1
|
Jia Q, Bowen RA, Masleša-Galić S, Horwitz MA. rLVS Δ capB/Yp F1-V single vector platform vaccine expressing Yersinia pestis F1 and LcrV antigens provides complete protection against lethal respiratory challenge with virulent plague bacilli. Hum Vaccin Immunother 2025; 21:2507475. [PMID: 40417828 PMCID: PMC12118393 DOI: 10.1080/21645515.2025.2507475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Revised: 05/08/2025] [Accepted: 05/13/2025] [Indexed: 05/27/2025] Open
Abstract
Yersinia pestis, the causative agent of plague, is classified as a Tier I Select Agent of bioterrorism and is among a few pathogens of high concern as a potential cause of a future pandemic. Currently, there is no licensed vaccine against plague. Previously, we developed a live attenuated vaccine candidate, rLVS ΔcapB/Yp F1-V, that utilizes a highly attenuated capB mutant of Francisella tularensis Live Vaccine Strain as a vector to express a fusion protein of Y. pestis F1 and LcrV antigens. We showed that homologous prime-boost vaccination with this vaccine provided potent protection in mice against lethal respiratory challenge with virulent Y. pestis. Here, we report on the immunogenicity and efficacy of rLVS ΔcapB/Yp F1-V and additional LVS ΔcapB-vectored vaccine candidates in mice. We demonstrate that three homologous prime-boost immunizations with an optimized dose of rLVS ΔcapB/Yp F1-V provided complete protection against pneumonic plague in a stringent mouse model, outperforming other candidates and matching the survival efficacy of the toxic and unlicensed live attenuated Y. pestis EV76 strain vaccine; moreover, mice immunized with the rLVS ΔcapB/Yp F1-V vaccine had minimal weight loss post-challenge that was significantly less than mice immunized with the EV76 vaccine. Protection induced by rLVS ΔcapB/Yp F1-V correlates with F1 and LcrV-specific serum antibody levels. Our results highlight the potential of rLVS ΔcapB/Yp F1-V to address the unmet need for a plague vaccine.
Collapse
MESH Headings
- Animals
- Plague/prevention & control
- Plague/immunology
- Plague Vaccine/immunology
- Plague Vaccine/administration & dosage
- Plague Vaccine/genetics
- Pore Forming Cytotoxic Proteins/immunology
- Pore Forming Cytotoxic Proteins/genetics
- Yersinia pestis/immunology
- Yersinia pestis/genetics
- Vaccines, Attenuated/immunology
- Vaccines, Attenuated/administration & dosage
- Vaccines, Attenuated/genetics
- Antigens, Bacterial/immunology
- Antigens, Bacterial/genetics
- Female
- Bacterial Proteins/immunology
- Bacterial Proteins/genetics
- Antibodies, Bacterial/blood
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Mice
- Mice, Inbred BALB C
- Disease Models, Animal
- Immunization, Secondary
- Genetic Vectors
- Survival Analysis
- Fimbriae Proteins/immunology
- Fimbriae Proteins/genetics
- Francisella tularensis/genetics
Collapse
Affiliation(s)
- Qingmei Jia
- Division of Infectious Diseases, Department of Medicine, 32-150 Center for Health Sciences, School of Medicine, University of California – Los Angeles, Los Angeles, CA, USA
| | - Richard A. Bowen
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Saša Masleša-Galić
- Division of Infectious Diseases, Department of Medicine, 32-150 Center for Health Sciences, School of Medicine, University of California – Los Angeles, Los Angeles, CA, USA
| | - Marcus A. Horwitz
- Division of Infectious Diseases, Department of Medicine, 32-150 Center for Health Sciences, School of Medicine, University of California – Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
2
|
Rashid M, Tachiyama S, Zhu S, Zhao H, McCaig WD, Sun J, Li H, Liu J, Thanassi DG. Outer membrane tube formation by Francisella novicida involves extensive envelope modifications and is linked with type VI secretion and alterations to the host phagosomal membrane. mBio 2025:e0106025. [PMID: 40387340 DOI: 10.1128/mbio.01060-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2025] [Accepted: 04/15/2025] [Indexed: 05/20/2025] Open
Abstract
Francisella tularensis is a gram-negative, intracellular pathogen that causes the zoonotic disease tularemia. Due to its ease of dissemination and high lethality, F. tularensis is classified as a tier 1 select agent with potential for misuse as a bioweapon. The mechanisms by which Francisella replicates intracellularly and interacts with the host during infection are not well understood. Francisella produces spherical outer membrane vesicles (OMVs) and novel tubular extensions of its cell surface that are also released extracellularly. These OMV and outer membrane tubes (OMTs) contain Francisella virulence factors and are produced in response to amino acid starvation and during infection of macrophages. To investigate how the OMTs are formed, we used cryogenic electron tomography to examine the model Francisella spp., Francisella novicida, during in vitro culture and within the macrophage phagosome. OMT formation involved progressive alterations of the bacterial envelope, resulting in extensions of both the inner and outer membranes. A dynamic cytoplasmic structure was present at the base of the OMT that extended into the tubes during elongation, together with cytoplasmic material. OMT produced within the macrophage phagosome was associated with changes to the phagosomal membrane, suggesting a role in phagosomal escape. Consistent with this, using confocal microscopy, we observed co-localization of the Francisella type VI secretion system with the OMT, both within bacteria and in released tubular vesicles. These findings reveal the cellular transformations that occur during membrane tubulation by Francisella and provide insights into the function of membrane-derived structures during host-pathogen interactions. IMPORTANCE Francisella tularensis is an intracellular bacterial pathogen that causes the zoonotic disease tularemia. Following uptake by host cells, the bacteria rapidly escape the phagosome and replicate intracellularly. In previous studies, we found that Francisella produces tubular extensions of its cell surface in response to specific cues and during macrophage infection. In the present study, we used cryogenic electron tomography to examine tube formation by the model Francisella sp., F. novicida. This analysis revealed that tube formation involves extensive bacterial envelope alterations and a dynamic cytoplasmic organelle. Furthermore, tubes produced by bacteria within infected macrophages were associated with the breakdown of the phagosomal membrane. In addition, we found that the Francisella type VI secretion system, which is essential for phagosomal escape, co-localized with the bacterial tubes. These findings reveal the cellular transformations that occur during membrane tubulation by Francisella and suggest a role for the tubes in phagosomal escape.
Collapse
Affiliation(s)
- Maheen Rashid
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA
- Center for Infectious Diseases, Stony Brook University, Stony Brook, New York, USA
| | - Shoichi Tachiyama
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, Connecticut, USA
- Microbial Sciences Institute, Yale University, New Haven, Connecticut, USA
| | - Shiwei Zhu
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, Connecticut, USA
- Microbial Sciences Institute, Yale University, New Haven, Connecticut, USA
| | - Hang Zhao
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, Connecticut, USA
- Microbial Sciences Institute, Yale University, New Haven, Connecticut, USA
| | - William D McCaig
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA
- Center for Infectious Diseases, Stony Brook University, Stony Brook, New York, USA
| | - Jingchuan Sun
- Department of Structural Biology, Van Andel Institute, Grand Rapids, Michigan, USA
| | - Huilin Li
- Department of Structural Biology, Van Andel Institute, Grand Rapids, Michigan, USA
| | - Jun Liu
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, Connecticut, USA
- Microbial Sciences Institute, Yale University, New Haven, Connecticut, USA
| | - David G Thanassi
- Department of Microbiology and Immunology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA
- Center for Infectious Diseases, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
3
|
Armstrong ZR, Alonso J, Stanton V, Patel N, Zogaj X, Cocioba SS, Klose KE. Mobilizable shuttle vectors with fluorescent markers functional across different species of bacteria. Appl Environ Microbiol 2025:e0004525. [PMID: 40353662 DOI: 10.1128/aem.00045-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 03/13/2025] [Indexed: 05/14/2025] Open
Abstract
Chromophore-containing proteins (CCPs), including fluorescent and non-fluorescent (chromoproteins), have been widely used in microbiological research. However, several roadblocks often limit their use in non-model bacterial species, including efficient transformation, suitable plasmid origins of replication, and optimal promoter choice. Here, we have engineered a set of 32 shuttle plasmids designed to overcome these roadblocks in an effort to streamline this process for future research. We have selected eight different CCPs: eforCP, YukonOFP, DasherGFP, tinsel Purple, aeBlue, FuGFP, super-folder GFP, and super-folder Cherry2. To broaden the potential host range, we utilized two distinct backbones with p15a either fused to a Francisella origin (FnOri) or to the broad host origin RSF1010 and included a transfer origin (oriT) to facilitate transformation via conjugation. Moreover, we have created versions of each vector, which confer resistance to either kanamycin or chloramphenicol. Lastly, to enable promoter-swapping, we engineered the constitutive pJ23100 promoter element to be flanked by BsaI sites, thereby enabling promoter exchange by the Golden Gate assembly to evaluate CCP expression with different host promoters. To demonstrate the usability of the pKEK-Chrom plasmid series, we evaluated their expression in Escherichia coli, Shewanella oneidensis, and Vibrio alginolyticus. We further demonstrated the utility of promoter swapping in Francisella novicida and validated the functionality of the RSF1010 origin in Acinetobacter baumannii. In summary, the pKEK-Chrom plasmid series provides a palette of different CCPs that streamline their use in non-model gram-negative bacteria. IMPORTANCE Chromophore-containing proteins (CCPs), including both fluorescent proteins and pigment-producing (non-fluorescent) chromoproteins, have become invaluable tools for microbial research. However, their successful implementation in understudied bacterial species lacking established genetic tools often requires substantial time and resources. Our goal was to develop a set of plasmid-based vectors that could streamline CCP expression in gram-negative bacteria. To do so, we developed a set of 32 plasmid vectors, the pKEK-Chrom plasmid series, specifically designed to facilitate CCP expression across different bacteria, including Escherichia coli, Vibrio alginolyticus, Shewanella oneidensis, Francisella novicida, and Acinetobacter baumannii.
Collapse
Affiliation(s)
- Zackary R Armstrong
- South Texas Center for Emerging Infectious Diseases and Department of Molecular Microbiology and Immunology, University of Texas San Antonio, San Antonio, Texas, USA
| | - Janie Alonso
- South Texas Center for Emerging Infectious Diseases and Department of Molecular Microbiology and Immunology, University of Texas San Antonio, San Antonio, Texas, USA
| | - Venus Stanton
- South Texas Center for Emerging Infectious Diseases and Department of Molecular Microbiology and Immunology, University of Texas San Antonio, San Antonio, Texas, USA
| | - Nikhil Patel
- South Texas Center for Emerging Infectious Diseases and Department of Molecular Microbiology and Immunology, University of Texas San Antonio, San Antonio, Texas, USA
| | - Xhavit Zogaj
- South Texas Center for Emerging Infectious Diseases and Department of Molecular Microbiology and Immunology, University of Texas San Antonio, San Antonio, Texas, USA
| | | | - Karl E Klose
- South Texas Center for Emerging Infectious Diseases and Department of Molecular Microbiology and Immunology, University of Texas San Antonio, San Antonio, Texas, USA
| |
Collapse
|
4
|
Hussain A, Ong EBB, Balaram P, Ismail A, Kien PK. TolC facilitates the intracellular survival and immunomodulation of Salmonella Typhi in human host cells. Virulence 2024; 15:2395831. [PMID: 39185619 PMCID: PMC11385165 DOI: 10.1080/21505594.2024.2395831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 06/29/2024] [Accepted: 08/19/2024] [Indexed: 08/27/2024] Open
Abstract
Salmonella enterica serovar Typhi (S. Typhi) causes typhoid fever, a systemic infection that affects millions of people worldwide. S. Typhi can invade and survive within host cells, such as intestinal epithelial cells and macrophages, by modulating their immune responses. However, the immunomodulatory capability of S. Typhi in relation to TolC-facilitated efflux pump function remains unclear. The role of TolC, an outer membrane protein that facilitates efflux pump function, in the invasion and immunomodulation of S. Typhi, was studied in human intestinal epithelial cells and macrophages. The tolC deletion mutant of S. Typhi was compared with the wild-type and its complemented strain in terms of their ability to invade epithelial cells, survive and induce cytotoxicity in macrophages, and elicit proinflammatory cytokine production in macrophages. The tolC mutant, which has a defective outer membrane, was impaired in invading epithelial cells compared to the wild-type strain, but the intracellular presence of the tolC mutant exhibited greater cytotoxicity and induced higher levels of proinflammatory cytokines (IL-1β and IL-8) in macrophages compared to the wild-type strain. These effects were reversed by complementing the tolC mutant with a functional tolC gene. Our results suggest that TolC plays a role in S. Typhi to efficiently invade epithelial cells and suppress host immune responses during infection. TolC may be a potential target for the development of novel therapeutics against typhoid fever.
Collapse
Affiliation(s)
- Ashraf Hussain
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang, Malaysia
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL USA
| | - Eugene Boon Beng Ong
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang, Malaysia
| | - Prabha Balaram
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang, Malaysia
| | - Asma Ismail
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang, Malaysia
| | - Phua Kia Kien
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang, Malaysia
| |
Collapse
|
5
|
Khalid N, Adams N, Cunha F, Taki AC, Le TG, Baell JB, Heine HS, Gasser RB, Eshraghi A. Tolfenpyrad Derivatives Exhibit Potent Francisella-Specific Antibacterial Activity without Toxicity to Mammalian Cells In Vitro. ACS Infect Dis 2024; 10:3902-3914. [PMID: 39356820 DOI: 10.1021/acsinfecdis.4c00547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
Tularemia is a deadly disease caused by Francisella tularensis, an emerging intracellular bacterial pathogen that can be disseminated rapidly through aerosols and vector-borne transmission. Recent surveillance data demonstrate an increasing incidence in several countries. Although clinical isolates of Francisella strains are sensitive to currently used antibiotics, engineered or horizontal acquisition of antibiotic resistance is a constant threat to public health. Therefore, the identification of antibiotics that target previously undrugged pathways is required to safeguard human health. An environmental pesticide that is registered for use in multiple countries, tolfenpyrad, shows promising activity to block Francisella growth; however, it is not a suitable antimicrobial candidate for use in vivo due to potential toxicity in humans and other animals. In this study, we applied a structure-activity relationship approach to tolfenpyrad to generate compounds with improved antibacterial activity and reduced toxicity. Through screening of a library of derivatives, we identified analogs with improved therapeutic windows compared with tolfenpyrad. Although structural diversity exists among these analogs, they inhibit the growth of Francisella species but not other Gram-negative or Gram-positive species. These compounds block intramacrophage growth of F. novicida and pathogenesis in an in vivo arthropod model of infection. Although the biochemical activity of these drugs is unknown, they appear to target the same pathway as the parent molecule because F. novicida mutants that are resistant to tolfenpyrad are also resistant to its analogs. Taken together, these findings suggest that these tolfenpyrad-derived compounds comprise a new class of Francisella-targeted antimicrobials and merit further evaluation and development.
