1
|
Dubois V, Locht C. Mucosal Immunization Against Pertussis: Lessons From the Past and Perspectives. Front Immunol 2021; 12:701285. [PMID: 34211481 PMCID: PMC8239240 DOI: 10.3389/fimmu.2021.701285] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 05/26/2021] [Indexed: 01/11/2023] Open
Abstract
Background Current vaccination strategies against pertussis are sub-optimal. Optimal protection against Bordetella pertussis, the causative agent of pertussis, likely requires mucosal immunity. Current pertussis vaccines consist of inactivated whole B. pertussis cells or purified antigens thereof, combined with diphtheria and tetanus toxoids. Although they are highly protective against severe pertussis disease, they fail to elicit mucosal immunity. Compared to natural infection, immune responses following immunization are short-lived and fail to prevent bacterial colonization of the upper respiratory tract. To overcome these shortcomings, efforts have been made for decades, and continue to be made, toward the development of mucosal vaccines against pertussis. Objectives In this review we systematically analyzed published literature on protection conferred by mucosal immunization against pertussis. Immune responses mounted by these vaccines are summarized. Method The PubMed Library database was searched for published studies on mucosal pertussis vaccines. Eligibility criteria included mucosal administration and the evaluation of at least one outcome related to efficacy, immunogenicity and safety. Results While over 349 publications were identified by the search, only 63 studies met the eligibility criteria. All eligible studies are included here. Initial attempts of mucosal whole-cell vaccine administration in humans provided promising results, but were not followed up. More recently, diverse vaccination strategies have been tested, including non-replicating and replicating vaccine candidates given by three different mucosal routes: orally, nasally or rectally. Several adjuvants and particulate formulations were tested to enhance the efficacy of non-replicating vaccines administered mucosally. Most novel vaccine candidates were only tested in animal models, mainly mice. Only one novel mucosal vaccine candidate was tested in baboons and in human trials. Conclusion Three vaccination strategies drew our attention, as they provided protective and durable immunity in the respiratory tract, including the upper respiratory tract: acellular vaccines adjuvanted with lipopeptide LP1569 and c-di-GMP, outer membrane vesicles and the live attenuated BPZE1 vaccine. Among all experimental vaccines, BPZE1 is the only one that has advanced into clinical development.
Collapse
Affiliation(s)
- Violaine Dubois
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Camille Locht
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| |
Collapse
|
2
|
Design of a Peptide-Carrier Vaccine Based on the Highly Immunogenic Fasciola hepatica Leucine Aminopeptidase. Methods Mol Biol 2021; 2137:191-204. [PMID: 32399930 DOI: 10.1007/978-1-0716-0475-5_14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Many studies have shown that the degree of organization and repetitiveness of an antigen correlates with its efficiency to induce a B-cell response and production of neutralizing antibodies. Here we describe the design of a chimeric protein based on the hexamer form of the highly immunogenic Fasciola hepatica leucine aminopeptidase as a carrier system of small peptides with potential use as a multiepitope vaccine.
Collapse
|
3
|
Harnessing the Membrane Translocation Properties of AB Toxins for Therapeutic Applications. Toxins (Basel) 2021; 13:toxins13010036. [PMID: 33418946 PMCID: PMC7825107 DOI: 10.3390/toxins13010036] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/28/2020] [Accepted: 01/01/2021] [Indexed: 12/31/2022] Open
Abstract
Over the last few decades, proteins and peptides have become increasingly more common as FDA-approved drugs, despite their inefficient delivery due to their inability to cross the plasma membrane. In this context, bacterial two-component systems, termed AB toxins, use various protein-based membrane translocation mechanisms to deliver toxins into cells, and these mechanisms could provide new insights into the development of bio-based drug delivery systems. These toxins have great potential as therapies both because of their intrinsic properties as well as the modular characteristics of both subunits, which make them highly amenable to conjugation with various drug classes. This review focuses on the therapeutical approaches involving the internalization mechanisms of three representative AB toxins: botulinum toxin type A, anthrax toxin, and cholera toxin. We showcase several specific examples of the use of these toxins to develop new therapeutic strategies for numerous diseases and explain what makes these toxins promising tools in the development of drugs and drug delivery systems.
