95 results on '"Thomas A. Ficht"'
Search Results
2. Evaluation of the safety profile of the vaccine candidate Brucella melitensis 16MΔvjbR strain in goats
- Author
-
Angela M. Arenas-Gamboa, Estefanía Maurizio, Martha E. Hensel, Allison C. Rice-Ficht, Daniel G. Garcia-Gonzalez, M. Raquel Castaño-Zubieta, Carlos A. Rossetti, and Thomas A. Ficht
- Subjects
Saliva ,Offspring ,030231 tropical medicine ,Brucella Vaccine ,Sheep Diseases ,Physiology ,Brucella ,Article ,Brucellosis ,03 medical and health sciences ,0302 clinical medicine ,Pregnancy ,Lactation ,Brucella melitensis ,medicine ,Animals ,Humans ,030212 general & internal medicine ,Sheep ,General Veterinary ,General Immunology and Microbiology ,biology ,business.industry ,Goats ,Public Health, Environmental and Occupational Health ,biology.organism_classification ,medicine.disease ,Vaccination ,Infectious Diseases ,medicine.anatomical_structure ,Molecular Medicine ,Gestation ,Female ,business - Abstract
Small ruminant brucellosis is caused by the Gram negative cocci-bacillus Brucella (B.) melitensis, the most virulent Brucella species for humans. In goats and sheep, middle to late-term gestation abortion, stillbirths and the delivery of weak infected offspring are the characteristic clinical signs of the disease. Vaccination with the currently available Rev. 1 vaccine is the best option to prevent and control the disease, although it is far from ideal. In this study, we investigate the safety of the B. melitensis 16MΔvjbR strain during a 15-month period beginning at vaccination of young goats, impregnation, delivery and lactation. Forty, 4 to 6 months old, healthy female crossbreed goats were randomly divided into four groups (n = 10) and immunized subcutaneously with a single vaccine dose containing 1x109 CFU of B. melitensis 16MΔvjbR delivered in alginate microcapsules or non-encapsulated. Controls received empty capsules or the commercially available Rev.1 vaccine. Seven months post-vaccination, when animals were sexually mature, all goats were naturally bred using brucellosis-free males, and allowed to carry pregnancies to term. Blood samples to assess the humoral immune response were collected throughout the study. At two months post-delivery, all dams and their offspring were euthanized and a necropsy was performed to collect samples for bacteriology and histology. Interestingly, none of the animals that received the vaccine candidate regardless of the formulation exhibited any clinical signs associated with vaccination nor shed the vaccine strain through saliva, vagina or the milk. Gross and histopathologic changes in all nannies and offspring were unremarkable with no evidence of tissue colonization or vertical transmission to fetuses. Altogether, these data demonstrate that vaccination with the mutant strain 16MΔvjbR is safe for use in the non-pregnant primary host.
- Published
- 2021
- Full Text
- View/download PDF
3. Brucella activates the host RIDD pathway to subvert BLOS1-directed immune defense
- Author
-
Kelsey Michelle Wells, Kai He, Aseem Pandey, Ana Cabello, Dongmei Zhang, Jing Yang, Gabriel Gomez, Yue Liu, Haowu Chang, Xueqiang Li, Hao Zhang, Xuehuang Feng, Luciana Fachini da Costa, Richard Metz, Charles D Johnson, Cameron Lee Martin, Jill Skrobarczyk, Luc R Berghman, Kristin L Patrick, Julian Leibowitz, Allison Ficht, Sing-Hoi Sze, Jianxun Song, Xiaoning Qian, Qing-Ming Qin, Thomas A Ficht, and Paul de Figueiredo
- Subjects
General Immunology and Microbiology ,General Neuroscience ,General Medicine ,General Biochemistry, Genetics and Molecular Biology - Abstract
The phagocytosis and destruction of pathogens in lysosomes constitute central elements of innate immune defense. Here, we show that Brucella, the causative agent of brucellosis, the most prevalent bacterial zoonosis globally, subverts this immune defense pathway by activating regulated IRE1α-dependent decay (RIDD) of Bloc1s1 mRNA encoding BLOS1, a protein that promotes endosome–lysosome fusion. RIDD-deficient cells and mice harboring a RIDD-incompetent variant of IRE1α were resistant to infection. Inactivation of the Bloc1s1 gene impaired the ability to assemble BLOC-1-related complex (BORC), resulting in differential recruitment of BORC-related lysosome trafficking components, perinuclear trafficking of Brucella-containing vacuoles (BCVs), and enhanced susceptibility to infection. The RIDD-resistant Bloc1s1 variant maintains the integrity of BORC and a higher-level association of BORC-related components that promote centrifugal lysosome trafficking, resulting in enhanced BCV peripheral trafficking and lysosomal destruction, and resistance to infection. These findings demonstrate that host RIDD activity on BLOS1 regulates Brucella intracellular parasitism by disrupting BORC-directed lysosomal trafficking. Notably, coronavirus murine hepatitis virus also subverted the RIDD–BLOS1 axis to promote intracellular replication. Our work establishes BLOS1 as a novel immune defense factor whose activity is hijacked by diverse pathogens.
- Published
- 2022
- Full Text
- View/download PDF
4. A metabolically engineered bacterium controls autoimmunity by remodeling the pro-inflammatory microenvironment
- Author
-
Jugal Kishore Das, Fengguang Guo, Carrie Hunt, Shelby Steinmeyer, Julia A Plocica, Koichi S. Kobayashi, Arul Jayaraman, Thomas A Ficht, Robert C. Alaniz, Paul de Figueiredo, and Jianxun Song
- Abstract
Immunotherapy has led to impressive advances in the treatment of autoimmune and pro-inflammatory disorders; yet, its clinical outcomes remain limited by a variety of factors including the pro-inflammatory microenvironment (IME). Discovering effective immunomodulatory agents, and the mechanisms by which they control disease, will lead to innovative strategies for enhancing the effectiveness of current immunotherapeutic approaches. We have metabolically engineered an attenuated bacterial strain (i.e., Brucella melitensis 16M ΔvjbR, BmΔvjbR) to produce indole, a tryptophan metabolite that controls the fate and function of regulatory T cells (Tregs). We demonstrated that treatment with this strain polarized M2 macrophages (Mφ) which produced anti-inflammatory cytokines (e.g., IL-10) and promoted Treg function; moreover, when combined with adoptive cell transfer (ACT) of Tregs, a single treatment with our engineered bacterial strain dramatically reduced the incidence and score of autoimmune arthritis and decreased joint damage. These findings show how a metabolically engineered bacterium can constitute a powerful vehicle for improving the efficacy of immunotherapy, defeating autoimmunity and reducing inflammation by remodeling the IME and augmenting Treg function.
- Published
- 2022
- Full Text
- View/download PDF
5. Author response: Brucella activates the host RIDD pathway to subvert BLOS1-directed immune defense
- Author
-
Kelsey Michelle Wells, Kai He, Aseem Pandey, Ana Cabello, Dongmei Zhang, Jing Yang, Gabriel Gomez, Yue Liu, Haowu Chang, Xueqiang Li, Hao Zhang, Xuehuang Feng, Luciana Fachini da Costa, Richard Metz, Charles D Johnson, Cameron Lee Martin, Jill Skrobarczyk, Luc R Berghman, Kristin L Patrick, Julian Leibowitz, Allison Ficht, Sing-Hoi Sze, Jianxun Song, Xiaoning Qian, Qing-Ming Qin, Thomas A Ficht, and Paul de Figueiredo
- Published
- 2022
- Full Text
- View/download PDF
6. Protective antibody response following oral vaccination with microencapsulated Bacillus Anthracis Sterne strain 34F2 spores
- Author
-
Allison C. Rice-Ficht, Yi Xu, Walter E. Cook, Jamie Benn Felix, Thomas A. Ficht, and Sankar P. Chaki
- Subjects
lcsh:Immunologic diseases. Allergy ,0301 basic medicine ,Live attenuated vaccines ,Immunology ,02 engineering and technology ,Biology ,complex mixtures ,lcsh:RC254-282 ,Article ,Microbiology ,03 medical and health sciences ,Post vaccination ,Fasciola hepatica ,Pharmacology (medical) ,Bacillus anthracis - Sterne strain ,Pharmacology ,fungi ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,021001 nanoscience & nanotechnology ,biology.organism_classification ,In vitro ,Bacillus anthracis ,Spore ,Vaccination ,030104 developmental biology ,Infectious Diseases ,Protective antibody ,Bacterial infection ,lcsh:RC581-607 ,0210 nano-technology - Abstract
An oral vaccine against anthrax (Bacillus anthracis) is urgently needed to prevent annual anthrax outbreaks that are causing catastrophic losses in free-ranging livestock and wildlife worldwide. The Sterne vaccine, the current injectable livestock vaccine, is a suspension of live attenuated B. anthracis Sterne strain 34F2 spores (Sterne spores) in saponin. It is not effective when administered orally and individual subcutaneous injections are not a practical method of vaccination for wildlife. In this study, we report the development of a microencapsulated oral vaccine against anthrax. Evaluating Sterne spore stability at varying pH’s in vitro revealed that spore exposure to pH 2 results in spore death, confirming that protection from the gastric environment is of main concern when producing an oral vaccine. Therefore, Sterne spores were encapsulated in alginate and coated with a protein shell containing poly-L-lysine (PLL) and vitelline protein B (VpB), a non-immunogenic, proteolysis resistant protein isolated from Fasciola hepatica. Capsule exposure to pH 2 demonstrated enhanced acid gel character suggesting that alginate microcapsules provided the necessary protection for spores to survive the gastric environment. Post vaccination IgG levels in BALBc/J mouse serum samples indicated that encapsulated spores induced anti-anthrax specific responses in both the subcutaneous and the oral vaccination groups. Furthermore, the antibody responses from both vaccination routes were protective against anthrax lethal toxin in vitro, suggesting that further optimization of this vaccine formulation may result in a reliable oral vaccine that will conveniently and effectively prevent anthrax in wildlife populations.
- Published
- 2020
- Full Text
- View/download PDF
7. Vaccine Candidate Brucella melitensis 16M
- Author
-
Martha E, Hensel, Daniel G, Garcia-Gonzalez, Sankar P, Chaki, Airn, Hartwig, Paul W, Gordy, Richard, Bowen, Thomas A, Ficht, and Angela M, Arenas-Gamboa
- Subjects
Sheep ,Vaccination ,Brucella Vaccine ,Sheep Diseases ,Therapeutics and Prevention ,Vaccines, Attenuated ,veterinary vaccine development ,Brucellosis ,Disease Models, Animal ,ovine ,Pregnancy ,brucellosis ,Brucella melitensis ,Animals ,Female ,pregnancy ,Conjunctiva ,Research Article - Abstract
Brucellosis is one of the most commonly reported zoonotic disease with a worldwide distribution. Of the 12 Brucella species, Brucella melitensis is considered the most virulent and causes reproductive failure (abortions/stillbirths) in small ruminants, which can spread the disease to other animals or to humans. Vaccination of small ruminants is a key measure used to protect both human and animal health. However, the commercially available live-attenuated vaccine for Brucella melitensis Rev. 1 retains virulence and can cause disease in animals and humans. In order to evaluate the safety and efficacy in sheep, we vaccinated pregnant sheep with 16MΔvjbR. Our results indicate that 16MΔvjbR was safer for use during pregnancy, provided a similar level of protection as Rev. 1, and could be considered an improved candidate for future vaccine trials., As a natural host species for Brucella melitensis, pregnant sheep offer an ideal model to evaluate vaccine candidates for safety. B. melitensis strain Rev. 1 has been used almost exclusively to prevent brucellosis in small ruminants, but it causes abortions when given to pregnant animals. To evaluate the comparative safety of the candidate Brucella melitensis 16MΔvjbR, pregnant sheep (n = 6) were vaccinated subcutaneously with 1 × 1010 CFU/ml of 16MΔvjbR or 1 × 109 CFU/ml Rev. 1 at a highly susceptible stage of gestation (approximately 70 days). 16MΔvjbR resulted in only 1 abortion (1 of 6) compared with 4 of 6 (66.7%) abortions in the Rev. 1 cohort. The placenta was evaluated by culture to determine if vaccination resulted in colonization. As another measure of safety, effects of B. melitensis on the fetus/offspring (vertical transmission) was evaluated by culture and histopathology of fetal tissues to determine if vaccination prevented infection of the fetus. Vaccination with 16MΔvjbR resulted in less vertical transmission than Rev. 1. To determine if vaccination was efficacious and could reduce tissue colonization in sheep, the same cohort of sheep were challenged 5 weeks postpartum by conjunctival inoculation with 1 × 107 CFU/ml B. melitensis. Protection was similar between Rev. 1 and 16MΔvjbR, with no statistical difference in colonization in the target organs. Overall, the 16MΔvjbR vaccine was considered safer than Rev. 1 based on a reduced number of abortions and limited infection in the offspring. Future experiments are needed to further refine the vaccine dose to increase the safety margin and to evaluate protection in pregnant ewes. IMPORTANCE Brucellosis is one of the most commonly reported zoonotic disease with a worldwide distribution. Of the 12 Brucella species, Brucella melitensis is considered the most virulent and causes reproductive failure (abortions/stillbirths) in small ruminants, which can spread the disease to other animals or to humans. Vaccination of small ruminants is a key measure used to protect both human and animal health. However, the commercially available live-attenuated vaccine for Brucella melitensis Rev. 1 retains virulence and can cause disease in animals and humans. In order to evaluate the safety and efficacy in sheep, we vaccinated pregnant sheep with 16MΔvjbR. Our results indicate that 16MΔvjbR was safer for use during pregnancy, provided a similar level of protection as Rev. 1, and could be considered an improved candidate for future vaccine trials.
