AcpA: A Francisella Acid Phosphatase Affecting Intramacrophage ...

2 downloads 0 Views 350KB Size Report
Oct 23, 2006 - 4 virulence, a deletion of acpA was constructed in Francisella novicida. The phosphatase. 5 ... MglA regulates several virulence factors within the. 20 .... For Southern blot analysis, PCR products were labeled with digoxigenin as. 16 ..... Saslaw S, Eigelsbach HT, Wilson HE, Prior JA, Carhart S. 1961. 13.
IAI Accepts, published online ahead of print on 23 October 2006 Infect. Immun. doi:10.1128/IAI.01226-06 Copyright © 2006, American Society for Microbiology and/or the Listed Authors/Institutions. All Rights Reserved.

1

AcpA: A Francisella Acid Phosphatase Affecting

2

Intramacrophage Survival and Virulence

3 4

Nrusingh P. Mohapatra, Ashwin Balagopal, Shilpa Soni, Larry S. Schlesinger, and John

5

S. Gunn*

6

The Center for Microbial Interface Biology; Department of Molecular Virology,

7

Immunology, and Medical Genetics; and Department of Internal Medicine, Division of

8

Infectious Diseases, The Ohio State University, Columbus, OH 43210 USA

T P

E C

9 10 11 12 13

D E

C A

Running title: Francisella acid phosphatase AcpA

14

*corresponding author

15

The Ohio State University

16

Tzagournis Medical Research Facility

17

420 W. 12th Ave.

18

Columbus, OH 43210-1214

19

Phone: 614-292-6036

20

Fax: 614-292-5495

21

E-mail: [email protected]

1

1 2

Abstract

3

AcpA of Francisella spp. is a respiratory burst-inhibiting acid phosphatase that also

4

exhibits phospholipase C activity. To better understand the molecular basis of AcpA in

5

virulence, a deletion of acpA was constructed in Francisella novicida. The phosphatase

6

and lipase activities were reduced 10-fold and 8-fold, respectively, in the acpA mutant

7

versus the wild type, and were found mostly associated with the outer membrane. The

8

acpA mutant was more susceptible to intracellular killing than the wild type strain in the

9

THP-1 human macrophage-like cell line. In addition, mice infected with the acpA mutant

D E

T P

10

survived longer than the wild type strain and were less fit than the wild type strain in

11

competition infection assays. Transmission electron microscopy showed that the acpA

12

mutant was delayed in escape from macrophage phagosomes, as more than 75% of acpA

13

mutant bacteria could still be found inside phagosomes after 12 hrs of infection in THP-1

14

cells and human monocyte-derived macrophages, whereas most of the wild type bacteria

15

had escaped from the phagosome by 6 hrs post-infection. Thus, AcpA affects intracellular

16

trafficking and the fate of Francisella within host macrophages.

E C

C A

2

Introduction 1

Francisella tularensis is a gram negative, facultative intracellular pathogen that causes

2

tularemia in humans and other mammals including rodents (38). The two primary human

3

pathogens are F. tularensis subspecies tularensis (type A strain) and F. tularensis

4

subspecies holarctica (type B strain). The type A strain is predominantly found in North

5

America, highly infectious and causes a life threatening disease in humans, especially

6

when inhaled (10). Type B strains are found primarily in Europe and are considered to be

7

less virulent for mammals than the type A strain (10). An attenuated live vaccine strain

8

(LVS) was derived from a type B strain (12) and it elicits a protective response in

9

humans, monkeys, guinea pigs and mice against systemic challenge with virulent type A

10

F. tularensis (9, 12, 13, 18, 35, 36). Francisella novicida (FnWT), a close relative of type

11

A F. tularensis, is a much less frequent cause of infection in humans than either type A or

12

type B (19). Nevertheless, F. novicida and F. tularensis subspecies tularensis and

13

holarctica all cause a lethal systemic infection in mice when inoculated by most routes

14

(19).

15

D E

T P

E C

C A

The virulence mechanisms of this bacterium are not clear, although the products

16

of several genes such as mglA and the pathogenicity island genes iglC, iglD and pdpA-D

17

have been identified that help Francisella to survive inside macrophages (6, 15, 17).

18

However, the exact functions of these genes are not known. MglA shares homology with

19

the SspA of E. coli, which regulates the stationary phase gene transcription by interacting

20

with RNA polymerase (40). MglA regulates several virulence factors within the

21

pathogenicity island including iglC (25), which is important for the ability of Francisella

22

spp. to escape from the phagosome. Several studies have shown that F. tularensis

3

1

resides inside a membrane-bound phagosome during its initial growth in a macrophage

2

and that it is released into the cytoplasm during a later phase of growth (2, 11, 16).

3

Acid phosphatases are ubiquitous in nature and are present in almost all bacteria.

