1
|
Lu T, Kramer ST, York MA, Zahan MN, Howlader DR, Dietz ZK, Whittier SK, Bivens NJ, Jurkevich A, Coghill LM, Picking WD, Picking WL. Spatial visualization provides insight into immune modulation by an L-DBF vaccine formulation against Shigella. Front Immunol 2025; 16:1577040. [PMID: 40336950 PMCID: PMC12056741 DOI: 10.3389/fimmu.2025.1577040] [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: 02/14/2025] [Accepted: 03/28/2025] [Indexed: 05/09/2025] Open
Abstract
Shigellosis remains a global public health problem, especially in regions with poor sanitation measures. Our prior work has demonstrated the protective efficacy of a three-dose regimen of L-DBF, a recombinant fusion of IpaD and IpaB from Shigella flexneri with the LTA1 moiety of enterotoxigenic E. coli labile toxin. Here, we investigate how a two-dose regimen (one prime and one booster) of L-DBF, formulated in an oil-in-water emulsion called ME, modulates immune responses in the lung using a spatial transcriptomics approach. Our findings show significant changes in the lung immune landscape following the vaccination, including increased expression of B cell markers, antigen presentation genes, and T cell-associated markers. Our analysis also revealed significant reprogramming of fibroblasts and cardiomyocytes, showing that fibroblasts are shifted from extracellular matrix production to immune modulation, while cardiomyocytes enhanced the signaling for immune cell recruitment and vascular stability. The communication between alveolar type 2 (AT2) cells and cardiomyocytes also increased, reflecting coordinated support for immune readiness and maintaining tissue integrity. These findings underscore the potential of L-DBF/ME vaccination to enhance both humoral and cellular immunity, as well as to reshape lung immune architecture while enhancing immune readiness, thereby offering a promising approach for effective protection against Shigella infections.
Collapse
Affiliation(s)
- Ti Lu
- Bond Life Sciences Center and Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
| | - Skyler T. Kramer
- Bioinformatics and Analytic Core, University of Missouri, Columbia, MO, United States
| | - Mary A. York
- Bioinformatics and Analytic Core, University of Missouri, Columbia, MO, United States
| | - Mst Nusrat Zahan
- Bond Life Sciences Center and Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
| | - Debaki R. Howlader
- Bond Life Sciences Center and Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
| | - Zackary K. Dietz
- Bond Life Sciences Center and Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
| | - Sean K. Whittier
- Bond Life Sciences Center and Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
| | - Nathan J. Bivens
- Genomics Technology Core, University of Missouri, Columbia, MO, United States
| | - Alexander Jurkevich
- Advanced Light Microscopy Core, University of Missouri, Columbia, MO, United States
| | - Lyndon M. Coghill
- Bond Life Sciences Center and Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
- Bioinformatics and Analytic Core, University of Missouri, Columbia, MO, United States
| | - William D. Picking
- Bond Life Sciences Center and Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
| | - Wendy L. Picking
- Bond Life Sciences Center and Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
| |
Collapse
|
2
|
Walker RI. Conserved antigens for enteric vaccines. Vaccine 2025; 50:126828. [PMID: 39914256 PMCID: PMC11878282 DOI: 10.1016/j.vaccine.2025.126828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 01/23/2025] [Accepted: 01/28/2025] [Indexed: 02/25/2025]
Abstract
Enterotoxigenic Escherichia coli (ETEC), Shigella, and Campylobacter have been identified as major causes of diarrheal diseases worldwide. In addition to overt disease and death, they are responsible for stunting in children with the risk of lifelong consequences on health and economic opportunities. All three of these bacterial pathogens, which collectively account for approximately 30 % of the cases of diarrheal diseases, are recognized as antimicrobial resistance (AMR) threats. In spite of the dangers these pathogens represent for both children and adults, there is as yet no licensed vaccine available for any of them. Fortunately, much has been accomplished to identify conserved antigens against each of these pathogens so that now relatively simple vaccines have the potential to be developed into multi-pathogen vaccines which could have a major impact on reduction of diarrheal diseases. Conserved antigens may be used even more efficiently if consolidated and expressed on a cellular vector or as part of a conjugate vaccine. A new mucosal adjuvant, double mutant heat-labile toxin (dmLT), has been shown to not only be among the conserved antigens against ETEC, but to also have properties which drive robust mucosal and systemic immune responses for antigens given orally or intramuscularly. Conserved antigens and the strategies for their use such as co-administration with dmLT will be presented in this review.
Collapse
Affiliation(s)
- Richard I Walker
- PATH, 455 Massachusetts Ave, Suite 1000, Washington, DC, 20001-2621, USA.
| |
Collapse
|
3
|
Halder S, Jaiswal N, Balajee SC, Mahata N. Efficient production of a novel recombinant fusion protein of EIEC effector IpaD and EGFP: Biophysical characterization and functional studies. BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2025; 1873:141066. [PMID: 40086498 DOI: 10.1016/j.bbapap.2025.141066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 02/27/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
The conserved invasion plasmid antigen D (IpaD) protein demonstrates broad protective capabilities against bacillary dysentery caused by Enteroinvasive Escherichia coli (EIEC) and Shigella. However, the instability of the IpaD protein at room temperature limits its therapeutic potential. The stabilization and efficient production of functional recombinant proteins remain critical challenges in therapeutic and vaccine development. This study presents a novel fluorescence fusion strategy for producing a stable IpaD-EGFP recombinant protein using a flexible linker (GGGGS)₃. The fusion technique enhances the expression level (∼53 %), solubility (∼77 %), and stability of the IpaD-EGFP fusion protein. Biophysical characterization studies suggest that the IpaD-EGFP fusion protein is stable at refrigerated temperatures for extended periods and up to 1 month at 25 °C. The IpaD-EGFP protein triggers apoptosis in Raw 267.4 cells through activation of caspases 3/7. The protein also induces antibody response in BALB/c mice indicating its immunogenicity. Together, these findings indicate that IpaD-EGFP generated in this study is a potential approach for the design and production of stable IpaD-based protein therapeutics, breaking the expensive "cold chain" of continuous refrigeration. Fusion approach significantly enhanced the solubility, yield, and stability of IpaD, while enabling efficient purification.
Collapse
Affiliation(s)
- Sudeshna Halder
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, 713209, India.
| | - Namita Jaiswal
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, 713209, India.
| | - Salari Charan Balajee
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, 713209, India.
| | - Nibedita Mahata
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, 713209, India.
| |
Collapse
|
4
|
Haldar R, Halder P, Koley H, Miyoshi SI, Das S. A newly developed oral infection mouse model of shigellosis for immunogenicity and protective efficacy studies of a candidate vaccine. Infect Immun 2025; 93:e0034624. [PMID: 39692481 PMCID: PMC11784180 DOI: 10.1128/iai.00346-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 11/06/2024] [Indexed: 12/19/2024] Open
Abstract
Shigella infection poses a significant public health challenge in the developing world. However, lack of a widely available mouse model that replicates human shigellosis creates a major bottleneck to better understanding of disease pathogenesis and development of newer drugs and vaccines. BALB/c mice pre-treated with streptomycin and iron (FeCl3) plus desferrioxamine intraperitoneally followed by oral infection with virulent Shigella flexneri 2a resulted in diarrhea, loss of body weight, bacterial colonization and progressive colitis characterized by disruption of epithelial lining, loss of crypt architecture with goblet cell depletion, increased polymorphonuclear infiltration into the mucosa, submucosal swelling (edema), and raised proinflammatory cytokines and chemokines in the large intestine. To evaluate the usefulness of the model for vaccine efficacy studies, mice were immunized intranasally with a recombinant protein vaccine containing Shigella invasion protein invasion plasmid antigen B (IpaB). Vaccinated mice conferred protection against Shigella, indicating that the model is suitable for testing of vaccine candidates. To protect both Shigella and Salmonella, a chimeric recombinant vaccine (rIpaB-T2544) was developed by fusing IpaB with Salmonella outer membrane protein T2544. Vaccinated mice developed antigen-specific serum IgG and IgA antibodies and a balanced Th1/Th2 response and were protected against oral challenge with Shigella (S. flexneri 2a, Shigella dysenteriae, and Shigella sonnei) using our present mouse model and Salmonella (Salmonella Typhi and Paratyphi) using an iron overload mouse model. We describe here the development of an oral Shigella infection model in wild-type mouse. This model was successfully used to demonstrate the immunogenicity and protective efficacy of a candidate protein subunit vaccine against Shigella.
Collapse
MESH Headings
- Animals
- Dysentery, Bacillary/prevention & control
- Dysentery, Bacillary/immunology
- Dysentery, Bacillary/microbiology
- Dysentery, Bacillary/pathology
- Disease Models, Animal
- Mice
- Shigella Vaccines/immunology
- Shigella Vaccines/administration & dosage
- Mice, Inbred BALB C
- Antibodies, Bacterial/blood
- Shigella flexneri/immunology
- Female
- Antigens, Bacterial/immunology
- Antigens, Bacterial/genetics
- Bacterial Proteins/immunology
- Bacterial Proteins/genetics
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Immunoglobulin G/blood
Collapse
Affiliation(s)
- Risha Haldar
- Division of Clinical Medicine, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India
| | - Prolay Halder
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India
| | - Hemanta Koley
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India
| | - Shin-ichi Miyoshi
- Division of Medicine, Dentistry and Pharmaceutical Sciences, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Okayama, Japan
| | - Santasabuj Das
- Division of Clinical Medicine, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India
- Division of Biological Science, ICMR-National Institute of Occupational Health, Ahmedabad, Gujarat, India
| |
Collapse
|
5
|
Hashemi P, Osanloo M, Farjadfar A, Nasiri-Ghiri M, Zarenezhad E, Mahmoodi S. A multi-epitope protein vaccine encapsulated in alginate nanoparticles as a candidate vaccine against Shigella sonnei. Sci Rep 2024; 14:22484. [PMID: 39341926 PMCID: PMC11438873 DOI: 10.1038/s41598-024-73105-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 09/13/2024] [Indexed: 10/01/2024] Open
Abstract
Shigellosis, caused by the Gram-negative bacterium Shigella, is a major global health challenge. Despite extensive research over the past two decades, no commercial vaccine is available to prevent Shigella infection. Developing multi-epitope vaccines offers a promising and innovative approach to tackling infectious diseases. In this study, we produced a multi-epitope vaccine candidate using E. coli BL21 (DE3) plysS bacteria and purified the vaccine protein with Ni-NTA affinity chromatography. We then prepared alginate nanoparticles containing the vaccine protein, with a particle size of 122 ± 6 nm, PDI 0.17, SPAN 0.83, and zeta potential of -27 ± 2 mV. Successful protein loading was confirmed through nanodrop and ATR-FTIR analyses. To evaluate the immunogenicity of the encapsulated vaccine, mice were orally vaccinated, and their serum was analyzed for IgG, IL-4, and IFN-γ levels cytokines. The results showed a significant increase in IgG level in the vaccinated group compared to controls. Additionally, the vaccinated group exhibited a notable increase in IL-4 and IFN-γ cytokines, indicating a robust Th-cell-mediated immune response essential for combating Shigella. Our nano-vaccine demonstrated high efficacy in activating both humoral and cellular immunity, effectively protecting against the bacteria. The alginate-based oral vaccine candidate thus emerges as a promising strategy for developing a multi-epitope vaccine candidate against Shigella.
Collapse
Affiliation(s)
- Parisa Hashemi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Mahmoud Osanloo
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Akbar Farjadfar
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Mahdi Nasiri-Ghiri
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Elham Zarenezhad
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Shirin Mahmoodi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran.
| |
Collapse
|
6
|
Li S, Zhang W. Mapping the functional B-cell epitopes of Shigella invasion plasmid antigen D (IpaD). Appl Environ Microbiol 2024; 90:e0098824. [PMID: 39082807 PMCID: PMC11337796 DOI: 10.1128/aem.00988-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 07/17/2024] [Indexed: 08/22/2024] Open
Abstract
Shigella bacteria utilize the type III secretion system (T3SS) to invade host cells and establish local infection. Invasion plasmid antigen D (IpaD), a component of Shigella T3SS, has garnered extensive interest as a vaccine target, primarily due to its pivotal role in the Shigella invasion, immunogenic property, and a high degree of conservation across Shigella species and serotypes. Currently, we are developing an epitope- and structure-based multivalent vaccine against shigellosis and require functional epitope antigens of key Shigella virulence determinants including IpaD. However, individual IpaD B-cell epitopes, their contributions to the overall immunogenicity, and functional activities attributing to bacteria invasion have not been fully characterized. In this study, we predicted continuous B-cell epitopes in silico and fused each epitope to a carrier protein. Then, we immunized mice intramuscularly with each epitope fusion protein, examined the IpaD-specific antibody responses, and measured antibodies from each epitope fusion for the activity against Shigella invasion in vitro. Data showed that all epitope fusion proteins induced similar levels of anti-IpaD IgG antibodies in mice, and differences were noted for antibody inhibition activity against Shigella invasion. IpaD epitope 1 (SPGGNDGNSV), IpaD epitope 2 (LGGNGEVVLDNA), and IpaD epitope 5 (SPNNTNGSSTET) induced antibodies significantly better in inhibiting invasion from Shigella flexneri 2a, and epitopes 1 and 5 elicited antibodies more effectively at preventing invasion of Shigella sonnei. These results suggest that IpaD epitopes 1 and 5 can be the IpaD representative antigens for epitope-based polyvalent protein construction and protein-based cross-protective Shigella vaccine development.IMPORTANCEShigella is a leading cause of diarrhea in children younger than 5 years in developing countries (children's diarrhea) and continues to be a major threat to public health. No licensed vaccines are currently available against the heterogeneous Shigella species and serotype strains. Aiming to develop a cross-protective multivalent vaccine against shigellosis and dysentery, we applied novel multiepitope fusion antigen (MEFA) technology to construct a broadly immunogenic polyvalent protein antigen, by presenting functional epitopes of multiple Shigella virulence determinants on a backbone protein. The functional IpaD epitopes identified from this study will essentially allow us to construct an optimal polyvalent Shigella immunogen, leading to the development of a cross-protective vaccine against shigellosis (and dysentery) and the improvement of global health. In addition, identifying functional epitopes from heterogeneous virulence determinants and using them as antigenic representatives for the development of cross-protective multivalent vaccines can be applied generally in vaccine development.
