1
|
Lowrey LC, Mote KB, Cotter PA. DNA duplication in Burkholderia thailandensis induces biofilm formation by activating a two-component regulatory system. PLoS Genet 2025; 21:e1011528. [PMID: 40392945 DOI: 10.1371/journal.pgen.1011528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 04/24/2025] [Indexed: 05/22/2025] Open
Abstract
Burkholderia thailandensis strain E264 (BtE264) and close relatives stochastically duplicate a 208.6 kb region of chromosome I via RecA-dependent recombination between two nearly identical insertion sequence elements. Because homologous recombination occurs at a constant, low level, populations of BtE264 are always heterogeneous, but cells containing two or more copies of the region (Dup+) have an advantage, and hence predominate, during biofilm growth, while those with a single copy (Dup-) are favored during planktonic growth. Moreover, only Dup+ bacteria form 'efficient' biofilms within 24 hours in liquid medium. We determined that duplicate copies of a subregion containing genes encoding an archaic chaperone-usher pathway pilus (csuFABCDE) and a two-component regulatory system (bfmSR) are necessary and sufficient for generating efficient biofilms and for conferring a selective advantage during biofilm growth. BfmSR functionality is required, as deletion of either bfmS or bfmR, or a mutation predicted to abrogate phosphorylation of BfmR, abrogates biofilm formation. However, duplicate copies of the csuFABCDE genes are not required. Instead, we found that BfmSR controls expression of csuFABCDE and bfmSR by activating a promoter upstream of csuF during biofilm growth or when the 208.6 kb region, or just bfmSR, are duplicated. Single cell analyses showed that duplication of the 208.6 kb region is sufficient to activate BfmSR in 75% of bacteria during planktonic (BfmSR 'OFF') growth conditions. Together, our data indicate that the combination of deterministic two-component signal transduction and stochastic, duplication-mediated activation of that TCS form a bet-hedging strategy that allows BtE264 to survive when conditions shift rapidly from those favoring planktonic growth to those requiring biofilm formation, such as may be encountered in the soils of Southeast Asia and Northern Australia. Our data highlight the positive impact that transposable elements can have on the evolution of bacterial populations.
Collapse
Affiliation(s)
- Lillian C Lowrey
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Katlyn B Mote
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Peggy A Cotter
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
2
|
Lowrey LC, Mote KB, Cotter PA. DNA duplication-mediated activation of a two-component regulatory system serves as a bet-hedging strategy for Burkholderia thailandensis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.09.627470. [PMID: 39713405 PMCID: PMC11661271 DOI: 10.1101/2024.12.09.627470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Burkholderia thailandensis strain E264 (BtE264) and close relatives stochastically duplicate a 208.6 kb region of chromosome I via RecA-dependent recombination between two nearly identical insertion sequence elements. Because homologous recombination occurs at a constant, low level, populations of BtE264 are always heterogeneous, but cells containing two or more copies of the region (Dup+) have an advantage, and hence predominate, during biofilm growth, while those with a single copy (Dup-) are favored during planktonic growth. Moreover, only Dup+ bacteria form 'efficient ' biofilms within 24 hours in liquid medium. We determined that duplicate copies of a subregion containing genes encoding an archaic chaperone-usher pilus (aplFABCDE) and a two-component regulatory system (bubSR) are necessary and sufficient for generating efficient biofilms and for conferring a selective advantage during biofilm growth. BubSR functionality is required, as deletion of either bubS or bubR, or a mutation predicted to abrogate phosphorylation of BubR, abrogates biofilm formation. However, duplicate copies of the aplFABCDE genes are not required. Instead, we found that BubSR controls expression of aplFABCDE and bubSR by activating a promoter upstream of aplF during biofilm growth or when the 208.6 kb region, or just bubSR, are duplicated. Single cell analyses showed that duplication of the 208.6 kb region is sufficient to activate BubSR in 75% of bacteria during planktonic (BubSR 'OFF') growth conditions. Together, our data indicate that the combination of deterministic two-component signal transduction and stochastic, duplication-mediated activation of that TCS form a bet-hedging strategy that allows BtE264 to survive when conditions shift rapidly from those favoring planktonic growth to those requiring biofilm formation, such as may be encountered in the soils of Southeast Asia and Northern Australia. Our data highlight the positive impact that transposable elements can have on the evolution of bacterial populations.
Collapse
Affiliation(s)
| | | | - Peggy A. Cotter
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, North Carolina, USA
| |
Collapse
|
3
|
Ghazali AK, Firdaus-Raih M, Uthaya Kumar A, Lee WK, Hoh CC, Nathan S. Transitioning from Soil to Host: Comparative Transcriptome Analysis Reveals the Burkholderia pseudomallei Response to Different Niches. Microbiol Spectr 2023; 11:e0383522. [PMID: 36856434 PMCID: PMC10100664 DOI: 10.1128/spectrum.03835-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 02/06/2023] [Indexed: 03/02/2023] Open
Abstract
Burkholderia pseudomallei, a soil and water saprophyte, is responsible for the tropical human disease melioidosis. A hundred years since its discovery, there is still much to learn about B. pseudomallei proteins that are essential for the bacterium's survival in and interaction with the infected host, as well as their roles within the bacterium's natural soil habitat. To address this gap, bacteria grown under conditions mimicking the soil environment were subjected to transcriptome sequencing (RNA-seq) analysis. A dual RNA-seq approach was used on total RNA from spleens isolated from a B. pseudomallei mouse infection model at 5 days postinfection. Under these conditions, a total of 1,434 bacterial genes were induced, with 959 induced in the soil environment and 475 induced in bacteria residing within the host. Genes encoding metabolism and transporter proteins were induced when the bacteria were present in soil, while virulence factors, metabolism, and bacterial defense mechanisms were upregulated during active infection of mice. On the other hand, capsular polysaccharide and quorum-sensing pathways were inhibited during infection. In addition to virulence factors, reactive oxygen species, heat shock proteins, siderophores, and secondary metabolites were also induced to assist bacterial adaptation and survival in the host. Overall, this study provides crucial insights into the transcriptome-level adaptations which facilitate infection by soil-dwelling B. pseudomallei. Targeting novel therapeutics toward B. pseudomallei proteins required for adaptation provides an alternative treatment strategy given its intrinsic antimicrobial resistance and the absence of a vaccine. IMPORTANCE Burkholderia pseudomallei, a soil-dwelling bacterium, is the causative agent of melioidosis, a fatal infectious disease of humans and animals. The bacterium has a large genome consisting of two chromosomes carrying genes that encode proteins with important roles for survival in diverse environments as well as in the infected host. While a general mechanism of pathogenesis has been proposed, it is not clear which proteins have major roles when the bacteria are in the soil and whether the same proteins are key to successful infection and spread. To address this question, we grew the bacteria in soil medium and then in infected mice. At 5 days postinfection, bacteria were recovered from infected mouse organs and their gene expression was compared against that of bacteria grown in soil medium. The analysis revealed a list of genes expressed under soil growth conditions and a different set of genes encoding proteins which may be important for survival, replication, and dissemination in an infected host. These proteins are a potential resource for understanding the full adaptation mechanism of this pathogen. In the absence of a vaccine for melioidosis and with treatment being reliant on combinatorial antibiotic therapy, these proteins may be ideal targets for designing antimicrobials to treat melioidosis.
Collapse
Affiliation(s)
- Ahmad-Kamal Ghazali
- Department of Biological Sciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Mohd Firdaus-Raih
- Department of Applied Physics, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
- Institute of Systems Biology, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Asqwin Uthaya Kumar
- Department of Biological Sciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | - Wei-Kang Lee
- Codon Genomics Sdn. Bhd., Seri Kembangan, Selangor, Malaysia
| | - Chee-Choong Hoh
- Codon Genomics Sdn. Bhd., Seri Kembangan, Selangor, Malaysia
| | - Sheila Nathan
- Department of Biological Sciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| |
Collapse
|
4
|
Abstract
The soil saprophyte, Burkholderia pseudomallei, is the causative agent of melioidosis, a disease endemic in South East Asia and northern Australia. Exposure to B. pseudomallei by either inhalation or inoculation can lead to severe disease. B. pseudomallei rapidly shifts from an environmental organism to an aggressive intracellular pathogen capable of rapidly spreading around the body. The expression of multiple virulence factors at every stage of intracellular infection allows for rapid progression of infection. Following invasion or phagocytosis, B. pseudomallei resists host-cell killing mechanisms in the phagosome, followed by escape using the type III secretion system. Several secreted virulence factors manipulate the host cell, while bacterial cells undergo a shift in energy metabolism allowing for overwhelming intracellular replication. Polymerisation of host cell actin into “actin tails” propels B. pseudomallei to the membranes of host cells where the type VI secretion system fuses host cells into multinucleated giant cells (MNGCs) to facilitate cell-to-cell dissemination. This review describes the various mechanisms used by B. pseudomallei to survive within cells.
Collapse
Affiliation(s)
- Nicole M Bzdyl
- The Marshall Centre for Infectious Diseases Research and Training, School of Biomedical Sciences, University of Western Australia, Perth, Western Australia, 6009, Australia
| | - Clare L Moran
- The Marshall Centre for Infectious Diseases Research and Training, School of Biomedical Sciences, University of Western Australia, Perth, Western Australia, 6009, Australia
| | - Justine Bendo
- The Marshall Centre for Infectious Diseases Research and Training, School of Biomedical Sciences, University of Western Australia, Perth, Western Australia, 6009, Australia
| | - Mitali Sarkar-Tyson
- The Marshall Centre for Infectious Diseases Research and Training, School of Biomedical Sciences, University of Western Australia, Perth, Western Australia, 6009, Australia
| |
Collapse
|
5
|
Quilici G, Berardi A, Fabris C, Ghitti M, Punta M, Gourlay LJ, Bolognesi M, Musco G. Solution Structure of the BPSL1445 Protein of Burkholderia pseudomallei Reveals the SYLF Domain Three-Dimensional Fold. ACS Chem Biol 2022; 17:230-239. [PMID: 34968022 DOI: 10.1021/acschembio.1c00886] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The SYLF domain is an evolutionary conserved protein domain with phosphatidylinositol binding ability, whose three-dimensional structure is unknown. Here, we present the solution structure and the dynamics characterization of the SYLF domain of the bacterial BPSL1445 protein. BPSL1445 is a seroreactive antigen and a diagnostic marker of Burkholderia pseudomallei, the etiological agent of melioidosis, a severe infectious disease in the tropics. The BPSL1445 SYLF domain (BPSL1445-SYLF) consists of a β-barrel core, with two flexible loops protruding out of the barrel and three helices packing on its surface. Our structure allows for a more precise definition of the boundaries of the SYLF domain compared to the previously reported one and suggests common ancestry with bacterial EipA domains. We also demonstrate by phosphatidyl-inositol phosphate arrays and nuclear magnetic resonance titrations that BPSL1445-SYLF weakly interacts with phosphoinositides, thus supporting lipid binding abilities of this domain also in prokaryotes.
Collapse
Affiliation(s)
- Giacomo Quilici
- Biomolecular NMR Laboratory, I.R.C.C.S. Ospedale San Raffaele, Via Olgettina 58, 20132 Milan, Italy
| | - Andrea Berardi
- Biomolecular NMR Laboratory, I.R.C.C.S. Ospedale San Raffaele, Via Olgettina 58, 20132 Milan, Italy
| | - Chantal Fabris
- Biomolecular NMR Laboratory, I.R.C.C.S. Ospedale San Raffaele, Via Olgettina 58, 20132 Milan, Italy
| | - Michela Ghitti
- Biomolecular NMR Laboratory, I.R.C.C.S. Ospedale San Raffaele, Via Olgettina 58, 20132 Milan, Italy
| | - Marco Punta
- Unit of Immunogenetics, Leukemia Genomics and Immunobiology and Center for Omics Sciences, I.R.C.C.S. Ospedale San Raffaele, Via Olgettina 58, 20132 Milan, Italy
| | - Louise J. Gourlay
- Department of Biosciences, University of Milano, Via Celoria 26, 20133 Milan, Italy
- Centro di Ricerca Pediatrica Romeo ed Enrica Invernizzi, Università degli Studi di Milano, 20133 Milan, Italy
| | - Martino Bolognesi
- Department of Biosciences, University of Milano, Via Celoria 26, 20133 Milan, Italy
- Centro di Ricerca Pediatrica Romeo ed Enrica Invernizzi, Università degli Studi di Milano, 20133 Milan, Italy
| | - Giovanna Musco
- Biomolecular NMR Laboratory, I.R.C.C.S. Ospedale San Raffaele, Via Olgettina 58, 20132 Milan, Italy
| |
Collapse
|
6
|
Shropshire H, Jones RA, Aguilo-Ferretjans MM, Scanlan DJ, Chen Y. Proteomics insights into the Burkholderia cenocepacia phosphorus stress response. Environ Microbiol 2021; 23:5069-5086. [PMID: 33684254 DOI: 10.1111/1462-2920.15451] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 03/02/2021] [Indexed: 11/26/2022]
Abstract
The Burkholderia cepacia complex is a group of Burkholderia species that are opportunistic pathogens causing high mortality rates in patients with cystic fibrosis. An environmental stress often encountered by these soil-dwelling and pathogenic bacteria is phosphorus limitation, an essential element for cellular processes. Here, we describe cellular and extracellular proteins differentially regulated between phosphate-deplete (0 mM, no added phosphate) and phosphate-replete (1 mM) growth conditions using a comparative proteomics (LC-MS/MS) approach. We observed a total of 128 and 65 unique proteins were downregulated and upregulated respectively, in the B. cenocepacia proteome. Of those downregulated proteins, many have functions in amino acid transport/metabolism. We have identified 24 upregulated proteins that are directly/indirectly involved in inorganic phosphate or organic phosphorus acquisition. Also, proteins involved in virulence and antimicrobial resistance were differentially regulated, suggesting B. cenocepacia experiences a dramatic shift in metabolism under these stress conditions. Overall, this study provides a baseline for further research into the biology of Burkholderia in response to phosphorus stress.
