NOTES. The Animal Pathogen-Like Type III Secretion System Is Required for the Intracellular Survival of Burkholderia mallei within J774.

Similar documents
Type III Secretion: a Virulence Factor Delivery System Essential for the Pathogenicity of Burkholderia mallei

TITLE: Anti-Inflammatory Cytokine Il-10 and Mammary Gland Development. CONTRACTING ORGANIZATION: University of Buffalo Buffalo, New York

Monoclonal Antibodies Passively Protect BALB/c Mice against Burkholderia mallei Aerosol Challenge

Burkholderia mallei Cluster 1 Type VI Secretion Mutants Exhibit Growth and Actin Polymerization Defects in RAW Murine Macrophages

Pathogenesis of Burkholderia pseudomallei and Burkholderia mallei

Nonlethal Small-Vessel Stopping With High-Power Microwave Technology

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland Approved for public release; distribution unlimited

Distribution Unlimited

AD (Leave blank) The Use of psychiatric Service Dogs in the Treatment of Veterans with PTSD. Craig Love, Ph.D.

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland X Approved for public release; distribution unlimited

Novel treatment opportunities for acute melioidosis and other infections caused by intracellular pathogens

TITLE: The Use of Psychiatric Service Dogs in the Treatment of Veterans with PTSD

Edinburgh Research Explorer

Diagnostic Microbiology and Infectious Disease 55 (2006)

TITLE: Polymicrobial Chronic Infection Including Acinetobacter baumannii in a Plated Segmental Defect in the Rat Femur

Biological Threat Fact Sheets

Exploring simvastatin, an antihyperlipidemic drug, as a potential topical antibacterial agent

PRINCIPAL INVESTIGATOR: Dr. Jetsumon (Sattabongkot) Prachumsri

LEQMYCES MALLEI AND MALLEOMYCES I SEUDOMALLEI

Inactivation of Burkholderia mallei in equine serum for laboratory use.

Antibiotic Resistance in Bacteria

Characterization of Clinically-Attenuated Burkholderia mallei by Whole Genome Sequencing: Candidate Strain for Exclusion from Select Agent Lists

Gliding Motility Assay for P. berghei Sporozoites

Guidelines for Laboratory Verification of Performance of the FilmArray BCID System

Impact of Spores on the Comparative Efficacies of Five Antibiotics. Pharmacodynamic Model

Evaluation of Carbohydrate-Derived Fulvic Acid (CHD-FA) as a Topical Broad-Spectrum Antimicrobial for Drug-Resistant Wound Infections.

Visit ABLE on the Web at:

Development and Characterization of Mouse Models of Infection with Aerosolized Brucella melitensis and Brucella suis

Methicillin-Resistant Staphylococcus aureus

Innate immune response to Burkholderia mallei

Typhoid fever - priorities for research and development of new treatments

Test Method Modified Association of Analytical Communities Test Method Modified Germicidal Spray Products as Disinfectants

SUPPLEMENTARY INFORMATION

The Disinfecting Effect of Electrolyzed Water Produced by GEN-X-3. Laboratory of Diagnostic Medicine, College of Medicine, Soonchunhyang University

Approved by the Food Safety Commission on September 30, 2004

Int.J.Curr.Microbiol.App.Sci (2017) 6(11):

Overview. There are commonly found arrangements of bacteria based on their division. Spheres, Rods, Spirals

Mesosomes are a definite event in antibiotic-treated Staphylococcus aureus ATCC 25923

Dual Antibiotic Delivery from Chitosan Sponges Prevents In Vivo Polymicrobial Biofilm Infections

Electron Microscopic Observations on Ciliated Epithelium of Tracheal Organ Cultures Infected with Bordetella bronchiseptica

Course: Microbiology in Health and Disease

Course: Microbiology in Health and Disease Office Hours: Before or after Class or by appointment

Introduction to Chemotherapeutic Agents. Munir Gharaibeh MD, PhD, MHPE School of Medicine, The university of Jordan November 2018

