31
1 The anti-apoptotic activity of the Coxiella burnetii effector protein AnkG is 1 controlled by p32-dependent trafficking 2 Rita A. Eckart 1 , Stephanie Bisle 1 , Jan Schulze-Luehrmann 1 , Irene Wittmann 1 , Jonathan 3 Jantsch 1 , Benedikt Schmid 2 , Christian Berens 3 and Anja Lührmann 1* 4 5 1 Mikrobiologisches Institut – Klinische Mikrobiologie, Immunologie und Hygiene, 6 Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, 7 Wasserturmstraße 3/5, D-91054 Erlangen, Germany 8 2 Lehrstuhl für Biotechnik, Department Biologie, Friedrich-Alexander-Universität 9 Erlangen-Nürnberg, Henkestrasse 91, D-91052 Erlangen, Germany 10 3 Lehrstuhl für Mikrobiologie, Department Biologie, Friedrich-Alexander-Universität 11 Erlangen-Nürnberg, Staudtstrasse 5, D-91058 Erlangen, Germany 12 13 *Corresponding author: 14 Anja Lührmann 1 15 Phone: (+49) 9131 85 22577; Fax: (+49) 9131 85 1001 16 Email: [email protected] 17 18 Running title: AnkG trafficking and apoptosis inhibition 19 20 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. on September 6, 2018 by guest http://iai.asm.org/ Downloaded from

IAI Accepts, published online ahead of print on 14 April ...iai.asm.org/content/early/2014/04/08/IAI.01204-13.full.pdf · 83 Xtreme Gene 9 Transfection Reagent were fr om Roche. Protein

  • Upload
    phamnhi

  • View
    212

  • Download
    0

Embed Size (px)

Citation preview

1

The anti-apoptotic activity of the Coxiella burnetii effector protein AnkG is 1

controlled by p32-dependent trafficking 2

Rita A. Eckart1, Stephanie Bisle1, Jan Schulze-Luehrmann1, Irene Wittmann1, Jonathan 3

Jantsch1, Benedikt Schmid2, Christian Berens3 and Anja Lührmann1* 4

5

1Mikrobiologisches Institut – Klinische Mikrobiologie, Immunologie und Hygiene, 6

Universitätsklinikum Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, 7

Wasserturmstraße 3/5, D-91054 Erlangen, Germany 8

2Lehrstuhl für Biotechnik, Department Biologie, Friedrich-Alexander-Universität 9

Erlangen-Nürnberg, Henkestrasse 91, D-91052 Erlangen, Germany 10

3Lehrstuhl für Mikrobiologie, Department Biologie, Friedrich-Alexander-Universität 11

Erlangen-Nürnberg, Staudtstrasse 5, D-91058 Erlangen, Germany 12

13

*Corresponding author: 14

Anja Lührmann1 15

Phone: (+49) 9131 85 22577; Fax: (+49) 9131 85 1001 16

Email: [email protected] 17

18

Running title: AnkG trafficking and apoptosis inhibition 19

20

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

on Septem

ber 6, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

2

Abstract 21

Intracellular bacterial pathogens frequently inhibit host cell apoptosis to ensure 22

survival of their host, thereby allowing bacterial propagation. The obligate intracellular 23

pathogen Coxiella burnetii displays anti-apoptotic activity which depends on a functional 24

type IV secretion system (T4SS). Accordingly, anti-apoptotic T4SS effector proteins, like 25

AnkG, have been identified. AnkG inhibits pathogen-induced apoptosis, possibly by 26

binding to the host cell mitochondrial protein p32 (gC1qR). However, the molecular 27

mechanism of AnkG activity remains unknown. 28

Here, we demonstrate that ectopically expressed AnkG associates with 29

mitochondria and traffics into the nucleus after apoptosis induction, although AnkG lacks 30

a predicted nuclear localization signal. We identified the p32-interaction region in AnkG 31

and constructed an AnkG-mutant (AnkGR22/23S) unable to bind to p32. By using this 32

mutant we found that intracellular localization and trafficking of AnkG into the nucleus is 33

dependent on binding to p32. Furthermore, we demonstrated that nuclear localization of 34

AnkG but not binding to p32 is required for apoptosis inhibition. Thus, the anti-apoptotic 35

activity of AnkG is controlled by p32-mediated intracellular trafficking, which in turn 36

seems to be regulated by host cell processes that sense stress. 37

38

on Septem

ber 6, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

3

Introduction 39

Coxiella burnetii is the obligate intracellular bacterial agent of human Q-fever, a 40

worldwide zoonotic disease (1). Infection in humans occurs by inhalation of infectious 41

material transmitted from domestic livestock, and as few as ten bacteria can result in 42

disease (2). After bacterial uptake into phagocytic cells, C. burnetii establishes a 43

phagolysosomal-like vacuole (3, 4, 5). Importantly, establishing this replicative niche 44

requires bacterial protein synthesis (6, 7), suggesting direct involvement of bacterial 45

proteins. In agreement with this assumption, the type IV secretion system (T4SS) was 46

shown to be essential for intracellular replication (8, 9). The presence of the replicative 47

C. burnetii-containing vacuole (CCV) within the cell most likely causes tremendous 48

stress for the infected cell, as the CCV almost completely fills the host cell lumen (10). 49

Eukaryotic cells often respond to intracellular pathogen invasion and stress induction by 50

initiating the intrinsic apoptotic pathway as part of the innate immune defense (11). 51

Apoptosis is a programmed cell death pathway crucial for immune system 52

maintenance and removal of damaged or infected cells (12). Two main pathways lead to 53

apoptosis. The extrinsic cell death pathway is launched in response to stimulation of 54

death receptor proteins at the cell surface by extracellular stimuli, while the intrinsic cell 55

death pathway is initiated in response to intracellular stimuli (13). 56

Apoptosis allows pathogen clearance without inflammation and additionally leads 57

to activation of the adaptive immune defense (14, 15). As a countermeasure intracellular 58

pathogens have developed multiple mechanisms to inhibit host cell apoptosis (16). C. 59

burnetii also interferes with host cell apoptosis (17, 18). How this occurs mechanistically 60

is incompletely understood, but effector proteins translocated into the host cell by the 61

T4SS are required for protection against apoptosis (8). Importantly, C. burnetii 62

on Septem

ber 6, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

4

possesses several anti-apoptotic effector proteins like CaeA, CaeB (19) and AnkG (20). 63

How exactly AnkG interferes with the host cell apoptotic machinery has been unknown 64

to date. However, the anti-apoptotic activity of AnkG correlates with binding to p32, 65

because only the N-terminal fragment of AnkG (amino acids (aa) 1-69), which interacts 66

with p32 and inhibits apoptosis, while the C-terminal fragment (aa 70-338) neither 67

interacts with p32 nor interferes with host cell death. Reducing the level of p32 in 68

mammalian cells made them more resistant to apoptosis, suggesting that p32 is a pro-69

apoptotic protein and that AnkG might function by interfering with this p32-mediated pro-70

apoptotic activity (20). 71

Several questions regarding AnkG´s function remained open: Does AnkG 72

influence p32 expression? Is the AnkG-p32 interaction direct or indirect? Is the binding 73

to p32 necessary for AnkG-mediated inhibition of apoptosis? 74

To address these questions we have defined the p32-binding pocket within AnkG 75

and created an AnkG mutant that does not bind to p32. Using this and several other 76

mutants we demonstrated that AnkG activity is controlled by p32-mediated trafficking, 77

which in turn seems to be regulated by cellular stress. 78

79

Materials and Methods. 80

Reagents, cell lines and bacterial strains. Unless otherwise noted, chemicals 81

were purchased from Sigma Aldrich. Complete Protease inhibitor cocktail mixture and 82

