• Aucun résultat trouvé

Production of large numbers of plasmacytoid dendritic cells with functional activities from CD34+ hematopoietic progenitor cells: use of IL-3

N/A
N/A
Protected

Academic year: 2021

Partager "Production of large numbers of plasmacytoid dendritic cells with functional activities from CD34+ hematopoietic progenitor cells: use of IL-3"

Copied!
13
0
0

Texte intégral

(1)

Production of large numbers of plasmacytoid dendritic cells with functional

activities from CD34

þ

hematopoietic progenitor cells: Use of interleukin-3

Q15

St

ephanie

Demoulin

,

Patrick

Roncarati

,

Philippe

Delvenne

, and

Pascale

Hubert

Laboratory of Experimental Pathology, GIGA-Cancer (Center for Experimental Cancer Research), University of Liege, Liege, Belgium (Received 10 August 2011; revised 20 December 2011; accepted 3 January 2012)

Plasmacytoid dendritic cells (pDC), a subset of dendritic cells characterized by a rapid and massive type-I interferon secretion through the Toll-like receptor pathway in response to viral infection, play important roles in the pathogenesis of several diseases, such as chronic viral infections (e.g., hepatitis C virus, human immunodeficiency virus), autoimmunity (e.g., psori-asis, systemic lupus erythematosus

Q2 ), and cancer. As pDC represent a rare cell type in the peripheral blood, the goal of this study was to develop a new method to efficiently generate large numbers of cells from a limited number of CD34+cord blood progenitors to provide a tool to resolve important questions about how pDC mediate tolerance, autoimmunity, and cancer. Human CD34+hematopoietic progenitor cells isolated from cord blood were cultured with a combination of Flt3-ligand (Flt3L), thrombopoietin (TPO), and one of the following cytokine: interleukin (IL)-3, interferon-b, or prostaglandin E2. Cells obtained in the different culture conditions were analyzed for their phenotype and functional characteristics. The addi-tion of IL-3 cooperates with Flt3L and TPO in the inducaddi-tion of pDC from CD34+ hematopoi-etic progenitor cells. Indeed, Flt3L/TPO alone or supplemented with prostaglandin E2 or interferon-b produced smaller amounts of pDC from hematopoietic progenitor cells. In addi-tion, pDC generated in Flt3L/TPO/IL-3 cultures exhibited morphological, immunohistochem-ical, and functional features of peripheral blood pDC. We showed that IL-3, in association with Flt3L and TPO, provides an advantageous tool for large-scale generation of pDC. This culture condition generated, starting from 23 105CD34+cells, up to 2.6 3 106 pDC presenting features of blood pDC. Ó 2012 ISEH - Society for Hematology and Stem Cells. Published by Elsevier Inc.

Dendritic

Q3 cells (DC) represent a major class of professional antigen-presenting cells characterized by their capacity to prime na€ıve T cells and to initiate primary immune responses [1]. In humans, two major lineages of DC can be distinguished based on differential developmental origins, phenotypes, and anatomical locations, myeloid DC (mDC) and plasmacytoid DC (pDC).

Human pDC represent a rare peripheral cell blood pop-ulation (0.20.8%), which can be distinguished from other blood cells based on selective expression of surface anti-gens BDCA-2 [2,3] and ILT7 [4,5]. Immature pDC also express CD4, CD45RA, CD123, and BDCA-4, but lack expression of the lineage markers CD3, CD19, CD14, CD16, and DC marker CD11c. pDC represent key effectors

in innate and adaptive immune responses. They selectively express two microbial pattern-recognition receptors (i.e., Toll-like receptor [TLR] 7 and TLR9) and play a major role in antiviral immunity by rapidly producing massive amounts of type-1 interferon (IFN-a/b) after viral stimula-tion through inducstimula-tion of the TLR pathway [6,7]. Other consequences of pDC activation include secretion of cyto-kines, such as tumor necrosis factora and interleukin (IL)-6, and acquisition of antigen presentation ability contributing to the recruitment/activation of other cell types, such as DC, natural killer cells, and T cells[8–10]. Q4

pDC fully develop in the bone marrow and migrate into T-cellrich areas of lymphoid organs through high endo-thelial venules under steady-state conditions. However, under pathological conditions, pDC are recruited from lymphoid organs to peripheral tissues through the action of different chemokines and adhesion molecules[6,11–13]. pDC have attracted a growing interest in recent years and fundamental questions remain concerning their Offprint requests to: Stephanie Demoulin, X.X.,

Q1 Laboratory of

Experi-mental Pathology, B^at.B23 Anatomie et Cytologie Pathologiques Tour 3þ4, Avenue de l’H^opital 3, 4000 Liege 1, Belgium; E-mail:stephanie. demoulin@ulg.ac.be 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77

(2)

regulation and activities, especially in tolerance, autoimmu-nity, and cancer. The obtention of pDC in large numbers has proven to be difficult, limiting progress in the under-standing of their functions. Here, we describe a simple method for the large-scale generation of pDC exhibiting morphological, immunohistochemical, and functional features of pDC present in the peripheral blood from a limited number of CD34þcord blood progenitors.

Several cytokines, such as IL-3, prostaglandin E2 (PGE2), and IFN-b have been shown to be implicated in

pDC development, proliferation, and/or survival. IL-3 induces the proliferation of pDC, inhibits their apoptosis

[14,15], and has a proliferative effect on CD34þprogenitor cells [16–18]. CD34þ cells cultured with mesenchymal stem cells or their conditioned medium have been shown to increase the number of cells generated and the percentage of pDC in culture through PGE2 production

[19]. Buelens et al. showed that monocytes cultured in IL-3 and IFN-b give rise to a population of DC showing several characteristics of pDC. Those cells express high levels of CD123 and secreted high levels of IL-6 and tumor necrosis factora [20]. To determine the best method to generate high amounts of pDC in vitro, we added one of those cytokines (i.e., IL-3, IFN-b, or PGE2) to Flt3 ligand

(Flt3L)/thrombopoietin (TPO) CD34þ hematopoietic progenitor cells (HPC) culture as previous studies showed that TPO acts in synergy with Flt3L for the differentiation and the expansion of HPC[21].

We showed that the conditions Flt3L/TPO, Flt3L/TPO/ PGE2, and Flt3L/TPO/IFN-b induced generation of low

numbers of pDC. In contrast, TPO, Flt3L, and IL-3 syner-gistically induce generation ofO2.6  106pDC presenting characteristics of peripheral blood pDC from a small initial number of precursor cells (2  105HPC) after 21 days in culture. These findings suggest that, unlike IFN-b and PGE2, IL-3 might represent a key factor in controlling

pDC development.