- Published
- 2020
8. Interactions between fungal hyaluronic acid and host CD44 promote internalization by recruiting host autophagy proteins to forming phagosomes
- Author
-
Rola Barhoumi Mouneimne, Shengli Ding, Kristin L. Patrick, Allison C. Rice-Ficht, Samantha L. Bell, Robert O. Watson, Xuehuan Feng, Thomas A. Ficht, Qing-Ming Qin, Paul de Figueiredo, Jing Yang, Luciana F. Costa, and Aseem Pandey
- Subjects
Cryptococcus neoformans ,Host cell surface ,0303 health sciences ,biology ,030306 microbiology ,media_common.quotation_subject ,Phagocytosis ,Autophagy ,biology.organism_classification ,Cell biology ,03 medical and health sciences ,Internalization ,Pathogen ,Intracellular ,030304 developmental biology ,media_common ,Phagosome - Abstract
SummaryPhagocytosis and autophagy play critical roles in immune defense. Cryptococcus neoformans (Cn), a fungal pathogen that causes fatal infection, subverts the host autophagy initiation complex (AIC) and its upstream regulatory proteins, to promote its phagocytosis and intracellular parasitism of host cells. The mechanisms by which the pathogen engages host AIC proteins remain obscure. Here, we show that the recruitment of host AIC proteins to forming phagosomes is dependent upon the activity of CD44, a host cell surface receptor that engages fungal hyaluronic acid (HA). This interaction elevates intracellular Ca2+ concentrations and activates CaMKKβ and its downstream target AMPKα, which results in activation of ULK1 and the recruitment of AIC components. Moreover, we demonstrate that HA-coated beads efficiently recruit AIC components to phagosomes. Taken together, these findings show that fungal HA plays a critical role in directing the internalization and productive intracellular membrane trafficking of a fungal pathogen of global importance.Graphical AbstractIn BriefDing et al. reveal that interactions between fungal hyaluronic acid (HA) and host CD44 activate a Ca2+ - CaMKKβ-AMPK-ULK1 signaling pathway that recruits autophagy initiation complex components to forming phagosomes to drive fungal internalization.HighlightsFungal HA interactions with host cells drive a novel non-canonical, ligand-induced, autophagy pathway in phagocytic cellsCryptococcus neoformans recruits host CD44, together with AIC components and regulatory proteins, to forming phagocytic cups to initiate host cell internalizationFungal HA interactions with CD44 on host cell surfaces elevate intracellular Ca2+ concentrations, leading to activation of CaMKKβA Ca2+-CaMKKβ-AMPK-ULK1 signaling axis is involved in HA and CD44 induced autophagy protein recruitment during Cn internalization
- Published
- 2020
- Full Text
- View/download PDF
9. A Tractable
- Author
-
Gabriel Gomez, Paul de Figueiredo, Allison C. Rice-Ficht, Jianwu Pei, Thomas A. Ficht, and Qing-Ming Qin
- Subjects
0301 basic medicine ,Microbiology (medical) ,Acinetobacter baumannii ,Drosophila S2 cells ,030106 microbiology ,Immunology ,lcsh:QR1-502 ,Microbiology ,lcsh:Microbiology ,03 medical and health sciences ,Cellular and Infection Microbiology ,Extracellular ,Animals ,Drosophila ,Pathogen ,030304 developmental biology ,0303 health sciences ,Cross Infection ,biology ,030306 microbiology ,Host (biology) ,Schneider 2 cells ,persistence ,Brief Research Report ,biology.organism_classification ,invasion ,host factors ,3. Good health ,Cell biology ,Anti-Bacterial Agents ,030104 developmental biology ,Infectious Diseases ,Organelle biogenesis ,Intracellular - Abstract
Acinetobacter baumannii is an important causative agent of nosocomial infections worldwide. The pathogen also readily acquires resistance to antibiotics, and pan-resistant strains have been reported. A. baumannii is widely regarded as an extracellular bacterial pathogen. However, accumulating evidence demonstrates that the pathogen can invade, survive or persist in infected mammalian cells. Unfortunately, the molecular mechanisms controlling these processes remain poorly understood. Here, we show that Drosophila S2 cells provide several attractive advantages as a model system for investigating the intracellular lifestyle of the pathogen, including susceptibility to bacterial intracellular replication and limited infection-induced host cell death. We also show that the Drosophila system can be used to rapidly identify host factors, including MAP kinase proteins, which confer susceptibility to intracellular parasitism. Finally, analysis of the Drosophila system suggested that host proteins that regulate organelle biogenesis and membrane trafficking contribute to regulating the intracellular lifestyle of the pathogen. Taken together, these findings establish a novel model system for elucidating interactions between A. baumannii and host cells, define new factors that regulate bacterial invasion or intracellular persistence, and identify subcellular compartments in host cells that interact with the pathogen.
- Published
- 2020
10. Live attenuated bacterium limits cancer resistance to CAR-T therapy by remodeling the tumor microenvironment
- Author
-
Fengguang Guo, Jugal K Das, Koichi S Kobayashi, Qing-Ming Qin, Thomas A Ficht, Robert C Alaniz, Jianxun Song, and Paul De Figueiredo
- Subjects
Pharmacology ,Cancer Research ,Receptors, Chimeric Antigen ,Bacteria ,Immunology ,adoptive ,Oncolytic and Local Immunotherapy ,Disease Models, Animal ,Mice ,Oncology ,Neoplasms ,Tumor Microenvironment ,Animals ,Humans ,Molecular Medicine ,Immunology and Allergy ,Immunotherapy ,Neoplasm Recurrence, Local - Abstract
The tumor microenvironment (TME) is characterized by the activation of immune checkpoints, which limit the ability of immune cells to attack the growing cancer. To overcome immune suppression in the clinic, antigen-expressing viruses and bacteria have been developed to induce antitumor immunity. However, the safety and targeting specificity are the main concerns of using bacteria in clinical practice as antitumor agents. In our previous studies, we have developed an attenuated bacterial strain (Brucella melitensis 16M ∆vjbR, henceforth Bm∆vjbR) for clinical use, which is safe in all tested animal models and has been removed from the select agent list by the Centers for Disease Control and Prevention. In this study, we demonstrated that Bm∆vjbR homed to tumor tissue and improved the TME in a murine model of solid cancer. In addition, live Bm∆vjbR promoted proinflammatory M1 polarization of tumor macrophages and increased the number and activity of CD8+ T cells in the tumor. In a murine colon adenocarcinoma model, when combined with adoptive transfer of tumor-specific carcinoembryonic antigen chimeric antigen receptor CD8+ T cells, tumor cell growth and proliferation was almost completely abrogated, and host survival was 100%. Taken together, these findings demonstrate that the live attenuated bacterial treatment can defeat cancer resistance to chimeric antigen receptor T-cell therapy by remodeling the TME to promote macrophage and T cell-mediated antitumor immunity.
- Published
- 2022
- Full Text
- View/download PDF
11. AOAC SMPR 2016.009:Standard Method Performance Requirements (SMPRs) for DNA-Based Methods of Detecting Brucella suis in Field-Deployable, Department of Defense Aerosol Collection Devices
- Author
-
Mikeljon P. Nikolich, Thomas R. Blank, Ryan Cahall, Katalin Kiss, Rebekah Tiller, Linda Beck, David A. Rozak, Steven C. Olsen, Shanmuga Sozhamannan, Jennifer S. Arce, Frank W. Schaefer, Francisco F. Roberto, Jeffrey T. Foster, Kenneth Damer, Scott G Coates, Thomas A. Ficht, and Rich Ozanich
- Subjects
Pharmacology ,Field (physics) ,Environmental Chemistry ,Brucella suis ,Environmental science ,Agronomy and Crop Science ,Food Science ,Analytical Chemistry ,Microbiology - Published
- 2017
- Full Text
- View/download PDF
12. Mucosal bacterial dissemination in a rhesus macaque model of experimental brucellosis
- Author
-
Thomas A. Ficht, Kasi E. Russell-Lodrigue, Stephanie Z. Killeen, and Chad J. Roy
- Subjects
Male ,0301 basic medicine ,Saliva ,030106 microbiology ,Brucellosis ,Article ,03 medical and health sciences ,biology.animal ,Brucella melitensis ,medicine ,Animals ,Primate ,Aerosols ,Mucous Membrane ,General Veterinary ,biology ,biology.organism_classification ,medicine.disease ,Macaca mulatta ,Virology ,Rhesus macaque ,030104 developmental biology ,Vagina ,Female ,Animal Science and Zoology ,Vaginal vault - Abstract
Animals were experimentally infected with Brucella melitensis via aerosol. B. melitensis was cultured from the saliva and vaginal vault of infected animals, corresponding to bacterial dissemination in other target tissues. This is the first report of bacterial dissemination to these mucosal surfaces in a non-human primate model of brucellosis.
- Published
- 2017
- Full Text
- View/download PDF
13. Brucella induction of RIDD activity promotes intracellular parasitism by subverting BLOS1-controlled immune defense
- Author
-
Paul de Figueiredo, Kelsey Wells, Kai He, Aseem Pandey, Ana-Lucia Cabello, Gabriel Gomez, Allison Rice-FIcht, Sing-hoi Sze, Xiaoning Qian, and Thomas A Ficht
- Subjects
Immunology ,Immunology and Allergy - Abstract
The intracellular bacterial pathogen and biothreat agent Brucella is the causative agent of brucellosis, a global zoonotic disease that is associated with acute and chronic symptoms in humans and animals. The mechanisms by which the pathogen colonizes host cells remain obscure. Here, we demonstrate that infection activates the regulated IRE1-dependent decay (RIDD) of mRNA encoding BLOS1 (biogenesis of lysosome-related organelles 1 subunit 1), a protein that promotes endosome-lysosome fusion. RIDD-deficient host cells as well as mice harboring a RIDD-incompetent variant of IRE1a displayed resistance to infection. Moreover, host cells carrying a RIDD resistant variant of BLOS1 displayed enhanced trafficking of the pathogen to lysosomes, and increased resistance to intracellular parasitism. Finally, cells harboring mutations in BLOS1 displayed increased susceptibility. Taken together, the data demonstrate that host RIDD activity on BLOS1 transcripts regulate Brucella intracellular parasitism by disrupting BLOS1-directed lysosomal trafficking activity, thereby promoting maintenance of the pathogen’s replicative niche in the endoplasmic reticulum.
- Published
- 2020
- Full Text
- View/download PDF
14. Bacillus anthracis Sterne Strain 34f2 Vaccine Antibody Dose Response by Subcutaneous and Oral Administration
- Author
-
Thomas A. Ficht, Allison C. Rice-Ficht, Walter E. Cook, Jamie Benn Felix, and Sankar P. Chaki
- Subjects
Anthrax vaccines ,biology ,business.industry ,fungi ,Antibody titer ,Outbreak ,biology.organism_classification ,Virology ,Bacillus anthracis ,Vaccination ,Subcutaneous injection ,Oral administration ,biology.protein ,Medicine ,Antibody ,business - Abstract
Anthrax (Bacillus anthracis) is a zoonotic disease endemic to environments worldwide. Spores, the dormant form of the bacteria, can survive for decades in nature’s harshest environments and maintain their viability to cause disease. Outbreaks are common in free-ranging livestock and wildlife, thus making anthrax an economically and ecologically important disease. The currently available vaccine to protect livestock is a suspension of B. anthracis Sterne Strain 34F2 spores in saponin (Sterne vaccine). However, it is only available as a subcutaneous injection which is an impractical method of prevention for wildlife. Oral vaccination is the ideal method for free-ranging wildlife, but the Sterne vaccine has never been thoroughly evaluated for oral administration. The current study evaluated the antibody titers induced in mice by subcutaneous or oral vaccination with three different doses of the Sterne vaccine. Results described here show a gradual increase in antibody titers at each time point following subcutaneous vaccination with all vaccine doses. In contrast, no antibody response was detected from any dose or any time point after oral vaccination. Taken together, these results suggest that the Sterne vaccine is only effective as a subcutaneous injection and that an alternate oral anthrax vaccine formulation must be developed to allow for efficient vaccination of free-ranging livestock and wildlife.
