JVI Accepted Manuscript Posted Online 10 June 2015 J. Virol. doi:10.1128/JVI.00722-15 Copyright © 2015, American Society for Microbiology. All Rights Reserved.

1

Human Papillomavirus Infectious Entry and Trafficking is a Rapid Process

2 3 4

Justyna Broniarczyk 1,2, Paola Massimi 1, Martina Bergant 3, Lawrence Banks 1

5 6 7 8 9 10 11

1. International Centre for Genetic Engineering and Biotechnology, Padriciano 99, I-34149 Trieste, Italy 2. Department of Molecular Virology, Adam Mickiewicz University, Umultowska 89, 61614 Poznan, Poland 3. Centre for Biomedical Sciences and Engineering, University of Nova Gorica, Vipavska 13, 5000 Nova Gorica, Slovenia

12 13

Running Title: HPV trafficking

14 15 16 17

Corresponding author:

18

Lawrence Banks

19

International Centre for Genetic Engineering and Biotechnology,

20

Padriciano 99,

21

I-34149 Trieste, Italy

22

[email protected]

23

Word count for main abstract: 152

24

Word count main text: 2,201

25

1

26

Abstract

27 28

Previous studies have indicated that Human Papillomavirus (HPV) infectious entry is slow

29

requiring many hours after initial infection for the virus to gain entry into the nucleus.

30

However intracellular transport pathways are typically very rapid and in the context of a

31

natural HPV infection in a wounded epithelium, such slow intracellular transport would seem

32

at odds with a normal viral infection. Using synchronised cell populations we show that HPV

33

trafficking can however be a rapid process. In cells that are infected in the late S-early G2/M

34

phase of the cell cycle, HPV16 pseudovirion (PsV) reporter DNA gene expression is

35

detectable by 8hrs post-infection. Likewise reporter DNA can be visualised within the nucleus

36

in conjunction with PML nuclear bodies 1-2hrs post-infection in cells that are infected with

37

PsVs just prior to mitotic entry. This demonstrates that endosomal trafficking of HPV is

38

rapid, with mitosis being the main restriction on nuclear entry.

39 40

Importance

41

HPV infectious entry appears to be slow and requires mitosis to occur before the incoming

42

viral DNA can access the nucleus. In this study we show that HPV trafficking in the cell is

43

actually very rapid. This demonstrates that in the context of a normal virus infection, the cell

44

cycle state will have a major influence on the time it takes for an incoming virus to enter the

45

nucleus and initiate viral gene expression.

46 47 48 49 50 2

51

Introduction

52

Papillomaviruses (PV) are small, non-enveloped DNA viruses that infect epithelial cells.

53

Their icosahedral capsid is formed by two structural proteins: L1 and L2. PVs have oncogenic

54

potential and are associated with multiple human cancers including cervical cancer, other

55

anogenital cancers, and a significant number of head and neck tumours [1]. The HPV life

56

cycle is intimately linked to epithelial differentiation of keratinocytes, with the virus believed

57

to enter the undifferentiated proliferating basal compartment of the epithelium through

58

microtraumas in the skin. As the cells differentiate the viral genome is amplified and

59

ultimately new infectious virus particles are assembled in the nucleus of differentiated spinous

60

and granular keratinocytes [1].

61

PVs enter basal cells via endocytosis, but the precise mechanism appears diverse and is

62

dependent on the virus type [2, 3]. Virus capsids then disassemble in the late endosomes

63

and/or lysosomes in a pH-dependent manner. L2 interacts with components of the cellular

64

sorting machinery, such as sorting nexin 17, which is required for the lysosomal escape of the

65

L2-DNA complex [4]. L2 then targets the viral DNA to the perinuclear region of the cell

66

through a pathway that requires Dynein-mediated transport [5] and endocytic retromer

67

components [6], where the L2-DNA complex eventually accumulates in the trans-Golgi

68

network [7, 8]. HPV entry into the nucleus requires mitosis, during which the barrier between

69

nucleoplasm and cytosol is removed and the trans-Golgi network becomes dispersed, thereby

70

allowing the L2-DNA complex to gain access to the nucleus [9,10], where it is found in close

71

proximity with PML bodies in which viral genome expression is believed to initiate [11].

