1
|
Ahmed A, Shimizu T, Shimoda H, Hosoi E, Uda A, Hotta A, Watarai M, Maeda K, Takano A. Molecular and serological investigation of Francisella tularensis among wild animals in Yamaguchi prefecture. Vet Res Commun 2024; 48:3397-3402. [PMID: 39066824 DOI: 10.1007/s11259-024-10474-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 07/20/2024] [Indexed: 07/30/2024]
Abstract
Francisella tularensis is an intracellular gram-negative bacterium known as the causative agent of tularemia, which can be transmitted to humans by direct contact with wild animals or by tick bites. Although F. tularensis is highly pathogenic, its recent prevalence in Japan is underreported due to the small number of reported cases. To clarify the current situation of F. tularensis in wild animals, we conducted surveillance on various species of wild animals in Yamaguchi prefecture. In this study, we screened 809 samples collected from 90 Japanese black bears, 105 Japanese monkeys, 168 sika deer, 205 wild boars, and 84 bats. For seroprevalence analysis, we tested 177 serum samples from 75 black bears and 102 monkeys using the microagglutination test. The results showed that serums from five black bears exhibited slight agglutination. Western blot was performed as a confirmatory test on these five samples, but no positive signals were detected. Additionally, molecular surveillance was conducted using DNA extracted from 464 whole blood and 168 tissues, targeting the gene encoding 23 KDa hypothetical protein by real-time PCR and outer membrane protein A gene by conventional PCR. No positive samples of F. tularensis were detected by either real-time or conventional PCR. Although we did not detect any F. tularensis-positive samples through serological and molecular analyses, continuous surveillance studies are necessary since sporadic human cases have been reported in Japan.
Collapse
Affiliation(s)
- Abdelrahman Ahmed
- Joint Graduate School of Veterinary Medicine, Yamaguchi University, Yamaguchi, 753-8515, Japan
| | - Takashi Shimizu
- Laboratory of Veterinary Public Health, Department of Veterinary Medicine, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, 753-8515, Japan
| | - Hiroshi Shimoda
- Laboratory of Veterinary Microbiology, Department of Veterinary Medicine, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, 753-8515, Japan
| | - Eiji Hosoi
- Graduate School of Sciences and Technology for Innovation, Yamaguchi University, Yamaguchi, 753-8515, Japan
| | - Akihiko Uda
- Department of Veterinary Science, National Institute of Infectious Diseases, Toyama, 162-8640, Tokyo, Japan
| | - Akitoyo Hotta
- Department of Veterinary Science, National Institute of Infectious Diseases, Toyama, 162-8640, Tokyo, Japan
| | - Masahisa Watarai
- Joint Graduate School of Veterinary Medicine, Yamaguchi University, Yamaguchi, 753-8515, Japan
- Laboratory of Veterinary Public Health, Department of Veterinary Medicine, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, 753-8515, Japan
| | - Ken Maeda
- Joint Graduate School of Veterinary Medicine, Yamaguchi University, Yamaguchi, 753-8515, Japan
- Department of Veterinary Science, National Institute of Infectious Diseases, Toyama, 162-8640, Tokyo, Japan
| | - Ai Takano
- Joint Graduate School of Veterinary Medicine, Yamaguchi University, Yamaguchi, 753-8515, Japan.
- Laboratory of Epidemiology, Department of Veterinary Medicine, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8515, Japan.
| |
Collapse
|
2
|
Adamu A, Reyer F, Lawal N, Hassan AJ, Imam MU, Bello MB, Kraiczy P. Aetiologies of bacterial tick-borne febrile illnesses in humans in Africa: diagnostic limitations and the need for improvement. Front Med (Lausanne) 2024; 11:1419575. [PMID: 39351006 PMCID: PMC11441061 DOI: 10.3389/fmed.2024.1419575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 08/19/2024] [Indexed: 10/04/2024] Open
Abstract
Tick-borne febrile illnesses caused by pathogens like Anaplasma spp., Bartonella spp., Borrelia spp., Ehrlichia spp., Coxiella burnetii, Francisella tularensis, and Rickettsia spp., are significant health concerns in Africa. The epidemiological occurrence of these pathogens is closely linked to the habitats of their vectors, prevalent in rural and semi-urban areas where humans and livestock coexist. The overlapping clinical presentations, non-specific symptoms, and limited access to commercially available in vitro diagnostics in resource-limited settings exacerbate the complexity of accurate diagnoses. This review aimed to systematically extract and analyze existing literature on tick-borne febrile illnesses in Africa, highlighting the diagnostic challenges and presenting an up-to-date overview of the most relevant pathogens affecting human populations. A comprehensive literature search from January 1990 to June 2024 using databases like PubMed, Cochrane Library, Science Direct, EMBASE, and Google Scholar yielded 13,420 articles, of which 70 met the inclusion criteria. Anaplasma spp. were reported in Morocco, Egypt, and South Africa; Francisella spp. in Kenya and Ethiopia; Ehrlichia spp. in Cameroon; Bartonella spp. in Senegal, Namibia, South Africa, and Ethiopia; Borrelia spp. in Senegal, Gabon, Tanzania, and Ethiopia; Coxiella burnetii in 10 countries including Senegal, Mali, and South Africa; and Rickettsia spp. in 14 countries including Senegal, Algeria, and Uganda. Data were analyzed using a fixed-effect model in R version 4.0.1 and visualized on an African map using Tableau version 2022.2. This review highlights the urgent need for improved diagnostics to better manage and control tick-borne febrile illnesses in Africa.
Collapse
Affiliation(s)
- Abdulrahman Adamu
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University Sokoto, Sokoto, Nigeria
- Department of Animal Health and Production Technology, Federal Polytechnic Bali, Taraba State, Nigeria
- Department of Veterinary Microbiology, Usmanu Danfodiyo University Sokoto, Sokoto, Nigeria
| | - Flavia Reyer
- Goethe University Frankfurt, Institute of Medical Microbiology and Infection Control, University Hospital of Frankfurt, Frankfurt, Germany
| | - Nafiú Lawal
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University Sokoto, Sokoto, Nigeria
- Department of Veterinary Microbiology, Usmanu Danfodiyo University Sokoto, Sokoto, Nigeria
| | - Abdurrahman Jibril Hassan
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University Sokoto, Sokoto, Nigeria
- Department of Veterinary Public and Preventive Medicine, Usmanu Danfodiyo University Sokoto, Sokoto, Nigeria
| | - Mustapha Umar Imam
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University Sokoto, Sokoto, Nigeria
- Department of Medical Biochemistry, Usmanu Danfodiyo University Sokoto, Sokoto, Nigeria
| | - Muhammad Bashir Bello
- Infectious Disease Research Department, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Peter Kraiczy
- Goethe University Frankfurt, Institute of Medical Microbiology and Infection Control, University Hospital of Frankfurt, Frankfurt, Germany
| |
Collapse
|
3
|
Matsumoto S, Shimizu T, Uda A, Watanabe K, Watarai M. Role of the JAK2/STAT3 pathway on infection of Francisella novicida. PLoS One 2024; 19:e0310120. [PMID: 39255287 PMCID: PMC11386456 DOI: 10.1371/journal.pone.0310120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 08/26/2024] [Indexed: 09/12/2024] Open
Abstract
Francisella tularensis is a causative agent of the zoonotic disease tularemia, and is highly pathogenic to humans. The pathogenicity of this bacterium is largely attributed to intracellular growth in host cells. Although several bacterial factors important for the intracellular growth have been elucidated, including the type VI secretion system, the host factors involved in the intracellular growth of F. tularensis are largely unknown. To identify the host factors important for F. tularensis infection, 368 compounds were screened for the negative regulation of F. tularensis subsp. novicida (F. novicida) infection. Consequently, 56 inhibitors were isolated that decreased F. novicida infection. Among those inhibitors, we focused on cucurbitacin I, an inhibitor of the JAK2/ STAT3 pathway. Cucurbitacin I and another JAK2/STAT3 inhibitor, Stattic, decreased the intracellular bacterial number of F. novicida. However, these inhibitors failed to affect the cell attachment or the intrasaccular proliferation of F. novicida. In addition, treatment with these inhibitors destabilized actin filaments. These results suggest that the JAK2/STAT3 pathway plays an important role in internalization of F. novicida into host cells through mechanisms involving actin dynamics, such as phagocytosis.
Collapse
Affiliation(s)
- Sonoko Matsumoto
- Laboratory of Veterinary Public Health, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Takashi Shimizu
- One Welfare Education and Research Center, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Akihiko Uda
- Department of Veterinary Science, National Institute of Infectious Diseases, Shinjuku, Tokyo, Japan
| | - Kenta Watanabe
- Laboratory of Veterinary Public Health, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Masahisa Watarai
- Laboratory of Veterinary Public Health, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| |
Collapse
|
4
|
Khaleque MA, Hossain SI, Ali MR, Aly Saad Aly M, Abuelmakarem HS, Al Mamun MS, Hossain Khan MZ. Bioreceptor modified electrochemical biosensors for the detection of life threating pathogenic bacteria: a review. RSC Adv 2024; 14:28487-28515. [PMID: 39247512 PMCID: PMC11378029 DOI: 10.1039/d4ra04038d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 08/23/2024] [Indexed: 09/10/2024] Open
Abstract
The lack of reliable and efficient techniques for early monitoring to stop long-term effects on human health is an increasing problem as the pathogenesis effect of infectious bacteria is growing continuously. Therefore, developing an effective early detection technique coupled with efficient and continuous monitoring of pathogenic bacteria is increasingly becoming a global public health prime target. Electrochemical biosensors are among the strategies that can be utilized for accomplishing that goal with promising potential. In recent years, identifying target biological analytes by interacting with bioreceptors modified electrodes is among the most commonly used detection techniques in electrochemical biosensing strategies. The commonly employed bioreceptors are nucleic acid molecules (DNA or RNA), proteins, antibodies, enzymes, organisms, tissues, and biomimetic components such as molecularly imprinted polymers. Despite the advancement in electrochemical biosensing, developing a reliable and effective biosensor for detecting pathogenic bacteria is still in the infancy stage with so much room for growth. A major milestone in addressing some of the issues and improving the detection pathway is the investigation of specific bacterial detection techniques. The present study covers the fundamental concepts of electrochemical biosensors, human PB illnesses, and the latest electrochemical biosensors based on bioreceptor elements that are designed to detect specific pathogenic bacteria. This study aims to assist researchers with the most up-to-date research work in the field of bio-electrochemical pathogenic bacteria detection and monitoring.
Collapse
Affiliation(s)
- Md Abdul Khaleque
- Dept. of Chemical Engineering, Jashore University of Science and Technology Jashore 7408 Bangladesh
- Laboratory of Nano-bio and Advanced Materials Engineering (NAME), Jashore University of Science and Technology Jashore 7408 Bangladesh
| | - Syed Imdadul Hossain
- Laboratory of Nano-bio and Advanced Materials Engineering (NAME), Jashore University of Science and Technology Jashore 7408 Bangladesh
- Centre for Sophisticated Instrumentation and Research Laboratory (CSIRL), Jashore University of Science and Technology Jashore 7408 Bangladesh
| | - Md Romzan Ali
- Dept. of Chemical Engineering, Jashore University of Science and Technology Jashore 7408 Bangladesh
- Laboratory of Nano-bio and Advanced Materials Engineering (NAME), Jashore University of Science and Technology Jashore 7408 Bangladesh
| | - Mohamed Aly Saad Aly
- Department of Electrical and Computer Engineering at Georgia Tech Shenzhen Institute (GTSI) Shenzhen Guangdong 518055 China
| | - Hala S Abuelmakarem
- Systems and Biomedical Engineering Department, The Higher Institute of Engineering El Shorouk Egypt
| | - Muhammad Shamim Al Mamun
- Chemistry Discipline, School of Science, Engineering and Technology, Khulna University Khulna 9208 Bangladesh
| | - Md Zaved Hossain Khan
- Dept. of Chemical Engineering, Jashore University of Science and Technology Jashore 7408 Bangladesh
- Laboratory of Nano-bio and Advanced Materials Engineering (NAME), Jashore University of Science and Technology Jashore 7408 Bangladesh
| |
Collapse
|
5
|
Rytter H, Roger K, Chhuon C, Ding X, Coureuil M, Jamet A, Henry T, Guerrera IC, Charbit A. Dual proteomics of infected macrophages reveal bacterial and host players involved in the Francisella intracellular life cycle and cell to cell dissemination by merocytophagy. Sci Rep 2024; 14:7797. [PMID: 38565565 PMCID: PMC10987565 DOI: 10.1038/s41598-024-58261-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 03/27/2024] [Indexed: 04/04/2024] Open
Abstract
Bacterial pathogens adapt and replicate within host cells, while host cells develop mechanisms to eliminate them. Using a dual proteomic approach, we characterized the intra-macrophage proteome of the facultative intracellular pathogen, Francisella novicida. More than 900 Francisella proteins were identified in infected macrophages after a 10-h infection. Biotin biosynthesis-related proteins were upregulated, emphasizing the role of biotin-associated genes in Francisella replication. Conversely, proteins encoded by the Francisella pathogenicity island (FPI) were downregulated, supporting the importance of the F. tularensis Type VI Secretion System for vacuole escape, not cytosolic replication. In the host cell, over 300 proteins showed differential expression among the 6200 identified during infection. The most upregulated host protein was cis-aconitate decarboxylase IRG1, known for itaconate production with antimicrobial properties in Francisella. Surprisingly, disrupting IRG1 expression did not impact Francisella's intracellular life cycle, suggesting redundancy with other immune proteins or inclusion in larger complexes. Over-representation analysis highlighted cell-cell contact and actin polymerization in macrophage deregulated proteins. Using flow cytometry and live cell imaging, we demonstrated that merocytophagy involves diverse cell-to-cell contacts and actin polymerization-dependent processes. These findings lay the groundwork for further exploration of merocytophagy and its molecular mechanisms in future research.Data are available via ProteomeXchange with identifier PXD035145.
