Characterization of the expression profiles of ligands to activating receptors on the HLA-null cell lines K562 and 721.221 and on HIV-1 infected or non-infected CD4+ T cells

Alexandra Tremblay-McLean Department of Experimental Medicine McGill University

A thesis submitted to the Faculty of Graduate Studies and Research In fulfilment of the requirements for the degree of Master of Science

© Alexandra Tremblay-McLean February 2017 1 Characterization of the expression profiles of ligands to activating Natural 2 Killer cell receptors on the HLA-null cell lines K562 and 721.221 and on 3 HIV-1 infected or non-infected CD4+ T cells 4 Alexandra Tremblay-McLean 5 Abstract 6 Natural Killer (NK) cells direct anti-viral responses through a process dependent on the 7 integration of signals received from inhibitory and activating NK receptors (iNKR and aNKR). 8 NK cells can be activated by autologous HIV-infected CD4 T cells (iCD4) to inhibit HIV 9 replication. While iNKR and the downregulation of their HLA ligands on iCD4s have been 10 investigated, the contribution of aNKR and their ligands on iCD4 to NK cell activation and 11 subsequent anti-viral responses remain unclear. Additionally, previous work from our lab showed 12 that the HLA-null cell lines 721.221 (721) and K562 activate different frequencies and functional 13 subsets of NK cells. These cell lines do not express iNKR ligands, but their aNKR ligand profile 14 may differ in a manner that explains the how they activate NK cell differentially. In this thesis, I 15 will describe experiments that characterized the aNKR profile of iCD4 and HLA null cells to 16 improve our understanding of the way in which these cells activate NK cells. Using flow 17 cytometry, I analyzed the expression of a panel of ligands to aNKR on iCD4, K562, and 721 cells. 18 This panel included ULBP-1, ULBP-2/5/6, ULBP-3, MIC-A, MIC-B, CD48, CD80, CD86, 19 CD112, CD155, ICAM-1, ICAM-2, HLA-E, HLA-F, the ligands to the aNKR NKp30, NKp44, 20 NKp46, and KIR3DS1. I also compared the expression of HLA-A2 and HLA-C in uninfected CD4 21 and iCD4, which are differentially impacted by HIV infection. I found that, while K562 and 721 22 cells shared the expression of several aNKR ligands, K562 cells were characterized by the 23 expression of ULBP-2/5/6, ULBP-3, CD112, CD155, and the ligand to NKp30 and 721 cells were 24 characterized by the expression of MIC-B, CD48, CD80, CD86, the ligand to NKp44, and HLA- 25 E. The aNKR ligand profile of 721 cells is capable of interacting with a greater breadth of aNKR 26 on NK cells than that of K562 cells, which may in part explain why 721 cells can stimulate greater 27 frequencies of NK cells. Additionally, I found that iCD4 T cells, compared to uninfected CD4 T 28 cells, trended towards expressing a higher frequency and intensity, of aNKR ligands. I also 29 observed that iCD4 that maintained CD4 expression at their surface expressed greater levels of 30 aNKR ligands than iCD4 where membrane CD4 levels were downmodulated by the HIV Nef and 31 Vpu (iCD4-). The levels of aNKR ligand expression in iCD4- were similar to those of uninfected 32 CD4 T cells for ULBP-1, ULBP-2/5/6, ULBP-3, MIC-A and CD80. In contrast, the levels of 33 aNKR ligand expression in iCD4- were below those of uninfected cells for MIC-B, CD48, CD112, 34 CD155, and ICAM-2. These findings suggest that ligands to aNKR are transiently upregulated in 35 T cells upon HIV infection and that this is followed by a downregulation in what is thought to be 36 the more productively infected iCD4-. The ligands that are downregulated to levels below those in 37 uninfected cells are of particular interest for future research, as reducing the expression of these 38 ligands and their subsequent interactions with NK cell receptors may confer a survival advantage 39 to HIV infected cells. i

40 Profils d’expression des ligands de récepteurs activateurs de cellules NK 41 sur des lignées HLA-nul et des cellules T CD4+ infectées ou non par le 42 VIH-1 43 Alexandra Tremblay-McLean 44 Abrégé 45 Les réponses antivirales dirigées par les cellules NK dépendent de l’intégration de signaux 46 transmis par les récepteurs inhibiteurs (iNKR) et activateurs (aNKR) qu’elles expriment. Les 47 cellules T CD4+ infectées par le VIH-1 (iCD4) sont capables d’activer les cellules NK qui vont 48 ensuite inhiber la réplication du virus. Alors que l’impact des iNKR et de leurs ligands HLA 49 (exprimés par les iCD4) sur la réplication virale est relativement bien connu, la contribution des 50 aNKR et leurs ligands demeure moins bien compris. De récents travaux menés au laboratoire ont 51 montré que les lignées cellulaires K562 et 721.221 activaient de manière distincte les cellules NK 52 tant au point de vu qualitatif que quantitatif. Dans ce travail, nous avons choisi d’étudier le profil 53 d’expression des ligands de aNKR sur les iCD4 et les lignées cellulaires HLA-nul afin de mieux 54 comprendre comment ces ligands stimulent les cellules NK. Pour ce faire, nous avons analysé par 55 cytométrie de flux l’expression d’un panel de ligands de aNKRs sur les iCD4, les K562 et les 56 721.221. Ce panel inclut ULBP-1, ULBP-2/5/6, ULBP-3, MIC-A, MIC-B, CD48, CD80, CD86, 57 CD112, CD155, ICAM-1, ICAM-2, HLA-E, HLA-F, les ligands de NKp30, NKp44, NKp46, 58 KIR3DS1 et HLA-A2 / HLA-C qui sont des ligands d’iNKR exprimés par les cellules NK. Nos 59 résultats montrent que les cellules K562 et 721 expriment des niveaux similaires de plusieurs 60 ligands de aNKR. Toutefois, les K562 se caractérisent par une expression plus importante de 61 ULBP-2/5/6, ULBP-3, CD112, CD155 et du ligand de NKp30; alors que les 721 se caractérisent 62 par une expression plus marquée de MIC-B, CD48, CD80, CD86, du ligand de NKp44 et de HLA- 63 E. Globalement, les cellules 721 expriment davantage de ligands susceptibles d’interagir avec les 64 aNKR présents sur les cellules NK, ce qui pourrait expliquer leur meilleure capacité à stimuler ces 65 cellules. Par ailleurs, nous avons observé que les cellules T CD4+ infectées par le VIH avaient 66 tendance à exprimer des niveaux plus élevés de ligands de aNKR que les cellules non-infectées, et 67 ceci en terme de fréquence de cellules positives et d’intensité moyenne de fluorescence. Nous 68 avons également observé que l’expression de ces ligands était plus élevée sur les cellules infectées 69 qui ont gardé le CD4 à la membrane (iCD4+), comparativement aux cellules infectées qui n’ont 70 plus de CD4 membranaire (iCD4-), suite probablement à son internalisation par la protéine Nef du 71 VIH. Une analyse plus détaillée montre que les iCD4- présentent des niveaux de ULBP-1, ULBP- 72 2/5/6, ULBP-3, MICA et CD80 semblables aux cellules non-infectées, et des niveaux de MICB, 73 CD48, CD112, CD155 et ICAM-2 encore plus réduit que les cellules non-infectées. L’ensemble 74 de nos données suggère un modèle selon lequel l’expression des ligands de aNKRs serait 75 transitoirement augmentée à la surface des cellules T CD4+ lors de l’infection précoce par le VIH, 76 puis réduite sur les cellules infectées de manière productive, à l’instar du CD4. L’impact de 77 l’infection VIH sur l’expression des ligands de aNKR est d’un intérêt particulier pour la recherche 78 future dans la mesure ou la réduction de leur expression membranaire et, par conséquent, la 79 diminution de leur capacité à activer les cellules NK pourrait accorder un avantage de survie aux 80 cellules productrices de virus qui échapperaient à la lyse par les cellules NK. 81 ii

82 Acknowledgements 83 First and foremost, I would like to acknowledge my supervisor Dr. Nicole Bernard, who 84 provided the seeds that would germinate into the research presented here and also the 85 environment, support, and guidance required to bring those ideas to fruition. Additionally, I owe 86 a debt of gratitude to many members of our lab, both past and present. First, to Drs. Gamze 87 Isitman and Irene Lisovsky, who provided vital training and help with the flow cytometric 88 aspects of this work. To Drs. Sandrina DaFonseca and Franck Dupuis for helping with various 89 troubleshooting issues and for help refining this work. To our technicians, Xiaoyan Ni and 90 Tsoarello Mabanga, who worked tirelessly to procure reagents and the cryopreserved cell 91 samples that were the foundation of this work. Finally, to all the lab members not listed here who 92 helped along the way. 93 94 Many thanks to Dr. Galit Alter for the kind gift of our 721.221 cell line and to our 95 colleagues responsible for maintaining and organizing the HIV seronegative donor cohorts and 96 establishing the infrastructure required to provide us with leukapheresis samples. In particular, 97 I’d like to thank Dr. Julie Bruneau, Dr. Betrand Lebouché, Dr. Jean Pierre Routy, Dr Cecile 98 Tremblay and the hard-working nurses Josée Girouard and Pascale Arlotto. Finally, and with 99 enormous gratitude, I would like to acknowledge those who generously donated samples and 100 cells, without whom none of this research would have been possible. These individuals provided 101 a constant and crucial reminder of the real people that I hope one day my research may positively 102 impact. Considering their contributions, this work is dedicated to them. 103 104 105 106 107 108 109

110 iii

111 Table of Contents

112 Abstract…………………………………………………………………………………………………...... i

113 Abrégé……………………………………………………………………………………………………... ii

114 Acknowledgements……………………………………………………………………………………...... iii

115 Table of contents………………………………………………………………………………...... iv

116 Abbreviations…………………………………………………………………………………………….. vii

117 List of Tables…………………………………………………………………………………………..…... x

118 List of Figures…………………………………………………………………………………...... xi

119 Chapter 1: Introduction and Literature Review…………………………………………….... 1

120 1.1: Rationale………………………………………………………………………………….…………... 2

121 1.2.: Objectives…………………………………………………………………………………….…….... 4

122 1.3: History of the HIV pandemic………………………………………………………………….……... 6

123 1.3.2: Viral structure………………………………………………………………………………….… 7

124 1.3.3: Kinetics of infection……………………………………………………………………………... 8

125 1.3.4: HIV immunopathogenesis………………………………………………………………....….... 11

126 1.3.5: Discovery and function of NK cells………………………………………………………….… 13

127 1.3.6: NK cells in HIV………………………………………………………………………….....…... 15

128 1.4: NK cell licensing and activation…………………………………………………………………..... 16

129 1.4.2: Models for the study of NK cell activation………………………………....………………...... 18

130 1.5: Characterization of activating NK cell receptors and their ligands…………………………....….... 19

131 1.5.2: NKG2 receptors………………………………………………………………………...……... 19

132 1.5.3: Natural cytotoxicity receptors……………………………………………………………...... 23

133 1.5.4: Additional activating receptors……………………………………………………...……….... 26

134 Linker Paragraph………………………………………………………………………………...……..... 31 iv

135 Chapter 2: Manuscript 1 – The HLA-null cell lines K562 and 721.221 express different ligand 136 profiles for activating NK cell receptors …………………………………………………….... 32

137 Title page………………………………………………………………………………………..……...… 33

138 2.1: Author contributions and acknowledgements…………………………………………...... ….…… 34

139 2.2: Abstract…………………………………………………………………………………….…….... 35

140 2.3: Introduction………………………………………………………………………………….…...... 36

141 2.4: Methods………………………………………………………………………………………….… 40

142 2.4.1: Origin and preparation of K562 and 721 cell lines…………………………………………… 40

143 2.4.2: Antibody staining and acquisition……………………………………………………………. 40

144 2.4.3: Statistical analysis……………………………………………………………………………. 42

145 2.5: Results……………………………………………………………………………………………... 43

146 2.5.1: Comparison of the frequencies of K562 and 721 expressing ligands to aNKR…………….... 43

147 2.5.2: Comparison of the expression score for ligands to aNKR on the K562 and 721 HLA-null cell 148 lines ……………………………………………………………………………………………….... 44

149 2.6: Discussion…………………………………………………………………………………………. 46

150 2.7: Figure Legends………………………………………………………………………………….…. 51

151 2.8: References……………………………………………………………………………………….… 54

152 Linker Paragraph……………………………………………………………………………………….… 58

153 Chapter 3: Manuscript 2 – Expression profiles of ligands for activating NK cell receptors on 154 HIV infected and uninfected CD4+ T cells………………………………………………….… 59

155 Title page…………………………………………………………………………………………….....… 60

156 3.1: Author contributions and acknowledgements……………………………………………………... 61

157 3.2: Abstract………………………………………………………………………………………….… 62

158 3.3: Introduction………………………………………………………………………………………... 63

159 3.4: Methods……………………………………………………………………………………………. 66

v

160 3.4.1: Ethics statement………………………………………………………………………….…… 66

161 3.4.2: Study population…………………………………………………………………………….... 66

162 3.4.3: CD4+ isolation………………………………………………………………………….. 66

163 3.4.4: HIV infection………………………………………………………………………………..... 67

164 3.4.5: Antibody staining and acquisition……………………………………………………….…… 67

165 3.4.6: Statistical analysis……………………………………………………………………...…….. 69

166 3.5: Results………………………………………………………………………………………...... 70

167 3.5.1: CD4+ T cell infection characteristics……………………………………….………….……... 70

168 3.5.2: Comparison of the expression profiles of ligands to aNKR on HIV-infected CD4+ T cells that 169 maintain or not cell surface expression of CD4………………………………………………….. 71

170 3.5.3: Comparison of the frequencies of HIV iCD4 and unCD4 T cells expressing ligands to aNKRs 171 ………………………………………………………………………………………………….... 71

172 3.5.4: Comparison of the aNKR ligand expression intensity on iCD4 and unCD4 T cells…………. 72

173 3.5.5: Expression profiles of HLA-E, HLA-A2, and HLA-C of iCD4 and unCD4……………….... 73

174 3.6: Discussion………………………………………………………………………………………..... 74

175 3.7: Figure Legends………………………………………………………………………………….…. 80

176 3.8: References…………………………………………………………………………………..……... 92

177 Chapter 4: Discussion………………………………………………………………………...... 97

178 4.1: Contributions of this work to the field………………………………………………………...... 98

179 4.2: Modulation of cell-surface ligands by HIV Nef………………………………………………….. 100

180 4.3: Limitations and future directions…………………………………………………………………. 105

181 4.4: Conclusion………………………………………………………………………………….…….. 108

182 Appendix A: Antibodies used in the analysis of ligand expression …………………...…………...... 109

183 Appendix B: Panel of the ligands studied and their respective activating NK cell receptors...... 110

184 References…………………………………………………………………………………………...... 111 vi

185 Abbreviations ER: Endoplasmic reticulum

186 Ab: Antibody ERK: Extracellular signal-regulated kinase

187 ADCC: Antibody-dependent cellular ERM: Ezrin/radixin/moesin

+ 188 cytotoxicity euCD4: HIV uninfected CD4 T cells that

189 AIDS: Acquired immune deficiency were exposed to virus

190 syndrome FBS: Fetal bovine serum

191 ARS: Acute retroviral syndrome Grb2: Growth factor receptor-bound protein

192 aNKR: Activating natural killer cell receptor 2

193 Bp: Base pair HA: Viral hemagglutinin

194 CA: Capsid HAART: Highly-active antiretroviral

195 CML: Chronic myelogenous leukemia therapy

196 CMV: Cytomegalovirus HLA: Human leukocyte antigen

197 CSK: C-terminal Src kinase HIV: Human immunodeficiency virus

198 CTL: Cytotoxic ICAM: Intercellular adhesion molecule

+ 199 CTLA-4: Cytotoxic T-lymphocyte- iCD4: HIV-infected CD4 T cells

+ + 200 associated protein 4 iCD4 : HIV-infected CD4 T cells that

201 DAP: DNAX-activating protein maintain CD4 at their surface

- + 202 DC: Dendritic cell iCD4 : HIV-infected CD4 T cells with

203 DNA: Deoxyribonucleic acid downregulated surface CD4

T 204 DNAM-1: DNAX accessory molecule-1 iCD4 : Total HIV-infected CD4 T cells,

205 dPBS: Dulbecco’s phosphate buffered saline regardless of surface CD4 expression

206 dsDNA: Double-stranded DNA IFN: Interferon

207 EBV: Epstein-Barr virus Ig: Immunoglobulin vii

208 iKIR: inhibitory KIR Nef: Negative regulatory factor

209 IL: Interleukin NK: Natural killer

210 ILC: Innate lymphoid cell NKG2: Natural-killer group 2 member

211 IN: Integrase receptors

212 iNKR: Inhibitory natural killer cell receptor NTB-A: NK-T- antigen

213 ITAM: Immunoreceptor tyrosine-based PBMC: Peripheral mononuclear cell

214 activating motif p-ERK: RNA-like endoplasmic reticulum

215 ITIM: Immunoreceptor tyrosine-based kinase

216 inhibitory motif PHA: Phytohemagglutinin

217 ITSM: Immunoreceptor tyrosine-based PIC: Pre-integration complex

218 switch motif PI3K: Phosphoinositide kinase-3

219 Kb: Kilobases pNK: Peripheral blood NK cell

220 KIR: Killer immunoglobulin-like receptor PR: protease

221 LFA-1: Lymphocyte function-associated Pyk2: Protein tyrosine kinase 2-β

222 antigen-1 RAET1: Retinoic acid early transcript 1

223 MA: Matrix protein Rev: Regulator of expression of virion

224 MAPK: Mitogen-activated protein kinase protein

225 MFI: Mean fluorescence intensity rhIL-2: Recombinant human interleukin-2

226 MHC: Major histocompatibility complex RNA: Ribonucleic acid

227 MIC: MHC class-I-chain-related protein RT: Reverse transcriptase

228 mRNA: Messenger ribonucleic acid R10: RPMI 1640 medium supplemented

229 NC: Nucleocapsid with 10% FBS; 2mM L-glutamine;

230 NCR: Natural cytotoxicity receptor

viii

100IU/mL penicillin; 100mg/mL Vpu: Viral protein unique

streptomycin Zap-70 : Zeta-chain-associated protein

231 SAP: SLAM-associated protein kinase 70

232 SC: Drosophila Schneider cell 3DL1: KIR3DL1

233 SHIP: SH2 domain-containing inositol 5'- 3DS1: KIR3DS1

234 phosphatase 721: 721.221 HLA-null cell line

235 SHP: Src homology region 2 domain-

236 containing phosphatase

237 SIV: Simian immunodeficiency virus

238 SLAM: Signaling lymphocyte activation

239 molecule

240 ssRNA: Single-stranded RNA

241 Syk: Spleen tyrosine kinase

242 Tat: Transactivator of transcription

243 TIGIT: T cell immunoreceptor with

244 immunoglobulin and ITIM motifs

245 ULBP: UL16-binding protein ligands

246 unCD4: HIV-uninfected CD4+ T cells

247 Vav1: Vav guanine nucleotide exchange

248 factor-1

249 Vif: Viral infectivity factor

250 VL: Viral load

251 Vpr: Viral protein R

ix

252 List of Tables

253 Chapter 3: Manuscript 2 - Expression profiles of ligands for activating NK cell receptors 254 on HIV infected and uninfected CD4+ T cells.

255 Table 1: Study population HLA genotypes………………………………………………….………….. 83

256 Table 2: Description of the aNKR ligands studied…………………………………………………...… 84

257 Appendix A: Antibodies used in the analysis of aNKR ligand expression……..………………………... 109

258

259

260

261

262

263

264

265

266

267

268

269

270

271

x

272 List of Figures

273 Chapter 1: Introduction and Literature Review

274 Figure 1: Schematic diagram of the HIV virion and genome…………………………………………..... 8

275 Figure 2: HIV replication cycle………………………………………………………...... 11

276 Figure 3: NKG2 receptor signaling………………………………………………………...... … 20

277 Figure 4: NKG2D ligands………………………………………………………………….…………… 21

278 Figure 5: Structure of natural cytotoxicity receptors…………………………………………………… 25

279 Chapter 2: Manuscript 1 - The HLA-null cell lines K562 and 721.221 express different ligand 280 profiles for activating NK cell receptors.

281 Figure 1: Different frequencies of K562 and 721 express ligands to activating NK cell receptors...... 52

282 Figure 2: K562 and 721 differ in the expression score of their ligands to activating NK cell receptor…...53

283 Chapter 3: Expression profiles of ligands for activating NK cell receptors on HIV infected 284 and uninfected CD4+ T cells.

285 Figure 1: Gating strategy………………………….……………………………………………………. 85

286 Figure 2: Frequencies of iCD4+ vs iCD4- expressing aNKR ligands……………….………………..… 86

287 Figure 3: Frequencies of all T cell subsets expressing aNKR ligands………………………………….. 87

288 Figure 4: Mean fluorescence intensity of aNKR ligand expressing T cell subsets……………………... 88

289 Figure 5: Expression profile of the HLA molecules HLA-E, HLA-A2, and HLA-C on CD4+ T cell 290 subsets………………………………………………………………………………………….……... 89

291 Supplemental Figure 1: NK cells preferentially target HIV-infected CD4 T cells that conserve cell-surface 292 CD4 expression…………………………………………………………………………...………….... 90

293 Supplemental Figure 2: The frequency and per-cell expression of ICAM-1 is correlated with HIV- 294 infection on all infected T cell subsets…………………………………………………………………. 91

295 Appendix B: Panel of the ligands studied and their respective activating NK cell receptors …………... 110

xi

296 Chapter 1: Introduction and Literature Review

297

298

299

300

301

302

303

304

305

306

307

308

309

310

311

312

313

314

315

316

317

- 1 -

318 1.1: Rationale

319 Natural Killer (NK) cells are a subset of that are crucial mediators of innate

320 immune responses against stressed or otherwise damaged, virally-infected, and transformed

321 cells. NK cells can directly kill target cells through the release of cytotoxic granules and

322 secretion of cytokines and chemokines. They can utilize their secretory abilities to interact with

323 T and B cells, effectively bridging the innate and adaptive immune systems (1). Activation of

324 NK cells by targets cells encountered is a process that depends on the integration of signals

325 received through inhibitory and activating NK receptors (iNKRs and aNKRs) (2, 3). NK cells

326 can be activated through abrogation of inhibitory signaling through iNKRs if they also receive

327 activating signals through aNKRs. A variety of cell types can be used in vitro or ex vivo for NK

328 cell stimulation assays to better understand the complex process of their activation. For example,

329 HLA-null cell lines are frequently used as NK cell stimuli. Human leukocyte antigen (HLA)-null

330 cells do not express classical major histocompatibility complex (MHC)-I HLA-A, -B, or -C,

331 which are ligands for iNKRs. They are therefore unable to engage iNKRs on NK cells,

332 abrogating inhibitory signaling through these receptors, but still express ligands for aNKR and

333 can stimulate NK activation. Previous work by our group comparing NK cell stimulation by the

334 HLA-null cell lines K562 and 721.221 (721) found that they stimulated NK cells differentially in

335 terms of secretion of the cytokine IFN-γ, the chemokine CCL4, and expression of the

336 degranulation marker CD107a (4). Additionally, 721 cells activated a greater fraction of NK

337 cells, compared to K562 cells. Although the mechanisms underlying these differences are

338 currently unclear, in the absence of ligands for iNKRs, examining the expression profiles of

- 2 -

339 ligands for aNKRs expressed by K562 and 721 is a promising way to address this knowledge gap

340 and further elucidate the ways in which NK cells can be activated.

341 HIV-infected CD4 T cells (iCD4s), which express reduced cell surface levels of the

342 iNKR ligands HLA-A and -B, can also stimulate NK cells ex vivo and have been shown to

343 activate NK cells to inhibit HIV-1 replication in autologous iCD4s (5). While the role of HLA-A

344 and -B downregulation in stimulating NK cells has been studied, it is unclear how ligands for

345 aNKRs contribute to NK cell activation. It is plausible that HIV infection can modulate aNKR

346 ligand expression in a similar manner to its noted effects on classical HLA expression,

347 subsequently influencing NK cell activation and anti-HIV responses. Recently, the RV144 Thai

348 trial demonstrated that a vaccine regimen that combined an ALVAC HIV vaccine prime and

349 AIDSVAX B/E vaccine boost could provide protection against subsequent HIV challenge (6).

350 Although the protection afforded was modest, this marked the first successful vaccine trial and

351 confirmed that development of an effective anti-HIV vaccine is possible. Secondary analyses of

352 samples obtained during the trial highlighted NK cell-mediated antibody-dependent cellular

353 cytotoxicity as a correlate of protection (7). These findings help emphasize the importance of a

354 thorough understanding of NK cell activation and NK-cell mediated anti-HIV responses in the

355 design of future anti-HIV vaccines and treatment. The work presented here analyses the aNKR

356 ligand expression profiles of HIV-infected and uninfected CD4+ T cells, with goal of clarifying

357 their different abilities to trigger NK cell activation and anti-HIV responses.

358

359

- 3 -

360 1.2: Objectives

361 Following our previous observation that the two HLA-null cell lines K562 and 721

362 activated different frequencies of total NK cells and stimulated different functional responses, we

363 were interested in investigating (i) whether K562 and 721 possessed different aNKR ligand

364 expression profiles that could explain the differences observed and (ii) whether HIV-1-infected

365 CD4+ T cells, which are also used ex vivo to stimulate NK cells, might also influence NK

366 activation through an aNKR ligand expression profile that is modulated by HIV-1 infection.

367 We compiled a comprehensive panel of ligands to aNKR that included ULBP-1, ULBP-

368 2/5/6, ULBP-3. MIC-A, MIC-B, CD48, CD80, CD86, CD112, CD155, ICAM-1, ICAM-2, the

369 still largely unidentified ligands to NKp30, NKp44, and NKp46, and HLA-E, which is a non-

370 classical MHC that previous work from our lab suggested is expressed on 721 cells. The recent

371 identification of HLA-F as the ligand to the activating KIR3DS1 receptor on NK cells also

372 prompted us to extend our analyses to include this ligand (8, 9). We chose to analyze the

373 expression of HLA-F directly with the use of an HLA-F specific antibody and indirectly with the

374 use of a KIR3DS1-Fc chimeric molecule. Two ligands to iNKRs, HLA-C and the HLA-A were

375 also included in our study. The reason for this was to corroborate the well-established

376 observation that HLA-A expression is modulated by HIV infection and the recent finding that

377 HLA-C expression on HIV-infected CD4+ T cells is influenced by the viral strain used to infect

378 them (10). Examining HLA-A expression in both K562 and 721 confirmed that our in-house cell

379 lines were indeed HLA-null as expected, corroborating previous work from our lab showing that

380 a pan-MHC-1 antibody failed to stain these cell lines.

- 4 -

381 The work presented here offers a more comprehensive characterization of the aNKR

382 ligand profiles of 721 and K562, as the antibody panel used in this thesis research probes a

383 broader range of aNKR ligands than has been done previously. This work also marks the first

384 head to head comparison of both HLA null cell lines.

385 Upon HIV infection, two clear populations of infected cells can be observed among

386 iCD4: infected cells that maintain surface expression of CD4 (iCD4+) and infected cells where

387 the cell surface expression of CD4 has been downregulated by the HIV-associated proteins Nef

388 and Vpu (iCD4-) (11, 12). Early work examining HIV-mediated CD4 downregulation suggested

389 that infected cells with downregulated CD4 were at a later stage of infection (12). It is also

390 plausible that these cells were more productively infected. Indeed, experiments in this thesis will

391 show that these cells expressed higher levels of the p24 viral capsid protein, which serves as a

392 marker of infection. Despite these differences, a review of the existing literature demonstrated a

393 lack of consensus in the way iCD4 should be examined. Some work analyzed iCD4- exclusively,

394 while the remainder disregarded the differences between both infected subsets entirely and

395 combined them to analyze total iCD4. As such, we were also interested in characterizing the

396 different aNKR ligand expression profiles of HIV-1-infected CD4+ T cells that maintained or

397 downregulated cell surface CD4. In so doing, we aimed to add to our current understanding of

398 how these two HIV-1-infected T cell subsets differed and further emphasize that they should be

399 considered separately.

400 Considering this, the ultimate goal of my thesis research was to compile a thorough

401 repertoire of the aNKR ligands expressed on the cell types commonly used to stimulate NK cells

402 in in vitro and ex vivo activation models, including K562, 721, and HIV-1-infected CD4+ T cells.

- 5 -

403 Literature Review

404 1.3: History of the HIV pandemic

405 The first cases of acquired immunodeficiency syndrome (AIDS) were observed in the

406 early 1980s and shortly after, in 1983, the human immunodeficiency virus subtype 1 (HIV) was

407 identified as its causative agent (13, 14). Since its discovery, HIV has continued to disseminate

408 globally and is responsible for what is now one of the deadliest viral pandemics in history. As of

409 2015, almost 37 million people globally are living with HIV, while 35 million have died of

410 AIDS-related illness since the beginning of the epidemic (15). Therapeutic advances allowing for

411 the administration of highly-active antiretroviral therapy (HAART) with increasing potency and

412 tolerability and reduced toxicity have been instrumental in limiting the spread of the virus.

413 However, attempts at designing an effective HIV vaccine have not been so successful. Despite

414 the continually increasing availability of highly effective treatment, HIV continues to target

415 vulnerable high-risk populations and 2015 saw an additional 2 million people become newly

416 infected (15). In Canada, the prevalence of HIV continues to increase and is disproportionately

417 represented in men who have sex with men (53% of all Canadians living with HIV), injection

418 drug users (19% of all Canadians living with HIV), and First Nations, Métis, and Inuit peoples

419 (9% of all Canadians living with HIV) (16-18). It is evident that treatment alone is insufficient to

420 control HIV transmission and that these methods need to be supplemented with new approaches

421 arising from continued preventative and curative research.

422 1.3.2: Viral Structure

- 6 -

423 HIV is a lentivirus of the Retroviridae family. The icosahedral HIV virion is enveloped

424 by a host cell-derived lipid bilayer that contains viral glycoproteins and some cellular membrane

425 proteins acquired from the host cell membrane (Figure 1). The virion contains a capsid-enclosed

426 genome composed of two copies of linear, negative-sense, single-stranded RNA (ssRNA)

427 molecules. The HIV genome has 3 major coding regions: gag, which encodes structural proteins

428 like the capsid protein p24, pol, which encodes the viral protease (PR), reverse transcriptase

429 (RT), and integrase (IN), and env, which encodes viral glycoproteins (Figure 1). The HIV

430 genome also contains several additional accessory genes, for a total of 9 open reading frames

431 within a genome of less than 10 kilobases (19, 20). In addition to the viral genome, the capsid

432 contains the viral nucleocapsid (NC), IN, RT, and the accessory protein Vpr.

433 As with most retroviruses, HIV is characterized by significant genetic heterogeneity and

434 these variants comprise three major phylogenetic groups: M (main), O (outlier), and N (non-M or

435 -O) (21-23). Group M variants make up the majority of global infection and can be further

436 subdivided into nine clades (A-K), of which clades A, B, and C are most prevalent (24).

437 Furthermore, there are two different strains of HIV, HIV-1 and HIV-2, which have similar

438 structures and genome, but are associated with different pathologies. HIV-2, largely confined to

439 West Africa, is less pathogenic and less transmissible than the pandemic strain HIV-1 (25).

- 7 -

440 Figure 1

441 Figure 1: Schematic diagram of the HIV virion and genome. The enveloped HIV virion contains an encapsidated 442 genome the codes for Gag, Pol, and Env, in additional to other accessory proteins (see text).

443 Image Source: adapted from Karlsson 2008 (26) and Peterlin 2003 (27).

444 1.3.3: Kinetics of infection

445 The majority of HIV infections arise as a result of sexual transmission through the genital

446 tract or rectal mucosa (28, 29). In vitro experiments using mucosal tissue and studies in non-

447 human primate models have helped elaborate how HIV infection is established. In this context,

448 HIV infection begins when free virions or cell-associated virus crosses the mucosal barrier into

449 the lamina propria (30-32). Within the lamina propria, HIV preferentially targets resting and

450 activated CD4+ T cells, but will also target macrophages and dendritic cells (DCs) (32-34). These

451 small populations of infected cells will propagate locally and produce virus until their numbers

452 are sufficient to disseminate into draining lymph nodes and other lymphoid tissues, establishing a

453 systemic infection (30-32). At this point, latently infected viral reservoirs have been established

454 and will sustain long-term self-propagating infection.

- 8 -

455 At the cellular level, the mature HIV virion will attach to target CD4 T cells through

456 interactions between the viral envelope (Env) proteins gp41 and gp120 and the amino-terminal

457 immunoglobulin (Ig) domain of CD4 (35). Binding to CD4 is insufficient for infection, which

458 also requires interactions with the additional cell surface proteins CCR5 and CXCR4 (Reviewed

459 in 36). Conformational changes in gp120 that are induced by CD4 binding permit its interaction

460 with these co-receptors (37, 38). Subsequently, gp41 will also undergo a conformational change

461 that results in the fusion of the viral and host cell membranes and release of the viral capsid into

462 the host cell (38).

463 Membrane fusion is followed by uncoating events that are incompletely understood and

464 by the RT-catalyzed reverse transcription of the ssRNA HIV genome into linear double-stranded

465 DNA (dsDNA) (35, 39). RT lacks proofreading ability and frequently introduces mutations into

466 the viral genome (40, 41). The low fidelity of RT in conjunction with the rapid in vivo viral

467 turnover drive HIV’s extensive genetic variation and ability to rapidly adapt to host- and drug-

468 mediated selection pressures (42, 43). RT-dependent DNA synthesis also requires viral NC,

469 which is a nucleic acid chaperone, and may be facilitated by the accessory protein Viral

470 Infectivity Factor (Vif) (44, 45). Newly synthesized viral DNA is transported to the nucleus as

471 part of a preintegration complex (PIC) that also includes IN, RT, matrix (MA) and Viral Protein

472 R (Vpr) proteins (46). Vpr targets the PIC to the nucleus through interactions with the cellular

473 nuclear import machinery and can also arrest infected cells in the G2 phase of the cell cycle (47,

474 48). The transported viral DNA can then be integrated into the host genome by IN. These steps

475 make up the early phase of HIV’s viral life cycle. During the late phase, infected cells can either

476 enter the productive cycle whereby new viruses are assembled and released to propagate

- 9 -

477 infection or the integrated viral DNA can remain transcriptionally silent. Infected cells

478 containing transcriptionally silent viral DNA form viral reservoirs that can persist in HAART-

479 treated individuals and that can be reactivated following treatment interruption, a major

480 impediment to achieving a sterilizing cure for this disease (49-51).

481 The late phase of the productive HIV replication cycle begins with the synthesis of viral

482 mRNA transcripts that are translocated back to the cytoplasm, where they are translated. The

483 viral proteins Transactivator of Transcription (Tat), Regulator of Expression of Virion protein

484 (Rev), and Negative Regulatory Factor (Nef) are synthesized first and facilitate the transcription

485 and nuclear export of unspliced or singly-spliced mRNAs encoding Gag, Gag-Pol, Env, Viral

486 Protein Unique (Vpu), Vif, and Vpr (35). Tat is a transcriptional activator that binds to the Tat-

487 responsive element on nascent RNA transcripts and stimulates transcription elongation (35, 52,

488 53). Rev binds to Rev-response elements on nascent unspliced mRNA and shuttles it out of the

489 nucleus, despite unspliced mRNAs normally being retained in the nucleus (35, 54, 55). Once in

490 the cytosol, HIV RNA will dimerize and two copies of full length viral RNA will be packaged

491 into new viral particles (35). Together with the Gag-Pol precursor, Gag and HIV RNA will

492 assemble into an immature viral particle at the plasma membrane (35, 56). The viral RNA

493 genome will be packaged into the forming viral particle and encapsidated. The newly formed

494 viral particle will bud off the infected host cell.

495 Following budding, PR cleaves Gag and Gag-Pol to produce MA, capsid (CA), and NC,

496 which will rearrange during maturation to form the infectious viral particle (35, 56). Efficient

497 budding relies on the function of the accessory proteins Vpu and Nef. The viral Env protein

498 gp160 (precursor to gp120) and host CD4 are both produced in the endoplasmic reticulum (ER)

- 10 -

499 and premature gp160-CD4 interactions can impair the transport of gp160 to the plasma

500 membrane and the formation of viral particles (57). To prevent this, Vpu targets CD4 for

501 degradation, thus removing it from the ER (58-60). Similarly, Nef downregulates cell surface

502 CD4, which prevents premature binding of Env to CD4 and reinfection of an infected cell by its

503 own budding virions (61).

504 Figure 2

505 Figure 2: HIV replication cycle. (1) Viral attachment and fusion require interactions between envelope 506 glycoproteins CD4, as well as the co-receptors CCR5 and CXCR4. (2) The uncoated ssRNA genome is reverse 507 transcribed into dsDNA by the viral reverse transcriptase. (3) Newly synthesized DNA is translocated to the as part 508 of the preintegration complex and integrated into the host genome. (4) Viral mRNA is transcribed in the nucleus and 509 transported to the cytoplasm. (5) Viral proteins are translated and the replicated viral genome is packaged into 510 nascent viral particles at the cell membrane. (6) Viral particles bud off from the cell, coating themselves in host 511 membrane, and will continue to mature into infectious virions. See text for more details.