Collapse
Affiliation(s)
- Nimra Khalid
- Department of Infectious Diseases & Immunology, University of Florida, Gainesville, Florida 32608, United States
| | - Nicole Adams
- Department of Infectious Diseases & Immunology, University of Florida, Gainesville, Florida 32608, United States
| | - Federico Cunha
- Department of Large Animal Clinical Sciences, University of Florida, Gainesville, Florida 32608, United States
| | - Aya C Taki
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Melbourne, Victoria 3010, Australia
| | - Thuy G Le
- Department of Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Melbourne, Victoria 3052, Australia
| | - Jonathan B Baell
- Department of Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Melbourne, Victoria 3052, Australia
| | - Henry S Heine
- Institute for Therapeutic Innovation, University of Florida, Orlando, Florida 32827, United States
| | - Robin B Gasser
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Melbourne, Victoria 3010, Australia
| | - Aria Eshraghi
- Department of Infectious Diseases & Immunology, University of Florida, Gainesville, Florida 32608, United States
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida 32610, United States
- Department of Oral Biology, University of Florida, Gainesville, Florida 32610, United States
| |
Collapse
|
6
|
Kopping EJ, Benziger PT, Thanassi DG. TolC and EmrA1 contribute to Francisella novicida multidrug resistance and modulation of host cell death. J Bacteriol 2024; 206:e0024624. [PMID: 39194223 PMCID: PMC11411944 DOI: 10.1128/jb.00246-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 07/29/2024] [Indexed: 08/29/2024] Open
Abstract
Francisella spp. are Gram-negative, facultative intracellular pathogens. Francisella tularensis causes the human disease tularemia and is considered a biological threat agent due to its high infectivity and virulence. A central aspect of Francisella virulence is its ability to dampen host immune responses. We previously identified the outer membrane channel (OMC) protein TolC as a critical F. tularensis virulence factor required for suppression of apoptotic and proinflammatory responses during macrophage infection. TolC functions as part of multidrug efflux systems and the type I secretion pathway that exports bacterial effector proteins. In these systems, TolC forms tripartite complexes together with an inner membrane transporter and periplasmic membrane fusion protein (MFP). To advance understanding of TolC function in Francisella, we analyzed OMC and MFP homologs in Francisella novicida, a widely used model species that causes a tularemia-like disease in mice. In agreement with the previous F. tularensis studies, all three OMCs present in F. novicida contributed to multidrug resistance, but only TolC was important for suppressing macrophage cell death. In addition, we identified the EmrA1 MFP as important for resisting antimicrobial compounds and dampening host cell death. In contrast to results obtained with F. tularensis, the cell death triggered during infection with the F. novicida tolC and emrA1 mutants was dominated by pyroptosis rather than apoptosis. These data expand our understanding of TolC function in Francisella and underscore both conserved and differential aspects of F. novicida and F. tularensis. IMPORTANCE Francisella tularensis is a Gram-negative intracellular bacterial pathogen and causative agent of tularemia. We previously identified the outer membrane channel protein TolC as contributing to antimicrobial resistance and subversion of host responses by F. tularensis. To advance understanding of TolC function in Francisella and to identify components that might work together with TolC, we took advantage of a transposon mutant library in F. novicida, a model species that causes a tularemia-like disease in mice. Our findings identify TolC and the membrane fusion protein EmrA1 as important for both antimicrobial resistance and suppression of macrophage cell death. This study also revealed differences in cell death pathways triggered by F. novicida versus F. tularensis infection that may relate to differences in virulence.
Collapse
Affiliation(s)
- Erik J Kopping
- Department of Microbiology and Immunology, School of Medicine, Stony Brook University, Stony Brook, New York, USA
- Center for Infectious Diseases, Stony Brook University, Stony Brook, New York, USA
| | - P Todd Benziger
- Department of Microbiology and Immunology, School of Medicine, Stony Brook University, Stony Brook, New York, USA
- Center for Infectious Diseases, Stony Brook University, Stony Brook, New York, USA
| | - David G Thanassi
- Department of Microbiology and Immunology, School of Medicine, Stony Brook University, Stony Brook, New York, USA
- Center for Infectious Diseases, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
7
|
Bedree JK, Bourgeois J, Balani P, Cen L, Hendrickson EL, Kerns KA, Camilli A, McLean JS, Shi W, He X. Identifying essential genes in Schaalia odontolytica using a highly-saturated transposon library. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.17.604004. [PMID: 39071323 PMCID: PMC11275721 DOI: 10.1101/2024.07.17.604004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
The unique epibiotic-parasitic relationship between Nanosynbacter lyticus type strain TM7x, a member of the newly identified Candidate Phyla Radiation, now referred to as Patescibacteria, and its basibiont, Schaalia odontolytica strain XH001 (formerly Actinomyces odontolyticus), require more powerful genetic tools for deeper understanding of the genetic underpinnings that mediate their obligate relationship. Previous studies have mainly characterized the genomic landscape of XH001 during or post TM7x infection through comparative genomic or transcriptomic analyses followed by phenotypic analysis. Comprehensive genetic dissection of the pair is currently cumbersome due to the lack of robust genetic tools in TM7x. However, basic genetic tools are available for XH001 and this study expands the current genetic toolset by developing high-throughput transposon insertion sequencing (Tn-seq). Tn-seq was employed to screen for essential genes in XH001 under laboratory conditions. A highly saturated Tn-seq library was generated with nearly 660,000 unique insertion mutations, averaging one insertion every 2-3 nucleotides. 203 genes, 10.5% of the XH001 genome, were identified as putatively essential.
Collapse
Affiliation(s)
- Joseph K Bedree
- Section of Oral Biology, Division of Oral Biology and Medicine, School of Dentistry, University of California-Los Angeles, Los Angeles, CA, 90095
- Department of Microbiology, The ADA Forsyth Institute; Cambridge, MA, 02142
| | - Jacob Bourgeois
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, USA
| | - Pooja Balani
- Department of Microbiology, The ADA Forsyth Institute; Cambridge, MA, 02142
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115
| | - Lujia Cen
- Department of Microbiology, The ADA Forsyth Institute; Cambridge, MA, 02142
| | - Erik L Hendrickson
- Department of Periodontics, School of Dentistry, University of Washington, Seattle, WA, 98195
| | - Kristopher A Kerns
- Department of Periodontics, School of Dentistry, University of Washington, Seattle, WA, 98195
| | - Andrew Camilli
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, USA
| | - Jeffrey S McLean
- Department of Periodontics, School of Dentistry, University of Washington, Seattle, WA, 98195
| | - Wenyuan Shi
- Department of Microbiology, The ADA Forsyth Institute; Cambridge, MA, 02142
| | - Xuesong He
- Department of Microbiology, The ADA Forsyth Institute; Cambridge, MA, 02142
| |
Collapse
|
8
|
Parkhill SL, Johnson EO. Integrating bacterial molecular genetics with chemical biology for renewed antibacterial drug discovery. Biochem J 2024; 481:839-864. [PMID: 38958473 PMCID: PMC11346456 DOI: 10.1042/bcj20220062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 07/04/2024]
Abstract
The application of dyes to understanding the aetiology of infection inspired antimicrobial chemotherapy and the first wave of antibacterial drugs. The second wave of antibacterial drug discovery was driven by rapid discovery of natural products, now making up 69% of current antibacterial drugs. But now with the most prevalent natural products already discovered, ∼107 new soil-dwelling bacterial species must be screened to discover one new class of natural product. Therefore, instead of a third wave of antibacterial drug discovery, there is now a discovery bottleneck. Unlike natural products which are curated by billions of years of microbial antagonism, the vast synthetic chemical space still requires artificial curation through the therapeutics science of antibacterial drugs - a systematic understanding of how small molecules interact with bacterial physiology, effect desired phenotypes, and benefit the host. Bacterial molecular genetics can elucidate pathogen biology relevant to therapeutics development, but it can also be applied directly to understanding mechanisms and liabilities of new chemical agents with new mechanisms of action. Therefore, the next phase of antibacterial drug discovery could be enabled by integrating chemical expertise with systematic dissection of bacterial infection biology. Facing the ambitious endeavour to find new molecules from nature or new-to-nature which cure bacterial infections, the capabilities furnished by modern chemical biology and molecular genetics can be applied to prospecting for chemical modulators of new targets which circumvent prevalent resistance mechanisms.
Collapse
Affiliation(s)
- Susannah L. Parkhill
- Systems Chemical Biology of Infection and Resistance Laboratory, The Francis Crick Institute, London, U.K
- Faculty of Life Sciences, University College London, London, U.K
| | - Eachan O. Johnson
- Systems Chemical Biology of Infection and Resistance Laboratory, The Francis Crick Institute, London, U.K
- Faculty of Life Sciences, University College London, London, U.K
- Department of Chemistry, Imperial College, London, U.K
- Department of Chemistry, King's College London, London, U.K
| |
Collapse
|
9
|
Ye C, Wu Q, Chen S, Zhang X, Xu W, Wu Y, Zhang Y, Yue Y. ECDEP: identifying essential proteins based on evolutionary community discovery and subcellular localization. BMC Genomics 2024; 25:117. [PMID: 38279081 PMCID: PMC10821549 DOI: 10.1186/s12864-024-10019-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 01/15/2024] [Indexed: 01/28/2024] Open
Abstract
BACKGROUND In cellular activities, essential proteins play a vital role and are instrumental in comprehending fundamental biological necessities and identifying pathogenic genes. Current deep learning approaches for predicting essential proteins underutilize the potential of gene expression data and are inadequate for the exploration of dynamic networks with limited evaluation across diverse species. RESULTS We introduce ECDEP, an essential protein identification model based on evolutionary community discovery. ECDEP integrates temporal gene expression data with a protein-protein interaction (PPI) network and employs the 3-Sigma rule to eliminate outliers at each time point, constructing a dynamic network. Next, we utilize edge birth and death information to establish an interaction streaming source to feed into the evolutionary community discovery algorithm and then identify overlapping communities during the evolution of the dynamic network. SVM recursive feature elimination (RFE) is applied to extract the most informative communities, which are combined with subcellular localization data for classification predictions. We assess the performance of ECDEP by comparing it against ten centrality methods, four shallow machine learning methods with RFE, and two deep learning methods that incorporate multiple biological data sources on Saccharomyces. Cerevisiae (S. cerevisiae), Homo sapiens (H. sapiens), Mus musculus, and Caenorhabditis elegans. ECDEP achieves an AP value of 0.86 on the H. sapiens dataset and the contribution ratio of community features in classification reaches 0.54 on the S. cerevisiae (Krogan) dataset. CONCLUSIONS Our proposed method adeptly integrates network dynamics and yields outstanding results across various datasets. Furthermore, the incorporation of evolutionary community discovery algorithms amplifies the capacity of gene expression data in classification.
Collapse
Affiliation(s)
- Chen Ye
- School of Information and Artificial Intelligence, Anhui Agricultural University, Hefei, Anhui, 230036, China
- Anhui Beidou Precision Agriculture Information Engineering Research Center, Anhui Agricultural University, Hefei, 230036, China
| | - Qi Wu
- School of Information and Artificial Intelligence, Anhui Agricultural University, Hefei, Anhui, 230036, China
- Anhui Beidou Precision Agriculture Information Engineering Research Center, Anhui Agricultural University, Hefei, 230036, China
| | - Shuxia Chen
- School of Information and Artificial Intelligence, Anhui Agricultural University, Hefei, Anhui, 230036, China
- Anhui Beidou Precision Agriculture Information Engineering Research Center, Anhui Agricultural University, Hefei, 230036, China
| | - Xuemei Zhang
- School of Information and Artificial Intelligence, Anhui Agricultural University, Hefei, Anhui, 230036, China
- Anhui Beidou Precision Agriculture Information Engineering Research Center, Anhui Agricultural University, Hefei, 230036, China
| | - Wenwen Xu
- School of Information and Artificial Intelligence, Anhui Agricultural University, Hefei, Anhui, 230036, China
- Anhui Beidou Precision Agriculture Information Engineering Research Center, Anhui Agricultural University, Hefei, 230036, China
| | - Yunzhi Wu
- School of Information and Artificial Intelligence, Anhui Agricultural University, Hefei, Anhui, 230036, China
- Anhui Beidou Precision Agriculture Information Engineering Research Center, Anhui Agricultural University, Hefei, 230036, China
| | - Youhua Zhang
- School of Information and Artificial Intelligence, Anhui Agricultural University, Hefei, Anhui, 230036, China
- Anhui Beidou Precision Agriculture Information Engineering Research Center, Anhui Agricultural University, Hefei, 230036, China
| | - Yi Yue
- School of Information and Artificial Intelligence, Anhui Agricultural University, Hefei, Anhui, 230036, China.
- Anhui Beidou Precision Agriculture Information Engineering Research Center, Anhui Agricultural University, Hefei, 230036, China.
| |
Collapse
|
10
|
van Hoek ML, Marchesani A, Rawat M. Diverse roles of low-molecular weight thiol GSH in Francisella's virulence, location sensing and GSH-stealing from host. CURRENT RESEARCH IN MICROBIAL SCIENCES 2023; 6:100218. [PMID: 38303966 PMCID: PMC10831187 DOI: 10.1016/j.crmicr.2023.100218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2024] Open
Abstract
Low-molecular weight (LMW) thiols, encompassing peptides and small proteins with active cysteine residue(s), are important to bacteria as they are involved in a wide range of redox reactions. They include the tripeptide glutathione (GSH) and the small redox proteins, thioredoxins and glutaredoxins. We review the low MW thiols and related molecules in Francisella species and what role they may play in growth and virulence. Genes for GSH biosynthesis, metabolism and thioredoxins are present in all strains of Francisella, including the fully human-virulent strains. GSH and cysteine (CSH) are the major LMW thiols in Francisella extracts. We explore the potential role of the LMW thiols to overcome the nutritional challenges of intracellular growth (high GSH conditions) as well as the nutritional challenges of planktonic growth (low GSH conditions), and their contribution to Francisella's sensing its environmental location. Francisella may also use GSH as a source of CSH, for which it is auxotrophic. "Glutathione stealing" from the host may be an important part of Francisella's success strategy as a facultative intracellular pathogen both to detect its location and obtain CSH. An understanding of GSH metabolism in Francisella provides insights into the interaction of this pathogen with its host and may reveal additional targets for therapeutic intervention for tularemia infections.
Collapse
Affiliation(s)
- Monique L. van Hoek
- School of Systems Biology, George Mason University, Manassas, VA, United States
| | | | - Mamta Rawat
- Biology Department, California State University, Fresno, CA, United States
| |
Collapse
|
11
|
Clarke A, Llabona IM, Khalid N, Hulvey D, Irvin A, Adams N, Heine HS, Eshraghi A. Tolfenpyrad displays Francisella-targeted antibiotic activity that requires an oxidative stress response regulator for sensitivity. Microbiol Spectr 2023; 11:e0271323. [PMID: 37800934 PMCID: PMC10848828 DOI: 10.1128/spectrum.02713-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 09/05/2023] [Indexed: 10/07/2023] Open
Abstract
IMPORTANCE Francisella species are highly pathogenic bacteria that pose a threat to global health security. These bacteria can be made resistant to antibiotics through facile methods, and we lack a safe and protective vaccine. Given their history of development as bioweapons, new treatment options must be developed to bolster public health preparedness. Here, we report that tolfenpyrad, a pesticide that is currently in use worldwide, effectively inhibits the growth of Francisella. This drug has an extensive history of use and a plethora of safety and toxicity data, making it a good candidate for development as an antibiotic. We identified mutations in Francisella novicida that confer resistance to tolfenpyrad and characterized a transcriptional regulator that is required for sensitivity to both tolfenpyrad and reactive oxygen species.
Collapse
Affiliation(s)
- Ashley Clarke
- Department of Infectious Diseases & Immunology, University of Florida, Gainesville, Florida, USA
| | - Isabelle M. Llabona
- Department of Infectious Diseases & Immunology, University of Florida, Gainesville, Florida, USA
| | - Nimra Khalid
- Department of Infectious Diseases & Immunology, University of Florida, Gainesville, Florida, USA
| | - Danielle Hulvey
- Department of Infectious Diseases & Immunology, University of Florida, Gainesville, Florida, USA
| | - Alexis Irvin
- Department of Infectious Diseases & Immunology, University of Florida, Gainesville, Florida, USA
| | - Nicole Adams
- Department of Infectious Diseases & Immunology, University of Florida, Gainesville, Florida, USA
| | - Henry S. Heine
- Institute for Therapeutic Innovation, University of Florida, Orlando, Florida, USA
| | - Aria Eshraghi
- Department of Infectious Diseases & Immunology, University of Florida, Gainesville, Florida, USA
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
- Department of Oral Biology, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
12
|
Välikangas T, Suomi T, Chandler CE, Scott AJ, Tran BQ, Ernst RK, Goodlett DR, Elo LL. Benchmarking tools for detecting longitudinal differential expression in proteomics data allows establishing a robust reproducibility optimization regression approach. Nat Commun 2022; 13:7877. [PMID: 36550114 PMCID: PMC9780321 DOI: 10.1038/s41467-022-35564-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
Quantitative proteomics has matured into an established tool and longitudinal proteomics experiments have begun to emerge. However, no effective, simple-to-use differential expression method for longitudinal proteomics data has been released. Typically, such data is noisy, contains missing values, and has only few time points and biological replicates. To address this need, we provide a comprehensive evaluation of several existing differential expression methods for high-throughput longitudinal omics data and introduce a Robust longitudinal Differential Expression (RolDE) approach. The methods are evaluated using over 3000 semi-simulated spike-in proteomics datasets and three large experimental datasets. In the comparisons, RolDE performs overall best; it is most tolerant to missing values, displays good reproducibility and is the top method in ranking the results in a biologically meaningful way. Furthermore, RolDE is suitable for different types of data with typically unknown patterns in longitudinal expression and can be applied by non-experienced users.
