PD-1+ CD4 and CD8 T cells 260

remained in the airways of influenza infected mice up to one month after infection. To 261

more precisely determine the expression pattern of PD-1 on different populations of 262

influenza virus-specific effector and memory T cells, influenza virus memory mice were 263

intranasally stimulated with poly (I:C) to induce non-specific activation of T cells in the 264

respiratory tract. We found that PD-1 was expressed on effector CD8 T cells but was 265

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from

13

absent from the cell surface of effector-memory and central-memory CD8 T cells as 266

defined by the expression of L-selectin and CD43-activation associated glycoform 267

(Figure 4). This pattern of expression was found on influenza virus NP366–374-specific 268

CD8 T cells as well as NP tetramer-negative CD8 T cells. Altogether, these data 269

indicate that the majority of influenza virus-specific effector CD8 T cells present in the 270

lung express PD-1. 271

PDL-1 expression on airway epithelial cells controls CD8 T cell function 272

To determine if PDL-1 expression on the airway epithelium modulated the function of T 273

cells we used a memory system that allowed us to analyze the function of virus-specific 274

T cells. We incubated T cells isolated from the spleens of day 30-40 influenza virus-275

infected mice with primary, polarized TECs in the presence or absence of PDL-1-276

blocking antibodies. TECs were first exposed to influenza virus for 24 h to induce the 277

up-regulation of PDL-1 on the epithelial cell surface, and then co-cultured with T cells 278

for an additional 36 h. T cell function was assessed by measuring the levels of 279

granzyme B secreted into the culture medium. As shown in Fig. 5, granzyme B was 280

undetectable in uninfected co-cultures regardless of the presence or not of anti-PDL-1 281

antibodies. However, in the presence of influenza virus, granzyme B was released into 282

the culture media, and PDL-1 blockade resulted in significantly higher levels of 283

granzyme B in culture supernatants. These data indicate that PDL-1 expressed on 284

airway epithelial cells can inhibit the cytotoxic activity of T cells through its interaction 285

with PD-1, and that this process can be reversed in vitro by blockade of PDL-1/PD-1 286

interaction. To address the functional role of PDL-1/PD-1 interactions in vivo during 287

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from

14

influenza virus infection, we performed PDL-1 blockade studies and monitored 288

parameters of T cell function in the airways. For these studies, we used influenza virus 289

memory mice to ensure that an elevated number of virus-specific T cells were present in 290

the respiratory tract very early after challenge. The memory mice were intranasally 291

challenged with a heterosubtypic influenza virus (PR8) infection, injected with PDL-1 292

blocking antibody or with isotype control antibody, and the levels of granzyme B and of 293

IFNγ were assayed in BAL fluid 3 days after viral challenge. As shown in Figure 6, 294

intraperitoneal administration of anti-PDL-1 blocking antibody resulted in a significant 295

increase in the concentration of granzyme B and IFNγ in the airways when compared to 296

mice treated with control IgG. 297

Although these in vivo experiments demonstrated that PDL-1/PD-1 interaction regulates 298

the effector functions during influenza virus infection, they did not specifically address 299

the role of the airway epithelium in this process. This is important because the PDL-300

1/PD-1 pathway has been shown to impact the priming of T cell responses (29-34). 301

Thus, in order to elucidate the role of the PDL-1/PD-1 axis in regulating intraepithelial T 302

cell function we administered anti-PDL-1 blocking antibodies intranasally to induce 303

blockade in the airways. Preliminary studies demonstrated that intranasal administration 304

of antibodies had a specific local effect in the airways but not in the lung parenchyma, 305

draining lymph nodes, or spleen (S2). The results show that intranasal administration of 306

anti-PDL-1 blocking antibodies resulted in significantly higher concentrations of 307

granzyme B and IFNγ in the airways 3 days after influenza virus infection (Figure 7 A-B). 308

By day 5 after influenza virus infection the levels of granzyme B and IFNγ in mice 309

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from

15

treated with anti-PDL1 antibodies were comparable to those of non-challenged mice 310

(Figure 7 C-D). These results indicate that intranasal PDL-1 blockade can enhance and 311

accelerate the cytotoxic function of immune cells in the airways. 312

We next analyzed the effect of intranasal PDL-1 blockade on CD8 T cell function. 313

Influenza virus-challenged mice treated with anti-PDL-1 antibodies had a statistically 314

significant higher frequency and absolute number of IFNγ CD107ab-double positive 315

cells than mice treated with control antibody or mock treated (Figure 7E-F). The 316

percentage of IFNγ and of CD107ab-single positive CD8 T cells was also higher in mice 317

treated with anti-PDL-1 antibodies but the differences were not statistically significant. 318

We did not observe any differences in CD8 T cell function in the draining lymph nodes 319

or the spleen between mice treated or not with anti-PDL-1 antibodies (data not shown). 320

Altogether, our data indicate that preventing the PDL-1/PD-1 interaction between the 321

airway epithelia and T cells results in enhanced IFNγ production and cytolytic function 322

by influenza virus-specific CD8 T cells. 323

PDL-1 blockade in the airways reduces influenza virus load and ameliorates 324

disease 325

We next analyzed the consequences of intranasal PDL-1 blockade on influenza virus 326

control on days 3 and 5 following a secondary viral challenge. The viral loads were 327

measured in lung homogenates using both qRT-PCR and infectious plaque assays. 328

The data show that PDL-1 blockade resulted in a slight reduction of influenza virus 329

nucleoprotein (NP) transcripts and of infectious viral titers by day three post infection 330

(Figure 8A). However, by day 5 post infection NP transcript levels and infectious virus 331

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from

16

titers were below the limit of detection in the anti-PDL-1 treated group (Figure 8B). 332