Collapse
Affiliation(s)
- Siqi Li
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Weiping Zhang
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
7
|
Lu T, Raju M, Howlader DR, Dietz ZK, Whittier SK, Varisco DJ, Ernst RK, Coghill LM, Picking WD, Picking WL. Vaccination with a Protective Ipa Protein-Containing Nanoemulsion Differentially Alters the Transcriptomic Profiles of Young and Elderly Mice following Shigella Infection. Vaccines (Basel) 2024; 12:618. [PMID: 38932347 PMCID: PMC11209624 DOI: 10.3390/vaccines12060618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 05/28/2024] [Accepted: 06/01/2024] [Indexed: 06/28/2024] Open
Abstract
Shigella spp. are responsible for bacillary dysentery or shigellosis transmitted via the fecal-oral route, causing significant morbidity and mortality, especially among vulnerable populations. There are currently no licensed Shigella vaccines. Shigella spp. use a type III secretion system (T3SS) to invade host cells. We have shown that L-DBF, a recombinant fusion of the T3SS needle tip (IpaD) and translocator (IpaB) proteins with the LTA1 subunit of enterotoxigenic E. coli labile toxin, is broadly protective against Shigella spp. challenge in a mouse lethal pulmonary model. Here, we assessed the effect of LDBF, formulated with a unique TLR4 agonist called BECC470 in an oil-in-water emulsion (ME), on the murine immune response in a high-risk population (young and elderly) in response to Shigella challenge. Dual RNA Sequencing captured the transcriptome during Shigella infection in vaccinated and unvaccinated mice. Both age groups were protected by the L-DBF formulation, while younger vaccinated mice exhibited more adaptive immune response gene patterns. This preliminary study provides a step toward identifying the gene expression patterns and regulatory pathways responsible for a protective immune response against Shigella. Furthermore, this study provides a measure of the challenges that need to be addressed when immunizing an aging population.
Collapse
Affiliation(s)
- Ti Lu
- Bond Life Sciences Center and Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA; (D.R.H.); (W.D.P.)
| | - Murugesan Raju
- Bioinformatics and Analytic Core, University of Missouri, Columbia, MO 65211, USA (L.M.C.)
- MU Institute for Data Science and Informatics, University of Missouri, Columbia, MO 65211, USA
| | - Debaki R. Howlader
- Bond Life Sciences Center and Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA; (D.R.H.); (W.D.P.)
| | - Zackary K. Dietz
- Bond Life Sciences Center and Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA; (D.R.H.); (W.D.P.)
| | - Sean K. Whittier
- Bond Life Sciences Center and Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA; (D.R.H.); (W.D.P.)
| | - David J. Varisco
- Department of Microbial Pathogenesis, University of Maryland, Baltimore, MD 21201, USA
| | - Robert K. Ernst
- Department of Microbial Pathogenesis, University of Maryland, Baltimore, MD 21201, USA
| | - Lyndon M. Coghill
- Bioinformatics and Analytic Core, University of Missouri, Columbia, MO 65211, USA (L.M.C.)
- MU Institute for Data Science and Informatics, University of Missouri, Columbia, MO 65211, USA
| | - William D. Picking
- Bond Life Sciences Center and Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA; (D.R.H.); (W.D.P.)
| | - Wendy L. Picking
- Bond Life Sciences Center and Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA; (D.R.H.); (W.D.P.)
| |
Collapse
|
8
|
Lu T, Das S, Howlader DR, Picking WD, Picking WL. Shigella Vaccines: The Continuing Unmet Challenge. Int J Mol Sci 2024; 25:4329. [PMID: 38673913 PMCID: PMC11050647 DOI: 10.3390/ijms25084329] [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: 02/27/2024] [Revised: 04/09/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Shigellosis is a severe gastrointestinal disease that annually affects approximately 270 million individuals globally. It has particularly high morbidity and mortality in low-income regions; however, it is not confined to these regions and occurs in high-income nations when conditions allow. The ill effects of shigellosis are at their highest in children ages 2 to 5, with survivors often exhibiting impaired growth due to infection-induced malnutrition. The escalating threat of antibiotic resistance further amplifies shigellosis as a serious public health concern. This review explores Shigella pathology, with a primary focus on the status of Shigella vaccine candidates. These candidates include killed whole-cells, live attenuated organisms, LPS-based, and subunit vaccines. The strengths and weaknesses of each vaccination strategy are considered. The discussion includes potential Shigella immunogens, such as LPS, conserved T3SS proteins, outer membrane proteins, diverse animal models used in Shigella vaccine research, and innovative vaccine development approaches. Additionally, this review addresses ongoing challenges that necessitate action toward advancing effective Shigella prevention and control measures.
Collapse
Affiliation(s)
- Ti Lu
- Department of Veterinary Pathobiology and Bond Life Science Center, University of Missouri, Columbia, MO 65201, USA; (D.R.H.); (W.D.P.)
| | - Sayan Das
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA;
| | - Debaki R. Howlader
- Department of Veterinary Pathobiology and Bond Life Science Center, University of Missouri, Columbia, MO 65201, USA; (D.R.H.); (W.D.P.)
| | - William D. Picking
- Department of Veterinary Pathobiology and Bond Life Science Center, University of Missouri, Columbia, MO 65201, USA; (D.R.H.); (W.D.P.)
| | - Wendy L. Picking
- Department of Veterinary Pathobiology and Bond Life Science Center, University of Missouri, Columbia, MO 65201, USA; (D.R.H.); (W.D.P.)
| |
Collapse
|
9
|
Lu T, Ericsson AC, Dietz ZK, Cato AK, Coghill LM, Picking WD, Picking WL. Impact of an intranasal L-DBF vaccine on the gut microbiota in young and elderly mice. Gut Microbes 2024; 16:2426619. [PMID: 39520707 PMCID: PMC11552291 DOI: 10.1080/19490976.2024.2426619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/21/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024] Open
Abstract
Shigella spp. cause bacillary dysentery (shigellosis) with high morbidity and mortality in low- and middle-income countries. Infection occurs through the fecal-oral route and can be devastating for vulnerable populations, including infants and the elderly. These bacteria invade host cells using a type III secretion system (T3SS). No licensed vaccine yet exists for shigellosis, but we have generated a recombinant fusion protein, L-DBF, combining the T3SS needle tip protein (IpaD), translocator protein (IpaB), and the LTA1 subunit of enterotoxigenic E. coli labile toxin, which offers broad protection in a mouse model of lethal pulmonary infection. The L-DBF vaccine protects high-risk groups, including young and elderly mice. Here, we investigated how the gut microbiota of young and elderly mice responds to intranasal L-DBF vaccination formulated in an oil-in-water emulsion (ME). Samples from lungs, small intestines, and feces were collected on day 14 after 2 or 3 doses of L-DBF in ME. 16S rRNA gene sequencing revealed age-dependent changes in gut microbiota post-vaccination. The vaccine-induced changes were more prominent in the elderly mice and were most significant in the intestinal tract, indicating that vaccination by the intranasal route can have a tremendous impact on the gut environment. These findings provide insight into the communication between the intranasal mucosal surface following subunit vaccination and the microbiota at a distant mucosal site, thereby highlighting the impact of vaccination and the host's microbiome.
Collapse
Affiliation(s)
- Ti Lu
- Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - Aaron C. Ericsson
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
- University of Missouri Metagenomics Center (MUMC), University of Missouri, Columbia, MO, USA
| | - Zackary K. Dietz
- Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - Alexa K. Cato
- Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - Lyndon M. Coghill
- Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - William D. Picking
- Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - Wendy L. Picking
- Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| |
Collapse
|
10
|
Clarkson KA, Porter CK, Talaat KR, Kapulu MC, Chen WH, Frenck RW, Bourgeois AL, Kaminski RW, Martin LB. Shigella-Controlled Human Infection Models: Current and Future Perspectives. Curr Top Microbiol Immunol 2024; 445:257-313. [PMID: 35616717 PMCID: PMC7616482 DOI: 10.1007/82_2021_248] [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] [Indexed: 10/18/2022]
Abstract
Shigella-controlled human infection models (CHIMs) are an invaluable tool utilized by the vaccine community to combat one of the leading global causes of infectious diarrhea, which affects infants, children and adults regardless of socioeconomic status. The impact of shigellosis disproportionately affects children in low- and middle-income countries (LMICs) resulting in cognitive and physical stunting, perpetuating a cycle that must be halted. Shigella-CHIMs not only facilitate the early evaluation of enteric countermeasures and up-selection of the most promising products but also provide insight into mechanisms of infection and immunity that are not possible utilizing animal models or in vitro systems. The greater understanding of shigellosis obtained in CHIMs builds and empowers the development of new generation solutions to global health issues which are unattainable in the conventional laboratory and clinical settings. Therefore, refining, mining and expansion of safe and reproducible infection models hold the potential to create effective means to end diarrheal disease and associated co-morbidities associated with Shigella infection.
Collapse
Affiliation(s)
- Kristen A Clarkson
- Department of Diarrheal Disease Research, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Chad K Porter
- Enteric Disease Department, Naval Medical Research Center, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Kawsar R Talaat
- Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, 624 North Broadway Street Hampton House, Baltimore, MD, 21205, USA
| | - Melissa C Kapulu
- Department of Biosciences, KEMRI-Wellcome Trust Research Programme, Kilifi County Hospital, Off Bofa Road, Kilifi, 80108, Kenya
| | - Wilbur H Chen
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD, 21201, USA
| | - Robert W Frenck
- Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| | - A Louis Bourgeois
- PATH Center for Vaccine Innovation and Access, 455 Massachusetts Avenue NW, Washington, DC, 20001, USA
| | - Robert W Kaminski
- Department of Diarrheal Disease Research, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD, 20910, USA
| | - Laura B Martin
- GSK Vaccines Institute for Global Health, Via Fiorentina 1, 53100, Siena, Italy.
| |
Collapse
|
11
|
Lu T, Howlader DR, Das S, Dietz ZK, Nagel AC, Whittier SK, Picking WD, Picking WL. The L-DBF vaccine cross protects mice against different Shigella serotypes after prior exposure to the pathogen. Microbiol Spectr 2023; 11:e0006223. [PMID: 37787548 PMCID: PMC10714971 DOI: 10.1128/spectrum.00062-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 08/20/2023] [Indexed: 10/04/2023] Open
Abstract
IMPORTANCE Shigellosis is endemic to low- and middle-income regions of the world where children are especially vulnerable. In many cases, there are pre-existing antibodies in the local population and the effect of prior exposure should be considered in the development and testing of vaccines against Shigella infection. Our study shows that L-DBF-induced immune responses are not adversely affected by prior exposure to this pathogen. Moreover, somewhat different cytokine profiles were observed in the lungs of vaccinated mice not having been exposed to Shigella, suggesting that the immune responses elicited by Shigella infection and L-DBF vaccination follow different pathways.
Collapse
Affiliation(s)
- Ti Lu
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas, USA
| | - Debaki R. Howlader
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas, USA
| | - Sayan Das
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas, USA
| | - Zackary K. Dietz
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas, USA
| | | | - Sean K. Whittier
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas, USA
| | - William D. Picking
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas, USA
| | - Wendy L. Picking
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas, USA
| |
Collapse
|
12
|
Li S, Anvari S, Ptacek G, Upadhyay I, Kaminski RW, Sack DA, Zhang W. A broadly immunogenic polyvalent Shigella multiepitope fusion antigen protein protects against Shigella sonnei and Shigella flexneri lethal pulmonary challenges in mice. Infect Immun 2023; 91:e0031623. [PMID: 37795982 PMCID: PMC10652900 DOI: 10.1128/iai.00316-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 08/21/2023] [Indexed: 10/06/2023] Open
Abstract
There are no licensed vaccines for Shigella, a leading cause of children's diarrhea and a common etiology of travelers' diarrhea. To develop a cross-protective Shigella vaccine, in this study, we constructed a polyvalent protein immunogen to present conserved immunodominant epitopes of Shigella invasion plasmid antigens B (IpaB) and D (IpaD), VirG, GuaB, and Shiga toxins on backbone protein IpaD, by applying an epitope- and structure-based multiepitope-fusion-antigen (MEFA) vaccinology platform, examined protein (Shigella MEFA) broad immunogenicity, and evaluated antibody function against Shigella invasion and Shiga toxin cytotoxicity but also protection against Shigella lethal challenge. Mice intramuscularly immunized with Shigella MEFA protein developed IgG responses to IpaB, IpaD, VirG, GuaB, and Shiga toxins 1 and 2; mouse sera significantly reduced invasion of Shigella sonnei, Shigella flexneri serotype 2a, 3a, or 6, Shigella boydii, and Shigella dysenteriae type 1 and neutralized cytotoxicity of Shiga toxins of Shigella and Shiga toxin-producing Escherichia coli in vitro. Moreover, mice intranasally immunized with Shigella MEFA protein (adjuvanted with dmLT) developed antigen-specific serum IgG, lung IgG and IgA, and fecal IgA antibodies, and survived from lethal pulmonary challenge with S. sonnei or S. flexneri serotype 2a, 3a, or 6. In contrast, the control mice died, became unresponsive, or lost 20% of body weight in 48 h. These results indicated that this Shigella MEFA protein is broadly immunogenic, induces broadly functional antibodies, and cross-protects against lethal pulmonary challenges with S. sonnei or S. flexneri serotypes, suggesting a potential application of this polyvalent MEFA protein in Shigella vaccine development.