Collapse
|
4
|
Jobling MG, Poole ST, Rasulova-Lewis F, O’Dowd A, McVeigh AL, Balakrishnan A, Sincock SA, Prouty MG, Holmes RK, Savarino SJ. Biochemical and immunological characterization of an ETEC CFA/I adhesin cholera toxin B subunit chimera. PLoS One 2020; 15:e0230138. [PMID: 32176708 PMCID: PMC7075575 DOI: 10.1371/journal.pone.0230138] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 02/21/2020] [Indexed: 12/12/2022] Open
Abstract
Surface-expressed colonization factors and their subunits are promising candidates for inclusion into a multivalent vaccine targeting enterotoxigenic Escherichia coli (ETEC), a leading cause of acute bacterial diarrhea in developing regions. However, soluble antigens are often poorly immunogenic in the absence of an adjuvant. We show here that the serum immune response to CfaE, the adhesin of the ETEC colonization factor CFA/I, can be enhanced in BALB/c mice by immunization with a chimeric antigen containing CfaE and pentameric cholera toxin B subunit (CTB) of cholera toxin from Vibrio cholerae. We constructed this antigen by replacing the coding sequence for the A1 domain of the cholera toxin A subunit (CTA) with the sequence of donor strand complemented CfaE (dscCfaE) within the cholera toxin operon, resulting in a dscCfaE-CTA2 fusion. After expression, via non-covalent interactions between CTA2 and CTB, the fusion and CTB polypeptides assemble into a complex containing a single dscCfaE-CTA2 protein bound to pentameric CTB (dscCfaE-CTA2/CTB). This holotoxin-like chimera retained the GM1 ganglioside binding activity of CTB, as well as the ability of CfaE to mediate the agglutination of bovine red blood cells when adsorbed to polystyrene beads. When administered intranasally to mice, the presence of CTB in the chimera significantly increased the serum immune response to CfaE compared to dscCfaE alone, stimulating a response similar to that obtained with a matched admixture of dscCfaE and CTB. However, by the orogastric route, immunization with the chimera elicited a superior functional immune response compared to an equivalent admixture of dscCfaE and CTB, supporting further investigation of the chimera as an ETEC vaccine candidate.
Collapse
Affiliation(s)
- Michael G. Jobling
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - Steven T. Poole
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States of America
| | - Fatima Rasulova-Lewis
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States of America
| | - Aisling O’Dowd
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States of America
| | - Annette L. McVeigh
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States of America
| | - Amit Balakrishnan
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States of America
| | - Stephanie A. Sincock
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD, United States of America
| | - Michael G. Prouty
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD, United States of America
| | - Randall K. Holmes
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - Stephen J. Savarino
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD, United States of America
- Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States of America
| |
Collapse
|
5
|
Gestal MC, Johnson HM, Harvill ET. Immunomodulation as a Novel Strategy for Prevention and Treatment of Bordetella spp. Infections. Front Immunol 2019; 10:2869. [PMID: 31921136 PMCID: PMC6923730 DOI: 10.3389/fimmu.2019.02869] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 11/22/2019] [Indexed: 12/13/2022] Open
Abstract
Well-adapted pathogens have evolved to survive the many challenges of a robust immune response. Defending against all host antimicrobials simultaneously would be exceedingly difficult, if not impossible, so many co-evolved organisms utilize immunomodulatory tools to subvert, distract, and/or evade the host immune response. Bordetella spp. present many examples of the diversity of immunomodulators and an exceptional experimental system in which to study them. Recent advances in this experimental system suggest strategies for interventions that tweak immunity to disrupt bacterial immunomodulation, engaging more effective host immunity to better prevent and treat infections. Here we review advances in the understanding of respiratory pathogens, with special focus on Bordetella spp., and prospects for the use of immune-stimulatory interventions in the prevention and treatment of infection.
Collapse
Affiliation(s)
- Monica C Gestal
- Department of Infectious Diseases, College of Veterinary Sciences, University of Georgia, Athens, GA, United States
| | - Hannah M Johnson
- Department of Infectious Diseases, College of Veterinary Sciences, University of Georgia, Athens, GA, United States
| | - Eric T Harvill
- Department of Infectious Diseases, College of Veterinary Sciences, University of Georgia, Athens, GA, United States
| |
Collapse
|
6
|
Abstract
Pertussis or whooping cough, mainly caused by Bordetella pertussis, is a severe respiratory disease that can affect all age groups but is most severe and can be life-threatening in young children. Vaccines against this disease are widely available since the 1950s. Despite high global vaccination coverage, the disease is not under control in any country, and its incidence is even increasing in several parts of the world. Epidemiological and experimental evidence has shown that the vaccines fail to prevent B. pertussis infection and transmission, although they are very effective in preventing disease. Given the high infection rate of B. pertussis, effective control of the disease likely requires prevention of infection and transmission in addition to protection against disease. With rare exceptions B. pertussis infections are restricted to the airways and do not usually disseminate beyond the respiratory epithelium. Therefore, protection at the level of the respiratory mucosa may be helpful for an improved control of pertussis. Yet, compared to systemic responses, mucosal immune responses have attracted relatively little attention in the context of pertussis vaccine development. In this review we summarize the available literature on the role of mucosal immunity in the prevention of B. pertussis infection. In contrast to vaccination, natural infection in humans and experimental infections in animals induce strong secretory IgA responses in the naso-pharynx and in the lungs. Several studies have shown that secretory IgA may be instrumental in the control of B. pertussis infection. Furthermore, studies in mouse models have revealed that B. pertussis infection, but not immunization with current acellular pertussis vaccines induces resident memory T cells, which may also contribute to protection against colonization by B. pertussis. As these resident memory T cells are long lived, vaccines that are able to induce them should provide long-lasting immunity. As of today, only one vaccine designed to induce potent mucosal immunity is in clinical development. This vaccine is a live attenuated B. pertussis strain delivered nasally in order to mimic the natural route of infection. Due to its ability to induce mucosal immunity it is expected that this approach will contribute to improved control of pertussis.
