induction of inflammatory cytokines by double- stranded and ...uncorrected proof induction of...

12
UNCORRECTED PROOF Induction of Inflammatory Cytokines by Double- stranded and Single-stranded siRNAs is Sequence- dependent and Requires Endosomal Localization Mouldy Sioud * Department of Immunology Molecular Medicine Group The Norwegian Radium Hospital, Montebello, N-0310 Oslo, Norway The potential induction of inflammatory cytokines and interferon responses by small-interfering RNAs (siRNAs) represents a major obstacle for their use as inhibitors of gene expression. Therapeutic applications of siRNAs will require a better understanding of the mechanisms that trigger such unwanted effects, especially in freshly isolated human cells. Surprisingly, the induction of tumor necrosis factor (TNF-a) and interleukin-6 (IL-6) in adherent peripheral blood mononuclear cells (PBMC) was not restricted to double-stranded siRNAs, because induction was also obtained with single-stranded siRNAs (sense or antisense strands). The immunostimulatory effects were sequence-dependent, since only certain sequences are prone to induce inflammatory responses while other not. The induction of TNF-a, IL-6 and interferon a (IFN-a) was chloroquine sensitive and dependent more likely on endosomal Toll-like receptor signaling. Indeed, no significant immunostimulatory effects were detected when either double or single-stranded siRNAs were delivered directly to cytoplasm via electroporation. Both RNA types activated a NF-kB promoter-driven luciferase gene in transiently transfected human adherent PBMC. Moreover, culture of immature dendritic cells with either double or single-stranded siRNAs stimulated interleukin-12 production and induced the expression of CD83, an activation marker. Interestingly, several double-stranded siRNAs did not induced TNF-a, IL-6 and IFN-a production, however, their single-stranded sense or antisense did. Taken together, the present data indicate for the first time that the induction inflammatory cytokines and IFN-a responses by either double-stranded or single-stranded siRNAs in adherent PBMC is sequence-dependent and requires endosomal intracellular signaling. The finding that endosomal localization of self-RNAs (sense strands) can trigger Toll-like receptor signaling in adherent human PBMC is intriguing because it indicates that endosomal self-RNAs can display a molecular pattern capable for activating innate immunity. q 2005 Elsevier Ltd. All rights reserved. Keywords: RNA interference; siRNAs; innate immunity; Toll-like receptors; inflammatory cytokines *Corresponding author Introduction RNA interference (RNAi) is an evolutionarily conserved post-transcriptional gene-silencing pro- cess, resulting in specific mRNA degradation in several organisms. 1,2 In this process, long double- stranded (ds) RNAs were processed by the RNase III-like enzyme Dicer to 21–25 nucleotides small- interfering RNAs (siRNAs), which are then incor- porated into a protein complex, the RNA-induced silencing complex (RISC). Thereafter, the RISC is remodulated into its active form, which contains the proteins necessary for cleaving the target mRNA at the site, where the guide antisense siRNA strand binds. 2 RNAi and the related phenomenon of 0022-2836/$ - see front matter q 2005 Elsevier Ltd. All rights reserved. Abbreviations used: DC, dendritic cells; iDC, immature dendritic cells; siRNA, small-interfering RNA; RNAi, RNA interference; PKR, protein kinase R; TLR, Toll-like receptor; IL, interleukin-; TNF-a, tumor necrosis factor a; NF-kB, nuclear factor NF-kappa B; INF, interferon; DOTAP, N-[1-(2,3-Dioleoyloxy)propyl]-N,N,N-trimethyl- ammoniunmmethyl-sulfate. E-mail address of the author [email protected] YJMBI 57125—21/3/2005—17:11—SFORSTER—141115—XML – pp. 1–12/APPS doi:10.1016/j.jmb.2005.03.013 J. Mol. Biol. (xxxx) xx, 1–12 DTD 5 ARTICLE IN PRESS 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 123 124 125 126

Upload: others

Post on 13-Feb-2021

7 views

Category:

Documents


0 download

TRANSCRIPT

  • doi:10.1016/j.jmb.2005.03.013 J. Mol. Biol. (xxxx) xx, 1–12

    DTD 5 ARTICLE IN PRESS

    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

    Induction of Inflammatory Cytokines by Double-stranded and Single-stranded siRNAs is Sequence-dependent and Requires Endosomal Localization

    Mouldy Sioud*

    78

    79

    80

    81

    82

    83

    Department of ImmunologyMolecular Medicine GroupThe Norwegian RadiumHospital, Montebello, N-0310Oslo, Norway

    UNC

    0022-2836/$ - see front matter q 2005 E

    Abbreviations used: DC, dendriticdendritic cells; siRNA, small-interfeRNA interference; PKR, protein kinreceptor; IL, interleukin-; TNF-a, tuNF-kB, nuclear factor NF-kappa B;DOTAP, N-[1-(2,3-Dioleoyloxy)propammoniunmmethyl-sulfate.E-mail address of the author mos

    YJMBI 57125—21/3/2005—17:11—SFORSTER

    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

    RE

    CTED

    PRO

    OF

    The potential induction of inflammatory cytokines and interferonresponses by small-interfering RNAs (siRNAs) represents a major obstaclefor their use as inhibitors of gene expression. Therapeutic applications ofsiRNAs will require a better understanding of the mechanisms that triggersuch unwanted effects, especially in freshly isolated human cells.Surprisingly, the induction of tumor necrosis factor (TNF-a) andinterleukin-6 (IL-6) in adherent peripheral blood mononuclear cells(PBMC) was not restricted to double-stranded siRNAs, because inductionwas also obtained with single-stranded siRNAs (sense or antisensestrands). The immunostimulatory effects were sequence-dependent, sinceonly certain sequences are prone to induce inflammatory responses whileother not. The induction of TNF-a, IL-6 and interferon a (IFN-a) waschloroquine sensitive and dependent more likely on endosomal Toll-likereceptor signaling. Indeed, no significant immunostimulatory effects weredetected when either double or single-stranded siRNAs were delivereddirectly to cytoplasm via electroporation. Both RNA types activated aNF-kB promoter-driven luciferase gene in transiently transfected humanadherent PBMC. Moreover, culture of immature dendritic cells with eitherdouble or single-stranded siRNAs stimulated interleukin-12 productionand induced the expression of CD83, an activation marker. Interestingly,several double-stranded siRNAs did not induced TNF-a, IL-6 and IFN-aproduction, however, their single-stranded sense or antisense did. Takentogether, the present data indicate for the first time that the inductioninflammatory cytokines and IFN-a responses by either double-stranded orsingle-stranded siRNAs in adherent PBMC is sequence-dependent andrequires endosomal intracellular signaling. The finding that endosomallocalization of self-RNAs (sense strands) can trigger Toll-like receptorsignaling in adherent human PBMC is intriguing because it indicates thatendosomal self-RNAs can display a molecular pattern capable foractivating innate immunity.

    q 2005 Elsevier Ltd. All rights reserved.

    Keywords: RNA interference; siRNAs; innate immunity; Toll-like receptors;inflammatory cytokines

    113

    R*Corresponding author

    114

    115

    116

    117

    OIntroductionRNA interference (RNAi) is an evolutionarily

    lsevier Ltd. All rights reserve

    cells; iDC, immaturering RNA; RNAi,ase R; TLR, Toll-likemor necrosis factor a;INF, interferon;yl]-N,N,N-trimethyl-

    [email protected]

    —141115—XML – pp. 1–12/APP

    118

    119

    120

    121

    122

    123

    124

    125

    126

    conserved post-transcriptional gene-silencing pro-cess, resulting in specific mRNA degradation inseveral organisms.1,2 In this process, long double-stranded (ds) RNAs were processed by the RNaseIII-like enzyme Dicer to 21–25 nucleotides small-interfering RNAs (siRNAs), which are then incor-porated into a protein complex, the RNA-inducedsilencing complex (RISC). Thereafter, the RISC isremodulated into its active form, which contains theproteins necessary for cleaving the target mRNA atthe site, where the guide antisense siRNA strandbinds.2 RNAi and the related phenomenon of

    d.