Materials and methods

Isolation of pDC from peripheral blood mononuclear cells Peripheral blood mononuclear cells (PBMC) were isolated from leukocyte-enriched Buffy-coats by centrifugation on Ficoll-Hypaque (Lymphoprep, Axis-Shield, Oslo, Norway). After washing PBMC at low centrifugation speed to discard a maximum of plate-lets, pDC were sorted by using a negative cell sorting kit following manufacturer’s instructions (Human Plasmacytoid DC Enrichment kit; StemCell Technologies, Vancouver, Canada).

Generation of pDC from CD34þHPC

CD34þHPC were isolated from human umbilical cord blood ob-tained during normal full-term deliveries. This study was approved by the Ethics Committee of the University Hospital of Liege and informed consent was obtained from donors. CD34þHPC were recovered by Ficoll-Hypaque density-gradient centrifugation (Lymphoprep; Axis-Shield). CD34þcells were isolated from other

mononucleated cells using the MACS Direct CD34 Progenitor Cell Isolation Kit (Miltenyi Biotec GmBH, Bergisch Gladbach, Germany) and MiniMacs separation columns (Miltenyi Biotec), according to manufacturer’s protocol. CD34þcells were cultured in 24-well plates (Nunc, Roskilde, Denmark) at 2 105cells/mL in RPMI medium supplemented with 10% fetal calf serum, 50mM b-mercaptoethanol and 1% penicillin-streptomycin, sodium pyru-vate, and nonessential amino acid and (all purchased from Invitro-gen, Merelbeke, Belgium). To obtain pDC differentiation, TPO (50 ng/mL), Flt3L (100 ng/mL), and one of the following re-combinant human cytokines: IL-3 (20 ng/mL), IFN-b (1000 U/ mL), or PGE2(15 ng/mL), were added to the medium. All

cyto-kines were purchased from Peprotech (Rocky Hill, NJ, USA) except for PGE2 (Cayman Chemicals, Ann Arbor, MI, USA).

Cell cultures were refreshed every 3 days with RPMI medium con-taining designated cytokines. pDC were isolated by using the Human Plasmacytoid DC Enrichment kit (StemCell Technolo-gies). In some experiments, pDC were stimulated during 24 hours with a CpG oligodeoxynucleotide (ODN) at 12mg/ml(CpG ODNQ5

2216: 50-ggGGGACGATCGTCgggggg-30; Eurogentec, Seraing, Belgium).

Flow cytometry analysis

Flow cytometry studies were performed by using procedures pub-lished previously[22]with the following antibodies: CD123-FITC (clone AC145; Miltenyi Biotec), CD11c-allophycocyanin (cloneQ6

B-ly6; BD Pharmingen, Franklin Lakes, NJ, USA), BDCA-4-phycoerythrin (PE) (clone AD5-17F6; Miltenyi Biotec), CD40-Q7

PE (clone 5C3; BD Pharmingen), CD83-PE (clone HB15e; BD Pharmingen), CD86-PE (clone c2331 (FUN-1); BD Pharmingen), CCR7-PE (clone 150503, R&D Systems, Minneapolis, MN, USA), and HLA-DR-PE (clone AB3; DAKO, Glostrup, Den-mark). Fluorescence intensity and positive cell percentages were measured on a FACSCanto (Becton Dickinson, NJ, USA) and data were analyzed using FACSDiva software V 6.1.2 (Becton Dickinson) and FlowJo software (TreeStar, Ashland, OR, USA). Reverse transcription polymerase chain reaction analysis One microgram total RNA extracted from cell cultures (RNeasy mini kit; Qiagen, Valencia, CA, USA) was reverse-transcribed using Superscript II reverse transcriptase (Invitrogen) according to manufacturer’s instructions. Reverse transcription polymerase chain reaction (RT-PCR) were performed using the following primer sequences: TLR9 F: ACTGGAGGTGGCCCCGGG; TLR9 R: CAGGGGTTGGGAGCGTGG; HPRT F: GTTGGATA TAGGCCAGACTTTG TTG; and HPRT R: CAGATGTTTCC AAACTCAACTTGAA (Eurogentec). The housekeeping gene HPRT was used as an internal control.

Cytokine production assays

Culture supernatants collected from pDC cultures were assessed for IFN-a levels by using an enzyme-linked immunosorbent assay kit according to manufacturer’s instructions (PBL Interferon-Source, Piscataway, NJ, USA).

pDC chemotaxis assay

pDC migration was evaluated using a chemotaxis microchamber technique (48-well Boyden microchamber; Neuroprobe, Cabin John, MD, USA) [23]. Human recombinant chemerin (10 pM, 100 pM, 10 nM, or 100 nM; R&D Systems) was added to the lower wells of the chamber. A nonconditioned medium was used as

78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 143 144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186 187

(3)

control for random migration. Conditioned medium of human fibroblasts was used as positive control. A polyvinylpyrollidone-free polycarbonate membrane 5-mM gelatin-coated

Q8 pore filter

(Poretics Corp., Livermore, CA, USA) was placed in the micro-chamber. After cell sorting, 55 mL pDC suspension (2  106 cells/mL) was applied into the upper wells of the chamber. The chamber was incubated for 5 hours at 37C. Cells having migrated to the underside of the filter were fixed and stained with Diff Quick Stain set (Baxter Diagnostics AG, D€udingen, Switzerland). The upper side of the filter was scraped to remove residual nonmigrating cells. One random field was counted per well using an eyepiece with a calibrated grid to evaluate the number of fully migrated cells. Mixed lymphocyte reaction assay

The stimulator population consisted of pDC sorted from periph-eral blood or Flt3L/TPO/IL-3 culture. Those cells were irradiated at 5000 rads and placed in RPMI 5% human pooled AB serum. Varying numbers of stimulator cells (31240,000 cells per well) were added to round-bottomed 96-well Nunclon plates containing 2 105allogeneic PBMC per well. A proliferative response was measured after 5 days of culture by adding 1mCi3H-thymidine to each well. Cells were harvested 18 hours later using an automated sample harvester (Packard, Canberra, Tilburg, The Netherlands) and counted using a liquid scintillation counter (Top Count, Pack-ard). DCs obtained by culturing adherent fraction of PBMC with IL-4 and

granulocyte-Q9 macrophage colony-stimulating factor, as described previously[22,24], were used as positive control. Morphology of pDC

Cytospins of pDC sorted from peripheral blood or Flt3L/TPO/IL-3 cultures were prepared by spinning (200 rpm, 3 minutes) 1 105 cells onto methanol-treated slides. MayGr€unwald Giemsa stain-ing was performed usstain-ing standard procedures and slides were examined using a FSX100 microscope (Olympus, Aartselaar, Belgium).