72

Whilst previous studies have shown that mitosis is an important element for completion of

73

infectious virus entry, expression of a HPV-16 Pseudovirion (PsV) reporter gene cannot be

74

detected earlier than 16h post-infection in asynchronously growing cells [3]. However,

75

classical endosomal cargo trafficking pathways are typically very quick [12], suggesting that 3

76

cells infected with HPVs when close to mitosis might allow nuclear entry and initiation of

77

viral gene expression at much earlier time points. To investigate this possibility we have

78

monitored HPV-16 PsV infection in cells following cell cycle synchronization. We now show

79

that intracellular transport of PsVs is remarkably quick, and depending upon the specific

80

phase of the cell cycle in which infection takes place, viral nuclear entry can occur within 1-

81

2h post infection and reporter gene expression can be detected as little as 8h post infection.

82

This demonstrates that the main restriction on nuclear entry of the L2-DNA complex and the

83

initiation of viral gene expression is completion of mitosis.

84 85

Materials and Methods

86

Cell lines

87

Spontaneously immortalized human keratinocyte cells (HaCaT) were maintained in

88

Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% fetal bovine serum

89

(FBS) penicillin-streptomycin (100U/ml) and glutamine (300mg/ml).

90 91

PsV production

92

HPV-16 PsVs containing a luciferase reporter plasmid were generated in 293TT cells as

93

previously described [4, 13]. PsVs containing packaged 5-ethynyl-2′-deoxyuridine (EdU)-

94

labeled plasmid DNA were prepared by addition of 25 μM EdU to the 293TT cells at 24h

95

post-transfection and harvested after a further 24h.

96 97

Cell synchronization and infection

98

HaCaT cells were seeded in a 12 well plate at a density of 0,75 x 105 cells/well (aphidicolin

99

treatment) or 0,5 x 105 cells/well (thymidine treatment). After adherence HaCaT cells were

100

incubated 24h with 1µg/ml aphidicolin (Sigma) to induce arrest at the G1/S boundary. The 4

101

cells were then released by washing with PBS and fresh DMEM was added. Cells were

102

infected with HPV16 PsVs (1h, 4°C) at a concentration of 12ng/ml at different times after the

103

release as indicated in the text. Cells were harvested at different time points (0h, 4h, 8h, 12h,

104

16h) and infection was monitored by luminometric analysis of firefly luciferase activity using

105

a Luciferase Assay System kit (Promega).

106

For synchronization experiments using double thymidine block, the cells were incubated with

107

2mM thymidine (Sigma) for 16h. The cells were then washed with PBS, and fresh DMEM

108

was added for a further 9h, after which 2mM thymidine (Sigma) was again added for 16h.

109

The cells were then released by washing with PBS and infected 3h post-release (S phase) with

110

HPV16 PsVs. 1h after infection at 4°C the cells were washed with PBS, fresh medium was

111

added and cells were incubated for further 8h. After that time cells were collected and

112

infection was monitored by luminometric analysis as above. Experiments were done in

113

triplicate and each sample was measured three times. Throughout, equal amounts of protein

114

was used in the assays which was ascertained by prior measurement of total protein

115

concentration in the different cell extracts.

116 117

Flow cytometry

118

Cell cycle analysis was done with FACS analysis by measuring DNA content using

119

propidium iodide staining as described previously [14].

120 121

PsV trafficking assay

122

HaCaT cells seeded on coverslips were grown overnight and synchronised using aphidicolin

123

block as described above. At different times after release from G1/S (0h, 10h) cells were

124

prechilled to 4°C, infected with EdU-labeled PsVs, and incubated for 1h at 4°C with agitation

125

to allow viral attachment. Cells were then washed, DMEM was replaced and cells were 5

126

transferred to 37°C. At different time points after attachment (1h, 3h), cells were washed with

127

PBS and fixed in PBS + 3.7 % paraformaldehyde for 15 min at room temperature. EdU and

128

PML staining were done as previously described using Click-iT EdU Imagining Kit

129

(Molecular Probes) and anti-PML primary antibodies (Santa Cruz, 1:100) [4]. Cells were

130

counterstained with DAPI, washed in water and mounted on glass slides. Slides were

131

visualized using a Zeiss Axiovert 100 M microscope (Zeiss) attached to a LSM 510 confocal

132

unit or a Leica DMLB fluorescence microscope equipped with a Leica photo camera

133

(A01M871016).

