Staphylococcal Alpha-Toxin Is Not Sufficient To Mediate Escape from Phagolysosomes in Upper-Airway...

15
INFECTION AND IMMUNITY, Sept. 2009, p. 3611–3625 Vol. 77, No. 9 0019-9567/09/$08.000 doi:10.1128/IAI.01478-08 Copyright © 2009, American Society for Microbiology. All Rights Reserved. Staphylococcal Alpha-Toxin Is Not Sufficient To Mediate Escape from Phagolysosomes in Upper-Airway Epithelial Cells Bernd Giese, 1 Silvia Dittmann, 1 Kerstin Paprotka, 1 Katja Levin, 1 Annett Weltrowski, 1 Diana Biehler, 1 Thie ˆn-Trí La ˆm, 2 Bhanu Sinha, 2 and Martin J. Fraunholz 1 * Competence Center for Functional Genomics, Ernst Moritz Arndt University, F. L. Jahnstrasse 15, D-17487 Greifswald, Germany, 1 and Institute of Hygiene and Microbiology, University of Wu ¨rzburg, Josef Schneider Str. 2, Bldg. E1, D-97080 Wu ¨rzburg, Germany 2 Received 3 December 2008/Returned for modification 11 May 2009/Accepted 8 June 2009 Intracellular Staphylococcus aureus has been implicated in the establishment of chronic infections. It is therefore imperative to understand by what means S. aureus is able to survive within cells. Here we use two expression systems with a fluorescent readout to assay alpha-toxin expression and function within phagoly- sosomes of infected upper-airway epithelial cells: avirulent Staphylococcus carnosus TM300 and phenotypically alpha-toxin-negative S. aureus laboratory strains. Data from CFU recovery assays suggest that the presence of alpha-toxin is not beneficial for the intracellular survival of recombinant Staphylococcus strains. This finding was corroborated by immunofluorescence studies: whereas S. carnosus and S. aureus are able to deliver S. aureus alpha-toxin to lumina of host cell phagolysosomes, the membrane integrity of these organelles was not affected. Alpha-toxin-expressing strains were detected exclusively within lysosome-associated membrane pro- tein 1 (LAMP1)-yellow fluorescent protein (YFP)-positive vesicles. Measurements of intraphagosomal pH illustrated that all infected phagolysosomes acidified regardless of alpha-toxin expression. In contrast, S. aureus expressing Listeria monocytogenes listeriolysin O leads to the breakdown of the phagolysosomal mem- brane, as indicated by staphylococci that are not associated with LAMP1-YFP-decorated vesicles and that do not reside within an acidic cellular environment. Thus, our results suggest that staphylococcal alpha-toxin is not sufficient to mediate phagolysosomal escape in upper-airway epithelial cells. Staphylococcus aureus is readily internalized by nonprofes- sional phagocytes including keratinocytes and endothelial or epithelial cells (2, 29, 39, 40, 50). Internalization is mediated by the cell wall-linked adhesins fibronectin-binding protein A (FnBPA) and FnBPB (50), via an 5 1 integrin-dependent pathway (51). Upon invasion, S. aureus-containing endosomes eventually fuse with phagolysosomes, where a large portion of the internalized bacteria is inactivated by host cells (34, 47). A fraction of bacteria is able to persist within the host cells as so-called small-colony variants, and the ability of S. aureus to persist within human host cells has been implicated in the establishment of chronic infections (e.g., reviewed in refer- ences 17, 42, 43, and 48). Cytotoxic strains of S. aureus seem to follow a different strategy: it was previously shown that these strains escape the phagolysosomal vesicles and ultimately lead to host cell death (6, 29, 30, 49). In order to understand the contributions of single virulence factors to intracellular survival strategies of S. aureus such as phagolysosomal escape, detailed analyses on a gene-by-gene basis are necessary. Previous stud- ies suggested that staphylococcal alpha-toxin is involved in the induction of host cell death (12, 24, 25, 56) and phagolysoso- mal escape (30). Furthermore, alpha-toxin has been described to be a key effector for intracellular pathogen survival within professional phagocytes (35). As a pore former that increases the permeability of cells or vesicles (14, 56) and exhibits en- hanced hemolytic activity at low pH (8, 28), staphylococcal alpha-toxin is a prime candidate for mediating phagolysosomal escape (30, 35, 49). The toxin is secreted by S. aureus in a monomeric 27-kDa form. On target membranes, heptamers form and ultimately perforate the membrane for ions and macromolecules (reviewed in references 9, 21, and 53). In order to investigate if alpha-toxin causes phagolysosomal escape, we used apathogenic Staphylococcus carnosus TM300 (20) as well as noncytotoxic S. aureus laboratory strains to deliver alpha-toxin to phagolysosomes of upper-airway epithe- lial cells. Data from CFU recovery assays suggest that the presence of alpha-toxin is not beneficial for the intracellular survival of recombinant staphylococci. Immunofluorescence assays as well as pH measurements of infected phagolysosomes with confocal fluorescence microscopy demonstrate that the organellar membrane is not disrupted by alpha-toxin. Our re- sults therefore suggest that staphylococcal alpha-toxin is not sufficient to mediate phagolysosomal escape in upper-airway epithelial cells. MATERIALS AND METHODS Bacterial culture. S. aureus and S. carnosus were grown in trypticase soy broth or B2 (45) using appropriate antibiotics unless stated otherwise. For observations of hemolysis, the strains were plated onto Columbia sheep blood agar (SBA) plates (Becton Dickinson, Heidelberg, Germany) supplied with the appropriate antibiotics as well as 200 ng/ml anhydrotetracycline (Acros Organics, Geel, Belgium). Recombinant Escherichia coli TOP10 (Invitrogen) was grown in LB using 100 g/ml ampicillin as a selective antibiotic. All strains used are listed in Table 1. * Corresponding author. Mailing address: FunGene, Competence Cen- ter for Functional Genomics, E. M. Arndt University, F. L. Jahnstrasse 15, D-17487 Greifswald, Germany. Phone: 49 (0)3834 864072. Fax: 49 (0)3834 864073. E-mail: [email protected]. † Supplemental material for this article may be found at http://iai .asm.org/. Published ahead of print on 29 June 2009. 3611 at UNIV BIBLIOTHEK GREIFSWALD on August 19, 2009 iai.asm.org Downloaded from

Transcript of Staphylococcal Alpha-Toxin Is Not Sufficient To Mediate Escape from Phagolysosomes in Upper-Airway...

INFECTION AND IMMUNITY, Sept. 2009, p. 3611–3625 Vol. 77, No. 90019-9567/09/$08.00�0 doi:10.1128/IAI.01478-08Copyright © 2009, American Society for Microbiology. All Rights Reserved.

Staphylococcal Alpha-Toxin Is Not Sufficient To Mediate Escape fromPhagolysosomes in Upper-Airway Epithelial Cells�†

Bernd Giese,1 Silvia Dittmann,1 Kerstin Paprotka,1 Katja Levin,1 Annett Weltrowski,1 Diana Biehler,1Thien-Trí Lam,2 Bhanu Sinha,2 and Martin J. Fraunholz1*

Competence Center for Functional Genomics, Ernst Moritz Arndt University, F. L. Jahnstrasse 15, D-17487 Greifswald,Germany,1 and Institute of Hygiene and Microbiology, University of Wurzburg, Josef Schneider Str. 2,

Bldg. E1, D-97080 Wurzburg, Germany2

Received 3 December 2008/Returned for modification 11 May 2009/Accepted 8 June 2009

Intracellular Staphylococcus aureus has been implicated in the establishment of chronic infections. It istherefore imperative to understand by what means S. aureus is able to survive within cells. Here we use twoexpression systems with a fluorescent readout to assay alpha-toxin expression and function within phagoly-sosomes of infected upper-airway epithelial cells: avirulent Staphylococcus carnosus TM300 and phenotypicallyalpha-toxin-negative S. aureus laboratory strains. Data from CFU recovery assays suggest that the presence ofalpha-toxin is not beneficial for the intracellular survival of recombinant Staphylococcus strains. This findingwas corroborated by immunofluorescence studies: whereas S. carnosus and S. aureus are able to deliver S.aureus alpha-toxin to lumina of host cell phagolysosomes, the membrane integrity of these organelles was notaffected. Alpha-toxin-expressing strains were detected exclusively within lysosome-associated membrane pro-tein 1 (LAMP1)-yellow fluorescent protein (YFP)-positive vesicles. Measurements of intraphagosomal pHillustrated that all infected phagolysosomes acidified regardless of alpha-toxin expression. In contrast, S.aureus expressing Listeria monocytogenes listeriolysin O leads to the breakdown of the phagolysosomal mem-brane, as indicated by staphylococci that are not associated with LAMP1-YFP-decorated vesicles and that donot reside within an acidic cellular environment. Thus, our results suggest that staphylococcal alpha-toxin isnot sufficient to mediate phagolysosomal escape in upper-airway epithelial cells.

Staphylococcus aureus is readily internalized by nonprofes-sional phagocytes including keratinocytes and endothelial orepithelial cells (2, 29, 39, 40, 50). Internalization is mediated bythe cell wall-linked adhesins fibronectin-binding protein A(FnBPA) and FnBPB (50), via an �5�1 integrin-dependentpathway (51). Upon invasion, S. aureus-containing endosomeseventually fuse with phagolysosomes, where a large portion ofthe internalized bacteria is inactivated by host cells (34, 47). Afraction of bacteria is able to persist within the host cells asso-called small-colony variants, and the ability of S. aureus topersist within human host cells has been implicated in theestablishment of chronic infections (e.g., reviewed in refer-ences 17, 42, 43, and 48). Cytotoxic strains of S. aureus seem tofollow a different strategy: it was previously shown that thesestrains escape the phagolysosomal vesicles and ultimately leadto host cell death (6, 29, 30, 49). In order to understand thecontributions of single virulence factors to intracellular survivalstrategies of S. aureus such as phagolysosomal escape, detailedanalyses on a gene-by-gene basis are necessary. Previous stud-ies suggested that staphylococcal alpha-toxin is involved in theinduction of host cell death (12, 24, 25, 56) and phagolysoso-mal escape (30). Furthermore, alpha-toxin has been describedto be a key effector for intracellular pathogen survival within

professional phagocytes (35). As a pore former that increasesthe permeability of cells or vesicles (14, 56) and exhibits en-hanced hemolytic activity at low pH (8, 28), staphylococcalalpha-toxin is a prime candidate for mediating phagolysosomalescape (30, 35, 49). The toxin is secreted by S. aureus in amonomeric 27-kDa form. On target membranes, heptamersform and ultimately perforate the membrane for ions andmacromolecules (reviewed in references 9, 21, and 53).

In order to investigate if alpha-toxin causes phagolysosomalescape, we used apathogenic Staphylococcus carnosus TM300(20) as well as noncytotoxic S. aureus laboratory strains todeliver alpha-toxin to phagolysosomes of upper-airway epithe-lial cells. Data from CFU recovery assays suggest that thepresence of alpha-toxin is not beneficial for the intracellularsurvival of recombinant staphylococci. Immunofluorescenceassays as well as pH measurements of infected phagolysosomeswith confocal fluorescence microscopy demonstrate that theorganellar membrane is not disrupted by alpha-toxin. Our re-sults therefore suggest that staphylococcal alpha-toxin is notsufficient to mediate phagolysosomal escape in upper-airwayepithelial cells.

