9
1 3 Transfection of BmCPV genomic dsRNA in silkmoth-derived Bm5 4 cells: Stability and interactions with the core RNAi machinery 5 6 7 Luc Swevers a,Q1 , Anna Kolliopoulou a , Zheng Li b , Maria Daskalaki c , Frederic Verret c , 8 Kriton Kalantidis c , Guy Smagghe d , Jingchen Sun b,9 a Insect Molecular Genetics and Biotechnology, Institute of Biosciences and Applications, National Centre for Scientific Research ‘‘Demokritos’’, P. Grigoriou & Neapoleos Str, Aghia 10 Paraskevi Attikis, 153 42 Athens, Greece 11 b Guangdong Engineering Research Center of Subtropical Sericulture and Mulberry Resources Protection and Safety, Guangdong Provincial Key Lab of Agro-animal Genomics and 12 Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, People’s Republic of China 13 c Department of Biology, University of Crete, Voutes University Campus, 700 13 Heraklion, Crete, Greece 14 d Department of Crop Protection, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium 15 16 17 19 article info 20 Article history: 21 Received 31 October 2013 22 Received in revised form 3 March 2014 23 Accepted 7 March 2014 24 Available online xxxx 25 Keywords: 26 Cytoplasmic polyhedrosis virus 27 BmCPV 28 dsRNA 29 RNAi 30 Bm5 cells 31 32 abstract 33 While several studies have been conducted to investigate the stability of dsRNA in the extracellular med- 34 ium (hemolymph, gut content, saliva), little is known regarding the persistence of dsRNA once it has been 35 introduced into the cell. Here, we investigate the stability of Bombyx mori cytoplasmic polyhedrosis virus 36 (BmCPV) genomic dsRNA fragments after transfection into Bombyx-derived Bm5 cells. Using RT-PCR as a 37 detection method, we found that dsRNA could persist for long periods (up to 8 days) in the intracellular 38 environment. While the BmCPV genomic dsRNA was processed by the RNAi machinery, its presence had 39 no effects on other RNAi processes, such as the silencing of a luciferase reporter by dsLuc. We also found 40 that transfection of BmCPV genomic dsRNA could not establish a viral infection in the Bm5 cells, even 41 when co-transfections were carried out with dsRNAs targeting Dicer and Argonaute genes, suggesting 42 that the neutralization by RNAi does not play a role in the establishment of an in vitro culture system. 43 The mechanism of the dsRNA stability in Bm5 cells is discussed, as well as the implications for the estab- 44 lishment for an in vitro culture system for BmCPV. 45 Ó 2014 Published by Elsevier Ltd. 46 47 48 49 1. Introduction 50 Today, RNA interference (RNAi) is used as a tool to achieve po- 51 tent and specific gene knockdown in eukaryotes (Carthew and 52 Sontheimer, 2009; Jinek and Doudna, 2009; Moazed, 2009; Siomi 53 and Siomi, 2009; Huvenne and Smagghe, 2010). The trigger of RNAi 54 is dsRNA which, in the canonical pathway, is processed by Dicer 55 enzymes to small interfering RNAs (siRNAs), typically consisting 56 of a 21–22 nucleotide (nt) dsRNA duplex with 2–3 nt 3 0 -overhangs 57 (Myers et al., 2003). The siRNAs are then assembled in RNA- 58 induced silencing complexes (RISCs) that retain one strand of the 59 duplex. RISC complexes, with Argonaute proteins functioning as 60 catalytic components, subsequently use the retained single- 61 stranded siRNAs as ‘guides’ to search RNA populations for comple- 62 mentary sequences as targets for cleavage (Liu et al., 2004). 63 In insects, exogenous dsRNA is preferentially processed by Di- 64 cer-2 in conjunction with a dsRNA-binding protein, R2D2, and gen- 65 erated siRNAs are loaded into RISC complexes with Ago-2 as 66 catalytic unit (Tomari et al., 2007). Although Dicer-2 by itself 67 should be able to directly (non-specifically) degrade RNA targets 68 with dsRNA structure, it is believed that the generation of small 69 RNAs and their loading in catalytic Argonaute-containing com- 70 plexes acts as a sequence-specific mechanism to achieve efficient 71 silencing (van Rij et al., 2006). 72 Despite its identification as the trigger of RNAi as defense mech- 73 anism, surprisingly little is known regarding the stability of dsRNA 74 in insects. DsRNA stability has been considered as a contributing 75 factor to explain differences in RNAi efficiency among insects 76 (Terenius et al., 2011; Garbutt et al., 2013; Scott et al., in press). 77 A few studies have investigated dsRNA stability in the hemolymph, 78 the gut content, the saliva and the food source in which it was 79 exogenously added (Li et al., 2011; Garbutt et al., 2013; Liu et al., 80 2013; Allen and Walker, 2012; Christiaens et al., in press). It has 81 also been demonstrated that dsRNA can be efficiently taken up 82 by cells (Saleh et al., 2006; Swevers and Smagghe, 2012). However, http://dx.doi.org/10.1016/j.jinsphys.2014.03.002 0022-1910/Ó 2014 Published by Elsevier Ltd. Correspon Q2 ding authors. Tel.: +30 2106503681 (L. Swevers). E-mail addresses: [email protected] (L. Swevers), [email protected] (J. Sun). Journal of Insect Physiology xxx (2014) xxx–xxx Contents lists available at ScienceDirect Journal of Insect Physiology journal homepage: www.elsevier.com/locate/jinsphys IP 3196 No. of Pages 9, Model 5G 20 March 2014 Please cite this article in press as: Swevers, L., et al. Transfection of BmCPV genomic dsRNA in silkmoth-derived Bm5 cells: Stability and interactions with the core RNAi machinery. Journal of Insect Physiology (2014), http://dx.doi.org/10.1016/j.jinsphys.2014.03.002

Transfection of BmCPV genomic dsRNA in silkmoth-derived Bm5 cells: Stability and interactions with the core RNAi machinery

Embed Size (px)

Citation preview

1

3

4

5

6

7 Q1

8

91011121314

151617

1 9

2021222324

25262728293031

3 2

48

49

50

51

52

53

54

55

56

57

58

59

60

61

62

Q2

Journal of Insect Physiology xxx (2014) xxx–xxx

IP 3196 No. of Pages 9, Model 5G

20 March 2014

Contents lists available at ScienceDirect

Journal of Insect Physiology

journal homepage: www.elsevier .com/ locate/ j insphys

Transfection of BmCPV genomic dsRNA in silkmoth-derived Bm5cells: Stability and interactions with the core RNAi machinery

http://dx.doi.org/10.1016/j.jinsphys.2014.03.0020022-1910/� 2014 Published by Elsevier Ltd.

⇑ Corresponding authors. Tel.: +30 2106503681 (L. Swevers).E-mail addresses: [email protected] (L. Swevers), [email protected]

(J. Sun).

Please cite this article in press as: Swevers, L., et al. Transfection of BmCPV genomic dsRNA in silkmoth-derived Bm5 cells: Stability and interactiothe core RNAi machinery. Journal of Insect Physiology (2014), http://dx.doi.org/10.1016/j.jinsphys.2014.03.002

Luc Swevers a,⇑, Anna Kolliopoulou a, Zheng Li b, Maria Daskalaki c, Frederic Verret c,Kriton Kalantidis c, Guy Smagghe d, Jingchen Sun b,⇑a Insect Molecular Genetics and Biotechnology, Institute of Biosciences and Applications, National Centre for Scientific Research ‘‘Demokritos’’, P. Grigoriou & Neapoleos Str, AghiaParaskevi Attikis, 153 42 Athens, Greeceb Guangdong Engineering Research Center of Subtropical Sericulture and Mulberry Resources Protection and Safety, Guangdong Provincial Key Lab of Agro-animal Genomics andMolecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, People’s Republic of Chinac Department of Biology, University of Crete, Voutes University Campus, 700 13 Heraklion, Crete, Greeced Department of Crop Protection, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium

a r t i c l e i n f o a b s t r a c t

333435363738394041424344

Article history:Received 31 October 2013Received in revised form 3 March 2014Accepted 7 March 2014Available online xxxx

Keywords:Cytoplasmic polyhedrosis virusBmCPVdsRNARNAiBm5 cells

4546

While several studies have been conducted to investigate the stability of dsRNA in the extracellular med-ium (hemolymph, gut content, saliva), little is known regarding the persistence of dsRNA once it has beenintroduced into the cell. Here, we investigate the stability of Bombyx mori cytoplasmic polyhedrosis virus(BmCPV) genomic dsRNA fragments after transfection into Bombyx-derived Bm5 cells. Using RT-PCR as adetection method, we found that dsRNA could persist for long periods (up to 8 days) in the intracellularenvironment. While the BmCPV genomic dsRNA was processed by the RNAi machinery, its presence hadno effects on other RNAi processes, such as the silencing of a luciferase reporter by dsLuc. We also foundthat transfection of BmCPV genomic dsRNA could not establish a viral infection in the Bm5 cells, evenwhen co-transfections were carried out with dsRNAs targeting Dicer and Argonaute genes, suggestingthat the neutralization by RNAi does not play a role in the establishment of an in vitro culture system.The mechanism of the dsRNA stability in Bm5 cells is discussed, as well as the implications for the estab-lishment for an in vitro culture system for BmCPV.