    S

  • 2

    DTD 5 ARTICLE IN PRESS

    127

    128

    129

    130

    131

    132

    133

    134

    135

    136

    137

    138

    139

    140

    141

    142

    143

    144

    145

    146

    147

    148

    149

    150

    151

    152

    153

    154

    155

    156

    157

    158

    159

    160

    161

    162

    163

    164

    165

    166

    167

    168

    169

    170

    171

    172

    173

    174

    175

    176

    177

    178

    179

    180

    181

    182

    183

    184

    185

    186

    187

    188

    189

    190

    191

    192

    193

    194

    195

    196

    197

    198

    199

    200

    201

    202

    203

    204

    205

    206

    207

    208

    209

    210

    211

    212

    213

    214

    215

    216

    217

    218

    219

    220

    221

    quelling and post-transcriptional gene silencing(PTGS) have been shown to exist in fungus(Neurospora), plants (Arabidopsis), invertebrates(Drosophila and C. elegans) and embryonic mouseand human cells.2,3 Notably, the cellular role ofRNAi is to maintain the integrity of the genome, todefend cells against viral infection, and to regulatethe expression of cellular genes.

    In general RNAi was difficult to detect in somaticmammalian cells, since dsRNA structures greaterthan 30 bp stimulate the IFN pathway, whichrepresents a host response to viral infection whereseveral genes are activated.4 Much of the interferonresponses are caused by the activation of thedsRNA-dependent protein kinase R (PKR). Theantiviral effect of PKR is in part mediated throughthe phosphorylation of the alpha subunit of theeukaryotic translation initiation factor eIF2a, whichresults in the sequestration of the recycling factoreIF-2b in an inactive complex together with eIF2a-GDP.5 The net effect is global inhibition of proteinsynthesis. However, it has recently been shown thatsynthetic 21 nt siRNAs, the effector in RNAi,transfected into human somatic cells can effectivelybypass activation of the interferon pathway.6 Sincethen siRNAs have become a powerful tool forgenetic analysis and might serve as a potenttherapeutic tool for gene silencing.7,8 In additionto mRNA cleavage, siRNAs can induce chromatinmodification in different organisms. Interestingly, arecent report specified a function for siRNAs in

    UNCO

    RREC

    T

    Table 1. Sequences of the siRNAs used in this study

    SiRNAs Sequence (5 0–3 0) (sense strand)

    1 GGCCUUCCUACCUUCAGACTT2 GAUCAUCUUCUCAAAAUUCTT3 GUUCACCUGAGCCUAAUAGTT4 CUGAUGACCAGCAACUUGATT5 GACAACCAACUAGUGGUGCTT9 GAGGCUGAGACAUAGGCACTT7 GAACUGAUGACAGGGAGGCTT8 GAAGAAGUCGUGCUGCCUUTT9 GGUGACAAGAACAUCUCCATT10 GACCUCAUGUACCACAUUCTT11 GCCAUUGCACUGUGAAUACTT12 GUGAUCAUUCAGAGCCAGCTT13 GGCAUCUGGCUUAAGGUGATT14 GACCCUCGAGUCAACAGAGTT15 GCAUGCCUUGGAAUUCCUUTT16 GGCCGAUUGAUCUCAGCGCTT17 GGCCAAAUUUACAUUCUCCTT18 CCAACUAUGACCAGGAUAATT19 GUAGAUCAAUACCCUACACTT20 GGAGCGCACCAUCUUCUUCTT21 GACUUGAGCGAGCGCUUUUTT22 GAGAUGAUACCACCUGAAATT23 GCAGAUCUGCGUCGGCCAGTT24 GGUUCCAUCGAAUCCUGCATT25 GAGGCAAUCACCAAUAGCATT26 GAAGAUUUGCGCAGUGGACTT27 GUCCGGGCAGGUCUACUUUTT28 GGCAUGGAUCUCAAAGACATT29 CCAACGGCAUGGAUCUCAATT30 UGCCCUUCUACAACCAGGATT31 GCUGGAGUACAACUACAACTT32 GCUGGAGAUCCUGAAGAACTT

    YJMBI 57125—21/3/2005—17:11—SFORSTER—141115—XML – pp. 1–12/A

    DNAmethylation in mammalian cells.9 Contrary towhat was thought; recent experiments indicate thatexogenously delivered siRNAs can activate theinterferon pathway.10–15 However, there are con-flicting reports regarding the extent of off-targeteffects and interferon induction by siRNA inmammalian cells. For example, Rossi and col-leagues noted that none of the three testedchemically made siRNAs induced interferonresponses in human cells.16 These differencescould be due to several factors, including differ-ences in the cell types, reagent preparations andsiRNA sequences. Notably, in all studies only a fewsiRNAs were tested using the same experimentalconditions. To further address the question of whatdetermines siRNA stimulatory function, a compre-hensive analysis of 32 siRNAs targeting differentgenes was performed. The results indicate that thestimulating capacities of either double-stranded,single-stranded sense or antisense siRNAs aresequence dependent and require endosomal com-partments for intracellular signaling.

    FResults and Discussion

    OOThe non-specific effects of siRNAs aresequence-dependent

    siRNA were initially thought to be small enoughto avoid double-stranded RNA responses.6

    ED P

    RTarget gene

    Mouse TNF-aMouse TNF-aHuman HIF-1Human HIF-1Mouse TNF-aMouse TNF-aScrambled siRNAScrambled siRNAHuman neuropilin-1Human PKC-aMouse Csf-1Mouse Csf-1 receptorMouse Csf-1 receptorMouse Csf-1Scrambled siRNAHuman TNF-aRat NG-2Human MMP-9Rat NG-2GFPHuman C20orf55Human CSPPHuman Frizzled-2Human Wnt-1Mouse BasiginHuman CSPPMouse TNF-aMouse TNF-aMouse TNF-aHuman AKT-1GFPHuman cDNA FLJ34902

    PPS

    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

  • 3

    DTD 5 ARTICLE IN PRESS

    253

    254

    255

    256

    257

    258

    259

    260

    261

    262

    263

    264

    265

    266

    267

    268

    269

    270

    271

    272

    273

    274

    275

    276

    277

    278

    279

    280

    281

    282

    283

    284

    285

    286

    287

    288

    289

    290

    291

    292

    293

    294

    295

    296

    297

    298

    299

    300

    301

    302

    303

    304

    305

    306

    307

    308

    309

    310

    311

    312

    313

    314

    315

    316

    317

    318

    319

    320

    321

    322

    323

    324

    325

    326

    However, recent studies indicated that they couldinduce interferons and cytokine responses.10–15

    Although the reported data provide a note ofcaution, the activation of the non-specific pathwaysby siRNAs is not well understood. We havepreviously examined the response of BALB/cmice to systemic delivery of siRNAs and foundinduction of TNF-a and IL-6 by larger RNAs andLPS, but not with a chemically synthesizedsiRNA.15 whilst the same siRNA preparationinduced inflammatory cytokine responses in

    UNCO

    RREC

    T

    YJMBI 57125—21/3/2005—17:11—SFORSTER—141115—XML – pp. 1–12/APP

    adherent PBMC in vitro.15 To assess whether thesiRNA non-specific effects are sequence-dependentand to uncover themolecular mechanisms bywhichsiRNA activate innate immuniy genes, the induc-tion of TNF-a and IL-6 secretion by 32 differentsiRNAs was examined in adherent PBMC, anenriched monocyte population. Freshly isolatedcells were treated with the various siRNAsequences (see Table 1) for 18 hours. SpecificELISA on culture supernatants revealed thataround 50% of the tested siRNAs induced the

    ED P

    ROOF

    Figure 1. Effects of siRNAs uponTNF-a and IL-6 production inadherent PBMC. Cells were trans-fected with the indicated siRNAsmolecules (100 nM) for 18 hoursand then TNF-a (a) and IL-6(b) were measured in the super-natants by ELISA. Results areshown as means of five indepen-dent experiments GSD. Thesequences of the used siRNAs areshown in Table 1. (c) Immunosti-mulatory siRNA induce TNF-aproduction in a dose dependentmanner. Adherent PBMC weretransfected with various concen-trations (5–200 nM) of eithersiRNA 27 for 18 hours. Subse-quently, TNF-a was measured inthe supernatants by ELISA. Allresults represent the mean of fouror more independent experiments.D, cells were incubated with onlyDOTAP and transfection buffer.