MATERIALS AND METHODS

Bacterial culture. S. aureus and S. carnosus were grown in trypticase soy brothor B2 (45) using appropriate antibiotics unless stated otherwise. For observationsof hemolysis, the strains were plated onto Columbia sheep blood agar (SBA)plates (Becton Dickinson, Heidelberg, Germany) supplied with the appropriateantibiotics as well as 200 ng/ml anhydrotetracycline (Acros Organics, Geel,Belgium). Recombinant Escherichia coli TOP10 (Invitrogen) was grown in LBusing 100 �g/ml ampicillin as a selective antibiotic. All strains used are listed inTable 1.

* Corresponding author. Mailing address: FunGene, Competence Cen-ter for Functional Genomics, E. M. Arndt University, F. L. Jahnstrasse 15,D-17487 Greifswald, Germany. Phone: 49 (0)3834 864072. Fax: 49(0)3834 864073. E-mail: [email protected].

† Supplemental material for this article may be found at http://iai.asm.org/.

� Published ahead of print on 29 June 2009.

3611

at UN

IV B

IBLIO

TH

EK

GR

EIF

SW

ALD

on August 19, 2009

iai.asm.org

Dow

nloaded from

Cell culture. S9 upper-airway epithelial cells (13) were grown in Dulbecco’smodified Eagle’s medium (DMEM)-F12 medium supplemented with 10% fetalcalf serum (Pan-Biotech, Munich, Germany) and penicillin-streptomycin (100U/ml and 100 �g/ml, respectively) in T75 or T175 tissue culture flasks (Gibco-BRL). Cell lines were grown Mycoplasma free. Mycoplasma testing was per-formed via Hoechst 34580 staining and via PCR using specific primers afterculturing of cells in the absence of antibiotics. Template DNA for Myco-plasma testing was isolated by use of the QiaAmp DNA isolation kit (Qiagen,Hilden, Germany).

CFU recovery assays. One day prior to infection, 1 � 105 S9 cells were seededonto 12-well plates. For microscopic analysis, the cells were seeded onto 18-mmglass coverslips. One hour prior to infection, the medium was changed fromDMEM-F12 medium containing penicillin and streptomycin to infection me-dium (DMEM containing 20 �g/ml chloramphenicol and 100 ng/ml anhydrotet-racycline). All media contained 10% fetal calf serum (Pan-Biotech, Munich,Germany). For the culture of transfected S9 cells, infection medium was sup-plemented with 3 �g/ml blasticidin. Recombinant S. carnosus and S. aureusstrains were grown overnight at 37°C with shaking in the appropriate broth. Theculture was then diluted to an optical density at 540 nm (OD540) of 0.2 in brothcontaining 200 ng/ml anhydrotetracycline (Acros Organics, Geel, Belgium) ex-cept for the cytotoxic S. aureus strain expressing Listeria monocytogenes listerio-lysin O (LLO), which was cultured in medium without an inducer. After reachingan OD540 of 0.6, bacteria were harvested by centrifugation at 3,000 � g for 10min, resuspended in infection medium, and added to the epithelial cells at amultiplicity of infection of 20. One hour after infection, the infection mediumwas replaced by DMEM containing 20 �g/ml chloramphenicol, 200 ng/ml anhy-drotetracycline, 20 �g/ml lysostaphin, and 100 �g/ml gentamicin for 30 min.After the removal of extracellular bacteria, the monolayers were rinsed twicewith phosphate-buffered saline (PBS, pH 7.4) and incubated in infection mediumsupplemented with 100 �g/ml gentamicin, 20 �g/ml chloramphenicol, and 200ng/ml anhydrotetracycline. For enumeration of intracellular bacteria, Host cellmonolayers were lysed 3 h postinfection (p.i.), 24 h p.i., and 4 and 6 days p.i.(dpi). Wells were rinsed three times with PBS followed by incubation in 300�l/well of 1% Triton X-100 in PBS for 2 min to release intracellular bacteria. Theresulting suspension was plated onto nutrient agar for enumeration of CFU.CFus of extracellular bacteria were determined from supernatants. In this case,cell culture medium was replaced with gentamycin-free medium 24 h prior tosampling. Colonies were counted after incubation at 37°C for 48 h (S. aureus) or72 h (S. carnosus).

Western blot analyses. Recombinant strains were inoculated from a culturegrown overnight in fresh medium containing anhydrotetracycline (200 ng/ml)and grown to an OD600 of 1. Ten milliliters of cultures was harvested by cen-trifugation for 10 min at 14,000 rpm. The culture supernatants were centrifugedand passaged through 0.22-�m-pore-size filters (Millipore, Schwalbach, Ger-many), and protein was precipitated overnight at 4°C by adding trichloroaceticacid to a final concentration of 10%. The precipitate was collected by centrifu-gation for 30 min at 14,000 rpm and at 4°C. The pellet was washed with acetoneand ethanol, air dried, and resuspended directly in sodium dodecyl sulfate (SDS)loading buffer.

Bacterial cell pellets were resuspended in 100 �l of a solution containing 50mM Tris-HCl (pH 8.0), 10 mM EDTA, and 100 �g/RNase A (Roth,Karlsruhe, Germany) supplemented with 20 �g/ml lysostaphin (Sigma-Al-drich, Taufkirchen, Germany). After incubation for 1 h at 37°C, 10% SDS wasadded, and the samples were boiled for 30 min. The samples were DNase Itreated (10 �g/ml; Roth, Karlsruhe, Germany) for 30 min at 37°C and spun downfor 20 min at 13,000 rpm. Soluble protein was mixed with SDS loading buffer andboiled for 20 min. After electrophoretic separation on 15% polyacrylamide gels,the bands were transferred onto polyvinylidene difluoride membranes (Roth,Karlsruhe, Germany) with a semidry blot transfer system (Pegasus, Lubeck,Germany) for 35 min at a constant current (0.8 mA/cm2).

For Western blot analyses of S. carnosus-derived proteins, the membrane wasblocked at 4°C overnight with a solution containing PBS, 5 ml/liter Tween 20, and50 g/liter nonfat dry milk. For S. aureus proteins, blocking solution was supple-mented with 2% human serum (Pan-Biotech, Germany) in order to inhibit thenonspecific binding of antibodies by staphylococcal protein A.

Primary antisera (sheep anti-alpha-toxin and rabbit anti-LLO [both fromAbcam, Cambridge, United Kingdom] or rabbit anti-alpha-toxin [Sigma,Taufkirchen, Germany]) were diluted 1:2,000 in PBS-Tween 20, and the blotmembrane was incubated overnight at 4°C. After the removal of primary anti-body by washing three times for 20 min in PBS-Tween 20 and 5 min with PBS,secondary antisera were applied for 1 h at room temperature, and membraneswere finally washed again. Horseradish peroxidase-conjugated antiserum wasdetected using a Lumi-Imager system (Roche Diagnostics, Darmstadt, Germany)using the Pierce SuperSignal West Pico chemiluminescent substrate (ThermoFisher Scientific, Schwerte, Germany) according to the manufacturer’s instruc-tions. Alkaline phosphatase-conjugated serum was detected by equilibration ofthe membrane in 20 ml of a solution containing 100 mM Tris-HCl (pH 9.3), 100mM NaCl, and 5 mM MgCl2 and adding 50 �l 5% (wt/vol) 5-bromo-4-chloro-3-indoxylphosphate in dimethylformamide (DMF) and 66 �l 7.5% (wt/vol) ni-troblue tetrazolium chloride in 70% DMF. The reaction was stopped with dis-tilled H2O, and the membrane was air dried.

Generation of a LAMP1-YFP-expressing epithelial cell line. Lysosome-asso-ciated membrane protein 1 (LAMP1) was amplified from a human cDNA library(kindly provided by Jorg Mostertz, Greifswald, Germany) using primers MF75Lamp1 s (5�-CGCCACCATGGCGGCCCCCGGCAGCGCC-3�) and MF76Lamp1 as (5�-GATAGTCTGGTAGCCTGCGTGACTCC-3�). The resultingLAMP1 PCR product was cloned into pcDNA6.2-YFP-Topo (Invitrogen) ac-cording to the manufacturer’s instructions and was checked by DNA sequencing.The vector was transfected into S9 cells with jetPEI (Biomol, Hamburg, Ger-many), and cells were continuously passaged in the presence of 3 �g/ml blasti-cidin (Sigma, Taufkirchen, Germany). The presence of LAMP1-yellow fluores-cent protein (YFP) was confirmed via fluorescence microscopy.

Generation of staphylococcal vector constructs. All PCRs were performedusing Phusion polymerase (Finnzymes). All enzymes were obtained from NewEngland Biolabs (Frankfurt am Main, Germany), except where explicitly statedotherwise. pALC2084, a Staphylococcus-E. coli shuttle vector expressing GFPuvr,a variant of GFP that is excitable with ultraviolet (uv) light, under the control ofa tetracycline-inducible promoter (4), was kindly provided by Ambrose Cheung

TABLE 1. Bacterial strains used in this study

Species and strain Description Source or reference

E. coli TOP10 �80 �(lacZ)M15 �(argF-lac)U169 endA1 recA1 hsdR17 (rK mK

�) deoR thi-1supE44 gyrA96 relA1

Invitrogen

S. aureus RN4220 8325-4 derivative phenotypically without alpha- and delta-hemolysin; acceptsforeign DNA

33

S. aureus GP269 8325-4 derivative; rsbU� 19S. aureus SA113 �spa 8325-4 derivative; deficient in staphylococcal protein A Christiane Wolz, Tubingen,

GermanyS. carnosus TM300 Apathogenic, food-safety-grade Staphylococcus 20S. carnosus GFP-FnbA S. carnosus expressing FnbA and GFPuvr This workS. carnosus HLA-GFP-FnbA S. carnosus expressing HLA, FnBPA, and GFPuvr This workS. carnosus HLAH35R-GFP-FnbA S. aureus expressing functionally inactive HLAH35R, FnBPA, and GFPuvr This workS. aureus HLA-Cerulean S. aureus RN4220 expressing HLA and Cerulean This workS. aureus HLAH35R-Cerulean S. aureus RN4220 expressing functionally inactive HLAH35R and Cerulean This workS. aureus LLO-Cerulean S. aureus RN4220 expressing L. monocytogenes LLO and Cerulean This workS. aureus HLA-Cerulean �spa S. aureus SA113 �spa expressing HLA and Cerulean This workS. aureus LLO-Cerulean �spa S. aureus SA113 �spa expressing L. monocytogenes LLO and Cerulean This work

3612 GIESE ET AL. INFECT. IMMUN.

at UN

IV B

IBLIO

TH

EK

GR

EIF

SW

ALD

on August 19, 2009

iai.asm.org

Dow

nloaded from

(Dartmouth, NH) and has been modified to adapt the multiple-cloning site. SacI,EcoRI, and SalI were eliminated from the multiple-cloning site. EcoRV,AvrII, AgeI, and PmeI restriction sites (underlined) were introduced viaPCR by amplifying GFPuvr with primers MF18bs (5�-GGTACCAGATCTCGATATCGGCATGCGCGTCGTAGGAGGTGTTTAAACCCTAGG-3�)and MF19as_2084mut (5�-GTGAATTCGGCTTAACTGCAGTTATTTG-3�)and inserting the KpnI/EcoRI restriction fragment into accordingly treatedpALC2084, yielding pGFP. pCerulean was created by replacing the open readingframe (ORF) for green fluorescent protein (GFP) with its cyan fluorescentspectral mutant Cerulean, which was codon adapted for S. aureus expression (ourunpublished data). To introduce the adhesin FnBPA, pGFP was opened at theunique EcoRI site, blunted via Klenow enzyme, and subsequently dephosphor-ylated by Antarctic phosphatase. The fnbA locus of S. aureus GP269 (kindlyprovided by Susanne Engelmann, Greifswald, Germany) was amplified with5�-phosphorylated primers MF109-FnbA-fwd (5�-CAAGCCTTGTGTGACAAGGGTTTCTGATG-3�) and MF110-FnbA-rev (5�-CAGGCTTACTTCATATAATTTATGAAATAAACCC-3�). The resulting PCR product was ligated into theblunted vector, thereby creating pGFP-FnbA. After transformation into E. coliTOP10 cells (Invitrogen), clones were isolated, and successful cloning as well asthe orientation of fnbA were verified via restriction digests.

