JVI Accepted Manuscript Posted Online 27 January 2016 J. Virol. doi:10.1128/JVI.02704-15 Copyright © 2016, American Society for Microbiology. All Rights Reserved.

1

ESCRT-0 Component Hrs Promotes Macropinocytosis of Kaposi’s Sarcoma-

2

Associated Herpesvirus in Human Dermal Microvascular Endothelial Cells

3 4

Mohanan Valiya Veettil#, Binod Kumar, Mairaj Ahmed Ansari, Dipanjan Dutta, Jawed

5

Iqbal, Olsi Gjyshi, Virginie Bottero and Bala Chandran

6 7

H. M. Bligh Cancer Research Laboratories, Department of Microbiology and

8

Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and

9

Science, North Chicago, Illinois 60064, USA.

10 11

Running title: Role of Hrs in macropinocytosis of KSHV

12 13

# Corresponding author.

14

Mailing Address:

15

Department of Microbiology and Immunology, Chicago Medical School,

16

Rosalind Franklin University of Medicine and Science,

17

3333 Green Bay Road, North Chicago, IL 60064.

18

Phone (847) 578-8700x7716; Fax (847) 578-3349.

19

E-mail: [email protected]

20 21 22 1

23

ABSTRACT

24

Kaposi’s sarcoma-associated herpesvirus (KSHV) enters human dermal microvascular

25

endothelial cells (HMVEC-d), its natural in vivo target cells, by lipid raft dependent

26

macropinocytosis. The internalized viral envelope fuses with the macropinocytic membrane and

27

released capsid is transported to the nuclear vicinity resulting in the nuclear entry of viral DNA.

28

The Endosomal Sorting Complexes Required for Transport (ESCRT) proteins which include

29

ESCRT-0, -I, -II, and -III play a central role in endosomal trafficking and sorting of internalized

30

and ubiquitinated receptors. Here, we examined the role of ESCRT-0 component Hrs

31

(Hepatocyte growth factor regulated tyrosine kinase substrate) in KSHV entry into HMVEC-d

32

cells by macropinocytosis. Knockdown of Hrs by shRNA transduction results in significant

33

decrease in KSHV entry and viral gene expression. Immunofluorescence analysis (IFA) and

34

plasma membrane isolation and proximity ligation assay (PLA) demonstrate the translocation of

35

Hrs from the cytosol to the plasma membranes of infected cells and association with α-actinin-4.

36

In addition, infection induces the plasma membrane translocation and activation of the

37

serine/threonine kinase ROCK1, a downstream target of the RhoA GTPase. Hrs knockdown

38

reduces these associations suggesting that the recruitment of ROCK1 is an Hrs mediated event.

39

Interaction between Hrs and ROCK1 is essential for the ROCK1 induced phosphorylation of

40

NHE1 (Na+/H+ exchanger 1) involved in the regulation of intracellular pH. Thus, our studies

41

demonstrate the plasma membrane association of ESCRT protein Hrs during macropinocytosis

42

and suggest that KSHV entry requires both Hrs and ROCK1 dependent mechanisms, and

43

ROCK1 mediated phosphorylation of NHE1 and pH change is an essential event required for the

44

macropinocytosis of KSHV.

45

2

46

IMPORTANCE

47

Macropinocytosis is the major entry pathway of KSHV in human dermal microvascular

48

endothelial cells, the natural target cells of KSHV. Although the role of ESCRT protein Hrs has

49

been extensively studied with respect to endosomal movement and sorting of ubiquitinated

50

proteins into lysosomes, its function in macropinocytosis is not known. In the present study, we

51

demonstrate for the first time that upon KSHV infection the endogenous Hrs localizes to the

52

plasma membrane and the membrane associated Hrs facilitates assembly of signaling molecules,

53

macropinocytosis and virus entry. Hrs recruits ROCK1 to the membrane which is required for

54

the activation of NHE1 and an increase in submembraneous intracellular pH occurring during

55

macropinocytosis. These studies demonstrate that the localization of Hrs from the cytosol to the

56

plasma membrane is important for coupling membrane dynamics to the cytosolic signaling

57

events during macropinocytosis of KSHV.

