86 results on '"Patricia L. Worsham"'
Search Results
2. Activation of Toll-Like Receptors by Live Gram-Negative Bacterial Pathogens Reveals Mitigation of TLR4 Responses and Activation of TLR5 by Flagella
- Author
-
Kei Amemiya, Jennifer L. Dankmeyer, Robert C. Bernhards, David P. Fetterer, David M. Waag, Patricia L. Worsham, and David DeShazer
- Subjects
TLRs ,gram-negative ,live pathogens ,innate immunity ,TLR4 suppression ,TLR5 activation ,Microbiology ,QR1-502 - Abstract
Successful bacterial pathogens have evolved to avoid activating an innate immune system in the host that responds to the pathogen through distinct Toll-like receptors (TLRs). The general class of biochemical components that activate TLRs has been studied extensively, but less is known about how TLRs interact with the class of compounds that are still associated with the live pathogen. Accordingly, we examined the activation of surface assembled TLR 2, 4, and 5 with live Tier 1 Gram-negative pathogens that included Yersinia pestis (plague), Burkholderia mallei (glanders), Burkholderia pseudomallei (melioidosis), and Francisella tularensis (tularemia). We found that Y. pestis CO92 grown at 28°C activated TLR2 and TLR4, but at 37°C the pathogen activated primarily TLR2. Although B. mallei and B. pseudomallei are genetically related, the former microorganism activated predominately TLR4, while the latter activated predominately TLR2. The capsule of wild-type B. pseudomallei 1026b was found to mitigate the activation of TLR2 and TLR4 when compared to a capsule mutant. Live F. tularensis (Ft) Schu S4 did not activate TLR2 or 4, although the less virulent Ft LVS and F. novicida activated only TLR2. B. pseudomallei purified flagellin or flagella attached to the microorganism activated TLR5. Activation of TLR5 was abolished by an antibody to TLR5, or a mutation of fliC, or elimination of the pathogen by filtration. In conclusion, we have uncovered new properties of the Gram-negative pathogens, and their interaction with TLRs of the host. Further studies are needed to include other microorganism to extend our observations with their interaction with TLRs, and to the possibility of leading to new efforts in therapeutics against these pathogens.
- Published
- 2021
- Full Text
- View/download PDF
3. Dysregulation of TNF-α and IFN-γ expression is a common host immune response in a chronically infected mouse model of melioidosis when comparing multiple human strains of Burkholderia pseudomallei
- Author
-
Kei Amemiya, Jennifer L. Dankmeyer, Jeremy J. Bearss, Xiankun Zeng, Spencer W. Stonier, Carl Soffler, Christopher K. Cote, Susan L. Welkos, David P. Fetterer, Taylor B. Chance, Sylvia R. Trevino, Patricia L. Worsham, and David M. Waag
- Subjects
Burkholderia pseudomallei ,Chronic infection ,BALB/c mice ,Pyogranulomatous lesion ,Inflammatory cytokines ,Intracellular TNFα and IFNγ ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Abstract Background Melioidosis is endemic in Southeast Asia and Northern Australia and is caused by the Gram-negative, facultative intracellular pathogen Burkholderia pseudomallei. Diagnosis of melioidosis is often difficult because of the protean clinical presentation of the disease, and it may mimic other diseases, such as tuberculosis. There are many different strains of B. pseudomallei that have been isolated from patients with melioidosis, but it was not clear if they could cause a similar disease in a chronic BALB/c murine model of melioidosis. Hence, we wanted to examine chronically infected mice exposed to different strains of B. pseudomallei to determine if there were differences in the host immune response to the pathogen. Results We identified common host immune responses exhibited in chronically infected BALB/c mice, although there was some heterogeneity in the host response in chronically infected mice after exposure to different strains of B. pseudomallei. They all displayed pyogranulomatous lesions in their spleens with a large influx of monocytes/macrophages, NK cells, and neutrophils identified by flow cytometry. Sera from chronically infected mice by ELISA exhibited elevated IgG titers to the pathogen, and we detected by Luminex micro-bead array technology a significant increase in the expression of inflammatory cytokines/chemokines, such as IFN-γ, IL-1α, IL-1β, KC, and MIG. By immunohistochemical and in situ RNA hybridization analysis we found that the increased expression of proinflammatory cytokines (IL-1α, IL-1β, TNF-α, IFN-γ) was confined primarily to the area with the pathogen within pyogranulomatous lesions. We also found that cultured splenocytes from chronically infected mice could express IFN-γ, TNF-α, and MIP-1α ex vivo without the need for additional exogenous stimulation. In addition by flow cytometry, we detected significant amounts of intracellular expression of TNF-α and IFN-γ without a protein transport blocker in monocytes/macrophages, NK cells, and neutrophils but not in CD4+ or CD8+ T cells in splenocytes from chronically infected mice. Conclusion Taken together the common features we have identified in chronically infected mice when 10 different human clinical strains of B. pseudomallei were examined could serve as biomarkers when evaluating potential therapeutic agents in mice for the treatment of chronic melioidosis in humans.
- Published
- 2020
- Full Text
- View/download PDF
4. Impact of Toll-Like Receptor-Specific Agonists on the Host Immune Response to the Yersinia pestis Plague rF1V Vaccine
- Author
-
Sergei Biryukov, Jennifer L. Dankmeyer, Zain Shamsuddin, Ivan Velez, Nathaniel O. Rill, Raysa Rosario-Acevedo, Christopher P. Klimko, Jennifer L. Shoe, Melissa Hunter, Michael D. Ward, Lisa H. Cazares, David P. Fetterer, Joel A. Bozue, Patricia L. Worsham, Christopher K. Cote, and Kei Amemiya
- Subjects
Yersinia pestis ,plague ,mice ,F1-V antigen ,vaccine ,CpG ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Relatively recent advances in plague vaccinology have produced the recombinant fusion protein F1-V plague vaccine. This vaccine has been shown to readily protect mice from both bubonic and pneumonic plague. The protection afforded by this vaccine is solely based upon the immune response elicited by the F1 or V epitopes expressed on the F1-V fusion protein. Accordingly, questions remain surrounding its efficacy against infection with non-encapsulated (F1-negative) strains. In an attempt to further optimize the F1-V elicited immune response and address efficacy concerns, we examined the inclusion of multiple toll-like receptor agonists into vaccine regimens. We examined the resulting immune responses and also any protection afforded to mice that were exposed to aerosolized Yersinia pestis. Our data demonstrate that it is possible to further augment the F1-V vaccine strategy in order to optimize and augment vaccine efficacy.
- Published
- 2021
- Full Text
- View/download PDF
5. Proteomic Analysis of Non-human Primate Peripheral Blood Mononuclear Cells During Burkholderia mallei Infection Reveals a Role of Ezrin in Glanders Pathogenesis
- Author
-
Chih-Yuan Chiang, Yang Zhong, Michael D. Ward, Douglas J. Lane, Tara Kenny, Raysa Rosario-Acevedo, Brett P. Eaton, Sylvia R. Treviño, Taylor B. Chance, Meghan Hu, Patricia L. Worsham, David M. Waag, Richard T. Moore, Lisa H. Cazares, Christopher K. Cote, Yingyao Zhou, and Rekha G. Panchal
- Subjects
Burkholderia mallei ,glanders ,innate immunity ,inflammatory responses ,biothreat agent ,proteomics ,Microbiology ,QR1-502 - Abstract
Burkholderia mallei, the causative agent of glanders, is a gram-negative intracellular bacterium. Depending on different routes of infection, the disease is manifested by pneumonia, septicemia, and chronic infections of the skin. B. mallei poses a serious biological threat due to its ability to infect via aerosol route, resistance to multiple antibiotics and to date there are no US Food and Drug Administration (FDA) approved vaccines available. Induction of innate immunity, inflammatory cytokines and chemokines following B. mallei infection, have been observed in in vitro and small rodent models; however, a global characterization of host responses has never been systematically investigated using a non-human primate (NHP) model. Here, using a liquid chromatography-tandem mass spectrometry (LC-MS/MS) approach, we identified alterations in expression levels of host proteins in peripheral blood mononuclear cells (PBMCs) originating from naïve rhesus macaques (Macaca mulatta), African green monkeys (Chlorocebus sabaeus), and cynomolgus macaques (Macaca fascicularis) exposed to aerosolized B. mallei. Gene ontology (GO) analysis identified several statistically significant overrepresented biological annotations including complement and coagulation cascade, nucleoside metabolic process, vesicle-mediated transport, intracellular signal transduction and cytoskeletal protein binding. By integrating an LC-MS/MS derived proteomics dataset with a previously published B. mallei host-pathogen interaction dataset, a statistically significant predictive protein-protein interaction (PPI) network was constructed. Pharmacological perturbation of one component of the PPI network, specifically ezrin, reduced B. mallei mediated interleukin-1β (IL-1β). On the contrary, the expression of IL-1β receptor antagonist (IL-1Ra) was upregulated upon pretreatment with the ezrin inhibitor. Taken together, inflammasome activation as demonstrated by IL-1β production and the homeostasis of inflammatory response is critical during the pathogenesis of glanders. Furthermore, the topology of the network reflects the underlying molecular mechanism of B. mallei infections in the NHP model.
- Published
- 2021
- Full Text
- View/download PDF
6. Protection Elicited by Attenuated Live Yersinia pestis Vaccine Strains against Lethal Infection with Virulent Y. pestis
- Author
-
Christopher K. Cote, Sergei S. Biryukov, Christopher P. Klimko, Jennifer L. Shoe, Melissa Hunter, Raysa Rosario-Acevedo, David P. Fetterer, Krishna L. Moody, Joshua R. Meyer, Nathaniel O. Rill, Jennifer L. Dankmeyer, Patricia L. Worsham, Joel A. Bozue, and Susan L. Welkos
- Subjects
plague ,Yersinia pestis ,vaccine ,mice ,bubonic ,pneumonic ,Medicine - Abstract
The etiologic agent of plague, Yersinia pestis, is a globally distributed pathogen which poses both a natural and adversarial threat. Due largely to the rapid course and high mortality of pneumonic plague, vaccines are greatly needed. Two-component protein vaccines have been unreliable and potentially vulnerable to vaccine resistance. We evaluated the safety and efficacy of eight live Y. pestis strains derived from virulent strains CO92 or KIM6+ and mutated in one or more virulence-associated gene(s) or cured of plasmid pPst. Stringent, single-dose vaccination allowed down-selection of the two safest and most protective vaccine candidates, CO92 mutants pgm- pPst- and ΔyscN. Both completely protected BALB/c mice against subcutaneous and aerosol challenge with Y. pestis. Strain CD-1 outbred mice were more resistant to bubonic (but not pneumonic) plague than BALB/c mice, but the vaccines elicited partial protection of CD-1 mice against aerosol challenge, while providing full protection against subcutaneous challenge. A ΔyscN mutant of the nonencapsulated C12 strain was expected to display antigens previously concealed by the capsule. C12 ΔyscN elicited negligible titers to F1 but comparable antibody levels to whole killed bacteria, as did CO92 ΔyscN. Although one dose of C12 ΔyscN was not protective, vaccination with two doses of either CO92 ΔyscN, or a combination of the ΔyscN mutants of C12 and CO92, protected optimally against lethal bubonic or pneumonic plague. Protection against encapsulated Y. pestis required inclusion of F1 in the vaccine and was associated with high anti-F1 titers.
- Published
- 2021
- Full Text
- View/download PDF
7. Binding Sites of Anti-Lcr V Monoclonal Antibodies Are More Critical than the Avidities and Affinities for Passive Protection against Yersinia pestis Infection in a Bubonic Plague Model
- Author
-
Kei Amemiya, Jennifer L. Dankmeyer, Sarah L. Keasey, Sylvia R. Trevino, Michael M. Wormald, Stephanie A. Halasohoris, Wilson J. Ribot, David P. Fetterer, Christopher K. Cote, Patricia L. Worsham, Jeffrey J. Adamovicz, and Robert G. Ulrich
- Subjects
monoclonal antibodies ,Yersinia pestis ,plague ,binding sites ,avidities ,affinities ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Plague is a zoonotic disease that is caused by Yersinia pestis. Monoclonal antibodies (mAbs) that bind to the V-antigen, a virulence factor that is produced by Y. pestis, can passively protect mice from plague. An analysis of protective mAbs that bind to V-antigen was made to assess binding sites, avidities, and affinities. Anti-V mAbs were screened for their efficacy in a murine model of plague. Antigen-binding sites of protective V mAbs were determined with a linear peptide library, V-antigen fragment, competitive binding, and surface plasmon resonance. The avidities to the V-antigen was determined by ELISA, and affinities of the mAbs to the V-antigen were determined by surface plasmon resonance. The most protective mAb 7.3 bound to a unique conformational site on the V-antigen, while a less protective mAb bound to a different conformational site located on the same V-antigen fragment as mAb 7.3. The avidity of mAb 7.3 for the V-antigen was neither the strongest overall nor did it have the highest affinity for the V-antigen. The binding site of the most protective mAb was critical in its ability to protect against a lethal plague challenge.