134 135

Neutralization assay

136

Neutralization assays were performed by incubation of PsVs with neutralising antibody

137

H16.V5 at a final concentration of 1:1000 or pre-immune antibody for 1h at 4°C. Neutralized

138

PsVs were then added to HaCaT cells and infectivity was measured at different time points as

139

described above. Mock infections were also performed as additional negative controls

140 141

Results and Discussion

142

Previous studies have shown that mitosis is needed to complete HPV infection and that the

143

expression of a PsV reporter gene cannot be detected earlier then 16h post infection in

144

asynchronously growing cells [3, 10-11]. However, intracellular transport mechanisms are

145

relatively fast [12], suggesting that cells infected with HPVs when close to mitosis might

146

allow detection of viral gene expression and nuclear entry at much earlier time points post

147

infection.

148

To first identify the earliest time point at which PsV reporter gene expression can be detected

149

in asynchronously growing HaCaT cells, HPV-16 PsVs carrying luciferase were used to

150

infect HaCaTs and cell extracts were collected at different times post-infection. Luciferase 6

151

activity was measured and the results in Figure 1A show the first detection of luciferase

152

activity at 16h post infection, increasing to a maximum at 26h post infection (Figure 1A).

153

These results are very much in agreement with previous studies [3] and indicate that in

154

asynchronous cultures infectious HPV entry and initiation of gene expression is a relatively

155

slow process.

156

We then proceeded to investigate whether reporter gene activity could be detected at earlier

157

time points post-infection, if cells were exposed to virions close to mitosis. HaCaT cells were

158

synchronized in G1/S with aphidicolin and were then released from the G1/S block. After 7h

159

the cells were infected with HPV16 PsVs (time 0h) and cells were harvested at different times

160

post-infection (0h, 4h, 8h, 12h, 14h and 16h) and analysed for luciferase activity and for the

161

corresponding cell cycle profiles. The results demonstrate that when cells are infected with

162

PsVs when the cells are predominantly in S phase (Figure 1C panel (II)) the luciferase activity

163

can first be detected by 8h post-infection (Figure 1B), which is significantly quicker than in

164

asynchronously infected cells. As can be seen, no luciferase activity could be detected at 4h

165

post infection, a time when the majority of the cells were still passing through mitosis (Figure

166

1C panel (III)), whilst at the 8h time point a significant proportion of the cell population had

167

passed back into the G1 phase of the cell cycle, consistent with completion of mitosis (Figure

168

1C panel (IV)).

169

To verify that the luciferase signal observed at 8h post-infection was a result of infectious

170

virus entry, the assay was repeated but the PsVs were pre-incubated with the anti-L1 PsV

171

neutralizing antibody H16.V5 [15]. As can be seen from Figure 1D, prior incubation of the

172

PsVs with H16.V5 antibody abolished the luciferase activity at the 8h time point. In order to

173

confirm these results using a different method of cell synchronization, the neutralization assay

174

was repeated and infections performed on HaCaT cells that had been synchronized following

175

a double thymidine block. The PsVs were incubated with H16.V5 antibody and used to infect 7

176

HaCaT cells 3h post-release from the thymidine block. Luciferase activity was then measured

177

again at the 8h time point post-infection, and as can be seen in Figure 2A there was readily

178

detectable levels of reporter gene activity at this time point, and this was abolished by prior

179

incubation of the PsVs with the neutralizing antibody. Consistent with the results in Figure 1,

180

PsV infection at 3h post-release from the thymidine block occurred whilst the majority of the

181

cell population was in S phase (Figure 2B panel (II)) and no luciferase activity was detectable

182

at 4h post infection (data not shown) when most of the cells were in G2M (Figure 2B panel

183

(III)). However luciferase was detected at 8h post infection when the majority of the cell

184

population had re-entered G1 (Figure 2B panel (IV)). These results demonstrate that HPV

185

PsV infectious entry and subsequent reporter gene expression is much quicker when cells are

186

infected during the late S or the early G2/M phase of the cell cycle. This is perfectly

187

consistent with previous reports suggesting that infectious entry into the nucleus requires

188

mitosis [9-10], but it emphasizes that the intracellular trafficking of the virus is much faster

189

than previously assumed.

