IAI Accepts, published online ahead of print on 14 April 2014 Infect. Immun. doi:10.1128/IAI.01204-13 Copyright © 2014, American Society for Microbiology. All Rights Reserved.

1

The anti-apoptotic activity of the Coxiella burnetii effector protein AnkG is

2

controlled by p32-dependent trafficking

3

Rita A. Eckart1, Stephanie Bisle1, Jan Schulze-Luehrmann1, Irene Wittmann1, Jonathan

4

Jantsch1, Benedikt Schmid2, Christian Berens3 and Anja Lührmann1*

5 6

1

7

Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg,

8

Wasserturmstraße 3/5, D-91054 Erlangen, Germany

9

2

Mikrobiologisches Institut – Klinische Mikrobiologie, Immunologie und Hygiene,

Lehrstuhl für Biotechnik, Department Biologie, Friedrich-Alexander-Universität

10

Erlangen-Nürnberg, Henkestrasse 91, D-91052 Erlangen, Germany

11

3

12

Erlangen-Nürnberg, Staudtstrasse 5, D-91058 Erlangen, Germany

Lehrstuhl für Mikrobiologie, Department Biologie, Friedrich-Alexander-Universität

13 14

*Corresponding author:

15

Anja Lührmann1

16

Phone: (+49) 9131 85 22577; Fax: (+49) 9131 85 1001

17

Email: [email protected]

18 19

Running title: AnkG trafficking and apoptosis inhibition

20

1

21

Abstract

22

Intracellular bacterial pathogens frequently inhibit host cell apoptosis to ensure

23

survival of their host, thereby allowing bacterial propagation. The obligate intracellular

24

pathogen Coxiella burnetii displays anti-apoptotic activity which depends on a functional

25

type IV secretion system (T4SS). Accordingly, anti-apoptotic T4SS effector proteins, like

26

AnkG, have been identified. AnkG inhibits pathogen-induced apoptosis, possibly by

27

binding to the host cell mitochondrial protein p32 (gC1qR). However, the molecular

28

mechanism of AnkG activity remains unknown.

29

Here, we demonstrate that ectopically expressed AnkG associates with

30

mitochondria and traffics into the nucleus after apoptosis induction, although AnkG lacks

31

a predicted nuclear localization signal. We identified the p32-interaction region in AnkG

32

and constructed an AnkG-mutant (AnkGR22/23S) unable to bind to p32. By using this

33

mutant we found that intracellular localization and trafficking of AnkG into the nucleus is

34

dependent on binding to p32. Furthermore, we demonstrated that nuclear localization of

35

AnkG but not binding to p32 is required for apoptosis inhibition. Thus, the anti-apoptotic

36

activity of AnkG is controlled by p32-mediated intracellular trafficking, which in turn

37

seems to be regulated by host cell processes that sense stress.

38

2

39

Introduction

40

Coxiella burnetii is the obligate intracellular bacterial agent of human Q-fever, a

41

worldwide zoonotic disease (1). Infection in humans occurs by inhalation of infectious

42

material transmitted from domestic livestock, and as few as ten bacteria can result in

43

disease (2). After bacterial uptake into phagocytic cells, C. burnetii establishes a

44

phagolysosomal-like vacuole (3, 4, 5). Importantly, establishing this replicative niche

45

requires bacterial protein synthesis (6, 7), suggesting direct involvement of bacterial

46

proteins. In agreement with this assumption, the type IV secretion system (T4SS) was

47

shown to be essential for intracellular replication (8, 9). The presence of the replicative

48

C. burnetii-containing vacuole (CCV) within the cell most likely causes tremendous

49

stress for the infected cell, as the CCV almost completely fills the host cell lumen (10).

50

Eukaryotic cells often respond to intracellular pathogen invasion and stress induction by

51

initiating the intrinsic apoptotic pathway as part of the innate immune defense (11).

52

Apoptosis is a programmed cell death pathway crucial for immune system

53

maintenance and removal of damaged or infected cells (12). Two main pathways lead to

54

apoptosis. The extrinsic cell death pathway is launched in response to stimulation of

55

death receptor proteins at the cell surface by extracellular stimuli, while the intrinsic cell

56

death pathway is initiated in response to intracellular stimuli (13).

57

Apoptosis allows pathogen clearance without inflammation and additionally leads

58

to activation of the adaptive immune defense (14, 15). As a countermeasure intracellular

59

pathogens have developed multiple mechanisms to inhibit host cell apoptosis (16). C.

60

burnetii also interferes with host cell apoptosis (17, 18). How this occurs mechanistically

61

is incompletely understood, but effector proteins translocated into the host cell by the

62

T4SS are required for protection against apoptosis (8). Importantly, C. burnetii 3

63

possesses several anti-apoptotic effector proteins like CaeA, CaeB (19) and AnkG (20).

64

How exactly AnkG interferes with the host cell apoptotic machinery has been unknown

65

to date. However, the anti-apoptotic activity of AnkG correlates with binding to p32,

66

because only the N-terminal fragment of AnkG (amino acids (aa) 1-69), which interacts

67

with p32 and inhibits apoptosis, while the C-terminal fragment (aa 70-338) neither

68

interacts with p32 nor interferes with host cell death. Reducing the level of p32 in

69

mammalian cells made them more resistant to apoptosis, suggesting that p32 is a pro-

70

apoptotic protein and that AnkG might function by interfering with this p32-mediated pro-

71

apoptotic activity (20).

72

Several questions regarding AnkG´s function remained open: Does AnkG

73

influence p32 expression? Is the AnkG-p32 interaction direct or indirect? Is the binding

74

to p32 necessary for AnkG-mediated inhibition of apoptosis?

75

To address these questions we have defined the p32-binding pocket within AnkG

76

and created an AnkG mutant that does not bind to p32. Using this and several other

77

mutants we demonstrated that AnkG activity is controlled by p32-mediated trafficking,

78

which in turn seems to be regulated by cellular stress.

79 80

Materials and Methods.

81

Reagents, cell lines and bacterial strains. Unless otherwise noted, chemicals

82

were purchased from Sigma Aldrich. Complete Protease inhibitor cocktail mixture and

83

Xtreme Gene 9 Transfection Reagent were from Roche. Protein A/G Sepharose was

84

from Santa Cruz. Staurosporine was from Cell Signaling. Cell lines were cultured at

85

37°C in 5% CO2 in media containing 10% heat-inactivated fetal bovine serum

86

(Biochrom) and 1% penicillin-streptomycin (Invitrogen). CHO-FcR cells were grown in 4

87

minimal essential medium alpha medium (Invitrogen), HeLa and HEK293 cells were

88

maintained in Dulbecco´s modified Eagle´s medium (Invitrogen). Bone marrow derived-

89

DCs from C57BL/6 mice were prepared as described (21). Escherichia coli strains DH5α

90

and BL21-DE3 were cultivated in Luria-Bertani (LB) broth supplemented with

91

kanamycin, or ampicillin where appropriate. L. pneumophila serogroup 1 ΔflaA strains

92

were grown as described (20).