    S

    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

  • T

    F

    Figure 2. Characterization ofsiRNA effects. (a) siRNA inducedTNF-a production required lipo-somal delivery of siRNAs. Adher-ent PBMC were incubated for 18hours with either siRNA liposomecomplexes or naked siRNA 27(100 nM) in X-VIVO 15 medium,a nuclease free medium. Subse-quently, TNF-a was measured inthe supernatants by ELISA.Results are shown as means offour independent experimentsGSD. (b) Inhibition of PKR.Adherent PBMC were pretreated,or not, with 2-aminopurine(10 mM) for one hour prior incu-bation with either siRNA 27 or 32(100 nM) for 18 hours. Subse-quently, TNF-a was measured inthe supernatants by ELISA.Results are shown as means offour independent experiments

    GSD. (c) siRNA induced TNF-a production in blood monocytes. Purified CD14-positive cells were incubated witheither siRNA 27 or 32 (100 nM) for 18 hours, and then TNF-awas measured in the supernatants using ELISA. All resultsrepresent the mean of four or more independent experiments

    4

    DTD 5 ARTICLE IN PRESS

    379

    380

    381

    382

    383

    384

    385

    386

    387

    388

    389

    390

    391

    392

    393

    394

    395

    396

    397

    398

    399

    400

    401

    402

    403

    404

    405

    406

    407

    408

    409

    410

    411

    412

    413

    414

    415

    416

    417

    418

    419

    420

    421

    422

    423

    424

    425

    426

    427

    428

    429

    430

    431

    432

    433

    434

    435

    436

    437

    438

    439

    440

    441

    442

    443

    444

    445

    446

    447

    448

    449

    450

    451

    452

    453

    454

    455

    456

    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

    UNCO

    RREC

    production of TNF-a(Figure 1(a)). Adherent PBMCproduced also IL-6 upon stimulation with siRNAscomplexed to DOTAP (Figure 1(b)). The profile ofIL-6 production is comparable to that of TNF-a.Notably, siRNAs varied dramatically in their abilityto induce TNF-a and IL-6 production, suggestingthat their activities are sequence-dependent. Onlysix of the siRNAs examined exhibited a strongimmunostimulatory effect, with siRNA-27 being themost effective under our experimental conditions.

    To determine the immunostimulatory capacity ofsiRNAs, cells were transfected with siRNA 27 at arange of concentrations (5–200 nM). After 18 hoursin culture, secreted TNF-a was assessed by ELISA(Figure 1(c)). As shown, siRNA 27 stimulatedTNF-a production even at low concentrations.Taken together these data indicate that siRNAscan activate the expression of inflammatory cyto-kines, but their affinity and specificity for a potentialcellular target are more likely to be influenced bythe siRNA base composition. Sequence alignmentof the siRNA sequences revealed no significanthomology between the immunostimulatory siR-NAs, suggesting the involvement of RNA tertiarystructures and/or specific dinucleotides.

    Intracellular siRNA delivery is required forcytokine induction

    Having found that the non-specific effects ofsiRNAs are sequence-dependent, I have nextinvestigated whether intracellular siRNA deliverywas required. Therefore siRNAs were addeddirectly to the culture medium without complexingthem to DOTAP. siRNA 27 activated adherentPMBC to secrete TNF-a when complexed toDOTAP. In contrast, free siRNA did not lead to

    YJMBI 57125—21/3/2005—17:11—SFORSTER—141115—XML – pp. 1–12/A

    ED P

    ROOsignificant cytokine production (Figure 2(a)).A nuclease resistant siRNA 27 gave similar results

    as its unmodified version (data not shown). Theseresults argue that siRNAs target mainly intra-cellular compounds. In this respect, a role for PKRin the activation of interferons by siRNAs wasrecently suggested.12

    To explore the involvement of the dsRNArecognition protein PKR in this process, the specificinhibitor of PKR, 2-aminopurine, was used. Cellswere pretreated with 2-aminopurine (10 mM), anormally used concentration, and subsequentlytreated with siRNA liposome complexes. Pretreat-ment with 2-aminopurine reduced, but did notabolish cytokine production by siRNAs (Figure2(b)). These results would indicate the involvementof other cellular targets in siRNA intracellularsignaling.

    siRNAs activate TNF-a production in purifiedhuman monocytes

    Monocytes are essential effector cells in chronicinflammatory disorders and infectious diseases. Toperform their function, monocytes need to beactivated, either via inflammatory cytokines pro-duced by the adaptive immune system or via directstimulation with microbial products. Although ahigh proportion of the adherent PBMC are mono-cytes, the capacity of siRNAs to activate TNF-aproduction in purified blood monocytes wasinvestigated. In these experiments, CD14-positivecell populations were prepared from PBMC bypositive selection with immunomagnetic beadscoated with anti-CD14 monoclonal antibodies(Dynal, Oslo, Norway), and the cells were subse-quently incubated with siRNAs (Figure 2(c)). There

    PPS

  • RREC

    TED

    PROO

    F

    Figure 3. Expression of TLR3 by human monocytes and iDC. (a) Flow cytometry analysis. Freshly isolated monocytesand iDC, monocytes cultured for six days in the presence of GM-CSF and IL-4, were stained either with FITC-labeledanti-TLR3 monoclonal antibody (eBioscience) or control mouse IgG1. After washing, cells were analyzed by flowcytometry. The mean fluorescence of stained cells is shown in the upper-right corner of the individual dot plots. (b) RT-PCR for TLR3 and actin mRNAwas performedwith monocytes and iDC as described in Experimental Procedures. TLR3was not detected in human monocytes.

    5

    DTD 5 ARTICLE IN PRESS

    505

    506

    507

    508

    509

    510

    511

    512

    513

    514

    515

    516

    517

    518

    519

    520

    521

    522

    523

    524

    525

    526

    527

    528

    529

    530

    531

    532

    533

    534

    535

    536

    537

    538

    539

    540

    541

    542

    543

    544

    545

    546

    547

    548

    549

    550

    551

    552

    553

    554

    555

    556

    557

    558

    559

    560

    561

    562

    563

    564

    565

    566

    567

    568

    569

    570

    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

    COwas a significant induction of TNF-a productionwith the immunostimulatory siRNA 27, whereas nosignificant activity was seen with the control siRNA32.