� 2014 Published by Elsevier Ltd.

47

63

64

65

66

67

68

69

70

71

72

73

74

75

76

77

1. Introduction

Today, RNA interference (RNAi) is used as a tool to achieve po-tent and specific gene knockdown in eukaryotes (Carthew andSontheimer, 2009; Jinek and Doudna, 2009; Moazed, 2009; Siomiand Siomi, 2009; Huvenne and Smagghe, 2010). The trigger of RNAiis dsRNA which, in the canonical pathway, is processed by Dicerenzymes to small interfering RNAs (siRNAs), typically consistingof a 21–22 nucleotide (nt) dsRNA duplex with 2–3 nt 30-overhangs(Myers et al., 2003). The siRNAs are then assembled in RNA-induced silencing complexes (RISCs) that retain one strand of theduplex. RISC complexes, with Argonaute proteins functioning ascatalytic components, subsequently use the retained single-stranded siRNAs as ‘guides’ to search RNA populations for comple-mentary sequences as targets for cleavage (Liu et al., 2004).

78

79

80

81

82

In insects, exogenous dsRNA is preferentially processed by Di-cer-2 in conjunction with a dsRNA-binding protein, R2D2, and gen-erated siRNAs are loaded into RISC complexes with Ago-2 ascatalytic unit (Tomari et al., 2007). Although Dicer-2 by itselfshould be able to directly (non-specifically) degrade RNA targetswith dsRNA structure, it is believed that the generation of smallRNAs and their loading in catalytic Argonaute-containing com-plexes acts as a sequence-specific mechanism to achieve efficientsilencing (van Rij et al., 2006).

Despite its identification as the trigger of RNAi as defense mech-anism, surprisingly little is known regarding the stability of dsRNAin insects. DsRNA stability has been considered as a contributingfactor to explain differences in RNAi efficiency among insects(Terenius et al., 2011; Garbutt et al., 2013; Scott et al., in press).A few studies have investigated dsRNA stability in the hemolymph,the gut content, the saliva and the food source in which it wasexogenously added (Li et al., 2011; Garbutt et al., 2013; Liu et al.,2013; Allen and Walker, 2012; Christiaens et al., in press). It hasalso been demonstrated that dsRNA can be efficiently taken upby cells (Saleh et al., 2006; Swevers and Smagghe, 2012). However,

ns with

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

2 L. Swevers et al. / Journal of Insect Physiology xxx (2014) xxx–xxx

IP 3196 No. of Pages 9, Model 5G

20 March 2014

very little is known regarding the stability and fate of dsRNA insidethe cells after uptake, although the fate of siRNA/dsRNA inside thecell is gaining more interest recently [see, for instance, Wang andHuang (2013)].

In this article, the stability inside the cells was investigated fornaturally occurring dsRNA fragments, corresponding to the seg-mented genome of Bombyx mori cytoplasmic polyhedrosis virus(BmCPV; Reoviridae). Besides the study of dsRNA stability, it wasalso of interest to observe whether transfection of the BmCPV gen-ome into B. mori (silkmoth)-derived Bm5 cells would result in thegeneration of a functional virus, as is routinely observed aftertransfection of the DNA genome of B. mori nuclear polyhedrosisvirus (BmNPV) (O’Reilly et al., 1992). BmCPV is one of the majorpathogens of the silkworm and causes enormous damage to thesericulture industry (Wu et al., 2009; Mori and Metcalf, 2010).Infection in larvae is initiated by ingestion of polyhedra which dis-solve in the alkaline environment of the gut to release viral parti-cles (Payne and Mertens, 1983). Usually the infection is limitedto the epithelial lining of the gut although spreading to other tis-sues has also been observed. Unfortunately, no in vitro cell culturesystem for BmCPV has been developed (Hill et al., 1999), and theavailability of such cell-based system which would be of great ben-efit to investigate and understand the pathogenicity of the virusand to develop methods to combat the disease.

In this study, the stability of the segmented dsRNA of BmCPVgenome was investigated after transfection in Bm5 cells in theabsence or presence of dsRNA targeting RNAi core machinerygenes, such as Dicer-2 (Dcr2) and Argonaute-2 (Ago2). Transfectionof BmCPV dsRNA genome in silkworm-derived Bm5 cells was alsocarried out to see whether an in vitro culture system for this path-ogen could be established. Simultaneously, we investigatedwhether transfection of BmCPV genomic dsRNA or expression oftwo genes of the BmCPV genome, RNA-dependent RNA polymerase(RdRp), and non-structural 5 (NS5) could affect RNAi efficiency ina luciferase-based silencing assay. Our results provide the basisfor a discussion of the mechanism of dsRNA stability in Bm5 cells.

176

177

178

179

180

181

182

183

184

185

186

187

188

189

190

191

2. Materials and methods

2.1. Isolation and purification of BmCPV polyhedra

BmCPV(C) was cultivated and purified by established protocolsat South China Agricultural University (Tan et al., 2001). SilkmothB. mori 4th instar larvae were orally infected with BmCPV usingsmall pieces (2 cm � 2 cm) of mulberry leaves that were soakedin BmCPV solution (106 polyhedra/ml) for a few minutes. Larvaewere left to feed for 6 h on BmCPV-soaked mulberry leaves andwere provided normal leaves after that period. At 6–10 days afterinfections, the midgut of diseased silkworm was dissected afterremoval of head, gut content and silk glands, homogenized indistilled water and centrifuged at 10000g for 10 min. The freevirions remained in the supernatant; the polyhedra were containedin the pellet. Polyhedra were purified by sucrose density ultracen-trifugation as described (Hayashi and Bird, 1969).

192

193

194

195

196

197

198

199

200

201

202

2.2. Preparation of BmCPV dsRNA genomic fragments

A purified polyhedra solution (108 polyhedra/ml) was mixed at1:1 (v:v) with urea/SDS lysis buffer (Drevet et al., 1995) and incu-bated at 37 �C for 30 min. Then nucleic acid was extracted by phe-nol/chloroform and precipitated with 1/10 volume of 2.5 Mammonium acetate and 2.5 volumes of ethanol. To remove con-taminating DNA, BmCPV dsRNA was treated with RNase-freeDNase I (Thermo Scientific) for 30 min at 37 �C (2 U per lg of

Please cite this article in press as: Swevers, L., et al. Transfection of BmCPV genthe core RNAi machinery. Journal of Insect Physiology (2014), http://dx.doi.org

nucleic acid) in the presence of RiboLock RNase inhibitor (ThermoScientific) (4 U per lg of nucleic acid).

2.3. Expression and reporter plasmids

BmCPV ORFs were amplified by RT-PCR using BmCPV genomeinformation available at the Reoviridae website (http://www.reoviridae.org/dsRNA_virus_proteins/Cypovirus.htm). For the iso-lation of BmCPV genes, BmCPV genomic dsRNA was first denaturedat 60 �C for 30 min in 50% DMSO. To isolate the ORF of the NS5 gene,reverse transcription reactions were carried out using the primer50-AGTAAATCCCAGGCGTAAACCG-30 (42 �C for 1 h) and PCR reac-tions were performed using the primers 50-ACGGATCATCATG-GAAGCCTTC-30 and 50-ATCAGCGACACTATTCTGATAC-30 (35 cyclesof 94 �C for 45 s, 55 �C for 45 s and 72 �C for 1 min). To isolate theORF of the RdRp gene, reverse transcription reactions were carriedout using the primer 50-CGTCTGAAATGTTACCGAACAC-30 and PCRreactions were performed using the primers 50-CGTCTGAAATGTTACCGAACAC-30 and 50-ACTCATCTACACGTTATCTGCC-30 (40 cyclesof 94 �C for 1 min, 55 �C for 1 min and 72 �C for 3 min). PCR reac-tions were performed using a mixture of Taq polymerase (HyTest;1.5 U per reaction) and Pfu polymerase (Thermo Scientific; 1 Uper reaction). PCR fragments were cloned in pBluescript vectorand their identities were confirmed by sequencing.