Collapse
Affiliation(s)
- Héloïse Rytter
- Université Paris CitéINSERM UMR-S1151, CNRS UMR-S8253Institut Necker Enfants Malades, 156-160 rue de Vaugirard, 75015, Paris, France
- INSERM U1151-CNRS UMR 8253, Team 7: Pathogénie des Infections Systémiques, 75015, Paris, France
| | - Kevin Roger
- INSERM US24/CNRS UAR3633, Proteomic Platform Necker, UniversitéParis-Cité, Federative Research Structure Necker, Paris, France
| | - Cerina Chhuon
- INSERM US24/CNRS UAR3633, Proteomic Platform Necker, UniversitéParis-Cité, Federative Research Structure Necker, Paris, France
| | - Xiongqi Ding
- Université Paris CitéINSERM UMR-S1151, CNRS UMR-S8253Institut Necker Enfants Malades, 156-160 rue de Vaugirard, 75015, Paris, France
- INSERM U1151-CNRS UMR 8253, Team 7: Pathogénie des Infections Systémiques, 75015, Paris, France
| | - Mathieu Coureuil
- Université Paris CitéINSERM UMR-S1151, CNRS UMR-S8253Institut Necker Enfants Malades, 156-160 rue de Vaugirard, 75015, Paris, France
- INSERM U1151-CNRS UMR 8253, Team 7: Pathogénie des Infections Systémiques, 75015, Paris, France
| | - Anne Jamet
- Université Paris CitéINSERM UMR-S1151, CNRS UMR-S8253Institut Necker Enfants Malades, 156-160 rue de Vaugirard, 75015, Paris, France
- INSERM U1151-CNRS UMR 8253, Team 7: Pathogénie des Infections Systémiques, 75015, Paris, France
| | - Thomas Henry
- CIRI, Centre International de Recherche en Infectiologie, Université Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, 69007, Lyon, France
| | - Ida Chiara Guerrera
- INSERM US24/CNRS UAR3633, Proteomic Platform Necker, UniversitéParis-Cité, Federative Research Structure Necker, Paris, France.
| | - Alain Charbit
- Université Paris CitéINSERM UMR-S1151, CNRS UMR-S8253Institut Necker Enfants Malades, 156-160 rue de Vaugirard, 75015, Paris, France.
- INSERM U1151-CNRS UMR 8253, Team 7: Pathogénie des Infections Systémiques, 75015, Paris, France.
| |
Collapse
|
6
|
Mlynek KD, Toothman RG, Martinez EE, Qiu J, Richardson JB, Bozue JA. Mutation of wbtJ, a N-formyltransferase involved in O-antigen synthesis, results in biofilm formation, phase variation and attenuation in Francisella tularensis. MICROBIOLOGY (READING, ENGLAND) 2024; 170:001437. [PMID: 38421161 PMCID: PMC10924466 DOI: 10.1099/mic.0.001437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 01/29/2024] [Indexed: 03/02/2024]
Abstract
Two clinically important subspecies, Francisella tularensis subsp. tularensis (type A) and F. tularensis subsp. holarctica (type B) are responsible for most tularaemia cases, but these isolates typically form a weak biofilm under in vitro conditions. Phase variation of the F. tularensis lipopolysaccharide (LPS) has been reported in these subspecies, but the role of variation is unclear as LPS is crucial for virulence. We previously demonstrated that a subpopulation of LPS variants can constitutively form a robust biofilm in vitro, but it is unclear whether virulence was affected. In this study, we show that biofilm-forming variants of both fully virulent F. tularensis subspecies were highly attenuated in the murine tularaemia model by multiple challenge routes. Genomic sequencing was performed on these strains, which revealed that all biofilm-forming variants contained a lesion within the wbtJ gene, a formyltransferase involved in O-antigen synthesis. A ΔwbtJ deletion mutant recapitulated the biofilm, O-antigen and virulence phenotypes observed in natural variants and could be rescued through complementation with a functional wbtJ gene. Since the spontaneously derived biofilm-forming isolates in this study were a subpopulation of natural variants, reversion events to the wbtJ gene were detected that eliminated the phenotypes associated with biofilm variants and restored virulence. These results demonstrate a role for WbtJ in biofilm formation, LPS variation and virulence of F. tularensis.
Collapse
Affiliation(s)
- Kevin D. Mlynek
- Bacteriology Division, US ARMY Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, USA
| | - Ronald G. Toothman
- Bacteriology Division, US ARMY Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, USA
| | - Elsie E. Martinez
- Bacteriology Division, US ARMY Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, USA
| | - Ju Qiu
- Regulated Research Administration Division, USAMRIID, Frederick, MD, USA
| | | | - Joel A. Bozue
- Bacteriology Division, US ARMY Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, USA
| |
Collapse
|
7
|
van Hoek ML, Marchesani A, Rawat M. Diverse roles of low-molecular weight thiol GSH in Francisella's virulence, location sensing and GSH-stealing from host. CURRENT RESEARCH IN MICROBIAL SCIENCES 2023; 6:100218. [PMID: 38303966 PMCID: PMC10831187 DOI: 10.1016/j.crmicr.2023.100218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2024] Open
Abstract
Low-molecular weight (LMW) thiols, encompassing peptides and small proteins with active cysteine residue(s), are important to bacteria as they are involved in a wide range of redox reactions. They include the tripeptide glutathione (GSH) and the small redox proteins, thioredoxins and glutaredoxins. We review the low MW thiols and related molecules in Francisella species and what role they may play in growth and virulence. Genes for GSH biosynthesis, metabolism and thioredoxins are present in all strains of Francisella, including the fully human-virulent strains. GSH and cysteine (CSH) are the major LMW thiols in Francisella extracts. We explore the potential role of the LMW thiols to overcome the nutritional challenges of intracellular growth (high GSH conditions) as well as the nutritional challenges of planktonic growth (low GSH conditions), and their contribution to Francisella's sensing its environmental location. Francisella may also use GSH as a source of CSH, for which it is auxotrophic. "Glutathione stealing" from the host may be an important part of Francisella's success strategy as a facultative intracellular pathogen both to detect its location and obtain CSH. An understanding of GSH metabolism in Francisella provides insights into the interaction of this pathogen with its host and may reveal additional targets for therapeutic intervention for tularemia infections.
Collapse
Affiliation(s)
- Monique L. van Hoek
- School of Systems Biology, George Mason University, Manassas, VA, United States
| | | | - Mamta Rawat
- Biology Department, California State University, Fresno, CA, United States
| |
Collapse
|
8
|
Zaman A, French JB, Carpino N. The Sts Proteins: Modulators of Host Immunity. Int J Mol Sci 2023; 24:8834. [PMID: 37240179 PMCID: PMC10218301 DOI: 10.3390/ijms24108834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/08/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
The suppressor of TCR signaling (Sts) proteins, Sts-1 and Sts-2, are a pair of closely related signaling molecules that belong to the histidine phosphatase (HP) family of enzymes by virtue of an evolutionarily conserved C-terminal phosphatase domain. HPs derive their name from a conserved histidine that is important for catalytic activity and the current evidence indicates that the Sts HP domain plays a critical functional role. Sts-1HP has been shown to possess a readily measurable protein tyrosine phosphatase activity that regulates a number of important tyrosine-kinase-mediated signaling pathways. The in vitro catalytic activity of Sts-2HP is significantly lower than that of Sts-1HP, and its signaling role is less characterized. The highly conserved unique structure of the Sts proteins, in which additional domains, including one that exhibits a novel phosphodiesterase activity, are juxtaposed together with the phosphatase domain, suggesting that Sts-1 and -2 occupy a specialized intracellular signaling niche. To date, the analysis of Sts function has centered predominately around the role of Sts-1 and -2 in regulating host immunity and other responses associated with cells of hematopoietic origin. This includes their negative regulatory role in T cells, platelets, mast cells and other cell types, as well as their less defined roles in regulating host responses to microbial infection. Regarding the latter, the use of a mouse model lacking Sts expression has been used to demonstrate that Sts contributes non-redundantly to the regulation of host immunity toward a fungal pathogen (C. albicans) and a Gram-negative bacterial pathogen (F. tularensis). In particular, Sts-/- animals demonstrate significant resistance to lethal infections of both pathogens, a phenotype that is correlated with some heightened anti-microbial responses of phagocytes derived from mutant mice. Altogether, the past several years have seen steady progress in our understanding of Sts biology.
Collapse
Affiliation(s)
- Anika Zaman
- Graduate Program in Molecular and Cellular Pharmacology, Stony Brook University, Stony Brook, NY 11794, USA;
| | - Jarrod B. French
- Hormel Institute, University of Minnesota, 801 16th Ave NE, Austin, MN 55912, USA;
| | - Nick Carpino
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
9
|
Dow J, Cytlak UM, Casulli J, McEntee CP, Smedley C, Hodge SH, D’Elia RV, Hepworth MR, Travis MA. Group 2 Innate Lymphoid Cells Are Detrimental to the Control of Infection with Francisella tularensis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:618-627. [PMID: 36602520 PMCID: PMC9946898 DOI: 10.4049/jimmunol.2100651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 12/02/2022] [Indexed: 01/06/2023]
Abstract
Innate lymphoid cells (ILCs) are capable of rapid response to a wide variety of immune challenges, including various respiratory pathogens. Despite this, their role in the immune response against the lethal intracellular bacterium Francisella tularensis is not yet known. In this study, we demonstrate that infection of the airways with F. tularensis results in a significant reduction in lung type 2 ILCs (ILC2s) in mice. Conversely, the expansion of ILC2s via treatment with the cytokine IL-33, or by adoptive transfer of ILC2s, resulted in significantly enhanced bacterial burdens in the lung, liver, and spleen, suggesting that ILC2s may favor severe infection. Indeed, specific reduction of ILC2s in a transgenic mouse model results in a reduction in lung bacterial burden. Using an in vitro culture system, we show that IFN-γ from the live vaccine strain-infected lung reduces ILC2 numbers, suggesting that this cytokine in the lung environment is mechanistically important in reducing ILC2 numbers during infection. Finally, we show Ab-mediated blockade of IL-5, of which ILC2s are a major innate source, reduces bacterial burden postinfection, suggesting that IL-5 production by ILC2s may play a role in limiting protective immunity. Thus, overall, we highlight a negative role for ILC2s in the control of infection with F. tularensis. Our work therefore highlights the role of ILC2s in determining the severity of potentially fatal airway infections and raises the possibility of interventions targeting innate immunity during infection with F. tularensis to benefit the host.
Collapse
Affiliation(s)
- Joshua Dow
- Lydia Becker Institute for Immunology and Inflammation, Manchester, United Kingdom
- Wellcome Trust Centre for Cell-Matrix Research, Manchester, United Kingdom
- Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, United Kingdom
| | - Urszula M. Cytlak
- Lydia Becker Institute for Immunology and Inflammation, Manchester, United Kingdom
- Wellcome Trust Centre for Cell-Matrix Research, Manchester, United Kingdom
- Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, United Kingdom
- Targeted Therapy Group, Division of Cancer Sciences, Manchester, United Kingdom
| | - Joshua Casulli
- Lydia Becker Institute for Immunology and Inflammation, Manchester, United Kingdom
- Wellcome Trust Centre for Cell-Matrix Research, Manchester, United Kingdom
- Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, United Kingdom
| | - Craig P. McEntee
- Lydia Becker Institute for Immunology and Inflammation, Manchester, United Kingdom
- Wellcome Trust Centre for Cell-Matrix Research, Manchester, United Kingdom
- Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, United Kingdom
| | - Catherine Smedley
- Lydia Becker Institute for Immunology and Inflammation, Manchester, United Kingdom
- Wellcome Trust Centre for Cell-Matrix Research, Manchester, United Kingdom
- Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, United Kingdom
| | - Suzanne H. Hodge
- Lydia Becker Institute for Immunology and Inflammation, Manchester, United Kingdom
- Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, United Kingdom
| | - Riccardo V. D’Elia
- Defence Science and Technology Laboratory, Porton Down, Salisbury, United Kingdom; and
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Matthew R. Hepworth
- Lydia Becker Institute for Immunology and Inflammation, Manchester, United Kingdom
- Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, United Kingdom
| | - Mark A. Travis
- Lydia Becker Institute for Immunology and Inflammation, Manchester, United Kingdom
- Wellcome Trust Centre for Cell-Matrix Research, Manchester, United Kingdom
- Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
10
|
Bachert BA, Bozue JA. Peptidoglycan enzymes of Francisella: Roles in cell morphology and pathogenesis, and potential as therapeutic targets. Front Microbiol 2023; 13:1099312. [PMID: 36713212 PMCID: PMC9877522 DOI: 10.3389/fmicb.2022.1099312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 12/28/2022] [Indexed: 01/13/2023] Open
Abstract
Peptidoglycan, found within the cell wall of bacteria, is a structure critical for maintaining cell morphology and providing a protective barrier in diverse environments. Peptidoglycan is a remarkably dynamic structure that is constantly remodeled during cell growth and division by various peptidoglycan enzymes. Numerous peptidoglycan enzymes have been characterized from diverse bacteria and are highly sought after as targets for therapeutics. However, very little is known about these enzymes within the biothreat agent Francisella tularensis. As the causative agent of tularemia, F. tularensis is classified as a category A biothreat pathogen, in part due to its low infectious dose and lack of FDA-approved vaccine. Many bacterial species encode multiple peptidoglycan enzymes with redundant functions that allow for compensation if one of the enzymes are inactivated. In contrast, F. tularensis appears to lack this redundancy, indicating peptidoglycan enzymes may be completely essential for growth and could be exploited as targets for medical countermeasures. Indeed, several peptidoglycan enzymes in F. tularensis have been shown to play important roles in cell division, cell morphology, virulence, and modulation of host response. The aim of this review is to summarize findings from the current literature on peptidoglycan enzymes present in Francisella and discuss areas where future research efforts might be directed. We conclude that Francisella harbors a distinct set of peptidoglycan enzymes important for cell growth and virulence and represent potentially valuable targets for the development of novel therapeutics.
Collapse
|
11
|
Välikangas T, Suomi T, Chandler CE, Scott AJ, Tran BQ, Ernst RK, Goodlett DR, Elo LL. Benchmarking tools for detecting longitudinal differential expression in proteomics data allows establishing a robust reproducibility optimization regression approach. Nat Commun 2022; 13:7877. [PMID: 36550114 PMCID: PMC9780321 DOI: 10.1038/s41467-022-35564-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
Quantitative proteomics has matured into an established tool and longitudinal proteomics experiments have begun to emerge. However, no effective, simple-to-use differential expression method for longitudinal proteomics data has been released. Typically, such data is noisy, contains missing values, and has only few time points and biological replicates. To address this need, we provide a comprehensive evaluation of several existing differential expression methods for high-throughput longitudinal omics data and introduce a Robust longitudinal Differential Expression (RolDE) approach. The methods are evaluated using over 3000 semi-simulated spike-in proteomics datasets and three large experimental datasets. In the comparisons, RolDE performs overall best; it is most tolerant to missing values, displays good reproducibility and is the top method in ranking the results in a biologically meaningful way. Furthermore, RolDE is suitable for different types of data with typically unknown patterns in longitudinal expression and can be applied by non-experienced users.