93

Plasmids and primers. Plasmid and primers used are listed in Tables 1 and 2.

94

Plasmid construction. For creation of the constructs AnkG1-91-pCMV-HA,

95

AnkG50-338-pCMV-HA, AnkG1-157-pCMV-HA and AnkF-pCMV-HA, the genes were

96

amplified from C. burnetii Nine Mile phase II clone 4 genomic DNA by PCR using the

97

primers listed in table 2, restricted with the enzymes indicated and ligated with likewise-

98

restricted pCMV-HA. For creation of the constructs AnkGR23S-pCMV-HA and

99

AnkGR22/23S-pCMV-HA the genes were amplified from AnkGFL-pCMV-HA with primers

100

listed in table 2 that were 5’ phosphorylated. The PCR constructs were gel-purified and

101

ligated. For cloning of the constructs AnkG1-69-pEGFP and AnkG70-338-pEGFP, the genes

102

were amplified from AnkG1-69-pJV400 or AnkG70-338-pJV400 using the primers listed in

103

table 2, restricted with the enzymes indicated and ligated with likewise-restricted

104

pEGFP. For creation of the constructs AnkGR22/23S-pJV400, NES-AnkG-pJV400 and

105

NLS-AnkGR22/23S-pJV400, the genes were amplified from AnkGR22/23S-pEGFP or NES-

106

AnkG-pEGFP using the primers listed in table 2, restricted as indicated and ligated with

107

likewise-restricted pJV400. For cloning the construct NES-AnkG-pGEFP, the gene was

108

amplified from AnkGFL-pEGFP using the primers listed in table 2, restricted with the

109

indicated enzymes and ligated with likewise restricted pEGFP. For cloning the construct

110

NLS-AnkGR22/23S-pEGFP, the gene was amplified from AnkGR22/23S-pCVM-HA using the 5

111

primers listed in table 2, restricted with the indicated enzymes and ligated with likewise

112

restricted pEGFP.

113

Confocal microscopy. CHO cells were plated on coverslips and were

114

transfected with the indicated plasmids. The cells were fixed with 4% paraformaldehyde

115

(Alfa Aeser) in PBS (Biochrom), permeabilized with ice-cold methanol, quenched with

116

50mM NH4Cl (Roth) in PBS. The cells were mounted using ProLong Gold with DAPI

117

(Invitrogen) to visualize the nucleus. For mitochondrial staining, the cells were incubated

118

using Mitotracker (Molecular Probes) before fixation. Confocal fluorescence microscopy

119

was performed using a Zeiss LSM 700 confocal microscope.

120

Nuclear fragmentation assays. Was performed as described (19).

121

Co-immunoprecipitation. HEK293 cells were transiently transfected with the

122

plasmids indicated. On the following day, the cells were washed with PBS and incubated

123

with lysis buffer (20mM HEPES (pH7.5), 200mM NaCl, 1mM EDTA, 0.1% (vol/vol)

124

Nonidet P-40, 10% (vol/vol) glycerol, 1x protease inhibitor, 1mM DTT) for 30min on ice.

125

After centrifugation the supernatants were incubated with anti-GFP rabbit serum from

126

Invitrogen for 2h at 4°C. Complexes were precipitated by adding protein A/G PLUS-

127

Agarose and incubated for 45min at 4°C. The beads were washed three times with

128

washing buffer (20mM HEPES (pH7.5), 100mM NaCl, 1mM EDTA, 0.1% (vol/vol)

129

Nonidet P-40) and samples were analyzed.

130

Protein purification. E. coli BL21 (DE3) cells transformed with plasmids

131

producing GST, GST-AnkG or His-p32 were grown in LB broth containing ampicillin.

132

IPTG was added to the media and samples were incubated for 4h at 30°C. The cells

133

were resuspended in PBS containing Protease inhibitor. After disruption by French

134

Press, the lysate was incubated in 1% Trition X-100 for 1h at 4°C. Lysates were clarified 6

135

by centrifugation at 15000x g for 30min. Proteins were purified using glutathione-

136

sepharose or Ni-NTA agarose colums respectively.

137

GST-tag pull-down. Purified GST or GST-AnkG were loaded onto glutathione–

138

sepharose columns (GE Healthcare), and purified His-p32 was added to the columns.

139

The columns were washed three times with PBS and bound proteins were eluted with

140

10mM glutathione in PBS (pH 9.0). The input, the eluate and the bead fractions were

141

analyzedas indicated

142

His-tag pull-down. Purified His-p32 was loaded onto Ni-NTA agarose columns

143

(GE Healthcare), and GST or GST-AnkG added to the columns. The columns were

144

washed with increasing concentrations of imidazol in lysis buffer (50mM Tris-HCl pH 7.5,

145

150mM NaCl, 1mM DTT) and bound proteins were eluted with 500mM imidazol in lysis

146

buffer. Eluate and input fractions were analyzed as indicated.

147

Statistical analysis. The unpaired Student’s t-test was used.

148

Legionella pneumophila ΔflaA infection. Dendritic cells derived from C57Bl/6

149

mice were infected with the L. pneumophila ΔflaA containing the indicated plasmid as

150

described (20). 2h and 10h after infection, cells were lysed and plated on charcoal yeast

151

extract plates. The plates were incubated for three days at 37°C and colony forming

152

units were counted. Colony number after 2h infection represents the infection efficiency,

153

after 10h the survival of the intracellular bacteria.

154 155

Results

156

AnkG binds p32 directly. To analyze the interaction of AnkG with p32, we first

157

determined whether the binding is direct or indirect. Typically, GST pull-down

158

experiments are used to verify direct interactions between two proteins. Thus, we 7

159

expressed and purified GST, GST-tagged AnkG and His-tagged p32 from Escherichia

160

coli. Purified GST or GST-AnkG was incubated with His-p32 and the putative protein

161

complex pulled-down with glutathione-coated sepharose beads. The eluate and bead

162

fractions were subjected to SDS-PAGE and stained with Coomassie blue (Fig. 1A).

163

Additionally, we analyzed eluate and bead fractions by immunoblot analysis (Fig. 1B).

164

As shown in Figs. 1A and 1B, His-p32 is pulled-down by GST-AnkG, but not by GST

165

alone. To confirm the direct interaction, we also performed the reverse experiment.

166

Thus, purified GST or GST-AnkG and His-p32 were incubated and His-coupled proteins

167

were pulled-down with nickel-NTA-coated agarose beads. Immunoblot analysis revealed

168

that His-p32 pulled-down GST-AnkG (Fig. 1C), but not GST (data not shown).