- Published
- 2019
- Full Text
- View/download PDF
15. Space: A Final Frontier for Vacuolar Pathogens
- Author
-
Thomas A. Ficht, Paul de Figueiredo, James E. Samuel, Elizabeth Di Russo Case, and Judith A. Smith
- Subjects
0301 basic medicine ,Intracellular parasite ,Lipid bilayer fusion ,Cell Biology ,Space (commercial competition) ,Biology ,Membrane Fusion ,Biochemistry ,Article ,Cell biology ,Cell membrane ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,Structural Biology ,Host-Pathogen Interactions ,Vacuoles ,Genetics ,medicine ,Compartment (development) ,Molecular Biology ,Space allocation ,Eukaryotic cell - Abstract
There is a fundamental gap in our understanding of how a eukaryotic cell apportions the limited space within its cell membrane. Upon infection, a cell competes with intracellular pathogens for control of this same precious resource. The struggle between pathogen and host provides us with an opportunity to uncover the mechanisms regulating subcel-lural space by understanding how pathogens modulate vesicular traffic and membrane fusion events to create a specialized compartment for replication. By comparing several important intracellular pathogens, we review the molecular mechanism and trafficking pathways that drive two space allocation strategies, the formation of tight and spacious pathogen-containing vacuoles. Additionally, we discuss the potential advantages of each pathogenic lifestyle, the broader implications these lifestyles might have for cellular biology and outline exciting opportunities for future investigation.
- Published
- 2016
- Full Text
- View/download PDF
16. Pathogenesis and Immunobiology of Brucellosis
- Author
-
Thomas A. Ficht, Carlos A. Rossetti, Allison C. Rice-Ficht, Paul de Figueiredo, and L. Garry Adams
- Subjects
Immune system ,biology ,Effector ,Intracellular parasite ,Antigen presentation ,Immunology ,Tissue tropism ,Brucella ,Dendritic cell ,biology.organism_classification ,Virology ,Pathology and Forensic Medicine ,Human morbidity - Abstract
This review of Brucella–host interactions and immunobiology discusses recent discoveries as the basis for pathogenesis-informed rationales to prevent or treat brucellosis. Brucella spp., as animal pathogens, cause human brucellosis, a zoonosis that results in worldwide economic losses, human morbidity, and poverty. Although Brucella spp. infect humans as an incidental host, 500,000 new human infections occur annually, and no patient-friendly treatments or approved human vaccines are reported. Brucellae display strong tissue tropism for lymphoreticular and reproductive systems with an intracellular lifestyle that limits exposure to innate and adaptive immune responses, sequesters the organism from the effects of antibiotics, and drives clinical disease manifestations and pathology. Stealthy brucellae exploit strategies to establish infection, including i) evasion of intracellular destruction by restricting fusion of type IV secretion system-dependent Brucella-containing vacuoles with lysosomal compartments, ii) inhibition of apoptosis of infected mononuclear cells, and iii) prevention of dendritic cell maturation, antigen presentation, and activation of naive T cells, pathogenesis lessons that may be informative for other intracellular pathogens. Data sets of next-generation sequences of Brucella and host time-series global expression fused with proteomics and metabolomics data from in vitro and in vivo experiments now inform interactive cellular pathways and gene regulatory networks enabling full-scale systems biology analysis. The newly identified effector proteins of Brucella may represent targets for improved, safer brucellosis vaccines and therapeutics.
- Published
- 2015
- Full Text
- View/download PDF
17. Activation of Host IRE1α-Dependent Signaling Axis Contributes the Intracellular Parasitism of
- Author
-
Aseem, Pandey, Furong, Lin, Ana L, Cabello, Luciana F, da Costa, Xuehuan, Feng, Hui-Qiang, Feng, Ming-Zhe, Zhang, Takao, Iwawaki, Allison, Rice-Ficht, Thomas A, Ficht, Paul, de Figueiredo, and Qing-Ming, Qin
- Subjects
autophagy ,inositol-requiring enzyme 1 (IRE1) ,Vesicular Transport Proteins ,Autophagy-Related Proteins ,Protein Serine-Threonine Kinases ,MAP Kinase Kinase Kinase 5 ,Microbiology ,Brucellosis ,Cell Line ,Mice ,Endoribonucleases ,Brucella melitensis ,Animals ,Autophagy-Related Protein-1 Homolog ,Phosphorylation ,unfolded protein response (UPR) ,bcl-2-Associated X Protein ,Original Research ,ULK1 ,Mice, Knockout ,JNK Mitogen-Activated Protein Kinases ,Membrane Proteins ,Drosophila melanogaster ,RAW 264.7 Cells ,bcl-2 Homologous Antagonist-Killer Protein ,Host-Pathogen Interactions ,Unfolded Protein Response ,Beclin-1 ,intracellular trafficking and replication ,Signal Transduction - Abstract
Brucella spp. are intracellular vacuolar pathogens that causes brucellosis, a worldwide zoonosis of profound importance. We previously demonstrated that the activity of host unfolded protein response (UPR) sensor IRE1α (inositol-requiring enzyme 1) and ER-associated autophagy confer susceptibility to Brucella melitensis and Brucella abortus intracellular replication. However, the mechanism by which host IRE1α regulates the pathogen intracellular lifestyle remains elusive. In this study, by employing a diverse array of molecular approaches, including biochemical analyses, fluorescence microscopy imaging, and infection assays using primary cells derived from Ern1 (encoding IRE1) conditional knockout mice, we address this gap in our understanding by demonstrating that a novel IRE1α to ULK1, an important component for autophagy initiation, signaling axis confers susceptibility to Brucella intracellular parasitism. Importantly, deletion or inactivation of key signaling components along this axis, including IRE1α, BAK/BAX, ASK1, and JNK as well as components of the host autophagy system ULK1, Atg9a, and Beclin 1, resulted in striking disruption of Brucella intracellular trafficking and replication. Host kinases in the IRE1α-ULK1 axis, including IRE1α, ASK1, JNK1, and/or AMPKα as well as ULK1, were also coordinately phosphorylated in an IRE1α-dependent fashion upon the pathogen infection. Taken together, our findings demonstrate that the IRE1α-ULK1 signaling axis is subverted by the bacterium to promote intracellular parasitism, and provide new insight into our understanding of the molecular mechanisms of intracellular lifestyle of Brucella.
- Published
- 2017
18. Vaccine safety studies of Brucella abortus S19 and S19ΔvjbR in pregnant swine
- Author
-
Omar H. Khalaf, Thomas A. Ficht, Angela M. Arenas-Gamboa, Sankar P. Chaki, Daniel G. Garcia-Gonzalez, Lance Wheeler, Allison C. Rice-Ficht, and Slim Zriba
- Subjects
Vaccine safety ,lcsh:Immunologic diseases. Allergy ,Litter (animal) ,Swine ,040301 veterinary sciences ,animal diseases ,Disease ,Brucella ,Abortion ,Brucellosis ,0403 veterinary science ,03 medical and health sciences ,Medicine ,reproductive and urinary physiology ,030304 developmental biology ,0303 health sciences ,Attenuated vaccine ,General Veterinary ,General Immunology and Microbiology ,biology ,business.industry ,Public Health, Environmental and Occupational Health ,04 agricultural and veterinary sciences ,B. abortus S19 ,B. abortus S19ΔvjbR ,medicine.disease ,biology.organism_classification ,3. Good health ,Vaccination ,Infectious Diseases ,Regular paper ,embryonic structures ,Immunology ,Molecular Medicine ,Brucella suis ,lcsh:RC581-607 ,business - Abstract
Highlights • Vaccination with Brucella abortus S19 or S19ΔvjbR in pregnant swine did not induce abortion, stillbirths or a reduction in litter size. • Gross and histopathological evaluation did not demonstrate any local or systemic side effect associated with either vaccine. • At the time of the delivery, there was no evidence of the presence of either vaccine strains in the fetuses, placentas or sows. • Both vaccine candidates are safe for use in pregnant swine., Brucellosis in swine is caused by Brucella suis, a bacterial infection of nearly worldwide distribution. Brucella suis is also transmissible to humans, dogs and cattle and is considered a reemerging disease of public health concern. To date, there is no effective vaccine for swine. This prompted us to investigate the potential use of the commercially available vaccine for cattle or the live attenuated vaccine candidate S19ΔvjbR. As the first step, we sought to study the safety of the vaccine candidates when administered in pregnant sows, since one of the major drawbacks associated with vaccination using Live Attenuated Vaccines (LAV) is the induction of abortions when administered in pregnant animals. Fifteen pregnant gilts at mid-gestation were divided into four groups and subsequently vaccinated subcutaneously using different formulations containing 2.0 ± 0.508 × 109 CFU of either S19 or S19ΔvjbR. Vaccination in pregnant animals with the vaccine candidates did not induce abortion, stillbirths or a reduction in litter size. Multiple tissues in the gilts and piglets were examined at the time of delivery to assess bacterial colonization and histopathological changes. There was no evidence of vaccine persistence in the gilts or bacterial colonization in the fetuses. Altogether, these data suggest that both vaccine candidates are safe for use in pregnant swine. Analysis of the humoral responses, specifically anti-Brucella IgG levels measured in serum, demonstrated a robust response induced by either vaccine, but of shorter duration (4–6 weeks post-inoculation) compared to that observed in cattle or experimentally infected mice. Such a transient humoral response may prove to be beneficial in cases where the vaccine is used in eradication campaigns and in the differentiation of vaccinated from infected animals. This study provides evidence to support future efficacy studies of both vaccine candidates in swine.
- Published
- 2019
- Full Text
- View/download PDF
19. Mechanisms controlling Cryptococcus Intracellular Parasitism
- Author
-
Paul de Figueiredo, Aseem Pandey, Sheng-Li Ding, Qing-Ming Qin, Rahul Gupta, Gabriel Gomez, Furong Lin, Xuehuan Feng, Luciana Fachini de Costa, Sankar P Chaki, Madhu Katepalli, Elizabeth Case, Erin Van Schaik, Tabasum Sidiq, Omar Khalaf, Angela Arenas, Koichi S Kobayashi, James E Samuel, Gonzalo Rivera, Robert Christopher Alaniz, Sing-Hoi Sze, Xiaoning Qian, William J Brown, Allison Rice-Ficht, William Russell, and Thomas A Ficht
- Subjects
Immunology ,Immunology and Allergy - Abstract
Cryptococcus neoformans (Cn) is a deadly fungal pathogen whose intracellular lifestyle is important for virulence. Host mechanisms controlling fungal phagocytosis and replication remain obscure. Here, we describe insights that have emerged from a global phosphoproteomic analysis of the host response to Cryptococcus infection. Our analysis revealed diverse host proteins that were differentially phosphorylated following fungal infection, indicating global reprogramming of host kinase signaling during this process. Notably, phagocytosis of the pathogen activated the host autophagy initiation complex (AIC) as well as regulatory components that reside upstream of this complex. Cn-containing vacuoles (CnCVs) were found to be decorated with the cell surface marker CD44, which colocalized with components of the AIC complex. Taken together, these findings suggest that associations between CD44 and AIC proteins confer susceptibility to infection, thereby implicating novel host mechanisms in regulating fungal intracellular parasitism.
- Published
- 2019
- Full Text
- View/download PDF
20. Global Reprogramming of Host Kinase Signaling in Response to Fungal Infection
- Author
-
Omar H. Khalaf, Aseem Pandey, Gonzalo M. Rivera, Thomas A. Ficht, Erin J. van Schaik, Robert C. Alaniz, Qing-Ming Qin, Luciana F. Costa, Rahul Gupta, James E. Samuel, William K. Russell, Angela Arenas, Xuehuan Feng, Paul de Figueiredo, Shengli Ding, Madhu Katepalli, Furong Lin, Sing-Hoi Sze, Koichi Kobayashi, Gabriel Gomez, Sankar P. Chaki, Elizabeth Di Russo Case, Xiaoning Qian, William J. Brown, Tabasum Sidiq, and Allison C. Rice-Ficht
- Subjects
0301 basic medicine ,Proteomics ,Host–pathogen interaction ,Virulence ,Vacuole ,Biology ,AMP-Activated Protein Kinases ,Protein Serine-Threonine Kinases ,Microbiology ,Monocytes ,Article ,Cell Line ,Fungal Proteins ,03 medical and health sciences ,Mice ,Phagocytosis ,Virology ,Autophagy ,Animals ,Autophagy-Related Protein-1 Homolog ,Pathogen ,Cryptococcus neoformans ,Macrophages ,Biological Transport ,Cryptococcosis ,biology.organism_classification ,Cell biology ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,RAW 264.7 Cells ,Coxiella burnetii ,Host-Pathogen Interactions ,Vacuoles ,Parasitology ,Female ,Apoptosis Regulatory Proteins ,Reprogramming ,Intracellular ,Signal Transduction - Abstract
Cryptococcus neoformans (Cn) is a deadly fungal pathogen whose intracellular lifestyle is important for virulence. Host mechanisms controlling fungal phagocytosis and replication remain obscure. Here, we perform a global phosphoproteomic analysis of the host response to Cryptococcus infection. Our analysis reveals numerous and diverse host proteins that are differentially phosphorylated following fungal ingestion by macrophages, thereby indicating global reprogramming of host kinase signaling. Notably, phagocytosis of the pathogen activates the host autophagy initiation complex (AIC) and the upstream regulatory components LKB1 and AMPKα, which regulate autophagy induction through their kinase activities. Deletion of Prkaa1, the gene encoding AMPKα1, in monocytes results in resistance to fungal colonization of mice. Finally, the recruitment of AIC components to nascent Cryptococcus-containing vacuoles (CnCVs) regulates the intracellular trafficking and replication of the pathogen. These findings demonstrate that host AIC regulatory networks confer susceptibility to infection and establish a proteomic resource for elucidating host mechanisms that regulate fungal intracellular parasitism.