- Published
- 2020
- Full Text
- View/download PDF
8. Laser Scanning Confocal Microscopy Was Used to Validate the Presence of Burkholderia pseudomallei or B. mallei in Formalin-Fixed Paraffin Embedded Tissues
- Author
-
Kei Amemiya, Xiankun Zeng, Jeremy J. Bearss, Christopher K. Cote, Carl Soffler, Robert C. Bernhards, Jennifer L. Dankmeyer, Wilson J. Ribot, Sylvia R. Trevino, Susan L. Welkos, Patricia L. Worsham, and David M. Waag
- Subjects
Burkholderia pseudomallei ,melioidosis ,Burkholderia mallei ,glanders ,laser scanning confocal microscopy ,formalin-fixed paraffin embedded tissue ,Medicine - Abstract
Burkholderia pseudomallei and B. mallei are Gram-negative, facultative intracellular bacteria that cause melioidosis and glanders, respectively. Currently, there are no vaccines for these two diseases. Animal models have been developed to evaluate vaccines and therapeutics. Tissues from infected animals, however, must be fixed in formalin and embedded in paraffin (FFPE) before analysis. A brownish staining material in infected tissues that represents the exopolysaccharide of the pathogen was seen by bright field microscopy but not the actual microorganism. Because of these results, FFPE tissue was examined by laser scanning confocal microscopy (LSCM) in an attempt to see the microorganism. Archival FFPE tissues were examined from ten mice, and five nonhuman primates after exposure to B. pseudomallei or B. mallei by LSCM. Additionally, a historical spleen biopsy from a human suspected of exposure to B. mallei was examined. B. pseudomallei was seen in many of the infected tissues from mice. Four out of five nonhuman primates were positive for the pathogen. In the human sample, B. mallei was seen in pyogranulomas in the spleen biopsy. Thus, the presence of the pathogen was validated by LSCM in murine, nonhuman primate, and human FFPE tissues.
- Published
- 2020
- Full Text
- View/download PDF
9. The Impact of Age and Sex on Mouse Models of Melioidosis
- Author
-
Christopher P. Klimko, Sylvia R. Treviño, Alicia M. Moreau, Michael J. Aponte Cuadrado, Joshua R. Meyer, David P. Fetterer, Susan L. Welkos, Patricia L. Worsham, Norman Kreiselmeier, Carl Soffler, and Christopher K. Cote
- Subjects
burkholderia pseudomallei ,melioidosis ,mice ,pathology ,median lethal dose ,inhalational ,intraperitoneal ,Medicine - Abstract
Mouse models have been used to generate critical data for many infectious diseases. In the case of Burkholderia pseudomallei, mouse models have been invaluable for bacterial pathogenesis studies as well as for testing novel medical countermeasures including both vaccines and therapeutics. Mouse models of melioidosis have also provided a possible way forward to better understand the chronicity associated with this infection, as it appears that BALB/c mice develop an acute infection with B. pseudomallei, whereas the C57BL/6 model is potentially more suggestive of a chronic infection. Several unanswered questions, however, persist around this model. In particular, little attention has been paid to the effect of age or sex on the disease outcome in these animal models. In this report, we determined the LD50 of the B. pseudomallei K96243 strain in both female and male BALB/c and C57BL/6 mice in three distinct age groups. Our data demonstrated a modest increase in susceptibility associated with sex in this model, and we documented important histopathological differences associated with the reproductive systems of each sex. There was a statistically significant inverse correlation between age and susceptibility. The older mice, in most cases, were more susceptible to the infection. Additionally, our retrospective analyses suggested that the impact of animal supplier on disease outcome in mice may be minimal. These observations were consistent regardless of whether the mice were injected with bacteria intraperitoneally or if they were exposed to aerosolized bacteria. All of these factors should be considered when designing experiments using mouse models of melioidosis.
- Published
- 2020
- Full Text
- View/download PDF
10. Assays for the rapid and specific identification of North American Yersinia pestis and the common laboratory strain CO92
- Author
-
Amy J. Vogler, Elizabeth M. Driebe, Judy Lee, Raymond K. Auerbach, Christopher J. Allender, Miles Stanley, Kristy Kubota, Gary L. Andersen, Lyndsay Radnedge, Patricia L. Worsham, Paul Keim, and David M. Wagner
- Subjects
Biology (General) ,QH301-705.5 - Abstract
We present TaqMan-minor groove binding (MGB) assays for an SNP that separates the Yersinia pestis strain CO92 from all other strains and for another SNP that separates North American strains from all other global strains.
- Published
- 2008
- Full Text
- View/download PDF
11. Multiple Roles of Myd88 in the Immune Response to the Plague F1-V Vaccine and in Protection against an Aerosol Challenge of Yersinia pestis CO92 in Mice
- Author
-
Jennifer L. Dankmeyer, Randy L. Fast, Christopher K. Cote, Patricia L. Worsham, David Fritz, Diana Fisher, Steven J. Kern, Tod Merkel, Carsten J. Kirschning, and Kei Amemiya
- Subjects
Immunologic diseases. Allergy ,RC581-607 - Abstract
The current candidate vaccine against Yersinia pestis infection consists of two subunit proteins: the capsule protein or F1 protein and the low calcium response V protein or V-antigen. Little is known of the recognition of the vaccine by the host’s innate immune system and how it affects the acquired immune response to the vaccine. Thus, we vaccinated Toll-like receptor (Tlr) 2, 4, and 2/4-double deficient, as well as signal adaptor protein Myd88-deficient mice. We found that Tlr4 and Myd88 appeared to be required for an optimal immune response to the F1-V vaccine but not Tlr2 when compared to wild-type mice. However, there was a difference between the requirement for Tlr4 and MyD88 in vaccinated animals. When F1-V vaccinated Tlr4 mutant (lipopolysaccharide tolerant) and Myd88-deficient mice were challenged by aerosol with Y. pestis CO92, all but one Tlr4 mutant mice survived the challenge, but no vaccinated Myd88-deficient mice survived the challenge. Spleens from these latter nonsurviving mice showed that Y. pestis was not cleared from the infected mice. Our results suggest that MyD88 appears to be important for both an optimal immune response to F1-V and in protection against a lethal challenge of Y. pestis CO92 in F1-V vaccinated mice.
- Published
- 2014
- Full Text
- View/download PDF
12. Activation of Toll-Like Receptors by Live Gram-Negative Bacterial Pathogens Reveals Mitigation of TLR4 Responses and Activation of TLR5 by Flagella
- Author
-
Patricia L. Worsham, David DeShazer, Robert C. Bernhards, Kei Amemiya, David P Fetterer, Jennifer L. Dankmeyer, and David M. Waag
- Subjects
Microbiology (medical) ,Immunology ,Microbiology ,Tularemia ,Burkholderia mallei ,Cellular and Infection Microbiology ,TLR5 activation ,medicine ,Animals ,TLRs ,innate immunity ,Francisella tularensis ,Original Research ,Innate immune system ,biology ,Burkholderia pseudomallei ,Toll-Like Receptors ,Glanders ,live pathogens ,TLR4 suppression ,biology.organism_classification ,medicine.disease ,bacterial infections and mycoses ,QR1-502 ,gram-negative ,Toll-Like Receptor 4 ,Toll-Like Receptor 5 ,Infectious Diseases ,Melioidosis ,Yersinia pestis ,Flagella ,biology.protein ,bacteria ,Flagellin - Abstract
Successful bacterial pathogens have evolved to avoid activating an innate immune system in the host that responds to the pathogen through distinct Toll-like receptors (TLRs). The general class of biochemical components that activate TLRs has been studied extensively, but less is known about how TLRs interact with the class of compounds that are still associated with the live pathogen. Accordingly, we examined the activation of surface assembled TLR 2, 4, and 5 with live Tier 1 Gram-negative pathogens that included Yersinia pestis (plague), Burkholderia mallei (glanders), Burkholderia pseudomallei (melioidosis), and Francisella tularensis (tularemia). We found that Y. pestis CO92 grown at 28°C activated TLR2 and TLR4, but at 37°C the pathogen activated primarily TLR2. Although B. mallei and B. pseudomallei are genetically related, the former microorganism activated predominately TLR4, while the latter activated predominately TLR2. The capsule of wild-type B. pseudomallei 1026b was found to mitigate the activation of TLR2 and TLR4 when compared to a capsule mutant. Live F. tularensis (Ft) Schu S4 did not activate TLR2 or 4, although the less virulent Ft LVS and F. novicida activated only TLR2. B. pseudomallei purified flagellin or flagella attached to the microorganism activated TLR5. Activation of TLR5 was abolished by an antibody to TLR5, or a mutation of fliC, or elimination of the pathogen by filtration. In conclusion, we have uncovered new properties of the Gram-negative pathogens, and their interaction with TLRs of the host. Further studies are needed to include other microorganism to extend our observations with their interaction with TLRs, and to the possibility of leading to new efforts in therapeutics against these pathogens.
- Published
- 2021
- Full Text
- View/download PDF
13. Protection Elicited by Attenuated Live Yersinia pestis Vaccine Strains Against Lethal Infection with Virulent Y. pestis
- Author
-
Joshua R. Meyer, Susan L. Welkos, Jennifer L. Shoe, Raysa Rosario-Acevedo, Joel A. Bozue, Nathaniel O Rill, Jennifer L. Dankmeyer, Sergei S. Biryukov, Melissa G. Hunter, Patricia L. Worsham, David P Fetterer, Christopher K. Cote, Krishna Moody, and Christopher P. Klimko
- Subjects
0301 basic medicine ,Pneumonic plague ,mice ,Yersinia pestis ,Immunology ,Virulence ,lcsh:Medicine ,bubonic ,Bubonic plague ,Article ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,vaccine ,Drug Discovery ,medicine ,Pharmacology (medical) ,030212 general & internal medicine ,pneumonic ,phage shock protein, PSP ,Pathogen ,Pharmacology ,live attenuated vaccine ,Attenuated vaccine ,biology ,lcsh:R ,biology.organism_classification ,medicine.disease ,Virology ,plague ,Vaccination ,030104 developmental biology ,Infectious Diseases ,phage shock protein (PSP) - Abstract
The etiologic agent of plague, Yersinia pestis, is a globally distributed pathogen which poses both a natural and adversarial threat. Due largely to the rapid course and high mortality of pneumonic plague, vaccines are greatly needed. Two-component protein vaccines have been unreliable and potentially vulnerable to vaccine resistance. We evaluated the safety and efficacy of eight live Y. pestis strains derived from virulent strains CO92 or KIM6+ and mutated in one or more virulence-associated gene(s) or cured of plasmid pPst. Stringent, single-dose vaccination allowed down-selection of the two safest and most protective vaccine candidates, CO92 mutants pgm- pPst- and ΔyscN. Both completely protected BALB/c mice against subcutaneous and aerosol challenge with Y. pestis. Strain CD-1 outbred mice were more resistant to bubonic (but not pneumonic) plague than BALB/c mice, but the vaccines elicited partial protection of CD-1 mice against aerosol challenge, while providing full protection against subcutaneous challenge. A ΔyscN mutant of the nonencapsulated C12 strain was expected to display antigens previously concealed by the capsule. C12 ΔyscN elicited negligible titers to F1 but comparable antibody levels to whole killed bacteria, as did CO92 ΔyscN. Although one dose of C12 ΔyscN was not protective, vaccination with two doses of either CO92 ΔyscN, or a combination of the ΔyscN mutants of C12 and CO92, protected optimally against lethal bubonic or pneumonic plague. Protection against encapsulated Y. pestis required inclusion of F1 in the vaccine and was associated with high anti-F1 titers.