Collapse
Affiliation(s)
- Tommi Välikangas
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520, Turku, Finland
| | - Tomi Suomi
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520, Turku, Finland
| | | | - Alison J Scott
- University of Maryland - Baltimore, Baltimore, MD, 21201, USA
| | - Bao Q Tran
- US Army 20th Support Command CBRNE Analytical and Remediation Activity, Baltimore, MD, 21010-5424, USA
| | - Robert K Ernst
- University of Maryland - Baltimore, Baltimore, MD, 21201, USA
| | - David R Goodlett
- University of Victoria, Victoria, BC, V8P 3E6, Canada
- International Centre for Cancer Vaccine Science, Gdansk, Poland
| | - Laura L Elo
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520, Turku, Finland.
- Institute of Biomedicine, University of Turku, FI-20520, Turku, Finland.
| |
Collapse
|
13
|
Goodall ECA, Morris FC, McKeand SA, Sullivan R, Warner IA, Sheehan E, Boelter G, Icke C, Cunningham AF, Cole JA, Banzhaf M, Bryant JA, Henderson IR. LI-Detector: a Method for Curating Ordered Gene-Replacement Libraries. Microbiol Spectr 2022; 10:e0083322. [PMID: 35856675 PMCID: PMC9431181 DOI: 10.1128/spectrum.00833-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 07/01/2022] [Indexed: 12/04/2022] Open
Abstract
In recent years the availability of genome sequence information has grown logarithmically resulting in the identification of a plethora of uncharacterized genes. To address this gap in functional annotation, many high-throughput screens have been devised to uncover novel gene functions. Gene-replacement libraries are one such tool that can be screened in a high-throughput way to link genotype and phenotype and are key community resources. However, for a phenotype to be attributed to a specific gene, there needs to be confidence in the genotype. Construction of large libraries can be laborious and occasionally errors will arise. Here, we present a rapid and accurate method for the validation of any ordered library where a gene has been replaced or disrupted by a uniform linear insertion (LI). We applied our method (LI-detector) to the well-known Keio library of Escherichia coli gene-deletion mutants. Our method identified 3,718 constructed mutants out of a total of 3,728 confirmed isolates, with a success rate of 99.7% for identifying the correct kanamycin cassette position. This data set provides a benchmark for the purity of the Keio mutants and a screening method for mapping the position of any linear insertion, such as an antibiotic resistance cassette in any ordered library. IMPORTANCE The construction of ordered gene replacement libraries requires significant investment of time and resources to create a valuable community resource. During construction, technical errors may result in a limited number of incorrect mutants being made. Such mutants may confound the output of subsequent experiments. Here, using the remarkable E. coli Keio knockout library, we describe a method to rapidly validate the construction of every mutant.
Collapse
Affiliation(s)
- Emily C. A. Goodall
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Faye C. Morris
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, United Kingdom
| | - Samantha A. McKeand
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, United Kingdom
| | - Rudi Sullivan
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, United Kingdom
| | - Isabel A. Warner
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Emma Sheehan
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, United Kingdom
| | - Gabriela Boelter
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, United Kingdom
| | - Christopher Icke
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Adam F. Cunningham
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Jeffrey A. Cole
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, United Kingdom
| | - Manuel Banzhaf
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, United Kingdom
| | - Jack A. Bryant
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, United Kingdom
| | - Ian R. Henderson
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
14
|
Bythrow GV, Farhat MF, Levendosky K, Mohandas P, Germain GA, Yoo B, Quadri LEN. Mycobacterium abscessus Mutants with a Compromised Functional Link between the Type VII ESX-3 System and an Iron Uptake Mechanism Reliant on an Unusual Mycobactin Siderophore. Pathogens 2022; 11:pathogens11090953. [PMID: 36145386 PMCID: PMC9505556 DOI: 10.3390/pathogens11090953] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/12/2022] [Accepted: 08/18/2022] [Indexed: 11/28/2022] Open
Abstract
The opportunistic pathogen Mycobacterium abscessus subsp. abscessus (Mab) has become an emerging public health threat due to the increasing number of Mab-associated chronic pulmonary disease cases. Treatment requires multiple drug courses and is often combined with surgical resection. Cure rates are only ~50% due to treatment failure and comorbidities. Deeper understanding of the biology of Mab is required to illuminate potential avenues for the development of better therapeutics against Mab infections. The ESX-3 type VII protein secretion system of Mab has an important role in host inflammatory and pathological responses during infection. In this work, we demonstrate a functional link between ESX-3 and an iron uptake system based on an unusual mycobactin-type siderophore (designated MBT Ab) and exploit this link to implement a large screen for transposon mutants with an impaired ESX-3. Most mutants we identified carry insertions in genes encoding predicted ESX-3 secretion machinery components or potential ESX-3 substrates. The mutants overproduce MBT Ab, a trait consistent with an iron uptake defect. Our characterization of MBT Ab revealed structural features reminiscent of nocardial mycobactin-like compounds with cytotoxicity. This finding raises the possibility that MBT Ab may play roles in pathogenesis unlinked to iron homeostasis. The mutants generated herein will facilitate research to better understand the role of ESX-3 and its interplay with the siderophore system.
Collapse
Affiliation(s)
- Glennon V. Bythrow
- Department of Biology, Brooklyn College, City University of New York, 2900 Bedford Avenue, Brooklyn, NY 11210, USA
- Biology Program, Graduate Center, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
| | - Manal F. Farhat
- Department of Biology, Brooklyn College, City University of New York, 2900 Bedford Avenue, Brooklyn, NY 11210, USA
- Biology Program, Graduate Center, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
| | - Keith Levendosky
- Department of Biology, Brooklyn College, City University of New York, 2900 Bedford Avenue, Brooklyn, NY 11210, USA
- Biology Program, Graduate Center, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
| | - Poornima Mohandas
- Department of Biology, Brooklyn College, City University of New York, 2900 Bedford Avenue, Brooklyn, NY 11210, USA
- Biology Program, Graduate Center, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
| | - Gabrielle A. Germain
- Department of Biology, Brooklyn College, City University of New York, 2900 Bedford Avenue, Brooklyn, NY 11210, USA
- Biology Program, Graduate Center, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
| | - Barney Yoo
- Department of Chemistry, Hunter College, City University of New York, 695 Park Avenue, New York, NY 10065, USA
| | - Luis E. N. Quadri
- Department of Biology, Brooklyn College, City University of New York, 2900 Bedford Avenue, Brooklyn, NY 11210, USA
- Biology Program, Graduate Center, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
- Biochemistry Program, Graduate Center, City University of New York, 365 Fifth Avenue, New York, NY 10016, USA
- Correspondence:
| |
Collapse
|
15
|
Ozanic M, Marecic V, Knezevic M, Kelava I, Stojková P, Lindgren L, Bröms JE, Sjöstedt A, Abu Kwaik Y, Santic M. The type IV pili component PilO is a virulence determinant of Francisella novicida. PLoS One 2022; 17:e0261938. [PMID: 35077486 PMCID: PMC8789160 DOI: 10.1371/journal.pone.0261938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 12/14/2021] [Indexed: 12/03/2022] Open
Abstract
Francisella tularensis is a highly pathogenic intracellular bacterium that causes the disease tularemia. While its ability to replicate within cells has been studied in much detail, the bacterium also encodes a less characterised type 4 pili (T4P) system. T4Ps are dynamic adhesive organelles identified as major virulence determinants in many human pathogens. In F. tularensis, the T4P is required for adherence to the host cell, as well as for protein secretion. Several components, including pilins, a pili peptidase, a secretin pore and two ATPases, are required to assemble a functional T4P, and these are encoded within distinct clusters on the Francisella chromosome. While some of these components have been functionally characterised, the role of PilO, if any, still is unknown. Here, we examined the role of PilO in the pathogenesis of F. novicida. Our results show that the PilO is essential for pilus assembly on the bacterial surface. In addition, PilO is important for adherence of F. novicida to human monocyte-derived macrophages, secretion of effector proteins and intracellular replication. Importantly, the pilO mutant is attenuated for virulence in BALB/c mice regardless of the route of infection. Following intratracheal and intradermal infection, the mutant caused no histopathology changes, and demonstrated impaired phagosomal escape and replication within lung liver as well as spleen. Thus, PilO is an essential virulence determinant of F. novicida.
Collapse
Affiliation(s)
- Mateja Ozanic
- Faculty of Medicine, Department of Microbiology and Parasitology, University of Rijeka, Rijeka, Croatia
| | - Valentina Marecic
- Faculty of Medicine, Department of Microbiology and Parasitology, University of Rijeka, Rijeka, Croatia
| | - Masa Knezevic
- Faculty of Medicine, Department of Microbiology and Parasitology, University of Rijeka, Rijeka, Croatia
| | - Ina Kelava
- Faculty of Medicine, Department of Microbiology and Parasitology, University of Rijeka, Rijeka, Croatia
| | - Pavla Stojková
- Department of Clinical Microbiology and Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
| | - Lena Lindgren
- Department of Clinical Microbiology and Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
| | - Jeanette E. Bröms
- Department of Clinical Microbiology and Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
| | - Anders Sjöstedt
- Department of Clinical Microbiology and Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
| | - Yousef Abu Kwaik
- Department of Microbiology and Immunology and Center for Predictive Medicine, College of Medicine, University of Louisville, Louisville, Kentucky, United States of America
| | - Marina Santic
- Faculty of Medicine, Department of Microbiology and Parasitology, University of Rijeka, Rijeka, Croatia
| |
Collapse
|
16
|
Neubert K, Zuchantke E, Leidenfrost RM, Wünschiers R, Grützke J, Malorny B, Brendebach H, Al Dahouk S, Homeier T, Hotzel H, Reinert K, Tomaso H, Busch A. Testing assembly strategies of Francisella tularensis genomes to infer an evolutionary conservation analysis of genomic structures. BMC Genomics 2021; 22:822. [PMID: 34773979 PMCID: PMC8590783 DOI: 10.1186/s12864-021-08115-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 10/12/2021] [Indexed: 02/08/2023] Open
Abstract
Background We benchmarked sequencing technology and assembly strategies for short-read, long-read, and hybrid assemblers in respect to correctness, contiguity, and completeness of assemblies in genomes of Francisella tularensis. Benchmarking allowed in-depth analyses of genomic structures of the Francisella pathogenicity islands and insertion sequences. Five major high-throughput sequencing technologies were applied, including next-generation “short-read” and third-generation “long-read” sequencing methods. Results We focused on short-read assemblers, hybrid assemblers, and analysis of the genomic structure with particular emphasis on insertion sequences and the Francisella pathogenicity island. The A5-miseq pipeline performed best for MiSeq data, Mira for Ion Torrent data, and ABySS for HiSeq data from eight short-read assembly methods. Two approaches were applied to benchmark long-read and hybrid assembly strategies: long-read-first assembly followed by correction with short reads (Canu/Pilon, Flye/Pilon) and short-read-first assembly along with scaffolding based on long reads (Unicyler, SPAdes). Hybrid assembly can resolve large repetitive regions best with a “long-read first” approach. Conclusions Genomic structures of the Francisella pathogenicity islands frequently showed misassembly. Insertion sequences (IS) could be used to perform an evolutionary conservation analysis. A phylogenetic structure of insertion sequences and the evolution within the clades elucidated the clade structure of the highly conservative F. tularensis. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-08115-x.
Collapse
Affiliation(s)
- Kerstin Neubert
- Department of Mathematics and Computer Science, Algorithmic Bioinformatics, Freie Universität Berlin, Institute of Computer Science, Takustr. 9, 14195, Berlin, Germany.,German Federal Institute for Risk Assessment, Diedersdorfer Weg 1, 12277, Berlin, Germany
| | - Eric Zuchantke
- Friedrich-Loeffler-Institut, Institute of Bacterial Infections and Zoonoses, Naumburger Str. 96a, 07749, Jena, Germany
| | - Robert Maximilian Leidenfrost
- Department of Biotechnology and Chemistry, Mittweida University of Applied Sciences, Technikumplatz 17a, 09648, Mittweida, Germany
| | - Röbbe Wünschiers
- Department of Biotechnology and Chemistry, Mittweida University of Applied Sciences, Technikumplatz 17a, 09648, Mittweida, Germany
| | - Josephine Grützke
- German Federal Institute for Risk Assessment, Diedersdorfer Weg 1, 12277, Berlin, Germany
| | - Burkhard Malorny
- German Federal Institute for Risk Assessment, Diedersdorfer Weg 1, 12277, Berlin, Germany
| | - Holger Brendebach
- German Federal Institute for Risk Assessment, Diedersdorfer Weg 1, 12277, Berlin, Germany
| | - Sascha Al Dahouk
- German Federal Institute for Risk Assessment, Diedersdorfer Weg 1, 12277, Berlin, Germany
| | - Timo Homeier
- Friedrich-Loeffler-Institut, Institute of Epidemiology, Südufer, 10 17493, Greifswald, Insel Riems, Germany
| | - Helmut Hotzel
- Friedrich-Loeffler-Institut, Institute of Bacterial Infections and Zoonoses, Naumburger Str. 96a, 07749, Jena, Germany
| | - Knut Reinert
- Department of Mathematics and Computer Science, Algorithmic Bioinformatics, Freie Universität Berlin, Institute of Computer Science, Takustr. 9, 14195, Berlin, Germany
| | - Herbert Tomaso
- Friedrich-Loeffler-Institut, Institute of Bacterial Infections and Zoonoses, Naumburger Str. 96a, 07749, Jena, Germany
| | - Anne Busch
- Friedrich-Loeffler-Institut, Institute of Bacterial Infections and Zoonoses, Naumburger Str. 96a, 07749, Jena, Germany. .,Department of Anaesthesiology and Intensive Care Medicine, University Hospital Jena, Jena, Germany.
| |
Collapse
|
17
|
The Biosynthetic Pathway of Ubiquinone Contributes to Pathogenicity of Francisella novicida. J Bacteriol 2021; 203:e0040021. [PMID: 34543102 DOI: 10.1128/jb.00400-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Francisella tularensis is the causative agent of tularemia. Because of its extreme infectivity and high mortality rate, this pathogen was classified as a biothreat agent. Francisella spp. are strict aerobes, and ubiquinone (UQ) has been previously identified in these bacteria. While the UQ biosynthetic pathways were extensively studied in Escherichia coli, allowing the identification of 15 Ubi proteins to date, little is known about Francisella spp. In this study, and using Francisella novicida as a surrogate organism, we first identified ubiquinone 8 (UQ8) as the major quinone found in the membranes of this bacterium. Next, we characterized the UQ biosynthetic pathway in F. novicida using a combination of bioinformatics, genetics, and biochemical approaches. Our analysis disclosed the presence in Francisella of 10 putative Ubi proteins, and we confirmed 8 of them by heterologous complementation in E. coli. The UQ biosynthetic pathways from F. novicida and E. coli share similar patterns. However, differences were highlighted: the decarboxylase remains unidentified in Francisella spp., and homologs of the Ubi proteins involved in the O2-independent UQ pathway are not present. This is in agreement with the strictly aerobic niche of this bacterium. Next, via two approaches, i.e., the use of an inhibitor (3-amino-4-hydroxybenzoic acid) and a transposon mutant, both of which strongly impair the synthesis of UQ, we demonstrated that UQ is essential for the growth of F. novicida in respiratory medium and contributes to its pathogenicity in Galleria mellonella used as an alternative animal model. IMPORTANCE Francisella tularensis is the causative bacterium of tularemia and is classified as a biothreat agent. Using multidisciplinary approaches, we investigated the ubiquinone (UQ) biosynthetic pathway that operates in F. novicida used as a surrogate. We show that UQ8 is the major quinone identified in the membranes of Francisella novicida. We identified a new competitive inhibitor that strongly decreased the biosynthesis of UQ. Our demonstration of the crucial roles of UQ for the respiratory metabolism of F. novicida and for the involvement in its pathogenicity in the Galleria mellonella model should stimulate the search for selective inhibitors of bacterial UQ biosynthesis.