512 Image Source: Adapted from Deeks 2015 (62)

513 1.3.4: HIV immunopathogenesis

514 HIV infection follows a well-defined time course that begins with primary infection.

515 Within 3-6 weeks following infection, between 50-70% of individuals will experience acute

516 retroviral syndrome (ARS) (19, 63). Although the clinical manifestations of ARS and their - 11 -

517 severity vary greatly between individuals, common symptoms include fever, fatigue and general

518 malaise, pharyngitis, maculopapular rash, and lymphadenopathy (64, 65). While ARS usually

519 resolves within two weeks, lymphadenopathy and malaise may persist (19, 66). During primary

520 infection, plasma viral loads increase dramatically and peak around 3 weeks following infection

521 (29, 65, 67). Increases in viral load are accompanied by reductions in the levels of peripheral

522 blood CD4+ T cells that are rarely completely reversed without treatment (68). Early T cell

523 depletion is particularly significant in the gut, where mucosal T cells serve as the initial target of

524 infection following vaginal or rectal transmission. As these T cells are responsible for

525 maintaining gut homeostasis, their loss results in enteropathy, bacterial translocation, and

526 inflammation, which recruits more T cells to the site of damage (Reviewed in 69). This cycle of

527 infection and inflammation continues to provide HIV with new targets to infect, permitting rapid

528 viral expansion and establishing a chronic state of immune activation. Unsurprisingly, primary

529 infection is associated with innate immune activation, characterized by an intense cytokine

530 response and activation of NK and NKT cells (70-72). HIV-specific cytotoxic lymphocyte (CTL)

531 responses can also be observed, with the first CD8+ T cell responses arising during peak viremia

532 and peaking 1-2 weeks later, as viremia declines (29, 73, 74). Anti-HIV antibody responses arise

533 more slowly, within approximately 3 months of infection and remain present for years following

534 infection (19, 74-76).

535 The initial viral load spike and the immune responses it induces are followed by a long

536 period of asymptomatic infection, during which time symptoms are mild, if present, and viral

537 load decreases to a set point. However, viral replication continues during this time and peripheral

538 CD4+ T cell numbers steadily decline (77-80). Although the latent stage can last decades, in the

- 12 -

539 absence of treatment, the majority of infected individuals will progress to AIDS with a median

540 time from infection of 10 years. The onset of AIDS is characterized by a drop in CD4+ T cell

541 numbers to below 500 cells/μL (19). This potent immune suppression predisposes infected

542 individuals to opportunistic infections that often prove fatal. Paradoxically, chronic immune

543 activation can also contribute to disease progression and render HIV-infected individuals more

544 vulnerable to inflammatory diseases (81, 82). HAART administration can halt disease

545 progression and reverse CD4+ T cell depletion, however, this treatment cannot eliminate non-

546 replicating viral reservoir cells and must be administered for life to prevent viral rebound. As

547 such, much of the research directed towards prevention has focused on modulating immune

548 responses that occur early in infection and precede the establishment of viral reservoirs,

549 highlighting the importance of innate immune responses.

550 1.3.5: Discovery and function of NK cells

551 NK cells are innate immune lymphocytes that were first identified by their ability to

552 spontaneously kill allogeneic tumor cells in vitro (83, 84). NK cells develop in the bone marrow

553 from the same common lymphoid progenitor that generates mature B and T cells (Reviewed in

554 85). As with all innate immune mediators, NK cells can lyse target cell without prior antigen

555 sensitization and their responses occur much quicker than adaptive immune responses, which

556 earned them the title of ‘natural killers’ (86, 87). The mechanism through which NK cells

557 targeted tumor cells was addressed by the ‘missing-self’ hypothesis, which theorizes that NK

558 cells can recognize self-proteins – specifically, MHC class I molecules – and are induced to kill

559 targets cells where surface expression of these proteins is downregulated (Reviewed in 88). It has

560 subsequently been confirmed that MHC-I molecules are downregulated in tumor cells and that

- 13 -

561 this results in NK cell activation upon interaction (89). MHC-I molecules are also downregulated

562 during viral infections and times of cellular stress. Correspondingly, NK cells are now thought of

563 as the first line of defense against virally-infected and transformed cells.

564 The primary way in which NK cell kill is through the contact-dependent release of

565 cytotoxic granules by a process called degranulation. Upon interaction with a target cell that

566 results in NK cell activation, an immunological synapse will form at the point of contact and

567 secretory lysosomes containing granzyme and perforin mobilize towards it (90-92). Following

568 granule exocytosis, perforin forms pores in the target cell that allow granzyme to enter and

569 cleave its targets, ultimately resulting in target cell death (Reviewed in 93). The rapid, non-

570 specific contact-dependent lysis of target cells is an important innate immune function, but NK

571 cell can also collaborate with adaptive immunity. Through their FcγRIIIA (CD16) cell surface

572 receptors that bind to the Fc portion of human IgG, NK cells can detect antibody-coated target

573 cells and mediated antibody-dependent cellular cytotoxicity (ADCC) (94). CD16

574 heterodimerizes with FcϵRIγ chains or CD3-ζ chains, whose cytoplasmic tails contain immune

575 tyrosine-based activating motifs (ITAM) (95). Ligation of CD16 triggers the phosphorylation of

576 these ITAM motifs and, subsequently, NK cell cytotoxicity (96). Finally, NK cells are also an

577 important cellular source of interferon-γ (IFN-γ) and can also produce and secrete several other

578 chemokines and cytokines (2, 97). Through the production of these signaling molecules and

579 direct interactions, NK cells can recruit other cell types to sites of inflammation and infection

580 and shape T and B cell responses (98-100). Because of their functional plasticity, NK cells serve

581 as a unique bridge between innate and adaptive immunity.

582 1.3.6: NK cells in HIV

- 14 -

583 NK responses are an important component of immunity against many viruses, including

584 HIV. Certain combinations of killer Ig-like receptors (KIR) on NK cells and their human MHC-I

585 or HLA ligands have been associated with protection against HIV infection and slower disease

586 progression (101-104). The HIV-derived peptide motifs presented by those HLA molecules may

587 also contribute to protection, as HLA/KIR interactions with specific viral peptides modulates NK

588 cell responses in vitro (105). Much like tumor cells, the cell surface expression of MHC-I

589 molecules is reduced on HIV-infected cells. Mediated by the viral accessory protein Nef, HLA

590 downregulation protects infected cells from CTL responses, but makes them targets for NK cell

591 missing-self responses (106, 107). However, HIV has evolved to evade such responses by

592 selectively downregulating HLA molecules. HLA-C and -E expression is maintained on the

593 surface of infected cells, preserving interactions with inhibitory NK cell receptors (iNKRs) and

594 abrogating NK cell anti-HIV responses (108, 109).

595 Additionally, secondary analyses of HIV vaccine trials have highlighted the NK cell

596 mediated-ADCC as an important correlate of vaccine-induced protection (7). ADCC activity has

597 also been associated with improved viral control in infected individuals (110, 111). More

598 recently, it has been proposed that NK cells are capable of memory-like responses and that

599 memory NK cells maintain prolonged anti-viral responses in a simian immunodeficiency virus

600 (SIV) model. Specifically, NK cells from macaques infected with SIV or a chimeric SIV with an

601 HIV envelope were observed to be capable of antigen-specific lysis of target cells that had been

602 pulsed with HIV protein components (112). Furthermore, NK cells from animals vaccinated

603 against adenovirus 26-vectored SIV elicited vaccine antigen-specific responses 5 years after

604 vaccination (112). While the existence of memory NK cells has not yet been confirmed in

- 15 -

605 humans, human cytomegalovirus (CMV) infection induces the preferential expansion of

606 CD94/NKG2C-expressing NK cells, which can mediate memory-like responses against CMV

607 (113-115).

608 1.4: NK cell licensing and activation

609 Anti-viral NK cell responses and NK cell functions as a whole are governed by the

610 balance of signals transmitted through aNKR and iNKR. In this context, NK cells can be

611 activated one of two ways. First, upon encountering target cells that do not express ligands to

612 iNKR, inhibitory signaling through these receptors will be abrogated, allowing for activation in

613 its absence. Secondly, NK cells interacting with target cells that do express ligands to aNKR will

614 experience an increase in activating signaling through those receptors. Physiologically, however,

615 most cells express ligands to both iNKR and aNKR and whether an NK cell will be activated

616 depends on the number and strength of each signal type. Additionally, NK cells can also be

617 activated by cytokines produced by other immune cells during infection and inflammation. For

618 example, interleukin (IL)-2 can promote NK cell survival, proliferation, and IFN-γ secretion,

619 while IL-12 and IL-18 act synergistically to stimulate NK cell IFN-γ production via promotion of

620 JAK/STAT signaling (116-122). Several other cytokines capable of triggering NK cell

621 cytotoxicity have been identified, but the mechanisms through which they activate NK cells have

622 not been conclusively defined.

623 The strength of mature NK cell responses is also influenced by a process known as

624 education, whereby HLA-KIR interactions during NK cell development dictate tolerance to self

625 and cytotoxicity against non-autologous and virally-infected or transformed cells (123). The

- 16 -

626 importance of HLA-KIR interactions in directing NK cell functional competency was first

627 suggested by the observation that NK cells developing in MHC-devoid environments lacked the

628 characteristic ability to lyse MHC-null cells (124, 125). KIRs comprise an important subset of

629 NKRs and recognize subsets of HLA molecules on the surface of putative target cells (126, 127).

630 The inhibitory KIRs (iKIRs) that govern NK cell education have immunoreceptor tyrosine-based

631 inhibition motifs (ITIMs) in their long cytoplasmic tails and are categorized by their number of

632 extracellular Ig-like domains (2D or 3D) (127). NK cells lacking iKIRs for self-HLA molecules

633 are hyporesponsive following in vitro stimulation and fail to lyse MHC-deficient bone marrow

634 transplants and mutations in KIR ITIMs confer hyporesponsiveness, even when those KIRs are

635 in the presence of their cognate ligands (123, 128, 129). Three different models for NK cell

636 education have been proposed. The first ‘arming’ model hypothesizes that NK cells are

637 constitutively unresponsive and uneducated and that iKIR engagement by their HLA ligands

638 during development confers functional competence (123). In contrast, a second ‘disarming’

639 model postulates that NK cells are constitutively and chronically responsive and will become

640 anergic in the absence of iKIR/HLA interactions (130). A third ‘rheostat’ model proposes that

641 education tunes NK cell responsiveness according to the number of inhibitory self-HLA

642 receptors on NK cells and the affinity of each receptor for their cognate ligand (131, 132).

643 According to this model, NK cells expressing two or more iKIRs would be more functional than

644 NK cells expressing only one. This functional enhancement could improve NK cell licensing

645 (arming model) or oppose chronic activation and anergy (disarming model).

646 1.4.2: Models for the study of NK cell activation

- 17 -

647 HLA-null cells are commonly used to study NK cell activation. HLA-null cells lack the

648 surface expression and, on occasion, the mRNA transcripts of the MHC-I molecules HLA-A, -B,

649 and -C. NK cell that interact with HLA-null cells will experience abrogation of inhibitory

650 signaling through the iKIRs that bind HLA-A, -B, and -C antigens. HLA-null cells do, however,

651 express ligands to aNKRs and activating signaling through these receptors, in tandem with the

652 suppression of inhibition through iKIRs, will activate NK cells and stimulate missing-self

653 responses. The two cell lines utilized by our lab for these purposes are the K562 and 721.221

654 (721) cell lines. The K562 cell line was first derived from a patient with chronic myelogenous

655 leukemia (CML) in blast crisis (133, 134). Phenotypically, K562 resemble non-adherent myeloid

656 progenitor cells and can spontaneously develop erythrocyte-, granulocyte-, and monocyte-like

657 characteristics (135). The karyotype of this cell line appears to vary according to its time in

658 culture, increasing from hypodiploid to near triploid in longer cultures (136-138). 721 is a B cell

659 lymphoblastoid cell line that was transformed and immortalized by Epstein-Barr virus (EBV)

660 infection and thus expresses the EBV genome (139-141). 721 are characterized by the complete

661 absence of class I HLA mRNA transcripts and α-chains as a result of gamma ray-induced

662 mutation in the HLA complex, but this cell line does express endogenous non-classical HLA-E

663 (139, 142, 143). While both cell lines lack HLA-A, -B, and -C antigens, we observed that they

664 stimulated NK cells differently and this likely reflects differences in the expression of ligands to

665 aNKR (4).

666 NK cells are activated during the course of viral infections and autologous HIV-infected

667 CD4+ T cells (iCD4) can also be used to activate NK cells in vitro. As has been mentioned,

668 downregulation of HLA-A and -B on the surface of iCD4, although this is impaired somewhat by

- 18 -

669 maintained expression of HLA-C, is partially responsible for activating NK cells and triggering

670 missing-self responses. Furthermore, HIV infection can also modulate the expression of ligands

671 to aNKRs and upregulation of those ligands on iCD4 likely also contributes to their ability to

672 activate NK cells (144-146). Following their contact-dependent activation, NK cells inhibit

673 further viral replication via the production of chemokines (5, 147). Activated NK cells can

674 secrete CC-chemokines CCL3, CCL4, and CCL5, which compete with HIV for binding of the

675 CCR5 co-receptor on CD4+ T cells and block viral entry (148, 149). However, NK cell-mediated

676 inhibition of HIV replication is incompletely understood and other NK cell functions may

677 contribute to this process.

678 1.5: Characterization of activating NK cell receptors (aNKRs) and their ligands

679 1.5.2: NKG2 receptors

680 One of the major aNKR is C-type lectin-like receptor family natural-killer group 2

681 member D (NKG2D), which is expressed on all NK cells and CD8+ T cells (150-152). This type

682 II transmembrane protein is encoded by a gene in the NKG2 complex on human chromosome 12

683 and contains no known signaling motif in its intracellular domain (153-155). Rather, this

684 receptor homodimerizes with the adaptor protein DNAX-activating protein of 10 kDa (DAP10)

685 for signal transduction and stability (152, 154, 156). Upon activation of the NKG2D and DAP10

686 homodimer, DAP10, which possesses a YXXM tyrosine-based motif, will recruit

687 phosphoinositide kinase-3 (PI3K) and growth factor receptor-bound protein 2 (Grb2) (152, 157).

688 In humans, NKG2D exclusively interacts with DAP10, however, a shorter isoform of NKG2D in

- 19 -

689 mice is also capable of binding to DNAX-activating protein of 12 kDa (DAP12) (158, 159). The

690 signaling pathways of NKG2D and other NKG2 receptors are illustrated in Figure 3.

691 Figure 3

692 Figure 3: NKG2 receptor signaling. Ligation of NKG2D and CD94/NKG2C, which associate with the adaptors 693 DAP10 and DAP12, respectively, results in NK cell activation. Interaction between HLA-E and CD94/NKG2A 694 results in inhibition of NK cell responses (see text).

695 Image source: Iwaszko 2012 (160)

696 NKG2D is notable in that several distinct MHC class-I-like ligands can bind to this single

697 invariant receptor. In humans, NKG2D recognizes members of the retinoic acid early transcript 1

698 (RAET1) or UL16-binding protein family (ULBP1-6; named for their ability to bind the UL16

699 glycoprotein of CMV) and the MHC class-I-chain-related proteins (MIC) A and B (161-165).

700 ULBP1-3 are glycosylphosphatidylinositol (GPI)-linked glycoproteins, while ULBP-4 and -5 are

701 transmembrane glycoproteins (163, 166). Like classical MHC-I molecules, MIC-A and -B are

702 highly polymorphic and have a structure that includes three external domains (α1-3), a

703 transmembrane domain, and a cytoplasmic domain (165, 167). Similarly, the Ig-like domains of

704 the ULBPs are comparable to the α1 and α2 domains of MHC-I molecules (168). The structure

705 of the NKG2D ligands is illustrated in Figure 4. However, ULBP and MIC proteins distinguish

- 20 -

706 themselves from their MHC-I counterparts in their inability to bind β2-microglobulin and

707 associate with transporter-associated proteins, making these proteins unable to bind peptide and

708 present antigen (169-172). Both ULBP and MIC proteins are classified as stress ligands, as they

709 are generally not expressed on healthy cells, but are upregulated by cellular stressors such as

710 viral and bacterial infection and cellular transformation (173-180). Engagement of NKG2D by

711 its cognate ligands on target cells triggers NK cell cytotoxicity through the regulation of NK cell

712 granule polarization and degranulation by NKG2D/DAP10 signaling (181). Furthermore,

713 signaling through NKG2D is potent enough to trigger NK cell activation despite concurrent

714 inhibitory self-recognition signaling (162, 182).

715 Figure 4

716 Figure 4: NKG2D ligands. The two families of NKG2D ligands, MIC and ULBP, have characteristic structures (see 717 text). TM+CYT = transmembrane and cytoplasmic domains. GPI = glycosyl-phosphatidyl-inositol anchor.

718 Image source: Fernández-Messina 2012 (183)

719 In addition to NKG2D, other members of the C-type lectin-like NKG2 family can

720 contribute to NK cell activation, including NKG2C, NKG2E, and NKG2H. These three NKG2

721 receptors bind to the non-classical MHC class Ib HLA-E (184, 185). Binding of HLA-E to a set

722 of nonamer peptides derived from the leader sequences of HLA class I molecules or HLA-G

723 allows for the stable expression of this ligand on the cell surface of a broad distribution of tissues

- 21 -

724 (186, 187). Through the binding of these leader peptides, HLA-E also functions to indirectly

725 monitor the expression of HLA class I molecules and can also be used by NK cells to monitor

726 cellular integrity through its binding to virally-, bacterially-, and stress-derived peptides (188-

727 191).

728 NKG2C, NKG2E, and NKG2H form heterodimers with the invariant CD94 glycoprotein,

729 which is disulfide-linked to an NKG2 glycoprotein (192, 193). NKG2C also noncovalently

730 associates with DAP12, whose presence is required for efficient cell surface expression of

731 CD94/NKG2C (194-196). NK cell activation upon ligation of DAP12 results in the

732 phosphorylation of its single cytoplasmic immunoreceptor tyrosine base activating motif (ITAM)

733 and ensuing recruitment of the tyrosine kinases spleen tyrosine kinase (Syk) and zeta-chain-

734 associated protein kinase 70 (ZAP-70) (194, 196, 197). NKG2C expression is associated with

735 terminal differentiation of NK cells and improved cytotoxicity (198). NKG2C-expressing NK

736 cells may be particularly important in the context of CMV infection, where this subset of NK

737 cells expands, enhances IFN-γ production, and acquires a mature phenotype (113, 115, 199,

738 200). Furthermore, NKG2C deletions have been associated with increased risk for HIV infection

739 and faster progression to AIDS and elevated frequencies of NKG2C+ NK cells are also observed

740 during HIV infection (114, 201, 202). However, considering that coincident HIV and CMV

741 infections are common and the interconnectedness of HIV progression and CMV reactivation, it

742 is difficult to conclude whether these findings are due to HIV infection alone or underlying CMV

743 infection (203-205). Whereas NKG2C is expressed on the surface of most NK cells, recent work

744 has shown that human NKG2E is retained in the cell cytoplasm where it forms intracellular

745 complexes with DAP12 and CD94 (206). In contrast, NKG2H, a splice variant of NKG2E, is

- 22 -

746 transported to the cell membrane in conjunction with CD94 and DAP12 (207). The contribution

747 of NKG2E and NKG2H signaling to NK-mediated anti-viral responses is currently unknown.

748 CD94/NKG2C and NKG2E share extensive homology with the inhibitory

749 CD94/NKG2A, to which HLA-E also binds and with a greater affinity (208, 209). The

750 cytoplasmic domain of NKG2A contains an ITIM that is phosphorylated upon engagement by

751 HLA-E and subsequently recruits Src homology region 2 domain-containing phosphatase (SHP)-

752 1 and SHP-2 to inhibit NK cell functions (210). Furthermore, co-stimulating NKG2A and

753 NKG2C when they are co-expressed on NK cells results in net inhibition (211). As such, it is

754 plausible that NKG2A signaling regulates NK cell stimulation by other activating NKG2 to limit

755 potential autoreactivity against self-HLA-E.

756 1.5.3: Natural cytotoxicity receptors (NCRs)

757 NCRs comprise the other major family of aNKRs and include the type I transmembrane

758 proteins NKp30, NKp44, and NKp46 (212-214). NCRs were first identified by their shared

759 ability to stimulate NK cell-mediated tumor cell lysis in vitro, although they share limited amino

760 acid or structural homology (214-217). Some similarities include the possession of one or two

761 extracellular ligand binding Ig-like domains and a transmembrane domain that can interact with

762 ITAM-containing signaling adaptor molecules (summarized in Figure 5).

763 NKp30 is encoded by the NKP30 gene, which is located in the class III region of the

764 human MHC locus on chromosome 6, and is expressed on all mature NK cells (214). NKp30 has

765 one extracellular domain that can recognize the tumor cell ligand B7-H6, the tegument protein

766 pp65 of human CMV, and the viral hemagglutinin (HA) of the orthopox family members

- 23 -

767 vaccinia virus and ectromelia virus (218-221). NKp30 can also bind heparin sulfate

768 proteoglycans on the surface of target cells, although the exact identity of its cellular ligand is

769 unknown (222-224). The molecule has an intracellular domain that lacks any signaling domain

770 and instead activates NK cells through the CD3ζ homodimers and CD3ζ/FcRγ heterodimers that

771 interact with the positively charged arginine residue in its transmembrane domain (218, 225).

772 FcRγ and CD3ζ have one and three intracellular ITAMs, respectively, which are phosphorylated

773 upon ligand binding and recruit Syk and ZAP-70 (226, 227). Ligation of NKp30 stimulates NK

774 cell cytotoxicity, degranulation, and IFN-γ production (228, 229). Interestingly, NKp30 has an

775 immunosuppressive splice variant known as NKp30c and signaling through this variant impairs

776 NK cell degranulation and TNF-α secretion, while stimulating IL-10 secretion (229).

777 NKp44 is also encoded in the class III region of the human MHC locus and possesses a

778 single V-type extracellular ligand-binding domain (217, 230-232). The transmembrane domain

779 of NKp44 contains a positively charged lysine residue and associates with and signals through

780 dimerized DAP12 (224, 233). NKp44 is unique among NCRs as it possesses an ITIM within its

781 cytoplasmic tail, although this ITIM is believed to be non-functional (231, 234). As with NKp30,

782 NKp44 has been shown to bind to heparin sulfate proteoglycans and a variety of tumor, viral,

783 and bacterial ligands (223, 235-238). Recently, proliferating cell nuclear antigen and NKp44L,

784 which shares sequence similarity with the mixed-lineage leukemia protein 5, have been

785 identified as cellular ligands for NKp44 (239, 240). However, these ligands are only expressed

786 intra-cellularly and cell surface ligands to NKp44 have yet to be identified (239, 241, 242).

787 NKp46 was the first NCR identified and is encoded by the NKP46 gene on chromosome

788 19 (213, 224). NKp46 is expressed on all NK cells, regardless of their activation state, and is also

- 24 -

789 expressed by some subsets of T cells and innate lymphoid cells (ILCs) (243-245). NKp44 has

790 two extracellular C2-type Ig domains that are connected by a hinge region, a single

791 transmembrane domain, and a cytoplasmic domain that lacks any signaling motifs (224, 246,

792 247). As with NKp30, the arginine residue in NKp46’s transmembrane domain interacts with the

793 FcRγ and CD3ζ adaptor proteins, which provide the signaling required for NK cell activation

794 (224). Additionally, homodimerization of NKp46, which would bring the transmembrane and

795 intracellular domains into close proximity, appears to be required for NK cell activation (248).

796 NKp46 can bind to the HA of influenza and certain poxviruses, as well as the hemagglutinin

797 neuraminidase of the Sendai virus and Newcastle disease virus (219, 220, 249, 250). NKp46 can

798 also bind to vimentin, a cellular ligand expressed on mycobacterium tuberculosis-infected cells,

799 targeting vimentin-expressing cells for NK cell lysis (251). As with all the NCRs, NKp46 can

800 bind to heparin sulfate proteoglycans, but more work will be required to determine which

801 specific cellular ligands that NKp46 and the other NCRs can engage (222, 223).

802 Figure 5

803 Figure 5: Structure of natural cytotoxicity receptors. Natural cytotoxicity receptors contain extracellular ligand- 804 binding domains and transmembrane domains that can interact with different adaptor molecules (see text).

805 Image source: Kruse 2014 (224)

- 25 -

806 1.5.4: Additional activating receptors

807 2B4

808 2B4, a member of the signaling lymphocyte activation molecule (SLAM) family of

809 receptors, is an NK receptor that can activate NK cells directly or provide co-stimulatory signals

810 that enhance NCR-, NKG2D-, and DNAM-1-mediated NK cell activation (252, 253). 2B4 is

811 expressed by all NK cells and some T cell subsets and binds to CD48, which is itself a SLAM

812 family receptor (254, 255). CD48 is broadly expressed on lymphoid and myeloid-derived cells,

813 where several viral infections can modulate its expression (144, 256-258). In humans,

814 stimulation of 2B4-expressing NK cells with CD48-expressing target cells induces NK cell

815 activation, cytotoxicity, and IFN-γ secretion (255, 259, 260). Upon ligation, 2B4 can also

816 transmit inhibitory signals and is thought to collaborate with MHC class I receptors to limit

817 autoreactive NK cell responses (254). The complexity of 2B4 signaling is due in part to the four

818 immunoreceptor tyrosine-based switch motifs (ITSM) in its intracellular domain. Following 2B4

819 engagement, 2B4 will be recruited into lipid rafts and its ITSM motifs will be phosphorylated

820 and recruit SLAM-associated protein (SAP) (261, 262). SAP inhibits the interactions between

821 2B4’s fourth ITSM and the negative regulatory molecules SHP-1, SHP-2, SH2 domain-

822 containing inositol 5'-phosphatase (SHIP), and C-terminal Src kinase (CSK), suppressing

823 inhibitory signaling (261, 263). Correspondingly, NK cells from patients with X-linked

824 lymphoproliferative disease – who lack a functional SAP protein – are inhibited upon 2B4

825 ligation (264-266). It has thus been proposed that the duality of 2B4 signaling is regulated by the

826 availability of SAP in the NK cell cytoplasm (267). The density of CD48 on target cells and

827 expression levels of 2B4, which is internalized and downregulated following ligation, are also

- 26 -

828 thought to influence the balance of inhibitory and activating 2B4 functions (267, 268).

829 Furthermore, 2B4 association with lipid rafts is essential for ITSM phosphorylation and 2B4-

830 mediated NK cell activation. Co-engagement of 2B4 and iNKR, such as KIRs and

831 CD94/NKG2A, interferes with lipid raft recruitment and phosphorylation of 2B4 and provides

832 another level of regulation (262, 269).

833 DNAX accessory molecule-1 (DNAM-1)

834 The leukocyte adhesion DNAM-1 (also known as CD226) is a transmembrane

835 glycoprotein, part of the Ig superfamily, and an aNKR (270). This receptor is encoded on

836 chromosome 18 and features two extracellular Ig-like domains and a cytoplasmic tail with three

837 tyrosine residues (271). DNAM-1 is expressed on monocytes, DCs, T cells, and virtually all

838 resting NK cells, where its engagement results in in Fyn-mediated phosphorylation of the

839 tyrosine residues and induction of NK cell cytotoxicity (270, 271). DNAM-1 can bind to both the

840 δ and α isoforms of CD112 (also known as Nectin-2), which differ from each other in their

841 transmembrane and cytoplasmic regions, and the nectin-like family member CD155 (also known

842 as the poliovirus receptor). However, binding to CD155 appears to predominate DNAM-1’s

843 interactions with its cognate ligands (271). The expression of both cellular ligands for DNAM-1

844 can be upregulated by cellular stress and are modulated by pathological conditions, including

845 cancer and infectious disease (272, 273). Interactions between DNAM-1 and its ligands regulate

846 NK cell adhesion and the induction phase of activation and cytolytic activity and contribute to

847 tumor immune surveillance (270, 271, 274, 275). DNAM-1 signaling is also a promoter of NK

848 cell cytokine secretion during inflammation, the induction of which can be reduced by

849 competition with other receptors for CD155 (276). The expression of CD112 and CD155 on DCs

- 27 -

850 has also been shown to be an important regulator of a selection process known as DC editing,

851 whereby NK cells can kill immature DCs after interacting with them (277-279). Interactions

852 between DNAM-1 and CD155, which is upregulated on T cells following activation of the T cell

853 receptor, may also be involved the NK cell-mediated elimination of activated T cells (280).

854 DNAM-1 signaling has also been implicated in the differentiation and expansion of the still

855 controversial memory NK cells during murine CMV infection and loss of DNAM-1 is associated

856 with impaired viral control and prolonged virus clearance (281, 282). Furthermore, cell surface

857 expression of CD112 and CD155 can be modulated by several viral proteins, impairing anti-viral

858 NK cell responses, and facilitating immune evasion (283-285).

859 Lymphocyte function-associated antigen-1 (LFA-1)

860 Interestingly, efficient DNAM-1 function requires the co-expression of the αLβ2 integrin

861 LFA-1, which is expressed on all NK cells (286). In NK cells isolated from patients who lack

862 LFA-1, DNAM- signaling is defective, but can be restored through genetic reconstitution of

863 LFA-1 expression (286). LFA-1 binds to intercellular adhesion molecule (ICAM)-1 and ICAM-

864 2, interactions that are critical for forming the immunological synapse and tight adhesion of NK

865 and target cells (287-292). Engagement of LFA-1 by ICAM-1 and -2 can also induce the

866 polarization of cytolytic granules in NK cells, which is a crucial early step in triggering contact-

867 dependent cytolysis (293-295). LFA-1-mediated NK cell activation occurs downstream of the

868 rapid phosphorylation of vav guanine nucleotide exchange factor-1 (Vav1) and the kinase protein

869 tyrosine kinase 2-β (Pyk2) and reorganization of the actin cytoskeleton (296-298). LFA-1

870 signaling and the ability of NK cells to form synapses with target cells requires the stable

- 28 -

871 tethering of ICAMs to α-actinin and to the ezrin/radixin/moesin (ERM) protein ezrin in the actin

872 cytoskeleton (289, 299-302).

873 CD28

874 Similarly, interaction with target cells expressing the B7 molecules CD80 and CD86 can

875 also trigger granule polarization and cytotoxicity in NK cells (303-306). The contribution of

876 CD80 and CD86 to T cell co-stimulation through their interactions with CD28 and cytotoxic T-

877 lymphocyte-associated protein 4 (CTLA-4) is well established (Reviewed in 307). Nevertheless,

878 it was believed that the majority of human peripheral blood NK cells (pNKs) did not express

879 CD28 and that interaction of CD80 and CD86 with NK cells would be independent of this

880 receptor (308). Furthermore, murine studies reported that the NK-mediated cytotoxicity induced

881 by stimulation with CD80- and CD86-expressing target cells could not be abrogated by blocking

882 CTLA-4 or CD28 (305, 306). However, human NK cells do express a variant of the T cell CD28

883 receptor, although expression levels are generally low and vary among individuals (303, 309).

884 Moreover, blocking of CD28 on human pNKs was able to completely eliminate NK cell-

885 mediated killing of target cells that expressed CD80 and CD86 (303). Despite these findings, the

886 nature of the mechanisms that underlie CD80- and CD86-induced NK cell activation remain

887 incompletely understood.

888 KIR3DS1

889 Although ITIM-containing KIRs are perhaps the most important family of iNKRs, a

890 smaller number contain ITAM motifs and can transmit activating signals upon ligation. One such

891 aNKR is encoded by the conserved allelic variant KIR3DS1, which shares its locus with alleles

- 29 -

892 encoding inhibitory KIR3DL1 allotypes (310). As an activating KIR, KIR3DS1 (3DS1) has a

893 characteristic short ITAM-containing cytoplasmic tail and transmembrane domain that associates

894 with DAP12 (310, 311). Signaling through 3DS1 induces NK cell cytotoxicity and IFN-γ

895 secretion (311). Carriage of 3DS1 has been associated with the acquisition risk and disease

896 outcome of a number of viral infections and most prominently with delayed disease progression

897 in HIV infected patients (101, 312-314). The Bernard laboratory reported that carriage of 3DS1

898 in its homozygous form was associated with protection from HIV infection in HIV exposed

899 seronegative subjects (104). Despite the extensive shared homology of the extra-cellular domains

900 of 3DS1 and KIR3DL1 (3DL1), while 3DL1 has been conclusively shown to bind to a subset of

901 HLA-A and -B molecules that contain a Bw4 motifs, attempts at identifying ligands to 3DS1

902 have been less successful (310, 315-317). Recently, however, the non-classical MHC class I

903 molecule HLA-F was identified as a high affinity ligand to 3DS1 (8, 9). Specifically, the open

904 conformer form of HLA-F, in which HLA class I heavy chains are not bound to β2m or peptide,

905 binds to 3DS1 on NK cells and triggers degranulation and production of pro-inflammatory

906 cytokines (8, 318). The cell surface expression of HLA-F is tightly regulated (319). HLA-F is

907 expressed at the surface of some tumors and B cell lines, but rarely in normal cells, where it is

908 thought to be retained in the endoplasmic reticulum (320-323). Although HLA-F is not expressed

909 on the surface of resting lymphocytes, its expression can be induced by activation (324).

910 Furthermore, HIV infection reduces the expression of HLA-F on previously activated T cells,

911 particularly on late stage infected CD4 T cells (8). Together these findings demonstrate that

912 HLA-F expression can be modulated during the course of inflammatory responses and infection.

- 30 -

913

914

915 Previous work from our lab that assessed the number of NK cells and NK cell functional

916 subsets activated by the two HLA-null cell lines K562 and 721.221 (721) found that K562 and 721

917 differed significantly in their capacities as NK cell stimuli. Specifically, we observed that 721

918 activated a larger fraction of NK cells and induced significantly greater numbers of cytokine and

919 chemokine secreting NK cells, as measured by IFN-γ and CCL4 production. We also observed

920 that, while K562 induced lower frequencies of all functional subsets relative to 721, they

921 preferentially induced degranulation, which was measured by CD107α expression. HLA-null cells

922 do not express the MHC-I human leukocyte antigens (HLA)-A, -B, and -C, which are ligands to

923 the inhibitory killer immunoglobulin-like receptors on NK cells, but do express ligands to

924 activating NK cell receptors. We were interested in studying whether, K562 and 721 had different

925 expression profiles of ligands to activating NK cell receptors and whether this might explain their

926 different abilities to stimulate NK cells.

927

928

- 31 -

929 Chapter 2: Manuscript 1 - The HLA-null cell lines K562 and 721.221 express different

930 ligand profiles for activating NK cell receptors.

931

932

933

934

935

936

937

938

939

940

941

942

- 32 -

943 The HLA-null cell lines K562 and 721.221 express different ligand profiles for activating NK 944 cell receptors. 945 946 947 948 949 Alexandra Tremblay-McLean1,2, Julie Bruneau3,4, Bertrand Lebouché1,5,6, and Nicole F. 950 Bernard1,2,6,7 951 952 953 1Research Institute of the McGill University Health Center, Montréal, Québec, Canada 954 2Division of Experimental Medicine, McGill University, Montréal, Québec, Canada 955 3Department of Family Medicine, Université de Montréal, Montréal, Québec, Canada 956 4Centre de Recherche de Centre Hospitalier de l’Université de Montréal, Montréal, Québec, 957 Canada 958 5Department of Family Medicine, McGill University, Montréal, Québec, Canada 959 6Chronic Viral Illness Service, McGill University Health Centre, Montréal, Québec, Canada 960 7Division of Clinical , McGill University Health Centre, Montréal, Québec, Canada 961 962 963 964 965 966 967 Mailing address: Dr. Nicole F. Bernard, Research Institute of the McGill University Health 968 Centre, Glen Site, 1001 Décarie Boulevard, Block E, Rm E03.3380, Montréal, Québec, Canada, 969 H4A 3J1. 970 Tel: (514) 934-1934 ext:42697 971 E-mail: [email protected] 972 973 974 Funding: This study received support from the Canadian Institutes for HIV Research grant #

975 MOP-142494. A. Tremblay-McLean was supported by Canadian Association for HIV Research

976 Abbvie Master’s Award in Basic Science.

977 978 Word Count: 4212

- 33 -

979 2.1: Author contributions and acknowledgments

980

981 ATM and NFB were responsible for study design, data analysis, and manuscript preparation. JB

982 and BL provided subject samples.

983

984 We would like to acknowledge the generosity of Dr. Galit Alter, who provided our 721 cell line,

985 and the assistance of several lab members. In particular, Xiaoyan Ni and Tsoarello Mabanga for

986 helping to maintain the HLA-null cell lines and Drs. Irene Lisovsky and Gamze Isitman for

987 helping with the development of the multi-parametric flow cytometry panels.