4

These enzymes have been identified and characterized from many eukaryotes and

5

prokaryotes and divided into subgroups according to their substrate specificity, molecular

6

weight and sensitivity to known inhibitors (30). Acid phosphatases catalyze the

7

hydrolysis of phospho-monoesters at an acidic pH. In several species they have been

8

implicated as virulence factors and help the bacteria to survive inside phagocytes (4, 7,

9

14, 24, 27, 28, 31), often by inhibiting the respiratory burst (4, 20, 24, 29, 31).

10

D E

T P

E C

The published genome sequence of F. tularensis Schu 4 revealed the presence of

11

four acid phosphatases (acpA, FTT0221; acpB, FTT0156; acpC, FTT0620; and hap,

12

FTT1662c [a pseudogene in Schu 4 but not LVS]) (21). AcpA (57 kDa) is a polyspecific

13

periplasmic acid phosphatase that is highly expressed by F. tularensis (7, 27) and shows

14

no significant global amino acid sequence similarity with any protein in the Protein Data

15

Bank (8). This protein is also unusual in that it exhibits phospholipase C activity (27).

16

Earlier studies reported that Francisella AcpA has respiratory burst inhibiting properties

17

and broad substrate specificity (27). It has also been shown that a transposon insertion in

18

the 3’ region of the acpA open reading frame did not result in an intramacrophage

19

survival defect or a loss of virulence (7). In the present study, we constructed a deletion

20

of the entire acpA gene in FnWT and analyzed its role in intracellular trafficking in

21

macrophages and virulence in mice.

C A

4

Materials and Methods 1

Bacterial Strain, Plasmid Construction and Molecular Biology Techniques

2

Bacterial strains, plasmids and primers used in this study are listed in Table-1. F.

3

novicida U112 was routinely grown at 37°C in cysteine heart agar (CHA) (Hi-Media

4

Laboratories, India) and modified Tryptic Soy Broth (TSB) (Difco Laboratories, Detroit,

5

Mich.) containing 135ug/ml ferric pyrophosphate and 0.1% cysteine hydrochloride. CHA

6

containing 5% defibrinated sheep blood and 135ug/ml ferric pyrophosphate was used for

7

transformation studies (Hemostat Laboratories, Dixon, Calif.). When required, the growth

8

medium for FnWT was supplemented with kanamycin (25 µg/ml) or tetracycline (12.5

9

µg/ml). All manipulations with Francisella spp. were performed in a class II biological

D E

T P

E C

10

safety laboratory. E. coli DH5α was grown at 37°C aerobically in Luria-Bertani (LB)

11

medium (Difco Laboratories, Detroit, Mich.) supplemented with kanamycin (15 µg/ml),

12

tetracycline (12.5 µg/ml), or ampicillin (100 µg/ml) when required. All antibiotics and

13

chemicals were purchased from Sigma-Aldrich (St. Louis, Mo.).

14

C A

Chromosomal DNA preparation, ligation, E. coli transformation, and Southern

15

blotting were performed according to methods described by Sambrook et al (32). To

16

construct the pAcpA-Kan plasmid, upstream and downstream regions of acpA were

17

generated by PCR with primers JG996/JG997 and JG998/JG999 respectively from FnWT

18

genomic DNA. The 772-bp upstream fragment was digested with SacI and KpnI, cloned

19

into dephosphorylated SacI and KpnI digested pUC18, and renamed pAcpUp. Similarly

20

871-bp downstream region of acpA was digested with PstI and SphI, cloned into

21

dephosphorylated PstI and SphI digested pAcpUp plasmid, and renamed pAcpUpDn. A

22

Kan cassette driven by the F. tularensis outer membrane protein (YP_169847) promoter

5

1

was amplified from plasmid pDSK519 using primers JG868 /JG1048, digested with KpnI

2

and PstI, ligated into KpnI/PstI digested and dephosphorylated pAcpUpDn vector, and

3

renamed pAcpA-Kan. Plasmid pAcpA-Kan was transformed into FnWT by

4

cryotransformation. In brief, 1 µg of plasmid was mixed with FnWT cells at an OD600 of

5

0.5 in transformation buffer (10mM MOPS, 75mM CaCl2, 10mM RbCl2 and 15%

6

glycerol, pH adjusted to 6.5 with 1N KOH), incubated on ice for 30 min. and flash frozen

7

in liquid nitrogen for 5 min. Cells were thawed at room temperature and plated on CHA-

8

Kan25 and incubated at 37° C. This resulted in the creation of strain JSG2660

9

(∆acpA::kan). Growth curves were identical for strains JSG2660 and FnWT. For

10

complementation in trans, plasmid pKK214 containing the groEL promoter of F.

11

tularensis LVS was used (1). The acpA gene was generated by PCR using primers

12

JG1016a/JG1017a and cloned into the EcoRI and PstI sites of pKK214groEL such that

13

acpA was expressed from the groEL promoter. The resulting plasmid pAcpA, was

14

introduced into JSG2660 by cryotransformation described as above creating strain

15

JSG2661.