- Published
- 2021
14. Comparative virulence of three different strains of Burkholderia pseudomallei in an aerosol non-human primate model
- Author
-
Teresa Krakauer, Sara I. Ruiz, Carl Soffler, Patricia L. Worsham, David M. Waag, Sylvia R. Trevino, Alicia M. Moreau, Jennifer L. Dankmeyer, Christopher P. Klimko, Kei Amemiya, David P Fetterer, Jo Lynne Raymond, and Christopher K. Cote
- Subjects
0301 basic medicine ,Bacterial Diseases ,Melioidosis ,Burkholderia pseudomallei ,Physiology ,Neutrophils ,Antibiotics ,RC955-962 ,Bacteremia ,Drug resistance ,White Blood Cells ,Medical Conditions ,Bone Marrow ,Animal Cells ,Immune Physiology ,Arctic medicine. Tropical medicine ,Chlorocebus aethiops ,Medicine and Health Sciences ,Telemetry ,Lung ,Immune Response ,Asia, Southeastern ,Innate Immune System ,biology ,Virulence ,Chemotaxis ,Thailand ,Cell Motility ,Infectious Diseases ,Liver ,Disease Progression ,Cytokines ,Chemokines ,Cellular Types ,Public aspects of medicine ,RA1-1270 ,Research Article ,medicine.drug_class ,Immune Cells ,030106 microbiology ,Immunology ,Research and Analysis Methods ,Microbiology ,Sepsis ,03 medical and health sciences ,Immune system ,Signs and Symptoms ,medicine ,Animals ,Humans ,Animal Models of Disease ,Aerosols ,Inflammation ,Blood Cells ,business.industry ,Euthanasia ,Macrophages ,Public Health, Environmental and Occupational Health ,Australia ,Biology and Life Sciences ,Cell Biology ,Molecular Development ,medicine.disease ,biology.organism_classification ,bacterial infections and mycoses ,Macaca mulatta ,Disease Models, Animal ,Animal Models of Infection ,030104 developmental biology ,Blood chemistry ,Immune System ,Animal Studies ,Clinical Medicine ,business ,Spleen ,Developmental Biology - Abstract
Melioidosis, caused by the Gram-negative bacterium Burkholderia pseudomallei, is a major cause of sepsis and mortality in endemic regions of Southeast Asia and Northern Australia. B. pseudomallei is a potential bioterrorism agent due to its high infectivity, especially via inhalation, and its inherent resistance to antimicrobials. There is currently no vaccine for melioidosis and antibiotic treatment can fail due to innate drug resistance, delayed diagnosis and treatment, or insufficient duration of treatment. A well-characterized animal model that mimics human melioidosis is needed for the development of new medical countermeasures. This study first characterized the disease progression of melioidosis in the African green monkey (AGM) and rhesus macaque (RM) for non-human primate model down-selection. All AGMs developed acute lethal disease similar to that described in human acute infection following exposure to aerosolized B. pseudomallei strain HBPUB10134a. Only 20% of RMs succumbed to acute disease. Disease progression, immune response and pathology of two other strains of B. pseudomallei, K96243 and MSHR5855, were also compared using AGMs. These three B. pseudomallei strains represent a highly virulent strain from Thailand (HBPUB101034a), a highly virulent strains from Australia (MSHR5855), and a commonly used laboratory strains originating from Thailand (K96243). Animals were observed for clinical signs of infection and blood samples were analyzed for cytokine responses, blood chemistry and leukocyte changes in order to characterize bacterial infection. AGMs experienced fever after exposure to aerosolized B. pseudomallei at the onset of acute disease. Inflammation, abscesses and/or pyogranulomas were observed in lung with all three strains of B. pseudomallei. Inflammation, abscesses and/or pyogranulomas were observed in lymph nodes, spleen, liver and/or kidney with B. pseudomallei, HBPUB10134a and K96243. Additionally, the Australian strain MSHR5855 induced brain lesions in one AGM similar to clinical cases of melioidosis seen in Australia. Elevated serum levels of IL-1β, IL-1 receptor antagonist, IL-6, MCP-1, G-CSF, HGF, IFNγ, MIG, I-TAC, and MIP-1β at terminal end points can be significantly correlated with non-survivors with B. pseudomallei infection in AGM. The AGM model represents an acute model of B. pseudomallei infection for all three strains from two geographical locations and will be useful for efficacy testing of vaccines and therapeutics against melioidosis. In summary, a dysregulated immune response leading to excessive persistent inflammation and inflammatory cell death is the key driver of acute melioidosis. Early intervention in these pathways will be necessary to counter B. pseudomallei and mitigate the pathological consequences of melioidosis., Author summary Melioidosis, caused by the Gram-negative bacterium Burkholderia pseudomallei, is endemic in tropical regions globally and is an emerging threat in non-tropical areas worldwide. Its mortality rate is high in endemic areas due to its high infectivity, antimicrobial resistance, lack of available vaccines and limited treatment options. Animal model development and pathogenicity studies of various isolates are critical for the development of countermeasures against this pathogen. In this study, we compared the virulence of three different isolates of B. pseudomallei from two geographical locations in an aerosol non-human primate model. We found that early elevations of both pro-inflammatory and anti-inflammatory mediators, as well as the persistence of these mediators in the terminal phase of bacterial infection correlate with mortality. Histopathological analysis showed that the severity of lesions in various organs also correlates with the virulence of the B. pseudomallei strains, HBPUB10134a, MSHR5855 and K96243. Thus, a dysregulated immune response leading to excessive IL-1β and IL-6 at terminal end points and necrosis are key drivers of acute melioidosis. Development of drugs targeting these host response processes will be necessary to counter B. pseudomallei and mitigate the pathological consequences of melioidosis. This non-human primate model will facilitate the screening of vaccines and novel therapeutics.
- Published
- 2021
15. Comparison of three non-human primate aerosol models for glanders, caused by Burkholderia mallei
- Author
-
Sylvia R. Trevino, Susan L. Welkos, Jennifer L. Dankmeyer, Christopher K. Cote, Patricia L. Worsham, Franco Rossi, Kei Amemiya, David M. Waag, Susham S. Ingavale, Xiankun Zeng, Taylor B. Chance, Steven J. Kern, David P Fetterer, and Steven A. Tobery
- Subjects
0301 basic medicine ,Melioidosis ,Burkholderia pseudomallei ,030106 microbiology ,Microbiology ,Burkholderia mallei ,03 medical and health sciences ,Antibiotic resistance ,Chlorocebus aethiops ,medicine ,Animals ,Horses ,Pathogen ,Subclinical infection ,Aerosols ,biology ,business.industry ,Glanders ,biology.organism_classification ,medicine.disease ,Macaca mulatta ,Rhesus macaque ,Disease Models, Animal ,030104 developmental biology ,Infectious Diseases ,Immunology ,business ,Pneumonia (non-human) - Abstract
Burkholderia mallei is a gram-negative obligate animal pathogen that causes glanders, a highly contagious and potentially fatal disease of solipeds including horses, mules, and donkeys. Humans are also susceptible, and exposure can result in a wide range of clinical forms, i.e., subclinical infection, chronic forms with remission and exacerbation, or acute and potentially lethal septicemia and/or pneumonia. Due to intrinsic antibiotic resistance and the ability of the organisms to survive intracellularly, current treatment regimens are protracted and complicated; and no vaccine is available. As a consequence of these issues, and since B. mallei is infectious by the aerosol route, B. mallei is regarded as a major potential biothreat agent. To develop optimal medical countermeasures and diagnostic tests, well characterized animal models of human glanders are needed. The goal of this study was to perform a head-to-head comparison of models employing three commonly used nonhuman primate (NHP) species, the African green monkey (AGM), Rhesus macaque, and the Cynomolgus macaque. The natural history of infection and in vitro clinical, histopathological, immunochemical, and bacteriological parameters were examined. The AGMs were the most susceptible NHP to B. mallei; five of six expired within 14 days. Although none of the Rhesus or Cynomolgus macaques succumbed, the Rhesus monkeys exhibited abnormal signs and clinical findings associated with B. mallei infection; and the latter may be useful for modeling chronic B. mallei infection. Based on the disease progression observations, gross and histochemical pathology, and humoral and cellular immune response findings, the AGM appears to be the optimal model of acute, lethal glanders infection. AGM models of infection by B. pseudomallei, the etiologic agent of melioidosis, have been characterized recently. Thus, the selection of the AGM species provides the research community with a single NHP model for investigations on acute, severe, inhalational melioidosis and glanders.
- Published
- 2021
16. Proteomic Analysis of Non-human Primate Peripheral Blood Mononuclear Cells During
- Author
-
Chih-Yuan, Chiang, Yang, Zhong, Michael D, Ward, Douglas J, Lane, Tara, Kenny, Raysa, Rosario-Acevedo, Brett P, Eaton, Sylvia R, Treviño, Taylor B, Chance, Meghan, Hu, Patricia L, Worsham, David M, Waag, Richard T, Moore, Lisa H, Cazares, Christopher K, Cote, Yingyao, Zhou, and Rekha G, Panchal
- Subjects
proteomics ,glanders ,inflammatory responses ,biothreat agent ,Microbiology ,Burkholderia mallei ,innate immunity ,Original Research - Abstract
Burkholderia mallei, the causative agent of glanders, is a gram-negative intracellular bacterium. Depending on different routes of infection, the disease is manifested by pneumonia, septicemia, and chronic infections of the skin. B. mallei poses a serious biological threat due to its ability to infect via aerosol route, resistance to multiple antibiotics and to date there are no US Food and Drug Administration (FDA) approved vaccines available. Induction of innate immunity, inflammatory cytokines and chemokines following B. mallei infection, have been observed in in vitro and small rodent models; however, a global characterization of host responses has never been systematically investigated using a non-human primate (NHP) model. Here, using a liquid chromatography-tandem mass spectrometry (LC-MS/MS) approach, we identified alterations in expression levels of host proteins in peripheral blood mononuclear cells (PBMCs) originating from naïve rhesus macaques (Macaca mulatta), African green monkeys (Chlorocebus sabaeus), and cynomolgus macaques (Macaca fascicularis) exposed to aerosolized B. mallei. Gene ontology (GO) analysis identified several statistically significant overrepresented biological annotations including complement and coagulation cascade, nucleoside metabolic process, vesicle-mediated transport, intracellular signal transduction and cytoskeletal protein binding. By integrating an LC-MS/MS derived proteomics dataset with a previously published B. mallei host-pathogen interaction dataset, a statistically significant predictive protein-protein interaction (PPI) network was constructed. Pharmacological perturbation of one component of the PPI network, specifically ezrin, reduced B. mallei mediated interleukin-1β (IL-1β). On the contrary, the expression of IL-1β receptor antagonist (IL-1Ra) was upregulated upon pretreatment with the ezrin inhibitor. Taken together, inflammasome activation as demonstrated by IL-1β production and the homeostasis of inflammatory response is critical during the pathogenesis of glanders. Furthermore, the topology of the network reflects the underlying molecular mechanism of B. mallei infections in the NHP model.
- Published
- 2020
17. Bacteriophage-associated genes responsible for the widely divergent phenotypes of variants of Burkholderia pseudomallei strain MSHR5848
- Author
-
Sean Lovett, Kathleen A. Kuehl, David DeShazer, Susan L. Welkos, Joshua Richardson, Kei Amemiya, G. I. Koroleva, Patricia L. Worsham, and Mei Sun
- Subjects
DNA, Bacterial ,0301 basic medicine ,Microbiology (medical) ,Burkholderia pseudomallei ,030106 microbiology ,Microbiology ,Bacteriophage ,03 medical and health sciences ,Gene Duplication ,Genes, Regulator ,Gene duplication ,Gene expression ,Humans ,Bacteriophages ,Cloning, Molecular ,Gene ,Type VI secretion system ,Regulator gene ,Genetics ,biology ,Sequence Analysis, RNA ,Gene Expression Profiling ,Computational Biology ,Gene Expression Regulation, Bacterial ,Sequence Analysis, DNA ,General Medicine ,biology.organism_classification ,Phenotype ,Microscopy, Electron ,RNA, Bacterial ,030104 developmental biology ,Melioidosis ,Multigene Family ,Myoviridae ,DNA, Viral ,Research Article - Abstract
Purpose. Burkholderia pseudomallei , the tier 1 agent of melioidosis, is a saprophytic microbe that causes endemic infections in tropical regions such as South-East Asia and Northern Australia. It is globally distributed, challenging to diagnose and treat, infectious by several routes including inhalation, and has potential for adversarial use. B. pseudomallei strain MSHR5848 produces two colony variants, smooth (S) and rough (R), which exhibit a divergent range of morphological, biochemical and metabolic phenotypes, and differ in macrophage and animal infectivity. We aimed to characterize two major phenotypic differences, analyse gene expression and study the regulatory basis of the variation. Methodology. Phenotypic expression was characterized by DNA and RNA sequencing, microscopy, and differential bacteriology. Regulatory genes were identified by cloning and bioinformatics. Results/Key findings. Whereas S produced larger quantities of extracellular DNA, R was upregulated in the production of a unique chromosome 1-encoded Siphoviridae-like bacteriophage, φMSHR5848. Exploratory transcriptional analyses revealed significant differences in variant expression of genes encoding siderophores, pili assembly, type VI secretion system cluster 4 (T6SS-4) proteins, several exopolysaccharides and secondary metabolites. A single 3 base duplication in S was the only difference that separated the variants genetically. It occurred upstream of a cluster of bacteriophage-associated genes on chromosome 2 that were upregulated in S. The first two genes were involved in regulating expression of the multiple phenotypes distinguishing S and R. Conclusion. Bacteriophage-associated proteins have a major role in the phenotypic expression of MSHR5848. The goals are to determine the regulatory basis of this phenotypic variation and its role in pathogenesis and environmental persistence of B. pseudomallei .
- Published
- 2019
- Full Text
- View/download PDF
18. Animal Models for Melioidosis
- Author
-
Joel A. Bozue, Susan L. Welkos, Carl Soffler, Kei Amemiya, Patricia L. Worsham, David DeShazer, and Christopher K. Cote
- Subjects
0301 basic medicine ,Melioidosis ,Burkholderia pseudomallei ,Diabetic mouse ,Computational biology ,Disease ,Biology ,medicine.disease ,biology.organism_classification ,03 medical and health sciences ,030104 developmental biology ,Infectious Diseases ,medicine ,Immunology and Allergy ,Disease process ,Identification (biology) ,Organism ,Large animal - Abstract
Development, testing, and evaluation of medical countermeasures for melioidosis are hampered by a lack of well-characterized and standardized animal models. Recent work has both refined existing animal models for this disease and identified new ones. Head-to-head comparisons of mouse strains with varying susceptibility to the organism and using different routes of infection highlighted and confirmed important similarities and differences between murine models and exposure routes. Diabetic mouse models provided insight into the disease process in humans having this major risk factor. Large animal models, both livestock and non-human primate, have been established. Alternative (non-mammalian) models have been useful in identification of virulence factors and screening of therapeutic candidates. They hold potential for large-scale screening that would not be appropriate or practical for mammalian species. Recent advances in animal and alternative modeling will enhance our understanding of the organism and the disease process, as well as accelerating the development of medical countermeasures.