Statistical analysis

Statistical evaluation of the results was performed using unpaired Student’s t test. Comparisons of means were studied by analysis of variance (ANOVA), followed by a NewmanKeuls multiple comparison test (one-way ANOVA) or a Bonferroni post-test (two-way ANOVA). ANOVA tests were performed on log-transformed data. Differences were considered as statistically significant when p! 0.05. Statistical tests were performed using the GraphPad Prism 5 software (Graph-Pad Software, La Jolla, CA, USA).

Results

IL-3 induces massive expansion of HPC and generation of large numbers of pDC from CD34þHPC cultivated in the presence of Flt3L and TPO

Previous works showed that TPO cooperates with Flt3L in the generation of pDC from CD34þHPC[21]. In this study, we tested the combinations of Flt3L (100 ng/mL) and TPO (50 ng/mL) with other growth factors that have been shown to be implicated in pDC development, proliferation, and/or

survival, such as IL-3 (20 ng/mL), IFN-b (1000 U/mL), and PGE2(15 ng/mL)[14,16,17,19,25].

In six different experiments, 2 105CD34þHPC were incubated in media containing Flt3L/TPO alone or with IL-3, IFN-b, or PGE2for 28 days. Total numbers of generated

cells were determined at periodic intervals between days 14 and 28. Interestingly, IL-3 together with Flt3L and TPO induced a massive expansion of cultured cell numbers from 1.6  107 6 2.4  107 cells at day 14, to 7.2  107 6 4.5  107 cells at day 21 (Fig. 1). ANOVA tests showed that the number of cells generated in Flt3L/TPO/ IL-3 condition was significantly higher at day 21 (p ! 0.001) and 28 (p ! 0.05) compared with the number of cells generated at day 14 in that condition. In addition, statistical test also showed that, after 21 days of culture, the number of cells generated in Flt3L/TPO/IL-3 condition was significantly higher compared with all the other culture conditions tested (Flt3L/TPO, p! 0.01; Flt3L/TPO/PGE2,

p ! 0.05; and Flt3L/TPO/IFN-b, p ! 0.001). After 28 days of culture, numbers of cells generated in Flt3L/TPO/ IL-3 condition was only significantly higher than the Flt3L/TPO/IFN-b (p ! 0.05) condition.

In three independent experiments, we also analyzed the percentages of hematopoietic cell types present in our culture conditions. Cells were examined for their expres-sion of markers related to pDC (BDCA-4þ/CD123þ/ CD11c), mDC (CD1aþ/CD11cþ), T cells (CD3þ), natural killer cells (CD3/CD16þ), B cells (CD19þ), granulocytesQ10 (CD3/CD11aþ/CD14þ and CD3/CD11aþ/CD14) and monocytes (CD3/CD11a/CD14þ) by flow cytometry. We showed that mDC, T cells, and granulocytes were the main cell types produced in the different culture conditions (Fig. 2). By using an anti-CD34 antibody we also showed that only a small number of CD34þHPC remained in the culture medium after 21 days of culture, as in Flt3L/TPO, Figure 1. IL-3 in combination with Flt3L and TPO promotes the prolifer-ation of CD34þHPC. CD34þHPC were cultured with Flt3L and TPO alone or with IL-3, PGE2, or IFN-b for 28 days. Total number of cells were

counted and analyzed at days 14, 21, and 28. Data shown represent mean 6 standard deviation of six independent experiments using different donors.

EQ2 188 189 190 191 192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 240 241 242 243 244 245 246 247 248 249 250 251 252 253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271 272 273 274 275 276 277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297

(4)

Figure 2. Various hematopoietic cell types are produced in Flt3L/TPO culture alone or supplemented with IL-3, PGE2, or IFN-b. Percentages of pDC

(BDCA-4þ/CD123þ/CD11c) (A), mDC (CD1aþ/CD11cþ) (B), T cells (CD3þ) (C), B cells (CD19þ) (D),natural killer cells (CD3/CD16þ) (E),granulo-Q14 cytes (CD3/CD11aþ/CD14þand CD3/CD11aþ/CD14) (F), and monocytes (CD3/CD11a/CD14þ) (G) in cultures, at days 14 (light gray), 21 (dark gray), and 28 (black), were determined by flow cytometry using their corresponding phenotypes. Data are from three different experiments using different donors and mean values are shown as percentages of positive cells6 standard deviation.

298 299 300 301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340 341 342 343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407

(5)

Flt3L/TPO/IL-3, Flt3L/TPO/PGE2, and Flt3L/TPO/IFN-b

conditions, only up to 6.2%, 0.8%, 2%, and 1.3% of cells were CD34þ HPC, respectively (data not shown). At day 14, the maximum yield of pDC was observed using IL-3 in the culture medium. With this condition, 4% 6 1.9% of cells showed a phenotype of fully differentiated pDC (BDCA-4þ/CD123þ/CD11c). The range of increase compared with pDC obtained at day 14 in other culture conditions was from O170% to 500%. After long-term cultures (28 days), the percentage of pDC increased and remained the highest in Flt3L/TPO/IL-3 culture (Fig. 2A). Next, we determined the number of immature pDC based on their percentages in culture and showed that the combination of Flt3L, TPO, and IL-3 induced an enhance-ment in pDC generation. At 14, 21, and 28 days of culture, the number of pDC was higher in Flt3L/TPO/IL-3 condi-tion compared with the other culture condicondi-tions (Fig. 3). The highest number of pDC was detected at day 21 in Flt3L/TPO/IL-3 culture (2.6  1066 1.2  106) and the number of pDC was significantly higher in Flt3L/TPO/IL-3 compared with FltFlt3L/TPO/IL-3L/TPO (p ! 0.05), Flt3L/TPO/ PGE2 (p ! 0.05), and Flt3L/TPO/IFN-b (p ! 0.01)

cultures. These results are concordant with apoptotic tests showing that the Flt3L/TPO/IL-3 condition presents the highest percentages of living cells in culture. Proliferation tests also showed that at day 14, 21, and 28 of culture, cell proliferation was higher in Flt3L/TPO/IL-3 condition than in the other conditions tested. Flt3L/TPO/PGE2

condi-tion was characterized by low percentages of proliferating cells. In addition, IFN-b strongly blocked pDC generation from HPC in Flt3/TPO culture, as this culture condition was characterized by low percentages of proliferating cells and high numbers of apoptotic cells (data not shown).