BIOL 2900 D 4.00 Microbiology in Health/Disease

on February 12, 2018 by guest

CONTRACTING ORGANIZATION: Rutgers, The State University of New Jersey Newark, NJ

COURSE SYLLABUS. (Clinical Bacteriology-1

Burn Infection & Laboratory Diagnosis

R-factor mediated trimethoprim resistance: result of two three-month clinical surveys

Penetration and Intracellular Growth of Brucella abortus in

DTIC I., I, I 8 8. N LD Lfl 0. N. IELECTE FEB2 8 89D Gordon R. Dreesman HTLV III VIRUS ISOLATION STUDIES ANNUAL REPORT. October 30, 1987.

Recommended for Implementation at Step 7 of the VICH Process on 15 December 2004 by the VICH Steering Committee

Bacteriology and Mycology Course Specifications ( ) A. BASIC INFORMATION B. PROFESSIONAL INFORMATION

Antimicrobial agents

Evaluation of a computerized antimicrobial susceptibility system with bacteria isolated from animals

VOL. XXIII NO. II THE JOURNAL OF ANTIBIOTICS 559. ANTIBIOTIC 6640.* Ill

Arrested oocyst maturation in Plasmodium parasites. lacking type II NADH:ubiquinone dehydrogenase

Please distribute a copy of this information to each provider in your organization.

USA Product Label CLINTABS TABLETS. Virbac. brand of clindamycin hydrochloride tablets. ANADA # , Approved by FDA DESCRIPTION

Supporting Online Material for

Isolation of antibiotic producing Actinomycetes from soil of Kathmandu valley and assessment of their antimicrobial activities

An#bio#cs and challenges in the wake of superbugs

Consequences of delayed ciprofloxacin and doxycycline. treatment regimens against F. tularensis airway infection

Informing Public Policy on Agricultural Use of Antimicrobials in the United States: Strategies Developed by an NGO

Medical Bacteriology- Lecture 14. Gram negative coccobacilli. Zoonosis. Brucella. Yersinia. Francesiella

Welcome to Pathogen Group 9

Intracellular Trafficking of Brucella abortus in J774 Macrophages

EXTENDED-SPECTRUM BETA-LACTAMASE (ESBL) TESTING

Course Offerings: Associate of Applied Science Veterinary Technology. Course Number Name Credits

Influence of ph on Adaptive Resistance of Pseudomonas aeruginosa to Aminoglycosides and Their Postantibiotic Effects

Virulent Brucella abortus Prevents Lysosome Fusion and Is Distributed within Autophagosome-Like Compartments

Mechanisms and Pathways of AMR in the environment

Use of a Recombinant Burkholderia Intracellular Motility A Protein for Immunodiagnosis of Glanders

VETERINARY BACTERIOLOGY FROM THE DARK AGES TO THE PRESENT DAY

Infecting Anopheles stephensi With Rodent Malaria Parasites Alida Coppi & Photini Sinnis

MICHAEL J. RYBAK,* ELLIE HERSHBERGER, TABITHA MOLDOVAN, AND RICHARD G. GRUCZ

Entry and Intracellular Localization of Brucella spp. in Vero Cells: Fluorescence and Electron Microscopy

HISTOLOGY OF MAMMARY GLAND DURING LACTATING AND NON-LACTATING PHASES OF MADRAS RED SHEEP WITH SPECIAL REFERENCE TO INVOLUTION

EUROPEAN REFERENCE LABORATORY (EU-RL) FOR BOVINE TUBERCULOSIS WORK-PROGRAMME PROPOSAL Version 2 VISAVET. Universidad Complutense de Madrid

EXPRESSION OF BACILLUS ANTHRACIS PROTECTIVE ANTIGEN IN VACCINE STRAIN BRUCELLA ABORTUS RB51. Sherry Poff

Lack of a Role for Natural Killer Cells in Early Control of Brucella abortus 2308 Infections in Mice