169

Therefore, binding of AnkG to p32 is direct, because no additional proteins were needed

170

for this interaction.

171

AnkG does not alter the p32 steady-state protein leveI. AnkG was suggested

172

to mediate its anti-apoptotic activity by blocking p32 function (20). Therefore, we first

173

analyzed whether the expression of AnkG results in a reduced p32 protein level. Thus,

174

the respective p32 protein level of cells ectopically expressing GFP or GFP-AnkG was

175

analyzed by immunoblot using an anti-p32 antibody. As shown in Fig. 1D, the p32

176

protein level was not altered by GFP-AnkG expression, suggesting that AnkG does not

177

act by changing the steady-state protein level of p32. Furthermore, AnkG expression did

178

not cause any changes in the intracellular distribution of p32 (Figure 1E).

179

AnkG associates with mitochondria and traffics into the nucleus after stress

180

induction. The host cell protein p32 is mainly found in the mitochondria (22, 23) and a

181

small fraction in the nucleus (24). In order to address the question where the interaction

182

between AnkG and p32 occurs within the cell, we analyzed the intracellular localization 8

183

of ectopically expressed GFP-AnkG. As demonstrated in Fig. 2A GFP-AnkG showed

184

vesicular staining with close association to host cell mitochondria.

185

The distribution of p32 is altered by perturbation of the physiological state of the

186

cell (23-25). As AnkG interacts with p32, we asked whether AnkG also alters its

187

intracellular localization after cellular stress induction. Thus, we treated GFP-AnkG

188

expressing cells for different time periods with staurosporine to cause cellular stress and

189

analyzed subsequently the intracellular localization of AnkG by immunofluorescence.

190

Before treatment the majority of GFP-AnkG was localized in close association with the

191

mitochondria and to a lesser degree in the nucleus, although AnkG does not contain a

192

predicted nuclear localization signal. After treatment with staurosporine, the intracellular

193

localization of GFP-AnkG changed. After 4h GFP-AnkG was mainly present within the

194

nucleus and only a minority remained in close association with the mitochondria (Fig.

195

2B). These results demonstrate that AnkG traffics into the nucleus after apoptosis-

196

induction. Furthermore, it suggests that AnkG requires binding to p32 or another host

197

cell protein to get transported into the nucleus, as AnkG does not contain a predicted

198

nuclear localization.

199

The amino-terminal fragment AnkG1–69 contains one or more regions necessary

200

for inhibition of apoptosis and for binding to p32, whereas AnkG70-338 neither binds to p32

201

nor inhibits apoptosis (20). If the change in intracellular localization depends on binding

202

to p32, the intracellular localization of AnkG70-338 should not change after staurosporine

203

treatment. Thus, we analyzed the intracellular localization of AnkG1-69 and AnkG70-338

204

after cellular perturbation. As shown in Fig. 2C AnkG1–69 was mainly localized in the

205

nucleus under healthy and apoptotic conditions. The nuclear localization of GFP-AnkG1-

206

69

under healthy conditions might be due to its small size of 34kDa. GFP-AnkG1-69 can 9

207

freely migrate into the nucleus and is most likely actively retained within the nucleus. In

208

contrast, AnkG70-338 was mainly localized in the cytoplasm and this localization did not

209

change after treatment with staurosporine. These results led to the hypothesis that

210

trafficking of AnkG might depend on binding to p32 and that nuclear localization might

211

be important for AnkG-mediated apoptosis-inhibition. However, to prove the first

212

hypothesis it was necessary to generate an AnkG mutant unable to bind to p32.

213

An arginine-rich region within AnkG is required for binding to p32. To

214

narrow down the region within AnkG required for binding to p32 we generated different

215

AnkG truncations. We expressed HA-tagged AnkG truncations and GFP-p32 in HEK293

216

cells, precipitated proteins from the cell lysates with an anti-GFP antibody and evaluated

217

the co-immunoprecipitation of the different AnkG truncations by immunoblot analysis. As

218

shown in Fig. 3A HA-AnkG, HA-AnkGΔAnk, HA-AnkG1-157, HA-AnkG1-91 and HA-AnkG1-69 ,

219

but not HA-AnkF, HA-AnkG70-338, HA-AnkG50-338 and HA-AnkG29-338 co-precipitated with

220

GFP-p32. Thus, the first 28 aa of AnkG are most likely required for binding to p32. This

221

N-terminal part contains seven arginine residues. Because p32 was shown to bind to

222

arginine-rich regions (26, 27), we generated point mutations within this arginine-rich N-

223

terminal part, replacing arginine with serine. To analyze binding of the AnkG mutants to

224

p32 co-immunoprecipitation was performed. While HA-AnkG and HA-AnkGR23S co-

225

precipitated with GFP-p32, HA-AnkGR22/23S did not (Fig. 3B). Hence, we identified the

226

p32-binding region within AnkG and generated an AnkG mutant unable to bind to p32.

227

AnkG intracellular localization and trafficking depends on p32 binding. Next,

228

we analyzed the intracellular localization of the AnkG mutant AnkGR22/23S by

229

immunofluorescence. As shown in Fig. 3C, ectopically expressed GFP-AnkGR22/23S co-

230

localized with αtubulin, suggesting that intracellular localization of AnkG is dependent on 10

231

p32 binding. To analyze whether intracellular trafficking of AnkG also depends on p32

232

binding, we treated GFP-AnkGR22/23S expressing cells for different time periods with

233

staurosporine

234

immunofluorescence. The majority of GFP-AnkGR22/23S co-localized with αtubulin and to

235

a lesser degree to the nucleus. Importantly, the intracellular localization of GFP-

236

AnkGR22/23S was not changed by treatment with staurosporine (Fig. 3D). Taken together,

237

nuclear localization and trafficking of AnkG depend on its binding to p32.

and

analyzed

the

intracellular

localization

of

AnkGR22/23S

by

238

AnkG has to migrate into the nucleus to inhibit apoptosis. Having

239

demonstrated that AnkG traffics into the nucleus after apoptosis induction, our goal was

240

to determine whether this nuclear localization is essential for the anti-apoptotic activity of

241

AnkG. Consequently, we expressed a chimera comprising GFP-AnkG fused to the

242

nuclear export signal of the HIV-1 Rev protein (GFP-NES-AnkG). This nuclear export

243

signal has been used successfully to prevent nuclear import of the Golgi vesicle

244

tethering protein p115 (28). GFP-NES-AnkG was excluded from the nucleus (Fig. 4A),

245

but still binds to p32 (Fig. 4B). As shown in Fig. 4C, GFP-NES-AnkG was present

246

exclusively in the cytoplasm and did not migrate into the nucleus after apoptosis-

247

induction (Fig. 4C). Thus, GFP-NES-AnkG can be used to analyze whether AnkG

248

nuclear localization is required for apoptosis inhibition. Therefore, we ectopically

249

produced GFP, GFP-AnkG and GFP-NES-AnkG transiently in CHO cells and treated the

250

cells with staurosporine to induce cell death. Nuclear fragmentation was visualized by

251

DAPI staining and counted to measure apoptosis. Whereas 35% of cells expressing

252

GFP had fragmented nuclei, this number was reduced to 20% in cells expressing GFP-

253

AnkG (Fig. 4D). Importantly, 40% of cells expressing GFP-NES-AnkG had fragmented

11

254

nuclei, demonstrating that NES-AnkG does not inhibit apoptosis. Thus, the anti-apoptotic

255

activity of AnkG strictly requires its translocation into the nucleus.