To construct the expression vectors, the NS5 and RdRp ORFswere amplified with the primer pairs NS5F: 50-AATTGGATCCCAACATGGAAGCCTTCTTACTC-30/NS5R: 50-ATCAGCGACACTATTCTGATAC-30 and RdRpF: 50-AATTGGATCCCAACATGTTACCGAACACAAAACTAC-30/RdRpR: 50-ACTCATCTACACGTTATCTGCC-30, respectively(stop codons in reverse primers in bold). In both cases, the forwardprimer contains a BamHI-site (underlined), a Kozak translation ini-tiation sequence (italic) upstream of the start codon (bold), and theconfiguration allows in-frame cloning with the N-terminal Myc se-quence of the pEA-Myc expression vector (Douris et al., 2006), thusgenerating the expression plasmids pEA-Myc-NS5 and pEA-Myc-RdRp.

For the isolation of the ORF encoding the B2 RNAi inhibitor pro-tein from nodavirus, RNA isolated from Hi5 cells was used for re-verse transcription reaction using primer Noda-U7 (Li et al.,2007). The ORF of B2 was subsequently amplified by primers 50-AAACGATGCCAAGCAAACTCGCG-30 and 50-CACCTACAGTTTTGCGGGTG-30 and cloned in pBluescript. For in-frame cloning with theN-terminal Flag tag of the pEA-Flag vector the primers 50-AATTG-GATCCCAACATGCCAAGCAAACTCGCG-30 and 50-AATTGGATCCCTACAGTTTTGCGGGTGGGGGGTC-30 were used.

2.4. Bm5 tissue culture cells and transfections

Bm5 cells, derived from ovarian tissue (Kolliopoulou and Swe-vers, 2013), were maintained in IPL-41 medium (Gibco) supple-mented with 10% fetal bovine serum (FBS) and cultured at 27 �C.Transfection was carried out according to established protocols(Johnson et al., 1992) using Escort IV (Sigma) as transfection agent.

To investigate the stability of dsRNA, Bm5 cells were transfec-ted with 0.5 lg/ml dsRNA of BmCPV in the presence of 0.5 lg/mldsRNA specific to malE (dsmalE), to Dicer-2 (dsDcr2) or to Argona-ute-2 (dsAgo2). Preparation of dsRNA targeting the RNAi machin-ery gene Dicer-2 (dsDcr2) and Argonaute-2 (dsAgo2) was carriedout as described before (Kolliopoulou and Swevers, 2013). In an-other series of experiments, 0.2 lg/ml of BmCPV genomic dsRNAwas transfected in the presence of 1.0 lg/ml of pEA-Myc-RdRp orpEA-PAC (negative control) expression vector and 0.2 lg/ml ofpBmIE1. Cells were collected at different times after transfection(from 2 h up until 8 days; see Section 3) for RNA extraction andpreparation for RT-PCR.

omic dsRNA in silkmoth-derived Bm5 cells: Stability and interactions with/10.1016/j.jinsphys.2014.03.002

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

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

L. Swevers et al. / Journal of Insect Physiology xxx (2014) xxx–xxx 3

IP 3196 No. of Pages 9, Model 5G

20 March 2014

For expression studies, 1.0 lg/ml of pEA-Myc-NS5, pEA-Myc-RdRp or pEA-Flag-B2 expression vector was used together with0.5 lg/ml of pBmIE1 helper plasmid encoding the i.e. -1 gene fromBmNPV (Lu et al., 1996). Cells were harvested at 2–3 days aftertransfection for expression analysis by Western blot.

To investigate the capacity of BmCPV genomic dsRNA to triggergene silencing, 1.0 lg/ml of pEA-Myc-NS5 expression vector and0.2 lg/ml of pBmIE1 were co-transfected with 0.05 lg/ml or0.01 lg/ml of BmCPV genomic dsRNA (the latter in the presenceof 0.04 lg/ml of dsmalE).

2.5. Nucleic acid extraction and RT-PCR

Cell pellets were dissolved in urea/SDS lysis buffer and total nu-cleic acid was extracted as described (Drevet et al., 1995). Thequantity of extracted nucleic acid was assessed with a NanoDrop1000 Spectrophotometer (Thermo Scientific) and/or by electropho-resis on 1% (w/v) agarose gels. First-strand complementary DNA(cDNA) synthesis was performed using RevertAid reversetranscriptase (Thermo Scientific) and oligo-dT or BmCPV segment10-specific primer 50-AGTAAAAGTCAGTATCTTACCGGC-30 (S10F).Oligo-dT primed cDNA was used to detect cellular actin mRNAby PCR using actin primers (Machado et al., 2007). BmCPVpolyhedrin gene was detected with the primers 50-AGGATCATGGCAGACGTAGC-30 and 50-GCAATCACTGACGGTTACTCAG-30 usingS10-primed cDNA as template. Cycling conditions were 94 �C for30 s, 55 �C for 30 s and 72 �C for 30 s for actin mRNA and were94 �C for 1 min, 55 �C for 1 min and 72 �C for 1 min for BmCPVpolyhedrin (25–40 cycles).

Quantification of PCR fragments in agarose gel was carried outby BioCapt image quantification software (Vilber Lourmat, Eber-hardzell, Germany). Quantities of PCR fragments correspondingto the housekeeping gene actin were used for normalization. Rela-tive amounts of the dsRNA fragments were compared with theirlevels at 2–2.5 h after transfection (set as 100%). Statistically signif-icant differences in normalized amounts of dsRNA in differentexperimental conditions or at different time points after transfec-tion were determined by ANOVA analysis.

2.6. Protein extracts preparation and Western blot

Transfected cells cultures were collected and separated as solu-ble cellular extracts and insoluble cellular pellets by centrifugationand freeze–thawing as described (Liu et al., 2012). Protein fractionswere electrophoresed on 10% polyacrylamide gels and subjected toWestern blot analysis as described (Georgomanolis et al., 2009).Proteins were transferred to Hybond ECL membranes (Amersham)and the membrane was blocked with 10% non-fat milk. To detect,Myc- and Flag-tagged proteins, Western blot analysis was carriedout using mouse anti-Myc (Cell Signalling; at 1:1000) or rabbitanti-Flag (Sigma; at 1:1000) as primary antibody and HRP-conju-gated anti-mouse or anti-rabbit (Chemicon; at 1:2000) as second-ary antibody. To detect the GW body marker Gawky, total cellextracts were prepared in cracking buffer (Georgomanolis et al.,2009) and subjected to SDS–PAGE and Western blot. Gawky pro-tein was detected using anti-Gawky (Schneider et al., 2006) andanti-guinea pig HRP (Chemicon) antibodies (both at 1:1000).

2.7. RNAi response assay in Bm5 cells

RNAi experiments were carried out as described before (Swe-vers et al., 2011; Kolliopoulou and Swevers, 2013).

To investigate possible interference of BmCPV dsRNA on RNAiefficiency, Bm5 cells were transfected with 3.3 lg/ml of ecdy-sone-responsive luminescence reporter pERE.bA.luc, 0.17 lg/mlof ecdysone-responsive fluorescence reporter pEcRE.bA.gfp, as well

Please cite this article in press as: Swevers, L., et al. Transfection of BmCPV genthe core RNAi machinery. Journal of Insect Physiology (2014), http://dx.doi.org

as with 0.8 lg/ml of BmCPV genomic dsRNA. DsMalE, which is spe-cific for maltose binding protein mRNA of Escherichia coli (Zhuet al., 2011), was used as non-specific control dsRNA. Moreover,all samples were transfected with 0.01–0.1 lg/ml of luciferase spe-cific dsRNA (dsLuc).

To investigate the expression of viral genes on RNAi efficiency,Bm5 cells were transfected with 0.9 lg/ml of pERE-Luc, 0.2 lg/mlof pERE-gfp, 0.9 lg/ml of expression plasmid pEA-PAC (negativecontrol), pEA-Myc-NS5, pEA-Myc-RdRp or pEA-Flag-B2, and0.05 lg/ml of dsLuc or non-specific dsMalE as a negative control.Two days after transfection, the ecdysone agonist tebufenozidewas added to a final concentration of 500 nM to induce the ecdy-sone reporter genes pERE-luc and pERE-gfp. Soluble cellularextracts of transfected cell populations were prepared as describedand directly used for fluorescence measurements or processed forluminescence measurements using the Steady-Glo� LuciferaseAssay System kit (Promega) according to the manufacturer’sinstructions. Specific luciferase activities were calculated as lumi-nescence/fluorescence ratios. Both fluorescence and luminescencemeasurements were carried out with an Infinite M200 lumino-meter (Tecan, Switzerland). Statistical analysis of differences innormalized luciferase activity was carried out by unpaired t-testanalysis.

2.8. Agro-infiltration

ORFs of BmCPV NS5 and nodavirus B2 were cloned as BamHI-fragments in the primary cloning vector pART7, downstream ofthe cauliflower mosaic virus 35S promoter. The 35S expression cas-sette was subsequently cloned as a NotI-fragment into the T-DNAvector pART27 (binary vector system; Gleave, 1992). Agro-infiltra-tion of GFP only or in combination with either BmCPV NS5, noda-virus B2 or P19 ORFs were carried out in leaves of Nicotianabenthamiana transgenic line 16C stably expressing a GFP trans-gene. Phenotype of co-suppression of the endogenous GFP trig-gered by the additional infiltrated GFP has been reported inHamilton et al. (2002). Co-infiltration of GFP with the well-knownsuppressor of RNA silencing P19 sustains GFP fluorescence and isused as a positive control.