The ORF for alpha-toxin (hla) was amplified using primers MF28-hla-as (5�-GTTTAAACCTCCGGATGTACTACCATTTGTCATTTCTTC-3�) and MF23-hla-s(5�-CCTAGGGAGGTTTTAAACATGAAAACACGTATAGTCAGC-3�) (re-striction sites are underlined). The fragment was cloned into pCR2.1-Topo(Invitrogen), and the sequence was verified by DNA sequencing. For site-di-rected mutagenesis of hla, the vector was isolated from a recombinant E. coliTOP10 strain via the Qiagen Spin minikit (Qiagen, Hilden, Germany). Mutagen-esis was performed via 17 cycles of PCR using antiparallel primer pairs containingthe desired point mutations (MF84-H35s [5�-GATAAAGAAAATGGCATGCGCAAAAAAGTATTTTATAG-3�] and MF85-H35as [5�-CTATAAAATACTTTTTTGCGCATGCCATTTTCTTTATC-3�]). PCR products were treated with 10 unitsof DpnI restriction endonuclease to degrade methylated and hemimethylatedtemplate DNA. The resulting plasmids were transformed into chemically com-petent E. coli TOP10 cells, and point mutations in the resulting colonies wereverified by DNA sequence analysis. The EcoRI-AvrII fragments of hla or itsfunctionally inactive H35R mutant were ligated into the accordingly linearizedpCerulean or pGFP-FnbA, resulting in plasmids pHLA-Cerulean, pHLAH35R-Cerulean, pHLA-GFP-FnbA, and pHLAH35R-GFP-FnbA, respectively.

pLLO-Cerulean was generated by amplifying L. monocytogenes LLO usingprimers LLOf (5�-GTTTAAACATGAAAAAAATAATGCTAGTTTTTATTACACTTATATTAG-3�) and LLOr (5�-CCTAGGTTATTCGATTGGATTATCTACTTTATTACTATATTTCGG-3�) (restriction sites are underlined) and cloningthe resulting PCR product into PmeI-AvrII-opened pHLA-Cerulean, therebyreplacing the ORF for HLA with that for LLO. L. monocytogenes chromosomalDNA was kindly provided by Jan Panne-Farre (Greifswald, Germany). Vectorswere introduced into S. carnosus TM300 (38), S. aureus SA113 �spa (kindlyprovided by Christiane Wolz, Tubingen, Germany), and S. aureus RN4220 (33)by electroporation (3, 38).

Flow cytometric invasion assays. Flow cytometric invasion assays were per-formed as described previously (23, 32), with minor modifications. Briefly,upper-airway epithelial cells were plated at a density of 0.6 � 106 cells in24-well plates. Cells were washed with DMEM-F12 medium, and 0.5 ml of asolution containing 1% human serum albumin and 10 mM HEPES (pH 7.4)in DMEM-F12 medium was added. Prior to infection, the cells were cooledon ice. Subsequently, 50 �l of fluorescein isothiocyanate-labeled (23) bacte-rial suspensions of S. carnosus TM300, S. carnosus GFP-FnbA, S. carnosusHLA-GFP-FnbA, S. carnosus HLAH35R-GFP-FnbA, or S. aureus strainCowan I (OD540 of 1) was added to the cells.

Preincubation for 1 h at 4°C allowed the sedimentation of the bacteria. Forsynchronized invasion, the culture dishes were incubated for an additional 3 h at37°C. The cells were harvested and were subjected to treatment with 20 �Mmonensin for 10 min at room temperature to neutralize fluorescence quenching.After propidium iodide (5 �g/ml) exclusion (23), the cells were analyzed by flowcytometry, and the results were normalized according to the mean fluorescenceintensity of the respective bacterial preparation (51). The invasiveness of labo-ratory S. aureus strain Cowan I was set as 100%.

Immunofluorescence. For fixation of monolayers, infected or uninfected cellsgrown on 18-mm glass coverslips were rinsed twice with PBS supplemented with1 mM MgCl2 and 0.1 mM CaCl2 (PBS2�) and were fixed using 3% formaldehydein PBS2� for 20 min. After fixation, cells were rinsed with PBS2�, incubated for5 min in PBS2� containing 50 mM NH4Cl, rinsed again with PBS2�, and finallyrinsed with water. Slides were then mounted with a solution containing 10%

(wt/vol) Mowiol (Calbiochem), 25% (wt/vol) glycerin, 25% water, and 45%(vol/vol) 0.1 M Tris-HCl (pH 8.5).

For immunofluorescence staining of infected and noninfected eukaryotic cells,the monolayers were permeabilized with PBS2� supplemented with 0.1% (wt/vol) Triton X-100 (PBS2�T) for 5 min immediately after fixation. After perme-abilization, the samples were incubated in PBS2�T containing 50 mM NH4Cl for5 min, washed with PBS2�T, and blocked with PBS2�T containing 2% (wt/vol)bovine serum albumin (BSA) for 1 h. Subsequently, the cells were treated withPBS2�T containing 0.1% (wt/vol) BSA and the primary antibody overnight at 4°Cor for 1 h at room temperature. Alpha-toxin was detected with a rabbit poly-clonal antiserum (Sigma-Aldrich, Taufkirchen, Germany). For the detection ofLLO, a polyclonal rabbit anti-LLO serum (Abcam, Cambridge, United King-dom) was used. After two washing steps with PBS2�T–0.1% (wt/vol) BSA, thecells were incubated with an MFP 631 (MoBiTec, Gottingen, Germany)- or anAlexa 647 (Invitrogen)-coupled secondary antibody in PBS2�T–0.1% (wt/vol)BSA for 1 h at room temperature. Subsequent washings with PBS2�T–0.1%(wt/vol) BSA, with PBS2�, and with deionized water completed the procedure.The slides were mounted with Mowiol. Secondary antibody specificity was con-trolled by the omission of primary antiserum.

Fluorescence microscopy. Fluorescence microscopy was performed using aZeiss LSM510 Meta confocal microscope (Carl Zeiss, Jena, Germany). Confocalimages, unless noted otherwise, represent 1-�m-thick confocal slices of thespecimen. The 405-nm line of the blue diode laser was used for the excitation ofCerulean. The beam path contained a 405-nm main dichroic mirror and a 420-to 480-nm-band-pass (BP) filter for the detection of the emitted fluorescence.GFPuvr fluorescence was detected using the 488-nm emission line of the argon-ion laser. The beam path contained a 488-nm dichroic mirror and a 505- to530-nm-BP filter. Enhanced YFP was excited by the 514-nm line of the argon-ionlaser in combination with a 514-nm dichroic mirror, and emitted fluorescencewas detected with the Meta detector using a detection range from 518 nm to 539nm. MFP 631 and Alexa 647 were imaged using the 633-nm line of the HeNelaser in combination with a 633-nm dichroic mirror and a detection range from646 nm to 753 nm of the Meta detector. Transmitted-light images were gener-ated by the longest excitation wavelength of the respective multitrack channelcombination and a transmitted-light detector below the specimen/focal plane.

Organellar pH measurements. For measurements of intraphagosomal pH, theratiometric pH-sensitive dye seminaphthorhodafluor-1 carboxylic acid acetate(SNARF1) conjugated to 10-kDa dextran or SNARF1-succinimidyl ester (SE)(Invitrogen) was used. One day prior to infection, 3 � 104 upper-airwayepithelial cells were seeded onto eight-well LabTekII chamber slides (Nunc).Cells were infected as described above. During the time of infection with bac-teria, medium was supplemented with 50 �M SNARF1-dextran. For directlabeling with SNARF1-SE, the bacteria were incubated in PBS containing 125�g/ml SNARF1-SE for 1 h at room temperature, harvested by centrifugation at3,000 � g for 10 min, resuspended in PBS, and incubated with shaking at 37°Cfor 10 min. After centrifugation, the bacterial pellet was resuspended in infectionmedium and added to the host cells. During image acquisition, SNARF1, GFPuvr, orCerulean fluorescence was imaged confocally in 8-bit mode using the line-switching multitrack mode of the LSM510 system. For the GFPuvr beam path, weused the 488-nm emission line of the argon-ion laser in combination with a405-nm/488-nm/561-nm dichroic mirror and a 505- to 530-nm-BP filter. Ceruleanwas imaged by excitation at 405 nm, a 405-nm/488-nm/561-nm dichroic mirror,and a 420- to 480-nm-BP filter. SNARF1 was excited by the 561-nm line of theyellow diode laser in combination with the 405-nm/488-nm/561-nm main dichroicmirror. The emission beam path contained a 571- to 603-nm-BP filter for theyellow part of the pH-dependent fluorescence spectrum (S2) and a 635- to753-nm-BP filter for the red part of the SNARF1 emission spectrum (S1). In theSNARF1 track, a transmitted-light image was collected in parallel. Each line forGFPuvr and SNARF1 fluorescence was scanned eight times, and data wereaveraged.

All ensuing image calculation steps were carried out using image-processingtools of LSM 510 software (Carl Zeiss GmbH, Jena, Germany) or ImageJ (1).For pH analysis, the acquired channels S1 and S2 of SNARF1 fluorescence (seeabove) were filtered using a 7- by 7-pixel low-pass-filter matrix. The lowest 25intensity values in the 8-bit-scaled images were subtracted to remove backgroundnoise. Both channel images were then multiplied by 256 and divided by them-selves to yield a binary mask for each channel. The masks were combined andmultiplied with the noise-reduced and low-pass-filtered S1 and S2 channel im-ages, respectively. In a subsequent step, S2 was divided by S1, and the result wasmultiplied by a factor of 10. The final-ratio image indicates the pH-sensitiveSNARF1 signal in regions with fluorescence above background levels. Only themean intensity of SNARF1 ratio signals in direct vicinity to GFPuvr or Ceruleanfluorescence of S. carnosus or S. aureus, respectively, was used for the analyses.