Collapse
Affiliation(s)
- Luis Solans
- Center of Infection and Immunity of Lille, Institut Pasteur de Lille, Lille, France
- Inserm U1019, Lille, France
- CNRS UMR8204, Lille, France
- Center for Infection and Immunity of Lille, Univ. Lille, Lille, France
| | - Camille Locht
- Center of Infection and Immunity of Lille, Institut Pasteur de Lille, Lille, France
- Inserm U1019, Lille, France
- CNRS UMR8204, Lille, France
- Center for Infection and Immunity of Lille, Univ. Lille, Lille, France
| |
Collapse
|
7
|
Immunization with the recombinant Cholera toxin B fused to Fimbria 2 protein protects against Bordetella pertussis infection. BIOMED RESEARCH INTERNATIONAL 2014; 2014:421486. [PMID: 24982881 PMCID: PMC4052895 DOI: 10.1155/2014/421486] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Revised: 04/14/2014] [Accepted: 04/22/2014] [Indexed: 11/18/2022]
Abstract
This study examined the immunogenic properties of the fusion protein fimbria 2 of Bordetella pertussis (Fim2)—cholera toxin B subunit (CTB) in the intranasal murine model of infection. To this end B. pertussis Fim2 coding sequence was cloned downstream of the cholera toxin B subunit coding sequence. The expression and assembly of the fusion protein into pentameric structures (CTB-Fim2) were evaluated by SDS-PAGE and monosialotetrahexosylgaglioside (GM1-ganglioside) enzyme-linked immunosorbent assay (ELISA). To evaluate the protective capacity of CTB-Fim2, an intraperitoneal or intranasal mouse immunization schedule was performed with 50 μg of CTB-Fim2. Recombinant (rFim2) or purified (BpFim2) Fim2, CTB, and phosphate-buffered saline (PBS) were used as controls. The results showed that mice immunized with BpFim2 or CTB-Fim2 intraperitoneally or intranasally presented a significant reduction in bacterial lung counts compared to control groups (P < 0.01 or P < 0.001 , resp.). Moreover, intranasal immunization with CTB-Fim2 induced significant levels of Fim2-specific IgG in serum and bronchoalveolar lavage (BAL) and Fim2-specific IgA in BAL. Analysis of IgG isotypes and cytokines mRNA levels showed that CTB-Fim2 results in a mixed Th1/Th2 (T-helper) response. The data presented here provide support for CTB-Fim2 as a promising recombinant antigen against Bordetella pertussis infection.
Collapse
|
8
|
Tinker JK, Yan J, Knippel RJ, Panayiotou P, Cornell KA. Immunogenicity of a West Nile virus DIII-cholera toxin A2/B chimera after intranasal delivery. Toxins (Basel) 2014; 6:1397-418. [PMID: 24759174 PMCID: PMC4014742 DOI: 10.3390/toxins6041397] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2013] [Revised: 04/09/2014] [Accepted: 04/09/2014] [Indexed: 12/19/2022] Open
Abstract
West Nile virus (WNV) causes potentially fatal neuroinvasive disease and persists at endemic levels in many parts of the world. Despite advances in our understanding of WNV pathogenesis, there remains a significant need for a human vaccine. The domain III (DIII) region of the WNV envelope protein contains epitopes that are the target of neutralizing antibodies. We have constructed a chimeric fusion of the non-toxic cholera toxin (CT) CTA2/B domains to DIII for investigation as a novel mucosally-delivered WNV vaccine. Purification and assembly of the chimera, as well as receptor-binding and antigen delivery, were verified by western blot, GM1 ELISA and confocal microscopy. Groups of BALB/c mice were immunized intranasally with DIII-CTA2/B, DIII, DIII mixed with CTA2/B, or CTA2/B control, and boosted at 10 days. Analysis of serum IgG after 14 and 45 days revealed that mucosal immunization with DIII-CTA2/B induced significant DIII-specific humoral immunity and drove isotype switching to IgG2a. The DIII-CTA2/B chimera also induced antigen-specific IgM and IgA responses. Bactericidal assays indicate that the DIII-CTA2/B immunized mice produced DIII-specific antibodies that can trigger complement-mediated killing. A dose escalation resulted in increased DIII-specific serum IgG titers on day 45. DIII antigen alone, in the absence of adjuvant, also induced significant systemic responses after intranasal delivery. Our results indicate that the DIII-CTA2/B chimera is immunogenic after intranasal delivery and merits further investigation as a novel WNV vaccine candidate.