256

Neither AnkGR22/23S nor NES-AnkG prevents pathogen-induced apoptosis.

257

Next, we asked whether AnkG delivered into the host cell by the T4SS also depends on

258

nuclear localization to exert its anti-apoptotic activity. Because C. burnetii harbors

259

several anti-apoptotic effector proteins, the construction of an ankG deletion-mutant

260

complemented or not with nes-ankG might not provide an answer to this question.

261

Instead we employed a gain-of-function analysis using Legionella pneumophila ΔflaA to

262

determine whether translocation of different AnkG mutants could prevent apoptosis. L.

263

pneumonia ΔflaA caused rapid apoptosis in mouse bone marrow-derived dendritic cells

264

(DCs) and, thus, could not replicate in these cells (21). These pathogen-induced

265

incidents were blocked by adding AnkG to the repertoire of L. pneumophila effector

266

proteins (20). Therefore, this model can be used to analyze whether AnkG has to bind to

267

p32 or whether AnkG has to migrate into the nucleus to inhibit pathogen-induced

268

apoptosis. We infected DCs with L. pneumophila ΔflaA containing either the empty

269

vector (pJV400), AnkG (pJV400-AnkG), NES-AnkG (pJV400-NES-AnkG) or AnkGR22/23S

270

(pJV400-AnkGR22/23S). At 2h and 10h post-infection, the cells were lysed and bacterial

271

colony forming units were counted. As shown in Fig. 5A, bacterial uptake was not

272

affected by the addition of AnkG or any of the AnkG mutants. At 10h post-infection, only

273

12% of the initial inoculum of L. pneumophila ΔflaA containing vector alone were

274

recovered, suggesting that these bacteria induce apoptosis in their host cells and, thus,

275

are not able to survive and replicate (Fig. 5B). In contrast, nearly 50% of the L.

276

pneumophila ΔflaA encoding AnkG were recovered, suggesting that AnkG delivered into

277

the host cell by the L. pneumophila T4SS is able to disrupt pathogen-induced apoptosis 12

278

in DC, in agreement with a previous report (20). Neither L. pneumophila ΔflaA encoding

279

NES-AnkG nor L. pneumophila ΔflaA encoding AnkGR22/23S seemed to inhibit pathogen-

280

induced apoptosis in DCs, as demonstrated by recovery rates of less than 10%. These

281

results support our previous findings and suggest that AnkG depends on binding to p32

282

for proper localization and trafficking into the nucleus and that the nuclear localization is

283

essential for inhibition of host cell apoptosis.

284

The intracellular trafficking, but not the anti-apoptotic activity of AnkG,

285

depends on binding to p32. The previous experiments did not clarify whether the

286

binding to p32 is also necessary for AnkG-mediated anti-apoptotic activity. To address

287

this question we constructed a chimera by fusing the SV40 large T antigen nuclear

288

localization signal (29) to the amino-terminus of AnkGR22/23S (GFP-NLS-AnkGR22/23S).

289

Ectopic expression of this construct displays nuclear localization (Fig. 5C). Next, we

290

ectopically produced GFP, GFP-AnkG and GFP-NLS-AnkGR22/23S transiently in CHO

291

cells and treated the cells with staurosporine to induce cell death. Nuclear fragmentation

292

was visualized by DAPI staining and counted to measure apoptosis. Whereas 35% of

293

cells expressing GFP had fragmented nuclei, this number was reduced to 23% in cells

294

expressing GFP-AnkG (Fig. 5D). Importantly, 22% of cells expressing GFP-NLS-

295

AnkGR22/23S had fragmented nuclei. Thus, AnkG depends on binding to p32 for proper

296

localization and trafficking, but not for anti-apoptotic activity.

297 298

Discussion

299

The elimination of infected cells via apoptosis is an evolutionarily conserved

300

defense mechanism (30). So it is not surprising that many intracellular pathogens have

301

developed mechanisms to counter apoptosis-induction by their host cells (16). Several 13

302

intracellular pathogens inject effector proteins into the host cell to prevent premature

303

host cell death. However, their molecular mechanisms of action are distinct (31). Here

304

we analyzed the anti-apoptotic activity of AnkG. We showed that the effector protein

305

AnkG localizes in association with the host cell mitochondria in unstressed cells (Fig.

306

2A). This is in contrast to a report showing that mCherry-AnkG co-localized with

307

microtubules (32). The difference in localization of AnkG cannot be explained by the cell

308

line used, because both studies used HeLa cells. The only other difference is the tag

309

used. However, we have not detected any tag-dependent differences in the intracellular

310

localization of AnkG so far. GFP-, HA- and myc-tagged AnkG all displayed the same

311

intracellular localization in HeLa and CHO-FcR cells (data not shown). Interestingly,

312

ectopically expressed GFP-AnkGR22/23S co-localized with tubulin (Fig. 3C), and thus

313

displays the same intracellular localization as reported for mCherry-AnkG (32). This

314

localization is surprising, as one would predict that AnkG unable to bind p32 would

315

display cytoplasmic localization. Furthermore, after staurosporine treatment GFP-NES-

316

AnkG displays partial co-localization with tubulin (data not shown). Therefore, it can be

317

speculated that microtubule-association might play a role in AnkG activity under certain

318

cellular conditions.

319

There are several anti-apoptotic type III or type IV secretion system effector

320

proteins that target the host cell mitochondria, the central organelle of the intrinsic

321

apoptotic pathway. Such targeting of the mitochondria by bacterial proteins seems to be

322

evolutionarily conserved, as plant pathogens also target the mitochondria to suppress

323

the hypersensitive response, a form of programmed cell death (33). As shown in Fig. 2A

324

AnkG only partially co-localized with mitochondria, suggesting that this effector protein is

325

not transported into the mitochondria, as it has been shown for Ats1 and PorB. Ats1 14

326

from Anaplasma phagocytophilum uses the mitochondrial import machinery to get

327

transported into the mitochondria (34), while the meningococcal PorB associate with a

328

porin located in the outer mitochondrial membrane (35). Importantly, the anti-apoptotic

329

activity of Ats1 correlates with mitochondrial import (34). AnkG, in clear contrast, has to

330

get transported into the nucleus to act anti-apoptotically (Fig. 4D). Interestingly, this

331

transport into the nucleus, which depends on the ability of AnkG to bind to p32, only

332

happens under apoptotic or stress conditions (Fig. 2B and 3D). This leads to the

333

hypothesis that AnkG primarily targets the mitochondria to sense host cell apoptotic

334

stress and then hitchhikes to the nucleus, the organelle of activity. As a consequence it

335

can be concluded that the activity of AnkG is adjusted by a host cell stress sensor which

336

regulates the transport process.