- Published
- 2016
21. A
- Author
-
Pedro F, Soler-Lloréns, Chris R, Quance, Sara D, Lawhon, Tod P, Stuber, John F, Edwards, Thomas A, Ficht, Suelee, Robbe-Austerman, David, O'Callaghan, and Anne, Keriel
- Subjects
DNA, Bacterial ,Lipopolysaccharides ,LPS ,Ochrobactrum ,Rhamnose ,Microbiology ,Brucellosis ,Cell Line ,Mice ,Bacterial Proteins ,Zoonoses ,evolution ,Animals ,Humans ,Child ,Phylogeny ,Original Research ,Base Sequence ,Virulence ,Macrophages ,O Antigens ,Epithelial Cells ,Biological Evolution ,Brucella ,Texas ,Carbon ,frog ,Phenotype ,motility ,Genes, Bacterial ,Multigene Family ,Female ,Anura ,metabolism ,Genome, Bacterial ,HeLa Cells - Abstract
Brucella are highly infectious bacterial pathogens responsible for brucellosis, a frequent worldwide zoonosis. The Brucella genus has recently expanded from 6 to 11 species, all of which were associated with mammals; The natural host range recently expanded to amphibians after some reports of atypical strains from frogs. Here we describe the first in depth phenotypic and genetic characterization of a Brucella strains isolated from a frog. Strain B13-0095 was isolated from a Pac-Man frog (Ceratophyrus ornate) at a veterinary hospital in Texas and was initially misidentified as Ochrobactrum anthropi. We found that B13-0095 belongs to a group of early-diverging brucellae that includes Brucella inopinata strain BO1 and the B. inopinata-like strain BO2, with traits that depart significantly from those of the “classical” Brucella spp. Analysis of B13-0095 genome sequence revealed several specific features that suggest that this isolate represents an intermediate between a soil associated ancestor and the host adapted “classical” species. Like strain BO2, B13-0095 does not possess the genes required to produce the perosamine based LPS found in classical Brucella, but has a set of genes that could encode a rhamnose based O-antigen. Despite this, B13-0095 has a very fast intracellular replication rate in both epithelial cells and macrophages. Finally, another major finding in this study is the bacterial motility observed for strains B13-0095, BO1, and BO2, which is remarkable for this bacterial genus. This study thus highlights several novel characteristics in strains belonging to an emerging group within the Brucella genus. Accurate identification tools for such atypical Brucella isolates and careful evaluation of their zoonotic potential, are urgently required.
- Published
- 2016
22. Serologic response in bottlenose dolphins Tursiops truncatus infected with Brucella sp. using a dolphin-specific indirect ELISA
- Author
-
Thomas A. Ficht, Inga F. Sidor, Jenny Meegan, Roberta Pugh, Cynthia R. Smith, Tracy A. Romano, L. Garry Adams, Stephanie Venn-Watson, William Van Bonn, Eric D. Jensen, Klaus Nielsen, and J. Lawrence Dunn
- Subjects
Male ,Time Factors ,Population ,Enzyme-Linked Immunosorbent Assay ,Brucella ,Aquatic Science ,Brucellosis ,Serology ,Marine mammal ,Species Specificity ,Animals ,Serologic Tests ,education ,Ecology, Evolution, Behavior and Systematics ,education.field_of_study ,biology ,Antibody titer ,Bottlenose dolphin ,biology.organism_classification ,Bottle-Nosed Dolphin ,Titer ,Immunology ,biology.protein ,Female ,Antibody ,human activities - Abstract
Marine-origin Brucella infections and serologic evidence of exposure have been doc- umented in multiple cetacean species. A dolphin-specific indirect enzyme-linked immunosorbent assay (ELISA) was developed to screen bottlenose dolphin sera for anti-Brucella antibodies. A total of 131 serum samples collected over a 2 to 18 yr period from 6 bottlenose dolphins Tursiops truncatus with confirmed Brucella infections were analyzed for the presence and magnitude of antibody titers against marine-origin Brucella to compare individual antibody responses to various disease manifestations. Additionally, an epidemiologic serologic survey of a managed population of 64 bottlenose dolphins was performed to evaluate for the presence of antibodies and to deter- mine whether there were any clinical pathology predictors for exposure or infection. The serologic results revealed that the dolphins with Brucella-associated abortions were seronegative for 7 to 18 yr until after the abortion and maintained positive titers for several years, with 2 of 3 animals returning to seronegative status. In contrast, the dolphins with Brucella-associated pulmonary or bone lesions maintained persistent positive titers for 2 to 18 yr. The population serosurvey revealed no significant differences in antibody levels among males and females, and dolphins between the ages of 17 and 25 yr were 6.8 times more likely to be Brucella antibody positive com- pared to those that were younger or older. Seropositive dolphins did not have significant inflam- mation compared to seronegative dolphins but were more likely to have higher levels of aspartate aminotransferase and gamma-glutamyl transpeptidase. Among 16 dolphins that tested seroposi- tive, 13 (81.3%) had previously been seropositive for at least 3 to 5 yr.
- Published
- 2012
- Full Text
- View/download PDF
23. Protective Efficacy and Safety of Brucella melitensis 16MΔ mucR against Intraperitoneal and Aerosol Challenge in BALB/c Mice
- Author
-
Thomas A. Ficht, Melissa Kahl-McDonagh, Allison C. Rice-Ficht, and Angela M. Arenas-Gamboa
- Subjects
Author's Correction ,Immunology ,Brucella Vaccine ,Brucella ,Vaccines, Attenuated ,Microbiology ,Brucellosis ,BALB/c ,Mice ,Immune system ,Bacterial Proteins ,Brucella melitensis ,Animals ,Medicine ,Pathogen ,Sequence Deletion ,Aerosols ,Mice, Inbred BALB C ,biology ,business.industry ,Vaccination ,biology.organism_classification ,medicine.disease ,Virology ,Infectious Diseases ,Microbial Immunity and Vaccines ,Mutation ,Female ,Parasitology ,business - Abstract
Brucellosis is a zoonosis of nearly worldwide distribution. Vaccination against this pathogen is an important control strategy to prevent the disease. Currently licensed vaccine strains used in animals are unacceptable for human use due to undesirable side effects and modest protection. Substantial progress has been made during the past 10 years toward the development of improved vaccines for brucellosis. In part, this has been achieved by the identification and characterization of live attenuated mutants that are safer in the host but still can stimulate an adequate immune response. In the present study, the identification and characterization of the mucR mutant (BMEI 1364) as a vaccine candidate for brucellosis was conducted. BALB/c mice were vaccinated intraperitoneally at a dose of 10 5 CFU with the mutant to evaluate safety and protective efficacy against intraperitoneal and aerosol challenge. All animals vaccinated with the vaccine candidate demonstrated a statistically significant degree of protection against both intraperitoneal and aerosol challenge. Safety was revealed by the absence of Brucella associated pathological changes, including splenomegaly, hepatomegaly, or granulomatous disease. These results suggest that the 16MΔ mucR vaccine is safe, elicits a strong protective immunity, and should be considered as a promising vaccine candidate for human use.
- Published
- 2011
- Full Text
- View/download PDF
24. Amplified Fragment Length Polymorphism Reveals Specific Epigenetic Distinctions between Mycobacterium avium Subspecies paratuberculosis Isolates of Various Isolation Types
- Author
-
Sangeeta Khare, Thomas A. Ficht, Allison C. Rice-Ficht, Allen J. Roussel, B. O'Shea, L. G. Adams, and Patricia E. Klein
- Subjects
Microbiology (medical) ,Genotype ,Sequence analysis ,Cattle Diseases ,Paratuberculosis ,Genetic analysis ,Epigenesis, Genetic ,Clinical Veterinary Microbiology ,Microbiology ,Feces ,Consensus sequence ,medicine ,Animals ,Cluster Analysis ,Humans ,Amplified Fragment Length Polymorphism Analysis ,Genetics ,biology ,biology.organism_classification ,medicine.disease ,Mycobacterium avium subspecies paratuberculosis ,Bacterial Typing Techniques ,Mycobacterium avium subsp. paratuberculosis ,Restriction site ,Restriction enzyme ,Cattle ,Amplified fragment length polymorphism - Abstract
Amplified fragment length polymorphism (AFLP) was employed as a genetic analysis tool for the study of the genetic relatedness of Mycobacterium avium subsp. paratuberculosis isolates harvested from bovine fecal samples and from bovine or human tissues. This analysis revealed genetic differences between these two isolate types that were confirmed through cluster analysis. Dendrogram analysis separated these two isolate types based on the isolation scheme (tissue-associated versus fecal M. avium subsp. paratuberculosis isolates). Further sequence analysis of unique genetic regions from each isolation type revealed no genetic sequence differences. However, Clustal DNA alignments identified AFLP restriction enzyme sites that were undigested in the tissue-associated isolates. AFLP analysis also disclosed that the same AFLP restriction sites were digested in all of the fecal isolates. Sequence analysis further revealed a consensus sequence upstream of the undigested restriction sites for possible methyltransferase recognition in the tissue-associated M. avium subsp. paratuberculosis isolates.
- Published
- 2011
- Full Text
- View/download PDF
25. Quantitative Yeast Genetic Interaction Profiling of Bacterial Effector Proteins Uncovers a Role for the Human Retromer in Salmonella Infection
- Author
-
Samantha L. Bell, Robert O. Watson, Jiewei Xu, Tao Jing, Sara Talmage, Pingwei Li, Pengbiao Xu, Jason A. Wojcechowskyj, Ana L. Cabello, James E. Samuel, Thomas A. Ficht, Kristin L. Patrick, David Jimenez-Morales, Michael Shales, Morgan N. Riba, Nevan J. Krogan, and Paul de Figueiredo
- Subjects
0301 basic medicine ,Retromer ,host-pathogen interaction ,Mice ,Yeasts ,2.2 Factors relating to the physical environment ,2.1 Biological and endogenous factors ,Gene Regulatory Networks ,Aetiology ,intracellular bacteria ,Effector ,Foodborne Illness ,Translocon ,Cell biology ,Infectious Diseases ,Host-Pathogen Interactions ,Salmonella Infections ,Microorganisms, Genetically-Modified ,Infection ,Signal Transduction ,Histology ,Host–pathogen interaction ,Microorganisms ,Salmonella enterica serovar Typhimurium ,Biology ,retromer complex ,Article ,Genetically-Modified ,Pathology and Forensic Medicine ,Vaccine Related ,03 medical and health sciences ,Bacterial Proteins ,Biodefense ,Genetics ,Animals ,Humans ,Secretion ,E-MAP ,bacterial effector protein ,genetic interaction profile ,Prevention ,Intracellular parasite ,Cell Biology ,Salmonella typhi ,Bacterial effector protein ,High-Throughput Screening Assays ,Retromer complex ,Emerging Infectious Diseases ,030104 developmental biology ,Hela Cells ,Multiprotein Complexes ,Mutation ,Biochemistry and Cell Biology ,HeLa Cells - Abstract
Intracellular bacterial pathogens secrete a repertoire of effector proteins into host cells that are required to hijack cellular pathways and cause disease. Despite decades of research, however, the molecular functions of most bacterial effectors remain unclear. To address this gap, we generated quantitative genetic interaction profiles between 36 validated and putative effectors from three evolutionarily divergent human bacterial pathogens and 4,190 yeast deletion strains. Correlating effector-generated profiles with those of yeast mutants, we recapitulated known biology for several effectors with remarkable specificity and predicted previously unknown functions for others. Biochemical and functional validation in human cells revealed a role for an uncharacterized component of the Salmonella SPI-2 translocon, SseC, in regulating maintenance of the Salmonella vacuole through interactions with components of the host retromer complex. These results exhibit the power of genetic interaction profiling to discover and dissect complex biology at the host-pathogen interface.