- Published
- 2017
- Full Text
- View/download PDF
19. Deletion of Two Genes in Burkholderia pseudomallei MSHR668 That Target Essential Amino Acids Protect Acutely Infected BALB/c Mice and Promote Long Term Survival
- Author
-
Christopher P. Klimko, Sherry M Mou, David P Fetterer, Sergei S. Biryukov, Patricia L. Worsham, Kei Amemiya, Sylvia R. Trevino, Christopher K. Cote, Jennifer L. Dankmeyer, David DeShazer, and Preston G Garnes
- Subjects
0301 basic medicine ,Melioidosis ,030106 microbiology ,Immunology ,Median lethal dose ,auxotroph ,Article ,Microbiology ,BALB/c ,03 medical and health sciences ,Immune system ,Antigen ,Drug Discovery ,medicine ,Pharmacology (medical) ,Pathogen ,Pharmacology ,live attenuated vaccine ,Attenuated vaccine ,biology ,Burkholderia pseudomallei ,bacterial infections and mycoses ,biology.organism_classification ,medicine.disease ,030104 developmental biology ,Infectious Diseases ,bacteria ,melioidosis - Abstract
Melioidosis is an emerging disease that is caused by the facultative intracellular pathogen Burkholderia pseudomallei. It is intrinsically resistant to many antibiotics and host risk factors play a major role in susceptibility to infection. Currently, there is no human or animal vaccine against melioidosis. In this study, multiple B. pseudomallei MSHR668 deletion mutants were evaluated as live attenuated vaccines in the sensitive BALB/c mouse model of melioidosis. The most efficacious vaccines after an intraperitoneal challenge with 50-fold over the 50% median lethal dose (MLD50) with B. pseudomallei K96243 were 668 &Delta, hisF and 668 &Delta, ilvI. Both vaccines completely protected mice in the acute phase of infection and showed significant protection (50% survivors) during the chronic phase of infection. The spleens of the survivors that were examined were sterile. Splenocytes from mice vaccinated with 668 &Delta, ilvI expressed higher amounts of IFN-&gamma, after stimulation with B. pseudomallei antigens than splenocytes from mice vaccinated with less protective candidates. Finally, we demonstrate that 668 &Delta, hisF is nonlethal in immunocompromised NOD/SCID mice. Our results show that 668 &Delta, ilvI provide protective cell-mediated immune responses in the acute phase of infection and promote long term survival in the sensitive BALB/c mouse model of melioidosis.
- Published
- 2019
20. Characterization of in vitro phenotypes of Burkholderia pseudomallei and Burkholderia mallei strains potentially associated with persistent infection in mice
- Author
-
Robert C. Bernhards, Susan L. Welkos, Kei Amemiya, Patricia L. Worsham, Christopher P. Klimko, David M. Waag, and Christopher K. Cote
- Subjects
Lipopolysaccharides ,0301 basic medicine ,Spleen isolates ,Burkholderia pseudomallei ,Melioidosis ,Mouse ,Burkholderia ,030106 microbiology ,Virulence ,Burkholderia mallei ,Biochemistry ,Microbiology ,Cell Line ,Mice ,03 medical and health sciences ,Genetics ,medicine ,Animals ,Molecular Biology ,Mice, Inbred BALB C ,Original Paper ,biology ,Macrophages ,Glanders ,General Medicine ,biology.organism_classification ,medicine.disease ,Antimicrobial ,Chronic infection ,Phenotype ,Female ,Spleen - Abstract
Burkholderia pseudomallei (Bp) and Burkholderia mallei (Bm), the agents of melioidosis and glanders, respectively, are Tier 1 biothreats. They infect humans and animals, causing disease ranging from acute and fatal to protracted and chronic. Chronic infections are especially challenging to treat, and the identification of in vitro phenotypic markers which signal progression from acute to persistent infection would be extremely valuable. First, a phenotyping strategy was developed employing colony morphotyping, chemical sensitivity testing, macrophage infection, and lipopolysaccharide fingerprint analyses to distinguish Burkholderia strains. Then mouse spleen isolates collected 3–180 days after infection were characterized phenotypically. Isolates from long-term infections often exhibited increased colony morphology differences and altered patterns of antimicrobial sensitivity and macrophage infection. Some of the Bp and Bm persistent infection isolates clearly displayed enhanced virulence in mice. Future studies will evaluate the potential role and significance of these phenotypic markers in signaling the establishment of a chronic infection. Electronic supplementary material The online version of this article (doi:10.1007/s00203-016-1303-8) contains supplementary material, which is available to authorized users.
- Published
- 2016
- Full Text
- View/download PDF
21. AOAC SMPR® 2016.008
- Author
-
Luther E. Lindler, Malcolm Johns, David Watson, Sanjiv Shah, Ken Gage, Mark Scheckelhoff, Patricia L. Worsham, Linda Beck, Shanmuga Sozhamannan, Jessica Appler, Jennifer Gibbons, Matthew Lesho, Jeff Ballin, Matthew Davenport, Robert Bull, Julie Boylan, Traci Pals, Timothy J. Bauer, Kenneth Damer, Ryan Cahall, Scott G Coates, Christina Egan, Ted L. Hadfield, Michael Retford, David M. Wagner, Laura Maple, and Robert D. Perry
- Subjects
Aerosols ,Pharmacology ,Information retrieval ,Yersinia pestis ,Computer science ,Environmental Microbiology ,Environmental Chemistry ,United States Department of Defense ,Agronomy and Crop Science ,United States ,Food Science ,Analytical Chemistry - Published
- 2016
- Full Text
- View/download PDF
22. Disease progression in mice exposed to low-doses of aerosolized clinical isolates of Burkholderia pseudomallei
- Author
-
Christopher K. Cote, David P Fetterer, Matthew C. Reed, Sharon P. Daye, Sergei S. Biryukov, Steven J. Kern, Kristen A. Fritts, Kei Amemiya, Ronald G. Toothman, Melissa G. Hunter, Christopher P. Klimko, Carl Soffler, Avery V. Quirk, Susan L. Welkos, Jennifer L. Shoe, Joel A. Bozue, Sylvia R. Trevino, Norman Kreiselmeier, Lara J. Kohler, Jennifer L. Dankmeyer, Michael J. Aponte-Cuadrado, Patricia L. Worsham, Joshua R. Meyer, Christopher W. Schellhase, and David M. Waag
- Subjects
0301 basic medicine ,Bacterial Diseases ,Melioidosis ,Burkholderia pseudomallei ,Physiology ,Disease ,Pathology and Laboratory Medicine ,Pathogenesis ,White Blood Cells ,0302 clinical medicine ,Animal Cells ,Immune Physiology ,Medicine and Health Sciences ,Immune Response ,Materials ,Mice, Inbred BALB C ,Innate Immune System ,Multidisciplinary ,biology ,Virulence ,T Cells ,Animal Models ,Thailand ,medicine.anatomical_structure ,Infectious Diseases ,Experimental Organism Systems ,Physical Sciences ,Disease Progression ,Medicine ,Cytokines ,Female ,Cellular Types ,Research Article ,Science ,Immune Cells ,Immunology ,Materials Science ,Spleen ,Bronchi ,Mouse Models ,Research and Analysis Methods ,Microbiology ,03 medical and health sciences ,Immune system ,Signs and Symptoms ,Model Organisms ,Diagnostic Medicine ,medicine ,Animals ,Humans ,Inflammation ,Aerosols ,Blood Cells ,Australia ,Biology and Life Sciences ,Cell Biology ,Molecular Development ,medicine.disease ,biology.organism_classification ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Immunoglobulin G ,Immune System ,Mixtures ,Antibody Formation ,Animal Studies ,Bacteria ,030215 immunology ,Developmental Biology - Abstract
Mouse models have been essential to generate supporting data for the research of infectious diseases. Burkholderia pseudomallei, the etiological agent of melioidosis, has been studied using mouse models to investigate pathogenesis and efficacy of novel medical countermeasures to include both vaccines and therapeutics. Previous characterization of mouse models of melioidosis have demonstrated that BALB/c mice present with an acute infection, whereas C57BL/6 mice have shown a tendency to be more resistant to infection and may model chronic disease. In this study, either BALB/c or C57BL/6 mice were exposed to aerosolized human clinical isolates of B. pseudomallei. The bacterial strains included HBPUB10134a (virulent isolate from Thailand), MSHR5855 (virulent isolate from Australia), and 1106a (relatively attenuated isolate from Thailand). The LD50 values were calculated and serial sample collections were performed in order to examine the bacterial burdens in tissues, histopathological features of disease, and the immune response mounted by the mice after exposure to aerosolized B. pseudomallei. These data will be important when utilizing these models for testing novel medical countermeasures. Additionally, by comparing highly virulent strains with attenuated isolates, we hope to better understand the complex disease pathogenesis associated with this bacterium.
- Published
- 2018
23. Characterization of Tetratricopeptide Repeat-Like Proteins in Francisella tularensis and Identification of a Novel Locus Required for Virulence
- Author
-
Sherry Mou, Joel A. Bozue, Adela Straskova, Todd K. Kijek, Petra Spidlova, Jiri Stulik, Vera Dankova, Lucie Balonova, Lukas Cerveny, Barbora Szotáková, Patricia L. Worsham, Christopher K. Cote, and Daniela Putzova
- Subjects
Virulence Factors ,Immunology ,Mutant ,Virulence ,Microbiology ,Virulence factor ,Tularemia ,Gene Knockout Techniques ,Cytosol ,Bacterial Proteins ,medicine ,Animals ,Francisella tularensis ,Gene ,Genetics ,Mice, Inbred BALB C ,biology ,Macrophages ,Bacterial Infections ,medicine.disease ,biology.organism_classification ,Disease Models, Animal ,Mutagenesis, Insertional ,Tetratricopeptide ,Infectious Diseases ,Genetic Loci ,Francisella ,Female ,Parasitology - Abstract
Francisella tularensis is a highly infectious bacterium that causes the potentially lethal disease tularemia. This extremely virulent bacterium is able to replicate in the cytosolic compartments of infected macrophages. To invade macrophages and to cope with their intracellular environment, Francisella requires multiple virulence factors, which are still being identified. Proteins containing tetratricopeptide repeat (TPR)-like domains seem to be promising targets to investigate, since these proteins have been reported to be directly involved in virulence-associated functions of bacterial pathogens. Here, we studied the role of the FTS_0201 , FTS_0778 , and FTS_1680 genes, which encode putative TPR-like proteins in Francisella tularensis subsp. holarctica FSC200. Mutants defective in protein expression were prepared by TargeTron insertion mutagenesis. We found that the locus FTS_1680 and its ortholog FTT_0166c in the highly virulent Francisella tularensis type A strain SchuS4 are required for proper intracellular replication, full virulence in mice, and heat stress tolerance. Additionally, the FTS_1680 -encoded protein was identified as a membrane-associated protein required for full cytopathogenicity in macrophages. Our study thus identifies FTS_1680/FTT_0166c as a new virulence factor in Francisella tularensis .
- Published
- 2014
- Full Text
- View/download PDF
24. Development of real-time PCR assays for specific detection of hmsH, hmsF, hmsR, and irp2 located within the 102-kb pgm locus of Yersinia pestis
- Author
-
Timothy D. Minogue, Patricia L. Worsham, Charla E. Gaddy, Laurie J. Hartman, Pedro F. Cuevas, and Gerald B. Howe
- Subjects
Genetics ,Virulence ,biology ,Yersinia pestis ,Membrane Proteins ,Reproducibility of Results ,Locus (genetics) ,Cell Biology ,Chromosomes, Bacterial ,Real-Time Polymerase Chain Reaction ,biology.organism_classification ,Polymerase Chain Reaction ,Pathogenicity island ,Microbiology ,Bacterial Proteins ,Gene Order ,Chromosomal region ,TaqMan ,Yersinia pseudotuberculosis ,Yersinia enterocolitica ,Iron Regulatory Protein 2 ,Molecular Biology ,Bacterial Outer Membrane Proteins - Abstract
Virulent isolates of three pathogenic Yersinia species (Yersinia pestis, Yersinia pseudotuberculosis, and Yersinia enterocolitica) harbor a 102-kb chromosomal region which encodes elements critical for virulence. A 35-kb high pathogenicity island is contained in this region, is a known virulence determinant, contains irp1 and irp2 iron-regulating genes. An additional segment, the 68-kb high pathogenicity island, contains genetic elements responsible for conferring the Y. pestis pigmentation phenotype on Congo red agar at 28 °C. Collectively, these contiguous segments are referred to as the pigmentation (pgm) locus, the absence of which results in strain attenuation and exemption from CDC Select Agent status. In this study, we developed a set of four real-time PCR assays to detect the presence or absence of multiple virulence genes located within this region. Specifically, we designed TaqMan(®) PCR assays to individually detect three hemin storage genes (hmsH, hmsF, and hmsR) which are genetic elements that confer the pigmentation phenotype, as well as the iron-regulating status of 25 Y. pestis isolates (representing 23 different strains), thus establishing a molecular based assay capable of determining the pgm status of candidate Y. pestis isolates. Included in the validation process, was a comparison of these real-time PCR assays and newly developed conventional PCR assays targeting much larger areas of the 102-kb region (including one assay spanning hmsR and hmsF, one spanning hmsH and hsmF, one targeting hmsF, and one targeting irp2). There was high concordance between the conventional and real-time PCR assays for all Y. pestis strains tested. The results from the comparative analysis document the specificity and sensitivity of the real-time PCR assays and further solidify the ostensible benefits of real-time PCR over conventional PCR.