Collapse
Affiliation(s)
- Tommi Välikangas
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520, Turku, Finland
| | - Tomi Suomi
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520, Turku, Finland
| | | | - Alison J Scott
- University of Maryland - Baltimore, Baltimore, MD, 21201, USA
| | - Bao Q Tran
- US Army 20th Support Command CBRNE Analytical and Remediation Activity, Baltimore, MD, 21010-5424, USA
| | - Robert K Ernst
- University of Maryland - Baltimore, Baltimore, MD, 21201, USA
| | - David R Goodlett
- University of Victoria, Victoria, BC, V8P 3E6, Canada
- International Centre for Cancer Vaccine Science, Gdansk, Poland
| | - Laura L Elo
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520, Turku, Finland.
- Institute of Biomedicine, University of Turku, FI-20520, Turku, Finland.
| |
Collapse
|
12
|
Mlynek KD, Bozue JA. Why vary what's working? Phase variation and biofilm formation in Francisella tularensis. Front Microbiol 2022; 13:1076694. [PMID: 36560950 PMCID: PMC9763628 DOI: 10.3389/fmicb.2022.1076694] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 11/17/2022] [Indexed: 12/12/2022] Open
Abstract
The notoriety of high-consequence human pathogens has increased in recent years and, rightfully, research efforts have focused on understanding host-pathogen interactions. Francisella tularensis has been detected in an impressively broad range of vertebrate hosts as well as numerous arthropod vectors and single-celled organisms. Two clinically important subspecies, F. tularensis subsp. tularensis (Type A) and F. tularensis subsp. holarctica (Type B), are responsible for the majority of tularemia cases in humans. The success of this bacterium in mammalian hosts can be at least partly attributed to a unique LPS molecule that allows the bacterium to avoid detection by the host immune system. Curiously, phase variation of the O-antigen incorporated into LPS has been documented in these subspecies of F. tularensis, and these variants often display some level of attenuation in infection models. While the role of phase variation in F. tularensis biology is unclear, it has been suggested that this phenomenon can aid in environmental survival and persistence. Biofilms have been established as the predominant lifestyle of many bacteria in the environment, though, it was previously thought that Type A and B isolates of F. tularensis typically form poor biofilms. Recent studies question this ideology as it was shown that alteration of the O-antigen allows robust biofilm formation in both Type A and B isolates. This review aims to explore the link between phase variation of the O-antigen, biofilm formation, and environmental persistence with an emphasis on clinically relevant subspecies and how understanding these poorly studied mechanisms could lead to new medical countermeasures to combat tularemia.
Collapse
|
13
|
Roth K, Chelikam N, Rathore H, Chittivelu S. An Uncommon Presentation of Pulmonary Tularemia: A Case Report and Literature Review. Cureus 2022; 14:e30379. [DOI: 10.7759/cureus.30379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2022] [Indexed: 11/05/2022] Open
|
14
|
Fever and Ulcerative Skin Lesions in a Patient Referred for Altered Mental Status: Clinical and Microbiological Diagnosis of Ulceroglandular Tularemia. Trop Med Infect Dis 2022; 7:tropicalmed7090220. [PMID: 36136631 PMCID: PMC9504304 DOI: 10.3390/tropicalmed7090220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 12/04/2022] Open
Abstract
Background: Tularemia is a devastating disease that affects multiple organ systems and can have several different presentations. In its most frequent form—that of ulceroglandular tularemia—a detailed history and physical examination can enable a physician to make the diagnosis clinically, leading to the prompt initiation of the appropriate antibiotic treatment. Detailed Case Description: A 63-year-old man was brought by ambulance to the emergency department for an evaluation of an altered mental status noted by his psychiatrist at a telehealth appointment. A physical examination revealed a fever and two ulcerative lesions with a central eschar on his left leg (of which the patient was unaware) with ipsilateral tender inguinal lymphadenopathy. When asked, the patient recalled visiting Martha’s Vineyard and having removed ticks from his legs. Gentamicin was administered on the clinical suspicion of ulceroglandular tularemia. Blood and skin lesion cultures grew Gram-negative rods, which were confirmed to be Francisella tularensis on hospital day eight, and the patient fully recovered. Conclusion: This case highlights the importance of clinician perception of altered mental status as a key alarm sign, the necessity of a thorough physical exam independent of the chief compliant in the emergency department, and the essential role of pattern recognition by front-line providers for the appropriate management of uncommon but serious infections such as tularemia.
Collapse
|
15
|
Tularemia Presenting Solely with Cervical Lymphadenopathy and Fever. Diagnostics (Basel) 2022; 12:diagnostics12082000. [PMID: 36010350 PMCID: PMC9407280 DOI: 10.3390/diagnostics12082000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/12/2022] [Accepted: 08/16/2022] [Indexed: 11/21/2022] Open
Abstract
A 52-year-old German female presented with cervical lymphadenopathy and fever. Despite the initial symptom-presentation leading to the consideration of sarcoidosis, lymphoma, tuberculosis, and toxoplasmosis, an extensive serologic and histo- and molecular pathologic workup eventually indicated a likely diagnosis of tularemia. This case brings to light that tularemia is a diagnostic challenge and requires high reliance on the epidemiological context thorough patient history, and an extensive interdisciplinary diagnostic workup.
Collapse
|
16
|
Xu JH, Kang L, Yuan B, Feng ZH, Li SQ, Wang J, Wang YR, Xin WW, Gao S, Li JX, Sun YS, Wang JL, Yuan Y. Development and evaluation of a rapid RPA/CRISPR-based detection of Francisella tularensis. Front Microbiol 2022; 13:901520. [PMID: 36033876 PMCID: PMC9399789 DOI: 10.3389/fmicb.2022.901520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Francisella tularensis is a dangerous pathogen that causes an extremely contagious zoonosis in humans named tularemia. Given its low-dose morbidity, the potential to be fatal, and aerosol spread, it is regarded as a severe threat to public health. The US Centers for Disease Control and Prevention (CDC) has classified it as a category A potential agent for bioterrorism and a Tier 1 Select Agent. Herein, we combined recombinase polymerase amplification (RPA) with CRISPR/Cas12a system to select the F. tularensis target gene (TUL4), creating a two-pronged rapid and ultrasensitive diagnostic method for detecting F. tularensis. The real-time RPA (RT-RPA) assay detected F. tularensis within 10 min at a sensitivity of 5 copies/reaction, F. tularensis genomic DNA of 5 fg, and F. tularensis of 2 × 102 CFU/ml; the RPA-CRISPR/Cas12a assay detects F. tularensis within 40 min at a sensitivity of 0.5 copies/reaction, F. tularensis genomic DNA of 1 fg, and F. tularensis of 2 CFU/ml. Furthermore, the evaluation of specificity showed that both assays were highly specific to F. tularensis. More importantly, in a test of prepared simulated blood and sewage samples, the RT-RPA assay results were consistent with RT-PCR assay results, and the RPA-CRISPR/Cas12a assay could detect a minute amount of F. tularensis genomic DNA (2.5 fg). There was no nonspecific detection with blood samples and sewage samples, giving the tests a high practical application value. For example, in on-site and epidemic areas, the RT-RPA was used for rapid screening and the RPA-CRISPR/Cas12a assay was used for more accurate diagnosis.
Collapse
Affiliation(s)
- Jian-Hao Xu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences (AMMS), Beijing, China
- School of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Lin Kang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences (AMMS), Beijing, China
| | - Bing Yuan
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences (AMMS), Beijing, China
| | - Zi-Han Feng
- Department of Disease Control and Prevention, The No. 96609 Hospital of Chinese People's Liberation Army, Yinchuan, China
| | - Shi-Qing Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences (AMMS), Beijing, China
| | - Jing Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences (AMMS), Beijing, China
| | - Ya-Ru Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences (AMMS), Beijing, China
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Wen-Wen Xin
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences (AMMS), Beijing, China
| | - Shan Gao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences (AMMS), Beijing, China
| | - Jia-Xin Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences (AMMS), Beijing, China
| | - Yan-Song Sun
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences (AMMS), Beijing, China
- *Correspondence: Yan-Song Sun
| | - Jing-Lin Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences (AMMS), Beijing, China
- School of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
- Jing-Lin Wang
| | - Yuan Yuan
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences (AMMS), Beijing, China
- Yuan Yuan
| |
Collapse
|
17
|
Kinkead LC, Krysa SJ, Allen LAH. Neutrophil Survival Signaling During Francisella tularensis Infection. Front Cell Infect Microbiol 2022; 12:889290. [PMID: 35873156 PMCID: PMC9299441 DOI: 10.3389/fcimb.2022.889290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 06/07/2022] [Indexed: 12/18/2022] Open
Abstract
Neutrophils are the most abundant and shortest-lived leukocytes in humans and tight regulation of neutrophil turnover via constitutive apoptosis is essential for control of infection and resolution of inflammation. Accordingly, aberrant neutrophil turnover is hallmark of many disease states. We have shown in previous work that the intracellular bacterial pathogen Francisella tularensis markedly prolongs human neutrophil lifespan. This is achieved, in part, by changes in neutrophil gene expression. Still unknown is the contribution of major neutrophil pro-survival signaling cascades to this process. The objective of this study was to interrogate the contributions of ERK and p38 MAP kinase, Class I phosphoinositide 3-kinases (PI3K), AKT, and NF-κB to neutrophil survival in our system. We demonstrate that both ERK2 and p38α were activated in F. tularensis-infected neutrophils, but only p38α MAPK was required for delayed apoptosis and the rate of cell death in the absence of infection was unchanged. Apoptosis of both infected and uninfected neutrophils was markedly accelerated by the pan-PI3K inhibitor LY2094002, but AKT phosphorylation was not induced, and neutrophil death was not enhanced by AKT inhibitors. In addition, isoform specific and selective inhibitors revealed a unique role for PI3Kα in neutrophil survival after infection, whereas only simultaneous inhibition of PI3Kα and PI3kδ accelerated death of the uninfected controls. Finally, we show that inhibition of NF-κB triggered rapid death of neutrophil after infection. Thus, we defined roles for p38α, PI3Kα and NF-κB delayed apoptosis of F. tularensis-infected cells and advanced understanding of Class IA PI3K isoform activity in human neutrophil survival.
Collapse
Affiliation(s)
- Lauren C. Kinkead
- Inflammation Program, University of Iowa, Iowa City, IA, United States,Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States,Iowa City VA Health Care System, Iowa City, IA, United States
| | - Samantha J. Krysa
- Inflammation Program, University of Iowa, Iowa City, IA, United States,Iowa City VA Health Care System, Iowa City, IA, United States,Molecular Medicine Training Program, University of Iowa, Iowa City, IA, United States
| | - Lee-Ann H. Allen
- Inflammation Program, University of Iowa, Iowa City, IA, United States,Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States,Iowa City VA Health Care System, Iowa City, IA, United States,Molecular Medicine Training Program, University of Iowa, Iowa City, IA, United States,Department of Medicine, Division of Infectious Diseases, University of Iowa, Iowa City, IA, United States,Harry S. Truman Memorial VA Hospital, Columbia, MO, United States,Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, United States,*Correspondence: Lee-Ann H. Allen,
| |
Collapse
|
18
|
Goel P, Panchal T, Kaushik N, Chauhan R, Saini S, Ahuja V, Thakur CJ. In silico functional and structural characterization revealed virulent proteins of Francisella tularensis strain SCHU4. MOLECULAR BIOLOGY RESEARCH COMMUNICATIONS 2022; 11:73-84. [PMID: 36059929 PMCID: PMC9336787 DOI: 10.22099/mbrc.2022.43128.1719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Francisella tularensis is a pathogenic, aerobic gram-negative coccobacillus bacterium. It is the causative agent of tularemia, a rare infectious disease that can attack skin, lungs, eyes, and lymph nodes. The genome of F. tularensis has been sequenced, and ~16% of the proteome is still uncharacterized. Characterizations of these proteins are essential to find new drug targets for better therapeutics. In silico characterization of proteins has become an extremely important approach to determine the functionality of proteins as experimental functional elucidation is unable to keep pace with the current growth of the sequence database. Initially, we have annotated 577 Hypothetical Proteins (HPs) of F. tularensis strain SCHU4 with seven bioinformatics tools which characterized them based on the family, domain and motif. Out of 577 HPs, 119 HPs were annotated by five or more tools and are further screened to predict their virulence properties, subcellular localization, transmembrane helices as well as physicochemical parameters. VirulentPred predicted 66 HPs out of 119 as virulent. These virulent proteins were annotated to find the interacting partner using STRING, and proteins with high confidence interaction scores were used to predict their 3D structures using Phyre2. The three virulent proteins Q5NH99 (phosphoserine phosphatase), Q5NG42 (Cystathionine beta-synthase) and Q5NG83 (Rrf2-type helix turn helix domain) were predicted to involve in modulation of cytoskeletal and innate immunity of host, H2S (hydrogen sulfide) based antibiotic tolerance and nitrite and iron metabolism of bacteria. The above predicted virulent proteins can serve as novel drug targets in the era of antibiotic resistance.