Collapse
Affiliation(s)
- Juliette K Tinker
- Department of Biological Sciences, Boise State University, Boise, ID 83725, USA.
| | - Jie Yan
- Department of Chemistry and Biochemistry, Boise State University, Boise, ID 83725, USA.
| | - Reece J Knippel
- Department of Chemistry and Biochemistry, Boise State University, Boise, ID 83725, USA.
| | - Panos Panayiotou
- Department of Chemistry and Biochemistry, Boise State University, Boise, ID 83725, USA.
| | - Kenneth A Cornell
- Department of Chemistry and Biochemistry, Boise State University, Boise, ID 83725, USA.
| |
Collapse
|
9
|
Martín R, Chain F, Miquel S, Natividad JM, Sokol H, Verdu EF, Langella P, Bermúdez-Humarán LG. Effects in the use of a genetically engineered strain of Lactococcus lactis delivering in situ IL-10 as a therapy to treat low-grade colon inflammation. Hum Vaccin Immunother 2014; 10:1611-21. [PMID: 24732667 DOI: 10.4161/hv.28549] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Irritable bowel syndrome (IBS) is a gastrointestinal disorder characterized by chronic abdominal pain, discomfort, and bloating. Interestingly, there is now evidence of the presence of a low-grade inflammatory status in many IBS patients, including histopathological and mucosal cytokine levels in the colon, as well as the presence of IBS-like symptoms in quiescent inflammatory bowel disease (IBD). The use of a genetically engineered food-grade bacterium, such as Lactococcus lactis, secreting the anti-inflammatory cytokine IL-10 has been proven by many pre-clinical studies to be a successful therapy to treat colon inflammation. In this study, we first reproduced the recovery-recurrence periods observed in IBS-patients in a new chronic model characterized by 2 episodes of DiNitro-BenzeneSulfonic-acid (DNBS)-challenge and we tested the effects of a recombinant strain of L. lactis secreting IL-10 under a Stress-Inducible Controlled Expression (SICE) system. In vivo gut permeability, colonic serotonin levels, cytokine profiles, and spleen cell populations were then measured as readouts of a low-grade inflammation. In addition, since there is increasing evidence that gut microbiota tightly regulates gut barrier function, tight junction proteins were also measured by qRT-PCR after administration of recombinant L. lactis in DNBS-treated mice. Strikingly, oral administration of L. lactis secreting active IL-10 in mice resulted in significant protective effects in terms of permeability, immune activation, and gut-function parameters. Although genetically engineered bacteria are, for now, used only as a "proof-of-concept," our study validates the interest in the use of the novel SICE system in L. lactis to express therapeutic molecules, such as IL-10, locally at mucosal surfaces.
Collapse
Affiliation(s)
- Rebeca Martín
- INRA; Commensal and Probiotics-Host Interactions Laboratory; UMR 1319 Micalis; Jouy-en-Josas, France; AgroParisTech; UMR1319 Micalis; Jouy-en-Josas, France
| | - Florian Chain
- INRA; Commensal and Probiotics-Host Interactions Laboratory; UMR 1319 Micalis; Jouy-en-Josas, France; AgroParisTech; UMR1319 Micalis; Jouy-en-Josas, France
| | - Sylvie Miquel
- INRA; Commensal and Probiotics-Host Interactions Laboratory; UMR 1319 Micalis; Jouy-en-Josas, France; AgroParisTech; UMR1319 Micalis; Jouy-en-Josas, France
| | - Jane M Natividad
- Farncombe Family Digestive Health Research Institute; McMaster University; Hamilton, ON Canada
| | - Harry Sokol
- INRA; Commensal and Probiotics-Host Interactions Laboratory; UMR 1319 Micalis; Jouy-en-Josas, France; AgroParisTech; UMR1319 Micalis; Jouy-en-Josas, France; Department of Gastroenterology and Nutrition; AP-HP; Hôpital Saint-Antoine F-75012 and UPMC Univ Paris; Paris, France; INSERM; Equipe AVENIR U1057 / UMR CNRS 7203; Paris, France
| | - Elena F Verdu
- Farncombe Family Digestive Health Research Institute; McMaster University; Hamilton, ON Canada
| | - Philippe Langella
- INRA; Commensal and Probiotics-Host Interactions Laboratory; UMR 1319 Micalis; Jouy-en-Josas, France; AgroParisTech; UMR1319 Micalis; Jouy-en-Josas, France
| | - Luis G Bermúdez-Humarán
- INRA; Commensal and Probiotics-Host Interactions Laboratory; UMR 1319 Micalis; Jouy-en-Josas, France; AgroParisTech; UMR1319 Micalis; Jouy-en-Josas, France
| |
Collapse
|
10
|
|
11
|
Gao Z, Zhao X, Lee S, Li J, Liao H, Zhou X, Wu J, Qi G. WH1fungin a surfactin cyclic lipopeptide is a novel oral immunoadjuvant. Vaccine 2013; 31:2796-803. [DOI: 10.1016/j.vaccine.2013.04.028] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Revised: 03/14/2013] [Accepted: 04/10/2013] [Indexed: 01/22/2023]
|
12
|
Locht C, Mielcarek N. New pertussis vaccination approaches: en route to protect newborns? ACTA ACUST UNITED AC 2012; 66:121-33. [PMID: 22574832 DOI: 10.1111/j.1574-695x.2012.00988.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2012] [Revised: 05/04/2012] [Accepted: 05/04/2012] [Indexed: 11/26/2022]
Abstract
Pertussis or whooping cough is a life-threatening childhood disease, particularly severe during the first months of life, although adolescent and adult pertussis is increasingly more noted. General vaccination has tremendously reduced its incidence but has failed to bring it completely under control. In fact, it remains one of the most poorly controlled vaccine-preventable diseases in the world. New vaccination strategies are thus being explored. These include vaccination of pregnant mothers to transmit protective antibodies to the offspring, a cocooning strategy to prevent the transmission of the disease from family members to the newborn and neonatal vaccination. All have their inherent limitations, and improved vaccines are urgently needed. Two types of pertussis vaccines are currently available, whole-cell, first-generation and second-generation, acellular vaccines, with an improved safety profile. Attempts have been made to discover additional protective antigens to the 1-5 currently included in the acellular vaccines or to include new adjuvants. Recently, a live attenuated nasal Bordetella pertussis vaccine has been developed and undergone first-in-man clinical trials. However, as promising as it may be, in order to protect infants against severe disease, a single approach may not be sufficient, and multiple strategies applied in a concerted fashion may ultimately be required.