Collapse
Affiliation(s)
- Holly Shropshire
- BBSRC Midlands Integrative Biosciences Training Partnership, University of Warwick, Coventry, CV4 7AL, UK.,School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
| | - Rebekah A Jones
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
| | | | - David J Scanlan
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
| | - Yin Chen
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
| |
Collapse
|
7
|
Trevino SR, Dankmeyer JL, Fetterer DP, Klimko CP, Raymond JLW, Moreau AM, Soffler C, Waag DM, Worsham PL, Amemiya K, Ruiz SI, Cote CK, Krakauer T. Comparative virulence of three different strains of Burkholderia pseudomallei in an aerosol non-human primate model. PLoS Negl Trop Dis 2021; 15:e0009125. [PMID: 33571211 PMCID: PMC7904162 DOI: 10.1371/journal.pntd.0009125] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 02/24/2021] [Accepted: 01/11/2021] [Indexed: 01/19/2023] Open
Abstract
Melioidosis, caused by the Gram-negative bacterium Burkholderia pseudomallei, is a major cause of sepsis and mortality in endemic regions of Southeast Asia and Northern Australia. B. pseudomallei is a potential bioterrorism agent due to its high infectivity, especially via inhalation, and its inherent resistance to antimicrobials. There is currently no vaccine for melioidosis and antibiotic treatment can fail due to innate drug resistance, delayed diagnosis and treatment, or insufficient duration of treatment. A well-characterized animal model that mimics human melioidosis is needed for the development of new medical countermeasures. This study first characterized the disease progression of melioidosis in the African green monkey (AGM) and rhesus macaque (RM) for non-human primate model down-selection. All AGMs developed acute lethal disease similar to that described in human acute infection following exposure to aerosolized B. pseudomallei strain HBPUB10134a. Only 20% of RMs succumbed to acute disease. Disease progression, immune response and pathology of two other strains of B. pseudomallei, K96243 and MSHR5855, were also compared using AGMs. These three B. pseudomallei strains represent a highly virulent strain from Thailand (HBPUB101034a), a highly virulent strains from Australia (MSHR5855), and a commonly used laboratory strains originating from Thailand (K96243). Animals were observed for clinical signs of infection and blood samples were analyzed for cytokine responses, blood chemistry and leukocyte changes in order to characterize bacterial infection. AGMs experienced fever after exposure to aerosolized B. pseudomallei at the onset of acute disease. Inflammation, abscesses and/or pyogranulomas were observed in lung with all three strains of B. pseudomallei. Inflammation, abscesses and/or pyogranulomas were observed in lymph nodes, spleen, liver and/or kidney with B. pseudomallei, HBPUB10134a and K96243. Additionally, the Australian strain MSHR5855 induced brain lesions in one AGM similar to clinical cases of melioidosis seen in Australia. Elevated serum levels of IL-1β, IL-1 receptor antagonist, IL-6, MCP-1, G-CSF, HGF, IFNγ, MIG, I-TAC, and MIP-1β at terminal end points can be significantly correlated with non-survivors with B. pseudomallei infection in AGM. The AGM model represents an acute model of B. pseudomallei infection for all three strains from two geographical locations and will be useful for efficacy testing of vaccines and therapeutics against melioidosis. In summary, a dysregulated immune response leading to excessive persistent inflammation and inflammatory cell death is the key driver of acute melioidosis. Early intervention in these pathways will be necessary to counter B. pseudomallei and mitigate the pathological consequences of melioidosis. Melioidosis, caused by the Gram-negative bacterium Burkholderia pseudomallei, is endemic in tropical regions globally and is an emerging threat in non-tropical areas worldwide. Its mortality rate is high in endemic areas due to its high infectivity, antimicrobial resistance, lack of available vaccines and limited treatment options. Animal model development and pathogenicity studies of various isolates are critical for the development of countermeasures against this pathogen. In this study, we compared the virulence of three different isolates of B. pseudomallei from two geographical locations in an aerosol non-human primate model. We found that early elevations of both pro-inflammatory and anti-inflammatory mediators, as well as the persistence of these mediators in the terminal phase of bacterial infection correlate with mortality. Histopathological analysis showed that the severity of lesions in various organs also correlates with the virulence of the B. pseudomallei strains, HBPUB10134a, MSHR5855 and K96243. Thus, a dysregulated immune response leading to excessive IL-1β and IL-6 at terminal end points and necrosis are key drivers of acute melioidosis. Development of drugs targeting these host response processes will be necessary to counter B. pseudomallei and mitigate the pathological consequences of melioidosis. This non-human primate model will facilitate the screening of vaccines and novel therapeutics.
Collapse
Affiliation(s)
- Sylvia R. Trevino
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America
| | - Jennifer L. Dankmeyer
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America
| | - David P. Fetterer
- Biostatistics Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America
| | - Christopher P. Klimko
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America
| | - Jo Lynne W. Raymond
- Pathology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America
| | - Alicia M. Moreau
- Pathology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America
| | - Carl Soffler
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America
| | - David M. Waag
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America
| | - Patricia L. Worsham
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America
| | - Kei Amemiya
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America
| | - Sara I. Ruiz
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America
| | - Christopher K. Cote
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America
- * E-mail: (CKC); (TK)
| | - Teresa Krakauer
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America
- * E-mail: (CKC); (TK)
| |
Collapse
|
8
|
Rao C, Mao C, Xia Y, Zhang M, Hu Z, Yuan S, Yang W, Yan J, Deng L, Cai X, Mao X, Li Q, Liao Y. Transcriptome Analysis Reveals Unfolded Protein Response Was Induced During the Early Stage of Burkholderia pseudomallei Infection in A549 Cells. Front Genet 2020; 11:585203. [PMID: 33363569 PMCID: PMC7753206 DOI: 10.3389/fgene.2020.585203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 11/09/2020] [Indexed: 12/17/2022] Open
Abstract
Burkholderia pseudomallei is a zoonotic pathogen that usually affects patients' lungs and causes serious melioidosis. The interaction of B. pseudomallei with its hosts is complex, and cellular response to B. pseudomallei infection in humans still remains to be elucidated. In this study, transcriptomic profiling of B. pseudomallei-infected human lung epithelial A549 cells was performed to characterize the cellular response dynamics during the early infection (EI) stage. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were performed by using the online databases DAVID 6.8 and KOBAS 3.0. Real-time quantitative PCR and western blot were used for validation experiments. Compared with the negative control group (NC), a set of 36 common genes varied over time with a cut-off level of 1.5-fold change, and a P-value < 0.05 was identified. Bioinformatics analysis indicated that the PERK-mediated unfolded protein response (UPR) was enriched as the most noteworthy biological process category, which was enriched as a branch of UPR in the signaling pathway of protein processing in the endoplasmic reticulum. Other categories, such as inflammatory responses, cell migration, and apoptosis, were also focused. The molecular chaperone Bip (GRP78), PERK, and PERK sensor-dependent phosphorylation of eIF2α (p-eIF2α) and ATF4 were verified to be increasing over time during the EI stage, suggesting that B. pseudomallei infection activated the PERK-mediated UPR in A549 cells. Collectively, these results provide important initial insights into the intimate interaction between B. pseudomallei and lung epithelial cells, which can be further explored toward the elucidation of the cellular mechanisms of B. pseudomallei infections in humans.
Collapse
Affiliation(s)
- Chenglong Rao
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, China
| | - Chan Mao
- Department of Pharmacy, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yupei Xia
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, China
| | - Meijuan Zhang
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhiqiang Hu
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, China
| | - Siqi Yuan
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, China
| | - Wenbo Yang
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jingmin Yan
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, China
| | - Ling Deng
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiaolian Cai
- Department of Cardiology, First Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xuhu Mao
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, China
| | - Qian Li
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yaling Liao
- Department of Clinical Microbiology and Immunology, College of Pharmacy and Medical Laboratory, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
9
|
Functional redundancy of Burkholderia pseudomallei phospholipase C enzymes and their role in virulence. Sci Rep 2020; 10:19242. [PMID: 33159122 PMCID: PMC7648637 DOI: 10.1038/s41598-020-76186-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 10/21/2020] [Indexed: 11/12/2022] Open
Abstract
Phospholipase C (PLC) enzymes are key virulence factors in several pathogenic bacteria. Burkholderia pseudomallei, the causative agent of melioidosis, possesses at least three plc genes (plc1, plc2 and plc3). We found that in culture medium plc1 gene expression increased with increasing pH, whilst expression of the plc3 gene was pH (4.5 to 9.0) independent. Expression of the plc2 gene was not detected in culture medium. All three plc genes were expressed during macrophage infection by B. pseudomallei K96243. Comparing B. pseudomallei wild-type with plc mutants revealed that plc2, plc12 or plc123 mutants showed reduced intracellular survival in macrophages and reduced plaque formation in HeLa cells. However, plc1 or plc3 mutants showed no significant differences in plaque formation compared to wild-type bacteria. These findings suggest that Plc2, but not Plc1 or Plc3 are required for infection of host cells. In Galleria mellonella, plc1, plc2 or plc3 mutants were not attenuated compared to the wild-type strain, but multiple plc mutants showed reduced virulence. These findings indicate functional redundancy of the B. pseudomallei phospholipases in virulence.
Collapse
|
10
|
Abstract
The regulation and timely expression of bacterial genes during infection is critical for a pathogen to cause an infection. Bacteria have multiple mechanisms to regulate gene expression in response to their environment, one of which is two-component systems (TCS). TCS have two components. One component is a sensory histidine kinase (HK) that autophosphorylates when activated by a signal. The activated sensory histidine kinase then transfers the phosphoryl group to the second component, the response regulator, which activates transcription of target genes. The genus Burkholderia contains members that cause human disease and are often extensively resistant to many antibiotics. The Burkholderia cepacia complex (BCC) can cause severe lung infections in patients with cystic fibrosis (CF) or chronic granulomatous disease (CGD). BCC members have also recently been associated with several outbreaks of bacteremia from contaminated pharmaceutical products. Separate from the BCC is Burkholderia pseudomallei, which is the causative agent of melioidosis, a serious disease that occurs in the tropics, and a potential bioterrorism weapon. Bioinformatic analysis of sequenced Burkholderia isolates predicts that most strains have at least 40 TCS. The vast majority of these TCS are uncharacterized both in terms of the signals that activate them and the genes that are regulated by them. This review will highlight TCS that have been described to play a role in virulence in either the BCC or B. pseudomallei Since many of these TCS are involved in virulence, TCS are potential novel therapeutic targets, and elucidating their function is critical for understanding Burkholderia pathogenesis.
Collapse
|
11
|
Duong LT, Schwarz S, Gross H, Breitbach K, Hochgräfe F, Mostertz J, Eske-Pogodda K, Wagner GE, Steinmetz I, Kohler C. GvmR - A Novel LysR-Type Transcriptional Regulator Involved in Virulence and Primary and Secondary Metabolism of Burkholderia pseudomallei. Front Microbiol 2018; 9:935. [PMID: 29867844 PMCID: PMC5964159 DOI: 10.3389/fmicb.2018.00935] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 04/23/2018] [Indexed: 01/22/2023] Open
Abstract
Burkholderia pseudomallei is a soil-dwelling bacterium able to survive not only under adverse environmental conditions, but also within various hosts which can lead to the disease melioidosis. The capability of B. pseudomallei to adapt to environmental changes is facilitated by the large number of regulatory proteins encoded by its genome. Among them are more than 60 uncharacterized LysR-type transcriptional regulators (LTTRs). Here we analyzed a B. pseudomallei mutant harboring a transposon in the gene BPSL0117 annotated as a LTTR, which we named gvmR (globally acting virulence and metabolism regulator). The gvmR mutant displayed a growth defect in minimal medium and macrophages in comparison with the wild type. Moreover, disruption of gvmR rendered B. pseudomallei avirulent in mice indicating a critical role of GvmR in infection. These defects of the mutant were rescued by ectopic expression of gvmR. To identify genes whose expression is modulated by GvmR, global transcriptome analysis of the B. pseudomallei wild type and gvmR mutant was performed using whole genome tiling microarrays. Transcript levels of 190 genes were upregulated and 141 genes were downregulated in the gvmR mutant relative to the wild type. Among the most downregulated genes in the gvmR mutant were important virulence factor genes (T3SS3, T6SS1, and T6SS2), which could explain the virulence defect of the gvmR mutant. In addition, expression of genes related to amino acid synthesis, glyoxylate shunt, iron-sulfur cluster assembly, and syrbactin metabolism (secondary metabolite) was decreased in the mutant. On the other hand, inactivation of GvmR increased expression of genes involved in pyruvate metabolism, ATP synthesis, malleobactin, and porin genes. Quantitative real-time PCR verified the differential expression of 27 selected genes. In summary, our data show that GvmR acts as an activating and repressing global regulator that is required to coordinate expression of a diverse set of metabolic and virulence genes essential for the survival in the animal host and under nutrient limitation.
