1
|
Jarząb A, Dąbrowska A, Naporowski P, Krasna K, Szmyt A, Świat M, Pawlik K, Witkowska D, Ziomek E, Gamian A. Outer membrane protein C is a protective and unique vaccine antigen against Shigella flexneri 3a. Sci Rep 2024; 14:25398. [PMID: 39455688 PMCID: PMC11511853 DOI: 10.1038/s41598-024-76745-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
The anti-Shigella vaccine is one of the WHO's top priorities. Every year the disease kills more than 200,000 people worldwide and poses a serious threat to children under 5 years of age and the elderly. Increasing antibiotic resistance and limitations in diagnostics emphasize the need to develop an effective vaccine. Recent research and clinical trials report multiple approaches used in Shigella-vaccine development. However, despite the efforts of researchers, pharmaceutical companies and health care organizations, there is no licensed vaccine against shigellosis available to the community. Here, we expressed, broadly characterized and demonstrated the protective properties of outer membrane protein C as an effective molecule serving as a universal antigen for Shigella vaccine. Most of the current approaches to the development of Shigella vaccine are based on the polysaccharide antigens, which are serotype specific and have always been challenging in terms of their high specificity, targeting the most exposed surface antigens identified for certain Shigella serotypes. Here, we confirm immunogenic and protective properties of the recombinant OmpC protein, which protects mice against a lethal dose of a virulent strain 2 weeks after active immunization.
Collapse
Affiliation(s)
- Anna Jarząb
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla Str. 12, 53-114, Wroclaw, Poland.
| | - Anna Dąbrowska
- Department of Animal Products Technology and Quality Management, Wroclaw University of Environmental and Life Sciences, Chelmonskiego Str. 37/41, 51-630, Wroclaw, Poland
| | - Piotr Naporowski
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla Str. 12, 53-114, Wroclaw, Poland
| | - Karina Krasna
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla Str. 12, 53-114, Wroclaw, Poland
| | - Agnieszka Szmyt
- Department of Animal Products Technology and Quality Management, Wroclaw University of Environmental and Life Sciences, Chelmonskiego Str. 37/41, 51-630, Wroclaw, Poland
| | - Michał Świat
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla Str. 12, 53-114, Wroclaw, Poland
| | - Krzysztof Pawlik
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla Str. 12, 53-114, Wroclaw, Poland
| | - Danuta Witkowska
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla Str. 12, 53-114, Wroclaw, Poland
| | - Edmund Ziomek
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla Str. 12, 53-114, Wroclaw, Poland
| | - Andrzej Gamian
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla Str. 12, 53-114, Wroclaw, Poland
| |
Collapse
|
2
|
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: 10] [Impact Index Per Article: 10.0] [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
|
3
|
Ali A, Waris A, Khan MA, Asim M, Khan AU, Khan S, Zeb J. Recent advancement, immune responses, and mechanism of action of various vaccines against intracellular bacterial infections. Life Sci 2023; 314:121332. [PMID: 36584914 DOI: 10.1016/j.lfs.2022.121332] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 12/29/2022]
Abstract
Emerging and re-emerging bacterial infections are a serious threat to human and animal health. Extracellular bacteria are free-living, while facultative intracellular bacteria replicate inside eukaryotic host cells. Many serious human illnesses are now known to be caused by intracellular bacteria such as Salmonella enterica, Escherichia coli, Staphylococcus aureus, Rickettsia massiliae, Chlamydia species, Brucella abortus, Mycobacterium tuberculosis and Listeria monocytogenes, which result in substantial morbidity and mortality. Pathogens like Mycobacterium, Brucella, MRSA, Shigella, Listeria, and Salmonella can infiltrate and persist in mammalian host cells, particularly macrophages, where they proliferate and establish a repository, resulting in chronic and recurrent infections. The current treatment for these bacteria involves the application of narrow-spectrum antibiotics. FDA-approved vaccines against obligate intracellular bacterial infections are lacking. The development of vaccines against intracellular pathogenic bacteria are more difficult because host defense against these bacteria requires the activation of the cell-mediated pathway of the immune system, such as CD8+ T and CD4+ T. However, different types of vaccines, including live, attenuated, subunit, killed whole cell, nano-based and DNA vaccines are currently in clinical trials. Substantial development has been made in various vaccine strategies against intracellular pathogenic bacteria. This review focuses on the mechanism of intracellular bacterial infection, host immune response, and recent advancements in vaccine development strategies against various obligate intracellular bacterial infections.
Collapse
Affiliation(s)
- Asmat Ali
- Department of Biotechnology and Genetic Engineering, Hazara University Mansehra, Pakistan
| | - Abdul Waris
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong.
| | - Muhammad Ajmal Khan
- Division of Life Sciences, Center for Cancer Research and State Key Laboratory of Molecular Neurosciences, The Hong Kong University of Science and Technology, Hong Kong
| | - Muhammad Asim
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong
| | - Atta Ullah Khan
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, China
| | - Sahrish Khan
- Department of Biotechnology, Quaid-i-Azam University, Islamabad, Pakistan
| | - Jehan Zeb
- Department of Infectious Diseases and Public Health, City University of Hong Kong, Hong Kong
| |
Collapse
|
4
|
ANAM K, ENDHARTI AT, POERANTO S, PRAWIRO SR. Peptide Sequence of Pili Subunit Protein 49.8 kDa Shigella flexneri as Antigenic Epitope for Shigellosis Vaccine Development. Turk J Pharm Sci 2022; 19:649-656. [PMID: 36544298 PMCID: PMC9780573 DOI: 10.4274/tjps.galenos.2021.75031] [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: 12/31/2022]
Abstract
Objectives This study investigates the amino acid sequence and identifies antigenic epitopes of 49.8 kilodalton (kDa) pili protein Shigella flexneri, which will be used as candidates for the shigellosis vaccine. Materials and Methods Our study is a prospectively descriptive laboratory. We used bacterial isolate of S. flexneri pili isolation was performed using a pili cutter and sodium dodecyl-sulfate polyacrylamide gel electrophoresis. The amino acid sequences were analyzed using liquid chromatography dual mass spectrometry (LC-MS/MS) method in the proteomic laboratory. The target epitope antigenicity analysis was tested using Kolaskar and Tongaonkar Antigenicity software. The Bepired Linear Epitope Prediction software is used for epitope mapping. PymOL software was used for the visualization of proteins and molecular docking. Peptides and antibodies were applied to hemagglutination test and immune response was tested using the dot blot method. Results LC-MS/MS analysis results from the mascot server showed that the 49.8 kDa pili protein is S. flexneri similar to the flagellin protein of S. flexneri 1235-66 (ID I6H2T2). The results of antigenicity analysis and epitope mapping showed that areas of protein that has the most potential and antigenic epitopes are the regions 98-111 and 263-290 with the amino acid sequences, QSSTGTNSQSDLDS (Q-S) and DTTITKAETKTVTKNQVVDTPVTTDAAK (D-K). The results of the molecular docking interaction test between the peptide and the B-cell receptor have a low binding energy. Peptide Q-S and peptide D-K antigens are hemagglutinin molecules because they can agglutinate erythrocytes. The immune response between peptide antigens and anti-peptide antibodies can react based on color gradations in the dotblot method. Conclusion The amino acid sequences Q-S and D-K are potentially antigenic epitopes. These peptides can be used to develop candidates for shigellosis vaccine.
Collapse
Affiliation(s)
- Khoirul ANAM
- Universitas Brawijaya, Faculty of Medicine, Doctoral Program in Medical Science, Malang, Indonesia,Institute of Technology and Health Science of Wiyata Husada Samarinda, Medical Laboratory Technology Study Program, Samarinda, Indonesia,* Address for Correspondence: Phone: +6281347175316 E-mail:
| | - Agustina Tri ENDHARTI
- Universitas Brawijaya, Faculty of Medicine, Department of Parasitology, Malang, Indonesia
| | - Sri POERANTO
- Universitas Brawijaya, Faculty of Medicine, Department of Parasitology, Malang, Indonesia
| | - Sumarno Reto PRAWIRO
- Universitas Brawijaya, Faculty of Medicine, Department of Clinical Microbiology, Malang, Indonesia
| |
Collapse
|
5
|
Sztein MB, Booth JS. Controlled human infectious models, a path forward in uncovering immunological correlates of protection: Lessons from enteric fevers studies. Front Microbiol 2022; 13:983403. [PMID: 36204615 PMCID: PMC9530043 DOI: 10.3389/fmicb.2022.983403] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
Enteric infectious diseases account for more than a billion disease episodes yearly worldwide resulting in approximately 2 million deaths, with children under 5 years old and the elderly being disproportionally affected. Enteric pathogens comprise viruses, parasites, and bacteria; the latter including pathogens such as Salmonella [typhoidal (TS) and non-typhoidal (nTS)], cholera, Shigella and multiple pathotypes of Escherichia coli (E. coli). In addition, multi-drug resistant and extensively drug-resistant (XDR) strains (e.g., S. Typhi H58 strain) of enteric bacteria are emerging; thus, renewed efforts to tackle enteric diseases are required. Many of these entero-pathogens could be controlled by oral or parenteral vaccines; however, development of new, effective vaccines has been hampered by lack of known immunological correlates of protection (CoP) and limited knowledge of the factors contributing to protective responses. To fully comprehend the human response to enteric infections, an invaluable tool that has recently re-emerged is the use of controlled human infection models (CHIMs) in which participants are challenged with virulent wild-type (wt) organisms. CHIMs have the potential to uncover immune mechanisms and identify CoP to enteric pathogens, as well as to evaluate the efficacy of therapeutics and vaccines in humans. CHIMs have been used to provide invaluable insights in the pathogenesis, host-pathogen interaction and evaluation of vaccines. Recently, several Oxford typhoid CHIM studies have been performed to assess the role of multiple cell types (B cells, CD8+ T, Tregs, MAIT, Monocytes and DC) during S. Typhi infection. One of the key messages that emerged from these studies is that baseline antigen-specific responses are important in that they can correlate with clinical outcomes. Additionally, volunteers who develop typhoid disease (TD) exhibit higher levels and more activated cell types (e.g., DC and monocytes) which are nevertheless defective in discrete signaling pathways. Future critical aspects of this research will involve the study of immune responses to enteric infections at the site of entry, i.e., the intestinal mucosa. This review will describe our current knowledge of immunity to enteric fevers caused byS. Typhi and S. Paratyphi A, with emphasis on the contributions of CHIMs to uncover the complex immunological responses to these organisms and provide insights into the determinants of protective immunity.
Collapse
Affiliation(s)
- Marcelo B. Sztein
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
- *Correspondence: Marcelo B. Sztein,
| | - Jayaum S. Booth
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
- Jayaum S. Booth,
| |
Collapse
|
6
|
Mohammad Shabani NR, Khairul Hisyam Ismail CM, Anthony AA, Leow CH, Chuah C, Abdul Majeed AB, Nor NM, He Y, Banga Singh KK, Leow CY. Mass spectrometry-based immunopeptidomics and computational vaccinology strategies for the identification of universal Shigella immunogenic candidates. Comput Biol Med 2022; 148:105900. [DOI: 10.1016/j.compbiomed.2022.105900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 06/26/2022] [Accepted: 07/16/2022] [Indexed: 11/16/2022]
|
7
|
Anam K, Endharti AT, Poeranto S, Sujuti H, Hidayati DYN, Prawiro SR. Shigella flexneri vaccine development: Oral administration of peptides derived from the 49.8 kDa pili protein subunit activates the intestinal immune response in mice. Vet World 2022; 15:281-287. [PMID: 35400957 PMCID: PMC8980390 DOI: 10.14202/vetworld.2022.281-287] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 01/05/2022] [Indexed: 11/16/2022] Open
Abstract
Background and Aim: The morbidity and mortality of Shigella infections remain a global challenge. Epitope-based vaccine development is an emerging strategy to prevent bacterial invasion. This study aimed to identify the ability of the 49.8 kDa pili subunit adhesin protein epitope of Shigella flexneri to induce an intestinal immune response in mice. Materials and Methods: Thirty adult male Balb/c mice were divided into a control group, cholera toxin B subunit (CTB) group, CTB+QSSTGTNSQSDLDS (pep_1) group, CTB+DTTITKAETKTVTKNQVVDTPVTTDAAK (pep_2) group, and CTB+ ATLGATLNRLDFNVNNK (pep_3). We performed immunization by orally administering 50 μg of antigen and 50 μl of adjuvant once a week over 4 weeks. We assessed the cellular immune response by quantifying T helper 2 (Th2) and Th17 using flow cytometry. In addition, we assessed the humoral immune response by quantifying interleukin (IL-4), IL-17, secretory immunoglobulin A (sIgA), and β-defensin using enzyme-linked immunoassay. Statistical analysis was performed using one-way analysis of variance and Kruskal–Wallis test. Results: Peptide oral immunization increases the cellular immune response as reflected by the increase of Th2 (p=0.019) and Th17 (p=0.004) cell counts, particularly in the CTB_pep_1 group. Humoral immune response activation was demonstrated by increased IL-4 levels, especially in the CTB+pep_3 group (p=0.000). The IL-17 level was increased significantly in the CTB+pep_1 group (p=0.042). The mucosal immune response was demonstrated by the sIgA levels increase in the CTB+pep_3 group (p=0.042) and the β-defensin protein levels (p=0.000). Conclusion: All selected peptides activated the cellular and humoral immune responses in the intestine of mice. Further studies are necessary to optimize antigen delivery and evaluate whether the neutralizing properties of these peptides allow them to prevent bacterial infection.