- Published
- 2014
- Full Text
- View/download PDF
25. The D-alanyl-d-alanine carboxypeptidase enzyme is essential for virulence in the Schu S4 strain of Francisella tularensis and a dacD mutant is able to provide protection against a pneumonic challenge
- Author
-
Sherry Mou, Beth A. Bachert, Sharon P. Daye, Kathleen A. Kuehl, Janice A. Williams, Joel A. Bozue, Todd M. Kijek, and Patricia L. Worsham
- Subjects
0301 basic medicine ,Penicillin binding proteins ,Virulence Factors ,030106 microbiology ,Mutant ,Virulence ,Vaccines, Attenuated ,medicine.disease_cause ,Microbiology ,Cell Line ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Bacterial Proteins ,Escherichia coli ,medicine ,Animals ,Penicillin-Binding Proteins ,Francisella tularensis ,Lung ,Tularemia ,Mice, Inbred BALB C ,Attenuated vaccine ,biology ,Escherichia coli Proteins ,Macrophages ,biology.organism_classification ,Serine-Type D-Ala-D-Ala Carboxypeptidase ,Complementation ,Disease Models, Animal ,030104 developmental biology ,Infectious Diseases ,chemistry ,Bacterial Vaccines ,Mutation ,Peptidoglycan - Abstract
Low molecular mass penicillin binding proteins (LMM PBP) are bacterial enzymes involved in the final steps of peptidoglycan biosynthesis. In Escherichia coli, most LMM PBP exhibit dd-carboxypeptidase activity, are not essential for growth in routine laboratory media, and contributions to virulent phenotypes remain largely unknown. The Francisella tularensis Schu S4 genome harbors the dacD gene (FTT_1029), which encodes a LMM PBP with homology to PBP6b of E. coli. Disruption of this locus in the fully virulent Schu S4 strain resulted in a mutant that could not grow in Chamberlain's Defined Medium and exhibited severe morphological defects. Further characterization studies demonstrated that the growth defects of the dacD mutant were pH-dependent, and could be partially restored by growth at neutral pH or fully restored by genetic complementation. Infection of murine macrophage-like cells showed that the Schu S4 dacD mutant is capable of intracellular replication. However, this mutant was attenuated in BALB/c mice following intranasal challenge (LD50 = 603 CFU) as compared to mice challenged with the parent (LD50 = 1 CFU) or complemented strain (LD50 = 1 CFU). Additionally, mice that survived infection with the dacD mutant showed significant protection against subsequent challenge with the parent strain. Collectively, these results indicate that the DacD protein of F. tularensis is essential for growth in low pH environments and virulence in vivo. These results also suggest that a PBP mutant could serve as the basis of a novel, live attenuated vaccine strain.
- Published
- 2019
- Full Text
- View/download PDF
26. In Vitro Antibiotic Susceptibilities of Yersinia pestis Determined by Broth Microdilution following CLSI Methods
- Author
-
Lynda Miller, Patricia L. Worsham, Bret K. Purcell, Stephanie Halasohoris, Charles L. Marchand, Jeremy R. Hershfield, and Henry S. Heine
- Subjects
Yersinia pestis ,medicine.drug_class ,Antibiotics ,Antibiotic susceptibilities ,Cephalosporin ,Population ,Colony Count, Microbial ,Microbial Sensitivity Tests ,Biology ,Microbiology ,Drug Resistance, Bacterial ,polycyclic compounds ,medicine ,Humans ,Pharmacology (medical) ,education ,Pharmacology ,Plague ,education.field_of_study ,Broth microdilution ,Temperature ,biochemical phenomena, metabolism, and nutrition ,biology.organism_classification ,In vitro ,Anti-Bacterial Agents ,Infectious Diseases ,Antibiotic Agents ,Susceptibility - Abstract
In vitro susceptibilities to 45 antibiotics were determined for 30 genetically and geographically diverse strains of Yersinia pestis by the broth microdilution method at two temperatures, 28°C and 35°C, following Clinical and Laboratory Standards Institute (CLSI) methods. The Y. pestis strains demonstrated susceptibility to aminoglycosides, quinolones, tetracyclines, β-lactams, cephalosporins, and carbapenems. Only a 1-well shift was observed for the majority of antibiotics between the two temperatures. Establishing and comparing antibiotic susceptibilities of a diverse but specific set of Y. pestis strains by standardized methods and establishing population ranges and MIC 50 and MIC 90 values provide reference information for assessing new antibiotic agents and also provide a baseline for use in monitoring any future emergence of resistance.
- Published
- 2015
- Full Text
- View/download PDF
27. Characterization of pathogenesis of and immune response to Burkholderia pseudomallei K96243 using both inhalational and intraperitoneal infection models in BALB/c and C57BL/6 mice
- Author
-
Joel A. Bozue, Susan L. Welkos, Ronald G. Toothman, Wendy M. Webster, Kei Amemiya, Chris H. Weaver, Christopher P. Klimko, Melissa G. Hunter, Avery V. Quirk, Jennifer L. Shoe, Jeremy J. Bearss, Patricia L. Worsham, Christopher K. Cote, David P. Fetterer, Jennifer L. Dankmeyer, and Kristen A. Fritts
- Subjects
0301 basic medicine ,Melioidosis ,Burkholderia pseudomallei ,Physiology ,Antibiotics ,Colony Count, Microbial ,lcsh:Medicine ,Drug resistance ,NK cells ,Pathology and Laboratory Medicine ,Monocytes ,Body Temperature ,White Blood Cells ,Mice ,Animal Cells ,Immune Physiology ,Medicine and Health Sciences ,lcsh:Science ,Immune Response ,Lung ,Mice, Inbred BALB C ,Multidisciplinary ,Animal Models ,medicine.anatomical_structure ,Experimental Organism Systems ,Physiological Parameters ,Liver ,Physical Sciences ,Cytokines ,Female ,Cellular Types ,Injections, Intraperitoneal ,Research Article ,medicine.drug_class ,Materials by Structure ,Immune Cells ,030106 microbiology ,Immunology ,Materials Science ,Spleen ,Mouse Models ,Biology ,Research and Analysis Methods ,Microbiology ,BALB/c ,03 medical and health sciences ,Immune system ,Antibiotic resistance ,Signs and Symptoms ,Model Organisms ,Species Specificity ,Diagnostic Medicine ,Administration, Inhalation ,medicine ,Animals ,Humans ,Inflammation ,Aerosols ,Blood Cells ,Body Weight ,lcsh:R ,Biology and Life Sciences ,Cell Biology ,medicine.disease ,biology.organism_classification ,Bacterial Load ,Immunity, Innate ,Lymphocyte Subsets ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Mixtures ,lcsh:Q ,Granulocytes - Abstract
Burkholderia pseudomallei, the etiologic agent of melioidosis, is a Gram negative bacterium designated as a Tier 1 threat. This bacterium is known to be endemic in Southeast Asia and Northern Australia and can infect humans and animals by several routes. Inhalational melioidosis has been associated with monsoonal rains in endemic areas and is also a significant concern in the biodefense community. There are currently no effective vaccines for B. pseudomallei and antibiotic treatment can be hampered by non-specific symptomology and also the high rate of naturally occurring antibiotic resistant strains. Well-characterized animal models will be essential when selecting novel medical countermeasures for evaluation prior to human clinical trials. Here, we further characterize differences between the responses of BALB/c and C57BL/6 mice when challenged with low doses of a low-passage and well-defined stock of B. pseudomallei K96243 via either intraperitoneal or aerosol routes of exposure. Before challenge, mice were implanted with a transponder to collect body temperature readings, and daily body weights were also recorded. Mice were euthanized on select days for pathological analyses and determination of the bacterial burden in selected tissues (blood, lungs, liver, and spleen). Additionally, spleen homogenate and sera samples were analyzed to better characterize the host immune response after infection with aerosolized bacteria. These clinical, pathological, and immunological data highlighted and confirmed important similarities and differences between these murine models and exposure routes.
- Published
- 2017
28. Bacillus anthracis comparative genome analysis in support of the Amerithrax investigation
- Author
-
Matthew N. Van Ert, Adam M. Phillippy, Steven L. Salzberg, Mark R. Wilson, Jacques Ravel, Timothy D. Read, Mihai Pop, Richard J. Langham, Scott T. Stanley, Terry G. Abshire, Patricia L. Worsham, Lingxia Jiang, R. Scott Decker, Jason D. Bannan, Leo J. Kenefic, David A. Rasko, Paul Keim, and Claire M. Fraser-Liggett
- Subjects
Whole genome sequencing ,Comparative genomics ,Genetics ,Multidisciplinary ,biology ,DNA Mutational Analysis ,Forensic Sciences ,Genome-wide association study ,Biological Sciences ,biology.organism_classification ,Bioterrorism ,Genome ,Bacillus anthracis ,Ames strain ,Genetic Loci ,Mutation ,Genotype ,Humans ,Genotyping ,Genome, Bacterial ,Genome-Wide Association Study - Abstract
Before the anthrax letter attacks of 2001, the developing field of microbial forensics relied on microbial genotyping schemes based on a small portion of a genome sequence. Amerithrax, the investigation into the anthrax letter attacks, applied high-resolution whole-genome sequencing and comparative genomics to identify key genetic features of the letters’ Bacillus anthracis Ames strain. During systematic microbiological analysis of the spore material from the letters, we identified a number of morphological variants based on phenotypic characteristics and the ability to sporulate. The genomes of these morphological variants were sequenced and compared with that of the B. anthracis Ames ancestor, the progenitor of all B. anthracis Ames strains. Through comparative genomics, we identified four distinct loci with verifiable genetic mutations. Three of the four mutations could be directly linked to sporulation pathways in B. anthracis and more specifically to the regulation of the phosphorylation state of Spo0F, a key regulatory protein in the initiation of the sporulation cascade, thus linking phenotype to genotype. None of these variant genotypes were identified in single-colony environmental B. anthracis Ames isolates associated with the investigation. These genotypes were identified only in B. anthracis morphotypes isolated from the letters, indicating that the variants were not prevalent in the environment, not even the environments associated with the investigation. This study demonstrates the forensic value of systematic microbiological analysis combined with whole-genome sequencing and comparative genomics.
- Published
- 2011
- Full Text
- View/download PDF
29. A strategy to verify the absence of the pgm locus in Yersinia pestis strain candidates for select agent exemption
- Author
-
Susan L. Welkos, Amy L. Jenkins, and Patricia L. Worsham
- Subjects
Microbiology (medical) ,Virulence Factors ,Yersinia pestis ,Virulence ,Locus (genetics) ,Select agent ,Biology ,Polymerase Chain Reaction ,Microbiology ,Bacterial Proteins ,Humans ,Molecular Biology ,Pcr analysis ,health care economics and organizations ,DNA Primers ,Genetics ,Plague ,fungi ,Pigments, Biological ,biology.organism_classification ,Enterobacteriaceae ,United States ,humanities ,Investigation methods ,Mutation ,United States Dept. of Health and Human Services - Abstract
Yersinia pestis is a Department of Health and Human Services select agent as defined in federal regulations. Certain attenuated strains of Y. pestis, such as the pgm(-) strain, are exempt from these regulations. Herein we describe a strategy to verify the absence of the pgm locus in Y. pestis strains being considered as candidates for select agent exemption by PCR analysis of virulence-associated genes.