Conversely, untreated or control IgG-treated mice had significantly greater levels of 333

expression of NP RNA in the lungs as measured by qRT-PCR, and several orders of 334

magnitude (102-105) higher viral titers measured by plaque assay. We also subjected 335

each mouse from each group to whole body plethysmography on day 5 post infection 336

with Influenza. Enhanced pause (Penh) values in Influenza memory mice exposed to a 337

heterosubtypic influenza challenge but treated with α-PDL-1 registered values 338

equivalent to Influenza memory mice that did not receive the secondary influenza 339

challenge. Conversely, Penh values were significantly elevated in influenza memory 340

mice that were exposed to a heterosubtypic influenza challenge but were left untreated 341

or treated with control α-IgG (S3). Lastly, to determine the effect of intranasal anti-PDL-342

1 treatment on acute respiratory disease we monitored weight loss. We observed that 343

mice treated with anti-PDL-1 antibodies during influenza virus challenge did not present 344

any significant weight loss between day 0 and day 5 of treatment, while untreated or 345

control IgG-treated mice had a significant decrease in weight loss (Figure 9) suggesting 346

that intranasal anti-PDL-1 treatment can influence acute respiratory disease severity. 347

Altogether, our data indicate that blockade of PDL-1/PD-1 interaction in the airways 348

during influenza virus infection can result in enhanced viral control and disease 349

recovery. 350

351

DISCUSSION 352

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from

17

In this report, we showed that influenza virus infection of primary airway epithelial cells 353

strongly enhances PDL-1 expression, and does so in an IFNAR signaling dependent 354

manner. Most importantly, we observed that intranasal administration of anti-PDL-1 355

blocking antibodies to induce local blockade of PDL-1 in the airways improved T cell 356

function in the acute phase of infection as defined by increased production of IFNγ, 357

granzyme B, and cell surface mobilization of CD107ab. PDL-1 blockade in the airways 358

resulted in accelerated influenza virus clearance and enhanced infection recovery as 359

measured by weight loss. These data indicate that at the onset of influenza virus 360

infection the PD-1/PDL-1 axis is available to limit effector T cell function in the airways. 361

Our findings showing that PDL-1 was constitutively expressed on airway epithelial cells 362

prompted us to hypothesize that the PDL-1/PD-1 axis could be regulating local lung 363

immune responses as a means to maintain the integrity of the epithelial lining of the 364

respiratory tract during infection. The epithelial surfaces of the lung have been shown to 365

express molecules such as CD200 among others to regulate the function of immune 366

cells and maintain lung homeostasis (9). The observation that expression of PDL-1 in 367

airway epithelial cells is controlled by viral infection and IFNAR signaling suggests that 368

the PDL-1/PD-1 pathway plays an important role during inflammatory responses in the 369

lung. Innate interferons are critical for the response of airway epithelial cells to 370

respiratory viral infection (28). These observations indicate that interferon signaling also 371

contributes to the regulation of inflammatory homeostasis in the lung by enhancing the 372

expression of PDL-1 in airway epithelial cells, adding another layer of complexity to the 373

regulatory pathways operated by interferon stimulated genes. Our findings with airway 374

epithelial cells are in accordance with observations in other systems. Mice lacking TLR3 375

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from

18

had reduced PDL-1 expression (35); and a major downstream product of TLR3 376

signaling, IFNβ has been shown to enhance PDL-1 expression on monocytes, dendritic 377

cells and hippocampal cultures (35, 36). Furthermore, synthetic dsRNA has been shown 378

to trigger PDL-1 expression on epithelial cells (37, 38). Taken together, the above 379

findings suggest that PDL-1 expression in the periphery during the effector phase of the 380

response to virus infection likely represents a generalized host regulatory response to 381

viral infection rather than a virus-specific immunoevasion strategy. 382

The PD-1/PDL-1 signaling cascade is a prominent mechanism by which T cell non-383

responsiveness occurs in the face of chronic viral infections and cancer. The role of PD-384

1 expressing T cells during acute infection has remained controversial (39), although 385

recent observations report increased primary T cell responses in the absence of PD-1 386

signaling (25, 27, 40). Additionally, a recent study showed that the majority of PD-1 387

expressing T cells in healthy human adults do not exhibit an exhausted phenotype (41). 388

Our data show that PD-1 is primarily expressed on effector T cells in the lung compared 389

to effector memory and central memory cells, and that shortly after influenza virus 390

infection an increased number of PD-1+ T cells are recruited to the airways. Zelinskyy et 391

al. (23) also demonstrated that during acute Friend retrovirus infection, the PD-1-392

expressing CD8 T cells were terminally differentiated cytotoxic effectors. These studies 393

indicate that PD-1 is not a marker of T cell exhaustion per se, and suggest a likely 394

scenario during acute infection in which the PD-1/PDL-1 axis modulates inflammation 395

and ultimately serves to prevent tissue damage from activated effector T cells. 396

Using co-cultures of primary airway epithelial cells and T cells, and a mouse model of 397

influenza virus infection, we determined that blockade of airway epithelial PDL-1 398

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from

19

improves CD8 T cell function defined by increased production of IFNγ, granzyme B, and 399

expression of CD107ab. Importantly, therapeutic PDL-1 blockade in the airways 400

accelerated influenza virus clearance and enhanced infection recovery as measured by 401

weight loss. Our results are in accordance with recent findings by Erickson (11) showing 402

that viral acute respiratory infections impair CD8 T cells through PD-1, and that 403

systemic inhibition of PD-1 prevented CD8 T cell impairment. Our study adds that PD-404