Collapse
Affiliation(s)
- Camille Locht
- Center for Infection and Immunity of Lille, Institut Pasteur de Lille, Lille, France.
| | | |
Collapse
|
13
|
Mucosal immunization with a Staphylococcus aureus IsdA-cholera toxin A2/B chimera induces antigen-specific Th2-type responses in mice. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2011; 18:1543-51. [PMID: 21734065 DOI: 10.1128/cvi.05146-11] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Staphylococcus aureus is a leading cause of opportunistic infection worldwide and a significant public health threat. The iron-regulated surface determinant A (IsdA) adhesin is essential for S. aureus colonization on human nasal epithelial cells and plays an important role in iron acquisition and resistance to human skin defenses. Here we investigated the murine immune response to intranasal administration of a cholera toxin A(2)/B (CTA(2)/B) chimera containing IsdA. Plasmids were constructed to express the IsdA-CTA(2)/B chimera and control proteins in Escherichia coli. Proper construction of the chimera was verified by SDS-PAGE, Western blotting, GM1 enzyme-linked immunosorbent assay (ELISA), and confocal microscopy. Groups of female BALB/c mice were mock immunized or immunized with IsdA-CTA(2)/B, IsdA mixed with CTA(2)/B, or IsdA alone, followed by one booster immunization at 10 days postpriming. Analysis of serum IgG and nasal, intestinal, and vaginal IgA suggested that mucosal immunization with IsdA-CTA(2)/B induces significant IsdA-specific humoral immunity. Functional in vitro assays revealed that immune serum significantly blocks the adherence of S. aureus to human epithelial cells. Splenocytes from mice immunized with IsdA-CTA(2)/B showed specific cellular proliferation and production of interleukin-4 (IL-4) after in vitro stimulation. Immunization with IsdA-CTA(2)/B drove isotype switching to IgG1, indicative of a Th2-type response. Our results suggest that the immunogenicity of the S. aureus IsdA-CTA(2)/B chimera merits further investigation as a potential mucosal vaccine candidate.
Collapse
|
14
|
Jinyong Z, Xiaoli Z, Weijun Z, Ying G, Gang G, Xuhu M, Quanming Z. Fusion expression and immunogenicity of Bordetella pertussis PTS1-FHA protein: implications for the vaccine development. Mol Biol Rep 2011; 38:1957-63. [PMID: 20878241 DOI: 10.1007/s11033-010-0317-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2010] [Accepted: 09/03/2010] [Indexed: 10/19/2022]
Abstract
Mutants of pertussis toxin (PT) S1 subunit and filamentous hemagglutinin (FHA) type I immunodominant domain from Bordetella pertussis (B. pertussis) are considered to be effective candidate antigens for acellular pertussis vaccines; however, the substantial progress is hampered in part for the lack of a suitable in vitro expression system. In this paper, the gene sequences of a S1 mutant C180-R9K/E129G (mS1) and a truncated peptide named Fs from FHA type I immunodominant domain were linked together and constructed to pET22b expression vector as a fusion gene; after inducing with IPTG, it was highly expressed in E. coli BL21 (DE3) as inclusion body. The fusion protein FsmS1 was purified from cell lysates and refolded successfully. The result of Western blotting indicate that it was able to react with both anti-S1 and anti-FHA McAbs; antiserum produced from New Zealand white rabbits immunized with this protein was able to recognize both native PT and FHA antigens as determined by western blotting. These data have provided a novel feasible method to produce PT S1 subunit and FHA type I immunodominant domain in large scale in vitro, which is implicated for the development of multivalent subunit vaccines candidate against B. pertussis infection.