190

These results indicate that PsV reporter DNA can gain access to the nucleus relatively quickly

191

post-infection, if infection occurs close to mitosis. To identify the minimum time required for

192

PsV DNA to gain access to the nucleus we used HPV16 PsVs containing reporter plasmid

193

DNA labelled with the thymidine analog, 5-ethynyl-2’deoxyuridine (EdU). HaCaT cells were

194

synchronized with aphidicolin as above, then released from the G1/S block and infected with

195

PsVs either immediately, when the cells were in G1 (Figure 1C panel (I) or after 10h when

196

the cells were still passing through G2M (Figure 1C panel (III)). The infected cells were fixed

197

at 1h post infection and EdU was detected in an azide–alkyne reaction. Cells were then

198

stained for PML, a major component of ND10 domains, where HPV genomes are typically

199

found in an established HPV infection [11]. As can be seen from Figure 3A EdU-labelled

200

DNA encapsidated by HPV-16 PsVs was detected only in the cytoplasm in cells infected 8

201

immediately after release from G1 and fixed 1h post-infection. In contrast, in cells that were

202

infected 10h post-release from G1 and fixed 1h post-infection, EdU staining was readily

203

detected within the nucleus and colocalision with PML could be observed (Figure 3B).

204

Similar results were also obtained when cells were analysed by confocal microscopy. As can

205

be seen from Figure 4A there is no co-localsiation of EdU with PML when cells are infected

206

immediately after release and fixed 1h post infection, whilst in cells infected 10h post –

207

release and analysed 1h post-infection there is clear co-localisation of PsV reporter DNA with

208

PML, as shown from the zeta-stack analysis (Figure 4B).

209

mitosis, PML bodies become fewer and larger [15], and in several instances, EdU labeled PsV

210

reporter DNA shows clear co-localisation with PML at such sites, using both epifluorescence

211

(Figure 3B) and confocal microscopy with zeta-stacks (Figure 5). Taken together these results

212

demonstrate that HPV PsV reporter DNA can gain access to the nucleus as soon as 1-2hrs

213

post infection if the infected cells are close to mitotic entry, and that association with PML

214

can also occur at this time.

215

Whilst previous studies have shown a requirement for mitosis for the L2-DNA complex to

216

gain entry into the nucleus, there has been little information on the speed with which HPV

217

virions and their packaged genomes are trafficked within the cell. We provide compelling

218

evidence that reporter gene expression can be detected as little as 8h post-infection if cells are

219

infected in late S phase. Likewise, PsV reporter DNA can be found associated with PML

220

nuclear bodies as little as 1-2h post-infection and, most strikingly, this can also be clearly

221

seen in cells which are undergoing mitosis and where nuclear envelope breakdown has

222

already occurred, suggesting that the association of the L2-DNA complex with PML nuclear

223

bodies first appears during mitosis. Current studies are focused on identifying the cellular

224

components that are required for recruiting the L2-DNA complex to these nuclear domains.

225

Acknowledgements

In cells actually undergoing

9

226

The authors are very grateful to Miranda Thomas for valuable comments on the manuscript.

227

Justyna Broniarczyk is a recipient of an Arturo Falaschi ICGEB Fellowship.

228 229

Statement of author contributions

230

Justyna Broniarczyk and Paola Massimi performed all the experiments and Martina Bergant

231

and Lawrence Banks helped with the study design and manuscript preparation.

232

10

233 234

References

235

1. Doorbar J, Quint W, Banks L, Bravo IG, Stoler M, Broker TR, Stanley MA. 2012.

236

The biology and life-cycle of human papillomaviruses. Vaccine Suppl 5: F55-70.

237

2. Schelhaas M, Shah B, Holzer M, Blattmann P, Kühling L, Day PM, Schiller JT,

238

Helenius A. 2012. Entry of human papillomavirus type 16 by actin-dependent, clathrin-

239

and lipid raft-independent endocytosis. PLoS Pathog 8(4):e1002657.