Collapse
Affiliation(s)
- Siqi Li
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Shaghayegh Anvari
- Department of Diagnostic Medicine/Pathobiology, Kansas State University College of Veterinary Medicine, Manhattan, Kansas, USA
| | - Galen Ptacek
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Ipshita Upadhyay
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Robert W. Kaminski
- Department of Enteric Infections, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - David A. Sack
- Department of International Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Weiping Zhang
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
13
|
Lu T, Das S, Howlader DR, Jain A, Hu G, Dietz ZK, Zheng Q, Ratnakaram SSK, Whittier SK, Varisco DJ, Ernst RK, Picking WD, Picking WL. Impact of the TLR4 agonist BECC438 on a novel vaccine formulation against Shigella spp. Front Immunol 2023; 14:1194912. [PMID: 37744341 PMCID: PMC10512073 DOI: 10.3389/fimmu.2023.1194912] [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: 03/27/2023] [Accepted: 08/18/2023] [Indexed: 09/26/2023] Open
Abstract
Shigellosis (bacillary dysentery) is a severe gastrointestinal infection with a global incidence of 90 million cases annually. Despite the severity of this disease, there is currently no licensed vaccine against shigellosis. Shigella's primary virulence factor is its type III secretion system (T3SS), which is a specialized nanomachine used to manipulate host cells. A fusion of T3SS injectisome needle tip protein IpaD and translocator protein IpaB, termed DBF, when admixed with the mucosal adjuvant double-mutant labile toxin (dmLT) from enterotoxigenic E. coli was protective using a murine pulmonary model. To facilitate the production of this platform, a recombinant protein that consisted of LTA-1, the active moiety of dmLT, and DBF were genetically fused, resulting in L-DBF, which showed improved protection against Shigella challenge. To extrapolate this protection from mice to humans, we modified the formulation to provide for a multivalent presentation with the addition of an adjuvant approved for use in human vaccines. Here, we show that L-DBF formulated (admix) with a newly developed TLR4 agonist called BECC438 (a detoxified lipid A analog identified as Bacterial Enzymatic Combinatorial Chemistry candidate #438), formulated as an oil-in-water emulsion, has a very high protective efficacy at low antigen doses against lethal Shigella challenge in our mouse model. Optimal protection was observed when this formulation was introduced at a mucosal site (intranasally). When the formulation was then evaluated for the immune response it elicits, protection appeared to correlate with high IFN-γ and IL-17 secretion from mucosal site lymphocytes.
Collapse
Affiliation(s)
- Ti Lu
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
| | - Sayan Das
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, MD, United States
| | - Debaki R. Howlader
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
| | - Akshay Jain
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
| | - Gang Hu
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
| | - Zackary K. Dietz
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
| | - Qi Zheng
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
| | | | - Sean K. Whittier
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
| | - David J. Varisco
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, MD, United States
| | - Robert K. Ernst
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, MD, United States
| | - William D. Picking
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
| | - Wendy L. Picking
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
| |
Collapse
|
14
|
Toward a Shigella Vaccine: Opportunities and Challenges to Fight an Antimicrobial-Resistant Pathogen. Int J Mol Sci 2023; 24:ijms24054649. [PMID: 36902092 PMCID: PMC10003550 DOI: 10.3390/ijms24054649] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 03/04/2023] Open
Abstract
Shigellosis causes more than 200,000 deaths worldwide and most of this burden falls on Low- and Middle-Income Countries (LMICs), with a particular incidence in children under 5 years of age. In the last decades, Shigella has become even more worrisome because of the onset of antimicrobial-resistant strains (AMR). Indeed, the WHO has listed Shigella as one of the priority pathogens for the development of new interventions. To date, there are no broadly available vaccines against shigellosis, but several candidates are being evaluated in preclinical and clinical studies, bringing to light very important data and information. With the aim to facilitate the understanding of the state-of-the-art of Shigella vaccine development, here we report what is known about Shigella epidemiology and pathogenesis with a focus on virulence factors and potential antigens for vaccine development. We discuss immunity after natural infection and immunization. In addition, we highlight the main characteristics of the different technologies that have been applied for the development of a vaccine with broad protection against Shigella.
Collapse
|
15
|
Baruah N, Ahamad N, Halder P, Koley H, Katti DS. Facile synthesis of multi-faceted, biomimetic and cross-protective nanoparticle-based vaccines for drug-resistant Shigella: a flexible platform technology. J Nanobiotechnology 2023; 21:34. [PMID: 36710326 PMCID: PMC9884485 DOI: 10.1186/s12951-023-01780-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 01/12/2023] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND No commercial vaccines are available against drug-resistant Shigella due to serotype-specific/narrow-range of protection. Nanoparticle-based biomimetic vaccines involving stable, conserved, immunogenic proteins fabricated using facile chemistries can help formulate a translatable cross-protective Shigella vaccine. Such systems can also negate cold-chain transportation/storage thus overcoming challenges prevalent in various settings. METHODS We explored facile development of biomimetic poly (lactide-co-glycolide)/PLGA 50:50 based nanovaccines (NVs), encapsulating conserved stabilized antigen(s)/immunostimulant of S. dysenteriae 1 origin surface-modified using simple chemistries. All encapsulants (IpaC/IpaB/LPS) and nanoparticles (NPs)-bare and modified (NV), were thoroughly characterized. Effect of IpaC on cellular uptake of NPs was assessed in-vitro. Immunogenicity of the NVs was assessed in-vivo in BALB/c mice by intranasal immunization. Cross-protective efficacy was assessed by intraperitoneally challenging the immunized groups with a high dose of heterologous S. flexneri 2a and observing for visible diarrhea, weight loss and survival. Passive-protective ability of the simplest NV was assessed in the 5-day old progeny of vaccinated mice. RESULTS All the antigens and immunostimulant to be encapsulated were successfully purified and found to be stable both before and after encapsulation into NPs. The ~ 300 nm sized NPs with a zeta potential of ~ - 25 mV released ~ 60% antigen by 14th day suggesting an appropriate delivery kinetics. The NPs could be successfully surface-modified with IpaC and/or CpG DNA. In vitro experiments revealed that the presence of IpaC can significantly increase cellular uptake of NPs. All NVs were found to be cytocompatible and highly immunogenic. Antibodies in sera of NV-immunized mice could recognize heterologous Shigella. Immunized sera also showed high antibody and cytokine response. The immunized groups were protected from diarrhea and weight loss with ~ 70-80% survival upon heterologous Shigella challenge. The simplest NV showed ~ 88% survival in neonates. CONCLUSIONS Facile formulation of biomimetic NVs can result in significant cross-protection. Further, passive protection in neonates suggest that parental immunization could protect infants, the most vulnerable group in context of Shigella infection. Non-invasive route of vaccination can also lead to greater patient compliance making it amenable for mass-immunization. Overall, our work contributes towards a yet to be reported platform technology for facile development of cross-protective Shigella vaccines.
Collapse
Affiliation(s)
- Namrata Baruah
- grid.417965.80000 0000 8702 0100Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016 Uttar Pradesh India ,grid.417965.80000 0000 8702 0100The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, 208016 Uttar Pradesh India
| | - Nadim Ahamad
- grid.417965.80000 0000 8702 0100Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016 Uttar Pradesh India
| | - Prolay Halder
- grid.419566.90000 0004 0507 4551Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, 700010 West Bengal India
| | - Hemanta Koley
- grid.419566.90000 0004 0507 4551Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, 700010 West Bengal India
| | - Dhirendra S. Katti
- grid.417965.80000 0000 8702 0100Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016 Uttar Pradesh India ,grid.417965.80000 0000 8702 0100The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, 208016 Uttar Pradesh India
| |
Collapse
|
16
|
Baruah N, Halder P, Koley H, Katti DS. Stable Recombinant Invasion Plasmid Antigen C (IpaC)-Based Single Dose Nanovaccine for Shigellosis. Mol Pharm 2022; 19:3884-3893. [PMID: 36122190 DOI: 10.1021/acs.molpharmaceut.2c00378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Shigellosis, caused by the bacteria Shigella, is the leading cause of bacterial diarrhea and the second leading cause of diarrheal death among children under the age of five. Unfortunately, Shigella strains have acquired resistance to antibiotics, and a commercial vaccine is yet to be available. We have previously demonstrated that Shigella dysenteriae serotype 1 (Sd1)-based recombinant, stabilized, "invasion plasmid antigen C" (IpaC; 42 kDa) protein can induce robust immune responses in BALB/c mice against a challenge of a high dose of heterologous Shigella when immunized via three intranasal doses of IpaC without an adjuvant. In this work, in order to reduce the frequency of dosing and increase possible patient compliance, based on our previous screening, the minimum protective dose of stabilized IpaC (20 μg) was encapsulated in biodegradable polymeric poly(lactide-co-glycolide) nanoparticles (∼370 nm) and intranasally administered in BALB/c mice in a single dose. Interestingly, a single intranasal dose of the developed vaccine particles encapsulating only 20 μg of Sd1 IpaC led to a temporal increase in the antibody production with an improved cytokine response compared to free IpaC administered three times as described in our previous report. Upon intraperitoneal challenge with a high dose of heterologous Shigella flexneri 2a (common in circulation), the immunized animals were protected from diarrhea, lethargy, and weight loss with ∼67% survival, while all the control animals died by 36 h of the challenge. Overall, the developed nanovaccine could be explored as a potential noninvasive, cross-protective, single-dose, single-antigen Shigella vaccine amenable for scale-up and eventual mass immunization.
Collapse
Affiliation(s)
- Namrata Baruah
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India.,The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
| | - Prolay Halder
- Division of Bacteriology, ICMR-National Institute of Cholera & Enteric Diseases, Kolkata, West Bengal 700010, India
| | - Hemanta Koley
- Division of Bacteriology, ICMR-National Institute of Cholera & Enteric Diseases, Kolkata, West Bengal 700010, India
| | - Dhirendra S Katti
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India.,The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
| |
Collapse
|
17
|
Bernshtein B, Ndungo E, Cizmeci D, Xu P, Kováč P, Kelly M, Islam D, Ryan ET, Kotloff KL, Pasetti MF, Alter G. Systems approach to define humoral correlates of immunity to Shigella. Cell Rep 2022; 40:111216. [PMID: 35977496 PMCID: PMC9396529 DOI: 10.1016/j.celrep.2022.111216] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 04/22/2022] [Accepted: 07/21/2022] [Indexed: 11/24/2022] Open
Abstract
Shigella infection is the second leading cause of death due to diarrheal disease in young children worldwide. With the rise of antibiotic resistance, initiatives to design and deploy a safe and effective Shigella vaccine are urgently needed. However, efforts to date have been hindered by the limited understanding of immunological correlates of protection against shigellosis. We applied systems serology to perform a comprehensive analysis of Shigella-specific antibody responses in sera obtained from volunteers before and after experimental infection with S. flexneri 2a in a series of controlled human challenge studies. Polysaccharide-specific antibody responses are infrequent prior to infection and evolve concomitantly with disease severity. In contrast, pre-existing antibody responses to type 3 secretion system proteins, particularly IpaB, consistently associate with clinical protection from disease. Linked to particular Fc-receptor binding patterns, IpaB-specific antibodies leverage neutrophils and monocytes, and complement and strongly associate with protective immunity. IpaB antibody-mediated functions improve with a subsequent rechallenge resulting in complete clinical protection. Collectively, our systems serological analyses indicate protein-specific functional correlates of immunity against Shigella in humans. Serological profiling of Shigella human challenge studies indicates protective markers Pre-existing IpaB-specific functional antibodies associate with less severe disease OPS immune responses post challenge are linked to less severe disease Shigella rechallenge boosts IpaB but not OPS functional antibody responses
Collapse
Affiliation(s)
| | - Esther Ndungo
- Department of Pediatrics, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Deniz Cizmeci
- Ragon Institute of MGH, Harvard and MIT, Cambridge, MA, USA
| | - Peng Xu
- NIDDK, LBC, National Institutes of Health, Bethesda, MD, USA
| | - Pavol Kováč
- NIDDK, LBC, National Institutes of Health, Bethesda, MD, USA
| | - Meagan Kelly
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA
| | - Dilara Islam
- Department of Pediatrics, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Edward T Ryan
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Karen L Kotloff
- Department of Pediatrics, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Marcela F Pasetti
- Department of Pediatrics, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Galit Alter
- Ragon Institute of MGH, Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
18
|
Li S, Han X, Upadhyay I, Zhang W. Characterization of Functional B-Cell Epitopes at the Amino Terminus of Shigella Invasion Plasmid Antigen B (IpaB). Appl Environ Microbiol 2022; 88:e0038422. [PMID: 35856689 PMCID: PMC9361828 DOI: 10.1128/aem.00384-22] [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: 03/01/2022] [Accepted: 06/29/2022] [Indexed: 01/22/2023] Open
Abstract
Shigella invasion plasmid antigen B (IpaB) plays an important role in causing shigellosis. While IpaB's protein structure, contribution to disease mechanism, and protective immunity against Shigella infection have been well studied, the significance of individual antigenic domains, especially at the N terminus, has not been systematically characterized. In an attempt to identify IpaB protein functional epitopes and to construct an optimized polyvalent multiepitope fusion antigen (MEFA) immunogen for development of a protein-based cross protective Shigella vaccine, in this study, we in silico identified immunodominant B-cell epitopes from the IpaB N terminus, fused each epitope to carrier protein CsaB (the major subunit of enterotoxigenic Escherichia coli CS4 adhesin) for epitope fusion proteins, immunized mice with each epitope fusion protein, examined IpaB-specific antibody responses, and assessed antibody functional activity against Shigella bacterial invasion. A total of 10 B-cell continuous epitopes were identified from IpaB N terminus, and after being fused to carrier protein CsaB, each epitope induced anti-IpaB IgG responses in the intramuscularly immunized mice. While in vitro antibody invasion inhibition assays demonstrated that antibodies derived from each identified epitope were functional, epitopes 1 (LAKILASTELGDNTIQAA), 2 (HSTSNILIPELKAPKSL), and 4 (QARQQKNLEFSDKI) induced antibodies to inhibit Shigella sonnei and Shigella flexneri invasion at levels similar to those of recombinant IpaB protein, suggesting that these three IpaB epitopes can be used potentially as IpaB-representing antigens to induce protective anti-IpaB antibodies and for construction of an epitope-based polyvalent MEFA protein immunogen for Shigella vaccine development. IMPORTANCE Currently, there are no effective measures for control or prevention of Shigella infection, the most common cause of diarrhea in children 3 to 5 years of age in developing countries. Challenges in developing Shigella vaccines include virulence heterogeneity among species and serotypes. To overcome virulence heterogeneity challenge and to develop a protein-based multivalent Shigella vaccine, we targeted a panel of virulence factors, including invasion plasmid antigens, identified functional antigenic domains or epitopes as representative antigens, and applied the novel epitope- and structure-based vaccinology platform multiepitope fusion antigen (MEFA) to integrate functional antigenic domains or epitopes into a backbone immunogen to produce a polyvalent immunogen for cross protective antibodies. Identification of functional IpaB epitopes from this study enhances our understanding of IpaB immunogenicity and allows us to directly utilize IpaB epitopes for construction of a cross protective polyvalent Shigella immunogen and to accelerate development of a protein-based Shigella vaccine.