- Published
- 2018
- Full Text
- View/download PDF
26. Polymeric particles in vaccine delivery
- Author
-
Allison C. Rice-Ficht, Thomas A. Ficht, Angela M. Arenas-Gamboa, and Melissa Kahl-McDonagh
- Subjects
Microbiology (medical) ,Vaccines ,Cellular immunity ,Polymers ,medicine.medical_treatment ,Vaccination ,Antigen presentation ,Inflammasome ,Biology ,Acquired immune system ,Microbiology ,Cell biology ,Infectious Diseases ,Adjuvants, Immunologic ,Antigen ,Immunity ,Delayed-Action Preparations ,Immunology ,medicine ,Humans ,Nanoparticles ,Antigen-presenting cell ,Adjuvant ,medicine.drug - Abstract
The tremendous power of the particulate vaccine delivery system has only recently been recognized and employed strategically in vaccine design. The entrapment of antigen in particles clearly alters its acquisition and processing by antigen presenting cells and ensuing adaptive immunity. The adjuvant activity of particles has recently been described at the molecular level as engaging the Nalp3 inflammasome and complementing the activity of toll-like receptor ligands. The inclusion of antigen within erodible particles and subsequent delivery to dendritic cells (DCs), enables antigen-specific cell-mediated immunity and extended antigen presentation with protective outcomes. Particles less than 1 microm in size with amphipathic coatings efficiently deliver antigen to and activate DCs with concomitant engagement of humoral and cellular immunity. The size and dissolution rates of particles as well as surface chemistry and charge appear to be central in tuning adaptive immunity.
- Published
- 2010
- Full Text
- View/download PDF
27. Brucellosis: The case for live, attenuated vaccines
- Author
-
Angela M. Arenas-Gamboa, Melissa Kahl-McDonagh, Allison C. Rice-Ficht, and Thomas A. Ficht
- Subjects
Cross Protection ,Brucella Vaccine ,Brucella abortus ,Brucella ,Biology ,Vaccines, Attenuated ,Article ,Brucellosis ,medicine ,Animals ,Humans ,Attenuated vaccine ,General Veterinary ,General Immunology and Microbiology ,Public Health, Environmental and Occupational Health ,medicine.disease ,biology.organism_classification ,Virology ,Vaccination ,Infectious Diseases ,Immunization ,Biological warfare ,Immunology ,Molecular Medicine ,Nasal administration ,Public Health - Abstract
The successful control of animal brucellosis and associated reduction in human exposure has limited the development of human brucellosis vaccines. However, the potential use of Brucella in bioterrorism or biowarfare suggests that direct intervention strategies are warranted. Although the dominant approach has explored the use of live attenuated vaccines, side-effects associated with their use has prevented widespread use in humans. Development of live, attenuated Brucella vaccines that are safe for use in humans has focused on the deletion of important genes required for survival. However, the enhanced safety of deletion mutants is most often associated with reduced efficacy. For this reason recent efforts have sought to combine the optimal features of a attenuated live vaccine that is safe, free of side effects and efficacious in humans with enhanced immune stimulation through microencapsulation. The competitive advantages and innovations of this approach are: (1) use of a highly attenuated, safe, gene knockout, live Brucella mutants; (2) manufacturing with unique disposable closed system technologies, and (3) oral/intranasal delivery in a novel microencapsulation-mediated controlled release formula to optimally provide the long term mucosal immunostimulation required for protective immunity. Based upon preliminary data, it is postulated that such vaccine delivery systems can be storage stable, administered orally or intranasally, and generally applicable to a number of agents.
- Published
- 2009
- Full Text
- View/download PDF
28. Analysis of Ten Brucella Genomes Reveals Evidence for Horizontal Gene Transfer Despite a Preferred Intracellular Lifestyle
- Author
-
Christine Munk, Joshua M. Shallom, J. Lu, Renée M. Tsolis, Ronald W. Kenyon, Allan W. Dickerman, Hyunseung Yoo, João C. Setubal, Stephen M. Boyle, Kelly P. Williams, David Bruce, Oswald Crasta, Nalvo F. Almeida, Alice R. Wattam, Eric E. Snyder, Thomas Brettin, Thomas A. Ficht, Maulik Shukla, Bruno W. S. Sobral, Roxanne Tapia, Cliff Han, and J. C. Detter
- Subjects
Genetics ,Brucella ovis ,Gene Transfer, Horizontal ,Models, Genetic ,biology ,Adipates ,Intracellular parasite ,Pseudogene ,Genetic transfer ,Computational Biology ,Brucella ,Chromosomes, Bacterial ,bacterial infections and mycoses ,biology.organism_classification ,Microbiology ,Phylogenetics ,Brucella ceti ,Horizontal gene transfer ,Molecular Biology ,Genome, Bacterial ,Phylogeny ,Pseudogenes ,Signal Transduction - Abstract
The facultative intracellular bacterial pathogen Brucella infects a wide range of warm-blooded land and marine vertebrates and causes brucellosis. Currently, there are nine recognized Brucella species based on host preferences and phenotypic differences. The availability of 10 different genomes consisting of two chromosomes and representing six of the species allowed for a detailed comparison among themselves and relatives in the order Rhizobiales . Phylogenomic analysis of ortholog families shows limited divergence but distinct radiations, producing four clades as follows: Brucella abortus-Brucella melitensis, Brucella suis-Brucella canis, Brucella ovis , and Brucella ceti . In addition, Brucella phylogeny does not appear to reflect the phylogeny of Brucella species' preferred hosts. About 4.6% of protein-coding genes seem to be pseudogenes, which is a relatively large fraction. Only B. suis 1330 appears to have an intact β-ketoadipate pathway, responsible for utilization of plant-derived compounds. In contrast, this pathway in the other species is highly pseudogenized and consistent with the “domino theory” of gene death. There are distinct shared anomalous regions (SARs) found in both chromosomes as the result of horizontal gene transfer unique to Brucella and not shared with its closest relative Ochrobactrum , a soil bacterium, suggesting their acquisition occurred in spite of a predominantly intracellular lifestyle. In particular, SAR 2-5 appears to have been acquired by Brucella after it became intracellular. The SARs contain many genes, including those involved in O-polysaccharide synthesis and type IV secretion, which if mutated or absent significantly affect the ability of Brucella to survive intracellularly in the infected host.
- Published
- 2009
- Full Text
- View/download PDF
29. ENHANCED IMMUNE RESPONSE OF RED DEER (CERVUS ELAPHUS) TO LIVE RB51 VACCINE STRAIN USING COMPOSITE MICROSPHERES
- Author
-
Allison C. Rice-Ficht, Donald S. Davis, Philip H. Elzer, Thomas A. Ficht, Alfredo Wong-Gonzalez, and Angela M. Arenas-Gamboa
- Subjects
Disease reservoir ,Injections, Subcutaneous ,Administration, Oral ,Brucella Vaccine ,Brucella abortus ,Animals, Wild ,Biology ,Vaccines, Attenuated ,Article ,Brucellosis ,Bison bison ,Random Allocation ,Risk Factors ,Zoonoses ,Prevalence ,medicine ,Animals ,symbols.heraldic_charge ,Ecology, Evolution, Behavior and Systematics ,Disease Reservoirs ,Immunity, Cellular ,Attenuated vaccine ,Cervus ,Ecology ,Deer ,Vaccination ,Abortion, Veterinary ,Vaccine efficacy ,biology.organism_classification ,medicine.disease ,Antibodies, Bacterial ,Virology ,Microspheres ,Treatment Outcome ,Antibody Formation ,symbols ,Female - Abstract
Brucellosis is an important zoonotic disease of nearly worldwide distribution. The occurrence of the infection in humans is largely dependent on the prevalence of brucellosis in animal reservoirs, including wildlife. The current vaccine used for cattle Brucella abortus strain RB51, has proven ineffective in protecting bison (Bison bison) and elk (Cervus nelsoni) from infection and abortion. To test possible improvements in vaccine efficacy, a novel approach of immunization was examined from April 2004 to November 2006 using alginate composite microspheres containing a nonimmunogenic, eggshell-precursor protein of the parasite Fasciola hepatica (Vitelline protein B, VpB) to deliver live vaccine strain RB51. Red deer (Cervus elaphus), used as a model for elk, were vaccinated orally (PO) or subcutaneously (SC) with 1.5x10(10) viable organisms per animal. Humoral responses postvaccination (immunoglobulin G [IgG] levels), assessed at different time points, indicated that capsules containing live RB51 elicited an anti-Brucella specific IgG response. Furthermore, the encapsulated vaccine elicited a cell-mediated response that the nonencapsulated vaccinates failed to produce. Finally, red deer were challenged with B. abortus strain 19 by conjunctival exposure. Only animals that received encapsulated RB51 vaccine by either route exhibited a significant reduction in bacterial counts in their spleens. These data suggest that alginate-VpB microspheres provide a method to enhance the RB51 vaccine performance in elk.
- Published
- 2009
- Full Text
- View/download PDF
30. Immunization with a Single Dose of a Microencapsulated Brucella melitensis Mutant Enhances Protection against Wild-Type Challenge
- Author
-
Allison C. Rice-Ficht, Angela M. Arenas-Gamboa, Thomas A. Ficht, and Melissa Kahl-McDonagh
- Subjects
Immunology ,Capsules ,Vaccines, Attenuated ,Microbiology ,Brucellosis ,Immunoglobulin G ,Mice ,Random Allocation ,Immunity ,Brucella melitensis ,Animals ,Mice, Inbred BALB C ,Virulence ,biology ,Macrophages ,Immunogenicity ,biology.organism_classification ,Vaccine efficacy ,Antibodies, Bacterial ,Virology ,Bacterial vaccine ,Infectious Diseases ,Immunization ,Delayed-Action Preparations ,Bacterial Vaccines ,Mutation ,Microbial Immunity and Vaccines ,biology.protein ,Cytokines ,Female ,Parasitology ,Cytokine secretion ,Injections, Intraperitoneal ,Spleen - Abstract
The development of safe and efficacious immunization systems to prevent brucellosis is needed to overcome the disadvantages of the currently licensed vaccine strains that restrict their use in humans. Alginate microspheres coated with a protein of the parasite Fasciola hepatica (vitelline protein B [VpB]) and containing live Brucella melitensis attenuated mutant vjbR ::Tn 5 (BMEII1116) were evaluated for vaccine efficacy and immunogenicity in mice. A single immunization dose in BALB/c mice with the encapsulated vjbR mutant improved protection against wild-type B. melitensis 16M challenge compared to the nonencapsulated vaccine strain ( P < 0.05). The encapsulated mutant was also shown to induce a sustained elevation of Immunoglobulin G levels. Cytokine secretion from spleen cells of mice vaccinated with the encapsulated vjbR ::Tn 5 revealed elevated secretion of gamma interferon and interleukin-12, but no interleukin-4, suggesting an induction of a T helper 1 response reflecting the enhanced immunity associated with microencapsulation. Together, these results suggest that microencapsulation of live attenuated organisms offers the ability to increase the efficacy of vaccine candidates.
- Published
- 2008
- Full Text
- View/download PDF
31. Evidence of Brucella abortus OPS dictating uptake and restricting NF-κB activation in murine macrophages
- Author
-
Jianwu Pei, Thomas A. Ficht, and Joshua E. Turse
- Subjects
medicine.medical_treatment ,Immunology ,Mutant ,Brucella abortus ,Receptors, Cell Surface ,Brucella ,Biology ,Microbiology ,Article ,Brucellosis ,Cell Line ,Mice ,medicine ,Animals ,Macrophage ,Receptor ,Lipid raft ,Macrophages ,NF-kappa B ,Macrophage Activation ,biology.organism_classification ,Cell biology ,Toll-Like Receptor 4 ,Infectious Diseases ,Cytokine ,TLR4 ,Intracellular - Abstract
Smooth Brucella abortus S2308 is virulent while rough derivatives are attenuated. Intracellular killing is often blamed for these differences. In the studies described, uptake kinetics and interaction of S2308 and S2308 manBA::Tn5 (CA180) rough mutants with macrophages were investigated. The results revealed that smooth B. abortus was rapidly internalized, achieving a maximum level in less than 5 minutes without additional uptake over the next 30 minutes. In contrast, continued uptake of the rough mutant was observed and only achieves a maximum level after 30 minutes. The results were confirmed by the differences in F-actin polymerization, lipid raft staining, early endosome colocalization and electron microscopic observations after smooth and rough Brucella infection. We also demonstrated for the first time that uptake of S2308, but not rough mutant CA180 was PI3-kinase and toll-like receptor 4 (TLR4) dependent. Differences in uptake were associated with differences in macrophage activation with regard to NF-κB translocation and cytokine production. These results provide evidence that the presence of B. abortus OPS dictates the interactions between Brucella and specific cell surface receptors minimizing macrophage activation and enhancing Brucella survival and/or persistence.