58 59 60 61 62 63 64 65 66 67 68 3

69

INTRODUCTION

70

KSHV entry into its in vitro adherent target cells is a multistage process which involves

71

binding of viral glycoproteins to cell surface heparan sulfate receptors, followed by interaction

72

with specific entry receptors, induction of cell signaling pathways and endocytosis. KSHV

73

exploits multiple host cell surface receptors including integrins-α3β1, αVβ3, αVβ5, α9β1, and

74

non-integrins CD98/xCT, and EphA2 to enter the adherent target cells such as HMVEC-d

75

(Human dermal microvascular endothelial) and HFF (Human foreskin fibroblasts) cells (1-6).

76

The interaction of KSHV with its specific entry receptors leads to the formation of a

77

multimolecular receptor complex consisting of integrins, xCT and EphA2 (2, 4, 7). Receptor

78

engagement and multimolecular receptor complex formation results in auto phosphorylation of

79

FAK, and the activation of Src, PI3-K and Rho GTPases, and all these molecules are targeted to

80

specific entry sites on the plasma membrane (8-10). Our previous studies have demonstrated that

81

the signal transduction pathways induced by KSHV and the consequent activation of their

82

downstream molecules plays a central role in coordinating the actin dynamics and the membrane

83

protein assembly required for the successful entry of the virus into the cytoplasm (8-10). The

84

endocytosed viral particles are then transported towards the nuclear periphery along the

85

microtubules by utilizing dynein motor proteins to deliver its DNA content into the nucleus (11).

86

KSHV utilizes different endocytic pathways to enter different cell types (12-17). In HFF

87

cells, primary B cells, and 293 cells, clathrin dependent endocytosis is the predominant pathway

88

of entry (12-14), whereas in HMVEC-d cells entry occurs by bleb associated macropinocytosis

89

(15, 16, 18). Bleb associated macropinocytosis begins with a remarkable set of events including

90

the formation of blebs, actomyosin contraction, bleb retraction, macropinosome formation and

91

eventually virus entry (18). Our studies have established that the adaptor protein c-Cbl and its

4

92

interaction with myosin IIA light chain (MLC) plays a significant role in blebbing and that

93

myosin IIA is required for both actomyosin contraction and retraction of the bleb (18). Our

94

subsequent studies proved that c-Cbl is also required for both translocation of the receptors into

95

the lipid raft and ubiquitination of the α3β1 and αVβ3 receptors which are critical determinants

96

of the macropinocytic entry, trafficking, and productive infection of KSHV (19).

97

Ubiquitination of receptors and the adaptor proteins such as c-Cbl serves to facilitate the

98

endocytosis of receptors and their sorting from membrane to lysosomes and subsequent

99

degradation (20, 21). In mammalian cells, the Endosomal Sorting Complexes Required for

100

Transport (ESCRT) helps in sorting ubiquitinated proteins and their delivery into lysosomes. The

101

ESCRT machinery consists a family of four complexes, ESCRT-0, -I, -II, and –III, which

102

sequentially assemble on the endosomal membrane. The ESCRT-0 component Hrs (Hepatocyte

103

growth factor (HGF)-regulated tyrosine kinase substrate) acts upstream to ESCRT complexes

104

and can recruit ESCRT-I to the endosomal membranes. This complex subsequently recruits

105

ESCRT-II and ESCRT-III complexes, two important complexes required for the sorting of

106

ubiquitinated proteins into late endosomes (22-25).