Collapse
MESH Headings
- Adhesins, Bacterial/chemistry
- Adhesins, Bacterial/immunology
- Animals
- Antibodies, Bacterial/biosynthesis
- Antibodies, Bacterial/immunology
- Blotting, Western
- Bordetella pertussis/immunology
- Cloning, Molecular
- Leukocytosis/immunology
- Leukocytosis/microbiology
- Leukocytosis/prevention & control
- Mice
- Mice, Inbred BALB C
- Pertussis Toxin/immunology
- Pertussis Vaccine/immunology
- Protein Refolding
- Protein Structure, Tertiary
- Rabbits
- Recombinant Fusion Proteins/immunology
- Recombinant Fusion Proteins/isolation & purification
- Virulence Factors, Bordetella/chemistry
- Virulence Factors, Bordetella/immunology
Collapse
Affiliation(s)
- Zhang Jinyong
- Department of Clinical Microbiology and Immunology, The Third Military Medical University, Chongqing Engineering Technology Research Center of Biopharmaceuticals, Chongqing 400038, China
| | | | | | | | | | | | | |
Collapse
|
15
|
Ayalew S, Confer AW, Payton ME, Garrels KD, Shrestha B, Ingram KR, Montelongo MA, Taylor JD. Mannheimia haemolytica chimeric protein vaccine composed of the major surface-exposed epitope of outer membrane lipoprotein PlpE and the neutralizing epitope of leukotoxin. Vaccine 2008; 26:4955-61. [DOI: 10.1016/j.vaccine.2008.07.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2007] [Revised: 06/26/2008] [Accepted: 07/08/2008] [Indexed: 11/26/2022]
|
16
|
Fry SR, Chen AY, Daggard G, Mukkur TKS. Parenteral immunization of mice with a genetically inactivated pertussis toxin DNA vaccine induces cell-mediated immunity and protection. J Med Microbiol 2008; 57:28-35. [PMID: 18065664 DOI: 10.1099/jmm.0.47527-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The immunogenicity and protective efficacy of a DNA vaccine encoding a genetically inactivated S1 domain of pertussis toxin was evaluated using a murine respiratory challenge model of Bordetella pertussis infection. It was found that mice immunized via the intramuscular route elicited a purely cell-mediated immune response to the DNA vaccine, with high levels of gamma interferon (IFN-gamma) and interleukin (IL)-2 detected in the S1-stimulated splenocyte supernatants and no serum IgG. Despite the lack of an antibody response, the lungs of DNA-immunized mice were cleared of B. pertussis at a significantly faster rate compared with mock-immunized mice following an aerosol challenge. To gauge the true potential of this S1 DNA vaccine, the immune response and protective efficacy of the commercial diphtheria-tetanus-acellular pertussis (DTaP) vaccine were included as the gold standard. Immunization with DTaP elicited a typically strong T-helper (Th)2-polarized immune response with significantly higher titres of serum IgG than in the DNA vaccine group, but a relatively weak Th1 response with low levels of IFN-gamma and IL-2 detected in the supernatants of antigen-stimulated splenocytes. DTaP-immunized mice cleared the aerosol challenge more efficiently than DNA-immunized mice, with no detectable pathogen after day 7 post-challenge.
Collapse
Affiliation(s)
- Scott R Fry
- Department of Biological and Physical Sciences, University of Southern Queensland, Toowoomba 4350, Queensland, Australia
| | - Austen Y Chen
- Department of Biological and Physical Sciences, University of Southern Queensland, Toowoomba 4350, Queensland, Australia
| | - Grant Daggard
- Department of Biological and Physical Sciences, University of Southern Queensland, Toowoomba 4350, Queensland, Australia
| | - Trilochan K S Mukkur
- Department of Biological and Physical Sciences, University of Southern Queensland, Toowoomba 4350, Queensland, Australia
| |
Collapse
|
17
|
Oliveira MLS, Arêas APM, Ho PL. Intranasal vaccines for protection against respiratory and systemic bacterial infections. Expert Rev Vaccines 2007; 6:419-29. [PMID: 17542756 DOI: 10.1586/14760584.6.3.419] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
More than 4 million deaths per year are due to respiratory diseases. Although licensed vaccines are available, bacteria, such as Streptococcus pneumoniae, Haemophilus influenzae, Mycobacterium tuberculosis, Bordetella pertussis and Neisseria meningiditis, among others, continue to be the major agents of diseases in young children, the elderly and/or immunocompromized individuals. Following respiratory tract infection, some microorganisms may also invade the epithelial tissue, achieving systemic circulation and/or other organs. Nasal administration of different antigen formulations has shown promising results in the induction of immune responses and the defeat of the pathogens at the site of infection. This review will focus on the main nasal vaccine strategies and technologies being investigated against the most common infections caused by respiratory bacteria.