- Published
- 2009
- Full Text
- View/download PDF
30. CpG oligodeoxynucleotides augment the murine immune response to the Yersinia pestis F1-V vaccine in bubonic and pneumonic models of plague
- Author
-
Sarah L. Norris, Anthony D. Bassett, Bradford S. Powell, Patricia L. Worsham, Randy L. Fast, Jeffrey J. Adamovicz, Arthur M. Krieg, Kei Amemiya, Jennifer L. Meyers, and Taralyn E. Rogers
- Subjects
Pore Forming Cytotoxic Proteins ,Yersinia pestis ,Ratón ,CpG Oligodeoxynucleotide ,Biology ,Bubonic plague ,Mice ,Immune system ,Adjuvants, Immunologic ,Bacterial Proteins ,medicine ,Animals ,Antigens, Bacterial ,Mice, Inbred BALB C ,Plague ,Plague Vaccine ,Vaccines, Synthetic ,General Veterinary ,General Immunology and Microbiology ,Vaccination ,Public Health, Environmental and Occupational Health ,Yersiniosis ,hemic and immune systems ,respiratory system ,medicine.disease ,biology.organism_classification ,Antibodies, Bacterial ,Virology ,Toll-Like Receptor 2 ,Infectious Diseases ,Oligodeoxyribonucleotides ,Immunology ,Molecular Medicine ,Plague vaccine ,Female - Abstract
The current U.S. Department of Defense candidate plague vaccine is a fusion between two Yersinia pestis proteins: the F1 capsular protein, and the low calcium response (Lcr) V-protein. We hypothesized that an immunomodulator, such as CpG oligodeoxynucleotide (ODN)s, could augment the immune response to the plague F1-V vaccine in a mouse model for plague. CpG ODNs significantly augmented the antibody response and efficacy of a single dose of the plague vaccine in murine bubonic and pneumonic models of plague. In the latter study, we also found an overall significant augmentation the immune response to the individual subunits of the plague vaccine by CpG ODN 2006. In a long-term, prime-boost study, CpG ODN induced a significant early augmentation of the IgG response to the vaccine. The presence of CpG ODN induced a significant increase in the IgG2a subclass response to the vaccine up to 5 months after the boost. Our studies showed that CpG ODNs significantly augmented the IgG antibody response to the plague vaccine, which increased the probability of survival in murine models of plague ( P
- Published
- 2009
- Full Text
- View/download PDF
31. Comparison of the early host immune response to two widely diverse virulent strains of Burkholderia pseudomallei that cause acute or chronic infections in BALB/c mice
- Author
-
David P Fetterer, Susan L. Welkos, Patricia L. Worsham, Christopher K. Cote, Jennifer L. Dankmeyer, and Kei Amemiya
- Subjects
Serum ,Chemokine ,Melioidosis ,Burkholderia pseudomallei ,Virulence ,Spleen ,Microbiology ,BALB/c ,Young Adult ,Immune system ,medicine ,Animals ,Humans ,Asia, Southeastern ,Mice, Inbred BALB C ,Innate immune system ,biology ,Australia ,biochemical phenomena, metabolism, and nutrition ,bacterial infections and mycoses ,biology.organism_classification ,medicine.disease ,Virology ,Bacterial Load ,Disease Models, Animal ,Infectious Diseases ,medicine.anatomical_structure ,Chronic Disease ,biology.protein ,Leukocytes, Mononuclear ,bacteria ,Cytokines ,Female - Abstract
Burkholderia pseudomallei is the etiologic agent of melioidosis, which is endemic in Southeast Asia and Northern Australia. We previously found by the intraperitoneal (IP) route that we could discern differences in virulence in mice amongst different strains of B. pseudomallei. We report an early immune response study comparing two strains in our collection which represent the least, B. pseudomallei 1106a, and one of the most, HBPUB10134a, virulent strains in BALB/c mice. B. pseudomallei HBPUB10134a infected mouse spleens contained a 2-3 log higher bacterial burden than mice infected with B. pseudomallei 1106a 3 days post-infection (PI). More and higher amounts of inflammatory cytokines/chemokines were detected in sera and spleen extracts from B. pseudomallei HBPUB10134a than B. pseudomallei 1106a infected mice. The most prominent were IFNγ, IL-1α, IL-1β, IL-6, IL-10, IP-10, and MIG. After 7 days PI, there was a decrease in bacterial burden in spleens from 1106a infected mice and a decrease in cytokines/chemokines in sera and spleen extracts from both sets of mice. By day 14 PI we saw an increase in monocytes/macrophages, NK cells, and granulocytes in spleens from both sets of mice. No B. pseudomallei HBPUB10134a infected mice survived after this time. In summary, B. pseudomallei HBPUB10134a was more virulent and induced host innate immune responses typical of a more acute-type infection than did B. pseudomallei 1106a which produced a more chronic infection in mice.
- Published
- 2015
32. Characterization of Burkholderia pseudomallei Strains Using a Murine Intraperitoneal Infection Model and In Vitro Macrophage Assays
- Author
-
Susan L. Welkos, Patricia L. Worsham, Kei Amemiya, David M. Waag, Christopher K. Cote, Christopher P. Klimko, Steven J. Kern, Joel A. Bozue, Sylvia R. Trevino, Jeremy J. Bearss, and Robert C. Bernhards
- Subjects
Lipopolysaccharides ,Melioidosis ,Burkholderia pseudomallei ,medicine.drug_class ,Science ,Antibiotics ,Virulence ,Microbiology ,Mice ,Antibiotic resistance ,In vivo ,medicine ,Macrophage ,Animals ,Multidisciplinary ,biology ,Intracellular parasite ,Macrophages ,medicine.disease ,biology.organism_classification ,Virology ,Abscess ,Disease Models, Animal ,Phenotype ,Medicine ,Research Article - Abstract
Burkholderia pseudomallei, the etiologic agent of melioidosis, is a gram-negative facultative intracellular bacterium. This bacterium is endemic in Southeast Asia and Northern Australia and can infect humans and animals by several routes. It has also been estimated to present a considerable risk as a potential biothreat agent. There are currently no effective vaccines for B. pseudomallei, and antibiotic treatment can be hampered by nonspecific symptomology, the high incidence of naturally occurring antibiotic resistant strains, and disease chronicity. Accordingly, there is a concerted effort to better characterize B. pseudomallei and its associated disease. Before novel vaccines and therapeutics can be tested in vivo, a well characterized animal model is essential. Previous work has indicated that mice may be a useful animal model. In order to develop standardized animal models of melioidosis, different strains of bacteria must be isolated, propagated, and characterized. Using a murine intraperitoneal (IP) infection model, we tested the virulence of 11 B. pseudomallei strains. The IP route offers a reproducible way to rank virulence that can be readily reproduced by other laboratories. This infection route is also useful in distinguishing significant differences in strain virulence that may be masked by the exquisite susceptibility associated with other routes of infection (e.g., inhalational). Additionally, there were several pathologic lesions observed in mice following IP infection. These included varisized abscesses in the spleen, liver, and haired skin. This model indicated that commonly used laboratory strains of B. pseudomallei (i.e., K96243 and 1026b) were significantly less virulent as compared to more recently acquired clinical isolates. Additionally, we characterized in vitro strain-associated differences in virulence for macrophages and described a potential inverse relationship between virulence in the IP mouse model of some strains and in the macrophage phagocytosis assay. Strains which were more virulent for mice (e.g., HBPU10304a) were often less virulent in the macrophage assays, as determined by several parameters such as intracellular bacterial replication and host cell cytotoxicity.
- Published
- 2015
33. Complete Genome Sequence of Yersinia pestis Strains Antiqua and Nepal516: Evidence of Gene Reduction in an Emerging Pathogen
- Author
-
Ping Hu, Lisa M. Vergez, May C. Chu, Patricia L. Worsham, Stephanie Malfatti, Gary L. Andersen, Frank W. Larimer, Patrick S. G. Chain, and Lyndsay Radnedge
- Subjects
Genetics ,Whole genome sequencing ,Genomics and Proteomics ,Phylogenetic tree ,Yersinia pestis ,Biovar ,Molecular Sequence Data ,Virulence ,Biology ,biology.organism_classification ,Polymorphism, Single Nucleotide ,Microbiology ,Genome ,Open Reading Frames ,Bacterial Proteins ,Species Specificity ,Yersinia pseudotuberculosis ,Insertion sequence ,Molecular Biology ,Gene Deletion ,Genome, Bacterial - Abstract
Yersinia pestis , the causative agent of bubonic and pneumonic plagues, has undergone detailed study at the molecular level. To further investigate the genomic diversity among this group and to help characterize lineages of the plague organism that have no sequenced members, we present here the genomes of two isolates of the “classical” antiqua biovar, strains Antiqua and Nepal516. The genomes of Antiqua and Nepal516 are 4.7 Mb and 4.5 Mb and encode 4,138 and 3,956 open reading frames, respectively. Though both strains belong to one of the three classical biovars, they represent separate lineages defined by recent phylogenetic studies. We compare all five currently sequenced Y. pestis genomes and the corresponding features in Yersinia pseudotuberculosis . There are strain-specific rearrangements, insertions, deletions, single nucleotide polymorphisms, and a unique distribution of insertion sequences. We found 453 single nucleotide polymorphisms in protein-coding regions, which were used to assess the evolutionary relationships of these Y. pestis strains. Gene reduction analysis revealed that the gene deletion processes are under selective pressure, and many of the inactivations are probably related to the organism's interaction with its host environment. The results presented here clearly demonstrate the differences between the two biovar antiqua lineages and support the notion that grouping Y. pestis strains based strictly on the classical definition of biovars (predicated upon two biochemical assays) does not accurately reflect the phylogenetic relationships within this species. A comparison of four virulent Y. pestis strains with the human-avirulent strain 91001 provides further insight into the genetic basis of virulence to humans.
- Published
- 2006
- Full Text
- View/download PDF
34. Design and Testing for a Nontagged F1-V Fusion Protein as Vaccine Antigen against Bubonic and Pneumonic Plague
- Author
-
Leonard A. Smith, Gerard P. Andrews, Harry B. Hines, Jeffrey J. Adamovicz, Jeffrey K. Pullen, Patricia L. Worsham, Jeffrey T. Enama, Wilson J. Ribot, David G. Heath, Bradford S. Powell, Chris R. Bolt, and Scott Jendrek
- Subjects
Pore Forming Cytotoxic Proteins ,Pneumonic plague ,Yersinia pestis ,Recombinant Fusion Proteins ,Protein subunit ,Drug Evaluation, Preclinical ,Protein Engineering ,Microbiology ,Mice ,Bacterial Proteins ,Antigen ,medicine ,Animals ,Humans ,Expressed Sequence Tags ,Antigens, Bacterial ,Plague ,Vaccines, Synthetic ,biology ,Edman degradation ,Protein primary structure ,biology.organism_classification ,medicine.disease ,Fusion protein ,Treatment Outcome ,Drug Design ,Vaccines, Subunit ,Plague vaccine ,Female ,Biotechnology - Abstract
A two-component recombinant fusion protein antigen was re-engineered and tested as a medical counter measure against the possible biological threat of aerosolized Yersinia pestis. The active component of the proposed subunit vaccine combines the F1 capsular protein and V virulence antigen of Y. pestis and improves upon the design of an earlier histidine-tagged fusion protein. In the current study, different production strains were screened for suitable expression and a purification process was optimized to isolate an F1-V fusion protein absent extraneous coding sequences. Soluble F1-V protein was isolated to 99% purity by sequential liquid chromatography including capture and refolding of urea-denatured protein via anion exchange, followed by hydrophobic interaction, concentration, and then transfer into buffered saline for direct use after frozen storage. Protein identity and primary structure were verified by mass spectrometry and Edman sequencing, confirming a purified product of 477 amino acids and removal of the N-terminal methionine. Purity, quality, and higher-order structure were compared between lots using RP-HPLC, intrinsic fluorescence, CD spectroscopy, and multi-angle light scattering spectroscopy, all of which indicated a consistent and properly folded product. As formulated with aluminum hydroxide adjuvant and administered in a single subcutaneous dose, this new F1-V protein also protected mice from wild-type and non-encapsulated Y. pestis challenge strains, modeling prophylaxis against pneumonic and bubonic plague. These findings confirm that the fusion protein architecture provides superior protection over the former licensed product, establish a foundation from which to create a robust production process, and set forth assays for the development of F1-V as the active pharmaceutical ingredient of the next plague vaccine.
- Published
- 2005
- Full Text
- View/download PDF
35. Genome plasticity in Yersinia pestis The GenBank accession numbers for the sequences reported in this paper can be found in Table 1 T1 ; the GenBank accession number for DFR4 is AF426171
- Author
-
Gary L. Andersen, Lyndsay Radnedge, Patricia L. Worsham, and Peter G. Agron
- Subjects
Comparative genomics ,Genetics ,biology ,Yersinia pestis ,Biovar ,Yersinia pseudotuberculosis ,Bacterial genome size ,Restriction fragment length polymorphism ,Insertion sequence ,biology.organism_classification ,Microbiology ,Genome - Abstract
Yersinia pestis, the causative agent of bubonic plague, emerged recently (
- Published
- 2002
- Full Text
- View/download PDF
36. Genetic Variability of Yersinia pestis Isolates as Predicted by PCR-Based IS 100 Genotyping and Analysis of Structural Genes Encoding Glycerol-3-Phosphate Dehydrogenase ( glpD )
- Author
-
Vladimir L. Motin, Emilio Garcia, Warren Regala, Bahrad A. Sokhansanj, Linda L. Ott, Tomas R. Slezak, Jeffrey M. Elliott, Ping Hu, Anca M. Georgescu, Robert R. Brubaker, and Patricia L. Worsham
- Subjects
DNA, Bacterial ,Glycerol ,Genotype ,Yersinia pestis ,Biovar ,Molecular Sequence Data ,Glycerolphosphate Dehydrogenase ,Yersinia ,Polymerase Chain Reaction ,Microbiology ,Bacterial Proteins ,Sequence Homology, Nucleic Acid ,Genetic variation ,Molecular Biology ,Genotyping ,Molecular Biology of Pathogens ,Genetics ,Base Sequence ,Phylogenetic tree ,biology ,Escherichia coli Proteins ,Structural gene ,food and beverages ,Genetic Variation ,biology.organism_classification ,Fructose-Bisphosphatase ,Mutagenesis, Insertional ,Genes, Bacterial ,DNA Transposable Elements ,bacteria ,DNA Probes - Abstract
A PCR-based genotyping system that detects divergence of IS 100 locations within the Yersinia pestis genome was used to characterize a large collection of isolates of different biovars and geographical origins. Using sequences derived from the glycerol-negative biovar orientalis strain CO92, a set of 27 locus-specific primers was designed to amplify fragments between the end of IS 100 and its neighboring gene. Geographically diverse members of the orientalis biovar formed a homogeneous group with identical genotype with the exception of strains isolated in Indochina. In contrast, strains belonging to the glycerol-positive biovar antiqua showed a variety of fingerprinting profiles. Moreover, strains of the biovar medievalis (also glycerol positive) clustered together with the antiqua isolates originated from Southeast Asia, suggesting their close phylogenetic relationships. Interestingly, a Manchurian biovar antiqua strain Nicholisk 51 displayed a genotyping pattern typical of biovar orientalis isolates. Analysis of the glycerol pathway in Y. pestis suggested that a 93-bp deletion within the glpD gene encoding aerobic glycerol-3-phosphate dehydrogenase might account for the glycerol-negative phenotype of the orientalis biovar. The glpD gene of strain Nicholisk 51 did not possess this deletion, although it contained two nucleotide substitutions characteristic of the glpD version found exclusively in biovar orientalis strains. To account for this close relationship between biovar orientalis strains and the antiqua Nicholisk 51 isolate, we postulate that the latter represents a variant of this biovar with restored ability to ferment glycerol. The fact that such a genetic lesion might be repaired as part of the natural evolutionary process suggests the existence of genetic exchange between different Yersinia strains in nature. The relevance of this observation on the emergence of epidemic Y. pestis strains is discussed.