16

D E

T P

E C

C A

For Southern blot analysis, PCR products were labeled with digoxigenin as

17

instructed by the manufacturer (Roche, Indianpolis, USA). Probes were hybridized to

18

EcoRI digested chromosomal DNA of FnWT and Fn∆acpA followed by anti-

19

digoxigenin-AP-conjugate antibody treatment. Membranes were developed with

20

NBT/BCIP solution.

21 22

Cell fractionation

6

1

Overnight stationary phase cultures of wild type (JSG1819) and mutant (JSG2660)

2

bacteria in TSB-0.1% cysteine hydrochloride were centrifuged at 8000 x g for 20 min at

3

4° C. The cell pellet was washed twice with PBS and sonicated at a constant output of

4

60 W for a total of 300 s. Cell debris and unbroken cells were removed by centrifugation

5

at 3000 x g for 15 min at 4° C. The supernatant was centrifuged at 25,000 x g for 24 h at

6

18° C in a 2.1::1.4::0.7 M sucrose gradient to separate the outer and inner membrane

7

fractions. The non-membrane containing fraction was used as the cytosolic fraction.

8

These fractions were assayed for phosphatase activity as described by Aragon et al. (3)

9

Western blot analysis was performed by standard protocols on protein fractions using

10

D E

T P

E C

anti-AcpA polyclonal sera (gift of Tom Reilly).

11 12

Intramacrophage Survival Assays

13

FnWT or Fn∆acpA strains were used to infect the following cell lines: J774.1 murine

14

macrophages, NR8383 rat alveolar macrophages, and PMA-induced (10 ng/ml) THP-1

15

human macrophages at an MOI of ~50:1. Wells were seeded with ~2 x105 macrophages

16

and ~1.0x107 bacteria were added to each well. After 2 h incubation at 37° C, 5% CO2,

17

gentamicin (50 µg/ml) was added to the medium to eliminate extracellular organisms.

18

Wells were washed twice with PBS and incubated with their respective media

19

supplemented with 10 µg/ml gentamicin. The macrophage cells were lysed with 0.1%

20

SDS at 2h, 12h and 24 h post infection, the lysates were immediately serially diluted in

21

PBS and plated on CHA plates for determination of viable counts. Experiments were

22

performed in triplicate on a minimum of three separate occasions with similar results.

C A

23

7

1

Transmission Electron Microscopy

2

Monolayers of monocyte-derived macrophages (MDMs) (23) or THP-1 cell lines were

3

incubated with FnWT (JSG1819) or Fn∆acpA (JSG2660) at an MOI of 500:1 in a plastic

4

four-well chamber slide. This high MOI was used to enhance the number of macrophages

5

in the population that were infected, which in turn aided visualization. After 2, 6 and 12

6

hrs of incubation at 370 C, 5% CO2, the wells were washed and fixed immediately with

7

2.5% warmed glutaraldehyde for 5 min followed by a combination of 2.5%

8

glutaraldehyde and 1% osmium tetroxide in 0.1M sodium cacodylate, pH 7.3 for 15 min

9

at 4° C (11). The cells were then stained with 0.25% uranyl acetate in a 0.1M sodium

D E

T P

E C

10

acetate buffer at pH 6.3 for 45 minutes. Monolayers were washed with ice cold normal

11

saline and the chambers were then removed. Slides were dehydrated through a graded

12

series of ethanol, rinsed in HPMA (hydroxypropylmethylacrylate) and infiltrated with

13

Polybed 812. They were embedded by up-ending resin-filled beam capsules over the

14

cells and polymerized at 60° C for 24 h. Thin sections cut with a Leica EM UC6

15

ultramicrotome were collected on Formvar coated copper grids, stained with uranyl

16

acetate and lead citrate and viewed by transmission electron microscopy in a Philips

17

CM12 at 60 kV. Multiple fields were examined for bacteria, and identified bacteria were

18

determined to be intraphagosomal or intracytosolic. The criterion for intraphagosomal

19

was the visualization of greater than 50% of the phagosomal membrane surrounding the

20

bacterium.

C A

21

8

1

Mouse Survival Studies

2

Groups of five female 4 to 6 week-old BALB/c mice (Harlan Sprague) were inoculated at

3

a dose of ~100 CFU delivered in 100 µl PBS by the intraperitoneal route. Actual bacterial

4

counts delivered were determined by plate count from each inoculum. Mice were

5

monitored for four days post-infection. For competition assays, wild type (JSG1819) and

6

mutant (JSG2660) bacteria were inoculated into BALB/c mice at a 1:1 ratio (100 CFU of

7

each in 100 µl total) and at four days post inoculation, organs were harvested, macerated,

8

and plated onto appropriate solid media to select for each of the competing strains. The

9

competitive index was calculated as the number of mutant CFU/FnWT CFU recovered.