107

Hrs protein contains several domains: an N-terminal VHS-domain (Vps27p, Hrs, STAM),

108

FYVE domain, an ubiquitin interacting motif, a proline-rich domain, two coiled-coil domains,

109

and a C-terminal clathrin binding domain (26). The functional role of the different domains of

110

Hrs includes protein-protein interaction, cellular localization, cargo sorting, and endosome

111

motility (24, 25, 27, 28). Although the function of Hrs in intracellular trafficking and signal

112

transduction is well established, its role in macropinocytosis remains unclear. In the present

113

study, we provide evidence that Hrs is recruited to the plasma membrane early during KSHV

114

infection which is required for the virus entry by macropinocytosis. Interestingly, we found that

5

115

Hrs interacts with the Rho-associated protein kinase ROCK1, which in turn activates MLC and

116

the intracellular pH-regulating protein NHE1 to promote actomyosin contraction and

117

intracellular pH changes required for macropinocytosis.

118 119 120 121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 136 137

6

138

MATERIALS AND METHODS

139

Cells and virus: Primary human dermal microvascular enodthelial cells (HMVEC-d CC-2543;

140

Clonetics, Walkersville, MD) were grown in EBM2 medium with growth factors (Cambrex,

141

Walkersville, MD). BCBL-1 cell culture, supernatant collection after TPA induction, and virus

142

purification procedures were performed as described previously (1). The isolation of KSHV

143

DNA from the virus, and the determination of viral copy numbers by real-time DNA PCR using

144

KSHV ORF73 gene-specific primers were performed as previously described (29). All infections

145

were carried out with 20 DNA copies/cell (multiplicity of infection-MOI) of KSHV, unless

146

stated otherwise.

147

Antibodies and reagents: Antibodies and reagents used for this study include: Anti-ROCK1

148

rabbit monoclonal antibody, anti-phospho MLC rabbit polyclonal antibody, anti-Na, K-ATPase

149

rabbit polyclonal antibody (Cell Signaling Technology); anti-Hrs rabbit polyclonal antibody

150

(Sigma); anti-NHE1 mouse monoclonal antibody (Santa Cruz); anti-calnexin rabbit monoclonal

151

antibody, mouse anti-phospho serine antibody (Abcam); anti-α-actinin-4 rabbit monoclonal

152

antibody (Millipore); rabbit anti-gB and mouse anti-gpK8.1A antibodies were created in Dr.

153

Chandran’s laboratory (30, 31); anti-rabbit and anti-mouse antibodies linked to horseradish

154

peroxidase (KPL Inc., Gaithersburg, Md.); Texas Red conjugated dextran, Alexa 594 conjugated

155

phalloidin and anti-rabbit and anti-mouse secondary antibodies conjugated to Alexa 488 or Alexa

156

594 (Invitrogen); protein A and G–Sepharose CL-4B beads (Amersham Pharmacia Biotech,

157

Piscataway, NJ).

158

Hrs shRNA knockdown: For shRNA knockdown of Hrs, five sets of bacterial glycerol stock

159

Hrs shRNA plasmid vectors (TRCN00000037894, -37895, -37896, -37897, and -37898) were

160

purchased from Thermoscientific. The Hrs shRNA lentiviral plasmids were purified from the

7

161

bacterial cultures (Invitrogen) and the lentivial particles were produced by transfection with a

162

four-plasmid system, as previously described (32). Briefly, HEK 293T cells were transiently

163

transfected with shRNA lentiviral constructs and the plasmid packaging system (Gag-Pol, Rev

164

and VSV-G), the supernatants were collected, and filtered. The lentiviral particles produced from

165

each plasmid vector were tested for knockdown efficiency by Western blot analysis. We found

166

that TRCN00000037897 and TRCN00000037898 had the highest knockdown efficiency among

167

the five shRNAs. Therefore, we used Hrs shRNAs created with these two plasmids (Hrs-

168

shRNA1 and Hrs-shRNA2) for the experiments in this manuscript. We used Hrs shRNA1 for all

169

experiments except figure 1A and 1B where we used both Hrs shRNA1 and 2. For negative

170

control, a nontargeting shRNA lentiviral pool was used.