1/PDL-1 regulation of CD8 T cell effector function is mediated locally by the epithelial 405

cells of the lung, it is regulated by IFNAR signaling, and can be therapeutically 406

manipulated in the airways by intranasal administration of anti-PDL-1 blocking 407

antibodies. As PD-1 signaling has been shown to impact dendritic cell maturation, 408

antigen presentation, and T cell priming and expansion (34, 42-44), it is important to 409

differentiate the systemic role of PD-1/PDL-1 during the generation of adaptive immune 410

responses from the its specific role at the mucosal site of infection during epithelial-T 411

cell interaction cross-talk. 412

Our findings are in line with several studies demonstrating that loss of PDL-1 during 413

acute viral infection can promote viral clearance (45, 46) and that PDL-1 blockade at the 414

onset of infection may serve as a potential therapeutic (27, 35). Clearly, further work is 415

needed to optimize the timing of therapy and to address role of other cell populations 416

expressing PD-1/PDL-1 in the lung. Nevertheless, our findings indicate that local 417

manipulation of the PDL-1/PD-1 pathway in the airways may represent a therapeutic 418

alternative during acute influenza virus infection. 419

420

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from

20

ACKNOWLEDGMENTS 421

We thank the Biomedical Genomics Core and Flow Cytometry Core Laboratory at 422

the Research Institute, and Dr. Mark Peeples and Sara Mertz for help with the HAE 423

cultures. This work was supported in part by NIH grant AI082962 and by The Research 424

Institute. 425

426

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from

21

REFERENCES 427

1. Taubenberger JK, Morens DM. 2008. The pathology of influenza virus 428

infections. Annu Rev Pathol 3:499-522. 429

2. Diamond G, Legarda D, Ryan LK. 2000. The innate immune response of the 430

respiratory epithelium. Immunol Rev 173:27-38. 431

3. Bals R, Hiemstra PS. 2004. Innate immunity in the lung: how epithelial cells fight 432

against respiratory pathogens. Eur Respir J 23:327-333. 433

4. Kato A, Schleimer RP. 2007. Beyond inflammation: airway epithelial cells are at 434

the interface of innate and adaptive immunity. Curr Opin Immunol 19:711-720. 435

5. Schleimer RP, Kato A, Kern R, Kuperman D, Avila PC. 2007. Epithelium: at 436

the interface of innate and adaptive immune responses. J Allergy Clin Immunol 437

120:1279-1284. 438

6. Shaykhiev R, Bals R. 2007. Interactions between epithelial cells and leukocytes 439

in immunity and tissue homeostasis. J Leukoc Biol 82:1-15. 440

7. Shornick LP, Wells AG, Zhang Y, Patel AC, Huang G, Takami K, Sosa M, 441

Shukla NA, Agapov E, Holtzman MJ. 2008. Airway epithelial versus immune 442

cell Stat1 Function for Innate Defense against Respiratory Viral Infection. J 443

Immunol 180:3319-3328. 444

8. Humlicek AL, Manzel LJ, Chin CL, Shi L, Excoffon KJ, Winter MC, Shasby 445

DM, Look DC. 2007. Paracellular permeability restricts airway epithelial 446

responses to selectively allow activation by mediators at the basolateral surface. 447

J Immunol 178:6395-6403. 448

9. Snelgrove RJ, Goulding J, Didierlaurent AM, Lyonga D, Vekaria S, Edwards 449

L, Gwyer E, Sedgwick JD, Barclay AN, Hussell T. 2008. A critical function for 450

CD200 in lung immune homeostasis and the severity of influenza infection. Nat 451

Immunol 9:1074-1083. 452

10. Ueno K, Koga T, Kato K, Golenbock DT, Gendler SJ, Kai H, Kim KC. 2008. 453

MUC1 mucin is a negative regulator of toll-like receptor signaling. Am J Respir 454

Cell Mol Biol 38:263-268. 455

11. Yamada C, Sano H, Shimizu T, Mitsuzawa H, Nishitani C, Himi T, Kuroki Y. 456

2006. Surfactant protein A directly interacts with TLR4 and MD-2 and regulates 457

inflammatory cellular response. Importance of supratrimeric oligomerization. J 458

Biol Chem 281:21771-21780. 459

12. Jia HP, Kline JN, Penisten A, Apicella MA, Gioannini TL, Weiss J, McCray 460

PB, Jr. 2004. Endotoxin responsiveness of human airway epithelia is limited by 461

low expression of MD-2. Am J Physiol Lung Cell Mol Physiol 287:L428-437. 462

13. Muir A, Soong G, Sokol S, Reddy B, Gomez MI, Van Heeckeren A, Prince A. 463

2004. Toll-like receptors in normal and cystic fibrosis airway epithelial cells. Am J 464

Respir Cell Mol Biol 30:777-783. 465

14. Greene CM, Carroll TP, Smith SG, Taggart CC, Devaney J, Griffin S, O'Neill 466

S J, McElvaney NG. 2005. TLR-induced inflammation in cystic fibrosis and non-467

cystic fibrosis airway epithelial cells. J Immunol 174:1638-1646. 468

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from

22

15. Butte MJ, Keir ME, Phamduy TB, Sharpe AH, Freeman GJ. 2007. 469

Programmed death-1 ligand 1 interacts specifically with the B7-1 costimulatory 470

molecule to inhibit T cell responses. Immunity 27:111-122. 471

16. Freeman GJ, Long AJ, Iwai Y, Bourque K, Chernova T, Nishimura H, Fitz LJ, 472

Malenkovich N, Okazaki T, Byrne MC, Horton HF, Fouser L, Carter L, Ling V, 473

Bowman MR, Carreno BM, Collins M, Wood CR, Honjo T. 2000. Engagement 474

of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to 475

negative regulation of lymphocyte activation. J Exp Med 192:1027-1034. 476

17. Francisco LM, Sage PT, Sharpe AH. 2010. The PD-1 pathway in tolerance and 477

autoimmunity. Immunol Rev 236:219-242. 478

18. Barber DL, Wherry EJ, Masopust D, Zhu B, Allison JP, Sharpe AH, Freeman 479

GJ, Ahmed R. 2006. Restoring function in exhausted CD8 T cells during chronic 480

viral infection. Nature 439:682-687. 481

19. Day CL, Kaufmann DE, Kiepiela P, Brown JA, Moodley ES, Reddy S, 482

Mackey EW, Miller JD, Leslie AJ, DePierres C, Mncube Z, Duraiswamy J, 483

Zhu B, Eichbaum Q, Altfeld M, Wherry EJ, Coovadia HM, Goulder PJ, 484

Klenerman P, Ahmed R, Freeman GJ, Walker BD. 2006. PD-1 expression on 485

HIV-specific T cells is associated with T-cell exhaustion and disease progression. 486