Federal Expert Select Agent Panel (FESAP) Deliberations

Monitoring of antimicrobial resistance in Campylobacter EURL AR activities in framework of the new EU regulation Lina Cavaco

2008 FELINE HEALTH GRANT AWARDS 10 projects funded for a total of $135,860

Antimicrobial agents. are chemicals active against microorganisms

The Type IV Pilin of Burkholderia mallei Is Highly Immunogenic but Fails To Protect against Lethal Aerosol Challenge in a Murine Model

Burton's Microbiology for the Health Sciences. Chapter 9. Controlling Microbial Growth in Vivo Using Antimicrobial Agents

Understanding and prevention of transmission of antibiotic resistance between bacterial populations and One Health reservoirs

Cell Wall Inhibitors. Assistant Professor Naza M. Ali. Lec 3 7 Nov 2017

Presence of extended spectrum β-lactamase producing Escherichia coli in

Drug resistance in relation to use of silver sulphadiazine cream in a burns unit

Antibiotic resistance of bacteria along the food chain: A global challenge for food safety

Hand washing, Asepsis, Precautions and Infection Control

Lactose-Fermenting Bacteria Isolated from

Mice Lacking Components of Adaptive Immunity Show Increased Brucella abortus virb Mutant Colonization

ELECTROPHORETIC ANALYSIS OF SERUM PROTEINS OF BIRDS AND MAMMALS

Characterization and Genetic Complementation of a Brucella abortus High-Temperature-Requirement A (htra) Deletion Mutant

PCR detection of Leptospira in. stray cat and

The Salmonella. Dr. Hala Al Daghisatni

SCANNING electron - microscopy has

Transcription:

INFECTION AND IMMUNITY, July 2006, p. 4349 4353 Vol. 74, No. 7 0019-9567/06/$08.00 0 doi:10.1128/iai.01939-05 NOTES The Animal Pathogen-Like Type III Secretion System Is Required for the Intracellular Survival of Burkholderia mallei within J774.2 Macrophages Wilson J. Ribot and Ricky L. Ulrich* Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland 21702-5011 Received 28 November 2005/Returned for modification 19 January 2006/Accepted 30 March 2006 Burkholderia mallei is a highly infectious gram-negative pathogen and is the causative agent of human and animal glanders. By generating polar mutations (disruption of bsaq and bsaz) in the B. mallei ATCC 23344 animal pathogen-like type III secretion system (TTS), we demonstrate that this bacterial protein delivery system is required for intracellular growth of B. mallei in J774.2 cells, formation of macrophage membrane protrusions, actin polymerization, and phagosomal escape. These findings suggest that TTS plays a role in the intracellular trafficking of B. mallei and may facilitate cell-to-cell spread via actin-based motility. * Corresponding author. Mailing address: Bacteriology Division, USAMRIID, 1425 Porter St., Fort Detrick, Frederick, MD 21702. Phone: (301) 619-8332. Fax: (301) 619-8351. E-mail: Ricky.Ulrich @AMEDD.ARMY.MIL. Burkholderia mallei is the etiologic agent of glanders and is a gram-negative, oxidase-positive, nonmotile bacillus that is a facultative intracellular pathogen (3). The natural hosts for B. mallei are horses, donkeys, and mules (solipeds), but other animals, including mice, hamsters, guinea pigs, monkeys, lions, and dogs, are susceptible to this pathogen (3, 7). Cases of human acquired glanders in the United States are uncommon since the implementation of quarantine strategies for imported animals in the 21st century (3). Both human and animal glanders can occur in acute and chronic forms, depending on the route of exposure. For humans, acute glanders is acquired via the inhalational route (without treatment, death results within days) while chronic forms of the disease occur following cutaneous exposure to the pathogen (3). Various gram-negative bacterial pathogens utilize type III secretion (TTS) for toxin delivery (i.e., effector proteins) into the cytoplasm of infected cells, which in turn subverts host immunological responses to the microbe (2, 6). Recent investigations have demonstrated that the B. mallei and Burkholderia pseudomallei animal pathogen-like TTSs, which are similar to the Salmonella Inv/Spa/Prg and Shigella Ipa/Mxi/Spa TTS networks, are important for in vitro and in vivo survival of these pathogenic Burkholderia species (9 12, 14, 15). These gram-negative bacterial TTSs function to translocate bacterial effector proteins directly into the host cell, which allows bacterial pathogens to evade or alter host defenses (2, 5, 6). We have recently shown that the B. mallei ATCC 23344 animal pathogen-like TTS is essential for full virulence in BALB/c mice and Syrian hamsters (14). In this investigation, we demonstrate that the B. mallei animal pathogen-like TTS is required for intracellular survival in J774.2 macrophages. Disruption of this TTS drastically reduced intracellular replication within J774.2 cells, prevented phagosomal escape into the host cell cytoplasm, and inhibited membrane protrusion and actin tail formation. B. mallei strains used in this study. The bacterial strains used in this investigation, RD01 (bsaq::ptsv3) and RD02 (bsaz::ptsv3), have been described by Ulrich and DeShazer (14). These B. mallei strains contain polar mutations (disrupted homologues of the Salmonella enterica serovar Typhimurium inva and spas genes) within the animal pathogen-like TTS and are avirulent in both hamsters and BALB/c mice (14). To analyze RD01 and RD02 in vitro, a broad-host-range expression plasmid (pbhr1) encoding the green fluorescent protein (GFP) was designed as previously described (8) and mobilized into RD01 and RD02 according to the methods of DeShazer et al. (4). Macrophage intracellular replication assays. To determine if disrupting the B. mallei animal pathogen-like TTS affects intracellular survival and growth, J774.2 macrophages were infected with either wild-type B. mallei, RD01, or RD02 at a multiplicity of infection of 10 bacteria per macrophage in sixwell plates. Cells were centrifuged for 5 min at 1,500 rpm and incubated for 1hat37 C with 5% CO 2. Cells were washed three times with phosphate-buffered saline (PBS) and overlaid with medium containing 250 g/ml kanamycin to prevent the growth of extracellular bacteria. At 3, 6, 12, and 18 h postinfection, macrophages were washed and lysed with 1 ml of cold sterile water containing 0.1% Triton X-100 (Sigma, St. Louis, Mo.). Cellular lysates were serially diluted 10-fold, and bacteria were enumerated on Luria-Bertani plates (Fisher Scientific, Pittsburgh, Pa.) supplemented with 4% glycerol containing 100 g/ml trimethoprim (Sigma). All assays were performed in duplicate. As Fig. 1 depicts, the B. mallei animal pathogen-like TTS is important for the intracellular survival of B. mallei in J774.2 macrophages. At 6 h postexposure (p.e.), both RD01 4349