Xtreme Gene 9 Transfection Reagent were from Roche. Protein A/G Sepharose was 83

from Santa Cruz. Staurosporine was from Cell Signaling. Cell lines were cultured at 84

37°C in 5% CO2 in media containing 10% heat-inactivated fetal bovine serum 85

(Biochrom) and 1% penicillin-streptomycin (Invitrogen). CHO-FcR cells were grown in 86

on Septem

ber 6, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

5

minimal essential medium alpha medium (Invitrogen), HeLa and HEK293 cells were 87

maintained in Dulbecco´s modified Eagle´s medium (Invitrogen). Bone marrow derived-88

DCs from C57BL/6 mice were prepared as described (21). Escherichia coli strains DH5α 89

and BL21-DE3 were cultivated in Luria-Bertani (LB) broth supplemented with 90

kanamycin, or ampicillin where appropriate. L. pneumophila serogroup 1 ΔflaA strains 91

were grown as described (20). 92

Plasmids and primers. Plasmid and primers used are listed in Tables 1 and 2. 93

Plasmid construction. For creation of the constructs AnkG1-91-pCMV-HA, 94

AnkG50-338-pCMV-HA, AnkG1-157-pCMV-HA and AnkF-pCMV-HA, the genes were 95

amplified from C. burnetii Nine Mile phase II clone 4 genomic DNA by PCR using the 96

primers listed in table 2, restricted with the enzymes indicated and ligated with likewise-97

restricted pCMV-HA. For creation of the constructs AnkGR23S-pCMV-HA and 98

AnkGR22/23S-pCMV-HA the genes were amplified from AnkGFL-pCMV-HA with primers 99

listed in table 2 that were 5’ phosphorylated. The PCR constructs were gel-purified and 100

ligated. For cloning of the constructs AnkG1-69-pEGFP and AnkG70-338-pEGFP, the genes 101

were amplified from AnkG1-69-pJV400 or AnkG70-338-pJV400 using the primers listed in 102

table 2, restricted with the enzymes indicated and ligated with likewise-restricted 103

pEGFP. For creation of the constructs AnkGR22/23S-pJV400, NES-AnkG-pJV400 and 104

NLS-AnkGR22/23S-pJV400, the genes were amplified from AnkGR22/23S-pEGFP or NES-105

AnkG-pEGFP using the primers listed in table 2, restricted as indicated and ligated with 106

likewise-restricted pJV400. For cloning the construct NES-AnkG-pGEFP, the gene was 107

amplified from AnkGFL-pEGFP using the primers listed in table 2, restricted with the 108

indicated enzymes and ligated with likewise restricted pEGFP. For cloning the construct 109

NLS-AnkGR22/23S-pEGFP, the gene was amplified from AnkGR22/23S-pCVM-HA using the 110

on Septem

ber 6, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

6

primers listed in table 2, restricted with the indicated enzymes and ligated with likewise 111

restricted pEGFP. 112

Confocal microscopy. CHO cells were plated on coverslips and were 113

transfected with the indicated plasmids. The cells were fixed with 4% paraformaldehyde 114

(Alfa Aeser) in PBS (Biochrom), permeabilized with ice-cold methanol, quenched with 115

50mM NH4Cl (Roth) in PBS. The cells were mounted using ProLong Gold with DAPI 116

(Invitrogen) to visualize the nucleus. For mitochondrial staining, the cells were incubated 117

using Mitotracker (Molecular Probes) before fixation. Confocal fluorescence microscopy 118

was performed using a Zeiss LSM 700 confocal microscope. 119

Nuclear fragmentation assays. Was performed as described (19). 120

Co-immunoprecipitation. HEK293 cells were transiently transfected with the 121

plasmids indicated. On the following day, the cells were washed with PBS and incubated 122

with lysis buffer (20mM HEPES (pH7.5), 200mM NaCl, 1mM EDTA, 0.1% (vol/vol) 123

Nonidet P-40, 10% (vol/vol) glycerol, 1x protease inhibitor, 1mM DTT) for 30min on ice. 124

After centrifugation the supernatants were incubated with anti-GFP rabbit serum from 125

Invitrogen for 2h at 4°C. Complexes were precipitated by adding protein A/G PLUS-126

Agarose and incubated for 45min at 4°C. The beads were washed three times with 127

washing buffer (20mM HEPES (pH7.5), 100mM NaCl, 1mM EDTA, 0.1% (vol/vol) 128

Nonidet P-40) and samples were analyzed. 129

Protein purification. E. coli BL21 (DE3) cells transformed with plasmids 130

producing GST, GST-AnkG or His-p32 were grown in LB broth containing ampicillin. 131

IPTG was added to the media and samples were incubated for 4h at 30°C. The cells 132

were resuspended in PBS containing Protease inhibitor. After disruption by French 133

Press, the lysate was incubated in 1% Trition X-100 for 1h at 4°C. Lysates were clarified 134

on Septem

ber 6, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

7

by centrifugation at 15000x g for 30min. Proteins were purified using glutathione-135

sepharose or Ni-NTA agarose colums respectively. 136

GST-tag pull-down. Purified GST or GST-AnkG were loaded onto glutathione–137

sepharose columns (GE Healthcare), and purified His-p32 was added to the columns. 138

The columns were washed three times with PBS and bound proteins were eluted with 139

10mM glutathione in PBS (pH 9.0). The input, the eluate and the bead fractions were 140

analyzedas indicated 141

His-tag pull-down. Purified His-p32 was loaded onto Ni-NTA agarose columns 142

(GE Healthcare), and GST or GST-AnkG added to the columns. The columns were 143

washed with increasing concentrations of imidazol in lysis buffer (50mM Tris-HCl pH 7.5, 144

150mM NaCl, 1mM DTT) and bound proteins were eluted with 500mM imidazol in lysis 145

buffer. Eluate and input fractions were analyzed as indicated. 146

Statistical analysis. The unpaired Student’s t-test was used. 147

Legionella pneumophila ΔflaA infection. Dendritic cells derived from C57Bl/6 148

mice were infected with the L. pneumophila ΔflaA containing the indicated plasmid as 149

described (20). 2h and 10h after infection, cells were lysed and plated on charcoal yeast 150

extract plates. The plates were incubated for three days at 37°C and colony forming 151

units were counted. Colony number after 2h infection represents the infection efficiency, 152

after 10h the survival of the intracellular bacteria. 153

154

Results 155

AnkG binds p32 directly. To analyze the interaction of AnkG with p32, we first 156

determined whether the binding is direct or indirect. Typically, GST pull-down 157

experiments are used to verify direct interactions between two proteins. Thus, we 158

on Septem

ber 6, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

8

expressed and purified GST, GST-tagged AnkG and His-tagged p32 from Escherichia 159

coli. Purified GST or GST-AnkG was incubated with His-p32 and the putative protein 160

complex pulled-down with glutathione-coated sepharose beads. The eluate and bead 161

fractions were subjected to SDS-PAGE and stained with Coomassie blue (Fig. 1A). 162