    623

    624

    625

    626

    627

    628

    629

    630

    UNTLR3 is not required for responsiveness toeither double-stranded or single-strandedsiRNAs in adherent PBMCNotably, the host sensors that initially detect viral

    and bacterial antigens and trigger cytokine pro-duction has been investigated by several groups,

    YJMBI 57125—21/3/2005—17:11—SFORSTER—141115—XML – pp. 1–12/APP

    some of which have indicated the involvement ofToll-like receptors (TLRs). These receptors, whichare mammalian homologous of the Drosophila Toll,recognize specific structural motifs expressed bymicrobes.17–19 So far, ten TLRs have been describedin humans, and ligands have been defined for nineof them. TLR1, K2 and K6 are triggered bypeptidoglycan and other microbial products, TLR3by dsRNA, TLR4 by LPS, TLR5 by flagellin, TLR7and K8 by imidazoquinolines, and TRL9 byunmethylated CpG DNA motifs. Although TLR3is a receptor for dsRNA and cellular mRNA,20,21 it

    S

  • 6

    DTD 5 ARTICLE IN PRESS

    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

    does not contain dsRNA binding motifs. While thiswork is underway, a recent study has indicated theinvolvement of TLR3 in siRNA signaling andsuggested the involvement of dsRNA-bindingprotein that has yet to be identified.14 In contrastto macrophages and dendritic cells, however,human monocytes do not express TLR3 as assessedby flow cytometry and RT-PCR (Figure 3(a) and (b)).These results agree with those of a recent studyreporting on the expression of TLR3 by humanmonocytes.22 In addition, treatment of either adher-ent PBMC or purified monocytes with anti-TLR3monoclonal antibody, known to blocks TLR3signaling,23 did not inhibit the production of TNF-a by immunostimulatory siRNAs (data not shown).

    709

    710

    711

    712

    713

    714

    715

    716

    717

    718

    719

    720

    721

    722

    723

    724

    725

    726

    Sense and antisense siRNA strands are highlystimulatory when compared to their double-stranded siRNA counterparts

    The data described above indicate that a sub-group of double-stranded siRNAs can induce theproduction of inflammatory cytokines. During thisstudy, however, I have been surprised by thefinding that both single-stranded sense and anti-sense siRNAs can trigger the production of pro-inflammatory cytokines (e.g. TNF-a) andinterferons (IFN-a) in adherent PBMC (Figure 4 asa representative example). This is in contrast towhat have been reported using various cancer celllines.12,13 As for the double-stranded siRNAs, theinduction of TNF-a by single-stranded siRNAs wassequence dependent. In the case of siRNA 27, the

    UNCO

    RREC

    T

    After 18 hours transfection time, TNF-awas measured in thestranded and single-strand siRNAs. The test molecules (100 nelectroporation. To test the involvement of endosomes in siRNtwo hours with chloroquine prior to transfection with DOTAPsupernatants by ELISA. All results represent the mean of thr

    YJMBI 57125—21/3/2005—17:12—SFORSTER—141115—XML – pp. 1–12/A

    sense strand exhibited the strongest effects, whereasin the case of siRNA 32 the antisense strand exertedthe strongest effect on TNF-a production (Figure4(a)). Notably, the single-stranded siRNAs werehighly immunostimulatory compared to theirdouble-stranded siRNA counterparts (e.g. siRNA32). As shown DOTAP alone did not induce eitherTNF-a or IFN-a production, indicating that theeffects are related to the RNA sequences. Directaddition of either naked double-stranded siRNAsor single-stranded siRNAs did not affect cytokineproduction (data not shown). Moreover, severaldouble-stranded siRNAs did not induce response,whereas their corresponding single-stranded senseor antisense strands did (Figure 4(a), as anillustration). It is worth noting that the samesingle-stranded siRNA preparations were used toprepare the annealed siRNAs. Taken together, theseobservations would indicate that the reportedsiRNA effects in adherent PBMC are related to thesiRNA sequences and ague against any possiblecontamination with endotoxins.

    ROOF

    Cytokine induction by double-stranded siRNAs,sense or antisense siRNA strands requiresendosomal acidification

    Cationic liposome-delivered siRNAs areexpected to enter the cell via endocytosis. Becauseseveral Toll-like receptors, in particular TLR7,TLR8, TLR9, are localized in the endosomes, Ihypothesized that endocytically introduced RNAcould trigger the activation of these receptors,

    ED P

    Figure 4. Effects of double-stranded and single-stranded siR-NAs upon TNF-a production inadherent PBMC. (a) Cells weretransfected with either double-stranded siRNAs (d), sensesiRNA strands (s) or antisensesiRNA strands (a) for 18 hours.Subsequently, TNF-a wasmeasured in the supernatants byELISA. All test RNA moleculeswere used at 100 nM and com-plexed with DOTAP at 10 mg/ml.(b) Inhibition of cytokine pro-duction by chloroquine in dose-dependent manner. Cells werepre-incubated with chloroquinefor two hours prior transfectionwith siRNAs (100 nM) for 18hours. Subsequently, TNF-a wasmeasured in the supernatants byELISA. (c) The test RNAmolecules(100 nM) were delivered to thecells either via DOTAP or electro-poration as indicated on the figure.

    supernatants by ELISA. (d) Induction of IFN-a by double-M) were delivered to adherent PBMC via either DOTAP orA induction of interferons, cells were also pretreated for

    . After transfection (20 hours), IFN-awas measured in theee or more independent experiments.

    PPS

    727

    728

    729

    730

    731

    732

    733

    734

    735

    736

    737

    738

    739

    740

    741

    742

    743

    744

    745

    746

    747

    748

    749

    750

    751

    752

    753

    754

    755

    756

  • 7

    DTD 5 ARTICLE IN PRESS

    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

    848

    849

    850

    851

    852

    853

    854

    855

    leading MAPK kinases and NF-kB activation. Totest the involvement of endosomes in siRNAsignaling, cells were treated with an inhibitor oflysosomal acidification, chloroquine for two hoursprior to transfection with either double-strandedor single-stranded siRNAs. TNF-a secretion byadherent PBMCwere inhibited in a dose-dependentmanner by chloroquine (Figure 4(b)), indicating thatthe acidification of the endosomes is importantfor cellular response to either double-stranded orsingle-stranded siRNAs. A similar inhibition wasobtained with Bafilomycin, an additional inhibitorof endosomal acidification (data not shown).

    Cationic liposomes such as DOTAP both protectthe siRNA and mediate internalization via endo-cytosis. After entry, the siRNA must escape theendolysosomes and enter the RNAi pathway.The requirement for acidification of endosomes inadherent PBMC was further examined via electro-poration of siRNAs into adherent PBMC. In contrastto DOTAP, the electroporation method open uppores in the cell, so siRNA molecules can enter thecell directly into the cytoplasm. As shown in Figure4(c) and (d), no TNF-a and IFN-a induction wasobtained when either double-stranded or single-stranded siRNAs were delivered via electropora-tion. Therefore, endosome compartments arerequired for both double-stranded and single-stranded siRNA intracellular signaling andsubsequent cytokines and interferon productionby adherent PBMC. Notably, pretreatment of PBMCwith chloroquine prior to transfection with siRNAabrogated IFN-a production (Figure 4(d)), indicat-ing for the first time the involvement of endosomesin siRNA induction of interferons.