Report Documentation Page Form Approved OMB No. 0704-0188 Public reporting burden for the collection of information is estimated to average 1 hour per response, including the time for reviewing instructions, searching existing data sources, gathering and maintaining the data needed, and completing and reviewing the collection of information. Send comments regarding this burden estimate or any other aspect of this collection of information, including suggestions for reducing this burden, to Washington Headquarters Services, Directorate for Information Operations and Reports, 1215 Jefferson Davis Highway, Suite 1204, Arlington VA 22202-4302. Respondents should be aware that notwithstanding any other provision of law, no person shall be subject to a penalty for failing to comply with a collection of information if it does not display a currently valid OMB control number. 1. REPORT DATE 1 JUL 2006 2. REPORT TYPE N/A 3. DATES COVERED - 4. TITLE AND SUBTITLE The animal pathogen-like type III secretion system is required for intracellular survival of Burkholderia mallei within J774.2 macrophages, Infection and Immunity 74:4349-4353 6. AUTHOR(S) Ribot, WJ Ulrich, RL 5a. CONTRACT NUMBER 5b. GRANT NUMBER 5c. PROGRAM ELEMENT NUMBER 5d. PROJECT NUMBER 5e. TASK NUMBER 5f. WORK UNIT NUMBER 7. PERFORMING ORGANIZATION NAME(S) AND ADDRESS(ES) United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 8. PERFORMING ORGANIZATION REPORT NUMBER RPP-05-461 9. SPONSORING/MONITORING AGENCY NAME(S) AND ADDRESS(ES) 10. SPONSOR/MONITOR S ACRONYM(S) 12. DISTRIBUTION/AVAILABILITY STATEMENT Approved for public release, distribution unlimited 13. SUPPLEMENTARY NOTES The original document contains color images. 11. SPONSOR/MONITOR S REPORT NUMBER(S) 14. ABSTRACT Burkholderia mallei is a highly infectious gram-negative pathogen and is the causative agent of human and animal glanders. By generating polar mutations (disruption of bsaq and bsaz ) in the B. mallei ATCC 23344 animal pathogen-like type III secretion system (TTS), we demonstrate that this bacterial protein delivery system is required for intracellular growth of B. mallei in J774.2 cells, formation of macrophage membrane protrusions, actin polymerization, and phagosomal escape. These findings suggest that TTS plays a role in the intracellular trafficking of B. mallei and may facilitate cell-to-cell spread via actin-based motility. 15. SUBJECT TERMS Burkholderia mallei, type III secretion system, glanders 16. SECURITY CLASSIFICATION OF: 17. LIMITATION OF ABSTRACT SAR a. REPORT unclassified b. ABSTRACT unclassified c. THIS PAGE unclassified 18. NUMBER OF PAGES 5 19a. NAME OF RESPONSIBLE PERSON Standard Form 298 (Rev. 8-98) Prescribed by ANSI Std Z39-18