Collapse
Affiliation(s)
- Khoirul Anam
- Doctoral Program in Medical Science, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia; Study Program of Medical Laboratory Technology, Institute of Health and Science Technology Wiyata Husada, Samarinda, Indonesia
| | - Agustina Tri Endharti
- Department of Parasitology, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Sri Poeranto
- Department of Parasitology, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Hidayat Sujuti
- Department of Biochemistry, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Dwi Yuni Nur Hidayati
- Department of Clinical Microbiology, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Sumarno Reto Prawiro
- Department of Clinical Microbiology, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| |
Collapse
|
8
|
Bengtsson RJ, Simpkin AJ, Pulford CV, Low R, Rasko DA, Rigden DJ, Hall N, Barry EM, Tennant SM, Baker KS. Pathogenomic analyses of Shigella isolates inform factors limiting shigellosis prevention and control across LMICs. Nat Microbiol 2022; 7:251-261. [PMID: 35102306 PMCID: PMC8813619 DOI: 10.1038/s41564-021-01054-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 12/17/2021] [Indexed: 12/17/2022]
Abstract
Shigella spp. are the leading bacterial cause of severe childhood diarrhoea in low- and middle-income countries (LMICs), are increasingly antimicrobial resistant and have no widely available licenced vaccine. We performed genomic analyses of 1,246 systematically collected shigellae sampled from seven countries in sub-Saharan Africa and South Asia as part of the Global Enteric Multicenter Study (GEMS) between 2007 and 2011, to inform control and identify factors that could limit the effectiveness of current approaches. Through contemporaneous comparison among major subgroups, we found that S. sonnei contributes ≥6-fold more disease than other Shigella species relative to its genomic diversity, and highlight existing diversity and adaptative capacity among S. flexneri that may generate vaccine escape variants in <6 months. Furthermore, we show convergent evolution of resistance against ciprofloxacin, the current WHO-recommended antimicrobial for the treatment of shigellosis, among Shigella isolates. This demonstrates the urgent need to integrate existing genomic diversity into vaccine and treatment plans for Shigella, providing a framework for the focused application of comparative genomics to guide vaccine development, and the optimization of control and prevention strategies for other pathogens relevant to public health policy considerations.
Collapse
Affiliation(s)
- Rebecca J Bengtsson
- Clinical Infection, Microbiology and Immunity, Institute of Infection, Veterinary and Ecological Sciences, The University of Liverpool, Liverpool, UK
| | - Adam J Simpkin
- Biochemistry and Systems Biology, Institute of Systems, Molecular and Systems Biology, The University of Liverpool, Liverpool, UK
| | - Caisey V Pulford
- Clinical Infection, Microbiology and Immunity, Institute of Infection, Veterinary and Ecological Sciences, The University of Liverpool, Liverpool, UK
- Gastrointestinal Infections and Food Safety (One Health), United Kingdom Health Security Agency, London, UK
| | - Ross Low
- Earlham Institute, Norwich Research Park, Norwich, UK
| | - David A Rasko
- Department of Microbiology and Immunology, Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Daniel J Rigden
- Biochemistry and Systems Biology, Institute of Systems, Molecular and Systems Biology, The University of Liverpool, Liverpool, UK
| | - Neil Hall
- Earlham Institute, Norwich Research Park, Norwich, UK
- School of Biological Sciences, University of East Anglia, Norwich, UK
| | - Eileen M Barry
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sharon M Tennant
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kate S Baker
- Clinical Infection, Microbiology and Immunity, Institute of Infection, Veterinary and Ecological Sciences, The University of Liverpool, Liverpool, UK.
| |
Collapse
|
9
|
Shigella Outer Membrane Vesicles as Promising Targets for Vaccination. Int J Mol Sci 2022; 23:ijms23020994. [PMID: 35055181 PMCID: PMC8781765 DOI: 10.3390/ijms23020994] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/11/2022] [Accepted: 01/14/2022] [Indexed: 12/17/2022] Open
Abstract
The clinical symptoms of shigellosis, a gastrointestinal infection caused by Shigella spp. range from watery diarrhea to fulminant dysentery. Endemic infections, particularly among children in developing countries, represent the majority of clinical cases. The situation is aggravated due to the high mortality rate of shigellosis, the rapid dissemination of multi-resistant Shigella strains and the induction of only serotype-specific immunity. Thus, infection prevention due to vaccination, encompassing as many of the circulating serotypes as possible, has become a topic of interest. However, vaccines have turned out to be ineffective so far. Outer membrane vesicles (OMVs) are promising novel targets for vaccination. OMVs are constitutively secreted by Gram-negative bacteria including Shigella during growth. They are composed of soluble luminal portions and an insoluble membrane and can contain toxins, bioactive periplasmic and cytoplasmic (lipo-) proteins, (phospho-) lipids, nucleic acids and/or lipopolysaccharides. Thus, OMVs play an important role in bacterial cell–cell communication, growth, survival and pathogenesis. Furthermore, they modulate the secretion and transport of biomolecules, the stress response, antibiotic resistance and immune responses of the host. Thus, OMVs serve as novel secretion machinery. Here, we discuss the current literature and highlight the properties of OMVs as potent vaccine candidates because of their immunomodulatory, antigenic and adjuvant properties.
Collapse
|
10
|
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
|
11
|
Recent Progress in Shigella and Burkholderia pseudomallei Vaccines. Pathogens 2021; 10:pathogens10111353. [PMID: 34832508 PMCID: PMC8621228 DOI: 10.3390/pathogens10111353] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/15/2021] [Accepted: 10/18/2021] [Indexed: 12/28/2022] Open
Abstract
Significant advancement has been made in the development of vaccines against bacterial pathogens. However, several roadblocks have been found during the evaluation of vaccines against intracellular bacterial pathogens. Therefore, new lessons could be learned from different vaccines developed against unrelated intracellular pathogens. Bacillary dysentery and melioidosis are important causes of morbidity and mortality in developing nations, which are caused by the intracellular bacteria Shigella and Burkholderia pseudomallei, respectively. Although the mechanisms of bacterial infection, dissemination, and route of infection do not provide clues about the commonalities of the pathogenic infectious processes of these bacteria, a wide variety of vaccine platforms recently evaluated suggest that in addition to the stimulation of antibodies, identifying protective antigens and inducing T cell responses are some additional required elements to induce effective protection. In this review, we perform a comparative evaluation of recent candidate vaccines used to combat these two infectious agents, emphasizing the common strategies that can help investigators advance effective and protective vaccines to clinical trials.
Collapse
|
12
|
Hu R, Liu H, Wang M, Li J, Lin H, Liang M, Gao Y, Yang M. An OMV-Based Nanovaccine Confers Safety and Protection against Pathogenic Escherichia coli via Both Humoral and Predominantly Th1 Immune Responses in Poultry. NANOMATERIALS 2020; 10:nano10112293. [PMID: 33233490 PMCID: PMC7699605 DOI: 10.3390/nano10112293] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 11/07/2020] [Accepted: 11/16/2020] [Indexed: 12/12/2022]
Abstract
Avian pathogenic Escherichia coli (APEC) infection in poultry causes enormous economic losses and public health risks. Bacterial outer membrane vesicles (OMVs) and nano-sized proteolipids enriched with various immunogenic molecules have gained extensive interest as novel nanovaccines against bacterial infections. In this study, after the preparation of APEC O2-derived OMVs (APEC_OMVs) using the ultracentrifugation method and characterization of them using electron microscopy and nanoparticle tracking analyses, we examined the safety and vaccination effect of APEC_OMVs in broiler chicks and investigated the underlying immunological mechanism of protection. The results showed that APEC_OMVs had membrane-enclosed structures with an average diameter of 89 nm. Vaccination with 50 μg of APEC_OMVs had no side effects and efficiently protected chicks against homologous infection. APEC_OMVs could be effectively taken up by chicken macrophages and activated innate immune responses in macrophages in vitro. APEC_OMV vaccination significantly improved activities of serum non-specific immune factors, enhanced the specific antibody response and promoted the proliferation of splenic and peripheral blood lymphocytes in response to mitogen. Furthermore, APEC_OMVs also elicited a predominantly IFN-γ-mediated Th1 response in splenic lymphocytes. Our data revealed the involvement of both non-specific immune responses and specific antibody and cytokine responses in the APEC_OMV-mediated protection, providing broader knowledge for the development of multivalent APEC_OMV-based nanovaccine with high safety and efficacy in the future.
Collapse
Affiliation(s)
- Rujiu Hu
- College of Animal Science and Technology, Northwest A&F University, No.22 Xinong Road, Yangling 712100, Shaanxi, China; (R.H.); (H.L.); (M.W.); (H.L.); (M.L.)
| | - Haojing Liu
- College of Animal Science and Technology, Northwest A&F University, No.22 Xinong Road, Yangling 712100, Shaanxi, China; (R.H.); (H.L.); (M.W.); (H.L.); (M.L.)
| | - Mimi Wang
- College of Animal Science and Technology, Northwest A&F University, No.22 Xinong Road, Yangling 712100, Shaanxi, China; (R.H.); (H.L.); (M.W.); (H.L.); (M.L.)
| | - Jing Li
- Department of Animal Engineering, Yangling Vocational and Technical College, No.24 Weihui Road, Yangling 712100, Shaanxi, China;
| | - Hua Lin
- College of Animal Science and Technology, Northwest A&F University, No.22 Xinong Road, Yangling 712100, Shaanxi, China; (R.H.); (H.L.); (M.W.); (H.L.); (M.L.)
| | - Mingyue Liang
- College of Animal Science and Technology, Northwest A&F University, No.22 Xinong Road, Yangling 712100, Shaanxi, China; (R.H.); (H.L.); (M.W.); (H.L.); (M.L.)
| | - Yupeng Gao
- College of Animal Science and Technology, Northwest A&F University, No.22 Xinong Road, Yangling 712100, Shaanxi, China; (R.H.); (H.L.); (M.W.); (H.L.); (M.L.)
- Correspondence: (Y.G.); (M.Y.)
| | - Mingming Yang
- College of Animal Science and Technology, Northwest A&F University, No.22 Xinong Road, Yangling 712100, Shaanxi, China; (R.H.); (H.L.); (M.W.); (H.L.); (M.L.)
- Correspondence: (Y.G.); (M.Y.)
| |
Collapse
|
13
|
Kim NH, Ha EJ, Ko DS, Choi KS, Kwon HJ. Comparison of Humoral Immune Responses to Different Forms of Salmonella enterica Serovar Gallinarum Biovar Gallinarum. Front Vet Sci 2020; 7:598610. [PMID: 33240965 PMCID: PMC7677237 DOI: 10.3389/fvets.2020.598610] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 10/19/2020] [Indexed: 02/03/2023] Open
Abstract
Fowl typhoid is caused by Salmonella enterica serovar Gallinarum biovar Gallinarum (SG), and live attenuated, rough vaccine strains have been used. Both humoral and cellular immune responses are involved in protection, but the humoral responses to different forms of SG antigens are unclear. In this study, we compared humoral responses to a killed oil-emulsion (OE) smooth vaccine (SG002) and its rough mutant vaccine (SR2-N6) strains using proteomics techniques. We identified two immunogenic outer membrane proteins (OmpA and OmpX), and the selected linear epitopes were successfully applied in peptide-ELISA. Our peptide- and total OMP-ELISAs were used to compare the temporal humoral responses to various SG antigens: OE SG002 and SR2-N6; live, killed [PBS-suspension (PS) and OE)] and mixed (live and PS) formulations of another rough vaccine strain (SG 9R); and orally challenge with a field strain. Serum antibodies to the linear epitopes of OmpA and OmpX lasted only for the first 2 weeks, but serum antibodies against OMPs increased over time. The rough strain (SR2-N6) and mixed SG 9R induced higher serum antibody titers than the smooth strain (SG002) and single SG 9R (OE, live and PS SG 9R), respectively. Infection with the field strain delayed the serum antibody response by ~2 weeks. Mucosal immunity was not induced by any formulation, except for infection with the field strain after SG 9R vaccination. Thus, our results may be useful to understand humoral immunity against various SG antigens and to improve vaccine programs and serological diagnosis in the field.