Collapse
|
18
|
Price GA, Masri HP, Hollander AM, Russell MW, Cornelissen CN. Gonococcal transferrin binding protein chimeras induce bactericidal and growth inhibitory antibodies in mice. Vaccine 2007; 25:7247-60. [PMID: 17720283 PMCID: PMC2225598 DOI: 10.1016/j.vaccine.2007.07.038] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2007] [Revised: 05/11/2007] [Accepted: 07/12/2007] [Indexed: 12/22/2022]
Abstract
We have previously demonstrated the full-length gonococcal transferrin binding proteins (TbpA and TbpB) to be promising antigens in the development of a protective vaccine against Neisseria gonorrhoeae. In the current study we employed a genetic chimera approach fusing domains from TbpA and TbpB to the A2 domain of cholera toxin, which naturally binds in a non-covalent fashion to the B subunit of cholera toxin during assembly. For one construct, the N-terminal half of TbpB (NB) was fused to the A2 subunit of cholera toxin. In a second construct, the loop 2 region (L2) of TbpA was genetically fused between the NB domain and the A2 domain, generating a double chimera. Both chimeras were immunogenic and induced serum bactericidal and vaginal growth-inhibiting antibodies. This study highlights the potential of using protective epitopes instead of full-length proteins in the development of an efficacious gonococcal vaccine.
Collapse
Affiliation(s)
- Gregory A. Price
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, Richmond, Virginia 23298 USA
| | - Heather P. Masri
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, Richmond, Virginia 23298 USA
| | - Aimee M. Hollander
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, Richmond, Virginia 23298 USA
| | - Michael W. Russell
- Departments of Oral Biology and Microbiology and Immunology, Witebsky Center for Microbial Pathogenesis and Immunology, University at Buffalo, Buffalo, New York
| | - Cynthia Nau Cornelissen
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, Richmond, Virginia 23298 USA
| |
Collapse
|
19
|
Mansour M, Brown RG, Morris A, Smith B, Halperin SA. Improved efficacy of a licensed acellular pertussis vaccine, reformulated in an adjuvant emulsion of liposomes in oil, in a murine model. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2007; 14:1381-3. [PMID: 17715327 PMCID: PMC2168125 DOI: 10.1128/cvi.00143-07] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2007] [Revised: 06/12/2007] [Accepted: 08/15/2007] [Indexed: 11/20/2022]
Abstract
The immunogenicities and efficacies of a licensed diphtheria, tetanus, acellular pertussis, and inactivated poliovirus vaccine and the same vaccine formulated in a liposome/oil emulsion adjuvant were compared in a mouse model of pertussis respiratory infection. A single dose of the liposome/oil emulsion-adjuvanted vaccine produced significantly higher antibody levels than one dose of the licensed vaccine and protected mice from Bordetella pertussis infection with an efficacy equivalent to that of three doses of the licensed vaccine.
Collapse
Affiliation(s)
- Marc Mansour
- ImmunoVaccine Technologies Inc., Dalhousie University, Halifax, Nova Scotia, Canada
| | | | | | | | | |
Collapse
|
20
|
Hussein AH, Davis EM, Halperin SA, Lee SF. Construction and characterization of single-chain variable fragment antibodies directed against the Bordetella pertussis surface adhesins filamentous hemagglutinin and pertactin. Infect Immun 2007; 75:5476-82. [PMID: 17724067 PMCID: PMC2168280 DOI: 10.1128/iai.00494-07] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A single-chain variable fragment (scFv) antibody library against Bordetella pertussis was constructed using M13 phage display. The library was enriched for phages surface displaying functional scFv by biopanning against B. pertussis immobilized on polystyrene plates. Two hundred eighty-eight individual clones from the enriched library were screened for binding to B. pertussis cells, filamentous hemagglutinin (FHA), and pertactin (PRN) in enzyme-linked immunosorbent assays (ELISAs). Based on the binding ability, the clones were put into eight groups. The scFv DNA inserts from the 288 clones were digested with BstOI, and 18 unique restriction patterns, named types 1 to 18, were found. Eight clones (types 1 to 7 and 18) were selected for further testing against FHA, PRN, and B. pertussis by ELISA. The results showed that types 1, 5, 7, and 18 bound strongly to B. pertussis cells as well as FHA and PRN. Type 3 bound strongly to the cells and FHA but weakly to PRN. Types 4 and 6 bound FHA only, and type 2 did not bind to the cells or antigens. The ability of the eight clones to inhibit B. pertussis from binding to HEp-2 cells was assayed. Types 1, 5, and 7, but not the remaining clones, inhibited the adherence of B. pertussis to HEp-2 cells. The scFvs were sequenced, and the deduced amino acid sequence showed that the scFvs were different antibodies. Maltose-binding protein (MBP) fusion proteins composed of three different regions of FHA (heparin-binding domain, carbohydrate recognition domain, and the RGD triplet motif) were constructed. The three fusion proteins and Mal85 (MBP-FHA type I domain) were used to map the binding sites for scFvs of types 1, 5, and 7 by ELISA. The results showed that all three scFvs bound to the heparin-binding domain fusion protein but not the other fusion proteins. BALB/c mice who received recombinant phage-treated B. pertussis had reduced bacterial counts in the nasal cavity, trachea, and lungs compared to the control groups.