VOL. 77, 2009 ALPHA-TOXIN IS NOT SUFFICIENT FOR PHAGOLYSOSOME ESCAPE 3613

at UN

IV B

IBLIO

TH

EK

GR

EIF

SW

ALD

on August 19, 2009

iai.asm.org

Dow

nloaded from

For calculations of organellar pH, a calibration curve ranging from pH 4 to 9was produced. To generate a pH calibration curve in the organellar context,SNARF1-dextran-loaded infected cells were incubated in infection medium con-taining 20 �M monensin and 0.1 �M bafilomycin (Sigma-Aldrich, Taufkirchen,Germany) for 50 min. Subsequently, the medium was changed to different-pHbuffers made of citric acid (pH 4 and 5), potassium phosphate (pH 6, 7, and 8),and Tris (pH 9) containing 20 �M monensin and 0.1 �M bafilomycin. Afterimage acquisition and processing, a standard curve for SNARF1 fluorescenceversus pH in phagosomes/lysosomes was determined. The data for the standardcurve were fit with Boltzmann regression analysis using MATLAB.

RESULTS

Expression of alpha-toxin by apathogenic S. carnosus andnoncytotoxic S. aureus. We investigated the ability of staphy-lococcal alpha-toxin to mediate bacterial escape from infectedphagolysosomes of upper-airway epithelial cells. For that pur-pose, we modified pALC2084, a tetracycline-inducible GFPexpression vector (4). In order to use a completely toxin-freebackground (44), we expressed alpha-toxin in the apathogenicaroma producer S. carnosus TM300 (20). We cloned the ORFfor alpha-toxin (hla) into pALC2084, yielding pHLA-GFP, inwhich toxin and the fluorescent marker are cotranscribed uponthe derepression of the tetracycline-inducible promoter. Thus,the expression of hla or its functionally inactive mutant as wellas the expression of cytoplasmic GFPuvr are driven by the samepromoter, yielding a fluorescent signal simultaneous to toxinproduction. To investigate if the effects observed for down-stream analyses are due to the activity of alpha-toxin, we fur-ther engineered hla to contain an H35R point mutation, whichcompletely abolishes alpha-toxin pore formation (31, 57), re-sulting in pHLAH35R-GFP. Since S. carnosus does not possessadhesins, we cloned the fnbA locus into the control and toxinexpression vectors, yielding pGFP-FnbA, pHLA-GFP-FnbA,and pHLAH35R-GFP-FnbA. pHLA-Cerulean and pHLAH35R-Cerulean were generated accordingly for toxin expressionwithin S. aureus strains because of better fluorescence signalseparation using standard filter sets. For alpha-toxin expres-sion in the S. aureus background, we used the noncytotoxic andphenotypically alpha-toxin-negative S. aureus strain RN4220(33). For immunofluorescence applications, we employed re-combinant S. aureus SA113 �spa (kindly provided by Chris-tiane Wolz, Tubingen, Germany).

To test whether recombinant S. carnosus secreted alpha-toxin or its mutant in the culture supernatant, we performedWestern blot analyses. The alpha-toxin antiserum detected bandswith an apparent molecular mass of 32 kDa in the culture super-natants, whereas GFP was detected as a band with an apparentmolecular mass of 27 kDa mainly in the pellet fraction of thesamples, indicating that alpha-toxin and its point mutant areexpressed and secreted by S. carnosus (Fig. 1A). The pheno-typic expression of alpha-hemolysin in S. carnosus and S. au-reus was observed using SBA (Fig. 1B). The expression ofwild-type alpha-toxin by S. carnosus led to beta-hemolyticzones on blood agar plates, indicating that a functional alpha-toxin is produced. However, the level of hemolysis was reducedin comparison to that of the recombinant S. aureus strain. Thiswas even more prominent in bacteria engineered to produceListeria monocytogenes LLO (see below), since the hemolysis ofrecombinant S. carnosus was not observed on SBA after over-night incubation. The alpha-toxin H35R point mutant as wellas the controls without the toxin ORF (data not shown) did not

produce beta-hemolysis on SBA, indicating that the hla ORFwas responsible for the observed hemolysis in staphylococciexpressing alpha-toxin (Fig. 1B).

Staphylococcal alpha-toxin is not beneficial for intracellularsurvival. The invasiveness of recombinant S. carnosus for upper-airway epithelial cells was qualitatively investigated by micros-copy, whereby the membrane-impermeable fluorescent dye Alexa647 was added to culture medium of living infected epithelialcells. The dye thus stained the medium surrounding the cells aswell as pinocytotic vesicles. Staphylococci within the unstainedhost cell bodies were internalized (Fig. 2A). The invasiveness ofthe different constructs was quantified by flow cytometric invasionassays (23, 32). We compared wild-type S. carnosus TM300 as well

FIG. 1. S. carnosus secretes S. aureus alpha-toxin. (A) Western blotanalysis of protein extracts from bacterial pellets (P) as well as culturesupernatants (S) of S. carnosus cultures expressing S. aureus alpha-toxin (HLA) or its point mutant HLAH35R in an operon with down-stream GFP. Cytoplasmic GFP is present only in the pellet fraction(right), whereas alpha-toxin is detected mainly in the culture superna-tants (left), indicating that supernatant protein is present due to pro-tein secretion rather than cellular lysis. Proteins were detected byincubation with anti-alpha-toxin or anti-GFP sera and subsequent de-tection with alkaline phosphatase-conjugated anti-immunoglobulin Gsera. S. carnosus GFP-FnbA without the toxin ORF as well as purifiedtoxin or a GFP preparation were included as controls. (B) Hemolyticpatterns on SBA plates revealed that recombinant S. carnosus TM300and S. aureus SA113 �spa secrete functional staphylococcal alpha-toxin. (Top) S. aureus HLA-GFP-FnbA produced a beta-hemolyticzone typical of alpha-toxin, whereas the H35R mutant does not. S.aureus LLO-Cerulean produces a sharp hemolytic zone. (Bottom) S.carnosus HLA-GFP-FnbA is slightly beta-hemolytic, whereas S. car-nosus HLAH35R-GFP-FnbA does not show hemolysis. (Right) S. au-reus LLO produces a sharp hemolytic zone, whereas S. carnosus LLOseems to be nonhemolytic and was not included in the experiments.Cerulean is a spectral variant of cyan fluorescent protein. Prior toinoculation, 200 ng/ml anhydrotetracycline was spread onto the bloodagar plates to allow the induction of alpha-toxin expression.

3614 GIESE ET AL. INFECT. IMMUN.

at UN

IV B

IBLIO

TH

EK

GR

EIF

SW

ALD

on August 19, 2009

iai.asm.org

Dow

nloaded from

as recombinant S. carnosus harboring plasmids pGFP-FnbA,pHLA-GFP-FnbA, and pHLAH35R-GFP-FnbA with S. aureusCowan I, whose invasiveness was set as 100%. With an invasive-ness of 42.34% � 2.19% (compared to Cowan I; errors are rep-resented as standard errors of the means), S. carnosus GFP-FnbAis strongly invasive for upper-airway epithelial cells, whereas wild-type S. carnosus TM300 is not invasive (0.94% � 0.16%). S.carnosus additionally expressing alpha-toxin (HLA) (invasiveness

of 46.72% � 2.03%) or its H35R point mutant (44.59% � 3.36%)were as invasive as S. carnosus GFP-FnbA (Fig. 2B).

Recombinant S. carnosus was bound by LAMP1-YFP-deco-rated membranes as early as 20 min after endocytosis andstayed in these phagolysosomal compartments over extendedperiods. Even 6 days after infection, green fluorescent S. car-nosus cells were identified exclusively within LAMP1-positiveorganelles (Fig. 3).

FIG. 2. Invasion assays. (A) Qualitative invasion assay using S. carnosus TM300 transformants with pGFP-FnbA and pALC2084. Shown areconfocal images of infections of upper-airway epithelial cells (gray) infected with recombinant S. carnosus expressing GFP (green). Counterstainingof the cell culture medium was achieved by adding membrane-impermeable Alexa 647 dye (red). Cells infected with S. carnosus GFP-FnbAharboring the locus for the adhesin FnbA are internalized by epithelial cells (top), whereas S. carnosus cells carrying parental plasmid pALC2084are excluded from the cell lumina (bottom). Intracellular dye accumulation is due to the endocytotic uptake of the Alexa dye. Bar, 20 �m.(B) Quantitative invasion assays of upper-airway epithelial cells by wild-type and recombinant S. carnosus TM300 strains. Internalized bacteriawere measured by flow cytometry. Results are the means of data from three independent experiments run in duplicates (� standard errors of themeans) and are expressed as relative invasiveness compared to strain Cowan I (set as 100% invasiveness). S. carnosus TM300 was not internalizedby the epithelial cells (0.94% � 0.16%), whereas S. carnosus carrying plasmid pGFP-FnbA showed an invasion efficiency of 42.34% � 2.19%. Thestrains expressing alpha-toxin, S. carnosus HLA-GFP-FnbA (46.72% � 2.03%), as well as its functional H35R mutant (44.59% � 3.36%) showsimilar invasivenesses. AFU, arbitrary fluorescence units.

VOL. 77, 2009 ALPHA-TOXIN IS NOT SUFFICIENT FOR PHAGOLYSOSOME ESCAPE 3615

at UN

IV B

IBLIO

TH

EK

GR

EIF

SW

ALD

on August 19, 2009

iai.asm.org

Dow

nloaded from

In order to establish the effects of alpha-toxin on the intra-cellular survival of staphylococci, we quantified the number ofviable bacteria within host cells by CFU recovery assays atdifferent time points during infection. Both S. carnosus TM300and S. aureus RN4220 were modified to express either alpha-toxin, its inactive H35R mutant, or merely fluorescent re-porter. No decrease in numbers of CFU per well was observedafter the first 24 h of infection (Fig. 4A and B). At 4 dpi, therecovered CFU dropped to values below 104 CFU for S. car-nosus and S. aureus strains except for S. aureus Cerulean, whichstill reached 32,313 � 7,852 CFU. At 6 dpi, the numbers ofintracellular bacteria decreased, further indicating that upper-airway epithelial cells efficiently degrade staphylococci. Inter-

estingly, S. carnosus and S. aureus strains expressing functionalalpha-toxin were consistently recovered with the least numbersof CFU/well (4 � 2 and 24 � 15 CFU/well, respectively),whereas the nonhemolytic strains were recovered in largeramounts.

Starting with bacterial loads of roughly 0.4 to 0.8 S. carnosusCFU and 1.3 to 6.7 S. aureus CFU per host cell, at 6 dpi, lessthan 1 in 1,000 host cells was infected (Fig. 4C and D). Theupper-airway epithelial cells themselves did not show signifi-cant differences in growth in the presence of recombinantstaphylococci expressing alpha-toxin, the mutant thereof, oronly the fluorescent protein marker (Fig. 4E and F).

We also assayed CFU in culture supernatant that was keptgentamicin free for 24 h prior to sampling in order to deter-mine numbers of staphylococci that had been released into themedium during this incubation period. (Fig. 4G and H). Ex-tracellular CFU were roughly recovered at rates reduced by 1to 2 orders of magnitude compared with intracellular CFU(Fig. 4A and B). These results indicated that the investigatedstrains do not escape from their host cells. Since we were inneed of a positive control for phagolysosomal escape in the S.aureus background, we engineered S. aureus to express LLO.The S. aureus LLO strain secretes cytolysin from Listeriamonocytogenes in an active form as assayed by Western analysis(not shown) and hemolysis on SBA plates (Fig. 1B).