Additionally, we analyzed eluate and bead fractions by immunoblot analysis (Fig. 1B). 163

As shown in Figs. 1A and 1B, His-p32 is pulled-down by GST-AnkG, but not by GST 164

alone. To confirm the direct interaction, we also performed the reverse experiment. 165

Thus, purified GST or GST-AnkG and His-p32 were incubated and His-coupled proteins 166

were pulled-down with nickel-NTA-coated agarose beads. Immunoblot analysis revealed 167

that His-p32 pulled-down GST-AnkG (Fig. 1C), but not GST (data not shown). 168

Therefore, binding of AnkG to p32 is direct, because no additional proteins were needed 169

for this interaction. 170

AnkG does not alter the p32 steady-state protein leveI. AnkG was suggested 171

to mediate its anti-apoptotic activity by blocking p32 function (20). Therefore, we first 172

analyzed whether the expression of AnkG results in a reduced p32 protein level. Thus, 173

the respective p32 protein level of cells ectopically expressing GFP or GFP-AnkG was 174

analyzed by immunoblot using an anti-p32 antibody. As shown in Fig. 1D, the p32 175

protein level was not altered by GFP-AnkG expression, suggesting that AnkG does not 176

act by changing the steady-state protein level of p32. Furthermore, AnkG expression did 177

not cause any changes in the intracellular distribution of p32 (Figure 1E). 178

AnkG associates with mitochondria and traffics into the nucleus after stress 179

induction. The host cell protein p32 is mainly found in the mitochondria (22, 23) and a 180

small fraction in the nucleus (24). In order to address the question where the interaction 181

between AnkG and p32 occurs within the cell, we analyzed the intracellular localization 182

on Septem

ber 6, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

9

of ectopically expressed GFP-AnkG. As demonstrated in Fig. 2A GFP-AnkG showed 183

vesicular staining with close association to host cell mitochondria. 184

The distribution of p32 is altered by perturbation of the physiological state of the 185

cell (23-25). As AnkG interacts with p32, we asked whether AnkG also alters its 186

intracellular localization after cellular stress induction. Thus, we treated GFP-AnkG 187

expressing cells for different time periods with staurosporine to cause cellular stress and 188

analyzed subsequently the intracellular localization of AnkG by immunofluorescence. 189

Before treatment the majority of GFP-AnkG was localized in close association with the 190

mitochondria and to a lesser degree in the nucleus, although AnkG does not contain a 191

predicted nuclear localization signal. After treatment with staurosporine, the intracellular 192

localization of GFP-AnkG changed. After 4h GFP-AnkG was mainly present within the 193

nucleus and only a minority remained in close association with the mitochondria (Fig. 194

2B). These results demonstrate that AnkG traffics into the nucleus after apoptosis-195

induction. Furthermore, it suggests that AnkG requires binding to p32 or another host 196

cell protein to get transported into the nucleus, as AnkG does not contain a predicted 197

nuclear localization. 198

The amino-terminal fragment AnkG1–69 contains one or more regions necessary 199

for inhibition of apoptosis and for binding to p32, whereas AnkG70-338 neither binds to p32 200

nor inhibits apoptosis (20). If the change in intracellular localization depends on binding 201

to p32, the intracellular localization of AnkG70-338 should not change after staurosporine 202

treatment. Thus, we analyzed the intracellular localization of AnkG1-69 and AnkG70-338 203

after cellular perturbation. As shown in Fig. 2C AnkG1–69 was mainly localized in the 204

nucleus under healthy and apoptotic conditions. The nuclear localization of GFP-AnkG1-205

69 under healthy conditions might be due to its small size of 34kDa. GFP-AnkG1-69 can 206

on Septem

ber 6, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

10

freely migrate into the nucleus and is most likely actively retained within the nucleus. In 207

contrast, AnkG70-338 was mainly localized in the cytoplasm and this localization did not 208

change after treatment with staurosporine. These results led to the hypothesis that 209

trafficking of AnkG might depend on binding to p32 and that nuclear localization might 210

be important for AnkG-mediated apoptosis-inhibition. However, to prove the first 211

hypothesis it was necessary to generate an AnkG mutant unable to bind to p32. 212

An arginine-rich region within AnkG is required for binding to p32. To 213

narrow down the region within AnkG required for binding to p32 we generated different 214

AnkG truncations. We expressed HA-tagged AnkG truncations and GFP-p32 in HEK293 215

cells, precipitated proteins from the cell lysates with an anti-GFP antibody and evaluated 216

the co-immunoprecipitation of the different AnkG truncations by immunoblot analysis. As 217

shown in Fig. 3A HA-AnkG, HA-AnkGΔAnk, HA-AnkG1-157, HA-AnkG1-91 and HA-AnkG1-69 , 218

but not HA-AnkF, HA-AnkG70-338, HA-AnkG50-338 and HA-AnkG29-338 co-precipitated with 219

GFP-p32. Thus, the first 28 aa of AnkG are most likely required for binding to p32. This 220

N-terminal part contains seven arginine residues. Because p32 was shown to bind to 221

arginine-rich regions (26, 27), we generated point mutations within this arginine-rich N-222

terminal part, replacing arginine with serine. To analyze binding of the AnkG mutants to 223

p32 co-immunoprecipitation was performed. While HA-AnkG and HA-AnkGR23S co-224

precipitated with GFP-p32, HA-AnkGR22/23S did not (Fig. 3B). Hence, we identified the 225

p32-binding region within AnkG and generated an AnkG mutant unable to bind to p32. 226

AnkG intracellular localization and trafficking depends on p32 binding. Next, 227

we analyzed the intracellular localization of the AnkG mutant AnkGR22/23S by 228

immunofluorescence. As shown in Fig. 3C, ectopically expressed GFP-AnkGR22/23S co-229

localized with αtubulin, suggesting that intracellular localization of AnkG is dependent on 230

on Septem

ber 6, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

11

p32 binding. To analyze whether intracellular trafficking of AnkG also depends on p32 231

binding, we treated GFP-AnkGR22/23S expressing cells for different time periods with 232

staurosporine and analyzed the intracellular localization of AnkGR22/23S by 233

immunofluorescence. The majority of GFP-AnkGR22/23S co-localized with αtubulin and to 234

a lesser degree to the nucleus. Importantly, the intracellular localization of GFP-235