Collapse
Affiliation(s)
- Prerna Goel
- Department of Bioinformatics, Goswami Ganesh Dutta Sanatan Dharma College, Sector 32 C, Chandigarh, India
| | - Tanya Panchal
- Department of Bioinformatics, Goswami Ganesh Dutta Sanatan Dharma College, Sector 32 C, Chandigarh, India
| | - Nandini Kaushik
- Department of Bioinformatics, Goswami Ganesh Dutta Sanatan Dharma College, Sector 32 C, Chandigarh, India
| | - Ritika Chauhan
- Department of Bioinformatics, Goswami Ganesh Dutta Sanatan Dharma College, Sector 32 C, Chandigarh, India
| | - Sandeep Saini
- Department of Bioinformatics, Goswami Ganesh Dutta Sanatan Dharma College, Sector 32 C, Chandigarh, India,Department of Biophysics, Panjab University, Sector 25, 160014, Chandigarh, India
| | - Vartika Ahuja
- Department of Bioinformatics, Goswami Ganesh Dutta Sanatan Dharma College, Sector 32 C, Chandigarh, India
| | - Chander Jyoti Thakur
- Department of Bioinformatics, Goswami Ganesh Dutta Sanatan Dharma College, Sector 32 C, Chandigarh, India,Corresponding Author: Department of Bioinformatics,Goswami Ganesh Dutta Sanatan Dharma College Sector 32 C, Chandigarh, India, 160030. Tel: +91 8699776533; Fax: +91 1722661077, E. mail:
| |
Collapse
|
19
|
Ozanic M, Marecic V, Knezevic M, Kelava I, Stojková P, Lindgren L, Bröms JE, Sjöstedt A, Abu Kwaik Y, Santic M. The type IV pili component PilO is a virulence determinant of Francisella novicida. PLoS One 2022; 17:e0261938. [PMID: 35077486 PMCID: PMC8789160 DOI: 10.1371/journal.pone.0261938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 12/14/2021] [Indexed: 12/03/2022] Open
Abstract
Francisella tularensis is a highly pathogenic intracellular bacterium that causes the disease tularemia. While its ability to replicate within cells has been studied in much detail, the bacterium also encodes a less characterised type 4 pili (T4P) system. T4Ps are dynamic adhesive organelles identified as major virulence determinants in many human pathogens. In F. tularensis, the T4P is required for adherence to the host cell, as well as for protein secretion. Several components, including pilins, a pili peptidase, a secretin pore and two ATPases, are required to assemble a functional T4P, and these are encoded within distinct clusters on the Francisella chromosome. While some of these components have been functionally characterised, the role of PilO, if any, still is unknown. Here, we examined the role of PilO in the pathogenesis of F. novicida. Our results show that the PilO is essential for pilus assembly on the bacterial surface. In addition, PilO is important for adherence of F. novicida to human monocyte-derived macrophages, secretion of effector proteins and intracellular replication. Importantly, the pilO mutant is attenuated for virulence in BALB/c mice regardless of the route of infection. Following intratracheal and intradermal infection, the mutant caused no histopathology changes, and demonstrated impaired phagosomal escape and replication within lung liver as well as spleen. Thus, PilO is an essential virulence determinant of F. novicida.
Collapse
Affiliation(s)
- Mateja Ozanic
- Faculty of Medicine, Department of Microbiology and Parasitology, University of Rijeka, Rijeka, Croatia
| | - Valentina Marecic
- Faculty of Medicine, Department of Microbiology and Parasitology, University of Rijeka, Rijeka, Croatia
| | - Masa Knezevic
- Faculty of Medicine, Department of Microbiology and Parasitology, University of Rijeka, Rijeka, Croatia
| | - Ina Kelava
- Faculty of Medicine, Department of Microbiology and Parasitology, University of Rijeka, Rijeka, Croatia
| | - Pavla Stojková
- Department of Clinical Microbiology and Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
| | - Lena Lindgren
- Department of Clinical Microbiology and Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
| | - Jeanette E. Bröms
- Department of Clinical Microbiology and Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
| | - Anders Sjöstedt
- Department of Clinical Microbiology and Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
| | - Yousef Abu Kwaik
- Department of Microbiology and Immunology and Center for Predictive Medicine, College of Medicine, University of Louisville, Louisville, Kentucky, United States of America
| | - Marina Santic
- Faculty of Medicine, Department of Microbiology and Parasitology, University of Rijeka, Rijeka, Croatia
| |
Collapse
|
20
|
Barbosa CHD, Lantier L, Reynolds J, Wang J, Re F. Critical role of IL-25-ILC2-IL-5 axis in the production of anti-Francisella LPS IgM by B1 B cells. PLoS Pathog 2021; 17:e1009905. [PMID: 34449811 PMCID: PMC8428711 DOI: 10.1371/journal.ppat.1009905] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 09/09/2021] [Accepted: 08/17/2021] [Indexed: 01/21/2023] Open
Abstract
B1 cells, a subset of B lymphocytes whose developmental origin, phenotype, and function differ from that of conventional B2 cells, are the main source of “natural” IgM but can also respond to infection by rapidly producing pathogen-specific IgM directed against T-independent antigens. Francisella tularensis (Ft) is a Gram-negative bacterium that causes tularemia. Infection with Ft Live Vaccine Strain activates B1 cells for production of IgM directed against the bacterial LPS in a process incompletely understood. Here we show that immunization with purified Ft LPS elicits production of LPS-specific IgM and IgG3 by B1 cells independently of TLR2 or MyD88. Immunization, but not infection, generated peritoneum-resident memory B1 cells that differentiated into LPS-specific antibody secreting cells (ASC) upon secondary challenge. IL-5 was rapidly induced by immunization with Ft LPS and was required for production of LPS-specific IgM. Antibody-mediated depletion of ILC2 indicated that these cells were the source of IL-5 and were required for IgM production. IL-25, an alarmin that strongly activates ILC2, was rapidly secreted in response to immunization or infection and its administration to mice significantly increased IgM production and B1 cell differentiation to ASC. Conversely, mice lacking IL-17RB, the IL-25 receptor, showed impaired IL-5 induction, IgM production, and B1 ASC differentiation in response to immunization. Administration of IL-5 to Il17rb-/- mice rescued these B1 cells-mediated responses. Il17rb-/- mice were more susceptible to infection with Ft LVS and failed to develop immunity upon secondary challenge suggesting that LPS-specific IgM is one of the protective adaptive immune mechanisms against tularemia. Our results indicated that immunization with Ft LPS triggers production of IL-25 that, through stimulation of IL-5 release by ILC2, promotes B1 cells activation and differentiation into IgM secreting cells. By revealing the existence of an IL-25-ILC2-IL-5 axis our results suggest novel strategies to improve vaccination against T-independent bacterial antigens. B1 cells are a subset of B lymphocytes that participate in the immune response to infection by producing antibodies of the IgM class. Here we investigate the mechanisms that control B1 cells activation and production of IgM directed against the lipopolysaccharide (LPS) of Francisella tularensis, a Gram-negative bacterium that causes tularemia. Using a mouse model of tularemia, our results revealed that Francisella LPS elicits production of the cytokine IL-25 that in turn activates blood cells called Innate Lymphoid Cells 2 (ILC2). Once activated, ILC2 produce the cytokine IL-5 that is required for activation of B1 cells and production of IgM. Mice unresponsive to IL-25 are more susceptible to F. tularensis infection. By revealing the existence of an IL-25-ILC2-IL-5 axis our results suggest novel strategies to improve vaccination against bacteria.
Collapse
Affiliation(s)
- Carlos Henrique D. Barbosa
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, United States of America
| | - Louis Lantier
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, United States of America
| | - Joseph Reynolds
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, United States of America
| | - Jinyong Wang
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, United States of America
| | - Fabio Re
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
21
|
Rytter H, Jamet A, Ziveri J, Ramond E, Coureuil M, Lagouge-Roussey P, Euphrasie D, Tros F, Goudin N, Chhuon C, Nemazanyy I, de Moraes FE, Labate C, Guerrera IC, Charbit A. The pentose phosphate pathway constitutes a major metabolic hub in pathogenic Francisella. PLoS Pathog 2021; 17:e1009326. [PMID: 34339477 PMCID: PMC8360588 DOI: 10.1371/journal.ppat.1009326] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 08/12/2021] [Accepted: 07/14/2021] [Indexed: 11/19/2022] Open
Abstract
Metabolic pathways are now considered as intrinsic virulence attributes of pathogenic bacteria and thus represent potential targets for antibacterial strategies. Here we focused on the role of the pentose phosphate pathway (PPP) and its connections with other metabolic pathways in the pathophysiology of Francisella novicida. The involvement of the PPP in the intracellular life cycle of Francisella was first demonstrated by studying PPP inactivating mutants. Indeed, we observed that inactivation of the tktA, rpiA or rpe genes severely impaired intramacrophage multiplication during the first 24 hours. However, time-lapse video microscopy demonstrated that rpiA and rpe mutants were able to resume late intracellular multiplication. To better understand the links between PPP and other metabolic networks in the bacterium, we also performed an extensive proteo-metabolomic analysis of these mutants. We show that the PPP constitutes a major bacterial metabolic hub with multiple connections to glycolysis, the tricarboxylic acid cycle and other pathways, such as fatty acid degradation and sulfur metabolism. Altogether our study highlights how PPP plays a key role in the pathogenesis and growth of Francisella in its intracellular niche.
Collapse
Affiliation(s)
- Héloise Rytter
- Université de Paris, Paris, France
- INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades. Team 7: Pathogénie des Infections Systémiques, Paris, France
| | - Anne Jamet
- Université de Paris, Paris, France
- INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades. Team 7: Pathogénie des Infections Systémiques, Paris, France
| | - Jason Ziveri
- Université de Paris, Paris, France
- INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades. Team 7: Pathogénie des Infections Systémiques, Paris, France
| | - Elodie Ramond
- Université de Paris, Paris, France
- INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades. Team 7: Pathogénie des Infections Systémiques, Paris, France
| | - Mathieu Coureuil
- Université de Paris, Paris, France
- INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades. Team 7: Pathogénie des Infections Systémiques, Paris, France
| | - Pauline Lagouge-Roussey
- Université de Paris, Paris, France
- INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades. Team 7: Pathogénie des Infections Systémiques, Paris, France
| | - Daniel Euphrasie
- Université de Paris, Paris, France
- INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades. Team 7: Pathogénie des Infections Systémiques, Paris, France
| | - Fabiola Tros
- Université de Paris, Paris, France
- INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades. Team 7: Pathogénie des Infections Systémiques, Paris, France
| | - Nicolas Goudin
- Pole Bio-analyse d’images, Structure Fédérative de Recherche Necker INSERM US24- CNRS UMS 3633, Paris, France
| | - Cerina Chhuon
- Université de Paris, Paris, France
- Plateforme Protéome Institut Necker, PPN, Structure Fédérative de Recherche Necker INSERM US24-CNRS UMS 3633, Paris, France
| | - Ivan Nemazanyy
- Université de Paris, Paris, France
- Plateforme Etude du métabolisme, Structure Fédérative de Recherche Necker INSERM US24-CNRS UMS 3633, Paris, France
| | - Fabricio Edgar de Moraes
- Laboratório Max Feffer de Genética de Plantas, Departamento de Genética, Escola Superior de Agricultura Luiz de Queiroz, Universidade de São Paulo, Piracicaba, Brazil
| | - Carlos Labate
- Laboratório Max Feffer de Genética de Plantas, Departamento de Genética, Escola Superior de Agricultura Luiz de Queiroz, Universidade de São Paulo, Piracicaba, Brazil
| | - Ida Chiara Guerrera
- Université de Paris, Paris, France
- Plateforme Protéome Institut Necker, PPN, Structure Fédérative de Recherche Necker INSERM US24-CNRS UMS 3633, Paris, France
- * E-mail: (ICG); (AC)
| | - Alain Charbit
- Université de Paris, Paris, France
- INSERM U1151 - CNRS UMR 8253, Institut Necker-Enfants Malades. Team 7: Pathogénie des Infections Systémiques, Paris, France
- * E-mail: (ICG); (AC)
| |
Collapse
|
22
|
Rotem S, Steinberger-Levy I, Israeli O, Zahavy E, Aloni-Grinstein R. Beating the Bio-Terror Threat with Rapid Antimicrobial Susceptibility Testing. Microorganisms 2021; 9:1535. [PMID: 34361970 PMCID: PMC8304332 DOI: 10.3390/microorganisms9071535] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 11/16/2022] Open
Abstract
A bioterror event using an infectious bacterium may lead to catastrophic outcomes involving morbidity and mortality as well as social and psychological stress. Moreover, a bioterror event using an antibiotic resistance engineered bacterial agent may raise additional concerns. Thus, preparedness is essential to preclude and control the dissemination of the bacterial agent as well as to appropriately and promptly treat potentially exposed individuals or patients. Rates of morbidity, death, and social anxiety can be drastically reduced if the rapid delivery of antimicrobial agents for post-exposure prophylaxis and treatment is initiated as soon as possible. Availability of rapid antibiotic susceptibility tests that may provide key recommendations to targeted antibiotic treatment is mandatory, yet, such tests are only at the development stage. In this review, we describe the recently published rapid antibiotic susceptibility tests implemented on bioterror bacterial agents and discuss their assimilation in clinical and environmental samples.
Collapse
Affiliation(s)
| | | | | | | | - Ronit Aloni-Grinstein
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 74100, Israel; (S.R.); (I.S.-L.); (O.I.); (E.Z.)
| |
Collapse
|
23
|
Abstract
Potassium is an essential mineral nutrient required by all living cells for normal physiological function. Therefore, maintaining intracellular potassium homeostasis during bacterial infection is a requirement for the survival of both host and pathogen. However, pathogenic bacteria require potassium transport to fulfill nutritional and chemiosmotic requirements, and potassium has been shown to directly modulate virulence gene expression, antimicrobial resistance, and biofilm formation. Host cells also require potassium to maintain fundamental biological processes, such as renal function, muscle contraction, and neuronal transmission; however, potassium flux also contributes to critical immunological and antimicrobial processes, such as cytokine production and inflammasome activation. Here, we review the role and regulation of potassium transport and signaling during infection in both mammalian and bacterial cells and highlight the importance of potassium to the success and survival of each organism.
Collapse
|
24
|
Maekawa S, Pulpipat T, Wang PC, Chen SC. Transcriptome analysis of immune- and iron-related genes after Francisella noatunensis subsp. orientalis infection in Nile tilapia (Oreochromis niloticus). FISH & SHELLFISH IMMUNOLOGY 2021; 111:36-48. [PMID: 33444737 DOI: 10.1016/j.fsi.2020.11.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 10/15/2020] [Accepted: 11/09/2020] [Indexed: 06/12/2023]
Abstract
Francisella noatunensis subsp. orientalis (Fno) is a gram-negative intracellular bacterium identified in many fish species worldwide, including cultured Nile tilapia (Oreochromis niloticus) in Taiwan. To investigate the gene expression responses to Fno infection, we performed transcriptome analysis of the head kidney and spleen in Nile tilapia using RNA-seq. Total RNA was extracted from the head kidney and spleen of infected (Fno-injected) and uninfected (control) tilapia at 1-day and 2-days post-infection, and RNA-seq was performed using the Illumina HiSeq™ 4000 platform. After de novo assembly, a total of 106,534 transcripts were detected. These transcripts were annotated and categorized into a total of 7171 genes based on the KEGG pathway database. Differentially expressed genes (DEGs) were significantly (2-fold difference comparing Fno and PBS groups at each time point) enriched in the immune-related pathways, including the following: complement and coagulation cascades, cytokine-cytokine receptor interaction, hematopoietic cell lineage, lysosome, phagosome. We identified the upregulation of inflammatory cytokine-, apoptosis-, and neutrophil-related genes, and downregulation of complement- and lymphocyte-related genes. Additionally, we found the induction of natural resistance-associated macrophage protein 1 (NRAMP1) and heme responsive gene-1 (HRG1). Anemia of inflammation, caused by intracellular iron storage in spleen after Fno infection, was also observed. This study provides natural disease control strategies against Fno infection in tilapia. It is suggested that intercellular iron storage is a host protection strategy.