Collapse
Affiliation(s)
- Nam-Hyung Kim
- Laboratory of Poultry Medicine, Seoul National University, Seoul, South Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, BK21 for Veterinary Science, Seoul, South Korea
| | - Eun-Jin Ha
- Laboratory of Poultry Medicine, Seoul National University, Seoul, South Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, BK21 for Veterinary Science, Seoul, South Korea
| | - Dae-Sung Ko
- Laboratory of Poultry Medicine, Seoul National University, Seoul, South Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, BK21 for Veterinary Science, Seoul, South Korea
| | - Kang-Seuk Choi
- Research Institute for Veterinary Science, College of Veterinary Medicine, BK21 for Veterinary Science, Seoul, South Korea
- Laboratory of Avian Diseases, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Hyuk-Joon Kwon
- Laboratory of Poultry Medicine, Seoul National University, Seoul, South Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, BK21 for Veterinary Science, Seoul, South Korea
- Farm Animal Clinical Training and Research Center (FACTRC), GBST, Seoul National University, Seoul, South Korea
| |
Collapse
|
14
|
León Y, Zapata L, Salas-Burgos A, Oñate A. In silico design of a vaccine candidate based on autotransporters and HSP against the causal agent of shigellosis, Shigella flexneri. Mol Immunol 2020; 121:47-58. [DOI: 10.1016/j.molimm.2020.02.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 02/10/2020] [Accepted: 02/12/2020] [Indexed: 12/19/2022]
|
15
|
Renu S, Markazi AD, Dhakal S, Lakshmanappa YS, Shanmugasundaram R, Selvaraj RK, Renukaradhya GJ. Oral Deliverable Mucoadhesive Chitosan- Salmonella Subunit Nanovaccine for Layer Chickens. Int J Nanomedicine 2020; 15:761-777. [PMID: 32099364 PMCID: PMC7006855 DOI: 10.2147/ijn.s238445] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 12/17/2019] [Indexed: 12/11/2022] Open
Abstract
PURPOSE Salmonellosis in poultry is a serious economic burden. A major concern is the public health hazard caused by consumption of Salmonella-contaminated poultry products. Currently used Salmonella vaccines are ineffective in combating poultry Salmonellosis warranting the need of a potent vaccine, especially an oral vaccine that can elicit robust local intestinal immunity. MATERIALS AND METHODS A Salmonella subunit chitosan nanoparticles (NPs)-based vaccine was prepared that contained immunogenic outer membrane proteins (OMPs) and -flagellin (F) protein (OMPs-F-CS NPs). OMPs-F-CS NPs were administered as an oral vaccine in layer chickens and the resultant humoral and cell-mediated immune responses and localization of NPs were examined using standard detection methods. RESULTS We demonstrated targeting of surface F-protein coated chitosan NPs to immune cells when delivered orally to layer chickens, the particles were localized in ileal Peyer's patches. The OMPs-F-CS NPs vaccinated layer chickens had significantly higher OMPs-specific mucosal IgA production and lymphocyte proliferation response. The candidate vaccine increased the expression of toll-like receptor (TLR)-2, TLR-4, IFN-γ, TGF-ß and IL-4 mRNA expression in chicken cecal tonsils. CONCLUSION Our study demonstrated that the chitosan-based oral Salmonella nanovaccine targets immune cells of chickens and induced antigen-specific B and T cell responses. This candidate oral Salmonella nanovaccine has the potential to mitigate Salmonellosis in poultry.
Collapse
Affiliation(s)
- Sankar Renu
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Wooster, OH 44691, USA and Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH43210, USA
| | - Ashley D Markazi
- Department of Animal Sciences, Ohio Agricultural Research and Development Center, The Ohio State University, Columbus, OH, USA
| | - Santosh Dhakal
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Wooster, OH 44691, USA and Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH43210, USA
| | - Yashavanth S Lakshmanappa
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Wooster, OH 44691, USA and Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH43210, USA
| | - Revathi Shanmugasundaram
- Department of Animal Sciences, Ohio Agricultural Research and Development Center, The Ohio State University, Columbus, OH, USA
| | - Ramesh K Selvaraj
- Department of Poultry Science, University of Georgia, Athens, GA30602, USA
| | - Gourapura J Renukaradhya
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, Wooster, OH 44691, USA and Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH43210, USA
| |
Collapse
|
16
|
Leow CY, Kazi A, Hisyam Ismail CMK, Chuah C, Lim BH, Leow CH, Banga Singh KK. Reverse vaccinology approach for the identification and characterization of outer membrane proteins of Shigella flexneri as potential cellular- and antibody-dependent vaccine candidates. Clin Exp Vaccine Res 2020; 9:15-25. [PMID: 32095437 PMCID: PMC7024733 DOI: 10.7774/cevr.2020.9.1.15] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 01/30/2020] [Indexed: 11/16/2022] Open
Abstract
Purpose In the developing world, bacillary dysentery is one of the most common communicable diarrheal infections. There are approximately 169 million cases of shigellosis reported worldwide. The disease is transmitted by a group of Gram-negative intracellular enterobacteria known as Shigella flexneri, S. sonnei, S. dysenteriae, and S. boydii. Conventional treatment regimens for Shigella have been less effective due to the development of resistant strains against antibiotics. Therefore, an effective vaccine for the long term control of Shigella transmission is urgently needed. Materials and Methods In this study, a reverse vaccinology approach was employed to identify most conserved and immunogenic outer membrane proteins (OMPs) of S. flexneri 2a. Results Five OMPs including fepA, ompC, nlpD_1, tolC, and nlpD_2 were identified as potential vaccine candidates. Protein-protein interactions analysis using STRING software (https://string-db.org/) revealed that five of these OMPs may potentially interact with other intracellular proteins which are involved in beta-lactam resistance pathway. B- and T-cell epitopes of the selected OMPs were predicted using BCPred as well as Propred I and Propred (http://crdd.osdd.net/raghava/propred/), respectively. Each of these OMPs contains regions which are capable to induce B- and T-cell immune responses. Conclusion Analysis acquired from this study showed that five selected OMPs have great potential for vaccine development against S. flexneri infection. The predicted immunogenic epitopes can also be used for development of peptide vaccines or multi-epitope vaccines against human shigellosis. Reverse vaccinology is a promising strategy for the discovery of potential vaccine candidates which can be used for future vaccine development against global persistent infections.
Collapse
Affiliation(s)
- Chiuan Yee Leow
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Ada Kazi
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | | | - Candy Chuah
- School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Boon Huat Lim
- School of Health Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Chiuan Herng Leow
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Gelugor, Malaysia
| | | |
Collapse
|
17
|
Kazi A, Hisyam Ismail CMK, Anthony AA, Chuah C, Leow CH, Lim BH, Banga Singh KK, Leow CY. Designing and evaluation of an antibody-targeted chimeric recombinant vaccine encoding Shigella flexneri outer membrane antigens. INFECTION GENETICS AND EVOLUTION 2020; 80:104176. [PMID: 31923724 DOI: 10.1016/j.meegid.2020.104176] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 01/03/2020] [Accepted: 01/05/2020] [Indexed: 11/16/2022]
Abstract
Shigellosis is one of the most common diseases found in the developing countries, especially those countries that are prone flood. The causative agent for this disease is the Shigella species. This organism is one of the third most common enteropathogens responsible for childhood diarrhea. Since Shigella can survive gastric acidity and is an intracellular pathogen, it becomes difficult to treat. Also, uncontrolled use of antibiotics has led to development of resistant strains which poses a threat to public health. Therefore, there is a need for long term control of Shigella infection which can be achieved by designing a proper and effective vaccine. In this study, emphasis was made on designing a candidate that could elicit both B-cell and T-cell immune response. Hence B- and T-cell epitopes of outer membrane channel protein (OM) and putative lipoprotein (PL) from S. flexneri 2a were computationally predicted using immunoinformatics approach and a chimeric construct (chimeric-OP) containing the immunogenic epitopes selected from OM and PL was designed, cloned and expressed in E. coli system. The immunogenicity of the recombinant chimeric-OP was assessed using Shigella antigen infected rabbit antibody. The result showed that the chimeric-OP was a synthetic peptide candidate suitable for the development of vaccine and immunodiagnostics against Shigella infection.
Collapse
Affiliation(s)
- Ada Kazi
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Kelantan, Malaysia
| | | | - Amy Amilda Anthony
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Candy Chuah
- School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Chiuan Herng Leow
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Penang, Malaysia
| | - Boon Huat Lim
- School of Health Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | | | - Chiuan Yee Leow
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Kelantan, Malaysia.
| |
Collapse
|
18
|
Evaluation of the protective immunity of Riemerella anatipestifer OmpA. Appl Microbiol Biotechnol 2019; 104:1273-1281. [PMID: 31865436 DOI: 10.1007/s00253-019-10294-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 11/21/2019] [Accepted: 12/03/2019] [Indexed: 10/25/2022]
Abstract
Riemerella anatipestifer is responsible for an economically important disease of commercially raised ducks. No or only few cross-protection was observed between different serotypes of R. anatipestifer strains, and so far no protective antigen in this bacterium has been identified. OmpA is a predominant immunogenic protein of R. anatipestifer, and within the 1467 bp ompA ORF (ompA1467), there is another 1164 bp ORF (ompA1164) with the same C-terminal. In this study, our results showed that the full sequence of ompA1467 from some R. anatipestifer strains with different serotypes shared the same amino acid sequence. Animal experiments showed that the soluble recombinant protein rOmpA1164, but not rOmpA1467, could provide partial protective immunity against challenge. Moreover, there was no significant difference in protective immunity between ducklings immunized with Th4△ompA bacterin and those immunized with Th4 bacterin. In addition, OmpA1467 was the main existing form of OmpA in R. anatipestifer cells by gel electrophoresis and western blot analyses. The results suggested that OmpA1467 was not a protective antigen of R. anatipestifer, and antibodies against proteins other than OmpA play a critical role in the process of anti-R. anatipestifer infection.
Collapse
|
19
|
Nosrati M, Hajizade A, Nazarian S, Amani J, Namvar Vansofla A, Tarverdizadeh Y. Designing a multi-epitope vaccine for cross-protection against Shigella spp: An immunoinformatics and structural vaccinology study. Mol Immunol 2019; 116:106-116. [PMID: 31634814 DOI: 10.1016/j.molimm.2019.09.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 07/27/2019] [Accepted: 09/14/2019] [Indexed: 12/28/2022]
Abstract
Shigellosis is a severe diarrheal disease with high mortality and morbidity rate. Until now, there is no approved vaccine against the disease. Therefore, the present study was planned to design a novel multi-epitope vaccine against Shigella spp., the causative agents of the disease based on the immunoinformatic tools. For this end, firstly seven conserved antigens of the bacteria, including IpaA, IpaB, IpaC, IpaD, OmpC, OmpF and VirG were selected. Then, linear B-cell epitope mapping of these proteins was carried out and top-ranked and shared epitopes were selected based on antigenicity, allergenicity, stability, toxicity and physicochemical properties for further analysis. In next step, B-cell derived T-cell epitopes were determined and appropriate epitopes were selected for incorporation into the final construct. Moreover, the selected epitopes and two mucosal adjuvants including ctxB and LT-IIc were joined using appropriate linkers. The three dimensional structure of the final construct was modeled and evaluated in term of structural quality and presence of conformational B-cell epitopes. Furthermore, binding affinity of the proposed vaccine to MHC I and II molecules were evaluated through molecular docking method using Hex 8.0. as well as the stability of the vaccine-MHC complexes was monitored by molecular dynamics method using the NAMD graphical user interface embedded in visual molecular dynamics. Finally, to evaluate the immunogenicity of the designed protein, the protein was administered to BALB/c mice and the serum IgG was determined by ELISA. The results indicated that the proposed vaccine has high structural quality and binding affinity to both MHC I and II molecules. Moreover, molecular dynamics studies confirmed that the vaccine-MHC docked complexes were stable during simulation time. Animal study showed that the proposed protein is able to evoke mice's humoral immune response. In sum, the results suggested that the proposed candidate vaccine could be considered as a promising anti-shigellosis vaccine.