- Published
- 2002
- Full Text
- View/download PDF
37. Plague
- Author
-
James R. Swearengen and Patricia L. Worsham
- Published
- 2014
- Full Text
- View/download PDF
38. Identification and Characterization of Variable-Number Tandem Repeats in the Yersinia pestis Genome
- Author
-
Patricia L. Worsham, James M. Schupp, Jane C. Y. Wong, Paul Keim, A. M. Klevytska, and Lance B. Price
- Subjects
DNA, Bacterial ,Microbiology (medical) ,Genetics ,Phylogenetic tree ,Yersinia pestis ,Biovar ,Minisatellite Repeat ,Genetic Variation ,Bacteriology ,Minisatellite Repeats ,Biology ,Multiple Loci VNTR Analysis ,bacterial infections and mycoses ,biology.organism_classification ,Polymerase Chain Reaction ,Genome ,Nucleotide diversity ,Variable number tandem repeat ,Tandem repeat ,Animals ,Humans ,Genome, Bacterial ,Phylogeny ,Plasmids - Abstract
Yersinia pestis , the infamous plague-causing pathogen, appears to have emerged in relatively recent history. Evidence of this fact comes from several studies that document a lack of nucleotide diversity in the Y. pestis genome. In contrast, we report that variable-number tandem repeat (VNTR) sequences are common in the Y. pestis genome and occur frequently in gene coding regions. Larger tandem repeat arrays, most useful for phylogenetic analysis, are present at an average of 2.18 arrays per 10 kbp and are distributed evenly throughout the genome and the two virulence plasmids, pCD1 and pMT1. We examined allelic diversity at 42 chromosomal VNTR loci in 24 selected isolates (12 globally distributed and 12 from Siskiyou County, Calif.). Vast differences in diversity were observed among the 42 VNTR loci, ranging from 2 to 11 alleles. We found that the maximum copy number of repeats in an array was highly correlated with diversity ( R = 0.86). VNTR-based phylogenetic analysis of the 24 strains successfully grouped isolates from biovar orientalis and most of the antiqua and mediaevalis strains. Hence, multiple-locus VNTR analysis (MLVA) appears capable of both distinguishing closely related strains and successfully classifying more distant relationships. Harnessing the power of MLVA to establish standardized databases will enable researchers to better understand plague ecology and evolution around the world.
- Published
- 2001
- Full Text
- View/download PDF
39. Identification of Nucleotide Sequences for the Specific and Rapid Detection of Yersinia pestis
- Author
-
Gary L. Andersen, Paula McCready, Silvia Gamez-Chin, Patricia L. Worsham, and Lyndsay Radnedge
- Subjects
DNA, Bacterial ,Time Factors ,Yersinia pestis ,Molecular Sequence Data ,Public Health Microbiology ,Yersinia ,Polymerase Chain Reaction ,Applied Microbiology and Biotechnology ,law.invention ,Nucleic acid thermodynamics ,Species Specificity ,law ,medicine ,Animals ,Polymerase chain reaction ,DNA Primers ,Genetics ,Base Sequence ,Ecology ,biology ,Nucleic Acid Hybridization ,Yersiniosis ,biology.organism_classification ,medicine.disease ,Virology ,Enterobacteriaceae ,Suppression subtractive hybridization ,Primer (molecular biology) ,Food Science ,Biotechnology - Abstract
Suppression subtractive hybridization, a cost-effective approach for targeting unique DNA, was used to identify a 41.7-kb Yersinia pestis -specific region. One primer pair designed from this region amplified PCR products from natural isolates of Y. pestis and produced no false positives for near neighbors, an important criterion for unambiguous bacterial identification.
- Published
- 2001
- Full Text
- View/download PDF
40. Isolation of an asporogenic (spoOA) protective antigen-producing strain of Bacillus anthracis
- Author
-
Patricia L Worsham and Michele R Sowers
- Subjects
Immunology ,Genetics ,General Medicine ,Molecular Biology ,Applied Microbiology and Biotechnology ,Microbiology - Abstract
We found that Congo red agar allows identification of sporulation-deficient Bacillus anthracis. Using Congo red agar, we isolated an asporogenic derivative of the protective antigen-producing strain B. anthracis deltaSterne-1(pPA102). Polymerase chain reaction and Southern hybridization analyses of DNA from the asporogenic mutant revealed that a deletion was present in spoOA, an essential gene for the initiation of sporulation. The deletion also encompassed the spoIVB homologue and a portion of the recN homologue. The avirulent spoOA strain deltaSterne-1(pPA102)CR4 is suitable for the safe production of protective antigen without endospore contamination of the vaccine production facility.Key words: Bacillus anthracis, protective antigen, spoOA, vaccine, Congo red.
- Published
- 1999
- Full Text
- View/download PDF
41. A Yersinia pestis tat mutant is attenuated in bubonic and small-aerosol pneumonic challenge models of infection but not as attenuated by intranasal challenge
- Author
-
Krishna Moody, Todd M. Bell, Christopher K. Cote, David Fritz, Patricia L. Worsham, Todd K. Kijek, Sherry Mou, Amy L. Jenkins, Joel A. Bozue, Camenzind G. Robinson, Jeffrey R. Kugelman, Taylor B. Chance, and Steven J. Kern
- Subjects
Signal peptide ,Yersinia pestis ,Science ,Mutant ,Veterinary Microbiology ,Virulence ,Pathogenesis ,Cell morphology ,Pathology and Laboratory Medicine ,Microbiology ,Bacterial genetics ,Mice ,Bacterial Proteins ,Pneumonia, Bacterial ,Medicine and Health Sciences ,Animals ,Aerosols ,Plague ,Multidisciplinary ,biology ,Biofilm ,Membrane Transport Proteins ,Biology and Life Sciences ,Bacteriology ,biology.organism_classification ,Veterinary Bacteriology ,Complementation ,Disease Models, Animal ,Veterinary Diseases ,Medical Microbiology ,Medicine ,Veterinary Science ,Gene Deletion ,Research Article - Abstract
Bacterial proteins destined for the Tat pathway are folded before crossing the inner membrane and are typically identified by an N-terminal signal peptide containing a twin arginine motif. Translocation by the Tat pathway is dependent on the products of genes which encode proteins possessing the binding site of the signal peptide and mediating the actual translocation event. In the fully virulent CO92 strain of Yersinia pestis, the tatA gene was deleted. The mutant was assayed for loss of virulence through various in vitro and in vivo assays. Deletion of the tatA gene resulted in several consequences for the mutant as compared to wild-type. Cell morphology of the mutant bacteria was altered and demonstrated a more elongated form. In addition, while cultures of the mutant strain were able to produce a biofilm, we observed a loss of adhesion of the mutant biofilm structure compared to the biofilm produced by the wild-type strain. Immuno-electron microscopy revealed a partial disruption of the F1 antigen on the surface of the mutant. The virulence of the ΔtatA mutant was assessed in various murine models of plague. The mutant was severely attenuated in the bubonic model with full virulence restored by complementation with the native gene. After small-particle aerosol challenge in a pneumonic model of infection, the mutant was also shown to be attenuated. In contrast, when mice were challenged intranasally with the mutant, very little difference in the LD50 was observed between wild-type and mutant strains. However, an increased time-to-death and delay in bacterial dissemination was observed in mice infected with the ΔtatA mutant as compared to the parent strain. Collectively, these findings demonstrate an essential role for the Tat pathway in the virulence of Y. pestis in bubonic and small-aerosol pneumonic infection but less important role for intranasal challenge.
- Published
- 2014
42. Two stable variants of Burkholderia pseudomallei strain MSHR5848 express broadly divergent in vitro phenotypes associated with their virulence differences
- Author
-
Robert C. Bernhards, Jeffrey W. Koehler, Sean Lovett, Ronald G. Toothman, Christopher P. Klimko, Joel A. Bozue, Jason T. Ladner, Susan L. Welkos, Steven J. Kern, David P Fetterer, G. I. Koroleva, Gustavo Palacios, Patricia L. Worsham, Mark Wolcott, David A. Rozak, C. J. Chase, Christopher K. Cote, and April A. Shea
- Subjects
Bacterial Diseases ,0301 basic medicine ,Burkholderia pseudomallei ,lcsh:Medicine ,Pathology and Laboratory Medicine ,Mice ,White Blood Cells ,Ribotyping ,Animal Cells ,Antibiotics ,Medicine and Health Sciences ,lcsh:Science ,Staining ,Mice, Inbred BALB C ,Multidisciplinary ,Virulence ,biology ,Antimicrobials ,Drugs ,Phenotype ,Bacterial Pathogens ,Membrane Staining ,Phenotypes ,Chemistry ,Infectious Diseases ,Medical Microbiology ,Physical Sciences ,Cellular Types ,Pathogens ,Research Article ,Chemical Elements ,Virulence Factors ,Burkholderia ,Immune Cells ,Immunology ,030106 microbiology ,Research and Analysis Methods ,Microbiology ,Cell Line ,03 medical and health sciences ,Microbial Control ,Drug Resistance, Bacterial ,Genetics ,Animals ,Microbial Pathogens ,Gene ,Pharmacology ,Polymorphism, Genetic ,Blood Cells ,Bacteria ,Macrophages ,lcsh:R ,Organisms ,Correction ,Biology and Life Sciences ,Phenotype microarray ,Cell Biology ,biology.organism_classification ,030104 developmental biology ,Genes, Bacterial ,Specimen Preparation and Treatment ,Burkholderia Infection ,Multilocus sequence typing ,lcsh:Q ,Gram Staining ,Sulfur - Abstract
Burkholderia pseudomallei (Bp), the agent of melioidosis, causes disease ranging from acute and rapidly fatal to protracted and chronic. Bp is highly infectious by aerosol, can cause severe disease with nonspecific symptoms, and is naturally resistant to multiple antibiotics. However, no vaccine exists. Unlike many Bp strains, which exhibit random variability in traits such as colony morphology, Bp strain MSHR5848 exhibited two distinct and relatively stable colony morphologies on sheep blood agar plates: a smooth, glossy, pale yellow colony and a flat, rough, white colony. Passage of the two variants, designated "Smooth" and "Rough", under standard laboratory conditions produced cultures composed of > 99.9% of the single corresponding type; however, both could switch to the other type at different frequencies when incubated in certain nutritionally stringent or stressful growth conditions. These MSHR5848 derivatives were extensively characterized to identify variant-associated differences. Microscopic and colony morphology differences on six differential media were observed and only the Rough variant metabolized sugars in selective agar. Antimicrobial susceptibilities and lipopolysaccharide (LPS) features were characterized and phenotype microarray profiles revealed distinct metabolic and susceptibility disparities between the variants. Results using the phenotype microarray system narrowed the 1,920 substrates to a subset which differentiated the two variants. Smooth grew more rapidly in vitro than Rough, yet the latter exhibited a nearly 10-fold lower lethal dose for mice than Smooth. Finally, the Smooth variant was phagocytosed and replicated to a greater extent and was more cytotoxic than Rough in macrophages. In contrast, multiple locus sequence type (MLST) analysis, ribotyping, and whole genome sequence analysis demonstrated the variants' genetic conservation; only a single consistent genetic difference between the two was identified for further study. These distinct differences shown by two variants of a Bp strain will be leveraged to better understand the mechanism of Bp phenotypic variability and to possibly identify in vitro markers of infection.
- Published
- 2017
- Full Text
- View/download PDF
43. Relationship Between Virulence and Immunity as Revealed in Recent Studies of the Fl Capsule of Yersinia pestis
- Author
-
Gerard P. Andrews, David G. Heath, Arthur M. Friedlander, James Estep, George W. Anderson, Patricia L. Worsham, Susan L. Welkos, Kelly J. Davis, and Margaret L. Pitt
- Subjects
Microbiology (medical) ,Bacterial capsule ,Virulence ,Biology ,medicine.disease ,Active immunization ,biology.organism_classification ,Bubonic plague ,Virology ,Microbiology ,Infectious Diseases ,Plasmid ,Yersinia pestis ,Immunity ,medicine ,Plague vaccine - Abstract
Yersinia pestis, the causative agent of plague, possesses multiple virulence determinants encoded on its three plasmids and on its chromosome. We evaluated the role of the protein capsule F1 in virulence an immunity against plague. Strains lacking F1, either those that are naturally occurring or those with genetically defined nonpolar mutations in the structural gene, retained their virulence for mice and nonhuman primates. However, both active immunization with F1, from either a recombinant vector or Y. pestis, and passive immunization with F1 monoclonal antibody protected mice from experimental infection with wild-type F1-positive organisms. These results suggest that protective immunogens like F1 need not be essential for virulence. The rare isolation of virulent F1-negative organisms from F1-immunized animals infected with F1-positive strains supports this conclusion and also suggests that, in addition to F1, an optimal vaccine against plague should include essential virulence factors as immunogens.