Collapse
|
18
|
Hansen JD, Ray K, Chen PJ, Yun S, Elliott DG, Conway CM, Calcutt MJ, Purcell MK, Welch TJ, Bellah JP, Davis EM, Greer JB, Soto E. Disruption of the Francisella noatunensis subsp. orientalis pdpA Gene Results in Virulence Attenuation and Protection in Zebrafish. Infect Immun 2021; 89:e0022021. [PMID: 34424748 PMCID: PMC8519269 DOI: 10.1128/iai.00220-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 08/10/2021] [Indexed: 12/20/2022] Open
Abstract
Several Francisella spp., including Francisella noatunensis, are regarded as important emerging pathogens of wild and farmed fish. However, very few studies have investigated the virulence factors that allow these bacterial species to be pathogenic in fish. The Francisella pathogenicity island (FPI) is a well-described, gene-dense region encoding major virulence factors for the genus Francisella. pdpA is a member of the pathogenicity-determining protein genes carried by the FPI that are implicated in the ability of the mammalian pathogen Francisella tularensis to escape and replicate in infected host cells. Using a sacB suicide approach, we generated pdpA knockouts to address the role of PdpA as a virulence factor for F. noatunensis. Because polarity can be an issue in gene-dense regions, we generated two different marker-based mutants in opposing polarity (the F. noatunensis subsp. orientalis ΔpdpA1 and ΔpdpA2 strains). Both mutants were attenuated (P < 0.0001) in zebrafish challenges and displayed impaired intracellular replication (P < 0.05) and cytotoxicity (P < 0.05), all of which could be restored to wild-type (WT) levels by complementation for the ΔpdpA1 mutant. Importantly, differences were found for bacterial burden and induction of acute-phase and proinflammatory genes for the F. noatunensis subsp. orientalis ΔpdpA1 and ΔpdpA2 mutants compared to the WT during acute infection. In addition, neither mutant resulted in significant histopathological changes. Finally, immunization with the F. noatunensis subsp. orientalis ΔpdpA1 mutant led to protection (P < 0.012) against an acute 40% lethal dose (LD40) challenge with WT F. noatunensis in the zebrafish model of infection. Taken together, the results from this study further demonstrate physiological similarities within the genus Francisella relative to their phylogenetic relationships and the utility of zebrafish for addressing virulence factors for the genus.
Collapse
Affiliation(s)
- John D. Hansen
- U.S. Geological Survey, Western Fisheries Research Center, Seattle, Washington, USA
| | - Karina Ray
- U.S. Geological Survey, Western Fisheries Research Center, Seattle, Washington, USA
| | - Po-Jui Chen
- U.S. Geological Survey, Western Fisheries Research Center, Seattle, Washington, USA
| | - Susan Yun
- Department of Medicine and Epidemiology, University of California—Davis, School of Veterinary Medicine, Davis, California, USA
| | - Diane G. Elliott
- U.S. Geological Survey, Western Fisheries Research Center, Seattle, Washington, USA
| | - Carla M. Conway
- U.S. Geological Survey, Western Fisheries Research Center, Seattle, Washington, USA
| | - Michael J. Calcutt
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, USA
| | - Maureen K. Purcell
- U.S. Geological Survey, Western Fisheries Research Center, Seattle, Washington, USA
| | - Timothy J. Welch
- National Center for Cool and Cold Water Aquaculture, Agricultural Research Service, United States Department of Agriculture, Kearneysville, West Virginia, USA
| | - John P. Bellah
- U.S. Geological Survey, Western Fisheries Research Center, Seattle, Washington, USA
| | - Ellie M. Davis
- U.S. Geological Survey, Western Fisheries Research Center, Seattle, Washington, USA
| | - Justin B. Greer
- U.S. Geological Survey, Western Fisheries Research Center, Seattle, Washington, USA
| | - Esteban Soto
- Department of Medicine and Epidemiology, University of California—Davis, School of Veterinary Medicine, Davis, California, USA
| |
Collapse
|
19
|
Li S, Zhang Z, Li X, Tan Y, Wang L, Chen Z. An iteration model for identifying essential proteins by combining comprehensive PPI network with biological information. BMC Bioinformatics 2021; 22:430. [PMID: 34496745 PMCID: PMC8425031 DOI: 10.1186/s12859-021-04300-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 07/08/2021] [Indexed: 11/10/2022] Open
Abstract
Background Essential proteins have great impacts on cell survival and development, and played important roles in disease analysis and new drug design. However, since it is inefficient and costly to identify essential proteins by using biological experiments, then there is an urgent need for automated and accurate detection methods. In recent years, the recognition of essential proteins in protein interaction networks (PPI) has become a research hotspot, and many computational models for predicting essential proteins have been proposed successively. Results In order to achieve higher prediction performance, in this paper, a new prediction model called TGSO is proposed. In TGSO, a protein aggregation degree network is constructed first by adopting the node density measurement method for complex networks. And simultaneously, a protein co-expression interactive network is constructed by combining the gene expression information with the network connectivity, and a protein co-localization interaction network is constructed based on the subcellular localization data. And then, through integrating these three kinds of newly constructed networks, a comprehensive protein–protein interaction network will be obtained. Finally, based on the homology information, scores can be calculated out iteratively for different proteins, which can be utilized to estimate the importance of proteins effectively. Moreover, in order to evaluate the identification performance of TGSO, we have compared TGSO with 13 different latest competitive methods based on three kinds of yeast databases. And experimental results show that TGSO can achieve identification accuracies of 94%, 82% and 72% out of the top 1%, 5% and 10% candidate proteins respectively, which are to some degree superior to these state-of-the-art competitive models. Conclusions We constructed a comprehensive interactive network based on multi-source data to reduce the noise and errors in the initial PPI, and combined with iterative methods to improve the accuracy of necessary protein prediction, and means that TGSO may be conducive to the future development of essential protein recognition as well.
Collapse
Affiliation(s)
- Shiyuan Li
- College of Computer Engineering and Applied Mathematics, Changsha University, Changsha, 410022, China.,Hunan Province Key Laboratory of Industrial Internet Technology and Security, Changsha University, Changsha, 410022, China
| | - Zhen Zhang
- College of Electronic Information and Electrical Engineering, Changsha University, Changsha, 410022, China
| | - Xueyong Li
- College of Computer Engineering and Applied Mathematics, Changsha University, Changsha, 410022, China.,Hunan Province Key Laboratory of Industrial Internet Technology and Security, Changsha University, Changsha, 410022, China
| | - Yihong Tan
- College of Computer Engineering and Applied Mathematics, Changsha University, Changsha, 410022, China. .,Hunan Province Key Laboratory of Industrial Internet Technology and Security, Changsha University, Changsha, 410022, China.
| | - Lei Wang
- College of Computer Engineering and Applied Mathematics, Changsha University, Changsha, 410022, China.,Hunan Province Key Laboratory of Industrial Internet Technology and Security, Changsha University, Changsha, 410022, China
| | - Zhiping Chen
- College of Computer Engineering and Applied Mathematics, Changsha University, Changsha, 410022, China. .,Hunan Province Key Laboratory of Industrial Internet Technology and Security, Changsha University, Changsha, 410022, China.
| |
Collapse
|
20
|
Xie J, Zhao C, Sun J, Li J, Yang F, Wang J, Nie Q. Prediction of Essential Genes in Comparison States Using Machine Learning. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2021; 18:1784-1792. [PMID: 32991286 DOI: 10.1109/tcbb.2020.3027392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Identifying essential genes in comparison states (EGS) is vital to understanding cell differentiation, performing drug discovery, and identifying disease causes. Here, we present a machine learning method termed Prediction of Essential Genes in Comparison States (PreEGS). To capture the alteration of the network in comparison states, PreEGS extracts topological and gene expression features of each gene in a five-dimensional vector. PreEGS also recruits a positive sample expansion method to address the problem of unbalanced positive and negative samples, which is often encountered in practical applications. Different classifiers are applied to the simulated datasets, and the PreEGS based on the random forests model (PreEGSRF) was chosen for optimal performance. PreEGSRF was then compared with six other methods, including three machine learning methods, to predict EGS in a specific state. On real datasets with four gene regulatory networks, PreEGSRF predicted five essential genes related to leukemia and five enriched KEGG pathways. Four of the predicted essential genes and all predicted pathways were consistent with previous studies and highly correlated with leukemia. With high prediction accuracy and generalization ability, PreEGSRF is broadly applicable for the discovery of disease-causing genes, driver genes for cell fate decisions, and complex biomarkers of biological systems.
Collapse
|
21
|
Chen W, Ren ZH, Tang N, Chai G, Zhang H, Zhang Y, Ma J, Wu Z, Shen X, Huang X, Luo GZ, Ji Q. Targeted genetic screening in bacteria with a Cas12k-guided transposase. Cell Rep 2021; 36:109635. [PMID: 34469724 DOI: 10.1016/j.celrep.2021.109635] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/26/2021] [Accepted: 08/09/2021] [Indexed: 12/26/2022] Open
Abstract
Microbes employ sophisticated cellular networks encoded by complex genomes to rapidly adapt to changing environments. High-throughput genome engineering methods are valuable tools for functionally profiling genotype-phenotype relationships and understanding the complexity of cellular networks. However, current methods either rely on special homologous recombination systems and are thus applicable in only limited bacterial species or can generate only nonspecific mutations and thus require extensive subsequent screening. Here, we report a site-specific transposon-assisted genome engineering (STAGE) method that allows high-throughput Cas12k-guided mutagenesis in various microorganisms, such as Pseudomonas aeruginosa and Klebsiella pneumoniae. Exploiting the powerful STAGE technique, we construct a site-specific transposon mutant library that focuses on all possible transcription factors (TFs) in P. aeruginosa, enabling the comprehensive identification of essential genes and antibiotic-resistance-related factors. Given its broad host range activity and easy programmability, this method can be widely adapted to diverse microbial species for rapid genome engineering and strain evolution.
Collapse
Affiliation(s)
- Weizhong Chen
- School of Physical Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Ze-Hui Ren
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, Guangdong, China
| | - Na Tang
- School of Physical Science and Technology, ShanghaiTech University, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guoshi Chai
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, Guangdong, China
| | - Hongyuan Zhang
- School of Physical Science and Technology, ShanghaiTech University, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yifei Zhang
- School of Physical Science and Technology, ShanghaiTech University, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiacheng Ma
- School of Physical Science and Technology, ShanghaiTech University, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhaowei Wu
- School of Physical Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xia Shen
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, Guangdong, China
| | - Xingxu Huang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; Guangzhou Laboratory, Guangzhou 510120, China
| | - Guan-Zheng Luo
- MOE Key Laboratory of Gene Function and Regulation, Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, Guangdong, China.
| | - Quanjiang Ji
- School of Physical Science and Technology, ShanghaiTech University, Shanghai 201210, China; Guangzhou Laboratory, Guangzhou 510120, China.
| |
Collapse
|
22
|
Rytter H, Jamet A, Ziveri J, Ramond E, Coureuil M, Lagouge-Roussey P, Euphrasie D, Tros F, Goudin N, Chhuon C, Nemazanyy I, de Moraes FE, Labate C, Guerrera IC, Charbit A. The pentose phosphate pathway constitutes a major metabolic hub in pathogenic Francisella. PLoS Pathog 2021; 17:e1009326. [PMID: 34339477 PMCID: PMC8360588 DOI: 10.1371/journal.ppat.1009326] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 08/12/2021] [Accepted: 07/14/2021] [Indexed: 11/19/2022] Open
Abstract
Metabolic pathways are now considered as intrinsic virulence attributes of pathogenic bacteria and thus represent potential targets for antibacterial strategies. Here we focused on the role of the pentose phosphate pathway (PPP) and its connections with other metabolic pathways in the pathophysiology of Francisella novicida. The involvement of the PPP in the intracellular life cycle of Francisella was first demonstrated by studying PPP inactivating mutants. Indeed, we observed that inactivation of the tktA, rpiA or rpe genes severely impaired intramacrophage multiplication during the first 24 hours. However, time-lapse video microscopy demonstrated that rpiA and rpe mutants were able to resume late intracellular multiplication. To better understand the links between PPP and other metabolic networks in the bacterium, we also performed an extensive proteo-metabolomic analysis of these mutants. We show that the PPP constitutes a major bacterial metabolic hub with multiple connections to glycolysis, the tricarboxylic acid cycle and other pathways, such as fatty acid degradation and sulfur metabolism. Altogether our study highlights how PPP plays a key role in the pathogenesis and growth of Francisella in its intracellular niche.
Collapse
Affiliation(s)
- Héloise Rytter
- Université de Paris, Paris, France
- INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades. Team 7: Pathogénie des Infections Systémiques, Paris, France
| | - Anne Jamet
- Université de Paris, Paris, France
- INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades. Team 7: Pathogénie des Infections Systémiques, Paris, France
| | - Jason Ziveri
- Université de Paris, Paris, France
- INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades. Team 7: Pathogénie des Infections Systémiques, Paris, France
| | - Elodie Ramond
- Université de Paris, Paris, France
- INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades. Team 7: Pathogénie des Infections Systémiques, Paris, France
| | - Mathieu Coureuil
- Université de Paris, Paris, France
- INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades. Team 7: Pathogénie des Infections Systémiques, Paris, France
| | - Pauline Lagouge-Roussey
- Université de Paris, Paris, France
- INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades. Team 7: Pathogénie des Infections Systémiques, Paris, France
| | - Daniel Euphrasie
- Université de Paris, Paris, France
- INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades. Team 7: Pathogénie des Infections Systémiques, Paris, France
| | - Fabiola Tros
- Université de Paris, Paris, France
- INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades. Team 7: Pathogénie des Infections Systémiques, Paris, France
| | - Nicolas Goudin
- Pole Bio-analyse d’images, Structure Fédérative de Recherche Necker INSERM US24- CNRS UMS 3633, Paris, France
| | - Cerina Chhuon
- Université de Paris, Paris, France
- Plateforme Protéome Institut Necker, PPN, Structure Fédérative de Recherche Necker INSERM US24-CNRS UMS 3633, Paris, France
| | - Ivan Nemazanyy
- Université de Paris, Paris, France
- Plateforme Etude du métabolisme, Structure Fédérative de Recherche Necker INSERM US24-CNRS UMS 3633, Paris, France
| | - Fabricio Edgar de Moraes
- Laboratório Max Feffer de Genética de Plantas, Departamento de Genética, Escola Superior de Agricultura Luiz de Queiroz, Universidade de São Paulo, Piracicaba, Brazil
| | - Carlos Labate
- Laboratório Max Feffer de Genética de Plantas, Departamento de Genética, Escola Superior de Agricultura Luiz de Queiroz, Universidade de São Paulo, Piracicaba, Brazil
| | - Ida Chiara Guerrera
- Université de Paris, Paris, France
- Plateforme Protéome Institut Necker, PPN, Structure Fédérative de Recherche Necker INSERM US24-CNRS UMS 3633, Paris, France
- * E-mail: (ICG); (AC)
| | - Alain Charbit
- Université de Paris, Paris, France
- INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades. Team 7: Pathogénie des Infections Systémiques, Paris, France
- * E-mail: (ICG); (AC)
| |
Collapse
|
23
|
Chin CY, Zhao J, Llewellyn AC, Golovliov I, Sjöstedt A, Zhou P, Weiss DS. Francisella FlmX broadly affects lipopolysaccharide modification and virulence. Cell Rep 2021; 35:109247. [PMID: 34133919 DOI: 10.1016/j.celrep.2021.109247] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 01/14/2021] [Accepted: 05/20/2021] [Indexed: 10/21/2022] Open
Abstract
The outer membrane protects Gram-negative bacteria from the host environment. Lipopolysaccharide (LPS), a major outer membrane constituent, has distinct components (lipid A, core, O-antigen) generated by specialized pathways. In this study, we describe the surprising convergence of these pathways through FlmX, an uncharacterized protein in the intracellular pathogen Francisella. FlmX is in the flippase family, which includes proteins that traffic lipid-linked envelope components across membranes. flmX deficiency causes defects in lipid A modification, core remodeling, and O-antigen addition. We find that an F. tularensis mutant lacking flmX is >1,000,000-fold attenuated. Furthermore, FlmX is required to resist the innate antimicrobial LL-37 and the antibiotic polymyxin. Given FlmX's central role in LPS modification and its conservation in intracellular pathogens Brucella, Coxiella, and Legionella, FlmX may represent a novel drug target whose inhibition could cripple bacterial virulence and sensitize bacteria to innate antimicrobials and antibiotics.