337

For intracellular trafficking of AnkG from the mitochondria to the nucleus and,

338

thus, for activity control, binding to p32 is essential. This is in agreement with a report

339

that proposed that p32 is involved in bridging a signaling pathway that extends from the

340

mitochondria to the cell nucleus (23). However, once AnkG is within the nucleus, binding

341

to p32 is not needed for anti-apoptotic activity (Fig. 5D). This result suggests that AnkG

342

must instead interfere with a nuclear function to prevent host cell death. There are

343

several effector proteins known to target the host cell nucleus. The Chlamydia

344

trachomatis effector protein NUE is a histone methyltransferase targeting histones (36).

345

AnkA from A. phagocytophilum mediates epigenetic changes at the CYBB promotor

346

(37), leading to a global down-regulation of host defense genes (38). How AnkG

347

modulates nuclear function has to be determined, but the activity of AnkG is clearly

348

regulated by host cell stress signaling and p32-dependent trafficking. This is, to our

15

349

knowledge the first example that an anti-apoptotic effector protein is regulated by host

350

cell protein-mediated trafficking.

351

The question how effector proteins are regulated has only rarely been

352

investigated. There are several avenues of regulation thinkable: 1) regulation by time

353

point and dosage of translocation; 2) regulation by modulation through other effector

354

proteins; 3) regulation by host cell dependent modification (phosphorylation, lipidation,

355

sumoylation etc.). Examples already exist for the latter scenario. It was shown that

356

intracellular localization, and thereby the function of Legionella pneumophila effector

357

proteins containing a CAAX motif are affected by lipidation through the host cell

358

farnesyltransferase and class I geranylgeranyltransferase (39). The Helicobacter pylori

359

T4SS effector protein CagA is phosphorylated by the host cell tyrosine kinases Src and

360

Abl. Phosphorylated CagA can then modulate various signaling cascades associated

361

with cell polarity, cell proliferation, actin-cytoskeletal rearrangements, cell elongation,

362

disruption of tight and adherence junctions, pro-inflammatory responses and apoptosis

363

inhibition (40). Here, we have identified a fourth possibility to regulate the activity of

364

effector proteins: regulation by stress sensing and intracellular trafficking. In our opinion,

365

more knowledge about host cell requirements for regulation of effector proteins is

366

needed. This knowledge will not only help to understand microbial pathogenesis better,

367

but will also allow us to develop new strategies for therapy. The first steps down this

368

avenue have already been made. An exemplified study showed that identifying host cell

369

signaling pathways required for bacterial survival might help to control infection (41).

370 371

Acknowledgements

16

372

This work was supported by the Deutsche Forschungsgemeinschaft (DFG) through the

373

Collaborative Research Initiative 796 (SFB796; to A.L. and C.B.) and through the Priority

374

Programme SPP1580 (to A.L.) as well as by the ERA-NET PathoGenoMics 3rd call (to

375

A.L.). We thank Dr. Christian Bogdan for his valuable comments on the manuscript.

376 377

References

378 379

1. Maurin M, Raoult D. 1999. Q fever. Clin. Microbiol. Rev. 12: 518-553.

380

2. Baca OG, Paretsky D. 1983. Q fever and Coxiella burnetii: a model for host-

381

parasite interactions. Microbiol. Rev. 47: 127-149.

382

3. Howe D, Melnicakova J, Barak I, Heinzen RA. 2003. Maturation of the Coxiella

383

burnetii parasitophorous vacuole requires bacterial protein synthesis but not

384

replication. Cell. Microbiol. 5: 469-80.

385

4. Howe D, Shannon JG, Winfree S, Dorward DW, Heinzen RA. 2010. Coxiella

386

burnetii phase I and II variants replicate with similar kinetics in degradative

387

phagolysosome-like compartments of human macrophages. Infect. Immun. 78:

388

3465-3474.

389

5. Maurin M, Benoliel AM, Bongrand P, Raoult D. (1992). Phagolysosomal

390

alkalinization and the bactericidal effect of antibiotics: the Coxiella burnetii

391

paradigm. J. Infect. Dis. 166: 1097- 1102.

392

6. Howe D, Mallavia LP. 2000. Coxiella burnetii exhibits morphological change and

393

delays phagolysosomal fusion after internalization by J774A.1 cells. Infect.

394

Immun. 68: 3815-3821.

17

395

7. Romano PS, Gutierrez MG, Beron W, Rabinovitch M, Colombo MI. 2007. The

396

autophagic pathway is actively modulated by phase II Coxiella burnetii to

397

efficiently replicate in the host cell. Cell. Microbiol. 9: 891-909.

398

8. Beare PA, Gilk SD, Larson CL, Hill J, Stead CM, Omsland A, Cockrell DC,

399

Howe D, Voth DE, Heinzen RA. 2011. Dot/Icm type IVB secretion system

400

requirements for Coxiella burnetii growth in human macrophages. MBio 2:

401

e00175-00111.

402

9. Carey KL, Newton HJ, Lührmann A, Roy CR. 2011. The Coxiella burnetii

403

Dot/Icm system delivers a unique repertoire of type IV effectors into host cells and

404

is required for intracellular replication. PLoS Pathog. 7: e1002056.

405

10. Van Schaik EJ, Chen C, Mertens K, Weber MM, Samuel JE. 2013. Molecular

406

pathogenesis of the obligate intracellular bacterium Coxiella burnetii. Nat. Rev.

407

Microbiol. 11: 561-573.

408 409 410 411 412 413 414 415 416 417

11. Lamkanfi M, Dixit VM. 2010. Manipulation of host cell death pathways during microbial infections. Cell Host Microbe. 8: 44-54. 12. Byrne GI, Ojcius DM. 2004. Chlamydia and apoptosis: life and death decisions of an intracellular pathogen. Nat. Rev. Microbiol. 2: 802-808. 13. Hotchkiss RS, Strasser A, McDunn JE, Swanson PE. 2009. Cell death. N. Engl. J. Med. 361: 1570-1583. 14. Gallucci S, Lolkema M, Matzinger P. 1999. Natural adjuvants: endogenous activators of dendritic cells. Nat. Med. 5: 1249-1255. 15. Elliott MR, Ravichandran KS. 2010. Clearance of apoptotic cells: implications in health and disease. J. Cell. Biol. 189: 1059-1070.