- Published
- 1995
- Full Text
- View/download PDF
44. Virulence of Yersinia pseudotuberculosis in aerosol models
- Author
-
Patricia L, Worsham, Sherry, Mou, Christopher K, Cote, and David, Fritz
- Subjects
Aerosols ,Mice ,Virulence ,Yersinia pseudotuberculosis ,Rats, Inbred BN ,Guinea Pigs ,Models, Animal ,Animals ,Rats - Published
- 2012
45. Cytotoxic necrotizing factor is an effective immunogen in a Yersinia pseudotuberculosis aerosol mouse model
- Author
-
Sherry, Mou, Christopher K, Cote, and Patricia L, Worsham
- Subjects
Aerosols ,Disease Models, Animal ,Mice ,Bacterial Toxins ,Vaccination ,Animals ,Yersinia pseudotuberculosis Infections - Published
- 2012
46. Cytotoxic Necrotizing Factor Is an Effective Immunogen in a Yersinia pseudotuberculosis Aerosol Mouse Model
- Author
-
Sherry Mou, Patricia L. Worsham, and Christopher K. Cote
- Subjects
Bordetella bronchiseptica ,Virulence ,Biology ,medicine.disease_cause ,biology.organism_classification ,Actin cytoskeleton ,Virology ,Microbiology ,Bordetella ,medicine ,Yersinia pseudotuberculosis ,Secretion ,Pathogen ,Escherichia coli - Abstract
There appears to be a link between bacteria producing certain Rho-activating toxins and respiratory disease. This class of toxins includes the cytotoxic necrotizing factor (CNF) of Escherichia coli, the dermonecrotic toxin (DNT) of certain Bordetella sp., and the CNF homologue of Yersinia pseudotuberculosis (CNFY). The host targets for these toxins are the GTP-binding proteins of the Rho family that function in a number of signaling pathways, including the modification of actin regulators. The toxins’ effect on Rho activity is a result of deamidation of the Rho proteins, leading to constitutive activation of Rho GTPases that are key regulators of the actin cytoskeleton. Because the cytoskeleton is involved in migration of phagocytes, phagocytosis, and the secretion of cytokines, this permanent activation of Rho proteins impairs host immune responses. Necrotoxic (CNF+) E. coli strains are associated with necrotizing pneumonia. Another member of this toxin family, DNT, is essential for virulence in a porcine pneumonia model (Bordetella bronchiseptica) and appears to be responsible for the severe lung damage in laboratory mice infected with this pathogen. The CNFY homologue is present in all strains of Y. pseudotuberculosis and Y. pestis that have been examined to date, although Y. pestis and many strains of Y. pseudotuberculosis do not produce the toxin due to a truncation in the cnf locus. We found that a Y. pseudotuberculosis strain expressing CNFY was virulent in the aerosol mouse model, while the CNF-negative isogenic strain did not cause mortality even at 100 LD50. We expressed CNFY in E. coli and vaccinated mice with the purified toxoided protein. Vaccinated animals were protected from >600 aerosol LD50 of a virulent CNFY+ strain but were not protected from a virulent strain carrying the truncated toxin gene. Thus, although CNFY is not required for aerosol virulence of all Y. pseudotuberculosis strains, it is an effective immunogen for CNFY+ strains. The Rho-activating toxin family may be an attractive target for development of medical countermeasures for necrotizing pneumonias.
- Published
- 2012
- Full Text
- View/download PDF
47. Virulence of Yersinia pseudotuberculosis in Aerosol Models
- Author
-
David Fritz, Sherry Mou, Patricia L. Worsham, and Christopher K. Cote
- Subjects
Serotype ,Yersinia pestis ,biology ,Strain (chemistry) ,Toxin ,medicine ,Virulence ,Yersinia pseudotuberculosis ,biology.organism_classification ,medicine.disease_cause ,Pathogenicity island ,Gene ,Microbiology - Abstract
Genetic analyses have determined that Yersinia pestis and Yersinia pseudotuberculosis are so closely related that they are a single species. Transitional strains of Y. pestis (also known as “Pestoides” strains) are ancient strains that lie very close to Y. pseudotuberculosis in the Y. pestis lineage. We previously demonstrated that the ability to infect mice by aerosol is exhibited by the Pestoides group and that the aerosol LD50 of these strains is comparable to that of “typical” isolates. In this study, we examined several Y. pseudotuberculosis strains of different serotypes in the mouse aerosol model and evaluated a subset of these in the guinea pig and rat. All of the strains of Y. pseudotuberculosis caused a fatal pneumonia in the aerosol mouse model. However, the range of virulence that we observed among natural isolates of Y. pseudotuberculosis was quite remarkable when compared to natural isolates of Y. pestis (including the Pestoides group), which have relatively uniform LD50 values in this model. For one strain of Y. pseudotuberculosis, the toxin cytotoxic necrotizing factor (CNF) was required for aerosol virulence. However, it appears that this is strain-specific as other naturally occurring strains carrying a deletion in CNF retained virulence. In general, isolates with the high pathogenicity island were more virulent than those lacking it. In our limited testing in guinea pigs, we found a similar variability in the virulence of Y. pseudotuberculosis strains, while rats were highly resistant to aerosols of the single Y. pseudotuberculosis strain evaluated to date. Although we have observed considerable variation in the virulence of Y. pestis natural isolates by parenteral routes of infection, LD50 values by aerosol do not vary like those of Y. pseudotuberculosis. This likely reflects the much greater genetic diversity of Y. pseudotuberculosis. A recent study indicates that gene acquisition from other species is a major factor that has influenced Y. pseudotuberculosis genome evolution. This surely played (and continues to play) a role in producing the constellation of virulence determinants observed in this organism and makes it likely that there are multiple independent mechanisms that contribute to pathogenicity by the aerosol route.
- Published
- 2012
- Full Text
- View/download PDF
48. Novel plasmids and resistance phenotypes in Yersinia pestis: unique plasmid inventory of strain Java 9 mediates high levels of arsenic resistance
- Author
-
Nicholas J. Vietri, Sherry Mou, Mark Eppinger, Grant Severson, Jacques Ravel, Gary L. Andersen, Patricia L. Worsham, and Lyndsay Radnedge
- Subjects
Transposable element ,Bacterial Diseases ,Genome evolution ,Arsenites ,Yersinia pestis ,Biovar ,Virulence ,lcsh:Medicine ,Microbiology ,Arsenic ,Mice ,Plasmid ,Drug Resistance, Bacterial ,Animals ,Yersinia enterocolitica ,lcsh:Science ,Biology ,Genetics ,Plague ,Multidisciplinary ,biology ,lcsh:R ,Computational Biology ,Genomics ,biology.organism_classification ,Rats ,Mutagenesis, Insertional ,Phenotype ,Infectious Diseases ,Genes, Bacterial ,Conjugation, Genetic ,DNA Transposable Elements ,Medicine ,lcsh:Q ,Mobilome ,Plasmids ,Research Article - Abstract
Growing evidence suggests that the plasmid repertoire of Yersinia pestis is not restricted to the three classical virulence plasmids. The Java 9 strain of Y. pestis is a biovar Orientalis isolate obtained from a rat in Indonesia. Although it lacks the Y. pestis-specific plasmid pMT, which encodes the F1 capsule, it retains virulence in mouse and non-human primate animal models. While comparing diverse Y. pestis strains using subtractive hybridization, we identified sequences in Java 9 that were homologous to a Y. enterocolitica strain carrying the transposon Tn2502, which is known to encode arsenic resistance. Here we demonstrate that Java 9 exhibits high levels of arsenic and arsenite resistance mediated by a novel promiscuous class II transposon, named Tn2503. Arsenic resistance was self-transmissible from Java 9 to other Y. pestis strains via conjugation. Genomic analysis of the atypical plasmid inventory of Java 9 identified pCD and pPCP plasmids of atypical size and two previously uncharacterized cryptic plasmids. Unlike the Tn2502-mediated arsenic resistance encoded on the Y. enterocolitica virulence plasmid; the resistance loci in Java 9 are found on all four indigenous plasmids, including the two novel cryptic plasmids. This unique mobilome introduces more than 105 genes into the species gene pool. The majority of these are encoded by the two entirely novel self-transmissible plasmids, which show partial homology and synteny to other enterics. In contrast to the reductive evolution in Y. pestis, this study underlines the major impact of a dynamic mobilome and lateral acquisition in the genome evolution of the plague bacterium.
- Published
- 2011
49. Human anti-plague monoclonal antibodies protect mice from Yersinia pestis in a bubonic plague model
- Author
-
Dimiter S. Dimitrov, Kei Amemiya, Patricia L. Worsham, Randy L. Fast, Christopher K. Cote, Jennifer L. Dankmeyer, Zhongyu Zhu, Xiaodong Xiao, and Michael M. Wormald
- Subjects
Yersinia pestis ,medicine.drug_class ,lcsh:Medicine ,Enzyme-Linked Immunosorbent Assay ,Biology ,Monoclonal antibody ,Bubonic plague ,Epitope ,Infectious Diseases/Bacterial Infections ,Mice ,03 medical and health sciences ,Antigen ,medicine ,Animals ,lcsh:Science ,030304 developmental biology ,Plague ,0303 health sciences ,Multidisciplinary ,Linear epitope ,030306 microbiology ,lcsh:R ,Antibodies, Monoclonal ,medicine.disease ,biology.organism_classification ,Virology ,3. Good health ,Disease Models, Animal ,Epitope mapping ,Immunology/Immune Response ,Immunology/Antigen Processing and Recognition ,biology.protein ,lcsh:Q ,Antibody ,Epitope Mapping ,Research Article - Abstract
Yersinia pestis is the etiologic agent of plague that has killed more than 200 million people throughout the recorded history of mankind. Antibiotics may provide little immediate relief to patients who have a high bacteremia or to patients infected with an antibiotic resistant strain of plague. Two virulent factors of Y. pestis are the capsid F1 protein and the low-calcium response (Lcr) V-protein or V-antigen that have been proven to be the targets for both active and passive immunization. There are mouse monoclonal antibodies (mAbs) against the F1- and V-antigens that can passively protect mice in a murine model of plague; however, there are no anti-Yersinia pestis monoclonal antibodies available for prophylactic or therapeutic treatment in humans. We identified one anti-F1-specific human mAb (m252) and two anti-V-specific human mAb (m253, m254) by panning a naïve phage-displayed Fab library against the F1- and V-antigens. The Fabs were converted to IgG1s and their binding and protective activities were evaluated. M252 bound weakly to peptides located at the F1 N-terminus where a protective mouse anti-F1 mAb also binds. M253 bound strongly to a V-antigen peptide indicating a linear epitope; m254 did not bind to any peptide from a panel of 53 peptides suggesting that its epitope may be conformational. M252 showed better protection than m253 and m254 against a Y, pestis challenge in a plague mouse model. A synergistic effect was observed when the three antibodies were combined. Incomplete to complete protection was achieved when m252 was given at different times post-challenge. These antibodies can be further studied to determine their potential as therapeutics or prophylactics in Y. pestis infection in humans.
- Published
- 2010
50. The role of the phoPQ operon in the pathogenesis of the fully virulent CO92 strain of Yersinia pestis and the IP32953 strain of Yersinia pseudotuberculosis
- Author
-
David Fritz, Joel A. Bozue, Sylvia R. Trevino, Patricia L. Worsham, Christopher K. Cote, Krishna Moody, and Sherry Mou
- Subjects
Yersinia Infections ,Operon ,Gastrointestinal Diseases ,Yersinia pestis ,Mutant ,Virulence ,Yersinia pseudotuberculosis Infections ,medicine.disease_cause ,Microbiology ,Mice ,Bacterial Proteins ,medicine ,Yersinia pseudotuberculosis ,Animals ,Humans ,Plague ,biology ,Macrophages ,Genetic Variation ,Pathogenic bacteria ,Gene Expression Regulation, Bacterial ,biology.organism_classification ,Virology ,Mice, Inbred C57BL ,Response regulator ,Infectious Diseases ,Mutation ,Gram-Negative Bacterial Infections ,Gene Deletion - Abstract
At the genomic level, Yersinia pestis and Yersinia pseudotuberculosis are nearly identical but cause very different diseases. Y. pestis is the etiologic agent of plague; whereas Y. pseudotuberculosis causes a gastrointestinal infection primarily after the consumption of contaminated food. In many gram-negative pathogenic bacteria, PhoP is part of a two-component global regulatory system in which PhoQ serves as the sensor kinase, and PhoP is the response regulator. PhoP is known to activate a number of genes in many bacteria related to virulence. To determine the role of the PhoPQ proteins in Yersinia infections, primarily using aerosol challenge models, the phoP gene was deleted from the chromosome of the CO92 strain of Y. pestis and the IP32953 strain of Y. pseudotuberculosis, leading to a polar mutation of the phoPQ operon. We demonstrated that loss of phoPQ from both strains leads to a defect in intracellular growth and/or survival within macrophages. These in vitro data would suggest that the phoPQ mutants would be attenuated in vivo. However, the LD(50) for the Y. pestis mutant did not differ from the calculated LD(50) for the wild-type CO92 strain for either the bubonic or pneumonic murine models of infection. In contrast, mice challenged by aerosol with the Y. pseudotuberculosis mutant had a LD(50) value 40× higher than the wild-type strain. These results demonstrate that phoPQ are necessary for full virulence by aerosol infection with the IP32953 strain of Y. pseudotuberculosis. However, the PhoPQ proteins do not play a significant role in infection with a fully virulent strain of Y. pestis.
- Published
- 2010
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.