AnkGR22/23S was not changed by treatment with staurosporine (Fig. 3D). Taken together, 236

nuclear localization and trafficking of AnkG depend on its binding to p32. 237

AnkG has to migrate into the nucleus to inhibit apoptosis. Having 238

demonstrated that AnkG traffics into the nucleus after apoptosis induction, our goal was 239

to determine whether this nuclear localization is essential for the anti-apoptotic activity of 240

AnkG. Consequently, we expressed a chimera comprising GFP-AnkG fused to the 241

nuclear export signal of the HIV-1 Rev protein (GFP-NES-AnkG). This nuclear export 242

signal has been used successfully to prevent nuclear import of the Golgi vesicle 243

tethering protein p115 (28). GFP-NES-AnkG was excluded from the nucleus (Fig. 4A), 244

but still binds to p32 (Fig. 4B). As shown in Fig. 4C, GFP-NES-AnkG was present 245

exclusively in the cytoplasm and did not migrate into the nucleus after apoptosis-246

induction (Fig. 4C). Thus, GFP-NES-AnkG can be used to analyze whether AnkG 247

nuclear localization is required for apoptosis inhibition. Therefore, we ectopically 248

produced GFP, GFP-AnkG and GFP-NES-AnkG transiently in CHO cells and treated the 249

cells with staurosporine to induce cell death. Nuclear fragmentation was visualized by 250

DAPI staining and counted to measure apoptosis. Whereas 35% of cells expressing 251

GFP had fragmented nuclei, this number was reduced to 20% in cells expressing GFP-252

AnkG (Fig. 4D). Importantly, 40% of cells expressing GFP-NES-AnkG had fragmented 253

on Septem

ber 6, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

12

nuclei, demonstrating that NES-AnkG does not inhibit apoptosis. Thus, the anti-apoptotic 254

activity of AnkG strictly requires its translocation into the nucleus. 255

Neither AnkGR22/23S nor NES-AnkG prevents pathogen-induced apoptosis. 256

Next, we asked whether AnkG delivered into the host cell by the T4SS also depends on 257

nuclear localization to exert its anti-apoptotic activity. Because C. burnetii harbors 258

several anti-apoptotic effector proteins, the construction of an ankG deletion-mutant 259

complemented or not with nes-ankG might not provide an answer to this question. 260

Instead we employed a gain-of-function analysis using Legionella pneumophila ΔflaA to 261

determine whether translocation of different AnkG mutants could prevent apoptosis. L. 262

pneumonia ΔflaA caused rapid apoptosis in mouse bone marrow-derived dendritic cells 263

(DCs) and, thus, could not replicate in these cells (21). These pathogen-induced 264

incidents were blocked by adding AnkG to the repertoire of L. pneumophila effector 265

proteins (20). Therefore, this model can be used to analyze whether AnkG has to bind to 266

p32 or whether AnkG has to migrate into the nucleus to inhibit pathogen-induced 267

apoptosis. We infected DCs with L. pneumophila ΔflaA containing either the empty 268

vector (pJV400), AnkG (pJV400-AnkG), NES-AnkG (pJV400-NES-AnkG) or AnkGR22/23S 269

(pJV400-AnkGR22/23S). At 2h and 10h post-infection, the cells were lysed and bacterial 270

colony forming units were counted. As shown in Fig. 5A, bacterial uptake was not 271

affected by the addition of AnkG or any of the AnkG mutants. At 10h post-infection, only 272

12% of the initial inoculum of L. pneumophila ΔflaA containing vector alone were 273

recovered, suggesting that these bacteria induce apoptosis in their host cells and, thus, 274

are not able to survive and replicate (Fig. 5B). In contrast, nearly 50% of the L. 275

pneumophila ΔflaA encoding AnkG were recovered, suggesting that AnkG delivered into 276

the host cell by the L. pneumophila T4SS is able to disrupt pathogen-induced apoptosis 277

on Septem

ber 6, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

13

in DC, in agreement with a previous report (20). Neither L. pneumophila ΔflaA encoding 278

NES-AnkG nor L. pneumophila ΔflaA encoding AnkGR22/23S seemed to inhibit pathogen-279

induced apoptosis in DCs, as demonstrated by recovery rates of less than 10%. These 280

results support our previous findings and suggest that AnkG depends on binding to p32 281

for proper localization and trafficking into the nucleus and that the nuclear localization is 282

essential for inhibition of host cell apoptosis. 283

The intracellular trafficking, but not the anti-apoptotic activity of AnkG, 284

depends on binding to p32. The previous experiments did not clarify whether the 285

binding to p32 is also necessary for AnkG-mediated anti-apoptotic activity. To address 286

this question we constructed a chimera by fusing the SV40 large T antigen nuclear 287

localization signal (29) to the amino-terminus of AnkGR22/23S (GFP-NLS-AnkGR22/23S). 288

Ectopic expression of this construct displays nuclear localization (Fig. 5C). Next, we 289

ectopically produced GFP, GFP-AnkG and GFP-NLS-AnkGR22/23S transiently in CHO 290

cells and treated the cells with staurosporine to induce cell death. Nuclear fragmentation 291

was visualized by DAPI staining and counted to measure apoptosis. Whereas 35% of 292

cells expressing GFP had fragmented nuclei, this number was reduced to 23% in cells 293

expressing GFP-AnkG (Fig. 5D). Importantly, 22% of cells expressing GFP-NLS-294

AnkGR22/23S had fragmented nuclei. Thus, AnkG depends on binding to p32 for proper 295

localization and trafficking, but not for anti-apoptotic activity. 296

297

Discussion 298

The elimination of infected cells via apoptosis is an evolutionarily conserved 299

defense mechanism (30). So it is not surprising that many intracellular pathogens have 300

developed mechanisms to counter apoptosis-induction by their host cells (16). Several 301

on Septem

ber 6, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

14

intracellular pathogens inject effector proteins into the host cell to prevent premature 302

host cell death. However, their molecular mechanisms of action are distinct (31). Here 303

we analyzed the anti-apoptotic activity of AnkG. We showed that the effector protein 304

AnkG localizes in association with the host cell mitochondria in unstressed cells (Fig. 305

2A). This is in contrast to a report showing that mCherry-AnkG co-localized with 306

microtubules (32). The difference in localization of AnkG cannot be explained by the cell 307

line used, because both studies used HeLa cells. The only other difference is the tag 308

used. However, we have not detected any tag-dependent differences in the intracellular 309

localization of AnkG so far. GFP-, HA- and myc-tagged AnkG all displayed the same 310

intracellular localization in HeLa and CHO-FcR cells (data not shown). Interestingly, 311

ectopically expressed GFP-AnkGR22/23S co-localized with tubulin (Fig. 3C), and thus 312

displays the same intracellular localization as reported for mCherry-AnkG (32). This 313

localization is surprising, as one would predict that AnkG unable to bind p32 would 314

display cytoplasmic localization. Furthermore, after staurosporine treatment GFP-NES-315

AnkG displays partial co-localization with tubulin (data not shown). Therefore, it can be 316

speculated that microtubule-association might play a role in AnkG activity under certain 317

cellular conditions. 318

There are several anti-apoptotic type III or type IV secretion system effector 319

proteins that target the host cell mitochondria, the central organelle of the intrinsic 320

apoptotic pathway. Such targeting of the mitochondria by bacterial proteins seems to be 321

evolutionarily conserved, as plant pathogens also target the mitochondria to suppress 322

the hypersensitive response, a form of programmed cell death (33). As shown in Fig. 2A 323