18

418 419

16. Faherty CS, Maurelli AT. 2008. Staying alive: bacterial inhibition of apoptosis during infection. Trends Microbiol. 16: 173-180.

420

17. Lührmann A, Roy CR. 2007. Coxiella burnetii inhibits activation of host cell

421

apoptosis through a mechanism that involves preventing cytochrome c release

422

from mitochondria. Infect. Immun. 75: 5282-5289.

423

18. Voth DE, Howe D, Heinzen RA. 2007. Coxiella burnetii inhibits apoptosis in

424

human THP-1 cells and monkey primary alveolar macrophages. Infect. Immun.

425

75: 4263-4271.

426

19. Klingenbeck L, Eckart RA, Berens C, Lührmann A. 2013. The Coxiella burnetii

427

type IV secretion system substrate CaeB inhibits intrinsic apoptosis at the

428

mitochondrial level. Cell. Microbiol. 15: 675-687.

429

20. Lührmann A, Nogueira CV, Carey KL, Roy CR. 2010. Inhibition of pathogen-

430

induced apoptosis by a Coxiella burnetii type IV effector protein. Proc. Natl. Acad.

431

Sci. U.S.A. 107: 18997-19001.

432

21. Dedio J, Jahnen-Dechent W, Bachmann M, Müller-Esterl W. 1998. The

433

multiligand-binding protein gC1qR, putative C1q receptor, is a mitochondrial

434

protein. J. Immunol. 160: 3524-3542.

435

22. Matthews DA, Russell WC. 1998. Adenovirus core protein V interacts with

436

p32—a protein which is associated with both the mitochondria and the nucleus. J.

437

Gen. Virol. 79: 1677-1685.

438

23. Brokstad KA, Kalland KH, Matthews DA, Russell WC. 2001. Mitochondrial

439

protein p32 can accumulate in the nucleus. Biochem. Biophys. Res. Commun.

440

281: 1161-1169.

19

441

24. Robles-Flores

M,

Rendon-Huerta

E,

Gonzalez-Aguilar

H,

Mendoza-

442

Hernandez G, Islas S, Mendoza V, Ponce-Castaneda MV, Gonzalez-Mariscal

443

L, Lopez-Casillas F. 2002. p32 (gC1qBP) is a general protein kinase C (PKC)-

444

binding protein; interaction and cellular localization of P32-PKC complexes in ray

445

hepatocytes. J. Biol. Chem. 277: 5247-5255.

446

25. Hall KT, Giles MS, Calderwood MA, Goodwin DJ, Matthews DA, Whitehouse

447

A. 2002. The Herpesvirus Saimiri open reading frame 73 gene product interacts

448

with the cellular protein p32. J. Virol. 76: 11612-11622.

449

26. Beatch MD, Everitt JC, Law LJ, Hobman TC. 2005. Interactions between

450

rubella virus capsid and host protein p32 are important for virus replication. J.

451

Virol. 79: 10807-10820.

452 453

27. Mukherjee S, Shields D. 2009. Nuclear import is required for the pro-apoptotic function of the Golgi protein p115. J. Biol. Chem. 284: 1709-1717.

454

28. Nogueira CV, Lindsten T, Jamieson AM, Case CL, Shin S, Thompson CB,

455

Roy CR. 2009. Rapid pathogen-induced apoptosis: a mechanism used by

456

dendritic cells to limit intracellular replication of Legionella pneumophila. PLoS

457

Pathog. 6: e1000478.

458 459

29. Kalderon D, Roberts BL, Richardson WD, Smith AE. 1984. A short amino acid sequence able to specify nuclear location. Cell. 39: 499-509.

460

30. Ashida H, Mimuro H, Ogawa M, Kobayashi T, Sanada T, Kim M, Sasakawa C.

461

2011. Cell death and infection: a double-edged sword for host and pathogen

462

survival. J. Cell Biol. 195: 931-942.

20

463

31. Raymond B, Young JC, Pallett M, Endres RG, Clements A, Frankel G. 2013.

464

Subversion of trafficking, apoptosis, and innate immunity by type III secretion

465

system effectors. Trends Microbiol. 21: 430-441.

466

32. Voth DE, Howe D, Beare PA, Vogel JP, Unsworth N, Samuel JE, Heinzen RA.

467

2009. The Coxiella burnetii ankyrin repeat domain-containing protein family is

468

heterogeneous, with C-terminal truncations that influence Dot/Icm-mediated

469

secretion. J. Bacteriol. 191: 4232-4242.

470

33. Rudel T, Kepp O, Kozjak-Pavlovic V. 2010. Interaction between bacterial

471

pathogens and mitochondrial cell death pathways. Nat. Rev. Microbiol. 8: 693-

472

34. Niu

H,

Kozjak-Pavlovic

V,

Rudel

T,

Rikihisa

Y.

2010.

Anaplasma

473

phagocytophilum Ats-1 is imported into host cell mitochondria and interferes with

474

apoptosis induction. PLoS Pathog. 6: e1000774.

475

35. Massari P, Ho Y, Wetzler LM. 2000. Neisseria meningitidis porin PorB interacts

476

with mitochondria and protects cells from apoptosis. Proc. Natl. Acad. Sci. U.S.A.

477

97: 9070-9075.

478

36. Pennini ME, Perrinet S, Dautry-Varsat A, Subtil A. 2010. Histone methylation

479

by NUE, a novel nuclear effector of the intracellular pathogen Chlamydia

480

trachomatis. PLoS Pathog. 6: e1000995.

481

37. Garcia-Garcia JC, Rennoll-Bankert KE, Pelly S, Milstone AM, Dumler JS.

482

2009. Silencing of host cell CYBB gene expression by the nuclear effector AnkA

483

of the intracellular pathogen Anaplasma phagocytophilum. Infect. Immun. 77:

484

2385-2391.

21

485

38. Garcia-Garcia JC, Barat NC, Trembley SJ, Dumler JS. 2009. Epigenetic

486

silencing of host cell defense genes enhances intracellular survival of the

487

rickettsial pathogen Anaplasma phagocytophilum. PLoS Pathog. 5: e1000488.

488

39. Ivanov SS, Charron G, Hang HC, Roy CR. 2010. Lipidation by the host

489

prenyltransferase machinery facilitates membrane localization of Legionella

490

pneumophila effector proteins. J. Biol. Chem. 285: 34686-34698.

491

40. Tegtmeyer N, Wessler S, Backert S. 2011 Role of the cag-pathogenicity island

492

encoded type IV secretion system in Helicobacter pylori pathogenesis. FEBS J

493

278: 1190-1202.

494

41. Kuijl C, Savage ND, Marsman M, Tuin AW, Janssen L, Egan DA, Ketema M,

495

van den Nieuwendijk R, van den Eeden SJ, Geluk A, Poot A, van der Marel

496

G, Beijersbergen RL, Overkleeft H, Ottenhoff TH, Neefjes J. 2007. Intracellular

497

bacterial growth is controlled by a kinase network around PKB/AKT1. Nature 450:

498

725-730.