We showed that Flt3L/TPO/IL-3 condition was respon-sible for the generation of the highest amount of pDC in vitro. We therefore decided to study more extensively cells obtained only with this condition. In addition, as pDC numbers were the highest at day 21 in Flt3L/TPO/ IL-3 culture, subsequent studies were performed on pDC obtained after 21 days of culture.

pDC derived from HPC in Flt3L/TPO/IL-3 culture differentiate into mature pDC after activation by CpG ODN

We performed a phenotypic analysis on pDC sorted from Flt3L/TPO/IL-3 culture at day 21. After pDC isolation, we showed that a majority of cells were CD11c. Only 2% of cells were CD11cþ. BDCA-4þ/CD123þpDC repre-sented 25.7% of CD11c cells. Interestingly, 11.2% of CD11cþ cells expressed the pDC markers BDCA-4þ and CD123þ (Fig. 4A). Sorted-pDC were then cultured 24 hours with IL-3 to assure their survival and with CpG ODN (a pDC stimulus recognized by their TLR9) to induce their maturation. Phenotype analysis of sorted cells cultured in medium complemented with IL-3 alone showed that the

number of CD11c and CD11cþ cells expressing the markers BDCA-4 and CD123 is increased in the presence of IL-3 from 25.7% to 53.4% and from 11.2% to 33.4%, respectively (Fig. 4B). Sorted cells cultured for 24 hours with IL-3 and CpG ODN showed a higher increase of BDCA-4þ/CD123þ/CD11c (from 25.7% to 91.5%) and BDCA-4þ/CD123þ/CD11cþ(from 11.2% to 77.5%) cells compared with sorted cells cultured with IL-3 alone (Fig. 4C). These results demonstrated that BDCA-4þ CD123þCD11ccells are immature pDC that differentiate into mature pDC expressing CD11c when cultured with IL-3, with or without CpG ODN. However, the combination of IL-3 and CpG ODN induced a higher number of mature pDC than IL-3 alone.

We were also interested in determining whether non-sorted pDC exposed to Flt3L, TPO, and IL-3 were capable of maturation with CpG ODN. Nonsorted pDC generated in Flt3L/TPO/IL-3 condition were assessed for their expres-sion of maturation markers (CD40, CD83, CD86, HLA-DR, CCR7, and CD11c) in the presence or absence of CpG ODN. After 24 hours of activation by CpG ODN, we showed that pDC stimulated with CpG ODN were matured. The fluorescence intensity of all maturation markers was increased in stimulated-pDC compared to non-stimulated pDC (Fig. 5).

pDC generated in Flt3L/TPO/IL-3 cultures display characteristics of peripheral blood pDC

In order to determine whether pDC generated in Flt3L/ TPO/IL-3 culture display characteristics of peripheral blood pDC, we sorted pDC from Flt3L/TPO/IL-3 cultures and from peripheral blood by magnetic bead isolation.

Sorted-pDC from Flt3L/TPO/IL-3 cultures (Fig. 6Aa) exhibited typical peripheral blood pDC plasma cell Figure 3. IL-3 induces the generation of large numbers of pDC from CD34þHPC cultivated with Flt3L and TPO. CD34þHPC were cultured with Flt3L and TPO alone (white) or with IL-3 (light gray), PGE2(dark

gray), or IFN-b (black) for 28 days. Numbers of pDC generated at day 14, 21, and 28 in the different culture conditions were determined based on their percentages in culture. Data shown represent mean 6 standard deviation of three independent experiments using different donors. 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517

(6)

morphology (Fig. 6Ab) with a high nucleuscytoplasm ratio on a MayGr€unwald Giemsastained cytospin.

By using a mixed lymphocyte reaction, we showed that pDC generated in Flt3L/TPO/IL-3 culture as well as periph-eral blood pDC have the capacity to provide accessory signals for an efficient proliferation of allogeneic T lymphocytes (Fig. 6B). pDC generated in culture or sorted from peripheral blood did not differ in their ability to stim-ulate an allogeneic response when the stimulator/responder ratio was the lowest (1:40 and 1:20), but the response was significantly more important for the population of pDC

generated in culture when the stimulator/responder ratio was higher (1:10 and 1:5) (p ! 0.05). In addition, as it has already been shown[21,26], the stimulatory capacity of pDC was significantly less important (p ! 0.01) than that of the mDC population used as control in the assay (Fig. 6B).

pDC function is associated with the expression of TLR different from that expressed by mDC[27]. We confirmed by quantitative real-time PCR that, unlike mDC, peripheral blood pDC and pDC produced in our culture system, express high levels of TLR9 messenger RNA (Fig. 6C).

print

&

w

eb

4C

=FPO

Figure 4. CD11cpDC are immature cells and differentiate into mature pDC expressing CD11c when cultured with IL-3, with or without CpG ODN. Sorted-pDC from Flt3L/TPO/IL-3 cultures were analyzed for their expression of CD11c directly after their sorting (A) or 24 hours after maturation with IL-3, with (C) or without (B) CpG ODN. The values correspond to percentages of BDCA-4þ/CD123þpDC in the population of CD11c(P3) and CD11cþcells (P2). Data are representative of three experiments showing similar results.

EQ1 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627

(7)

This messenger RNA expression was correlated to the TLR9 protein level detected in pDC by intracellular stain-ing (Fig. 6D).

We also showed that in pDC sorted from the Flt3L/TPO/ IL-3 culture, TLR9 stimulation by CpG ODN induces the secretion of high amounts of IFN-a (Fig. 6E) compared with unstimulated pDC.

As pDC migrate in response to chemerin due to their expression of CMKLR1 [28–30], we investigated the ability of pDC generated in Flt3L/TPO/IL-3 cultures to migrate in the presence of human recombinant chemerin by using a Boyden chamber assay. A significantly increased migration of pDC was observed in the presence of human fibroblasts media (p! 0.001) compared with a noncondi-tioned medium used as control. We also showed that different concentrations of chemerin (100 pM, 10 nM, and 100 nM) induced a significantly increased migration of pDC, with a peak observed at 10 nM, compared with the nonconditioned medium (Fig. 6F).

These results demonstrated that pDC generated in vitro have characteristics and functional properties similar to those of peripheral blood pDC.

Discussion

Because of their unique ability to secrete IFN-a and their central roles in innate and adaptive immunity, pDC have at-tracted growing interest for several years. pDC investigation is also of potential interest because of their implication in the pathogenesis of several diseases in which they often present a pathogenic/tolerogenic role mainly related to either the increase or the reduction of their function [13]. Sustained overproduction of type-I IFN by pDC in response to host-derived self-nucleic acid is associated with the development of autoimmunity in systemic lupus erythematosus[31]andQ11 psoriasis[32]. On the other hand, pDC have also been shown to present a limited responsiveness to certain viruses (e.g., hepatitis C virus and human immunodeficiency virus Q12)

[33,34]. In addition, in the microenvironment of certain tumors[35–37], pDC are often responsible for the develop-ment of a immunosuppressive/tolerogenic response[38].