D E

T P

E C

C A

9

1

Results

2

Construction of a Fn∆acpA mutant

3

A modified cryotransformation technique was developed that increased the efficiency of

4

transformation 100-1000 fold in comparison to the standard Francisella MgCl2-KCl

5

cryotransformation or electroporation. This technique was used to introduce pAcpA-Kan,

6

a non-replicating suicide vector carrying a DNA fragment with a kanamycin cassette in

7

the place of a complete deletion of acpA, into FnWT. After 5-days growth on CHA-Kan25

8

plates, several colonies were screened by PCR and Southern blot resulting in the

9

identification of more than 85% of the clones with the correct deletion and loss of the

10

D E

T P

E C

vector sequences (Figure 1A).

11 12

Comparative measurements of acid phosphatase and lipase activity

13

FnWT and Fn∆acpA cells were disrupted and separated by sucrose gradient to recover

14

inner and outer membrane fractions. Western blot analysis was performed which showed

15

the presence of AcpA in whole cell lysates and the outer membrane fraction of FnWT

16

(Figure 1B). AcpA was absent in all fractions analyzed from the Fn∆acpA strain. A

17

single band of similar molecular weight was detected in a Schu 4 whole cell lysate,

18

demonstrating its expression in Type A virulent Francisella. Acid phosphatase activities

19

of the Fn cell-free extracts and inner and outer membrane fractions were measured.

20

Using 6,8-difluoro-4-methylumbelliferyl phosphate as the substrate, we found no

21

difference in phosphatase activities of cell-free extracts/cytosolic fractions or inner

22

membrane fractions between the mutant and wild-type strains, but a 10-fold decrease in

23

phosphatase activity of the mutant outer membrane fraction (Figure 2A). This was

C A

10

1

consistent with the observed location of the enzyme in the outer membrane fraction

2

(Figure 1B). Complementation of Fn∆acpA with a plasmid containing the acpA gene

3

restored outer membrane phosphatase activity to the wild-type level (Figure 2A). Using

4

p-nitrophenyl palmitate as the substrate, we also measured an 8-fold decrease in

5

phospholipase activity recovered from the outer membrane fraction of Fn∆acpA

6

compared to FnWT (Figure 2B). These results are consistent with the predicted secretion

7

of AcpA into the periplasmic space and suggest a tight association with the outer

8

membrane. In addition, these data demonstrate that Fn∆acpA has significantly reduced

9

enzymatic activities that are associated with AcpA.

D E

T P

E C

10 11

Fn∆acpA is defective in intramacrophage survival/replication

12

The primary cellular target of F. tularensis during infection is the macrophage (26, 39).

13

To examine the role that AcpA may play in protecting Francisella from phagocytic

14

killing and intracellular trafficking, we measured survival of Fn∆acpA in the THP-1 cell

15

line. While the FnWT and Fn∆acpA complemented strains replicated within the THP-1

16

cells, the Fn∆AcpA mutant showed about a half-log reduction over the 12 hours post

17

infection, with a slight recovery to the initial infection numbers by 24 hrs (Figure 3). The

18

experiment was not continued beyond 24 hrs due to a loss in macrophage viability.

19

Similar results were observed in murine and rat macrophage cell lines (data not shown).

20

These results demonstrate that AcpA plays a role in intramacrophage survival and/or

21

replication of FnWT.

C A

22 23

Electron microscopy demonstrates delayed phagosomal escape of the acpA mutant

11

1

To further delineate the function of AcpA in intramacrophage survival, we used

2

transmission electron microscopy to visualize PMA-stimulated THP-1 cells and MDMs

3

infected with either FnWT or Fn∆acpA (Figures 4 & 5). At two hours post-infection in

4

both macrophages, nearly all of the mutant and wild-type bacteria were contained in

5

membrane-bound vacuoles, consistent with the known early compartmentalization of the

6

pathogen in phagosomes (Figures 4A & 5A). After six hours, over half of FnWT had

7

escaped from phagosomes (Figures 4B & 5B) and by 12 hours post infection, almost

8

none of the wild-type pathogen was contained inside vacuoles with distinct membranes

9

boundaries in THP-1 cells (Figures 4C). Studies with MDMs revealed similar findings,

D E

T P

E C

10

except that the escape of FnWT at 12 hours was slower, such that 40% were still in intact

11

membranes (versus 75% of the Fn∆acpA, Figure 5C). In sharp contrast, we observed

12

very little degradation of vacuoles containing Fn∆acpA during the 12-hour time frame of

13

the experiments. After 12 hours, approximately 75 % of Fn∆acpA was still contained in

14

vacuoles having distinct membrane borders (Figures 4D & 5D). These results suggest

15

that AcpA plays a role in intramacrophage survival/replication via disruption of the

16

phagosomal membrane that allows access of Francisella to the host cell cytosol.

17

C A

18

The Fn∆acpA mutant has reduced virulence in the mouse model

19

To examine the role of AcpA in an animal model of infection, we measured survival rates

20

of mice infected with FnWT and Fn∆acpA. After 36 hours following intraperitoneal

21

infection, only 20 % of the mice infected with FnWT had survived, while 80 % of the

22

mice infected with Fn∆acpA survived (Figure 6). At the 48-hour time point, all mice

23

infected with FnWT were dead, while 60 % of those infected with Fn∆acpA survived.