2.9. Subcellular localization of fluorescent dsRNA andimmunofluorescence

DsRNA specific for GFP (dsGFP) was synthesized as described(Liu et al., 2013) and labelled with fluorescein using the SilencerSiRNA Labeling Kit (Ambion) according to the instructions of themanufacturer. Based on absorbance measurements at 260 nmand 492 nm, the base:dye ratio was calculated as 132. This indi-cates the presence of approximately 5–6 fluorescein groups oneach dsGFP molecule. Fluorescein-labelled dsGFP (2.5 lg/ml) wastransfected in Bm5 cells and cells were collected after 16 h for fluo-rescence and immunofluorescence staining (Labropoulou et al.,2008). Cells were fixed with 4% formaldehyde in PBS for 20 minand subsequently permeabilized with PBS supplemented with0.1% (v/v) Triton X-100 (PBS-T) for 10 min. Cells were stained over-night at 4 �C with anti-Gawky antibody (Schneider et al., 2006) at1:500. Following five washes in PBS, AlexaFluor-conjugated goatanti-guinea pig secondary antibody (Invitrogen; at 1:2000) wasadded for 1 h at room temperature. Cells were mounted in Mowiol4–88 (Sigma) and examined under a fluorescence microscope(Zeiss Axiovert 25 inverted microscope). For confocal microscopy,the cells were treated as described above and observed in a Bio-Rad confocal microscope (MRC 1024 ES) equipped with Lasersharpsoftware (Bio-Rad) and a kryptonargon laser.

omic dsRNA in silkmoth-derived Bm5 cells: Stability and interactions with/10.1016/j.jinsphys.2014.03.002

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

4 L. Swevers et al. / Journal of Insect Physiology xxx (2014) xxx–xxx

IP 3196 No. of Pages 9, Model 5G

20 March 2014

3. Results

3.1. Stability of BmCPV dsRNA in transfected Bm5 cells

Analysis of nucleic acid extracts of purified BmCPV polyhedraby agarose gel electrophoresis revealed the 10 dsRNA segmentsthat constitute the BmCPV genome and range in size from 4.2 kbuntil 0.95 kb (sequences available at http://www.reoviridae.org/dsRNA_virus_proteins/Cypovirus.htm) (Fig. 1A). BmCPV dsRNAgenome was transfected at 0.5 lg/ml into Bm5 cells in the pres-ence of dsRNA that targets the RNAi core machinery genes Dcr2and Ago2 (also at 0.5 lg/ml). Our previous studies demonstratedthat these dsRNAs could specifically knock-down the expressionof the targeted genes and influence the RNAi efficiency in reporterassays (Kolliopoulou and Swevers, 2013). DsRNA targeting malE(the maltose-binding protein of E. coli) was used as a negative con-trol. At different times after transfection (2.5 h, 2.5 days, 5 days,8 days), aliquots were collected from the transfected cells and pro-cessed for RNA extraction (N = 3). As shown in Fig. 1B, RT-PCRexperiments showed that the polyhedrin gene of BmCPV could bedetected by RT-PCR efficiently for a period of up to 8 days aftertransfection. While the amount of BmCPV dsRNA has dropped sig-nificantly between 2.5 h and 2.5 days post infection (to 43 ± 16%,69 ± 6% and 39 ± 8% for the dsmalE, dsDcr2 and dsAgo2 conditions,respectively), the intracellular levels remained stable between2.5 days and 8 days (Fig. 1B). Statistically significant differencesin the amount of BmCPV dsRNA were found between samplesco-transfected with dsDcr2 and dsAgo2 at 2.5 days after

2.5 hr

2.5 days

5 days

8 days

dsmalE- BmCPV polh

(757 bp)

Actin(379 bp)

Lambda D

NA

HincII/H

indIII

Bm

CPV

dsRN

Agenom

e

A

B

Fig. 1. Stability of BmCPV dsRNA genome in transfected Bm5 cells. Panel A: Ethidium broand a preparation of BmCPV dsRNA genome. Panel B: BmCPV dsRNA genome at 0.5 lg/dsAgo2, as indicated. BmCPV polyhedrin dsRNA and cellular actin mRNA was detected bethidium bromide-stained agarose gel showing the amplified PCR fragments. Amplificagraph with the quantification of relative amounts of dsRNA normalized to actin mRNA (three different conditions (co-transfection with dsmalE, dsDcr2 and dsAgo2) is set at 100%amount.

Please cite this article in press as: Swevers, L., et al. Transfection of BmCPV genthe core RNAi machinery. Journal of Insect Physiology (2014), http://dx.doi.org

transfection (P = 0.006) and between samples co-transfected withdsmalE and dsDcr2 at 5 days after transfection (P = 0.037). No sta-tistically significant differences in the amount of BmCPV dsRNAwere observed in the different experimental conditions at 8 daysafter transfection (Fig. 1B).

3.2. Interactions of BmCPV genomic dsRNA with the core RNAimachinery

We also tested whether co-transfection of BmCPV dsRNA gen-ome could interfere with dsLuc-mediated silencing of the lucifer-ase reporter. In the presence of 0.8 lg/ml of BmCPV dsRNA,dsLuc silenced luciferase activity to 18.7 ± 3.5% (0.01 lg/ml) and2.8 ± 0.5% (0.1 lg/ml) (Fig. 2A). However, similar values were ob-tained when 0.8 lg/ml of control dsRNA (dsmalE) was co-transfec-ted (reduction to 21.6 ± 0.6% at 0.01 lg/ml of dsLuc and to2.1 ± 0.3% at 0.1 lg/ml of dsLuc). Statistical analysis showed no dif-ference between the two types of dsRNA that were co-transfected(P values of 0.25 and 0.47).

However, transfected BmCPV dsRNA was recognized by theRNAi machinery and could induce a silencing response. Whenlow amounts of BmCPV dsRNA genome were co-transfected withthe expression plasmid for Myc-tagged NS5 (encoded by segment9 of the BmCPV dsRNA genome), clear interference with theexpression of the protein was observed (Fig. 2B). The silencing ofotherwise efficiently expressed Myc-NS5 (Fig. 4A) was dose-dependent and occurred at a similar sensitivity as that was ob-served for dsLuc-mediated silencing of the luciferase reporter.

2.5 hr

2.5 days

5 days

8 days

2.5 hr

2.5 days

5 days

8 days

dsDcr2 dsAgo2

mide-stained agarose gel showing Lambda DNA double-digested with HincII/HindIIIml was transfected to Bm5 cells in the presence of 0.5 lg/ml of dsmalE, dsDcr2 ory RT-PCR at different times after transfection. At the top is shown a representativetion cycles were 35� for BmCPV polyhedrin and 28� for actin. Below is shown theN = 3). In the graph, the amount of dsRNA detected at 2.5 h after transfection in the

. The quantities of dsRNA detected at later time points are expressed relative to this

omic dsRNA in silkmoth-derived Bm5 cells: Stability and interactions with/10.1016/j.jinsphys.2014.03.002

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

0.1

0.2

0.3

0.4

0.5

0.6

0.7

0.01 0.1- 0.01 0.1- dsLuc

dsmalE BmCPV dsRNA

luminescence/fluorescence

A

tubulin

Myc-NS535

kDa

65kDa

dsmalE(50 ng)

dsmalE(40 ng)

+ CPV

(10 ng)CPV

(50 ng)

B

Fig. 2. Interaction of BmCPV dsRNA with the RNAi machinery in Bm5 cells. Panel A: BmCPV dsRNA does not interfere with silencing of a luciferase reporter by dsLuc. Doses ofdsLuc were 0.01 or 0.1 lg/ml while the concentration of dsmalE or BmCPV dsRNA was 0.8 lg/ml. Panel B: Co-transfection of BmCPV genomic dsRNA at the indicated doses(10–50 ng/ml) interferes with expression of Myc-NS5 protein from the pEA-Myc-NS5 expression vector. Western blot shows expression of Myc-NS5 and tubulin.