Although the function and clinical manipulation of pDC have become topics of great interest, progress in the under-standing of pDC is limited by their low percentage in peripheral blood and the difficulty producing large quanti-ties of these cells in culture. Several groups have already Figure 5. Immature pDC differentiate into mature pDC on activation by CpG ODN. Nonsorted pDC were analyzed for their expression of the maturation markers CD40, CD83, CD86, HLA-DR, CCR7, and CD11c after stimulation with CpG ODN for 24 hours. Fluorescence intensity (x-axis) is plotted against the percent of Max (y-axis), where the maximum y-axis value in absolute count becomes 100% of total. Patterns of isotype-matched control antibodies (full lines), unstimulated pDC (long dashed lines), and CpG ODN-stimulated pDC (dotted lines) are included in each histogram. Data are representative of six independent experiments showing similar results.

628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737

(8)

induced the differentiation of pDC in vitro from CD34þ HPC [14,19,21,39,40]. However, these approaches gener-ated relatively small amounts of pDC. Here, we describe an original procedure using a combination of Flt3L, TPO,

and IL-3 for large-scale generation of pDC exhibiting morphological, immunohistochemical, and functional features of pDC present in the peripheral blood from a limited number of CD34þHPC.

web

4

C

=FPO

Figure 6. pDC generated with Flt3L/TPO/IL-3 cultures present several characteristics of peripheral blood pDC. (A) pDC morphology on a cytospin (MayGr€unwald Giemsa coloration). (a) Sorted pDC from a Flt3L/TPO/IL-3 culture at day 21, (b) sorted pDC from peripheral blood. Magnification  400. (B) Mixed lymphocyte reaction of sorted pDC from peripheral blood or generated in Flt3L/TPO/IL-3 cultures with allogeneic T lymphocytes. Responder cells: lymphocytes T; stimulator cells: pDC. mDC were used as positive control. Data represent mean6 standard deviation of three independent experiments using different donors. (C) Real-time quantitative analysis of TLR9 messenger RNA expression in CpG ODN-stimulated pDC generated in Flt3L/TPO/IL-3 culture (dark gray), peripheral blood pDC (light gray), and mDC (black). Data represent mean6 standard deviation of three independent experiments. (D) TLR9 protein expression analysis on CpG ODN-stimulated pDC generated in Flt3/TPO/IL-3 culture or sorted from peripheral blood. Fluorescence intensity (x-axis) is plotted against the percent of Max (y-axis). Patterns of isotype-matched control antibody (full line), stimulated-pDC generated in Flt3L/TPO/IL-3 culture (long dashed line) and stimulated-pDC sorted from peripheral blood (dotted line) are included in the histogram. (E) Secretion level of IFN-a with (þCpG) or without (CpG) CpG ODN in culture supernatants of sorted pDC from Flt3L/TPO/IL-3 culture. Data represent mean 6 standard deviation (***p ! 0.001) of three independent experiments using different donors. (F) Influence of recombinant chemerin on pDC migration in a Boyden chamber assay. HFM5 conditioned medium of human fibroblasts (positive control); NC 5 nonconditioned medium (negative control). NC was supplemented with four different concentrations (10 pM, 100 pM, 10 nM, and 100 nM) of human recombinant chemerin. Data represent mean6 standard deviation (*p ! 0.05; ***p! 0.001) of three independent experiments using different donors.

738 739 740 741 742 743 744 745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780 781 782 783 784 785 786 787 788 789 790 791 792 793 794 795 796 797 798 799 800 801 802 803 804 805 806 807 808 809 810 811 812 813 814 815 816 817 818 819 820 821 822 823 824 825 826 827 828 829 830 831 832 833 834 835 836 837 838 839 840 841 842 843 844 845 846 847

(9)

Originally, pDC were thought to be in the lymphoid lineage. Interestingly, recent studies indicate that these cells have diverse origins and can develop from lymphoid or myeloid precursors[41,42]. Even if the origin and affilia-tion of pDC is controversial, partly because pDC show features of lymphocytes and DC, clear evidences indicate that Flt3L and TPO might represent major regulators of pDC development. First, previous reports showed that Flt3L is responsible for development of pDC in vitro

[21,39,43,44] and in vivo [45]. Interestingly, mice and human subjects injected with Flt3L have an increase in pDC number [46–48]. Second, TPO has been shown to support the proliferation and long-term expansion of HPC and to induce their differentiation into pDC in synergy with

EQ3 Flt3L [21,49,50]. As IL-3 was reported to induce the

proliferation of pDC, to inhibit their apoptosis [14,15]

and to have a proliferative effect on CD34þHPC in vitro

[16–18], we postulated that this cytokine in combination with Flt3L and TPO could have an impact on pDC genera-tion from CD34þ HPC in culture. Other cytokines (e.g., IFN-b and PGE2) potentially implicated in pDC

develop-ment and proliferation [19,20,25] were also tested in combination with Flt3L and TPO for their induction of pDC from CD34þHPC.

We showed that IL-3 together with Flt3L and TPO al-lowed the generation after 21 days of culture ofO2.6  106pDC from 2 105HPC. Flt3L/TPO alone or in combi-nation with PGE2 induced the generation of smaller

amounts of pDC compared with Flt3L/TPO/IL-3 culture. At day 21, the combination of Flt3L, TPO, and IL-3 was associated with a mean increase of 152% and 158% in pDC production, compared with a culture without IL-3 or with a culture where IL-3 was replaced by PGE2,

respec-tively. In addition, the addition of IFN-b to Flt3L/TPO culture induced a very low proliferation of HPC, high number of apoptotic cells, and inhibited pDC differentia-tion. Our results indicate that, in contrast to PGE2 and

IFN-b, IL-3 is an efficient cofactor in triggering develop-ment and survival of human pDC from CD34þHPC. The effects of IFN-b on pDC production could be progenitor specific. Indeed, the poor pDC differentiation/proliferation effect of IFN-b on CD34þcells might be due to the fact that this cytokine only acts on monocytes to induce their differentiation in DC with pDC characteristics as shown by Buelens et al.[20]and Huang et al.[25].

Interestingly, we demonstrated that CD11cpDC sorted from our culture system at day 21 and stimulated with IL-3 alone or with IL-3 and CpG ODN acquire the expression of the CD11c marker. This implies that IL-3 presents the ability to induce the maturation of a fraction of pDC, as already shown by Grouard et al. [15] and Defays et al.

[51]. It must be emphasized that CD11cþ cells maintain their expression of the pDC markers BDCA-4 and CD123 and do not differentiate into DC. Even if the manipulation of a pure population of immature pDC might require cell

sorting after their generation, our culture system only requires a small quantity (2 105) of HPC, allowing func-tional studies on large quantities of pDC coming from the same donor and does not require pooling pDC obtained from different patients due to their very low frequency in the peripheral blood. We found that BDCA-4/CD123 and BDCA-4/CD123þ cells sorted from cultures might be pDC precursors, as a fraction of those cells differentiated into BDCA4þ/CD123þpDC in the presence of IL-3 alone or in combination with CpG ODN. We also observed an increased expression of the BDCA-4 and CD123 markers in the pDC population after their maturation with CpG ODN and IL-3 when cells were not sorted (data not shown). This implies that although cells generated in culture have been exposed to IL-3 for 21 days, they maintain their capacity to be maturated by CpG ODN.