171

Western blotting: Cells lysates were prepared in RIPA lysis buffer (15 mM NaCl, 1 mM

172

MgCl2, 1 mM MnCl2, 2 mM CaCl2, 2 mM phenylmethylsulfonyl fluoride, and protease

173

inhibitor mixture (Sigma). The lysates were centrifuged at 12,000 rpm for 15 min at 4°C and the

174

protein concentration was measured with BCA reagent. Equal amounts of proteins from each

175

sample were separated on SDS-PAGE and transferred to nitrocellulose membranes. Membranes

176

were probed with the indicated primary antibodies and detected by incubation with species-

177

specific HRP-conjugated secondary antibodies. Immunoreactive protein bands were visualized

178

and imaged by enhanced chemiluminescent HRP substrate kit (Pierce, Rockford, IL). The bands

179

were scanned and densitometric quantification was performed using the FluorChem FC2 and

180

Alpha-Imager Systems (Alpha Innotech Corporation, San Leonardo, CA).

181

Immunoprecipitation: Cell lysates were immunoprecipitated for 2h with appropriate antibodies

182

and the immune complexes were captured by protein A or G-Sepharose. The immune complexes

183

were then eluted in sample buffer for SDS-PAGE and run on 10% gels and transferred to

8

184

membranes. The membranes were tested by Western blot with specific primary and secondary

185

antibodies.

186

Immunofluorescence microscopy: Cells grown in 8-well chamber slides were fixed with 4%

187

paraformaldehyde after infection and permeabilized with 0.2% Triton X-100. The cells were

188

blocked with Image-iT FX signal enhancer (Invitrogen) for 20 min, and then incubated with

189

primary antibodies against the specific proteins and subsequently stained with secondary

190

antibodies conjugated to Alexa 488 or 594. The stained cells were mounted in mounting medium

191

with DAPI (4′,6′-diamidino-2-phenylindole) (Invitrogen) and visualized with a Nikon 80i

192

fluorescent microscope equipped with Metamorph digital imaging software.

193

Proximity Ligation Assay: Proximity Ligation Assay (PLA) was performed using the Duolink

194

in situ detection kit (Sigma) as per the manufacturer’s instructions (33, 34). Briefly, uninfected

195

and infected cells fixed with 4% paraformaldehyde solution for 15 min, permeabilized with

196

Triton X- 100 for 5 min and washed with PBS, and incubated for 1h with the Duolink blocking

197

buffer. The cells were then incubated with pairs of primary antibodies in Duolink antibody

198

diluent solution and then labeled with Duolink PLA PLUS and MINUS probes for 1h at 37°C.

199

This was followed by hybridization, ligation, amplification, and detection using a red fluorescent

200

probe. The red fluorescent fluorophore-tagged oligonucleotides were visualized using a Nikon

201

Eclipse 80i microscope equipped with Metamorph digital imaging software.

202

Determination of KSHV entry by real-time DNA PCR: Target cells were infected with 20

203

MOI of KSHV at 37°C for 2h. After infection, the cells were washed with HBSS, and treated

204

with 0.25% trypsin-EDTA for 5 min at 37°C to remove the bound but non-internalized virus.

205

Total DNA was isolated from the uninfected and infected cells using DNeasy kit (QIAGEN,

206

Valencia, CA) and real time DNA PCR was carried out using primer sets described previously

9

207

(29). To calculate the percent inhibition of KSHV entry, internalized KSHV DNA was

208

quantitated by amplification of the ORF73 gene by real-time DNA PCR. The KSHV ORF73

209

gene cloned in the pGEM-T vector (Promega) was used for the external standard.

210

Determination of KSHV gene expression by real-time RT-PCR: Isolation of total RNA from

211

cells, reverse transcription, and qPCR analysis were carried out using primer sets described

212

previously. Briefly, total RNA was isolated from the lysate using RNeasy kit (QIAGEN),

213

quantified, and the ORF73 expression was detected by real-time reverse transcription PCR using

214

specific

215

dehydrogenase) gene expression was used to normalize the expression levels of ORF73.