12

1

However by 72 hours post infection, all mice had died. Competition assays corroborated

2

the apparent decrease in Fn∆acpA virulence, demonstrating a consistent competitive

3

index of ~0.17 in both liver and spleen. These data indicate that loss of AcpA activity

4

results in a less virulent pathogen, which is presumably due to decreased

5

intramacrophage survival. Because mice infected with Fn∆acpA eventually died, it is

6

likely that AcpA acts in concert with other factors to disrupt normal intracellular

7

trafficking and that the influence of AcpA is largest in the initial stages of infection.

D E

T P

8 9

Discussion

E C

10

Francisella AcpA possesses physical and chemical properties unlike other bacterial acid

11

phosphatases (20, 31). This 57–kDa enzyme hydrolyzes a wide variety of physiologically

12

meaningful substrates, including phosphorylated tyrosine peptides, inositol phosphates,

13

phospholipid-like molecules, AMP, ATP, fructose 1,6-bisphosphate, glucose and fructose

14

6-phosphosphates, NADP and ribose 5-phosphate, but fails to possess significant

15

similarity to other acid phosphatases in the protein database (27-28). It has been recently

16

crystallized (14) and the completed structure of this enzyme will likely provide important

17

clues to its biological functions.

18

C A

Many acid phosphatases have been shown or predicted to play a role in virulence,

19

most often in intracellular pathogens, by the inhibition of a respiratory burst. Such

20

activity has been reported for AcpA of FnWT (27), as well as for Coxiella burnetii (4),

21

Legionella spp. (31) and Leishmania (29). In this study, we described the role of

22

Francisella AcpA, which has dual acid phosphatase and phospholipase C activities, in

23

virulence and phagosomal trafficking in macrophages.

13

1

In this work, we constructed a complete deletion of the acpA open reading frame.

2

A previous study of AcpA in FnWT concluded that, upon the chromosomal deletion of a

3

300 bp 3’ region and the replacement of this region by an erythromycin cassette, there

4

was no effect upon intramacrophage survival or virulence (7). We were therefore

5

somewhat surprised by the phenotypes observed due to the complete deletion of acpA. It

6

is possible that, since the entire acpA orf was not deleted in the previous study, a

7

functional truncated protein was still produced. With regard to acid phosphatase activity,

8

this may be unlikely, as the peak of phosphatase activity in protein fractions associated

9

with AcpA was absent in their mutant. However, it is possible that the truncated protein

D E

T P

E C

10

retained phospholipase activity, and it is this activity that is important in phagosomal

11

trafficking.

12

It was shown that the phosphatase and phospholipase activities of the Fn∆acpA

C A

13

mutant had 10-fold and 8-fold less activity than FnWT respectively, indicating that AcpA

14

contributed to the overall activity but is not the only phosphatase or phospholipase in

15

FnWT. Indeed, enzyme assays have shown at least two peaks of phosphatase activity in

16

crude protein fractions (7) and genome scanning shows three other acid phosphatases in

17

FnWT (unpublished observations). The future characterization and mutagenesis of these

18

additional acid phosphatases will help to define the individual contributions of each

19

enzyme to the biology of Francisella.

20

We determined the growth kinetics of the acpA mutant versus the FnWT in

21

various macrophage cell lines where we showed that the acpA mutant exhibited a 10- to

22

20-fold reduction in survival in THP-1 cells. This phenotype was due to the loss of acpA,

23

as the complemented mutant strain revealed intramacrophage growth kinetics identical to

14

1

the FnWT strain. To explore the survival or growth of the acpA mutant in vivo, mice were

2

infected intraperitoneally (and intranasally with similar results, data not shown). We

3

found that mice infected with the acpA mutant survived longer than mice infected with

4

the FnWT strain and there was a corresponding reduction in organ bacterial load. When

5

mice were infected with a mixture of FnWT and acpA mutant strains at a ratio of 1:1, the

6

acpA mutant strain was less competitive than the FnWT strain (competitive index of

7

~0.17 in both liver and spleen). These data suggest that AcpA contributes to virulence. It

8

is possible that a greater virulence defect will be observed upon the deletion of multiple

9

acid phosphatase genes.

D E

T P

E C

10

It has been shown that Francisella spp are initially contained within a phagosome

11

but later escape into cytoplasm (11, 34-33). The factors involved in and the mechanism of

12

phagosomal escape are largely unknown, but IglC and MglA have been shown to play a

13

role (33). We found that in THP-1 cells and MDMs, the Fn∆acpA mutant was primarily

14

still located inside the phagosome at 6 hrs post infection and only poorly escaped into the

15

cytoplasm after 12 hrs of infection, whereas, FnWT was nearly 100% and 60% escaped

16

from phagosomes by 12 hrs of infection in THP-1s and MDMs, respectively. These data

17

suggest that AcpA plays a role in intramacrophage trafficking, and the inability of the

18

mutant to escape from the phagosome likely contributes to its decreased intramacrophage

19

survival and virulence.