Collapse
Affiliation(s)
- Chui-Yoke Chin
- Emory Antibiotic Resistance Center, Emory University School of Medicine, Atlanta, GA 30329, USA; Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30329, USA; Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, GA 30329, USA; Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30329, USA
| | - Jinshi Zhao
- Department of Biochemistry, Duke University School of Medicine, Durham, NC 27710, USA
| | - Anna C Llewellyn
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30329, USA; Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, GA 30329, USA; Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30329, USA
| | - Igor Golovliov
- Clinical Bacteriology, and Laboratory for Molecular Infection Medicine Sweden, Department of Clinical Microbiology, Umeå University, 90185 Umeå, Sweden
| | - Anders Sjöstedt
- Clinical Bacteriology, and Laboratory for Molecular Infection Medicine Sweden, Department of Clinical Microbiology, Umeå University, 90185 Umeå, Sweden
| | - Pei Zhou
- Department of Biochemistry, Duke University School of Medicine, Durham, NC 27710, USA
| | - David S Weiss
- Emory Antibiotic Resistance Center, Emory University School of Medicine, Atlanta, GA 30329, USA; Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30329, USA; Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, GA 30329, USA; Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30329, USA; Research Service, Atlanta VA Medical Center, Decatur, GA 30033, USA.
| |
Collapse
|
24
|
Kassinger SJ, van Hoek ML. Genetic Determinants of Antibiotic Resistance in Francisella. Front Microbiol 2021; 12:644855. [PMID: 34054749 PMCID: PMC8149597 DOI: 10.3389/fmicb.2021.644855] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 03/25/2021] [Indexed: 12/21/2022] Open
Abstract
Tularemia, caused by Francisella tularensis, is endemic to the northern hemisphere. This zoonotic organism has historically been developed into a biological weapon. For this Tier 1, Category A select agent, it is important to expand our understanding of its mechanisms of antibiotic resistance (AMR). Francisella is unlike many Gram-negative organisms in that it does not have significant plasmid mobility, and does not express AMR mechanisms on plasmids; thus plasmid-mediated resistance does not occur naturally. It is possible to artificially introduce plasmids with AMR markers for cloning and gene expression purposes. In this review, we survey both the experimental research on AMR in Francisella and bioinformatic databases which contain genomic and proteomic data. We explore both the genetic determinants of intrinsic AMR and naturally acquired or engineered antimicrobial resistance as well as phenotypic resistance in Francisella. Herein we survey resistance to beta-lactams, monobactams, carbapenems, aminoglycosides, tetracycline, polymyxins, macrolides, rifampin, fosmidomycin, and fluoroquinolones. We also highlight research about the phenotypic AMR difference between planktonic and biofilm Francisella. We discuss newly developed methods of testing antibiotics against Francisella which involve the intracellular nature of Francisella infection and may better reflect the eventual clinical outcomes for new antibiotic compounds. Understanding the genetically encoded determinants of AMR in Francisella is key to optimizing the treatment of patients and potentially developing new antimicrobials for this dangerous intracellular pathogen.
Collapse
Affiliation(s)
| | - Monique L. van Hoek
- School of Systems Biology, George Mason University, Manassas, VA, United States
| |
Collapse
|
25
|
Shao S, Wei L, Xia F, Zhang Y, Wang AQ. Defined Mutant Library Sequencing (DML-Seq) for Identification of Conditional Essential Genes. Bio Protoc 2021; 11:e3943. [PMID: 33796617 DOI: 10.21769/bioprotoc.3943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 01/12/2021] [Accepted: 01/16/2021] [Indexed: 11/02/2022] Open
Abstract
Transposon insertion sequencing (TIS) is an emerging technique which utilizes a massive transposon mutant library to screen specific phenotype and determine the conditional essential genetic requirements for bacterial fitness under distinct conditions combined with high-throughput parallel sequencing technology. Compared with a massive mutant library in traditional TIS, the defined mutant library sequencing (DML-Seq) has advantages as: 1) efficient mutagenesis; 2) low bottleneck effects; 3) avoid hotpots caused by screening; 4) can be directly used in the following experiments. Here, we described an optimized procedure of DML-Seq for fitness screen to supply classical TIS using the marine pathogenic bacterium Edwardsiella piscicida as an example.
Collapse
Affiliation(s)
- Shuai Shao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Lifan Wei
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China.,Department of Endodontics and Operative Dentistry, Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Xia
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Yuanxing Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China.,Shanghai Collaborative Innovation Center for Biomanufacturing Technology, 130 Meilong Road, Shanghai 200237, China.,Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai 200237, China
| | - And Qiyao Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China.,Shanghai Collaborative Innovation Center for Biomanufacturing Technology, 130 Meilong Road, Shanghai 200237, China.,Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai 200237, China
| |
Collapse
|
26
|
Sheshko V, Link M, Golovliov I, Balonova L, Stulik J. Utilization of a tetracycline-inducible system for high-level expression of recombinant proteins in Francisella tularensis LVS. Plasmid 2021; 115:102564. [PMID: 33610608 DOI: 10.1016/j.plasmid.2021.102564] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 01/14/2021] [Accepted: 01/27/2021] [Indexed: 12/28/2022]
Abstract
Francisella tularensis is a Gram-negative intracellular pathogen causing tularemia. A number of its potential virulence factors have been identified, but their biology and functions are not precisely known. Understanding the biological and immunological functions of these proteins requires adequate genetic tools for homologous and heterologous expression of cloned genes, maintaining both original structure and post-translational modifications. Here, we report the construction of a new multipurpose shuttle plasmid - pEVbr - which can be used for high-level expression in F. tularensis. The pEVbr plasmid has been constructed by modifying the TetR-regulated expression vector pEDL17 (LoVullo, 2012) that includes (i) a strong F. tularensis bfr promoter, and (ii) two tet operator sequences cloned into the promoter. The cloned green fluorescent protein (GFP), used as a reporter, demonstrated almost undetectable basal expression level under uninduced conditions and a highly dynamic dose-dependent response to the inducer. The utility of the system was further confirmed by cloning the gapA and FTT_1676 genes into the pEVbr vector and quantifying proteins expression in F. tularensis LVS, as well as by studying post-translational modification of the cloned genes. This study demonstrates that high levels of recombinant native-like Francisella proteins can be produced in Francisella cells. Hence, this system may be beneficial for the analysis of protein function and the development of new treatments and vaccines.
Collapse
Affiliation(s)
- Valeria Sheshko
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, Hradec Kralove, Czech Republic.
| | - Marek Link
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, Hradec Kralove, Czech Republic
| | - Igor Golovliov
- Department of Clinical Microbiology, Umeå University, SE-901 85 Umeå, Sweden
| | - Lucie Balonova
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, Hradec Kralove, Czech Republic
| | - Jiri Stulik
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence, Hradec Kralove, Czech Republic
| |
Collapse
|
27
|
Luo H, Lin Y, Liu T, Lai FL, Zhang CT, Gao F, Zhang R. DEG 15, an update of the Database of Essential Genes that includes built-in analysis tools. Nucleic Acids Res 2021; 49:D677-D686. [PMID: 33095861 PMCID: PMC7779065 DOI: 10.1093/nar/gkaa917] [Citation(s) in RCA: 123] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 09/30/2020] [Accepted: 10/06/2020] [Indexed: 12/21/2022] Open
Abstract
Essential genes refer to genes that are required by an organism to survive under specific conditions. Studies of the minimal-gene-set for bacteria have elucidated fundamental cellular processes that sustain life. The past five years have seen a significant progress in identifying human essential genes, primarily due to the successful use of CRISPR/Cas9 in various types of human cells. DEG 15, a new release of the Database of Essential Genes (www.essentialgene.org), has provided major advancements, compared to DEG 10. Specifically, the number of eukaryotic essential genes has increased by more than fourfold, and that of prokaryotic ones has more than doubled. Of note, the human essential-gene number has increased by more than tenfold. Moreover, we have developed built-in analysis modules by which users can perform various analyses, such as essential-gene distributions between bacterial leading and lagging strands, sub-cellular localization distribution, enrichment analysis of gene ontology and KEGG pathways, and generation of Venn diagrams to compare and contrast gene sets between experiments. Additionally, the database offers customizable BLAST tools for performing species- and experiment-specific BLAST searches. Therefore, DEG comprehensively harbors updated human-curated essential-gene records among prokaryotes and eukaryotes with built-in tools to enhance essential-gene analysis.
Collapse
Affiliation(s)
- Hao Luo
- Department of Physics, School of Science, Tianjin University, Tianjin 300072, China
| | - Yan Lin
- Department of Physics, School of Science, Tianjin University, Tianjin 300072, China
| | - Tao Liu
- Department of Physics, School of Science, Tianjin University, Tianjin 300072, China
| | - Fei-Liao Lai
- Department of Physics, School of Science, Tianjin University, Tianjin 300072, China
| | - Chun-Ting Zhang
- Department of Physics, School of Science, Tianjin University, Tianjin 300072, China
| | - Feng Gao
- Department of Physics, School of Science, Tianjin University, Tianjin 300072, China.,Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China
| | - Ren Zhang
- Center for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, MI 48201, USA
| |
Collapse
|
28
|
The Sensor Kinase QseC Regulates the Unlinked PmrA Response Regulator and Downstream Gene Expression in Francisella. J Bacteriol 2020; 202:JB.00321-20. [PMID: 32839173 DOI: 10.1128/jb.00321-20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 08/13/2020] [Indexed: 12/22/2022] Open
Abstract
The facultative intracellular bacterial pathogen Francisella tularensis is the causative agent of tularemia in humans and animals. Gram-negative bacteria utilize two-component regulatory systems (TCS) to sense and respond to their changing environment. No classical, tandemly arranged sensor kinase and response regulator TCS genes exist in the human virulent Francisella tularensis subsp. tularensis, but orphaned members are present. PmrA is an orphan response regulator responsible for intramacrophage growth and virulence; however, the regulation of PmrA activity is not understood. We and others have shown that PmrA represses the expression of priM, described to encode an antivirulence determinant. By screening a mutant library for increased priM promoter activity, we identified the sensor kinase homolog QseC as an upstream regulator of priM expression, and this regulation is in part dependent upon the aspartate phosphorylation site of PmrA (D51). Several examined environmental signals, including epinephrine, which is reported to activate QseC in other bacteria, do not affect priM expression in a manner dependent on PmrA. Intramacrophage survival assays also question the finding that PriM is an antivirulence factor. Thus, these data suggest that the PmrA-regulated gene priM is modulated by the QseC-PmrA (QseB) TCS in Francisella IMPORTANCE The disease tularemia is caused by the highly infectious Gram-negative pathogen Francisella tularensis This bacterium encodes few regulatory factors (e.g., two-component systems [TCS]). PmrA, required for intramacrophage survival and virulence in the mouse model, is encoded by an orphan TCS response regulator gene. It is unclear how PmrA is responsive to environmental signals to regulate loci, including the PmrA-repressed gene priM We identify an orphan sensor kinase (QseC) that is required for priM repression and further explore both environmental signals that might regulate the QseC-PmrA TCS and the function of PriM.
Collapse
|
29
|
Zhang Y, Chen J, Wang Y, Li Y, Rui W, Zhang J, Luo D. Expression and protease characterization of a conserved protein YgjD in Vibrio harveyi. PeerJ 2020; 8:e9061. [PMID: 32477834 PMCID: PMC7241418 DOI: 10.7717/peerj.9061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 04/04/2020] [Indexed: 11/20/2022] Open
Abstract
The glycopeptidase GCP and its homologue proteins are conserved and essential for survival of bacteria. The ygjD gene (Glycopeptidase homologue) was cloned from Vibrio harveyi strain SF-1. The gene consisted of 1,017 bp, which encodes a 338 amino acid polypeptide. The nucleotide sequence similarity of the ygjD gene with that of V. harveyi FDAARGOS 107 was 95%. The ygjD gene also showed similarities of 68%, 67% and 50% with those of Salmonella enterica, Escherichia coli and Bacillus cereus. The ygjD gene was expressed in E. coli BL21 (DE3) and the recombinant YgjD was purified by Ni2+ affinity chromatography column. The purified YgjD showed a specific 37 kDa band on sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and exhibited protease activities of 59,000 units/mg, 53,700 units/mg and 8,100 units/mg, respectively, on N-Acetyl-L-tyrosine ethyl ester monohydrate (ATEE), N-Benzoyl-L-tyrosine ethyl ester (BTEE) and N-Benzoyl-DL-arginine-4-nitroanilide hydrochloride (BAPNA) substrates. When the conserved amino acids of His111, Glu113 and His115 in the YgjD were replaced with alanine, respectively, the protease activities of the mutants were partly decreased. The two conserved His111 and His115 of YgjD were mutated and the protein lost the protease activity, which implied that the two amino acid played very important roles in maintaining its protease activity. The addition of the purified YgjD to the culture medium of V. harveyi strain SF-1 can effectively promote the bacteria growth. These results indicated that the protease activities may be involved in the survival of bacteria.
Collapse
Affiliation(s)
- Yayuan Zhang
- School of Petrochemical Engineering, Lanzhou University of Technology, Lanzhou, China
| | - Jixiang Chen
- School of Petrochemical Engineering, Lanzhou University of Technology, Lanzhou, China
| | - Yonggang Wang
- School of life science and enginerring, Lanzhou University of Technology, Lanzhou, China
| | - Yanlin Li
- Chongqing Key Laboratory of Environmental Materials & Remediation Technologies, Chongqing University of Arts and Sciences, Chongqing, China
| | - Wenhong Rui
- School of Petrochemical Engineering, Lanzhou University of Technology, Lanzhou, China
| | - Jiyi Zhang
- School of Petrochemical Engineering, Lanzhou University of Technology, Lanzhou, China
| | - Dan Luo
- School of Petrochemical Engineering, Lanzhou University of Technology, Lanzhou, China
| |
Collapse
|
30
|
Li X, Li W, Zeng M, Zheng R, Li M. Network-based methods for predicting essential genes or proteins: a survey. Brief Bioinform 2020; 21:566-583. [PMID: 30776072 DOI: 10.1093/bib/bbz017] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 01/21/2019] [Accepted: 01/22/2019] [Indexed: 01/03/2025] Open
Abstract
Genes that are thought to be critical for the survival of organisms or cells are called essential genes. The prediction of essential genes and their products (essential proteins) is of great value in exploring the mechanism of complex diseases, the study of the minimal required genome for living cells and the development of new drug targets. As laboratory methods are often complicated, costly and time-consuming, a great many of computational methods have been proposed to identify essential genes/proteins from the perspective of the network level with the in-depth understanding of network biology and the rapid development of biotechnologies. Through analyzing the topological characteristics of essential genes/proteins in protein-protein interaction networks (PINs), integrating biological information and considering the dynamic features of PINs, network-based methods have been proved to be effective in the identification of essential genes/proteins. In this paper, we survey the advanced methods for network-based prediction of essential genes/proteins and present the challenges and directions for future research.