3 days

7 days

3 days

7 days

polh(757 bp)

actin(379 bp)

2 hr

2 hr

PAC RdRpA

125kDa65

kDa

IS ISSOL SOL

PAC Myc-RdRp

RdRp

tubulin

B

C

0.00

0.25

0.50

0.75

1.00

PAC RdRp

dsmalE

dsmalE

dsLuc

dsLuc

luminescence/fluorescence

Fig. 3. Expression of viral RdRp in Bm5 cells and functional assays. Panel A: Western blot detection of Myc-RdRp in soluble and insoluble extracts of the Bm5 cells. Tubulindetection is used as control in the Western blot analysis. Panel B: Stimulation of BmCPV dsRNA amplification by co-expressed Myc-RdRp. BmCPV polyhedrin is amplified byRT-PCR (40 cycles) and actin amplification (25 cycles) is used as control. At the top is shown a representative ethidium bromide-stained agarose gel showing the amplifiedPCR fragments. Below is shown the graph with the quantification of relative amounts of dsRNA normalized to actin mRNA (N = 3). In the graph, the amount of dsRNA detectedat 2 h after transfection in the different conditions (co-transfection with pEA-PAC and pEA-Myc-RdRp) is set at 100%. The quantities of dsRNA detected at later time points areexpressed relative to this amount. Panel C: Expression of Myc-RdRp does not interfere with silencing of a luciferase reporter by dsLuc (at 50 ng/ml). In all cases, Bm5 cellstransfected with EA-PAC were used as negative control for Myc-RdRp expression and functional analysis.

L. Swevers et al. / Journal of Insect Physiology xxx (2014) xxx–xxx 5

IP 3196 No. of Pages 9, Model 5G

20 March 2014

Silencing of Myc-NS5 was indeed observed at concentrations of10–50 ng/ml of BmCPV dsRNA which corresponds to 0.5–2.5 ng/ml of segment 9 dsRNA (total length of BmCPV dsRNA geno-me = 24758 bp; length of segment 9 dsRNA = 1186 bp, of which963 bp correspond to the NS5 ORF; http://www.reoviridae.org/dsRNA_virus_proteins/Cypovirus.htm).

3.3. Expression of viral RdRp and investigating its capacity to amplifyBmCPV dsRNA and to modulate dsRNA-mediated gene silencing

The viral RdRp was cloned in the pEA-N-Myc expression vectorwhich was subsequently used to transfect Bm5 cells in the pres-ence of the pBmIE1 helper plasmid (Douris et al., 2006). Westernblot analysis revealed a cross-reacting protein to the Myc antibodythat is somewhat smaller (approximately125 kDa) than the pre-dicted size (138.6 kDa) in the soluble extracts of the transfectedbut not the untransfected cells (Fig. 3A).

Please cite this article in press as: Swevers, L., et al. Transfection of BmCPV genthe core RNAi machinery. Journal of Insect Physiology (2014), http://dx.doi.org

To test whether expression of viral RdRp could stimulate ampli-fication of BmCPV dsRNA genome, co-transfections were carriedout in the presence of 0.2 lg/ml of BmCPV genomic dsRNA. At3 days after transfection, BmCPV dsRNA was detected at 3.1(±0.3) -fold higher amounts in cells co-transfected with the expres-sion vector for viral RdRp than in the cells transfected with a con-trol construct (Fig. 3B; P = 0.0002). Also at 7 days after transfection,higher amounts of BmCPV dsRNA were detected in cells co-trans-fected with RdRp expression vector (P = 0.005).

Finally, we investigated whether expression of viral RdRp couldinterfere with the RNAi process, using assays in which a luciferasereporter was silenced by co-transfected dsRNA targeting the lucif-erase ORF (dsLuc). Low amounts of dsLuc (0.05 lg/ml; achievingapproximately a knockdown of 50%) were used in the experimentto reveal more clearly possible modulations of the silencing re-sponse. In the presence of control expression plasmid (pEA-PAC),dsLuc silenced the luciferase reporter to 50.7 ± 2.7%, while in the

omic dsRNA in silkmoth-derived Bm5 cells: Stability and interactions with/10.1016/j.jinsphys.2014.03.002

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

A

35kDa

65kDa

IS ISSOL SOL

control

Myc-NS5

tubulin

Flag-B2

transfected

12kDa

B

0.0

0.2

0.4

0.6

0.8

dsmalE

dsmalE

dsLuc

dsLuc

dsmalE

dsLuc

PAC B2 NS5

luminescence/fluorescence

Fig. 4. Expression of viral NS5 in Bm5 cells and functional assay. Panel A: Western blot detection of Myc-NS5 and nodaviral Flag-B2 in soluble and insoluble extracts of theBm5 cells. Tubulin detection is used as control in the Western blot analysis. Panel B: Expression of Myc-NS5 or Flag-B2 does not interfere with silencing of a luciferasereporter by dsLuc (at 50 ng/ml). In all cases, Bm5 cells transfected with EA-PAC were used as negative control for Myc-NS5 expression and functional analysis.

6 L. Swevers et al. / Journal of Insect Physiology xxx (2014) xxx–xxx

IP 3196 No. of Pages 9, Model 5G

20 March 2014

presence of RdRP expression vector luciferase was reduced to49.0 ± 8.4%, indicating no modulation of the RNAi response byRdRp (P = 0.86, Fig. 3C).

3.4. Expression of viral NS5 and investigating its capacity to modulatedsRNA-mediated gene silencing in Bm5 cells

Transfection of their respective expression vectors in Bm5 cellsresulted in efficient expression of BmCPV-derived Myc-tagged NS5and nodavirus-derived Flag-tagged B2 proteins (Fig. 4A). The B2protein is derived from the nodavirus that persistently infectsHi5 cells (Li et al., 2007). It was demonstrated that B2 functionsas an inhibitor of RNAi because of its propensity to bind dsRNAand siRNA (Lingel et al., 2005). On the other hand, NS5 is a non-structural protein that is encoded by BmCPV genomic dsRNA seg-ment 9 and for which dsRNA-binding activity has been demon-strated (Hagiwara et al., 1998).

When expressed in Bm5 cells, we found that neither nodaviralB2 nor BmCPV-derived NS5 could interfere with the silencing ofa luciferase reporter by co-transfected dsLuc (Fig. 4B). DsLuc at0.05 lg/ml suppressed luciferase activity to 32.9 ± 3.4% in controlcells, to 24.7 ± 3.0% in B2-expressing cells, and to 29.3 ± 0.8% inNS5-expressing cells. Neither for B2 (P = 0.14) nor for NS5(P = 0.36), the repression level differed significantly from thecontrol.

3.5. Investigating the capacity of viral NS5 to modulate dsRNA-mediated gene silencing in agro-infiltrated tobacco leaves

Agro-infiltration in N. benthamiana leaves is a powerful ap-proach to characterize putative suppressors of RNA silencing(Helm et al., 2011). Co-expression of NS5 in agro-infiltrated leavesdid not delay silencing of the GFP reporter, in contrast to expres-sion of P19 of tombusvirus (Fig. 5A). On the other hand, nodavirusB2 could function as a strong RNAi suppressor in plants (Fig. 5B).Similar results were found for the tagged versions of B2 (N-termi-nally Flag-tagged and C-terminally MycHis-tagged; data notshown).

3.6. Distribution of transfected dsRNA in the intracellular environment

Although amplification of dsRNA can be considered as a factor(see Section 4), the most important factor for the stability of dsRNAin transfected cells likely is the inefficiency of the intracellular RNAdegradation machinery to recognize and degrade transfecteddsRNA. In Drosophila and mammalian cells, cytoplasmic bodieshave been identified that are implicated in the regulation of mRNA

Please cite this article in press as: Swevers, L., et al. Transfection of BmCPV genthe core RNAi machinery. Journal of Insect Physiology (2014), http://dx.doi.org

stability, translation and the RNAi pathway (Schneider et al., 2006).These cytoplasmic bodies (called P-bodies in mammalian cells;GW-bodies in Drosophila cells) can be visualized by antibodies di-rected against GW182 protein, which is an effector in the RNAipathway (Jakymiw et al., 2005).

To investigate whether transfected dsRNA could localize to GWbodies, we transfected Bm5 cells with FITC-labeled dsRNA and per-formed co-localization studies using an antibody directed againstGawky, the Drosophila homolog of GW182 (Schneider et al.,2006). When used in Western blot, the anti-Gawky antibody recog-nized a protein of approximately 160 kDa in Drosophila-derived S2cells, and a protein of 120 kDa in Bm5 cells (Fig. 6A). The size dif-ferences are consistent with predicted sequence information: Dro-sophila Gawky isoforms consist of around 1380–1385 AA while thehomolog in the monarch butterfly, Danaus plexippus, is predicted tobe considerably smaller (1088 AA; accession EHJ71059). While thehomolog in Bombyx has not been completely annotated yet, it issafe to assume that it will correspond in size to the protein pre-dicted in Danaus.

In co-staining experiments, transfected FITC-dsRNA and Gawkyshowed a different intracellular localization: Gawky-reactivebodies showed mostly a peripheral localization in the cell whiledsRNA was detected more centrally (Fig. 6B). While some dsRNAstaining overlapped with that of Gawky at the periphery, it is clearthat the majority of transfected dsRNA had a different localizationin the cell.