C A

20

Because AcpA has numerous enzymatic targets, a challenge for the future is to

21

determine which of these enzymatic activities underlies the observed respiratory burst

22

suppression and altered phagosomal escape due to AcpA. One possibility is that AcpA

23

hydrolyzes phospholipids of the phagosomal inner membrane, which might compromise

15

1

membrane integrity. Alternatively, AcpA might affect host signaling pathways by

2

dephosphorylation of host proteins, inositol phosphates or phosphoinositides, the latter

3

being critically important for phagosome formation (37) and respiratory burst activation

4

(5). A third hypothesis is that AcpA activity within the bacterium helps to induce or

5

amplify the activity of other proteins that are essential for phagosome disruption, such as

6

IglC and its regulator MglA (33, 22). In this context, the polyspecificity and high

7

abundance of AcpA may point to a phosphate scavenger role for the enzyme (41). Based

8

on structural, enzymatic and phenotypic properties, inhibitors of AcpA and other acid

9

phosphatases may be developed that, when used in combination with other antimicrobial

10

D E

T P

E C

agents, would provide alternative therapy against F. tularensis infection.

11

C A

12

Acknowledgements

13

This work was supported by National Institutes of Health grants: U56AI057164

14

(Midwest Center of Excellence (MWCE): Transmission/Pathogenesis of Bioterrorism

15

Agents to LSS), U54AI057153 (Region V "Great Lakes" Regional Center of Excellence

16

in Biodefense and Emerging Infectious Diseases Consortium (GLRCE) to LSS and JSG),

17

U54AI057156 (Western Regional Center of Excellence for Biodefense and Emerging

18

Infectious Diseases to JSG) and an NIH institutional training grant AI 0155411 (to AB).

19

We thank Dr. Karen Elkins and Fran Nano for providing guidance and Francisella

20

strains, and Jack Tanner and Tom Reilly for their advice and critical reading of this

21

manuscript.

22

16

1

References

2

1. Abd H, Johansson T, Golovliov I, Sandström G, Forsman M. 2003.

3

Survival and Growth of Francisella tularensis in Acanthamoeba castellanii.

4

Appl Environ Microbiol 69: 600-06.

5 6

D E

2. Anthony LD, Burke RD, Nano FE. 1991. Growth of Francisella spp. in rodent macrophages. Infect. Immun 59: 3291-3296.

T P

7

3. Aragon V, Kurtz S, Flieger A, Neumeister B, Cianciotto NP. 2000.

8

Secreted enzymatic activities of wild-type and pilD-deficient Legionella

9

pneumophila. Infect Immun 68: 1855-63.

E C

10

4. Baca OG, Roman MJ, Glew RH, Christner RF, Buhler JE, Aragon AS.

11

1993. Acid phosphatase activity in Coxiella burnetii: a possible virulence

12 13 14 15 16

C A

factor. Infect Immun 61: 4232-9.

5. Baggiolini M, Wymann MP. 1990. Turning on the respiratory burst. Trends Biochem Sci 15: 69-72.

6. Baron, GS & Nano, FE. 1998. MglA and MglB are required for the intramacrophage growth of Francisella novicida. Mol Microbiol. 29: 247-59.

17

7. Baron GS, Reilly TJ, Nano FE. 1999. The respiratory burst-inhibiting acid

18

phosphatase AcpA is not essential for the intramacrophage growth or

19

virulence of Francisella novicida. FEMS Microbiol Lett 176: 85-90.

17

1

8. Berman HM, Westbrook J, Feng Z, Gilliland G, Bhat TN, Weissig H,

2

Shindyalov IN, Boune PE. 2001. The Protein Data Bank. Nucleic Acids Res.

3

28: 235-42.

4

9. Chen W, Shen H, Webb A, KuoLee R, Conlan JW. 2003. Tularemia in

5

BALB/c and C57BL/6 mice vaccinated with Francisella tularensis LVS and

6

challenged intradermally, or by aerosol with virulent isolates of the pathogen;

7

protection varies depending on pathogen virulence, route of exposure, and

8

host genetic background. Vaccine 21: 3690-3700.

D E

T P

9

10. Chu, MC, and Weyant R. 2003. Francisella and Brucella, Manual of clinical

10

microbiology, 8th ed. Washington, D.C: American Society for Microbiology.

11

789-797p.

12 13 14 15 16

E C

C A

11. Clemens DL, Lee BY, Horwitz MA. 2004. Virulent and avirulent strains of Francisella tularensis prevent acidification and maturation of their

phagosomes and escape into the cytoplasm in human macrophages. Infect Immun 72: 3204-17.

12. Eigelsbach HT, Downs CM. 1961. Prophylactic effectiveness of live and

17

killed tularemia vaccines. I. Production of vaccine and evaluation in the white

18

mouse and guinea pig. J Immunol 87: 415-425.