Collapse
Affiliation(s)
- Linh Tuan Duong
- Friedrich Loeffler Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Sandra Schwarz
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, University of Tübingen, Tübingen, Germany
| | - Harald Gross
- Department of Pharmaceutical Biology, Pharmaceutical Institute, Eberhard Karls University of Tübingen, Tübingen, Germany.,German Centre for Infection Research, Partner Site Tübingen, Tübingen, Germany
| | - Katrin Breitbach
- Friedrich Loeffler Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Falko Hochgräfe
- Competence Center Functional Genomics, Junior Research Group Pathoproteomics, University of Greifswald, Greifswald, Germany
| | - Jörg Mostertz
- Competence Center Functional Genomics, Junior Research Group Pathoproteomics, University of Greifswald, Greifswald, Germany
| | - Kristin Eske-Pogodda
- Friedrich Loeffler Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Gabriel E Wagner
- Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Graz, Austria
| | - Ivo Steinmetz
- Friedrich Loeffler Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany.,Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Graz, Austria
| | - Christian Kohler
- Friedrich Loeffler Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
12
|
Norris MH, Heacock-Kang Y, Zarzycki-Siek J, Bluhm AP, McMillan IA, Schweizer HP, Hoang TT. Burkholderia pseudomallei natural competency and DNA catabolism: Identification and characterization of relevant genes from a constructed fosmid library. PLoS One 2017; 12:e0189018. [PMID: 29253888 PMCID: PMC5734746 DOI: 10.1371/journal.pone.0189018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Accepted: 11/18/2017] [Indexed: 11/19/2022] Open
Abstract
Burkholderia spp. are genetically and physiologically diverse. Some strains are naturally transformable and capable of DNA catabolism. Burkholderia pseudomallei (Bp) strains 1026b and K96243 and B. thailandensis strain E264 are able to utilize DNA as a sole carbon source for growth, while only strains 1026b and E264 are naturally transformable. In this study, we constructed low-copy broad-host-range fosmid library, containing Bp strain 1026b chromosomal DNA fragments, and employed a novel positive selection approach to identify genes responsible for DNA uptake and DNA catabolism. The library was transferred to non-competent Bp K96243 and B. cenocepacia (Bc) K56-2, harboring chromosomally-inserted FRT-flanked sacB and pheS counter-selection markers. The library was incubated with DNA encoding Flp recombinase, followed by counter-selection on sucrose and chlorinated phenylalanine, to select for clones that took up flp-DNA, transiently expressed Flp, and excised the sacB-pheS cassette. Putative clones that survived the counter-selection were subsequently incubated with gfp-DNA and bacteria were visualized via fluorescent microscopy to confirm natural competency. Fosmid sequencing identified several 1026b genes implicated in DNA uptake, which were validated using chromosomal mutants. One of the naturally competent clones selected in Bc K56-2 enabled Bc, Bp and B. mallei to utilize DNA as a sole carbon source, and all fosmids were used to successfully create mutations in non-naturally-competent B. mallei and Bp strains.
Collapse
Affiliation(s)
- Michael H. Norris
- Department of Microbiology, University of Hawaii at Manoa, Honolulu, HI, United States of America
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, HI, United States of America
- Department of Infectious Diseases and Pathology, University of Florida, Gainesville, Florida, United States of America
| | - Yun Heacock-Kang
- Department of Microbiology, University of Hawaii at Manoa, Honolulu, HI, United States of America
| | - Jan Zarzycki-Siek
- Department of Microbiology, University of Hawaii at Manoa, Honolulu, HI, United States of America
| | - Andrew P. Bluhm
- Department of Microbiology, University of Hawaii at Manoa, Honolulu, HI, United States of America
| | - Ian A. McMillan
- Department of Microbiology, University of Hawaii at Manoa, Honolulu, HI, United States of America
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, HI, United States of America
| | - Herbert P. Schweizer
- Department of Molecular Genetics and Microbiology, College of Medicine and Emerging Pathogens Institute, University of Florida, Gainesville, Florida, United States of America
| | - Tung T. Hoang
- Department of Microbiology, University of Hawaii at Manoa, Honolulu, HI, United States of America
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, HI, United States of America
- * E-mail:
| |
Collapse
|
13
|
|
14
|
Mariappan V, Vellasamy KM, Vadivelu J. Host-Adaptation of Burkholderia pseudomallei Alters Metabolism and Virulence: a Global Proteome Analysis. Sci Rep 2017; 7:9015. [PMID: 28827633 PMCID: PMC5567125 DOI: 10.1038/s41598-017-09373-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 07/26/2017] [Indexed: 11/24/2022] Open
Abstract
Little is known about the evolution, adaptation and pathogenesis of Burkholderia pseudomallei within host during acute melioidosis infection. Melioidosis is a potential life threatening disease contracted through inhalation, ingestion, inoculation or direct entry of the organism into the blood stream via wounds or skin abrasions from contaminated soil and water. Environmental B. pseudomallei strain (BpMARAN), isolated during a melioidosis outbreak in Pahang, Malaysia was injected intra-peritoneally into a mouse and passaged strain was recovered from spleen (Bpmouse-adapted). A gel-based comparative proteomics profiling approach was used, to map and identify differentially expressed proteins (fold-change ≥ 2; p-value ≤ 0.05) between the strains. A total of 730 and 685 spots were visualised in the BpMARAN and Bpmouse-adapted strains, respectively. Of the 730 spots (BpMARAN as reference gel), 87 spots were differentially regulated (44 up- and 43 down-regulated). The identified proteins were classified as proteins related to metabolism, stress response, virulence, signal transduction, or adhesion. In comparison, it was found that those proteins related to adhesins, virulence factors and stress- response were up-regulated and could possibly explain the adaptation of the bacteria in the host. Investigating the differentially expressed proteins may provide better perspective of bacterial factors which aid survivability of B. pseudomallei in host.
Collapse
Affiliation(s)
- Vanitha Mariappan
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Kumutha Malar Vellasamy
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Jamuna Vadivelu
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia.
| |
Collapse
|
15
|
Relationships Between Resistance and Virulence in Burkholderia pseudomallei. CURRENT TROPICAL MEDICINE REPORTS 2017. [DOI: 10.1007/s40475-017-0119-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
16
|
Borlee GI, Plumley BA, Martin KH, Somprasong N, Mangalea MR, Islam MN, Burtnick MN, Brett PJ, Steinmetz I, AuCoin DP, Belisle JT, Crick DC, Schweizer HP, Borlee BR. Genome-scale analysis of the genes that contribute to Burkholderia pseudomallei biofilm formation identifies a crucial exopolysaccharide biosynthesis gene cluster. PLoS Negl Trop Dis 2017; 11:e0005689. [PMID: 28658258 PMCID: PMC5507470 DOI: 10.1371/journal.pntd.0005689] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 07/11/2017] [Accepted: 06/08/2017] [Indexed: 11/19/2022] Open
Abstract
Burkholderia pseudomallei, the causative agent of melioidosis, is an important public health threat due to limited therapeutic options for treatment. Efforts to improve therapeutics for B. pseudomallei infections are dependent on the need to understand the role of B. pseudomallei biofilm formation and its contribution to antibiotic tolerance and persistence as these are bacterial traits that prevent effective therapy. In order to reveal the genes that regulate and/or contribute to B. pseudomallei 1026b biofilm formation, we screened a sequence defined two-allele transposon library and identified 118 transposon insertion mutants that were deficient in biofilm formation. These mutants include transposon insertions in genes predicted to encode flagella, fimbriae, transcriptional regulators, polysaccharides, and hypothetical proteins. Polysaccharides are key constituents of biofilms and B. pseudomallei has the capacity to produce a diversity of polysaccharides, thus there is a critical need to link these biosynthetic genes with the polysaccharides they produce to better understand their biological role during infection. An allelic exchange deletion mutant of the entire B. pseudomallei biofilm-associated exopolysaccharide biosynthetic cluster was decreased in biofilm formation and produced a smooth colony morphology suggestive of the loss of exopolysaccharide production. Conversely, deletion of the previously defined capsule I polysaccharide biosynthesis gene cluster increased biofilm formation. Bioinformatics analyses combined with immunoblot analysis and glycosyl composition studies of the partially purified exopolysaccharide indicate that the biofilm-associated exopolysaccharide is neither cepacian nor the previously described acidic exopolysaccharide. The biofilm-associated exopolysaccharide described here is also specific to the B. pseudomallei complex of bacteria. Since this novel exopolysaccharide biosynthesis cluster is retained in B. mallei, it is predicted to have a role in colonization and infection of the host. These findings will facilitate further advances in understanding the pathogenesis of B. pseudomallei and improve diagnostics and therapeutic treatment strategies.
Collapse
Affiliation(s)
- Grace I. Borlee
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Brooke A. Plumley
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Kevin H. Martin
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Nawarat Somprasong
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida, United States of America
| | - Mihnea R. Mangalea
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - M. Nurul Islam
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Mary N. Burtnick
- Department of Microbiology and Immunology, University of South Alabama, Mobile, Alabama, United States of America
| | - Paul J. Brett
- Department of Microbiology and Immunology, University of South Alabama, Mobile, Alabama, United States of America
| | - Ivo Steinmetz
- Institute of Hygiene, Microbiology, and Environmental Medicine, Medical University of Graz, Graz, Austria
| | - David P. AuCoin
- Department of Molecular Microbiology and Immunology, University of Nevada-Reno, School of Medicine Reno, Nevada, United States of America
| | - John T. Belisle
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Dean C. Crick
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Herbert P. Schweizer
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida, United States of America
| | - Bradley R. Borlee
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| |
Collapse
|
17
|
Robbe-Saule M, Babonneau J, Sismeiro O, Marsollier L, Marion E. An Optimized Method for Extracting Bacterial RNA from Mouse Skin Tissue Colonized by Mycobacterium ulcerans. Front Microbiol 2017; 8:512. [PMID: 28392785 PMCID: PMC5364165 DOI: 10.3389/fmicb.2017.00512] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 03/13/2017] [Indexed: 11/13/2022] Open
Abstract
Bacterial transcriptome analyses during host colonization are essential to decipher the complexity of the relationship between the bacterium and its host. RNA sequencing (RNA-seq) is a promising approach providing valuable information about bacterial adaptation, the host response and, in some cases, mutual tolerance underlying crosstalk, as recently observed in the context of Mycobacterium ulcerans infection. Buruli ulcer is caused by M. ulcerans. This neglected disease is the third most common mycobacterial disease worldwide. Without treatment, M. ulcerans provokes massive skin ulcers. A healing process may be observed in 5% of Buruli ulcer patients several months after the initiation of disease. This spontaneous healing process suggests that some hosts can counteract the development of the lesions caused by M. ulcerans. Deciphering the mechanisms involved in this process should open up new treatment possibilities. To this end, we recently developed the first mouse model for studies of the spontaneous healing process. We have shown that the healing process is based on mutual tolerance between the bacterium and its host. In this context, RNA-seq seems to be the most appropriate method for deciphering bacterial adaptation. However, due to the low bacterial load in host tissues, the isolation of mycobacterial RNA from skin tissue for RNA-seq analysis remains challenging. We developed a method for extracting and purifying mycobacterial RNA whilst minimizing the amount of host RNA in the sample. This approach was based on the extraction of bacterial RNA by a differential lysis method. The challenge in the development of this method was the choice of a lysis system favoring the removal of host RNA without damage to the bacterial cells. We made use of the thick, resistant cell wall of M. ulcerans to achieve this end.
Collapse
Affiliation(s)
- Marie Robbe-Saule
- Center for Research in Cancerology and Immunology Nantes-Angers, Institut National de la Santé et de la Recherche Médicale, Université de Nantes, Université d'AngersAngers, France; Equipe Atip-Avenir, Center for Research in Cancerology and Immunology Nantes-Angers, Institut National de la Santé et de la Recherche Médicale, Centre Hospitalier Universitaire et Université d'AngersAngers, France
| | - Jérémie Babonneau
- Center for Research in Cancerology and Immunology Nantes-Angers, Institut National de la Santé et de la Recherche Médicale, Université de Nantes, Université d'AngersAngers, France; Equipe Atip-Avenir, Center for Research in Cancerology and Immunology Nantes-Angers, Institut National de la Santé et de la Recherche Médicale, Centre Hospitalier Universitaire et Université d'AngersAngers, France
| | - Odile Sismeiro
- Transcriptome and Epigenome Platform, Biomics, Center for Innovation and Technological Research, Institut Pasteur Paris, France
| | - Laurent Marsollier
- Center for Research in Cancerology and Immunology Nantes-Angers, Institut National de la Santé et de la Recherche Médicale, Université de Nantes, Université d'AngersAngers, France; Equipe Atip-Avenir, Center for Research in Cancerology and Immunology Nantes-Angers, Institut National de la Santé et de la Recherche Médicale, Centre Hospitalier Universitaire et Université d'AngersAngers, France
| | - Estelle Marion
- Center for Research in Cancerology and Immunology Nantes-Angers, Institut National de la Santé et de la Recherche Médicale, Université de Nantes, Université d'AngersAngers, France; Equipe Atip-Avenir, Center for Research in Cancerology and Immunology Nantes-Angers, Institut National de la Santé et de la Recherche Médicale, Centre Hospitalier Universitaire et Université d'AngersAngers, France
| |
Collapse
|
18
|
Eyedrop Inoculation Causes Sublethal Leptospirosis in Mice. Infect Immun 2017; 85:IAI.01050-16. [PMID: 28115508 DOI: 10.1128/iai.01050-16] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 01/18/2017] [Indexed: 01/17/2023] Open
Abstract
Leptospirosis is potentially a fatal zoonosis acquired by contact of skin and mucosal surfaces with soil and water contaminated with infected urine. We analyzed the outcome of infection of C3H/HeJ mice with Leptospira interrogans serovar Copenhageni using an enzootic mode of transmission, the conjunctival route. Infection led to weight loss and L. interrogans dissemination from blood to urine, and spirochetes were detected in blood and urine simultaneously. The infectious dose that led to consistent dissemination to kidney after conjunctival infection was ∼108 leptospires. Interestingly, a lower number of spirochetes appeared to colonize the kidney, given that we quantified ∼105 and ∼10 leptospires per μl of urine and per μg of kidney, respectively. Leptospira-specific IgM and IgG were detected at 15 days postinfection, and isotyping of the Ig subclass showed that the total IgG response switched from an IgG1 response to an IgG3 response after infection with L. interrogans Histological periodic acid-Schiff D staining of infected kidney showed interstitial nephritis, mononuclear cell infiltrates, and reduced size of glomeruli. Quantification of proinflammatory immunomediators in kidney showed that keratinocyte-derived chemokine, macrophage inflammatory protein 2, RANTES, tumor necrosis factor alpha, gamma interferon, and interleukin-10 were upregulated in infected mice. We show that the kinetics of disease progression after infection via the ocular conjunctiva is delayed compared with infection via the standard intraperitoneal route. Differences may be related to the number of L. interrogans spirochetes that succeed in overcoming the natural defenses of the ocular conjunctiva and transit through tissue.