Collapse
Affiliation(s)
- Siqi Li
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Xinfeng Han
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Sichuan Agricultural University College of Veterinary Medicine, Chengdu, Sichuan, People’s Republic of China
| | - Ipshita Upadhyay
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Weiping Zhang
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
19
|
Nogueira WG, Gois BVA, Pinheiro KDC, Aragão ADO, Queiroz ALC, da Silva AL, Folador AC, Ramos RTJ. Viral Metagenomics Reveals Widely Diverse Viral Community of Freshwater Amazonian Lake. Front Public Health 2022; 10:869886. [PMID: 35548089 PMCID: PMC9081339 DOI: 10.3389/fpubh.2022.869886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 03/24/2022] [Indexed: 11/28/2022] Open
Abstract
Despite the importance of understanding the ecology of freshwater viruses, there are not many studies on the subject compared to marine viruses. The microbiological interactions in these environments are still poorly known, especially between bacteriophages and their host bacteria and between cyanophages and cyanobacteria. Lake Bologna, Belém, capital of the Brazilian State of Pará, is a water source that supplies the city and its metropolitan region. However, it remains unexplored regarding the contents of its virome and viral diversity composition. Therefore, this work aims to explore the taxonomic diversity of DNA viruses in this lake, especially bacteriophages and cyanophages, since they can act as transducers of resistance genes and reporters of water quality for human consumption. We used metagenomic sequencing data generated by previous studies. We analyzed it at the taxonomic level using the tools Kraken2, Bracken, and Pavian; later, the data was assembled using Genome Detective, which performs the assembly of viruses. The results observed here suggest the existence of a widely diverse viral community and established microbial phage-regulated dynamics in Lake Bolonha. This work is the first ever to describe the virome of Lake Bolonha using a metagenomic approach based on high-throughput sequencing, as it contributes to the understanding of water-related public health concerns regarding the spreading of antibiotic resistance genes and population control of native bacteria and cyanobacteria.
Collapse
Affiliation(s)
| | | | | | - Andressa de Oliveira Aragão
- Laboratory of Genomic and Bioinformatics, Center of Genomics and System Biology, Federal University of Pará, Belém, Brazil
| | | | - Artur Luiz da Silva
- Laboratory of Genomic and Bioinformatics, Center of Genomics and System Biology, Federal University of Pará, Belém, Brazil
| | - Adriana Carneiro Folador
- Laboratory of Genomic and Bioinformatics, Center of Genomics and System Biology, Federal University of Pará, Belém, Brazil
| | - Rommel Thiago Jucá Ramos
- Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
- *Correspondence: Rommel Thiago Jucá Ramos
| |
Collapse
|
20
|
From Concept to Clinical Product: A Brief History of the Novel Shigella Invaplex Vaccine’s Refinement and Evolution. Vaccines (Basel) 2022; 10:vaccines10040548. [PMID: 35455297 PMCID: PMC9025769 DOI: 10.3390/vaccines10040548] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/12/2022] [Accepted: 03/23/2022] [Indexed: 01/13/2023] Open
Abstract
The Shigella invasin complex or Invaplex vaccine is a unique subunit approach to generate a protective immune response. Invaplex is a large, macromolecular complex consisting of the major Shigella antigens: lipopolysaccharide (LPS) and the invasion plasmid antigen (Ipa) proteins B and C. Over the past several decades, the vaccine has progressed from initial observations through pre-clinical studies to cGMP manufacture and clinical evaluations. The Invaplex product maintains unique biological properties associated with the invasiveness of virulent shigellae and also presents both serotype-specific epitopes, as well as highly conserved invasin protein epitopes, to the immunized host. The vaccine product has evolved from a native product isolated from wild-type shigellae (native Invaplex) to a more defined vaccine produced from purified LPS and recombinant IpaB and IpaC (artificial Invaplex). Each successive “generation” of the vaccine is derived from earlier versions, resulting in improved immunogenicity, homogeneity and effectiveness. The current vaccine, detoxified artificial Invaplex (InvaplexAR-Detox), was developed for parenteral administration by incorporating LPS with under-acylated lipid A. InvaplexAR-Detox has demonstrated an excellent safety and immunogenicity profile in initial clinical studies and is advancing toward evaluations in the target populations of children and travelers to endemic countries.
Collapse
|
21
|
Palit P, Chowdhury FT, Baruah N, Sarkar B, Mou SN, Kamal M, Siddiqua TJ, Noor Z, Ahmed T. A Comprehensive Computational Investigation into the Conserved Virulent Proteins of Shigella species Unveils Potential Small-Interfering RNA Candidates as a New Therapeutic Strategy against Shigellosis. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27061936. [PMID: 35335300 PMCID: PMC8950558 DOI: 10.3390/molecules27061936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/19/2021] [Accepted: 12/27/2021] [Indexed: 11/16/2022]
Abstract
Shigella species account for the second-leading cause of deaths due to diarrheal diseases among children of less than 5 years of age. The emergence of multi-drug-resistant Shigella isolates and the lack of availability of Shigella vaccines have led to the pertinence in the efforts made for the development of new therapeutic strategies against shigellosis. Consequently, designing small-interfering RNA (siRNA) candidates against such infectious agents represents a novel approach to propose new therapeutic candidates to curb the rampant rise of anti-microbial resistance in such pathogens. In this study, we analyzed 264 conserved sequences from 15 different conserved virulence genes of Shigella sp., through extensive rational validation using a plethora of first-generation and second-generation computational algorithms for siRNA designing. Fifty-eight siRNA candidates were obtained by using the first-generation algorithms, out of which only 38 siRNA candidates complied with the second-generation rules of siRNA designing. Further computational validation showed that 16 siRNA candidates were found to have a substantial functional efficiency, out of which 11 siRNA candidates were found to be non-immunogenic. Finally, three siRNA candidates exhibited a sterically feasible three-dimensional structure as exhibited by parameters of nucleic acid geometry such as: the probability of wrong sugar puckers, bad backbone confirmations, bad bonds, and bad angles being within the accepted threshold for stable tertiary structure. Although the findings of our study require further wet-lab validation and optimization for therapeutic use in the treatment of shigellosis, the computationally validated siRNA candidates are expected to suppress the expression of the virulence genes, namely: IpgD (siRNA 9) and OspB (siRNA 15 and siRNA 17) and thus act as a prospective tool in the RNA interference (RNAi) pathway. However, the findings of our study require further wet-lab validation and optimization for regular therapeutic use for treatment of shigellosis.
Collapse
Affiliation(s)
- Parag Palit
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh; (P.P.); (M.K.); (T.J.S.); (T.A.)
| | - Farhana Tasnim Chowdhury
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka 1000, Bangladesh; (F.T.C.); (B.S.); (S.N.M.)
| | - Namrata Baruah
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, Uttar Pradesh, India;
| | - Bonoshree Sarkar
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka 1000, Bangladesh; (F.T.C.); (B.S.); (S.N.M.)
| | - Sadia Noor Mou
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka 1000, Bangladesh; (F.T.C.); (B.S.); (S.N.M.)
| | - Mehnaz Kamal
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh; (P.P.); (M.K.); (T.J.S.); (T.A.)
| | - Towfida Jahan Siddiqua
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh; (P.P.); (M.K.); (T.J.S.); (T.A.)
| | - Zannatun Noor
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh; (P.P.); (M.K.); (T.J.S.); (T.A.)
- Correspondence:
| | - Tahmeed Ahmed
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka 1212, Bangladesh; (P.P.); (M.K.); (T.J.S.); (T.A.)
| |
Collapse
|
22
|
Efficient production of immunologically active Shigella invasion plasmid antigens IpaB and IpaH using a cell-free expression system. Appl Microbiol Biotechnol 2021; 106:401-414. [PMID: 34932164 PMCID: PMC8688910 DOI: 10.1007/s00253-021-11701-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/15/2021] [Accepted: 11/18/2021] [Indexed: 11/16/2022]
Abstract
Abstract Shigella spp. invade the colonic epithelium and cause bacillary dysentery in humans. Individuals living in areas that lack access to clean water and sanitation are the most affected. Even though infection can be treated with antibiotics, Shigella antimicrobial drug resistance complicates clinical management. Despite decades of effort, there are no licensed vaccines to prevent shigellosis. The highly conserved invasion plasmid antigens (Ipa), which are components of the Shigella type III secretion system, participate in bacterial epithelial cell invasion and have been pursued as vaccine targets. However, expression and purification of these proteins in conventional cell-based systems have been challenging due to solubility issues and extremely low recovery yields. These difficulties have impeded manufacturing and clinical advancement. In this study, we describe a new method to express Ipa proteins using the Xpress+TM cell-free protein synthesis (CFPS) platform. Both IpaB and the C-terminal domain of IpaH1.4 (IpaH-CTD) were efficiently produced with this technology at yields > 200 mg/L. Furthermore, the expression was linearly scaled in a bioreactor under controlled conditions, and proteins were successfully purified using multimode column chromatography to > 95% purity as determined by SDS-PAGE. Biophysical characterization of the cell-free synthetized IpaB and IpaH-CTD using SEC-MALS analysis showed well-defined oligomeric states of the proteins in solution. Functional analysis revealed similar immunoreactivity as compared to antigens purified from E. coli. These results demonstrate the efficiency of CFPS for Shigella protein production; the practicality and scalability of this method will facilitate production of antigens for Shigella vaccine development and immunological analysis. Key points • First report of Shigella IpaB and IpaH produced at high purity and yield using CFPS • CFPS-IpaB and IpaH perform similarly to E. coli–produced proteins in immunoassays • CFPS-IpaB and IpaH react with Shigella-specific human antibodies and are immunogenic in mice. Graphical abstract ![]()
Collapse
|
23
|
Lu T, Das S, Howlader DR, Zheng Q, Ratnakaram SSK, Whittier SK, Picking WD, Picking WL. L-DBF Elicits Cross Protection Against Different Serotypes of Shigella spp. FRONTIERS IN TROPICAL DISEASES 2021. [DOI: 10.3389/fitd.2021.729731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Shigellosis is a severe diarrheal disease caused by members of the genus Shigella, with at least 80 million cases and 700,000 deaths annually around the world. The type III secretion system (T3SS) is the primary virulence factor used by the shigellae, and we have previously demonstrated that vaccination with the type T3SS proteins IpaB and IpaD, along with an IpaD/IpaB fusion protein (DBF), protects mice from Shigella infection in a lethal pulmonary model. To simplify the formulation and development of the DBF Shigella vaccine, we have genetically fused LTA1, the active subunit of heat-labile toxin from enterotoxigenic E. coli, with DBF to produce the self-adjuvanting antigen L-DBF. Here we immunized mice with L-DBF via the intranasal, intramuscular, and intradermal routes and challenged them with a lethal dose of S. flexneri 2a. While none of the mice vaccinated intramuscularly or intradermally were protected, mice vaccinated with L-DBF intranasally were protected from lethal challenges with S. flexneri 2a, S. flexneri 1b, S. flexneri 3a, S. flexneri 6, and S. sonnei. Intranasal L-DBF induced both B cell and T cell responses that correlated with protection against Shigella infection. Our results suggest that L-DBF is a candidate for developing an effective serotype-independent vaccine against Shigella spp.
Collapse
|
24
|
Baruah N, Ahamad N, Maiti S, Howlader DR, Bhaumik U, Patil VV, Chakrabarti MK, Koley H, Katti DS. Development of a Self-Adjuvanting, Cross-Protective, Stable Intranasal Recombinant Vaccine for Shigellosis. ACS Infect Dis 2021; 7:3182-3196. [PMID: 34734708 DOI: 10.1021/acsinfecdis.1c00345] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
With the acquirement of antibiotic resistance, Shigella has resulted in multiple epidemics of shigellosis, an infectious diarrheal disease, causing thousands of deaths per year. Unfortunately, there are no licensed vaccines, primarily due to low or serotype-specific immunogenicity. Thus, conserved subunit vaccines utilizing recombinant invasion plasmid antigens (Ipa) have been explored as cross-protective vaccine candidates. However, achieving cross-protection against Shigella dysenteriae 1, which caused multiple pandemics/epidemics in the recent past, has been difficult. Therefore, a rational approach to improve cross-protection in the preparation for a possible pandemic should involve conserved proteins from S. dysenteriae 1 (Sd1). IpaC is one such conserved immunogenic protein that is less explored as an independent vaccine due to its instability/aggregation. Therefore, to improve cross-protection and potential immunogenicity and to be prepared for a future epidemic/pandemic, herein, we stabilized recombinant Sd1 IpaC, expressed without its chaperone, using a previously reported stabilizing detergent (LDAO) in a modified protocol and assessed its vaccine potential without an adjuvant. The protein assembled into heterogeneous complex spherical structures in the presence of LDAO and showed improved stability at storage temperatures of -80, -20, 4, 25, and 37 °C while providing enhanced yield and concentration. The protein could also be stably lyophilized and reconstituted, increasing the convenience of transportation and storage. Upon intranasal administration in BALB/c mice, the stabilized-IpaC-immunized groups generated significant antibody response and were not only protected against a high intraperitoneal dose of homologous S. dysenteriae 1 but also showed 100% survival against heterologous Shigella flexneri 2a without an adjuvant, while the control animals showed visible diarrhea (bloody-Sd1 challenge), lethargy, and weight loss with 0% survival. Overall, this work demonstrates that stabilized IpaC can be explored as a minimalist, self-adjuvanting, cross-protective, intranasal, single-antigen Shigella vaccine.