Collapse
Affiliation(s)
- Shun Maekawa
- Department of Veterinary Medicine, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, Taiwan; International Degree Program of Ornamental Fish Technology and Aquatic Animal Health, International College, National Pingtung University of Science and Technology, Pingtung, Taiwan; General Research Service Centre, National Pingtung University of Science and Technology, Pingtung, Taiwan.
| | - Theeraporn Pulpipat
- Department of Veterinary Medicine, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Pei-Chi Wang
- Department of Veterinary Medicine, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Shih-Chu Chen
- Department of Veterinary Medicine, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, Taiwan; International Degree Program of Ornamental Fish Technology and Aquatic Animal Health, International College, National Pingtung University of Science and Technology, Pingtung, Taiwan.
| |
Collapse
|
25
|
Jiang J, Wang W, Sun F, Zhang Y, Liu Q, Yang D. Bacterial infection reinforces host metabolic flux from arginine to spermine for NLRP3 inflammasome evasion. Cell Rep 2021; 34:108832. [PMID: 33691113 DOI: 10.1016/j.celrep.2021.108832] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 01/11/2021] [Accepted: 02/16/2021] [Indexed: 11/27/2022] Open
Abstract
Hosts recognize cytosolic microbial infection via the nucleotide-binding domain-like receptor (NLR) protein family, triggering inflammasome complex assembly to provoke pyroptosis or cytokine-related caspase-1-dependent antimicrobial responses. Pathogens have evolved diverse strategies to antagonize inflammasome activation. Here, Edwardsiella piscicida gene-defined transposon library screening for lactate dehydrogenase (LDH) release in nlrc4-/- bone marrow-derived macrophages (BMDMs) demonstrates that genes clustered in the bacterial arginine metabolism pathway participate in NLRP3 inflammasome inhibition. Blocking arginine uptake or putrescine export significantly relieves NLRP3 inflammasome inhibition, indicating that this bacterium rewires its arginine metabolism network during infection. Moreover, intracellular E. piscicida recruits the host arginine importer (mCAT-1) and putrescine exporter (Oct-2) to bacterium-containing vacuoles, accompanied by reduced arginine and accumulated cytosolic spermine. Neutralizing E. piscicida-induced cytosolic spermine enhancement by spermine synthetase or extracellular spermine significantly alters NLRP3 inflammasome activation. Importantly, accumulated cytosolic spermine inhibits K+ efflux-dependent NLRP3 inflammasome activation. These data highlight the mechanism of bacterial gene-mediated arginine metabolism control for NLRP3 inflammasome evasion.
Collapse
Affiliation(s)
- Jiatiao Jiang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Wenwen Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Fei Sun
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Yuanxing Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai 519000, China
| | - Qin Liu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, China; Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai 200237, China
| | - Dahai Yang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China; Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai 200237, China.
| |
Collapse
|
26
|
Nakamura T, Shimizu T, Inagaki F, Okazaki S, Saha SS, Uda A, Watanabe K, Watarai M. Identification of Membrane-Bound Lytic Murein Transglycosylase A (MltA) as a Growth Factor for Francisella novicida in a Silkworm Infection Model. Front Cell Infect Microbiol 2021; 10:581864. [PMID: 33553001 PMCID: PMC7862118 DOI: 10.3389/fcimb.2020.581864] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 12/04/2020] [Indexed: 12/22/2022] Open
Abstract
Francisella tularensis, the causative agent of tularemia, is transmitted by arthropod vectors within mammalian hosts. The detailed mechanisms contributing to growth and survival of Francisella within arthropod remain poorly understood. To identify novel factors supporting growth and survival of Francisella within arthropods, a transposon mutant library of F. tularensis subsp. novicida (F. novicida) was screened using an F. novicida-silkworm infection model. Among 750 transposon mutants screened, the mltA-encoding membrane-bound lytic murein transglycosylase A (MltA) was identified as a novel growth factor of F. novicida in silkworms. Silkworms infection with an mltA deletion mutant (ΔmltA) resulted in a reduction in the number of bacteria and prolonged survival. The ΔmltA strain exhibited limited intracellular growth and cytotoxicity in BmN4 silkworm ovary cells. Moreover, the ΔmltA strain induced higher expression of the antimicrobial peptide in silkworms compared to the wild-type strain. These results suggest that F. novicida MltA contributes to the survival of F. novicida in silkworms via immune suppression-related mechanisms. Intracellular growth of the ΔmltA strain was also reduced in human monocyte THP-1 cells. These results also suggest the contribution of MltA to pathogenicity in humans and utility of the F. novicida-silkworm infection model to explore Francisella infection.
Collapse
Affiliation(s)
- Takemasa Nakamura
- Laboratory of Veterinary Public Health, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Takashi Shimizu
- Laboratory of Veterinary Public Health, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Fumiya Inagaki
- Laboratory of Veterinary Public Health, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Shoma Okazaki
- Laboratory of Veterinary Public Health, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Shib Shankar Saha
- Department of Pathology and Parasitology, Patuakhali Science and Technology University, Barisal, Bangladesh
| | - Akihiko Uda
- Department of Veterinary Science, National Institute of Infectious Diseases, Tokyo, Japan
| | - Kenta Watanabe
- Laboratory of Veterinary Public Health, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Masahisa Watarai
- Laboratory of Veterinary Public Health, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| |
Collapse
|
27
|
The Comparative Virulence of Francisella tularensis Subsp. mediasiatica for Vaccinated Laboratory Animals. Microorganisms 2020; 8:microorganisms8091403. [PMID: 32932593 PMCID: PMC7564995 DOI: 10.3390/microorganisms8091403] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/08/2020] [Accepted: 09/11/2020] [Indexed: 11/17/2022] Open
Abstract
Tularemia is a severe infectious disease caused by the Gram-negative bacteria Fracisella tularensis. There are four subspecies of F.tularensis: holarctica, tularensis, mediasiatica, and novicida, which differ in their virulence and geographic distribution. One of them, subsp. mediasiatica remains extremely poorly studied, primarily due to the fact that it is found only in the sparsely populated regions of Central Asia and Russia. In particular there is little information in the literature on the virulence and pathogenicity of subsp. mediasiatica. In the present article, we evaluated the comparative virulence of subsp. mediasiatica in vaccinated laboratory animals which we infected with virulent strains: subsp. mediasiatica 678, subsp. holarctica 503, and subsp. tularensis SCHU within 60 to 180 days after vaccination. We found that subsp. mediasiatica is comparable in pathogenicity in mice with subsp. tularensis and in guinea pigs with subsp. holarctica. We also found that the live vaccine does not fully protect mice from subsp. mediasiatica but completely protects guinea pigs for at least six months. In general, our data suggest that subsp. mediasiatica occupies an intermediate position in virulence between spp. tularensis and holarctica.
Collapse
|
28
|
Using proteomics to identify host cell interaction partners for VgrG and IglJ. Sci Rep 2020; 10:14612. [PMID: 32884055 PMCID: PMC7471685 DOI: 10.1038/s41598-020-71641-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 07/23/2020] [Indexed: 11/17/2022] Open
Abstract
Francisella tularensis is a highly virulent intracellular bacterium and the causative agent of tularemia. The disease is characterized by the suboptimal innate immune response and consequently by the impaired adaptive immunity. The virulence of this pathogen depends on proteins encoded by a genomic island termed the Francisella Pathogenicity Island (FPI). However, the precise biological roles of most of the FPI-encoded proteins remain to be clarified. In this study, we employed stable isotope labeling by amino acids in cell culture (SILAC) in combination with affinity protein purification coupled with liquid chromatography–mass spectrometry to identify potential protein-effector binding pairs for two FPI virulence effectors IglJ and VgrG. Our results may indicate that while the IglJ protein interactions primarily affect mitochondria, the VgrG interactions affect phagosome and/or autophagosome biogenesis via targeting components of the host’s exocyst complex.
Collapse
|
29
|
Parashar K, Carpino N. A role for the Sts phosphatases in negatively regulating IFNγ-mediated production of nitric oxide in monocytes. IMMUNITY INFLAMMATION AND DISEASE 2020; 8:523-533. [PMID: 32841534 PMCID: PMC7654413 DOI: 10.1002/iid3.336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 07/01/2020] [Accepted: 07/20/2020] [Indexed: 12/17/2022]
Abstract
Introduction The atypical Sts phosphatases negatively regulate signaling pathways in diverse immune cell types, with two of their molecular targets being the related kinases Syk and Zap‐70. Mice lacking Sts expression (Sts−/−) are resistant to infection by the live vaccine strain (LVS) of Francisella tularensis. Although the mechanisms underlying the enhanced resistance of Sts−/− mice have not been definitively established, Sts−/− bone marrow‐derived monocytes (BMMs) demonstrate greater clearance of intracellular LVS following ex vivo infection, relative to wild type cells. To determine how the Sts proteins regulate monocyte bactericidal properties, we analyzed responses of infected cells. Methods Monocyte bacterial clearance was assayed using ex vivo coculture infections followed by colony‐forming unit analysis of intracellular bacteria. Levels of gene expression were quantified by quantitative reverse‐transcription polymerase chain reaction, levels of Nos2 protein levels were quantified by Western blot analysis, and levels of nitric oxide (NO) were quantified directly using the Griess reagent. We characterized monocyte cytokine production via enzyme‐linked immunosorbent assay. Results We demonstrate that Sts−/− monocyte cultures produce elevated levels of interferon‐γ (IFNγ) after infection, relative to wild type cultures. Sts−/− monocytes also demonstrate heightened responsiveness to IFNγ. Specifically, Sts−/− monocytes produce elevated levels of antimicrobial NO following IFNγ stimulation, and this NO plays an important role in LVS restriction. Additional IFNγ‐stimulated genes, including Ip10 and members of the Gbp gene family, also display heightened upregulation in Sts−/− cells. Both Sts‐1 and Sts‐2 contribute to the regulation of NO production, as evidenced by the responses of monocytes lacking each phosphatase individually. Finally, we demonstrate that the elevated production of IFNγ‐induced NO in Sts−/− monocytes is abrogated following chemical inhibition of Syk kinase. Conclusion Our results indicate a novel role for the Sts enzymes in regulating monocyte antibacterial responses downstream of IFNγ.
Collapse
Affiliation(s)
- Kaustubh Parashar
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York
| | - Nicholas Carpino
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, New York
| |
Collapse
|
30
|
Metabolic Profiling of Volatile Organic Compounds (VOCs) Emitted by the Pathogens Francisella tularensis and Bacillus anthracis in Liquid Culture. Sci Rep 2020; 10:9333. [PMID: 32518249 PMCID: PMC7283342 DOI: 10.1038/s41598-020-66136-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 05/05/2020] [Indexed: 11/08/2022] Open
Abstract
We conducted comprehensive (untargeted) metabolic profiling of volatile organic compounds (VOCs) emitted in culture by bacterial taxa Francisella tularensis (F. tularensis) subspecies novicida and Bacillus anthracis (B. anthracis) Sterne, surrogates for potential bacterial bioterrorism agents, as well as selective measurements of VOCs from their fully virulent counterparts, F. tularensis subspecies tularensis strain SCHU S4 and B. anthracis Ames. F. tularensis and B. anthracis were grown in liquid broth for time periods that covered logarithmic growth, stationary, and decline phases. VOCs emitted over the course of the growth phases were collected from the headspace above the cultures using solid phase microextraction (SPME) and were analyzed using gas chromatography-mass spectrometry (GC-MS). We developed criteria for distinguishing VOCs originating from bacteria versus background VOCs (originating from growth media only controls or sampling devices). Analyses of collected VOCs revealed methyl ketones, alcohols, esters, carboxylic acids, and nitrogen- and sulfur-containing compounds that were present in the bacterial cultures and absent (or present at only low abundance) in control samples indicating that these compounds originated from the bacteria. Distinct VOC profiles where observed for F. tularensis when compared with B. anthracis while the observed profiles of each of the two F. tularensis and B. anthracis strains exhibited some similarities. Furthermore, the relative abundance of VOCs was influenced by bacterial growth phase. These data illustrate the potential for VOC profiles to distinguish pathogens at the genus and species-level and to discriminate bacterial growth phases. The determination of VOC profiles lays the groundwork for non-invasive probes of bacterial metabolism and offers prospects for detection of microbe-specific VOC biomarkers from two potential biowarfare agents.
Collapse
|
31
|
Obaidat MM, Malania L, Bani Salman AE, Arner RJ, Roess AA. Seroepidemiology, Spatial Distribution, and Risk Factors of Francisella tularensis in Jordan. Am J Trop Med Hyg 2020; 103:659-664. [PMID: 32524955 DOI: 10.4269/ajtmh.19-0335] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
There is a paucity of data on Francisella tularensis in the Middle East and North Africa. This is the first countrywide study to determine the seroprevalence, spatial distribution, and risk factors for F. tularensis in Jordan. A total of 828 Jordanians were serologically tested for F. tularensis by ELISA. These individuals filled out a self-administered questionnaire to collect demographic and risk factor information. Bivariate and multivariate logistic regressions were performed to determine which variables are associated with seropositivity. The overall seroprevalence of F. tularensis was 7.7% (95% CI: 6.10-9.75). The bivariate analyses showed that age, region of residence, small ruminant ownership, and practicing horticulture were significantly associated with seropositivity, and these variables were controlled for in the multivariate analysis. The multivariate analysis showed an increased odds of seropositivity among individuals living in northern desert, middle, and northern highland areas, compared with individuals living in the drier southern area, as 7.27 (95% CI: 2.49-21.19), 3.79 (95% CI: 1.53-9.39), and 3.52 (95% CI: 1.45-388.55), respectively. Individuals owning a small ruminant had 1.86 (95% CI: 1.02-3.40) greater odds for seropositivity than individuals who do not own a small ruminant. Individuals practicing horticulture had 2.10 (95% CI: 1.20-3.66) greater odds for seropositivity than individuals who do not practice horticulture. This is the first study to address the seroprevalence of F. tularensis in Jordan and the Middle East. Further research is needed to identify clinical cases of tularemia in Jordan and to determine the circulating F. tularensis subspecies.