988

989

990

991

992

993

- 34 -

994 2.2: Abstract

995 Natural Killer (NK) cells mediate innate immune responses to stressed, virally-infected,

996 and transformed cells. NK cell activation depends on the integration of signals received through

997 inhibitory (iNKR) and activating NK receptors (aNKR). HLA-null cells can be used in vitro to

998 stimulate and study NK cell activation. Previous work by our group comparing NK cell

999 stimulation by the HLA-null cell lines K562 and 721.221 (721) found that they stimulated

1000 different frequencies of NK cell functional subsets. As both these cell lines lack surface

1001 expression of classical MHC-1 antigens, a subset of which are ligands to iNKR, differences in

1002 NK function following K562 and 721 stimulation may reflect differences in the expression of

1003 ligands for aNKR. Using multiparametric flow cytometry we analyzed the expression of the

1004 following aNKR ligands on these HLA-null cell lines: ULBP-1, ULBP-2/5/6, ULBP-3, MIC-A,

1005 MIC-B, CD48, CD80, CD86, CD112, CD155, ICAM-1, ICAM-2, HLA-F, HLA-E, and the

1006 ligands to NKp30, NKp44, NKp46, and KIR3DS1. Expression was measured as the frequency of

1007 cells expressing each ligand and as an expression score, which reflected the frequency of positive

1008 cells multiplied by the mean fluorescence intensity of staining for each ligand. We found that,

1009 both cell lines expressed ICAM-1, ICAM-2, and HLA-F. K562 exclusively expressed ULBP-

1010 2/5/6, ULBP-3, CD112, CD155, and the ligand to NKp30, while 721 expressed characteristically

1011 high levels of MIC-B, CD48, CD80, CD86, HLA-E, and the ligand to NKp44. The ligands

1012 expressed on 721 can engage a larger number of aNKR on NK cells, which may, in part, explain

1013 their ability to activate a larger fraction of these cells and elicit different functions, compared to

1014 K562.

- 35 -

1015 2.3: Introduction

1016 Natural Killer (NK) cells are a subset of lymphocytes that direct innate immune

1017 responses to kill stressed, virally-infected, and transformed cells (Reviewed in 1). NK cells can

1018 interact with target cells and lyse them directly via the release of cytotoxic granules containing

1019 perforin and granzyme (2, 3). Activated NK cells can also secrete a broad range of cytokines and

1020 chemokines that can activate adaptive immune cells to lyse target cells, bridging the innate and

1021 adaptive immune systems (4-6). Regardless of the method of lysis, an NK cell must first be

1022 activated to elicit a response. The activation state of an NK cell results from the integration of

1023 different signals transmitted through its activating (aNKR) and inhibitory (iNKR) receptors (6,

1024 7). Activation can result from the loss of inhibitory signaling, when there are no ligands for

1025 iNKR to engage, in conjunction with sustained aNKR signaling, or from the engagement of

1026 aNKR by their ligands that overwhelms inhibitory signaling through iNKR (1, 6, 8, 9). However,

1027 in vivo, NK cells interacting with target cells will receive a variety of signals through both

1028 receptor types and whether this results in activation or not depends on the number and strength of

1029 each type of signal transmitted.

1030 A common method to activate NK cells is by co-culturing them with HLA-null cells.

1031 The most frequently used HLA-null cells include the myelogenous leukemia K562 and B-

1032 lymphoblastoid 721.221 (721) cells lines, which do not express the major histocompatibility

1033 complex class I (MHC-I) human leukocyte antigens (HLA) -A, -B, or -C antigens on their

1034 surface (10-12). As these cells are incapable of engaging the inhibitory killer immunoglobulin-

1035 like receptors (KIR) on NK cells, which recognize subsets of HLA-A, -B, and -C, inhibitory

1036 signaling through these receptors is abrogated (1, 6). As signals from these inhibitory receptors

- 36 -

1037 oppose those from aNKR, their removal allows the engagement of aNKR by their ligands to

1038 activate NK cells (13).

1039 Previous work from our lab demonstrated that the K562 and 721 HLA-null cell lines

1040 stimulated NK cells differentially to secrete the cytokine IFN-γ, the chemokine CCL4, and to

1041 express the degranulation marker CD107a. Specifically, we observed that 721 activated a greater

1042 fraction of NK cells than did K562 and that stimulation with 721 preferentially induced IFN-γ

1043 and CCL4 secretion, while K562 more potently stimulated degranulation (14). In the absence of

1044 the expression of the major ligands for iNKR on the surface of K562 and 721, it is likely that

1045 ligands to aNKR regulate NK cell activation and differences in their expression profiles might

1046 explain the different capacities of these two cell lines to activate NK cells. Indeed, the expression

1047 of some ligands to aNKR was shown to differ on both cell lines, although these differences

1048 remain incompletely characterized (15-20).

1049 There are two major classes of aNKR. The first is the C-type lectin NKG2D receptor that

1050 binds to a family of MHC-I like molecules expressed on healthy cells only after periods of

1051 cellular stress (15, 21). These include the human cytomegalovirus UL-16 binding proteins

1052 (ULBP) and MHC-I related chain (MIC) proteins (17, 22, 23). The second includes the natural

1053 cytotoxicity receptors (NCR) NKp30, NKp44, and NKp46, which can bind to membrane-

1054 associated heparan sulfate glycosaminoglycans and viral hemagglutinin (24-31). Despite these

1055 findings, the cellular ligands to NCRs remain incompletely identified.

1056 In addition to the two major aNKR families, signaling through several other receptors can

1057 contribute to NK cell activation. These include CD244/2B4 and the NK-T-B cell antigen (NTB-

1058 A), which are CD2 family receptors that engage CD48 to trigger NK cell cytotoxicity (32).

- 37 -

1059 Another receptor, expressed on virtually all NK cells, is the leukocyte adhesion molecule DNAX

1060 accessory molecule-1 (DNAM-1) that binds to CD112 (Nectin-2) and CD155 (poliovirus

1061 receptor) ligands (16, 33, 34). Signaling through DNAM-1 can stimulate NK cells when it is co-

1062 expressed with the lymphocyte function-associated antigen 1 (LFA-1). LFA-1 can bind the

1063 integrins ICAM-1 and ICAM-2 on target cells, bridging NK and target cells and forming the

1064 immunological synapse (35-39). Additionally, target cells expressing the T cell co-stimulatory

1065 B7 molecules CD80 and CD86 can stimulate NK cells (40-42). The nature of this signaling is

1066 still poorly understood, but it has been suggested that NK cell activation by CD80 and CD86

1067 depends on a CD28 variant expressed on NK cells (41, 43).

1068 In contrast to HLA-A, -B, and -C, which signal through iNKRs, the non-classical HLA-E

1069 and F can contribute to NK cell activation through the engagement of aNKRs. Much like its

1070 classical MHC-I counterparts, HLA-E interacts with the inhibitory C-type lectin-like receptor

1071 NKG2A, which is heterodimerized with CD94, and with the aNKRs NKG2E and -C (44-47).

1072 HLA-F is a more recent addition to the family of ligands to aNKR and has been shown to

1073 stimulate degranulation and cytokine production through its binding to KIR3DS1 on NK cells

1074 (48, 49).

1075 To determine whether the differences observed in frequency and function of NK cell

1076 subsets stimulated by K562 and 721 are related to differences in their expression profiles of

1077 ligands to aNKRs, we analyzed the expression of a comprehensive panel of aNKR ligands on

1078 both HLA-null cell lines by multi-parametric flow cytometry. As ligands to inhibitory KIR are

1079 not expressed on HLA-null cell lines, it is likely that expression of ligands to aNKRs plays a role

1080 in modulating differential NK cell responses to these cells. This research sought to better

- 38 -

1081 characterize the phenotype of two NK cell stimuli, the HLA-null cell lines K562 and 721, in

1082 order to better understand the mechanisms governing their ability to activate NK cells.

- 39 -

1083 2.4: Methods

1084 2.4.1: Origin and preparation of K562 and 721 cell lines

1085 K562 cells (American Type Culture Collection, Manassas, VA) and 721 cells (a kind gift

1086 from Dr. Galit Alter; Harvard University) were cryopreserved in in 10% dimethyl sulfoxide

1087 (DMSO; Sigma-Aldrich, St. Louis, MO) with 90% fetal bovine serum (FBS; Wisent, Inc. St.

1088 Bruno, QC, Canada) were thawed and cultured in RPMI 1640 medium supplemented with 10%

1089 FBS; 2mM L-glutamine; 100IU/mL penicillin; 100mg/mL streptomycin (R10, all from Wisent)

1090 for at least one full passage before staining. 721 and K562 that were cultured in R10

1091 supplemented with 400IU/mL of recombinant human IL-2 (rhIL-2; Chiron Corp., Emeryville,

1092 CA) for 3 days served as positive controls.

1093 2.4.2: Antibody staining and acquisition

1094 Both HLA-null cell lines were stained within two passages of thawing and in triplicate

1095 with UV Live/Dead ® Fixable Blue cell stain kit, as per manufacturer’s directions

1096 (Thermofisher, Waltham, MA) and then were stained with the following antibody-fluorochrome

1097 conjugates (monoclonal antibody clones) specific for the following cell surface antigens, which

1098 were organized into 4 panels for the purpose of analysis: (Panel 1) ULBP-1-PerCP (170818),

1099 ULBP-2/5/6-APC (165903), ULBP-3-PE (166510), MIC-A-APC (159227), MIC-B-AlexaFluor

1100 700 (236511; all from R&D Systems, Minneapolis, MN), (Panel 2) CD48-PE (BJ40), CD80-

1101 BV421 (2D10), CD86-PE-Dazzle594 (IT2.2; all from BioLegend, San Diego, CA), CD112-

1102 AlexaFluor 700 (610603; R&D), CD155-PE-Cy7 (SKII.4), (Panel 3) ICAM-1-Pacific Blue

1103 (HCD54), and ICAM-2-PE (CBR-1C2/2; all from BioLegend). Cells were also stained with

- 40 -

1104 recombinant human IgG1 Fc chimeras NKp30-Fc, NKp44-Fc, and NKp46-Fc (all from R&D) as

1105 a part of Panel 3. A final HLA Panel comprised the conjugated antibody HLA-E-PE-Cy7 (3D12;

1106 BioLegend), as well as an unconjugated recombinant human IgG1 Fc chimera KIR3DS1-Fc

1107 (R&D) and 3D11, an unconjugated primary antibody against HLA-F (a kind gift from Dr. Daniel

1108 Geraghty, Fred Hutchison Research Institute, Seattle, WA). Briefly, cells were resuspended in a

1109 96-well V bottom plate (Sarstedt, Nümbrecht, Germany) at a concentration of 1x106 cells per

1110 100μL of Dulbecco’s phosphate buffered saline (dPBS; Wisent) and stained with the Live/Dead

1111 reagent. TruStain FcX reagent (BioLegend) was used to minimize non-specific Fc receptor

1112 interactions and cells were stained with conjugated antibodies from Panel 1, 2, 3, and HLA for

1113 30 min in the dark at room temperature (RT). For staining with any of the recombinant human

1114 IgG1 Fc chimeras or 3D11, cells were prepared as previously described, with the exception that

1115 they were stained with protein chimeras or primary antibody on ice for 40 min. After washing,

1116 binding of Fc chimera proteins was detected using a polyclonal anti-human IgG (Fc gamma-

1117 specific)-PE secondary antibody, while 3D11 binding was detected using a polyclonal F(ab’)2

1118 anti-mouse IgG-APC secondary antibody (both from eBioscience) for 20 min on ice. Following

1119 staining, all cells were washed and fixed with 2% paraformaldehyde (PFA; Santa Cruz, Dallas

1120 TX). Between 400,000 and 600,000 events were acquired on an LSRFortessa x20 within 24hrs

1121 (BD). Unstained, single stained controls (CompBead; BD), fluorescence minus one, and

1122 secondary antibody alone controls were used for multi-colour compensation and gating purposes.

1123 Flow cytometric analysis was performed using FlowJo software version 10 (TreeStar, Ashland

1124 OR). All the antibodies used and their manufacturers are listed in Appendix A.

1125 Results are presented as the frequency of cells expressing each aNKR ligand and the

1126 expression score for each of these aNKR ligands. The expression score for a ligand was

- 41 -

1127 calculated by multiplying the frequency and mean fluorescence intensity (MFI) of cells

1128 expressing that ligand. As K562 and 721 have different levels of autofluorescence, the MFI was

1129 first background corrected against the MFI of unstained cells.

1130 2.4.3: Statistical analysis

1131 Statistical analysis was performed using GraphPad Prism version 6 (GraphPad Software,

1132 Inc., La Jolla, CA). Mann-Whitney tests were used to determine the significance of differences in

1133 the frequency or expression score of a cell surface ligand between 2 unpaired groups. Data on the

1134 frequency and expression score of ligand expression is represented as median (range), with those

1135 three values representing each of the triplicate values. P-values less than 0.05 were considered

1136 significant.

- 42 -

1137 2.5: Results

1138 2.5.1: Comparison of the frequencies of K562 and 721 expressing ligands to aNKR

1139 We hypothesized that differences in aNKR ligands expressed by K562 and 721 could

1140 explain their different abilities to stimulate NK cells. We used flow cytometry to determine the

1141 frequency of K562 and 721 cells expressing ligands to aNKR. Figure 1 shows the percentage of

1142 cells expressing ligands detected by antibody panels 1, 2, 3, and HLA. As IL-2 can upregulate

1143 the surface expression of these ligands, HLA-null cells cultured for 3 days in rhIL-2-containing

1144 media served as positive controls for the comparison of ligand expression on unstimulated cells.

1145 For Panel 1 (Fig. 1A), we observed that a median of 64.9(63.1, 66.7)%, 21.3(19.5, 28.4)%, and

1146 24.3(24.2, 28.1)% of K562 cells expressed ULBP-2/5/6, ULBP-3, and MIC-B, respectively. Less

1147 than 10% of these cells expressed the other ligands in panel 1. On the other hand, less than 10%

1148 of 721 expressed the stress ligands in this panel except for MIC-B, which was present on

1149 76.9(66.2, 77.9)% of these cells. Thus, a higher frequency of K562 than 721 cells expressed

1150 ULBP-2/5/6 and ULBP-3, while a lower frequency of K562 than 721 expressed MIC-B. As

1151 shown in Figure 1B, 15.4(11.3, 21.1)%, 88.6(88.2, 88.7)%, 0.4(0.3, 0.5)%, 67.5(66.5, 68.6)%

1152 and 99.1(99.0, 99.2)% of K562 expressed CD48, CD80, CD86, CD112, and CD155,

1153 respectively. Virtually all 721 cells were positive for CD48, CD80, and CD86, while 3.8(3.4,

1154 3.8)% and 6.8(6.0, 9.9)% expressed CD112 and CD155. Therefore, CD48 and CD86 was present

1155 on a lower frequency of K562 than 721, while a higher frequency of K562 than 721 cells

1156 expressed CD112 and CD155.

1157 All K562 and 721 cells expressed ICAM-1 and ICAM-2 (Fig. 1C). We found that

1158 73.0(71.4, 74.5)% and 24.7(20.1, 40.5)% of K562 expressed the ligand to NKp30 and NKp44,

- 43 -

1159 whereas 0.8(0.4, 1.0)% and 41.7(39.5, 42.8)% of 721 expressed these ligands. As such,

1160 compared to 721, a higher frequency of K562 expressed the ligand to NKp30, while a lower

1161 frequency expressed the ligand to NKp44. Neither cell line expressed the ligand to NKp46.

1162 HLA-E was present on 21.1(12.9, 21.7)% of K562 and 42.7(40.4, 48.5)% of 721 cells (Fig. 1D).

1163 HLA-F was detected on 55.6(50.9, 59.0)% and 60.2(47.6, 60.3)% of K562 cells and 80.9(75.1,

1164 85.4)% and 66.8(60.5, 69.6)% of 721 cells stained with 3D11 and the KIR3DS1-Fc chimera

1165 protein, respectively. Thus, both K562 and 721 expressed HLA-E and HLA-F, though there was

1166 a trend towards the frequency of expression of these markers being lower on the former cell line.

1167 In sum, we observed that variable frequencies of K562 cells expressed ULBP-2/5/6, ULBP-3,

1168 MIC-B, CD48, CD80, CD112, CD155, ICAM-1, ICAM-2, the ligands to NKp30 and NKp44,

1169 HLA-E, and HLA-F. In contrast, 721 expressed MIC-B, CD48, CD80, CD86, ICAM-1, ICAM-

1170 2, the ligand to NKp44, HLA-E, and HLA-F. K562, but not 721, expressed ULBP-2/5/6, ULBP-

1171 3, CD112, CD155, and the ligand to NKp30. 721, but not K562, expressed CD86 and a higher

1172 frequency of 721 than K562 cells expressed MIC-B, CD48, CD80, and HLA-E.

1173 2.5.2: Comparison of the expression score for ligands to aNKR on the K562 and 721 HLA-

1174 null cell lines

1175 In addition to determining the frequency with which each HLA null cell line expressed

1176 the aNKR ligands studied, we also examined the intensity with which these ligands were

1177 expressed by measuring the MFI of staining by the antibodies specific for each ligand. We then

1178 multiplied the frequency of cells positive for each ligand by the MFI of ligand staining to obtain

1179 a fluorescence expression score.

- 44 -

1180 The expression score for panel 1 ligands had a pattern similar to that observed for the

1181 frequency of these ligands on K562 and 721 cells. As shown in Figure 2A, the expression score

1182 for ULBP-2/5/6 was 252(240, 258) on K562, compared to 0(0, 0) on 721. Additionally,

1183 compared to 721, ULBP-3 and MIC-B expression was 10.8-fold greater and 2.4-fold lower on

1184 K562 cells. For panel 2 ligands, K562 had expression scores for CD48, CD80, and CD86 that

1185 were almost 300-fold, 2.4-fold, and almost 800-fold lower than those of 721 (Fig. 2B).

1186 Meanwhile, the expression scores of CD112 and CD155 were 65-fold and 122-fold greater on

1187 K562 than on 721. Both cell lines had similar expression scores for ICAM-1, ICAM-2, and the

1188 ligand to NKp46 (Fig. 2C). However, K562 had an expression score for the ligand to NKp30 of

1189 216(203, 234), while 721 did not express this ligand, and expressed the ligand to NKp44 at

1190 levels 2.3-fold less than those observed on 721. Of the HLA ligands studied, only HLA-E was

1191 expressed differentially on K562 and 721, with K562 having an expression score for this ligand

1192 that was 2-fold lower than that of 721 (Fig. 2D. Overall, these findings mirror the trends

1193 observed in frequency of aNKR ligand expression. Namely, that K562 exclusively express the

1194 aNKR ligands ULBP-2/5/6, ULBP-3, CD112, CD155, and the ligand to NKp30, whereas 721

1195 express MIC-B, CD48, CD80, CD86, the ligand to NKp44, and HLA-E at levels greater than

1196 those observed on K562.

- 45 -

1197 2.6: Discussion

1198 A subset of the MHC-I HLA-A, -B, and -C antigens bind to inhibitory KIRs on NK cells.

1199 In the absence of any cell-surface iNKR ligands, differential expression of ligands to aNKR on

1200 HLA-null cell lines plays a more important role in regulating their ability to interact with and

1201 activate NK cells. Previous work from our lab found that the two HLA-null cell lines K562 and

1202 721 activated different frequencies of NK cells exhibiting any permutation of IFN-γ and/or

1203 CCL4 secretion or CD107α expression, as well as several of the functional subsets defined by

1204 these activities (14). A complete characterization of the ligands to aNKR expressed on different

1205 NK cell stimuli is crucial to predict their stimulatory activity and to understanding the

1206 mechanisms that underlie that activation. Here, we performed phenotypic analyses to examine

1207 the expression profiles of ligands to aNKRs on K562 and 721. We observed that while both cell

1208 lines shared expression of some of these ligands, they each possessed a characteristic expression

1209 profile. Although prior studies have partially characterized the expression profiles of ligands to

1210 aNKRs on K562 and 721, in this study, we included a larger and more comprehensive panel of

1211 ligands and, for the first time, compared the ligand profiles on these HLA-null cells at the same

1212 tie and in the same experiments.

1213 We observed that both HLA-null cell lines expressed the adhesion molecules ICAM-1

1214 and ICAM-2 and HLA-F, the ligand for the activating KIR3DS1 receptor on NK cells. This

1215 discovery was unexpected, as recent work that identified HLA-F as the ligand for KIR3DS1 did

1216 not find that K562 expressed HLA-F (48, 49). K562 cells exclusively expressed ULBP-2/5/6,

1217 ULBP-3, CD112, CD155, and the ligand for the activating NCR NKp30. These findings are

1218 consistent with previous reports that ULBP ligands are preferentially expressed on K562 and

- 46 -

1219 with the observation that K562 express the tumor ligand B7-H6, which can bind NKp30 (19, 50).

1220 The stress ligands ULBP-2/5/6 and ULBP-3 bind to the C-type lectin NKG2D and CD112 and

1221 CD155 bind to DNAM-1 (16, 22, 23, 34). As such, K562 has a ligand profile that is consistent

1222 with being able to bind to and activate NK cells through NKG2D, DNAM-1 and NKp30, while

1223 721 are incapable of engaging these aNKRs. NKG2D signaling has been shown to play a crucial

1224 role in NK cell cytotoxic granule polarization, degranulation, and cytotoxicity (51). The ability

1225 of K562 to induce signaling through this pathway may explain why this cell line preferentially

1226 stimulates NK cell degranulation, rather than cytokine or chemokine secretion (14).

1227 Unfortunately, no antibody currently exists that can discriminate between ULBP-2, -5, and -6

1228 and we are incapable of determining which combinations of these ligands are expressed on

1229 K562. However, it is possible that K562 express all three of these ligands, in addition to ULBP-

1230 3, and could, therefore, induce more potent signaling through NKG2D than 721, which express a

1231 single NKG2D ligand (MIC-B). Moreover, expression of NKp30 on NK cells has been

1232 correlated with both perforin expression and degranulation and engagement of this aNKR by

1233 K562 may also have contributed to their induction of degranulation (52).

1234 In contrast, 721 exclusively expressed CD48 and CD86 and expressed characteristically

1235 high levels of MIC-B, CD80, the ligand for NKp44, and HLA-E. Our findings confirm that

1236 HLA-E, the ligand to the aNKRs NKG2E and NKG2C and iNKR NKG2A on NK cells, is

1237 expressed by 721 cells (53). Stable cell-surface expression of HLA-E usually requires binding to

1238 a set of nonamer peptides derived from the leader sequences of HLA class I molecules or HLA-

1239 G, which are absent in the HLA-null 721 (54, 55). However, the HLA-E peptidome was recently

1240 shown to be less restricted that previously thought. HLA-E can also bind to an array of self-

1241 peptides in the absence of HLA class I signal peptides, permitting its stable expression and

- 47 -

1242 induction of NK cell cytotoxicity (56, 57). In addition, HLA-E is also capable of presenting

1243 Epstein-Barr virus (EBV)-derived peptides, like BZLF1, which would be present in the EBV

1244 transformed 721 cell line (58, 59). Yet, HLA-E/BZLF1 complexes are poorly recognized by NK

1245 cells and it is likely that HLA-E molecules presenting noncanonical self-make greater

1246 contributions to 721-induced NK cell activation (58).

1247 Engagement of the aNKR 2B4 by its ligand CD48 on 721 can induce low levels of IFN-γ

1248 production and concurrent signaling through 2B4 and other aNKRs can drive high levels of

1249 cytokine and chemokine production (6, 18). Although, the exact NKR that recognizes the co-

1250 stimulatory molecules CD80 and CD86 has not been identified, expression of these ligands on

1251 target cells can trigger NK cell-mediated cytolysis (41). Furthermore, signaling through NKp44,

1252 which is expressed on activated NK cells, induces the production of cytokines, like IFN-γ (25,

1253 60). Together, the ligands that are expressed on 721 are capable of engaging receptors that are

1254 important for the production of cytokines and chemokine, which may explain why 721 stimulate

1255 larger numbers of IFN-γ and CCL4 secreting NK cells (14). Additionally, the aNKR ligand

1256 expression profile of 721 is associated with binding to NK cells through a larger number of

1257 aNKR than K562. This may contribute to the observation that 721 activate greater numbers of

1258 NK cells, compared to K562 (14).

1259 For the purpose of this study, the ligands analyzed were included on the basis of their

1260 ability to stimulate NK cell responses through the engagement of aNKRs. However, it is

1261 important to consider that several of these ligands are capable of signaling through both aNKRs

1262 and iNKRs. CD112 and CD155, which signal through the activating DNAM-1, can also bind to

1263 the iNKR, T cell immunoreceptor with immunoglobulin and ITIM motifs (TIGIT) (61, 62).

- 48 -

1264 While both DNAM-1 and TIGIT are widely expressed on NK cells, the affinity of CD155 for

1265 TIGIT is greater than for DNAM-1 and TIGIT expression can reduce DNAM-1/CD155

1266 interactions in a dose-dependent manner (62-64). TIGIT has also been shown to compete with

1267 DNAM-1 for the binding of CD112. Furthermore, when transfected into the NK cell line YTS,

1268 TIGIT greatly limits NK-medicated cytotoxicity by disrupting cytotoxic granule polarization (64,

1269 65). Considering this, it is possible that CD112 and CD155, which are exclusively expressed on

1270 K562, contribute more to NK cell inhibition than activation and may be an additional reason why

1271 K562 activate a smaller fraction of NK cells, compared to 721 (14). Another aNKR ligand,

1272 HLA-E, similarly contributes to both NK cell activation and inhibition. HLA-E binds to the

1273 CD94/NKG2 family of NK cell receptors, which includes the activating NKG2E and -C and the

1274 inhibitory NKG2A and -B NKRs (44, 45). Interactions between NKG2A, which is expressed on

1275 the majority of NK cells, and HLA-E have been shown to predominate over interactions with

1276 NKG2C and surface expression of HLA-E is sufficient to save those cells from lysis by

1277 NKG2A+ NK cells (44, 45, 66). Despite this, work assessing NK cell stimulation by autologous

1278 HIV-infected CD4 T cells, which express HLA-E, found that carriage of NKG2A on NK cells

1279 was associated with improved activation (67). It is plausible that, while interactions between

1280 HLA-E on 721 and NKG2A on NK cells can impair NK cell activation, signaling through the

1281 other aNKR engaged by characteristic 721 ligands can compensate for this inhibitory input.

1282 In conclusion, we found that the two HLA-null cell lines K562 and 721 differed in their

1283 expression of ligands to activating NK cell receptors. Together, these experiments provide a

1284 more systematic assessment of the mechanisms underlying the stimulus properties of two cell

1285 types commonly used to study NK cell activation. The different aNKR ligand expression profiles

1286 of K562 and 721 are associated with the induction of different NK cell responses, however, more

- 49 -

1287 work that examines the specific contribution of each ligand-aNKR pair to different stages of NK

1288 cell activation will be required to confirm the functional effects of these phenotypic differences.

- 50 -

1289 2.7: Figure legends

1290 Figure 1: Different frequencies of K562 and 721 express ligands to activating NK cell

1291 receptors. Comparison of the frequency of K562 (white bars) and 721 (grey bars) expressing

1292 (A) Panel 1, (B) Panel 2, (C) Panel 3, and (D) HLA aNKR ligands. The frequency (%) of HLA-

1293 null cells expressing the indicated ligand is represented on the y-axis. Bar height and error bars

1294 represent the median and range for the data set. Each data point represents one of three replicates

1295 run concurrently.

1296 Figure 2: K562 and 721 differ in the expression score of their ligands to activating NK cell

1297 receptor. Comparison of the expression score of (A) Panel 1, (B) Panel 2, (C) Panel 3, and (D)

1298 HLA aNKR ligands on K562 (white bars) and 721 (grey bars). The expression score was

1299 calculated by multiplying the frequency and mean fluorescence intensity of the indicated ligand

1300 expression and is represented on the y-axis. Bar height and error bars represent the median and

1301 range for the data set. Each data point represents one of three replicates run concurrently.

- 51 -

1302 Figure 1: Different frequencies of K562 and 721 express ligands to activating NK cell

1303 receptors.

- 52 -

1304 Figure 2: K562 and 721 differ in the expression score of their ligands to activating NK cell

1305 receptor.

- 53 -

2.8: References 1. Vivier E, Tomasello E, Baratin M, Walzer T, Ugolini S. 2008. Functions of natural killer cells. Nat Immunol 9:503-510. 2. Lettau M, Schmidt H, Kabelitz D, Janssen O. 2007. Secretory lysosomes and their cargo in T and NK cells. Immunol Lett 108:10-19. 3. Orange JS, Harris KE, Andzelm MM, Valter MM, Geha RS, Strominger JL. 2003. The mature activating natural killer cell immunologic synapse is formed in distinct stages. Proc Natl Acad Sci U S A 100:14151-14156. 4. Vivier E, Raulet DH, Moretta A, Caligiuri MA, Zitvogel L, Lanier LL, Yokoyama WM, Ugolini S. 2011. Innate or Adaptive Immunity? The Example of Natural Killer Cells. Science 331:44-49. 5. Cheng M, Chen Y, Xiao W, Sun R, Tian Z. 2013. NK cell-based immunotherapy for malignant diseases. Cell Mol Immunol 10:230-252. 6. Fauriat C, Long EO, Ljunggren H-G, Bryceson YT. 2010. Regulation of human NK-cell cytokine and chemokine production by target cell recognition. Blood 115:2167-2176. 7. Lanier LL. 2004. NK CELL RECOGNITION. Annual Review of Immunology 23:225-274. 8. Vivier E, Ugolini S, Blaise D, Chabannon C, Brossay L. 2012. Targeting natural killer cells and natural killer T cells in cancer. Nat Rev Immunol 12:239-252. 9. Lanier LL. 2005. NK cell recognition. Annu Rev Immunol 23:225-274. 10. Klein E, Ben-Bassat H, Neumann H, Ralph P, Zeuthen J, Polliack A, Vánky F. 1976. Properties of the K562 cell line, derived from a patient with chronic myeloid leukemia. International Journal of Cancer 18:421-431. 11. Lozzio C, Lozzio B. 1975. Human chronic myelogenous leukemia cell-line with positive Philadelphia chromosome. Blood 45:321-334. 12. Shimizu Y, DeMars R. 1989. Production of human cells expressing individual transferred HLA- A,-B,-C genes using an HLA-A,-B,-C null human cell line. J Immunol 142:3320-3328. 13. Bryceson YT, March ME, Ljunggren H-G, Long EO. 2006. Activation, co–activation, and co– stimulation of resting human NK cells. Immunological reviews 214:10.1111/j.1600- 1065X.2006.00457.x. 14. Lisovsky I, Isitman G, Bruneau J, Bernard NF. 2015. Functional analysis of NK cell subsets activated by 721.221 and K562 HLA-null cells. J Leukoc Biol 97:761-767. 15. Bauer S, Groh V, Wu J, Steinle A, Phillips JH, Lanier LL, Spies T. 1999. Activation of NK cells and T cells by NKG2D, a receptor for stress-inducible MICA. Science 285:727-729. 16. Bottino C, Castriconi R, Pende D, Rivera P, Nanni M, Carnemolla B, Cantoni C, Grassi J, Marcenaro S, Reymond N, Vitale M, Moretta L, Lopez M, Moretta A. 2003. Identification of PVR (CD155) and Nectin-2 (CD112) as cell surface ligands for the human DNAM-1 (CD226) activating molecule. J Exp Med 198:557-567. 17. Chalupny NJ, Sutherland CL, Lawrence WA, Rein-Weston A, Cosman D. 2003. ULBP4 is a novel ligand for human NKG2D. Biochem Biophys Res Commun 305:129-135. 18. Sandusky MM, Messmer B, Watzl C. 2006. Regulation of 2B4 (CD244)-mediated NK cell activation by ligand-induced receptor modulation. Eur J Immunol 36:3268-3276. 19. Brandt CS, Baratin M, Yi EC, Kennedy J, Gao Z, Fox B, Haldeman B, Ostrander CD, Kaifu T, Chabannon C, Moretta A, West R, Xu W, Vivier E, Levin SD. 2009. The B7 family member B7-H6 is a tumor cell ligand for the activating natural killer cell receptor NKp30 in humans. J Exp Med 206:1495-1503. 20. Sanchez-Correa B, Gayoso I, Bergua JM, Casado JG, Morgado S, Solana R, Tarazona R. 2012. Decreased expression of DNAM-1 on NK cells from acute myeloid leukemia patients. Immunol Cell Biol 90:109-115. 21. Gasser S, Orsulic S, Brown EJ, Raulet DH. 2005. The DNA damage pathway regulates innate immune system ligands of the NKG2D receptor. Nature 436:1186-1190.

54

22. Moretta A, Bottino C, Vitale M, Pende D, Cantoni C, Mingari MC, Biassoni R, Moretta L. 2001. Activating receptors and coreceptors involved in human natural killer cell-mediated cytolysis. Annu Rev Immunol 19:197-223. 23. Cosman D, Mullberg J, Sutherland CL, Chin W, Armitage R, Fanslow W, Kubin M, Chalupny NJ. 2001. ULBPs, novel MHC class I-related molecules, bind to CMV glycoprotein UL16 and stimulate NK cytotoxicity through the NKG2D receptor. Immunity 14:123-133. 24. Sivori S, Vitale M, Morelli L, Sanseverino L, Augugliaro R, Bottino C, Moretta L, Moretta A. 1997. p46, a Novel Natural Killer Cell–specific Surface Molecule That Mediates Cell Activation. The Journal of Experimental Medicine 186:1129-1136. 25. Vitale M, Bottino C, Sivori S, Sanseverino L, Castriconi R, Marcenaro E, Augugliaro R, Moretta L, Moretta A. 1998. NKp44, a novel triggering surface molecule specifically expressed by activated natural killer cells, is involved in non-major histocompatibility complex-restricted tumor cell lysis. J Exp Med 187:2065-2072. 26. Pende D, Parolini S, Pessino A, Sivori S, Augugliaro R, Morelli L, Marcenaro E, Accame L, Malaspina A, Biassoni R, Bottino C, Moretta L, Moretta A. 1999. Identification and molecular characterization of NKp30, a novel triggering receptor involved in natural cytotoxicity mediated by human natural killer cells. J Exp Med 190:1505-1516. 27. Mandelboim O, Lieberman N, Lev M, Paul L, Arnon TI, Bushkin Y, Davis DM, Strominger JL, Yewdell JW, Porgador A. 2001. Recognition of haemagglutinins on virus-infected cells by NKp46 activates lysis by human NK cells. Nature 409:1055-1060. 28. Bloushtain N, Qimron U, Bar-Ilan A, Hershkovitz O, Gazit R, Fima E, Korc M, Vlodavsky I, Bovin NV, Porgador A. 2004. Membrane-associated heparan sulfate proteoglycans are involved in the recognition of cellular targets by NKp30 and NKp46. J Immunol 173:2392-2401. 29. Arnon TI, Lev M, Katz G, Chernobrov Y, Porgador A, Mandelboim O. 2001. Recognition of viral hemagglutinins by NKp44 but not by NKp30. Eur J Immunol 31:2680-2689. 30. Kruse PH, Matta J, Ugolini S, Vivier E. 2014. Natural cytotoxicity receptors and their ligands. Immunol Cell Biol 92:221-229. 31. Hecht ML, Rosental B, Horlacher T, Hershkovitz O, De Paz JL, Noti C, Schauer S, Porgador A, Seeberger PH. 2009. Natural cytotoxicity receptors NKp30, NKp44 and NKp46 bind to different heparan sulfate/heparin sequences. J Proteome Res 8:712-720. 32. Brown MH, Boles K, van der Merwe PA, Kumar V, Mathew PA, Barclay AN. 1998. 2B4, the natural killer and T cell immunoglobulin superfamily surface protein, is a ligand for CD48. J Exp Med 188:2083-2090. 33. Pende D, Bottino C, Castriconi R, Cantoni C, Marcenaro S, Rivera P, Spaggiari GM, Dondero A, Carnemolla B, Reymond N, Mingari MC, Lopez M, Moretta L, Moretta A. 2005. PVR (CD155) and Nectin-2 (CD112) as ligands of the human DNAM-1 (CD226) activating receptor: involvement in tumor cell lysis. Mol Immunol 42:463-469. 34. Tahara-Hanaoka S, Shibuya K, Onoda Y, Zhang H, Yamazaki S, Miyamoto A, Honda S, Lanier LL, Shibuya A. 2004. Functional characterization of DNAM-1 (CD226) interaction with its ligands PVR (CD155) and nectin-2 (PRR-2/CD112). Int Immunol 16:533-538. 35. Shibuya A, Campbell D, Hannum C, Yssel H, Franz-Bacon K, McClanahan T, Kitamura T, Nicholl J, Sutherland GR, Lanier LL, Phillips JH. 1996. DNAM-1, a novel adhesion molecule involved in the cytolytic function of T lymphocytes. Immunity 4:573-581. 36. Shibuya K, Lanier LL, Phillips JH, Ochs HD, Shimizu K, Nakayama E, Nakauchi H, Shibuya A. 1999. Physical and functional association of LFA-1 with DNAM-1 adhesion molecule. Immunity 11:615-623. 37. Gross CC, Brzostowski JA, Liu D, Long EO. 2010. Tethering of intercellular adhesion molecule on target cells is required for LFA-1-dependent NK cell adhesion and granule polarization. J Immunol 185:2918-2926. 38. Schleinitz N, March ME, Long EO. 2008. Recruitment of Activation Receptors at Inhibitory NK Cell Immune Synapses. PLOS ONE 3:e3278.