Collapse
Affiliation(s)
- Namrata Baruah
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
- The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
| | - Nadim Ahamad
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
| | - Suhrid Maiti
- Division of Bacteriology, ICMR-National Institute of Cholera & Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata 700010, West Bengal, India
| | - Debaki R. Howlader
- Division of Bacteriology, ICMR-National Institute of Cholera & Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata 700010, West Bengal, India
| | - Ushasi Bhaumik
- Division of Bacteriology, ICMR-National Institute of Cholera & Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata 700010, West Bengal, India
| | - Vinod V. Patil
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
| | - Manoj K. Chakrabarti
- Division of Bacteriology, ICMR-National Institute of Cholera & Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata 700010, West Bengal, India
| | - Hemanta Koley
- Division of Bacteriology, ICMR-National Institute of Cholera & Enteric Diseases, P-33 CIT Road, Scheme-XM, Beliaghata, Kolkata 700010, West Bengal, India
| | - Dhirendra S. Katti
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
- The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
| |
Collapse
|
25
|
Howlader DR, Das S, Lu T, Hu G, Varisco DJ, Dietz ZK, Walton SP, Ratnakaram SSK, Gardner FM, Ernst RK, Picking WD, Picking WL. Effect of Two Unique Nanoparticle Formulations on the Efficacy of a Broadly Protective Vaccine Against Pseudomonas Aeruginosa. Front Pharmacol 2021; 12:706157. [PMID: 34483911 PMCID: PMC8416447 DOI: 10.3389/fphar.2021.706157] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 08/04/2021] [Indexed: 11/23/2022] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen responsible for a wide range of infections in humans. In addition to its innate antibiotic resistance, P. aeruginosa is very effective in acquiring resistance resulting in the emergence of multi-drug resistance strains and a licensed vaccine is not yet available. We have previously demonstrated the protective efficacy of a novel antigen PaF (Pa Fusion), a fusion of the type III secretion system (T3SS) needle tip protein, PcrV, and the first of two translocator proteins, PopB. PaF was modified to provide a self-adjuvanting activity by fusing the A1 subunit of the heat-labile enterotoxin from Enterotoxigenic E. coli to its N-terminus to give L-PaF. In addition to providing protection against 04 and 06 serotypes of P. aeruginosa, L-PaF elicited opsonophagocytic killing and stimulated IL-17A secretion, which have been predicted to be required for a successful vaccine. While monomeric recombinant subunit vaccines can be protective in mice, this protection often does not transfer to humans where multimeric formulations perform better. Here, we use two unique formulations, an oil-in-water (o/w) emulsion and a chitosan particle, as well as the addition of a unique TLR4 agonist, BECC438 (a detoxified lipid A analogue designated Bacterial Enzymatic Combinatorial Chemistry 438), as an initial step in optimizing L-PaF for use in humans. The o/w emulsion together with BECC438 provided the best protective efficacy, which correlated with high levels of opsonophagocytic killing and IL-17A secretion, thereby reducing the lung burden among all the vaccinated groups tested.
Collapse
Affiliation(s)
- Debaki R Howlader
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
| | - Sayan Das
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
| | - Ti Lu
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
| | - Gang Hu
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
| | - David J Varisco
- Department of Microbial Pathogenesis, University of Maryland, Baltimore, MD, United States
| | - Zackary K Dietz
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
| | - Sierra P Walton
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
| | | | - Francesca M Gardner
- Department of Microbial Pathogenesis, University of Maryland, Baltimore, MD, United States
| | - Robert K Ernst
- Department of Microbial Pathogenesis, University of Maryland, Baltimore, MD, United States
| | - William D Picking
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
| | - Wendy L Picking
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
| |
Collapse
|
26
|
Walker R, Kaminski RW, Porter C, Choy RKM, White JA, Fleckenstein JM, Cassels F, Bourgeois L. Vaccines for Protecting Infants from Bacterial Causes of Diarrheal Disease. Microorganisms 2021; 9:1382. [PMID: 34202102 PMCID: PMC8303436 DOI: 10.3390/microorganisms9071382] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/16/2021] [Accepted: 06/21/2021] [Indexed: 12/22/2022] Open
Abstract
The global diarrheal disease burden for Shigella, enterotoxigenic Escherichia coli (ETEC), and Campylobacter is estimated to be 88M, 75M, and 75M cases annually, respectively. A vaccine against this target trio of enteric pathogens could address about one-third of diarrhea cases in children. All three of these pathogens contribute to growth stunting and have demonstrated increasing resistance to antimicrobial agents. Several combinations of antigens are now recognized that could be effective for inducing protective immunity against each of the three target pathogens in a single vaccine for oral administration or parenteral injection. The vaccine combinations proposed here would result in a final product consistent with the World Health Organization's (WHO) preferred product characteristics for ETEC and Shigella vaccines, and improve the vaccine prospects for support from Gavi, the Vaccine Alliance, and widespread uptake by low- and middle-income countries' (LMIC) public health stakeholders. Broadly protective antigens will enable multi-pathogen vaccines to be efficiently developed and cost-effective. This review describes how emerging discoveries for each pathogen component of the target trio could be used to make vaccines, which could help reduce a major cause of poor health, reduced cognitive development, lost economic productivity, and poverty in many parts of the world.
Collapse
Affiliation(s)
- Richard Walker
- Center for Vaccine Innovation and Access, PATH, Washington, DC 20001, USA;
| | - Robert W. Kaminski
- Department of Diarrheal Disease Research, Walter Reed Institute of Research, Silver Spring, MD 20910, USA;
| | - Chad Porter
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD 20910, USA;
| | - Robert K. M. Choy
- Center for Vaccine Innovation and Access, PATH, San Francisco, CA 94108, USA;
| | - Jessica A. White
- Center for Vaccine Innovation and Access, PATH, Seattle, WA 98121, USA; (J.A.W.); (F.C.)
| | - James M. Fleckenstein
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA;
- Medicine Service, Saint Louis VA Health Care System, St. Louis, MO 63106, USA
| | - Fred Cassels
- Center for Vaccine Innovation and Access, PATH, Seattle, WA 98121, USA; (J.A.W.); (F.C.)
| | - Louis Bourgeois
- Center for Vaccine Innovation and Access, PATH, Washington, DC 20001, USA;
| |
Collapse
|
27
|
Bazhenova A, Gao F, Bolgiano B, Harding SE. Glycoconjugate vaccines against Salmonella enterica serovars and Shigella species: existing and emerging methods for their analysis. Biophys Rev 2021; 13:221-246. [PMID: 33868505 PMCID: PMC8035613 DOI: 10.1007/s12551-021-00791-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 02/25/2021] [Indexed: 12/26/2022] Open
Abstract
The global spread of enteric disease, the increasingly limited options for antimicrobial treatment and the need for effective eradication programs have resulted in an increased demand for glycoconjugate enteric vaccines, made with carbohydrate-based membrane components of the pathogen, and their precise characterisation. A set of physico-chemical and immunological tests are employed for complete vaccine characterisation and to ensure their consistency, potency, safety and stability, following the relevant World Health Organization and Pharmacopoeia guidelines. Variable requirements for analytical methods are linked to conjugate structure, carrier protein nature and size and O-acetyl content of polysaccharide. We investigated a key stability-indicating method which measures the percent free saccharide of Salmonella enterica subspecies enterica serovar Typhi capsular polysaccharide, by detergent precipitation, depolymerisation and HPAEC-PAD quantitation. Together with modern computational approaches, a more precise design of glycoconjugates is possible, allowing for improvements in solubility, structural conformation and stability, and immunogenicity of antigens, which may be applicable to a broad spectrum of vaccines. More validation experiments are required to establish the most effective and suitable methods for glycoconjugate analysis to bring uniformity to the existing protocols, although the need for product-specific approaches will apply, especially for the more complex vaccines. An overview of current and emerging analytical approaches for the characterisation of vaccines against Salmonella Typhi and Shigella species is described in this paper. This study should aid the development and licensing of new glycoconjugate vaccines aimed at the prevention of enteric diseases.
Collapse
Affiliation(s)
- Aleksandra Bazhenova
- School of Biosciences, University of Nottingham, Sutton Bonington, Loughborough, LE12 5RD UK
| | - Fang Gao
- Division of Bacteriology, National Institute for Biological Standards and Control (NIBSC), Blanche Lane, South Mimms, Potters Bar, EN6 3QG UK
| | - Barbara Bolgiano
- Division of Bacteriology, National Institute for Biological Standards and Control (NIBSC), Blanche Lane, South Mimms, Potters Bar, EN6 3QG UK
| | - Stephen E. Harding
- School of Biosciences, University of Nottingham, Sutton Bonington, Loughborough, LE12 5RD UK
- Museum of Cultural History, University of Oslo, Postboks 6762 St. Olavs plass, 0130 Oslo, Norway
| |
Collapse
|
28
|
The Shigella Type III Secretion System: An Overview from Top to Bottom. Microorganisms 2021; 9:microorganisms9020451. [PMID: 33671545 PMCID: PMC7926512 DOI: 10.3390/microorganisms9020451] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/16/2021] [Accepted: 02/18/2021] [Indexed: 12/16/2022] Open
Abstract
Shigella comprises four species of human-restricted pathogens causing bacillary dysentery. While Shigella possesses multiple genetic loci contributing to virulence, a type III secretion system (T3SS) is its primary virulence factor. The Shigella T3SS nanomachine consists of four major assemblies: the cytoplasmic sorting platform; the envelope-spanning core/basal body; an exposed needle; and a needle-associated tip complex with associated translocon that is inserted into host cell membranes. The initial subversion of host cell activities is carried out by the effector functions of the invasion plasmid antigen (Ipa) translocator proteins, with the cell ultimately being controlled by dedicated effector proteins that are injected into the host cytoplasm though the translocon. Much of the information now available on the T3SS injectisome has been accumulated through collective studies on the T3SS from three systems, those of Shigella flexneri, Salmonella typhimurium and Yersinia enterocolitica/Yersinia pestis. In this review, we will touch upon the important features of the T3SS injectisome that have come to light because of research in the Shigella and closely related systems. We will also briefly highlight some of the strategies being considered to target the Shigella T3SS for disease prevention.
Collapse
|
29
|
Hotinger JA, Pendergrass HA, May AE. Molecular Targets and Strategies for Inhibition of the Bacterial Type III Secretion System (T3SS); Inhibitors Directly Binding to T3SS Components. Biomolecules 2021; 11:biom11020316. [PMID: 33669653 PMCID: PMC7922566 DOI: 10.3390/biom11020316] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 01/01/2023] Open
Abstract
The type III secretion system (T3SS) is a virulence apparatus used by many Gram-negative pathogenic bacteria to cause infections. Pathogens utilizing a T3SS are responsible for millions of infections yearly. Since many T3SS knockout strains are incapable of causing systemic infection, the T3SS has emerged as an attractive anti-virulence target for therapeutic design. The T3SS is a multiprotein molecular syringe that enables pathogens to inject effector proteins into host cells. These effectors modify host cell mechanisms in a variety of ways beneficial to the pathogen. Due to the T3SS’s complex nature, there are numerous ways in which it can be targeted. This review will be focused on the direct targeting of components of the T3SS, including the needle, translocon, basal body, sorting platform, and effector proteins. Inhibitors will be considered a direct inhibitor if they have a binding partner that is a T3SS component, regardless of the inhibitory effect being structural or functional.
Collapse
|
30
|
Das S, Howlader DR, Zheng Q, Ratnakaram SSK, Whittier SK, Lu T, Keith JD, Picking WD, Birket SE, Picking WL. Development of a Broadly Protective, Self-Adjuvanting Subunit Vaccine to Prevent Infections by Pseudomonas aeruginosa. Front Immunol 2020; 11:583008. [PMID: 33281815 PMCID: PMC7705240 DOI: 10.3389/fimmu.2020.583008] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 10/16/2020] [Indexed: 12/19/2022] Open
Abstract
Infections caused by the opportunistic pathogen Pseudomonas aeruginosa can be difficult to treat due to innate and acquired antibiotic resistance and this is exacerbated by the emergence of multi-drug resistant strains. Unfortunately, no licensed vaccine yet exists to prevent Pseudomonas infections. Here we describe a novel subunit vaccine that targets the P. aeruginosa type III secretion system (T3SS). This vaccine is based on the novel antigen PaF (Pa Fusion), a fusion of the T3SS needle tip protein, PcrV, and the first of two translocator proteins, PopB. Additionally, PaF is made self-adjuvanting by the N-terminal fusion of the A1 subunit of the mucosal adjuvant double-mutant heat-labile enterotoxin (dmLT). Here we show that this triple fusion, designated L-PaF, can activate dendritic cells in vitro and elicits strong IgG and IgA titers in mice when administered intranasally. This self-adjuvanting vaccine expedites the clearance of P. aeruginosa from the lungs of challenged mice while stimulating host expression of IL-17A, which may be important for generating a protective immune response in humans. L-PaF's protective capacity was recapitulated in a rat pneumonia model, further supporting the efficacy of this novel fusion vaccine.
Collapse
Affiliation(s)
- Sayan Das
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Kansas, Lawrence, KS, United States
| | - Debaki R Howlader
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Kansas, Lawrence, KS, United States
| | - Qi Zheng
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Kansas, Lawrence, KS, United States
| | - Siva Sai Kumar Ratnakaram
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Kansas, Lawrence, KS, United States
| | - Sean K Whittier
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Kansas, Lawrence, KS, United States.,Hafion LLC, Lawrence, KS, United States
| | - Ti Lu
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Kansas, Lawrence, KS, United States
| | - Johnathan D Keith
- Department of Medicine and Gregory Fleming James Cystic Fibrosis Research Center, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - William D Picking
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Kansas, Lawrence, KS, United States
| | - Susan E Birket
- Department of Medicine and Gregory Fleming James Cystic Fibrosis Research Center, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Wendy L Picking
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Kansas, Lawrence, KS, United States.,Hafion LLC, Lawrence, KS, United States
| |
Collapse
|
31
|
Harutyunyan S, Neuhauser I, Mayer A, Aichinger M, Szijártó V, Nagy G, Nagy E, Girardi P, Malinoski FJ, Henics T. Characterization of ShigETEC, a Novel Live Attenuated Combined Vaccine against Shigellae and ETEC. Vaccines (Basel) 2020; 8:vaccines8040689. [PMID: 33207794 PMCID: PMC7712393 DOI: 10.3390/vaccines8040689] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 11/16/2022] Open
Abstract
Background: Shigella spp. and enterotoxigenic Escherichia coli (ETEC) remain the two leading bacterial causes of diarrheal diseases worldwide. Attempts to develop preventive vaccines against Shigella and ETEC have not yet been successful. The major challenge for a broad Shigella vaccine is the serotype-specific immune response to the otherwise protective LPS O-antigen. ETEC vaccines mainly rely on the heat-labile enterotoxin (LT), while heat-stable toxin (ST) has also been shown to be an important virulence factor. Methods: We constructed a combined Shigella and ETEC vaccine (ShigETEC) based on a live attenuated Shigella strain rendered rough and non-invasive with heterologous expression of two ETEC antigens, LTB and a detoxified version of ST (STN12S). This new vaccine strain was characterized and tested for immunogenicity in relevant animal models. Results: Immunization with ShigETEC resulted in serotype independent protection in the mouse lung shigellosis model and induced high titer IgG and IgA antibodies against bacterial lysates, and anti-ETEC toxin antibodies with neutralizing capacity. Conclusions: ShigETEC is a promising oral vaccine candidate against Shigella and ETEC infections and currently in Phase 1 testing.