240

3. Spoden G, Kühling L, Cordes N, Frenzel B, Sapp M, Boller K, Florin L, Schelhaas

241

M.

242

requirements for entry. J Virol 87(13):7765-73.

243 244

2013. Human papillomavirus types 16, 18, and 31 share similar endocytic

4. Bergant M, Banks L. 2013. SNX17 facilitates infection with diverse papillomavirus types. J Virol 87(2):1270-3.

245

5. Florin L, Becker KA, Lambert C, Nowak T, Sapp C, Strand D, Streeck RE, Sapp M.

246

2006. Identification of a dynein interacting domain in the papillomavirus minor capsid

247

protein L2. J Virol 80(13):6691-6.

248

6. Popa A, Zhang W, Harrison MS, Goodner K, Kazakov T, Goodwin EC, Lipovsky A,

249

Burd CG, DiMaio D. 2015. Direct binding of retromer to human papillomavirus type 16

250

minor capsid protein L2 mediates endosome exit during viral infection. PLoS Pathog

251

11(2):e1004699.

252

7. Day PM, Thompson CD, Schowalter RM, Lowy DR, Schiller JT. 2013. Identification

253

of a role for the trans-Golgi network in human papillomavirus 16 pseudovirus infection. J

254

Virol 87(7):3862-70.

255

8. Zhang W, Kazakov T, Popa A, DiMaio D. 2014. Vesicular trafficking of incoming

256

human papillomavirus 16 to the Golgi apparatus and endoplasmic reticulum requires γ-

257

secretase activity. MBio 5(5):e01777-14. 11

258

9. Aydin I, Weber S, Snijder B, Samperio Ventayol P, Kühbacher A, Becker M, Day

259

PM, Schiller JT, Kann M, Pelkmans L, Helenius A, Schelhaas M. 2014. Large scale

260

RNAi reveals the requirement of nuclear envelope breakdown for nuclear import of

261

human papillomaviruses. PLoS Pathog 10(5):e1004162.

262

10. Pyeon D, Pearce SM, Lank SM, Ahlquist P, Lambert PF. 2009. Establishment of

263

human papillomavirus infection requires cell cycle progression. PLOS Pathog 5(2):

264

e1000318.

265

11. Day PM, Baker CC, Lowy DR, Schiller JT. 2004. Establishment of papillomavirus

266

infection is enhanced by promyelocytic leukemia protein (PML) expression. Proc Natl

267

Acad Sci U S A 101(39):14252-7.

268

12. Huotari J, Helenius A. 2011. Endosome maturation. EMBO J 30(17):3481-500.

269

13. Buck CB, Pastrana DV, Lowy DR, Schiller JT.

2005 Generation of HPV

270

pseudovirions using transfection and their use in neutralization assays. Methods Mol Med

271

119: 445–462.

272 273

14. Banks L, Barnett SC, Crook T. 1990. HPV-16 E7 functions at the G1 to S phase transition in the cell cycle. Oncogene 5:833–7.

274

15. Christensen ND, Dillner J, Eklund C, Carter JJ, Wipf GC, Reed CA, Cladel NM,

275

Galloway DA. 1996. Surface conformational and linear epitopes on HPV-16 and HPV-

276

18L1 virus-like particles as defined by monoclonal antibodies. Virology 223:174–184.

277

16. Bernardi R, Pandolfi PP. 2007. Structure, dynamics and functions of promyelocytic

278

leukaemia nuclear bodies. Nat Rev Mol Cell Biol 8(12):1006-16.

279 280

12

281

Figure Legends

282 283

Figure 1. Comparative rate of HPV16 PsVs infectious entry in asynchronous and

284

synchronous HaCAT cells. Panel A. Asynchronously growing HaCaT cells were infected

285

with HPV-16 PsVs carrying a luciferase reporter plasmid and were harvested at different

286

time points post-infection (0h, 4h, 8h, 13h,16h, 19h, 22h, 26, 48h) and the luciferase activity

287

was measured. The results are expressed as the means of at least three independent

288

experiments and the standard deviations are shown. Panel B. Aphidicolin synchronised

289

HaCaT cells were infected with HPV16 PsVs carrying a luciferase reporter plasmid 7h post-