Collapse
Affiliation(s)
- Mokhtar Nosrati
- Department of Biotechnology, Faculty of Advanced Sciences and Technologies, University of Isfahan, Isfahan, Iran
| | - Abbas Hajizade
- Biology Research Center, Faculty of Basic Sciences, Imam Hossein University, Tehran, Iran.
| | - Shahram Nazarian
- Biology Research Center, Faculty of Basic Sciences, Imam Hossein University, Tehran, Iran
| | - Jafar Amani
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Amir Namvar Vansofla
- Biology Research Center, Faculty of Basic Sciences, Imam Hossein University, Tehran, Iran
| | - Yousof Tarverdizadeh
- Biology Research Center, Faculty of Basic Sciences, Imam Hossein University, Tehran, Iran
| |
Collapse
|
20
|
Yagnik B, Sharma D, Padh H, Desai P. Oral immunization with LacVax® OmpA induces protective immune response against Shigella flexneri 2a ATCC 12022 in a murine model. Vaccine 2019; 37:3097-3105. [PMID: 31047673 PMCID: PMC7115592 DOI: 10.1016/j.vaccine.2019.04.053] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 04/15/2019] [Accepted: 04/18/2019] [Indexed: 02/05/2023]
Abstract
Shigellosis is an acute invasive disease of the lower intestine, which afflicts millions of people worldwide with an estimated one million fatalities per annum. Despite of extensive research during the last two decades, a vaccine against multi-drug resistant Shigella is not yet available in the market. To provide a safe, effective and broad-spectrum vaccine against Shigella, we explored food grade bacteria Lactococcus lactis (L. lactis) for the delivery of conserved antigenic protein; Outer membrane protein A (OmpA) to the mucosal sites for effective elicitation of systemic and mucosal immunity. We have previously confirmed the immunogenic potential of recombinant L. lactis expressing OmpA (LacVax® OmpA) in BALB/c mice. In the present study, we have characterized the humoral and cellular immune profile of LacVax® OmpA and assessed its protective efficacy using a newly developed human like murine shigellosis model. The significant increase in OmpA specific serum IgG, fecal sIgA and a Th1 dominant immune response (indicated by high INF-γ/IL-4 ratio) in LacVax® OmpA immunized mice revealed successful activation of humoral and cellular immunity. The LacVax® OmpA immunized animals were also protected from human-like shigellosis when challenged with S. flexneri 2a ATCC 12022. The antigen specific serum IgG, fecal sIgA, INF-γ and IL-10 levels were found to be the significant correlates of protection. Collectively these results suggest that the LacVax® OmpA is a promising prophylactic candidate against shigellosis. However, the protective efficacy of LacVax® OmpA in the higher animals would further strengthen its future application in humans.
Collapse
Affiliation(s)
- Bhrugu Yagnik
- Department of Cell and Molecular Biology, B. V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Ahmedabad, Gujarat, India; B. R. D. School of Biosciences, Sardar Patel University, Vallabh Vidhyanagar, Gujarat, India
| | - Drashya Sharma
- Department of Cell and Molecular Biology, B. V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Ahmedabad, Gujarat, India; B. R. D. School of Biosciences, Sardar Patel University, Vallabh Vidhyanagar, Gujarat, India
| | - Harish Padh
- Sardar Patel University, Vallabh Vidhyanagar, Gujarat, India
| | - Priti Desai
- Department of Cell and Molecular Biology, B. V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Ahmedabad, Gujarat, India; Department of Biological Sciences and Biotechnology, Institute of Advanced Research (IAR), Koba, Gandhinagar, Gujarat, India.
| |
Collapse
|
21
|
Li Y, Tang X, Zhao Z, Wang H, Wang X, Shang X, Liu P, Kou Z, Jiang Y, Li Y. Intranasal immunization with recombinant outer membrane protein A induces protective immune response against Stenotrophomonas maltophilia infection. PLoS One 2019; 14:e0214596. [PMID: 30934008 PMCID: PMC6443155 DOI: 10.1371/journal.pone.0214596] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 03/17/2019] [Indexed: 11/18/2022] Open
Abstract
Stenotrophomonas maltophilia (S. maltophilia), a multi-drug resistant opportunistic pathogen, is associated with nosocomial and community-acquired infections. Preventive and therapeutic strategies for such infections are greatly needed. In this study, sequence alignment analysis revealed that Outer membrane protein A (OmpA) was highly conserved among S. maltophilia strains but shared no significant similarity with human and mouse proteomes. In mice, intranasal immunization with S. maltophilia recombinant OmpA (rOmpA) without additional adjuvant induced sustained mucosal and systemic rOmpA-specific antibody responses. Treatment with rOmpA stimulated significantly higher levels of secretion of IFN-γ, IL-2, and IL-17A (All P<0.05) from the primary splenocytes isolated from rOmpA-immunized mice than from the primary splenocytes isolated from PBS-immunized mice. Furthermore, mice immunized with rOmpA showed significantly reduced bacterial burden in the lung and reduced levels of pro-inflammatory cytokines (TNF-α and IL-6) in bronchoalveolar lavage fluid (BALF) 24 hours after intranasal S. maltophilia infection, indicating that immunization with rOmpA may have protective effects against S. maltophilia challenge in mice. Our findings suggest that intranasal immunization with rOmpA may induce mucosal and systemic immune responses in mice, trigger Th1- and Th17-mediated cellular immune responses, and thus stimulate host immune defense against S. maltophilia infection. These results also demonstrate that intranasal vaccination may offer an alternative approach to current strategies since it induces a mucosal as well as a systemic immune response.
Collapse
Affiliation(s)
- Yan Li
- No 307 Hospital of PLA of Anhui Medical University, Hefei, China.,Department of Critical Care Medicine, No 307 Hospital of PLA, Beijing, China.,Department of Respiratory and Digestive, Fengyang First People's Hospital, Fengyang, Anhui, China
| | - Xueping Tang
- Department of Critical Care Medicine, No 307 Hospital of PLA, Beijing, China
| | - Zunquan Zhao
- The Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Hui Wang
- The Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Xin Wang
- Department of Critical Care Medicine, No 307 Hospital of PLA, Beijing, China
| | - Xueyi Shang
- Department of Critical Care Medicine, No 307 Hospital of PLA, Beijing, China
| | - Peng Liu
- The Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Zhihua Kou
- The Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Yongqiang Jiang
- The Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Yan Li
- No 307 Hospital of PLA of Anhui Medical University, Hefei, China.,Department of Critical Care Medicine, No 307 Hospital of PLA, Beijing, China
| |
Collapse
|
22
|
Koestler BJ, Ward CM, Fisher CR, Rajan A, Maresso AW, Payne SM. Human Intestinal Enteroids as a Model System of Shigella Pathogenesis. Infect Immun 2019; 87:e00733-18. [PMID: 30642906 PMCID: PMC6434139 DOI: 10.1128/iai.00733-18] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 01/06/2019] [Indexed: 01/01/2023] Open
Abstract
The enteric bacterium and intracellular human pathogen Shigella causes hundreds of millions of cases of the diarrheal disease shigellosis per year worldwide. Shigella is acquired by ingestion of contaminated food or water; upon reaching the colon, the bacteria invade colonic epithelial cells, replicate intracellularly, spread to adjacent cells, and provoke an intense inflammatory response. There is no animal model that faithfully recapitulates human disease; thus, cultured cells have been used to model Shigella pathogenesis. However, the use of transformed cells in culture does not provide the same environment to the bacteria as the normal human intestinal epithelium. Recent advances in tissue culture now enable the cultivation of human intestinal enteroids (HIEs), which are derived from human intestinal stem cells, grown ex vivo, and then differentiated into "mini-intestines." Here, we demonstrate that HIEs can be used to model Shigella pathogenesis. We show that Shigella flexneri invades polarized HIE monolayers preferentially via the basolateral surface. After S. flexneri invades HIE monolayers, S. flexneri replicates within HIE cells and forms actin tails. S. flexneri also increases the expression of HIE proinflammatory signals and the amino acid transporter SLC7A5. Finally, we demonstrate that disruption of HIE tight junctions enables S. flexneri invasion via the apical surface.
Collapse
Affiliation(s)
- Benjamin J Koestler
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas, USA
| | - Cara M Ward
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas, USA
| | - C R Fisher
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas, USA
| | - Anubama Rajan
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Anthony W Maresso
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Shelley M Payne
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
23
|
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: 0.6] [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
|
24
|
Q.S. Medeiros PH, Ledwaba SE, Bolick DT, Giallourou N, Yum LK, Costa DV, Oriá RB, Barry EM, Swann JR, Lima AÂM, Agaisse H, Guerrant RL. A murine model of diarrhea, growth impairment and metabolic disturbances with Shigella flexneri infection and the role of zinc deficiency. Gut Microbes 2019; 10:615-630. [PMID: 30712505 PMCID: PMC6748602 DOI: 10.1080/19490976.2018.1564430] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Shigella is one of the major enteric pathogens worldwide. We present a murine model of S. flexneri infection and investigate the role of zinc deficiency (ZD). C57BL/6 mice fed either standard chow (HC) or ZD diets were pretreated with an antibiotic cocktail and received S. flexneri strain 2457T orally. Antibiotic pre-treated ZD mice showed higher S. flexneri colonization than non-treated mice. ZD mice showed persistent colonization for at least 50 days post-infection (pi). S. flexneri-infected mice showed significant weight loss, diarrhea and increased levels of fecal MPO and LCN in both HC and ZD fed mice. S. flexneri preferentially colonized the colon, caused epithelial disruption and inflammatory cell infiltrate, and promoted cytokine production which correlated with weight loss and histopathological changes. Infection with S. flexneri ΔmxiG (critical for type 3 secretion system) did not cause weight loss or diarrhea, and had decreased stool shedding duration and tissue burden. Several biochemical changes related to energy, inflammation and gut-microbial metabolism were observed. Zinc supplementation increased weight gains and reduced intestinal inflammation and stool shedding in ZD infected mice. In conclusion, young antibiotic-treated mice provide a new model of oral S. flexneri infection, with ZD promoting prolonged infection outcomes.
Collapse
Affiliation(s)
- Pedro Henrique Q.S. Medeiros
- Center for Global Health, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, USA,Institute of Biomedicine, Federal University of Ceara, Fortaleza, Brazil,CONTACT Pedro Henrique Q.S. Medeiros Center for Global Health, Division of Infectious Diseases and International Health, University of Virginia, 345 Crispell Drive, MR6 Room 2711, Charlottesville, VA, USA
| | - Solanka E. Ledwaba
- Center for Global Health, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, USA
| | - David T. Bolick
- Center for Global Health, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, USA
| | - Natasa Giallourou
- Division of Computational and Systems Medicine, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Lauren K. Yum
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, USA
| | - Deiziane V.S. Costa
- Center for Global Health, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, USA,Institute of Biomedicine, Federal University of Ceara, Fortaleza, Brazil
| | - Reinaldo B. Oriá
- Center for Global Health, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, USA,Institute of Biomedicine, Federal University of Ceara, Fortaleza, Brazil
| | - Eileen M. Barry
- Center for Vaccine Development, University of Maryland, Baltimore, USA
| | - Jonathan R. Swann
- Division of Computational and Systems Medicine, Department of Surgery and Cancer, Imperial College London, London, UK
| | | | - Hervé Agaisse
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, USA
| | - Richard L. Guerrant
- Center for Global Health, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, USA
| |
Collapse
|
25
|
Renu S, Markazi AD, Dhakal S, Lakshmanappa YS, Gourapura SR, Shanmugasundaram R, Senapati S, Narasimhan B, Selvaraj RK, Renukaradhya GJ. Surface engineered polyanhydride-based oral Salmonella subunit nanovaccine for poultry. Int J Nanomedicine 2018; 13:8195-8215. [PMID: 30555234 PMCID: PMC6280892 DOI: 10.2147/ijn.s185588] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
PURPOSE Salmonellosis is a severe economic threat in poultry and a public health concern. Currently available vaccines are ineffective, and thus, developing effective oral Salmonella vaccine is warranted. Especially, a potent oral vaccine such as the mucoadhesive polyanhydride nanoparticle (PNP) protects the vaccine cargo and delivers to intestinal immune sites to elicit robust mucosal immunity and mitigate Salmonella colonization and shedding. MATERIALS AND METHODS We designed a Salmonella subunit vaccine using PNP containing immunogenic Salmonella outer membrane proteins (OMPs) and flagellar (F) protein-entrapped and surface F-protein-coated PNPs (OMPs-F-PNPs) using a solvent displacement method. Using high-throughput techniques, we characterized the OMPs-F-PNPs physicochemical properties and analyzed its efficacy in layer birds vaccinated orally. RESULTS The candidate vaccine was resistant in acidic microenvironment and had ideal physicochemical properties for oral delivery in terms of particle size, charge, morphology, biocompatibility, and pH stability. In vitro, in vivo, and ex vivo studies showed that F-protein surface-anchored nanoparticles were better targeted to chicken immune cells in peripheral blood and splenocytes and intestinal Peyer's patch sites. In layer chickens inoculated orally with OMPs-F-PNPs, substantially higher OMPs-specific IgG response and secretion of Th1 cytokine IFN-γ in the serum, enhanced CD8+/CD4+ cell ratio in spleen, and increased OMPs-specific lymphocyte proliferation were observed. OMPs-F-PNPs vaccination also upregulated the expression of toll-like receptor (TLR)-2 and -4, TGF-β, and IL-4 cytokines' genes in chicken cecal tonsils (lymphoid tissues). Importantly, OMPs-F-PNPs vaccine cleared Salmonella cecal colonization in 33% of vaccinated birds. CONCLUSION This pilot in vivo study demonstrated the targeted delivery of OMPs-F-PNPs to ileum mucosal immune sites of chickens and induced specific immune response to mitigate Salmonella colonization in intestines.