In contrast to staphylococci expressing alpha-toxin, host cellnumbers in our infection assays with S. aureus LLO dropped byone-half during the first 24 h of infection, indicating the cyto-toxicity of LLO (5.34 � 104 � 2.48 � 104 cells at 3 h p.i. versus2.16 � 104 � 1.57 � 104 cells at 24 h p.i.) (Fig. 4F). Owing tothis cytotoxicity of the S. aureus LLO strain, experiments in-volving this recombinant strain were stopped at 1 dpi.

We observed increasing numbers of CFU in cell culturesupernatants at 1 dpi when upper-airway epithelial cells wereinfected with the S. aureus LLO strain (P � 0.001), whereasnumbers of CFU in the supernatant at 3 h p.i. were comparableto those obtained with the remainder of the S. aureus strainsused (Fig. 4H). The high numbers of bacteria in the culturesupernatant indicate that the S. aureus LLO strain is releasedinto the medium and, most likely, replicated after release.However, a large proportion of S. aureus LLO can still berecovered from the intracellular environment (Fig. 4B), indi-cating that only a fraction of intracellular staphylococci couldbe stimulated to escape from the endosomes. Normalizingintracellular CFU with respect to host cells and comparing the24-h and 3-h time points suggested that by using the currentexpression vector, only about one in seven intracellular S. au-reus LLO CFU escapes from the phagosome/phagolysosomeafter the induction of LLO expression. This was also observedmicroscopically (see below).

Alpha-toxin is delivered to phagolysosomes by recombinantstaphylococci, but disruption of the organellar membrane wasnot observed. As no clear effects of alpha-toxin expressionwere observed in the CFU recovery assays regardless of thebacterial species or strain used, we chose to investigate toxinexpression as well as host-pathogen interactions on a subcel-lular level. The subcellular location of alpha-toxin in upper-airway epithelial cells infected with recombinant S. carnosusand S. aureus SA113 �spa (kindly provided by Christiane Wolz,Tubingen, German) was determined by immunofluorescence

FIG. 3. S. carnosus GFP-FnbA resides within LAMP1-positivevesicles throughout a 6-day infection. Shown are representativeimages of fixed airway epithelial cells expressing LAMP1-YFP in-fected with S. carnosus GFP-FnbA. The localization in LAMP1-positive vesicles starts as early as 20 min after infection and laststhroughout the duration of the experiment. Yellow, LAMP1-YFP;green, GFPuvr expressed by the staphylococci; gray scale, noncon-focal transmitted-light image. Bar, 5 �m.

3616 GIESE ET AL. INFECT. IMMUN.

at UN

IV B

IBLIO

TH

EK

GR

EIF

SW

ALD

on August 19, 2009

iai.asm.org

Dow

nloaded from

FIG. 4. CFU recovery assays of recombinant S. carnosus (S. c.) (left) and S. aureus (S. a.) (right) cells in upper-airway epithelial cells. At 3 h p.i., 1dpi, 4 dpi, and 6 dpi, we enumerated CFU per well from host cell pellet (A and B) and CFU per host cell (C and D). At 1 dpi, the numbers of intracellularS. aureus LLO-Cerulean cells were significantly reduced compared to those of alpha-toxin-expressing strains. Viable S. carnosus GFP-FnbA (A and C)as well as S. aureus Cerulean (B and D) cells were still detectable in host cells 6 days after infection. After high recovery rates at the beginning of theexperiment, the alpha-toxin-expressing strains were outperformed by nonhemolytic recombinant strains of S. carnosus and S. aureus. The numbers of S.carnosus (E)- and S. aureus (F)-infected cells indicate that except for LLO-expressing S. aureus strains, the recombinant strains were not cytotoxicregardless of HLA production. Enumeration of S. carnosus (G) and S. aureus (H) CFU from the supernatant revealed a significant increase in numbersof CFU, hinting at an escape from host cells only for LLO-expressing S. aureus strains. Results are presented as means of data from three independentexperiments performed in duplicates (� standard deviations). Black, S. carnosus GFP-FnbA/S. aureus Cerulean; white, S. carnosus HLA-GFP-FnbA/S.aureus HLA-Cerulean; light gray, S. carnosus HLAH35R-GFP-FnbA/S. aureus HLAH35R-Cerulean; dark gray, S. aureus LLO-Cerulean; thatching,uninfected host cells. Significance levels are given where appropriate. *, P � 0.05; **, P � 0.01; ***, P � 0.001.

3617

at UN

IV B

IBLIO

TH

EK

GR

EIF

SW

ALD

on August 19, 2009

iai.asm.org

Dow

nloaded from

assays. S. aureus SA113 �spa was used to avoid nonspecificimmunoglobulin binding by staphylococcal protein A (spa).

Staphylococci were found exclusively within LAMP1-YFP-positive vesicles. Alpha-toxin is clearly detected with immuno-

fluorescence, and the toxin is present in lumina of staphylo-coccus-containing vesicles. Thus, recombinant S. carnosus aswell as S. aureus deliver alpha-toxin to the phagolysosomes ofnonprofessional phagocytes (Fig. 5A). However, in all strains

FIG. 5. Alpha-toxin is not sufficient to disrupt phagolysosomal membranes. (A) Alpha-toxin immunofluorescence analyses demonstrate that staph-ylococci (green, S. carnosus; blue, S. aureus SA113 �spa) expressing alpha-toxin (red) are found exclusively within LAMP1-YFP-positive vesicles (yellow),whereas cells infected with the S. aureus LLO strain show bacteria without such a fluorescent envelope. (B) Membrane-impermeable fluorescent Alexa647 dye (red) was applied at the time of infection and was coingested by upper-airway epithelial cells. Staphylococcus-infected endosomes were thuslabeled with an additional fluorescent cargo. The loss of Alexa 647 and LAMP1-YFP signals (arrows), compatible with phagolysosomal escape, wasobserved only when S. aureus LLO cells were used (see also Movies S1 to S3 in the supplemental material). Images were acquired 3 h p.i. S. aureus SA113�spa was used to eliminate the immunoglobulin-binding activity of staphylococcal protein A. Scale bar, 10 �m.

3618 GIESE ET AL. INFECT. IMMUN.

at UN

IV B

IBLIO

TH

EK

GR

EIF

SW

ALD

on August 19, 2009

iai.asm.org

Dow

nloaded from

of S. carnosus and S. aureus investigated, staphylococci werebound by LAMP1-YFP-stained membranes, indicating aphagolysosomal location throughout the course of the experi-ment. Contrasting our findings with these strains, fluorescencemicroscopy with the S. aureus LLO strain repeatedly revealedstaphylococci that were not surrounded by a phagolysosomal

membrane (Fig. 5A). In additional dye uptake assays, the flu-orescent membrane-impermeable dye Alexa 647 was appliedat the time of infection and was coingested by upper-airwayepithelial cells, leading to Staphylococcus-infected endosomeswith an additional fluorescent cargo. A loss of Alexa 647 andLAMP1-YFP signal was observed only with LLO-expressing S.

FIG. 5—Continued.

VOL. 77, 2009 ALPHA-TOXIN IS NOT SUFFICIENT FOR PHAGOLYSOSOME ESCAPE 3619

at UN

IV B

IBLIO

TH

EK

GR

EIF

SW

ALD

on August 19, 2009

iai.asm.org

Dow

nloaded from

aureus strains (Fig. 5B; see Movies S1 to S3 in the supplemen-tal material).

Alpha-toxin does not disrupt the phagolysosomal pH gra-dient. The continuous localization of alpha-toxin-expressingstaphylococci within LAMP1-decorated vesicles was unexpected,since the pore former is hypothesized to mediate phagolysosomalescape. Because alpha-toxin is known to produce membranepores with a size exclusion limit of up to 3 kDa (7), we set outto investigate if the toxin permeabilizes the membrane forsmaller molecules without an actual disruption of the phagoly-sosomal membrane. As permeabilized phagosomes would failto acidify, we performed intraphagosomal pH measurementsusing the ratiometric pH-sensitive dye SNARF-1 conjugated to10-kDa dextran. By using defined pH ranges in culture mediaof epithelial cells that had been infected with S. aureus in thepresence of SNARF1-dextran, a pH calibration curve was ob-tained (Fig. 6A). The calibration curve was used to measurethe pH of phagolysosomes infected with various engineeredstaphylococcal strains. Two hours after infection, S. carnosusGFP-FnbA, S. carnosus HLA-GFP-FnbA, as well as S. carno-sus HLAH35R-GFP-FnbA strains were less acidified (pH rang-ing from 5.77 � 0.17 to 6.02 � 0.09) than the S. aureus HLA-Cerulean (pH 5.32 � 0.18) and S. aureus HLAH35R-Cerulean(pH 5.13 � 0.30) strains. At 6 h p.i., all investigated strains,regardless of whether they were in the S. aureus or S. carnosusbackground, acidified to a pH of 5.3 to 5.5 and stayed at thisacidification level up to 24 h p.i. (Fig. 6B). With the exceptionof the 2-h time point, no significant differences between thetwo strains were observed. Control experiments were per-formed with infected host cells that were additionally treatedwith the V-type ATPase inhibitor bafilomycin. During the timecourse, pH values of infected phagolysosomes of the controlwere in the neutral-pH range (Fig. 6B).

The S. aureus LLO strain is able to break down the phagoly-sosomal membrane (see above). Since this would result in adiffusion-based dilution of the fluorescent reporter SNARF1-dextran and, thus, a failure to measure pH, we labeled staph-ylococci with the membrane-permeant dye SNARF1-SE. Byusing this technique, however, the fluorescence signal frombacterial cytoplasmic pH contributes to the overall measure-ments so that pH calibration was not advisable (data notshown). Furthermore, as CFU recovery assays and immuno-fluorescence assays suggested, only a small fraction of S. aureusLLO cells (about 1 in 10) escaped from phagolysosomes. Wetherefore performed an analysis in which we enumerated S.aureus CFU in highly acidic, mildly acidic, or near-neutralphagolysosomes. Classification was performed on the basis ofthe ratiometric fluorescence signal in comparison to a bafilo-mycin control (Fig. 7).

Figure 7 shows that at 2 h p.i., the largest proportion of S.aureus LLO cells (57.3%) is in an environment with near-neutral pH, whereas only 18% of S. aureus HLA and 28.8% ofS. aureus Cerulean cells were in vesicles with that acidificationstatus.

At 6 h, 47.4% of S. aureus LLO cells are not acidified,whereas S. aureus HLA is present in a highly acidic environ-ment with 55.2% of all investigated staphylococcal clusters.The acidification thus seems to progress faster than that invesicles carrying S. aureus Cerulean, whose main fraction isfound in mildly acidified environments (60.6%). At 24 h,

mainly acidified staphylococci were identified, including S. au-reus LLO, which failed to escape from the phagolysosome(53.6% in a highly acidic environment). Notably, noncytotoxicS. aureus Cerulean cells, to a large extent (74.1%), is found inonly mildly acidified vesicles. All bafilomycin controls wereessentially located in a near-neutral environment at 2 and 6 hp.i.; however, at extended incubation times, acidified vesiclesaccumulated (Fig. 7).

Together, the results of pH measurements indicate that al-pha-toxin is not sufficient to induce a breakdown of phagoly-sosomal acidification, whereas a large fraction of LLO-express-ing bacteria is effective in neutralizing the environmental pH ininitial stages of intracellular infection (2 and 6 h p.i., respec-tively). At a later infection stage (24 h), S. aureus LLO-Ceru-lean strain that failed to disrupt phagolysosomal membraneswas acidified just like the HLA-expressing strains under scru-tiny.