AnkG only partially co-localized with mitochondria, suggesting that this effector protein is 324

not transported into the mitochondria, as it has been shown for Ats1 and PorB. Ats1 325

on Septem

ber 6, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

15

from Anaplasma phagocytophilum uses the mitochondrial import machinery to get 326

transported into the mitochondria (34), while the meningococcal PorB associate with a 327

porin located in the outer mitochondrial membrane (35). Importantly, the anti-apoptotic 328

activity of Ats1 correlates with mitochondrial import (34). AnkG, in clear contrast, has to 329

get transported into the nucleus to act anti-apoptotically (Fig. 4D). Interestingly, this 330

transport into the nucleus, which depends on the ability of AnkG to bind to p32, only 331

happens under apoptotic or stress conditions (Fig. 2B and 3D). This leads to the 332

hypothesis that AnkG primarily targets the mitochondria to sense host cell apoptotic 333

stress and then hitchhikes to the nucleus, the organelle of activity. As a consequence it 334

can be concluded that the activity of AnkG is adjusted by a host cell stress sensor which 335

regulates the transport process. 336

For intracellular trafficking of AnkG from the mitochondria to the nucleus and, 337

thus, for activity control, binding to p32 is essential. This is in agreement with a report 338

that proposed that p32 is involved in bridging a signaling pathway that extends from the 339

mitochondria to the cell nucleus (23). However, once AnkG is within the nucleus, binding 340

to p32 is not needed for anti-apoptotic activity (Fig. 5D). This result suggests that AnkG 341

must instead interfere with a nuclear function to prevent host cell death. There are 342

several effector proteins known to target the host cell nucleus. The Chlamydia 343

trachomatis effector protein NUE is a histone methyltransferase targeting histones (36). 344

AnkA from A. phagocytophilum mediates epigenetic changes at the CYBB promotor 345

(37), leading to a global down-regulation of host defense genes (38). How AnkG 346

modulates nuclear function has to be determined, but the activity of AnkG is clearly 347

regulated by host cell stress signaling and p32-dependent trafficking. This is, to our 348

on Septem

ber 6, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

16

knowledge the first example that an anti-apoptotic effector protein is regulated by host 349

cell protein-mediated trafficking. 350

The question how effector proteins are regulated has only rarely been 351

investigated. There are several avenues of regulation thinkable: 1) regulation by time 352

point and dosage of translocation; 2) regulation by modulation through other effector 353

proteins; 3) regulation by host cell dependent modification (phosphorylation, lipidation, 354

sumoylation etc.). Examples already exist for the latter scenario. It was shown that 355

intracellular localization, and thereby the function of Legionella pneumophila effector 356

proteins containing a CAAX motif are affected by lipidation through the host cell 357

farnesyltransferase and class I geranylgeranyltransferase (39). The Helicobacter pylori 358

T4SS effector protein CagA is phosphorylated by the host cell tyrosine kinases Src and 359

Abl. Phosphorylated CagA can then modulate various signaling cascades associated 360

with cell polarity, cell proliferation, actin-cytoskeletal rearrangements, cell elongation, 361

disruption of tight and adherence junctions, pro-inflammatory responses and apoptosis 362

inhibition (40). Here, we have identified a fourth possibility to regulate the activity of 363

effector proteins: regulation by stress sensing and intracellular trafficking. In our opinion, 364

more knowledge about host cell requirements for regulation of effector proteins is 365

needed. This knowledge will not only help to understand microbial pathogenesis better, 366

but will also allow us to develop new strategies for therapy. The first steps down this 367

avenue have already been made. An exemplified study showed that identifying host cell 368

signaling pathways required for bacterial survival might help to control infection (41). 369

370

Acknowledgements 371

on Septem

ber 6, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

17

This work was supported by the Deutsche Forschungsgemeinschaft (DFG) through the 372

Collaborative Research Initiative 796 (SFB796; to A.L. and C.B.) and through the Priority 373

Programme SPP1580 (to A.L.) as well as by the ERA-NET PathoGenoMics 3rd call (to 374

A.L.). We thank Dr. Christian Bogdan for his valuable comments on the manuscript. 375

376

References 377

378

1. Maurin M, Raoult D. 1999. Q fever. Clin. Microbiol. Rev. 12: 518-553. 379

2. Baca OG, Paretsky D. 1983. Q fever and Coxiella burnetii: a model for host-380

parasite interactions. Microbiol. Rev. 47: 127-149. 381

3. Howe D, Melnicakova J, Barak I, Heinzen RA. 2003. Maturation of the Coxiella 382

burnetii parasitophorous vacuole requires bacterial protein synthesis but not 383

replication. Cell. Microbiol. 5: 469-80. 384

4. Howe D, Shannon JG, Winfree S, Dorward DW, Heinzen RA. 2010. Coxiella 385

burnetii phase I and II variants replicate with similar kinetics in degradative 386

phagolysosome-like compartments of human macrophages. Infect. Immun. 78: 387

3465-3474. 388

5. Maurin M, Benoliel AM, Bongrand P, Raoult D. (1992). Phagolysosomal 389

alkalinization and the bactericidal effect of antibiotics: the Coxiella burnetii 390

paradigm. J. Infect. Dis. 166: 1097- 1102. 391

6. Howe D, Mallavia LP. 2000. Coxiella burnetii exhibits morphological change and 392

delays phagolysosomal fusion after internalization by J774A.1 cells. Infect. 393

Immun. 68: 3815-3821. 394

on Septem

ber 6, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

18

7. Romano PS, Gutierrez MG, Beron W, Rabinovitch M, Colombo MI. 2007. The 395

autophagic pathway is actively modulated by phase II Coxiella burnetii to 396

efficiently replicate in the host cell. Cell. Microbiol. 9: 891-909. 397

8. Beare PA, Gilk SD, Larson CL, Hill J, Stead CM, Omsland A, Cockrell DC, 398

Howe D, Voth DE, Heinzen RA. 2011. Dot/Icm type IVB secretion system 399

requirements for Coxiella burnetii growth in human macrophages. MBio 2: 400

e00175-00111. 401

9. Carey KL, Newton HJ, Lührmann A, Roy CR. 2011. The Coxiella burnetii 402

Dot/Icm system delivers a unique repertoire of type IV effectors into host cells and 403

is required for intracellular replication. PLoS Pathog. 7: e1002056. 404

10. Van Schaik EJ, Chen C, Mertens K, Weber MM, Samuel JE. 2013. Molecular 405

pathogenesis of the obligate intracellular bacterium Coxiella burnetii. Nat. Rev. 406