19

13. Eigelsbach HT, Tulis J, Overholt EL, Griffith WR. 1961. Aerogenic

20

immunization of the monkey and guinea pig with live tularemia vaccine. Proc

21

Soc Exp Biol Med 108: 732-734.

18

1

14. Felts RL, Reilly TJ, Tanner JJ. 2005. Crystallization of AcpA, a respiratory

2

burst-inhibiting acid phosphatase from Francisella tularensis. Biochim

3

Biophys Acta 1752: 107-10.

4

15. Golovliov, I, Baranov V, Krocova Z, Kovarova H, Sjostedt A. 2003. An

5

attenuated strain of the facultative intracellular bacterium Francisella

6

tularensis can escape the phagosome of monocytic cells. Infect. Immun 71:

7

5940-5950.

D E

T P

8

16. Golovliov I, Sjostedt A, Mokrievich A, Pavlov VA. 2003. Method for allelic

9

replacement in Francisella tularensis. FEMS Microbiol Lett. 222: 273-80.

10

17. Gray, CG, Cowley, SC, Cheung, KK, Nano, FE. 2002. The identification of

11

five genetic loci of Francisella novicida associated with intracellular growth.

12

FEMS Microbiol Lett. 215: 53-6.

13 14 15 16 17

E C

C A

18. Hodge FA, Leif WR, Silverman MS. 1968. Susceptibility to infection with Pasteurella tularensis and the immune response of mice exposed to continuous low dose rate gamma radiation. Natl Radiol Defense Lab Trans 68: 1-28.

19. Kieffer T, Cowley S, Nano FE, Elkins KL. 2003. Francisella novicida LPS

18

has greater immunobiological activity in mice than F. tularensis LPS, and

19

contributes to F. novicida murine pathogenesis. Microbes Infect 5: 397-403.

20

20. Kondo E, Kurata T, Naigowit P, Kanai K. 1996. Evolution of cell-surface

21

acid phosphatase of Burkholderia pseudomallei. Southeast Asian J Trop Med

22

Public Health 27: 592-9.

19

1

21. Larsson P, Oyston PCF, Chain P, Chu MC, Duffield M, Fuxelius HH,

2

Garcia E, Hälltorp G, Johansson D, Isherwood KE, Karp PD, Larsson E,

3

Ying Liu, Michell S, Prior J, Prior R, Malfatti S, Sjöstedt A, Svensson K,

4

Thompson N, Vergez L, Wagg JK, Wren BW, Lindler LE, Andersson

5

SGE, Forsman M and Titball RW. 2005. The complete genome sequence of

6

Francisella tularensis, the causative agent of tularemia. Nat Genet 37: 153-9.

D E

T P

7

22. Lauriano CM, Barker JR, Yoon SS, Nano FE, Arulanandam BP, Hassett

8

DJ, Klose KE. 2004. MglA regulates transcription of virulence factors

9

necessary for Francisella tularensis intraamoebae and intramacrophage

10 11 12 13 14 15 16

E C

survival. Proc Natl Acad Sci U S A 101: 4246-9.

23. McCarthy TR, Torrelles JB, MacFarlane AS, Katawczik M, Kutzbach B,

C A

DesJardin LE, Clegg S, Goldberg JB and Schlesinger LS. 2005. Overexpression

of

phosphomannomutase

Mycobacterium

that

increases

tuberculosis phosphatidylinositol

manB,

a

mannoside

biosynthesis in Mycobacterium smegmatis and mycobacterial association with human macrophages. Mol Microbiol. 58: 774-90.

17

24. Markova N, Kussovski V, Radoucheva T. 1998. Killing of Pseudomonas

18

pseudomallei by polymorphonuclear leukocytes and peritoneal macrophages

19

from chicken, sheep, swine and rabbits. Zentralbl Bakteriol 288: 103-10.

20

25. Nano FE, Zhang N, Cowley SC, Klose KE, Cheung KKM, Roberts MJ,

21

Ludu JS, Letendre GW, Meierovics AI, Stephens G and Elkins KL. 2004.

20

1

A Francisella tularensis pathogenicity island required for intramacrophage

2

growth. J Bacteriol 186: 6430-6.

3 4

26. Oyston PC, Sjostedt A, Titball RW. 2004. Tularaemia: bioterrorism defence renews interest in Francisella tularensis. Nat Rev Microbiol 2: 967-78.

D E

5

27. Reilly TJ, Baron GS, Nano FE, Kuhlenschmidt MS. 1996. Characterization

6

and sequencing of a respiratory burst-inhibiting acid phosphatase from

7

Francisella tularensis. J Biol Chem 271: 10973-83.

T P

8

28. Reilly TJ, Felts RL, Henzl MT, Calcutt MJ, Tanner JJ. 2006.

9

Characterization of recombinant Francisella tularensis acid phosphatase A.

10 11 12 13 14 15

E C

Protein Expr Purif 45: 132-41.

C A

29. Remaley AT, Glew RH, Kuhns DB, Basford RE, Waggoner AS, Ernst LA, Pope M. 1985. Leishmania donovani: surface membrane acid phosphatase blocks neutrophil oxidative metabolite production. Exp Parasitol 60: 331-41.