Nature 443:350-354. 487

20. Trautmann L, Janbazian L, Chomont N, Said EA, Wang G, Gimmig S, 488

Bessette B, Boulassel MR, Delwart E, Sepulveda H, Balderas RS, Routy JP, 489

Haddad EK, Sekaly RP. 2006. Upregulation of PD-1 expression on HIV-specific 490

CD8 + T cells leads to reversible immune dysfunction. Nat Med. 491

21. Urbani S, Amadei B, Tola D, Massari M, Schivazappa S, Missale G, Ferrari 492

C. 2006. PD-1 expression in acute hepatitis C virus (HCV) infection is associated 493

with HCV-specific CD8 exhaustion. J Virol 80:11398-11403. 494

22. Wherry EJ, Blattman JN, Murali-Krishna K, van der Most R, Ahmed R. 2003. 495

Viral persistence alters CD8 T-cell immunodominance and tissue distribution and 496

results in distinct stages of functional impairment. J Virol 77:4911-4927. 497

23. Zelinskyy G, Myers L, Dietze KK, Gibbert K, Roggendorf M, Liu J, Lu M, 498

Kraft AR, Teichgraber V, Hasenkrug KJ, Dittmer U. 2011. Virus-specific CD8+ 499

T cells upregulate programmed death-1 expression during acute friend retrovirus 500

infection but are highly cytotoxic and control virus replication. J Immunol 501

187:3730-3737. 502

24. Bowen DG, Shoukry NH, Grakoui A, Fuller MJ, Cawthon AG, Dong C, 503

Hasselschwert DL, Brasky KM, Freeman GJ, Seth NP, Wucherpfennig KW, 504

Houghton M, Walker CM. 2008. Variable patterns of programmed death-1 505

expression on fully functional memory T cells after spontaneous resolution of 506

hepatitis C virus infection. Journal of virology 82:5109-5114. 507

25. Allie SR, Zhang W, Fuse S, Usherwood EJ. 2011. Programmed death 1 508

regulates development of central memory CD8 T cells after acute viral infection. 509

J Immunol 186:6280-6286. 510

26. Telcian AG, Laza-Stanca V, Edwards MR, Harker JA, Wang H, Bartlett NW, 511

Mallia P, Zdrenghea MT, Kebadze T, Coyle AJ, Openshaw PJ, Stanciu LA, 512

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from

23

Johnston SL. 2011. RSV-induced bronchial epithelial cell PD-L1 expression 513

inhibits CD8+ T cell nonspecific antiviral activity. J Infect Dis 203:85-94. 514

27. Erickson JJ, Gilchuk P, Hastings AK, Tollefson SJ, Johnson M, Downing 515

MB, Boyd KL, Johnson JE, Kim AS, Joyce S, Williams JV. 2012. Viral acute 516

lower respiratory infections impair CD8+ T cells through PD-1. J Clin Invest 517

122:2967-2982. 518

28. Ioannidis I, McNally B, Willette M, Peeples ME, Chaussabel D, Durbin JE, 519

Ramilo O, Mejias A, Flano E. 2012. Plasticity and Virus Specificity of the Airway 520

Epithelial Cell Immune Response during Respiratory Virus Infection. J Virol 521

86:5422-5436. 522

29. Probst HC, McCoy K, Okazaki T, Honjo T, van den Broek M. 2005. Resting 523

dendritic cells induce peripheral CD8+ T cell tolerance through PD-1 and CTLA-524

4. Nat Immunol 6:280-286. 525

30. Keir ME, Freeman GJ, Sharpe AH. 2007. PD-1 regulates self-reactive CD8+ T 526

cell responses to antigen in lymph nodes and tissues. J Immunol 179:5064-5070. 527

31. Martin-Orozco N, Wang YH, Yagita H, Dong C. 2006. Cutting Edge: 528

Programmed death (PD) ligand-1/PD-1 interaction is required for CD8+ T cell 529

tolerance to tissue antigens. J Immunol 177:8291-8295. 530

32. Wei S, Shreiner AB, Takeshita N, Chen L, Zou W, Chang AE. 2008. Tumor-531

induced immune suppression of in vivo effector T-cell priming is mediated by the 532

B7-H1/PD-1 axis and transforming growth factor beta. Cancer research 68:5432-533

5438. 534

33. Wahl C, Bochtler P, Chen L, Schirmbeck R, Reimann J. 2008. B7-H1 on 535

hepatocytes facilitates priming of specific CD8 T cells but limits the specific recall 536

of primed responses. Gastroenterology 135:980-988. 537

34. Goldberg MV, Maris CH, Hipkiss EL, Flies AS, Zhen L, Tuder RM, Grosso 538

JF, Harris TJ, Getnet D, Whartenby KA, Brockstedt DG, Dubensky TW, Jr., 539

Chen L, Pardoll DM, Drake CG. 2007. Role of PD-1 and its ligand, B7-H1, in 540

early fate decisions of CD8 T cells. Blood 110:186-192. 541

35. Lafon M, Megret F, Meuth SG, Simon O, Velandia Romero ML, Lafage M, 542

Chen L, Alexopoulou L, Flavell RA, Prehaud C, Wiendl H. 2008. Detrimental 543

contribution of the immuno-inhibitor B7-H1 to rabies virus encephalitis. J Immunol 544