4350 NOTES INFECT. IMMUN. FIG. 1. Disruption of the B. mallei ATCC 23344 animal pathogen-like TTS prevents intracellular replication of B. mallei within J774.2 murine macrophage-like cells. Infected macrophages were lysed at 3, 6, 12, and 18 h p.e., and bacterial loads were determined in duplicate experiments. RD01 contains a disrupted bsaq gene (BMAA1542), RD02 harbors a mutation in the bsaz allele (BMAA1533), and ATCC 23344 represents wild-type B. mallei. and RD02 were cleared from macrophages while the number (9.8 10 4 CFU) of wild-type B. mallei organisms remained consistent (Fig. 1). At 18 h p.e., the number of wild-type B. mallei organisms reached 1.8 10 7 CFU while RD01 and RD02 remained undetected, suggesting that TTS is essential for the intracellular survival of B. mallei within J774.2 cells (Fig. 1). Interestingly, compared to B. pseudomallei macrophage intracellular growth ( 10 5 CFU at 12 h p.e.) (11), it appears that B. mallei proliferation is more pronounced in murine macrophage-like cells (1.8 10 7 CFU by 12 h p.e.) (data not shown). Since polar mutations were generated in RD01 and RD02 TTS gene complementation studies were not performed. Mutagenesis of the B. mallei bsaq and bsaz TTS genes disrupts host cell actin tail formation. J774.2 murine macrophages were infected with wild-type B. mallei, RD01, or RD02 (each strain constitutively expresses GFP) as described above, with the exception that kanamycin was replaced with 30 g/ml gentamicin (Sigma) to retard the growth of extracellular bacteria (pbhr1 encodes kanamycin resistance). After 6 h of incubation, cells were washed once with PBS and fixed with 4% paraformaldehyde in PBS overnight at 4 C. Cells were permeabilized with 1% Triton X-100, and filamentous actin was stained red by overnight incubation at 4 C with Alexa Fluor 568 -phalloidin diluted to 5 U/ml in PBS (Molecular Probes, Eugene, Oreg.). Microscopic visualization (Nikon Eclipse TE2000-S inverted microscope equipped with a Spot-RT digital camera; Image Systems, Columbia, Md.) of J774.2 cells infected with wild-type B. mallei or each TTS mutant clearly demonstrated that the B. mallei TTS is involved in the induction of host cell actin polymerization (Fig. 2). Consistent with a recent investigation (8), Fig. 2A and D show that wild-type B. mallei ATCC 23344 is capable of causing host cellular actin rearrangements whereas RD01 and RD02 fail to promote actin tail formation. These findings suggest that B. mallei employs the Bsa TTS for intracellular trafficking and cell-to-cell spread. Association of B. mallei with lysosome-associated membrane glycoprotein 1 (LAMP-1). To determine if the B. mallei animal pathogen-like TTS is involved in confining B. mallei within host cell phagosomes, J774.2 macrophages were infected as described for actin polymerization analysis and the association of LAMP-1 with wild-type B. mallei, RD01, and RD02 was determined at 6 h postinfection in duplicate experiments. B. mallei strains constitutively expressing GFP were visualized by their green fluorescence, and LAMP-1 was detected with rat anti-lamp-1 (1D4B; Department of Biological Sciences, The University of Iowa, Iowa City) and an anti-rat immunoglobulin-alexa 568 (red; Molecular Probes) antibody. Bacteria were considered LAMP-1 associated if any region of the bacterial cell was colocalized with LAMP-1. For quantitative analysis (percent association), bacteria localized with LAMP-1 were determined for 500 bacilli of wild-type B. mallei, RD01, and RD02 and scored as LAMP-1 positive if the green fluorescent bacterial cells were surrounded by red fluorescent membranes (data not shown) (1). As depicted in Fig. 3, wild-type B. mallei was infrequently (18% of the bacterial cells analyzed) colocalized with LAMP-1 staining. In contrast, RD01 (97% of the cells) and RD02 (94% of the bacteria) were heavily colocalized with LAMP-1-positive phagosomes. In the majority of the macrophages observed, the pattern of LAMP-1 distribution differed between cells infected with wild-type B. mallei and those infected with either RD01 or RD02 (Fig. 3). The LAMP-1 distribution in cells infected with wild-type B. mallei was perinuclear, whereas in the RD01 and RD02 infections, the LAMP-1 distribution was more uniformly distributed throughout the cell (Fig. 3). A possible explanation for these differences in LAMP-1 distribution is (i) that LAMP-1 has not yet been recruited to the membrane or (ii) that the phagocytosed organisms, in particular, wild-type B. mallei, are able to escape

VOL. 74, 2006 NOTES 4351 FIG. 2. The B. mallei ATCC 23344 animal pathogen-like TTS is involved in the induction of actin-based host cell membrane protrusions. J774.2 cells were infected wild-type B. mallei (A and D), RD01 (B), and RD02 (C) expressing GFP at a multiplicity of infection of 10 bacteria to one macrophage. At 6 h p.e., cells were fixed and cellular actin was stained with Alexa Fluor 568 -phalloidin and viewed at a magnification of 630. into the host cell cytosol (Fig. 3). The LAMP-1 staining pattern(s), in parallel with our actin polymerization assays, suggests that the B. mallei bsa TTS is involved in the access of this pathogen to the host cell cytosol, which, in turn, likely contributes to the differences in the LAMP-1 distribution patterns within macrophages infected with wild-type B. mallei and each TTS mutant (Fig. 2 and 3). These results suggest that wild-type B. mallei is able to escape from phagosomal compartments, whereas RD01 and RD02 remain within the maturing phagolysosomes. TTS and B. mallei phagosomal escape. To further analyze the role of the B. mallei animal pathogen-like TTS, transmission electron microscopy (TEM) was employed to determine the intracellular location of wild-type B. mallei, RD01, and RD02 within J774.2 cells. For TEM, macrophages were infected with wild-type B. mallei, RD01, or RD02 (as described for intracellular replication assays) for 6 h and fixed at 25 C with 2.5% glutaraldehyde in a modified Millonig buffer for 16 h. Cells were stained en bloc with 0.5% uranyl acetate in ethanol. Samples were dehydrated in graded ethanol and propylene oxide and embedded in Poly/Bed 812 resin (Polysciences, Inc., Warrington, Pa.). Ultrathin sections were placed on 200-mesh nickel grids and stained with 5% uranyl acetate and 0.2% lead citrate. Figure 4 demonstrates that wild-type B. mallei is capable of endocytic escape, whereas RD01 and RD02 are confined within cellular phagosomes containing intact membranes. These findings imply that the B. mallei animal pathogen-like TTS is required for phagosomal membrane lysis and bacterial escape into the macrophage cytoplasm. Several virulence determinants have recently been identified