Collapse
Affiliation(s)
- Xingyi Li
- School of Computer Science and Engineering, Central South University, Changsha, Hunan, China
| | - Wenkai Li
- School of Computer Science and Engineering, Central South University, Changsha, Hunan, China
| | - Min Zeng
- School of Computer Science and Engineering, Central South University, Changsha, Hunan, China
| | - Ruiqing Zheng
- School of Computer Science and Engineering, Central South University, Changsha, Hunan, China
| | - Min Li
- School of Computer Science and Engineering, Central South University, Changsha, Hunan, China
| |
Collapse
|
31
|
Dean SN, Milton ME, Cavanagh J, van Hoek ML. Francisella novicida Two-Component System Response Regulator BfpR Modulates iglC Gene Expression, Antimicrobial Peptide Resistance, and Biofilm Production. Front Cell Infect Microbiol 2020; 10:82. [PMID: 32232010 PMCID: PMC7082314 DOI: 10.3389/fcimb.2020.00082] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 02/18/2020] [Indexed: 12/20/2022] Open
Abstract
Response regulators are a critical part of the two-component system of gene expression regulation in bacteria, transferring a signal from a sensor kinase into DNA binding activity resulting in alteration of gene expression. In this study, we investigated a previously uncharacterized response regulator in Francisella novicida, FTN_1452 that we have named BfpR (Biofilm-regulating Francisella protein Regulator, FTN_1452). In contrast to another Francisella response regulator, QseB/PmrA, BfpR appears to be a negative regulator of biofilm production, and also a positive regulator of antimicrobial peptide resistance in this bacterium. The protein was crystallized and X-ray crystallography studies produced a 1.8 Å structure of the BfpR N-terminal receiver domain revealing interesting insight into its potential interaction with the sensor kinase. Structural analysis of BfpR places it in the OmpR/PhoP family of bacterial response regulators along with WalR and ResD. Proteomic and transcriptomic analyses suggest that BfpR overexpression affects expression of the critical Francisella virulence factor iglC, as well as other proteins in the bacterium. We demonstrate that mutation of bfpR is associated with an antimicrobial peptide resistance phenotype, a phenotype also associated with other response regulators, for the human cathelicidin peptide LL-37 and a sheep antimicrobial peptide SMAP-29. F. novicida with mutated bfpR replicated better than WT in intracellular infection assays in human-derived macrophages suggesting that the down-regulation of iglC expression in bfpR mutant may enable this intracellular replication to occur. Response regulators have been shown to play important roles in the regulation of bacterial biofilm production. We demonstrate that F. novicida biofilm formation was highly increased in the bfpR mutant, corresponding to altered glycogen synthesis. Waxworm infection experiments suggest a role of BfpR as a negative modulator of iglC expression with de-repression by Mg2+. In this study, we find that the response regulator BfpR may be a negative regulator of biofilm formation, and a positive regulator of antimicrobial peptide resistance in F. novicida.
Collapse
Affiliation(s)
- Scott N Dean
- National Center for Biodefense and Infectious Diseases, and School of Systems Biology, George Mason University, Manassas, VA, United States
| | - Morgan E Milton
- Department of Biochemistry and Molecular Biology, The Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - John Cavanagh
- Department of Biochemistry and Molecular Biology, The Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Monique L van Hoek
- National Center for Biodefense and Infectious Diseases, and School of Systems Biology, George Mason University, Manassas, VA, United States
| |
Collapse
|
32
|
May HC, Yu JJ, Shrihari S, Seshu J, Klose KE, Cap AP, Chambers JP, Guentzel MN, Arulanandam BP. Thioredoxin Modulates Cell Surface Hydrophobicity in Acinetobacter baumannii. Front Microbiol 2019; 10:2849. [PMID: 31921031 PMCID: PMC6927278 DOI: 10.3389/fmicb.2019.02849] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 11/25/2019] [Indexed: 12/04/2022] Open
Abstract
Acinetobacter baumannii, a Gram-negative coccobacillus, has become a prevalent nosocomial health threat affecting the majority of hospitals both in the U.S. and around the globe. Microbial cell surface hydrophobicity (CSH) has previously been correlated with virulence, uptake by immune cells, and attachment to epithelial cells. A mutant strain of A. baumannii (ΔtrxA) lacking the redox protein thioredoxin A was found to be more hydrophobic than its wild type (WT) and complemented counterparts, as measured by both Microbial Adhesion to Hydrocarbon (MATH) and salt aggregation. The hydrophobicity of the mutant could be abrogated through treatment with sodium cyanoborohydride (SCBH). This modulation correlated with reduction of disulfide bonds, as SCBH was able to reduce 5,5′-dithio-bis-[2-nitrobenzoic acid] and treatment with the known disulfide reducer, β-mercaptoethanol, also decreased ΔtrxA CSH. Additionally, the ΔtrxA mutant was more readily taken up than WT by J774 macrophages and this differential uptake could be abrogated though SCBH treatment. When partitioned into aqueous and hydrophobic phases, ΔtrxA recovered from the hydrophobic partition was phagocytosed more readily than from the aqueous phase further supporting the contribution of CSH to A. baumannii uptake by phagocytes. A second Gram-negative bacterium, Francisella novicida, also showed the association of TrxA deficiency (FnΔtrxA) with increased hydrophobicity and uptake by J774 cells. We previously have demonstrated that modification of the type IV pilus system (T4P) was associated with the A. baumannii ΔtrxA phenotype, and the Francisella FnΔtrxA mutant also was found to have a marked T4P deficiency. Interestingly, a F. novicida mutant lacking pilT also showed increased hydrophobicity over FnWT. Collective evidence presented in this study suggests that Gram-negative bacterial thioredoxin mediates CSH through multiple mechanisms including disulfide-bond reduction and T4P modulation.
Collapse
Affiliation(s)
- Holly C May
- Department of Biology, South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, San Antonio, TX, United States
| | - Jieh-Juen Yu
- Department of Biology, South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, San Antonio, TX, United States
| | - Swathi Shrihari
- Department of Biology, South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, San Antonio, TX, United States
| | - Janakiram Seshu
- Department of Biology, South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, San Antonio, TX, United States
| | - Karl E Klose
- Department of Biology, South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, San Antonio, TX, United States
| | - Andrew P Cap
- Acute Combat Casualty Care Research Division, U.S. Army Institute for of Surgical Research, JBSA-Fort Sam Houston, San Antonio, TX, United States
| | - James P Chambers
- Department of Biology, South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, San Antonio, TX, United States
| | - M Neal Guentzel
- Department of Biology, South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, San Antonio, TX, United States
| | - Bernard P Arulanandam
- Department of Biology, South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, San Antonio, TX, United States
| |
Collapse
|
33
|
Wei L, Qiao H, Sit B, Yin K, Yang G, Ma R, Ma J, Yang C, Yao J, Ma Y, Xiao J, Liu X, Zhang Y, Waldor MK, Wang Q. A Bacterial Pathogen Senses Host Mannose to Coordinate Virulence. iScience 2019; 20:310-323. [PMID: 31605945 PMCID: PMC6817725 DOI: 10.1016/j.isci.2019.09.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 08/19/2019] [Accepted: 09/20/2019] [Indexed: 11/21/2022] Open
Abstract
Bacterial pathogens are thought to activate expression of virulence genes upon detection of host-associated cues, but identification of the nature of such signals has proved difficult. We generated a genome-scale defined transposon mutant library in Edwardsiella piscicida, an important fish pathogen, to quantify the fitness of insertion mutants for intracellular growth in macrophages and in turbot (Scophthalmus maximus). These screens identified EvrA, a transcription activator that induces expression of esrB, a key virulence regulator. EvrA is directly bound and activated by mannose-6-phosphate (man-6P) derived from actively imported mannose. Mutants lacking EvrA or expressing an EvrA unable to bind man-6P were similarly attenuated in turbot. Exogenously added mannose promoted E. piscicida virulence, and high levels of mannose were detected in fish tissue. Together, these observations reveal that binding of a host-derived sugar to a transcription factor can facilitate pathogen sensing of the host environment and trigger virulence programs.
Collapse
Affiliation(s)
- Lifan Wei
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Haoxian Qiao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Brandon Sit
- Division of Infectious Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Kaiyu Yin
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Guanhua Yang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Ruiqing Ma
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Jiabao Ma
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Chun Yang
- State Key Laboratory of Genetic Engineering, Department of Physiology and Biophysics, School of Life Sciences, Fudan University, Shanghai, 200433, China
| | - Jun Yao
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433, China
| | - Yue Ma
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China; Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai 200237, China
| | - Jingfan Xiao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Xiaohong Liu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China; Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai 200237, China
| | - Yuanxing Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China; Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai 200237, China
| | - Matthew K Waldor
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China; Division of Infectious Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Boston, MA 02115, USA
| | - Qiyao Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, China; Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai 200237, China.
| |
Collapse
|
34
|
Kong X, Zhu B, Stone VN, Ge X, El-Rami FE, Donghai H, Xu P. ePath: an online database towards comprehensive essential gene annotation for prokaryotes. Sci Rep 2019; 9:12949. [PMID: 31506471 PMCID: PMC6737131 DOI: 10.1038/s41598-019-49098-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 08/15/2019] [Indexed: 02/01/2023] Open
Abstract
Experimental techniques for identification of essential genes (EGs) in prokaryotes are usually expensive, time-consuming and sometimes unrealistic. Emerging in silico methods provide alternative methods for EG prediction, but often possess limitations including heavy computational requirements and lack of biological explanation. Here we propose a new computational algorithm for EG prediction in prokaryotes with an online database (ePath) for quick access to the EG prediction results of over 4,000 prokaryotes ( https://www.pubapps.vcu.edu/epath/ ). In ePath, gene essentiality is linked to biological functions annotated by KEGG Ortholog (KO). Two new scoring systems, namely, E_score and P_score, are proposed for each KO as the EG evaluation criteria. E_score represents appearance and essentiality of a given KO in existing experimental results of gene essentiality, while P_score denotes gene essentiality based on the principle that a gene is essential if it plays a role in genetic information processing, cell envelope maintenance or energy production. The new EG prediction algorithm shows prediction accuracy ranging from 75% to 91% based on validation from five new experimental studies on EG identification. Our overall goal with ePath is to provide a comprehensive and reliable reference for gene essentiality annotation, facilitating the study of those prokaryotes without experimentally derived gene essentiality information.
Collapse
Affiliation(s)
- Xiangzhen Kong
- Philips Institute for Oral Health Research, Virginia Commonwealth University, Richmond, Virginia, 23298, United States of America
| | - Bin Zhu
- Philips Institute for Oral Health Research, Virginia Commonwealth University, Richmond, Virginia, 23298, United States of America
| | - Victoria N Stone
- Philips Institute for Oral Health Research, Virginia Commonwealth University, Richmond, Virginia, 23298, United States of America
| | - Xiuchun Ge
- Philips Institute for Oral Health Research, Virginia Commonwealth University, Richmond, Virginia, 23298, United States of America
| | - Fadi E El-Rami
- Philips Institute for Oral Health Research, Virginia Commonwealth University, Richmond, Virginia, 23298, United States of America
| | - Huangfu Donghai
- Application Services, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Ping Xu
- Philips Institute for Oral Health Research, Virginia Commonwealth University, Richmond, Virginia, 23298, United States of America.
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, Virginia, United States of America.
- Center for Biological Data Science, Virginia Commonwealth University, Richmond, Virginia, United States of America.
| |
Collapse
|
35
|
Wei L, Qiao H, Liu B, Yin K, Liu Q, Zhang Y, Ma Y, Wang Q. MarTrack: A versatile toolbox of mariner transposon derivatives used for functional genetic analysis of bacterial genomes. Microbiol Res 2019; 228:126306. [PMID: 31422233 DOI: 10.1016/j.micres.2019.126306] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 07/16/2019] [Accepted: 08/02/2019] [Indexed: 11/24/2022]
Abstract
The mariner transposon family of Himar1 has been widely used for the random mutagenesis of bacteria to generate single insertions into the chromosome. Here, a versatile toolbox of mariner transposon derivatives was generated and applied to the functional genomics investigation of fish pathogen Edwardsiella piscicida. In this study, we combined the merits of the random mutagenesis of mariner transposon and common efficient reporter marker genes or regulatory elements, mCherry, gfp, luxAB, lacZ, sacBR, and PBAD and antibiotic resistance cassettes to construct a series of derivative transposon vectors, pMmch, pMKGR, pMCGR, pMXKGR, pMLKGR, pMSGR, and pMPR, based on the initial transposon pMar2xT7. The function and effectiveness of the modified transposons were verified by introducing them into E. piscicida EIB202. Based on the toolbox, a transposon insertion mutant library containing approximately 3.0 × 105 distinct mutants was constructed to explore the upstream regulators of esrB, the master regulator of the type III and type VI secretion systems (T3/T6SS) in E. piscicida. Following analysis by Con-ARTIST, ETAE_3474, annotated as fabR and involved in fatty acid metabolism, was screened out and identified as a novel regulator mediating T3SS and T6SS expression. In addition, the fabR mutants displayed critical virulence attenuation in turbot. Due to the broad-range host compatibility of mariner transposons, the newly built transposon toolbox can be applied for functional genomics studies in various bacteria.
Collapse
Affiliation(s)
- Lifan Wei
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, PR China
| | - Haoxian Qiao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, PR China
| | - Bing Liu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, PR China
| | - Kaiyu Yin
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, PR China; Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai, PR China
| | - Qin Liu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, PR China; Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai, PR China
| | - Yuanxing Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, PR China; Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai, PR China
| | - Yue Ma
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, PR China; Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai, PR China.
| | - Qiyao Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, PR China; Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai, PR China.
| |
Collapse
|
36
|
Lin Y, Zhang FZ, Xue K, Gao YZ, Guo FB. Identifying Bacterial Essential Genes Based on a Feature-Integrated Method. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2019; 16:1274-1279. [PMID: 28212095 DOI: 10.1109/tcbb.2017.2669968] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Essential genes are those genes of an organism that are considered to be crucial for its survival. Identification of essential genes is therefore of great significance to advance our understanding of the principles of cellular life. We have developed a novel computational method, which can effectively predict bacterial essential genes by extracting and integrating homologous features, protein domain feature, gene intrinsic features, and network topological features. By performing the principal component regression (PCR) analysis for Escherichia coli MG1655, we established a classification model with the average area under curve (AUC) value of 0.992 in ten times 5-fold cross-validation tests. Furthermore, when employing this new model to a distantly related organism-Streptococcus pneumoniae TIGR4, we still got a reliable AUC value of 0.788. These results indicate that our feature-integrated approach could have practical applications in accurately investigating essential genes from broad bacterial species, and also provide helpful guidelines for the minimal cell.