Collapse
|
19
|
Chen YS, Lin HH, Hsueh PT, Ni WF, Liu PJ, Chen PS, Chang HH, Sun DS, Chen YL. Involvement of L-selectin expression in Burkholderia pseudomallei-infected monocytes invading the brain during murine melioidosis. Virulence 2016; 8:751-766. [PMID: 27646437 DOI: 10.1080/21505594.2016.1232239] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
The development of neurologic melioidosis was linked to the elicitation of Burkholderia pseudomallei-infected L-selectinhiCD11b+ BALB/c cells in our previous study. However, whether monocytic L-selectin (CD62L, encoded by the sell gene) is a key factor remains uncertain. In the present study, after establishing multi-organ foci via hematogenous routes, we demonstrated that B. pseudomallei GFP steadily persisted in blood, splenic, hepatic and bone marrow (BM) Ly6C monocytes; however, the circulating CD16/32+CD45hiGFP+ brain-infiltrating leukocytes (BILs) derived from the blood Ly6C monocytes were expanded in BALB/c but not in C57BL/6 bacteremic melioidosis. Consistent with these results, 60% of BALB/c mice but only 10% of C57BL/6 mice exhibited neurologic melioidosis. In a time-dependent manner, B. pseudomallei invaded C57BL/6 BM-derived phagocytes and monocytic progenitors by 2 d. The number of Ly6C+CD62L+GFP+ inflamed cells that had expanded in the BM and that were ready for emigration peaked on d 21 post-infection. Hematogenous B. pseudomallei-loaded sell+/+Ly6C monocytes exacerbated the bacterial loads and the proportion of Ly6C+GFP+ BILs in the recipient brains compared to sell-/- infected Ly6C cells when adoptively transferred. Moreover, a neutralizing anti-CD62L antibody significantly depleted the bacterial colonization of the brain following adoptive transfer of B. pseudomallei-loaded C57BL/6 or BALB/c Ly6C cells. Our data thus suggest that Ly6C+CD62L+ infected monocytes served as a Trojan horse across the cerebral endothelium to induce brain infection. Therefore, CD62L should be considered as not only a temporally elicited antigen but also a disease-relevant leukocyte marker during the development of neurologic melioidosis.
Collapse
Affiliation(s)
- Yao-Shen Chen
- a Department of Internal Medicine , Kaohsiung Veterans General Hospital , Kaohsiung , Taiwan.,b Department of Internal Medicine , National Yang-Ming University , Taipei , Taiwan
| | - Hsi-Hsun Lin
- c Section of Infectious Disease , Department of Medicine , E-Da Hospital and University , Kaohsiung Taiwan
| | - Pei-Tan Hsueh
- d Department of Biological Science , National Sun Yat-sen University , Kaohsiung , Taiwan.,e Department of Biotechnology , National Kaohsiung Normal University , Kaohsiung , Taiwan
| | - Wei-Fen Ni
- e Department of Biotechnology , National Kaohsiung Normal University , Kaohsiung , Taiwan
| | - Pei-Ju Liu
- e Department of Biotechnology , National Kaohsiung Normal University , Kaohsiung , Taiwan
| | - Pei-Shih Chen
- f Department of Public Health , College of Health Science, Kaohsiung Medical University , Kaohsiung , Taiwan.,g Institute of Environmental Engineering, National Sun Yat-sen University , Kaohsiung , Taiwan
| | - Hsin-Hou Chang
- h Department of Molecular Biology and Human Genetics , Tzu Chi University , Hualien , Taiwan
| | - Der-Shan Sun
- h Department of Molecular Biology and Human Genetics , Tzu Chi University , Hualien , Taiwan
| | - Ya-Lei Chen
- e Department of Biotechnology , National Kaohsiung Normal University , Kaohsiung , Taiwan
| |
Collapse
|
20
|
Abstract
Bacterial sphingomyelinases and phospholipases are a heterogeneous group of esterases which are usually surface associated or secreted by a wide variety of Gram-positive and Gram-negative bacteria. These enzymes hydrolyze sphingomyelin and glycerophospholipids, respectively, generating products identical to the ones produced by eukaryotic enzymes which play crucial roles in distinct physiological processes, including membrane dynamics, cellular signaling, migration, growth, and death. Several bacterial sphingomyelinases and phospholipases are essential for virulence of extracellular, facultative, or obligate intracellular pathogens, as these enzymes contribute to phagosomal escape or phagosomal maturation avoidance, favoring tissue colonization, infection establishment and progression, or immune response evasion. This work presents a classification proposal for bacterial sphingomyelinases and phospholipases that considers not only their enzymatic activities but also their structural aspects. An overview of the main physiopathological activities is provided for each enzyme type, as are examples in which inactivation of a sphingomyelinase- or a phospholipase-encoding gene impairs the virulence of a pathogen. The identification of sphingomyelinases and phospholipases important for bacterial pathogenesis and the development of inhibitors for these enzymes could generate candidate vaccines and therapeutic agents, which will diminish the impacts of the associated human and animal diseases.
Collapse
|
21
|
Lazar Adler NR, Allwood EM, Deveson Lucas D, Harrison P, Watts S, Dimitropoulos A, Treerat P, Alwis P, Devenish RJ, Prescott M, Govan B, Adler B, Harper M, Boyce JD. Perturbation of the two-component signal transduction system, BprRS, results in attenuated virulence and motility defects in Burkholderia pseudomallei. BMC Genomics 2016; 17:331. [PMID: 27147217 PMCID: PMC4855414 DOI: 10.1186/s12864-016-2668-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 04/26/2016] [Indexed: 02/08/2023] Open
Abstract
Background Burkholderia pseudomallei is the causative agent of melioidosis, a severe invasive disease of humans and animals. Initial screening of a B. pseudomallei signature-tagged mutagenesis library identified an attenuated mutant with a transposon insertion in a gene encoding the sensor component of an uncharacterised two-component signal transduction system (TCSTS), which we designated BprRS. Results Single gene inactivation of either the response regulator gene (bprR) or the sensor histidine kinase gene (bprS) resulted in mutants with reduced swarming motility and reduced virulence in mice. However, a bprRS double mutant was not attenuated for virulence and displayed wild-type levels of motility. The transcriptomes of the bprS, bprR and bprRS mutants were compared with the transcriptome of the parent strain K96243. Inactivation of the entire BprRS TCSTS (bprRS double mutant) resulted in altered expression of only nine genes, including both bprR and bprS, five phage-related genes and bpss0686, encoding a putative 5, 10-methylene tetrahydromethanopterin reductase involved in one carbon metabolism. In contrast, the transcriptomes of each of the bprR and bprS single gene mutants revealed more than 70 differentially expressed genes common to both mutants, including regulatory genes and those required for flagella assembly and for the biosynthesis of the cytotoxic polyketide, malleilactone. Conclusions Inactivation of the entire BprRS TCSTS did not alter virulence or motility and very few genes were differentially expressed indicating that the definitive BprRS regulon is relatively small. However, loss of a single component, either the sensor histidine kinase BprS or its cognate response regulator BprR, resulted in significant transcriptomic and phenotypic differences from the wild-type strain. We hypothesize that the dramatically altered phenotypes of these single mutants are the result of cross-regulation with one or more other TCSTSs and concomitant dysregulation of other key regulatory genes.
Collapse
Affiliation(s)
- Natalie R Lazar Adler
- Department of Microbiology, Monash University, 19 Innovation Walk, Clayton, Victoria, 3800, Australia.,Core Biotechnology Services, University of Leicester, Leicester, LE1 9HN, UK
| | - Elizabeth M Allwood
- Department of Microbiology, Monash University, 19 Innovation Walk, Clayton, Victoria, 3800, Australia
| | - Deanna Deveson Lucas
- Department of Microbiology, Monash University, 19 Innovation Walk, Clayton, Victoria, 3800, Australia.,Infection and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Victoria, Australia
| | - Paul Harrison
- Victorian Bioinformatics Platform, Monash University, Victoria, Australia
| | - Stephen Watts
- Department of Microbiology, Monash University, 19 Innovation Walk, Clayton, Victoria, 3800, Australia
| | - Alexandra Dimitropoulos
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Victoria, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Victoria, Australia
| | - Puthayalai Treerat
- Department of Microbiology, Monash University, 19 Innovation Walk, Clayton, Victoria, 3800, Australia.,Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Priyangi Alwis
- Department of Microbiology, Monash University, 19 Innovation Walk, Clayton, Victoria, 3800, Australia.,Faculty of Dentistry, National University of Singapore, Singapore, Singapore
| | - Rodney J Devenish
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Victoria, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Victoria, Australia
| | - Mark Prescott
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Victoria, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Victoria, Australia
| | - Brenda Govan
- Department of Microbiology and Immunology, James Cook University, Townsville, Queensland, Australia
| | - Ben Adler
- Department of Microbiology, Monash University, 19 Innovation Walk, Clayton, Victoria, 3800, Australia.,Infection and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Victoria, Australia
| | - Marina Harper
- Department of Microbiology, Monash University, 19 Innovation Walk, Clayton, Victoria, 3800, Australia.,Infection and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Victoria, Australia
| | - John D Boyce
- Department of Microbiology, Monash University, 19 Innovation Walk, Clayton, Victoria, 3800, Australia. .,Infection and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Victoria, Australia.
| |
Collapse
|
22
|
Gierok P, Kohler C, Steinmetz I, Lalk M. Burkholderia pseudomallei Colony Morphotypes Show a Synchronized Metabolic Pattern after Acute Infection. PLoS Negl Trop Dis 2016; 10:e0004483. [PMID: 26943908 PMCID: PMC4778764 DOI: 10.1371/journal.pntd.0004483] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Accepted: 02/02/2016] [Indexed: 11/18/2022] Open
Abstract
Background Burkholderia pseudomallei is a water and soil bacterium and the causative agent of melioidosis. A characteristic feature of this bacterium is the formation of different colony morphologies which can be isolated from environmental samples as well as from clinical samples, but can also be induced in vitro. Previous studies indicate that morphotypes can differ in a number of characteristics such as resistance to oxidative stress, cellular adhesion and intracellular replication. Yet the metabolic features of B. pseudomallei and its different morphotypes have not been examined in detail so far. Therefore, this study aimed to characterize the exometabolome of B. pseudomallei morphotypes and the impact of acute infection on their metabolic characteristics. Methods and Principal Findings We applied nuclear magnetic resonance spectroscopy (1H-NMR) in a metabolic footprint approach to compare nutrition uptake and metabolite secretion of starvation induced morphotypes of the B. pseudomallei strains K96243 and E8. We observed gluconate production and uptake in all morphotype cultures. Our study also revealed that among all morphotypes amino acids could be classified with regard to their fast and slow consumption. In addition to these shared metabolic features, the morphotypes varied highly in amino acid uptake profiles, secretion of branched chain amino acid metabolites and carbon utilization. After intracellular passage in vitro or murine acute infection in vivo, we observed a switch of the various morphotypes towards a single morphotype and a synchronization of nutrient uptake and metabolite secretion. Conclusion To our knowledge, this study provides first insights into the basic metabolism of B. pseudomallei and its colony morphotypes. Furthermore, our data suggest, that acute infection leads to the synchronization of B. pseudomallei colony morphology and metabolism through yet unknown host signals and bacterial mechanisms. Melioidosis is a common disease in Northern Australia and East Asia, with regional mortality rates of up to 40%. Clinical manifestations range from soft tissue infections to severe sepsis. It is caused by the Gram negative saprophytic water and soil bacterium Burkholderia pseudomallei, which forms a variety of colony morphologies on solid agar. Various morphotypes appear after the bacterium is exposed to physiological stress conditions or underwent the process of infection, yet the physiological function is unclear. Metabolism is closely linked to virulence in many pathogens, and since metabolic data are not available so far for this bacterium, we monitored the nutrition uptake and metabolite secretion of B. pseudomallei morphotypes. Interestingly, despite typical genes responsible for gluconate production are missing in the B. pseudomallei genome, we observed high amounts of gluconate in the extracellular space. Furthermore, we were able to investigate metabolic differences among the morphotypes and identified synchronization in morphology and metabolism after infection as an adaptation to the host environment.