180:7506-7515. 545

36. Schreiner B, Mitsdoerffer M, Kieseier BC, Chen L, Hartung HP, Weller M, 546

Wiendl H. 2004. Interferon-beta enhances monocyte and dendritic cell 547

expression of B7-H1 (PD-L1), a strong inhibitor of autologous T-cell activation: 548

relevance for the immune modulatory effect in multiple sclerosis. Journal of 549

neuroimmunology 155:172-182. 550

37. Heinecke L, Proud D, Sanders S, Schleimer RP, Kim J. 2008. Induction of B7-551

H1 and B7-DC expression on airway epithelial cells by the Toll-like receptor 3 552

agonist double-stranded RNA and human rhinovirus infection: In vivo and in vitro 553

studies. J Allergy Clin Immunol 121:1155-1160. 554

38. Tsuda M, Matsumoto K, Inoue H, Matsumura M, Nakano T, Mori A, Azuma 555

M, Nakanishi Y. 2005. Expression of B7-H1 and B7-DC on the airway epithelium 556

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from

24

is enhanced by double-stranded RNA. Biochem Biophys Res Commun 330:263-557

270. 558

39. Brown KE, Freeman GJ, Wherry EJ, Sharpe AH. 2010. Role of PD-1 in 559

regulating acute infections. Curr Opin Immunol 22:397-401. 560

40. Channappanavar R, Twardy BS, Suvas S. 2012. Blocking of PDL-1 interaction 561

enhances primary and secondary CD8 T cell response to herpes simplex virus-1 562

infection. PLoS ONE 7:e39757. 563

41. Duraiswamy J, Ibegbu CC, Masopust D, Miller JD, Araki K, Doho GH, Tata 564

P, Gupta S, Zilliox MJ, Nakaya HI, Pulendran B, Haining WN, Freeman GJ, 565

Ahmed R. 2011. Phenotype, function, and gene expression profiles of 566

programmed death-1(hi) CD8 T cells in healthy human adults. J Immunol 567

186:4200-4212. 568

42. Talay O, Shen CH, Chen L, Chen J. 2009. B7-H1 (PD-L1) on T cells is required 569

for T-cell-mediated conditioning of dendritic cell maturation. Proc Natl Acad Sci U 570

S A 106:2741-2746. 571

43. Yao S, Wang S, Zhu Y, Luo L, Zhu G, Flies S, Xu H, Ruff W, Broadwater M, 572

Choi IH, Tamada K, Chen L. 2009. PD-1 on dendritic cells impedes innate 573

immunity against bacterial infection. Blood 113:5811-5818. 574

44. Rowe JH, Johanns TM, Ertelt JM, Way SS. 2008. PDL-1 blockade impedes T 575

cell expansion and protective immunity primed by attenuated Listeria 576

monocytogenes. J Immunol 180:7553-7557. 577

45. Iwai Y, Terawaki S, Ikegawa M, Okazaki T, Honjo T. 2003. PD-1 inhibits 578

antiviral immunity at the effector phase in the liver. J Exp Med 198:39-50. 579

46. Kirchberger S, Majdic O, Steinberger P, Bluml S, Pfistershammer K, 580

Zlabinger G, Deszcz L, Kuechler E, Knapp W, Stockl J. 2005. Human 581

rhinoviruses inhibit the accessory function of dendritic cells by inducing 582

sialoadhesin and B7-H1 expression. J Immunol 175:1145-1152. 583

584

585

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from

25

FIGURE LEGENDS 586

Figure 1. PDL-1 is constitutively expressed in the human airway epithelium and is 587

up-regulated by influenza infection in HAEs. (Panel A) mRNA expression of PDL-1 588

in mock treated HAE cultures or in HAE cultures 24 h post exposure to 2x105 PFU 589

influenza virus (Udorn strain). (Panel B) Relative mRNA levels of PDL-1 in Influenza 590

infected HAE cells validated by qRT-PCR. (Panel C) Immunofluorescent analysis of 591

PDL-1 expression (red) in a paraffin embedded human trachea section. The box inset 592

represents a high magnification detail of the apical side of the epithelium. (Panel D) 593

Immunofluorescent analysis of PDL-1 expression (red) in paraffin embedded HAE 594

cultures either mock treated (upper panel) or infected with influenza virus (lower panel). 595

In all immunofluorescent images, the nucleus is counterstained with DAPI (blue). Error 596

bars indicate SD, (*** p ≤ 0.0005). 597

598

Figure 2. IFNAR signaling regulates PDL-1 expression in TEC cultures. (Panel A) 599

PDL-1 mRNA expression analyzed in TEC cultures derived from balb/c (wt) and IFNAR-600

/- mice 24 h after +/- exposure to 2x105 PFU influenza virus (WSN). (Panels B & C) Cell 601

surface expression of PDL-1 on wt or IFNAR-/- TEC cultures infected with 0.1 PFU/cell 602

or 1.0 PFU/cell Influenza virus 24 and 60 hour post infection. Panel B: Representative 603

histograms. Panel C: Median fluorescent intensity (MFI) in which values used for the bar 604

graph represent the MFI of PD1L minus the MFI of the isotype control. All panels are 605

representative of 2-3 separate experiments in which TEC cultures were performed in 606

triplicate. Error bars indicate SD, (*** p ≤ 0.0005). 607

608

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from

26

609

Figure 3. Influenza virus infection induces early, non-specific recruitment of PD-1+ 610

effector T cells into the lung airways. Cohorts of naïve C57BL/6 mice were infected 611

with influenza virus X31 and the T cell phenotype was assessed at the indicated time 612

points. Representative dot plots show the frequency of PD-1+ CD44+ T cells present in 613

the CD4 and CD8 subpopulations of the BAL (Panel A) and Lung parenchyma (Panel B) 614

at the indicated time points after infection. Bar diagrams show the number of antigen 615

experienced PD1+ CD4 & PD-1+ CD8 T cells present in the BAL (Panel C), and the lung 616

parenchyma (Panel D). Data are representative of 3 independent experiments with 3 617

mice per group, error bars indicate SD, (* p ≤ 0.05, ** p ≤ 0.005). 618

619

Figure 4. PD-1 is mostly expressed on effector CD8 T cells. Cohorts of mice 620

previously infected with influenza virus X31 received intranasal administration of 50 ug 621

poly (I:C). (Panel A) Representative dot plots and histograms show PD-1 expression on 622

effector and memory T cell subsets within NP tetramer positive and negative CD8 T cell 623

populations in the lung parenchyma 3 days post stimulation. Bar diagrams show the 624