    UNCO

    RREC

    T

    YJMBI 57125—21/3/2005—17:12—SFORSTER—141115—XML – pp. 1–12/APP

    PRO

    OF

    Endosomal siRNA signaling supports theinvolvement of the Toll-like receptor 8

    Toll-like receptors (TLRs) represent a class ofpattern recognition receptors (PPRs) that detectmicrobes or their components.19 TLRs are pre-dominantly expressed on the cell surface; however,a subset (TLR7, TLR8, TLR9, and in some cases,TLR3) is retained in intracellular compartments.Notably, TLR9, TLR7 and TLR8 require endosomalacidification for signaling.19 Because the inductionof inflammatory cytokines by siRNAs is chloro-quine-sensitive, the data would support the invol-vement of these receptors in siRNA intracellularsignaling. The recent identification of single-stranded RNAs as a ligand for human TLR824,25

    would support our initial observation15 and thepresent elaborated results suggesting the involve-ment of endosomal TLRs in single-stranded siRNAsignaling. This novel observation invites the ques-tion of what serves as the endosomal receptors fordouble-stranded siRNAs. In acidic microenviron-ment such as the endosomes, double-strandedsiRNA might dissociate and generate single-stranded siRNAs for intracellular signaling. Insupport of this idea is the observation that certaindouble-stranded siRNAs are very less effective ininducing inflammatory cytokine production thantheir corresponding single-stranded sense or anti-sense strand counterparts (Figure 4). Depending ofthe relative internal stability, certain siRNAduplexes may exist in equilibrium with their singlestrand forms, which can activate TRL8 and/or otherendosomal TLRs (Figure 5). Alternatively, double-stranded siRNAs may signal through endosomalTLR3, known to bind double-stranded RNAs.21

    ED

    Figure 5. A schematic diagramillustrating the intracellular siRNAsignaling. A complex of eitherdouble-strand or single-strand siR-NAs or cationic lipids bind to theplasma membrane electrostaticallyand are internalized via endocyto-sis. Within the endosomes RNAmolecules can bind to endosomalToll-like receptors, in particularTLR8 and 7, which can activatethe transcription factor NF-kB,leading to inflammatory cytokineproduction. TLR7, 8 and 9 useMyD88 as primary adaptor toactivate interferon production viaundefined signaling intermediates.TLR3 uses the adaptor proteinTRIF to activate interferon regulat-ory factor-3 (IRF-3). The activationof Toll receptors may activate PKRdirectly.19,11

    S

    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

  • T

    ROOF

    Figure 6. Immunostimulatory siRNAs activate theNF-kB transcription factor. A NF-kB-dependent luciferasereporter gene was co-transfected in human adherentPBMC with a control plasmid containing the Renillaluciferase gene under the control of the CMV promoterfor 24 hours. Subsequently; cells were treated with eithersiRNA 27 or 32. The sense (S) and the antisense strand (A)of siRNA 27 and 32, respectively, were also tested.Eighteen hours later, luciferase activity was determinedusing Dual-Luciferase Reporter Assay System (Promega).The data show the relative firefly luciferase activitynormalized to that of Renilla luciferase. Results areshown as means of three independent experiments GSD.

    8

    DTD 5 ARTICLE IN PRESS

    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

    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

    ORR

    EC

    Given that single-stranded siRNA are highlyimmunostimulatory compared to their correspond-ing double-stranded siRNAs, during siRNAannealing it is important to make sure that allsiRNA molecules exist as siRNA duplexes.Although all double-stranded siRNA preparationsused in this study were checked by PAGE for thepresence of unannealed single-stranded siRNAs,some of the double-stranded siRNA effects couldoriginate from tiny traces of free single-strandedsiRNAs. Whatever the nature of double-strandedsiRNA receptors; however, the present data indicatefor the first time that induction of inflammatorycytokines and interferon-a by either double-stranded or single-stranded siRNAs requires endo-somal acidification in fleshly isolated PBMC. There-fore, the development of carriers that deliver siRNAdirectly into the cytoplasm should overcome theproblem of inflammatory cytokine induction bychemically synthesized siRNA, a major obstacle forsystemic administration of siRNA in patients.Similarly to siRNA liposome complexes, nakedsiRNAs delivered via other techniques in vitro andin vivo are more likely to be taken up by cells viaendocytosis.

    The finding that several siRNA sense strands(mRNA sequences) can be highly immunostimula-tory indicates that endosomal Toll-like receptorscan recognize self-RNA. Thus, how can the immunesystem distinguish between self and non-self RNAs(e.g. viral RNAs,) and how to mount and amplify animmune response against invading RNA in aspecific manner? The present data argue that theimmune system uses the endosomes or lysosomalcompartments as molecular recognition signaturefor RNA in general (whether self or non-self).In contrast to viral RNAs, self-RNAs exist in thecytoplasm and therefore cannot enter the endoso-mal compartments; hence in normal situation theimmune system cannot be activated by self-RNAs.Similar to single-stranded siRNAs, liposomaldelivery of short single-stranded RNA (21 nt)derived from HIV-1 TAT mRNA or human TNF-amRNA also induced TNF-a and IFN-a productionin adherent PBMC (data not shown). Thus, endo-somal location of RNA seems to function as a“danger signal” for TLR recognition, and sub-sequent activation of innate immunity genes. Theobservation that certain sequences are more stimu-latory than others would indicate that endosomalTLRs, in particular TLR8, have preferences for aparticular nucleotides and/or structures that needfurther investigation.

    999

    1000

    1001

    1002

    1003

    1004

    1005

    1006

    1007

    1008

    UNCsiRNA treatment can activate the NF-kB

    promoter

    The core TLR signaling pathway uses myeloiddifferentiation factor 88 (MyD88) as the primaryadaptor protein and results in NF-kB activation,cytokine production and expression of costimula-tory molecules, such as the class II major histocom-patibility complex. To address the involvement of

    YJMBI 57125—21/3/2005—17:12—SFORSTER—141115—XML – pp. 1–12/A

    ED PNF-kB, promoter activation studies using fireflyluciferase as a reporter gene was performed.

    Adherent PBMC were transiently transfected withNF-kB-luciferase gene along with plasmid DNAwith the Renilla luciferase gene for 24 hours.Subsequently, the cells were either mock transfectedor transfected with a stimulatory siRNA, and 18hours later, cells were harvested, and equalamounts of cell extracts were subjected to luciferaseassay (Figure 6). The data showed that double-stranded siRNA 27 could induce around 8-foldincrease in NF-kB activity, whereas 22-fold increasewere obtained with the single-stranded siRNA(S27), again identifying single-stranded siRNAs asan important inducer of NF-kB activation.

    Single-stranded and double-stranded siRNAsstimulate monocyte-derived immature dendriticcells to produce cytokines and up-regulatecostimulatory molecules

    If we view the induction of inflammatorycytokines as a beneficial mediator in cancer andinfectious diseases, then stimulatory siRNAs couldemerge as a viable agent to knockdown specificgenes and activate innate and acquired immunityagainst tumor cells. Notably, stimulation of imma-ture dendritic cells (iDC) by microbial productsinduces the production of inflammatory cytokines

    PPS

  • T

    Figure 7. Immature DC activatedwith siRNA upregulated theexpression of CD83 and secretedIL-12. Immature dendritic cellswere culture for 48 hours in thepresence of either siRNA 27 or 32(100 nM) and then stained withFITC-labeled monoclonal anti-bodies specific for CD83 (a). Cellswere also stained with the corre-sponding isotype control. Themean fluorescence of stained cellsis shown in the upper-right cornerof the individual dot plots. Resultsare shown as means of threeindependent experiments GSD.

    (b) After 48 hours stimulation with siRNAs, IL-12 was measured in the supernatants using ELISA. Results are shownas means of three independent experiments GSD.

    9

    DTD 5 ARTICLE IN PRESS

    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

    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

    UNCO

    RREC

    such as TNF-a and IL-12, which can inducedifferentiation of T cells into a T helper cell type1.26 In addition, these stimuli are known to up-regulate certain costimulatory molecules such asCD40, CD80, CD83 and CD86. This processunderlies DC maturation, and it strongly poten-tiates the ability of DC to activate naive T cells.26,27

    Therefore, the effects of siRNAs upon the matu-ration of DC were investigated. To generate iDCs,freshly isolated monocytes were cultured for sixdays in the presence of GM-CSF (50 ng/ml) andIL-4 (100 ng/ml). Subsequently, they were bothtransfected with siRNA 27 or siRNA 32 and thenanalyzed by flow cytometry (Figure 7(a)). Addingstimulatory siRNA 27 to the culture mediumupregulated the expression of CD83, whereas nosignificant effects were obtained with the controlsiRNA 32. Treatment with siRNA 27 also upregu-lated the expression of CD86 (data not shown).Furthermore, siRNA 27-treatment resulted in IL-12production (Figure 7(b)). Similar results wereobtained with single-stranded siRNA 27. Theseresults suggest that stimulatory siRNAs canenhance immunity by inducing cytokine pro-duction and DC maturation. Thus, the use ofstimulatory double-stranded or single-strandedsiRNAs as a new generation of adjuvants mayfacilitate the design of effective vaccines.