Collapse
Affiliation(s)
- Sankar Renu
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, USA,
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA,
| | - Ashley D Markazi
- Department of Animal Sciences, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, USA
| | - Santosh Dhakal
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, USA,
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA,
| | - Yashavanth S Lakshmanappa
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, USA,
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA,
| | - Suren R Gourapura
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, USA,
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA,
| | - Revathi Shanmugasundaram
- Department of Animal Sciences, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, USA
| | - Sujata Senapati
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011, USA
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011, USA
| | - Ramesh K Selvaraj
- Department of Poultry Science, University of Georgia, Athens, GA 30602, USA,
| | - Gourapura J Renukaradhya
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH 44691, USA,
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA,
| |
Collapse
|
26
|
Yagnik B, Sharma D, Padh H, Desai P. In vivo delivery of pPERDBY to BALB/c mice by LacVax ® DNA-I and comparison of elicited immune response with conventional immunization methods. Gene Ther 2018; 25:485-496. [PMID: 30108273 DOI: 10.1038/s41434-018-0033-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 05/29/2018] [Accepted: 05/30/2018] [Indexed: 01/17/2023]
Abstract
The non-invasive food grade Lactococcus lactis (L. lactis) represents a safe and attractive alternative to invasive pathogens for the delivery of plasmid DNA at mucosal sites. We have earlier shown the DNA delivery potential of r-L. lactis harboring DNA vaccine reporter plasmid; pPERDBY in vitro. In the present work, we examined in vivo delivery potential of food grade non-invasive r-L. lactis::pPERDBY (LacVax® DNA-I) in BALB/c mice. Moreover, using EGFP as a model antigen, we also characterized and compared the immune response elicited by LacVax® DNA-I with other conventional vaccination approaches using protein and naked DNA immunization. The presence of antigen-specific serum IgG and fecal secretory IgA (sIgA) antibodies demonstrated in vivo DNA delivery and immune elicitation potential of the developed LacVax® DNA-I. As compared with intramuscular injection, oral delivery of pPERDBY via L. lactis resulted in a significantly rapid increase in IgG and higher sIgA titers, indicating the immunogenic and immunostimulatory properties of the LacVax® DNA-I. The needle-free immunization with LacVax® DNA-I led to increased production of IL-4, an indicator of Th2 screwed response. To the best of our knowledge, this report for the first time outlines comparison of orally administered LacVax® DNA-I with other conventional vaccination approaches.
Collapse
Affiliation(s)
- Bhrugu Yagnik
- Department of Cell and Molecular Biology, B. V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Ahmedabad, Gujarat, India.,B. R. D. School of Biosciences, Sardar Patel University, Vallabh Vidhyanagar, Gujarat, India
| | - Drashya Sharma
- Department of Cell and Molecular Biology, B. V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Ahmedabad, Gujarat, India.,B. R. D. School of Biosciences, Sardar Patel University, Vallabh Vidhyanagar, Gujarat, India
| | - Harish Padh
- Sardar Patel University, Vallabh Vidhyanagar, Gujarat, India
| | - Priti Desai
- Department of Cell and Molecular Biology, B. V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Ahmedabad, Gujarat, India. .,School of Biological Sciences & Biotechnology, Institute of Advanced Research, Koba, Gandhinagar, Gujarat, India.
| |
Collapse
|
27
|
Jarząb A, Witkowska D, Ziomek E, Setner B, Czajkowska A, Dorot M, Szewczuk Z, Gamian A. Cyclic OmpC peptidic epitope conjugated to tetanus toxoid as a potential vaccine candidate against shigellosis. Vaccine 2018; 36:4641-4649. [PMID: 29960802 DOI: 10.1016/j.vaccine.2018.06.037] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 05/22/2018] [Accepted: 06/17/2018] [Indexed: 11/29/2022]
Abstract
In earlier works we have described that mice immunized with outer membrane protein OmpC survive the challenge with live Shigella flexnerii 3a. We have also identified conformational epitope of this protein, that was recognized by mice antibodies. The aim of current work was to investigate whether synthetic OmpC epitope homologs can elicit immunological response sufficient in protecting mice against shigellosis. Several linear peptides containing RYDERY motif were synthesized and conjugated to poly-lysine. These conjugates appeared to be poor immunogens and to boost the immunological response an addition of the adjuvant (MPL) was required. Unfortunately, the MPL alone caused a very high immunological reaction that was masking response to peptidic epitope. Under those circumstances we used tetanus toxoid (TT) as the carrier protein for the peptides and the agent stimulating immunological response. Series of cyclic peptides, homologs of the OmpC main epitope were synthesized and conjugated to TT. The loop size in cyclic peptides varied by number of glycine residues, i.e., 1-3 residues added to the GLNRYDERYIGK motif. The linear GLNRYDERYIGC-TT was also prepared as the control. The latter conjugate gave the highest immunological response, followed by the cyclic-GGLNRYDERYIGC-TT and cyclic-GLNRYDERYIGC-TT. The third peptide, cyclic-GGGLNRYDERYIGC-TT, gave a very low response, although it was the most resistant to proteolysis. However, antibodies obtained against cyclic-GGLNRYDERYIGC-TT were more potent to recognize both OmpC and Shigella flexnerii 3a cells than the antibodies against linear GLNRYDERYIGC-TT. Furthermore, the monoclonal antibodies raised against linear GLNRYDERYIGC-TT showed 20-fold lower dissociation constant (KD) than the naturally occurring polyclonal antibodies from umbilical cord sera. Monoclonal antibodies also gave a weaker signal in electron microscope than mice and human polyclonal antibodies. In overall, our results point to cyclic peptides as better candidates for a vaccine development, since they are eliciting production of the higher affinity antibodies against Shigella cells and OmpC.
Collapse
Affiliation(s)
- Anna Jarząb
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland; Wroclaw Research Center EIT+, Wroclaw, Poland
| | - Danuta Witkowska
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland; Wroclaw Research Center EIT+, Wroclaw, Poland.
| | | | - Bartosz Setner
- Faculty of Chemistry, University of Wroclaw, Wroclaw, Poland
| | - Aleksandra Czajkowska
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Małgorzata Dorot
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | | | - Andrzej Gamian
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland; Wroclaw Research Center EIT+, Wroclaw, Poland
| |
Collapse
|
28
|
Farhani I, Nezafat N, Mahmoodi S. Designing a Novel Multi-epitope Peptide Vaccine Against Pathogenic Shigella spp. Based Immunoinformatics Approaches. Int J Pept Res Ther 2018. [DOI: 10.1007/s10989-018-9698-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
29
|
Cheng ZX, Chu X, Wang SN, Peng XX, Li H. Six genes of ompA family shuffling for development of polyvalent vaccines against Vibrio alginolyticus and Edwardsiella tarda. FISH & SHELLFISH IMMUNOLOGY 2018; 75:308-315. [PMID: 29438846 DOI: 10.1016/j.fsi.2018.02.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 02/08/2018] [Accepted: 02/09/2018] [Indexed: 06/08/2023]
Abstract
Polyvalent vaccines against more than one species of pathogens are especially important due to the complex ecosystem in aquaculture. We have previously shown that the development of polyvalent vaccines by shuffling six ompA genes from different bacteria with V. parahaemolyticus VP0764 primers. Here, we used the same 6 genes, V. alginolyticus VA0764 and VA1186, V. parahaemolyticus VP0764 and VP1186, E. tarda ompA and E. coli ompA, but with E. tarda ompA primers to develop new polyvalent vaccines. By this approach, we identified 7 potential polyvalent vaccines that were effective against both V. alginolyticus and E. tarda infections. Furthermore, the innate immunity triggered by the vaccines were also explored in three groups, no protection (group I), protection against V. alginolyticus (group II), and protection against both V. alginolyticus and E. tarda (group III). The transcription of IL-1β, IL-6, IL-8, C3b and NF-kB were significantly increased in group II and group III but not group I, where the expression level of group III was higher than group II. In addition, differential activities of succinate dehydrogenase were detected among the three groups. These results indicate the expansion of polyvalent vaccine reservoir with the same shuffling genes but different primers, and promote the understanding of the mechanisms of polyvalent vaccines based on vaccine-induced innate immunity.
Collapse
Affiliation(s)
- Zhi-Xue Cheng
- Center for Proteomics and Metabolomics, State Key Laboratory of Bio-Control, Guangdong Province Key Laboratory for Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, University City, Guangzhou, 510006, PR China
| | - Xiao Chu
- Center for Proteomics and Metabolomics, State Key Laboratory of Bio-Control, Guangdong Province Key Laboratory for Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, University City, Guangzhou, 510006, PR China
| | - Sheng-Nan Wang
- Center for Proteomics and Metabolomics, State Key Laboratory of Bio-Control, Guangdong Province Key Laboratory for Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, University City, Guangzhou, 510006, PR China
| | - Xuan-Xian Peng
- Center for Proteomics and Metabolomics, State Key Laboratory of Bio-Control, Guangdong Province Key Laboratory for Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, University City, Guangzhou, 510006, PR China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, PR China.
| | - Hui Li
- Center for Proteomics and Metabolomics, State Key Laboratory of Bio-Control, Guangdong Province Key Laboratory for Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, University City, Guangzhou, 510006, PR China.
| |
Collapse
|
30
|
Assembly, Biochemical Characterization, Immunogenicity, Adjuvanticity, and Efficacy of Shigella Artificial Invaplex. mSphere 2018; 3:mSphere00583-17. [PMID: 29600284 PMCID: PMC5874444 DOI: 10.1128/msphere.00583-17] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 02/02/2018] [Indexed: 11/20/2022] Open
Abstract
The native Invaplex (InvaplexNAT) vaccine and adjuvant is an ion exchange-purified product derived from the water extract of virulent Shigella species. The key component of InvaplexNAT is a high-molecular-mass complex (HMMC) consisting of the Shigella lipopolysaccharide (LPS) and the invasin proteins IpaB and IpaC. To improve product purity and immunogenicity, artificial Invaplex (InvaplexAR) was developed using recombinant IpaB and IpaC proteins and purified Shigella LPS to assemble an HMMC consisting of all three components. Characterization of InvaplexAR by various methods demonstrated similar characteristics as the previously reported HMMC in InvaplexNAT. The well-defined InvaplexAR vaccine consistently contained greater quantities of IpaB, IpaC, and LPS than InvaplexNAT. InvaplexAR and InvaplexNAT immunogenicities were compared in mouse and guinea pig dose escalation studies. In both models, immunization induced antibody responses specific for InvaplexNAT and LPS while InvaplexAR induced markedly higher anti-IpaB and -IpaC serum IgG and IgA endpoint titers. In the murine model, homologous protection was achieved with 10-fold less InvaplexAR than InvaplexNAT and mice receiving InvaplexAR lost significantly less weight than mice receiving the same amount of InvaplexNAT. Moreover, mice immunized with InvaplexAR were protected from challenge with both homologous and heterologous Shigella serotypes. Guinea pigs receiving approximately 5-fold less InvaplexAR compared to cohorts immunized with InvaplexNAT were protected from ocular challenge. Furthermore, adjuvanticity previously attributed to InvaplexNAT was retained with InvaplexAR. The second-generation Shigella Invaplex vaccine, InvaplexAR, offers significant advantages over InvaplexNAT in reproducibility, flexible yet defined composition, immunogenicity, and protective efficacy. IMPORTANCEShigella species are bacteria that cause severe diarrheal disease worldwide, primarily in young children. Treatment of shigellosis includes oral fluids and antibiotics, but the high burden of disease, increasing prevalence of antibiotic resistance, and long-term health consequences clearly warrant the development of an effective vaccine. One Shigella vaccine under development is termed the invasin complex or Invaplex and is designed to drive an immune response to specific antigens of the bacteria in an effort to protect an individual from infection. The work presented here describes the production and evaluation of a new generation of Invaplex. The improved vaccine stimulates the production of antibodies in immunized mice and guinea pigs and protects these animals from Shigella infection. The next step in the product's development will be to test the safety and immune response induced in humans immunized with Invaplex.