Collapse
Affiliation(s)
- Philipp Gierok
- Institute of Biochemistry, Ernst-Moritz-Arndt-University, Greifswald, Germany
| | - Christian Kohler
- Friedrich Loeffler Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Ivo Steinmetz
- Friedrich Loeffler Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Michael Lalk
- Institute of Biochemistry, Ernst-Moritz-Arndt-University, Greifswald, Germany
- * E-mail:
| |
Collapse
|
23
|
Lau SKP, Lee KC, Lo GCS, Ding VSY, Chow WN, Ke TYH, Curreem SOT, To KKW, Ho DTY, Sridhar S, Wong SCY, Chan JFW, Hung IFN, Sze KH, Lam CW, Yuen KY, Woo PCY. Metabolomic Profiling of Plasma from Melioidosis Patients Using UHPLC-QTOF MS Reveals Novel Biomarkers for Diagnosis. Int J Mol Sci 2016; 17:307. [PMID: 26927094 PMCID: PMC4813170 DOI: 10.3390/ijms17030307] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 02/15/2016] [Accepted: 02/22/2016] [Indexed: 12/22/2022] Open
Abstract
To identify potential biomarkers for improving diagnosis of melioidosis, we compared plasma metabolome profiles of melioidosis patients compared to patients with other bacteremia and controls without active infection, using ultra-high-performance liquid chromatography-electrospray ionization-quadruple time-of-flight mass spectrometry. Principal component analysis (PCA) showed that the metabolomic profiles of melioidosis patients are distinguishable from bacteremia patients and controls. Using multivariate and univariate analysis, 12 significant metabolites from four lipid classes, acylcarnitine (n = 6), lysophosphatidylethanolamine (LysoPE) (n = 3), sphingomyelins (SM) (n = 2) and phosphatidylcholine (PC) (n = 1), with significantly higher levels in melioidosis patients than bacteremia patients and controls, were identified. Ten of the 12 metabolites showed area-under-receiver operating characteristic curve (AUC) >0.80 when compared both between melioidosis and bacteremia patients, and between melioidosis patients and controls. SM(d18:2/16:0) possessed the largest AUC when compared, both between melioidosis and bacteremia patients (AUC 0.998, sensitivity 100% and specificity 91.7%), and between melioidosis patients and controls (AUC 1.000, sensitivity 96.7% and specificity 100%). Our results indicate that metabolome profiling might serve as a promising approach for diagnosis of melioidosis using patient plasma, with SM(d18:2/16:0) representing a potential biomarker. Since the 12 metabolites were related to various pathways for energy and lipid metabolism, further studies may reveal their possible role in the pathogenesis and host response in melioidosis.
Collapse
Affiliation(s)
- Susanna K P Lau
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong, China.
- Research Centre of Infection and Immunology, The University of Hong Kong, Pokfulam, Hong Kong, China.
- Carol Yu Centre for Infection, The University of Hong Kong, Pokfulam, Hong Kong, China.
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Kim-Chung Lee
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - George C S Lo
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Vanessa S Y Ding
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Wang-Ngai Chow
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Tony Y H Ke
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Shirly O T Curreem
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Kelvin K W To
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong, China.
- Research Centre of Infection and Immunology, The University of Hong Kong, Pokfulam, Hong Kong, China.
- Carol Yu Centre for Infection, The University of Hong Kong, Pokfulam, Hong Kong, China.
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Deborah T Y Ho
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Siddharth Sridhar
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Sally C Y Wong
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Jasper F W Chan
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Ivan F N Hung
- Research Centre of Infection and Immunology, The University of Hong Kong, Pokfulam, Hong Kong, China.
- Department of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Kong-Hung Sze
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Ching-Wan Lam
- Department of Pathology, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Kwok-Yung Yuen
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong, China.
- Research Centre of Infection and Immunology, The University of Hong Kong, Pokfulam, Hong Kong, China.
- Carol Yu Centre for Infection, The University of Hong Kong, Pokfulam, Hong Kong, China.
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Patrick C Y Woo
- State Key Laboratory of Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong, China.
- Research Centre of Infection and Immunology, The University of Hong Kong, Pokfulam, Hong Kong, China.
- Carol Yu Centre for Infection, The University of Hong Kong, Pokfulam, Hong Kong, China.
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong, China.
| |
Collapse
|
24
|
Mobley HLT. Measuring Escherichia coli Gene Expression during Human Urinary Tract Infections. Pathogens 2016; 5:pathogens5010007. [PMID: 26784237 PMCID: PMC4810128 DOI: 10.3390/pathogens5010007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 01/13/2016] [Accepted: 01/13/2016] [Indexed: 11/25/2022] Open
Abstract
Extraintestinal Escherichiacoli (E. coli) evolved by acquisition of pathogenicity islands, phage, plasmids, and DNA segments by horizontal gene transfer. Strains are heterogeneous but virulent uropathogenic isolates more often have specific fimbriae, toxins, and iron receptors than commensal strains. One may ask whether it is the virulence factors alone that are required to establish infection. While these virulence factors clearly contribute strongly to pathogenesis, bacteria must survive by metabolizing nutrients available to them. By constructing mutants in all major metabolic pathways and co-challenging mice transurethrally with each mutant and the wild type strain, we identified which major metabolic pathways are required to infect the urinary tract. We must also ask what else is E. coli doing in vivo? To answer this question, we examined the transcriptome of E. coli CFT073 in the murine model of urinary tract infection (UTI) as well as for E. coli strains collected and analyzed directly from the urine of patients attending either a urology clinic or a university health clinic for symptoms of UTI. Using microarrays and RNA-seq, we measured in vivo gene expression for these uropathogenic E. coli strains, identifying genes upregulated during murine and human UTI. Our findings allow us to propose a new definition of bacterial virulence.
Collapse
Affiliation(s)
- Harry L T Mobley
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
25
|
Vander Broek CW, Chalmers KJ, Stevens MP, Stevens JM. Quantitative proteomic analysis of Burkholderia pseudomallei Bsa type III secretion system effectors using hypersecreting mutants. Mol Cell Proteomics 2015; 14:905-16. [PMID: 25635268 PMCID: PMC4390269 DOI: 10.1074/mcp.m114.044875] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 01/07/2015] [Indexed: 11/06/2022] Open
Abstract
Burkholderia pseudomallei is an intracellular pathogen and the causative agent of melioidosis, a severe disease of humans and animals. One of the virulence factors critical for early stages of infection is the Burkholderia secretion apparatus (Bsa) Type 3 Secretion System (T3SS), a molecular syringe that injects bacterial proteins, called effectors, into eukaryotic cells where they subvert cellular functions to the benefit of the bacteria. Although the Bsa T3SS itself is known to be important for invasion, intracellular replication, and virulence, only a few genuine effector proteins have been identified and the complete repertoire of proteins secreted by the system has not yet been fully characterized. We constructed a mutant lacking bsaP, a homolog of the T3SS "gatekeeper" family of proteins that exert control over the timing and magnitude of effector protein secretion. Mutants lacking BsaP, or the T3SS translocon protein BipD, were observed to hypersecrete the known Bsa effector protein BopE, providing evidence of their role in post-translational control of the Bsa T3SS and representing key reagents for the identification of its secreted substrates. Isobaric Tags for Relative and Absolute Quantification (iTRAQ), a gel-free quantitative proteomics technique, was used to compare the secreted protein profiles of the Bsa T3SS hypersecreting mutants of B. pseudomallei with the isogenic parent strain and a bsaZ mutant incapable of effector protein secretion. Our study provides one of the most comprehensive core secretomes of B. pseudomallei described to date and identified 26 putative Bsa-dependent secreted proteins that may be considered candidate effectors. Two of these proteins, BprD and BapA, were validated as novel effector proteins secreted by the Bsa T3SS of B. pseudomallei.
Collapse
Affiliation(s)
- Charles W Vander Broek
- From the ‡The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, Scotland, UK
| | - Kevin J Chalmers
- §Dundee Cell Products, James Lindsay Place, Dundee Technopole, Dundee, DD1 5JJ, Scotland, UK
| | - Mark P Stevens
- From the ‡The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, Scotland, UK
| | - Joanne M Stevens
- From the ‡The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, Scotland, UK.;
| |
Collapse
|
26
|
Challacombe JF, Stubben CJ, Klimko CP, Welkos SL, Kern SJ, Bozue JA, Worsham PL, Cote CK, Wolfe DN. Interrogation of the Burkholderia pseudomallei genome to address differential virulence among isolates. PLoS One 2014; 9:e115951. [PMID: 25536074 PMCID: PMC4275268 DOI: 10.1371/journal.pone.0115951] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 12/01/2014] [Indexed: 11/18/2022] Open
Abstract
Infection by the Gram-negative pathogen Burkholderia pseudomallei results in the disease melioidosis, acquired from the environment in parts of southeast Asia and northern Australia. Clinical symptoms of melioidosis range from acute (fever, pneumonia, septicemia, and localized infection) to chronic (abscesses in various organs and tissues, most commonly occurring in the lungs, liver, spleen, kidney, prostate and skeletal muscle), and persistent infections in humans are difficult to cure. Understanding the basic biology and genomics of B. pseudomallei is imperative for the development of new vaccines and therapeutic interventions. This formidable task is becoming more tractable due to the increasing number of B. pseudomallei genomes that are being sequenced and compared. Here, we compared three B. pseudomallei genomes, from strains MSHR668, K96243 and 1106a, to identify features that might explain why MSHR668 is more virulent than K96243 and 1106a in a mouse model of B. pseudomallei infection. Our analyses focused on metabolic, virulence and regulatory genes that were present in MSHR668 but absent from both K96243 and 1106a. We also noted features present in K96243 and 1106a but absent from MSHR668, and identified genomic differences that may contribute to variations in virulence noted among the three B. pseudomallei isolates. While this work contributes to our understanding of B. pseudomallei genomics, more detailed experiments are necessary to characterize the relevance of specific genomic features to B. pseudomallei metabolism and virulence. Functional analyses of metabolic networks, virulence and regulation shows promise for examining the effects of B. pseudomallei on host cell metabolism and will lay a foundation for future prediction of the virulence of emerging strains. Continued emphasis in this area will be critical for protection against melioidosis, as a better understanding of what constitutes a fully virulent Burkholderia isolate may provide for better diagnostic and medical countermeasure strategies.
Collapse
Affiliation(s)
- Jean F. Challacombe
- Los Alamos National Laboratory, Bioscience Division, Los Alamos, NM, United States of America
- * E-mail:
| | - Chris J. Stubben
- Los Alamos National Laboratory, Bioscience Division, Los Alamos, NM, United States of America
| | - Christopher P. Klimko
- US Army Medical Research Institute of Infectious Diseases, Bacteriology Division, Fort Detrick, MD, United States of America
| | - Susan L. Welkos
- US Army Medical Research Institute of Infectious Diseases, Bacteriology Division, Fort Detrick, MD, United States of America
| | - Steven J. Kern
- US Army Medical Research Institute of Infectious Diseases, Biostatistics Division, Fort Detrick, MD, United States of America
| | - Joel A. Bozue
- US Army Medical Research Institute of Infectious Diseases, Bacteriology Division, Fort Detrick, MD, United States of America
| | - Patricia L. Worsham
- US Army Medical Research Institute of Infectious Diseases, Bacteriology Division, Fort Detrick, MD, United States of America
| | - Christopher K. Cote
- US Army Medical Research Institute of Infectious Diseases, Bacteriology Division, Fort Detrick, MD, United States of America
| | - Daniel N. Wolfe
- Defense Threat Reduction Agency, Chemical and Biological Technologies Department, Fort Belvoir, VA, United States of America
| |
Collapse
|
27
|
The environment of "Mycobacterium avium subsp. hominissuis" microaggregates induces synthesis of small proteins associated with efficient infection of respiratory epithelial cells. Infect Immun 2014; 83:625-36. [PMID: 25422262 DOI: 10.1128/iai.02699-14] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
"Mycobacterium avium subsp. hominissuis" is an opportunistic environmental pathogen that causes respiratory illness in immunocompromised patients, such as those with cystic fibrosis as well as other chronic respiratory diseases. Currently, there is no efficient approach to prevent or treat M. avium subsp. hominissuis infection in the lungs. During initial colonization of the airways, M. avium subsp. hominissuis forms microaggregates composed of 3 to 20 bacteria on human respiratory epithelial cells, which provides an environment for phenotypic changes leading to efficient mucosal invasion in vitro and in vivo. DNA microarray analysis was employed to identify genes associated with the microaggregate phenotype. The gene encoding microaggregate-binding protein 1 (MBP-1) (MAV_3013) is highly expressed during microaggregate formation. When expressed in noninvasive Mycobacterium smegmatis, MBP-1 increased the ability of the bacteria to bind to HEp-2 epithelial cells. Using anti-MBP-1 immune serum, microaggregate binding to HEp-2 cells was significantly reduced. By far-Western blotting, and verified by coimmunoprecipitation, we observed that MBP-1 interacts with the host cytoskeletal protein vimentin. As visualized by confocal microscopy, microaggregates, as well as MBP-1, induced vimentin polymerization at the site of bacterium-host cell contact. Binding of microaggregates to HEp-2 cells was inhibited by treatment with an antivimentin antibody, suggesting that MBP-1 expression is important for M. avium subsp. hominissuis adherence to the host cell. MBP-1 immune serum significantly inhibited M. avium subsp. hominissuis infection throughout the respiratory tracts of mice. This study characterizes a pathogenic mechanism utilized by M. avium subsp. hominissuis to bind and invade the host respiratory epithelium, suggesting new potential targets for the development of antivirulence therapy.