Cytometry analysis showed that stimulated-pDC ob-tained in Flt3L/TPO/IL-3 cultures exhibit a matured pheno-type as shown by the increased fluorescence intensity of their maturation markers HLA-DR, CD83, CD86, CD40, CCR7, and CD11c. This full pDC matured phenotype was obtained with an IL-3 concentration of 20 ng/mL in culture. We also tested two other concentrations of IL-3 (40 ng/mL and 60 ng/mL) in our Flt3L/TPO culture system in order to determine if we could obtain more benefit for pDC generation or shorten the time frame needed for successful pDC generation. Our results showed that even if we obtained more pDC at days 14 and 21 of culture with 60 ng/mL than with 20 ng/mL of IL-3, the use of 20 ng/mL IL-3 is more appropriate to generate pDC, which could be used to perform functional studies, as pDC gener-ated with 60 ng/mL IL-3 could not be fully matured by CpG ODN in culture (data not shown).

pDC generated in culture displayed characteristics of peripheral blood pDC as demonstrated by their plasma cell morphology, their stimulation of allogeneic T lympho-cytes proliferation in a mixed lymphocyte reaction assay, and their expression of TLR9 messenger RNA. We also showed that pDC generated in the Flt3L/TPO/IL-3 condi-tion keep, after 21 days of culture, their main characteristic, including the secretion of IFN-b, after their stimulation with CpG ODN. Also, pDC generated in culture were che-moattracted in a Boyden chamber assay by chemerin, their chemoattractant.

In conclusion, we have described a simple and reproduc-ible method for the generation, from small amounts of progenitor cells, of a large number of cells with phenotyp-ical, structural, and functional features of pDC. The number of pDC was twice as important as the maximal number of pDC generated in a previous report using a combination of Flt3L and TPO [21]. This ratio is still underestimated, as Chen et al. used a double staining (HLA-DR/CD123) toQ13 identify pDC, which is less specific for pDC than the double staining BDCA-4/CD123 used in this study. Indeed, in the Flt3L/TPO/IL3 culture system, the number of 848 849 850 851 852 853 854 855 856 857 858 859 860 861 862 863 864 865 866 867 868 869 870 871 872 873 874 875 876 877 878 879 880 881 882 883 884 885 886 887 888 889 890 891 892 893 894 895 896 897 898 899 900 901 902 903 904 905 906 907 908 909 910 911 912 913 914 915 916 917 918 919 920 921 922 923 924 925 926 927 928 929 930 931 932 933 934 935 936 937 938 939 940 941 942 943 944 945 946 947 948 949 950 951 952 953 954 955 956 957

(10)

CD123 cells was 1.7 fold more important than the number of BDCA4þ/CD123þcells.

Achievement of generation of high amounts of pDC in vitro opens up an exciting possibilities for the initiation of functional studies allowing for better comprehension of pDC regulation and function in homeostatic and patholog-ical conditions, and also for the development of therapeutic approaches targeting human pDC.

Funding disclosure

This work was supported by a grant from the FNRS (Bourse Televie) and the GIGA imaging and flow cytome-try platform.

Conflict of interest disclosure

No financial interest/relationships with financial interest relating to the topic of this article have been declared.

References

1. Banchereau J, Briere F, Caux C, et al. Immunobiology of dendritic cells. Annu Rev Immunol. 2000;18:767–811.

2. Dzionek A, Sohma Y, Nagafune J, et al. BDCA-2, a novel plasmacy-toid dendritic cell-specific type II C-type lectin, mediates antigen capture and is a potent inhibitor of interferon alpha/beta induction. J Exp Med. 2001;194:1823–1834.

3. Dzionek A, Fuchs A, Schmidt P, et al. 2, 3, and BDCA-4: three markers for distinct subsets of dendritic cells in human periph-eral blood. J Immunol. 2000;165:6037–6046.

4. Rissoan MC, Duhen T, Bridon JM, et al. Subtractive hybridization reveals the expression of immunoglobulin-like transcript 7, Eph-B1, granzyme B, and 3 novel transcripts in human plasmacytoid dendritic cells. Blood. 2002;100:3295–3303.

5. Cho M, Ishida K, Chen J, et al. SAGE library screening reveals ILT7 as a specific plasmacytoid dendritic cell marker that regulates type I IFN production. Int Immunol. 2008;20:155–164.

6. Reizis B, Bunin A, Ghosh HS, Lewis KL, Sisirak V. Plasmacytoid dendritic cells: recent progress and open questions. Annu Rev Immu-nol. 2011;29:163–183.

7. Liu YJ. IPC: professional type 1 interferon-producing cells and plas-macytoid dendritic cell precursors. Annu Rev Immunol. 2005;23: 275–306.

8. Swiecki M, Colonna M. Unraveling the functions of plasmacytoid dendritic cells during viral infections, autoimmunity, and tolerance. Immunol Rev. 2010;234:142–162.

9. Zhang Z, Wang FS. Plasmacytoid dendritic cells act as the most competent cell type in linking antiviral innate and adaptive immune responses. Cell Mol Immunol. 2005;2:411–417.

10. Gilliet M, Cao W, Liu YJ. Plasmacytoid dendritic cells: sensing nu-cleic acids in viral infection and autoimmune diseases. Nat Rev Immu-nol. 2008;8:594–606.

11. Colonna M, Trinchieri G, Liu YJ. Plasmacytoid dendritic cells in immunity. Nat Immunol. 2004;5:1219–1226.

12. McKenna K, Beignon AS, Bhardwaj N. Plasmacytoid dendritic cells: linking innate and adaptive immunity. J Virol. 2005;79:17–27. 13. Sozzani S, Vermi W, Del Prete A, Facchetti F. Trafficking properties of

plasmacytoid dendritic cells in health and disease. Trends Immunol. 2010;31:270–277.

14. Encabo A, Solves P, Mateu E, Sepulveda P, Carbonell-Uberos F, Minana MD. Selective generation of different dendritic cell precursors

from CD34þ cells by interleukin-6 and interleukin-3. Stem Cells. 2004;22:725–740.

15. Grouard G, Rissoan MC, Filgueira L, Durand I, Banchereau J, Liu YJ. The enigmatic plasmacytoid T cells develop into dendritic cells with interleukin (IL)-3 and CD40-ligand. J Exp Med. 1997;185:1101–1111. 16. Saeland S, Caux C, Favre C, et al. Combined and sequential effects of human IL-3 and GM-CSF on the proliferation of CD34þ hematopoi-etic cells from cord blood. Blood. 1989;73:1195–1201.