216

Isolation of membrane fraction: Subcellular fractionation and cell membrane preparation were

217

performed as described previously (18, 35). Briefly, cells were homogenized with a Dounce-

218

homogenizer in homogenization buffer (250 mM sucrose, 20 mM HEPES, 10 mM KCl, 1 mM

219

EDTA, 1 mM EGTA and protease inhibitors). The homogenate was centrifuged at 3,000 rpm for

220

5 min, and the post-nuclear supernatant was centrifuged at 8,000 rpm for 5 min at 4ºC. The

221

supernatant was again centrifuged at 40,000 rpm for 1h at 4ºC, and the cytosolic (supernatant)

222

and membrane (pellet) fractions were separated. The membrane fractions were lysed in RIPA

223

buffer and used for Western blot.

224

Analysis of dextran uptake: For the dextran uptake study, HMVEC-d cells were incubated with

225

Texas Red labeled dextran (40 kDa, 0.5 mg/ml; Invitrogen) and KSHV for 30 min. The cells

226

were then washed twice in HBSS, fixed with 4% paraformaldehyde for 15 min, and washed three

227

times with PBS. Internalized dextran was observed under immunofluorescence microscope.

primers

and

TaqMan

probes

(29).

GAPDH

(glyceraldehyde-3-phosphate

228

To perform quantitative analysis of dextran uptake, the dextran positive cells were counted

229

under immunofluorescence microscope. At least 10 different microscopic fields of 25-30 cells

10

230

each were counted for each experiment and the results displayed as percentage of dextran

231

positive cells.

232

Intracellular pH measurement: An equal number of cells in 96 well plates were loaded with

233

BCECF-AM (bis-(2-carboxy-ethyl)-5-(and-6)-carboxyfluorescein, acetoxy-methyl, 10 μM) in

234

HBSS for 30 min. The cells were washed with HBSS to remove the free dye, and then infected

235

with KSHV for 2, 5, and 10 min at 37°C. Immediately after infection, BCECF fluorescence

236

excited with a 485/20 filter and the emission was measured with a 528/20 filter on a BioTek

237

Synergy2 HT microplate reader. The average readings for each time point were calculated and

238

the changes in pH expressed as relative fluorescent units.

239

Statistical analyses.

240

Statistical significance was calculated using a two tailed Student’s t-test. P < 0.05 was

241

considered significant.

242 243 244 245 246 247 248 249 250 251 252

11

253

RESULTS

254

Hrs knockdown inhibits KSHV entry and gene expression in HMVEC-d cells.

255

To explore the role of Hrs in KSHV infection, we performed Hrs knockdown in

256

HMVEC-d cells using two different lentiviral Hrs shRNAs (Hrs shRNA1 and 2). HMVEC-d

257

cells were transduced with control and Hrs shRNAs and the efficiency of knockdown was

258

determined 48h after transduction by Western blotting with anti-Hrs antibody. As shown in

259

figure 1A, both the Hrs shRNA1 and Hrs shRNA2 completely knockdown endogenous Hrs

260

expression. To address whether Hrs inhibition affects the entry of KSHV, control and Hrs

261

shRNA transduced HMVEC-d cells were infected with KSHV for 2h, DNA was isolated and the

262

internalized viral DNA copy numbers were determined by measuring viral ORF73 DNA copy

263

numbers by real-time DNA PCR. Compared to the control shRNA transduced cells (copy

264

number 18009 ± 938), the entry of KSHV was significantly reduced (~65-70 %) in both Hrs

265

shRNA1 (4427 ± 49) and Hrs shRNA2 (5330± 214) knockdown cells. Heparin-treated virus,

266

which cannot bind and enter the cells, was used as a control (Fig. 1B). We next investigated

267

whether Hrs knockdown also results in decreased viral gene expression. Control and Hrs shRNA

268

transduced HMVEC-d cells were infected with KSHV for 24h, and viral gene expression of the

269

latency associated ORF73 gene was determined by real-time RT-PCR analysis. Similar to the

270

entry results, the expression of ORF73 gene in both Hrs shRNA1 (copy number 7742 ± 2180)

271

and shRNA2 (8364 ± 963) was reduced compared to control cells (41135 ± 680). Hrs shRNA

272

cells showed 70–80% inhibition of ORF73 gene expression (Fig. 1C), suggesting that the

273

decreased viral entry results in decreased gene expression in Hrs knockdown cells.