MFI and frequency of PD-1+ effector (CD43+CD62L-), effector-memory (CD43-CD62L-) 625

and central-memory (CD43-CD62L+) CD8 T cells residing in the NP366–374/Db tetramer-626

positive subset (Panel B) and in the tetramer-negative subset (Panel C). Data are 627

representative of 2 independent experiments with 3 mice per group. Error bars indicate 628

SD, (*** p ≤ 0.0005). 629

630

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from

27

Figure 5. PDL-1 blockade on airway epithelial cells enhances CD8 T cell function. 631

T cells isolated from the spleens of influenza-infected mice were co-cultured with TEC 632

cells infected or not with influenza virus WSN in the presence of anti-PDL-1 blocking 633

antibody or control IgG antibody. 36 hours after co-culture, supernatants were 634

harvested for the determination of granzyme B secretion by ELISA. Data are combined 635

from 2 separate experiments employing triplicate wells per group. Error bars indicate 636

SD, (* p ≤ 0.05). 637

638

Figure 6. PDL-1 blockade increases the concentration of granzyme B and IFNγ in 639

the airways during recall. Influenza-memory mice were secondarily challenged with 640

heterosubtypic influenza virus PR8, and intra-peritoneally treated with anti-mouse PDL-641

1 blocking antibody or rat IgG control. 3 days post-infection, BAL samples were assayed 642

for the presence of granzyme B (Panel A) or interferon-γ (Panel B) by ELISA. Data are 643

representative of 3 independent experiments (3 mice per group). Error bars indicate SD, 644

(* p ≤ 0.05). 645

646

Figure 7. Intranasal PDL-1 blockade enhances the secretion of granzyme B and 647

IFNγ in the airways during influenza virus recall. Influenza virus memory mice were 648

challenged with influenza virus PR8 (or mock challenged with HBSS), and treated 649

intranasally with anti-mouse PDL-1 blocking antibody or IgG control or HBSS (Flu). 650

ELISA was used to determine the levels of Granzyme B or IFN� present in the BAL on 651

days 3 & 5 post PDL-1 blockade (panels A-D). One-way ANOVA was used to compare 652

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from

28

the differences between means, and when these differences were significant (p ≤ 0.05), 653

the Tukey test was used to correct for multiple comparisons and to generate the 654

multiplicity adjusted P values shown. (Panel A) Granzyme B, day 3 (ANOVA, **p<.01); 655

(Panel B) IFNγ, day 3 (ANOVA, **p<.01); (Panel C) Granzyme B, day 5 (ANOVA, 656

**p<.01); (Panel D) IFNγ, day 5 (ANOVA, ****p<.0001). (Panel E) Frequency of IFNγ 657

and CD107ab-expressing NP366–374-specific CD8 T cells in lung. (Panel F) Number of 658

IFNγ and CD107ab expressing NP366–374-specific CD8 T cells in lung. Data are 659

representative of 2-4 independent experiments (3 mice per group). Error bars indicate 660

SD, (* p ≤ 0.05). 661

662

Figure 8. Reduced viral load in anti-PDL-1 treated mice. Influenza memory mice (30-663

40 days post intranasal inoculation with the Influenza X-31 strain) were intranasally 664

challenged with 1x102 PFU/ml influenza virus PR8 (or mock challenged with HBSS) and 665

treated with intra-nasal instillations of anti-mouse PDL-1 blocking antibody, rat IgG or 666

HBSS (Flu). On days 3 and 5 after initiation of PDL-1 blockade, influenza virus load in 667

lung homogenates was determined by qRT-PCR for the NP transcript (left panels) and 668

by plaque assay (right panels). (Panel A) Day 3. (Panel B) Day 5. Values for plaque 669

assay represent the means (+/- SD) of RSV log10 PFU/ml where 1.7 log10 PFU/ml 670

represents the limit of detection. Comparisons were made by one-way ANOVA 671

(****p<0.0001 for all panels), and the Tukey post-test was used to correct for multiple 672

comparisons and to generate the multiplicity adjusted P values shown. Data are 673

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from

29

representative of at least 2 independent experiments (3 mice per group). Error bars 674

indicate SD, (* p ≤ 0.05). 675

676

Figure 9. Intranasal PDL-1 blockade prevents influenza-associated weight loss. 677

Percent weight loss in mice inoculated and treated as in Figure 8 was determined by 678

dividing the average starting weight of each group (day 0, pre inoculation) by the 679

average weight of each treatment group on day 5 post-infection. Data are 680

representative of 3 independent experiments with 3 mice per group each. Error bars 681

indicate SD, (* p ≤ 0.05, *** p ≤0.0005). 682

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from

0

10

20

30

40

50

60

Mock Flu

Mock

Flu

A

D

54.3x

***

0

2000

4000

6000

8000

10000

Mock Flu

PD

-L1 R

ela

tive e

xpre

ssio

n

B

1 PD

-L1 R

ela

tive e

xpre

ssio

n 89.9x

***

1

C

Figure 1. PDL-1 is cons titu tive ly expres s ed in the human a irway ep ithe lium and is up-regula ted by influenza infec tion in HAEs . (Pane l A)

mRNA expression of PDL-1 in mock treated HAE cultures or in HAE cultures

24 h post exposure to 2x105 PFU influenza virus (Udorn strain). (Pane l B)

Relative mRNA levels of PDL-1 in Influenza infected HAE cells validated by

qRT-PCR. (Pane l C) Immunofluorescent analysis of PDL-1 expression (red) in

a paraffin embedded human trachea section. The box inset represents a high

magnification detail of the apical side of the epithelium. (Pane l D)

Immunofluorescent analysis of PDL-1 expression (red) in paraffin embedded

HAE cultures either mock treated (upper panel) or infected with influenza virus

(lower panel). In all immunofluorescent images, the nucleus is counterstained

with DAPI (blue). Error bars indicate SD, (*** p ≤ 0.0005).