Collapse
|
28
|
Stone JK, DeShazer D, Brett PJ, Burtnick MN. Melioidosis: molecular aspects of pathogenesis. Expert Rev Anti Infect Ther 2014; 12:1487-99. [PMID: 25312349 DOI: 10.1586/14787210.2014.970634] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Burkholderia pseudomallei is a gram-negative bacterium that causes melioidosis, a multifaceted disease that is highly endemic in southeast Asia and northern Australia. This facultative intracellular pathogen possesses a large genome that encodes a wide array of virulence factors that promote survival in vivo by manipulating host cell processes and disarming elements of the host immune system. Antigens and systems that play key roles in B. pseudomallei virulence include capsular polysaccharide, lipopolysaccharide, adhesins, specialized secretion systems, actin-based motility and various secreted factors. This review provides an overview of the current and steadily expanding knowledge regarding the molecular mechanisms used by this organism to survive within a host and their contribution to the pathogenesis of melioidosis.
Collapse
Affiliation(s)
- Joshua K Stone
- Department of Microbiology and Immunology, University of South Alabama, 610 Clinic Drive, Mobile, AL 36688, USA
| | | | | | | |
Collapse
|
29
|
Chirakul S, Bartpho T, Wongsurawat T, Taweechaisupapong S, Karoonutaisiri N, Talaat AM, Wongratanacheewin S, Ernst RK, Sermswan RW. Characterization of BPSS1521 (bprD), a regulator of Burkholderia pseudomallei virulence gene expression in the mouse model. PLoS One 2014; 9:e104313. [PMID: 25111708 PMCID: PMC4128674 DOI: 10.1371/journal.pone.0104313] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 07/07/2014] [Indexed: 11/19/2022] Open
Abstract
The Gram-negative saprophytic bacterium Burkholderia pseudomallei is the causative agent of melioidosis, a severe infectious disease of both humans and animals. Severity of the disease is thought to be dependent on both the health status of the host, including diabetes mellitus and kidney disease, and bacterial-derived factors. To identify the bacterial factors important during an acute infection, gene expression profiles in the spleen, lung, and liver of BALB/c (Th2 prototype) and C57BL/6 mice (Th1 prototype) were determined using DNA microarrays. This analysis identified BPSS1521 (bprD), a predicted transcriptional regulator located in the type III secretion system (T3SS-3) operon, to be up regulated, specifically in C57BL/6 mice. BALB/c mice infected with a bprD mutant showed a shorter time to death and increased inflammation, as determined by histopathological analysis and enumeration of bacteria in the spleen. Elevated numbers of multinucleated giant cells (MNGCs), which is the hallmark of melioidosis, were detected in both the wild-type and the bprD mutants; a similar elevation occurs in melioidosis patients. One striking observation was the increased expression of BPSS1520 (bprC), located downstream of bprD, in the bprD mutant. BprC is a regulator of T6SS-1 that is required for the virulence of B. pseudomallei in murine infection models. Deletion of bprD led to the overexpression of bprC and a decreased time to death. bprD expression was elevated in C57BL/6--as compared to BALB/c--mice, suggesting a role for BprD in the natural resistance of C57BL/6 mice to B. pseudomallei. Ultimately, this analysis using mice with different immune backgrounds may enhance our understanding of the outcomes of infection in a variety of models.
Collapse
Affiliation(s)
- Sunisa Chirakul
- Melioidosis Research Center, Khon Kaen University, Khon Kaen, Thailand
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | | | - Thidathip Wongsurawat
- National Center for Genetic Engineering and Biotechnology (BIOTEC), Pathumthani, Thailand
| | | | - Nitsara Karoonutaisiri
- National Center for Genetic Engineering and Biotechnology (BIOTEC), Pathumthani, Thailand
| | - Adel M. Talaat
- Department of Pathobiology, SVM, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Surasakdi Wongratanacheewin
- Melioidosis Research Center, Khon Kaen University, Khon Kaen, Thailand
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Robert K. Ernst
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland-Baltimore, Baltimore, Maryland, United States of America
| | - Rasana W. Sermswan
- Melioidosis Research Center, Khon Kaen University, Khon Kaen, Thailand
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
30
|
Memišević V, Zavaljevski N, Pieper R, Rajagopala SV, Kwon K, Townsend K, Yu C, Yu X, DeShazer D, Reifman J, Wallqvist A. Novel Burkholderia mallei virulence factors linked to specific host-pathogen protein interactions. Mol Cell Proteomics 2013; 12:3036-51. [PMID: 23800426 PMCID: PMC3820922 DOI: 10.1074/mcp.m113.029041] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 06/10/2013] [Indexed: 11/09/2022] Open
Abstract
Burkholderia mallei is an infectious intracellular pathogen whose virulence and resistance to antibiotics makes it a potential bioterrorism agent. Given its genetic origin as a commensal soil organism, it is equipped with an extensive and varied set of adapted mechanisms to cope with and modulate host-cell environments. One essential virulence mechanism constitutes the specialized secretion systems that are designed to penetrate host-cell membranes and insert pathogen proteins directly into the host cell's cytosol. However, the secretion systems' proteins and, in particular, their host targets are largely uncharacterized. Here, we used a combined in silico, in vitro, and in vivo approach to identify B. mallei proteins required for pathogenicity. We used bioinformatics tools, including orthology detection and ab initio predictions of secretion system proteins, as well as published experimental Burkholderia data to initially select a small number of proteins as putative virulence factors. We then used yeast two-hybrid assays against normalized whole human and whole murine proteome libraries to detect and identify interactions among each of these bacterial proteins and host proteins. Analysis of such interactions provided both verification of known virulence factors and identification of three new putative virulence proteins. We successfully created insertion mutants for each of these three proteins using the virulent B. mallei ATCC 23344 strain. We exposed BALB/c mice to mutant strains and the wild-type strain in an aerosol challenge model using lethal B. mallei doses. In each set of experiments, mice exposed to mutant strains survived for the 21-day duration of the experiment, whereas mice exposed to the wild-type strain rapidly died. Given their in vivo role in pathogenicity, and based on the yeast two-hybrid interaction data, these results point to the importance of these pathogen proteins in modulating host ubiquitination pathways, phagosomal escape, and actin-cytoskeleton rearrangement processes.
Collapse
Affiliation(s)
- Vesna Memišević
- From the ‡Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Materiel Command, Fort Detrick, Maryland 21702
| | - Nela Zavaljevski
- From the ‡Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Materiel Command, Fort Detrick, Maryland 21702
| | | | | | - Keehwan Kwon
- §J. Craig Venter Institute, Rockville, Maryland 20850
| | | | - Chenggang Yu
- From the ‡Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Materiel Command, Fort Detrick, Maryland 21702
| | - Xueping Yu
- From the ‡Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Materiel Command, Fort Detrick, Maryland 21702
| | - David DeShazer
- ¶Bacteriology Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland 21702
| | - Jaques Reifman
- From the ‡Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Materiel Command, Fort Detrick, Maryland 21702
| | - Anders Wallqvist
- From the ‡Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Materiel Command, Fort Detrick, Maryland 21702
| |
Collapse
|
31
|
Chieng S, Carreto L, Nathan S. Burkholderia pseudomallei transcriptional adaptation in macrophages. BMC Genomics 2012; 13:328. [PMID: 22823543 PMCID: PMC3418162 DOI: 10.1186/1471-2164-13-328] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Accepted: 07/23/2012] [Indexed: 11/10/2022] Open
Abstract
Background Burkholderia pseudomallei is a facultative intracellular pathogen of phagocytic and non-phagocytic cells. How the bacterium interacts with host macrophage cells is still not well understood and is critical to appreciate the strategies used by this bacterium to survive and how intracellular survival leads to disease manifestation. Results Here we report the expression profile of intracellular B. pseudomallei following infection of human macrophage-like U937 cells. During intracellular growth over the 6 h infection period, approximately 22 % of the B. pseudomallei genome showed significant transcriptional adaptation. B. pseudomallei adapted rapidly to the intracellular environment by down-regulating numerous genes involved in metabolism, cell envelope, motility, replication, amino acid and ion transport system and regulatory function pathways. Reduced expression in catabolic and housekeeping genes suggested lower energy requirement and growth arrest during macrophage infection, while expression of genes encoding anaerobic metabolism functions were up regulated. However, whilst the type VI secretion system was up regulated, expression of many known virulence factors was not significantly modulated over the 6hours of infection. Conclusions The transcriptome profile described here provides the first comprehensive view of how B. pseudomallei survives within host cells and will help identify potential virulence factors and proteins that are important for the survival and growth of B. pseudomallei within human cells.
Collapse
Affiliation(s)
- Sylvia Chieng
- School of Biosciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi, 43600, Malaysia
| | | | | |
Collapse
|
32
|
O'Grady EP, Sokol PA. Burkholderia cenocepacia differential gene expression during host-pathogen interactions and adaptation to the host environment. Front Cell Infect Microbiol 2011; 1:15. [PMID: 22919581 PMCID: PMC3417382 DOI: 10.3389/fcimb.2011.00015] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Accepted: 11/20/2011] [Indexed: 01/08/2023] Open
Abstract
Members of the Burkholderia cepacia complex (Bcc) are important in medical, biotechnological, and agricultural disciplines. These bacteria naturally occur in soil and water environments and have adapted to survive in association with plants and animals including humans. All Bcc species are opportunistic pathogens including Burkholderia cenocepacia that causes infections in cystic fibrosis and chronic granulomatous disease patients. The adaptation of B. cenocepacia to the host environment was assessed in a rat chronic respiratory infection model and compared to that of high cell-density in vitro grown cultures using transcriptomics. The distribution of genes differentially expressed on chromosomes 1, 2, and 3 was relatively proportional to the size of each genomic element, whereas the proportion of plasmid-encoded genes differentially expressed was much higher relative to its size and most genes were induced in vivo. The majority of genes encoding known virulence factors, components of types II and III secretion systems and chromosome 2-encoded type IV secretion system were similarly expressed between in vitro and in vivo environments. Lower expression in vivo was detected for genes encoding N-acyl-homoserine lactone synthase CepI, orphan LuxR homolog CepR2, zinc metalloproteases ZmpA and ZmpB, LysR-type transcriptional regulator ShvR, nematocidal protein AidA, and genes associated with flagellar motility, Flp type pilus formation, and type VI secretion. Plasmid-encoded type IV secretion genes were markedly induced in vivo. Additional genes induced in vivo included genes predicted to be involved in osmotic stress adaptation or intracellular survival, metal ion, and nutrient transport, as well as those encoding outer membrane proteins. Genes identified in this study are potentially important for virulence during host–pathogen interactions and may be associated with survival and adaptation to the host environment during chronic lung infections.
Collapse
Affiliation(s)
- Eoin P O'Grady
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, Canada
| | | |
Collapse
|
33
|
Conejero L, Patel N, de Reynal M, Oberdorf S, Prior J, Felgner PL, Titball RW, Salguero FJ, Bancroft GJ. Low-dose exposure of C57BL/6 mice to burkholderia pseudomallei mimics chronic human melioidosis. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:270-80. [PMID: 21703409 DOI: 10.1016/j.ajpath.2011.03.031] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2010] [Revised: 03/23/2011] [Accepted: 03/29/2011] [Indexed: 12/15/2022]
Abstract
Burkholderia pseudomallei is the etiological agent of human melioidosis, a disease with a broad spectrum of clinical manifestations ranging from fatal septicemia to chronic localized infection or asymptomatic latent infection. Most clinical and immunological studies to date have focused on the acute disease process; however, little is known about pathology and immune response in chronic melioidosis. Here, we have developed a murine model of chronic disease by challenging C57BL/6 mice intranasally with a low dose of B. pseudomallei and monitoring them up to 100 days postinfection. Bacterial burdens were heterogeneous in different animals at all time points, consistent with the spectrum of clinical severity observed in humans. Proinflammatory cytokines such as gamma interferon (IFN-γ), interleukin-6 (IL-6), monocyte chemotactic protein-1 (MCP-1), and tumor necrosis factor-α (TNF-α) were induced during chronic infection, and histopathological analysis showed features in common with human melioidosis. Interestingly, many of these features were similar to those induced by Mycobacterium tuberculosis in humans, such as development of a collagen cord that encapsulates the lesions, the presence of multinucleated giant cells, and granulomas with a caseous necrotic center, which may explain why chronic melioidosis is often misdiagnosed as tuberculosis. Our model now provides a relevant and practical tool to define the immunological features of chronic melioidosis and aid in the development of more effective treatment of this disease in humans.
Collapse
Affiliation(s)
- Laura Conejero
- Department of Immunology and Infection, Faculty of Infectious & Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Genome-wide analysis reveals loci encoding anti-macrophage factors in the human pathogen Burkholderia pseudomallei K96243. PLoS One 2010; 5:e15693. [PMID: 21203527 PMCID: PMC3008741 DOI: 10.1371/journal.pone.0015693] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Accepted: 11/22/2010] [Indexed: 11/19/2022] Open
Abstract
Burkholderia pseudomallei is an important human pathogen whose infection biology is still poorly understood. The bacterium is endemic to tropical regions, including South East Asia and Northern Australia, where it causes melioidosis, a serious disease associated with both high mortality and antibiotic resistance. B. pseudomallei is a Gram-negative facultative intracellular pathogen that is able to replicate in macrophages. However despite the critical nature of its interaction with macrophages, few anti-macrophage factors have been characterized to date. Here we perform a genome-wide gain of function screen of B. pseudomallei strain K96243 to identify loci encoding factors with anti-macrophage activity. We identify a total of 113 such loci scattered across both chromosomes, with positive gene clusters encoding transporters and secretion systems, enzymes/toxins, secondary metabolite, biofilm, adhesion and signal response related factors. Further phenotypic analysis of four of these regions shows that the encoded factors cause striking cellular phenotypes relevant to infection biology, including apoptosis, formation of actin 'tails' and multi-nucleation within treated macrophages. The detailed analysis of the remaining host of loci will facilitate genetic dissection of the interaction of this important pathogen with host macrophages and thus further elucidate this critical part of its infection cycle.