Microbiol. 11: 561-573. 407

11. Lamkanfi M, Dixit VM. 2010. Manipulation of host cell death pathways during 408

microbial infections. Cell Host Microbe. 8: 44-54. 409

12. Byrne GI, Ojcius DM. 2004. Chlamydia and apoptosis: life and death decisions 410

of an intracellular pathogen. Nat. Rev. Microbiol. 2: 802-808. 411

13. Hotchkiss RS, Strasser A, McDunn JE, Swanson PE. 2009. Cell death. N. 412

Engl. J. Med. 361: 1570-1583. 413

14. Gallucci S, Lolkema M, Matzinger P. 1999. Natural adjuvants: endogenous 414

activators of dendritic cells. Nat. Med. 5: 1249-1255. 415

15. Elliott MR, Ravichandran KS. 2010. Clearance of apoptotic cells: implications in 416

health and disease. J. Cell. Biol. 189: 1059-1070. 417

on Septem

ber 6, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

19

16. Faherty CS, Maurelli AT. 2008. Staying alive: bacterial inhibition of apoptosis 418

during infection. Trends Microbiol. 16: 173-180. 419

17. Lührmann A, Roy CR. 2007. Coxiella burnetii inhibits activation of host cell 420

apoptosis through a mechanism that involves preventing cytochrome c release 421

from mitochondria. Infect. Immun. 75: 5282-5289. 422

18. Voth DE, Howe D, Heinzen RA. 2007. Coxiella burnetii inhibits apoptosis in 423

human THP-1 cells and monkey primary alveolar macrophages. Infect. Immun. 424

75: 4263-4271. 425

19. Klingenbeck L, Eckart RA, Berens C, Lührmann A. 2013. The Coxiella burnetii 426

type IV secretion system substrate CaeB inhibits intrinsic apoptosis at the 427

mitochondrial level. Cell. Microbiol. 15: 675-687. 428

20. Lührmann A, Nogueira CV, Carey KL, Roy CR. 2010. Inhibition of pathogen-429

induced apoptosis by a Coxiella burnetii type IV effector protein. Proc. Natl. Acad. 430

Sci. U.S.A. 107: 18997-19001. 431

21. Dedio J, Jahnen-Dechent W, Bachmann M, Müller-Esterl W. 1998. The 432

multiligand-binding protein gC1qR, putative C1q receptor, is a mitochondrial 433

protein. J. Immunol. 160: 3524-3542. 434

22. Matthews DA, Russell WC. 1998. Adenovirus core protein V interacts with 435

p32—a protein which is associated with both the mitochondria and the nucleus. J. 436

Gen. Virol. 79: 1677-1685. 437

23. Brokstad KA, Kalland KH, Matthews DA, Russell WC. 2001. Mitochondrial 438

protein p32 can accumulate in the nucleus. Biochem. Biophys. Res. Commun. 439

281: 1161-1169. 440

on Septem

ber 6, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

20

24. Robles-Flores M, Rendon-Huerta E, Gonzalez-Aguilar H, Mendoza-441

Hernandez G, Islas S, Mendoza V, Ponce-Castaneda MV, Gonzalez-Mariscal 442

L, Lopez-Casillas F. 2002. p32 (gC1qBP) is a general protein kinase C (PKC)-443

binding protein; interaction and cellular localization of P32-PKC complexes in ray 444

hepatocytes. J. Biol. Chem. 277: 5247-5255. 445

25. Hall KT, Giles MS, Calderwood MA, Goodwin DJ, Matthews DA, Whitehouse 446

A. 2002. The Herpesvirus Saimiri open reading frame 73 gene product interacts 447

with the cellular protein p32. J. Virol. 76: 11612-11622. 448

26. Beatch MD, Everitt JC, Law LJ, Hobman TC. 2005. Interactions between 449

rubella virus capsid and host protein p32 are important for virus replication. J. 450

Virol. 79: 10807-10820. 451

27. Mukherjee S, Shields D. 2009. Nuclear import is required for the pro-apoptotic 452

function of the Golgi protein p115. J. Biol. Chem. 284: 1709-1717. 453

28. Nogueira CV, Lindsten T, Jamieson AM, Case CL, Shin S, Thompson CB, 454

Roy CR. 2009. Rapid pathogen-induced apoptosis: a mechanism used by 455

dendritic cells to limit intracellular replication of Legionella pneumophila. PLoS 456

Pathog. 6: e1000478. 457

29. Kalderon D, Roberts BL, Richardson WD, Smith AE. 1984. A short amino acid 458

sequence able to specify nuclear location. Cell. 39: 499-509. 459

30. Ashida H, Mimuro H, Ogawa M, Kobayashi T, Sanada T, Kim M, Sasakawa C. 460

2011. Cell death and infection: a double-edged sword for host and pathogen 461

survival. J. Cell Biol. 195: 931-942. 462

on Septem

ber 6, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

21

31. Raymond B, Young JC, Pallett M, Endres RG, Clements A, Frankel G. 2013. 463

Subversion of trafficking, apoptosis, and innate immunity by type III secretion 464

system effectors. Trends Microbiol. 21: 430-441. 465

32. Voth DE, Howe D, Beare PA, Vogel JP, Unsworth N, Samuel JE, Heinzen RA. 466

2009. The Coxiella burnetii ankyrin repeat domain-containing protein family is 467

heterogeneous, with C-terminal truncations that influence Dot/Icm-mediated 468

secretion. J. Bacteriol. 191: 4232-4242. 469

33. Rudel T, Kepp O, Kozjak-Pavlovic V. 2010. Interaction between bacterial 470

pathogens and mitochondrial cell death pathways. Nat. Rev. Microbiol. 8: 693- 471

34. Niu H, Kozjak-Pavlovic V, Rudel T, Rikihisa Y. 2010. Anaplasma 472

phagocytophilum Ats-1 is imported into host cell mitochondria and interferes with 473

apoptosis induction. PLoS Pathog. 6: e1000774. 474

35. Massari P, Ho Y, Wetzler LM. 2000. Neisseria meningitidis porin PorB interacts 475

with mitochondria and protects cells from apoptosis. Proc. Natl. Acad. Sci. U.S.A. 476

97: 9070-9075. 477

36. Pennini ME, Perrinet S, Dautry-Varsat A, Subtil A. 2010. Histone methylation 478

by NUE, a novel nuclear effector of the intracellular pathogen Chlamydia 479

trachomatis. PLoS Pathog. 6: e1000995. 480

37. Garcia-Garcia JC, Rennoll-Bankert KE, Pelly S, Milstone AM, Dumler JS. 481

2009. Silencing of host cell CYBB gene expression by the nuclear effector AnkA 482

of the intracellular pathogen Anaplasma phagocytophilum. Infect. Immun. 77: 483

2385-2391. 484

on Septem

ber 6, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

22

38. Garcia-Garcia JC, Barat NC, Trembley SJ, Dumler JS. 2009. Epigenetic 485

silencing of host cell defense genes enhances intracellular survival of the 486

rickettsial pathogen Anaplasma phagocytophilum. PLoS Pathog. 5: e1000488. 487

39. Ivanov SS, Charron G, Hang HC, Roy CR. 2010. Lipidation by the host 488

prenyltransferase machinery facilitates membrane localization of Legionella 489

pneumophila effector proteins. J. Biol. Chem. 285: 34686-34698. 490

40. Tegtmeyer N, Wessler S, Backert S. 2011 Role of the cag-pathogenicity island 491

encoded type IV secretion system in Helicobacter pylori pathogenesis. FEBS J 492

278: 1190-1202. 493

41. Kuijl C, Savage ND, Marsman M, Tuin AW, Janssen L, Egan DA, Ketema M, 494

van den Nieuwendijk R, van den Eeden SJ, Geluk A, Poot A, van der Marel 495

G, Beijersbergen RL, Overkleeft H, Ottenhoff TH, Neefjes J. 2007. Intracellular 496

bacterial growth is controlled by a kinase network around PKB/AKT1. Nature 450: 497