DISCUSSION

In order to establish if staphylococcal alpha-toxin is suffi-cient to mediate phagolysosomal escape in upper-airway epi-thelial cells, we used the apathogenic aroma producer S. car-nosus as well as noncytotoxic laboratory S. aureus strains toexpress alpha-toxin, its functionally inactive H35R point mu-tant, or merely a fluorescent reporter and monitored the effectson host cells upon phagocytosis. As a positive control, weengineered an S. aureus strain to express LLO from Listeriamonocytogenes.

S. carnosus delivers alpha-toxin to phagolysosomes of airwayepithelial cells. We initially sought to microscopically ana-lyze the influence of alpha-toxin on phagolysosomal escapewithin upper-airway epithelial cells by expressing the toxinin the completely hemolysin-free and apathogenic Staphylo-coccus carnosus TM300 (20). We previously showed that S.carnosus TM300 harboring plasmid pFNBA4 (22) was effi-ciently internalized by 293 cells (50). However, staphylo-cocci are often hard to detect within their respective hostcells by light microscopy. To improve detection and enabledownstream fluorescence-based studies, we therefore choseto integrate the locus for FnBPA in an established GFPexpression vector, pALC2084 (4). To adapt pGFP-FnbA foralpha-hemolysin expression, the ORF for alpha-toxin (hla)was cloned to be expressed in an operon together with thedownstream GFP gene. Thus, upon the activation of thetetracycline-inducible promoter of the operon, cytoplasmicgreen fluorescent signal was available for the simultaneousproduction of staphylococcal alpha-toxin.

Figure 1A shows that alpha-toxin is released into the culturesupernatant by recombinant S. carnosus via secretion and notvia bacterial lysis. However, a significant fraction of alpha-toxinis also found in the pellet fraction and most likely is cell wallassociated. S. carnosus HLA-GFP-FnbA expresses functionalalpha-toxin, although the relatively weak hemolytic zone de-picted in Fig. 1B indicates that S. carnosus is not capable ofsecreting functional alpha-toxin with the same efficiency as thatof S. aureus. However, hemolysin is produced at levels that areeasily detectable by immunofluorescence (see below). LLO,which was used as a positive control for the phagolysosomalescape of S. aureus later in this study, did not form hemolytic

3620 GIESE ET AL. INFECT. IMMUN.

at UN

IV B

IBLIO

TH

EK

GR

EIF

SW

ALD

on August 19, 2009

iai.asm.org

Dow

nloaded from

zones after overnight incubation of recombinant S. carnosus, incontrast to the clear hemolysis by LLO when expressed in S.aureus.

Internalized staphylococci were easily detectable by fluores-cence microscopy via their cytoplasmic GFPuvr expression.Employing green fluorescent staphylococci as well as a fluo-rescent cell culture medium counterstain provides an efficient

and fast technique to qualitatively assay bacterial invasion byconfocal microscopy of living cells without the need fordifferential antibody staining or fixation (Fig. 2A). Qualita-tive invasion assays using flow cytometry demonstrated thatall recombinant S. carnosus strains carrying the various plas-mids are efficiently internalized by upper-airway epithelialcells (Fig. 2B).

FIG. 6. Acidification of phagolysosomes infected with recombinant S. carnosus and S. aureus. (A) A calibration curve was generated for S.aureus-infected upper-airway epithelial cells by supplying media with different pH values in the presence of bafilomycin and monensin. TheSNARF1 fluorescence emission signal in the short-wavelength (yellow) and long-wavelength (red) ranges is recorded in the direct vicinity ofintracellular staphylococci (inset, arrows). Based on the calibration curve, channel ratios were converted into pH values in subsequent microscopicanalyses. (Inset) Short-wavelength (yellow) and long-wavelength (red) ranges of SNARF1 fluorescence. Green, GFPuvr expressed by the staph-ylococci (here, exemplary, S. carnosus HLA-GFP-FnbA); gray scale, nonconfocal transmitted-light channel. Bar, 5 �m. (B) Using the ratiometricdye SNARF1-dextran, the pH in infected phagolysosomes of upper-airway epithelial cells was determined at 2, 6, and 24 h p.i. Open circles, S.carnosus GFP-FnbA; open squares, S. carnosus HLA-GFP-FnbA; open triangles, S. carnosus HLAH35R-GFP-FnbA; crosses, S. aureus HLA-Cerulean; dashes, S. aureus HLAH35R-Cerulean. Control experiments were performed by applying the V-type ATPase inhibitor bafilomycin (closeddiamonds). All measurements were obtained from three independent experiments. Values are represented as pH � standard deviations.

VOL. 77, 2009 ALPHA-TOXIN IS NOT SUFFICIENT FOR PHAGOLYSOSOME ESCAPE 3621

at UN

IV B

IBLIO

TH

EK

GR

EIF

SW

ALD

on August 19, 2009

iai.asm.org

Dow

nloaded from

We further examined the possibility of microscopically mon-itoring infection of upper-airway epithelial cells with recombi-nant S. carnosus over a period of 6 days. For that purpose, weinfected epithelial cells expressing LAMP1-YFP with S. carno-

sus GFP-FnbA and fixed the samples for subsequent micro-scopic examinations (Fig. 3). As early as 20 min after theaddition of bacteria to the tissue culture medium and even atthe last sampling time point of our analysis, 6 days after infec-tion, green fluorescent S. carnosus GFP-FnbA is found exclu-sively in LAMP1-positive vesicles. The expression of the fluo-rescent reporter protein GFP at later infection stages and theisolation of culturable staphylococci in CFU recovery assays(see below) suggest that the bacteria are viable and metabol-ically active. A long-term association with LAMP1-positivevesicles has also been observed for noncytotoxic strains of S.aureus and S. carnosus pFNBA4 (47) in human umbilical veinendothelial cells. One might speculate that staphylococcal per-sistence within eukaryotic nonprofessional phagocytes is en-abled by the tolerance of staphylococci to extreme environ-mental conditions.

In a subsequent analysis, we identified alpha-toxin in phagoly-sosomes of airway epithelial cells infected with the S. carnosusHLA-GFP-FnbA strain by immunofluorescence (Fig. 5A), dem-onstrating that S. carnosus is able to deliver the toxin to this hostcell organelle.

In order to establish the impact of staphylococcal alpha-toxin on phagolysosomal localization, we also analyzed S. au-reus RN4220 harboring various toxin expression plasmids inorder to exclude effects of lower levels of hemolysin productionwithin S. carnosus.

Expression of alpha-toxin is not beneficial for staphylococciin intracellular infections. All investigated alpha-toxin-ex-pressing strains in both species backgrounds, S. carnosus and S.aureus, showed a similar development of CFU recovery ratesduring 6-day infection experiments: at 1 dpi, close to 100% ofthe initially internalized staphylococci (as determined at 3 hp.i.) were recovered for all strains investigated, indicating thatthe presence of the toxin ORF does not have an influence atthis stage (Fig. 4A and B). The recovered CFU dropped byapproximately 2 log units per well after 4 dpi, whereas extra-cellular CFU did not increase. This demonstrates that upper-airway epithelial cells efficiently degrade staphylococci. Thiseffective removal was described previously for a variety ofnoncytotoxic strains (34). However, even at 6 dpi, viablestaphylococci were recovered from host cells. At this time,the nonhemolytic S. carnosus strains outperform S. carnosusHLA-GFP-FnbA (4 � 2 CFU/well). Similarly, all nonhemo-lytic S. aureus strains used in this study were recovered athigher CFU/well than the hemolytic S. aureus HLA-Ceru-lean strain.

It was reported previously that S. carnosus pFNBA4 cellscould be recovered from human umbilical vein endothelial cellmonolayers at extended infection times and with comparablerecovery rates (47). However, the additional expression offunctional alpha-toxin seems to have a detrimental effect onthe intracellular survival of the staphylococci, as we demon-strated in both species backgrounds, S. carnosus and S. aureus(Fig. 4). Whereas the lowest numbers of CFU were recoveredconsistently for strains expressing functional alpha-toxin, theprobability of errors in these analyses is not below the 5% levelowing to the variability in CFU numbers (P 0.101 and 0.067for S. carnosus and S. aureus, respectively, as determined by atwo-tailed Student’s t test in a comparison of alpha-toxin-ex-pressing strains with their toxin-less counterparts).

FIG. 7. Histogram of the distribution of intracellular SNARF1-SE-labeled staphylococci within areas with highly acidic, mildly acidic, ornear-neutral pH (see pseudocolored range indicator). The percentages ofbacterial clusters within either one of the classes are indicated by bars:black, S. aureus (S. a.) Cerulean; white, S. aureus HLA-Cerulean; darkgray, S. aureus LLO; thatching, bafilomycin-treated control.

3622 GIESE ET AL. INFECT. IMMUN.

at UN

IV B

IBLIO

TH

EK

GR

EIF

SW

ALD

on August 19, 2009

iai.asm.org

Dow

nloaded from

Our findings are consistent with previous reports that small-colony variants of S. aureus are generally defective in the agrquorum-sensing system, which results in a decreased level ofproduction of virulence factors such as alpha-toxin. Also, al-pha-toxin was previously found to be downregulated in globaltranscriptomic or proteomic analyses of staphylococci recov-ered from intracellular infections (11, 16). One can only spec-ulate why alpha-toxin appears to be detrimental to the intra-cellular survival of staphylococci. The expression of anadditional ORF is not responsible for this phenomenon, asHLAH35R constructs are of identical lengths and are consis-tently recovered at higher numbers of CFU (Fig. 4). Interest-ingly, a previously reported in vivo study of experimental en-docarditis found a “paradoxically reduced virulence” in S.aureus strains hyperproducing alpha-toxin (5). Whereas thisstudy suggested an increased release of platelet microbicidalproteins for the reduced virulence of toxin overexpressors,there could be an additional mechanism on the cellular levelrecognizing alpha-toxin and leading to an enhanced clearanceof hemolytic bacteria. Such mechanisms could be based onToll-like receptors, which are also present in phagosomes andare able to mount cellular defenses against vacuolar pathogenssuch as Listeria monocytogenes (36).

Since our results from CFU recovery experiments were notcompatible with phagolysosomal escape, we infected upper-airway epithelial cells with an S. aureus strain expressing LLOfrom Listeria monocytogenes. L. monocytogenes escapes fromphagolysosomes (15, 54), mainly by the action of LLO (10, 15).We observed a decrease in numbers of intracellular CFU/wellof the S. aureus strain expressing LLO by 1 order of magnitudeduring the first 24 h p.i., which can be attributed mainly to thedeath of host cells due to the high level of cytotoxicity of thisstrain (Fig. 4F). However, a reduction of the bacterial load perhost cell during the first 24 h of infection was observed only forthe S. aureus LLO strain (Fig. 4D). Our results thus suggestthat about one in seven staphylococci disappears from theintracellular environment. In parallel, increasing numbers ofCFU of extracellular S. aureus LLO illustrate that the decreasein the number of CFU/host cell is due to escape rather than tothe degradation of staphylococci.