Collapse
Affiliation(s)
- Shushan Harutyunyan
- Eveliqure Biotechnologies GmbH, Karl-Farkas-Gasse 22, 1030 Vienna, Austria; (S.H.); (I.N.); (A.M.); (M.A.); (V.S.); (G.N.); (E.N.); (P.G.); (F.J.M.)
| | - Irene Neuhauser
- Eveliqure Biotechnologies GmbH, Karl-Farkas-Gasse 22, 1030 Vienna, Austria; (S.H.); (I.N.); (A.M.); (M.A.); (V.S.); (G.N.); (E.N.); (P.G.); (F.J.M.)
| | - Alexandra Mayer
- Eveliqure Biotechnologies GmbH, Karl-Farkas-Gasse 22, 1030 Vienna, Austria; (S.H.); (I.N.); (A.M.); (M.A.); (V.S.); (G.N.); (E.N.); (P.G.); (F.J.M.)
| | - Michael Aichinger
- Eveliqure Biotechnologies GmbH, Karl-Farkas-Gasse 22, 1030 Vienna, Austria; (S.H.); (I.N.); (A.M.); (M.A.); (V.S.); (G.N.); (E.N.); (P.G.); (F.J.M.)
| | - Valéria Szijártó
- Eveliqure Biotechnologies GmbH, Karl-Farkas-Gasse 22, 1030 Vienna, Austria; (S.H.); (I.N.); (A.M.); (M.A.); (V.S.); (G.N.); (E.N.); (P.G.); (F.J.M.)
| | - Gábor Nagy
- Eveliqure Biotechnologies GmbH, Karl-Farkas-Gasse 22, 1030 Vienna, Austria; (S.H.); (I.N.); (A.M.); (M.A.); (V.S.); (G.N.); (E.N.); (P.G.); (F.J.M.)
- CEBINA GmbH, Karl-Farkas-Gasse 22, 1030 Vienna, Austria
| | - Eszter Nagy
- Eveliqure Biotechnologies GmbH, Karl-Farkas-Gasse 22, 1030 Vienna, Austria; (S.H.); (I.N.); (A.M.); (M.A.); (V.S.); (G.N.); (E.N.); (P.G.); (F.J.M.)
- CEBINA GmbH, Karl-Farkas-Gasse 22, 1030 Vienna, Austria
| | - Petra Girardi
- Eveliqure Biotechnologies GmbH, Karl-Farkas-Gasse 22, 1030 Vienna, Austria; (S.H.); (I.N.); (A.M.); (M.A.); (V.S.); (G.N.); (E.N.); (P.G.); (F.J.M.)
| | - Frank J. Malinoski
- Eveliqure Biotechnologies GmbH, Karl-Farkas-Gasse 22, 1030 Vienna, Austria; (S.H.); (I.N.); (A.M.); (M.A.); (V.S.); (G.N.); (E.N.); (P.G.); (F.J.M.)
| | - Tamás Henics
- Eveliqure Biotechnologies GmbH, Karl-Farkas-Gasse 22, 1030 Vienna, Austria; (S.H.); (I.N.); (A.M.); (M.A.); (V.S.); (G.N.); (E.N.); (P.G.); (F.J.M.)
- Correspondence: ; Tel.: +43-1-909-2208-3313
| |
Collapse
|
32
|
Abstract
Enteric viral and bacterial infections continue to be a leading cause of mortality and morbidity in young children in low-income and middle-income countries, the elderly, and immunocompromised individuals. Vaccines are considered an effective and practical preventive approach against the predominantly fecal-to-oral transmitted gastroenteritis particularly in the resource-limited countries or regions where implementation of sanitation systems and supply of safe drinking water are not quickly achievable. While vaccines are available for a few enteric pathogens including rotavirus and cholera, there are no vaccines licensed for many other enteric viral and bacterial pathogens. Challenges in enteric vaccine development include immunological heterogeneity among pathogen strains or isolates, a lack of animal challenge models to evaluate vaccine candidacy, undefined host immune correlates to protection, and a low protective efficacy among young children in endemic regions. In this article, we briefly updated the progress and challenges in vaccines and vaccine development for the leading enteric viral and bacterial pathogens including rotavirus, human calicivirus, Shigella, enterotoxigenic Escherichia coli (ETEC), cholera, nontyphoidal Salmonella, and Campylobacter, and introduced a novel epitope- and structure-based vaccinology platform known as MEFA (multiepitope fusion antigen) and the application of MEFA for developing broadly protective multivalent vaccines against heterogenous pathogens.
Collapse
Affiliation(s)
- Hyesuk Seo
- University of Illinois at Urbana-Champaign, Department of Pathobiology, Urbana, Illinois, USA
| | - Qiangde Duan
- University of Yangzhou, Institute of Comparative Medicine, Yangzhou, PR China
| | - Weiping Zhang
- University of Illinois at Urbana-Champaign, Department of Pathobiology, Urbana, Illinois, USA,CONTACT Weiping Zhang, University of Illinois at Urbana-Champaign, Department of Pathobiology, Urbana, Illinois, USA
| |
Collapse
|
33
|
Jain A, Hu G, Kumar Ratnakaram SS, Johnson DK, Picking WD, Picking WL, Middaugh CR. Preformulation Characterization and the Effect of Ionic Excipients on the Stability of a Novel DB Fusion Protein. J Pharm Sci 2020; 110:108-123. [PMID: 32916136 PMCID: PMC7750262 DOI: 10.1016/j.xphs.2020.09.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 09/04/2020] [Accepted: 09/04/2020] [Indexed: 12/17/2022]
Abstract
Shigella ssp cause bacillary dysentery (shigellosis) which has high global morbidity in young children and the elderly. The virulence of Shigella relies upon a type III secretion system (T3SS) which injects host altering effector proteins into targeted intestinal cells. The Shigella T3SS contains two components, invasion plasmid antigen D (IpaD) and invasion plasmid antigen B (IpaB), that were previously identified as broadly protective antigens. When IpaD and IpaB were co-expressed to give the DB fusion (DBF) protein, vaccine efficacy was further improved. Biophysical characterization under various pH conditions showed that DBF is most stable at pH 7 and 8 and loses its conformational integrity at 48 and 50 °C respectively. Forced degradation studies revealed significant effects on the secondary structure, tertiary structure and conformational stability of DBF. In the presence of phosphate buffers as well as other anionic excipients, DBF demonstrated a concentration dependent conformational stabilization. Molecular docking revealed potential polyanion binding sites in DBF that may interact with phytic acid. These sites can be exploited to stabilize the DBF protein. This work highlights potential destabilizing and stabilizing factors, which not only improves our understanding of the DBF protein but helps in future development of a stable Shigella vaccine.
Collapse
Affiliation(s)
- Akshay Jain
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS 66047, USA; Macromolecule and Vaccine Stabilization Center, University of Kansas, Lawrence, KS 66047, USA
| | - Gang Hu
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS 66047, USA; Macromolecule and Vaccine Stabilization Center, University of Kansas, Lawrence, KS 66047, USA
| | | | - David K Johnson
- Computational Chemical Biology Laboratory, University of Kansas, Lawrence, KS 66047, USA
| | - William D Picking
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS 66047, USA
| | - Wendy L Picking
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS 66047, USA.
| | - Charles Russell Middaugh
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS 66047, USA; Macromolecule and Vaccine Stabilization Center, University of Kansas, Lawrence, KS 66047, USA.
| |
Collapse
|
34
|
Sawa T, Kinoshita M, Inoue K, Ohara J, Moriyama K. Immunoglobulin for Treating Bacterial Infections: One More Mechanism of Action. Antibodies (Basel) 2019; 8:antib8040052. [PMID: 31684203 PMCID: PMC6963986 DOI: 10.3390/antib8040052] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 10/17/2019] [Accepted: 10/28/2019] [Indexed: 02/08/2023] Open
Abstract
The mechanisms underlying the effects of immunoglobulins on bacterial infections are thought to involve bacterial cell lysis via complement activation, phagocytosis via bacterial opsonization, toxin neutralization, and antibody-dependent cell-mediated cytotoxicity. Nevertheless, recent advances in the study of the pathogenicity of Gram-negative bacteria have raised the possibility of an association between immunoglobulin and bacterial toxin secretion. Over time, new toxin secretion systems like the type III secretion system have been discovered in many pathogenic Gram-negative bacteria. With this system, the bacterial toxins are directly injected into the cytoplasm of the target cell through a special secretory apparatus without any exposure to the extracellular environment, and therefore with no opportunity for antibodies to neutralize the toxin. However, antibodies against the V-antigen, which is located on the needle-shaped tip of the bacterial secretion apparatus, can inhibit toxin translocation, thus raising the hope that the toxin may be susceptible to antibody targeting. Because multi-drug resistant bacteria are now prevalent, inhibiting this secretion mechanism is an attractive alternative or adjunctive therapy against lethal bacterial infections. Thus, it is not unreasonable to define the blocking effect of anti-V-antigen antibodies as the fifth mechanism for immunoglobulin action against bacterial infections.
Collapse
Affiliation(s)
- Teiji Sawa
- Department of Anesthesiology, School of Medicine, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan.
| | - Mao Kinoshita
- Department of Anesthesiology, School of Medicine, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan.
| | - Keita Inoue
- Department of Anesthesiology, School of Medicine, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan.
| | - Junya Ohara
- Department of Anesthesiology, School of Medicine, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan.
| | - Kiyoshi Moriyama
- Department of Anesthesiology, Kyorin University School of Medicine, Tokyo 181-8611, Japan.
| |
Collapse
|
35
|
In-silico design and production of a novel antigenic chimeric Shigella IpaB fused to C-terminal of Clostridium perfringens enterotoxin. Mol Biol Rep 2019; 46:6105-6115. [PMID: 31473892 DOI: 10.1007/s11033-019-05046-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 08/27/2019] [Indexed: 10/26/2022]
Abstract
The emergence of antibiotic-resistant phenotypes in Shigella serotypes and the high mortality rate, approximately one million dead annually, in affected patients announce a global demand for an effective serotype-independent vaccine against Shigella. This study aims to design, express, and purify a novel chimeric protein, as a serotype-independent vaccine candidate against Shigella containing full-length Shigella invasion plasmid antigen B (IpaB) and a C-terminal fragment (residues 194-319) of Clostridium perfringens enterotoxin (C-CPE) as a mucosal adjuvant. Several online databases and bioinformatics software were utilized to design the chimeric protein and the relative recombinant gene. The recombinant gene encoding IpaB-CPE194-319 was synthesized, cloned into pACYCDuet-1 expression vector, and transferred to E. coli Bl21 (DE3) cells. IpaB-CPE194-319 was then expressed in auto-induction medium, purified and characterized using MALDI-TOF-TOF mass spectrometry. Followed by subcutaneous injection of the purified IpaB-CPE194-319 to BALB/c mice, antigenicity of this chimeric protein was determined through performing dot-blot immunoassay on nitrocellulose membrane using mice sera. The outcomes of this study show the successful design, efficient expression, and purification of IpaB-CPE194-319 divalent chimeric protein under mentioned conditions. The obtained results also demonstrate the intrinsic antigenic property of IpaB-CPE194-319.
Collapse
|
36
|
Seo H, Lu T, Mani S, Bourgeois AL, Walker R, Sack DA, Zhang W. Adjuvant effect of enterotoxigenic Escherichia coli (ETEC) double-mutant heat-labile toxin (dmLT) on systemic immunogenicity induced by the CFA/I/II/IV MEFA ETEC vaccine: Dose-related enhancement of antibody responses to seven ETEC adhesins (CFA/I, CS1-CS6). Hum Vaccin Immunother 2019; 16:419-425. [PMID: 31361177 PMCID: PMC7062417 DOI: 10.1080/21645515.2019.1649555] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Double-mutant heat-labile toxin (dmLT, LTR192G/L211A) of enterotoxigenic Escherichia coli (ETEC) is an effective mucosal adjuvant. Recent studies have shown that dmLT also exhibits adjuvanticity for antigens administered parenterally. In this study, we subcutaneously (SC) immunized mice with the ETEC adhesin-based vaccine, CFA/I/II/IV MEFA (multiepitope fusion antigen), adjuvanted with dmLT and examined the impact of dmLT on antibody responses specific to the seven adhesins in the vaccine construction [CFA/I, CFA/II (CS1, CS2, CS3) and CFA/IV (CS4, CS5, CS6)]. Mice were immunized with a fixed dose of CFA/I/II/IV MEFA and ascending doses of dmLT adjuvant (0, 0.05, 0.1, 0.5 or 1.0 µg) to assess the potential dmLT dose response relationship. Data showed that dmLT enhanced systemic antibody responses to all seven antigens (CFA/I, CS1-CS6) targeted by MEFA in a dose-dependent way. The adjuvant effect of dmLT on the MEFA construct plateaued at a dose of 0.1 µg. Results also indicated that dmLT is an effective parenteral adjuvant when given by the SC route with the ETEC adhesin MEFA vaccine and that antibody enhancement was achieved with relatively low doses. These observations suggest the potential usefulness of dmLT for parenteral ETEC vaccine candidates and also perhaps for vaccines against other pathogens.