Collapse
|
31
|
Wang SN, Cheng ZX, Ling XP, Chu X, Peng XX, Li H. Construction, immune protection and innate immune response of shuffled polyvalent ompAs vaccines. FISH & SHELLFISH IMMUNOLOGY 2018; 74:325-331. [PMID: 29289655 DOI: 10.1016/j.fsi.2017.12.048] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 12/15/2017] [Accepted: 12/22/2017] [Indexed: 06/07/2023]
Abstract
Our previous studies demonstrated that molecular breeding via DNA shuffling directs the evolution of polyvalent vaccines with desired traits, which leads to generation of polyvalent ompA vaccines using Vibrio alginolyticus VA0764 primers. Here, we replaced VA0764 primers with Edwardsiella tarda ompA primers to generate new polyvalent ompA vaccines by DNA shuffling of the same five ompA genes from four species of bacteria E. tarda, V. parahaemolyticus, V. alginolyticus and Escherichia coli. We identified four polyvalent vaccine candidates from a eukaryotic expressing library EompAs-FE containing 82 ompAs using active immune protection against V. alginolyticus and E. tarda. Furthermore, we explored mechanisms of polyvalent vaccine candidates by investigation of the innate immune response to these ompAs, and found that expression of IL-1β, IL-8, IL-15, COX-2, IFN-γ, TLR-1, TLR-3 and C3b genes was elevated as a characteristic feature of these polyvalent vaccine candidates. These results indicate that use of different primers to construct a DNA library selects new evolution of polyvalent vaccines with desired traits, and polyvalent ompA vaccines elicit high innate immune response.
Collapse
Affiliation(s)
- Sheng-Nan Wang
- Center for Proteomics and Metabolomics, State Key Laboratory of Bio-Control, Guangdong Province Key Laboratory for Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, University City, Guangzhou 510006, People's Republic of China
| | - Zhi-Xue Cheng
- Center for Proteomics and Metabolomics, State Key Laboratory of Bio-Control, Guangdong Province Key Laboratory for Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, University City, Guangzhou 510006, People's Republic of China
| | - Xiao-Peng Ling
- Center for Proteomics and Metabolomics, State Key Laboratory of Bio-Control, Guangdong Province Key Laboratory for Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, University City, Guangzhou 510006, People's Republic of China
| | - Xiao Chu
- Center for Proteomics and Metabolomics, State Key Laboratory of Bio-Control, Guangdong Province Key Laboratory for Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, University City, Guangzhou 510006, People's Republic of China
| | - Xuan-Xian Peng
- Center for Proteomics and Metabolomics, State Key Laboratory of Bio-Control, Guangdong Province Key Laboratory for Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, University City, Guangzhou 510006, People's Republic of China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, People's Republic of China.
| | - Hui Li
- Center for Proteomics and Metabolomics, State Key Laboratory of Bio-Control, Guangdong Province Key Laboratory for Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, University City, Guangzhou 510006, People's Republic of China.
| |
Collapse
|
32
|
Characterization, genetic regulation and production of cyanobacterial exopolysaccharides and its applicability for heavy metal removal. Carbohydr Polym 2018; 179:228-243. [DOI: 10.1016/j.carbpol.2017.09.091] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 09/15/2017] [Accepted: 09/26/2017] [Indexed: 11/18/2022]
|
33
|
Harikrishnan H, Banga Singh KK, Ismail A. Outer membrane proteins analysis of Shigella sonnei and evaluation of their antigenicity in Shigella infected individuals. PLoS One 2017; 12:e0182878. [PMID: 28846684 PMCID: PMC5573271 DOI: 10.1371/journal.pone.0182878] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 07/26/2017] [Indexed: 11/22/2022] Open
Abstract
Bacillary dysentery caused by infection with Shigella spp. remains as serious and common health problem throughout the world. It is a highly multi drug resistant organism and rarely identified from the patient at the early stage of infection. S. sonnei is the most frequently isolated species causing shigellosis in industrialized countries. The antigenicity of outer membrane protein of this pathogen expressed during human infection has not been identified to date. We have studied the antigenic outer membrane proteins expressed by S. sonnei, with the aim of identifying presence of specific IgA and IgG in human serum against the candidate protein biomarkers. Three antigenic OMPs sized 33.3, 43.8 and 100.3 kDa were uniquely recognized by IgA and IgG from patients with S. sonnei infection, and did not cross-react with sera from patients with other types of infection. The antigenic proteome data generated in this study are a first for OMPs of S. sonnei, and they provide important insights of human immune responses. Furthermore, numerous prime candidate proteins were identified which will aid the development of new diagnostic tools for the detection of S. sonnei.
Collapse
Affiliation(s)
- Hemavathy Harikrishnan
- Department of Medical Microbiology & Parasitology, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Kirnpal Kaur Banga Singh
- Department of Medical Microbiology & Parasitology, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, Kelantan, Malaysia
- * E-mail:
| | - Asma Ismail
- Office of the Vice-Chancellor, Universiti Sains Malaysia, Penang, Malaysia
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Kelantan, Malaysia
| |
Collapse
|
34
|
Van Goor A, Stromberg ZR, Mellata M. A recombinant multi-antigen vaccine with broad protection potential against avian pathogenic Escherichia coli. PLoS One 2017; 12:e0183929. [PMID: 28837660 PMCID: PMC5570496 DOI: 10.1371/journal.pone.0183929] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 08/14/2017] [Indexed: 11/18/2022] Open
Abstract
Chickens are a major source of protein worldwide, yet infectious diseases continue to threaten the poultry industry. Avian pathogenic Escherichia coli (APEC), a subgroup of extraintestinal pathogenic E. coli (ExPEC), causes colibacillosis in chickens resulting in economic loss because of treatment, condemnation of products, and death. In this study, we evaluated a recombinant antigens (rAg) vaccine combining common ExPEC surface proteins EtsC, OmpA, OmpT, and TraT for broad protective potential against APEC infections in chickens. The specific objectives were to evaluate antibody (serum) and cytokines (lymphoid organs) responses to vaccination; in vitro bactericidal ability of serum and splenocytes against multiple APEC serotypes; and in vivo protection against APEC challenge in chickens. Groups of four-day old chickens (N = 10) were vaccinated twice (two-week interval) subcutaneously with rAgs alone or in combination and CpG adjuvant or PBS (control). IgY antibody in the serum and mRNA expression of IL-1β, IL-6, IL-18, IFN-γ, IL-4, IFN-β, and IL-8 in bursa, spleen, and thymus were measured using ELISA and RT-qPCR, respectively. Serum and splenocytes were tested for their bactericidal ability in vitro against multiple APEC isolates. Vaccinated and non-vaccinated chickens were challenged with 108 CFU of APEC-O2 via air sac at 31 days post first vaccination. Vaccine protection was determined by the decrease of bacterial loads in blood and organs (lung, heart, spleen, and liver), as well as gross colibacillosis lesion scores in air sac, heart, and liver. Vaccination significantly (P < 0.05) elicited IgY against specific antigens, induced immune related mRNA expression in the spleen and bursa, reduced in vitro growth of multiple APEC serotypes, and decreased bacterial loads in the heart and spleen, and gross lesion scores of the air sac, heart and liver in chickens. The vaccine reported may be used to provide broad protection against APEC strains, increasing animal welfare and food production.
Collapse
Affiliation(s)
- Angelica Van Goor
- Department of Food Science and Human Nutrition, Iowa State University, Ames, Iowa, United States of America
| | - Zachary R. Stromberg
- Department of Food Science and Human Nutrition, Iowa State University, Ames, Iowa, United States of America
| | - Melha Mellata
- Department of Food Science and Human Nutrition, Iowa State University, Ames, Iowa, United States of America
- * E-mail:
| |
Collapse
|
35
|
In silico analysis to identify vaccine candidates common to multiple serotypes of Shigella and evaluation of their immunogenicity. PLoS One 2017; 12:e0180505. [PMID: 28767653 PMCID: PMC5540609 DOI: 10.1371/journal.pone.0180505] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 06/18/2017] [Indexed: 12/20/2022] Open
Abstract
Shigellosis or bacillary dysentery is an important cause of diarrhea, with the majority of the cases occurring in developing countries. Considering the high disease burden, increasing antibiotic resistance, serotype-specific immunity and the post-infectious sequelae associated with shigellosis, there is a pressing need of an effective vaccine against multiple serotypes of the pathogen. In the present study, we used bio-informatics approach to identify antigens shared among multiple serotypes of Shigella spp. This approach led to the identification of many immunogenic peptides. The five most promising peptides based on MHC binding efficiency were a putative lipoprotein (EL PGI I), a putative heat shock protein (EL PGI II), Spa32 (EL PGI III), IcsB (EL PGI IV) and a hypothetical protein (EL PGI V). These peptides were synthesized and the immunogenicity was evaluated in BALB/c mice by ELISA and cytokine assays. The putative heat shock protein (HSP) and the hypothetical protein elicited good humoral response, whereas putative lipoprotein, Spa32 and IcsB elicited good T-cell response as revealed by increased IFN-γ and TNF-α cytokine levels. The patient sera from confirmed cases of shigellosis were also evaluated for the presence of peptide specific antibodies with significant IgG and IgA antibodies against the HSP and the hypothetical protein, bestowing them as potential future vaccine candidates. The antigens reported in this study are novel and have not been tested as vaccine candidates against Shigella. This study offers time and cost-effective way of identifying unprecedented immunogenic antigens to be used as potential vaccine candidates. Moreover, this approach should easily be extendable to find new potential vaccine candidates for other pathogenic bacteria.
Collapse
|
36
|
Pastor Y, Camacho A, Gil AG, Ramos R, Ceráin ALD, Peñuelas I, Irache JM, Gamazo C. Effective protection of mice against Shigella flexneri with a new self-adjuvant multicomponent vaccine. J Med Microbiol 2017; 66:946-958. [PMID: 28721849 DOI: 10.1099/jmm.0.000527] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
PURPOSE The aim of this study was to develop an immunogenic protective product against Shigella flexneri by employing a simple and safe heat treatment-based strategy. METHODOLOGY The physicochemical characteristics of naturally produced (OMV) and heat-induced (HT) outer-membrane vesicles from S. flexneri were examined, including a comparison of the protein content of the products. Toxicological and biodistribution studies, and a preliminary experiment to examine the protective effectiveness of HT in a murine model of S. flexneri infection, were also included. RESULTS This method simultaneously achieves complete bacterial inactivation and the production of the HT vaccine product, leading to a safe working process. The obtained HT complex presented a similar morphology (electron microscopy) and chemical composition to the classical OMV, although it was enriched in some immunogens, such as lipoproteins, OmpA or OmpC, among others. The HT formulation was not toxic and biodistribution studies performed in mice demonstrated that the vaccine product remained in the small intestine after nasal administration. Finally, a single dose of HT administered nasally was able to protect mice against S. flexneri 2a. CONCLUSION The convenient and safe manufacturing process, and the preliminary biological evaluation, support the use of the self-adjuvanted HT complex as a new vaccine candidate to face shigellosis. Further development is required, such as additional immune analyses, to evaluate whether this new subunit vaccine can be useful in achieving full protection against Shigella.
Collapse
Affiliation(s)
- Yadira Pastor
- Department of Microbiology, Institute of Tropical Health, University of Navarra, 31008 Pamplona, Spain
| | - Ana Camacho
- Department of Microbiology, Institute of Tropical Health, University of Navarra, 31008 Pamplona, Spain
| | - Ana Gloria Gil
- Department of Toxicology, Institute of Tropical Health, University of Navarra, 31008 Pamplona, Spain
| | - Rocío Ramos
- Department of Nuclear Medicine, Clínica Universidad de Navarra, Institute of Tropical Health, University of Navarra 31008, Pamplona, Spain
| | - Adela López de Ceráin
- Department of Toxicology, Institute of Tropical Health, University of Navarra, 31008 Pamplona, Spain
| | - Iván Peñuelas
- Department of Nuclear Medicine, Clínica Universidad de Navarra, Institute of Tropical Health, University of Navarra 31008, Pamplona, Spain
| | - Juan M Irache
- Department of Pharmaceutical Technology, Institute of Tropical Health, University of Navarra, 31008 Pamplona, Spain
| | - Carlos Gamazo
- Department of Microbiology, Institute of Tropical Health, University of Navarra, 31008 Pamplona, Spain
| |
Collapse
|
37
|
Novinrooz A, Zahraei Salehi T, Firouzi R, Arabshahi S, Derakhshandeh A. In-silico design, expression, and purification of novel chimeric Escherichia coli O157:H7 OmpA fused to LTB protein in Escherichia coli. PLoS One 2017; 12:e0173761. [PMID: 28296951 PMCID: PMC5351874 DOI: 10.1371/journal.pone.0173761] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 02/27/2017] [Indexed: 01/03/2023] Open
Abstract
E. coli O157:H7, one of the major EHEC serotypes, is capable of developing bloody diarrhea, hemorrhagic colitis (HC), and fatal hemolytic uremic syndrome (HUS) and is accompanied by high annual economic loss worldwide. Due to the increased risk of HC and HUS development following antibiotic therapy, the prevention of infections caused by this pathogen is considered to be one of the most effective ways of avoiding the consequences of this infection. The main aim of the present study was to design, express, and purify a novel chimeric protein to develope human vaccine candidate against E. coli O157:H7 containing loop 2–4 of E. coli O157:H7, outer membrane protein A (OmpA), and B subunit of E. coli heat labile enterotoxin (LTB) which are connected by a flexible peptide linker. Several online databases and bioinformatics software were utilized to choose the peptide linker among 537 analyzed linkers, design the chimeric protein, and optimize the codon of the relative gene encoding this protein. Subsequently, the recombinant gene encoding OmpA-LTB was synthesized and cloned into pET-24a (+) expression vector and transferred to E. coli BL21(DE3) cells. The expression of OmpA-LTB chimeric protein was then carried out by induction of cultured E. coli Bl21 (DE3) cells with 1mM isopropyl-β-D-thiogalactopyranoside (IPTG). The purification of OmpA-LTB was then performed by nickel affinity chromatography. Expression and purification were analyzed by sodium dodecyl sulphate poly acrylamide gel electrophoresis. Moreover, the identity of the expressed protein was analyzed by western blotting. SDS-PAGE and western immunoblotting confirmed the successful expression of a 27 KDa recombinant protein after 24 hours at 37°C post-IPTG induction. OmpA-LTB was then successfully purified, using nickel affinity chromatography under denaturing conditions. The yield of purification was 12 mg per liter of culture media. Ultimately, we constructed the successful design and efficient expression and purification of OmpA-LTB divalent under the above-mentioned conditions.