- Published
- 2008
- Full Text
- View/download PDF
32. Cytotoxicity in Macrophages Infected with Rough Brucella Mutants Is Type IV Secretion System Dependent
- Author
-
Melissa Kahl-McDonagh, Qingmin Wu, Jianwu Pei, and Thomas A. Ficht
- Subjects
Immunology ,Mutant ,Mutagenesis (molecular biology technique) ,Brucella ,medicine.disease_cause ,Microbiology ,Cell Line ,Mice ,Necrosis ,Bacterial Proteins ,Brucella melitensis ,medicine ,Animals ,Secretion ,Cytotoxicity ,Mutation ,biology ,Macrophages ,Gene Expression Regulation, Bacterial ,biology.organism_classification ,Molecular Pathogenesis ,Infectious Diseases ,Parasitology ,Transposon mutagenesis ,Apoproteins ,Gene Deletion - Abstract
Smooth Brucella spp. inhibit macrophage apoptosis, whereas rough Brucella mutants induce macrophage oncotic and necrotic cell death. However, the mechanisms and genes responsible for Brucella cytotoxicity have not been identified. In the current study, a random mutagenesis approach was used to create a mutant bank consisting of 11,354 mutants by mariner transposon mutagenesis using Brucella melitensis rough mutant 16MΔ manBA as the parental strain. Subsequent screening identified 56 mutants (0.49% of the mutant bank) that failed to cause macrophage cell death (release of 10% or less of the lactate dehydrogenase). The absence of cytotoxicity during infection with these mutants was independent of demonstrable defects in in vitro bacterial growth or uptake and survival in macrophages. Interrupted genes in 51 mutants were identified by DNA sequence analysis, and the mutations included interruptions in virB encoding the type IV secretion system (T4SS) ( n = 36) and in vjbR encoding a LuxR-like regulatory element previously shown to be required for virB expression ( n = 3), as well as additional mutations ( n = 12), one of which also has predicted roles in virB expression. These results suggest that the T4SS is associated with Brucella cytotoxicity in macrophages. To verify this, deletion mutants were constructed in B. melitensis 16M by removing genes encoding phosphomannomutase/phosphomannoisomerase (Δ manBA ) and the T4SS (Δ virB ). As predicted, deletion of virB from 16MΔ manBA and 16M resulted in a complete loss of cytotoxicity in rough strains, as well as the low level cytotoxicity observed with smooth strains at extreme multiplicities of infection (>1,000). Taken together, these results demonstrate that Brucella cytotoxicity in macrophages is T4SS dependent.
- Published
- 2008
- Full Text
- View/download PDF
33. Aerosol Infection of BALB/c Mice with Brucella melitensis and Brucella abortus and Protective Efficacy against Aerosol Challenge
- Author
-
Melissa Kahl-McDonagh, Angela M. Arenas-Gamboa, and Thomas A. Ficht
- Subjects
Ratón ,Immunology ,Colony Count, Microbial ,Brucella Vaccine ,Brucella abortus ,Heterologous ,Brucellaceae ,Vaccines, Attenuated ,Microbiology ,Brucellosis ,BALB/c ,Mice ,Brucella melitensis ,medicine ,Animals ,Lung ,Aerosols ,Mice, Inbred BALB C ,biology ,biology.organism_classification ,medicine.disease ,Vaccine efficacy ,Virology ,Chronic infection ,Infectious Diseases ,Liver ,Microbial Immunity and Vaccines ,Bacterial Vaccines ,Female ,Parasitology ,Gene Deletion ,Spleen - Abstract
Brucellosis is a zoonotic disease with a worldwide distribution that can be transmitted via intentional or accidental aerosol exposure. In order to engineer superior vaccine strains against Brucella species for use in animals as well as in humans, the possibility of challenge infection via aerosol needs to be considered to properly evaluate vaccine efficacy. In this study, we assessed the use of an aerosol chamber to infect deep lung tissue of mice to elicit systemic infections with either Brucella abortus or B. melitensis at various doses. The results reveal that B. abortus causes a chronic infection of lung tissue in BALB/c mice and peripheral organs at low doses. In contrast, B. melitensis infection diminishes more rapidly, and higher infectious doses are required to obtain infection rates in animals similar to those of B. abortus . Whether this difference translates to severity of human infection remains to be elucidated. Despite these differences, unmarked deletion mutants BAΔ asp 24 and BMΔ asp 24 consistently confer superior protection to mice against homologous and heterologous aerosol challenge infection and should be considered viable candidates as vaccine strains against brucellosis.
- Published
- 2007
- Full Text
- View/download PDF
34. Evaluation of novel Brucella melitensis unmarked deletion mutants for safety and efficacy in the goat model of brucellosis
- Author
-
Renée M. Tsolis, Christopher M. Seabury, Joel V. Walker, Melissa Kahl-McDonagh, Philip H. Elzer, Thomas A. Ficht, Andreas B. den Hartigh, Quinesha L. Perry, Donald S. Davis, Sue D. Hagius, and L. Garry Adams
- Subjects
Protective immunity ,Deletion mutant ,Fetal tissue ,Brucella Vaccine ,Brucellaceae ,Abortion ,Brucellosis ,Vaccine strain ,Pregnancy ,Brucella melitensis ,medicine ,Animals ,reproductive and urinary physiology ,Vaccines, Synthetic ,General Veterinary ,General Immunology and Microbiology ,biology ,Goats ,Vaccination ,Public Health, Environmental and Occupational Health ,biology.organism_classification ,medicine.disease ,Virology ,Disease Models, Animal ,Infectious Diseases ,Mutation ,Molecular Medicine ,Female - Abstract
Pregnant goats were employed to assess unmarked deletion mutant vaccine candidates BMDeltaasp24, BMDeltacydBA, and BMDeltavirB2, as the target host species naturally infected with Brucella melitensis. Goats were assessed for the degree of pathology associated with the vaccine strains as well as the protective immunity afforded by each strain against abortion and infection after challenge with wild-type Brucella melitensis 16M. Both BMDeltaasp24 and BMDeltavirB2 were considered safe vaccine candidates in the pregnant goat model because they did not cause abortion or colonize fetal tissues. BMDeltaasp24 was isolated from the maternal tissues only, indicating a slower rate of clearance of the vaccine strain than for BMDeltavirB2, which was not isolated from any maternal or fetal tissues. Both strains were protective against abortion and against infection in the majority of pregnant goats, although BMDeltaasp24 was more efficacious than BMDeltavirB2 against challenge infection.
- Published
- 2006
- Full Text
- View/download PDF
35. Brucella abortus Rough Mutants Induce Macrophage Oncosis That Requires Bacterial Protein Synthesis and Direct Interaction with the Macrophage
- Author
-
Joshua E. Turse, Qingmin Wu, Thomas A. Ficht, and Jianwu Pei
- Subjects
Programmed cell death ,Necrosis ,media_common.quotation_subject ,Immunology ,Brucella abortus ,Apoptosis ,Brucella ,Microbiology ,Bacterial cell structure ,Cell Line ,Mice ,Bacterial Proteins ,medicine ,Animals ,Macrophage ,Internalization ,media_common ,Cellular Microbiology: Pathogen-Host Cell Molecular Interactions ,L-Lactate Dehydrogenase ,biology ,Macrophages ,Macrophage Activation ,biology.organism_classification ,Infectious Diseases ,Phosphotransferases (Phosphomutases) ,Protein Biosynthesis ,Parasitology ,Tumor necrosis factor alpha ,medicine.symptom ,Brucella melitensis - Abstract
Previous studies suggest that smooth Brucella organisms inhibit macrophage apoptosis. In contrast, necrotic cell death of macrophages infected with rough Brucella organisms in vitro has been reported, which may in part explain the failure of some rough organisms to thrive. To characterize these potential macrophage killing mechanisms, J774.A1 murine macrophages were infected with Brucella abortus S2308-derived rough mutant CA180. Electron microscopic analysis and polyethylene glycol protection assays revealed that the cells were killed as a result of necrosis and oncosis. This killing was shown to be unaffected by treatment with carbenicillin, an inhibitor of bacterial cell wall biosynthesis and, indirectly, replication. In contrast, chloramphenicol treatment of macrophages infected at multiplicities of infection exceeding 10,000 prevented cell death, despite internalization of large numbers of bacteria. Similarly, heat-killed and gentamicin-killed CA180 did not induce cytopathic effects in the macrophage. These results suggested that killing of infected host cells requires active bacterial protein synthesis. Cytochalasin D treatment revealed that internalization of the bacteria was necessary to initiate killing. Transwell experiments demonstrated that cell death is not mediated by a diffusible product, including tumor necrosis factor alpha and nitric oxide, but does require direct contact between host and pathogen. Furthermore, macrophages preinfected with B. abortus S2308 or pretreated with B. abortus O polysaccharide did not prevent rough CA180-induced cell death. In conclusion, Brucella rough mutant infection induces necrotic and oncotic macrophage cell death that requires bacterial protein synthesis and direct interaction of bacteria with the target cells.
- Published
- 2006
- Full Text
- View/download PDF
36. Amplified Fragment Length Polymorphism Reveals Genomic Variability among Mycobacterium avium subsp. paratuberculosis Isolates
- Author
-
B. O'Shea, Allison C. Rice-Ficht, Thomas A. Ficht, Patricia E. Klein, L. G. Adams, K. Bliss, and Sangeeta Khare
- Subjects
DNA, Bacterial ,Microbiology (medical) ,Restriction Mapping ,Paratuberculosis ,Biology ,Genome ,Microbiology ,law.invention ,Restriction map ,law ,Genetic variation ,medicine ,Humans ,Polymerase chain reaction ,DNA Primers ,Genetics ,Polymorphism, Genetic ,Base Sequence ,Gene Amplification ,Genetic Variation ,Mycobacteriology and Aerobic Actinomycetes ,bacterial infections and mycoses ,medicine.disease ,biology.organism_classification ,Housekeeping gene ,Mycobacterium avium subsp. paratuberculosis ,bacteria ,Amplified fragment length polymorphism ,Mycobacterium avium ,Mycobacterium - Abstract
Ninety-six primer sets were used for amplified fragment length polymorphism (AFLP) to characterize the genomes of 20 Mycobacterium avium subsp. paratuberculosis field isolates, 1 American Type Culture Collection (ATCC) M. avium subsp. paratuberculosis isolate (ATCC 19698), and 2 M. avium subsp. avium isolates (ATCC 35716 and Mac 104). AFLP analysis revealed a high degree of genomic polymorphism among M. avium subsp. paratuberculosis isolates that may be used to establish diagnostic patterns useful for the epidemiological tracking of M. avium subsp. paratuberculosis isolates. Four M. avium subsp. paratuberculosis -polymorphic regions revealed by AFLP were cloned and sequenced. Primers were generated internal to these regions for use in PCR analysis and applied to the M. avium subsp. paratuberculosis field isolates. An appropriate PCR product was obtained in 79 of 80 reactions, while the M. avium subsp. avium isolates failed to act as templates for PCR amplification in seven of eight reactions. This work revealed the presence of extensive polymorphisms in the genomes of M. avium subsp. paratuberculosis and M. avium subsp. avium , many of which are based on deletions. Of the M. avium subsp. paratuberculosis -specific sequences studied, one revealed a 5,145-bp region with no homologue in the M. avium subsp. avium genome. Within this region are genes responsible for integrase-recombinase function. Three additional M. avium subsp. paratuberculosis -polymorphic regions were cloned, revealing a number of housekeeping genes; all were evaluated for their diagnostic and epidemiological value.
- Published
- 2004
- Full Text
- View/download PDF
37. Intracellular survival of Brucella: defining the link with persistence
- Author
-
Thomas A. Ficht
- Subjects
Phagocyte ,Cattle Diseases ,Brucellaceae ,Brucella ,Microbiology ,Brucellosis ,Mice ,Pregnancy ,medicine ,Animals ,Humans ,Pathogen ,Virulence ,General Veterinary ,biology ,Intracellular parasite ,General Medicine ,Mononuclear phagocyte system ,Abortion, Veterinary ,biology.organism_classification ,Disease Models, Animal ,Chronic infection ,medicine.anatomical_structure ,Carrier State ,Immunology ,Cattle ,Female ,Intracellular - Abstract
Brucellosis is caused by a facultative intracellular pathogen that invades both professional and non-professional phagocytic cells. Resistance to killing in professional phagocytic cells controls survival and chronic infection. Resistance of the organism to killing appears to derive from altered intracellular trafficking of Brucella containing vacuoles to the endoplasmic reticulum via the autophagic pathway. Acute infection is observed in pregnant ruminants in which invasion of the chorionic trophoblasts results in abortion. Following abortion persistence of the organism is observed in the mammary gland and lymph nodes of ruminants. The risk of multiple abortions and subsequent shedding of the organism in the milk has resulted in the culling of infected animals. Persistence of the organism in the reticuloendothelial system is a primary symptom in human infection and may persist over several decades. We have employed the mouse model of brucellosis to characterize genes responsible for persistent infection in an effort to identify potential drug targets for elimination of infection or to attenuate potential vaccine candidates. The results suggest that Brucella utilizes a battery of metabolic functions to sustain itself in intracellular environments in conjunction with altering the intracellular course of infection.