4352 NOTES INFECT. IMMUN. FIG. 3. The Bsa TTS is required for phagosomal escape of B. mallei from infected J774.2 cells. Macrophage-like cells were infected as described for intracellular replication and actin polymerization assays. Wild-type (WT) B. mallei, RD01, and RD02 constitutively express GFP (green). The phagosome-associated protein LAMP-1 was detected with rat anti-lamp-1 and anti-rat immunoglobulin Alexa 568 antibodies (red) at 6 h following infection at a magnification of 630. Host cell nuclei were visualized with 4,6 -diamidino-2-phenylindole (DAPI) stain (blue). Images are from representative slides obtained from duplicate experiments. The Detail column demonstrates that disruption of the Bsa TTS enhances the colocalization of LAMP-1 with RD01 and RD02 at 6 h p.e., whereas the majority of wild-type B. mallei cells (WT B) did not colocalize with LAMP-1 staining. The LAMP-1 staining pattern observed in the wild-type experiments and the low levels of bacterial colocalization are suggestive of bacterial escape into the cytosol. Furthermore, the wild-type bacteria are able to polymerize actin, resulting in membrane protrusions (WT A Detail), which is possible only if the bacteria have gained access to the cytosol.

VOL. 74, 2006 NOTES 4353 FIG. 4. Representative TEM image depicting the intracellular location of wild-type B. mallei (A), RD01 (B), and RD02 (C) at 6 h postinfection. Panel A demonstrates that wild-type B. mallei disrupts the phagosomal membrane, whereas RD01 and RD02 remain within phagosomes containing intact membranes (membranes are labeled with the letter M). The host cell nucleus is labeled with the letter N. Magnification, 25,000. for B. mallei, with the most attenuated strains resulting from disruption of genes involved in B. mallei capsule biosynthesis (wcbb) (4) and the TTS (bsaq and bsaz) (14). In addition, we have shown that a B. mallei branched-chain amino acid auxotroph was significantly attenuated in an aerosol BALB/c and hamster model of acute glanders (13). This investigation further shows the importance of the B. mallei animal pathogenlike TTS and how this bacterial protein delivery system is required for the intracellular replication of B. mallei in J774.2 murine macrophage-like cells, induction of host cell actinbased membrane protrusions, and phagosomal escape. Additional work is needed to further characterize the B. mallei animal pathogen-like TTS and to identify the secreted effector molecule(s). We thank Melanie Ulrich, David Heath, and Katheryn Kenyon for reviewing the manuscript and Kathy Kuehl for performing the electron microscopy. The rat anti-lamp-1 (1D4B) monoclonal antibody developed by J. T. August was obtained from the Developmental Studies Hybridoma Bank developed under the auspices of the NICHD and maintained by the Department of Biological Sciences, The University of Iowa, Iowa City. The research described herein was sponsored by NIAID interagency agreement Y1-AI-5004-01. The opinions, interpretations, conclusions, and recommendations presented here are ours and are not necessarily endorsed by the U.S. Army. REFERENCES 1. Bellaire, B. H., R. M. Roop II, and J. A. Cardelli. 2005. Opsonized virulent Brucella abortus replicates within nonacidic, endoplasmic reticulum-negative, LAMP-1-positive phagosomes in human monocytes. Infect. Immun. 