The phagosome-specific lysin LLO (46) was heterologouslyexpressed previously in a variety of organisms such as Listeriainnocua (52), Bacillus subtilis (10), Salmonella enterica serovarDublin (18), and Mycobacterium bovis BCG (26). In all thoseexperiments, the heterologous expression of the cytolysin re-sulted in the phagosomal escape of the respective bacterialspecies, although significant intracellular replication was dem-onstrated only for recombinant B. subtilis (10). Judging by thereduction in numbers of S. aureus LLO CFU per host cell at 1dpi in our experiments, the increase in levels of extracellular S.aureus LLO stems from the replication of bacteria in the cul-ture supernatant during the gentamicin-free treatment in theinfection assay. Nevertheless, since the replication of staphy-lococci within the host cell cytoplasm was previously described(30), some genetic backgrounds of S. aureus seem to be capa-ble of intracellular growth once the phagolysosomal barrier isbreached. The cytotoxicity of the S. aureus strain expressingLLO possibly precluded such observations in our currentsetup.

In conclusion, alpha-toxin alone is not sufficient to mediateescape from host cells.

Delivery of staphylococcal alpha-toxin to phagolysosomes ofairway epithelial cells does not lead to organellar membranedisruption or permeabilization. In upper-airway epithelialcells infected with staphylococci expressing alpha-toxin (S. car-nosus HLA-GFP-FnbA and S. aureus HLA-Cerulean strains)or the functionally inactive toxin (S. carnosus HLAH35R-GFP-FnbA and S. aureus HLAH35R-Cerulean strains), the alpha-toxin immunofluorescence signal was present exclusively in thelumina of infected phagolysosomes (Fig. 5A). However, allstaphylococci resided in LAMP1-YFP-positive vesicles at alltime points investigated, which implies that phagolysosomalescape did not take place despite the presence of the poreformer. S. aureus expressing LLO serves as an “escape” posi-tive control: 6 h after infection, a subset of S. aureus LLO-infected phagolysosomes of upper-airway epithelial cells is nolonger bound by LAMP1-YFP-positive membranes (Fig. 5A;see Movie S3 in the supplemental material) or leads to a lossof soluble fluorescent cargo that had been coapplied duringinfection (Fig. 5B). Both sets of experiments suggest thephagolysosomal escape of the S. aureus strain expressing LLO.

Using a Salmonella strain expressing LLO, Gentschev et al.previously found that 8% of bacteria escaped from the phago-some (18), whereas we estimate that about 10 to 15% of S.aureus LLO bacteria are able to disrupt the phagolysosomalmembrane. It still has to be established whether higher escaperates can be achieved by using alternative inducible promotersor different concentrations of inducer. Summarizing our im-munofluorescence-based observations, we concluded that al-pha-toxin did not have any obvious effect on phagolysosomalmembrane integrity.

Since alpha-toxin pores have an exclusion limit of about 3kDa (7), we investigated whether the phagolysosomal mem-brane might be perforated without an actual disruption of theorganelle. Thus, the ratiometric fluorescent pH sensorSNARF1–10-kDa dextran was administrated together with re-combinant staphylococci during infection. The pH of phagoly-sosomes infected with staphylococci expressing either alpha-toxin, its H35R mutant, or merely GFP did show acidificationof the organelle at a pH of 5.3 to 5.5 regardless of the strainused. This indicated that alpha-toxin does not permeabilize thephagolysosomal membrane of upper-airway epithelial cells(Fig. 6). Using SNARF1-SE-labeled recombinant S. aureusstrains, again, no indication of permeabilization was found forS. aureus strains expressing alpha-toxin. The histograms shownin Fig. 7 suggest, however, that over multiple hours after in-fection, the S. aureus strain expressing LLO is in a near-neutralenvironment within the cells, whereas after 24 h, S. aureus LLOcells that failed to escape from the phagolysosomes were foundin highly acidic environments. It has to be noted that owing tothe cytotoxicity of the LLO strain, expression was induced onlyafter the phagocytosis of staphylococci. In contrast, all otherstrains were already induced in the main bacterial culture priorto infection. Thus, the delayed induction and reduced LLOconcentrations could lead to the phagolysosomal fraction of S.aureus LLO cells.

Taken together, these measurements suggest that alpha-toxin is not sufficient to mediate phagolysosomal escape inupper-airway epithelial cells. This finding is compatible with

VOL. 77, 2009 ALPHA-TOXIN IS NOT SUFFICIENT FOR PHAGOLYSOSOME ESCAPE 3623

at UN

IV B

IBLIO

TH

EK

GR

EIF

SW

ALD

on August 19, 2009

iai.asm.org

Dow

nloaded from

previously reported findings that demonstrated the inability ofthe alpha-toxin-producing strain Wood 46 to induce apoptosisin endothelial cells (24). Since phagolysosomal escape hasbeen observed by multiple groups (6, 30, 34), the disruption ofthe organellar membrane either must be caused by the coop-erative action of multiple virulence factors, including alpha-toxin (24, 30, 49), or must be dependent on a different viru-lence factor. Also, the ability of S. aureus to disrupt thephagolysosomal membrane is certainly influenced by the ge-netic background of the host cell (30, 55). Furthermore, caveo-lin-1 as well as �1-integrin were previously described to bepossible alpha-toxin receptors on the plasma membrane (37,41). Since caveolin-1 and integrins are normally not routed tothe lysosome, an apt receptor for heptamer assembly would bemissing in this organelle. Thus, just as alpha-toxin-insensitivecell lines exist (27, 55), phagolysosomes of upper-airway epi-thelial cells may constitute alpha-toxin-resistant organelles.

ACKNOWLEDGMENTS

Funding was provided by the German Ministry for Science andResearch (BMBF) within the program Entrepreneurial Regions: Com-petence Centers, under code ZIK011 (M.J.F.), and German ResearchCouncil (DFG) SFB-TR34 project C6 (B.S.).

We are indebted to following colleagues: Ambrose Cheung (Dart-mouth, NH) for pALC2084, Susanne Engelmann (Greifswald, Ger-many) for purified alpha-toxin as well as S. aureus strains GP269 andRN4220, Fritz Gotz (Tubingen, Germany) for S. carnosus TM300, JorgMostertz (Greifswald, Germany) for human cDNA, Jan Panne-Farre(Greifswald, Germany) for Listeria monocytogenes DNA and anti-ClpLantiserum, Christiane Wolz (Tubingen, Germany) for S. aureus SA113�spa, and Olivier Tabary and Jacky Jacquot (Inserm, Paris, France) forupper-airway epithelial cells. We also thank Matthias Scholz for helpwith MATLAB and Tobias Steiner for assistance.

REFERENCES

1. Abramoff, M. D., P. J. Magelhaes, and S. J. Ram. 2004. Image processingwith Image J. Biophoton. Int. 11:36–42.

2. Almeida, R. A., K. R. Matthews, E. Cifrian, A. J. Guidry, and S. P. Oliver.1996. Staphylococcus aureus invasion of bovine mammary epithelial cells. J.Dairy Sci. 79:1021–1026.

3. Augustin, J., and F. Gotz. 1990. Transformation of Staphylococcus epidermi-dis and other staphylococcal species with plasmid DNA by electroporation.FEMS Microbiol. Lett. 54:203–207.

4. Bateman, B. T., N. P. Donegan, T. M. Jarry, M. Palma, and A. L. Cheung.2001. Evaluation of a tetracycline-inducible promoter in Staphylococcus au-reus in vitro and in vivo and its application in demonstrating the role of sigBin microcolony formation. Infect. Immun. 69:7851–7857.

5. Bayer, A. S., M. D. Ramos, B. E. Menzies, M. R. Yeaman, A. J. Shen, andA. L. Cheung. 1997. Hyperproduction of alpha-toxin by Staphylococcus au-reus results in paradoxically reduced virulence in experimental endocarditis:a host defense role for platelet microbicidal proteins. Infect. Immun. 65:4652–4660.

6. Bayles, K. W., C. A. Wesson, L. E. Liou, L. K. Fox, G. A. Bohach, and W. R.Trumble. 1998. Intracellular Staphylococcus aureus escapes the endosomeand induces apoptosis in epithelial cells. Infect. Immun. 66:336–342.

7. Bhakdi, S., H. Bayley, A. Valeva, I. Walev, B. Walker, M. Kehoe, and M.Palmer. 1996. Staphylococcal alpha-toxin, streptolysin-O, and Escherichiacoli hemolysin: prototypes of pore-forming bacterial cytolysins. Arch. Micro-biol. 165:73–79.

8. Bhakdi, S., M. Muhly, and R. Fussle. 1984. Correlation between toxin bind-ing and hemolytic activity in membrane damage by staphylococcal alpha-toxin. Infect. Immun. 46:318–323.

9. Bhakdi, S., and J. Tranum-Jensen. 1991. Alpha-toxin of Staphylococcusaureus. Microbiol. Rev. 55:733–751.

10. Bielecki, J., P. Youngman, P. Connelly, and D. A. Portnoy. 1990. Bacillussubtilis expressing a haemolysin gene from Listeria monocytogenes can growin mammalian cells. Nature 345:175–176.

11. Burlak, C., C. H. Hammer, M. A. Robinson, A. R. Whitney, M. J. McGavin,B. N. Kreiswirth, and F. R. Deleo. 2007. Global analysis of community-associated methicillin-resistant Staphylococcus aureus exoproteins revealsmolecules produced in vitro and during infection. Cell. Microbiol. 9:1172–1190.

12. Essmann, F., H. Bantel, G. Totzke, I. H. Engels, B. Sinha, K. Schulze-Osthoff, and R. U. Janicke. 2003. Staphylococcus aureus alpha-toxin-inducedcell death: predominant necrosis despite apoptotic caspase activation. CellDeath Differ. 10:1260–1272.

13. Flotte, T. R., S. A. Afione, C. Conrad, S. A. McGrath, R. Solow, H. Oka, P. L.Zeitlin, W. B. Guggino, and B. J. Carter. 1993. Stable in vivo expression ofthe cystic fibrosis transmembrane conductance regulator with an adeno-associated virus vector. Proc. Natl. Acad. Sci. USA 90:10613–10617.

14. Forti, S., and G. Menestrina. 1989. Staphylococcal alpha-toxin increases thepermeability of lipid vesicles by cholesterol- and pH-dependent assembly ofoligomeric channels. Eur. J. Biochem. 181:767–773.

15. Gaillard, J. L., P. Berche, J. Mounier, S. Richard, and P. Sansonetti. 1987.In vitro model of penetration and intracellular growth of Listeria monocyto-genes in the human enterocyte-like cell line Caco-2. Infect. Immun. 55:2822–2829.

16. Garzoni, C., P. Francois, A. Huyghe, S. Couzinet, C. Tapparel, Y. Charbon-nier, A. Renzoni, S. Lucchini, D. P. Lew, P. Vaudaux, W. L. Kelley, and J.Schrenzel. 2007. A global view of Staphylococcus aureus whole genomeexpression upon internalization in human epithelial cells. BMC Genomics8:171.

17. Garzoni, C., and W. L. Kelley. 2009. Staphylococcus aureus: new evidence forintracellular persistence. Trends Microbiol. 17:59–65.

18. Gentschev, I., Z. Sokolovic, H.-J. Mollenkopf, J. Hess, S. H. Kaufmann, M.Kuhn, G. F. Krohne, and W. Goebel. 1995. Salmonella strain secreting activelisteriolysin changes its intracellular localization. Infect. Immun. 63:4202–4205.