274

Since macropinocytosis is the major pathway of KSHV entry leading to a productive

275

infection in HMVEC-d cells (16, 18), we asked whether the decreased entry in Hrs shRNA cells

12

276

is correlated with macropinocytosis. The macropinocytosis of KSHV in the control and Hrs

277

shRNA cells was assessed by incubating the cells with the macropinocytosis marker dextran

278

(Texas Red labeled) in the presence or absence of virus for 30 min. Compared to the uninfected

279

cells, KSHV infection resulted in a considerable increase in dextran uptake in the control shRNA

280

transduced cells (Fig. 1D, upper and middle panel). However, Hrs shRNA cells incubated with

281

dextran and KSHV showed a substantial decrease in dextran uptake compared to the control

282

shRNA cells (Fig. 1D, middle and lower panel). Quantitative analysis performed by counting the

283

number of dextran positive cells in the control and Hrs shRNA cells confirmed the decreased

284

dextran uptake in Hrs shRNA cells (Fig. 1E). This result suggested that Hrs plays a role in

285

macropinocytosis mediated entry of KSHV into endothelial cells.

286

In addition to macropinocytic mode of entry in endothelial cells, KSHV is known to enter

287

by clathrin mediated endocytosis in other target cells such as HFF, HEK293 and B-cells. To

288

assess whether Hrs is involved in clathrin mediated endocytosis of KSHV, we examined the

289

effect of Hrs knockdown on the entry of KSHV in HFF cells (Fig.1.F). HFF cells transduced

290

with control or lentiviral Hrs shRNA were infected with KSHV for 2h, and the entry of KSHV

291

was determined by measuring viral ORF73 DNA copy numbers by real-time DNA PCR.

292

Interestingly, real-time PCR analysis demonstrated that Hrs knockdown had no significant effect

293

on the entry of virus in HFF cells (Fig.1.F). These results suggest that Hrs plays a specific role in

294

regulating macropinocytosis-mediated entry of KSHV, but not in clathrin mediated entry.

295

Hrs localizes to the plasma membrane of KSHV infected cells.

296

To ascertain the mechanism of Hrs function in the entry process of KSHV, we next

297

analyzed the cellular localization of endogenous Hrs protein in the infected cells by

298

immunofluorescent labeling. HMVEC-d cells were infected with KSHV for 5, 10 and 30 min

13

299

and then stained for the KSHV envelope glycoprotein gB and host Hrs protein. As shown in

300

figure 2A, Hrs was distributed throughout the cytoplasm in the uninfected cells. In contrast, at 5

301

and 10 min post-KSHV infection, Hrs localized predominantly in the plasma membranes (Fig.

302

2A, arrow heads), and the KSHV gB staining (Fig. 2A, block arrows) was also observed in

303

similar regions of the infected cell membrane. After 10 and 30 min of infection, in addition to

304

membrane staining, we observed cytoplasmic staining as well as colocalization of Hrs with

305

KSHV-gB in the infected cells (Fig. 2A, yellow spots).