- Published
- 2003
- Full Text
- View/download PDF
38. The myth of Brucella L-forms and possible involvement of Brucella penicillin binding proteins (PBPs) in pathogenicity
- Author
-
Thomas A. Ficht, L. G. Adams, Menachem Banai, M. Frey, and Roberta Pugh
- Subjects
Penicillin binding proteins ,Meticillin ,medicine.drug_class ,Antibiotics ,Brucellaceae ,Brucella ,Muramoylpentapeptide Carboxypeptidase ,Microbiology ,Methicillin ,Bacterial Proteins ,Brucella melitensis ,polycyclic compounds ,medicine ,Animals ,Penicillin-Binding Proteins ,Sheep ,General Veterinary ,biology ,Macrophages ,Genetic Variation ,General Medicine ,biology.organism_classification ,Virology ,Penicillin ,Hexosyltransferases ,Peptidyl Transferases ,Carrier Proteins ,Bacteria ,medicine.drug - Abstract
Brucella spp. L-forms have been proposed to be stationary phase organisms in the evolution of new variants and enduring entities in the host in complicated cases of brucellosis and during latent brucellosis. In vitro formation of Brucella L-forms has been achieved by treating the cells with sub-lethal doses of penicillin. Interestingly, Brucella spp. have classified during the evolution into two groups, penicillin susceptible or penicillin resistant, yet both types grow on 20 microg/ml of methicillin. Strains proven susceptible to penicillin grew in the presence of methicillin as L-forms as demonstrated by light and electron microscopy. In addition, the B. melitensis vaccine strain Rev.1, a penicillin susceptible organism, responded to sheep serum by development of L-form-like structures unlike wild type, strain 16M. The two strains grew normally in sheep macrophages. We propose, for the first time, a model that associates Brucella pathogenicity with the structure and activity of two of their penicillin binding proteins (PBPs). According to the model, PBP1 has evolved as the major cell wall synthesizing enzyme of the genus, capable of responding to host serum growth factor(s) necessary for Brucella survival in the host. This property is associated with high avidity to beta-lactam antibiotics. PBP2 complements the activity of PBP1. New beta-lactam antibiotics and improved vaccines might be developed based on this property.
- Published
- 2002
- Full Text
- View/download PDF
39. Brucella attenuation and relevance to vaccine properties
- Author
-
M Banai, L. G. Adams, L.J Dangott, M. Frey, and Thomas A. Ficht
- Subjects
biology ,Strain (chemistry) ,Intracellular parasite ,Wild type ,Virulence ,Brucella ,biology.organism_classification ,Virology ,Microbiology ,Food Animals ,Animal Science and Zoology ,Gene ,Bacteria ,Brucella melitensis - Abstract
Brucellosis is a zoonotic disease caused by Gram negative, facultative intracellular bacteria. The genus includes six species, the major three being Brucella melitensis , B. abortus and B. suis. These three species cause abortions in their primary livestock hosts resulting in major economic losses worldwide. Although attenuated live vaccine strains have been derived from the three important species and used extensively, there have been adverse side-effects. To study the virulence properties of Brucella , we targeted intracellular survival genes using macrophage survival and mouse virulence. We first evaluated the method using B. abortus 2308 wild type strain. Brucella genes were randomly inactivated following electroporation with a DNA construct that included a transposon element associated with antibiotic resistance marker. Transformed clones were then screened for survival in mice or in monocyte-derived macrophages, and were compared with the parental wild type strain. Clones showing reduced survival in the mouse or the macrophage model, respectively, were considered deleted in an essential virulence gene. In an alternative approach the B. melitensis vaccine strain Rev.1 has been compared to its wild type prototype strain, B. melitensis 16M. The study included 2-D gel analysis of the protein profiles of these strains reveal similar patterns when grown under normal growth conditions. Electron microscopic comparison has shown, however, that strain Rev.1 changes its ultrastructural morphology according to the surrounding environmental conditions.
- Published
- 2002
- Full Text
- View/download PDF
40. The Brucella abortus Lon functions as a generalized stress response protease and is required for wild-type virulence in BALB/c mice
- Author
-
Michael E. Kovach, Gregory T. Robertson, R. Martin Roop, Chris A. Allen, and Thomas A. Ficht
- Subjects
Protease ,medicine.medical_treatment ,Mutant ,Wild type ,lac operon ,Virulence ,Biology ,bacterial infections and mycoses ,biology.organism_classification ,medicine.disease_cause ,Microbiology ,BALB/c ,law.invention ,law ,medicine ,Recombinant DNA ,bacteria ,Molecular Biology ,Escherichia coli - Abstract
The gene encoding a Lon protease homologue has been cloned from Brucella abortus. The putative Brucella abortus Lon shares > 60% amino acid identity with its Escherichia coli counterpart and the recombinant form of this protein restores the capacity of an Escherichia coli lon mutant to resist killing by ultraviolet irradiation and regulate the expression of a cpsB::lacZ fusion to wild-type levels. A σ32 type promoter was identified upstream of the predicted lon coding region and Northern analysis revealed that transcription of the native Brucella abortus lon increases in response to heat shock and other environmental stresses. ATP-dependent proteolytic activity was also demonstrated for purified recombinant Lon. To evaluate the capacity of the Brucella abortus Lon homologue to function as a stress response protease, the majority of the lon coding region was removed from virulent strain Brucella abortus 2308 via allelic exchange. In contrast to the parent strain, the Brucella abortus lon mutant, designated GR106, was impaired in its capacity to form isolated colonies on solid medium at 41°C and displayed an increased sensitivity to killing by puromycin and H2O2. GR106 also displayed reduced survival in cultured murine macrophages and significant attenuation in BALB/c mice at 1 week post infection compared with the virulent parental strain. Beginning at 2 weeks and continuing for 6 weeks post infection, however, GR106 and 2308 displayed equivalent spleen and liver colonization levels in mice. These findings suggest that the Brucella abortus Lon homologue functions as a stress response protease that is required for wild-type virulence during the initial stages of infection in the mouse model, but is not essential for the establishment and maintenance of chronic infection in this host.
- Published
- 2002
- Full Text
- View/download PDF
41. SiRNA screens using Drosophila cells to identify host factors required for infection
- Author
-
Aseem, Pandey, Sheng Li, Ding, Thomas A, Ficht, and Paul, de Figueiredo
- Subjects
Host-Pathogen Interactions ,Animals ,Drosophila ,Brucella ,Cell Line - Abstract
Drosophila melanogaster offers a powerful model system for interrogating interactions between host cells and human bacterial pathogens. Brucella, a gram-negative, facultative intracellular bacterium is the causative agent of brucellosis, a zoonotic disease of global consequence. Over the past several decades, pathogen factors that mediate Brucella infection have been identified. However, host factors that mediate infection have remained obscure. We have used the power of the Drosophila S2 cell system to identify and characterize host factors that support infection by Brucella melitensis. Host protein inositol-requiring enzyme 1 (IRE1α), a transmembrane kinase and master regulator of the eukaryotic unfolded protein response, was shown to play an important role in regulating Brucella infection, thereby providing the first glimpse of host mechanisms that are subverted by the pathogen to support its intracellular lifestyle. Furthermore, our study also established the Drosophila S2 cell as a powerful system for elucidating Brucella host factors. Here, we describe a protocol for using the Drosophila S2 cell system for studying the Brucella-host interaction.
- Published
- 2014
42. Pathogenesis and immunobiology of brucellosis: review of Brucella-host interactions
- Author
-
Paul, de Figueiredo, Thomas A, Ficht, Allison, Rice-Ficht, Carlos A, Rossetti, and L Garry, Adams
- Subjects
Host-Pathogen Interactions ,Animals ,Humans ,Review ,Brucella ,Brucellosis - Abstract
This review of Brucella–host interactions and immunobiology discusses recent discoveries as the basis for pathogenesis-informed rationales to prevent or treat brucellosis. Brucella spp., as animal pathogens, cause human brucellosis, a zoonosis that results in worldwide economic losses, human morbidity, and poverty. Although Brucella spp. infect humans as an incidental host, 500,000 new human infections occur annually, and no patient-friendly treatments or approved human vaccines are reported. Brucellae display strong tissue tropism for lymphoreticular and reproductive systems with an intracellular lifestyle that limits exposure to innate and adaptive immune responses, sequesters the organism from the effects of antibiotics, and drives clinical disease manifestations and pathology. Stealthy brucellae exploit strategies to establish infection, including i) evasion of intracellular destruction by restricting fusion of type IV secretion system-dependent Brucella-containing vacuoles with lysosomal compartments, ii) inhibition of apoptosis of infected mononuclear cells, and iii) prevention of dendritic cell maturation, antigen presentation, and activation of naive T cells, pathogenesis lessons that may be informative for other intracellular pathogens. Data sets of next-generation sequences of Brucella and host time-series global expression fused with proteomics and metabolomics data from in vitro and in vivo experiments now inform interactive cellular pathways and gene regulatory networks enabling full-scale systems biology analysis. The newly identified effector proteins of Brucella may represent targets for improved, safer brucellosis vaccines and therapeutics.
- Published
- 2014
43. Salmonella-Induced Cell Death Is Not Required for Enteritis in Calves
- Author
-
Andreas J. Bäumler, Shuping Zhang, Renée M. Tsolis, Renato L. Santos, L. G. Adams, and Thomas A. Ficht
- Subjects
Diarrhea ,Male ,Salmonella typhimurium ,medicine.medical_treatment ,Immunology ,Apoptosis ,Inflammation ,Biology ,Microbiology ,Proinflammatory cytokine ,Enteritis ,Bacterial Proteins ,medicine ,Animals ,Cellular Microbiology: Pathogen-Host Cell Molecular Interactions ,TUNEL assay ,Monocyte ,medicine.disease ,Disease Models, Animal ,B vitamins ,Infectious Diseases ,medicine.anatomical_structure ,Cytokine ,Mutagenesis ,Salmonella Infections ,Cattle ,Parasitology ,medicine.symptom - Abstract
Salmonellaenterica serovar Typhimurium causes cell death in bovine monocyte-derived and murine macrophages in vitro by asipB-dependent mechanism. During this process, SipB binds and activates caspase-1, which in turn activates the proinflammatory cytokine interleukin-1β through cleavage. We used bovine ileal ligated loops to address the role of serovar Typhimurium-induced cell death in induction of fluid accumulation and inflammation in this diarrhea model. Twelve perinatal calves had 6- to 9-cm loops prepared in the terminal ileum. They were divided into three groups: one group received an intralumen injection of Luria-Bertani broth as a control in 12 loops. The other two groups (four calves each) were inoculated with 0.75 × 109CFU of either wild-type serovar Typhimurium (strain IR715) or asopBmutant per loop in 12 loops. Hematoxylin and eosin-stained sections were scored for inflammation, and terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end labeling (TUNEL)-positive cells were detected in situ. Fluid accumulation began at 3 h postinfection (PI). Inflammation was detected in all infected loops at 1 h PI. The area of TUNEL-labeled cells in the wild-type infected loops was significantly higher than that of the controls at 12 h PI, when a severe inflammatory response and tissue damage had already developed. ThesopBmutant induced the same amount of TUNEL-positive cells as the wild type, but it was attenuated for induction of fluid secretion and inflammation. Our results indicate that serovar Typhimurium-induced cell death is not required to trigger an early inflammatory response and fluid accumulation in the ileum.
- Published
- 2001
- Full Text
- View/download PDF
44. A Multiplex Approach to Molecular Detection of Brucella abortus and/or Mycobacterium bovis Infection in Cattle
- Author
-
Srikrishna Isloor, Raj R. Barathur, Thomas A. Ficht, Jack Bain Bookout, Srinand Sreevatsan, V. S. Perumaalla, M. Rajasekhar, Betsy J. Bricker, Philip H. Elzer, L. Garry Adams, Fidel M. Ringpis, Girish K. Kumar, and Sue D. Hagius
- Subjects
DNA, Bacterial ,Microbiology (medical) ,Chaperonins ,Brucella abortus ,Tuberculin ,Brucellaceae ,Polymerase Chain Reaction ,Sensitivity and Specificity ,law.invention ,Microbiology ,Serology ,Brucellosis, Bovine ,Bacterial Proteins ,law ,Multiplex polymerase chain reaction ,medicine ,Animals ,Multiplex ,Polymerase chain reaction ,Mycobacterium bovis ,biology ,Mycobacteriology and Aerobic Actinomycetes ,Brucellosis ,Chaperonin 60 ,biology.organism_classification ,medicine.disease ,Milk ,Cattle ,Nasal Cavity ,Tuberculosis, Bovine - Abstract
A multiplex amplification and detection platform for the diagnosis of Mycobacterium bovis and Brucella abortus infection simultaneously in bovine milk and nasal secretions was developed. This system (designated the bovine pathogen detection assay [BPDA]-PCR) consists of duplex amplification of species-specific targets (a region of the BCSP31K gene of B. abortus and a repeat-sequence region in the hsp65 gene of M. bovis , respectively). This is followed by a solid-phase probe capture hybridization of amplicons for detection. On the basis of spiking experiments with normal milk, the analytical sensitivity of the assay was 800 CFU equivalents/ml of milk for B. abortus and as low as 4 CFU equivalents per ml of milk for M. bovis . BPDA-PCR was validated with 45 liver samples from lemmings experimentally infected with B. abortus . The assay sensitivity, based on culture status as a “gold standard,” was 93.9%. In this experiment, BPDA-PCR also identified five culture-negative liver samples as positive (41.7%). Field studies for the evaluation of BPDA-PCR were performed with samples from dairy animals from geographically distinct regions (India, Mexico, and Argentina). A high prevalence of shedding of B. abortus (samples from India) and M. bovis (samples from Mexico) was identified by BPDA-PCR. In samples from India, B. abortus shedding was identified in 86% of milk ring test-positive animals ( n = 15) and 80% of milk ring test-negative cows ( n = 5). In samples from Mexico, M. bovis was identified by PCR in 32.6% of pools ( n = 46) of milk that each contained milk from 10 animals and in 56.2% of nasal swabs ( n = 121) from cattle from tuberculin test-positive herds. In contrast, the Argentine cattle ( n = 70) had a modest prevalence of M. bovis shedding in nasal swabs (2.9%) and milk (1.4%) and of B. abortus in milk (11.4%). On the basis of these analyses, we identify BPDA-PCR as an optimal tool for both screening of herds and testing of individual animals in a disease eradication program. A combination of the duplex assay, screening of milk samples in pools, and the proposed algorithm provides a highly sensitive, cost-effective, and economically viable alternative to serological testing.