17. Kawano Y, Takaue Y, Hirao A, et al. Synergistic effect of recombinant interferon-gamma and interleukin-3 on the growth of immature human hematopoietic progenitors. Blood. 1991;77:2118–2121.

18. Astori G, Malangone W, Adami V, et al. A novel protocol that allows short-term stem cell expansion of both committed and pluripotent hematopoietic progenitor cells suitable for clinical use. Blood Cells Mol Dis. 2001;27:715–724. discussion 725717.

19. Chen L, Zhang W, Yue H, et al. Effects of human mesenchymal stem cells on the differentiation of dendritic cells from CD34þ cells. Stem Cells Dev. 2007;16:719–731.

20. Buelens C, Bartholome EJ, Amraoui Z, et al. Interleukin-3 and inter-feron beta cooperate to induce differentiation of monocytes into dendritic cells with potent helper T-cell stimulatory properties. Blood. 2002;99:993–998.

21. Chen W, Antonenko S, Sederstrom JM, et al. Thrombopoietin cooper-ates with FLT3-ligand in the generation of plasmacytoid dendritic cell precursors from human hematopoietic progenitors. Blood. 2004;103: 2547–2553.

22. Hubert P, Greimers R, Franzen-Detrooz E, et al. In vitro propagated dendritic cells from patients with human-papilloma virus-associated preneoplastic lesions of the uterine cervix: use of Flt3 ligand. Cancer Immunol Immunother. 1998;47:81–89.

23. Herfs M, Herman L, Hubert P, et al. High expression of PGE2 enzy-matic pathways in cervical (pre)neoplastic lesions and functional consequences for antigen-presenting cells. Cancer Immunol Immun-other. 2009;58:603–614.

24. Sallusto F, Lanzavecchia A. Efficient presentation of soluble antigen by cultured human dendritic cells is maintained by granulocyte/macro-phage colony-stimulating factor plus interleukin 4 and downregulated by tumor necrosis factor alpha. J Exp Med. 1994;179:1109–1118. 25. Huang YM, Hussien Y, Yarilin D, Xiao BG, Liu YJ, Link H.

Inter-feron-beta induces the development of type 2 dendritic cells. Cyto-kine. 2001;13:264–271.

26. Kohrgruber N, Halanek N, Groger M, et al. Survival, maturation, and function of CD11c- and CD11cþ peripheral blood dendritic cells are differentially regulated by cytokines. J Immunol. 1999;163:3250–3259. 27. Hornung V, Rothenfusser S, Britsch S, et al. Quantitative expression of toll-like receptor 1-10 mRNA in cellular subsets of human peripheral blood mononuclear cells and sensitivity to CpG oligodeoxynucleoti-des. J Immunol. 2002;168:4531–4537.

28. Zabel BA, Silverio AM, Butcher EC. Chemokine-like receptor 1 expression and chemerin-directed chemotaxis distinguish plasmacy-toid from myeloid dendritic cells in human blood. J Immunol. 2005; 174:244–251.

29. Vermi W, Riboldi E, Wittamer V, et al. Role of ChemR23 in directing the migration of myeloid and plasmacytoid dendritic cells to lymphoid organs and inflamed skin. J Exp Med. 2005;201:509–515.

30. Skrzeczynska-Moncznik J, Wawro K, Stefanska A, et al. Potential role of chemerin in recruitment of plasmacytoid dendritic cells to diseased skin. Biochem Biophys Res Commun. 2009;380:323–327.

31. Farkas L, Beiske K, Lund-Johansen F, Brandtzaeg P, Jahnsen FL. Plas-macytoid dendritic cells (natural interferon- alpha/beta-producing cells) accumulate in cutaneous lupus erythematosus lesions. Am J Pathol. 2001;159:237–243.

32. Nestle FO, Conrad C, Tun-Kyi A, et al. Plasmacytoid predendritic cells initiate psoriasis through interferon-alpha production. J Exp Med. 2005;202:135–143. 958 959 960 961 962 963 964 965 966 967 968 969 970 971 972 973 974 975 976 977 978 979 980 981 982 983 984 985 986 987 988 989 990 991 992 993 994 995 996 997 998 999 1000 1001 1002 1003 1004 1005 1006 1007 1008 1009 1010 1011 1012 1013 1014 1015 1016 1017 1018 1019 1020 1021 1022 1023 1024 1025 1026 1027 1028 1029 1030 1031 1032 1033 1034 1035 1036 1037 1038 1039 1040 1041 1042 1043 1044 1045 1046 1047 1048 1049 1050 1051 1052 1053 1054 1055 1056 1057 1058 1059 1060 1061 1062 1063 1064 1065 1066 1067

(11)

33. O’Brien M, Manches O, Sabado RL, et al. Spatiotemporal trafficking of HIV in human plasmacytoid dendritic cells defines a persistently IFN-alpha-producing and partially matured phenotype. J Clin Invest. 2011;121:1088–1101.

34. Lai WK, Curbishley SM, Goddard S, et al. Hepatitis C is associated with perturbation of intrahepatic myeloid and plasmacytoid dendritic cell function. J Hepatol. 2007;47:338–347.

35. Zou W, Machelon V, Coulomb-L’Hermin A, et al. Stromal-derived factor-1 in human tumors recruits and alters the function of plasmacy-toid precursor dendritic cells. Nat Med. 2001;7:1339–1346. 36. Bekeredjian-Ding I, Schafer M, Hartmann E, et al. Tumour-derived

prostaglandin E and transforming growth factor-beta synergize to inhibit plasmacytoid dendritic cell-derived interferon-alpha. Immu-nology. 2009;128:439–450.

37. Treilleux I, Blay JY, Bendriss-Vermare N, et al. Dendritic cell infiltra-tion and prognosis of early stage breast cancer. Clin Cancer Res. 2004; 10:7466–7474.

38. Hirsch I, Caux C, Hasan U, Bendriss-Vermare N, Olive D. Impaired Toll-like receptor 7 and 9 signaling: from chronic viral infections to cancer. Trends Immunol. 2010;31:391–397.

39. Olivier A, Lauret E, Gonin P, Galy A. The Notch ligand delta-1 is a hematopoietic development cofactor for plasmacytoid dendritic cells. Blood. 2006;107:2694–2701.

40. Blom B, Ho S, Antonenko S, Liu YJ. Generation of interferon alpha-producing predendritic cell (Pre-DC)2 from human CD34(þ) hemato-poietic stem cells. J Exp Med. 2000;192:1785–1796.