Collapse
|
35
|
Galyov EE, Brett PJ, DeShazer D. Molecular insights into Burkholderia pseudomallei and Burkholderia mallei pathogenesis. Annu Rev Microbiol 2010; 64:495-517. [PMID: 20528691 DOI: 10.1146/annurev.micro.112408.134030] [Citation(s) in RCA: 196] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Burkholderia pseudomallei and Burkholderia mallei are closely related gram-negative bacteria that can cause serious diseases in humans and animals. This review summarizes the current and rapidly expanding knowledge on the specific virulence factors employed by these pathogens and their roles in the pathogenesis of melioidosis and glanders. In particular, the contributions of recently identified virulence factors are described in the context of the intracellular lifestyle of these pathogens. Throughout this review, unique and shared virulence features of B. pseudomallei and B. mallei are discussed.
Collapse
Affiliation(s)
- Edouard E Galyov
- Department of Infection, Immunity and Inflammation, MSB, University of Leicester, Leicester LE1 9HN, United Kingdom.
| | | | | |
Collapse
|
36
|
Reckseidler-Zenteno SL, Viteri DF, Moore R, Wong E, Tuanyok A, Woods DE. Characterization of the type III capsular polysaccharide produced by Burkholderia pseudomallei. J Med Microbiol 2010; 59:1403-1414. [DOI: 10.1099/jmm.0.022202-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Burkholderia pseudomallei has been shown to produce more than one capsular polysaccharide (CPS). Analysis of the B. pseudomallei genome has revealed that the organism contains four CPS operons (I–IV). One of these operons (CPS III) was selected for further study. Comparative sequencing analysis revealed that the genes encoding CPS III are present in B. pseudomallei and Burkholderia thailandensis but not in Burkholderia mallei. In this study, CPS III was not found to contribute to the virulence of B. pseudomallei. Strains containing mutations in CPS III had the same LD50 value as the wild-type when tested in an animal infection model. Production of CPS III was shown to be induced in water but inhibited in 30 % normal human serum using a lux reporter fusion assay. Microarray analysis of capsule gene expression in infected hamsters revealed that the genes encoding CPS III were not significantly expressed in vivo compared with the genes encoding the previously characterized mannoheptose capsule (CPS I), which is an important virulence factor in B. pseudomallei. Glycosyl-composition analysis by combined GC/MS indicated that the CPS III genes are involved in the synthesis of a capsule composed of galactose, glucose, mannose and xylose.
Collapse
Affiliation(s)
- Shauna L. Reckseidler-Zenteno
- Centre for Science, Athabasca University, Athabasca, AB T9S 9Z9, Canada
- Department of Microbiology and Infectious Diseases, University of Calgary Health Sciences Center, Calgary, AB T2N 4N1, Canada
| | - Duber-Frey Viteri
- Department of Microbiology and Infectious Diseases, University of Calgary Health Sciences Center, Calgary, AB T2N 4N1, Canada
| | - Richard Moore
- Department of Microbiology and Infectious Diseases, University of Calgary Health Sciences Center, Calgary, AB T2N 4N1, Canada
| | - Erica Wong
- Department of Microbiology and Infectious Diseases, University of Calgary Health Sciences Center, Calgary, AB T2N 4N1, Canada
| | - Apichai Tuanyok
- Department of Microbiology and Infectious Diseases, University of Calgary Health Sciences Center, Calgary, AB T2N 4N1, Canada
| | - Donald E. Woods
- Department of Microbiology and Infectious Diseases, University of Calgary Health Sciences Center, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
37
|
Rossetti CA, Galindo CL, Garner HR, Adams LG. Selective amplification of Brucella melitensis mRNA from a mixed host-pathogen total RNA. BMC Res Notes 2010; 3:244. [PMID: 20920187 PMCID: PMC2954846 DOI: 10.1186/1756-0500-3-244] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2010] [Accepted: 09/28/2010] [Indexed: 11/26/2022] Open
Abstract
Background Brucellosis is a worldwide anthropozoonotic disease caused by an in vivo intracellular pathogen belonging to genus Brucella. The characterization of brucelae transcriptome's during host-pathogen interaction has been limited due to the difficulty of obtaining an adequate quantity of good quality eukaryotic RNA-free pathogen RNA for downstream applications. Findings Here, we describe a combined protocol to prepare RNA from intracellular B. melitensis in a quantity and quality suitable for pathogen gene expression analysis. Initially, B. melitensis total RNA was enriched from a host:pathogen mixed RNA sample by reducing the eukaryotic RNA..Then, to increase the Brucella RNA concentration and simultaneously minimize the contaminated host RNA in the mixed sample, a specific primer set designed to anneal to all B. melitensis ORF allows the selective linear amplification of sense-strand prokaryotic transcripts in a previously enriched RNA sample. Conclusion The novelty of the method we present here allows analysis of the gene expression profile of B. melitensis when limited amounts of pathogen RNA are present, and is potentially applicable to both in vivo and in vitro models of infection, even at early infection time points.
Collapse
Affiliation(s)
- Carlos A Rossetti
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, TX, USA.
| | | | | | | |
Collapse
|
38
|
A Burkholderia pseudomallei deltapurM mutant is avirulent in immunocompetent and immunodeficient animals: candidate strain for exclusion from select-agent lists. Infect Immun 2010; 78:3136-43. [PMID: 20404077 DOI: 10.1128/iai.01313-09] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Burkholderia pseudomallei causes the disease melioidosis in humans and is classified as a category B select agent. Research utilizing this pathogen is highly regulated in the United States, and even basic studies must be conducted in biosafety level 3 (BSL-3) facilities. There is currently no attenuated B. pseudomallei strain available that is excluded from select-agent regulations and can be safely handled at BSL-2 facilities. To address this need, we created Bp82 and Bp190, which are DeltapurM derivatives of B. pseudomallei strains 1026b and K96243 that are deficient in adenine and thiamine biosynthesis but replication competent in vitro in rich medium. A series of animal challenge studies was conducted to ensure that these strains were fully attenuated. Whereas the parental strains 1026b and K96243 and the complemented mutants Bp410 and Bp454 were virulent in BALB/c mice following intranasal inoculation, the DeltapurM mutants Bp82 and Bp190 were avirulent even when they were administered at doses 4 logs higher than the doses used for the parental strains. Animals challenged with high doses of the DeltapurM mutants rapidly cleared the bacterium from tissues (lung, liver, and spleen) and remained free of culturable bacteria for the duration of the experiments (up to 60 days postinfection). Moreover, highly susceptible 129/SvEv mice and immune incompetent mice (IFN-gamma-/-, SCID) were resistant to challenges with DeltapurM mutant Bp82. This strain was also avirulent in the Syrian hamster challenge model. We concluded that DeltapurM mutant Bp82 is fully attenuated and safe for use under BSL-2 laboratory conditions and thus is a candidate for exclusion from the select-agent list.
Collapse
|
39
|
Leroy Q, Raoult D. Review of microarray studies for host-intracellular pathogen interactions. J Microbiol Methods 2010; 81:81-95. [PMID: 20188126 DOI: 10.1016/j.mimet.2010.02.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2009] [Revised: 02/12/2010] [Accepted: 02/16/2010] [Indexed: 12/17/2022]
Abstract
Obligate intracellular bacteria are privileged soldiers on the battlefield that represent host-pathogen interactions. Microarrays are a powerful technology that can increase our knowledge about how bacteria respond to and interact with their hosts. This review summarizes the limitations inherent to host-pathogen interaction studies and essential strategies to improve microarray investigations of intracellular bacteria. We have compiled the comparative genomic and gene expression analyses of obligate intracellular bacteria currently available from microarrays. In this review we explore ways in which microarrays can be used to identify polymorphisms in different obligate intracellular bacteria such as Coxiella burnetii, Chlamydia trachomatis, Ehrlichia chaffeensis, Rickettsia prowazekii and Tropheryma whipplei. These microarray studies reveal that, while genomic content is highly conserved in obligate intracellular bacteria, genetic polymorphisms can potentially occur to increase bacterial pathogenesis. Additionally, changes in the gene expression of C. trachomatis throughout its life cycle, as well as changes in the gene expression profile of the pathogens R. prowazekii, Rickettsia rickettsii, Rickettsia typhi, T. whipplei and C. trachomatis following environmental changes, are discussed. Finally, an in vivo model of Rickettsia conorii within the skin is discussed. The gene expression analyses highlight the capacity of obligate intracellular bacteria to adapt to environmental changes and potentially to thwart the host response.
Collapse
Affiliation(s)
- Quentin Leroy
- Université de la Méditerranée, URMITE IRD-CNRS 6236, Faculté de Médecine, 27 Bd Jean Moulin, 13385 Marseille cedex 05, France
| | | |
Collapse
|
40
|
Titball RW, Russell P, Cuccui J, Easton A, Haque A, Atkins T, Sarkar-Tyson M, Harley V, Wren B, Bancroft GJ. Burkholderia pseudomallei: animal models of infection. Trans R Soc Trop Med Hyg 2009; 102 Suppl 1:S111-6. [PMID: 19121670 DOI: 10.1016/s0035-9203(08)70026-9] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
A range of animal models of Burkholderia pseudomallei infection have been reported, and the host species differ widely both in their susceptibility to infection and in the pathogenesis of disease. In mice, and depending on the route of infection, dose, and mouse strain, the disease can range from a chronic, and in some cases, an apparently latent infection to an acute fulminant disease. Alternative small animal models of infection include diabetic rats or hamsters. Larger animal models of disease have not yet been fully developed. It is not clear which of the small animal models of melioidosis most accurately reflect disease in humans. However, the findings that diabetic rats are susceptible to infection, that some strains of mice can develop persistent subclinical infections that can spontaneously reactivate, and that inhalation exposure generally results in more acute disease suggest that these different models mimic different aspects of human melioidosis.
Collapse
Affiliation(s)
- Richard W Titball
- School of Biosciences, Geoffrey Pope Building, University of Exeter, Exeter EX4 4QD, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Lazar Adler NR, Govan B, Cullinane M, Harper M, Adler B, Boyce JD. The molecular and cellular basis of pathogenesis in melioidosis: how does Burkholderia pseudomallei cause disease? FEMS Microbiol Rev 2009; 33:1079-99. [PMID: 19732156 DOI: 10.1111/j.1574-6976.2009.00189.x] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Melioidosis, a febrile illness with disease states ranging from acute pneumonia or septicaemia to chronic abscesses, was first documented by Whitmore & Krishnaswami (1912). The causative agent, Burkholderia pseudomallei, was subsequently identified as a motile, gram-negative bacillus, which is principally an environmental saprophyte. Melioidosis has become an increasingly important disease in endemic areas such as northern Thailand and Australia (Currie et al., 2000). This health burden, plus the classification of B. pseudomallei as a category B biological agent (Rotz et al., 2002), has resulted in an escalation of research interest. This review focuses on the molecular and cellular basis of pathogenesis in melioidosis, with a comprehensive overview of the current knowledge on how B. pseudomallei can cause disease. The process of B. pseudomallei movement from the environmental reservoir to attachment and invasion of epithelial and macrophage cells and the subsequent intracellular survival and spread is outlined. Furthermore, the diverse assortment of virulence factors that allow B. pseudomallei to become an effective opportunistic pathogen, as well as to avoid or subvert the host immune response, is discussed. With the recent increase in genomic and molecular studies, the current understanding of the infection process of melioidosis has increased substantially, yet, much still remains to be elucidated.
Collapse
|
42
|
A Burkholderia pseudomallei protein microarray reveals serodiagnostic and cross-reactive antigens. Proc Natl Acad Sci U S A 2009; 106:13499-504. [PMID: 19666533 DOI: 10.1073/pnas.0812080106] [Citation(s) in RCA: 144] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Understanding the way in which the immune system responds to infection is central to the development of vaccines and many diagnostics. To provide insight into this area, we fabricated a protein microarray containing 1,205 Burkholderia pseudomallei proteins, probed it with 88 melioidosis patient sera, and identified 170 reactive antigens. This subset of antigens was printed on a smaller array and probed with a collection of 747 individual sera derived from 10 patient groups including melioidosis patients from Northeast Thailand and Singapore, patients with different infections, healthy individuals from the USA, and from endemic and nonendemic regions of Thailand. We identified 49 antigens that are significantly more reactive in melioidosis patients than healthy people and patients with other types of bacterial infections. We also identified 59 cross-reactive antigens that are equally reactive among all groups, including healthy controls from the USA. Using these results we were able to devise a test that can classify melioidosis positive and negative individuals with sensitivity and specificity of 95% and 83%, respectively, a significant improvement over currently available diagnostic assays. Half of the reactive antigens contained a predicted signal peptide sequence and were classified as outer membrane, surface structures or secreted molecules, and an additional 20% were associated with pathogenicity, adaptation or chaperones. These results show that microarrays allow a more comprehensive analysis of the immune response on an antigen-specific, patient-specific, and population-specific basis, can identify serodiagnostic antigens, and contribute to a more detailed understanding of immunogenicity to this pathogen.