Collapse
Affiliation(s)
- Mohammad M Obaidat
- Department of Veterinary Pathology and Public Health, Faculty of Veterinary Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Lile Malania
- National Center for Disease Control and Public Health, Tbilisi, Georgia
| | - Alaa E Bani Salman
- Department of Veterinary Pathology and Public Health, Faculty of Veterinary Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Ryan J Arner
- Ryan Arner Science Consulting, LLC, Freeport, Pennsylvania
| | - Amira A Roess
- Department of Global and Community Health, College of Health and Human Services, George Mason University, Fairfax, Virginia.,Department of Global Health, Milken Institute School of Public Health, George Washington University, Washington, District of Columbia
| |
Collapse
|
32
|
Freudenberger Catanzaro KC, Inzana TJ. The Francisella tularensis Polysaccharides: What Is the Real Capsule? Microbiol Mol Biol Rev 2020; 84:e00065-19. [PMID: 32051235 PMCID: PMC7018499 DOI: 10.1128/mmbr.00065-19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Francisella tularensis is a tier 1 select agent responsible for tularemia in humans and a wide variety of animal species. Extensive research into understanding the virulence factors of the bacterium has been ongoing to develop an efficacious vaccine. At least two such virulence factors are described as capsules of F. tularensis: the O-antigen capsule and the capsule-like complex (CLC). These two separate entities aid in avoiding host immune defenses but have not been clearly differentiated. These components are distinct and differ in composition and genetic basis. The O-antigen capsule consists of a polysaccharide nearly identical to the lipopolysaccharide (LPS) O antigen, whereas the CLC is a heterogeneous complex of glycoproteins, proteins, and possibly outer membrane vesicles and tubes (OMV/Ts). In this review, the current understanding of these two capsules is summarized, and the historical references to "capsules" of F. tularensis are clarified. A significant amount of research has been invested into the composition of each capsule and the genes involved in synthesis of the polysaccharide portion of each capsule. Areas of future research include further exploration into the molecular regulation and pathways responsible for expression of each capsule and further elucidating the role that each capsule plays in virulence.
Collapse
Affiliation(s)
- Kelly C Freudenberger Catanzaro
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
| | - Thomas J Inzana
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, USA
- Virginia Tech Carilion School of Medicine, Roanoke, Virginia, USA
- College of Veterinary Medicine, Long Island University, Brookville, New York, USA
| |
Collapse
|
33
|
Autotransporter-Mediated Display of Complement Receptor Ligands by Gram-Negative Bacteria Increases Antibody Responses and Limits Disease Severity. Pathogens 2020; 9:pathogens9050375. [PMID: 32422907 PMCID: PMC7281241 DOI: 10.3390/pathogens9050375] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/06/2020] [Accepted: 05/11/2020] [Indexed: 12/13/2022] Open
Abstract
The targeting of immunogens/vaccines to specific immune cells is a promising approach for amplifying immune responses in the absence of exogenous adjuvants. However, the targeting approaches reported thus far require novel, labor-intensive reagents for each vaccine and have primarily been shown as proof-of-concept with isolated proteins and/or inactivated bacteria. We have engineered a plasmid-based, complement receptor-targeting platform that is readily applicable to live forms of multiple gram-negative bacteria, including, but not limited to, Escherichia coli, Klebsiella pneumoniae, and Francisella tularensis. Using F. tularensis as a model, we find that targeted bacteria show increased binding and uptake by macrophages, which coincides with increased p38 and p65 phosphorylation. Mice vaccinated with targeted bacteria produce higher titers of specific antibody that recognizes a greater diversity of bacterial antigens. Following challenge with homologous or heterologous isolates, these mice exhibited less weight loss and/or accelerated weight recovery as compared to counterparts vaccinated with non-targeted immunogens. Collectively, these findings provide proof-of-concept for plasmid-based, complement receptor-targeting of live gram-negative bacteria.
Collapse
|
34
|
Nakamura T, Shimizu T, Uda A, Watanabe K, Watarai M. Soluble lytic transglycosylase SLT of Francisella novicida is involved in intracellular growth and immune suppression. PLoS One 2019; 14:e0226778. [PMID: 31877174 PMCID: PMC6932806 DOI: 10.1371/journal.pone.0226778] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 12/03/2019] [Indexed: 12/12/2022] Open
Abstract
Francisella tularensis, a category-A bioterrorism agent causes tularemia. F. tularensis suppresses the immune response of host cells and intracellularly proliferates. However, the detailed mechanisms of immune suppression and intracellular growth are largely unknown. Here we developed a transposon mutant library to identify novel pathogenic factors of F. tularensis. Among 750 transposon mutants of F. tularensis subsp. novicida (F. novicida), 11 were isolated as less cytotoxic strains, and the genes responsible for cytotoxicity were identified. Among them, the function of slt, which encodes soluble lytic transglycosylase (SLT) was investigated in detail. An slt deletion mutant (Δslt) was less toxic to the human monocyte cell line THP-1 vs the wild-type strain. Although the wild-type strain proliferated in THP-1 cells, the number of intracellular Δslt mutant decreased in comparison. The Δslt mutant escaped from phagosomes during the early stages of infection, but the mutant was detected within the autophagosome, followed by degradation in lysosomes. Moreover, the Δslt mutant induced host cells to produce high levels of cytokines such as tumor necrosis factor-α, interleukin (IL)-6, and IL-1β, compared with the wild-type strain. These results suggest that the SLT of F. novicida is required for immune suppression and escape from autophagy to allow its survival in host cells.
Collapse
Affiliation(s)
- Takemasa Nakamura
- Joint Faculty of Veterinary Medicine, Laboratory of Veterinary Public Health, Yamaguchi University, Yamaguchi, Japan
| | - Takashi Shimizu
- Joint Faculty of Veterinary Medicine, Laboratory of Veterinary Public Health, Yamaguchi University, Yamaguchi, Japan
| | - Akihiko Uda
- Department of Veterinary Science, National Institute of Infectious Diseases, Shinjuku, Tokyo, Japan
| | - Kenta Watanabe
- Joint Faculty of Veterinary Medicine, Laboratory of Veterinary Public Health, Yamaguchi University, Yamaguchi, Japan
| | - Masahisa Watarai
- Joint Faculty of Veterinary Medicine, Laboratory of Veterinary Public Health, Yamaguchi University, Yamaguchi, Japan
- * E-mail:
| |
Collapse
|
35
|
Ziveri J, Chhuon C, Jamet A, Rytter H, Prigent G, Tros F, Barel M, Coureuil M, Lays C, Henry T, Keep NH, Guerrera IC, Charbit A. Critical Role of a Sheath Phosphorylation Site On the Assembly and Function of an Atypical Type VI Secretion System. Mol Cell Proteomics 2019; 18:2418-2432. [PMID: 31578219 PMCID: PMC6885697 DOI: 10.1074/mcp.ra119.001532] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 09/03/2019] [Indexed: 12/12/2022] Open
Abstract
The bacterial pathogen Francisella tularensis possesses a noncanonical type VI secretion system (T6SS) that is required for phagosomal escape in infected macrophages. KCl stimulation has been previously used to trigger assembly and secretion of the T6SS in culture. By differential proteomics, we found here that the amounts of the T6SS proteins remained unchanged upon KCl stimulation, suggesting involvement of post-translational modifications in T6SS assembly. A phosphoproteomic analysis indeed identified a unique phosphorylation site on IglB, a key component of the T6SS sheath. Substitutions of Y139 with alanine or phosphomimetics prevented T6SS formation and abolished phagosomal escape whereas substitution with phenylalanine delayed but did not abolish phagosomal escape in J774-1 macrophages. Altogether our data demonstrated that the Y139 site of IglB plays a critical role in T6SS biogenesis, suggesting that sheath phosphorylation could participate to T6SS dynamics.Data are available via ProteomeXchange with identifier PXD013619; and on MS-Viewer, key lkaqkllxwx.
Collapse
Affiliation(s)
- Jason Ziveri
- Université Paris Descartes, Sorbonne Paris Cité, INSERM U1151-CNRS UMR 8253, Institut Necker-Enfants Malades. Team 7: Pathogenesis of Systemic Infections, Paris 75015, France; Plateforme protéomique 3P5-Necker, Universit[c33c]zpi;● Paris Descartes - Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS3633, Paris 75015, France
| | - Cerina Chhuon
- Plateforme protéomique 3P5-Necker, Universit[c33c]zpi;● Paris Descartes - Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS3633, Paris 75015, France; Plateforme protéomique 3P5-Necker, Universit[c33c]zpi;● Paris Descartes - Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS3633, Paris 75015, France
| | - Anne Jamet
- Université Paris Descartes, Sorbonne Paris Cité, INSERM U1151-CNRS UMR 8253, Institut Necker-Enfants Malades. Team 7: Pathogenesis of Systemic Infections, Paris 75015, France; Plateforme protéomique 3P5-Necker, Universit[c33c]zpi;● Paris Descartes - Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS3633, Paris 75015, France
| | - Héloïse Rytter
- Université Paris Descartes, Sorbonne Paris Cité, INSERM U1151-CNRS UMR 8253, Institut Necker-Enfants Malades. Team 7: Pathogenesis of Systemic Infections, Paris 75015, France; Plateforme protéomique 3P5-Necker, Universit[c33c]zpi;● Paris Descartes - Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS3633, Paris 75015, France
| | - Guénolé Prigent
- Université Paris Descartes, Sorbonne Paris Cité, INSERM U1151-CNRS UMR 8253, Institut Necker-Enfants Malades. Team 7: Pathogenesis of Systemic Infections, Paris 75015, France; Plateforme protéomique 3P5-Necker, Universit[c33c]zpi;● Paris Descartes - Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS3633, Paris 75015, France
| | - Fabiola Tros
- Université Paris Descartes, Sorbonne Paris Cité, INSERM U1151-CNRS UMR 8253, Institut Necker-Enfants Malades. Team 7: Pathogenesis of Systemic Infections, Paris 75015, France; Plateforme protéomique 3P5-Necker, Universit[c33c]zpi;● Paris Descartes - Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS3633, Paris 75015, France
| | - Monique Barel
- Université Paris Descartes, Sorbonne Paris Cité, INSERM U1151-CNRS UMR 8253, Institut Necker-Enfants Malades. Team 7: Pathogenesis of Systemic Infections, Paris 75015, France; Plateforme protéomique 3P5-Necker, Universit[c33c]zpi;● Paris Descartes - Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS3633, Paris 75015, France
| | - Mathieu Coureuil
- Université Paris Descartes, Sorbonne Paris Cité, INSERM U1151-CNRS UMR 8253, Institut Necker-Enfants Malades. Team 7: Pathogenesis of Systemic Infections, Paris 75015, France; Plateforme protéomique 3P5-Necker, Universit[c33c]zpi;● Paris Descartes - Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS3633, Paris 75015, France
| | - Claire Lays
- CIRI, Centre International de Recherche en Infectiologie, Université Lyon, Inserm, U1111, University Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Labex ECOFECT, Eco-evolutionary dynamics of infectious diseases, F-69007, LYON, France
| | - Thomas Henry
- CIRI, Centre International de Recherche en Infectiologie, Université Lyon, Inserm, U1111, University Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Labex ECOFECT, Eco-evolutionary dynamics of infectious diseases, F-69007, LYON, France
| | - Nicholas H Keep
- Crystallography, Institute for Structural and Molecular Biology, Department of Biological Sciences Birkbeck, University of London, United Kingdom
| | - Ida Chiara Guerrera
- Plateforme protéomique 3P5-Necker, Universit[c33c]zpi;● Paris Descartes - Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS3633, Paris 75015, France; Plateforme protéomique 3P5-Necker, Universit[c33c]zpi;● Paris Descartes - Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS3633, Paris 75015, France.
| | - Alain Charbit
- Université Paris Descartes, Sorbonne Paris Cité, INSERM U1151-CNRS UMR 8253, Institut Necker-Enfants Malades. Team 7: Pathogenesis of Systemic Infections, Paris 75015, France; Plateforme protéomique 3P5-Necker, Universit[c33c]zpi;● Paris Descartes - Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMS3633, Paris 75015, France.
| |
Collapse
|
36
|
Champion AE, Catanzaro KCF, Bandara AB, Inzana TJ. Formation of the Francisella tularensis Biofilm is Affected by Cell Surface Glycosylation, Growth Medium, and a Glucan Exopolysaccharide. Sci Rep 2019; 9:12252. [PMID: 31439876 PMCID: PMC6706388 DOI: 10.1038/s41598-019-48697-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 07/22/2019] [Indexed: 01/05/2023] Open
Abstract
Biofilms are matrix-associated communities that enable bacteria to colonise environments unsuitable for free-living bacteria. The facultative intracellular pathogen Francisella tularensis can persist in water, amoebae, and arthropods, as well as within mammalian macrophages. F. tularensis Types A and B form poor biofilms, but F. tularensis mutants lacking lipopolysaccharide O-antigen, O-antigen capsule, and capsule-like complex formed up to 15-fold more biofilm than fully glycosylated cells. The Type B live vaccine strain was also 50% less capable of initiating surface attachment than mutants deficient in O-antigen and capsule-like complex. However, the growth medium of all strains tested also influenced the formation of biofilm, which contained a novel exopolysaccharide consisting of an amylose-like glucan. In addition, the surface polysaccharide composition of the bacterium affected the protein:DNA:polysaccharide composition of the biofilm matrix. In contrast, F. novicida attached to surfaces more efficiently and made a more robust biofilm than Type A or B strains, but loss of O-antigen or capsule-like complex did not significantly affect F. novicida biofilm formation. These results indicated that suppression of surface polysaccharides may promote biofilm formation by F. tularensis Types A and B. Whether biofilm formation enhances survival of F. tularensis in aquatic or other environmental niches has yet to be determined.
Collapse
Affiliation(s)
- Anna E Champion
- Center for Molecular Medicine and Infectious Diseases, Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Kelly C Freudenberger Catanzaro
- Center for Molecular Medicine and Infectious Diseases, Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Aloka B Bandara
- Center for Molecular Medicine and Infectious Diseases, Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Thomas J Inzana
- Center for Molecular Medicine and Infectious Diseases, Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, 24061, USA.