    The ability to enhance or augment the innateimmune response clearly represents a potentialpowerful way to prevent or treat infections as wellas a way to develop cancer vaccines. Recently, it hasbeen discovered that unmethylated CpG dinucleo-tides in particular base contexts are recognized bythe immune system as danger signals.28 In animalmodels, deoxyoligonucleotides with stimulatoryCpG motifs have been shown to be of therapeuticvalue as adjuvants for conventional and therapeuticvaccines against infectious diseases and tumors.29

    As shown in the present study, the stimulatoryRNAs, in particular single-stranded siRNAs, acti-vated human monocytes and dendritic cells toproduce TNF-a and IL-12, two key cytokines inimmune regulation. In addition, the induction of

    YJMBI 57125—21/3/2005—17:12—SFORSTER—141115—XML – pp. 1–12/APP

    D PR

    OOF

    other cytokines such as IL-6 and interferons is anessential response for the clearance of viral infec-tions.30 Natural killer cells are activated by cyto-kines and chemokines, including type I IFN andIL-12, which are secreted by dendritic cells.31

    Therefore, treatment of cancer and infectious dis-eases might benefit from the induction of cytokineand dendritic cell activation by immunostimulatorysiRNAs. TLR activators are already used asadjutants to boost immune responses in vaccines.However, the success of a given vaccine maydepend on appropriate activation of TLR. There-fore, single-stranded sense RNA-27 should beexplored as an adjuvant for vaccination andimmunotherapy. On the other hand, the develop-ment of TLR antagonist holds promise as a newclass of anti-inflammatory agents. Although furtherstudy is required, some of the non-stimulatoryRNAs may function as antagonists.

    ESelection of siRNA against human TNF-a

    One of the key cytokines crucial to manybiological processes is TNF-a. Low levels of TNF-a produced by monocytes and tissue macrophagesunder physiological conditions are expected to beinvolved in maintaining cellular and tissue homeo-static. The production of TNF-a increases inresponse to microbial infection and tissue injury.Because of its important role in the pathogenesis ofa variety of inflammatory and immune diseases,TNF-a has been identified as a key target forpharmacological modulation.32 Therefore, thedevelopment of agents that specifically inhibitTNF-a may provide clinician with a valuablealternative to traditional disease-modifying anti-inflammatory drugs. In this respect, we have shownthat siRNA-targeting mouse TNF-a can delay theonset of LPS-induced sepsis.33 Out of 12 recentlytested siRNA targeting mouse TNF-a, siRNA 29(see Table 1), exhibited the greatest protective effect.To select, siRNAs against human TNF-a, several

    siRNAs were designed (Table 2) and tested for theirinhibitory effect on siRNA 27-induced TNF-a

    S

  • Table 2. Sequences of siRNAs targeting human TNF-a

    SiRNAs Sequence (5 0–3 0) (Sense strand)

    H1 GAGUGACAAGCCUGUAGCCTTH2 CUCAGCGCUGAGAUCAAUCTTH3 CUAGUGGUGCCAGCCGAUGTTH4 GCGUGGAGCUGAGAGAUAATT

    10

    DTD 5 ARTICLE IN PRESS

    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

    1164

    1165

    1166

    1167

    1168

    1169

    1170

    1171

    1172

    1173

    1174

    1175

    1176

    1177

    1178

    1179

    1180

    1181

    1182

    1183

    1184

    1185

    1186

    1187

    1188

    1189

    1190

    1191

    1192

    1193

    1194

    1195

    1196

    1197

    1198

    1199

    1200

    1201

    1202

    1203

    1204

    1205

    1206

    1207

    1208

    1209

    1210

    1211

    1212

    1213

    1214

    1215

    1216

    1217

    1218

    1219

    1220

    1221

    1222

    1223

    1224

    1225

    1226

    1227

    1228

    1229

    1230

    1231

    production in adherent PBMC (Figure 8(a)). SomesiRNAs were effective in inhibiting TNF-a pro-duction, whereas others had no effects, thussuggesting the specificity of the inhibitory effects.When tested alone, the siRNA inhibitors signifi-cantly reduced LPS-induced TNF-a production byadherent human PBMC (Figure 8(b)). The utility ofthis selection assay may be extended to otherinflammatory cytokines and type I interferon toselect effective siRNAs in human blood cells. It isworth noting that the immunostimulatory effects ofsiRNAs are more pronounced in human PBMC ascompared to other human cell lines. This maydepend on the expression level of Toll-like recep-tors, which is under investigation.

    Collectively, the present study underscores forthe first time the importance of the endosomecompartments in siRNA intracellular signaling inadherent PBMC. The results can easily be explainedby the interaction of RNA molecules with endo-somal Toll-like receptors and subsequent inductionof inflammatory and interferon responses (seeFigure 5). The finding that endosomal localizationof self RNA can trigger Toll-like receptors signalingin adherent PBMC is intriguing because it indicates

    UNCO

    RREC

    T

    Figure 8. Selection of siRNAs against human TNF-a.(a) Inhibitory effects of TNF-a siRNAs on TNF-aproduction by the immunostimulatory siRNA 27. Adher-ent PBMC cells were co transfected with siRNA 27 andvarious siRNA targeting the human TNF-a (H1-5).Eighteen hours later supernatants were analyzed forTNF-a contents by ELISA. Results are shown as means ofthree independent experiments GSD. (b) Blockage ofLPS-induced TNF-a expression by the inhibitory siRNAs.The cells were transfected for 18 hours with either siRNA32, H2, H3 or H4 (50 nM). Subsequently, cells werestimulated with LPS (20 ng/ml) for eight hours and thensupernatants were analyzed for TNF-a contents byELISA. Results are shown as means of three independentexperiments GSD.

    YJMBI 57125—21/3/2005—17:12—SFORSTER—141115—XML – pp. 1–12/A

    that endosomal self RNA can display a molecularpattern capable for triggering innate immunityactivation.

    1232

    1233

    1234

    1235

    1236

    1237

    1238

    1239

    1240

    1241

    1242

    1243

    1244

    1245

    1246

    1247

    1248

    1249

    1250

    1251

    1252

    1253

    1254

    1255

    1256

    1257

    1258

    1259

    1260

    ED P

    ROOF

    Experimental Procedures

    siRNAs

    The siRNAs used in this study correspond to variousmolecules targeting genes related to other ongoingprojects. All siRNAs were chemically synthesized byEurogentec (Seraing, Belgium), dissolved in water andannealed in the transfection buffer (20 mM Hepes,150 mM NaCl, pH 7.4) at 20 mM. Analysis of LPS/endo-toxin levels in siRNA stocks was found to be less than0.01 EU/ml (PyrogentR, CAMBREX). The sequencesof the sense strands and the target genes are shown inTable 1.

    Preparation of human cells

    Human mononuclear cells were prepared from buffycoats by density gradient centrifugation (Lymphoprep,Nycomed Pharm, Oslo), washed, and then resuspendedin RPMI 1640 containing 10% heated inactivated fetal calfserum (FCS). Enriched monocyte populations wereisolated by plastic adherence. After three hours incu-bation at 37 8C, non-adherent cells were removed byrepeated gentle washing with warm medium. More than75% of the obtained cells by this technique are CD14Ccells. In addition, CD14C cell population was preparedfrom PBMC by positive selection with immunomagneticbeads coated with anti-CD14 monoclonal antibodies(Dynal, Oslo, Norway).