Collapse
|
43
|
Vellasamy KM, Vasu C, Puthucheary SD, Vadivelu J. Comparative analysis of extracellular enzymes and virulence exhibited by Burkholderia pseudomallei from different sources. Microb Pathog 2009; 47:111-7. [PMID: 19524661 DOI: 10.1016/j.micpath.2009.06.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2007] [Revised: 06/02/2009] [Accepted: 06/04/2009] [Indexed: 10/20/2022]
Abstract
To evaluate the potential role of extracellular proteins in the pathogenicity and virulence of Burkholderia pseudomallei, the activities of several enzymes in the culture filtrates of nine clinical and six environmental isolates were investigated in vitro and in vivo in ICR strain of mice. The production of protease, phosphatase, phospholipase C, superoxide dismutase, catalase and peroxidase were detected in the culture filtrates of all the 15 isolates at different time points of growth 4-24h. Over time, activity of each enzyme at each time point varied. Profile of secretion was similar among the 15 isolates irrespective of source, that is clinical or environmental. Catalase, phosphatase and phospholipase C were found to be increased in 60-100% of the isolates post-passage in mice. In vivo inoculation studies in ICR mice demonstrated a wide difference in their ability to cause bacteraemia, splenic or external abscesses and mortality rate ranged from few days to several weeks.
Collapse
|
44
|
Tuanyok A, Leadem BR, Auerbach RK, Beckstrom-Sternberg SM, Beckstrom-Sternberg JS, Mayo M, Wuthiekanun V, Brettin TS, Nierman WC, Peacock SJ, Currie BJ, Wagner DM, Keim P. Genomic islands from five strains of Burkholderia pseudomallei. BMC Genomics 2008; 9:566. [PMID: 19038032 PMCID: PMC2612704 DOI: 10.1186/1471-2164-9-566] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2008] [Accepted: 11/27/2008] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Burkholderia pseudomallei is the etiologic agent of melioidosis, a significant cause of morbidity and mortality where this infection is endemic. Genomic differences among strains of B. pseudomallei are predicted to be one of the major causes of the diverse clinical manifestations observed among patients with melioidosis. The purpose of this study was to examine the role of genomic islands (GIs) as sources of genomic diversity in this species. RESULTS We found that genomic islands (GIs) vary greatly among B. pseudomallei strains. We identified 71 distinct GIs from the genome sequences of five reference strains of B. pseudomallei: K96243, 1710b, 1106a, MSHR668, and MSHR305. The genomic positions of these GIs are not random, as many of them are associated with tRNA gene loci. In particular, the 3' end sequences of tRNA genes are predicted to be involved in the integration of GIs. We propose the term "tRNA-mediated site-specific recombination" (tRNA-SSR) for this mechanism. In addition, we provide a GI nomenclature that is based upon integration hotspots identified here or previously described. CONCLUSION Our data suggest that acquisition of GIs is one of the major sources of genomic diversity within B. pseudomallei and the molecular mechanisms that facilitate horizontally-acquired GIs are common across multiple strains of B. pseudomallei. The differential presence of the 71 GIs across multiple strains demonstrates the importance of these mobile elements for shaping the genetic composition of individual strains and populations within this bacterial species.
Collapse
Affiliation(s)
- Apichai Tuanyok
- Department of Biological Sciences, Northern Arizona University, Flagstaff, AZ 86011-5640, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Su YC, Wan KL, Mohamed R, Nathan S. A genome level survey of Burkholderia pseudomallei immunome expressed during human infection. Microbes Infect 2008; 10:1335-45. [PMID: 18761419 DOI: 10.1016/j.micinf.2008.07.034] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2008] [Revised: 07/22/2008] [Accepted: 07/30/2008] [Indexed: 11/26/2022]
Abstract
Burkholderia pseudomallei is the etiological agent of melioidosis, a severe infectious disease of humans and animals. The role of the bacterium's proteins expressed in vivo during human melioidosis continues to remain an enigma. This study's aim was to identify B. pseudomallei target proteins that elicit the humoral immune response in infected humans. A small insert genomic expression library was constructed and immunoscreened to identify peptides that reacted exclusively with melioidosis patients' sera. Sero-positive clones expressing immunogenic peptides were sequenced and annotated, and shown to represent 109 proteins involved in bacterial cell envelope biogenesis, cell motility and secretion, transcription, amino acid, ion and protein metabolism, energy production, DNA repair and unknown hypothetical proteins. Western blot analysis of three randomly selected full-length immunogenic polypeptides with patients' sera verified the findings of the immunome screening. The patients' humoral immune response to the 109 proteins suggests the induction or significant upregulation of these proteins in vivo during human infection and thus may play a role in the pathogenesis of B. pseudomallei. Identification of B. pseudomallei immunogens has shed new light on the elucidation of the bacterium's pathogenesis mechanism and disease severity. These immunogens can be further evaluated as prophylactic and serodiagnostic candidates as well as drug targets.
Collapse
Affiliation(s)
- Yu-Ching Su
- School of Biosciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, 43600 UKM Bangi, Selangor Darul Ehsan, Malaysia
| | | | | | | |
Collapse
|
46
|
van Schaik E, Tom M, DeVinney R, Woods DE. Development of novel animal infection models for the study of acute and chronic Burkholderia pseudomallei pulmonary infections. Microbes Infect 2008; 10:1291-9. [PMID: 18707015 DOI: 10.1016/j.micinf.2008.07.028] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2008] [Revised: 07/07/2008] [Accepted: 07/18/2008] [Indexed: 11/27/2022]
Abstract
Burkholderia pseudomallei causes the disease melioidosis. The most common clinical presentation of melioidosis is pneumonia which can occur in acute and chronic forms. The tsunami of 2004 demonstrated a new risk factor for the acquisition of melioidosis and resulted in the proposal that direct delivery of B. pseudomallei into the lungs may result in the enhanced ability of this pathogen to cause disease. In the present studies, we present the development and characterization of rat models of acute and chronic pulmonary melioidosis, and we have utilized these models to demonstrate that direct delivery of B. pseudomallei into the lungs does indeed result in the enhanced ability of this pathogen to cause disease. Importantly, the rat lung infection models for melioidosis can quantify differences in virulence between individual B. pseudomallei wild type strains during both acute and chronic infections. Further, the histopathology associated with pulmonary melioidosis in the rat resembles that seen in tuberculosis. B. pseudomallei microarrays were used to characterize gene expression patterns during chronic pulmonary infections. Transcriptional profiling at several time points during chronic infection revealed that a wide range of genes associated with virulence and metabolic functions are differentially regulated in vivo during chronic infections.
Collapse
Affiliation(s)
- Erin van Schaik
- Department of Microbiology and Infectious Diseases, Faculty of Medicine, University of Calgary Health Sciences Centre, 3330 Hospital Drive, NW, Calgary, Alberta T2N 4N1, Canada
| | | | | | | |
Collapse
|
47
|
La MV, Raoult D, Renesto P. Regulation of whole bacterial pathogen transcription within infected hosts. FEMS Microbiol Rev 2008; 32:440-60. [PMID: 18266740 DOI: 10.1111/j.1574-6976.2008.00103.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
DNA microarrays are a powerful and promising approach to gain a detailed understanding of the bacterial response and the molecular cross-talk that can occur as a consequence of host-pathogen interactions. However, published studies mainly describe the host response to infection. Analysis of bacterial gene regulation in the course of infection has confronted many challenges. This review summarizes the different strategies used over the last few years to investigate, at the genomic scale, and using microarrays, the alterations in the bacterial transcriptome in response to interactions with host cells. Thirty-seven studies involving 19 different bacterial pathogens were compiled and analyzed. Our in silico comparison of the transcription profiles of bacteria grown in broth or in contact with eukaryotic cells revealed some features commonly observed when bacteria interact with host cells, including stringent response and cell surface remodeling.
Collapse
Affiliation(s)
- My-Van La
- Unité des Rickettsies, Faculté de Médecine, 27 Boulevard Jean Moulin, Marseille, France
| | | | | |
Collapse
|
48
|
Comparative genomics and an insect model rapidly identify novel virulence genes of Burkholderia mallei. J Bacteriol 2008; 190:2306-13. [PMID: 18223084 DOI: 10.1128/jb.01735-07] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Burkholderia pseudomallei and its host-adapted deletion clone Burkholderia mallei cause the potentially fatal human diseases melioidosis and glanders, respectively. The antibiotic resistance profile and ability to infect via aerosol of these organisms and the absence of protective vaccines have led to their classification as major biothreats and select agents. Although documented infections by these bacteria date back over 100 years, relatively little is known about their virulence and pathogenicity mechanisms. We used in silico genomic subtraction to generate their virulome, a set of 650 putative virulence-related genes shared by B. pseudomallei and B. mallei but not present in five closely related nonpathogenic Burkholderia species. Although most of these genes are clustered in putative operons, the number of targets for mutant construction and verification of reduced virulence in animal models is formidable. Therefore, Galleria mellonella (wax moth) larvae were evaluated as a surrogate host; we found that B. pseudomallei and B. mallei, but not other phylogenetically related bacteria, were highly pathogenic for this insect. More importantly, four previously characterized B. mallei mutants with reduced virulence in hamsters or mice had similarly reduced virulence in G. mellonella larvae. Site-specific inactivation of selected genes in the computationally derived virulome identified three new potential virulence genes, each of which was required for rapid and efficient killing of larvae. Thus, this approach may provide a means to quickly identify high-probability virulence genes in B. pseudomallei, B. mallei, and other pathogens.
Collapse
|
49
|
Barnes JL, Ketheesan N. Development of protective immunity in a murine model of melioidosis is influenced by the source of
Burkholderia pseudomallei
antigens. Immunol Cell Biol 2007; 85:551-7. [PMID: 17563759 DOI: 10.1038/sj.icb.7100084] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Melioidosis is a potentially fatal disease caused by the bacterium, Burkholderia pseudomallei. The current study was carried out to determine the mechanisms involved in the development of protective immunity in a murine model of melioidosis. Following intravenous infection with B. pseudomallei, both C57BL/6 and BALB/c mice demonstrated delayed-type hypersensitivity responses and lymphocyte proliferation towards B. pseudomallei antigens, indicating the generation of B. pseudomallei-specific lymphocytes. Adoptive transfer of these lymphocytes to naïve C57BL/6 mice was demonstrated by a delayed-type hypersensitivity response. Mice were not protected from a subsequent lethal challenge with a highly virulent strain of B. pseudomallei, suggesting that a single intravenous dose of the bacterium is insufficient to induce a protective adaptive immune response. Attempts to induce resistance in susceptible BALB/c mice used repetitive low-dose exposure to live B. pseudomallei. Immune responses and resistance following subcutaneous immunization with live B. pseudomallei were compared with exposure to heat-killed, culture filtrate and sonicated B. pseudomallei antigens. Compared to heat-killed B. pseudomallei, significant protection was generated in BALB/c mice following immunization with live bacteria. Our studies also demonstrate that the type of immune response generated in vivo is influenced by the antigenic preparation of B. pseudomallei used for immunization.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antigens, Bacterial/administration & dosage
- Antigens, Bacterial/immunology
- Antigens, Bacterial/therapeutic use
- Burkholderia pseudomallei/immunology
- Cell Proliferation
- Cells, Cultured
- Disease Models, Animal
- Female
- Hypersensitivity, Delayed/immunology
- Immunity, Innate/immunology
- Immunity, Innate/physiology
- Injections, Intravenous
- Lymphocytes/cytology
- Macrophages/immunology
- Macrophages/metabolism
- Male
- Melioidosis/immunology
- Melioidosis/pathology
- Melioidosis/therapy
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Vaccination
Collapse
Affiliation(s)
- Jodie L Barnes
- Department of Microbiology and Immunology, School of Veterinary and Biomedical Sciences, James Cook University, Townsville, Queensland, Australia.
| | | |
Collapse
|
50
|
Korbsrisate S, Tomaras AP, Damnin S, Ckumdee J, Srinon V, Lengwehasatit I, Vasil ML, Suparak S. Characterization of two distinct phospholipase C enzymes from Burkholderia pseudomallei. MICROBIOLOGY (READING, ENGLAND) 2007; 153:1907-1915. [PMID: 17526847 DOI: 10.1099/mic.0.2006/003004-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Burkholderia pseudomallei is a serious bacterial pathogen that can cause a lethal infection in humans known as melioidosis. In this study two of its phospholipase C (PLC) enzymes (Plc-1 and Plc-2) were characterized. Starting with a virulent strain, two single mutants were constructed, each with one plc gene inactivated, and one double mutant with both plc genes inactivated. The single plc mutants exhibited decreased extracellular PLC activity in comparison to the wild-type strain, thereby demonstrating that the two genes encoded functional extracellular PLCs. Growth comparisons between the wild-type and PLC mutants in egg-yolk-supplemented medium indicated that both PLCs contributed to egg-yolk phospholipid utilization. Both PLCs hydrolysed phosphatidylcholine and sphingomyelin but neither was haemolytic for human erythrocytes. Experimental infections of eukaryotic cells demonstrated that Plc-1 itself had no effect on plaque-forming efficiency but it had an additive effect on increasing the efficiency of Plc-2 to form plaques. Only Plc-2 had a significant role in host cell cytotoxicity. In contrast, neither Plc-1 nor Plc-2 appeared to play any role in multinucleated giant cell (MNGC) formation or induction of apoptotic death in the cells studied. These data suggested that PLCs contribute, at least in part, to B. pseudomallei virulence and support the view that Plc-1 and Plc-2 are not redundant virulence factors.
Collapse
Affiliation(s)
- Sunee Korbsrisate
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Andrew P Tomaras
- Department of Microbiology, University of Colorado at Denver and Health Sciences Center Aurora, CO 80045, USA
| | - Suwat Damnin
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Jutturong Ckumdee
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Varintip Srinon
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | | | - Michael L Vasil
- Department of Microbiology, University of Colorado at Denver and Health Sciences Center Aurora, CO 80045, USA
| | - Supaporn Suparak
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| |
Collapse
|