Collapse
Affiliation(s)
- Hyesuk Seo
- Diagnostic Medicine/Pathobiology Department, Kansas State University College of Veterinary Medicine, Manhattan, KS, USA.,Department of Pathobiology, University of Illinois at Urbana-Champaign, Illinois, Il, USA
| | - Ti Lu
- Diagnostic Medicine/Pathobiology Department, Kansas State University College of Veterinary Medicine, Manhattan, KS, USA
| | - Sachin Mani
- PATH, Center for Vaccine Innovation and Access, Washington, DC, USA
| | | | - Richard Walker
- PATH, Center for Vaccine Innovation and Access, Washington, DC, USA
| | - David A Sack
- Department of International Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Weiping Zhang
- Diagnostic Medicine/Pathobiology Department, Kansas State University College of Veterinary Medicine, Manhattan, KS, USA.,Department of Pathobiology, University of Illinois at Urbana-Champaign, Illinois, Il, USA
| |
Collapse
|
37
|
Arizmendi O, Kumar P, Zheng Q, Stewart JP, Picking WD, Picking W, Martinez-Becerra FJ. Vaccination With Mouse Dendritic Cells Loaded With an IpaD-IpaB Fusion Provides Protection Against Shigellosis. Front Immunol 2019; 10:192. [PMID: 30800131 PMCID: PMC6376248 DOI: 10.3389/fimmu.2019.00192] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 01/22/2019] [Indexed: 12/14/2022] Open
Abstract
Diarrheal diseases are a major cause of morbidity and mortality worldwide. They are most prevalent in settings with inadequate sanitation, poor hygiene and contaminated water. An important diarrheal pathogen in such settings is Shigella. No commercially available vaccine exists against shigellosis and immunity to the pathogen is serotype-restricted. We have previously shown that a polypeptide fusion of the Type Three Secretion Apparatus (T3SA) proteins IpaB and IpaD (named DBF) was efficacious as a vaccine against Shigella. Vaccination using different administration routes indicated that protection conferred by DBF did not fully correlate with antibodies. To define the immune responses involved in protection, we studied cellular responses to intranasal immunization with the DBF and the adjuvant dmLT. We found dendritic cell (DC) activation at the nasal associated lymphoid tissue (NALT). Activation markers CD86 and MHCII significantly increase in cells from immunized mice. Antigen exposure in vitro further confirmed the upregulation of CD80 and CD40 in primary dendritic cells. Animals immunized with antigen-primed dendritic cells were protected against Shigella infection, at levels comparable to the efficacy of immunization with the protein vaccine formulation. Therefore, we show that antigen-primed DCs are enough to provide immunity, and propose a mechanism of protection against Shigella spp. based on DC-mediated antigen presentation to T cells.
Collapse
Affiliation(s)
- Olivia Arizmendi
- Higuchi Biosciences Center, University of Kansas, Lawrence, KS, United States
| | - Prashant Kumar
- Higuchi Biosciences Center, University of Kansas, Lawrence, KS, United States
| | - Qi Zheng
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
| | - Jason P Stewart
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
| | - William D Picking
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
| | - Wendy Picking
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, United States
| | | |
Collapse
|
38
|
Kim MJ, Moon YH, Kim H, Rho S, Shin YK, Song M, Walker R, Czerkinsky C, Kim DW, Kim JO. Cross-Protective Shigella Whole-Cell Vaccine With a Truncated O-Polysaccharide Chain. Front Microbiol 2018; 9:2609. [PMID: 30429838 PMCID: PMC6220597 DOI: 10.3389/fmicb.2018.02609] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 10/12/2018] [Indexed: 11/13/2022] Open
Abstract
Shigella is a highly prevalent bacterium causing acute diarrhea and dysentery in developing countries. Shigella infections are treated with antibiotics but Shigellae are increasingly resistant to these drugs. Vaccination can be a countermeasure against emerging antibiotic-resistant shigellosis. Because of the structural variability in Shigellae O-antigen polysaccharides (Oag), cross-protective Shigella vaccines cannot be derived from single serotype-specific Oag. We created an attenuated Shigella flexneri 2a strain with one rather than multiple Oag units by disrupting the Oag polymerase gene (Δwzy), which broadened protective immunogenicity by exposing conserved surface proteins. Inactivated Δwzy mutant cells combined with Escherichia coli double mutant LT(R192G/L211A) as adjuvant, induced potent antibody responses to outer membrane protein PSSP-1, and type III secretion system proteins IpaB and IpaC. Intranasal immunization with the vaccine preparation elicited cross-protective immunity against S. flexneri 2a, S. flexneri 3a, S. flexneri 6, and Shigella sonnei in a mouse pneumonia model. Thus, S. flexneri 2a Δwzy represents a promising candidate strain for a universal Shigella vaccine.
Collapse
Affiliation(s)
- Min Jung Kim
- Clinical Research Lab, International Vaccine Institute, Seoul National University Research Park, Seoul, South Korea.,Laboratory of Molecular Pathology and Cancer Genomics, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Young-Hye Moon
- Clinical Research Lab, International Vaccine Institute, Seoul National University Research Park, Seoul, South Korea
| | - Heejoo Kim
- Clinical Research Lab, International Vaccine Institute, Seoul National University Research Park, Seoul, South Korea
| | - Semi Rho
- Clinical Research Lab, International Vaccine Institute, Seoul National University Research Park, Seoul, South Korea
| | - Young Kee Shin
- Laboratory of Molecular Pathology and Cancer Genomics, College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Manki Song
- Clinical Research Lab, International Vaccine Institute, Seoul National University Research Park, Seoul, South Korea
| | | | - Cecil Czerkinsky
- Clinical Research Lab, International Vaccine Institute, Seoul National University Research Park, Seoul, South Korea.,Institut de Pharmacologie Moléculaire & Cellulaire CNRS-INSERM-University of Nice Sophia Antipolis, Valbonne, France
| | - Dong Wook Kim
- Clinical Research Lab, International Vaccine Institute, Seoul National University Research Park, Seoul, South Korea
| | - Jae-Ouk Kim
- Clinical Research Lab, International Vaccine Institute, Seoul National University Research Park, Seoul, South Korea
| |
Collapse
|
39
|
Fleckenstein JM. Providing Structure to Enterotoxigenic Escherichia coli Vaccine Development. J Infect Dis 2018; 216:1-3. [PMID: 28541516 DOI: 10.1093/infdis/jix146] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 03/17/2017] [Indexed: 01/09/2023] Open
Affiliation(s)
- James M Fleckenstein
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine.,Medicine Service, Veterans Affairs Medical Center, St. Louis, Missouri
| |
Collapse
|
40
|
Huang J, Duan Q, Zhang W. Significance of Enterotoxigenic Escherichia coli (ETEC) Heat-Labile Toxin (LT) Enzymatic Subunit Epitopes in LT Enterotoxicity and Immunogenicity. Appl Environ Microbiol 2018; 84:e00849-18. [PMID: 29802193 PMCID: PMC6052278 DOI: 10.1128/aem.00849-18] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 05/19/2018] [Indexed: 01/28/2023] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) strains producing heat-labile toxin (LT) and/or heat-stable toxin (STa) are a top cause of children's diarrhea and travelers' diarrhea. Holotoxin-structured GM1-binding LT is a strong immunogen and an effective adjuvant, and can serve a carrier or a platform for multivalent vaccine development. However, the significance of peptide domains or epitopes of LT particularly enzymatic LTA subunit in association with LT enterotoxicity and immunogenicity has not been characterized. In this study, we identified B-cell epitopes in silico from LTA subunit and examined epitopes for immunogenicity and association with LT enterotoxicity. Epitopes identified from LTA subunit were individually fused to a modified chicken ovalbumin carrier protein, and each epitope-ovalbumin fusion was used to immunize mice. Data showed all 11 LTA epitopes were immunogenic; epitope 7 (105SPHPYEQEVSA115) induced greater titers of anti-LT antibodies which neutralized LT enterotoxicity more effectively. To examine these epitopes for the significance in LT enterotoxicity, we constructed LT mutants by substituting each of 10 epitopes at the toxic A1 domain of LTA subunit with a foreign epitope and examined LT mutants for enterotoxicity and GM1-binding activity. Data showed that LT mutants exhibited no enterotoxicity but retained GM1-binding activity. The results from this study indicated that while not all immunodominant LTA epitopes were neutralizing, LT mutants with an individual epitope substituted lost enterotoxicity but retained GM1-binding activity. These results provided additional information to understand LT immunogenicity and enterotoxicity and suggested the potential application of LT platform for multivalent vaccines against ETEC diarrhea and other diseases.IMPORTANCE No vaccine is licensed for enterotoxigenic Escherichia coli (ETEC) strains, which remain a leading cause of diarrhea in children from developing countries and international travelers. GM1-binding heat-labile toxin (LT) which is a key virulence factor of ETEC diarrhea is a strong vaccine antigen and a self-adjuvant. LT can also serve a backbone or platform for MEFA (multiepitope fusion antigen), a newly developed structural vaccinology technology, to present heterogeneous epitopes (by replacing LT epitopes) and to mimic epitope antigenicity for development of broadly protective vaccines. Data from this study identified neutralizing LT epitopes and demonstrated that substitution of LT epitopes eliminated LT enterotoxicity without altering GM1-binding activity, suggesting LT is potentially a versatile MEFA platform to present heterogeneous epitopes for multivalent vaccines against ETEC and other pathogens.
Collapse
Affiliation(s)
- Jiachen Huang
- Department of Diagnostic Medicine/Pathobiology, Kansas State University College of Veterinary Medicine, Manhattan, Kansas, USA
| | - Qiangde Duan
- Department of Diagnostic Medicine/Pathobiology, Kansas State University College of Veterinary Medicine, Manhattan, Kansas, USA
| | - Weiping Zhang
- Department of Diagnostic Medicine/Pathobiology, Kansas State University College of Veterinary Medicine, Manhattan, Kansas, USA
| |
Collapse
|
41
|
Characterization and Protective Efficacy of Type III Secretion Proteins as a Broadly Protective Subunit Vaccine against Salmonella enterica Serotypes. Infect Immun 2018; 86:IAI.00473-17. [PMID: 29311233 DOI: 10.1128/iai.00473-17] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 12/17/2017] [Indexed: 12/20/2022] Open
Abstract
Nontyphoidal Salmonella enterica serotypes (NTS) are the leading cause of hospitalization and death due to foodborne illnesses. NTS are the costliest of the foodborne pathogens and cause ∼$4 billion annually in health care costs. In Africa, new invasive NTS are the leading cause of bacteremia, especially in HIV-positive children and adults. Current vaccines against S. enterica are not broadly protective and most are directed at the typhoid-causing serotypes, not the NTS. All S. enterica strains require two type III secretion systems (T3SS) for virulence. The T3SS needle tip protein and the first translocator are localized to the T3SS needle tip and are required for pathogenesis of S. enterica Collectively they are 95 to 98% conserved at the amino acid sequence level among all S. enterica strains. The Salmonella pathogenicity island 1 or 2 tip and first translocator proteins were genetically fused to produce the S1 and S2 fusion proteins, respectively, as potential vaccine candidates. S1 and S2 were then characterized using spectroscopic techniques to understand their structural and biophysical properties. Formulated at the proper pH, S1, S2, or S1 plus S2 (S1S2), admixed with adjuvant, was used to immunize mice followed by a lethal challenge with S. enterica serotype Typhimurium or S. enterica serotype Enteritidis. The S1S2 formulation provided the highest protective efficacy, thus demonstrating that an S1S2 subunit vaccine can provide broad, serotype-independent protection, possibly against all S. enterica serotypes. Such a finding would be transformative in improving human health.
Collapse
|
42
|
Arabshahi S, Nayeri Fasaei B, Derakhshandeh A, Novinrooz A. In silico design of a novel chimeric shigella IpaB fused to C terminal of clostridium perfringens enterotoxin as a vaccine candidate. Bioengineered 2017; 9:170-177. [PMID: 29091543 PMCID: PMC5972921 DOI: 10.1080/21655979.2017.1373535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
This study aimed to design a novel chimeric protein in silico to serve as a serotype-independent vaccine candidate against Shigella. The chimera contains amino acid residues 240–460 of Shigella invasion plasmid antigen B (IpaB) and the C-terminus of Clostridium perfringens enterotoxin (C-CPE). Amino acid sequences of 537 peptide linkers were obtained from two protein linker databases. 3D structures of IpaB-CPE290–319, IpaB-CPE184–319, IpaB-CPE194–319 and 537 newly designed IpaB-linker-CPE290–319 constructs with varying linker regions were predicted. These predicted 3D structures were merged with the 3D structures of native IpaB240–460, CPE194–319, CPE184–319 and CPE290–319 to select the structure most similar to native IpaB and C-CPE. Several in silico tools were used to determine the suitability of the selected IpaB-C-CPE structure as a vaccine candidate. None of the 537 linkers was capable of preserving the native structure of CPE290–319 within the IpaB-linker-CPE290–319 structure. In silico analysis determined that the IpaB-CPE194–319 3D structure was the most similar to the 3D structure of the respective native CPE domain and that it was a stable chimeric protein exposing multiple B-cell epitopes. IpaB-CPE194–319 was designed for its capability to bind to human intestinal epithelial and M cells and to accumulate on these cells. The predicted B-cell epitopes are likely to be capable of inducing a mucosal antibody response in the human intestine against Shigella IpaB. This study also showed that the higher binding affinities of CPE184–319 and CPE194–319 to claudin molecules than those of CPE290–319 is the result of preserving the 3D structures of CPE184–319 and CPE194–319 when they are linked to the C-termini of other proteins.
Collapse
Affiliation(s)
- Sina Arabshahi
- a Department of Pathobiology , School of Veterinary Medicine, Shiraz University , Shiraz , Iran
| | - Bahar Nayeri Fasaei
- b Department of Microbiology and Immunology, Faculty of Veterinary Medicine , University of Tehran , Tehran , Iran
| | - Abdollah Derakhshandeh
- a Department of Pathobiology , School of Veterinary Medicine, Shiraz University , Shiraz , Iran
| | - Aytak Novinrooz
- a Department of Pathobiology , School of Veterinary Medicine, Shiraz University , Shiraz , Iran
| |
Collapse
|
43
|
Vander Broek CW, Stevens JM. Type III Secretion in the Melioidosis Pathogen Burkholderia pseudomallei. Front Cell Infect Microbiol 2017; 7:255. [PMID: 28664152 PMCID: PMC5471309 DOI: 10.3389/fcimb.2017.00255] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 05/31/2017] [Indexed: 02/03/2023] Open
Abstract
Burkholderia pseudomallei is a Gram-negative intracellular pathogen and the causative agent of melioidosis, a severe disease of both humans and animals. Melioidosis is an emerging disease which is predicted to be vastly under-reported. Type III Secretion Systems (T3SSs) are critical virulence factors in Gram negative pathogens of plants and animals. The genome of B. pseudomallei encodes three T3SSs. T3SS-1 and -2, of which little is known, are homologous to Hrp2 secretion systems of the plant pathogens Ralstonia and Xanthomonas. T3SS-3 is better characterized and is homologous to the Inv/Mxi-Spa secretion systems of Salmonella spp. and Shigella flexneri, respectively. Upon entry into the host cell, B. pseudomallei requires T3SS-3 for efficient escape from the endosome. T3SS-3 is also required for full virulence in both hamster and murine models of infection. The regulatory cascade which controls T3SS-3 expression and the secretome of T3SS-3 have been described, as well as the effect of mutations of some of the structural proteins. Yet only a few effector proteins have been functionally characterized to date and very little work has been carried out to understand the hierarchy of assembly, secretion and temporal regulation of T3SS-3. This review aims to frame current knowledge of B. pseudomallei T3SSs in the context of other well characterized model T3SSs, particularly those of Salmonella and Shigella.