4. Discussion

In this work, evidence is provided for long-term stability ofdsRNA in the intracellular environment after its initial transfectioninto silkmoth-derived Bm5 cells. At 8 days after transfection,considerable amounts of dsRNA could still be detected by RT-PCR,and the amount of intracellular dsRNA remained remarkablystable between 2.5 days and 8 days after transfection (Figs. 1 and2). Although in these experiments, a special type of dsRNA samplewas used, corresponding to the dsRNA genome of BmCPV, anotherstudy has also demonstrated rather long-term persistence (up toat least 4 days) of dsGFP in another lepidopteran cell line, Hi5 (Liuet al., 2012).

Although the cause for the persistence remains unknown, it canbe speculated that the cells do not express many nucleases that candegrade long perfect duplexes of dsRNA efficiently, in contrast tossRNA. However, it is well established that dsRNA is recognizedby Dicer enzymes and becomes incorporated in RISC complexessince it will trigger homologous gene silencing (Figs. 2–4; Swevers

omic dsRNA in silkmoth-derived Bm5 cells: Stability and interactions with/10.1016/j.jinsphys.2014.03.002

492

493

494

495

496

497

498

499

500

501

502

503

504

505

506

507

508

509

510

511

512

513

514

515

516

517

518

519

520

521

522

523

524

525

526

527

528

529

530

531

532

533

534

535

536

537

GFP+B2

GFP + p19

GFPGFP + p19

GFP + NS5

GFP

A B

Fig. 5. Assay for RNAi suppressor activity in agro-infiltrated leaves. Panel A: Suppression of RNAi is observed in agro-infiltrated leaf areas with tombus-virus P19 expression,but not in agro-infiltrated leaf areas with BmCPV NS5 expression. Panel B: Strong suppression of RNAi in agro-infiltrated leaf areas with nodaviral B2 expression. In bothpanels, ‘‘GFP’’ shows co-suppression by additional infiltrated GFP in the absence of additionally expressed protein (P19, NS5 or B2).

Gawky

tubulin

S2 Bm5

65kDa

160kDa

120kDa

FITC-dsRNA anti-Gawky overlayA B

Fig. 6. Detection of Gawky protein in Bm5 cells and co-localization with transfected FITC-labeled dsRNA. Panel A: Western blot analysis of Gawky expression in Drosophila S2cells and Bombyx Bm5 cells. Tubulin expression is shown as control. Panel B: Immunofluorescence detection of Gawky protein (AlexaFluor) and fluorescence detection ofFITC-dsRNA in Bm5 cells. Shown are three representative stained cells. Magnification = 200�.

L. Swevers et al. / Journal of Insect Physiology xxx (2014) xxx–xxx 7

IP 3196 No. of Pages 9, Model 5G

20 March 2014

et al., 2011; Liu et al., 2012; Kolliopoulou and Swevers, 2013). It isnoted that efficient silencing can be triggered by very smallamounts of dsRNA (5–10 ng/ml) and only small fractions of trans-fected dsRNA in our experiments therefore need to be processed byDicer-2 to cause efficient silencing by RISC complexes. Processingby Dicer-2 may therefore be a minor factor that affects dsRNA sta-bility. This result is corroborated by the observation that co-trans-fection with dsRNA targeting Dicer-2 has only a minor impact onthe abundance of dsRNA (Fig. 1B). The suggestion that dsRNA-degrading enzymes are limiting in lepidopteran cell lines is alsostrengthened by the observation that expression of dsRNA-degrad-ing enzymes can significantly accelerate the degradation of dsRNA(Liu et al., 2012).

Another possible factor may be amplification by intracellularunknown RNA-dependent RNA polymerases which could counter-balance degradation activity. This is expected to occur since trans-fected cells are dividing which would lead to loss of dsRNA unless acompensatory mechanism occurs. An extra factor playing in ourexperiments relates to the use of BmCPV dsRNA which could trig-ger expression of BmCPV proteins that can affect amplification ofdsRNA, such as the viral RdRp. Although we observed that viralRdRp could stimulate BmCPV replication up to 3-fold (Fig. 4), itshould be kept in mind that the polymerase was expressed from

Please cite this article in press as: Swevers, L., et al. Transfection of BmCPV genthe core RNAi machinery. Journal of Insect Physiology (2014), http://dx.doi.org

an expression vector plasmid and that expression from transfecteddsRNA likely has much lower efficiency.

It is known that dsRNA readily can undergo editing andunwinding by RNA-editing enzymes such as adenosine deaminasesthat act on RNA (ADARs; Keegan et al., 2011). Since edited dsRNA,but not non-edited dsRNA, is a target for degradation by Tudor-SNenzymes, editing can be considered as the trigger for an alternativedegradation pathway for dsRNA (Scadden, 2005). However, thecytoplasm of Bm5 cells does not contain unwinding (and thereforeediting) activity although it exists in the nuclei (Skeiky and Iatrou,1991). Thus, the majority of cell’s volume may in this case be de-void of this degradation pathway and this could be a contributingfactor to explain the stability of dsRNA.

It is possible that cellular or viral dsRNA-binding factors protectdsRNA from degradation. For instance, Hi5 cells are persistently in-fected with a nodavirus which produces the B2 antiviral suppres-sor (Li et al., 2007; Lingel et al., 2005). B2 protein can sequestratedsRNA and siRNA from viral origin to prevent their further process-ing by the RNAi machinery (Lingel et al., 2005). However, Bm5 cellsdo not seem to be infected by nodavirus, in contrast to Hi5 cells(unpublished results), and the accumulation of B2 protein cantherefore not explain dsRNA persistence in this cell line (see alsobelow for further discussion regarding B2).

omic dsRNA in silkmoth-derived Bm5 cells: Stability and interactions with/10.1016/j.jinsphys.2014.03.002

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

628

629

630

631

632

633

634

635

636

637

638

639

640

641

642

643

644

645

646Q3

647Q5

648

649

650

651

652

653

654

655

656

657

658

659

660661662663664665

8 L. Swevers et al. / Journal of Insect Physiology xxx (2014) xxx–xxx

IP 3196 No. of Pages 9, Model 5G

20 March 2014

In the experiments presented in this work, dsRNA is introducedinto the cells by transfection. Transfection can be considered as anunnatural way to introduce dsRNA into the cells. By contrast,dsRNA from the extracellular medium enters the cells naturallyby endocytosis and evidence has accumulated that late endo-somes/multivesicular bodies constitute a critical point in the endo-cytosis pathway where RNAi is activated (Lee et al., 2009;Rocheleau, 2012). Evidence exists that only a very small amountof dsRNA (1–2%) can escape from the endosomes while the remain-der will undergo exocytosis or degradation in lysosomes (Wangand Huang, 2013). Since the RNAi machinery is associated withthe endosome pathway, dsRNA that has escaped from the endo-some may be processed immediately and not allowed to spread in-side the cells. A similar argumentation can be applied to othernatural sources of dsRNA such as viral infection and productionof RNA hairpins at genomic loci: locally produced dsRNA structuresmay not have the opportunity to spread throughout the cell be-cause of strict surveillance. By contrast, transfected dsRNA occursthroughout the cell (Fig. 6); as an example, very little overlap is ob-served with GW-bodies, where the RNA-degradation machinery(including RISC components) is concentrated (Schneider et al.,2006).

The presence of BmCPV genomic dsRNA clearly does not affectRNAi efficiency, despite its recognition by the RNAi machinery(Fig. 2). This suggests that the RNAi machinery in Bm5 cells has arather large capacity to respond to dsRNA which cannot be satu-rated easily. Transfection of BmCPV dsRNA also represents a spe-cial case because of its encoding of ORFs which, if expressed,could interfere with RNAi at the protein level. To test this possibil-ity, two ORFs of the BmCPV genome, RdRp and NS5, were cloned inexpression vectors to check for possible interference with RNAi.Although expression was observed, no effect on silencing of a lucif-erase reporter could be observed (Figs. 3 and 4). However, no effectin this Bm5 cell-based assay was also observed for B2, a knownRNAi inhibitor from nodavirus (Lingel et al., 2005; Singh et al.,2009). When tested in another assay based on agro-infiltration ofN. benthamiana leaves, high suppressor activity of B2 (but not ofNS5) was observed (Fig. 5). The different outcomes between thetwo assays (Bm5 cell transfection and N. benthamiana leaf agro-infiltration) can be explained by two mechanisms: (1) the relativeamount of the targeting transgene in agro-infiltrated tissues ishigher than in transfected Bm5 cells, due to multiple insertionsof T-DNA, and strong and constitutive transgene expression con-trolled by the p35S promoter; and (2) amplification of and expres-sion from agro-infiltration vectors occur locally and geneticelements as RNA hairpin and RNAi inhibitor are more likely tointeract. To achieve better results in RNAi inhibition experimentsemploying Bm5 cells, we have tried to stimulate expression ofRNAi inhibitors by co-transfection with pIE1 plasmid (Douriset al., 2006), but were unsuccessful. Regarding the second possibil-ity, there is evidence that the B2 protein acts locally rather thanthroughout the cell during nodavirus infection; it may preferen-tially interact with viral RdRp and through this interaction seques-trate emerging dsRNA structures during viral replication andtranscription (Aliyari et al., 2008). Thus, protein inhibitors of RNAimay affect the RNAi efficiency only at high concentrations, whichcan be achieved only locally during persistent viral replicationand only globally in special circumstances.