30. Rossolini GM, Schippa S, Riccio ML, Berlutti F, Macaskie LE, Thaller

16

MC. 1998. Bacterial nonspecific acid phosphohydrolases: physiology,

17

evolution and use as tools in microbial biotechnology. Cell Mol Life Sci 54:

18

833-50.

19

31. Saha AK, Dowling JN, LaMarco KL, Das S, Remaley AT, Olomu N, Pope

20

MT and Glew RH.1985. Properties of an acid phosphatase from Legionella

21

1

micdadei which blocks superoxide anion production by human neutrophils.

2

Arch Biochem Biophys 243: 150-60.

3 4

32. Sambrook J, Fritsch EF, Maniatis T. 1989. Molecular cloning, 2nd ed. Cold Spring Harbor, N.Y: Cold Spring Harbor Laboratory Press.

D E

5

33. Santic M, Molmeret M, Abu Kwaik Y. 2005. Modulation of biogenesis of

6

the Francisella tularensis subsp. novicida-containing phagosome in quiescent

7

human macrophages and its maturation into a phagolysosome upon activation

8

by IFN-gamma. Cell Microbiol 7: 957-67

T P

E C

9

34. Santic M, Molmeret M, Klose KE, Jones S, Kwaik YA. 2005. The

10

Francisella tularensis pathogenicity island protein IglC and its regulator

11

MglA are essential for modulating phagosome biogenesis and subsequent

12 13 14 15

C A

bacterial escape into the cytoplasm. Cell Microbiol 7: 969-79.

35. Saslaw S, Eigelsbach HT, Wilson HE, Prior JA, Carhart S. 1961. Tularemia vaccine study. I. Intracutaneous challenge. Arch Int Med 107: 689701.

16

36. Shen H, Chen W, Conlan JW. 2004. Susceptibility of various mouse strains

17

to systemically- or aerosol-initiated tularemia by virulent type A Francisella

18

tularensis before and after immunization with the attenuated live vaccine

19

strain of the pathogen. Vaccine 22: 2116–21.

20

37. Simonsen A, Wurmser AE, Emr SD, Stenmark H. 2001. The role of

21

phosphoinositides in membrane transport. Curr Opin Cell Biol 13: 485-92.

22

1 2

38. Tärnvik A. 1989. Nature of protective immunity to Francisella tularensis. Rev Infect Dis 11: 440-451.

3

39. Titball RW, Johansson A, Forsman M. 2003. Will the enigma of

4

Francisella tularensis virulence soon be solved? Trends Microbiol 11: 118-

5

23.

D E

6

40. Williams MD, Ouyang TX, Flickinger MC. 1994. Glutathione S-

7

transferase-sspA fusion binds to E. coli RNA polymerase and complements

8

delta sspA mutation allowing phage P1 replication. Biochem Biophys Res

9

Commun. 201: 123-7.

T P

E C

10

41. Yang K, Metcalf WW. 2004. A new activity for an old enzyme: Escherichia

11

coli bacterial alkaline phosphatase is a phosphite-dependent hydrogenase.

12

C A

Proc Natl Acad Sci U S A 101: 7919-24

23

1

Figure Legends

2

Figure 1. Construction and confirmation of the ∆acpA mutant. (A) Southern blot

3

analysis of the chromosomal DNA of the ∆acpA mutant vs. the FnWT. DNAs were

4

digested with EcoRI and probed with the acpA orf (lanes 1 and 2) or the kan cassette

5

(lanes 3 and 4) showing the expected differential hybridization of the probe. An EcoRI

6

site within the acpA orf results in two hybridizing bands, while no EcoRI sites are within

7

the acpA orf. Lane 1 and 3, F. novicida; lane 2 and 4, F. novicida ∆acpA. (B) Western

8

blot analysis of whole cell lysates (WC), outer membrane fractions (OM) and inner

9

membrane fractions (IM) of FnWT, Fn∆acpA, and Schu 4 detecting the 57 kDa AcpA

10

D E

T P

E C

protein. Rabbit polyclonal anti-AcpA sera was used as the primary antibody.

11 12

Figure 2. Acid phosphatase (A) and lipase activity (B) of inner membrane (IM), outer

13

membrane (OM) and cytoplasmic (Cyt) fractions of Fn∆acpA (gray bars) versus the

14

Fn∆acpA complemented (white bars) and parental strains (black bars). Data represents

15

the mean +/- the standard deviation of three independent experiments each with duplicate

16

wells.

17

C A

18

Figure 3. Intramacrophage survival assays performed in THP-1 human macrophage-like

19

cell lines with Fn∆acpA (square), ∆acpA complemented (triangle) and parental strains

20

(diamond). Data represents the mean +/- the standard deviation of three independent

21

experiments each with duplicate wells for each time point. Asterisks denote significant

22

differences of the ∆acpA mutant vs. the FnWTat 12 and 24 hrs. post-infection (p