73:3702 3713. 2. Cornelis, G. R., and F. Van Gijsegem. 2000. Assembly and function of type III secretory systems. Annu. Rev. Microbiol. 54:735 774. 3. DeShazer, D., and D. Waag. 2004. Glanders: new insights into an old disease, p. 209 237. In L. Lindler, F. Lebeda, and G. W. Korch (ed.), Biological weapons defense: infectious diseases and counterbioterrorism. The Humana Press, Inc., Totowa, N.J. 4. DeShazer, D., D. M. Waag, D. L. Fritz, and D. E. Woods. 2001. Identification of a Burkholderia mallei polysaccharide gene cluster by subtractive hybridization and demonstration that the encoded capsule is an essential virulence determinant. Microb. Pathog. 30:253 269. 5. Galan, J. E. 2001. Salmonella interactions with host cells: type III secretion at work. Annu. Rev. Cell Dev. Biol. 17:53 86. 6. Hueck, C. J. 1998. Type III protein secretion systems in bacterial pathogens of animals and plants. Microbiol. Mol. Biol. Rev. 62:379 433. 7. Miller, W. R., L. Pannell, L. Cravitz, W. A. Tanner, and T. Rosebury. 1947. Studies on certain biological characteristics of Malleomyces mallei and Malleomyces pseudomallei. J. Bacteriol. 55:115 126. 8. Stevens, J. M., R. L. Ulrich, L. A. Taylor, M. W. Wood, D. Deshazer, M. P. Stevens, and E. E. Galyov. 2005. Actin-binding proteins from Burkholderia mallei and Burkholderia thailandensis can functionally compensate for the actin-based motility defect of a Burkholderia pseudomallei bima mutant. J. Bacteriol. 187:7857 7862. 9. Stevens, M. P., A. Friebel, L. A. Taylor, M. W. Wood, P. J. Brown, W. D. Hardt, and E. E. Galyov. 2003. A Burkholderia pseudomallei type III secreted protein, BopE, facilitates bacterial invasion of epithelial cells and exhibits guanine nucleotide exchange factor activity. J. Bacteriol. 185:4992 4996. 10. Stevens, M. P., A. Haque, T. Atkins, J. Hill, M. W. Wood, A. Easton, M. Nelson, C. Underwood-Fowler, R. W. Titball, G. J. Bancroft, and E. E. Galyov. 2004. Attenuated virulence and protective efficacy of a Burkholderia pseudomallei bsa type III secretion mutant in murine models of melioidosis. Microbiology 150:2669 2676. 11. Stevens, M. P., M. W. Wood, L. A. Taylor, P. Monaghan, P. Hawes, P. W. Jones, T. S. Wallis, and E. E. Galyov. 2002. An Inv/Mxi-Spa-like type III protein secretion system in Burkholderia pseudomallei modulates intracellular behaviour of the pathogen. Mol. Microbiol. 46:649 659. 12. Suparak, S., W. Kespichayawattana, A. Haque, A. Easton, S. Damnin, G. Lertmemongkolchai, G. J. Bancroft, and S. Korbsrisate. 2005. Multinucleated giant cell formation and apoptosis in infected host cells is mediated by Burkholderia pseudomallei type III secretion protein BipB. J. Bacteriol. 187: 6556 6560. 13. Ulrich, R. L., K. Amemiya, D. Waag, C. J. Roy, and D. DeShazer. 2005. Aerogenic vaccination with a Burkholderia mallei auxotroph protects against aerosol-initiated glanders in mice. Vaccine 23:1986 1992. 14. Ulrich, R. L., and D. DeShazer. 2004. Type III secretion: a virulence factor delivery system essential for the pathogenicity of Burkholderia mallei. Infect. Immun. 72:1150 1154. 15. Warawa, J., and D. E. Woods. 2005. Type III secretion system cluster 3 is required for maximal virulence of Burkholderia pseudomallei in a hamster infection model. FEMS Microbiol. Lett. 242:101 108. Editor: V. J. DiRita