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from

0

1000

2000

3000

4000

5000

6000

27.6x

*** A

C

B

Isotype control

wt

mock

0.1 PFU / cell 24h

1 PFU / cell 24h

0.1 PFU / cell 60h

1 PFU / cell 60h

0 10 2 10 3 10 4 10 5 0

20

40

60

80

100

0 10 2 10 3 10 4 10 5 0

20

40

60

80

100

0 10 2 10 3 10 4 10 5 0

20

40

60

80

100

0 10 2 10 3 10 4 10 5 0

20

40

60

80

100

0 10 2 10 3 10 4 10 5 0

20

40

60

80

100

0 10 2 10 3 10 4 10 5 0

20

40

60

80

100

0 10 2 10 3 10 4 10 5 0

20

40

60

80

100

0 10 2 10 3 10 4 10 5 0

20

40

60

80

100

0 10 2 10 3 10 4 10 5

0

20

40

60

80

100

0 10 2 10 3 10 4 10 5 0

20

40

60

80

100

PDL-1

IFNAR -/-

-50

0

50

100

150

200

250

mock

0.1

PF

U /

cell

24h

0.1

PF

U /

cell

60h

1.0

PF

U /

cell

24h

1.0

PF

U /

cell

60h

PD

L-1

MF

I

IFNAR-/-

wt

Wt m

ock

Wt F

lu

IFN

AR

-/-

Flu

IFN

AR

-/-

mock P

D-L

1 R

ela

tive e

xpre

ssio

n

Figure 2. IFNAR signaling regulates PDL-1 expression in TEC cultures.

(Panel A) PDL-1 mRNA expression analyzed in TEC cultures derived from

balb/c (wt) and IFNAR-/- mice 24 h after +/- exposure to 2x105 PFU influenza

virus (WSN). (Panels B & C ) Cell surface expression of PDL-1 on wt or

IFNAR-/- TEC cultures infected with 0.1 PFU/cell or 1.0 PFU/cell Influenza

virus 24 and 60 hour post infection. Panel B: Representative histograms.

Panel C: Median fluorescent intensity (MFI) in which values used for the bar

graph represent the MFI of PD1L minus the MFI of the isotype control. All

panels are representative of 2-3 separate experiments in which TEC cultures

were performed in triplicate. Error bars indicate SD, (*** p ≤ 0.0005).

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from

Figure 3. Influenza virus infection induces early, non-specific

recruitm ent of PD-1+ effector T cells into the lung airw ays. Cohorts of

naïve C57BL/6 mice were infected with influenza virus X31 and the T cell

phenotype was assessed at the indicated time points. Representative dot

plots show the frequency of PD-1+ CD44+ T cells present in the CD4 and

CD8 subpopulations of the BAL (Panel A) and Lung parenchyma (Panel

B) at the indicated time points after infection. Bar diagrams show the

number of antigen experienced PD1+ CD4 & PD-1+ CD8 T cells present

in the BAL (Panel C), and the lung parenchyma (Panel D ). Data are

representative of 3 independent experiments with 3 mice per group, error

bars indicate SD, (* p ≤ 0.05, ** p ≤ 0.005).

PD

-1

CD44

PD

-1

CD44

BAL

CD8

CD4

d0 d1 d3 d7

A

Lung

CD8

CD4

d0 d1 d3 d7

B

BAL

Lung

C

D

1

10

100

1000

10000

100000

1

10

100

1000

10000

100000

1

10

100

1000

10000

100000

1000000

1

10

100

1000

10000

100000

*

* *

* ** **

*

*

PD

-1+

CD

8 T

cells

1 3 7 12 30 0 1 3 7 12 30 0

1 3 7 12 30 0 1 3 7 12 30 0

day day

PD

-1+

CD

4 T

cells

PD

-1+

CD

8 T

cells

PD

-1+

CD

4 T

cells

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from

0 10 2 10 3 10 4 10 5

CD8

0

10 2

10 3

10 4

10 5

NP

te

tram

er

25.8

1.35

0 10 2 10 3 10 4 10 5

0

10 2

10 3

10 4

10 5 5.41 4.39

37.5 52.7

0 10 2 10 3 10 4 10 5

CD43

0

10 2

10 3

10 4

10 5

CD

62L

61.5 1.89

9.81 26.7

0 10 2 10 3 10 4 10 5

0

20

40

60

80

100

0 10 2 10 3 10 4 10 5

PD-1

0

20

40

60

80

100

% o

f M

ax

CD43- CD62L+

CD43- CD62L-

CD43+ CD62L- 0

500

1000

1500

2000

2500

3000

3500

MF

I

PD

-1

0

20

40

60

80

100

Fre

quency

P

D-1

0

1000

2000

3000

4000

5000

0

20

40

60

80

100 A B

Fre

quency

P

D-1

CD

62L

MF

I

PD

-1

CD43

% o

f M

ax

PD-1

C

*** ***

*** ***

Figure 4. PD-1 is m ostly expressed on effector CD8 T cells. Cohorts of

mice previously infected with influenza virus X31 received intranasal

administration of 50 mg poly (I:C). (Panel A) Representative dot plots and

histograms show PD-1 expression on effector and memory T cell subsets

within NP tetramer positive and negative CD8 T cell populations in the lung

parenchyma 3 days post stimulation. Bar diagrams show the MFI and

frequency of PD-1+ effector (CD43+CD62L-), effector-memory (CD43-CD62L-)

and central-memory (CD43-CD62L+) CD8 T cells residing in the NP366–374/Db

tetramer-positive subset (Panel B) and in the tetramer-negative subset (Panel

C ). Data are representative of 2 independent experiments with 3 mice per

group. Error bars indicate SD, (*** p ≤ 0.0005).