    Culture of dendritic cells

    Enriched population of CD14 positive monocytes wereisolated from human PBMC as described above andcultured for five days in RPMI 1640 supplemented with10% FCS and antibiotics, in the presence of 50 ng/ml GM-CSF and 100 ng/ml IL-4 to obtain monocyte- derivedimmature dendritic cells (iDC). After five days in culture,the cells were either mock transfected or transfected witheither siRNA 27 or 32 for 48 hours, and then cells werestained with FITC-conjugated monoclonal antibodiesspecific for CD83 (Immunotech). Cells were also stainedwith FITC-conjugated normal mouse IgG1, an isotypecontrol.

    FACS staining and analysis

    Briefly, 2!105 cells were incubated in the recom-mended FITC-labeled antibody dilutions for 30 minutesat 4 8C in staining buffer (PBS containing 0.5% FCS and0.1% azide). Cells were also stained with the correspond-ing Ab isotype controls. Subsequently, the cells werewashed twice with the staining buffer and then analyzedon a FACScan using Cell-Quest software (BD Bio-sciences). Cells were gated in forward/side scatter andthe quadrants were set on the relative isotype controls.

    Transfection and ELISA

    Cells were seeded at 5!104/well/200 ml in 96-wellplates. After an overnight incubation with siRNAs/lipo-some complexes, culture supernatants were collected and

    PPS

  • 11

    DTD 5 ARTICLE IN PRESS

    1261

    1262

    1263

    1264

    1265

    1266

    1267

    1268

    1269

    1270

    1271

    1272

    1273

    1274

    1275

    1276

    1277

    1278

    1279

    1280

    1281

    1282

    1283

    1284

    1285

    1286

    1287

    1288

    1289

    1290

    1291

    1292

    1293

    1294

    1295

    1296

    1297

    1298

    1299

    1300

    1301

    1302

    1303

    1304

    1305

    1306

    1307

    1308

    1309

    1310

    1311

    1312

    1313

    1314

    1315

    1316

    1317

    1318

    1319

    1320

    1321

    1322

    1323

    1324

    1325

    1326

    1327

    1328

    1329

    1330

    1331

    1332

    1333

    1334

    1335

    1336

    1337

    1338

    1339

    1340

    cytokine contents were assessed by ELISA. Cells weretransfected in RPMI 1640 medium (PAA LaboratoriesGmbH) supplemented with 10% heat-inactivated FCS,2 mM L-glutamine, 100 mg/ml penicillin and 100 mg/mlstreptomycin using DOTAP (10 mg/ml) as described.33

    Each assay was carried out in triplicate. When nakedsiRNAs were used, cells were incubated in X-VIVO 15medium without adding FCS (CAMBREX). In the caseof monocytes, 2!104 cells were used per well. Theinactivation of PKR was carried out as follows: Cells wereincubated with 2-aminopurine (10 mM), a specific inhibi-tor of PKR, for one hour prior to addition of the siRNA/liposome complexes. After an overnight transfectionwith the tested molecules, the levels of TNF-a, IL-6 andIL-12p70 in the culture supernatants were measured byavailable ELISA kits according to the manufacture’sinstructions (R&D Pharmingen). Samples were run intriplicate. Human IFN-a was measured using ELISA kit(PBL Biomedical Laboratories).

    1341

    1342

    1343

    1344

    1345

    1346

    1347

    1348

    1349

    1350

    1351

    1352

    1353

    1354

    Transfection and reporter assays

    An enriched monocyte population (2!106 cells) wereseeded onto a 24-well plate and transfected the followingday with firefly luciferase gene (1 mg) under the control ofthe NF-kB-promoter and pRL-TK DNA (1 mg, Promega)encoding the Renilla luciferase gene. After 24 hours, thecells were transfected with siRNAs and then cells wereharvested 18 hours later. Luciferase activities weremeasured using the Dual-Luciferase Reporter AssaySystem according to the manufacture’s instructions(Promega).

    T

    1355

    1356

    1357

    1358

    1359

    1360

    1361

    1362

    1363

    1364

    1365

    RT-PCR analysis

    Total RNA was extracted from freshly isolated mono-cytes and monocytes-derived iDC using total RNAisolation kit (Pharmacia Biotech). DNAse I-treated RNA(10 mg) was reverse-transcribed with NotI-d(T)18 primerusing the first-strand cDNA synthesis Kit (PharmaciaBiotech) according to the manufacturer’s instructions.Polymerase chain reaction (PCR) was performed on 3 mlof cDNA in 50 ml of final volume. The TLR3 primers are:

    1366

    1367

    YJM

    1368

    C5 0-ATTGGGTCTGGGAACATTTCTCTTC-3 0 (forwardprimer)

    1369

    1370

    1371

    1372

    RE

    5 0-GTGAGATTTAAACATTCCTCTTCGC-3 0 (reverseprimer)

    As a control, b actin mRNA was also amplified usingthe following primers:

    1373

    1374

    1375

    R5 0-ATCTGGCACCACACCTTCTACAATGAGCTGCG-3 0 (forward primer)

    1376

    1377

    1378

    1379

    1380

    NCO50-CGTCATACTCCTGCTTGGTGATCCACATCTGC-30

    After 35 cycles of amplification (one minute at 92 8C;one minute at 56 8C; one minute at 72 8C, samples wereanalyzed by electrophoresis on 1.5% agarose gel andvisualized by ethidium bromide staining.

    U 13811382

    1383

    1384

    1385

    1386

    Uncited Reference

    34.

    BI 57125—21/3/2005—17:12—SFORSTER—141115—XML – pp. 1–12/APP

    Acknowledgements

    I thank Drs Per Ole Iversen and Anne Dybwadfor critical reading of the manuscript, and LiseForfang for excellent technical assistance with flowcytometry. This work was supported by theNorwegian Cancer Society.

    ED P

    ROOF

    References

    1. Fire, A., Xu, S., Montgomery, M. K., Kostas, S. A.,Driver, S. E. & Mello, C. C. (1998). Potent and specificgenetic interference by double-stranded RNA inCaenorhabditis elegans. Nature, 391, 806–811.

    2. Hannon, G. J. (2002). RNA interference. Nature, 418,244–251.

    3. Hamilton, A. J. & Baulcombe, D. C. (1999). A speciesof small antisense RNA in posttranscriptional genesilencing in plants. Science, 286, 950–952.

    4. Sen, G. C. (2001). Viruses and interferons. Annu. Rev.Microbiol. 55, 255–281.

    5. Saunders, L. R. & Barber, G. N. (2003). The dsRNAbinding protein family: critical roles, diverse cellularfunctions. FASEB J. 17, 961–983.

    6. Elbashir, S. M., Harborth, M. J., Ledecknel, W., Yalcin,A., Weber, K. & Tuchl, T. (2001). Duplexes of 21-nucleotide RNAs mediate RNA interference incultured mammalian cells. Nature, 411, 494–498.

    7. Sioud, M. (2004). Therapeutic siRNAs. Trends Pharma-col. Sci. 25, 22–24.

    8. Hannon, G. J. & Rossi, J. J. (2004). Unlocking thepotential of the human genome with RNA inter-ference. Nature, 431, 371–378.

    9. Kawasaki, H. & Taira, K. (2004). Induction of DNAmethylation and gene silencing by short interferingRNAs in human cells. Nature, 431, 211–217.

    10. Semizarov, D. et al. (2003). Specificity of shortinterfering RNA determined through gene expressionsignatures. Proc. Natl Acad. Sci. USA, 100, 6347–6352.

    11. Persengiev, S. P., Zhu, X. & Green, M. R. (2004).Nonspecific, concentration-dependent stimulationand repression of mammalian gene expression bysmall interfering RNAs (siRNAs). RNA, 10, 12–18.