306

To confirm the membrane localization of Hrs in the infected cells, plasma membranes

307

were isolated from uninfected and infected cells and tested for the presence of Hrs using a

308

specific anti-Hrs antibody. As shown in figure 2B, first panel, lanes 1-5, detection of Hrs in the

309

plasma membrane fractions confirmed the membrane localization with a pattern similar to that of

310

the IFA results in figure 2A. However, in the cells infected with heparin treated virus in which

311

KSHV cannot bind and enter the cells (36), we did not observe the induction of membrane

312

translocation (Fig. 2B, lane 6). This indicates that KSHV binding and interaction with receptors

313

are essential for the translocation of Hrs to the membrane. The purity of the membrane fraction

314

was assessed by the detection of membrane positive marker Na, K-ATPase in the purified

315

membrane fractions (Fig. 2B, second panel). Additionally, the absence of ER membrane marker

316

calnexin by Western blot indicated that the fraction was not contaminated with the internal

317

membrane proteins (Fig. 2B, third panel). Western blot of whole cell lysate using Hrs antibody

318

showed no variation in the total Hrs protein level (Fig. 2B, fourth panel). The recruitment of Hrs

319

to the plasma membrane of the infected cells indicated that Hrs may be involved in the initial

320

stages of macropinosome formation and cytoplasmic entry of the virus.

14

321

To further evaluate whether Hrs affects the cytoplasmic entry of KSHV, we examined the

322

localization of viral particles in control and Hrs shRNA cells. Cells infected with KSHV for 30

323

min were stained with rhodamine-phalloidin for filamentous actin and KSHV-gB to visualize the

324

localization of KSHV. In control cells infected with KSHV, virus particles were detected at the

325

nuclear periphery by 30 min post-infection, whereas in Hrs shRNA cells the virus particles failed

326

to enter the cells and were found stuck at the cell surface (Fig. 2C). This result indicated that

327

there is a significant correlation between the membrane recruitment of Hrs and the early stages

328

of KSHV entry by macropinocytosis.

329

Hrs localizes to the membrane by interacting with α-actinin-4 in the infected cells.

330

Macropinocytosis is an actin driven process that depends on actin polymerization and

331

membrane ruffling (37). As membrane associated Hrs affects the macropinocytosis of KSHV, we

332

rationalized that membrane localization may be achieved through the interaction of Hrs with an

333

actin binding protein that is localized to the plasma membrane. It has been shown that Hrs is a

334

binding partner of α-actinin-4 (38), a cytoskeleton scaffolding protein that transiently associates

335

with macropinosomes (39). Moreover, we have reported before that α-actinin-4 formed a

336

complex with the membrane associated proteins EphA2, myosin IIA, and CIB1 in KSHV

337

infected cells (15). Since α-actinin-4 primarily cross-links F-actin and forms α-actinin-4-F-actin

338

network surrounding macropinosomes, we hypothesized that the interaction of α-actinin-4 with

339

Hrs may localize Hrs to the membrane and influence macropinosome formation. To test this

340

hypothesis, we first determined whether KSHV infection results in the recruitment of α-actinin-4

341

to the membrane of infected cells. Immunofluorescence analysis of uninfected and KSHV

342

infected cells with α-actinin-4 and KSHV envelope glycoprotein gpK8.1A antibody

343

demonstrated that the virus particles colocalize with α-actinin-4 at 10 min post infection at the

15

344

membrane of infected cells (Fig. 3A). To evaluate whether α-actinin-4 and Hrs could associate

345

with each other at the membrane, we examined the colocalization of Hrs and α-actinin-4 in

346

KSHV infected cells. Immunofluorescent analysis of uninfected and cells infected with KSHV

347

for 10 min demonstrated that Hrs colocalizes with α-actinin-4 at the membrane of infected cells

348

(Fig. 3B).

349

To visualize that the interaction of α-actinin-4 with Hrs leads to the recruitment of Hrs to

350

the cell membrane of infected cells, we used a proximity ligation assay (PLA) with antibodies

351

against Hrs and α-actinin-4. PLA enables the identification of the association between two

352

proteins which are located in close proximity (

ESCRT-0 Component Hrs Promotes Macropinocytosis of Kaposi's Sarcoma-Associated Herpesvirus in Human Dermal Microvascular Endothelial Cells.

Kaposi's sarcoma-associated herpesvirus (KSHV) enters human dermal microvascular endothelial cells (HMVEC-d), its naturalin vivotarget cells, by lipid...
7MB Sizes 0 Downloads 8 Views