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from

0

50

100

150

200

250

300

control aPDL1

Gra

nzym

e B

(p

g/m

l)

*

control aPDL1

Influenza Mock

Figure 5. PDL-1 blockade on airw ay epithelial cells enhances CD8 T cell

function. T cells isolated from the spleens of influenza-infected mice were co-

cultured with TEC cells infected or not with influenza virus WSN in the

presence of anti-PDL-1 blocking antibody or control IgG antibody. 36 hours

after co-culture, supernatants were harvested for the determination of

granzyme B secretion by ELISA. Data are combined from 2 separate

experiments employing triplicate wells per group. Error bars indicate SD, (* p ≤ 0.05).

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from

0

200

400

600

800

1000

1200

1400

1600

IgG aPDL-1

Gra

nzym

e B

(p

g/m

L)

A

B

0

500

1000

1500

2000

2500

IgG aPDL-1

IFN

g (p

g/m

L)

*

*

Figure 6. PDL-1 blockade increases the concentration of granzym e B and

IFNg in the airw ays during recall. Influenza-memory mice were secondarily

challenged with heterosubtypic influenza virus PR8, and intra-peritoneally

treated with anti-mouse PDL-1 blocking antibody or rat IgG control. 3 days

post-infection, BAL samples were assayed for the presence of granzyme B

(Panel A) or interferon-g (Panel B) by ELISA. Data are representative of 3

independent experiments (3 mice per group). Error bars indicate SD, (* p ≤ 0.05).

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from

0

400

800

1200

1600

Mock Flu IgG aPDL1

0

2000

4000

6000

8000

Mock Flu IgG aPDL1 Gra

nzym

e B

(pg/m

l) *

*

A

C

0

400

800

1200

1600

Mock Flu IgG aPDL1

IFN

g (p

g/m

L)

*

IFN

g (p

g/m

L)

0

2000

4000

6000

8000

Mock Flu IgG aPDL1 Gra

nzym

e B

(pg/m

l)

B

D

CD10

7ab

CD10

7ab/

IFNg

IFNg

0

2

4

6

IgG

PDL-1

% C

D8 T

cells

#

CD

8 T

cells

E F

*

*

0

500

1000

1500

* *

*** ****

Figure 7. Intranasal PDL-1 blockade enhances the secretion of granzym e

B and IFNg in the airw ays during influenza virus recall. Influenza virus

memory mice were challenged with influenza virus PR8 (or mock challenged

with HBSS), and treated intranasally with anti-mouse PDL-1 blocking antibody

or IgG control or HBSS (Flu). ELISA was used to determine the levels of

Granzyme B or IFNg present in the BAL on days 3 & 5 post PDL-1 blockade

(panels A-D). One-way ANOVA was used to compare the differences between

means, and when these differences were significant (p ≤ 0.05), the Tukey test

was used to correct for multiple comparisons and to generate the multiplicity

adjusted P values shown. (Panel A) Granzyme B, day 3 (ANOVA, **p<.01);

(Panel B) IFNg, day 3 (ANOVA, **p<.01); (Panel C ) Granzyme B, day 5

(ANOVA, **p<.01); (Panel D ) IFNg, day 5 (ANOVA, ****p<.0001). (Panel E)

Frequency of IFNg and CD107ab-expressing NP366–374-specific CD8 T cells in

lung. (Panel F) Number of IFNg and CD107ab expressing NP366–374-specific

CD8 T cells in lung. Data are representative of 2-4 independent experiments (3

mice per group). Error bars indicate SD, (* p ≤ 0.05).

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from

A

B

0

5000

10000

15000

20000

25000

Mock Flu IgG aPDL1

d 3

NP

RN

A

Re

lative E

xpre

ssio

n

0 200 400 600 800

1000 1200 1400 1600 1800

Mock Flu IgG aPDL1

d 5

NP

RN

A

Re

lative E

xpre

ssio

n

**** ****

1

2

4

8

Mock Flu IgG PD1L

d3 V

iral lo

ad

Log

10 P

FU

/ml

1

2

4

8

Mock Flu IgG PD1L

d5 V

iral lo

ad

Log

10 P

FU

/ml ****

****

Figure 8. Reduced viral load in anti-PDL-1 treated m ice. Influenza memory

mice (30-40 days post intranasal inoculation with the Influenza X-31 strain)

were intranasally challenged with 1x102 PFU/ml influenza virus PR8 (or mock

challenged with HBSS) and treated with intra-nasal instillations of anti-mouse

PDL-1 blocking antibody, rat IgG or HBSS (Flu). On days 3 and 5 after

initiation of PDL-1 blockade, influenza virus load in lung homogenates was

determined by qRT-PCR for the NP transcript (left panels) and by plaque

assay (right panels). (Panel A) Day 3. (Panel B) Day 5. Values for plaque

assay represent the means (+/- SD) of RSV log10 PFU/ml where 1.7 log10

PFU/ml represents the limit of detection. Comparisons were made by one-way

ANOVA (****p<0.0001 for all panels), and the Tukey post test was used to

correct for multiple comparisons and to generate the multiplicity adjusted P

values shown. Data are representative of at least 2 independent experiments

(3 mice per group). Error bars indicate SD, (* p ≤ 0.05).

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from

0

20

40

60

80

100

120

Mock Flu IgG PDL1

% s

tart

ing w

eig

ht

day 0

day 5

* ***

Figure 9. Intranasal PDL-1 blockade prevents influenza-associated

w eight loss. Percent weight loss in mice inoculated and treated as in Figure 8

was determined by dividing the average starting weight of each group (day 0,

pre inoculation) by the average weight of each treatment group on day 5 post-

infection. Data are representative of 3 independent experiments with 3 mice

per group each. Error bars indicate SD, (* p ≤ 0.05, *** p ≤0.0005).

on April 4, 2019 by guest

http://jvi.asm.org/

Dow

nloaded from