Collapse
|
37
|
The Lipid A 1-Phosphatase, LpxE, Functionally Connects Multiple Layers of Bacterial Envelope Biogenesis. mBio 2019; 10:mBio.00886-19. [PMID: 31213552 PMCID: PMC6581854 DOI: 10.1128/mbio.00886-19] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Dephosphorylation of the lipid A 1-phosphate by LpxE in Gram-negative bacteria plays important roles in antibiotic resistance, bacterial virulence, and modulation of the host immune system. Our results demonstrate that in addition to removing the 1-phosphate from lipid A, LpxEs also dephosphorylate undecaprenyl pyrophosphate, an important metabolite for the synthesis of the essential envelope components, peptidoglycan and O-antigen. Therefore, LpxEs participate in multiple layers of biogenesis of the Gram-negative bacterial envelope and increase antibiotic resistance. This discovery marks an important step toward understanding the regulation and biogenesis of the Gram-negative bacterial envelope. Although distinct lipid phosphatases are thought to be required for processing lipid A (component of the outer leaflet of the outer membrane), glycerophospholipid (component of the inner membrane and the inner leaflet of the outer membrane), and undecaprenyl pyrophosphate (C55-PP; precursors of peptidoglycan and O antigens of lipopolysaccharide) in Gram-negative bacteria, we report that the lipid A 1-phosphatases, LpxEs, functionally connect multiple layers of cell envelope biogenesis in Gram-negative bacteria. We found that Aquifex aeolicus LpxE structurally resembles YodM in Bacillus subtilis, a phosphatase for phosphatidylglycerol phosphate (PGP) with a weak in vitro activity on C55-PP, and rescues Escherichia coli deficient in PGP and C55-PP phosphatase activities; deletion of lpxE in Francisella novicida reduces the MIC value of bacitracin, indicating a significant contribution of LpxE to the native bacterial C55-PP phosphatase activity. Suppression of plasmid-borne lpxE in F. novicida deficient in chromosomally encoded C55-PP phosphatase activities results in cell enlargement, loss of O-antigen repeats of lipopolysaccharide, and ultimately cell death. These discoveries implicate LpxE as the first example of a multifunctional regulatory enzyme that orchestrates lipid A modification, O-antigen production, and peptidoglycan biogenesis to remodel multiple layers of the Gram-negative bacterial envelope.
Collapse
|
38
|
Bachert BA, Biryukov SS, Chua J, Rodriguez SA, Toothman RG, Cote CK, Klimko CP, Hunter M, Shoe JL, Williams JA, Kuehl KA, Biot FV, Bozue JA. A Francisella novicida Mutant, Lacking the Soluble Lytic Transglycosylase Slt, Exhibits Defects in Both Growth and Virulence. Front Microbiol 2019; 10:1343. [PMID: 31258523 PMCID: PMC6587636 DOI: 10.3389/fmicb.2019.01343] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 05/29/2019] [Indexed: 01/28/2023] Open
Abstract
Francisella tularensis is the causative agent of tularemia and has gained recent interest as it poses a significant biothreat risk. F. novicida is commonly used as a laboratory surrogate for tularemia research due to genetic similarity and susceptibility of mice to infection. Currently, there is no FDA-approved tularemia vaccine, and identifying therapeutic targets remains a critical gap in strategies for combating this pathogen. Here, we investigate the soluble lytic transglycosylase or Slt in F. novicida, which belongs to a class of peptidoglycan-modifying enzymes known to be involved in cell division. We assess the role of Slt in biology and virulence of the organism as well as the vaccine potential of the slt mutant. We show that the F. novicida slt mutant has a significant growth defect in acidic pH conditions. Further microscopic analysis revealed significantly altered cell morphology compared to wild-type, including larger cell size, extensive membrane protrusions, and cell clumping and fusion, which was partially restored by growth in neutral pH or genetic complementation. Viability of the mutant was also significantly decreased during growth in acidic medium, but not at neutral pH. Furthermore, the slt mutant exhibited significant attenuation in a murine model of intranasal infection and virulence could be restored by genetic complementation. Moreover, we could protect mice using the slt mutant as a live vaccine strain against challenge with the parent strain; however, we were not able to protect against challenge with the fully virulent F. tularensis Schu S4 strain. These studies demonstrate a critical role for the Slt enzyme in maintaining proper cell division and morphology in acidic conditions, as well as replication and virulence in vivo. Our results suggest that although the current vaccination strategy with F. novicida slt mutant would not protect against Schu S4 challenges, the Slt enzyme could be an ideal target for future therapeutic development.
Collapse
Affiliation(s)
- Beth A Bachert
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, United States
| | - Sergei S Biryukov
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, United States
| | - Jennifer Chua
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, United States
| | - Sabrina A Rodriguez
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, United States
| | - Ronald G Toothman
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, United States
| | - Christopher K Cote
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, United States
| | - Christopher P Klimko
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, United States
| | - Melissa Hunter
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, United States
| | - Jennifer L Shoe
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, United States
| | - Janice A Williams
- Pathology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, United States
| | - Kathleen A Kuehl
- Pathology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, United States
| | - Fabrice V Biot
- Unité de Bactériologie/UMR_MD1, Département de Biologie des Agents Transmissibles, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France
| | - Joel A Bozue
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, United States
| |
Collapse
|
39
|
Global Analysis of Genes Essential for Francisella tularensis Schu S4 Growth In Vitro and for Fitness during Competitive Infection of Fischer 344 Rats. J Bacteriol 2019; 201:JB.00630-18. [PMID: 30642993 PMCID: PMC6416918 DOI: 10.1128/jb.00630-18] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 01/02/2019] [Indexed: 01/02/2023] Open
Abstract
The highly virulent intracellular pathogen Francisella tularensis is a Gram-negative bacterium that has a wide host range, including humans, and is the causative agent of tularemia. To identify new therapeutic drug targets and vaccine candidates and investigate the genetic basis of Francisella virulence in the Fischer 344 rat, we have constructed an F. tularensis Schu S4 transposon library. This library consists of more than 300,000 unique transposon mutants and represents a transposon insertion for every 6 bp of the genome. A transposon-directed insertion site sequencing (TraDIS) approach was used to identify 453 genes essential for growth in vitro Many of these essential genes were mapped to key metabolic pathways, including glycolysis/gluconeogenesis, peptidoglycan synthesis, fatty acid biosynthesis, and the tricarboxylic acid (TCA) cycle. Additionally, 163 genes were identified as required for fitness during colonization of the Fischer 344 rat spleen. This in vivo selection screen was validated through the generation of marked deletion mutants that were individually assessed within a competitive index study against the wild-type F. tularensis Schu S4 strain.IMPORTANCE The intracellular bacterial pathogen Francisella tularensis causes a disease in humans characterized by the rapid onset of nonspecific symptoms such as swollen lymph glands, fever, and headaches. F. tularensis is one of the most infectious bacteria known and following pulmonary exposure can have a mortality rate exceeding 50% if left untreated. The low infectious dose of this organism and concerns surrounding its potential as a biological weapon have heightened the need for effective and safe therapies. To expand the repertoire of targets for therapeutic development, we initiated a genome-wide analysis. This study has identified genes that are important for F. tularensis under in vitro and in vivo conditions, providing candidates that can be evaluated for vaccine or antibacterial development.
Collapse
|
40
|
Wei L, Qiao H, Liu B, Yin K, Liu Q, Zhang Y, Ma Y, Wang Q. MarTrack: A versatile toolbox of mariner transposon derivatives used for functional genetic analysis of bacterial genomes. Microbiol Res 2019; 219:84-93. [PMID: 30642470 DOI: 10.1016/j.micres.2018.11.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 11/22/2018] [Accepted: 11/23/2018] [Indexed: 11/28/2022]
Abstract
The mariner transposon family of Himar1 has been widely used for the random mutagenesis of bacteria to generate single insertions into the chromosome. Here, a versatile toolbox of mariner transposon derivatives was generated and applied to the functional genomics investigation of fish pathogen Edwardsiella piscicida. In this study, we combined the merits of the random mutagenesis of mariner transposon and common efficient reporter marker genes or regulatory elements, mcherry, gfp, luxAB, lacZ, sacBR, and PBAD and antibiotic resistance cassettes to construct a series of derivative transposon vectors, pMmch, pMKGR, pMCGR, pMXKGR, pMLKGR, pMSGR, and pMPR, based on the initial transposon pMar2xT7. The function and effectiveness of the modified transposons were verified by introducing them into E. piscicida EIB202. Based on the toolbox, a transposon insertion mutant library containing approximately 3.0 × 105 separated mutants was constructed to explore the upstream regulators of esrB, the master regulator of the type III and type VI secretion systems (T3/T6SS) in E. piscicida. Following analysis by Con-ARTIST, ETAE_2184 (renamed as EsrR) was screened out and identified as a novel regulator mediating T3SS expression. In addition, the esrR mutants displayed critical virulence attenuation. Due to the broad-range host compatibility of mariner transposons, the newly built transposon toolbox can be broadly applied for functional genomics studies in various bacteria.
Collapse
Affiliation(s)
- Lifan Wei
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, PR China
| | - Haoxian Qiao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, PR China
| | - Bing Liu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, PR China
| | - Kaiyu Yin
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, PR China; Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai, PR China
| | - Qin Liu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, PR China; Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai, PR China
| | - Yuanxing Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, PR China; Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai, PR China
| | - Yue Ma
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, PR China; Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai, PR China.
| | - Qiyao Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, PR China; Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai, PR China.
| |
Collapse
|
41
|
Ijaq J, Malik G, Kumar A, Das PS, Meena N, Bethi N, Sundararajan VS, Suravajhala P. A model to predict the function of hypothetical proteins through a nine-point classification scoring schema. BMC Bioinformatics 2019; 20:14. [PMID: 30621574 PMCID: PMC6325861 DOI: 10.1186/s12859-018-2554-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 11/30/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Hypothetical proteins [HP] are those that are predicted to be expressed in an organism, but no evidence of their existence is known. In the recent past, annotation and curation efforts have helped overcome the challenge in understanding their diverse functions. Techniques to decipher sequence-structure-function relationship, especially in terms of functional modelling of the HPs have been developed by researchers, but using the features as classifiers for HPs has not been attempted. With the rise in number of annotation strategies, next-generation sequencing methods have provided further understanding the functions of HPs. RESULTS In our previous work, we developed a six-point classification scoring schema with annotation pertaining to protein family scores, orthology, protein interaction/association studies, bidirectional best BLAST hits, sorting signals, known databases and visualizers which were used to validate protein interactions. In this study, we introduced three more classifiers to our annotation system, viz. pseudogenes linked to HPs, homology modelling and non-coding RNAs associated to HPs. We discuss the challenges and performance of these classifiers using machine learning heuristics with an improved accuracy from Perceptron (81.08 to 97.67), Naive Bayes (54.05 to 96.67), Decision tree J48 (67.57 to 97.00), and SMO_npolyk (59.46 to 96.67). CONCLUSION With the introduction of three new classification features, the performance of the nine-point classification scoring schema has an improved accuracy to functionally annotate the HPs.
Collapse
Affiliation(s)
- Johny Ijaq
- Department of Biotechnology, Osmania University, Hyderabad, 500007 India
- Bioclues.org, Kukatpally, Hyderabad, 500072 India
| | - Girik Malik
- Department of Pediatrics, The Battelle Center for Mathematical Medicine, The Research Institute at Nationwide Children’s Hospital, The Ohio State University, Columbus, OH USA
- Bioclues.org, Kukatpally, Hyderabad, 500072 India
- Labrynthe, New Delhi, India
| | - Anuj Kumar
- Bioclues.org, Kukatpally, Hyderabad, 500072 India
- Advanced Center for Computational and Applied Biotechnology, Uttarakhand Council for Biotechnology, Dehradun, 248007 India
| | - Partha Sarathi Das
- Bioclues.org, Kukatpally, Hyderabad, 500072 India
- Department of Microbiology, Bioinformatics Infrastructure Facility, Vidyasagar University, Midnapore, India
| | - Narendra Meena
- Department of Biotechnology and Bioinformatics, Birla Institute of Scientific Research, Statue Circle, RJ 302001 India
| | - Neeraja Bethi
- Department of Biotechnology, Osmania University, Hyderabad, 500007 India
| | | | - Prashanth Suravajhala
- Bioclues.org, Kukatpally, Hyderabad, 500072 India
- Department of Biotechnology and Bioinformatics, Birla Institute of Scientific Research, Statue Circle, RJ 302001 India
| |
Collapse
|
42
|
Metabolic models and gene essentiality data reveal essential and conserved metabolism in prokaryotes. PLoS Comput Biol 2018; 14:e1006556. [PMID: 30444863 PMCID: PMC6283598 DOI: 10.1371/journal.pcbi.1006556] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 12/06/2018] [Accepted: 10/09/2018] [Indexed: 01/13/2023] Open
Abstract
Essential metabolic reactions are shaping constituents of metabolic networks, enabling viable and distinct phenotypes across diverse life forms. Here we analyse and compare modelling predictions of essential metabolic functions with experimental data and thereby identify core metabolic pathways in prokaryotes. Simulations of 15 manually curated genome-scale metabolic models were integrated with 36 large-scale gene essentiality datasets encompassing a wide variety of species of bacteria and archaea. Conservation of metabolic genes was estimated by analysing 79 representative genomes from all the branches of the prokaryotic tree of life. We find that essentiality patterns reflect phylogenetic relations both for modelling and experimental data, which correlate highly at the pathway level. Genes that are essential for several species tend to be highly conserved as opposed to non-essential genes which may be conserved or not. The tRNA-charging module is highlighted as ancestral and with high centrality in the networks, followed closely by cofactor metabolism, pointing to an early information processing system supplied by organic cofactors. The results, which point to model improvements and also indicate faults in the experimental data, should be relevant to the study of centrality in metabolic networks and ancient metabolism but also to metabolic engineering with prokaryotes. If we tried to list every known chemical reaction within an organism–human, plant or even bacteria–we would get quite a long and confusing read. But when this information is represented in so-called genome-scale metabolic networks, we have the means to access computationally each of those reactions and their interconnections. Some parts of the network have alternatives, while others are unique and therefore can be essential for growth. Here, we simulate growth and compare essential reactions and genes for the simplest type of unicellular species–prokaryotes–to understand which parts of their metabolism are universally essential and potentially ancestral. We show that similar patterns of essential reactions echo phylogenetic relationships (this makes sense, as the genome provides the building plan for the enzymes that perform those reactions). Our computational predictions correlate strongly with experimental essentiality data. Finally, we show that a crucial step of protein synthesis (tRNA charging) and the synthesis and transformation of small molecules that enzymes require (cofactors) are the most essential and conserved parts of metabolism in prokaryotes. Our results are a step further in understanding the biology and evolution of prokaryotes but can also be relevant in applied studies including metabolic engineering and antibiotic design.
Collapse
|
43
|
Alam A, Golovliov I, Javed E, Sjöstedt A. ClpB mutants of Francisella tularensis subspecies holarctica and tularensis are defective for type VI secretion and intracellular replication. Sci Rep 2018; 8:11324. [PMID: 30054549 PMCID: PMC6063899 DOI: 10.1038/s41598-018-29745-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 07/17/2018] [Indexed: 12/13/2022] Open
Abstract
Francisella tularensis, a highly infectious, intracellular bacterium possesses an atypical type VI secretion system (T6SS), which is essential for the virulence of the bacterium. Recent data suggest that the HSP100 family member, ClpB, is involved in T6SS disassembly in the subspecies Francisella novicida. Here, we investigated the role of ClpB for the function of the T6SS and for phenotypic characteristics of the human pathogenic subspecies holarctica and tularensis. The ∆clpB mutants of the human live vaccine strain, LVS, belonging to subspecies holarctica, and the highly virulent SCHU S4 strain, belonging to subspecies tularensis, both showed extreme susceptibility to heat shock and low pH, severely impaired type VI secretion (T6S), and significant, but impaired intracellular replication compared to the wild-type strains. Moreover, they showed essentially intact phagosomal escape. Infection of mice demonstrated that both ΔclpB mutants were highly attenuated, but the SCHU S4 mutant showed more effective replication than the LVS strain. Collectively, our data demonstrate that ClpB performs multiple functions in the F. tularensis subspecies holarctica and tularensis and its function is important for T6S, intracellular replication, and virulence.