499 500

Figure legends

501

Figure 1: AnkG binds directly to the host cell protein p32 and does not alter its steady

502

state protein level. (A) Glutathione–sepharose columns with GST-AnkG or GST alone

503

were incubated with His-p32. Eluate (E1-E4) and bead (beads) fractions were resolved

504

by SDS-PAGE and stained with Coomassie blue. (B) Glutathione–sepharose columns

505

with GST-AnkG or GST alone were incubated with His-p32. Input, eluate (E1-E4) and

506

bead (beads) fractions were subjected to immunoblot analysis using anti-GST and anti-

507

p32 antibodies. (C) Ni-NTA agarose columns with His-p32 were incubated with GST or

508

with GST-AnkG. Eluate (E1-E4) and input were subjected to immunoblot analysis using 22

509

anti-GST and anti-His antibodies. (D) HEK293 cells were transfected with plasmids

510

encoding GFP or GFP-tagged AnkG. Protein extracts were separated by SDS-PAGE,

511

transferred to a PVDF membrane and probed with antibodies directed against GFP, p32

512

and actin. One representative immunoblot out of at least three independent experiments

513

is shown. (E) HeLa cells were transiently transfected with plasmids encoding GFP or

514

GFP-tagged AnkG. The cells were treated with Mitotracker (red) followed by fixation and

515

permeabilization. P32 was stained with a specific primary antibody and a secondary

516

dye405 labeled antibody (blue). Figure 2: Intracellular localization of AnkG. (A)

517

Representative

518

transfected with a plasmid encoding GFP-tagged AnkG (green). The cells were treated

519

with Mitotracker (red) followed by fixation, permeabilization and staining of the nuclei

520

with DAPI (blue). (B) CHO-FcR cells transiently transfected with GFP-tagged AnkG were

521

incubated with 2µM staurosporine. After the indicated time-points cells were fixed and

522

the intracellular localization of AnkG was analyzed in at least 100 transfected cells per

523

sample using confocal microscopy from eight independent experiments. * p< 0.001, n.s.

524

not significant (p=0.055) (C) Representative immunofluorescence micrographs show

525

CHO-FcR cells expressing GFP-tagged AnkG, -AnkG1-69 or -AnkG70-338 (green). The

526

cells were incubated with staurosporine followed by fixation, permeabilization and

527

staining of the nuclei with DAPI (blue).

528

Figure 3: Identification of the p32 binding site. (A and B) HEK293 cells were co-

529

transfected with plasmids encoding GFP-tagged p32 and the indicated HA-tagged AnkG

530

mutants (HA-tagged AnkF was used as negative control). The proteins were precipitated

531

from the cell lysates with an anti-GFP antibody. Immunoblot analysis was used to detect

532

p32 (anti-GFP) and Ank-proteins (anti-HA) in the lysates (pre-IP) and precipitates (IP).

immunofluorescence

micrographs

show

HeLa

cells

transiently

23

533

(C) Representative immunofluorescence micrographs show HeLa cells expressing GFP-

534

tagged AnkGR22/23S (green). The cells were fixed, permeabilized and stained with anti-

535

tubulin antibody (red) and DAPI (blue). (D) CHO-FcR cells expressing GFP-tagged

536

AnkGR22/23S were incubated with 2µM staurosporine. After the indicated time points cells

537

were fixed and the localization of AnkG was analyzed in at least 100 transfected cells

538

per sample from four independent experiments using confocal microscopy. n.s. not

539

significant

540

Figure 4: AnkG has to migrate into the nucleus to inhibit staurosporine-induced

541

apoptosis. (A) Representative immunofluorescence micrograph show CHO-FcR cells

542

expressing GFP-tagged NES-AnkG (green). The cells were treated with Mitotracker

543

(red) followed by fixation, permeabilization and staining of the nuclei with DAPI (blue).

544

(B) HEK293 cells were co-transfected with plasmids encoding the indicated GFP-tagged

545

AnkG mutants or GFP as negative control. The proteins were precipitated from the cell

546

lysates with an anti-GFP antibody. Immunoblot analysis was used to detect endogenous

547

p32 (anti-p32) or AnkG (anti-AnkG) in the lysates (pre-IP) and precipitates (IP). (C)

548

CHO-FcR cells expressing GFP-tagged NES-AnkG were incubated with 2µM

549

staurosporine. After the indicated time-points cells were fixed and the localization of

550

AnkG was analyzed in at least 100 transfected cells per sample from three independent

551

experiments using confocal microscopy. n.s. not significant (D) CHO-FcR cells

552

expressing GFP, GFP- AnkG or -NES-AnkG were treated with staurosporine for 4h. The

553

cells were fixed, permeabilized and the nuclei were stained with DAPI. The nuclear

554

morphology of at least 100 GFP expressing cells was scored in four independent

555

experiments. n.s. not significant * p< 0.02.

24

556

Figure 5: Neither AnkGR22/23S nor NES-AnkG can prevent pathogen-induced apoptosis

557

(A/B) Dendritic cells were infected with Legionella pneumophila ΔflaA containing the

558

indicated plasmids. The data shown are from one representative experiment of three

559

experiments with similar results. Shown is the bacterial uptake after 2 hours infection (A)

560

and the relative number of intracellular bacteria 10 hours after infection compared to the

561

2 hour value. **p= 0.001 (C) Representative immunofluorescence micrograph shows

562

CHO-FcR cells expressing GFP-NLS-AnkGR22/23S (green) incubated with mitotracker

563

(red) followed by fixation, permeabilization and staining of the nuclei with DAPI (blue).

564

(D) CHO-FcR cells expressing GFP, GFP-AnkG or GFP-NLS-AnkGR22/23S were treated

565

with 2µM staurosporine for 4h. After treatment the cells were fixed, permeabilized and

566

the nuclei were stained with DAPI. The nuclear morphology was scored of at least 100

567

GFP expressing cells in three independent experiments. * p< 0.01.

568 569

Tables

570 Plasmid AnkGFL-pCMV-HA AnkG∆Ank-pCMV-HA AnkG1-69-pCMV-HA AnkG70-338-pCMV-HA AnkG1-91-pCMV-HA AnkG50-338-pCMV-HA AnkG1-157-pCMV-HA AnkGR23S-pCMV-HA AnkGR22/23S-pCMV-HA AnkF-pCMV-HA pGEX-5X AnkG-pGEX-5X p32-pET16b pEGFP AnkG-pEGFP p32-pEGFP AnkG1-69-pEGFP AnkG70-338-pEGFP

Primer

329/339 331/340 307/310 590/591 590/592 79/229 184/185

329/338 332/340

Reference 20 20 20 20 This study This study This study This study This study This study Amersham 20 This study Clontech 20 20 This study This study

25

AnkGR22/23S-pEGFP NES-AnkG-pEGFP NLS-AnkGR22/23S-pEGFP pJV400 pJV400-AnkG pJV400-AnkGR22/23S pJV400-NES-AnkG

571

665/34 746/400 373/26 696/26

This study This study This study 20 20 This study This study

Table 1: Primer numbers are as in Table 2.