Collapse
Affiliation(s)
- Charles W Vander Broek
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of EdinburghMidlothian, United Kingdom
| | - Joanne M Stevens
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of EdinburghMidlothian, United Kingdom
| |
Collapse
|
44
|
Functional and Antigen-Specific Serum Antibody Levels as Correlates of Protection against Shigellosis in a Controlled Human Challenge Study. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2017; 24:CVI.00412-16. [PMID: 27927680 PMCID: PMC5299116 DOI: 10.1128/cvi.00412-16] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 11/29/2016] [Indexed: 11/29/2022]
Abstract
Shigella is an important cause of diarrheal disease in young children living in developing countries. No approved vaccines are available, and the development of vaccine candidates has been hindered by the lack of firm immunological correlates of protection, among other reasons. To address this gap in knowledge, we established quantitative assays to measure Shigella-specific serum bactericidal antibody (SBA) and opsonophagocytic killing antibody (OPKA) activities and investigated their potential association with protection against disease in humans. SBA, OPKA, and Ipa-, VirG (IscA)-, and Shigella flexneri 2a lipopolysaccharide-specific serum IgG titers were determined in adult volunteers who received Shigella vaccine candidate EcSf2a-2 and in unvaccinated controls, all of whom were challenged with virulent Shigella flexneri 2a. Prechallenge antibody titers were compared with disease severity after challenge. SBA and OPKA, as well as IpaB- and VirG-specific IgG, significantly correlated with reduced illness. SBA and OPKA assays were also used to evaluate the immunogenicity of leading live attenuated vaccine candidates Shigella CVD 1204 and CVD 1208S in humans. A single oral immunization with CVD 1204 or CVD 1208S resulted in SBA seroconversion rates of 71% and 47% and OPKA seroconversion rates of 57% and 35%, respectively. Higher functional antibody responses were induced by CVD 1204, which is consistent with its lower attenuation. This is the first demonstration of SBA, OPKA, and IpaB- and VirG-specific IgG levels as potential serological correlates of protection against shigellosis in humans. These results warrant further studies to establish their capacity to predict protective immunity and vaccine efficacy.
Collapse
|
45
|
Stinson JA, Raja WK, Lee S, Kim HB, Diwan I, Tutunjian S, Panilaitis B, Omenetto FG, Tzipori S, Kaplan DL. Silk Fibroin Microneedles for Transdermal Vaccine Delivery. ACS Biomater Sci Eng 2017; 3:360-369. [PMID: 33465933 DOI: 10.1021/acsbiomaterials.6b00515] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Microneedles represent an exciting departure from the existing parenteral injection paradigm for drug delivery, particularly for the administration of vaccines. While the benefit of delivering vaccine antigens to immunocompetent tissue in the skin has been established, there have been varying degrees of success using microneedles to do so. Here, we investigate the use of silk fibroin protein to produce microneedles and evaluate their ability to deliver vaccines against influenza, Clostridium difficile, and Shigella. Fibroin protein from the silkworm Bombyx mori possesses suitable properties for use in a microneedle system, including all-aqueous processing, mechanical strength in dried formats, biocompatibility, and the ability to temperature stabilize biomacromolecules. As such, this biomaterial combines the processing and biocompatibility advantages of a dissolving microneedle system with the product stability and mechanical strength of coated insoluble microneedles. Through successful vaccination of mice against three separate antigens, we establish that silk fibroin is well-suited for use as a solid-coated microneedle delivery system and offers long-term potential similar to that of the leading microneedle biomaterials.
Collapse
Affiliation(s)
- Jordan A Stinson
- Department of Biomedical Engineering, 4 Colby Street, Tufts University, Medford, Massachusetts 02155, United States
| | - Waseem K Raja
- Department of Biomedical Engineering, 4 Colby Street, Tufts University, Medford, Massachusetts 02155, United States.,Department of Brain and Cognitive Sciences, 43 Vassar Street, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Sangun Lee
- Department of Infectious Disease and Global Health, 200 Westboro Road, Tufts University Cummings School of Veterinary Medicine, North Grafton, Massachusetts 01536, United States
| | - Hyeun Bum Kim
- Department of Animal Resources Science, Dankook University, Cheonan 31116, South Korea
| | - Izzuddin Diwan
- Department of Biomedical Engineering, 4 Colby Street, Tufts University, Medford, Massachusetts 02155, United States
| | - Stephen Tutunjian
- Department of Biomedical Engineering, 4 Colby Street, Tufts University, Medford, Massachusetts 02155, United States
| | - Bruce Panilaitis
- Department of Biomedical Engineering, 4 Colby Street, Tufts University, Medford, Massachusetts 02155, United States
| | - Fiorenzo G Omenetto
- Department of Biomedical Engineering, 4 Colby Street, Tufts University, Medford, Massachusetts 02155, United States
| | - Saul Tzipori
- Department of Infectious Disease and Global Health, 200 Westboro Road, Tufts University Cummings School of Veterinary Medicine, North Grafton, Massachusetts 01536, United States
| | - David L Kaplan
- Department of Biomedical Engineering, 4 Colby Street, Tufts University, Medford, Massachusetts 02155, United States
| |
Collapse
|
46
|
Nandre R, Ruan X, Duan Q, Zhang W. Enterotoxigenic Escherichia coli heat-stable toxin and heat-labile toxin toxoid fusion 3xSTaN12S-dmLT induces neutralizing anti-STa antibodies in subcutaneously immunized mice. FEMS Microbiol Lett 2016; 363:fnw246. [PMID: 27810884 PMCID: PMC10101651 DOI: 10.1093/femsle/fnw246] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 09/02/2016] [Accepted: 11/01/2016] [Indexed: 12/31/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) bacteria producing heat-stable toxin (STa) and/or heat-labile toxin (LT) are among top causes of children's diarrhea and travelers' diarrhea. Currently no vaccines are available for ETEC associated diarrhea. A major challenge in developing ETEC vaccines is the inability to stimulate protective antibodies against the key STa toxin that is potently toxic and also poorly immunogenic. A recent study suggested toxoid fusion 3xSTaN12S-dmLT, which consists of a monomer LT toxoid (LTR192G/L211A) and three copies of STa toxoid STaN12S, may represent an optimal immunogen inducing neutralizing antibodies against STa toxin [IAI 2014, 82(5):1823-32]. In this study, we immunized mice with this fusion protein following a different parenteral route and using different adjuvants to further characterize immunogenicity of this toxoid fusion. Data from this study showed that 3xSTaN12S-dmLT toxoid fusion induced neutralizing anti-STa antibodies in the mice following subcutaneous immunization, as effectively as in the mice under intraperitoneal route. Data also indicated that double mutant LT (dmLT) can be an effective adjuvant for this toxoid fusion in mice subcutaneous immunization. Results from this study affirmed that toxoid fusion 3xSTaN12S-dmLT induces neutralizing antibodies against STa toxin, suggesting this toxoid fusion is potentially a promising immunogen for ETEC vaccine development.
Collapse
|
47
|
Development of recombinant vaccine candidate molecule against Shigella infection. Vaccine 2016; 34:5376-5383. [DOI: 10.1016/j.vaccine.2016.08.034] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 07/28/2016] [Accepted: 08/12/2016] [Indexed: 11/22/2022]
|
48
|
Epigenetics changes caused by the fusion of human embryonic stem cell and ovarian cancer cells. Biosci Rep 2016; 36:BSR20160104. [PMID: 27377320 PMCID: PMC5025808 DOI: 10.1042/bsr20160104] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Accepted: 07/01/2016] [Indexed: 12/21/2022] Open
Abstract
To observe the effect of gene expression and tumorigenicity in hybrid cells of human embryonic stem cells (hESCs) and ovarian cancer cells in vitro and in vivo using a mouse model, and to determine its feasibility in reprogramming tumour cells growth and apoptosis, for a potential exploration of the role of hESCs and tumour cells fusion in the management of ovarian cancer. Stable transgenic hESCs (H1) and ovarian cancer cell line OVCAR-3 were established before fusion, and cell fusion system was established to analyse the related indicators. PTEN expression in HO-H1 cells was higher than those in the parental stem cells and lower than those in parental tumour cells; the growth of OV-H1 (RFP+GFP) hybrid cells with double fluorescence expressions were obviously slower than that of human embryonic stem cells and OVCAR-3 ovarian cancer cells. The apoptosis signal of the OV-H1 hybrid cells was significantly higher than that of the hESCs and OVCAR-3 ovarian cancer cells. In vivo results showed that compared with 7 days, 28 days and 35 days after inoculation of OV-H1 hybrid cells; also, apoptotic cell detection indicated that much stronger apoptotic signal was found in OV-H1 hybrid cells inoculated mouse. The hESCs can inhibit the growth of OVCAR-3 cells in vitro by suppressing p53 and PTEN expression to suppress the growth of tumour that may be achieved by inducing apoptosis of OVCAR-3 cells. The change of epigenetics after fusion of ovarian cancer cells and hESCs may become a novel direction for treatment of ovarian cancer.
Collapse
|
49
|
Circulating Gut-Homing (α4β7+) Plasmablast Responses against Shigella Surface Protein Antigens among Hospitalized Patients with Diarrhea. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2016; 23:610-7. [PMID: 27193041 DOI: 10.1128/cvi.00205-16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 05/09/2016] [Indexed: 11/20/2022]
Abstract
Developing countries are burdened with Shigella diarrhea. Understanding mucosal immune responses associated with natural Shigella infection is important to identify potential correlates of protection and, as such, to design effective vaccines. We performed a comparative analysis of circulating mucosal plasmablasts producing specific antibodies against highly conserved invasive plasmid antigens (IpaC, IpaD20, and IpaD120) and two recently identified surface protein antigens, pan-Shigella surface protein antigen 1 (PSSP1) and PSSP2, common to all virulent Shigella strains. We examined blood and stool specimens from 37 diarrheal patients admitted to the Infectious Diseases & Beliaghata General Hospital, Kolkata, India. The etiological agent of diarrhea was investigated in stool specimens by microbiological methods and real-time PCR. Gut-homing (α4β7 (+)) antibody-secreting cells (ASCs) were isolated from patient blood by means of combined magnetic cell sorting and two-color enzyme-linked immunosorbent spot (ELISPOT) assay. Overall, 57% (21 of 37) and 65% (24 of 37) of the patients were positive for Shigella infection by microbiological and real-time PCR assays, respectively. The frequency of α4β7 (+) IgG ASC responders against Ipas was higher than that observed against PSSP1 or PSSP2, regardless of the Shigella serotype isolated from these patients. Thus, α4β7 (+) ASC responses to Ipas may be considered an indirect marker of Shigella infection. The apparent weakness of ASC responses to PSSP1 is consistent with the lack of cross-protection induced by natural Shigella infection. The finding that ASC responses to IpaD develop in patients with recent-onset shigellosis indicates that such responses may not be protective or may wane too rapidly and/or be of insufficient magnitude.
Collapse
|
50
|
Bulir DC, Liang S, Lee A, Chong S, Simms E, Stone C, Kaushic C, Ashkar A, Mahony JB. Immunization with chlamydial type III secretion antigens reduces vaginal shedding and prevents fallopian tube pathology following live C. muridarum challenge. Vaccine 2016; 34:3979-85. [PMID: 27325352 DOI: 10.1016/j.vaccine.2016.06.046] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 04/25/2016] [Accepted: 06/14/2016] [Indexed: 11/26/2022]
Abstract
Chlamydia trachomatis infections in women are often asymptomatic and if left untreated can lead to significant late sequelae including pelvic inflammatory disease and tubal factor infertility. Vaccine development efforts over the past three decades have been unproductive and there is no vaccine approved for use in humans. The existence of serologically distinct strains or serovars of C. trachomatis mandates a vaccine that will provide protection against multiple serovars. Chlamydia spp. use a highly conserved type III secretion system (T3SS) composed of both structural and effector proteins which is an essential virulence factor for infection and intracellular replication. In this study we evaluated a novel fusion protein antigen (BD584) which consists of three T3SS proteins from C. trachomatis (CopB, CopD, and CT584) as a potential chlamydial vaccine candidate. Intranasal immunization with BD584 elicited serum neutralizing antibodies that inhibited C. trachomatis infection in vitro. Following intravaginal challenge with C. muridarum, immunized mice had a 95% reduction in chlamydial shedding from the vagina at the peak of infection and cleared the infection sooner than control mice. Immunization with BD584 also reduced the rate of hydrosalpinx by 87.5% compared to control mice. Together, these results suggest that highly conserved proteins of the chlamydial T3SS may represent good candidates for a Chlamydia vaccine.
Collapse
Affiliation(s)
- David C Bulir
- M. G. DeGroote Institute for Infectious Disease Research, Canada; St. Joseph's Research Institute, St. Joseph's Healthcare, Hamilton, Ontario, Canada
| | - Steven Liang
- M. G. DeGroote Institute for Infectious Disease Research, Canada; St. Joseph's Research Institute, St. Joseph's Healthcare, Hamilton, Ontario, Canada
| | - Amanda Lee
- St. Joseph's Research Institute, St. Joseph's Healthcare, Hamilton, Ontario, Canada
| | - Sylvia Chong
- St. Joseph's Research Institute, St. Joseph's Healthcare, Hamilton, Ontario, Canada
| | - Elizabeth Simms
- St. Joseph's Research Institute, St. Joseph's Healthcare, Hamilton, Ontario, Canada
| | - Christopher Stone
- St. Joseph's Research Institute, St. Joseph's Healthcare, Hamilton, Ontario, Canada
| | - Charu Kaushic
- M. G. DeGroote Institute for Infectious Disease Research, Canada; Department of Pathology and Molecular Medicine, McMaster University, Canada
| | - Ali Ashkar
- M. G. DeGroote Institute for Infectious Disease Research, Canada; Department of Pathology and Molecular Medicine, McMaster University, Canada
| | - James B Mahony
- M. G. DeGroote Institute for Infectious Disease Research, Canada; Department of Pathology and Molecular Medicine, McMaster University, Canada; St. Joseph's Research Institute, St. Joseph's Healthcare, Hamilton, Ontario, Canada.
| |
Collapse
|