Collapse
Affiliation(s)
- Athar Alam
- Department of Clinical Microbiology, Umeå University, SE-901 85, Umeå, Sweden
| | - Igor Golovliov
- Department of Clinical Microbiology, Umeå University, SE-901 85, Umeå, Sweden
| | - Eram Javed
- Department of Clinical Microbiology, Umeå University, SE-901 85, Umeå, Sweden
| | - Anders Sjöstedt
- Department of Clinical Microbiology, Umeå University, SE-901 85, Umeå, Sweden.
| |
Collapse
|
44
|
Single vector platform vaccine protects against lethal respiratory challenge with Tier 1 select agents of anthrax, plague, and tularemia. Sci Rep 2018; 8:7009. [PMID: 29725025 PMCID: PMC5934503 DOI: 10.1038/s41598-018-24581-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 04/04/2018] [Indexed: 01/26/2023] Open
Abstract
Bacillus anthracis, Yersinia pestis, and Francisella tularensis are the causative agents of Tier 1 Select Agents anthrax, plague, and tularemia, respectively. Currently, there are no licensed vaccines against plague and tularemia and the licensed anthrax vaccine is suboptimal. Here we report F. tularensis LVS ΔcapB (Live Vaccine Strain with a deletion in capB)- and attenuated multi-deletional Listeria monocytogenes (Lm)-vectored vaccines against all three aforementioned pathogens. We show that LVS ΔcapB- and Lm-vectored vaccines express recombinant B. anthracis, Y. pestis, and F. tularensis immunoprotective antigens in broth and in macrophage-like cells and are non-toxic in mice. Homologous priming-boosting with the LVS ΔcapB-vectored vaccines induces potent antigen-specific humoral and T-cell-mediated immune responses and potent protective immunity against lethal respiratory challenge with all three pathogens. Protection against anthrax was far superior to that obtained with the licensed AVA vaccine and protection against tularemia was comparable to or greater than that obtained with the toxic and unlicensed LVS vaccine. Heterologous priming-boosting with LVS ΔcapB- and Lm-vectored B. anthracis and Y. pestis vaccines also induced potent protective immunity against lethal respiratory challenge with B. anthracis and Y. pestis. The single vaccine platform, especially the LVS ΔcapB-vectored vaccine platform, can be extended readily to other pathogens.
Collapse
|
45
|
Cui G, Wang J, Qi X, Su J. Transcription Elongation Factor GreA Plays a Key Role in Cellular Invasion and Virulence of Francisella tularensis subsp. novicida. Sci Rep 2018; 8:6895. [PMID: 29720697 PMCID: PMC5932009 DOI: 10.1038/s41598-018-25271-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 04/18/2018] [Indexed: 01/24/2023] Open
Abstract
Francisella tularensis is a facultative intracellular Gram-negative bacterium that causes the zoonotic disease tularemia. We identified the transcription elongation factor GreA as a virulence factor in our previous study, but its role was not defined. Here, we investigate the effects of the inactivation of the greA gene, generating a greA mutant of F. tularensis subsp. novicida. Inactivation of greA impaired the bacterial invasion into and growth within host cells, and subsequently virulence in mouse infection model. A transcriptomic analysis (RNA-Seq) showed that the loss of GreA caused the differential expression of 196 bacterial genes, 77 of which were identified as virulence factors in previous studies. To confirm that GreA regulates the expression of virulence factors involved in cell invasion by Francisella, FTN_1186 (pepO) and FTN_1551 (ampD) gene mutants were generated. The ampD deletion mutant showed reduced invasiveness into host cells. These results strongly suggest that GreA plays an important role in the pathogenesis of Francisella by affecting the expression of virulence genes and provide new insights into the complex regulation of Francisella infection.
Collapse
Affiliation(s)
- Guolin Cui
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Jun Wang
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Xinyi Qi
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Jingliang Su
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
46
|
Haymond A, Dowdy T, Johny C, Johnson C, Ball H, Dailey A, Schweibenz B, Villarroel K, Young R, Mantooth CJ, Patel T, Bases J, Dowd CS, Couch RD. A high-throughput screening campaign to identify inhibitors of DXP reductoisomerase (IspC) and MEP cytidylyltransferase (IspD). Anal Biochem 2018; 542:63-75. [PMID: 29180070 PMCID: PMC5817008 DOI: 10.1016/j.ab.2017.11.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 11/20/2017] [Accepted: 11/22/2017] [Indexed: 11/17/2022]
Abstract
The rise of antibacterial resistance among human pathogens represents a problem that could change the landscape of healthcare unless new antibiotics are developed. The methyl erythritol phosphate (MEP) pathway represents an attractive series of targets for novel antibiotic design, considering each enzyme of the pathway is both essential and has no human homologs. Here we describe a pilot scale high-throughput screening (HTS) campaign against the first and second committed steps in the pathway, catalyzed by DXP reductoisomerase (IspC) and MEP cytidylyltransferase (IspD), using compounds present in the commercially available LOPAC1280 library as well as in an in-house natural product extract library. Hit compounds were characterized to deduce their mechanism of inhibition; most function through aggregation. The HTS workflow outlined here is useful for quickly screening a chemical library, while effectively identifying false positive compounds associated with assay constraints and aggregation.
Collapse
Affiliation(s)
- Amanda Haymond
- Department of Chemistry and Biochemistry, George Mason University, Manassas, VA 20110, USA
| | - Tyrone Dowdy
- Department of Chemistry and Biochemistry, George Mason University, Manassas, VA 20110, USA
| | - Chinchu Johny
- Department of Chemistry and Biochemistry, George Mason University, Manassas, VA 20110, USA
| | - Claire Johnson
- Department of Chemistry and Biochemistry, George Mason University, Manassas, VA 20110, USA
| | - Haley Ball
- Department of Chemistry and Biochemistry, George Mason University, Manassas, VA 20110, USA
| | - Allyson Dailey
- Department of Chemistry and Biochemistry, George Mason University, Manassas, VA 20110, USA
| | - Brandon Schweibenz
- Department of Chemistry and Biochemistry, George Mason University, Manassas, VA 20110, USA
| | - Karen Villarroel
- Department of Chemistry and Biochemistry, George Mason University, Manassas, VA 20110, USA
| | - Richard Young
- Department of Chemistry and Biochemistry, George Mason University, Manassas, VA 20110, USA
| | - Clark J Mantooth
- Department of Chemistry and Biochemistry, George Mason University, Manassas, VA 20110, USA
| | - Trishal Patel
- Department of Chemistry and Biochemistry, George Mason University, Manassas, VA 20110, USA
| | - Jessica Bases
- Department of Chemistry and Biochemistry, George Mason University, Manassas, VA 20110, USA
| | - Cynthia S Dowd
- Department of Chemistry, George Washington University, Washington DC 20052, USA.
| | - Robin D Couch
- Department of Chemistry and Biochemistry, George Mason University, Manassas, VA 20110, USA.
| |
Collapse
|
47
|
Zinc Acquisition Mechanisms Differ between Environmental and Virulent Francisella Species. J Bacteriol 2018; 200:JB.00587-17. [PMID: 29109188 PMCID: PMC5786701 DOI: 10.1128/jb.00587-17] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 10/27/2017] [Indexed: 12/29/2022] Open
Abstract
Zinc is an essential nutrient for bacterial growth. Because host cells can restrict pathogen access to zinc as an antimicrobial defense mechanism, intracellular pathogens such as Francisella must sense their environment and acquire zinc in response. In many bacteria, the conserved transcription factor Zur is a key regulator of zinc acquisition. To identify mechanisms of zinc uptake in Francisella novicida U112, transcriptome sequencing of wild-type and putative zur mutant bacteria was performed. Only three genes were confirmed as directly regulated by Zur and zinc limitation by quantitative reverse transcription-PCR. One of these genes, FTN_0879, is predicted to encode a protein with similarity to the zupT family of zinc transporters, which are not typically regulated by Zur. While a putative znuACB operon encoding a high-affinity zinc transporter was identified in U112, expression of this operon was not controlled by Zur or zinc concentration. Disruption of zupT but not znuA in U112 impaired growth under zinc limitation, suggesting that ZupT is the primary mechanism of zinc acquisition under these conditions. In the virulent Francisella tularensis subsp. tularensis Schu S4 strain, zupT is a pseudogene, and attempts to delete znuA were unsuccessful, suggesting that it is essential in this strain. A reverse TetR repression system was used to knock down the expression of znuA in Schu S4, revealing that znuA is required for growth under zinc limitation and contributes to intracellular growth within macrophages. Overall, this work identifies genes necessary for adaptation to zinc limitation and highlights nutritional differences between environmental and virulent Francisella strains. IMPORTANCEFrancisella tularensis is a tier 1 select agent with a high potential for lethality and no approved vaccine. A better understanding of Francisella virulence factors is required for the development of therapeutics. While acquisition of zinc has been shown to be required for the virulence of numerous intracellular pathogens, zinc uptake has not been characterized in Francisella. This work characterizes the Zur regulon in F. novicida and identifies two transporters that contribute to bacterial growth under zinc limitation. In addition, these data identify differences in mechanisms of zinc uptake and tolerance to zinc limitation between F. tularensis and F. novicida, highlighting the role of znuA in the growth of Schu S4 under zinc limitation.
Collapse
|
48
|
Sampath V, McCaig WD, Thanassi DG. Amino acid deprivation and central carbon metabolism regulate the production of outer membrane vesicles and tubes by Francisella. Mol Microbiol 2018; 107:523-541. [PMID: 29240272 DOI: 10.1111/mmi.13897] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 12/07/2017] [Accepted: 12/11/2017] [Indexed: 12/23/2022]
Abstract
Francisella tularensis is a highly virulent Gram-negative bacterial pathogen that causes the zoonotic disease tularemia. F. novicida, a model tularemia strain, produces spherical outer membrane vesicles (OMV), as well as novel tubular vesicles and extensions of the cell surface. These OMV and tubes (OMV/T) are produced in a regulated manner and contain known virulence factors. Mechanisms by which bacterial vesicles are produced and regulated are not well understood. We performed a genetic screen in F. novicida to decipher the molecular basis for regulated OMV/T formation, and identified both hypo- and hyper-vesiculating mutants. Mutations in fumA and tktA, involved in central carbon metabolism, and in FTN_0908 and FTN_1037, of unknown function, resulted in severe defects in OMV/T production. Cysteine deprivation was identified as the signal that triggers OMV/T formation in F. novicida during growth in rich medium. We also found that fully virulent F. tularensis produces OMV/T in a similarly regulated manner. Further analysis revealed that OMV/T production is responsive to deprivation of essential amino acids in addition to cysteine, and that the hypo-vesiculating mutants are defective in responding to this signal. Thus, amino acid starvation, such as encountered by Francisella during host cell invasion, regulates the production of membrane-derived structures.
Collapse
Affiliation(s)
- Vinaya Sampath
- Department of Molecular Genetics and Microbiology, Center for Infectious Diseases, Stony Brook University, Stony Brook, NY 11794, USA
| | - William D McCaig
- Department of Molecular Genetics and Microbiology, Center for Infectious Diseases, Stony Brook University, Stony Brook, NY 11794, USA
| | - David G Thanassi
- Department of Molecular Genetics and Microbiology, Center for Infectious Diseases, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
49
|
Kinkead LC, Whitmore LC, McCracken JM, Fletcher JR, Ketelsen BB, Kaufman JW, Jones BD, Weiss DS, Barker JH, Allen LAH. Bacterial lipoproteins and other factors released by Francisella tularensis modulate human neutrophil lifespan: Effects of a TLR1 SNP on apoptosis inhibition. Cell Microbiol 2017; 20. [PMID: 29063667 PMCID: PMC5764820 DOI: 10.1111/cmi.12795] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 10/10/2017] [Accepted: 10/15/2017] [Indexed: 12/21/2022]
Abstract
Francisella tularensis infects several cell types including neutrophils, and aberrant neutrophil accumulation contributes to tissue destruction during tularaemia. We demonstrated previously that F. tularensis strains Schu S4 and live vaccine strain markedly delay human neutrophil apoptosis and thereby prolong cell lifespan, but the bacterial factors that mediate this aspect of virulence are undefined. Herein, we demonstrate that bacterial conditioned medium (CM) can delay apoptosis in the absence of direct infection. Biochemical analyses show that CM contained F. tularensis surface factors as well as outer membrane components. Our previous studies excluded roles for lipopolysaccharide and capsule in apoptosis inhibition, and current studies of [14C] acetate‐labelled bacteria argue against a role for other bacterial lipids in this process. At the same time, studies of isogenic mutants indicate that TolC and virulence factors whose expression requires FevR or MglA were also dispensable, demonstrating that apoptosis inhibition does not require Type I or Type VI secretion. Instead, we identified bacterial lipoproteins (BLPs) as active factors in CM. Additional studies of isolated BLPs demonstrated dose‐dependent neutrophil apoptosis inhibition via a TLR2‐dependent mechanism that is significantly influenced by a common polymorphism, rs5743618, in human TLR1. These data provide fundamental new insight into pathogen manipulation of neutrophil lifespan and BLP function.
Collapse
Affiliation(s)
- Lauren C Kinkead
- Inflammation Program, University of Iowa, Iowa City, Iowa, USA.,Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Laura C Whitmore
- Inflammation Program, University of Iowa, Iowa City, Iowa, USA.,Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Jenna M McCracken
- Inflammation Program, University of Iowa, Iowa City, Iowa, USA.,Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Joshua R Fletcher
- Graduate Program in Genetics, University of Iowa, Iowa City, Iowa, USA
| | - Brandi B Ketelsen
- Inflammation Program, University of Iowa, Iowa City, Iowa, USA.,Iowa City VA Health Care System, Iowa City, Iowa, USA
| | - Justin W Kaufman
- Inflammation Program, University of Iowa, Iowa City, Iowa, USA.,Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Bradley D Jones
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA.,Graduate Program in Genetics, University of Iowa, Iowa City, Iowa, USA
| | - David S Weiss
- Emory Vaccine Center, Emory University, Atlanta, Georgia, USA
| | - Jason H Barker
- Inflammation Program, University of Iowa, Iowa City, Iowa, USA.,Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA.,Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Lee-Ann H Allen
- Inflammation Program, University of Iowa, Iowa City, Iowa, USA.,Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA.,Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA.,Iowa City VA Health Care System, Iowa City, Iowa, USA
| |
Collapse
|
50
|
Hoang KV, Adcox HE, Fitch JR, Gordon DM, Curry HM, Schlesinger LS, White P, Gunn JS. AR-13, a Celecoxib Derivative, Directly Kills Francisella In Vitro and Aids Clearance and Mouse Survival In Vivo. Front Microbiol 2017; 8:1695. [PMID: 28955308 PMCID: PMC5600997 DOI: 10.3389/fmicb.2017.01695] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 08/23/2017] [Indexed: 12/15/2022] Open
Abstract
Francisella tularensis (F. tularensis) is the causative agent of tularemia and is classified as a Tier 1 select agent. No licensed vaccine is currently available in the United States and treatment of tularemia is confined to few antibiotics. In this study, we demonstrate that AR-13, a derivative of the cyclooxygenase-2 inhibitor celecoxib, exhibits direct in vitro bactericidal killing activity against Francisella including a type A strain of F. tularensis (SchuS4) and the live vaccine strain (LVS), as well as toward the intracellular proliferation of LVS in macrophages, without causing significant host cell toxicity. Identification of an AR-13-resistant isolate indicates that this compound has an intracellular target(s) and that efflux pumps can mediate AR-13 resistance. In the mouse model of tularemia, AR-13 treatment protected 50% of the mice from lethal LVS infection and prolonged survival time from a lethal dose of F. tularensis SchuS4. Combination of AR-13 with a sub-optimal dose of gentamicin protected 60% of F. tularensis SchuS4-infected mice from death. Taken together, these data support the translational potential of AR-13 as a lead compound for the further development of new anti-Francisella agents.
Collapse
Affiliation(s)
- Ky V Hoang
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, ColumbusOH, United States
| | - Haley E Adcox
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, ColumbusOH, United States
| | - James R Fitch
- The Institute for Genomic Medicine, Nationwide Children's Hospital, ColumbusOH, United States
| | - David M Gordon
- The Institute for Genomic Medicine, Nationwide Children's Hospital, ColumbusOH, United States
| | - Heather M Curry
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, ColumbusOH, United States
| | - Larry S Schlesinger
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, ColumbusOH, United States
| | - Peter White
- The Institute for Genomic Medicine, Nationwide Children's Hospital, ColumbusOH, United States.,Department of Pediatrics, The Ohio State University College of Medicine, ColumbusOH, United States
| | - John S Gunn
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, ColumbusOH, United States
| |
Collapse
|