- Virginia Tech Carilion School of Medicine, Roanoke, VA, 24016, USA.
- Long Island University, College of Veterinary Medicine, 216 Roth Hall, Brookville, NY, 11548, USA.
| |
Collapse
|
37
|
Bachert BA, Biryukov SS, Chua J, Rodriguez SA, Toothman RG, Cote CK, Klimko CP, Hunter M, Shoe JL, Williams JA, Kuehl KA, Biot FV, Bozue JA. A Francisella novicida Mutant, Lacking the Soluble Lytic Transglycosylase Slt, Exhibits Defects in Both Growth and Virulence. Front Microbiol 2019; 10:1343. [PMID: 31258523 PMCID: PMC6587636 DOI: 10.3389/fmicb.2019.01343] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 05/29/2019] [Indexed: 01/28/2023] Open
Abstract
Francisella tularensis is the causative agent of tularemia and has gained recent interest as it poses a significant biothreat risk. F. novicida is commonly used as a laboratory surrogate for tularemia research due to genetic similarity and susceptibility of mice to infection. Currently, there is no FDA-approved tularemia vaccine, and identifying therapeutic targets remains a critical gap in strategies for combating this pathogen. Here, we investigate the soluble lytic transglycosylase or Slt in F. novicida, which belongs to a class of peptidoglycan-modifying enzymes known to be involved in cell division. We assess the role of Slt in biology and virulence of the organism as well as the vaccine potential of the slt mutant. We show that the F. novicida slt mutant has a significant growth defect in acidic pH conditions. Further microscopic analysis revealed significantly altered cell morphology compared to wild-type, including larger cell size, extensive membrane protrusions, and cell clumping and fusion, which was partially restored by growth in neutral pH or genetic complementation. Viability of the mutant was also significantly decreased during growth in acidic medium, but not at neutral pH. Furthermore, the slt mutant exhibited significant attenuation in a murine model of intranasal infection and virulence could be restored by genetic complementation. Moreover, we could protect mice using the slt mutant as a live vaccine strain against challenge with the parent strain; however, we were not able to protect against challenge with the fully virulent F. tularensis Schu S4 strain. These studies demonstrate a critical role for the Slt enzyme in maintaining proper cell division and morphology in acidic conditions, as well as replication and virulence in vivo. Our results suggest that although the current vaccination strategy with F. novicida slt mutant would not protect against Schu S4 challenges, the Slt enzyme could be an ideal target for future therapeutic development.
Collapse
Affiliation(s)
- Beth A Bachert
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, United States
| | - Sergei S Biryukov
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, United States
| | - Jennifer Chua
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, United States
| | - Sabrina A Rodriguez
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, United States
| | - Ronald G Toothman
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, United States
| | - Christopher K Cote
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, United States
| | - Christopher P Klimko
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, United States
| | - Melissa Hunter
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, United States
| | - Jennifer L Shoe
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, United States
| | - Janice A Williams
- Pathology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, United States
| | - Kathleen A Kuehl
- Pathology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, United States
| | - Fabrice V Biot
- Unité de Bactériologie/UMR_MD1, Département de Biologie des Agents Transmissibles, Institut de Recherche Biomédicale des Armées, Brétigny-sur-Orge, France
| | - Joel A Bozue
- Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, MD, United States
| |
Collapse
|
38
|
Role of peroxiredoxin of the AhpC/TSA family in antioxidant defense mechanisms of Francisella tularensis. PLoS One 2019; 14:e0213699. [PMID: 30870480 PMCID: PMC6417708 DOI: 10.1371/journal.pone.0213699] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 02/23/2019] [Indexed: 12/16/2022] Open
Abstract
Francisella tularensis is a Gram-negative, facultative intracellular pathogen and the causative agent of a lethal human disease known as tularemia. Due to its extremely high virulence and potential to be used as a bioterror agent, F. tularensis is classified by the CDC as a Category A Select Agent. As an intracellular pathogen, F. tularensis during its intracellular residence encounters a number of oxidative and nitrosative stresses. The roles of the primary antioxidant enzymes SodB, SodC and KatG in oxidative stress resistance and virulence of F. tularensis live vaccine strain (LVS) have been characterized in previous studies. However, very fragmentary information is available regarding the role of peroxiredoxin of the AhpC/TSA family (annotated as AhpC) of F. tularensis SchuS4; whereas the role of AhpC of F. tularensis LVS in tularemia pathogenesis is not known. This study was undertaken to exhaustively investigate the role of AhpC in oxidative stress resistance of F. tularensis LVS and SchuS4. We report that AhpC of F. tularensis LVS confers resistance against a wide range of reactive oxygen and nitrogen species, and serves as a virulence factor. In highly virulent F. tularensis SchuS4 strain, AhpC serves as a key antioxidant enzyme and contributes to its robust oxidative and nitrosative stress resistance, and intramacrophage survival. We also demonstrate that there is functional redundancy among primary antioxidant enzymes AhpC, SodC, and KatG of F. tularensis SchuS4. Collectively, this study highlights the differences in antioxidant defense mechanisms of F. tularensis LVS and SchuS4.
Collapse
|
39
|
Glycoconjugate vaccine using a genetically modified O antigen induces protective antibodies to Francisella tularensis. Proc Natl Acad Sci U S A 2019; 116:7062-7070. [PMID: 30872471 PMCID: PMC6452683 DOI: 10.1073/pnas.1900144116] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Francisella tularensis is the causative agent of tularemia, a category A bioterrorism agent. The lipopolysaccharide (LPS) O antigen (OAg) of F. tularensis has been considered for use in a glycoconjugate vaccine, but conjugate vaccines tested so far have failed to confer protection necessary against aerosolized pulmonary bacterial challenge. When F. tularensis OAg was purified under standard conditions, the antigen had a small molecular size [25 kDa, low molecular weight (LMW)]. Using milder extraction conditions, we found the native OAg had a larger molecular size [80 kDa, high molecular weight (HMW)], and in a mouse model of tularemia, a glycoconjugate vaccine made with the HMW polysaccharide coupled to tetanus toxoid (HMW-TT) conferred better protection against intranasal challenge than a conjugate made with the LMW polysaccharide (LMW-TT). To further investigate the role of OAg size in protection, we created an F. tularensis live vaccine strain (LVS) mutant with a significantly increased OAg size [220 kDa, very high molecular weight (VHMW)] by expressing in F. tularensis a heterologous chain-length regulator gene (wzz) from the related species Francisella novicida Immunization with VHMW-TT provided markedly increased protection over that obtained with TT glycoconjugates made using smaller OAgs. We found that protective antibodies recognize a length-dependent epitope better expressed on HMW and VHMW antigens, which bind with higher affinity to the organism.
Collapse
|
40
|
Zhou W, Yin Y, Smith E, Chou J, Shumate J, Scampavia L, Spicer TP, Carpino N, French JB. Discovery and Characterization of Two Classes of Selective Inhibitors of the Suppressor of the TCR Signaling Family of Proteins. ACS Infect Dis 2019; 5:250-259. [PMID: 30485744 DOI: 10.1021/acsinfecdis.8b00238] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The suppressor of T-cell receptor signaling (Sts) proteins, Sts-1, has recently emerged as a potential immunostimulatory target for drug development. Genetic inactivation of the Sts proteins dramatically increases host survival of systemic infection and leads to improved pathogen clearance. The protein tyrosine phosphatase (PTP) activity of these proteins arises from a C-terminal 2-histidine phosphatase (HP) domain. To identify new inhibitors of the HP activity of Sts-1, we miniaturized a phosphatase assay to a 1536-well format and conducted a 20 580 compound screen. Among the hits were two classes of structurally related compounds, tetracycline variants and sulfonated azo dyes. These hits had low micromolar to nanomolar IC50 values. Orthogonal screening confirmed the validity of these inhibitors and demonstrated that both act competitively on Sts-1 phosphatase activity. When tested on other PTPs, PTP1B and SHP1, it was found that the tetracycline PTP1B, SHP1, the tetracycline variant (doxycycline), and the sulfonated azo dye (Congo red) are selective inhibitors of Sts-1HP, with selectivity indices ranging from 19 to as high as 200. The planar polyaromatic moieties present in both classes of compounds suggested a common binding mode. The mutation of either tryptophan 494 or tyrosine 596, located near the active site of the protein, reduced the Ki of the inhibitors from 3- to 18-fold, indicating that these residues may help to promote the binding of substrates with aromatic groups. This work provides new insights into substrate selectivity mechanisms and describes two classes of compounds that can serve as probes of function or as a basis for future drug discovery.
Collapse
Affiliation(s)
| | | | - Emery Smith
- Department of Molecular Medicine, Scripps Research Molecular Screening Center, Scripps Research, 130 Scripps Way, Jupiter, Florida 33458, United States
| | | | - Justin Shumate
- Department of Molecular Medicine, Scripps Research Molecular Screening Center, Scripps Research, 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Louis Scampavia
- Department of Molecular Medicine, Scripps Research Molecular Screening Center, Scripps Research, 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Timothy P. Spicer
- Department of Molecular Medicine, Scripps Research Molecular Screening Center, Scripps Research, 130 Scripps Way, Jupiter, Florida 33458, United States
| | | | | |
Collapse
|
41
|
Raasch M, Fritsche E, Kurtz A, Bauer M, Mosig AS. Microphysiological systems meet hiPSC technology - New tools for disease modeling of liver infections in basic research and drug development. Adv Drug Deliv Rev 2019; 140:51-67. [PMID: 29908880 DOI: 10.1016/j.addr.2018.06.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 06/01/2018] [Accepted: 06/12/2018] [Indexed: 02/08/2023]
Abstract
Complex cell culture models such as microphysiological models (MPS) mimicking human liver functionality in vitro are in the spotlight as alternative to conventional cell culture and animal models. Promising techniques like microfluidic cell culture or micropatterning by 3D bioprinting are gaining increasing importance for the development of MPS to address the needs for more predictivity and cost efficiency. In this context, human induced pluripotent stem cells (hiPSCs) offer new perspectives for the development of advanced liver-on-chip systems by recreating an in vivo like microenvironment that supports the reliable differentiation of hiPSCs to hepatocyte-like cells (HLC). In this review we will summarize current protocols of HLC generation and highlight recently established MPS suitable to resemble physiological hepatocyte function in vitro. In addition, we are discussing potential applications of liver MPS for disease modeling related to systemic or direct liver infections and the use of MPS in testing of new drug candidates.
Collapse
|
42
|
O’Malley KJ, Bowling JL, Stinson E, Cole KS, Mann BJ, Namjoshi P, Hazlett KRO, Barry EM, Reed DS. Aerosol prime-boost vaccination provides strong protection in outbred rabbits against virulent type A Francisella tularensis. PLoS One 2018; 13:e0205928. [PMID: 30346998 PMCID: PMC6197691 DOI: 10.1371/journal.pone.0205928] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 10/03/2018] [Indexed: 12/15/2022] Open
Abstract
Tularemia, also known as rabbit fever, is a severe zoonotic disease in humans caused by the gram-negative bacterium Francisella tularensis (Ft). While there have been a number of attempts to develop a vaccine for Ft, few candidates have advanced beyond experiments in inbred mice. We report here that a prime-boost strategy with aerosol delivery of recombinant live attenuated candidate Ft S4ΔaroD offers significant protection (83% survival) in an outbred animal model, New Zealand White rabbits, against aerosol challenge with 248 cfu (11 LD50) of virulent type A Ft SCHU S4. Surviving rabbits given two doses of the attenuated strains by aerosol did not exhibit substantial post-challenge fevers, changes in erythrocyte sedimentation rate or in complete blood counts. At a higher challenge dose (3,186 cfu; 139 LD50), protection was still good with 66% of S4ΔaroD-vaccinated rabbits surviving while 50% of S4ΔguaBA vaccinated rabbits also survived challenge. Pre-challenge plasma IgG titers against Ft SCHU S4 corresponded with survival time after challenge. Western blot analysis found that plasma antibody shifted from predominantly targeting Ft O-antigen after the prime vaccination to other antigens after the boost. These results demonstrate the superior protection conferred by a live attenuated derivative of virulent F. tularensis, particularly when given in an aerosol prime-boost regimen.
Collapse
Affiliation(s)
- Katherine J. O’Malley
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Jennifer L. Bowling
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Elizabeth Stinson
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Kelly S. Cole
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Barbara J. Mann
- Department of Medicine, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, United States of America
| | - Prachi Namjoshi
- Department for Immunology & Microbial Diseases, Albany Medical College, Albany, NY, United States of America
| | - Karsten R. O. Hazlett
- Department for Immunology & Microbial Diseases, Albany Medical College, Albany, NY, United States of America
| | - Eileen M. Barry
- Center for Vaccine Development, University of Maryland Baltimore, Baltimore, MD, United States of America
| | - Douglas S. Reed
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, United States of America
- * E-mail:
| |
Collapse
|
43
|
Trottein F, Paget C. Natural Killer T Cells and Mucosal-Associated Invariant T Cells in Lung Infections. Front Immunol 2018; 9:1750. [PMID: 30116242 PMCID: PMC6082944 DOI: 10.3389/fimmu.2018.01750] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 07/16/2018] [Indexed: 12/13/2022] Open
Abstract
The immune system has been traditionally divided into two arms called innate and adaptive immunity. Typically, innate immunity refers to rapid defense mechanisms that set in motion within minutes to hours following an insult. Conversely, the adaptive immune response emerges after several days and relies on the innate immune response for its initiation and subsequent outcome. However, the recent discovery of immune cells displaying merged properties indicates that this distinction is not mutually exclusive. These populations that span the innate-adaptive border of immunity comprise, among others, CD1d-restricted natural killer T cells and MR1-restricted mucosal-associated invariant T cells. These cells have the unique ability to swiftly activate in response to non-peptidic antigens through their T cell receptor and/or to activating cytokines in order to modulate many aspects of the immune response. Despite they recirculate all through the body via the bloodstream, these cells mainly establish residency at barrier sites including lungs. Here, we discuss the current knowledge into the biology of these cells during lung (viral and bacterial) infections including activation mechanisms and functions. We also discuss future strategies targeting these cell types to optimize immune responses against respiratory pathogens.