A second observation in this work relates to the generation ofan in vitro culture system for BmCPV virus. Such in vitro culturesystem would be of great value for the genetic analysis of BmCPVfunction and the generation of recombinant virus. However, nofunctional virus was obtained after transfection with BmCPV geno-mic dsRNA. The failure is not likely caused by the degradation ofgenomic dsRNA or viral transcripts by the RNAi machinery(Fig. 1B), since co-transfection with dsRNA targeting Dicers and

Please cite this article in press as: Swevers, L., et al. Transfection of BmCPV genthe core RNAi machinery. Journal of Insect Physiology (2014), http://dx.doi.org

Argonaute proteins did not result in an increase in dsRNA abun-dance. In these experiments, it is assumed that Dicer enzymes willdegrade dsRNA directly while Argonaute proteins will silence theviral mRNAs after the generation of viral siRNAs (Siomi and Siomi,2009). In our previous study, efficient silencing of RNAi machinerygenes by similar doses of dsRNA was achieved that resulted in clearinhibition of RNAi in a luciferase reporter assay (Kolliopoulou andSwevers, 2013).

Two additional hypotheses are therefore considered to explainthe failure to generate an in vitro culture system for BmCPV: first,it is possible that BmCPV only replicates efficiently in specializedcells of the midgut, its main infection site (Wu et al., 2011). How-ever, BmCPV infection is known to spread to other tissues in somecases (unpublished results; Payne and Mertens, 1983) and is there-fore not restricted to one cell type. A second possibility is that geneexpression from transfected dsRNA is very inefficient and thatdsRNA amplification and virion formation only can be achievedby co-expression of high amounts of viral proteins. While a smallincrease in dsRNA abundance was observed after co-expressionwith viral RdRp (Fig. 3), it is probably necessary to express mostof the 10 proteins of the BmCPV genome at sufficiently high levelstogether with the efficient introduction of the 10 dsRNA genomicsegments in order to establish a culture system. This strategy hasalready been used to establish an in vitro production system forinfluenza virus, the genome of which consists of 8 segments of(-)-ssRNA. In this approach, 8 plasmids are transfected in a mam-malian cell line which generate 8 viral mRNAs by RNA polymeraseII concomitant with the production of the 8 genomic (-)-ssRNAs byRNA polymerase I (Hoffmann et al., 2002).

In summary, our study records the remarkable stability ofdsRNA in the intracellular environment after transfection, whichcontrasts to the reported instability of dsRNA in some extracellularmedia, such as gut content, hemolymph and saliva (Li et al., 2011;Garbutt et al., 2013; Liu et al., 2013; Christiaens et al., in press).This stability is possibly explained by the transfection procedurein which dsRNA is spread throughout the cell volume, while themachinery that directs RNA degradation, including RNAi, has alocal distribution. It is also established that a BmCPV culture sys-tem cannot be established by simple transfection of genomicdsRNA (in contrast to transfection of DNA of BmNPV), even in con-ditions of knock-down of RNAi genes.

Acknowledgements

This work was supported by the National Natural Science Foun-dation of China (grant number 31172263), Guangdong Natural Sci-ence Foundation of China grant number (S2013010016750), theFund for Scientific Research (FWO-Vlaanderen, Belgium), and bythe General Secretariat for Research and Technology, HellenicRepublic Ministry of National Education and Religious Affairs, inGreece. Thanks are due to D. Stefanou for technical support inmolecular cloning and Western blot techniques and to Dr. T. The-odossiou and Dr. M. Sagnou for expert technical support at the La-ser Confocal Subunit, Institute of Biosciences & Applications, NCSR‘‘Demokritos’’. We also thank Dr. A. Simmonds (University of Alber-ta, Canada) for a sample of the polyclonal antibody against Dro-sophila GW (CG31992).

References

Aliyari, R., Wu, Q., Li, W.-H., Wang, X.-H., Li, F., Green, L.D., Han, C.S., Li, W.-X., Ding,S.-W., 2008. Mechanism of induction and suppression of antiviral immunitydirected by virus-derived small RNAs in Drosophila. Cell Host & Microbe 4, 387–397.

Allen, M.L., Walker, W.B., 2012. Saliva of Lygus lineolaris digests double strandedribonucleic acids. J. Insect Physiol. 58, 391–396.

omic dsRNA in silkmoth-derived Bm5 cells: Stability and interactions with/10.1016/j.jinsphys.2014.03.002

666667668 Q4669670671672673674675676677678679680681682683684685686687688689690691692693694695696697698699700701702703704705706707708709710711712713714715716717718719720721722723724725726727728729730731732733734735736737738

739740741742743744745746747748749750751752753754755756757758759760761762763764765766767768769770771772773774775776777778779780781782783784785786787788789790791792793794795796797798799800801802803804805806807808809810

811

L. Swevers et al. / Journal of Insect Physiology xxx (2014) xxx–xxx 9

IP 3196 No. of Pages 9, Model 5G

20 March 2014

Carthew, R.W., Sontheimer, E.J., 2009. Origins and mechanisms of miRNAs andsiRNAs. Cell 136, 642–655.

Christiaens, O., Swevers, L., Smagghe, G., in press. DsRNA degradation in the peaaphid (Acyrthosiphon pisum) associated with lack of response in RNAi feedingand injection assay. Peptides.

Douris, V., Swevers, L., Labropoulou, V., Andronopoulou, E., Georgoussi, Z., Iatrou, K.,2006. Stably transformed insect cell lines: tools for expression of secreted andmembrane-anchored proteins and high-throughput screening platforms fordrug and insecticide discovery. Adv. Virus Res. 68, 113–156.

Drevet, J.R., Swevers, L., Iatrou, K., 1995. Developmental regulation of a silkwormgene encoding ‘‘GATA’’ – type transcription factors by alternative splicing. J.Mol. Biol. 246, 43–53.

Garbutt, J.S., Bellés, X., Richards, E.H., Reynolds, S.E., 2013. Persistence of double-stranded RNA in hemolymph as a potential determiner of RNA interferencesuccess: evidence from Manduca sexta and Blatella germanica. J. Insect Physiol.59, 171–178.

Georgomanolis, T., Iatrou, K., Swevers, L., 2009. BmCAP, a silkmoth gene encodingmultiple protein isoforms characterized by SoHo and SH3 domains: expressionanalysis during ovarian follicular development. Insect Biochem. Mol. Biol. 39,892–902.

Gleave, A.P., 1992. A versatile binary vector system with a T-DNA organisationalstructure conducive to efficient integration of cloned DNA into the plantgenome. Plant Mol. Biol. 20, 1203–1207.

Hagiwara, K., Tomita, M., Nakai, K., Kobayashi, J., Miyajima, S., Yoshimura, T., 1998.Determination of the nucleotide sequence of Bombyx mori cytoplasmicpolyhedrosis virus segment 9 and its expression in BmN4 cells. J. Virol. 72,5762–5768.

Hamilton, A., Voinnet, O., Chappell, L., Baulcombe, D., 2002. Two classes of shortinterfering RNA in RNA silencing. EMBO J. 21, 4671–4679.

Hayashi, Y., Bird, F.T., 1969. The isolation of cytoplasmic polyhedrosis virus from thewhite-marked tussock moth, Orgyia leucostigma (Smith). Can. J. Microbiol. 16,695–701.

Helm, J.M., Dadami, E., Kalantidis, K., 2011. Local RNA silencing mediated byAgroinfiltration. Methods Mol. Biol. 744, 97–108.

Hill, C.J., Booth, T.F., Stuart, D.I., Mertens, P.P.C., 1999. Lipofectin increases thespecific activity of cypovirus particles for cultured insect cells. J. Virol. Meth. 78,177–189.

Hoffmann, E., Mahmood, K., Yang, C.-F., Webster, R.G., Greenvberg, H.B., Kemble, G.,2002. Rescue of influenza B virus from eight plasmids. Proc. Natl. Acad. Sci. USA99, 11411–11416.

Huvenne, H., Smagghe, G., 2010. Mechanisms of dsRNA uptake in insects andpotential of RNAi for pest control: a review. J. Insect Physiol. 56, 227–235.

Jakymiw, A., Lian, S., Eystathioy, T., Li, S., Satoh, M., Hamel, J.C., Fritzler, M.J., Chan,E.K., 2005. Disruption of GW bodies impairs mammalian RNA interference. Nat.Cell Biol. 7, 1267–1274.

Jinek, M., Doudna, J.A., 2009. A three-dimensional view of the molecular machineryof RNA interference. Nature 457, 405–412.