55

39. Culley FJ, Johnson M, Evans JH, Kumar S, Crilly R, Casasbuenas J, Schnyder T, Mehrabi M, Deonarain MP, Ushakov DS, Braud V, Roth G, Brock R, Kohler K, Davis DM. 2009. Natural killer cell signal integration balances synapse symmetry and migration. PLoS Biol 7:e1000159. 40. Luque I, Reyburn H, Strominger JL. 2000. Expression of the CD80 and CD86 molecules enhances cytotoxicity by human natural killer cells. Hum Immunol 61:721-728. 41. Wilson JL, Charo J, Martin-Fontecha A, Dellabona P, Casorati G, Chambers BJ, Kiessling R, Bejarano MT, Ljunggren HG. 1999. NK cell triggering by the human costimulatory molecules CD80 and CD86. J Immunol 163:4207-4212. 42. Costa C, Barber DF, Fodor WL. 2002. Human NK cell-mediated cytotoxicity triggered by CD86 and Gal alpha 1,3-Gal is inhibited in genetically modified porcine cells. J Immunol 168:3808-3816. 43. Galea-Lauri J, Darling D, Gan SU, Krivochtchapov L, Kuiper M, Gaken J, Souberbielle B, Farzaneh F. 1999. Expression of a variant of CD28 on a subpopulation of human NK cells: implications for B7-mediated stimulation of NK cells. J Immunol 163:62-70. 44. Kaiser BK, Barahmand-Pour F, Paulsene W, Medley S, Geraghty DE, Strong RK. 2005. Interactions between NKG2x immunoreceptors and HLA-E ligands display overlapping affinities and thermodynamics. J Immunol 174:2878-2884. 45. Braud VM, Allan DS, O'Callaghan CA, Soderstrom K, D'Andrea A, Ogg GS, Lazetic S, Young NT, Bell JI, Phillips JH, Lanier LL, McMichael AJ. 1998. HLA-E binds to natural killer cell receptors CD94/NKG2A, B and C. Nature 391:795-799. 46. Wada H, Matsumoto N, Maenaka K, Suzuki K, Yamamoto K. 2004. The inhibitory NK cell receptor CD94/NKG2A and the activating receptor CD94/NKG2C bind the top of HLA-E through mostly shared but partly distinct sets of HLA-E residues. Eur J Immunol 34:81-90. 47. Lee N, Llano M, Carretero M, Ishitani A, Navarro F, López-Botet M, Geraghty DE. 1998. HLA-E is a major ligand for the natural killer inhibitory receptor CD94/NKG2A. Proceedings of the National Academy of Sciences 95:5199-5204. 48. Garcia-Beltran WF, Holzemer A, Martrus G, Chung AW, Pacheco Y, Simoneau CR, Rucevic M, Lamothe-Molina PA, Pertel T, Kim TE, Dugan H, Alter G, Dechanet-Merville J, Jost S, Carrington M, Altfeld M. 2016. Open conformers of HLA-F are high-affinity ligands of the activating NK-cell receptor KIR3DS1. Nat Immunol 17:1067-1074. 49. Burian A, Wang KL, Finton KAK, Lee N, Ishitani A, Strong RK, Geraghty DE. 2016. HLA- F and MHC-I Open Conformers Bind Natural Killer Cell Ig-Like Receptor KIR3DS1. PLoS ONE 11:e0163297. 50. Pende D, Rivera P, Marcenaro S, Chang CC, Biassoni R, Conte R, Kubin M, Cosman D, Ferrone S, Moretta L, Moretta A. 2002. Major histocompatibility complex class I-related chain A and UL16-binding protein expression on tumor cell lines of different histotypes: analysis of tumor susceptibility to NKG2D-dependent natural killer cell cytotoxicity. Cancer Res 62:6178- 6186. 51. Chen X, Trivedi PP, Ge B, Krzewski K, Strominger JL. 2007. Many NK cell receptors activate ERK2 and JNK1 to trigger microtubule organizing center and granule polarization and cytotoxicity. Proc Natl Acad Sci U S A 104:6329-6334. 52. Golden-Mason L, Cox AL, Randall JA, Cheng L, Rosen HR. 2010. Increased NK cytotoxicity and NKp30 expression protects against HCV infection in high-risk individuals and inhibits replication in vitro. Hepatology (Baltimore, Md) 52:1581-1589. 53. Monaco EL, Tremante E, Cerboni C, Melucci E, Sibilio L, Zingoni A, Nicotra MR, Natali PG, Giacomini P. 2011. Human Leukocyte Antigen E Contributes to Protect Tumor Cells from Lysis by Natural Killer Cells. Neoplasia (New York, NY) 13:822-830. 54. Maier S, Grzeschik M, Weiss EH, Ulbrecht M. 2000. Implications of HLA-E allele expression and different HLA-E ligand diversity for the regulation of NK cells. Hum Immunol 61:1059- 1065.

56

55. Lee N, Goodlett DR, Ishitani A, Marquardt H, Geraghty DE. 1998. HLA-E surface expression depends on binding of TAP-dependent peptides derived from certain HLA class I signal sequences. J Immunol 160:4951-4960. 56. Kraemer T, Celik AA, Huyton T, Kunze-Schumacher H, Blasczyk R, Bade-Döding C. 2015. HLA-E: Presentation of a Broader Peptide Repertoire Impacts the Cellular Immune Response— Implications on HSCT Outcome. Stem Cells International 2015:346714. 57. Celik AA, Kraemer T, Huyton T, Blasczyk R, Bade-Döding C. 2016. The diversity of the HLA-E-restricted peptide repertoire explains the immunological impact of the Arg107Gly mismatch. Immunogenetics 68:29-41. 58. Jørgensen PB, Livbjerg AH, Hansen HJ, Petersen T, Höllsberg P. 2012. Epstein-Barr virus Peptide Presented by HLA-E is Predominantly Recognized by CD8(bright) Cells in multiple Sclerosis Patients. PLoS ONE 7:e46120. 59. Romagnani C, Pietra G, Falco M, Millo E, Mazzarino P, Biassoni R, Moretta A, Moretta L, Mingari MC. 2002. Identification of HLA-E-specific alloreactive T lymphocytes: a cell subset that undergoes preferential expansion in mixed lymphocyte culture and displays a broad cytolytic activity against allogeneic cells. Proc Natl Acad Sci U S A 99:11328-11333. 60. Castriconi R, Dondero A, Cantoni C, Della Chiesa M, Prato C, Nanni M, Fiorini M, Notarangelo L, Parolini S, Moretta L, Notarangelo L, Moretta A, Bottino C. 2007. Functional characterization of natural killer cells in type I leukocyte adhesion deficiency. Blood 109:4873-4881. 61. Stanietsky N, Simic H, Arapovic J, Toporik A, Levy O, Novik A, Levine Z, Beiman M, Dassa L, Achdout H, Stern-Ginossar N, Tsukerman P, Jonjic S, Mandelboim O. 2009. The interaction of TIGIT with PVR and PVRL2 inhibits human NK cell cytotoxicity. Proc Natl Acad Sci U S A 106:17858-17863. 62. Yu X, Harden K, C Gonzalez L, Francesco M, Chiang E, Irving B, Tom I, Ivelja S, Refino CJ, Clark H, Eaton D, Grogan JL. 2009. The surface protein TIGIT suppresses T cell activation by promoting the generation of mature immunoregulatory dendritic cells. Nat Immunol 10:48-57. 63. Wang F, Hou H, Wu S, Tang Q, Liu W, Huang M, Yin B, Huang J, Mao L, Lu Y, Sun Z. 2015. TIGIT expression levels on human NK cells correlate with functional heterogeneity among healthy individuals. European Journal of Immunology 45:2886-2897. 64. Stanietsky N, Rovis TL, Glasner A, Seidel E, Tsukerman P, Yamin R, Enk J, Jonjic S, Mandelboim O. 2013. Mouse TIGIT inhibits NK-cell cytotoxicity upon interaction with PVR. Eur J Immunol 43:2138-2150. 65. Liu S, Zhang H, Li M, Hu D, Li C, Ge B, Jin B, Fan Z. 2013. Recruitment of Grb2 and SHIP1 by the ITT-like motif of TIGIT suppresses granule polarization and cytotoxicity of NK cells. Cell Death Differ 20:456-464. 66. Valés-Gómez M, Reyburn HT, Erskine RA, López-Botet M, Strominger JL. 1999. Kinetics and peptide dependency of the binding of the inhibitory NK receptor CD94/NKG2-A and the activating receptor CD94/NKG2-C to HLA-E. The EMBO Journal 18:4250-4260. 67. Lisovsky I, Isitman G, Song R, DaFonseca S, Tremblay-McLean A, Lebouche B, Routy JP, Bruneau J, Bernard NF. 2015. A Higher Frequency of NKG2A+ than of NKG2A- NK Cells Responds to Autologous HIV-Infected CD4 Cells irrespective of Whether or Not They Coexpress KIR3DL1. J Virol 89:9909-9919.

57

1306

1307

1308

1309

1310 NK cells can be activated by autologous HIV-infected CD4+ T cells and will

1311 subsequently inhibit viral replication. It has been well established that the HIV viral accessory

1312 proteins Nef and Vpu can internalize the major histocompatibility complex (MHC)-1 molecules

1313 human leukocyte antigens (HLA)-A and -B, which are ligands to the inhibitory killer

1314 immunoglobulin-like receptors on NK cells. Similarly, the influence of HIV on inhibitory

1315 receptor expression on NK cells and the impact of these changes on NK-mediated anti-HIV

1316 responses has been well studied. In contrast, modulation of ligands to activating NK cell

1317 receptors by HIV infection and its contribution to anti-viral NK cell responses remains

1318 incompletely characterized. We wanted to address this knowledge gap by assessing the

1319 expression of a comprehensive panel of ligands to activating NK cell receptors on HIV-infected

1320 and -uninfected CD4+ T cells.

1321

1322

1323

1324 - 58 -

1325 Chapter 3: Manuscript 2 – Expression profiles of ligands for activating NK cell receptors

1326 on HIV infected and uninfected CD4+ T cells.

1327

1328

1329

1330

1331

1332

1333

1334

1335

1336

1337

1338

1339

1340 - 59 -

1341 Expression profiles of ligands for activating NK cell receptors on HIV infected and 1342 uninfected CD4+ T cells. 1343 1344 1345 1346 Alexandra Tremblay-McLean1,2, Julie Bruneau3,4, Bertrand Lebouché1,5,6, and Nicole F. 1347 Bernard1,2,6,7 1348 1349 1350 1Research Institute of the McGill University Health Center, Montréal, Québec, Canada 1351 2Division of Experimental Medicine, McGill University, Montréal, Québec, Canada 1352 3Department of Family Medicine, Université de Montréal, Montréal, Québec, Canada 1353 4Centre de Recherche de Centre Hospitalier de l’Université de Montréal, Montréal, Québec, 1354 Canada 1355 5Department of Family Medicine, McGill University, Montréal, Québec, Canada 1356 6Chronic Viral Illness Service, McGill University Health Centre, Montréal, Québec, Canada 1357 7Division of Clinical Immunology, McGill University Health Centre, Montréal, Québec, Canada 1358 1359 1360 1361 1362 1363 1364 Mailing address: Dr. Nicole F. Bernard, Research Institute of the McGill University Health 1365 Centre, Glen Site, 1001 Décarie Boulevard, Block E, Rm E03.3380, Montréal, Québec, Canada, 1366 H4A 3J1. 1367 Tel: (514) 934-1934 ext:42697 1368 E-mail: [email protected] 1369 1370 1371 Funding: This study received support from the Canadian Institutes for HIV Research grant #

1372 MOP-142494. A. Tremblay-McLean was supported by Canadian Association for HIV Research

1373 Abbvie Master’s Award in Basic Science.

1374 1375 Word Count: 5769 1376

- 60 -

1377 3.1: Author contributions and acknowledgements

1378

1379 ATM and NFB were responsible for study design, data analysis, and manuscript preparation. JB

1380 and BL provided subject samples.

1381

1382 We would like to acknowledge the assistance of several lab members. In particular, Ms. Xiaoyan

1383 Ni and Ms. Tsoarello Mabanga who KIR and HLA typed the study subjects. Dr. Irene Lisovsky

1384 and Dr. Gamze Isitman helped with the development of the multi-parametric flow cytometry

1385 panels. Dr. Irene Lisovsky and Dr. Rujun Song who provided the raw data for the analysis of

1386 inhibition of viral replication.

1387

1388

1389

1390

1391

1392

1393

- 61 -

1394 3.2: Abstract

1395 Natural Killer (NK) cells play a crucial role in the clearance of virally-infected cells, an

1396 ability that depends on the integration of signals received through inhibitory and activating NK

1397 receptors (iNKR and aNKR). HIV-infected CD4 T cells (iCD4) activate NK cells to inhibit HIV

1398 replication. HIV infection-dependent changes in the HLA ligands for iNKR on iCD4 are well

1399 documented. In contrast, less is known regarding the HIV infection related changes in ligands for

1400 aNKR on iCD4 T cells. Here, we compared the aNKR ligand profiles of early and late stage HIV

1401 iCD4s to uninfected CD4 (unCD4) T cells. We activated and infected CD4 T cells isolated from

1402 17 HIV-1 seronegative donors with HIVJR-CSF for 7 days. UnCD4 served as a control for the

1403 effect of HIV infection on aNKR. On day 7, the expression of ULBP-1, ULBP-2/5/6, ULBP-3,

1404 MIC-A, MIC-B, CD48, CD80, CD86, CD112, CD155, ICAM-1, ICAM-2, HLA-E, HLA-A2,

1405 HLA-C was analyzed by flow cytometry. Intracellular p24 staining was also used to distinguish

1406 iCD4 from uninfected CD4 cells present in the same culture (euCD4). We found that the iCD4

1407 that maintained CD4 surface expression (iCD4+) showed higher expression of aNKR ligands

1408 compared to unCD4, euCD4, and iCD4 where CD4 expression had been downregulated by HIV

1409 Nef. Meanwhile, the expression of aNKR ligands on iCD4- was similar to (ULBP-1, ULBP-

1410 2/5/6, ULBP-3, MIC-A, CD80, and ICAM-1) or significantly lower than (MIC-B, CD48,

1411 CD112, CD155, and ICAM-2) what was observed on unCD4 and euCD4. This suggests that HIV

1412 infection initially results in increased expression of aNKR ligands, but that this increase is

1413 transient and decreases to baseline or below concomitantly with the HIV-mediated

1414 downregulation of CD4 on the surface of infected cells. Additionally, the aNKR ligand profiles

1415 of unCD4 and euCD4 T cells did not differ, suggesting that exposure to virus of uninfected cells

1416 alone is insufficient for aNKR ligand modulation.

- 62 -

1417 3.3: Introduction

1418 Natural Killer (NK) cells are innate immune cells that mediate cytotoxic responses to

1419 stressed, virally-infected, and transformed cells (Reviewed in 1). NK cells can lyse target cells

1420 directly through the production of granzyme and perforin, or indirectly through antibody (Ab)-

1421 dependent mechanisms, through the production of cytokines and chemokines, and through

1422 activation of adaptive immune cells (2-4). The induction and potency of these responses depend

1423 on the activation state of the NK cell, which is regulated by signals transmitted through both

1424 activating and inhibitory NK cell receptors (aNKR and iNKRs) (4, 5). Signals from iNKR often

1425 dominate by interrupting signals from aNKR, the consequences of which explain tolerance to

1426 self (6). When signals from iNKR are interrupted NK cell activation can occur if target cells

1427 express ligands to aNKR (1, 4, 7, 8). However, in vivo most cells co-express variable

1428 combinations of ligands to both NKR types and NK cell activation will depend on the integration

1429 of the number and strength of all activating and inhibitory signals transmitted.

1430 NK cells express a variety of receptors. One of the major aNKRs is the C-type lectin

1431 NKG2D, which binds to a family of major histocompatibility complex (MHC)-I like molecules

1432 that includes the human cytomegalovirus UL-16 binding proteins (ULBP) and MHC-I related

1433 chain (MIC) proteins (9-11). The expression of these ligands is induced by cellular stress and

1434 they are otherwise not found on the surface of healthy cells (12). The other major family of

1435 aNKR comprises the natural cytotoxicity receptors (NCR) NKp30, NKp44, and NKp46 (9, 13-

1436 15). Although NCRs can bind membrane-associated heparan sulfate glycosaminoglycans and

1437 viral hemagglutinin, their cellular ligands remain incompletely identified (16-20).

- 63 -

1438 Several additional receptors that contribute to NK cell activation by target cells have been

1439 identified. Two members of the CD2 subfamily of receptors, CD244/2B4 and the NK-T-B cell

1440 antigen (NTB-A), can bind to CD48 and trigger NK cell cytotoxicity (21). The leukocyte

1441 adhesion molecule DNAX accessory molecule-1 (DNAM-1) is widely expressed on NK cells

1442 and engages both CD112 (Nectin-2) and CD155 (poliovirus receptor) expressed on target cells

1443 (22-24). DNAM-1’s ability to trigger NK cell responses requires co-expression of the

1444 lymphocyte function-associated antigen 1 (LFA-1), which interacts with the integrins ICAM-1

1445 and ICAM-2 on target cells to form the immunological synapse (25-29). The T cell co-

1446 stimulatory B7 molecules CD80 and CD86 have also been shown to stimulate NK cell-mediated

1447 lysis through a mechanism that is currently undefined, but may depend on a CD28 variant

1448 expressed on NK cells (30-33).

1449 While a subset of the classical MHC-I human leukocyte antigens (HLA) -A, -B, and -C

1450 are ligands to the inhibitory killer immunoglobulin-like receptor (KIR) family, the non-classical

1451 HLA-E and -F interact with aNKR (Reviewed in 34). HLA-E can signal through the inhibitory

1452 C-type lectin-like receptor NKG2A and the activating NKG2E and -C, all of which can form

1453 heterodimers with CD94 (35-37).

1454 It was previously shown that autologous HIV-infected CD4+ T cells are capable of

1455 activating NK cells ex vivo and that these activated NK cells will inhibit further viral replication

1456 (38-40). The HIV accessory proteins Nef and Vpu have well documented effects on expression

1457 of ligands to iNKR on the surface of infected cells. Specifically, both viral proteins contribute to

1458 the internalization and downregulation of HLA-A and -B, abrogating inhibitory signaling

1459 through their cognate iNKR (41-43). As NK cells are directed to kill cells with low expression of

- 64 -

1460 MHC-1 antigens, the downregulation of HLA-A and -B by HIV should make infected cells more

1461 susceptible to NK cell lysis (44). However, a third MHC-1 antigen, HLA-C, and the non-

1462 classical HLA-E antigen remain expressed on the surface of infected cells, where they can

1463 engage the inhibitory KIR2DL1, -2, and -3 and NKG2A/CD94, respectively, to inhibit NK cell

1464 activation (45).

1465 The influence of HIV on iNKR ligand expression and its impact on NK cell activation

1466 and subsequent antiviral responses has been well studied. The contribution of ligands to aNKR,

1467 however, is less well characterized. In this work, we determined whether HIV-I infection can

1468 modulate the expression profile of ligands to aNKRs on infected cells and to identify aNKR

1469 ligands that are preferentially modulated by infection.

- 65 -

1470 3.4: Methods

1471 3.4.1: Ethics Statement

1472 This study was conducted in accordance with the principles expressed in the Declaration

1473 of Helsinki. It was approved by the Institutional Review Boards of the Comité d’Éthique de la

1474 Recheche du Centre Hospitalier de l’Université de Montréal and the Research Ethics Committee

1475 of the McGill University Health Centre - Montreal General Hospital. Informed consent was

1476 obtained for all study participants.

1477 3.4.2: Study population

1478 This study included a total of 17 HIV seronegative donors. The HLA-A, -B, and -C

1479 genotypes of the study participants is presented in Table 1. 11 additional HIV seronegative

1480 donors were included in in the supplemental analysis of viral inhibition.

1481 3.4.3: CD4+ T cell isolation

1482 Peripheral blood mononuclear cells (PBMC) were isolated from blood by density

1483 gradient centrifugation (Ficoll-Paque; Pharmacia, Uppsala, Sweden) and cryopreserved in 10%

1484 dimethyl sulfoxide (DMSO; Sigma-Aldrich, St. Louis, MO) with 90% fetal bovine serum (FBS;

1485 Wisent, Inc. St. Bruno, QC, Canada). CD4+ T cells from 17 HIV seronegative individuals were

1486 positively selected from thawed PBMCs using a commercially available kit, as per

1487 manufacturer’s instructions (Stemcell Technologies, Inc. Vancouver, BC, Canada). Briefly, an

1488 anti-CD4 selection cocktail was added to PBMCs resuspended at 100x106 cells/mL in

1489 Dulbecco’s phosphate buffered saline (dPBS); 2% FBS (Wisent, Inc. St. Bruno, QC, Canada);

- 66 -

1490 1mM EDTA (Sigma-Aldrich, Oakville, ON, Canada) for 15 mins at room temperature (RT). Ab-

1491 bound cells were then mixed with magnetic beads for 10 mins at RT. Cell suspensions were

1492 placed in a magnet for 5 min, after which the supernatant was removed; this process was

1493 repeated for a total of 3 cycles. The purity of the isolated cells was determined by flow cytometry

1494 and averaged 93.6±3.5%.

1495 3.4.4: HIV infection

1496 Following isolation, CD4+ T cells were incubated at 37°C at a concentration of 2x106

1497 cells/mL in RPMI 1640 medium; 10%FBS; 2mM L-glutamine; 100IU/mL penicillin; 100mg/mL

1498 streptomycin (R10; all from Wisent) supplemented with 100IU/mL of recombinant human IL-2

1499 (R10-rhIL-2; Chiron Corp., Emeryville, CA) and activated with 1μg/mL of phytohemagglutinin

1500 (PHA; MP Biomedicals, Santa Ana, CA) for 24 hrs. After activation, cells were washed and

1501 incubated at 37°C in R10-IL2 for 72 hrs. Following this incubation, at least 2x106 cells were

1502 infected with the CCR5-tropic HIV-1JR-CSF at a multiplicity of infection of 0.01 and subsequently

1503 cultured at 37°C at a concentration of 1x106 cells/mL of R10-IL-2 for 7 days. Uninfected CD4+ T

1504 cells (unCD4) were cultured in parallel with their infected counterparts.

1505 3.4.5: Antibody staining and acquisition

1506 On day 7 post-infection, HIV-infected CD4+ T cells (iCD4) and unCD4 were stained with

1507 UV Live/Dead ® Fixable Blue cell stain kit (Thermofisher, Waltham, MA) and then with anti-

1508 CD3-BV785 (OKT3; BioLegend, San Diego, CA) and anti-CD4-BUV737 (SK3; BD Bioscience,

1509 San Jose, CA) in conjunction with fluorochrome conjugated monoclonal antibodies (Abs) with

1510 clone designations shown in parentheses to the following cell surface markers, which were

- 67 -

1511 organized into 4 panels for the purpose of analysis: (Panel 1) ULBP-1-PerCP (170818), ULBP-

1512 2/5/6-APC (165903), ULBP-3-PE (166510), MIC-A-APC (159227), MIC-B-AlexaFluor 700

1513 (236511; all from R&D Systems, Minneapolis, MN), (Panel 2) CD48-PE (BJ40), CD80-BV421

1514 (2D10), CD86-PE-Dazzle594 (IT2.2; all from BioLegend), CD112-AlexaFluor 700 (610603;

1515 R&D), CD155-PE-Cy7 (SKII.4; BioLegend), (Panel 3) ICAM-1-Pacific Blue (HCD54), and

1516 ICAM-2-PE (CBR-1C2/2; both from BioLegend). A final HLA Panel comprised the conjugated

1517 antibodies HLA-E-PE-Cy7 (3D12; BioLegend) and HLA-A2-APC (BB7.2; eBioscience, San

1518 Diego, CA), as well as DT9, an unconjugated Ab against HLA-C (DT9; EMD Millipore,

1519 Billerica, MA). Briefly, cells were resuspended in a 96-well V bottom plate (Sarstedt,

1520 Nümbrecht, Germany) at a concentration of 1x106 cells per 100μL of Dulbecco’s phosphate

1521 buffered saline (dPBS; Wisent) and stained with the Live/Dead reagent. TruStain FcX reagent

1522 (BioLegend) was used to minimize non-specific Fc receptor interactions and cells were stained

1523 with conjugated antibodies from Panel 1, 2, 3, and HLA for 30 min in the dark at room

1524 temperature (RT). Following staining, cells were washed, fixed, and permeabilized using the

1525 reagents in the Fix & Perm Cell fixation and permeabilization kit (Thermofisher), as per

1526 manufacturer’s instruction. Intracellular staining with anti-p24-FITC (KC57; Beckman-Coulter,

1527 Mississauga, ON, Canada) was used to determine the percentage of HIV-infected (iCD4), with

1528 unCD4 providing background values.

1529 iCD4 and unCD4 stained with DT9 were included in the HLA panel and were prepared

1530 as above. They were incubated with the primary Ab on ice for 40 min. After washing, anti-HLA-

1531 C binding was detected with APC-conjugated polyclonal goat F(ab’)2 anti-mouse IgG secondary

1532 Ab (eBioscience) for 20 min on ice. Following this, cells were washed and stained with Abs to

1533 CD3, CD4, and intracellular p24 as previously described. After staining, all cells were washed

- 68 -

1534 and fixed with 2% paraformaldehyde (PFA; Santa Cruz, Dallas TX). Between 400,000 and

1535 600,000 events were acquired on an LSRFortessa x20 (BD) within 24hrs. Unstained, single

1536 stained control (CompBead; BD), fluorescence minus one, and secondary Ab alone controls were

1537 used for multi-colour compensation and gating purposes. Flow cytometric analysis was

1538 performed using FlowJo software version 10 (TreeStar, Ashland OR). All the Abs used and their

1539 manufacturers are listed in Appendix A.

1540 3.4.6: Statistical analysis

1541 Statistical analysis was performed using GraphPad Prism version 6 (GraphPad, San

1542 Diego, CA). Wilcoxon tests or Friedman tests with Dunn’s post-test comparisons were used to

1543 compare the significance of differences in the frequency or mean fluorescence intensity (MFI) of

1544 a cell surface ligand between 2 or more than 2 matched groups, respectively. Spearman

1545 correlation tests were used to determine the significance of correlations between ligand

1546 expression and HIV infection. P-values less than 0.05 were considered significant.

- 69 -

1547 3.5: Results

1548 3.5.1: CD4+ T cell infection characteristics

1549 To investigate the expression of aNKR ligands on CD4+ T cells and the modulatory

1550 effects of HIV-1 infection on aNKR expression, we used 4 Ab panels specific for aNKR ligands.

1551 As the infection protocol requires culturing cells in rhIL-2-containing media, which can

1552 upregulate aNKR ligands, we cultured unCD4 T cells under the same conditions as an internal

1553 control to observe the effects of HIV infection on aNKR profiles irrespective of the effects of IL-

1554 2. The gating strategy in Figure 1A was used to identify T cells as single, live, CD3+

1555 lymphocytes. As illustrated in Figure 1B and C, the expression of CD4 and intracellular p24 – a

1556 measure of HIV-1 infection – was used to delineate 5 different T cell subsets. The first subset

1557 was HIV-uninfected p24-CD4+ T cells that had never been exposed to virus (unCD4s; Fig. 1B).

1558 Similarly, from the population of cells that were cultured with HIV, we identified a subset of

1559 p24-CD4+ T cells that remained HIV-uninfected despite being exposed to virus (euCD4) (Fig.

1560 1C). Total infected p24+ T cells comprise the iCD4T subset. However, consistent with reports

1561 that the HIV-1 viral protein Nef can downregulate surface expression of CD4, two distinct

1562 subsets could be observed within iCD4T (41, 46). We observed an HIV-infected p24+CD4+ T cell

1563 (iCD4+) subset and an HIV-infected p24+CD4- T cells (iCD4-) subset in which cell-surface CD4

1564 was downregulated (Fig. 1B).

1565 3.5.2: Comparison of the expression profiles of ligands to aNKR on HIV-infected CD4+ T

1566 cells that maintain or downregulate CD4

- 70 -

1567 It has been proposed that HIV iCD4- are more productively infected than iCD4+ (46, 47).

1568 In line with this, we observed that iCD4-, compared to iCD4+ cells, have higher expression levels

1569 of p24 on a per cell basis (p≤0.0001, paired t-test, Fig. 1D). To assess the potential phenotypic

1570 implications of these differences in infection kinetics, we compared the frequencies of iCD4+ and

1571 iCD4- that expressed ligands to the Abs in Panels 1, 2, and 3 (Fig. 2). We found that, apart from

1572 cells expressing ULBP-2/5/6, CD86, and ICAM-1, the frequency of iCD4- expressing all other

1573 aNKR ligands detected by these Abs was significantly lower than that of iCD4+ cells (p≤0.008,

1574 for all comparisons, Wilcoxon test).

1575 3.5.3: Comparison of the frequencies of HIV iCD4 and unCD4 T cells expressing ligands to

1576 aNKRs

1577 We next compared the expression profiles of uninfected cells to iCD4T, iCD4+ and iCD4-

1578 cells; Fig. 3). As we observed no significant differences in the expression of each of the aNKR

1579 ligands studied between unCD4 and euCD4 (data not shown), all comparisons are made against

1580 unCD4. We observed that HIV infection altered the frequency of T cells expressing all the aNKR

1581 ligands studied, except CD86. Specifically, we found that a greater frequency of iCD4+ than

1582 unCD4 expressed ULBP-1, ULBP-2/5/6, ULBP-3, MIC-A, CD80, and ICAM-1 (p≤0.0009, for

1583 all comparisons, Friedman with Dunn’s post-test). While a higher frequency of iCD4T than

1584 unCD4 were positive for these aNKR ligands, few of these comparisons reached statistical

1585 significance. Furthermore, the frequency of iCD4- and unCD4 expressing ULBP-1, ULBP-2/5/6,

1586 ULBP-3, MIC-A, and CD80 did not differ significantly (Figure 3A-C). The exception to this was

1587 ICAM-1, where frequencies of all infected subsets (iCD4T, iCD4+, and iCD4-) expressing this

1588 ligand were higher than those of unCD4 (p ≤ 0.0032, for all comparisons). The frequency of

- 71 -

1589 iCD4+ cells expressing MIC-B, CD48, CD112, CD155, and ICAM-2 was not significantly higher

1590 than that of unCD4 (Fig. 3A-C). However, the frequencies of iCD4- cells expressing these aNKR

1591 ligands were less than those of unCD4, although this trend only reached significance for MIC-B

1592 expression (p≤0.0115). Together these results show that HIV infection lead to the upregulated

1593 expression of several aNKR ligands. The lower frequency of several of these ligands on iCD4-

1594 compared to iCD4+ cells suggests that they are downregulated concomitantly with CD4

1595 downregulation.

1596 3.5.4: Comparison of the aNKR ligand expression intensity on iCD4 and unCD4 T cells

1597 To further characterize the extent of ligand modulation in HIV-1-infected CD4+ T cells,

1598 we next assessed potential changes to the per-cell expression of the Panel 1, 2, and 3 aNKR

1599 ligands studied by measuring the mean fluorescence intensity (MFI) of aNKR ligand expression

1600 (Fig. 4). We observed that the MFI of ULBP-1, ULBP-2/5/6, ULBP-3, MIC-A, CD48, CD80,

1601 and CD155 was significantly elevated in iCD4+, but not on iCD4-, compared to unCD4

1602 (p≤0.0232, for all comparisons, Friedman with Dunn’s post-test). The MFI of these ligands was

1603 also elevated in iCD4T, but was only significantly higher than unCD4 for ULBP-3, MIC-A, and

1604 CD80 expression (p=0.0032, p=0.0061, and p=0.0115, respectively). While infection had no

1605 effect on the frequency of T cells expressing CD86, we observed that the expression of this

1606 ligand on iCD4+ and iCD4T was significantly increased, relative to unCD4 (p=0.002 and

1607 p=0.0061, respectively). In contrast, HIV infection was associated with a sustained increase in

1608 the MFI of ICAM-1 on all infected cells, regardless of CD4 expression (p≤0.0077, for all

1609 comparisons). In sum, we observed that HIV infection is associated with changes to the aNKR

1610 ligand expression profile and that changes in the MFI of ligand expression often mirrored the

- 72 -

1611 changes observed in the frequencies of T cells expressing that ligand. Specifically, we found that

1612 compared to HIV-unCD4 the expression of ULBP-1, ULBP-2/5/6, ULBP-3, MIC-A, CD48,

1613 CD80, CD86, CD155, and ICAM-1 was elevated on iCD4+, whereas expression of MIC-B,

1614 CD112, and ICAM-2 was reduced on iCD4-.

1615 3.5.5: Expression profiles of HLA-E, HLA-A2, and HLA-C of iCD4 and unCD4

1616 In addition to the iNKR/aNKR ligand HLA-E, we analysed the expression of two iNKR

1617 ligands, HLA-A2 and HLA-C (Fig. 5). Although HIV Nef can downregulate the expression of

1618 HLA-A and B antigens on the surface of infected cells, cell-surface expression of HLA-E and

1619 HLA-C is maintained (45). While a lower frequency of iCD4-, compared to iCD4+, expressed

1620 HLA-E (p = 0.078, Wilcoxon, Fig. 5A), the frequency and MFI of HLA-E expression on iCD4T

1621 was comparable to that of unCD4 (Fig. 5B and C), consistent with the observation that HLA-E is

1622 retained at the surface of bulk HIV-infected cells (45, 48). Similarly, we found that HIV

1623 infection did not appear to impact HLA-C expression on iCD4T or iCD4-, compared to unCD4

1624 (Fig. 5B and C). Consistent with previous reports, the frequencies of HLA-A2+ iCD4- and iCD4T

1625 were significantly lower than that of unCD4 (p = 0.0061 and p = 0.0372, respectively, Friedman

1626 with Dunn’s post-test) and, while just above the threshold for significance (p=0.0637), the MFI

1627 of HLA-A2 expression on iCD4- was two-fold less than that of unCD4 (49, 50). Overall, our

1628 findings support previous observations that the HIV viral protein Nef downmodulates expression

1629 of HLA-A, while leaving surface expression of HLA-E and HLA-C unmodulated.

- 73 -

1630 3.6: Discussion

1631 Since the surface expression of HLA-A and -B antigens are downregulated over the

1632 course of HIV-infection, it is plausible that modulation of the expression of ligands to aNKRs on

1633 the surface of infected cells may have important regulatory effects on the NK cell responses they

1634 elicit. As such, elucidating HIV-induced regulation of the aNKR ligand expression profile on

1635 infected cells would help further characterize the NK cell stimulating potential of these cells. In

1636 this study, we report that HIV infection is associated with significant changes to the expression

1637 profiles of ligands to aNKRs. We observed two general patterns of HIV-induced ligand

1638 modulation. Specifically, we found that, compared to unCD4, the expression of ULBP-1, ULBP-

1639 2/5/6, ULBP-3, MIC-A, and CD80 was increased exclusively on HIV-infected T cells that

1640 maintained cell-surface CD4 expression (iCD4+), whereas infected cells with downregulated

1641 CD4 (iCD4-) demonstrated reduced expression of MIC-B, CD48, CD112, CD155, and ICAM-2.

1642 Furthermore, ICAM-1 was found to be broadly upregulated on all HIV infected cell subsets

1643 examined.

1644 The influence of HIV infection on the expression of some ligands to aNKR has been

1645 previously investigated in different infection models. While ULBP-1, ULBP-2/5/6, and ULBP-3

1646 are rarely expressed on healthy primary CD4+ T cells, it has been well established that infection

1647 with several laboratory strains of HIV can induce their surface expression (51-54). In contrast,

1648 reports on HIV-mediated regulation of the expression of other NKG2D ligands, i.e. MIC-A and

1649 MIC-B, on CD4+ T cells are conflicting (51, 53, 54). HIV infection induces the loss of surface

1650 expression of CD155 on the Jurkat T cell line, however, this could not be reproduced in bulk

+ 1651 p24 primary HIVNL4-3-infected T cells, which found CD155 to be modestly upregulated (55-57).

- 74 -

1652 Infection of the human monocytic cell line U937 with primary isolates of an Indian HIV-1

1653 subtype C induced the downregulation of CD80 and CD86 (58). Finally, HIV infection has also

1654 been suggested to trigger the cell surface loss of CD48 on infected CD4+ T cells (59). In addition

1655 to these analyses of surface expression, an RNA microarray of primary human CD4+ T cells

1656 infected with the HIV-1-based reporter virus NL4-3 BAL-IRES-HSA found that ULBP-1, CD80,

1657 and ICAM-1 expression was upregulated, compared to mock-infected cells, consistent with our

1658 observations of increased surface expression of these ligands (60). Moreover, the RNA

1659 expression profiles of these ligands did not differ between mock infected cells and uninfected

1660 bystander cells, which are the equivalent of the euCD4 studied here (60).

1661 The results presented here add to this body of work by including aNKR ligands not

1662 previously studied, including CD112 and ICAM-2. Additionally, in vitro HIV-infected CD4+ T

1663 cells can be easily divided into two distinct populations (iCD4+ and iCD4-), depending on their

1664 expression of the CD4 co-receptor. In infected CD4 T cells, CD4 surface expression is

1665 downregulated and internalized by the HIV viral protein Nef (41-43, 46). Regardless, most of the

1666 existing literature assessed the phenotype of bulk p24+ infected cells, regardless of CD4

1667 expression. It has, however, been postulated that iCD4+ and iCD4- differ in the burden and

1668 timing of HIV infection and we also observed that the intensity of staining for the viral capsid

1669 protein p24, which serves as a marker of infection, differed significantly between these two

1670 subsets. HIV-infected CD4+ T cells have been shown in vitro to be capable of activating NK

1671 cells, which in turn can block viral replication (38, 39). The ligands studied here and their

1672 engagement of aNKR likely contribute to anti-HIV NK cell responses and differences in the

1673 expression of these ligands on both subsets of infected cells may alter their ability to activate NK

1674 cells. To address this knowledge gap, we devised a gating strategy that allowed us to examine the

- 75 -

1675 expression profiles of aNKR ligands on both subsets of infected T cells separately, including

1676 iCD4+ and iCD4- cells, and compare them to those of euCD4 and unCD4. As hypothesized

1677 aNKR ligand expression did differ on iCD4+ and iCD4-, with iCD4+ expressing higher levels of

1678 the aNKR ligands studied, with the exception of ULBP-2/5/6 and CD86. Co-culture of NK cells

1679 with autologous iCD4 for 7 days revealed that iCD4+ cells were preferentially depleted, leaving

1680 iCD4- virtually untouched, compared to the co-culture of NK cells with unCD4 (Fig. S1).