41. Reizis B. Regulation of plasmacytoid dendritic cell development. Curr Opin Immunol. 2010;22:206–211.

42. Karsunky H, Merad M, Mende I, Manz MG, Engleman EG, Weissman IL. Developmental origin of interferon-alpha-producing dendritic cells from hematopoietic precursors. Exp Hematol. 2005;33:173–181.

43. Gilliet M, Boonstra A, Paturel C, et al. The development of murine plasmacytoid dendritic cell precursors is differentially regulated by FLT3-ligand and granulocyte/macrophage colony-stimulating factor. J Exp Med. 2002;195:953–958.

44. Onai N, Obata-Onai A, Tussiwand R, Lanzavecchia A, Manz MG. Activation of the Flt3 signal transduction cascade rescues and enhances type I interferon-producing and dendritic cell development. J Exp Med. 2006;203:227–238.

45. Maraskovsky E, Daro E, Roux E, et al. In vivo generation of human dendritic cell subsets by Flt3 ligand. Blood. 2000;96:878–884. 46. Maraskovsky E, Brasel K, Teepe M, et al. Dramatic increase in the

numbers of functionally mature dendritic cells in Flt3 ligand-treated mice: multiple dendritic cell subpopulations identified. J Exp Med. 1996;184:1953–1962.

47. Bjorck P. Isolation and characterization of plasmacytoid dendritic cells from Flt3 ligand and granulocyte-macrophage colony-stimulating factor-treated mice. Blood. 2001;98:3520–3526.

48. Pulendran B, Banchereau J, Burkeholder S, et al. Flt3-ligand and gran-ulocyte colony-stimulating factor mobilize distinct human dendritic cell subsets in vivo. J Immunol. 2000;165:566–572.

49. Piacibello W, Sanavio F, Garetto L, et al. Extensive amplification and self-renewal of human primitive hematopoietic stem cells from cord blood. Blood. 1997;89:2644–2653.

50. Arrighi JF, Hauser C, Chapuis B, Zubler RH, Kindler V. Long-term culture of human CD34(þ) progenitors with FLT3-ligand, thrombo-poietin, and stem cell factor induces extensive amplification of a CD34(-)CD14(-) and a CD34(-)CD14(þ) dendritic cell precursor. Blood. 1999;93:2244–2252.

51. Defays A, David A, de Gassart A, et al. BAD-LAMP is a novel biomarker of non-activated human plasmacytoid dendritic cells. Blood. 2011;118:609–617. 1068 1069 1070 1071 1072 1073 1074 1075 1076 1077 1078 1079 1080 1081 1082 1083 1084 1085 1086 1087 1088 1089 1090 1091 1092 1093 1094 1095 1096 1097 1098 1099 1100 1101 1102 1103 1104 1105 1106 1107 1108 1109 1110 1111 1112 1113 1114 1115 1116 1117 1118 1119 1120 1121 1122 1123 1124 1125 1126 1127 1128 1129 1130 1131 1132 1133 1134 1135 1136 1137 1138 1139 1140 1141 1142 1143 1144 1145 1146 1147 1148 1149 1150 1151 1152 1153 1154 1155 1156 1157 1158 1159 1160 1161 1162 1163

(12)

AUTHOR QUERY FORM

Journal:

EXPHEM

Article Number:

2843

Please e-mail or fax your responses and any corrections to:

E-mail:mannd@cadmus.com

Fax: 717-738-9479 or 717-738-9478

Dear Author,

Please check your proof carefully and mark all corrections at the appropriate place in the proof (e.g., by using on-screen annotation in the PDF file) or compile them in a separate list. Note: if you opt to annotate the file with software other than Adobe Reader then please also highlight the appropriate place in the PDF file. To ensure fast publication of your paper please return your corrections within 48 hours.

For correction or revision of any artwork, please consulthttp://www.elsevier.com/artworkinstructions.

Any queries or remarks that have arisen during the processing of your manuscript are listed below and highlighted by flags in the proof.

Location in article

Query / Remark: Click on the Q link to find the query’s location in text Please insert your reply or correction at the corresponding line in the proof

Q1 Please provide degree initials for corresponding author.

Q2 SLE expanded to systemic lupus erythematosus Please correct if necessary.

Q3 Many of the Greek letters in your text had to be re-input. Please check them carefully as you review your proofs.

Q4 NK expanded to natural killer Please correct if necessary.

Q5 In line “In some experiments,...” please check whether the unit is correct or not after 2.

Q6 APC has been expanded to« allophycocyanin” Please correct if necessary

Q7 PE has been defined as phycoerythrin Please correct if necessary.

Q8 gelatin-coated” moved here from after parentheses. Please correct if necessary.

Q9 GM-CSF has been expanded to granulocyte-macrophage colony-stimulating factor Please correct if necessary.

Q10 NK expanded to« natural killer » please correct if necessary.

Q11 SLE expanded to systemic lupus erythematosus Please correct if necessary.

Q12 HCV and HIV expanded to hepatitis C virus and human immunodeficiency virus” Please correct if necessary.

Q13 Please provide reference number for Chen et al.

Q14 NK expanded to natural killer Please correct if necessary.

(13)

EQ2 We have typesetted figures 1 and 3 in single column. Please check and confirm.

EQ3 We have changed the reference citation 51 to 15 and 52 to 51. Please check and confirm.

Figure

Figure 1. IL-3 in combination with Flt3L and TPO promotes the prolifer- prolifer-ation of CD34 þ HPC
Figure 2. Various hematopoietic cell types are produced in Flt3L/TPO culture alone or supplemented with IL-3, PGE 2 , or IFN- b
Figure 3. IL-3 induces the generation of large numbers of pDC from CD34 þ HPC cultivated with Flt3L and TPO
Figure 4. CD11c  pDC are immature cells and differentiate into mature pDC expressing CD11c when cultured with IL-3, with or without CpG ODN.
+3

Références

Documents relatifs

endothelial biomarkers of damage and repair, this study identifies the mobilisation of CD34 + CD45 − EPCs and high levels of s-Fractalkine as specific features of SSc-

In order to explore how this shock tool could affect the penetration rate, he simulated the drilling process of two hydraulic pulsing drilling tools (1- AGT 2- Hydropulse

Objectives: Simulate random variables and illustrate the law of large numbers and the Central Limit Theorem..

Any integer greater than 1 can be written as a product of primes, moreover this expression as a product of primes is unique up to the order of the prime factors.

A real

The use of algebra to simulate the divine foreknowledge of such events, as Bernoulli suggested, was for Leibniz unfeasible: there was no predetermined finite number of

In addition to the cutting face forces and the chamfer forces, it has been shown 16 using an elasto plastic rock behaviour model that some deformation appeared on the back of the

has been obtained in Kabluchko and Wang (2012)... Theorem 1.7 indicates that the construction of polygonal line process ζ n is not adapted to the structure of H¨ older topology