Collapse
Affiliation(s)
- François Trottein
- Univ. Lille, U1019 – UMR 8204 – CIIL – Centre d’Infection et d’Immunité de Lille, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale U1019, Lille, France
- Centre Hospitalier Universitaire de Lille, Lille, France
- Institut Pasteur de Lille, Lille, France
| | - Christophe Paget
- Institut National de la Santé et de la Recherche Médicale U1100, Centre d’Etude des Pathologies Respiratoires (CEPR), Tours, France
- Université de Tours, Tours, France
| |
Collapse
|
44
|
Rastawicki W, Formińska K, Zasada AA. Development and Evaluation of a Latex Agglutination Test for the Identification of Francisella tularensis Subspecies Pathogenic for Human. Pol J Microbiol 2018; 67:241-244. [PMID: 30015465 PMCID: PMC7256844 DOI: 10.21307/pjm-2018-030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/28/2018] [Indexed: 11/11/2022] Open
Abstract
Francisella tularensis are highly infectious bacteria causing a zoonotic disease called tularemia. Identification of this bacterium is based on antigen detection or PCR. The paper presents a latex agglutination test (LAT) for rapid identification of clinically relevant F. tularensis subspecies. The test can be performed within three minutes with live or inactivated bacteria. The possibility to test the inactivated samples reduces the risk of laboratory acquired infection and allows performing the test under BSL-2 conditions.
Collapse
Affiliation(s)
- Waldemar Rastawicki
- Department of Bacteriology, National Institute of Public Health - National Institute of Hygiene,Warsaw,Poland
| | - Kamila Formińska
- Department of Sera and Vaccines Evaluation, National Institute of Public Health - National Institute of Hygiene,Warsaw,Poland
| | - Aleksandra A Zasada
- Department of Sera and Vaccines Evaluation, National Institute of Public Health - National Institute of Hygiene,Warsaw,Poland
| |
Collapse
|
45
|
Nualnoi T, Kirosingh A, Basallo K, Hau D, Gates-Hollingsworth MA, Thorkildson P, Crump RB, Reed DE, Pandit S, AuCoin DP. Immunoglobulin G subclass switching impacts sensitivity of an immunoassay targeting Francisella tularensis lipopolysaccharide. PLoS One 2018; 13:e0195308. [PMID: 29630613 PMCID: PMC5890998 DOI: 10.1371/journal.pone.0195308] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 03/20/2018] [Indexed: 01/15/2023] Open
Abstract
The CDC Tier 1 select agent Francisella tularensis is a small, Gram-negative bacterium and the causative agent of tularemia, a potentially life-threatening infection endemic in the United States, Europe and Asia. Currently, there is no licensed vaccine or rapid point-of-care diagnostic test for tularemia. The purpose of this research was to develop monoclonal antibodies (mAbs) specific to the F. tularensis surface-expressed lipopolysaccharide (LPS) for a potential use in a rapid diagnostic test. Our initial antigen capture ELISA was developed using murine IgG3 mAb 1A4. Due to the low sensitivity of the initial assay, IgG subclass switching, which is known to have an effect on the functional affinity of a mAb, was exploited for the purpose of enhancing assay sensitivity. The ELISA developed using the IgG1 or IgG2b mAbs from the subclass-switch family of 1A4 IgG3 yielded improved assay sensitivity. However, surface plasmon resonance (SPR) demonstrated that the functional affinity was decreased as a result of subclass switching. Further investigation using direct ELISA revealed the potential self-association of 1A4 IgG3, which could explain the higher functional affinity and higher assay background seen with this mAb. Additionally, the higher assay background was found to negatively affect assay sensitivity. Thus, enhancement of the assay sensitivity by subclass switching is likely due to the decrease in assay background, simply by avoiding the self-association of IgG3.
Collapse
Affiliation(s)
- Teerapat Nualnoi
- Department of Microbiology and Immunology, University of Nevada, Reno School of Medicine, Reno, Nevada, United States of America
| | - Adam Kirosingh
- Department of Microbiology and Immunology, University of Nevada, Reno School of Medicine, Reno, Nevada, United States of America
| | - Kaitlin Basallo
- Department of Microbiology and Immunology, University of Nevada, Reno School of Medicine, Reno, Nevada, United States of America
| | - Derrick Hau
- Department of Microbiology and Immunology, University of Nevada, Reno School of Medicine, Reno, Nevada, United States of America
| | | | - Peter Thorkildson
- Department of Microbiology and Immunology, University of Nevada, Reno School of Medicine, Reno, Nevada, United States of America
| | - Reva B. Crump
- Department of Microbiology and Immunology, University of Nevada, Reno School of Medicine, Reno, Nevada, United States of America
| | - Dana E. Reed
- Department of Microbiology and Immunology, University of Nevada, Reno School of Medicine, Reno, Nevada, United States of America
| | - Sujata Pandit
- Department of Microbiology and Immunology, University of Nevada, Reno School of Medicine, Reno, Nevada, United States of America
| | - David P. AuCoin
- Department of Microbiology and Immunology, University of Nevada, Reno School of Medicine, Reno, Nevada, United States of America
- * E-mail:
| |
Collapse
|
46
|
Simultaneous Immunodetection of Anthrax, Plague, and Tularemia from Blood Cultures by Use of Multiplexed Suspension Arrays. J Clin Microbiol 2018; 56:JCM.01479-17. [PMID: 29386263 DOI: 10.1128/jcm.01479-17] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 01/11/2018] [Indexed: 01/10/2023] Open
Abstract
Multiplexed detection technologies are becoming increasingly important given the possibility of bioterrorism attacks, for which the range of suspected pathogens can vary considerably. In this work, we describe the use of Luminex MagPlex magnetic microspheres for the construction of two multiplexed diagnostic suspension arrays, enabling antibody-based detection of bacterial pathogens and their related disease biomarkers directly from blood cultures. The first 4-plex diagnostic array enabled the detection of both anthrax and plague infections using soluble disease biomarkers, including protective antigen (PA) and anthrax capsular antigen for anthrax detection and the capsular F1 and LcrV antigens for plague detection. The limits of detection (LODs) ranged between 0.5 and 5 ng/ml for the different antigens. The second 2-plex diagnostic array facilitated the detection of Yersinia pestis (LOD of 1 × 106 CFU/ml) and Francisella tularensis (LOD of 1 × 104 CFU/ml) from blood cultures. Inoculated, propagated blood cultures were processed (15 to 20 min) via 2 possible methodologies (Vacutainer or a simple centrifugation step), allowing the direct detection of bacteria in each sample, and the entire assay could be performed in 90 min. While detection of bacteria and soluble markers from blood cultures using PCR Luminex suspension arrays has been widely described, to our knowledge, this study is the first to demonstrate the utility of the Luminex system for the immunodetection of both bacteria and soluble markers directly from blood cultures. Targeting both the bacterial pathogens as well as two different disease biomarkers for each infection, we demonstrated the benefit of the multiplexed developed assays for enhanced, reliable detection. The presented arrays could easily be expanded to include antibodies for the detection of other pathogens of interest in hospitals or labs, demonstrating the applicability of this technology for the accurate detection and confirmation of a wide range of potential select agents.
Collapse
|
47
|
|
48
|
Kinkead LC, Whitmore LC, McCracken JM, Fletcher JR, Ketelsen BB, Kaufman JW, Jones BD, Weiss DS, Barker JH, Allen LAH. Bacterial lipoproteins and other factors released by Francisella tularensis modulate human neutrophil lifespan: Effects of a TLR1 SNP on apoptosis inhibition. Cell Microbiol 2017; 20. [PMID: 29063667 PMCID: PMC5764820 DOI: 10.1111/cmi.12795] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 10/10/2017] [Accepted: 10/15/2017] [Indexed: 12/21/2022]
Abstract
Francisella tularensis infects several cell types including neutrophils, and aberrant neutrophil accumulation contributes to tissue destruction during tularaemia. We demonstrated previously that F. tularensis strains Schu S4 and live vaccine strain markedly delay human neutrophil apoptosis and thereby prolong cell lifespan, but the bacterial factors that mediate this aspect of virulence are undefined. Herein, we demonstrate that bacterial conditioned medium (CM) can delay apoptosis in the absence of direct infection. Biochemical analyses show that CM contained F. tularensis surface factors as well as outer membrane components. Our previous studies excluded roles for lipopolysaccharide and capsule in apoptosis inhibition, and current studies of [14C] acetate‐labelled bacteria argue against a role for other bacterial lipids in this process. At the same time, studies of isogenic mutants indicate that TolC and virulence factors whose expression requires FevR or MglA were also dispensable, demonstrating that apoptosis inhibition does not require Type I or Type VI secretion. Instead, we identified bacterial lipoproteins (BLPs) as active factors in CM. Additional studies of isolated BLPs demonstrated dose‐dependent neutrophil apoptosis inhibition via a TLR2‐dependent mechanism that is significantly influenced by a common polymorphism, rs5743618, in human TLR1. These data provide fundamental new insight into pathogen manipulation of neutrophil lifespan and BLP function.
Collapse
Affiliation(s)
- Lauren C Kinkead
- Inflammation Program, University of Iowa, Iowa City, Iowa, USA.,Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Laura C Whitmore
- Inflammation Program, University of Iowa, Iowa City, Iowa, USA.,Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Jenna M McCracken
- Inflammation Program, University of Iowa, Iowa City, Iowa, USA.,Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Joshua R Fletcher
- Graduate Program in Genetics, University of Iowa, Iowa City, Iowa, USA
| | - Brandi B Ketelsen
- Inflammation Program, University of Iowa, Iowa City, Iowa, USA.,Iowa City VA Health Care System, Iowa City, Iowa, USA
| | - Justin W Kaufman
- Inflammation Program, University of Iowa, Iowa City, Iowa, USA.,Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Bradley D Jones
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA.,Graduate Program in Genetics, University of Iowa, Iowa City, Iowa, USA
| | - David S Weiss
- Emory Vaccine Center, Emory University, Atlanta, Georgia, USA
| | - Jason H Barker
- Inflammation Program, University of Iowa, Iowa City, Iowa, USA.,Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA.,Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Lee-Ann H Allen
- Inflammation Program, University of Iowa, Iowa City, Iowa, USA.,Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, USA.,Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA.,Iowa City VA Health Care System, Iowa City, Iowa, USA
| |
Collapse
|
49
|
The metabolic enzyme fructose-1,6-bisphosphate aldolase acts as a transcriptional regulator in pathogenic Francisella. Nat Commun 2017; 8:853. [PMID: 29021545 PMCID: PMC5636795 DOI: 10.1038/s41467-017-00889-7] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 08/02/2017] [Indexed: 12/31/2022] Open
Abstract
The enzyme fructose-bisphosphate aldolase occupies a central position in glycolysis and gluconeogenesis pathways. Beyond its housekeeping role in metabolism, fructose-bisphosphate aldolase has been involved in additional functions and is considered as a potential target for drug development against pathogenic bacteria. Here, we address the role of fructose-bisphosphate aldolase in the bacterial pathogen Francisella novicida. We demonstrate that fructose-bisphosphate aldolase is important for bacterial multiplication in macrophages in the presence of gluconeogenic substrates. In addition, we unravel a direct role of this metabolic enzyme in transcription regulation of genes katG and rpoA, encoding catalase and an RNA polymerase subunit, respectively. We propose a model in which fructose-bisphosphate aldolase participates in the control of host redox homeostasis and the inflammatory immune response.The enzyme fructose-bisphosphate aldolase (FBA) plays central roles in glycolysis and gluconeogenesis. Here, Ziveri et al. show that FBA of the pathogen Francisella novicida acts, in addition, as a transcriptional regulator and is important for bacterial multiplication in macrophages.
Collapse
|
50
|
Timofeev V, Bakhteeva I, Titareva G, Kopylov P, Christiany D, Mokrievich A, Dyatlov I, Vergnaud G. Russian isolates enlarge the known geographic diversity of Francisella tularensis subsp. mediasiatica. PLoS One 2017; 12:e0183714. [PMID: 28873421 PMCID: PMC5584958 DOI: 10.1371/journal.pone.0183714] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 08/09/2017] [Indexed: 11/18/2022] Open
Abstract
Francisella tularensis, a small Gram-negative bacterium, is capable of infecting a wide range of animals, including humans, and causes a plague-like disease called tularemia—a highly contagious disease with a high mortality rate. Because of these characteristics, F. tularensis is considered a potential agent of biological terrorism. Currently, F. tularensis is divided into four subspecies, which differ in their virulence and geographic distribution. Two of them, subsp. tularensis (primarily found in North America) and subsp. holarctica (widespread across the Northern Hemisphere), are responsible for tularemia in humans. Subsp. novicida is almost avirulent in humans. The fourth subspecies, subsp. mediasiatica, is the least studied because of its limited distribution and impact in human health. It is found only in sparsely populated regions of Central Asia. In this report, we describe the first focus of naturally circulating F. tularensis subsp. mediasiatica in Russia. We isolated and characterized 18 strains of this subspecies in the Altai region. All strains were highly virulent in mice. The virulence of subsp. mediasiatica in a vaccinated mouse model is intermediate between that of subsp. tularensis and subsp. holarctica. Based on a multiple-locus variable number tandem repeat analysis (MLVA), we show that the Altaic population of F. tularensis subsp. mediasiatica is genetically distinct from the classical Central Asian population, and probably is endemic to Southern Siberia. We propose to subdivide the mediasiatica subspecies into three phylogeographic groups, M.I, M.II and M.III.
Collapse
Affiliation(s)
- Vitalii Timofeev
- State Research Center for Applied Microbiology and Biotechnology (SRCAMB), Obolensk, Moscow Region, Russia
- * E-mail: (VT); (GV)
| | - Irina Bakhteeva
- State Research Center for Applied Microbiology and Biotechnology (SRCAMB), Obolensk, Moscow Region, Russia
| | - Galina Titareva
- State Research Center for Applied Microbiology and Biotechnology (SRCAMB), Obolensk, Moscow Region, Russia
| | - Pavel Kopylov
- State Research Center for Applied Microbiology and Biotechnology (SRCAMB), Obolensk, Moscow Region, Russia
| | - David Christiany
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, Gif-sur-Yvette cedex, France
| | - Alexander Mokrievich
- State Research Center for Applied Microbiology and Biotechnology (SRCAMB), Obolensk, Moscow Region, Russia
| | - Ivan Dyatlov
- State Research Center for Applied Microbiology and Biotechnology (SRCAMB), Obolensk, Moscow Region, Russia
| | - Gilles Vergnaud
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Univ. Paris-Sud, Université Paris-Saclay, Gif-sur-Yvette cedex, France
- * E-mail: (VT); (GV)
| |
Collapse
|