1681 Together, these findings suggest that HIV-infected T cells expressing CD4 likely constitute

1682 better stimuli for NK cell activation, as they express higher levels of the aNKR ligands studied

1683 here. Their preferential depletion suggests that they are also targeted more effectively by NK cell

1684 than are iCD4- cells

1685 Additionally, we observed that these two infected T cell subsets differed phenotypically

1686 from unCD4, further emphasizing the ability of HIV infection to modulate aNKR ligand

1687 expression. We found that HIV-induced modulation was ligand specific. The expression of the

1688 stress ligands ULBP-1, ULBP-2/5/6, ULBP-3, and MIC-A and CD80 was increased on iCD4+,

1689 but their expression on iCD4- cells decreased to levels comparable to those observed on unCD4.

1690 On the other hand, HIV-infection did not upregulate the expression of MIC-B, CD112, CD155,

1691 and ICAM-2 on total p24+ HIV-infected cells or iCD4+, compared to unCD4, but reduced

1692 expression of these ligands on iCD4- to levels below those observed on unCD4. This may be a

1693 Nef-dependent event. Nef is responsible for the downregulation of CD4 on these iCD4 and has

1694 previously been demonstrated to influence certain aNKR ligands on a variety of HIV-infected

1695 cell types (41, 43, 46). It is well supported that Nef is required for the selective downregulation

1696 of the MHC-I molecules HLA-A and HLA-B and can also block the trafficking of newly

1697 synthesized MHC-I proteins to the cell surface (45, 49, 61-63). Nef is also instrumental in

- 76 -

1698 inducing the loss of cell surface CD155 in T cell lines and CD80 and CD86 in the human

1699 monocytic cell line U937 (55, 58). While HIV infection has been associated with increased

1700 expression of ULBP-1, ULBP-2, and ULBP-3, Nef expression is associated with the

1701 downregulation of NKG2D ligands and expression of ULBP-1, ULBP-2, and MIC-A increases

1702 more dramatically in cells infected with Nef-deficient virus (51, 54). These findings are

1703 consistent with our observation that the increased expression of ULBP-1, ULBP-2/5/6, ULBP-3,

1704 and MIC-A in iCD4+ is lost in parallel with the Nef-mediated downregulation of CD4.

1705 Furthermore, both Nef expression and blocking of interactions between NKG2D and its ligands

1706 have been shown to reduce NK cell-mediated lysis of HIV-infected Jurkat cells (54).

1707 Considering that downmodulation of activating ligands may thus protect HIV-infected cells from

1708 NK cell targeting, our observation that expression of MIC-B, CD112, CD155, and ICAM-2 is

1709 reduced on iCD4- to levels below those observed on unCD4 is of particular interest. Much like

1710 NKG2D signaling, engagement of the aNKR DNAM-1 by its cognate ligand CD155 has recently

1711 been demonstrated to partially regulate NK-cell mediated lysis of HIV-infected CD4+ T cells, a

1712 process that could be disrupted by the downregulation of CD155 (57). It is therefore plausible

1713 that the downregulation of these ligands to aNKRon the surface of iCD4- may confer a survival

1714 advantage to these cells by dampening their ability to activate NK-cell mediated anti-HIV

1715 responses. Nevertheless, the regulation of these ligands by Nef and other viral proteins is a

1716 complex process and, at this time, the specific contribution of these aNKR ligands to NK

1717 activation and antiviral NK cell responses and the ways in which infection might influence them

1718 remains unclear.

1719 Notably, we found that only ICAM-1 expression was consistently increased across all

1720 infected cell subsets, regardless of CD4 expression. ICAM-1 and ICAM-2 are adhesion

- 77 -

1721 molecules that both contribute to the formation of the immunological synapse between an NK

1722 and target cell, which facilitates NK cell activation (27-29). However, expression of ICAM-1,

1723 but not ICAM-2, was also shown to be required for efficient dendritic cell-mediated HIV

1724 transmission and increases infectivity by almost 10-fold (64, 65). It is possible that the consistent

1725 upregulation of ICAM-1 on all infected cell subsets contributes to improved viral transmission

1726 and may therefore be favorable to viral propagation, despite its effects on NK cell activation.

1727 Furthermore, ICAM-1 was the only aNKR ligand studied whose frequency and intensity of

1728 expression positively correlated with the intensity of p24 staining in both iCD4 subsets and in

1729 CD4T cells (Fig. S2). Nef has been demonstrated to increase p24 levels in HIV-infected primary

1730 CD4+ T cells and ICAM-1 was also observed to be overexpressed in Nef+ HIV-infected

1731 endothelial cells (66, 67). These findings and our observations that HIV induces significantly

1732 different aNKR ligand expression phenotypes on iCD4+ and iCD4- cells, suggest that the activity

1733 of the viral protein Nef may be the underlying factor regulating aNKR ligand expression in HIV-

1734 infected CD4+ T cells. It is also possible that iCD4+ and iCD4- may be at different stages of

1735 infection, as the time from infection to the production of Nef has been shown to vary within

1736 infected CD4+ T cells (68). However, the exact relationship between the timing of Nef

1737 production and CD4 downregulation remains incompletely understood and additional work

1738 characterizing the activity of this viral protein in both iCD4+ and iCD4- and its association with

1739 aNKR ligand expression will be required to confirm these possibilities.

1740 Although our study focuses on these ligands in the context of NK cell activation, it is

1741 important to consider the contribution of several of the ligands studied here to inhibitory

1742 signaling. For example, both CD112 and CD155 are also able to engage the iNKR, T cell

1743 immunoreceptor with immunoglobulin and ITIM motifs (TIGIT) on NK cells (69, 70). The

- 78 -

1744 affinity of TIGIT, which is widely expressed on human NK cells, for CD112 and CD155 is

1745 greater than that of the activating NK cell receptor DNAM-1 (70-72). Furthermore, HLA-E, the

1746 expression of which was not significantly modulated by HIV infection, can signal through both

1747 inhibitory (NKG2A) and activating (NKG2E and -C) members of the NKG2x/CD94 family of

1748 NK cell receptors (35). Finally, HLA-C, which we and others have observed remains expressed

+ 1749 on HIV-infected CD4 T cells, engages the inhibitory killer immunoglobulin receptors

1750 KIR2DL1, -2, and -3, limiting NK cell activation by infected cells (45).

1751 Activation of NK cells both in vitro and in vivo is a complex process that depends on the

1752 balance and potency of the different activating and inhibitory signals transmitted upon

1753 interaction with a potential target cell. While our findings provide the foundation for an

1754 improved understanding of the way HIV infection can impact NK cell activation through the

1755 modulation of the expression of activating ligands, the complete picture of how these changes

1756 contribute to activating signaling and how this might integrate with potential inhibitory signaling

1757 remains to be discovered.

- 79 -

1758 3.7: Figure legends

1759 Figure 1: Gating strategy. Gating strategy identifying live singlet CD3+ T lymphocytes (A).

1760 HIV-uninfected CD4+ T cells were identified from this gate (B), The different subsets of HIV-

1761 infected CD4+ T cells were also identified from this gate (C). (D) Comparison of the mean

1762 fluorescence intensity of p24 expression on HIV-infected p24+CD4+ (iCD4+) and HIV-infected

1763 p24+CD4- (iCD4-) T cells (n=17). A Wilcoxon test was used to determine significance of within

1764 subject differences for the indicated T cell subsets. Each data point represents a separate

1765 infection of CD4+ T cells isolated from one individual. Bar height and error bars represent the

1766 mean and standard deviation for the data set. Significant values are shown; “****” = p< 0.0001.

1767 Figure 2: Frequencies of iCD4+ vs iCD4- expressing aNKR ligands. Comparison of the

1768 frequency of HIV-infected p24+CD4+ (iCD4+) and p24+CD4- (iCD4-) T cells expressing ligands

1769 specific for Panel 1, 2, and 3 Abs. Panel 1 (A) includes ULBP-1 (n=11), ULBP-2/5/6 (n=11),

1770 ULBP-3 (n=11), MIC-A (n=9), and MIC-B (n=9). Panel 2 (B) includes CD48, CD80, CD86,

1771 CD112, and CD155 (n=9, for all). Panel 3 (C) includes ICAM-1 and ICAM-2 (n=9, both). The

1772 percent of cells expressing the indicated ligand is represented on the y-axis. A Wilcoxon test was

1773 used to determine significance of within subject differences for the indicated T cell subsets. Each

1774 data point represents T cells isolated from a separate individual. Significant values are shown;

1775 “**” = p< 0.01; “***” = p< 0.001.

1776 Figure 3: Frequencies of all T cell subsets expressing aNKR ligands. Comparison of the

1777 frequency of HIV-infected p24+CD4+ T cells (iCD4+), p24+CD4- T cells (iCD4-), p24+ T cells

1778 (iCD4T), and HIV-uninfected p24-CD4+ T cells (unCD4) expressing Panel 1, 2, and 3 ligands.

1779 Panel 1 (A) includes ULBP-1 (n=11), ULBP-2/5/6 (n=11), ULBP-3 (n=11), MIC-A (n=9), and

- 80 -

1780 MIC-B (n=9). Panel 2 (B) includes CD48, CD80, CD86, CD112, and CD155 (n=9, for all).

1781 Panel 3 (C) includes ICAM-1 and ICAM-2 (n=9, for both). The percent of cells expressing the

1782 indicated ligand is represented on the y-axis. Friedman (PFriedman) and Dunn’s post (*) tests were

1783 used to determine significance of within subject differences between data sets. Each data point

1784 represents T cells isolated from a separate individual. Bar height and error bars represent the

1785 mean and standard deviation for the data set. Significant values are shown; “*” = p< 0.05; “**” =

1786 p< 0.01; “***” = p< 0.001; “****” = p< 0.0001.

1787 Figure 4: Mean fluorescence intensity of aNKR ligand expressing T cell subsets.

1788 Comparison of the per-cell expression of Panel 1, 2, and 3 ligands to aNKR on HIV-infected

1789 p24+CD4+ T cells (iCD4+), p24+CD4- T cells (iCD4-), total p24+ T cells (iCD4T), and HIV-

1790 uninfected p24-CD4+ T cells (unCD4). Panel 1 (A) includes ULBP-1 (n=11), ULBP-2/5/6

1791 (n=11), ULBP-3 (n=11), MIC-A (n=9), and MIC-B (n=9). Panel 2 (B) includes CD48, CD80,

1792 CD86, CD112, and CD155 (n=9, for all). Panel 3 (C) includes ICAM-1 and ICAM-2 (n=9, for

1793 both). The mean fluorescence intensity (MFI) of the expression of the indicated ligand is

1794 represented on the y-axis. Friedman (PFriedman) and Dunn’s post (*) tests were used to determine

1795 significance of within subject differences between data sets. Each data point represents T cells

1796 isolated from a separate individual. Bar height and error bars represent the mean and standard

1797 deviation for the data set. Significant values are shown; “*” = p< 0.05; “**” = p< 0.01; “***” =

1798 p< 0.001; “****” = p< 0.0001.

1799 Figure 5: Expression profile of the HLA molecules HLA-E, HLA-A2, and HLA-C on CD4+

1800 T cell subsets. Panel A shows the frequency of HIV-infected p24+CD4+ (iCD4+) and p24+CD4-

1801 (iCD4-) T cells expressing HLA-E (n=9), HLA-A2 (n=9), and HLA-C (n=4). The frequency (B)

1802 and mean fluorescent intensity (C) of iCD4+, iCD4-, total HIV-infected p24+ T cells (iCD4T) and

- 81 -

1803 uninfected CD4 (unCD4) cells expressing the indicated ligands for the same study subjects as in

1804 (A). The percent and mean fluorescence intensity (MFI) of cells expressing the indicated ligand

1805 is represented on the y-axis. A Wilcoxon test was used to determine significance of within

+ - 1806 subject differences between iCD4 and iCD4 T cell subsets shown in (A). Friedman (PFriedman)

1807 and Dunn’s post (*) tests were used to determine significance of within subject differences

1808 between the data sets shown in panels (B) and (C). Each data point represents T cells isolated

1809 from a separate individual. Bar height and error bars represent the mean and standard deviation

1810 for the data set. Significant values are shown; “*” = p< 0.05; “**” = p< 0.01; “***” = p< 0.001;

1811 “****” = p< 0.0001.

1812 Supplemental Figure 1: NK cells preferentially target HIV-infected CD4 T cells that

1813 conserve cell-surface CD4 expression. The frequency of HIV-infected (p24+) cells within both

1814 iCD4+ and iCD4- T cell subsets from iCD4 cultured alone (white bars) or co-cultured with NK

1815 cells (grey bars) for 10 days (n=11). Friedman (PFriedman) and Dunn’s post (*) tests were used to

1816 determine significance of within subject differences between data sets. Each data point

1817 represents T cells isolated from a separate individual. Significant values are shown; “*” = p<

1818 0.05; “**” = p< 0.01.

1819 Supplemental Figure 2: The frequency and per-cell expression of ICAM-1 is correlated

1820 with HIV-infection on all infected T cell subsets. Correlation of the frequency (%) (top panel)

1821 and mean fluorescence intensity (MFI) (bottom panel) of ICAM-1 expression with the MFI of

1822 p24 expression in (A) iCD4T, (B) iCD4+, and (C) iCD4- HIV infected T cell subsets. Each data

1823 point represents T cells isolated from a separate individual (n=11). Dotted lines represent 95%

1824 confidence intervals. Significant values are shown.

- 82 -

1825 Table 1. Study population HLA genotypes.

HLA-A HLA-B HLA-C

1 01:01, 03:01 44:03, 49 07:01, 16:01

2 24:01, 26:01 15:01, 57:01 05:01, 06:02

3 03:01, 32:01 13:02, 53:01 04:01, 6

4 01:01, 31:01 49:01 07:01

5 02:01, 03:01 07:02, 27:05 01:02, 07:02

6 02:01, 24:02 38:01, 78:01 07:02

7 02:01 07:02, 57:01 05:01, 06:02

8 03:01, 11:01 40, 57:01 02:02, 04:01

9 01:01, 23:01 14:02, 38:01 08:02, 12:02

10 02:01, 29:02 07:02, 44:03 07:02,16:01

11 02:01, 03:01 27:05, 47:01 02:02, 06:02

12 02:01, 03:01 07:02, 50:01 07:02, 16:02

13 01:01, 02:01 08:01, 40:01 03:02, 07:01

14 02:01 07:02, 08:01 07:01, 07:02

15 02:01, 30:02 07:02, 35:01 04:01, 07:02

16 03:01, 11:01 07:02, 35:01 04:01, 07:02

17 02:01, 24:02 44:02, 01:01 05:01, 08:02

- 83 -

1826 Table 2: Description of the aNKR ligands studied.

Number of Patients Panel Ligand Analyzed ULBP-1 11 ULBP-2/5/6 11 Panel 1 ULBP-3 11 MIC-A 9 MIC-B 9 CD48 9 CD80 9 Panel 2 CD86 9 CD112 9 CD155 9

Panel 3 ICAM-1 11 ICAM-2 9 HLA-E 9 HLA HLA-A2 9 HLA-C 4

a

- 84 -

1827 Figure 1: Gating strategy.

- 85 -

1828 Figure 2: Frequencies of iCD4+ vs iCD4- expressing aNKR ligands.

- 86 -

1829 Figure 3: Frequencies of all T cell subsets expressing aNKR ligands.

- 87 -

1830 Figure 4: Mean fluorescence intensity of aNKR ligand expressing T cell subsets.

a

- 88 -

1831 Figure 5: Expression profile of the HLA molecules HLA-E, HLA-A2, and HLA-C on CD4+

1832 T cell subsets.

a

- 89 -

1833 Figure S1: NK cells preferentially target HIV-infected CD4 T cells that conserve cell-

1834 surface CD4 expression.

- 90 -

1835 Figure S2: The frequency and per-cell expression of ICAM-1 is correlated with HIV-

1836 infection on all infected T cell subsets.

- 91 -

3.8: References

1. Vivier E, Tomasello E, Baratin M, Walzer T, Ugolini S. 2008. Functions of natural killer cells. Nat Immunol 9:503-510. 2. Vivier E, Raulet DH, Moretta A, Caligiuri MA, Zitvogel L, Lanier LL, Yokoyama WM, Ugolini S. 2011. Innate or Adaptive Immunity? The Example of Natural Killer Cells. Science 331:44-49. 3. Cheng M, Chen Y, Xiao W, Sun R, Tian Z. 2013. NK cell-based immunotherapy for malignant diseases. Cell Mol Immunol 10:230-252. 4. Fauriat C, Long EO, Ljunggren H-G, Bryceson YT. 2010. Regulation of human NK-cell cytokine and chemokine production by target cell recognition. Blood 115:2167-2176. 5. Lanier LL. 2004. NK CELL RECOGNITION. Annual Review of Immunology 23:225-274. 6. Watzl C, Stebbins CC, Long EO. 2000. NK cell inhibitory receptors prevent tyrosine phosphorylation of the activation receptor 2B4 (CD244). J Immunol 165:3545-3548. 7. Vivier E, Ugolini S, Blaise D, Chabannon C, Brossay L. 2012. Targeting natural killer cells and natural killer T cells in cancer. Nat Rev Immunol 12:239-252. 8. Lanier LL. 2005. NK cell recognition. Annu Rev Immunol 23:225-274. 9. Moretta A, Bottino C, Vitale M, Pende D, Cantoni C, Mingari MC, Biassoni R, Moretta L. 2001. Activating receptors and coreceptors involved in human natural killer cell-mediated cytolysis. Annu Rev Immunol 19:197-223. 10. Bauer S, Groh V, Wu J, Steinle A, Phillips JH, Lanier LL, Spies T. 1999. Activation of NK cells and T cells by NKG2D, a receptor for stress-inducible MICA. Science 285:727-729. 11. Cosman D, Mullberg J, Sutherland CL, Chin W, Armitage R, Fanslow W, Kubin M, Chalupny NJ. 2001. ULBPs, novel MHC class I-related molecules, bind to CMV glycoprotein UL16 and stimulate NK cytotoxicity through the NKG2D receptor. Immunity 14:123-133. 12. Gasser S, Orsulic S, Brown EJ, Raulet DH. 2005. The DNA damage pathway regulates innate immune system ligands of the NKG2D receptor. Nature 436:1186-1190. 13. Sivori S, Vitale M, Morelli L, Sanseverino L, Augugliaro R, Bottino C, Moretta L, Moretta A. 1997. p46, a Novel Natural Killer Cell–specific Surface Molecule That Mediates Cell Activation. The Journal of Experimental Medicine 186:1129-1136. 14. Vitale M, Bottino C, Sivori S, Sanseverino L, Castriconi R, Marcenaro E, Augugliaro R, Moretta L, Moretta A. 1998. NKp44, a novel triggering surface molecule specifically expressed by activated natural killer cells, is involved in non-major histocompatibility complex-restricted tumor cell lysis. J Exp Med 187:2065-2072. 15. Pende D, Parolini S, Pessino A, Sivori S, Augugliaro R, Morelli L, Marcenaro E, Accame L, Malaspina A, Biassoni R, Bottino C, Moretta L, Moretta A. 1999. Identification and molecular characterization of NKp30, a novel triggering receptor involved in natural cytotoxicity mediated by human natural killer cells. J Exp Med 190:1505-1516. 16. Mandelboim O, Lieberman N, Lev M, Paul L, Arnon TI, Bushkin Y, Davis DM, Strominger JL, Yewdell JW, Porgador A. 2001. Recognition of haemagglutinins on virus-infected cells by NKp46 activates lysis by human NK cells. Nature 409:1055-1060. 17. Bloushtain N, Qimron U, Bar-Ilan A, Hershkovitz O, Gazit R, Fima E, Korc M, Vlodavsky I, Bovin NV, Porgador A. 2004. Membrane-associated heparan sulfate proteoglycans are involved in the recognition of cellular targets by NKp30 and NKp46. J Immunol 173:2392-2401. 18. Arnon TI, Lev M, Katz G, Chernobrov Y, Porgador A, Mandelboim O. 2001. Recognition of viral hemagglutinins by NKp44 but not by NKp30. Eur J Immunol 31:2680-2689. 19. Kruse PH, Matta J, Ugolini S, Vivier E. 2014. Natural cytotoxicity receptors and their ligands. Immunol Cell Biol 92:221-229. 20. Hecht ML, Rosental B, Horlacher T, Hershkovitz O, De Paz JL, Noti C, Schauer S, Porgador A, Seeberger PH. 2009. Natural cytotoxicity receptors NKp30, NKp44 and NKp46 bind to different heparan sulfate/heparin sequences. J Proteome Res 8:712-720.

92

21. Brown MH, Boles K, van der Merwe PA, Kumar V, Mathew PA, Barclay AN. 1998. 2B4, the natural killer and T cell immunoglobulin superfamily surface protein, is a ligand for CD48. J Exp Med 188:2083-2090. 22. Bottino C, Castriconi R, Pende D, Rivera P, Nanni M, Carnemolla B, Cantoni C, Grassi J, Marcenaro S, Reymond N, Vitale M, Moretta L, Lopez M, Moretta A. 2003. Identification of PVR (CD155) and Nectin-2 (CD112) as cell surface ligands for the human DNAM-1 (CD226) activating molecule. J Exp Med 198:557-567. 23. Pende D, Bottino C, Castriconi R, Cantoni C, Marcenaro S, Rivera P, Spaggiari GM, Dondero A, Carnemolla B, Reymond N, Mingari MC, Lopez M, Moretta L, Moretta A. 2005. PVR (CD155) and Nectin-2 (CD112) as ligands of the human DNAM-1 (CD226) activating receptor: involvement in tumor cell lysis. Mol Immunol 42:463-469. 24. Tahara-Hanaoka S, Shibuya K, Onoda Y, Zhang H, Yamazaki S, Miyamoto A, Honda S, Lanier LL, Shibuya A. 2004. Functional characterization of DNAM-1 (CD226) interaction with its ligands PVR (CD155) and nectin-2 (PRR-2/CD112). Int Immunol 16:533-538. 25. Shibuya A, Campbell D, Hannum C, Yssel H, Franz-Bacon K, McClanahan T, Kitamura T, Nicholl J, Sutherland GR, Lanier LL, Phillips JH. 1996. DNAM-1, a novel adhesion molecule involved in the cytolytic function of T lymphocytes. Immunity 4:573-581. 26. Shibuya K, Lanier LL, Phillips JH, Ochs HD, Shimizu K, Nakayama E, Nakauchi H, Shibuya A. 1999. Physical and functional association of LFA-1 with DNAM-1 adhesion molecule. Immunity 11:615-623. 27. Gross CC, Brzostowski JA, Liu D, Long EO. 2010. Tethering of intercellular adhesion molecule on target cells is required for LFA-1-dependent NK cell adhesion and granule polarization. J Immunol 185:2918-2926. 28. Schleinitz N, March ME, Long EO. 2008. Recruitment of Activation Receptors at Inhibitory NK Cell Immune Synapses. PLOS ONE 3:e3278. 29. Culley FJ, Johnson M, Evans JH, Kumar S, Crilly R, Casasbuenas J, Schnyder T, Mehrabi M, Deonarain MP, Ushakov DS, Braud V, Roth G, Brock R, Kohler K, Davis DM. 2009. Natural killer cell signal integration balances synapse symmetry and migration. PLoS Biol 7:e1000159. 30. Luque I, Reyburn H, Strominger JL. 2000. Expression of the CD80 and CD86 molecules enhances cytotoxicity by human natural killer cells. Hum Immunol 61:721-728. 31. Wilson JL, Charo J, Martin-Fontecha A, Dellabona P, Casorati G, Chambers BJ, Kiessling R, Bejarano MT, Ljunggren HG. 1999. NK cell triggering by the human costimulatory molecules CD80 and CD86. J Immunol 163:4207-4212. 32. Galea-Lauri J, Darling D, Gan SU, Krivochtchapov L, Kuiper M, Gaken J, Souberbielle B, Farzaneh F. 1999. Expression of a variant of CD28 on a subpopulation of human NK cells: implications for B7-mediated stimulation of NK cells. J Immunol 163:62-70. 33. Costa C, Barber DF, Fodor WL. 2002. Human NK cell-mediated cytotoxicity triggered by CD86 and Gal alpha 1,3-Gal is inhibited in genetically modified porcine cells. J Immunol 168:3808-3816. 34. Fauci AS, Mavilio D, Kottilil S. 2005. NK cells in HIV infection: paradigm for protection or targets for ambush. Nat Rev Immunol 5:835-843. 35. Kaiser BK, Barahmand-Pour F, Paulsene W, Medley S, Geraghty DE, Strong RK. 2005. Interactions between NKG2x immunoreceptors and HLA-E ligands display overlapping affinities and thermodynamics. J Immunol 174:2878-2884. 36. Wada H, Matsumoto N, Maenaka K, Suzuki K, Yamamoto K. 2004. The inhibitory NK cell receptor CD94/NKG2A and the activating receptor CD94/NKG2C bind the top of HLA-E through mostly shared but partly distinct sets of HLA-E residues. Eur J Immunol 34:81-90. 37. Lee N, Llano M, Carretero M, Ishitani A, Navarro F, López-Botet M, Geraghty DE. 1998. HLA-E is a major ligand for the natural killer inhibitory receptor CD94/NKG2A. Proceedings of the National Academy of Sciences 95:5199-5204.

93

38. Alter G, Martin MP, Teigen N, Carr WH, Suscovich TJ, Schneidewind A, Streeck H, Waring M, Meier A, Brander C, Lifson JD, Allen TM, Carrington M, Altfeld M. 2007. Differential natural killer cell–mediated inhibition of HIV-1 replication based on distinct KIR/HLA subtypes. The Journal of Experimental Medicine 204:3027-3036. 39. Song R, Lisovsky I, Lebouche B, Routy JP, Bruneau J, Bernard NF. 2014. HIV protective KIR3DL1/S1-HLA-B genotypes influence NK cell-mediated inhibition of HIV replication in autologous CD4 targets. PLoS Pathog 10:e1003867. 40. Garcia-Beltran WF, Holzemer A, Martrus G, Chung AW, Pacheco Y, Simoneau CR, Rucevic M, Lamothe-Molina PA, Pertel T, Kim TE, Dugan H, Alter G, Dechanet-Merville J, Jost S, Carrington M, Altfeld M. 2016. Open conformers of HLA-F are high-affinity ligands of the activating NK-cell receptor KIR3DS1. Nat Immunol 17:1067-1074. 41. Chaudhuri R, Lindwasser OW, Smith WJ, Hurley JH, Bonifacino JS. 2007. Downregulation of CD4 by human immunodeficiency virus type 1 Nef is dependent on clathrin and involves direct interaction of Nef with the AP2 clathrin adaptor. J Virol 81:3877-3890. 42. Bresnahan PA, Yonemoto W, Ferrell S, Williams-Herman D, Geleziunas R, Greene WC. 1998. A dileucine motif in HIV-1 Nef acts as an internalization signal for CD4 downregulation and binds the AP-1 clathrin adaptor. Curr Biol 8:1235-1238. 43. Lindwasser OW, Chaudhuri R, Bonifacino JS. 2007. Mechanisms of CD4 downregulation by the Nef and Vpu proteins of primate immunodeficiency viruses. Curr Mol Med 7:171-184. 44. Huard B, Fruh K. 2000. A role for MHC class I down-regulation in NK cell lysis of herpes virus-infected cells. Eur J Immunol 30:509-515. 45. Cohen GB, Gandhi RT, Davis DM, Mandelboim O, Chen BK, Strominger JL, Baltimore D. 1999. The selective downregulation of class I major histocompatibility complex proteins by HIV- 1 protects HIV-infected cells from NK cells. Immunity 10:661-671. 46. Tanaka M, Ueno T, Nakahara T, Sasaki K, Ishimoto A, Sakai H. 2003. Downregulation of CD4 is required for maintenance of viral infectivity of HIV-1. Virology 311:316-325. 47. Lundquist CA, Tobiume M, Zhou J, Unutmaz D, Aiken C. 2002. Nef-mediated downregulation of CD4 enhances human immunodeficiency virus type 1 replication in primary T lymphocytes. J Virol 76:4625-4633. 48. Nattermann J, Nischalke HD, Hofmeister V, Kupfer B, Ahlenstiel G, Feldmann G, Rockstroh J, Weiss EH, Sauerbruch T, Spengler U. 2005. HIV-1 infection leads to increased HLA-E expression resulting in impaired function of natural killer cells. Antivir Ther 10:95-107. 49. Brown A, Gartner S, Kawano T, Benoit N, Cheng-Mayer C. 2005. HLA-A2 down-regulation on primary human macrophages infected with an M-tropic EGFP-tagged HIV-1 reporter virus. J Leukoc Biol 78:675-685. 50. Apps R, Meng Z, Del Prete GQ, Lifson JD, Zhou M, Carrington M. 2015. Relative Expression Levels of the HLA Class-I Proteins in Normal and HIV-Infected Cells. The Journal of Immunology 194:3594-3600. 51. Ward J, Bonaparte M, Sacks J, Guterman J, Fogli M, Mavilio D, Barker E. 2007. HIV modulates the expression of ligands important in triggering natural killer cell cytotoxic responses on infected primary T-cell blasts. Blood 110:1207-1214. 52. Fogli M, Mavilio D, Brunetta E, Varchetta S, Ata K, Roby G, Kovacs C, Follmann D, Pende D, Ward J, Barker E, Marcenaro E, Moretta A, Fauci AS. 2008. Lysis of endogenously infected CD4+ T cell blasts by rIL-2 activated autologous natural killer cells from HIV-infected viremic individuals. PLoS Pathog 4:e1000101. 53. Ward J, Davis Z, DeHart J, Zimmerman E, Bosque A, Brunetta E, Mavilio D, Planelles V, Barker E. 2009. HIV-1 Vpr triggers natural killer cell-mediated lysis of infected cells through activation of the ATR-mediated DNA damage response. PLoS Pathog 5:e1000613. 54. Cerboni C, Neri F, Casartelli N, Zingoni A, Cosman D, Rossi P, Santoni A, Doria M. 2007. Human immunodeficiency virus 1 Nef protein downmodulates the ligands of the activating receptor NKG2D and inhibits natural killer cell-mediated cytotoxicity. J Gen Virol 88:242-250.

94

55. Bolduan S, Reif T, Schindler M, Schubert U. 2014. HIV-1 Vpu mediated downregulation of CD155 requires alanine residues 10, 14 and 18 of the transmembrane domain. Virology 464- 465:375-384. 56. Matusali G, Potesta M, Santoni A, Cerboni C, Doria M. 2012. The human immunodeficiency virus type 1 Nef and Vpu proteins downregulate the natural killer cell-activating ligand PVR. J Virol 86:4496-4504. 57. Davis ZB, Sowrirajan B, Cogswell A, Ward JP, Planelles V, Barker E. 2016. CD155 on HIV- Infected Cells Is Not Modulated by HIV-1 Vpu and Nef but Synergizes with NKG2D Ligands to Trigger NK Cell Lysis of Autologous Primary HIV-Infected Cells. AIDS Res Hum Retroviruses doi:10.1089/aid.2015.0375. 58. Chaudhry A, Das SR, Hussain A, Mayor S, George A, Bal V, Jameel S, Rath S. 2005. The Nef protein of HIV-1 induces loss of cell surface costimulatory molecules CD80 and CD86 in APCs. J Immunol 175:4566-4574. 59. Ward J, Bonaparte M, Sacks J, Guterman J, Fogli M, Mavilio D, Barker E. 2007. HIV modulates the expression of ligands important in triggering natural killer cell cytotoxic responses on infected primary T-cell blasts. Blood 110:1207-1214. 60. Imbeault M, Giguère K, Ouellet M, Tremblay MJ. 2012. Exon Level Transcriptomic Profiling of HIV-1-Infected CD4+ T Cells Reveals Virus-Induced Genes and Host Environment Favorable for Viral Replication. PLOS Pathogens 8:e1002861. 61. Schwartz O, Marechal V, Le Gall S, Lemonnier F, Heard JM. 1996. Endocytosis of major histocompatibility complex class I molecules is induced by the HIV-1 Nef protein. Nat Med 2:338-342. 62. Le Gall S, Erdtmann L, Benichou S, Berlioz-Torrent C, Liu L, Benarous R, Heard J-M, Schwartz O. 1998. Nef Interacts with the μ Subunit of Clathrin Adaptor Complexes and Reveals a Cryptic Sorting Signal in MHC I Molecules. Immunity 8:483-495. 63. Swann SA, Williams M, Story CM, Bobbitt KR, Fleis R, Collins KL. 2001. HIV-1 Nef blocks transport of MHC class I molecules to the cell surface via a PI 3-kinase-dependent pathway. Virology 282:267-277. 64. Wang JH, Kwas C, Wu L. 2009. Intercellular adhesion molecule 1 (ICAM-1), but not ICAM-2 and -3, is important for dendritic cell-mediated human immunodeficiency virus type 1 transmission. J Virol 83:4195-4204. 65. Fortin JF, Cantin R, Lamontagne G, Tremblay M. 1997. Host-derived ICAM-1 glycoproteins incorporated on human immunodeficiency virus type 1 are biologically active and enhance viral infectivity. J Virol 71:3588-3596. 66. Malbec M, Sourisseau M, Guivel-Benhassine F, Porrot F, Blanchet F, Schwartz O, Casartelli N. 2013. HIV-1 Nef promotes the localization of Gag to the cell membrane and facilitates viral cell-to-cell transfer. Retrovirology 10:80. 67. Yang F, Liu CQ, Qin XL, Shi JJ. 2010. [HIV-1 Nef regulates ICAM-1 expression on endothelial cells via Erk /Mapk signaling pathway]. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi 26:44-46. 68. Petravic J, Ellenberg P, Chan ML, Paukovics G, Smyth RP, Mak J, Davenport MP. 2014. Intracellular dynamics of HIV infection. J Virol 88:1113-1124. 69. Stanietsky N, Rovis TL, Glasner A, Seidel E, Tsukerman P, Yamin R, Enk J, Jonjic S, Mandelboim O. 2013. Mouse TIGIT inhibits NK-cell cytotoxicity upon interaction with PVR. Eur J Immunol 43:2138-2150. 70. Stanietsky N, Simic H, Arapovic J, Toporik A, Levy O, Novik A, Levine Z, Beiman M, Dassa L, Achdout H, Stern-Ginossar N, Tsukerman P, Jonjic S, Mandelboim O. 2009. The interaction of TIGIT with PVR and PVRL2 inhibits human NK cell cytotoxicity. Proc Natl Acad Sci U S A 106:17858-17863.

95

71. Liu S, Zhang H, Li M, Hu D, Li C, Ge B, Jin B, Fan Z. 2013. Recruitment of Grb2 and SHIP1 by the ITT-like motif of TIGIT suppresses granule polarization and cytotoxicity of NK cells. Cell Death Differ 20:456-464. 72. Wang F, Hou H, Wu S, Tang Q, Liu W, Huang M, Yin B, Huang J, Mao L, Lu Y, Sun Z. 2015. TIGIT expression levels on human NK cells correlate with functional heterogeneity among healthy individuals. European Journal of Immunology 45:2886-2897.

96

1837 Chapter 4: Discussion

1838

1839

1840

1841

1842

1843

1844

1845

1846

1847

1848

1849

1850

- 97 -

1851 4.1: Contributions of this work to the field

1852 The results presented in this thesis are, to our knowledge, the first time that the

1853 expression profile of ligands to activating NK cell receptors (aNKR) on the two HLA null cell

1854 lines, K562 and 721, has been compared head to head. Studies that assessed the expression of

1855 individual aNKR ligands on either cell line are present in the literature. However, the incomplete

1856 characterization of the activating ligand phenotypes of K562 and 721 limited our ability to

1857 explain the differences we previously observed in their ability to activate NK cells. Here we

1858 confirmed that K562 and 721 differ in their expression of aNKR ligands and identified profiles

1859 that were characteristic of each cell line. We also pinpointed specific ligand-receptor pairs that

1860 should be studied further, so as to better understand their individual contribution to NK cell

1861 activation and how they shape the overall stimulatory potential of each cell line.

1862 A significant portion of this thesis is also dedicated to studying the modulation of aNKR

1863 ligand expression by HIV infection. Historically, there is a lack of consensus on how to analyze

1864 HIV-infected CD4 T cells (iCD4) by flow cytometry, with the majority of the literature treating

1865 iCD4 as one homogenous population. However, iCD4 clearly comprise two distinct subsets of

1866 infected cells that can be differentiated by their expression (iCD4+) or not (iCD4-) of cell-surface

1867 CD4. Considering this, the results presented in this thesis offer a unique perspective, as they

1868 directly compare the expression of aNKR ligands on both subsets of infected T cells. While it

1869 has been postulated that both iCD4 subsets differ in stage and productivity of HIV infection, to

1870 our knowledge, our findings provide the first concrete evidence that iCD4+ and iCD4- differ

1871 phenotypically.

- 98 -

1872 Advances in NK cell biology in recent years have provided the community with new

1873 avenues to address different areas of HIV treatment and cure. The NK cell-mediated antibody-

1874 dependent cellular cytotoxicity has been identified as an important contributor to the protection

1875 offered by effective HIV vaccines (7). Other NK cell responses have also been associated with a

1876 reduction in the risk of infection and with slowed disease progression (101, 103, 325-327).

1877 Recent work has shown promising evidence that NK cell responses can be directed (328, 329).

1878 To adequately direct NK cell responses and to translate this into efficient therapy, however,

1879 requires a holistic characterization of NK receptor signaling and activation. Despite concerted

1880 efforts, the determinants of a specific NK cell response to challenge with allogeneic,

1881 transformed, or virally-infected cells remain unclear. The research presented in this thesis offers

1882 insight into the ligand receptor interactions that may be important in governing individual NK

1883 cell responses and activation as a whole. We believe that this research will assist future studies of

1884 NK cell activation and contribute to eventually harnessing the therapeutic potential of NK cells

1885 in context of HIV infection and beyond.