    12. Sledz, C. A., Holko, M., de Veer, M. J., Silverman, R. H.& Williams, B. R. G. (2003). Activation of theinterferon system by short-interfering RNAs. NatureCell. Biol. 5, 834–8340.

    13. Bridge, A. J., Pebernard, S., Ducraux, A., Nicoulaz,A.-L. & Iggio, R. (2003). Induction of an interferonresponse by RNAi vectors in mammalian cells.NatureGen. 34, 263–264.

    14. Kariko, K., Bhuyan, P., Capodici, J. & Weissman, D.(2004). Small interfering RNAs mediate sequence-independent gene suppression and induce immuneactivation by signaling through Toll-like receptor 3.J. Immunol. 172, 6545–6549.

    15. Sioud, M. & Sørensen, D. R. (2003). Cationic liposome-mediated delivery of siRNAs in adult mice. Biochem.Biophys. Res. Commun. 312, 1220–1225.

    16. Kim, D-H. , Longo, M., Han, Y., Lundberg, P., Cantin,E. & Rossi, J. J. (2004). Interferon induction by siRNAsand ssRNAs synthesized by phage polymerase.Nature Biotechnol. 22, 312–325.

    17. Medzhitov, R., Preston-Hurlburt, P. & Janeway, C. A.,Jr (1997). A human homologue of drosophila Tollprotein signals activation of adaptive immunity.Nature, 388, 394–397.

    S

  • 12

    DTD 5 ARTICLE IN PRESS

    1387

    1388

    1389

    1390

    1391

    1392

    1393

    1394

    1395

    1396

    1397

    1398

    1399

    1400

    1401

    1402

    1403

    1404

    1405

    1406

    1407

    1408

    1409

    1410

    1411

    1412

    1413

    1414

    1415

    1416

    1417

    1418

    1419

    1420

    1421

    1422

    1423

    1424

    1425

    1426

    1427

    1428

    1429

    1430

    1431

    1432

    1433

    1434

    1435

    1436

    1437

    1438

    1439

    1440

    1441

    1442

    1443

    1444

    1445

    1446

    1447

    1448

    1449

    1450

    1451

    1452

    1453

    1454

    1455

    1456

    1457

    1458

    1459

    1460

    1461

    1462

    1463

    1464

    1465

    1466

    1467

    1468

    1469

    1470

    1471

    1472

    1473

    1474

    1475

    1476

    1477

    18. Aderem, A. & Ulevitch, R. J. (2000). Toll-like receptorsin the induction of the innate immune response.Nature, 406, 782–787.

    19. Armant, M. A. & Fenton, M. J. (2002). Toll-likereceptors: a family of pattern recognition receptorsin mammals. Gen. Biol. 3, 3011.

    20. Alexopoulou, L., Holt, A. C., Medzhitov, R. & Flavell,R. A. (2001). Recognition of double-stranded RNAand activation of NF- kappa B by Toll-like receptor 3.Nature, 413, 732–738.

    21. Kariko, K., Ni, H., Capodici, J., Lamphier, M. &Weissman, D. (2004). mRNA is an endogenous ligandfor Toll-like receptor 3. J. Biol. Chem. 279, 12542–12550.

    22. Matsumoto, M., Funami, K., Tanabe, M., Oshiumi, H.,Shingai, M., Seto, Y. et al. (2003). Subcellular localiz-ation of Toll-like receptor 3 in human dendritic cells.J. Immunol. 171, 3154–3162.

    23. Matsumoto, M., Kikkawa, S., Kohase, M., Miyake, K.& Seya, T. (2002). Establishment of a monoclonalantibody against human Toll-like receptor 3 blocksdouble stranded RNA-mediated signaling. Biochem.Biophys. Res. Commun. 293, 1364–1369.

    24. Heil, F., Hemmi, H., Hochrein, H., Ampenberger, F.,Kirschning, C. & Akira, S. (2004). Species-specificrecognition of single-stranded RNA via toll-likereceptor 7 and 8. Science, 303, 1526–1529.

    25. Diebold, S. S., Kaisho, T., Hemmi, H., Akira, S. &Sousa, C. R. (2004). Innate antiviral responses bymeans of TLR7-mediated recognition of single-stranded RNA. Science, 303, 1529–1531.

    26. Langenkamp, A., Messi, M., Lanzavecchia, A. &

    UNCO

    RREC

    T

    YJMBI 57125—21/3/2005—17:12—SFORSTER—141115—XML – pp. 1–12/A

    OF

    Sallusto, F. (2000). Kinetics of dendritic cell activation:impact on priming of TH1, TH2 and nonpolarized Tcells. Nature Immunol. 1, 311–316.

    27. Lutz, M. B. & Schuler, G. (2002). Immature, semi-mature and fully mature dendritic cells: which signalsinduce tolerance or immunity? Trends Immunol. 23,445–449.

    28. Krieg, A. M. (1996). Lymphocyte activation by CpGdinucleotide motifs in prokaryotic DNA. TrendsMicrobiol. 4, 73–76.

    29. Krieg, A. M., Love-Homan, L., Yi, A. K. & Harty, J. T.(1998). CpG DNA induces sustained IL-12 expressionin vivo and resistance to Listeria monocytogeneschallenge. J. Immunol. 161, 2428–2434.

    30. Netea, M. G., van der Graaf, C., Van der Meer, J. W. M.& Kullberg, B. J. (2004). Toll-like receptors and thehost defense against microbial pathogens: bringingspecificity to the innate-immune system. J. Leukoc.Biol. 75, 749–755.

    31. Raulet, D. H. (2004). Interplay of natural killer cellsand their receptors with the adaptive immuneresponse. Nature Immunol. 5, 996–1002.

    32. Tracey, K. J. & Cerami, A. (1994). Tumour necrosisfactor: a pleiotropic cytokine and therapeutic targets.Ann. Rev. Med. 45, 491–503.

    33. Sørensen, R. & Leirdal, M. (2003). Sioud M Genesilencing by systemic delivery of synthetic siRNAs inadult mice. J. Mol. Biol. 327, 761–766.

    34. Sioud, M. (1994). Interaction between tumor necrosisfactor a ribozyme and cellular proteins. J. Mol. Biol.242, 619–629.

    1478

    1479

    1480

    O

    Edited by J. Karn

    R 14811482

    1483

    (Received 20 December 2004; received in revised form 3 March 2005; accepted 4 March 2005)

    ED P

    PPS

    1484

    1485

    1486

    1487

    1488

    1489

    1490

    1491

    1492

    1493

    1494

    1495

    1496

    1497

    1498

    1499

    1500

    1501

    1502

    1503

    1504

    1505

    1506

    1507

    1508

    1509

    1510

    1511

    1512

    Induction of Inflammatory Cytokines by Double-stranded and Single-stranded siRNAs is Sequence-dependent and Requires Endosomal LocalizationIntroductionResults and DiscussionThe non-specific effects of siRNAs are sequence-dependentIntracellular siRNA delivery is required for cytokine inductionsiRNAs activate TNF-alpha production in purified human monocytesTLR3 is not required for responsiveness to either double-stranded or single-stranded siRNAs in adherent PBMCSense and antisense siRNA strands are highly stimulatory when compared to their double-stranded siRNA counterpartsCytokine induction by double-stranded siRNAs, sense or antisense siRNA strands requires endosomal acidificationEndosomal siRNA signaling supports the involvement of the Toll-like receptor 8siRNA treatment can activate the NF-kappaB promoterSingle-stranded and double-stranded siRNAs stimulate monocyte-derived immature dendritic cells to produce cytokines and up-regulate costimulatory moleculesSelection of siRNA against human TNF-alpha

    Experimental ProceduressiRNAsPreparation of human cellsCulture of dendritic cellsFACS staining and analysisTransfection and ELISATransfection and reporter assaysRT-PCR analysis

    Uncited ReferenceAcknowledgementsReferences