Johnson, R., Meidinger, R.G., Iatrou, K., 1992. A cellular promoter-based expressioncassette for generating recombinant baculoviruses directing rapid expression ofpassenger genes in infected insects. Virology 190, 815–823.

Keegan, L.P., McGurk, L., Palavicini, J.P., Brindle, J., Paro, S., Li, X., Rosenthal, J.J.C.,O’Connell, M.A., 2011. Functional conservation in human and Drosophila ofMetazoan ADAR2 involved in RNA editing: loss of ADAR1 in insects. NucleicAcids Res. 39, 7249–7262.

Kolliopoulou, A., Swevers, L., 2013. Functional analysis of the RNAi response inovary-derived silkmoth Bm5 cells. Insect Biochem. Mol. Biol. 43, 654–663.

Labropoulou, V., Douris, V., Stefanou, D., Magrioti, C., Swevers, L., Iatrou, K., 2008.Endoparasitoid wasp bracovirus-mediated inhibition of hemolin function andlepidopteran host immunosuppression. Cell Microbiol. 10, 2118–2128.

Lee, Y.S., Pressman, S., Andress, A.P., Kim, K., White, J.L., Cassidy, J.J., et al., 2009.Silencing by small RNAs is linked to endosomal trafficking. Nat. Cell Biol. 11,1150–1156.

Li, T.C., Scotti, P.D., Miyamura, T., Takeda, N., 2007. Latent infection of a newalphanodavirus in an insect cell line. J. Virol. 81, 10890–10896.

Li, J., Chen, Q., Lin, Y., Jiang, T., Wu, G., Hua, H., 2011. RNA interference in Nilaparvatalugens (Homoptera: Delphacidae) based on dsRNA ingestion. Pest Manag. Sci.67, 852–859.

Lingel, A., Simon, B., Izaurralde, E., Sattler, M., 2005. The structure of the flock housevirus B2 protein, a viral suppressor of RNA interference, shows a novel mode ofdouble-stranded RNA recognition. EMBO Rep. 6, 1149–1155.

Liu, J., Carmell, M.A., Rivas, F.V., Marsden, C.G., Thomson, J.M., Song, J.-J., Hammond,S.M., Joshua-Tor, L., Hannon, G.J., 2004. Argonaute2 is the catalytic engine ofmammalian RNAi. Science 305, 1437–1441.

Please cite this article in press as: Swevers, L., et al. Transfection of BmCPV genthe core RNAi machinery. Journal of Insect Physiology (2014), http://dx.doi.org

Liu, J., Smagghe, G., Swevers, L., 2013. Transcriptional response of BmToll9-1 andRNAi machinery genes to exogenous dsRNA in the midgut of Bombyx mori. J.Insect Physiol. 59, 646–654.

Liu, J., Swevers, L., Iatrou, K., Huvenne, H., Smagghe, G., 2012. Bombyx mori DNA/RNA non-specific nuclease: expression of isoforms in insect culture cells,subcellular localization and functional assays. J. Insect Physiol. 58, 1166–1176.

Lu, M., Johnson, R.R., Iatrou, K., 1996. Trans-Activation of a cell housekeeping genepromoter by the IE1 gene product of baculoviruses. Virology 218, 103–113.

Machado, E., Swevers, L., Sdralia, N., Medeiros, M.N., Fernando, G., Mello, F.G., Iatrou,K., 2007. Prostaglandin signaling and ovarian follicle development in thesilkmoth, Bombyx mori. Insect Biochem. Mol. Biol. 37, 876–885.

Moazed, D., 2009. Small RNAs in transcriptional gene silencing and genome defense.Nature 457, 413–420.

Mori, H., Metcalf, P., 2010. Cypoviruses. In: Asgari, Sassan, Johnson, Karyn (Eds.),Insect Virology. Caister Academic Press, Norfolk, UK, pp. 307–323.

Myers, J.W., Jones, J.T., Meyer, T., Ferrell Jr., J.E., 2003. Recombinant Dicer efficientlyconverts large dsRNAs into siRNAs suitable for gene silencing. Nat. Biotechnol.21, 324–328.

O’Reilly, D.R., Miller, L.K., Luckow, V.A., 1992. Baculovirus Expression Vectors – ALaboratory Manual. W.H. Freeman and Co., New York.

Payne, C.C., Mertens, P.P.C., 1983. Cytoplasmic polyhedrosis viruses. In: Joklik, W.K.,Fraenkel-Conrat, H., Wagner, R.R. (Eds.), The Reoviridae. Plenum press, NewYork, pp. 425–504.

Rocheleau, C.E., 2012. RNA interference: systemic RNAi SIDes with endosomes. Curr.Biol. 22, R873–R875.

Saleh, M.C., van Rij, R.P., Hekele, A., Gillis, A., Foley, E., O’Farrell, P.H., Andino, R.,2006. The endocytic pathway mediates cell entry of dsRNA to induce RNAisilencing. Nat. Cell Biol. 8, 793–802.

Scadden, A.D., 2005. The RISC subunit Tudor-SN binds to hyper-edited double-stranded RNA and promotes its cleavage. Nat. Struct. Mol. Biol. 12, 489–496.

Schneider, M.D., Najand, N., Chaker, S., Pare, J.M., Haskins, J., Hughes, S.C., Hobman,T.C., Locke, J., Simmonds, A.J., 2006. Gawky is a component of cytoplasmicmRNA processing bodies for early Drosophila development. J. Cell Biol. 174,349–358.

Scott, J.G., Michel, K., Bartholomay, L., Siegfried, B.D., Hunter, W.B., Smagghe, G., Zhu,K.Y., and Douglas, A.E., in press. Towards the elements of successful insect RNAi.J. Insect Physiol. http://dx.doi.org/10.1016/j.jinsphys.2013.08.014.

Singh, G., Popli, S., Hari, Y., Malhotra, P., Mukherjee, S., Bhatnagar, R.K., 2009.Suppression of RNA silencing by Flock house virus B2 protein is mediatedthrough its interaction with the PAZ domain of Dicer. FASEB J. 23, 1845–1857.

Siomi, H., Siomi, M.C., 2009. On the road to reading the RNA-interference code.Nature 457, 396–404.

Skeiky, Y.A., Iatrou, K., 1991. Developmental regulation of covalent modification ofdouble-stranded RNA during silkmoth oogenesis. J. Mol. Biol. 218, 517–527.

Swevers, L., Liu, J., Huvenne, H., Smagghe, G., 2011. Search for limiting factors in theRNAi pathway in silkmoth tissues and Bm5 cells: the RNA-binding proteinsR2D2 and Translin. PLoS One 6, e20250.

Swevers, L., Smagghe, G., 2012. Use of RNAi for control of insect crop pests. In:Smagghe, G., Diaz, I. (Eds.), Progress in Biological Control. Springer-Verlag,Dordrecht, pp. 177–197.

Tan, P.C., Xu, X.Y., Sun, J.C., 2001. General technique about inoculation andpurification of silkworm pathogen. Guangdong Sericulture 35, 26–28.

Terenius, O., Papanicolaou, A., Garbutt, J.S., Eleftherianos, I., Huvenne, H.,Kanginakudru, S., et al., 2011. RNA interference in Lepidoptera: an overviewof successful and unsuccessful studies and implications for experimentaldesign. J. Insect Physiol. 57, 231–245.

Tomari, Y., Du, T., Zamore, P.D., 2007. Sorting of Drosophila small silencing RNAs.Cell 130, 299–308.

van Rij, R.P., Saleh, M.-C., Berry, B., Foo, C., Hook, A., Antoniewski, C., Andino, R.,2006. The RNA silencing endonuclease Argonaute 2 mediates specific antiviralimmunity in Drosophila melanogaster. Genes Dev. 20, 2985–2995.

Wang, Y., Huang, L., 2013. A window into siRNA delivery. Nat. Biotechol. 31, 611–612.

Wu, P., Li, M.-W., Wang, X., Zhao, P., Wang, X.-Y., Liu, T., Qin, G.-X., Guo, X.-J., 2009.Differentially expressed genes in the midgut of silkworm infected withcytoplasmic polyhedrosis virus. African J. Biotechnol. 8, 3711–3720.

Wu, P., Wang, X., Qin, G.X., Liu, T., Jiang, Y.F., et al., 2011. Microarray analysis of thegene expression profile in the midgut of silkworm infected with cytoplasmicpolyhedrosis virus. Mol. Biol. Rep. 38, 333–341.

Zhu, F., Xu, J., Palli, R., Ferguson, J., Palli, S.R., 2011. Ingested RNA interference formanaging the populations of the Colorado potato beetle, Leptinotarsadecemlineata. Pest Manag. Sci. 67, 175–182.

omic dsRNA in silkmoth-derived Bm5 cells: Stability and interactions with/10.1016/j.jinsphys.2014.03.002