- 99 -

1886 4.2: Modulation of cell-surface ligands by HIV Nef

1887 Nef is a myristoylated ~27 kilodalton HIV accessory protein whose production facilitates

1888 the transcription and nuclear export of unspliced or singly-spliced mRNAs encoding Gag, Gag-

1889 Pol, Env, Vpu, Vif, and Vpr (35). Nef is considered to be an HIV early protein and Nef mRNA

1890 can first be detected 20 hours following infection, although a broad distribution of times from

1891 infection to Nef protein production can be observed within HIV-infected CD4+ T cells (330,

1892 331). The Nef protein comprises three general domains: an N-terminal anchor and flexible loop,

1893 a central core, and a C-terminal flexible loop (Reviewed in 332). Both N-terminal myristoylation

1894 and other sequences within the N terminal anchor are crucial for membrane association and, in

1895 conjunction with the central core, are also required for the protein interactions the underlie Nef’s

1896 broad array of functions (333-335). In addition to contributing to viral genomic transcription,

1897 Nef can also influence T cell activation, although these effects are pleiomorphic and may depend

1898 on the cellular context of Nef expression, and increase p24 levels in HIV-infected primary CD4+

1899 T cells, stimulating HIV infectivity (336-338). However, arguably the most well-known function

1900 of Nef is its ability to downregulate the cell-surface expression of the T cell coreceptor CD4

1901 (339). Nef-mediated CD4 downregulation is achieved through the direct interaction of Nef with

1902 the AP-2 clathrin adaptor complex, which induces CD4 endocytosis and degradation in

1903 lysosomes, and requires sequences in the Nef central core that bind to AP-2 and the highly

1904 conserved LL163, 164 and DD174, 175 motifs in the membrane-proximal region of CD4’s

1905 cytoplasmic tail (339-345). This process is facilitated by Vpu, another accessory protein, which

1906 promotes the entry of newly synthesized CD4 into the endoplasmic-reticulum-associated

1907 degradation pathway (11, 58, 59, 346). HIV Nef is also instrumental in the downregulation of the

- 100 -

1908 additional coreceptors CCR5 and CXCR4, which are required for HIV viral entry (347, 348).

1909 Reduced expression of these entry receptors on the surface of infected T cells promotes viral

1910 replication and persistence by preventing superinfection and subsequent cytotoxicity resulting

1911 from the accumulation of multiple integrated viral genomes, by reducing signaling that could

1912 influence viral transcription or induce apoptosis, and by protecting infected cells from antibody-

1913 mediated cell lysis (45, 346-351).

1914 It is well supported that Nef is required for the selective downregulation of the MHC-I

1915 molecules HLA-A and HLA-B, an ability that relies on sequences in the Nef N-terminal alpha

1916 helix and an interdomain binding site (107, 108, 352-356). The mechanism by which Nef

1917 orchestrates MHC-I downregulation is similar to that of Nef-mediated CD4 downregulation,

1918 although the two are functionally dissociated (353, 355). Nef complexes with the cytoplasmic

1919 tail of MHC-I molecules and the μ1 subunit of the AP-1 clathrin adaptor complex to either

1920 disrupt MHC-I trafficking by re-routing HLA-A and -B from the trans-Golgi network to

1921 lysosomes or induce clathrin-dependent endocytosis (107, 357-363). However, the relative

1922 contribution of each of these pathways to MHC-I modulation remains controversial (Reviewed in

1923 364). Moreover, the cytoplasmic domains of HLA-B appear to be more resistant to Nef-mediated

1924 downregulation and so may not be as reliably downregulated (365, 366). Due to the presence of

1925 unique residues at codons 320 and 327 in the cytoplasmic domain of HLA-C molecules, their

1926 surface expression remains unaltered by Nef activity (108). Maintenance of HLA-C at the

1927 surface of infected cells prevents them from being totally void of MHC-I molecules and of, thus,

1928 being eliminated by missing-self NK cell responses (108). While this is true of laboratory strains

1929 of HIV, as is used in the work presented here, HLA-C can be downregulated by primary HIV

- 101 -

1930 clones and, paradoxically, this impairs T cell inhibition of viral replication (10). This is

1931 accomplished by the viral accessory protein Vpu, which selectively downregulates HLA-C

1932 through a mechanism that may resemble HLA-A and -B downregulation by Nef (10). It remains

1933 unclear why laboratory strains and primary isolates differ in this manner, although it has been

1934 suggested that viral targeting of HLA-C reflects an adaptation to immune pressure and that

1935 laboratory-adapted virus propagates in vitro in the absence of such pressure (10). Similarly, a

1936 single amino acid substitution in the cytoplasmic tail of the non-classical MHC HLA-E confers

1937 resistance to Nef-mediated downregulation (108). As was initially believed of HLA-C, sustained

1938 HLA-E expression is thought to ensure continued signaling through its iNKR NKG2A,

1939 preventing NK cell-mediated cytolysis of infected cells (108, 187, 367).

1940 It’s clear that a large part of Nef-mediated modulation of surface protein expression

1941 serves to impair anti-HIV responses, including those of NK cells. In line with this, Nef has also

1942 been shown to modulate the expression of certain aNKR ligands and NK cell activation, by

1943 extension. While primary CD4+ T cells respond to HIV infection by increasing surface

1944 expression of the DNAM-1 ligand, CD155, this is attenuated by Nef, which promotes CD155

1945 downregulation and reduces the NK cell-mediated lysis of infected cells (283). In the Jurkat T

1946 cell line, the downregulation of CD155 by Nef shares many similarities with the Nef-mediated

1947 downregulation of MHC-I molecules, including reduced intracellular accumulation of CD155,

1948 requiring similar conserved residues, and being functionally separate from the Nef-mediated

1949 downregulation of CD4 (283). Notably, the downregulation of CD155 appears to be facilitated

1950 by Vpu, which functions with Nef to localize CD155 to perinuclear compartments, and this

1951 ability requires conserved alanine residues in the Vpu transmembrane domain (283, 368). The

- 102 -

1952 extent of CD155 downregulation, however, may depend on the virus used and on the cell types

1953 infected, as these findings were not observed when using several other HIV strains or when

1954 infecting primary CD4+ T cells (368, 369). Similarly, the expression of NKG2D ligands is

1955 increased by HIV infection, but the surface expression of ULBP-1, ULBP-2, and MIC-A is

1956 downregulated by Nef (144, 176). Downregulation of ULBP-2 and MIC-A appears to be a

1957 conserved function of the laboratory-adapted HIVNL4-3 Nef and from variants obtained from

1958 clinical isolates, however this was not observed for ULBP-1 (176). Additionally, Nef-induced

1959 downregulation of NKG2D ligands likely proceeds through a unique mechanism, as mutations in

1960 residues critical for the downregulation of CD4 and MHC-I molecules had little effect on

1961 NKG2D ligand expression (176). Finally, HIV Nef was observed to upregulate ICAM-1

1962 expression on an endothelial cell line, which requires the protein kinase RNA-like endoplasmic

1963 reticulum kinase (p-ERK) and likely proceeds through extracellular signal-regulated kinase

1964 (ERK)/mitogen-activated protein kinase (MAPK) signaling (370).

1965 Considering these precedents, it seems plausible that, as discussed in Chapter 3, the

1966 different aNKR ligand expression profiles observed on iCD4+ and iCD4- may reflect differences

1967 in the production of Nef. As Nef is responsible for CD4 downregulation and iCD4- are

1968 characterized by their lack of CD4 cell-surface expression, this subset is often considered to be at

+ 1969 a later stage of infection than iCD4 , which have presumably not begun to produce HIV viral

1970 proteins. Considering that Nef expression has not been studied in these individual HIV-infected

1971 T cell subsets, it is possible that Nef protein is being produced in iCD4+, however the sustained

1972 expression of CD4 suggests that Nef is not active in these cells. As such, we speculate that Nef is

- 103 -

1973 capable of modulating the expression of aNKR ligands and that increased expression of Nef in

1974 iCD4- could explain why aNKR ligand expression on these cells differs from iCD4+.

- 104 -

1975 4.3: Limitations and future directions

1976 One limitation of this study is that the ligands studied are only considered in the context

1977 of their capacity to bind aNKRs. The potential for several of the ligands studied here to

1978 potentiate inhibitory signaling, as discussed in Chapters 2 and 3, complicates our ability to

1979 extrapolate how ligand expression might influence NK cell activation. Indeed, the obvious

1980 direction for future work in this vein is to address how the aNKR ligand phenotype of the two

1981 HLA-null cell lines studied, K562 and 721, and of iCD4 contributes to NK cell activation. This

1982 can be addressed by additional functional analyses using an NK cell stimulation assay,

1983 previously employed by our lab, to determine the minimal requirements for NK cell activation

1984 (5). Briefly, the percentages of all combinations of cytokine secreting (measured by IFN-γ

1985 expression), chemokine secreting (measured by CCL4 expression), and degranulating (measured

1986 by CD107α expression) NK cells would be assessed by flow cytometry following stimulation

1987 with K562, 721, and iCD4 in the presence or absence of blocking reagents that prevent the

1988 interaction of defined aNKR/ligand pairs. In this context, a significant reduction of a functional

1989 subset in the presence of defined combinations of blocking reagents would be interpreted as

1990 evidence that the targeted aNKR-ligand pairs are required for that NK cell activation stage and

1991 function. Such studies would be particularly useful in confirming that the aNKR ligand profiles

1992 that we observed to be characteristic of K562 or 721 (Figure 1 and 2, Chapter 2) are directly

1993 related to the ability of each HLA-null cell line to activate different numbers and functional

1994 subsets of NK cells.

1995 Furthermore, an experimental framework exists that would allow us to determine which

1996 aNKR-ligand pairs regulate individual stages of NK cell activation, including adhesion, granule

- 105 -

1997 polarization, actin mobilization, and degranulation. Such experiments would complement the

1998 work described above by contributing more direct data on the aNKR-ligand requirements for

1999 NK-cell mediated cytolysis. Briefly, this method, which was pioneered by Eric O. Long’s group,

2000 involves using Drosophila Schneider cell (SC) 2 cells transfected with individual or

2001 combinations of ligands as target cells (294). This method overcomes the complexity of NK cell

2002 signaling by reducing the multiplicity of receptor-ligand interactions that is observed with

2003 mammalian target cells to one receptor-ligand pair. This approach also dissects total NK cell

2004 responsiveness by using cell-surface expression of CD107α as a marker of degranulation, but

2005 also measures ADCC – for which we have existing in house protocols – and uses confocal

2006 microscopy to analyze the polarization of perforin-containing granules (294). Additionally, the

2007 ability of receptor-ligand pairs to promote adhesion can be observed by measuring their ability to

2008 induce LFA-1 conformational change through what are known as “inside-out” signals (371).

2009 Such methodology has already provided some insight into the importance of certain aNKR-

2010 ligand pairs. Target cells individually expressing ICAM-1 and ULBP-1 were found to induce

2011 granule polarization, but not degranulation, while CD48-2B4 signaling induced weak

2012 polarization and no degranulation (294, 372). Furthermore, concurrent ULBP-1-NKG2D, CD48-

2013 2B4, and ICAM-1-LFA-1 signaling defined a minimal requirement for NK cell cytotoxicity,

2014 although none of these receptor-ligand pairs could induce degranulation individually (371, 373).

2015 However, we believe that valuable data could be gained by using this method to study the ways

2016 in which the aNKR-ligand pairs that were studied here, but absent from previous studies,

2017 influence the distinct stages of NK cell activation, which remains undefined.

- 106 -

2018 One of the chief findings of this research is that HIV-infected T cells that maintain cell

2019 surface CD4 (iCD4+) or not (iCD4-) differ in their expression of the aNKR ligands studied

2020 (Figure 2 and 4, Chapter 3). Specifically, we observed that iCD4- expressed lower levels of

2021 aNKR ligands, compared to iCD4+. While we can speculate that these differences reflect

2022 variations in the activity of the HIV viral accessory protein Nef, which also downregulates cell

2023 surface CD4, we are limited by the lack of data characterizing these two unique HIV-infected T

2024 cell subsets. Transcriptomic analyses using microarrays and Western blots have previously been

2025 utilized to assess viral gene expression in HIV infected cells (374, 375). A similar approach to

+ - 2026 analyzing iCD4 and iCD4 , could help confirm the mechanism by which HIV modulates aNKR

2027 ligand expression on both infected T cell subsets. Additionally, we observed that iCD4+ were

2028 preferentially targeted by NK cell-mediated inhibition of viral replication (Supplemental Figure

2029 1, Chapter 3). We hypothesize that the susceptibility of iCD4+ to NK cell anti-HIV responses

2030 results from their increased expression of aNKR ligands and, therefore, superior ability to

2031 activate NK cells. However, future experiments that measure NK cell activation induced by

+ - 2032 sorted iCD4 and iCD4 – as could be accomplished by the experimental set up described above

2033 – would help support our theory.

- 107 -

2034 4.4: Conclusion

2035 The results presented here elaborate a more complete phenotypic characterization of the

2036 cell types commonly used to stimulate NK cell activity, including the HLA-null cell lines K562

2037 and 721 and HIV-infected CD4+ T cells. We confirmed that K562 and 721 each have

2038 characteristic expression profiles of ligands to aNKRs. Furthermore, each profile was capable of

2039 engaging different numbers and subsets of aNKRs, further supporting our hypothesis that

2040 underlying differences in aNKR ligand expression explained the differences in the NK cell

2041 stimulating capacities of K562 and 721 that we previously observed. We have also demonstrated

2042 that HIV infection significantly influenced the expression profile of aNKR ligands of infected

2043 CD4+ T cells in a ligand specific manner. Our head to head comparisons of infected T cells that

2044 maintained CD4 expression (iCD4+) or not (iCD4-) also showed that aNKR ligand expression

2045 differed between these two infected subsets. This finding suggested that differences in the

2046 kinetics of HIV infection may also be important in regulating ligand expression on the surface of

2047 infected cells and would be strengthened by further characterizations of iCD4+ and iCD4-. While

2048 our work offers important insight into how these three cell types might stimulate NK cells,

2049 additional work needs to be done to confirm the contribution of each ligand-aNKR pair to NK

2050 cell activation and anti-HIV responses. Clarifying the mechanisms of NK cell activation,

2051 particularly in the context of HIV infection, will improve our understanding of the mechanisms

2052 of protection against HIV and may identify novel therapeutic targets.

- 108 -

Appendix A: Antibodies used in the analysis of aNKR ligand expression

Antibody Conjugation Clone Manufacturer Live/Dead Blue/Indo-Violet N/A Thermofisher Anti-human CD3 BV785 OKT3 BioLegend Anti-human CD4 BUV737 SK3 BD Biosciences Anti-human p24 FITC KC57 Beckman-Coulter Anti-human ULBP-1 PerCP 170818 R&D Systems Anti-human ULBP-2/5/6 APC 165903 R&D Systems Anti-human ULBP-3 PE 166510 R&D Systems Anti-human HLA-E PE-Cy7 3D12 BioLegend Anti-human ICAM-1 Pacific Blue HCD54 BioLegend Anti-human HLA-A2 APC BB7.2 eBioscience Anti-human CD80 BV421 2D10 BioLegend Anti-human CD86 PE-Dazzle594 IT2.2 BioLegend Anti-human CD112 Alexa Fluor 700 610603 Cedarlane Anti-human CD155 PE-Cy7 SKII.4 BioLegend Anti-human ICAM-2 PE CBR-1C2/2 BioLegend Anti-human MICA APC 159227 R&D Systems Anti-human MICB Alexa Fluor 700 236511 R&D Systems Anti-human CD48 PE BJ40 BioLegend Anti-human HLA-C N/A DT9 EMD Millipore Anti-human HLA-F N/A 3D11 N/A Recombinant human KIR3DS1-Fc N/A N/A R&D Systems Recombinant human NKp30-Fc N/A N/A R&D Systems Recombinant human NKp44-Fc N/A N/A R&D Systems Recombinant human NKp46-Fc N/A N/A R&D Systems Anti-human IgG (Fc gamma-specific) PE Polyclonal eBioscience F(ab’)2 anti-mouse IgG APC Polyclonal eBioscience

- 109 -

Appendix B: Panel of the ligands studied and their respective activating NK cell receptors

- 110 -

References 1. Vivier E, Tomasello E, Baratin M, Walzer T, Ugolini S. 2008. Functions of natural killer cells. Nat Immunol 9:503-510. 2. Fauriat C, Long EO, Ljunggren H-G, Bryceson YT. 2010. Regulation of human NK-cell cytokine and chemokine production by target cell recognition. Blood 115:2167-2176. 3. Lanier LL. 2005. NK cell recognition. Annu Rev Immunol 23:225-274. 4. Lisovsky I, Isitman G, Bruneau J, Bernard NF. 2015. Functional analysis of NK cell subsets activated by 721.221 and K562 HLA-null cells. J Leukoc Biol 97:761-767. 5. Song R, Lisovsky I, Lebouche B, Routy JP, Bruneau J, Bernard NF. 2014. HIV protective KIR3DL1/S1-HLA-B genotypes influence NK cell-mediated inhibition of HIV replication in autologous CD4 targets. PLoS Pathog 10:e1003867. 6. Rerks-Ngarm S, Pitisuttithum P, Nitayaphan S, Kaewkungwal J, Chiu J, Paris R, Premsri N, Namwat C, de Souza M, Adams E, Benenson M, Gurunathan S, Tartaglia J, McNeil JG, Francis DP, Stablein D, Birx DL, Chunsuttiwat S, Khamboonruang C, Thongcharoen P, Robb ML, Michael NL, Kunasol P, Kim JH. 2009. Vaccination with ALVAC and AIDSVAX to Prevent HIV-1 Infection in Thailand. New England Journal of Medicine 361:2209-2220. 7. Haynes BF, Gilbert PB, McElrath MJ, Zolla-Pazner S, Tomaras GD, Alam SM, Evans DT, Montefiori DC, Karnasuta C, Sutthent R, Liao H-X, DeVico AL, Lewis GK, Williams C, Pinter A, Fong Y, Janes H, DeCamp A, Huang Y, Rao M, Billings E, Karasavvas N, Robb ML, Ngauy V, de Souza MS, Paris R, Ferrari G, Bailer RT, Soderberg KA, Andrews C, Berman PW, Frahm N, De Rosa SC, Alpert MD, Yates NL, Shen X, Koup RA, Pitisuttithum P, Kaewkungwal J, Nitayaphan S, Rerks-Ngarm S, Michael NL, Kim JH. 2012. Immune-Correlates Analysis of an HIV-1 Vaccine Efficacy Trial. New England Journal of Medicine 366:1275-1286. 8. Garcia-Beltran WF, Holzemer A, Martrus G, Chung AW, Pacheco Y, Simoneau CR, Rucevic M, Lamothe-Molina PA, Pertel T, Kim TE, Dugan H, Alter G, Dechanet-Merville J, Jost S, Carrington M, Altfeld M. 2016. Open conformers of HLA-F are high-affinity ligands of the activating NK-cell receptor KIR3DS1. Nat Immunol 17:1067-1074. 9. Burian A, Wang KL, Finton KAK, Lee N, Ishitani A, Strong RK, Geraghty DE. 2016. HLA- F and MHC-I Open Conformers Bind Natural Killer Cell Ig-Like Receptor KIR3DS1. PLoS ONE 11:e0163297. 10. Apps R, Del Prete GQ, Chatterjee P, Lara A, Brumme ZL, Brockman MA, Neil S, Pickering S, Schneider DK, Piechocka-Trocha A, Walker BD, Thomas R, Shaw GM, Hahn BH, Keele BF, Lifson JD, Carrington M. 2016. HIV-1 Vpu Mediates HLA-C Downregulation. Cell host & microbe 19:686-695. 11. Lindwasser OW, Chaudhuri R, Bonifacino JS. 2007. Mechanisms of CD4 downregulation by the Nef and Vpu proteins of primate immunodeficiency viruses. Curr Mol Med 7:171-184. 12. Chen BK, Gandhi RT, Baltimore D. 1996. CD4 down-modulation during infection of human T cells with human immunodeficiency virus type 1 involves independent activities of vpu, env, and nef. J Virol 70:6044-6053. 13. Gottlieb MS, Schroff R, Schanker HM, Weisman JD, Fan PT, Wolf RA, Saxon A. 1981. Pneumocystis carinii pneumonia and mucosal candidiasis in previously healthy homosexual men: evidence of a new acquired cellular immunodeficiency. N Engl J Med 305:1425-1431. 14. Barre-Sinoussi F, Chermann JC, Rey F, Nugeyre MT, Chamaret S, Gruest J, Dauguet C, Axler-Blin C, Vezinet-Brun F, Rouzioux C, Rozenbaum W, Montagnier L. 1983. Isolation of a T-lymphotropic retrovirus from a patient at risk for acquired immune deficiency syndrome (AIDS). Science 220:868-871. 15. UNAIDS. 2016. Fact Sheet: November 2016. http://www.unaids.org/en/resources/fact-sheet. Accessed - 111 -

16. CATIE. 2014. The epidemiology of HIV in Canada. http://www.catie.ca/en/fact- sheets/epidemiology/epidemiology-hiv-canada Accessed 17. PHAC. 2014. Summary: Estimates of HIV Incidence, Prevalence and Proportion Undiagnosed in Canada, 2014. http://healthycanadians.gc.ca/publications/diseases-conditions-maladies- affections/hiv-aids-estimates-2014-vih-sida-estimations/index-eng.php. Accessed 18. PHAC. 2014. HIV/AIDS Epi Updates Chapter 8: HIV/AIDS among Aboriginal people in Canada. http://www.phac-aspc.gc.ca/aids-sida/publication/epi/2010/8-eng.php. Accessed 19. Coffin J, Hughes S, Varmus H. 1997. Retroviruses. Cold Spring Harbor Laboratory Press. 20. Frankel AD, Young JA. 1998. HIV-1: fifteen proteins and an RNA. Annu Rev Biochem 67:1- 25. 21. Ayouba A, Souquieres S, Njinku B, Martin PM, Muller-Trutwin MC, Roques P, Barre- Sinoussi F, Mauclere P, Simon F, Nerrienet E. 2000. HIV-1 group N among HIV-1- seropositive individuals in Cameroon. Aids 14:2623-2625. 22. Gurtler LG, Hauser PH, Eberle J, von Brunn A, Knapp S, Zekeng L, Tsague JM, Kaptue L. 1994. A new subtype of human immunodeficiency virus type 1 (MVP-5180) from Cameroon. J Virol 68:1581-1585. 23. Simon F, Mauclere P, Roques P, Loussert-Ajaka I, Muller-Trutwin MC, Saragosti S, Georges-Courbot MC, Barre-Sinoussi F, Brun-Vezinet F. 1998. Identification of a new human immunodeficiency virus type 1 distinct from group M and group O. Nat Med 4:1032-1037. 24. Buonaguro L, Tornesello ML, Buonaguro FM. 2007. Human Immunodeficiency Virus Type 1 Subtype Distribution in the Worldwide Epidemic: Pathogenetic and Therapeutic Implications. Journal of Virology 81:10209-10219. 25. Nyamweya S, Hegedus A, Jaye A, Rowland-Jones S, Flanagan KL, Macallan DC. 2013. Comparing HIV-1 and HIV-2 infection: Lessons for viral immunopathogenesis. Rev Med Virol 23:221-240. 26. Karlsson Hedestam GB, Fouchier RA, Phogat S, Burton DR, Sodroski J, Wyatt RT. 2008. The challenges of eliciting neutralizing antibodies to HIV-1 and to influenza virus. Nat Rev Microbiol 6:143-155. 27. Peterlin BM, Trono D. 2003. Hide, shield and strike back: how HIV-infected cells avoid immune eradication. Nat Rev Immunol 3:97-107. 28. Royce RA, Seña A, Cates W, Cohen MS. 1997. Sexual Transmission of HIV. New England Journal of Medicine 336:1072-1078. 29. McMichael AJ, Borrow P, Tomaras GD, Goonetilleke N, Haynes BF. 2010. The immune response during acute HIV-1 infection: clues for vaccine development. Nat Rev Immunol 10:11- 23. 30. Miller CJ, Alexander NJ, Sutjipto S, Lackner AA, Gettie A, Hendrickx AG, Lowenstine LJ, Jennings M, Marx PA. 1989. Genital mucosal transmission of simian immunodeficiency virus: animal model for heterosexual transmission of human immunodeficiency virus. J Virol 63:4277- 4284. 31. Miller CJ, Li Q, Abel K, Kim EY, Ma ZM, Wietgrefe S, La Franco-Scheuch L, Compton L, Duan L, Shore MD, Zupancic M, Busch M, Carlis J, Wolinsky S, Haase AT. 2005. Propagation and dissemination of infection after vaginal transmission of simian immunodeficiency virus. J Virol 79:9217-9227. 32. Zhang Z, Schuler T, Zupancic M, Wietgrefe S, Staskus KA, Reimann KA, Reinhart TA, Rogan M, Cavert W, Miller CJ, Veazey RS, Notermans D, Little S, Danner SA, Richman DD, Havlir D, Wong J, Jordan HL, Schacker TW, Racz P, Tenner-Racz K, Letvin NL, Wolinsky S, Haase AT. 1999. Sexual transmission and propagation of SIV and HIV in resting and activated CD4+ T cells. Science 286:1353-1357. - 112 -

33. Izmailova E, Bertley FM, Huang Q, Makori N, Miller CJ, Young RA, Aldovini A. 2003. HIV-1 Tat reprograms immature dendritic cells to express chemoattractants for activated T cells and macrophages. Nat Med 9:191-197. 34. Collman RG, Perno CF, Crowe SM, Stevenson M, Montaner LJ. 2003. HIV and cells of macrophage/dendritic lineage and other non-T cell reservoirs: new answers yield new questions. J Leukoc Biol 74:631-634. 35. Turner BG, Summers MF. 1999. Structural biology of HIV1. Journal of Molecular Biology 285:1-32. 36. Moore JP. 1997. Coreceptors: implications for HIV pathogenesis and therapy. Science 276:51- 52. 37. Sullivan N, Sun Y, Sattentau Q, Thali M, Wu D, Denisova G, Gershoni J, Robinson J, Moore J, Sodroski J. 1998. CD4-Induced Conformational Changes in the Human Immunodeficiency Virus Type 1 gp120 Glycoprotein: Consequences for Virus Entry and Neutralization. Journal of Virology 72:4694-4703. 38. Jones PLSJ, Korte T, Blumenthal R. 1998. Conformational Changes in Cell Surface HIV-1 Envelope Glycoproteins Are Triggered by Cooperation between Cell Surface CD4 and Co- receptors. Journal of Biological Chemistry 273:404-409. 39. Goff SP. 1990. Retroviral reverse transcriptase: synthesis, structure, and function. J Acquir Immune Defic Syndr 3:817-831. 40. Op de Coul EL, Prins M, Cornelissen M, van der Schoot A, Boufassa F, Brettle RP, Hernandez-Aguado L, Schiffer V, McMenamin J, Rezza G, Robertson R, Zangerle R, Goudsmit J, Coutinho RA, Lukashov VV. 2001. Using phylogenetic analysis to trace HIV-1 migration among western European injecting drug users seroconverting from 1984 to 1997. Aids 15:257-266. 41. Roberts JD, Bebenek K, Kunkel TA. 1988. The accuracy of reverse transcriptase from HIV-1. Science 242:1171-1173. 42. Ho DD, Neumann AU, Perelson AS, Chen W, Leonard JM, Markowitz M. 1995. Rapid turnover of plasma virions and CD4 lymphocytes in HIV-1 infection. Nature 373:123-126. 43. Michael NL. 1999. Host genetic influences on HIV-1 pathogenesis. Curr Opin Immunol 11:466- 474. 44. Levin JG, Mitra M, Mascarenhas A, Musier-Forsyth K. 2010. Role of HIV-1 nucleocapsid protein in HIV-1 reverse transcription. RNA Biol 7:754-774. 45. Goncalves J, Korin Y, Zack J, Gabuzda D. 1996. Role of Vif in human immunodeficiency virus type 1 reverse transcription. J Virol 70:8701-8709. 46. Miller MD, Farnet CM, Bushman FD. 1997. Human immunodeficiency virus type 1 preintegration complexes: studies of organization and composition. J Virol 71:5382-5390. 47. Jowett JB, Planelles V, Poon B, Shah NP, Chen ML, Chen IS. 1995. The human immunodeficiency virus type 1 vpr gene arrests infected T cells in the G2 + M phase of the cell cycle. Journal of Virology 69:6304-6313. 48. Nie Z, Bergeron D, Subbramanian RA, Yao XJ, Checroune F, Rougeau N, Cohen EA. 1998. The putative alpha helix 2 of human immunodeficiency virus type 1 Vpr contains a determinant which is responsible for the nuclear translocation of proviral DNA in growth-arrested cells. J Virol 72:4104-4115. 49. Chun T-W, Carruth L, Finzi D, Shen X, DiGiuseppe JA, Taylor H, Hermankova M, Chadwick K, Margolick J, Quinn TC, Kuo Y-H, Brookmeyer R, Zeiger MA, Barditch- Crovo P, Siliciano RF. 1997. Quantification of latent tissue reservoirs and total body viral load in HIV-1 infection. Nature 387:183-188. 50. Siliciano JD, Kajdas J, Finzi D, Quinn TC, Chadwick K, Margolick JB, Kovacs C, Gange SJ, Siliciano RF. 2003. Long-term follow-up studies confirm the stability of the latent reservoir for HIV-1 in resting CD4+ T cells. Nat Med 9:727-728. - 113 -

51. Ruiz L, Martinez-Picado J, Romeu J, Paredes R, Zayat MK, Marfil S, Negredo E, Sirera G, Tural C, Clotet B. 2000. Structured treatment interruption in chronically HIV-1 infected patients after long-term viral suppression. Aids 14:397-403. 52. Wei P, Garber ME, Fang SM, Fischer WH, Jones KA. 1998. A novel CDK9-associated C- type cyclin interacts directly with HIV-1 Tat and mediates its high-affinity, loop-specific binding to TAR RNA. Cell 92:451-462. 53. Herrmann CH, Rice AP. 1995. Lentivirus Tat proteins specifically associate with a cellular protein kinase, TAK, that hyperphosphorylates the carboxyl-terminal domain of the large subunit of RNA polymerase II: candidate for a Tat cofactor. J Virol 69:1612-1620. 54. Emerman M, Malim MH. 1998. HIV-1 regulatory/accessory genes: keys to unraveling viral and host cell biology. Science 280:1880-1884. 55. Zapp ML, Green MR. 1989. Sequence-specific RNA binding by the HIV-1 Rev protein. Nature 342:714-716. 56. Freed EO. 2015. HIV-1 assembly, release and maturation. Nat Rev Micro 13:484-496. 57. Hoxie JA, Alpers JD, Rackowski JL, Huebner K, Haggarty BS, Cedarbaum AJ, Reed JC. 1986. Alterations in T4 (CD4) protein and mRNA synthesis in cells infected with HIV. Science 234:1123-1127. 58. Schubert U, Anton LC, Bacik I, Cox JH, Bour S, Bennink JR, Orlowski M, Strebel K, Yewdell JW. 1998. CD4 glycoprotein degradation induced by human immunodeficiency virus type 1 Vpu protein requires the function of proteasomes and the ubiquitin-conjugating pathway. J Virol 72:2280-2288. 59. Margottin F, Bour SP, Durand H, Selig L, Benichou S, Richard V, Thomas D, Strebel K, Benarous R. 1998. A novel human WD protein, h-beta TrCp, that interacts with HIV-1 Vpu connects CD4 to the ER degradation pathway through an F-box motif. Mol Cell 1:565-574. 60. Crise B, Buonocore L, Rose JK. 1990. CD4 is retained in the endoplasmic reticulum by the human immunodeficiency virus type 1 glycoprotein precursor. J Virol 64:5585-5593. 61. Goldsmith MA, Warmerdam MT, Atchison RE, Miller MD, Greene WC. 1995. Dissociation of the CD4 downregulation and viral infectivity enhancement functions of human immunodeficiency virus type 1 Nef. J Virol 69:4112-4121. 62. Deeks SG, Overbaugh J, Phillips A, Buchbinder S. 2015. HIV infection. Nature Reviews Disease Primers 1:15035. 63. Tindall B, Cooper DA. 1991. Primary HIV infection: host responses and intervention strategies. Aids 5:1-14. 64. Braun DL, Kouyos RD, Balmer B, Grube C, Weber R, Gunthard HF. 2015. Frequency and Spectrum of Unexpected Clinical Manifestations of Primary HIV-1 Infection. Clin Infect Dis 61:1013-1021. 65. Kahn JO, Walker BD. 1998. Acute human immunodeficiency virus type 1 infection. N Engl J Med 339:33-39. 66. Schacker T, Collier AC, Hughes J, Shea T, Corey L. 1996. Clinical and epidemiologic features of primary HIV infection. Ann Intern Med 125:257-264. 67. Chakrabarti L, Hurtrel M, Maire MA, Vazeux R, Dormont D, Montagnier L, Hurtrel B. 1991. Early viral replication in the brain of SIV-infected rhesus monkeys. Am J Pathol 139:1273- 1280. 68. Gaines H, von Sydow MA, von Stedingk LV, Biberfeld G, Bottiger B, Hansson LO, Lundbergh P, Sonnerborg AB, Wasserman J, Strannegaard OO. 1990. Immunological changes in primary HIV-1 infection. Aids 4:995-999. 69. Brenchley JM, Douek DC. 2008. HIV infection and the gastrointestinal immune system. Mucosal immunology 1:23-30.

- 114 -

70. Alter G, Teigen N, Ahern R, Streeck H, Meier A, Rosenberg ES, Altfeld M. 2007. Evolution of innate and adaptive effector cell functions during acute HIV-1 infection. J Infect Dis 195:1452-1460. 71. Borrow P, Bhardwaj N. 2008. Innate immune responses in primary HIV-1 infection. Curr Opin HIV AIDS 3:36-44. 72. Stacey AR, Norris PJ, Qin L, Haygreen EA, Taylor E, Heitman J, Lebedeva M, DeCamp A, Li D, Grove D, Self SG, Borrow P. 2009. Induction of a striking systemic cytokine cascade prior to peak viremia in acute human immunodeficiency virus type 1 infection, in contrast to more modest and delayed responses in acute hepatitis B and C virus infections. J Virol 83:3719- 3733. 73. Fiebig EW, Wright DJ, Rawal BD, Garrett PE, Schumacher RT, Peddada L, Heldebrant C, Smith R, Conrad A, Kleinman SH, Busch MP. 2003. Dynamics of HIV viremia and antibody seroconversion in plasma donors: implications for diagnosis and staging of primary HIV infection. Aids 17:1871-1879. 74. Koup RA, Safrit JT, Cao Y, Andrews CA, McLeod G, Borkowsky W, Farthing C, Ho DD. 1994. Temporal association of cellular immune responses with the initial control of viremia in primary human immunodeficiency virus type 1 syndrome. J Virol 68:4650-4655. 75. Richman DD, Wrin T, Little SJ, Petropoulos CJ. 2003. Rapid evolution of the neutralizing antibody response to HIV type 1 infection. Proceedings of the National Academy of Sciences of the United States of America 100:4144-4149. 76. Gray ES, Moore PL, Choge IA, Decker JM, Bibollet-Ruche F, Li H, Leseka N, Treurnicht F, Mlisana K, Shaw GM, Karim SS, Williamson C, Morris L. 2007. Neutralizing antibody responses in acute human immunodeficiency virus type 1 subtype C infection. J Virol 81:6187- 6196. 77. Pantaleo G, Graziosi C, Demarest JF, Butini L, Montroni M, Fox CH, Orenstein JM, Kotler DP, Fauci AS. 1993. HIV infection is active and progressive in lymphoid tissue during the clinically latent stage of disease. Nature 362:355-358. 78. Piatak M, Jr., Saag MS, Yang LC, Clark SJ, Kappes JC, Luk KC, Hahn BH, Shaw GM, Lifson JD. 1993. High levels of HIV-1 in plasma during all stages of infection determined by competitive PCR. Science 259:1749-1754. 79. Wang S, Hottz P, Schechter M, Rong L. 2016. Modeling the Slow CD4+ T Cell Decline in HIV-Infected Individuals. PLOS Computational Biology 11:e1004665. 80. Lang W, Perkins H, Anderson RE, Royce R, Jewell N, Winkelstein W, Jr. 1989. Patterns of T lymphocyte changes with human immunodeficiency virus infection: from seroconversion to the development of AIDS. J Acquir Immune Defic Syndr 2:63-69. 81. Liu Z, Cumberland WG, Hultin LE, Prince HE, Detels R, Giorgi JV. 1997. Elevated CD38 antigen expression on CD8+ T cells is a stronger marker for the risk of chronic HIV disease progression to AIDS and death in the Multicenter AIDS Cohort Study than CD4+ cell count, soluble immune activation markers, or combinations of HLA-DR and CD38 expression. J Acquir Immune Defic Syndr Hum Retrovirol 16:83-92. 82. Deeks SG, Kitchen CM, Liu L, Guo H, Gascon R, Narvaez AB, Hunt P, Martin JN, Kahn JO, Levy J, McGrath MS, Hecht FM. 2004. Immune activation set point during early HIV infection predicts subsequent CD4+ T-cell changes independent of viral load. Blood 104:942- 947. 83. Sinkovics JG, Dreyer DA, Shirato E, Cabiness JR, Shullenberger CC. 1971. Cytotoxic lymphocytes. I. Destruction of neoplastic cells by lymphocytes in cultures of human origin. Tex Rep Biol Med 29:227-242. 84. Sinkovics JG, Pienta RJ, Ahearn MJ, Trujillo JM, Mikulik FM. 1970. Activities of immune lymphoid cells against leukemia virus-carrier murine neoplastic cells. Bibl Haematol:618-623.