572 Number 26 34 79 184 185 229 307 310 329 330 331 332 338 339 340 373 400 590 591 592 665 696 746

573

Sequence 5' AAGGCGCGCCTCACCGAGGACTAGACAG 5’ AAGGATCCTCACCGAGGACTAGACAGA 5’ CCGGTACCCTACCGCTGGAAGCCGC 5’ CCCATATGCTGCACACCGACGGAGAC 5’ CCGGATCCCTACTGGCTCTTGACAAAACT 5’ CCGAATTCATGTGCAATACCAACATGTCT 5’ CCGGTACCATGAGTAGACGTGAGACTCC 5’ CCGGTACCTTATTTATATTTGATTTTCACATCAGC 5' CCAAGATCTCTATGAGTAGACGTGAGACTCC 5’ CCAAGATCTCTATGGGACATCCTGTAAGAAGAAG 5’ CCAAGATCTCTATGTCGTTTGAAATACTCATAAATGC 5’ CCAAGATCTCTATGCTTCGCGGGGATTCTTTTCA 5' CCGGTACCTCAGTAGTTTTTTATTATGCTCAAGCT 5’ CCGGTACCTCAGAAATCCGTCTTTGGCGGTA 5' CCGGTACCTCACCGAGGACTAGACAGA 5' CCGGCCGGCCATGAGTAGACGTGAGACTCC 5' CCGGTACCTCACCGAGGACTAGACAGA P-5’ CGTTGAGGATATTGTGCTAGTGGGAGTCTACGTCTAC TCAT P-5’ CGACAGGAACTCGAACGCCGAGAAGTAGATTGAGCC GAAAA P-5’CGACAGGAACTCGAACGCCGAGTAGTAGATTGAGC CGAAAA 5’ CCGGTACCGCCTCCAGCAGCCTCCCCTGGAGGACTGACCCT GAGTAGACGTGAGACTCCCACTAGC 5’ CCGGCCGGCCATGCTCCAGCTGCCTCCCC 5’ CCACTCAGATCTCTCCTAAGAAGAAAAGGAAGGTTAGT AGACGTGAGACTCCCACTAGCACAA

Site AscI BamHI KpnI NdeI BamHI EcoRI KpnI KpnI BglII BglII BglII BglII KpnI KpnI KpnI FseI KpnI KpnI FseI BglII

Table 2. Underlined denotes the location of the restriction site.

26

GST-AnkG

E1

E2

E3

E4

anti-GST anti-GFP

anti-HIS

HIS-p32 GST

D

nk G

GST-AnkG + HIS-p32

G FP -A

C

GST + His-p32

G FP

GST-AnkG + His-p32

E1 E2 E3 E4 Beads E1 E2 E3 E4 Beads

G ST -A n H IS kG -p 32

A

HIS-tag pull-down

anti-p32

input

anti-actin GST-tag pull-down

E G ST H is -p 3

2

B

p32

GFP

Mitotracker

Merge

p32

GFP-AnkG

Mitotracker

Merge

E1 E2 E3 E4 Beads anti-GST anti-p32 GST-tag pull-down 10µm

G ST H Ank is -p G 32

input

E1 E2 E3 E4 Beads anti-GST anti-p32

input

GST-tag pull-down

10µm

A GFP-AnkG

Mitotracker

Merge

Zoom

10µm

B % phenotype

125

*** ***

100

n.s.

75 50

Mitochondrial associa!on

25

nuclear

0 Mock

30

120

240

min staurosporine

GFP-AnkG

GFP-AnkG1-69

GFP-AnkG 70-338

Mock

C

10µm

10µm

Staurosporine

10µm

10µm

10µm

10µm

22

S

A

-A

nk G

R

23 R

W T

-A H

H

A

A H

H

nk G

nk G

nk F

-A

-A A

-A A

/2 3

S

S /2 3 22

S

nk G

R

22 R

W T

nk G

nk G

-A A

-A

nk F A

-A H

A H

H

50

nk G -A

A H

H

38 -3

38

B

70

-3

-3 29

-A A H

H

A

-A

nk G

nk G -A

A H

nk G

1-

91 1-

1-

H

A

-A

nk G

nk G -A

A H

69

7 15

nk ∆A

W T

nk G

nk G H

A

-A

-A A H

38

A

pre-IP

anti-HA anti-HA anti-GFP

pre-IP amino acid AnkG AnkG R23S AnkG R22/23S

18 19 20 21 22 23 24 25 26 27 28 T R T P R R R L S R K T R T P R S R L S R K T R T P S S R L S R K

IP

anti-HA

IP

D

n.s.

anti-GFP

GFP-AnkGR22/23S

αTubulin

100

% phenotype

C Merge

n.s. n.s.

75 50

Co-localiza!on with Tubulin

25

nuclear

0 Mock 10µm

30

120

240

min staurosporine

A GFP-NES-AnkG

Mitotracker

Merge

G FP -

G FP

G FP -

A nk G G FP -A nk G G FP R 22 -N / ES 23S -A nk G G FP

B

A nk G G FP -A nk G G FP R 22 -N / ES 23S -A nk G

10µm 10µm

anti-AnkG anti-p32

pre-IP

IP

C

n.s. n.s. n.s.

% phenotype

100 75 50 cytosolic 25

nuclear

0 Mock

30

120

min staurosporine

D

n.s.

% fragmented nuclei

50

*

40 30 20 10

240

-A ES -N FP G G nk

-A FP G

FP G

G nk

5

10 0

0

* * 20

10

0

Merge Mitotracker GFP-NLS-AnkG

G nk -A ES /23S -N 22 00 GR V4 nk pJ 0-A 0 G V4 nk pJ 0-A 0 V4 pJ 0 40

C

30

20

% survival

40

bacterial *103/well

10

V pJ

G nk -A ES /23S -N 22 00 GR V4 nk pJ 0-A 0 G V4 nk pJ 0-A 0 V4 pJ 0 40 V pJ

40

D

30

% fragmented nuclei

15

* 50 20

60

B n.s. 25

A

-A LS -N FP kG G An FP G FP G

G nk

R

22

3 /2

S

Antiapoptotic activity of Coxiella burnetii effector protein AnkG is controlled by p32-dependent trafficking.

Intracellular bacterial pathogens frequently inhibit host cell apoptosis to ensure survival of their host, thereby allowing bacterial propagation. The...
3MB Sizes 0 Downloads 3 Views