725-730. 498

499

Figure legends 500

Figure 1: AnkG binds directly to the host cell protein p32 and does not alter its steady 501

state protein level. (A) Glutathione–sepharose columns with GST-AnkG or GST alone 502

were incubated with His-p32. Eluate (E1-E4) and bead (beads) fractions were resolved 503

by SDS-PAGE and stained with Coomassie blue. (B) Glutathione–sepharose columns 504

with GST-AnkG or GST alone were incubated with His-p32. Input, eluate (E1-E4) and 505

bead (beads) fractions were subjected to immunoblot analysis using anti-GST and anti-506

p32 antibodies. (C) Ni-NTA agarose columns with His-p32 were incubated with GST or 507

with GST-AnkG. Eluate (E1-E4) and input were subjected to immunoblot analysis using 508

on Septem

ber 6, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

23

anti-GST and anti-His antibodies. (D) HEK293 cells were transfected with plasmids 509

encoding GFP or GFP-tagged AnkG. Protein extracts were separated by SDS-PAGE, 510

transferred to a PVDF membrane and probed with antibodies directed against GFP, p32 511

and actin. One representative immunoblot out of at least three independent experiments 512

is shown. (E) HeLa cells were transiently transfected with plasmids encoding GFP or 513

GFP-tagged AnkG. The cells were treated with Mitotracker (red) followed by fixation and 514

permeabilization. P32 was stained with a specific primary antibody and a secondary 515

dye405 labeled antibody (blue). Figure 2: Intracellular localization of AnkG. (A) 516

Representative immunofluorescence micrographs show HeLa cells transiently 517

transfected with a plasmid encoding GFP-tagged AnkG (green). The cells were treated 518

with Mitotracker (red) followed by fixation, permeabilization and staining of the nuclei 519

with DAPI (blue). (B) CHO-FcR cells transiently transfected with GFP-tagged AnkG were 520

incubated with 2µM staurosporine. After the indicated time-points cells were fixed and 521

the intracellular localization of AnkG was analyzed in at least 100 transfected cells per 522

sample using confocal microscopy from eight independent experiments. * p< 0.001, n.s. 523

not significant (p=0.055) (C) Representative immunofluorescence micrographs show 524

CHO-FcR cells expressing GFP-tagged AnkG, -AnkG1-69 or -AnkG70-338 (green). The 525

cells were incubated with staurosporine followed by fixation, permeabilization and 526

staining of the nuclei with DAPI (blue). 527

Figure 3: Identification of the p32 binding site. (A and B) HEK293 cells were co-528

transfected with plasmids encoding GFP-tagged p32 and the indicated HA-tagged AnkG 529

mutants (HA-tagged AnkF was used as negative control). The proteins were precipitated 530

from the cell lysates with an anti-GFP antibody. Immunoblot analysis was used to detect 531

p32 (anti-GFP) and Ank-proteins (anti-HA) in the lysates (pre-IP) and precipitates (IP). 532

on Septem

ber 6, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

24

(C) Representative immunofluorescence micrographs show HeLa cells expressing GFP-533

tagged AnkGR22/23S (green). The cells were fixed, permeabilized and stained with anti-534

tubulin antibody (red) and DAPI (blue). (D) CHO-FcR cells expressing GFP-tagged 535

AnkGR22/23S were incubated with 2µM staurosporine. After the indicated time points cells 536

were fixed and the localization of AnkG was analyzed in at least 100 transfected cells 537

per sample from four independent experiments using confocal microscopy. n.s. not 538

significant 539

Figure 4: AnkG has to migrate into the nucleus to inhibit staurosporine-induced 540

apoptosis. (A) Representative immunofluorescence micrograph show CHO-FcR cells 541

expressing GFP-tagged NES-AnkG (green). The cells were treated with Mitotracker 542

(red) followed by fixation, permeabilization and staining of the nuclei with DAPI (blue). 543

(B) HEK293 cells were co-transfected with plasmids encoding the indicated GFP-tagged 544

AnkG mutants or GFP as negative control. The proteins were precipitated from the cell 545

lysates with an anti-GFP antibody. Immunoblot analysis was used to detect endogenous 546

p32 (anti-p32) or AnkG (anti-AnkG) in the lysates (pre-IP) and precipitates (IP). (C) 547

CHO-FcR cells expressing GFP-tagged NES-AnkG were incubated with 2µM 548

staurosporine. After the indicated time-points cells were fixed and the localization of 549

AnkG was analyzed in at least 100 transfected cells per sample from three independent 550

experiments using confocal microscopy. n.s. not significant (D) CHO-FcR cells 551

expressing GFP, GFP- AnkG or -NES-AnkG were treated with staurosporine for 4h. The 552

cells were fixed, permeabilized and the nuclei were stained with DAPI. The nuclear 553

morphology of at least 100 GFP expressing cells was scored in four independent 554

experiments. n.s. not significant * p< 0.02. 555

on Septem

ber 6, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

25

Figure 5: Neither AnkGR22/23S nor NES-AnkG can prevent pathogen-induced apoptosis 556

(A/B) Dendritic cells were infected with Legionella pneumophila ΔflaA containing the 557

indicated plasmids. The data shown are from one representative experiment of three 558

experiments with similar results. Shown is the bacterial uptake after 2 hours infection (A) 559

and the relative number of intracellular bacteria 10 hours after infection compared to the 560

2 hour value. **p= 0.001 (C) Representative immunofluorescence micrograph shows 561

CHO-FcR cells expressing GFP-NLS-AnkGR22/23S (green) incubated with mitotracker 562

(red) followed by fixation, permeabilization and staining of the nuclei with DAPI (blue). 563

(D) CHO-FcR cells expressing GFP, GFP-AnkG or GFP-NLS-AnkGR22/23S were treated 564

with 2µM staurosporine for 4h. After treatment the cells were fixed, permeabilized and 565

the nuclei were stained with DAPI. The nuclear morphology was scored of at least 100 566