- 115 -

85. Sun JC, Lanier LL. 2011. NK cell development, homeostasis and function: parallels with CD8(+) T cells. Nat Rev Immunol 11:645-657. 86. Herberman RB, Nunn ME, Lavrin DH. 1975. Natural cytotoxic reactivity of mouse lymphoid cells against syngeneic acid allogeneic tumors. I. Distribution of reactivity and specificity. Int J Cancer 16:216-229. 87. Kiessling R, Klein E, Wigzell H. 1975. "Natural" killer cells in the mouse. I. Cytotoxic cells with specificity for mouse Moloney leukemia cells. Specificity and distribution according to genotype. Eur J Immunol 5:112-117. 88. Karre K. 2008. Natural killer cell recognition of missing self. Nat Immunol 9:477-480. 89. Stern P, Gidlund M, Orn A, Wigzell H. 1980. Natural killer cells mediate lysis of embryonal carcinoma cells lacking MHC. Nature 285:341-342. 90. Lettau M, Schmidt H, Kabelitz D, Janssen O. 2007. Secretory lysosomes and their cargo in T and NK cells. Immunol Lett 108:10-19. 91. Stinchcombe JC, Majorovits E, Bossi G, Fuller S, Griffiths GM. 2006. Centrosome polarization delivers secretory granules to the immunological synapse. Nature 443:462-465. 92. Orange JS, Harris KE, Andzelm MM, Valter MM, Geha RS, Strominger JL. 2003. The mature activating natural killer cell immunologic synapse is formed in distinct stages. Proc Natl Acad Sci U S A 100:14151-14156. 93. Lieberman J. 2003. The ABCs of granule-mediated cytotoxicity: new weapons in the arsenal. Nat Rev Immunol 3:361-370. 94. Lanier LL, Ruitenberg JJ, Phillips JH. 1988. Functional and biochemical analysis of CD16 antigen on natural killer cells and granulocytes. J Immunol 141:3478-3485. 95. Lanier LL. 2008. Up on the tightrope: natural killer cell activation and inhibition. Nat Immunol 9:495-502. 96. Smyth MJ, Cretney E, Kelly JM, Westwood JA, Street SE, Yagita H, Takeda K, van Dommelen SL, Degli-Esposti MA, Hayakawa Y. 2005. Activation of NK cell cytotoxicity. Mol Immunol 42:501-510. 97. Vivier E, Raulet DH, Moretta A, Caligiuri MA, Zitvogel L, Lanier LL, Yokoyama WM, Ugolini S. 2011. Innate or Adaptive Immunity? The Example of Natural Killer Cells. Science 331:44-49. 98. Moretta A, Marcenaro E, Sivori S, Della Chiesa M, Vitale M, Moretta L. 2005. Early liaisons between cells of the innate immune system in inflamed peripheral tissues. Trends Immunol 26:668-675. 99. Robbins SH, Bessou G, Cornillon A, Zucchini N, Rupp B, Ruzsics Z, Sacher T, Tomasello E, Vivier E, Koszinowski UH, Dalod M. 2007. Natural killer cells promote early CD8 T cell responses against cytomegalovirus. PLoS Pathog 3:e123. 100. Krebs P, Barnes MJ, Lampe K, Whitley K, Bahjat KS, Beutler B, Janssen E, Hoebe K. 2009. NK-cell-mediated killing of target cells triggers robust antigen-specific T-cell-mediated and humoral responses. Blood 113:6593-6602. 101. Martin MP, Gao X, Lee JH, Nelson GW, Detels R, Goedert JJ, Buchbinder S, Hoots K, Vlahov D, Trowsdale J, Wilson M, O'Brien SJ, Carrington M. 2002. Epistatic interaction between KIR3DS1 and HLA-B delays the progression to AIDS. Nat Genet 31:429-434. 102. Martin MP, Qi Y, Gao X, Yamada E, Martin JN, Pereyra F, Colombo S, Brown EE, Shupert WL, Phair J, Goedert JJ, Buchbinder S, Kirk GD, Telenti A, Connors M, O'Brien SJ, Walker BD, Parham P, Deeks SG, McVicar DW, Carrington M. 2007. Innate partnership of HLA-B and KIR3DL1 subtypes against HIV-1. Nat Genet 39:733-740. 103. Boulet S, Kleyman M, Kim JY, Kamya P, Sharafi S, Simic N, Bruneau J, Routy JP, Tsoukas CM, Bernard NF. 2008. A combined genotype of KIR3DL1 high expressing alleles and HLA-B*57 is associated with a reduced risk of HIV infection. Aids 22:1487-1491.

- 116 -

104. Boulet S, Sharafi S, Simic N, Bruneau J, Routy JP, Tsoukas CM, Bernard NF. 2008. Increased proportion of KIR3DS1 homozygotes in HIV-exposed uninfected individuals. Aids 22:595-599. 105. van Teijlingen NH, Holzemer A, Korner C, Garcia-Beltran WF, Schafer JL, Fadda L, Suscovich TJ, Brander C, Carrington M, Evans DT, van Baarle D, Altfeld M. 2014. Sequence variations in HIV-1 p24 Gag-derived epitopes can alter binding of KIR2DL2 to HLA- C*03:04 and modulate primary natural killer cell function. Aids 28:1399-1408. 106. Collins KL, Chen BK, Kalams SA, Walker BD, Baltimore D. 1998. HIV-1 Nef protein protects infected primary cells against killing by cytotoxic T lymphocytes. Nature 391:397-401. 107. Schwartz O, Marechal V, Le Gall S, Lemonnier F, Heard JM. 1996. Endocytosis of major histocompatibility complex class I molecules is induced by the HIV-1 Nef protein. Nat Med 2:338-342. 108. Cohen GB, Gandhi RT, Davis DM, Mandelboim O, Chen BK, Strominger JL, Baltimore D. 1999. The selective downregulation of class I major histocompatibility complex proteins by HIV- 1 protects HIV-infected cells from NK cells. Immunity 10:661-671. 109. Specht A, DeGottardi MQ, Schindler M, Hahn B, Evans DT, Kirchhoff F. 2008. Selective downmodulation of HLA-A and -B by Nef alleles from different groups of primate lentiviruses. Virology 373:229-237. 110. Wren LH, Chung AW, Isitman G, Kelleher AD, Parsons MS, Amin J, Cooper DA, Stratov I, Navis M, Kent SJ. 2013. Specific antibody-dependent cellular cytotoxicity responses associated with slow progression of HIV infection. Immunology 138:116-123. 111. Lambotte O, Ferrari G, Moog C, Yates NL, Liao HX, Parks RJ, Hicks CB, Owzar K, Tomaras GD, Montefiori DC, Haynes BF, Delfraissy JF. 2009. Heterogeneous neutralizing antibody and antibody-dependent cell cytotoxicity responses in HIV-1 elite controllers. Aids 23:897-906. 112. Reeves RK, Li H, Jost S, Blass E, Li H, Schafer JL, Varner V, Manickam C, Eslamizar L, Altfeld M, von Andrian UH, Barouch DH. 2015. Antigen-specific NK cell memory in rhesus macaques. Nat Immunol 16:927-932. 113. Guma M, Budt M, Saez A, Brckalo T, Hengel H, Angulo A, Lopez-Botet M. 2006. Expansion of CD94/NKG2C+ NK cells in response to human cytomegalovirus-infected fibroblasts. Blood 107:3624-3631. 114. Guma M, Cabrera C, Erkizia I, Bofill M, Clotet B, Ruiz L, Lopez-Botet M. 2006. Human cytomegalovirus infection is associated with increased proportions of NK cells that express the CD94/NKG2C receptor in aviremic HIV-1-positive patients. J Infect Dis 194:38-41. 115. Foley B, Cooley S, Verneris MR, Curtsinger J, Luo X, Waller EK, Anasetti C, Weisdorf D, Miller JS. 2012. Human cytomegalovirus (CMV)-induced memory-like NKG2C(+) NK cells are transplantable and expand in vivo in response to recipient CMV antigen. J Immunol 189:5082- 5088. 116. Lotzova E, Savary CA, Herberman RB. 1987. Induction of NK cell activity against fresh human leukemia in culture with interleukin 2. J Immunol 138:2718-2727. 117. Fehniger TA, Bluman EM, Porter MM, Mrozek E, Cooper MA, VanDeusen JB, Frankel SR, Stock W, Caligiuri MA. 2000. Potential mechanisms of human natural killer cell expansion in vivo during low-dose IL-2 therapy. J Clin Invest 106:117-124. 118. Shimoda K, Tsutsui H, Aoki K, Kato K, Matsuda T, Numata A, Takase K, Yamamoto T, Nukina H, Hoshino T, Asano Y, Gondo H, Okamura T, Okamura S, Nakayama K, Nakanishi K, Niho Y, Harada M. 2002. Partial impairment of interleukin-12 (IL-12) and IL-18 signaling in Tyk2-deficient mice. Blood 99:2094-2099. 119. Yu CR, Young HA, Ortaldo JR. 1998. Characterization of cytokine differential induction of STAT complexes in primary human T and NK cells. J Leukoc Biol 64:245-258.

- 117 -

120. Nakahira M, Ahn HJ, Park WR, Gao P, Tomura M, Park CS, Hamaoka T, Ohta T, Kurimoto M, Fujiwara H. 2002. Synergy of IL-12 and IL-18 for IFN-gamma gene expression: IL-12-induced STAT4 contributes to IFN-gamma promoter activation by up-regulating the binding activity of IL-18-induced activator protein 1. J Immunol 168:1146-1153. 121. Orange JS, Biron CA. 1996. An absolute and restricted requirement for IL-12 in natural killer cell IFN-gamma production and antiviral defense. Studies of natural killer and T cell responses in contrasting viral infections. J Immunol 156:1138-1142. 122. Gherardi MM, Ramirez JC, Esteban M. 2003. IL-12 and IL-18 act in synergy to clear vaccinia virus infection: involvement of innate and adaptive components of the immune system. J Gen Virol 84:1961-1972. 123. Kim S, Poursine-Laurent J, Truscott SM, Lybarger L, Song Y-J, Yang L, French AR, Sunwoo JB, Lemieux S, Hansen TH, Yokoyama WM. 2005. Licensing of natural killer cells by host major histocompatibility complex class I molecules. Nature 436:709-713. 124. Furukawa H, Yabe T, Watanabe K, Miyamoto R, Miki A, Akaza T, Tadokoro K, Tohma S, Inoue T, Yamamoto K, Juji T. 1999. Tolerance of NK and LAK activity for HLA class I- deficient targets in a TAP1-deficient patient (bare lymphocyte syndrome type I). Hum Immunol 60:32-40. 125. Hoglund P, Ohlen C, Carbone E, Franksson L, Ljunggren HG, Latour A, Koller B, Karre K. 1991. Recognition of beta 2-microglobulin-negative (beta 2m-) T-cell blasts by natural killer cells from normal but not from beta 2m- mice: nonresponsiveness controlled by beta 2m- bone marrow in chimeric mice. Proc Natl Acad Sci U S A 88:10332-10336. 126. Biassoni R, Ugolotti E, De Maria A. 2009. NK cell receptors and their interactions with MHC. Curr Pharm Des 15:3301-3310. 127. Moretta A, Bottino C, Vitale M, Pende D, Biassoni R, Mingari MC, Moretta L. 1996. Receptors for HLA class-I molecules in human natural killer cells. Annu Rev Immunol 14:619- 648. 128. Fernandez NC, Treiner E, Vance RE, Jamieson AM, Lemieux S, Raulet DH. 2005. A subset of natural killer cells achieves self-tolerance without expressing inhibitory receptors specific for self-MHC molecules. Blood 105:4416-4423. 129. Anfossi N, Andre P, Guia S, Falk CS, Roetynck S, Stewart CA, Breso V, Frassati C, Reviron D, Middleton D, Romagne F, Ugolini S, Vivier E. 2006. Human NK cell education by inhibitory receptors for MHC class I. Immunity 25:331-342. 130. Gasser S, Raulet DH. 2006. Activation and self-tolerance of natural killer cells. Immunol Rev 214:130-142. 131. Brodin P, Karre K, Hoglund P. 2009. NK cell education: not an on-off switch but a tunable rheostat. Trends Immunol 30:143-149. 132. Raulet DH, Vance RE. 2006. Self-tolerance of natural killer cells. Nat Rev Immunol 6:520-531. 133. Lozzio C, Lozzio B. 1975. Human chronic myelogenous leukemia cell-line with positive Philadelphia chromosome. Blood 45:321-334. 134. Klein E, Ben-Bassat H, Neumann H, Ralph P, Zeuthen J, Polliack A, Vanky F. 1976. Properties of the K562 cell line, derived from a patient with chronic myeloid leukemia. Int J Cancer 18:421-431. 135. Lozzio BB, Lozzio CB, Bamberger EG, Feliu AS. 1981. A Multipotential Leukemia Cell Line (K-562) of Human Origin. Experimental Biology and Medicine 166:546-550. 136. Dimery IW, Ross DD, Testa JR, Gupta SK, Felsted RL, Pollak A, Bachur NR. 1983. Variation amongst K562 cell cultures. Exp Hematol 11:601-610. 137. Collins SJ, Groudine MT. 1983. Rearrangement and amplification of c-abl sequences in the human chronic myelogenous leukemia cell line K-562. Proc Natl Acad Sci U S A 80:4813-4817. 138. Naumann S, Reutzel D, Speicher M, Decker H-J. 2001. Complete karyotype characterization of the K562 cell line by combined application of G-banding, multiplex-fluorescence in situ - 118 -

hybridization, fluorescence in situ hybridization, and comparative genomic hybridization. Leukemia Research 25:313-322. 139. Shimizu Y, DeMars R. 1989. Production of human cells expressing individual transferred HLA- A,-B,-C genes using an HLA-A,-B,-C null human cell line. J Immunol 142:3320-3328. 140. Kavathas P, Bach FH, DeMars R. 1980. Gamma ray-induced loss of expression of HLA and glyoxalase I alleles in lymphoblastoid cells. Proc Natl Acad Sci U S A 77:4251-4255. 141. Baichwal VR, Sugden B. 1987. Posttranslational processing of an Epstein-Barr virus-encoded membrane protein expressed in cells transformed by Epstein-Barr virus. J Virol 61:866-875. 142. Lee N, Goodlett DR, Ishitani A, Marquardt H, Geraghty DE. 1998. HLA-E surface expression depends on binding of TAP-dependent peptides derived from certain HLA class I signal sequences. J Immunol 160:4951-4960. 143. Braud VM, Allan DSJ, McMichael AJ. 1999. Functions of nonclassical MHC and non-MHC- encoded class I molecules. Current Opinion in Immunology 11:100-108. 144. Ward J, Bonaparte M, Sacks J, Guterman J, Fogli M, Mavilio D, Barker E. 2007. HIV modulates the expression of ligands important in triggering natural killer cell cytotoxic responses on infected primary T-cell blasts. Blood 110:1207-1214. 145. Richard J, Sindhu S, Pham TNQ, Belzile J-P, Cohen ÉA. 2010. HIV-1 Vpr up-regulates expression of ligands for the activating NKG2D receptor and promotes NK cell–mediated killing. Blood 115:1354-1363. 146. Cerboni C, Zingoni A, Cippitelli M, Piccoli M, Frati L, Santoni A. 2007. Antigen-activated human T lymphocytes express cell-surface NKG2D ligands via an ATM/ATR-dependent mechanism and become susceptible to autologous NK- cell lysis. Blood 110:606-615. 147. Alter G, Martin MP, Teigen N, Carr WH, Suscovich TJ, Schneidewind A, Streeck H, Waring M, Meier A, Brander C, Lifson JD, Allen TM, Carrington M, Altfeld M. 2007. Differential natural killer cell–mediated inhibition of HIV-1 replication based on distinct KIR/HLA subtypes. The Journal of Experimental Medicine 204:3027-3036. 148. Oliva A, Kinter AL, Vaccarezza M, Rubbert A, Catanzaro A, Moir S, Monaco J, Ehler L, Mizell S, Jackson R, Li Y, Romano JW, Fauci AS. 1998. Natural killer cells from human immunodeficiency virus (HIV)-infected individuals are an important source of CC-chemokines and suppress HIV-1 entry and replication in vitro. J Clin Invest 102:223-231. 149. Cocchi F, DeVico AL, Garzino-Demo A, Arya SK, Gallo RC, Lusso P. 1995. Identification of RANTES, MIP-1 alpha, and MIP-1 beta as the major HIV-suppressive factors produced by CD8+ T cells. Science 270:1811-1815. 150. Ehrlich LI, Ogasawara K, Hamerman JA, Takaki R, Zingoni A, Allison JP, Lanier LL. 2005. Engagement of NKG2D by cognate ligand or antibody alone is insufficient to mediate costimulation of human and mouse CD8+ T cells. J Immunol 174:1922-1931. 151. Jamieson AM, Diefenbach A, McMahon CW, Xiong N, Carlyle JR, Raulet DH. 2002. The role of the NKG2D immunoreceptor in immune cell activation and natural killing. Immunity 17:19-29. 152. Wu J, Song Y, Bakker AB, Bauer S, Spies T, Lanier LL, Phillips JH. 1999. An activating immunoreceptor complex formed by NKG2D and DAP10. Science 285:730-732. 153. Glienke J, Sobanov Y, Brostjan C, Steffens C, Nguyen C, Lehrach H, Hofer E, Francis F. 1998. The genomic organization of NKG2C, E, F, and D receptor genes in the human natural killer gene complex. Immunogenetics 48:163-173. 154. Garrity D, Call ME, Feng J, Wucherpfennig KW. 2005. The activating NKG2D receptor assembles in the membrane with two signaling dimers into a hexameric structure. Proc Natl Acad Sci U S A 102:7641-7646. 155. Houchins JP, Yabe T, McSherry C, Bach FH. 1991. DNA sequence analysis of NKG2, a family of related cDNA clones encoding type II integral membrane proteins on human natural killer cells. J Exp Med 173:1017-1020. - 119 -

156. Rosen DB, Araki M, Hamerman JA, Chen T, Yamamura T, Lanier LL. 2004. A Structural basis for the association of DAP12 with mouse, but not human, NKG2D. J Immunol 173:2470- 2478. 157. Chang C, Dietrich J, Harpur AG, Lindquist JA, Haude A, Loke YW, King A, Colonna M, Trowsdale J, Wilson MJ. 1999. Cutting edge: KAP10, a novel transmembrane adapter protein genetically linked to DAP12 but with unique signaling properties. J Immunol 163:4651-4654. 158. Gilfillan S, Ho EL, Cella M, Yokoyama WM, Colonna M. 2002. NKG2D recruits two distinct adapters to trigger NK cell activation and costimulation. Nat Immunol 3:1150-1155. 159. Diefenbach A, Tomasello E, Lucas M, Jamieson AM, Hsia JK, Vivier E, Raulet DH. 2002. Selective associations with signaling proteins determine stimulatory versus costimulatory activity of NKG2D. Nat Immunol 3:1142-1149. 160. Iwaszko M, Gębura K, Bogunia-Kubik K. 2012. Non-Classical MHC Class Ib Molecules and their Receptors-Role in Allogeneic Transplantation of Hematopoietic Stem Cells. Journal of Transplant Techologies and Research 2:1-8. 161. Chalupny NJ, Sutherland CL, Lawrence WA, Rein-Weston A, Cosman D. 2003. ULBP4 is a novel ligand for human NKG2D. Biochem Biophys Res Commun 305:129-135. 162. Cosman D, Mullberg J, Sutherland CL, Chin W, Armitage R, Fanslow W, Kubin M, Chalupny NJ. 2001. ULBPs, novel MHC class I-related molecules, bind to CMV glycoprotein UL16 and stimulate NK cytotoxicity through the NKG2D receptor. Immunity 14:123-133. 163. Radosavljevic M, Cuillerier B, Wilson MJ, Clement O, Wicker S, Gilfillan S, Beck S, Trowsdale J, Bahram S. 2002. A cluster of ten novel MHC class I related genes on human chromosome 6q24.2-q25.3. Genomics 79:114-123. 164. Steinle A, Li P, Morris DL, Groh V, Lanier LL, Strong RK, Spies T. 2001. Interactions of human NKG2D with its ligands MICA, MICB, and homologs of the mouse RAE-1 protein family. Immunogenetics 53:279-287. 165. Bauer S, Groh V, Wu J, Steinle A, Phillips JH, Lanier LL, Spies T. 1999. Activation of NK cells and T cells by NKG2D, a receptor for stress-inducible MICA. Science 285:727-729. 166. Bacon L, Eagle RA, Meyer M, Easom N, Young NT, Trowsdale J. 2004. Two human ULBP/RAET1 molecules with transmembrane regions are ligands for NKG2D. J Immunol 173:1078-1084. 167. Bahram S, Bresnahan M, Geraghty DE, Spies T. 1994. A second lineage of mammalian major histocompatibility complex class I genes. Proc Natl Acad Sci U S A 91:6259-6263. 168. Radaev S, Sun PD. 2003. Structure and function of natural killer cell surface receptors. Annu Rev Biophys Biomol Struct 32:93-114. 169. Zwirner NW, Fernandez-Vina MA, Stastny P. 1998. MICA, a new polymorphic HLA-related antigen, is expressed mainly by keratinocytes, endothelial cells, and monocytes. Immunogenetics 47:139-148. 170. Groh V, Bahram S, Bauer S, Herman A, Beauchamp M, Spies T. 1996. Cell stress-regulated human major histocompatibility complex class I gene expressed in gastrointestinal epithelium. Proc Natl Acad Sci U S A 93:12445-12450. 171. Sutherland CL, Chalupny NJ, Cosman D. 2001. The UL16-binding proteins, a novel family of MHC class I-related ligands for NKG2D, activate natural killer cell functions. Immunol Rev 181:185-192. 172. Matsumura M, Fremont DH, Peterson PA, Wilson IA. 1992. Emerging principles for the recognition of peptide antigens by MHC class I molecules. Science 257:927-934. 173. Vilarinho S, Ogasawara K, Nishimura S, Lanier LL, Baron JL. 2007. Blockade of NKG2D on NKT cells prevents hepatitis and the acute immune response to hepatitis B virus. Proceedings of the National Academy of Sciences 104:18187-18192.

- 120 -

174. Li J, Rabinovich BA, Hurren R, Cosman D, Miller RG. 2005. Survival versus neglect: redefining subsets based on expression of NKG2D ligand(s) and MHC class I. Eur J Immunol 35:439-448. 175. Nomura M, Zou Z, Joh T, Takihara Y, Matsuda Y, Shimada K. 1996. Genomic structures and characterization of Rae1 family members encoding GPI-anchored cell surface proteins and expressed predominantly in embryonic mouse brain. J Biochem 120:987-995. 176. Cerboni C, Neri F, Casartelli N, Zingoni A, Cosman D, Rossi P, Santoni A, Doria M. 2007. Human immunodeficiency virus 1 Nef protein downmodulates the ligands of the activating receptor NKG2D and inhibits natural killer cell-mediated cytotoxicity. J Gen Virol 88:242-250. 177. Ward J, Bonaparte M, Sacks J, Guterman J, Fogli M, Mavilio D, Barker E. 2007. HIV modulates the expression of ligands important in triggering natural killer cell cytotoxic responses on infected primary T-cell blasts. Blood 110:1207-1214. 178. Diefenbach A, Jamieson AM, Liu SD, Shastri N, Raulet DH. 2000. Ligands for the murine NKG2D receptor: expression by tumor cells and activation of NK cells and macrophages. Nat Immunol 1:119-126. 179. Siren J, Sareneva T, Pirhonen J, Strengell M, Veckman V, Julkunen I, Matikainen S. 2004. Cytokine and contact-dependent activation of natural killer cells by influenza A or Sendai virus- infected macrophages. J Gen Virol 85:2357-2364. 180. Gasser S, Orsulic S, Brown EJ, Raulet DH. 2005. The DNA damage pathway regulates innate immune system ligands of the NKG2D receptor. Nature 436:1186-1190. 181. Chen X, Trivedi PP, Ge B, Krzewski K, Strominger JL. 2007. Many NK cell receptors activate ERK2 and JNK1 to trigger microtubule organizing center and granule polarization and cytotoxicity. Proc Natl Acad Sci U S A 104:6329-6334. 182. Cerwenka A, Baron JL, Lanier LL. 2001. Ectopic expression of retinoic acid early inducible-1 gene (RAE-1) permits natural killer cell-mediated rejection of a MHC class I-bearing tumor in vivo. Proc Natl Acad Sci U S A 98:11521-11526. 183. Fernández-Messina L, Reyburn H, Vales-Gomez M. 2012. Human NKG2D-ligands: cell biology strategies to ensure immune recognition. Frontiers in Immunology 3. 184. Geraghty DE. 1993. Structure of the HLA class I region and expression of its resident genes. Curr Opin Immunol 5:3-7. 185. Braud VM, Allan DS, O'Callaghan CA, Soderstrom K, D'Andrea A, Ogg GS, Lazetic S, Young NT, Bell JI, Phillips JH, Lanier LL, McMichael AJ. 1998. HLA-E binds to natural killer cell receptors CD94/NKG2A, B and C. Nature 391:795-799. 186. Maier S, Grzeschik M, Weiss EH, Ulbrecht M. 2000. Implications of HLA-E allele expression and different HLA-E ligand diversity for the regulation of NK cells. Hum Immunol 61:1059- 1065. 187. Lee N, Llano M, Carretero M, Ishitani A, Navarro F, López-Botet M, Geraghty DE. 1998. HLA-E is a major ligand for the natural killer inhibitory receptor CD94/NKG2A. Proceedings of the National Academy of Sciences 95:5199-5204. 188. Marin R, Ruiz-Cabello F, Pedrinaci S, Mendez R, Jimenez P, Geraghty DE, Garrido F. 2003. Analysis of HLA-E expression in human tumors. Immunogenetics 54:767-775. 189. van Hall T, Oliveira CC, Joosten SA, Ottenhoff TH. 2010. The other Janus face of Qa-1 and HLA-E: diverse peptide repertoires in times of stress. Microbes Infect 12:910-918. 190. Michaelsson J, Teixeira de Matos C, Achour A, Lanier LL, Karre K, Soderstrom K. 2002. A signal peptide derived from hsp60 binds HLA-E and interferes with CD94/NKG2A recognition. J Exp Med 196:1403-1414. 191. Ulbrecht M, Martinozzi S, Grzeschik M, Hengel H, Ellwart JW, Pla M, Weiss EH. 2000. Cutting edge: the human cytomegalovirus UL40 gene product contains a ligand for HLA-E and prevents NK cell-mediated lysis. J Immunol 164:5019-5022.

- 121 -

192. Carretero M, Cantoni C, Bellon T, Bottino C, Biassoni R, Rodriguez A, Perez-Villar JJ, Moretta L, Moretta A, Lopez-Botet M. 1997. The CD94 and NKG2-A C-type lectins covalently assemble to form a natural killer cell inhibitory receptor for HLA class I molecules. Eur J Immunol 27:563-567. 193. Lazetic S, Chang C, Houchins JP, Lanier LL, Phillips JH. 1996. Human natural killer cell receptors involved in MHC class I recognition are disulfide-linked heterodimers of CD94 and NKG2 subunits. J Immunol 157:4741-4745. 194. Lanier LL, Corliss B, Wu J, Phillips JH. 1998. Association of DAP12 with activating CD94/NKG2C NK cell receptors. Immunity 8:693-701. 195. Cheng X, Im W. 2012. NMR observable-based structure refinement of DAP12-NKG2C activating immunoreceptor complex in explicit membranes. Biophys J 102:L27-29. 196. Call ME, Wucherpfennig KW, Chou JJ. 2010. The structural basis for intramembrane assembly of an activating immunoreceptor complex. Nat Immunol 11:1023-1029. 197. Lanier LL. 2008. Up on the tightrope: natural killer cell activation and inhibition. Nature immunology 9:495-502. 198. Beziat V, Dalgard O, Asselah T, Halfon P, Bedossa P, Boudifa A, Hervier B, Theodorou I, Martinot M, Debre P, Bjorkstrom NK, Malmberg KJ, Marcellin P, Vieillard V. 2012. CMV drives clonal expansion of NKG2C+ NK cells expressing self-specific KIRs in chronic hepatitis patients. Eur J Immunol 42:447-457. 199. Della Chiesa M, Muccio L, Moretta A. 2013. CMV induces rapid NK cell maturation in HSCT recipients. Immunol Lett 155:11-13. 200. Lopez-Verges S, Milush JM, Schwartz BS, Pando MJ, Jarjoura J, York VA, Houchins JP, Miller S, Kang SM, Norris PJ, Nixon DF, Lanier LL. 2011. Expansion of a unique CD57(+)NKG2Chi natural killer cell subset during acute human cytomegalovirus infection. Proc Natl Acad Sci U S A 108:14725-14732. 201. Brunetta E, Fogli M, Varchetta S, Bozzo L, Hudspeth KL, Marcenaro E, Moretta A, Mavilio D. 2010. Chronic HIV-1 viremia reverses NKG2A/NKG2C ratio on natural killer cells in patients with human cytomegalovirus co-infection. Aids 24:27-34. 202. Thomas R, Low HZ, Kniesch K, Jacobs R, Schmidt RE, Witte T. 2012. NKG2C deletion is a risk factor of HIV infection. AIDS Res Hum Retroviruses 28:844-851. 203. Adland E, Klenerman P, Goulder P, Matthews PC. 2015. Ongoing burden of disease and mortality from HIV/CMV coinfection in Africa in the antiretroviral therapy era. Frontiers in Microbiology 6:1016. 204. Paiardini M, Müller-Trutwin M. 2013. HIV-associated chronic immune activation. Immunological reviews 254:78-101. 205. Malmberg KJ, Beziat V, Ljunggren HG. 2012. Spotlight on NKG2C and the human NK-cell response to CMV infection. Eur J Immunol 42:3141-3145. 206. Orbelyan GA, Tang F, Sally B, Solus J, Meresse B, Ciszewski C, Grenier J-C, Barreiro LB, Lanier LL, Jabri B. 2014. Human NKG2E is expressed and forms an intracytoplasmic complex with CD94 and DAP12. Journal of immunology (Baltimore, Md : 1950) 193:610-616. 207. Bellón T, Heredia ABd, Llano M, Minguela A, Rodriguez A, López-Botet M, Aparicio P. 1999. Triggering of Effector Functions on a CD8+ T Cell Clone Upon the Aggregation of an Activatory CD94/kp39 Heterodimer. The Journal of Immunology 162:3996-4002. 208. Valés-Gómez M, Reyburn HT, Erskine RA, López-Botet M, Strominger JL. 1999. Kinetics and peptide dependency of the binding of the inhibitory NK receptor CD94/NKG2-A and the activating receptor CD94/NKG2-C to HLA-E. The EMBO Journal 18:4250-4260. 209. Vance RE, Jamieson AM, Raulet DH. 1999. Recognition of the Class Ib Molecule Qa-1(b) by Putative Activating Receptors Cd94/Nkg2c and Cd94/Nkg2e on Mouse Natural Killer Cells. The Journal of Experimental Medicine 190:1801-1812.

- 122 -

210. Houchins JP, Lanier LL, Niemi EC, Phillips JH, Ryan JC. 1997. Natural killer cell cytolytic activity is inhibited by NKG2-A and activated by NKG2-C. J Immunol 158:3603-3609. 211. Beziat V, Hervier B, Achour A, Boutolleau D, Marfain-Koka A, Vieillard V. 2011. Human NKG2A overrides NKG2C effector functions to prevent autoreactivity of NK cells. Blood 117:4394-4396. 212. Vitale M, Bottino C, Sivori S, Sanseverino L, Castriconi R, Marcenaro E, Augugliaro R, Moretta L, Moretta A. 1998. NKp44, a novel triggering surface molecule specifically expressed by activated natural killer cells, is involved in non-major histocompatibility complex-restricted tumor cell lysis. J Exp Med 187:2065-2072. 213. Sivori S, Vitale M, Morelli L, Sanseverino L, Augugliaro R, Bottino C, Moretta L, Moretta A. 1997. p46, a Novel Natural Killer Cell–specific Surface Molecule That Mediates Cell Activation. The Journal of Experimental Medicine 186:1129-1136. 214. Pende D, Parolini S, Pessino A, Sivori S, Augugliaro R, Morelli L, Marcenaro E, Accame L, Malaspina A, Biassoni R, Bottino C, Moretta L, Moretta A. 1999. Identification and molecular characterization of NKp30, a novel triggering receptor involved in natural cytotoxicity mediated by human natural killer cells. J Exp Med 190:1505-1516. 215. Castriconi R, Dondero A, Augugliaro R, Cantoni C, Carnemolla B, Sementa AR, Negri F, Conte R, Corrias MV, Moretta L, Moretta A, Bottino C. 2004. Identification of 4Ig-B7-H3 as a neuroblastoma-associated molecule that exerts a protective role from an NK cell-mediated lysis. Proc Natl Acad Sci U S A 101:12640-12645. 216. Byrd A, Hoffmann SC, Jarahian M, Momburg F, Watzl C. 2007. Expression analysis of the ligands for the Natural Killer cell receptors NKp30 and NKp44. PLoS One 2:e1339. 217. Joyce MG, Sun PD. 2011. The structural basis of ligand recognition by natural killer cell receptors. J Biomed Biotechnol 2011:203628. 218. Li Y, Wang Q, Mariuzza RA. 2011. Structure of the human activating natural cytotoxicity receptor NKp30 bound to its tumor cell ligand B7-H6. J Exp Med 208:703-714. 219. Chisholm SE, Reyburn HT. 2006. Recognition of Vaccinia Virus-Infected Cells by Human Natural Killer Cells Depends on Natural Cytotoxicity Receptors. Journal of Virology 80:2225- 2233. 220. Jarahian M, Fiedler M, Cohnen A, Djandji D, Hammerling GJ, Gati C, Cerwenka A, Turner PC, Moyer RW, Watzl C, Hengel H, Momburg F. 2011. Modulation of NKp30- and NKp46-mediated natural killer cell responses by poxviral hemagglutinin. PLoS Pathog 7:e1002195. 221. Arnon TI, Achdout H, Levi O, Markel G, Saleh N, Katz G, Gazit R, Gonen-Gross T, Hanna J, Nahari E, Porgador A, Honigman A, Plachter B, Mevorach D, Wolf DG, Mandelboim O. 2005. Inhibition of the NKp30 activating receptor by pp65 of human cytomegalovirus. Nat Immunol 6:515-523. 222. Bloushtain N, Qimron U, Bar-Ilan A, Hershkovitz O, Gazit R, Fima E, Korc M, Vlodavsky I, Bovin NV, Porgador A. 2004. Membrane-associated heparan sulfate proteoglycans are involved in the recognition of cellular targets by NKp30 and NKp46. J Immunol 173:2392-2401. 223. Hecht ML, Rosental B, Horlacher T, Hershkovitz O, De Paz JL, Noti C, Schauer S, Porgador A, Seeberger PH. 2009. Natural cytotoxicity receptors NKp30, NKp44 and NKp46 bind to different heparan sulfate/heparin sequences. J Proteome Res 8:712-720. 224. Kruse PH, Matta J, Ugolini S, Vivier E. 2014. Natural cytotoxicity receptors and their ligands. Immunol Cell Biol 92:221-229. 225. Joyce MG, Tran P, Zhuravleva MA, Jaw J, Colonna M, Sun PD. 2011. Crystal structure of human natural cytotoxicity receptor NKp30 and identification of its ligand binding site. Proc Natl Acad Sci U S A 108:6223-6228. 226. Love PE, Hayes SM. 2010. ITAM-mediated Signaling by the T-Cell Antigen Receptor. Cold Spring Harbor Perspectives in Biology 2:a002485. - 123 -

227. Hamerman JA, Ni M, Killebrew JR, Chu C-L, Lowell CA. 2009. The expanding roles of ITAM adapters FcRγ and DAP12 in myeloid cells. Immunological reviews 232:42-58. 228. Golden-Mason L, Cox AL, Randall JA, Cheng L, Rosen HR. 2010. Increased NK cytotoxicity and NKp30 expression protects against HCV infection in high-risk individuals and inhibits replication in vitro. Hepatology (Baltimore, Md) 52:1581-1589. 229. Delahaye NF, Rusakiewicz S, Martins I, Menard C, Roux S, Lyonnet L, Paul P, Sarabi M, Chaput N, Semeraro M, Minard-Colin V, Poirier-Colame V, Chaba K, Flament C, Baud V, Authier H, Kerdine-Romer S, Pallardy M, Cremer I, Peaudecerf L, Rocha B, Valteau- Couanet D, Gutierrez JC, Nunes JA, Commo F, Bonvalot S, Ibrahim N, Terrier P, Opolon P, Bottino C, Moretta A, Tavernier J, Rihet P, Coindre J-M, Blay J-Y, Isambert N, Emile J- F, Vivier E, Lecesne A, Kroemer G, Zitvogel L. 2011. Alternatively spliced NKp30 isoforms affect the prognosis of gastrointestinal stromal tumors. Nat Med 17:700-707. 230. Cantoni C, Bottino C, Vitale M, Pessino A, Augugliaro R, Malaspina A, Parolini S, Moretta L, Moretta A, Biassoni R. 1999. NKp44, a triggering receptor involved in tumor cell lysis by activated human natural killer cells, is a novel member of the immunoglobulin superfamily. J Exp Med 189:787-796. 231. Cantoni C, Ponassi M, Biassoni R, Conte R, Spallarossa A, Moretta A, Moretta L, Bolognesi M, Bordo D. 2003. The three-dimensional structure of the human NK cell receptor NKp44, a triggering partner in natural cytotoxicity. Structure 11:725-734. 232. Moretta A, Bottino C, Vitale M, Pende D, Cantoni C, Mingari MC, Biassoni R, Moretta L. 2001. Activating receptors and coreceptors involved in human natural killer cell-mediated cytolysis. Annu Rev Immunol 19:197-223. 233. Joyce MG, Sun PD. 2011. The Structural Basis of Ligand Recognition by Natural Killer Cell Receptors. Journal of Biomedicine and Biotechnology 2011. 234. Campbell KS, Yusa S, Kikuchi-Maki A, Catina TL. 2004. NKp44 triggers NK cell activation through DAP12 association that is not influenced by a putative cytoplasmic inhibitory sequence. J Immunol 172:899-906. 235. Arnon TI, Lev M, Katz G, Chernobrov Y, Porgador A, Mandelboim O. 2001. Recognition of viral hemagglutinins by NKp44 but not by NKp30. Eur J Immunol 31:2680-2689. 236. Hershkovitz O, Rosental B, Rosenberg LA, Navarro-Sanchez ME, Jivov S, Zilka A, Gershoni-Yahalom O, Brient-Litzler E, Bedouelle H, Ho JW, Campbell KS, Rager-Zisman B, Despres P, Porgador A. 2009. NKp44 receptor mediates interaction of the envelope glycoproteins from the West Nile and dengue viruses with NK cells. J Immunol 183:2610-2621. 237. Esin S, Batoni G, Counoupas C, Stringaro A, Brancatisano FL, Colone M, Maisetta G, Florio W, Arancia G, Campa M. 2008. Direct binding of human NK cell natural cytotoxicity receptor NKp44 to the surfaces of mycobacteria and other bacteria. Infect Immun 76:1719-1727. 238. Esin S, Counoupas C, Aulicino A, Brancatisano FL, Maisetta G, Bottai D, Di Luca M, Florio W, Campa M, Batoni G. 2013. Interaction of Mycobacterium tuberculosis cell wall components with the human natural killer cell receptors NKp44 and Toll-like receptor 2. Scand J Immunol 77:460-469. 239. Baychelier F, Sennepin A, Ermonval M, Dorgham K, Debre P, Vieillard V. 2013. Identification of a cellular ligand for the natural cytotoxicity receptor NKp44. Blood 122:2935- 2942. 240. Rosental B, Brusilovsky M, Hadad U, Oz D, Appel MY, Afergan F, Yossef R, Rosenberg LA, Aharoni A, Cerwenka A, Campbell KS, Braiman A, Porgador A. 2011. Proliferating cell nuclear antigen is a novel inhibitory ligand for the natural cytotoxicity receptor NKp44. J Immunol 187:5693-5702. 241. Funato H, Yoshimura M, Ito Y, Okeda R, Ihara Y. 1996. Proliferating cell nuclear antigen (PCNA) expressed in human leptomeninges. J Histochem Cytochem 44:1261-1265.