290

release from G1/S (time point 0h). Cells were harvested at different time points post-infection

291

(0h, 4h, 8h, 12h, 16h) and luciferase activity measured. The results are expressed as the means

292

of at least three independent experiments and the standard deviations are shown. Panel C. Cell

293

cycle analysis of the cells harvested in Panel B stained with propidium iodide and analyzed by

294

flow cytometry. Note that the cells were mostly arrested in G1 following aphidicolin

295

treatment (panel I), at the time of infection 7h post-release they were in S phase (panel II), and

296

had entered mitosis by 4h post-infection (panel III). A siginficant proportion of cells had re-

297

entered G1 at the 8h time point (panel IV) when luciferase activity was first detected. Panel

298

D. HPV16 PsV infections were performed as in Panel B except that PsVs were pre-incubated

299

with H16.V5 neutralising antibody or pre-immune (PI) antibody prior to infection and

300

luciferase activity measured 8h post-infection. The results are expressed as the means of at

301

least three independent experiments and the standard deviations are shown. Also shown are

302

mock infected cells.

303 304 305 13

306

Figure 2. HPV16 PsV reporter gene expression can be detected 8h post-infection.

307

Panel A. Double thymidine block synchronised HaCaT cells were infected with HPV16 PsVs

308

carrying a luciferase reporter plasmid 3h post-release from G1 arrest using PsVs that were

309

either pre-incubated with H16.V5 neutralising antibody or pre-immune (PI) antibody. After

310

8h the cells were harvested and luciferase activity measured. The results are expressed as the

311

means of at least three independent experiments and the standard deviations are shown and a

312

mock infection control was also included. Panel B. Cell cycle analysis of the HaCaT cells

313

used in Panel A. Note growth arrest in G1 following the double thymidine block (panel I),

314

entry into S phase 3h post-release (panel II) at the time of infection (time 0h), entry into G2M

315

4h post-infection (panel III) and re-entry into G1 8h post-infection (panel IV) at the time of

316

detection of luciferase activity in Panel A.

317 318

Figure 3. Detection of HPV16 PsV reporter DNA in proximity with PML 1h post-

319

infection. Aphidicolin synchronised HaCaT cells were infected with HPV16 PsVs carrying

320

EdU labelled luciferase reporter plasmid 1h (Panel A) or 10h (Panel B) post-release from G1/

321

S. After a further 1h at 37°C the cells were fixed and processed for the detection of EdU-

322

labelled DNA (red), PML (in green) and DAPI (blue). Note appearence of perinuclear

323

accumulation of EdU labelled DNA in Panel A but co-localisation with nuclear PML in Panel

324

B as indicated by the arrows and the expanded field.

325 326

Figure 4. Confocal image analysis showing HPV16 PsVs reporter DNA in conjunction

327

with PML. Aphidicolin synchronised HaCaT cells were infected with HPV16 PsVs carrying

328

EdU labelled luciferase reporter plasmid 1h (Panel A) or 10h (Panel B) post-release from G1/

329

S. After a further 1h at 37°C the cells were fixed and processed for the detection of EdU-

330

labelled DNA (red) and

PML (in green). Shown are the zeta-stacks demonstrating 14

331

cytoplasmic PsV EdU labelled DNA in Panel A and co-localisation with PML in two different

332

fields (upper and lower panels) in Panel B, with an expanded view of one of those fields.

333 334

Figure 5. Confocal image analysis showing HPV16 PsVs reporter DNA in conjunction

335

with PML in mitotic cells. Aphidicolin synchronised HaCaT cells were infected with HPV16

336

PsVs carrying EdU labelled luciferase reporter plasmid 10h post-release from G1/S. After a

337

further 3h at 37°C the cells were fixed and processed for the detection of EdU-labelled DNA

338

(red) and PML (in green). Shown are the zeta-stacks demonstrating PsV EdU labelled DNA

339

and co-localsiation with PML in two different fields (upper and lower panel) in cells that are

340

undergoing mitosis.

15

Human Papillomavirus Infectious Entry and Trafficking Is a Rapid Process.

Previous studies have indicated that human papillomavirus (HPV) infectious entry is slow, requiring many hours after initial infection for the virus t...
793KB Sizes 2 Downloads 8 Views