GFP expressing cells in three independent experiments. * p< 0.01. 567

568

Tables 569

570 Plasmid Primer Reference AnkGFL-pCMV-HA 20 AnkG∆Ank-pCMV-HA 20 AnkG1-69-pCMV-HA 20 AnkG70-338-pCMV-HA 20 AnkG1-91-pCMV-HA 329/339 This study AnkG50-338-pCMV-HA 331/340 This study AnkG1-157-pCMV-HA 307/310 This study AnkGR23S-pCMV-HA 590/591 This study AnkGR22/23S-pCMV-HA 590/592 This study AnkF-pCMV-HA 79/229 This study pGEX-5X Amersham AnkG-pGEX-5X 20 p32-pET16b 184/185 This study pEGFP Clontech AnkG-pEGFP 20 p32-pEGFP 20 AnkG1-69-pEGFP 329/338 This study AnkG70-338-pEGFP 332/340 This study

on Septem

ber 6, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

26

AnkGR22/23S-pEGFP - This study NES-AnkG-pEGFP 665/34 This study NLS-AnkGR22/23S-pEGFP 746/400 This study pJV400 20 pJV400-AnkG 20 pJV400-AnkGR22/23S 373/26 This study pJV400-NES-AnkG 696/26 This study Table 1: Primer numbers are as in Table 2. 571

572 Number Sequence Site 26 5' AAGGCGCGCCTCACCGAGGACTAGACAG AscI 34 5’ AAGGATCCTCACCGAGGACTAGACAGA BamHI 79 5’ CCGGTACCCTACCGCTGGAAGCCGC KpnI 184 5’ CCCATATGCTGCACACCGACGGAGAC NdeI 185 5’ CCGGATCCCTACTGGCTCTTGACAAAACT BamHI 229 5’ CCGAATTCATGTGCAATACCAACATGTCT EcoRI 307 5’ CCGGTACCATGAGTAGACGTGAGACTCC KpnI 310 5’ CCGGTACCTTATTTATATTTGATTTTCACATCAGC KpnI 329 5' CCAAGATCTCTATGAGTAGACGTGAGACTCC BglII 330 5’ CCAAGATCTCTATGGGACATCCTGTAAGAAGAAG BglII 331 5’ CCAAGATCTCTATGTCGTTTGAAATACTCATAAATGC BglII 332 5’ CCAAGATCTCTATGCTTCGCGGGGATTCTTTTCA BglII 338 5' CCGGTACCTCAGTAGTTTTTTATTATGCTCAAGCT KpnI 339 5’ CCGGTACCTCAGAAATCCGTCTTTGGCGGTA KpnI 340 5' CCGGTACCTCACCGAGGACTAGACAGA KpnI 373 5' CCGGCCGGCCATGAGTAGACGTGAGACTCC FseI 400 5' CCGGTACCTCACCGAGGACTAGACAGA KpnI 590 P-5’ CGTTGAGGATATTGTGCTAGTGGGAGTCTACGTCTAC

TCAT -

591 P-5’ CGACAGGAACTCGAACGCCGAGAAGTAGATTGAGCC GAAAA

-

592 P-5’CGACAGGAACTCGAACGCCGAGTAGTAGATTGAGC CGAAAA

-

665 5’ CCGGTACCGCCTCCAGCAGCCTCCCCTGGAGGACTGACCCTGAGTAGACGTGAGACTCCCACTAGC

KpnI

696 5’ CCGGCCGGCCATGCTCCAGCTGCCTCCCC FseI 746 5’ CCACTCAGATCTCTCCTAAGAAGAAAAGGAAGGTTAGT

AGACGTGAGACTCCCACTAGCACAA

BglII

Table 2. Underlined denotes the location of the restriction site. 573

on Septem

ber 6, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

A GST-AnkG + His-p32

E1 E2 E3 E4 Beads

GST + His-p32

GST-AnkG

HIS-p32

GST

GST-tag pull-down

B

E1 E2 E3 E4 BeadsGST

His

-p32

anti-GST

anti-p32

GST-tag pull-downinput

GST-tag pull-downinput

anti-GST

anti-p32

GST-

AnkG

His

-p32

E1 E2 E3 E4 Beads

HIS-tag pull-down input

anti-GST

anti-HIS

GST-

AnkG

HIS

-p32

E1 E2 E3 E4

GST-AnkG + HIS-p32C D

anti-GFP

anti-p32

anti-actin

GFP

GFP

-AnkG

E1 E2 E3 E4 Beads

EMerge

Merge

p32

p32 GFP-AnkG

GFP

Mitotracker

Mitotracker

10µm

10µm

on Septem

ber 6, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

0

25

50

75

100

125

Mock 30 120 240

% p

he

no

typ

e

min staurosporine

Mitochondrial

associa!on

nuclear

***

A

B

C

GFP-AnkG Mitotracker

Merge Zoom

GFP-AnkG GFP-AnkG1-69 GFP-AnkG70-338

Mock

Staurosporine

***n.s.

10µm

10µm10µm 10µm

10µm10µm10µm

on Septem

ber 6, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

0

25

50

75

100

Mock 30 120 240

% p

he

no

typ

e

min staurosporine

Co-localiza!on

with Tubulin

nuclear

amino acid 18 19 20 21 22 23 24 25 26 27 28

AnkG T R T P R R R L S R K

AnkG T R T P R S R L S R K

AnkG T R T P S S R L S R K

R23S

R22/23S

HA-AnkG

HA-AnkG

HA-AnkG

HA-AnkG

HA-AnkG

HA-AnkG

HA-AnkG

HA-AnkG

HA-AnkF

HA-AnkG

HA-AnkG

HA-AnkG

HA-AnkF

HA-AnkG

HA-AnkG

HA-AnkG

pre-IP

IP

anti-HA

anti-HA

anti-GFP

WT

∆Ank

1-157

1-91

1-69

29-338

50-338

70-338

anti-HA

anti-GFP

D

C

BA

GFP-AnkG αTubulin MergeR22/23S

R22S

R22/23S

R23S

R22/23S

WT

WT

pre-IP IP

n.s.n.s.

n.s.

10µm

on Septem

ber 6, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

0

25

50

75

100

Mock 30 120 240

% p

he

no

typ

e

min staurosporine

cytosolic

nuclear

GFP-NES-AnkG Mitotracker Merge

anti-AnkG

anti-p32

IPpre-IP

GFP

GFP-NES-AnkG

GFP-AnkG

GFP-AnkG

GFP

GFP-AnkG

GFP-AnkG

GFP-NES-AnkG

R22/23S

R22/23S

n.s.

10

20

30

40

50

% fr

agm

en

ted

nu

clei

GFP

GFP-AnkG

GFP-NES-AnkG

D

C

B

A

*

n.s.

n.s.n.s.

10µm10µm

on Septem

ber 6, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

0

10

20

30

40

50

60

% s

urvi

val

0

5

10

15

20

25

bact

eria

l *10

3 /wel

l

pJV400

pJV400-A

nkG

pJV400-A

nkG

pJV400-N

ES-AnkG

R22/23S

pJV400

pJV400-A

nkG

pJV400-A

nkG

pJV400-N

ES-AnkG

R22/23S

GFP-NLS-AnkG Mitotracker Merge

0

10

20

30

40

% f

rag

me

nte

d n

ucl

ei

GFPGFP-AnkG

GFP-NLS-AnkG

R22/23S

n.s.

*

* *

D

C

A B

on Septem

ber 6, 2018 by guesthttp://iai.asm

.org/D

ownloaded from