- 124 -

242. Bouayad D, Pederzoli-Ribeil M, Mocek J, Candalh C, Arlet J-B, Hermine O, Reuter N, Davezac N, Witko-Sarsat V. 2012. Nuclear-to-cytoplasmic Relocalization of the Proliferating Cell Nuclear Antigen (PCNA) during Differentiation Involves a Chromosome Region Maintenance 1 (CRM1)-dependent Export and Is a Prerequisite for PCNA Antiapoptotic Activity in Mature Neutrophils. The Journal of Biological Chemistry 287:33812-33825. 243. Moretta L, Bottino C, Pende D, Castriconi R, Mingari MC, Moretta A. 2006. Surface NK receptors and their ligands on tumor cells. Semin Immunol 18:151-158. 244. Yu J, Mitsui T, Wei M, Mao H, Butchar JP, Shah MV, Zhang J, Mishra A, Alvarez- Breckenridge C, Liu X, Liu S, Yokohama A, Trotta R, Marcucci G, Jr., Benson DM, Loughran TP, Jr., Tridandapani S, Caligiuri MA. 2011. NKp46 identifies an NKT cell subset susceptible to leukemic transformation in mouse and human. J Clin Invest 121:1456-1470. 245. Spits H, Artis D, Colonna M, Diefenbach A, Di Santo JP, Eberl G, Koyasu S, Locksley RM, McKenzie AN, Mebius RE, Powrie F, Vivier E. 2013. Innate lymphoid cells--a proposal for uniform nomenclature. Nat Rev Immunol 13:145-149. 246. Ponassi M, Cantoni C, Biassoni R, Conte R, Spallarossa A, Pesce A, Moretta A, Moretta L, Bolognesi M, Bordo D. 2003. Structure of the human NK cell triggering receptor NKp46 ectodomain. Biochem Biophys Res Commun 309:317-323. 247. Foster CE, Colonna M, Sun PD. 2003. Crystal structure of the human natural killer (NK) cell activating receptor NKp46 reveals structural relationship to other leukocyte receptor complex immunoreceptors. J Biol Chem 278:46081-46086. 248. Jaron-Mendelson M, Yossef R, Appel MY, Zilka A, Hadad U, Afergan F, Rosental B, Engel S, Nedvetzki S, Braiman A, Porgador A. 2012. Dimerization of NKp46 receptor is essential for NKp46-mediated lysis: characterization of the dimerization site by epitope mapping. J Immunol 188:6165-6174. 249. Mandelboim O, Lieberman N, Lev M, Paul L, Arnon TI, Bushkin Y, Davis DM, Strominger JL, Yewdell JW, Porgador A. 2001. Recognition of haemagglutinins on virus-infected cells by NKp46 activates lysis by human NK cells. Nature 409:1055-1060. 250. Jarahian M, Watzl C, Fournier P, Arnold A, Djandji D, Zahedi S, Cerwenka A, Paschen A, Schirrmacher V, Momburg F. 2009. Activation of natural killer cells by newcastle disease virus hemagglutinin-neuraminidase. J Virol 83:8108-8121. 251. Garg A, Barnes PF, Porgador A, Roy S, Wu S, Nanda JS, Griffith DE, Girard WM, Rawal N, Shetty S, Vankayalapati R. 2006. Vimentin expressed on Mycobacterium tuberculosis- infected human monocytes is involved in binding to the NKp46 receptor. J Immunol 177:6192- 6198. 252. Sivori S, Parolini S, Falco M, Marcenaro E, Biassoni R, Bottino C, Moretta L, Moretta A. 2000. 2B4 functions as a co-receptor in human NK cell activation. Eur J Immunol 30:787-793. 253. Cannons JL, Tangye SG, Schwartzberg PL. 2011. SLAM family receptors and SAP adaptors in immunity. Annu Rev Immunol 29:665-705. 254. McNerney ME, Lee KM, Kumar V. 2005. 2B4 (CD244) is a non-MHC binding receptor with multiple functions on natural killer cells and CD8+ T cells. Mol Immunol 42:489-494. 255. Waggoner SN, Kumar V. 2012. Evolving role of 2B4/CD244 in T and NK cell responses during virus infection. Frontiers in Immunology 3:377. 256. Yokoyama S, Staunton D, Fisher R, Amiot M, Fortin JJ, Thorley-Lawson DA. 1991. Expression of the Blast-1 activation/adhesion molecule and its identification as CD48. J Immunol 146:2192-2200. 257. Zarama A, Perez-Carmona N, Farre D, Tomic A, Borst EM, Messerle M, Jonjic S, Engel P, Angulo A. 2014. Cytomegalovirus m154 hinders CD48 cell-surface expression and promotes viral escape from host natural killer cell control. PLoS Pathog 10:e1004000.

- 125 -

258. Chibueze-Nnorom CE, White Y, Yoshimitsu M, Arima N. 2013. Role of CD48 in regulation of T-cell mediated immuntiy in HTLV-1 infection. Frontiers in Immunology Conference Abstract: 15th International Congress of Immunology 259. Tangye SG, Cherwinski H, Lanier LL, Phillips JH. 2000. 2B4-mediated activation of human natural killer cells. Mol Immunol 37:493-501. 260. Chuang SS, Kumaresan PR, Mathew PA. 2001. 2B4 (CD244)-Mediated Activation of Cytotoxicity and IFN-γ Release in Human NK Cells Involves Distinct Pathways. The Journal of Immunology 167:6210-6216. 261. Veillette A, Dong Z, Perez-Quintero LA, Zhong MC, Cruz-Munoz ME. 2009. Importance and mechanism of 'switch' function of SAP family adapters. Immunol Rev 232:229-239. 262. Watzl C, Long EO. 2003. Natural killer cell inhibitory receptors block actin cytoskeleton- dependent recruitment of 2B4 (CD244) to lipid rafts. J Exp Med 197:77-85. 263. Eissmann P, Beauchamp L, Wooters J, Tilton JC, Long EO, Watzl C. 2005. Molecular basis for positive and negative signaling by the natural killer cell receptor 2B4 (CD244). Blood 105:4722-4729. 264. Nakajima H, Cella M, Bouchon A, Grierson HL, Lewis J, Duckett CS, Cohen JI, Colonna M. 2000. Patients with X-linked lymphoproliferative disease have a defect in 2B4 receptor- mediated NK cell cytotoxicity. Eur J Immunol 30:3309-3318. 265. Parolini S, Bottino C, Falco M, Augugliaro R, Giliani S, Franceschini R, Ochs HD, Wolf H, Bonnefoy JY, Biassoni R, Moretta L, Notarangelo LD, Moretta A. 2000. X-linked lymphoproliferative disease. 2B4 molecules displaying inhibitory rather than activating function are responsible for the inability of natural killer cells to kill Epstein-Barr virus-infected cells. J Exp Med 192:337-346. 266. Tangye SG, Phillips JH, Lanier LL, Nichols KE. 2000. Functional requirement for SAP in 2B4-mediated activation of human natural killer cells as revealed by the X-linked lymphoproliferative syndrome. J Immunol 165:2932-2936. 267. Chlewicki LK, Velikovsky CA, Balakrishnan V, Mariuzza RA, Kumar V. 2008. Molecular basis of the dual functions of 2B4 (CD244). J Immunol 180:8159-8167. 268. Sandusky MM, Messmer B, Watzl C. 2006. Regulation of 2B4 (CD244)-mediated NK cell activation by ligand-induced receptor modulation. European Journal of Immunology 36:3268- 3276. 269. Watzl C, Stebbins CC, Long EO. 2000. NK cell inhibitory receptors prevent tyrosine phosphorylation of the activation receptor 2B4 (CD244). J Immunol 165:3545-3548. 270. Shibuya A, Campbell D, Hannum C, Yssel H, Franz-Bacon K, McClanahan T, Kitamura T, Nicholl J, Sutherland GR, Lanier LL, Phillips JH. 1996. DNAM-1, a novel adhesion molecule involved in the cytolytic function of T lymphocytes. Immunity 4:573-581. 271. Bottino C, Castriconi R, Pende D, Rivera P, Nanni M, Carnemolla B, Cantoni C, Grassi J, Marcenaro S, Reymond N, Vitale M, Moretta L, Lopez M, Moretta A. 2003. Identification of PVR (CD155) and Nectin-2 (CD112) as cell surface ligands for the human DNAM-1 (CD226) activating molecule. J Exp Med 198:557-567. 272. Chan CJ, Smyth MJ, Martinet L. 2014. Molecular mechanisms of natural killer cell activation in response to cellular stress. Cell Death Differ 21:5-14. 273. Sakisaka T, Takai Y. 2004. Biology and pathology of nectins and nectin-like molecules. Curr Opin Cell Biol 16:513-521. 274. Carlsten M, Norell H, Bryceson YT, Poschke I, Schedvins K, Ljunggren HG, Kiessling R, Malmberg KJ. 2009. Primary human tumor cells expressing CD155 impair tumor targeting by down-regulating DNAM-1 on NK cells. J Immunol 183:4921-4930. 275. de Andrade LF, Smyth MJ, Martinet L. 2014. DNAM-1 control of natural killer cells functions through nectin and nectin-like proteins. Immunol Cell Biol 92:237-244.

- 126 -

276. Chan CJ, Martinet L, Gilfillan S, Souza-Fonseca-Guimaraes F, Chow MT, Town L, Ritchie DS, Colonna M, Andrews DM, Smyth MJ. 2014. The receptors CD96 and CD226 oppose each other in the regulation of natural killer cell functions. Nat Immunol 15:431-438. 277. Walzer T, Dalod M, Robbins SH, Zitvogel L, Vivier E. 2005. Natural-killer cells and dendritic cells: "l'union fait la force". Blood 106:2252-2258. 278. Pende D, Castriconi R, Romagnani P, Spaggiari GM, Marcenaro S, Dondero A, Lazzeri E, Lasagni L, Martini S, Rivera P, Capobianco A, Moretta L, Moretta A, Bottino C. 2006. Expression of the DNAM-1 ligands, Nectin-2 (CD112) and poliovirus receptor (CD155), on dendritic cells: relevance for natural killer-dendritic cell interaction. Blood 107:2030-2036. 279. Seth S, Georgoudaki AM, Chambers BJ, Qiu Q, Kremmer E, Maier MK, Czeloth N, Ravens I, Foerster R, Bernhardt G. 2009. Heterogeneous expression of the adhesion receptor CD226 on murine NK and T cells and its function in NK-mediated killing of immature dendritic cells. J Leukoc Biol 86:91-101. 280. Ardolino M, Zingoni A, Cerboni C, Cecere F, Soriani A, Iannitto ML, Santoni A. 2011. DNAM-1 ligand expression on Ag-stimulated T lymphocytes is mediated by ROS-dependent activation of DNA-damage response: relevance for NK-T cell interaction. Blood 117:4778-4786. 281. Nabekura T, Kanaya M, Shibuya A, Fu G, Gascoigne NR, Lanier LL. 2014. Costimulatory molecule DNAM-1 is essential for optimal differentiation of memory natural killer cells during mouse cytomegalovirus infection. Immunity 40:225-234. 282. Welch MJ, Teijaro JR, Lewicki HA, Colonna M, Oldstone MB. 2012. CD8 T cell defect of TNF-alpha and IL-2 in DNAM-1 deficient mice delays clearance in vivo of a persistent virus infection. Virology 429:163-170. 283. Matusali G, Potesta M, Santoni A, Cerboni C, Doria M. 2012. The human immunodeficiency virus type 1 Nef and Vpu proteins downregulate the natural killer cell-activating ligand PVR. J Virol 86:4496-4504. 284. Tomasec P, Wang ECY, Davison AJ, Vojtesek B, Armstrong M, Griffin C, McSharry BP, Morris RJ, Llewellyn-Lacey S, Rickards C, Nomoto A, Sinzger C, Wilkinson GWG. 2005. Downregulation of natural killer cell-activating ligand CD155 by human cytomegalovirus UL141. Nat Immunol 6:181-188. 285. Prod'homme V, Sugrue DM, Stanton RJ, Nomoto A, Davies J, Rickards CR, Cochrane D, Moore M, Wilkinson GW, Tomasec P. 2010. Human cytomegalovirus UL141 promotes efficient downregulation of the natural killer cell activating ligand CD112. J Gen Virol 91:2034- 2039. 286. Shibuya K, Lanier LL, Phillips JH, Ochs HD, Shimizu K, Nakayama E, Nakauchi H, Shibuya A. 1999. Physical and functional association of LFA-1 with DNAM-1 adhesion molecule. Immunity 11:615-623. 287. Springer TA. 1994. Traffic signals for lymphocyte recirculation and leukocyte emigration: the multistep paradigm. Cell 76:301-314. 288. Krensky AM, Sanchez-Madrid F, Robbins E, Nagy JA, Springer TA, Burakoff SJ. 1983. The functional significance, distribution, and structure of LFA-1, LFA-2, and LFA-3: cell surface antigens associated with CTL-target interactions. J Immunol 131:611-616. 289. Gross CC, Brzostowski JA, Liu D, Long EO. 2010. Tethering of intercellular adhesion molecule on target cells is required for LFA-1-dependent NK cell adhesion and granule polarization. J Immunol 185:2918-2926. 290. Almeida CR, Davis DM. 2006. Segregation of HLA-C from ICAM-1 at NK cell immune synapses is controlled by its cell surface density. J Immunol 177:6904-6910. 291. Liu D, Bryceson YT, Meckel T, Vasiliver-Shamis G, Dustin ML, Long EO. 2009. Integrin- dependent organization and bidirectional vesicular traffic at cytotoxic immune synapses. Immunity 31:99-109.

- 127 -

292. Schleinitz N, March ME, Long EO. 2008. Recruitment of Activation Receptors at Inhibitory NK Cell Immune Synapses. PLOS ONE 3:e3278. 293. Barber DF, Long EO. 2003. Coexpression of CD58 or CD48 with intercellular adhesion molecule 1 on target cells enhances adhesion of resting NK cells. J Immunol 170:294-299. 294. Bryceson YT, March ME, Barber DF, Ljunggren HG, Long EO. 2005. Cytolytic granule polarization and degranulation controlled by different receptors in resting NK cells. J Exp Med 202:1001-1012. 295. Barber DF, Faure M, Long EO. 2004. LFA-1 contributes an early signal for NK cell cytotoxicity. J Immunol 173:3653-3659. 296. Gismondi A, Jacobelli J, Mainiero F, Paolini R, Piccoli M, Frati L, Santoni A. 2000. Cutting edge: functional role for proline-rich tyrosine kinase 2 in NK cell-mediated natural cytotoxicity. J Immunol 164:2272-2276. 297. Mace EM, Zhang J, Siminovitch KA, Takei F. 2010. Elucidation of the integrin LFA-1– mediated signaling pathway of actin polarization in natural killer cells. Blood 116:1272-1279. 298. Riteau B, Barber DF, Long EO. 2003. Vav1 Phosphorylation Is Induced by β2 Integrin Engagement on Natural Killer Cells Upstream of Actin Cytoskeleton and Lipid Raft Reorganization. The Journal of Experimental Medicine 198:469-474. 299. Carpen O, Pallai P, Staunton DE, Springer TA. 1992. Association of intercellular adhesion molecule-1 (ICAM-1) with actin-containing cytoskeleton and alpha-actinin. J Cell Biol 118:1223-1234. 300. Helander TS, Carpen O, Turunen O, Kovanen PE, Vaheri A, Timonen T. 1996. ICAM-2 redistributed by ezrin as a target for killer cells. Nature 382:265-268. 301. Heiska L, Alfthan K, Gronholm M, Vilja P, Vaheri A, Carpen O. 1998. Association of ezrin with intercellular adhesion molecule-1 and -2 (ICAM-1 and ICAM-2). Regulation by phosphatidylinositol 4, 5-bisphosphate. J Biol Chem 273:21893-21900. 302. Celli L, Ryckewaert JJ, Delachanal E, Duperray A. 2006. Evidence of a functional role for interaction between ICAM-1 and nonmuscle alpha-actinins in leukocyte diapedesis. J Immunol 177:4113-4121. 303. Chen X, Allan DS, Krzewski K, Ge B, Kopcow H, Strominger JL. 2006. CD28-stimulated ERK2 phosphorylation is required for polarization of the microtubule organizing center and granules in YTS NK cells. Proc Natl Acad Sci U S A 103:10346-10351. 304. Luque I, Reyburn H, Strominger JL. 2000. Expression of the CD80 and CD86 molecules enhances cytotoxicity by human natural killer cells. Hum Immunol 61:721-728. 305. Wilson JL, Charo J, Martin-Fontecha A, Dellabona P, Casorati G, Chambers BJ, Kiessling R, Bejarano MT, Ljunggren HG. 1999. NK cell triggering by the human costimulatory molecules CD80 and CD86. J Immunol 163:4207-4212. 306. Chambers BJ, Salcedo M, Ljunggren HG. 1996. Triggering of natural killer cells by the costimulatory molecule CD80 (B7-1). Immunity 5:311-317. 307. Sansom DM. 2000. CD28, CTLA-4 and their ligands: who does what and to whom? Immunology 101:169-177. 308. Nagler A, Lanier LL, Cwirla S, Phillips JH. 1989. Comparative studies of human FcRIII- positive and negative natural killer cells. J Immunol 143:3183-3191. 309. Galea-Lauri J, Darling D, Gan SU, Krivochtchapov L, Kuiper M, Gaken J, Souberbielle B, Farzaneh F. 1999. Expression of a variant of CD28 on a subpopulation of human NK cells: implications for B7-mediated stimulation of NK cells. J Immunol 163:62-70. 310. Parham P, Norman PJ, Abi-Rached L, Guethlein LA. 2011. Variable NK cell receptors exemplified by human KIR3DL1/S1. J Immunol 187:11-19. 311. Carr WH, Rosen DB, Arase H, Nixon DF, Michaelsson J, Lanier LL. 2007. Cutting Edge: KIR3DS1, a gene implicated in resistance to progression to AIDS, encodes a DAP12-associated receptor expressed on NK cells that triggers NK cell activation. J Immunol 178:647-651. - 128 -

312. Jiang Y, Chen O, Cui C, Zhao B, Han X, Zhang Z, Liu J, Xu J, Hu Q, Liao C, Shang H. 2013. KIR3DS1/L1 and HLA-Bw4-80I are associated with HIV disease progression among HIV typical progressors and long-term nonprogressors. BMC Infect Dis 13:405. 313. Trydzenskaya H, Juerchott K, Lachmann N, Kotsch K, Kunert K, Weist B, Schonemann C, Schindler R, Nickel P, Melzig MF, Hugo C, Thomusch O, Neumann AU, Reinke P, Babel N. 2013. The genetic predisposition of natural killer cell to BK virus-associated nephropathy in renal transplant patients. Kidney Int 84:359-365. 314. Rivero-Juarez A, Gonzalez R, Camacho A, Manzanares-Martin B, Caruz A, Martinez- Peinado A, Torre-Cisneros J, Pineda JA, Pena J, Rivero A. 2013. Natural killer KIR3DS1 is closely associated with HCV viral clearance and sustained virological response in HIV/HCV patients. PLoS One 8:e61992. 315. O'Connor GM, Guinan KJ, Cunningham RT, Middleton D, Parham P, Gardiner CM. 2007. Functional polymorphism of the KIR3DL1/S1 receptor on human NK cells. J Immunol 178:235- 241. 316. Cella M, Longo A, Ferrara GB, Strominger JL, Colonna M. 1994. NK3-specific natural killer cells are selectively inhibited by Bw4-positive HLA alleles with isoleucine 80. J Exp Med 180:1235-1242. 317. Gillespie GM, Bashirova A, Dong T, McVicar DW, Rowland-Jones SL, Carrington M. 2007. Lack of KIR3DS1 binding to MHC class I Bw4 tetramers in complex with CD8+ T cell epitopes. AIDS Res Hum Retroviruses 23:451-455. 318. Arosa FA, Santos SG, Powis SJ. 2007. Open conformers: the hidden face of MHC-I molecules. Trends Immunol 28:115-123. 319. Boyle LH, Gillingham AK, Munro S, Trowsdale J. 2006. Selective export of HLA-F by its cytoplasmic tail. J Immunol 176:6464-6472. 320. Lepin EJ, Bastin JM, Allan DS, Roncador G, Braud VM, Mason DY, van der Merwe PA, McMichael AJ, Bell JI, Powis SH, O'Callaghan CA. 2000. Functional characterization of HLA-F and binding of HLA-F tetramers to ILT2 and ILT4 receptors. Eur J Immunol 30:3552- 3561. 321. Lee N, Geraghty DE. 2003. HLA-F surface expression on B cell and monocyte cell lines is partially independent from tapasin and completely independent from TAP. J Immunol 171:5264- 5271. 322. Shimizu Y, Geraghty DE, Koller BH, Orr HT, DeMars R. 1988. Transfer and expression of three cloned human non-HLA-A,B,C class I major histocompatibility complex genes in mutant lymphoblastoid cells. Proc Natl Acad Sci U S A 85:227-231. 323. Wainwright SD, Biro PA, Holmes CH. 2000. HLA-F is a predominantly empty, intracellular, TAP-associated MHC class Ib protein with a restricted expression pattern. J Immunol 164:319- 328. 324. Lee N, Ishitani A, Geraghty DE. 2010. HLA-F is a surface marker on activated lymphocytes. Eur J Immunol 40:2308-2318. 325. Naranbhai V, Altfeld M, Abdool Karim Q, Ndung'u T, Abdool Karim SS, Carr WH. 2012. Natural killer cell function in women at high risk for HIV acquisition: insights from a microbicide trial. Aids 26:1745-1753. 326. Kamya P, Boulet S, Tsoukas CM, Routy J-P, Thomas R, Côté P, Boulassel M-R, Baril J-G, Kovacs C, Migueles SA, Connors M, Suscovich TJ, Brander C, Tremblay CL, Bernard N, for the Canadian Cohort of HIVISP. 2011. Receptor-Ligand Requirements for Increased NK Cell Polyfunctional Potential in Slow Progressors Infected with HIV-1 Coexpressing KIR3DL1*h/*y and HLA-B*57. Journal of Virology 85:5949-5960. 327. Tiemessen CT, Shalekoff S, Meddows-Taylor S, Schramm DB, Papathanasopoulos MA, Gray GlE, Sherman GG, Coovadia AH, Kuhn L. 2010. Natural killer cells that respond to

- 129 -

HIV-1 peptides are associated with control of HIV-1 infection. The Journal of infectious diseases 202:1444-1453. 328. Jost S, Tomezsko PJ, Rands K, Toth I, Lichterfeld M, Gandhi RT, Altfeld M. 2014. CD4+ T-cell help enhances NK cell function following therapeutic HIV-1 vaccination. J Virol 88:8349- 8354. 329. Tomescu C, Mavilio D, Montaner LJ. 2015. Lysis of HIV-1-infected autologous CD4+ primary T cells by interferon-alpha-activated NK cells requires NKp46 and NKG2D. Aids 29:1767-1773. 330. Kim SY, Byrn R, Groopman J, Baltimore D. 1989. Temporal aspects of DNA and RNA synthesis during human immunodeficiency virus infection: evidence for differential gene expression. Journal of Virology 63:3708-3713. 331. Petravic J, Ellenberg P, Chan ML, Paukovics G, Smyth RP, Mak J, Davenport MP. 2014. Intracellular dynamics of HIV infection. J Virol 88:1113-1124. 332. Geyer M, Fackler OT, Peterlin BM. 2001. Structure–function relationships in HIV-1 Nef. EMBO Reports 2:580-585. 333. Fackler OT, Luo W, Geyer M, Alberts AS, Peterlin BM. 1999. Activation of Vav by Nef induces cytoskeletal rearrangements and downstream effector functions. Mol Cell 3:729-739. 334. Giese SI, Woerz I, Homann S, Tibroni N, Geyer M, Fackler OT. 2006. Specific and distinct determinants mediate membrane binding and lipid raft incorporation of HIV-1(SF2) Nef. Virology 355:175-191. 335. Das SR, Jameel S. 2005. Biology of the HIV Nef protein. Indian J Med Res 121:315-332. 336. Malbec M, Sourisseau M, Guivel-Benhassine F, Porrot F, Blanchet F, Schwartz O, Casartelli N. 2013. HIV-1 Nef promotes the localization of Gag to the cell membrane and facilitates viral cell-to-cell transfer. Retrovirology 10:80. 337. Luria S, Chambers I, Berg P. 1991. Expression of the type 1 human immunodeficiency virus Nef protein in T cells prevents antigen receptor-mediated induction of interleukin 2 mRNA. Proc Natl Acad Sci U S A 88:5326-5330. 338. Baur AS, Sawai ET, Dazin P, Fantl WJ, Cheng-Mayer C, Peterlin BM. 1994. HIV-1 Nef leads to inhibition or activation of T cells depending on its intracellular localization. Immunity 1:373-384. 339. Aiken C, Konner J, Landau NR, Lenburg ME, Trono D. 1994. Nef induces CD4 endocytosis: Requirement for a critical dileucine motif in the membrane-proximal CD4 cytoplasmic domain. Cell 76:853-864. 340. Greenberg ME, Bronson S, Lock M, Neumann M, Pavlakis GN, Skowronski J. 1997. Co‐ localization of HIV‐1 Nef with the AP‐2 adaptor protein complex correlates with Nef‐induced CD4 down‐regulation. The EMBO Journal 16:6964-6976. 341. Ren X, Park SY, Bonifacino JS, Hurley JH. 2014. How HIV-1 Nef hijacks the AP-2 clathrin adaptor to downregulate CD4. eLife 3:e01754. 342. Chaudhuri R, Lindwasser OW, Smith WJ, Hurley JH, Bonifacino JS. 2007. Downregulation of CD4 by human immunodeficiency virus type 1 Nef is dependent on clathrin and involves direct interaction of Nef with the AP2 clathrin adaptor. J Virol 81:3877-3890. 343. Lindwasser OW, Chaudhuri R, Bonifacino JS. 2007. Mechanisms of CD4 downregulation by the Nef and Vpu proteins of primate immunodeficiency viruses. Curr Mol Med 7. 344. Chaudhuri R, Mattera R, Lindwasser OW, Robinson MS, Bonifacino JS. 2009. A basic patch on alpha-adaptin is required for binding of human immunodeficiency virus type 1 Nef and cooperative assembly of a CD4-Nef-AP-2 complex. J Virol 83:2518-2530. 345. Lindwasser OW, Smith WJ, Chaudhuri R, Yang P, Hurley JH, Bonifacino JS. 2008. A diacidic motif in human immunodeficiency virus type 1 Nef is a novel determinant of binding to AP-2. J Virol 82:1166-1174.

- 130 -

346. Pham TN, Lukhele S, Hajjar F, Routy J-P, Cohen ÉA. 2014. HIV Nef and Vpu protect HIV- infected CD4+ T cells from antibody-mediated cell lysis through down-modulation of CD4 and BST2. Retrovirology 11:15. 347. Michel N, Allespach I, Venzke S, Fackler OT, Keppler OT. 2005. The Nef Protein of Human Immunodeficiency Virus Establishes Superinfection Immunity by a Dual Strategy to Downregulate Cell-Surface CCR5 and CD4. Current Biology 15:714-723. 348. Venzke S, Michel N, Allespach I, Fackler OT, Keppler OT. 2006. Expression of Nef Downregulates CXCR4, the Major Coreceptor of Human Immunodeficiency Virus, from the Surfaces of Target Cells and Thereby Enhances Resistance to Superinfection. Journal of Virology 80:11141-11152. 349. Robinson HL, Zinkus DM. 1990. Accumulation of human immunodeficiency virus type 1 DNA in T cells: results of multiple infection events. Journal of Virology 64:4836-4841. 350. Juan L. 2003. The Physiological Relevance of CD4 Receptor Down-Modulation During HIV Infection. Current HIV Research 1:167-184. 351. Veillette M, Désormeaux A, Medjahed H, Gharsallah N-E, Coutu M, Baalwa J, Guan Y, Lewis G, Ferrari G, Hahn BH, Haynes BF, Robinson JE, Kaufmann DE, Bonsignori M, Sodroski J, Finzi A. 2014. Interaction with Cellular CD4 Exposes HIV-1 Envelope Epitopes Targeted by Antibody-Dependent Cell-Mediated Cytotoxicity. Journal of Virology 88:2633- 2644. 352. Brown A, Gartner S, Kawano T, Benoit N, Cheng-Mayer C. 2005. HLA-A2 down-regulation on primary human macrophages infected with an M-tropic EGFP-tagged HIV-1 reporter virus. J Leukoc Biol 78:675-685. 353. Mangasarian A, Piguet V, Wang JK, Chen YL, Trono D. 1999. Nef-induced CD4 and major histocompatibility complex class I (MHC-I) down-regulation are governed by distinct determinants: N-terminal alpha helix and proline repeat of Nef selectively regulate MHC-I trafficking. J Virol 73:1964-1973. 354. Dikeakos JD, Thomas L, Kwon G, Elferich J, Shinde U, Thomas G. 2012. An interdomain binding site on HIV-1 Nef interacts with PACS-1 and PACS-2 on endosomes to down-regulate MHC-I. Mol Biol Cell 23:2184-2197. 355. Akari H, Arold S, Fukumori T, Okazaki T, Strebel K, Adachi A. 2000. Nef-Induced Major Histocompatibility Complex Class I Down-Regulation Is Functionally Dissociated from Its Virion Incorporation, Enhancement of Viral Infectivity, and CD4 Down-Regulation. Journal of Virology 74:2907-2912. 356. Piguet V, Wan L, Borel C, Mangasarian A, Demaurex N, Thomas G, Trono D. 2000. HIV-1 Nef protein binds to the cellular protein PACS-1 to downregulate class I major histocompatibility complexes. Nat Cell Biol 2:163-167. 357. Le Gall S, Erdtmann L, Benichou S, Berlioz-Torrent C, Liu L, Benarous R, Heard J-M, Schwartz O. 1998. Nef Interacts with the μ Subunit of Clathrin Adaptor Complexes and Reveals a Cryptic Sorting Signal in MHC I Molecules. Immunity 8:483-495. 358. Williams M, Roeth JF, Kasper MR, Fleis RI, Przybycin CG, Collins KL. 2002. Direct binding of human immunodeficiency virus type 1 Nef to the major histocompatibility complex class I (MHC-I) cytoplasmic tail disrupts MHC-I trafficking. J Virol 76:12173-12184. 359. Blagoveshchenskaya AD, Thomas L, Feliciangeli SF, Hung CH, Thomas G. 2002. HIV-1 Nef downregulates MHC-I by a PACS-1- and PI3K-regulated ARF6 endocytic pathway. Cell 111:853-866. 360. Kasper MR, Collins KL. 2003. Nef-Mediated Disruption of HLA-A2 Transport to the Cell Surface in T Cells. Journal of Virology 77:3041-3049. 361. Lubben NB, Sahlender DA, Motley AM, Lehner PJ, Benaroch P, Robinson MS. 2007. HIV- 1 Nef-induced Down-Regulation of MHC Class I Requires AP-1 and Clathrin but Not PACS-1 and Is Impeded by AP-2. Molecular Biology of the Cell 18:3351-3365. - 131 -

362. Swann SA, Williams M, Story CM, Bobbitt KR, Fleis R, Collins KL. 2001. HIV-1 Nef blocks transport of MHC class I molecules to the cell surface via a PI 3-kinase-dependent pathway. Virology 282:267-277. 363. Jia X, Singh R, Homann S, Yang H, Guatelli J, Xiong Y. 2012. Structural basis of evasion of cellular adaptive immunity by HIV-1 Nef. Nat Struct Mol Biol 19:701-706. 364. Roeth JF, Collins KL. 2006. Human Immunodeficiency Virus Type 1 Nef: Adapting to Intracellular Trafficking Pathways. Microbiology and Molecular Biology Reviews 70:548-563. 365. Mahiti M, Toyoda M, Jia X, Kuang XT, Mwimanzi F, Mwimanzi P, Walker BD, Xiong Y, Brumme ZL, Brockman MA, Ueno T. 2016. Relative Resistance of HLA-B to Downregulation by Naturally Occurring HIV-1 Nef Sequences. mBio 7. 366. Rajapaksa US, Li D, Peng YC, McMichael AJ, Dong T, Xu XN. 2012. HLA-B may be more protective against HIV-1 than HLA-A because it resists negative regulatory factor (Nef) mediated down-regulation. Proc Natl Acad Sci U S A 109:13353-13358. 367. Wada H, Matsumoto N, Maenaka K, Suzuki K, Yamamoto K. 2004. The inhibitory NK cell receptor CD94/NKG2A and the activating receptor CD94/NKG2C bind the top of HLA-E through mostly shared but partly distinct sets of HLA-E residues. Eur J Immunol 34:81-90. 368. Bolduan S, Reif T, Schindler M, Schubert U. 2014. HIV-1 Vpu mediated downregulation of CD155 requires alanine residues 10, 14 and 18 of the transmembrane domain. Virology 464- 465:375-384. 369. Davis ZB, Sowrirajan B, Cogswell A, Ward JP, Planelles V, Barker E. 2016. CD155 on HIV- Infected Cells Is Not Modulated by HIV-1 Vpu and Nef but Synergizes with NKG2D Ligands to Trigger NK Cell Lysis of Autologous Primary HIV-Infected Cells. AIDS Res Hum Retroviruses doi:10.1089/aid.2015.0375. 370. Yang F, Liu CQ, Qin XL, Shi JJ. 2010. [HIV-1 Nef regulates ICAM-1 expression on endothelial cells via Erk /Mapk signaling pathway]. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi 26:44-46. 371. Bryceson YT, Ljunggren HG, Long EO. 2009. Minimal requirement for induction of natural cytotoxicity and intersection of activation signals by inhibitory receptors. Blood 114:2657-2666. 372. Das A, Long EO. 2010. Lytic Granule Polarization, rather than Degranulation, is the Preferred Target of Inhibitory Receptors in NK Cells(). Journal of immunology (Baltimore, Md : 1950) 185:10.4049/jimmunol.1001220. 373. Bryceson YT, March ME, Ljunggren H-G, Long EO. 2006. Activation, co–activation, and co– stimulation of resting human NK cells. Immunological reviews 214:10.1111/j.1600- 1065X.2006.00457.x. 374. Aqil M, Mallik S, Bandyopadhyay S, Maulik U, Jameel S. 2015. Transcriptomic Analysis of mRNAs in Human Monocytic Cells Expressing the HIV-1 Nef Protein and Their Exosomes. BioMed Research International 2015:492395. 375. Kishi M, Nishino Y, Ohki K, Kimura T, Ikuta K. 1993. Persistently human immunodeficiency virus type 1-infected T cell clone expressing only doubly spliced mRNA exhibits reduced cell surface CD4 expression. Jpn J Cancer Res 84:153-162.

- 132 -