19. Giachino, P., S. Engelmann, and M. Bischoff. 2001. �B activity depends onRsbU in Staphylococcus aureus. J. Bacteriol. 183:1843–1852.

20. Gotz, F., B. Kreutz, and K. H. Schleifer. 1983. Protoplast transformation ofStaphylococcus carnosus by plasmid DNA. Mol. Gen. Genet. 189:340–342.

21. Gouaux, E. 1998. Alpha-hemolysin from Staphylococcus aureus: an archetypeof beta-barrel, channel-forming toxins. J. Struct. Biol. 121:110–122.

22. Greene, C., D. McDevitt, P. Francois, P. E. Vaudaux, D. P. Lew, and T. J.Foster. 1995. Adhesion properties of mutants of Staphylococcus aureus de-fective in fibronectin-binding proteins and studies on the expression of fnbgenes. Mol. Microbiol. 17:1143–1152.

23. Grundmeier, M., M. Hussain, P. Becker, C. Heilmann, G. Peters, and B.Sinha. 2004. Truncation of fibronectin-binding proteins in Staphylococcusaureus strain Newman leads to deficient adherence and host cell invasion dueto loss of the cell wall anchor function. Infect. Immun. 72:7155–7163.

24. Haslinger-Loffler, B., B. C. Kahl, M. Grundmeier, K. Strangfeld, B. Wagner,U. Fischer, A. L. Cheung, G. Peters, K. Schulze-Osthoff, and B. Sinha. 2005.Multiple virulence factors are required for Staphylococcus aureus-inducedapoptosis in endothelial cells. Cell. Microbiol. 7:1087–1097.

25. Haslinger-Loffler, B., B. Wagner, M. Bruck, K. Strangfeld, M. Grundmeier,U. Fischer, W. Volker, G. Peters, K. Schulze-Osthoff, and B. Sinha. 2006.Staphylococcus aureus induces caspase-independent cell death in humanperitoneal mesothelial cells. Kidney Int. 70:1089–1098.

26. Hess, J., D. Miko, A. Catic, V. Lehmensiek, D. G. Russell, and S. H. E.Kaufmann. 1998. Mycobacterium bovis bacille Calmette-Guerin strains se-creting listeriolysin of Listeria monocytogenes. Proc. Natl. Acad. Sci. USA95:5299–5304.

27. Husmann, M., E. Beckmann, K. Boller, N. Kloft, S. Tenzer, W. Bobkiewicz,C. Neukirch, H. Bayley, and S. Bhakdi. 2009. Elimination of a bacterialpore-forming toxin by sequential endocytosis and exocytosis. FEBS Lett.583:337–344.

28. Ikigai, H., and T. Nakae. 1985. Conformational alteration in alpha-toxinfrom Staphylococcus aureus concomitant with the transformation of the wa-ter-soluble monomer to the membrane oligomer. Biochem. Biophys. Res.Commun. 130:175–181.

29. Jarry, T. M., and A. L. Cheung. 2006. Staphylococcus aureus escapes moreefficiently from the phagosome of a cystic fibrosis bronchial epithelial cellline than from its normal counterpart. Infect. Immun. 74:2568–2577.

30. Jarry, T. M., G. Memmi, and A. L. Cheung. 2008. The expression of alpha-haemolysin is required for Staphylococcus aureus phagosomal escape afterinternalization in CFT-1 cells. Cell. Microbiol. 10:1801–1814.

31. Jursch, R., A. Hildebrand, G. Hobom, J. Tranum-Jensen, R. Ward, M.Kehoe, and S. Bhakdi. 1994. Histidine residues near the N terminus ofstaphylococcal alpha-toxin as reporters of regions that are critical for oli-gomerization and pore formation. Infect. Immun. 62:2249–2256.

32. Juuti, K. M., B. Sinha, C. Werbick, G. Peters, and P. I. Kuusela. 2004.Reduced adherence and host cell invasion by methicillin-resistant Staphylo-coccus aureus expressing the surface protein Pls. J. Infect. Dis. 189:1574–1584.

33. Kreiswirth, B. N., S. Lofdahl, M. J. Betley, M. O’Reilly, P. M. Schlievert,M. S. Bergdoll, and R. P. Novick. 1983. The toxic shock syndrome exotoxinstructural gene is not detectably transmitted by a prophage. Nature 305:709–712.

34. Krut, O., O. Utermohlen, X. Schlossherr, and M. Kronke. 2003. Strain-specific association of cytotoxic activity and virulence of clinical Staphylococ-cus aureus isolates. Infect. Immun. 71:2716–2723.

35. Kubica, M., K. Guzik, J. Koziel, M. Zarebski, W. Richter, B. Gajkowska, A.

3624 GIESE ET AL. INFECT. IMMUN.

at UN

IV B

IBLIO

TH

EK

GR

EIF

SW

ALD

on August 19, 2009

iai.asm.org

Dow

nloaded from

Golda, A. Maciag-Gudowska, K. Brix, L. Shaw, T. Foster, and J. Potempa.2008. A potential new pathway for Staphylococcus aureus dissemination: thesilent survival of S. aureus phagocytosed by human monocyte-derived mac-rophages. PLoS ONE 3:e1409.

36. Leber, J. H., G. T. Crimmins, S. Raghavan, N. P. Meyer-Morse, J. S. Cox,and D. A. Portnoy. 2008. Distinct TLR- and NLR-mediated transcriptionalresponses to an intracellular pathogen. PLoS Pathog. 4:e6.

37. Liang, X., and Y. Ji. 2006. Alpha-toxin interferes with integrin-mediatedadhesion and internalization of Staphylococcus aureus by epithelial cells.Cell. Microbiol. 8:1656–1668.

38. Lofblom, J., N. Kronqvist, M. Uhlen, S. Ståhl, and H. Wernerus. 2007.Optimization of electroporation-mediated transformation: Staphylococcuscarnosus as model organism. J. Appl. Microbiol. 102:736–747.

39. Lowy, F. D., J. Fant, L. L. Higgins, S. K. Ogawa, and V. B. Hatcher. 1988.Staphylococcus aureus-human endothelial cell interactions. J. Ultrastruct.Mol. Struct. Res. 98:137–146.

40. Mempel, M., C. Schnopp, M. Hojka, H. Fesq, S. Weidinger, M. Schaller,H. C. Korting, J. Ring, and D. Abeck. 2002. Invasion of human keratinocytesby Staphylococcus aureus and intracellular bacterial persistence representhaemolysin-independent virulence mechanisms that are followed by featuresof necrotic and apoptotic keratinocyte cell death. Br. J. Dermatol. 146:943–951.

41. Pany, S., R. Vijayvargia, and M. V. Krishnasastry. 2004. Caveolin-1 bindingmotif of alpha-hemolysin: its role in stability and pore formation. Biochem.Biophys. Res. Commun. 322:29–36.

42. Proctor, R. A., D. M. Bates, and P. McNamara. 2001. Electron transport-deficient Staphylococcus aureus small-colony variants as emerging pathogens.ASM Press, Washington, DC.

43. Proctor, R. A., C. von Eiff, B. C. Kahl, K. Becker, P. McNamara, M. Her-rmann, and G. Peters. 2006. Small colony variants: a pathogenic form ofbacteria that facilitates persistent and recurrent infections. Nat. Rev. Micro-biol. 4:295–305.

44. Rosenstein, R., C. Nerz, L. Biswas, A. Resch, G. Raddatz, S. C. Schuster, andF. Gotz. 2009. Genome analysis of the meat starter culture bacterium Staph-ylococcus carnosus TM300. Appl. Environ. Microbiol. 75:811–822.

45. Schenk, S., and R. A. Laddaga. 1992. Improved method for electroporationof Staphylococcus aureus. FEMS Microbiol. Lett. 73:133–138.

46. Schnupf, P., and D. A. Portnoy. 2007. Listeriolysin O: a phagosome-specificlysin. Microbes Infect. 9:1176–1187.

47. Schroder, A., R. Kland, A. Peschel, C. von Eiff, and M. Aepfelbacher. 2006.Live cell imaging of phagosome maturation in Staphylococcus aureus infectedhuman endothelial cells: small colony variants are able to survive in lyso-somes. Med. Microbiol. Immunol. 195:185–194.

48. Sendi, P., and R. A. Proctor. 2009. Staphylococcus aureus as an intracellularpathogen: the role of small colony variants. Trends Microbiol. 17:54–58.

49. Shompole, S., K. T. Henon, L. E. Liou, K. Dziewanowska, G. A. Bohach, andK. W. Bayles. 2003. Biphasic intracellular expression of Staphylococcus au-reus virulence factors and evidence for Agr-mediated diffusion sensing. Mol.Microbiol. 49:919–927.

50. Sinha, B., P. Francois, Y. A. Que, M. Hussain, C. Heilmann, P. Moreillon, D.Lew, K. H. Krause, G. Peters, and M. Herrmann. 2000. Heterologouslyexpressed Staphylococcus aureus fibronectin-binding proteins are sufficientfor invasion of host cells. Infect. Immun. 68:6871–6878.

51. Sinha, B., P. P. Francois, O. Nuße, M. Foti, O. M. Hartford, P. Vaudaux,T. J. Foster, D. P. Lew, M. Herrmann, and K.-H. Krause. 1999. Fibronectin-binding protein acts as Staphylococcus aureus invasin via fibronectin bridgingto integrin �5�1. Cell. Microbiol. 1:101–117.

52. Slaghuis, J., M. Gotz, F. Engelbrecht, and W. Goebel. 2004. Inefficientreplication of Listeria innocua in the cytosol of mammalian cells. J. Infect.Dis. 189:393–401.

53. Song, L., M. R. Hobaugh, C. Shustak, S. Cheley, H. Bayley, and J. E.Gouaux. 1996. Structure of staphylococcal alpha-hemolysin, a heptamerictransmembrane pore. Science 274:1859–1866.

54. Tilney, L. G., and D. A. Portnoy. 1989. Actin filaments and the growth,movement, and spread of the intracellular bacterial parasite, Listeria mono-cytogenes. J. Cell Biol. 109:1597–1608.

55. Valeva, A., I. Walev, M. Pinkernell, B. Walker, H. Bayley, M. Palmer, and S.Bhakdi. 1997. Transmembrane beta-barrel of staphylococcal alpha-toxinforms in sensitive but not in resistant cells. Proc. Natl. Acad. Sci. USA94:11607–11611.

56. Walev, I., E. Martin, D. Jonas, M. Mohamadzadeh, W. Muller-Klieser, L.Kunz, and S. Bhakdi. 1993. Staphylococcal alpha-toxin kills human kerati-nocytes by permeabilizing the plasma membrane for monovalent ions. Infect.Immun. 61:4972–4979.

57. Walker, B., and H. Bayley. 1995. Key residues for membrane binding, oli-gomerization, and pore forming activity of staphylococcal alpha-hemolysinidentified by cysteine scanning mutagenesis and targeted chemical modifi-cation. J. Biol. Chem. 270:23065–23071.

Editor: J. B. Bliska

VOL. 77, 2009 ALPHA-TOXIN IS NOT SUFFICIENT FOR PHAGOLYSOSOME ESCAPE 3625

at UN

IV B

IBLIO

TH

EK

GR

EIF

SW

ALD

on August 19, 2009

iai.asm.org

Dow

nloaded from