- Published
- 2000
- Full Text
- View/download PDF
45. SspA Is Required for Lethal Salmonella enterica Serovar Typhimurium Infections in Calves but Is Not Essential for Diarrhea
- Author
-
Michael J. Hantman, Christina A. Scherer, Thomas A. Ficht, L. G. Adams, Andreas J. Bäumler, Renée M. Tsolis, Samuel I. Miller, T. Kimbrough, and Robert A. Kingsley
- Subjects
Diarrhea ,Salmonella typhimurium ,Serotype ,Salmonella ,Immunology ,Cattle Diseases ,Virulence ,medicine.disease_cause ,Microbiology ,medicine ,Animals ,Adhesins, Bacterial ,Salmonella Infections, Animal ,biology ,Bacterial Infections ,biology.organism_classification ,Virology ,Enterobacteriaceae ,Pathogenicity island ,Intestines ,Infectious Diseases ,Genes, Bacterial ,Salmonella enterica ,Mutation ,Fluid Therapy ,Cattle ,Parasitology ,medicine.symptom ,Bacteria - Abstract
Salmonella pathogenicity island 1 (SPI-1) encodes virulence determinants, which are important for enteropathogenicity in calves. To determine whether the Salmonella enterica serovar Typhimurium SPI-1 effector proteins SspA and SptP are important for enteropathogenicity, strains lacking these proteins were tested during oral infection of calves. Calves infected with a sptP mutant or its isogenic parent developed diarrhea and lethal morbidity. In contrast, calves infected with an sspA mutant developed diarrhea, which resolved within 10 days but did not result in mortality. The sspA mutant was recovered from bovine intestinal tissues at numbers similar to those obtained for its isogenic parent and caused marked intestinal lesions. Thus, the severity of pathological changes caused by serovar Typhimurium strains or their ability to cause diarrhea were not predictive of their ability to cause lethal morbidity in calves. We conclude that factors other than or in addition to bacterial colonization, intestinal lesions, or electrolyte loss contribute to lethal morbidity in calves infected with serovar Typhimurium.
- Published
- 2000
- Full Text
- View/download PDF
46. Impact of Horizontal Gene Transfer on the Evolution of Salmonella Pathogenesis
- Author
-
Renée M. Tsolis, Stacy M. Townsend, Robert A. Kingsley, Tracy L. Norris, Thomas A. Ficht, Andreas J. Bäumler, and L. Garry Adams
- Subjects
Salmonella bongori ,Serotype ,Salmonella ,biology ,Aroa ,Salmonella enterica ,Horizontal gene transfer ,medicine ,Virulence ,biology.organism_classification ,medicine.disease_cause ,Pathogen ,Microbiology - Abstract
The establishment of a phylogenetic tree is a prerequisite for studying the evolution of virulence. The current nomenclature of the genus Salmonella is based on this phylogenetic tree and distinguishes only two species: Salmonella enterica and Salmonella bongori. If acquisition of SPI-1 introduced a virulence factor required for the pathogenesis of diarrheal disease, then mutational inactivation of this determinant should attenuate Salmonella serotypes in animal models of gastroenteritis. The contribution of the invasion-associated type III secretion system to serotype Typhimurium pathogenesis in this animal model has recently been investigated using strains carrying mutations in hilA and prgH. The majority of antibodies elicited by immunization with heat-killed serotype Typhimurium or with a live-attenuated serotype Typhimurium aroA vaccine is directed against the immunodominant O-antigen. Mathematical models predict that in this between-serotype competition, the serotype with higher transmissibility will dominate and eventually eliminate its competitor. The generation of O-antigen polymorphism through horizontal gene transfer was therefore a likely mechanism that allowed Salmonella serotypes to adapt to the enhanced immune memory encountered in warm-blooded hosts. The fljB gene is present in biphasic S. enterica subspecies but absent from monophasic S. enterica subspecies and Escherichia coli, suggesting its acquisition by horizontal gene transfer. A primary pathogen can be defined as an organism capable of entering a host and finding a unique niche to multiply and avoid or subvert the host defenses, the outcome of which may be clinical disease manifestations.
- Published
- 2000
- Full Text
- View/download PDF
47. Identification of a Putative Salmonella enterica Serotype Typhimurium Host Range Factor with Homology to IpaH and YopM by Signature-Tagged Mutagenesis
- Author
-
Renée M. Tsolis, Thomas A. Ficht, Stacy M. Townsend, Andreas J. Bäumler, Edward A. Miao, L. Garry Adams, and Samuel I. Miller
- Subjects
DNA, Bacterial ,Salmonella typhimurium ,Molecular Sequence Data ,Immunology ,Mutant ,Cattle Diseases ,Virulence ,Microbiology ,Mice ,Shigella flexneri ,Bacterial Proteins ,Animals ,Amino Acid Sequence ,Antigens, Bacterial ,Salmonella Infections, Animal ,Signature-tagged mutagenesis ,Base Sequence ,Sequence Homology, Amino Acid ,biology ,Wild type ,Sequence Analysis, DNA ,biology.organism_classification ,Pathogenicity island ,Virology ,Mutagenesis, Insertional ,Infectious Diseases ,Salmonella enterica ,DNA Transposable Elements ,Molecular and Cellular Pathogenesis ,Cattle ,Parasitology ,Host adaptation ,Bacterial Outer Membrane Proteins - Abstract
The genetic basis for the host adaptation of Salmonella serotypes is currently unknown. We have explored a new strategy to identify Salmonella enterica serotype Typhimurium ( S. typhimurium ) genes involved in host adaptation, by comparing the virulence of 260 randomly generated signature-tagged mutants during the oral infection of mice and calves. This screen identified four mutants, which were defective for colonization of only one of the two host species tested. One mutant, which only displayed a colonization defect during the infection of mice, was further characterized. During competitive infection experiments performed with the S. typhimurium wild type, the mutant was defective for colonization of murine Peyer's patches but colonized bovine Peyer's patches at the wild-type level. No difference in virulence between wild type and mutant was observed when calves were infected orally with 10 10 CFU/animal. In contrast, the mutant possessed a sixfold increase in 50% lethal morbidity dose when mice were infected orally. The transposon in this mutant was inserted in a 2.9-kb pathogenicity islet, which is located between uvrB and yphK on the S. typhimurium chromosome. This pathogenicity islet contained a single gene, termed slrP , with homology to ipaH of Shigella flexneri and yopM of Yersinia pestis . These data show that comparative screening of signature-tagged mutants in two animal species can be used for scanning the S. typhimurium genome for genes involved in host adaptation.
- Published
- 1999
- Full Text
- View/download PDF
48. siRNA Screens Using Drosophila Cells to Identify Host Factors Required for Infection
- Author
-
Aseem Pandey, Thomas A. Ficht, Shengli Ding, and Paul de Figueiredo
- Subjects
biology ,Host (biology) ,Intracellular parasite ,Unfolded protein response ,Brucella ,Drosophila melanogaster ,biology.organism_classification ,Pathogen ,Transmembrane protein ,Brucella melitensis ,Cell biology - Abstract
Drosophila melanogaster offers a powerful model system for interrogating interactions between host cells and human bacterial pathogens. Brucella, a gram-negative, facultative intracellular bacterium is the causative agent of brucellosis, a zoonotic disease of global consequence. Over the past several decades, pathogen factors that mediate Brucella infection have been identified. However, host factors that mediate infection have remained obscure. We have used the power of the Drosophila S2 cell system to identify and characterize host factors that support infection by Brucella melitensis. Host protein inositol-requiring enzyme 1 (IRE1α), a transmembrane kinase and master regulator of the eukaryotic unfolded protein response, was shown to play an important role in regulating Brucella infection, thereby providing the first glimpse of host mechanisms that are subverted by the pathogen to support its intracellular lifestyle. Furthermore, our study also established the Drosophila S2 cell as a powerful system for elucidating Brucella host factors. Here, we describe a protocol for using the Drosophila S2 cell system for studying the Brucella-host interaction.
- Published
- 2014
- Full Text
- View/download PDF
49. Synergistic effect of mutations in invA and lpfC on the ability of Salmonella typhimurium to cause murine typhoid
- Author
-
Renée M. Tsolis, Thomas A. Ficht, Andreas J. Bäumler, Fred Heffron, and Peter J. Valentine
- Subjects
Salmonella typhimurium ,Salmonella ,Operon ,Immunology ,Mutant ,Virulence ,Biology ,medicine.disease_cause ,Microbiology ,Mice ,Bacterial Proteins ,Intestinal mucosa ,medicine ,Animals ,Mesenteric lymph nodes ,Typhoid Fever ,Mice, Inbred BALB C ,biology.organism_classification ,Enterobacteriaceae ,Peyer Patch ,Infectious Diseases ,medicine.anatomical_structure ,Genes, Bacterial ,Mutation ,Female ,Parasitology ,Research Article - Abstract
Penetration of the intestinal mucosa at areas of Peyer's patches is an important first step for Salmonella typhimurium to produce lethal systemic disease in mice. However, mutations in genes that are important for intestinal invasion result in only moderately decreased virulence of S. typhimurium for mice. Here we report that combining mutations in invA and lpfC, two genes necessary for entry into Peyer's patches, results in a much stronger attenuation of S. typhimurium than inactivation of either of these genes alone. An S. typhimurium invA lpfC mutant was 150-fold attenuated by the oral route of infection but was fully virulent when the intestine was bypassed by intraperitoneal challenge of mice. During mixed-infection experiments, the S. typhimurium invA lpfC mutant showed a strong defect in colonizing Peyer's patches and mesenteric lymph nodes. These data suggest that mutations in invA and lpfC deactivate distinct pathways for intestinal penetration and colonization of Peyer's patches. While the inv-mediated pathway is widely distributed, the lpf operon is absent from many phylogenetic groups within the genus Salmonella. To investigate how acquisition of the lpf-mediated pathway for mucosal penetration contributed to evolution of virulence, we studied the relationship between the presence of the lpf operon and the pathogenicity for mice of 18 isolates representing 14 Salmonella serotypes. Only strains possessing the lpf operon were able to cause lethal infection in mice. These data show that both the invA- and lpfC-mediated pathways of intestinal perforation are conserved in mouse virulent Salmonella serotypes.
- Published
- 1997
- Full Text
- View/download PDF
50. Immunological response to the Brucella abortus GroEL homolog
- Author
-
Thomas A. Ficht, L. G. Adams, and J Lin
- Subjects
animal diseases ,Blotting, Western ,Immunology ,Brucella abortus ,Virulence ,Brucellaceae ,Biology ,complex mixtures ,Microbiology ,Serology ,Mice ,Immune system ,Western blot ,Immunity ,medicine ,Animals ,medicine.diagnostic_test ,Vaccination ,Chaperonin 60 ,bacterial infections and mycoses ,biology.organism_classification ,Antibodies, Bacterial ,GroEL ,Virology ,Infectious Diseases ,Humoral immunity ,Cattle ,Female ,Parasitology ,Rabbits ,Research Article - Abstract
Western blot (immunoblot) analysis of sera from cattle vaccinated with Brucella abortus S19 exhibit an elevated serologic response to Hsp62, the GroEL homolog (BaGroEL). Serologic screening of individual cows vaccinated with B. abortus S19 revealed no correlation between the immune response to BaGroEL and protection against a challenge with virulent organisms. The humoral immune response to BaGroEL was restricted to a region of the mature protein which mapped to amino acids 317 to 355 and may represent a useful diagnostic tool for monitoring exposure to B. abortus. Immunity to a challenge with virulent B. abortus S2308 was not observed in the BaGroEL vaccinated mouse model.
- Published
- 1996
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.