Collapse
Affiliation(s)
- Aytak Novinrooz
- Department of Pathobiology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Taghi Zahraei Salehi
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
- * E-mail:
| | - Roya Firouzi
- Department of Pathobiology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Sina Arabshahi
- Department of Pathobiology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Abdollah Derakhshandeh
- Department of Pathobiology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| |
Collapse
|
38
|
Yagnik B, Sharma D, Padh H, Desai P. Immunization with r-Lactococcus lactis expressing outer membrane protein A of Shigella dysenteriae type-1: evaluation of oral and intranasal route of administration. J Appl Microbiol 2016; 122:493-505. [PMID: 27860045 DOI: 10.1111/jam.13353] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 10/24/2016] [Accepted: 11/11/2016] [Indexed: 02/07/2023]
Abstract
AIMS To evaluate the comparative immunogenic potential of food grade Lactococcus lactis expressing outer membrane protein A (OmpA) of Shigella dysenteriae type-1 (SD-1) when administered either orally or intranasally. METHODS AND RESULTS OmpA of SD-1 was cloned and expressed first in Escherichia coli and then in L. lactis. Presence of recombinant gene was confirmed by restriction enzyme digestion and immunoblot analysis. Using immobilized metal affinity chromatography, OmpA was purified from recombinant E. coliBL21 (DE3) and subcutaneously administered in BALB/c mice. Detection of OmpA-specific IgG antibodies by enzyme-linked immunosorbent assay (ELISA) confirmed the immunogenicity of OmpA. In order to establish r-L. lactis as a mucosal delivery vehicle, it was administered orally and nasally in BALB/c mice. Serum IgG and faecal IgA were assessed through ELISA to compare the relative potential of immunization routes and immunogenic potential of r-L. lactis. Immunization via the oral route proved superior to intranasal exposure. CONCLUSION Recombinant L. lactis expressing OmpA of SD-1 was found to be immunogenic. Oral administration of r-L. lactis elicited higher systemic and mucosal immune response when compared with the nasal route. SIGNIFICANCE AND IMPACT OF THE STUDY Using food grade recombinant L. lactis has implications in the development of a prophylactic against multidrug-resistant Shigella, which can be used as a prospective vaccine candidate. Evaluating mucosal routes of immunization demonstrated that the oral route of administration elicited better immune response against OmpA of Shigella.
Collapse
Affiliation(s)
- B Yagnik
- Department of Cell and Molecular Biology, B. V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Ahmedabad, 380054, Gujarat, India.,B. R. D School of Biosciences, Sardar Patel University, Vallabh Vidhyanagar, 388120, Gujarat, India
| | - D Sharma
- Department of Cell and Molecular Biology, B. V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Ahmedabad, 380054, Gujarat, India.,B. R. D School of Biosciences, Sardar Patel University, Vallabh Vidhyanagar, 388120, Gujarat, India
| | - H Padh
- Sardar Patel University, Vallabh Vidhyanagar, 388120, Gujarat, India
| | - P Desai
- Department of Cell and Molecular Biology, B. V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Ahmedabad, 380054, Gujarat, India
| |
Collapse
|
39
|
Abstract
Shigellosis remains a serious issue throughout the developing countries, particularly in children under the age of 5. Numerous strategies have been tested to develop vaccines targeting shigellosis; unfortunately despite several years of extensive research, no safe, effective, and inexpensive vaccine against shigellosis is available so far. Here, we illustrate in detail an approach to identify and establish immunogenic outer membrane proteins from Shigella flexneri 2a as subunit vaccine candidates.
Collapse
|
40
|
Dubey S, Avadhani K, Mutalik S, Sivadasan SM, Maiti B, Girisha SK, Venugopal MN, Mutoloki S, Evensen Ø, Karunasagar I, Munang’andu HM. Edwardsiella tarda OmpA Encapsulated in Chitosan Nanoparticles Shows Superior Protection over Inactivated Whole Cell Vaccine in Orally Vaccinated Fringed-Lipped Peninsula Carp (Labeo fimbriatus). Vaccines (Basel) 2016; 4:vaccines4040040. [PMID: 27827990 PMCID: PMC5192360 DOI: 10.3390/vaccines4040040] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 10/21/2016] [Accepted: 10/28/2016] [Indexed: 12/13/2022] Open
Abstract
The use of oral vaccination in finfish has lagged behind injectable vaccines for a long time as oral vaccines fall short of injection vaccines in conferring protective immunity. Biodegradable polymeric nanoparticles (NPs) have shown potential to serve as antigen delivery systems for oral vaccines. In this study the recombinant outer membrane protein A (rOmpA) of Edwardsiella tarda was encapsulated in chitosan NPs (NP-rOmpA) and used for oral vaccination of Labeo fimbriatus. The rOmpA purity was 85%, nanodiameter <500 nm, encapsulation efficiency 60.6%, zeta potential +19.05 mV, and there was an in vitro release of 49% of encapsulated antigen within 48 h post incubation in phosphate-buffered saline. Empty NPs and a non-formulated, inactivated whole cell E. tarda (IWC-ET) vaccine were used as controls. Post-vaccination antibody levels were significantly (p = 0.0458) higher in the NP-rOmpA vaccinated fish (Mean OD450 = 2.430) than in fish vaccinated with inactivated whole cell E. tarda (IWC-ET) vaccine (Mean OD450 = 1.735), which corresponded with post-challenge survival proportions (PCSP) of 73.3% and 48.28% for the NP-rOmpA and IWC-ET groups, respectively. Serum samples from NP-rOmpA-vaccinated fish had a higher inhibition rate for E. tarda growth on tryptic soy agar (TSA) than the IWC-ET group. There was no significant difference (p = 0.989) in PCSPs between fish vaccinated with empty NPs and the unvaccinated control fish, while serum from both groups showed no detectable antibodies against E. tarda. Overall, these data show that the NP-rOmpA vaccine produced higher antibody levels and had superior protection over the IWC-ET vaccine, showing that encapsulating OmpA in chitosan NPs confer improved protection against E. tarda mortality in L. fimbriatus. There is a need to elucidate the possible adjuvant effects of chitosan NPs and the immunological mechanisms of protective immunity induced by OMPs administered orally to fish.
Collapse
Affiliation(s)
- Saurabh Dubey
- Department of Fisheries Microbiology, Karnataka Veterinary, Animal & Fisheries Sciences University, College of Fisheries, Mangalore 575002, India.
- Norwegian University of Life Sciences, Faculty of Veterinary Medicine, Department of Basic Sciences and Aquatic Medicine, Section of Aquatic Medicine and Nutrition, Adamstuen Campus, Ullevålseveien 72, P.O. Box 8146, NO-0033 Dep, Oslo 0454, Norway.
| | - Kiran Avadhani
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal University, Manipal 576104, Karnataka State, India.
| | - Srinivas Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal University, Manipal 576104, Karnataka State, India.
| | - Sangeetha Madambithara Sivadasan
- Department of Fisheries Microbiology, Karnataka Veterinary, Animal & Fisheries Sciences University, College of Fisheries, Mangalore 575002, India.
| | - Biswajit Maiti
- UNESCO MIRCEN for Marine Biotechnology, Nitte University Centre for Science Education and Research, Paneer Campus, Deralakatte, Mangalore 575018, India.
| | - Shivani Kallappa Girisha
- Department of Fisheries Microbiology, Karnataka Veterinary, Animal & Fisheries Sciences University, College of Fisheries, Mangalore 575002, India.
| | - Moleyur Nagarajappa Venugopal
- Department of Fisheries Microbiology, Karnataka Veterinary, Animal & Fisheries Sciences University, College of Fisheries, Mangalore 575002, India.
| | - Stephen Mutoloki
- Norwegian University of Life Sciences, Faculty of Veterinary Medicine, Department of Basic Sciences and Aquatic Medicine, Section of Aquatic Medicine and Nutrition, Adamstuen Campus, Ullevålseveien 72, P.O. Box 8146, NO-0033 Dep, Oslo 0454, Norway.
| | - Øystein Evensen
- Norwegian University of Life Sciences, Faculty of Veterinary Medicine, Department of Basic Sciences and Aquatic Medicine, Section of Aquatic Medicine and Nutrition, Adamstuen Campus, Ullevålseveien 72, P.O. Box 8146, NO-0033 Dep, Oslo 0454, Norway.
| | - Indrani Karunasagar
- UNESCO MIRCEN for Marine Biotechnology, Nitte University Centre for Science Education and Research, Paneer Campus, Deralakatte, Mangalore 575018, India.
| | - Hetron Mweemba Munang’andu
- Norwegian University of Life Sciences, Faculty of Veterinary Medicine, Department of Basic Sciences and Aquatic Medicine, Section of Aquatic Medicine and Nutrition, Adamstuen Campus, Ullevålseveien 72, P.O. Box 8146, NO-0033 Dep, Oslo 0454, Norway.
| |
Collapse
|
41
|
Li H, Chu X, Peng B, Peng XX. DNA shuffling approach for recombinant polyvalent OmpAs against V. alginolyticus and E. tarda infections. FISH & SHELLFISH IMMUNOLOGY 2016; 58:508-513. [PMID: 27697557 DOI: 10.1016/j.fsi.2016.09.058] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 09/26/2016] [Accepted: 09/30/2016] [Indexed: 06/06/2023]
Abstract
Molecular breeding via DNA shuffling directs the evolution of vaccines with desired traits. In the present study, polyvalent OmpA vaccines were generated by DNA shuffling of five ompA genes from four species of bacteria Vibrio parahaemolyticus, V. alginolyticus, Edwardsiella tarda and Escherichia coli. First, a new hybrid OmpA was constructed using VA0764 primers and used for construction of a prokaryotic expressing library PompAs-FV containing 84 ompAs, which were validated by PCR and SDS/PAGE. Then, the 84 ompAs were used to construct a eukaryotic expressing library EompAs-FV for preparing DNA vaccines. Third, extracellular bacterium V. alginolyticus challenge post active immunization using these DNA vaccines was carried out to identify genes with high immunoprotection. Among the 84 ompAs, 17 showed higher or equal immune protection against infection caused by V. alginolyticus than control VA0764. Finally, immune protection against infection caused by intracellular bacterium Edwardsiella tarda was assessed further using the top seven out of the 17 ompAs. This led to identification of three efficient polyvalent vaccines against infections caused by the extracellular bacterium V. alginolyticus and intracellular bacterium E. tarda. In addition, we sequenced genes for understanding mechanisms of the polyvalent vaccines, but association of immune protection with mutation of gene and amino acids is not determined. These results indicate that DNA shuffling is an efficient way to develop polyvalent vaccines against microbial infections.
Collapse
Affiliation(s)
- Hui Li
- Center for Proteomics and Metabolomics, State Key Laboratory of Bio-Control, MOE Key Lab Aquatic Food Safety, Guangdong Province Key Laboratory for Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, University City, Guangzhou 510006, People's Republic of China.
| | - Xiao Chu
- Center for Proteomics and Metabolomics, State Key Laboratory of Bio-Control, MOE Key Lab Aquatic Food Safety, Guangdong Province Key Laboratory for Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, University City, Guangzhou 510006, People's Republic of China
| | - Bo Peng
- Center for Proteomics and Metabolomics, State Key Laboratory of Bio-Control, MOE Key Lab Aquatic Food Safety, Guangdong Province Key Laboratory for Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, University City, Guangzhou 510006, People's Republic of China
| | - Xuan-Xian Peng
- Center for Proteomics and Metabolomics, State Key Laboratory of Bio-Control, MOE Key Lab Aquatic Food Safety, Guangdong Province Key Laboratory for Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, University City, Guangzhou 510006, People's Republic of China
| |
Collapse
|
42
|
Shigellosis murine model established by intraperitoneal and intranasal route of administration: a comparative comprehension overview. Microbes Infect 2016; 19:47-54. [PMID: 27664045 DOI: 10.1016/j.micinf.2016.09.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 09/05/2016] [Accepted: 09/07/2016] [Indexed: 11/22/2022]
Abstract
Shigellosis, a major cause of mortality and morbidity, requires development of effective intervention strategy for which animal model mimicking human pathology is essential. Among various animal models for shigellosis, mice being more convenient have been used wherein intraperitoneal and intranasal routes are preferred. With the aim to comprehend the comparative pathophysiological indicators, we have examined relatively high and low dose of Shigella flexneri administered through intraperitoneal and intranasal routes in mice. Characterization of these two models along with the resulting pathophysiology of shigellosis adds to our understanding and offers suitable models appropriate to the objectives of the study.
Collapse
|
43
|
Construction of a recombinant Lactococcus lactis strain expressing a fusion protein of Omp22 and HpaA from Helicobacter pylori for oral vaccine development. Biotechnol Lett 2016; 38:1911-1916. [PMID: 27406731 DOI: 10.1007/s10529-016-2173-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Accepted: 07/04/2016] [Indexed: 12/22/2022]
Abstract
OBJECTIVES To develop orally administrated anti-Helicobacter pylori vaccination, a Lactococcus lactis strain was genetically constructed for fusion expression of H. pylori protective antigens HpaA and Omp22. RESULTS The fusion gene of omp22 and hpaA with an adapter encoding three glycines was cloned from a plasmid pMAL-c2x-omp22-hpaA into Escherichia coli MC1061 and L. lactis NZ3900 successively using a shutter vector pNZ8110. Expression of the fusion gene in L. lactis was induced with nisin resulting in production of proteins with molecular weights of 50 and 28 kDa. Both of them were immunoreactive with mouse anti-H. pylori sera as determined via western blotting. Oral vaccination of BALB/c mice using the L. lactis strain carrying pNZ8110-omp22-hpaA elicited significant systematic humoral immune response (P < 0.05). CONCLUSIONS This is the first report showing that a fusion protein of two H. pylori antigens was efficiently expressed in L. lactis with immunogenicity. This is a considerable step towards H. pylori vaccines.
Collapse
|
44
|
Yadav SK, Meena JK, Sharma M, Dixit A. Recombinant outer membrane protein C of Aeromonas hydrophila elicits mixed immune response and generates agglutinating antibodies. Immunol Res 2016; 64:1087-99. [DOI: 10.1007/s12026-016-8807-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
45
|
|
46
|
Li H, Chu X, Li D, Zeng ZH, Peng XX. Construction and immune protection evaluation of recombinant polyvalent OmpAs derived from genetically divergent ompA by DNA shuffling. FISH & SHELLFISH IMMUNOLOGY 2016; 49:230-236. [PMID: 26707781 DOI: 10.1016/j.fsi.2015.12.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 12/10/2015] [Accepted: 12/14/2015] [Indexed: 06/05/2023]
Abstract
A wide variety of bacterial infections is a major challenge in aquaculture. Development of polyvalent vaccines that can fight against as many pathogens as possible is especially necessary. The present study uses DNA shuffling to create a new hybrid OmpA with improved cross-protection against Vibrio alginolyticus and Edwardsiella tarda through the recombination of six OmpA genes from Vibrio parahaemolyticus, V. alginolyticus, E. tarda and Escherichia coli. Out of the 43 recombinant chimeras genes constructed using VA0764 primers, EompAs-19 was demonstrated as an ideal polyvalent vaccine against infections caused V. alginolyticus and E. tarda. Compared with VA0764, OmpAs-19 had three mutations, which may be a molecular basis of EompAs-19 as an efficient polyvalent vaccine against both V. alginolyticus and E. tarda infections. These results develop a polyvalent vaccine that prevents the infections caused by extracellular and intracellular bacteria. Thus, the present study highlights the way to develop polyvalent vaccines against microbial infections by DNA shuffling.
Collapse
Affiliation(s)
- Hui Li
- Center for Proteomics and Metabolomics, State Key Laboratory of Bio-Control, MOE Key Lab Aquatic Food Safety, School of Life Sciences, Sun Yat-sen University, University City, Guangzhou 510006, People's Republic of China.
| | - Xiao Chu
- Center for Proteomics and Metabolomics, State Key Laboratory of Bio-Control, MOE Key Lab Aquatic Food Safety, School of Life Sciences, Sun Yat-sen University, University City, Guangzhou 510006, People's Republic of China
| | - Dan Li
- Center for Proteomics and Metabolomics, State Key Laboratory of Bio-Control, MOE Key Lab Aquatic Food Safety, School of Life Sciences, Sun Yat-sen University, University City, Guangzhou 510006, People's Republic of China
| | - Zao-Hai Zeng
- Center for Proteomics and Metabolomics, State Key Laboratory of Bio-Control, MOE Key Lab Aquatic Food Safety, School of Life Sciences, Sun Yat-sen University, University City, Guangzhou 510006, People's Republic of China
| | - Xuan-Xian Peng
- Center for Proteomics and Metabolomics, State Key Laboratory of Bio-Control, MOE Key Lab Aquatic Food Safety, School of Life Sciences, Sun Yat-sen University, University City, Guangzhou 510006, People's Republic of China.
| |
Collapse
|
47
|
Abstract
The original scientific strategy behind vaccinology has historically been to “isolate, inactivate, and inject,” first invoked by Louis Pasteur.
Collapse
|
48
|
Nag D, Sinha R, Mitra S, Barman S, Takeda Y, Shinoda S, Chakrabarti MK, Koley H. Heat killed multi-serotype Shigella immunogens induced humoral immunity and protection against heterologous challenge in rabbit model. Immunobiology 2015. [PMID: 26210044 DOI: 10.1016/j.imbio.2015.07.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Recently we have shown the homologous protective efficacy of heat killed multi-serotype Shigella (HKMS) immunogens in a guinea pig colitis model. In our present study, we have advanced our research by immunizing rabbits with a reduced number of oral doses and evaluating the host's adaptive immune responses. The duration of immunogenicity and subsequently protective efficacy was determined against wild type heterologous Shigella strains in a rabbit luminal model. After three successive oral immunizations with HKMS immunogens, serum and lymphocyte supernatant antibody titer against the heterologous shigellae were reciprocally increased and remained at an elevated level up to 180 days. Serogroup and serotype specific O-antigen of lipopolysaccharide and immunogenic proteins of heterologous challenge strains were detected by immunoblot assay. Up-regulation of IL-12p35, IFN-γ and IL-10 mRNA expression was detected in immunized rabbit peripheral blood mononuclear cells (PBMC) after stimulation with HKMS in vitro. HKMS-specific plasma cell response was confirmed by production of a relatively higher level of HKMS-specific IgG in immunized PBMC supernatant compared to control group. Furthermore, the immunized groups of rabbits exhibited complete protection against wild type heterologous shigellae challenge. Thus HKMS immunogens induced humoral and Th1-mediated adaptive immunity and provided complete protection in a rabbit model. These immunogens could be a broad spectrum non-living vaccine candidate for human use in the near future.
Collapse
Affiliation(s)
- Dhrubajyoti Nag
- Division of Bacteriology, National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Ritam Sinha
- Division of Bacteriology, National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Soma Mitra
- Division of Bacteriology, National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Soumik Barman
- Division of Bacteriology, National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Yoshifumi Takeda
- Collaborative Research Centre of Okayama University for Infectious Diseases, National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Sumio Shinoda
- Collaborative Research Centre of Okayama University for Infectious Diseases, National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - M K Chakrabarti
- Division of Pathophysiology, National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Hemanta Koley
- Division of Bacteriology, National Institute of Cholera and Enteric Diseases, Kolkata, India.
| |
Collapse
|
49
|
Kämpf MM, Braun M, Sirena D, Ihssen J, Thöny-Meyer L, Ren Q. In vivo production of a novel glycoconjugate vaccine against Shigella flexneri 2a in recombinant Escherichia coli: identification of stimulating factors for in vivo glycosylation. Microb Cell Fact 2015; 14:12. [PMID: 25612741 PMCID: PMC4308876 DOI: 10.1186/s12934-015-0195-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 01/12/2015] [Indexed: 11/26/2022] Open
Abstract
Background Glycoconjugated vaccines composed of polysaccharide antigens covalently linked to immunogenic carrier proteins have proved to belong to the most effective and safest vaccines for combating bacterial pathogens. The functional transfer of the N-glycosylation machinery from Campylobacter jejuni to the standard prokaryotic host Escherichia coli established a novel bioconjugation methodology termed bacterial glycoengineering. Results In this study, we report on the production of a new recombinant glycoconjugate vaccine against Shigella flexneri 2a representing the major serotype for global outbreaks of shigellosis. We demonstrate that S. flexneri 2a O-polysaccharides can be transferred to a detoxified variant of Pseudomonas aeruginosa carrier protein exotoxin A (EPA) by the C. jejuni oligosaccharyltransferase PglB, resulting in glycosylated EPA-2a. Moreover, we optimized the in vivo production of this novel vaccine by identification and quantitative analysis of critical process parameters for glycoprotein synthesis. It was found that sequential induction of oligosaccharyltransferase PglB and carrier protein EPA increased the specific productivity of EPA-2a by a factor of 1.6. Furthermore, by the addition of 10 g/L of the monosaccharide N-acetylglucosamine during induction, glycoconjugate vaccine yield was boosted up to 3.1-fold. The optimum concentration of Mg2+ ions for N-glycan transfer was determined to be 10 mM. Finally, optimized parameters were transferred to high cell density cultures with a 46-fold increase of overall yield of glycoconjugate compared to the one in initial shake flask production. Conclusion The present study is the first attempt to identify stimulating parameters for improved productivity of S. flexneri 2a bioconjugates. Optimization of glycosylation efficiency will ultimately foster the transfer of lab-scale expression to a cost-effective in vivo production process for a glycoconjugate vaccine against S. flexneri 2a in E. coli. This study is an important step towards this goal and provides a starting point for further optimization studies.
Collapse
Affiliation(s)
- Michael M Kämpf
- Laboratory for Biointerfaces, Swiss Federal Laboratories for Materials Science and Technology (Empa), Lerchenfeldstrasse 5, CH-9014, St. Gallen, Switzerland. .,GlycoVaxyn AG, Grabenstrasse 3, 8952, Schlieren, Switzerland.
| | - Martin Braun
- GlycoVaxyn AG, Grabenstrasse 3, 8952, Schlieren, Switzerland.
| | | | - Julian Ihssen
- Laboratory for Biointerfaces, Swiss Federal Laboratories for Materials Science and Technology (Empa), Lerchenfeldstrasse 5, CH-9014, St. Gallen, Switzerland.
| | - Linda Thöny-Meyer
- Laboratory for Biointerfaces, Swiss Federal Laboratories for Materials Science and Technology (Empa), Lerchenfeldstrasse 5, CH-9014, St. Gallen, Switzerland.
| | - Qun Ren
- Laboratory for Biointerfaces, Swiss Federal Laboratories for Materials Science and Technology (Empa), Lerchenfeldstrasse 5, CH-9014, St. Gallen, Switzerland.
| |
Collapse
|
50
|
Recombinant outer membrane protein A induces a protective immune response against Escherichia coli infection in mice. Appl Microbiol Biotechnol 2015; 99:5451-60. [PMID: 25567514 DOI: 10.1007/s00253-014-6339-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 12/12/2014] [Accepted: 12/17/2014] [Indexed: 01/19/2023]
Abstract
Pathogenic Escherichia coli (E. coli) is an important infectious Gram-negative bacterium causing millions of death every year. Outer membrane protein A (OmpA) has been suggested as a potential vaccine candidate for conferring protection against bacterial infection. In this study, a universal vaccine candidate for E. coli infection was developed and evaluated. Bioinformatics analysis revealed the OmpA protein from E. coli shares 96~100%, 90~94%, and 45% identity with Shigella, Salmonella, and Pseudomonas strains, respectively. The ompA gene was cloned from the genomic DNA of E. coli, and then the OmpA protein was expressed in BL21 (DE3) using the auto-induction method. The recombinant OmpA (rOmpA) protein had an average molecular weight of 36 kDa with the purity of 93.5%. Immunological analysis indicated that the titers of anti-rOmpA sera against rOmpA and whole cells were 1:642,000 and 1:140,000, respectively. Moreover, rOmpA not only conferred a high level of immunogenicity to protect mice against the challenge of E. coli, but also generated cross-protection against Shigella and Salmonella. The anti-rOmpA sera could enhance the phagocytic activity of neutrophils against E. coli. The survive ratios of mice immunized with rOmpA and PBS were 50% and 20% after 48 h post-challenge, indicating mice were protected from E. coli infection after immunization with rOmpA. All these results clearly indicate that rOmpA may be a promising candidate for the development of a subunit vaccine to prevent E. coli infection.
Collapse
|