Collapse
MESH Headings
- Adhesins, Bacterial/genetics
- Adhesins, Bacterial/immunology
- Amino Acid Sequence
- Animals
- Antibodies, Bacterial/chemistry
- Antibodies, Bacterial/genetics
- Antibodies, Bacterial/immunology
- Antibodies, Bacterial/therapeutic use
- Bacterial Adhesion/immunology
- Bacterial Outer Membrane Proteins/immunology
- Bacteriophage M13
- Bordetella pertussis/immunology
- Cell Line
- Colony Count, Microbial
- DNA, Bacterial/chemistry
- DNA, Bacterial/genetics
- Enzyme-Linked Immunosorbent Assay
- Epitope Mapping
- Female
- Humans
- Mice
- Mice, Inbred BALB C
- Molecular Sequence Data
- Peptide Library
- Protein Binding
- Recombinant Proteins/chemistry
- Recombinant Proteins/genetics
- Recombinant Proteins/immunology
- Recombinant Proteins/therapeutic use
- Virulence Factors, Bordetella/genetics
- Virulence Factors, Bordetella/immunology
- Whooping Cough/prevention & control
Collapse
Affiliation(s)
- Ahmad H Hussein
- Department of Applied Oral Sciences, Faculty of Dentistry, Dalhousie University Halifax, Nova Scotia B3H 3J5, Canada
| | | | | | | |
Collapse
|
21
|
Kamachi K, Arakawa Y. Development of safer pertussis DNA vaccine expressing non-toxic C180 polypeptide of pertussis toxin S1 subunit. Vaccine 2006; 25:1000-6. [PMID: 17050047 DOI: 10.1016/j.vaccine.2006.09.077] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2006] [Revised: 09/05/2006] [Accepted: 09/26/2006] [Indexed: 11/18/2022]
Abstract
A toxic N-terminal 180-amino-acid fragment (C180) of pertussis toxin S1 subunit has the most potent ability to induce protective immunity against pertussis toxin (PT) following DNA-based immunization [Kamachi K, Arakawa Y. Infect Immun 2004;72:4293-6]. For the development of a safer pertussis DNA vaccine, three plasmids encoding mutant C180 (C180-R9K, C180-E129G and C180-R9K/E129G) were constructed and tested for their protective immunogenicity and cytotoxicity. All of the gene gun delivery of the plasmid, performed by inserting the mutant C180 gene into a mammalian expression vector pcDNA3.1, successfully induced anti-PT IgG antibody production without the loss of immunogenicity in mice. The immunizations of mice with the plasmids significantly inhibited leukocytosis-promoting activity by PT. Among stably transfected Chinese hamster ovary (CHO) cells expressing mutant C180, the expression of C180-R9K and C180-R9K/E129G was non-toxic to the transfectants, confirming that these mutant C180s have no cytotoxicity to mammalian cells. These results indicate that C180-R9K and C180-R9K/E129G genes, especially C180-R9K/E129G, are candidates for safe and effective antigen DNAs in the development of pertussis DNA vaccine.
Collapse
Affiliation(s)
- Kazunari Kamachi
- Department of Bacterial Pathogenesis and Infection Control, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashimurayama, Tokyo 208-0011, Japan.
| | | |
Collapse
|
22
|
Mallaley PP, Halperin SA, Morris A, MacMillan A, Lee SF. Expression of a pertussis toxin S1 fragment by inducible promoters in oral Streptococcus and the induction of immune responses during oral colonization in mice. Can J Microbiol 2006; 52:436-44. [PMID: 16699568 DOI: 10.1139/w05-151] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Previous work aimed at developing a live oral vaccine expressing pertussis toxin S1 fragment on the surface of the bacterium Streptococcus gordonii elicited a lower than expected antibody response, perhaps because of low antigen expression. In this study, in-frame promoter fusions were constructed to investigate whether an increase in antigen production by the streptococcal vaccine strain results in a better antibody response. The promoters tested were (i) the Streptococcus mutans sucrose-inducible fructosyltransferase (ftf) promoter and (ii) the Bacillus subtilis/Escherichia coli chimeric tetracycline-inducible xyl/tetO promoter. Each of these two promoters was placed upstream of the spaP/s1 fusion gene to drive its expression. The constructs were introduced into S. gordonii DL1 and S. mutans 834. The inducibility of the promoters was confirmed through the determination of SpaP/S1 production via Western blottings. Induced production of SpaP/S1 was observed in S. gordonii and S. mutans with each of the promoters, but the level of expression was the highest in S. mutans, using the xyl/tetO promoter. Thus, S. mutans carrying the xyl/tetO/spaP/s1 construct (S. mutans PM14) was used in oral colonization studies in BALB/c mice. Streptococccus mutans PM14 was able to colonize the animals for the 14-week duration of experimentation. A mucosal IgA response was observed in all the treatment groups but was highest in mice receiving tetracycline induction. In the mouse model of Bordetella pertussis respiratory infection, animals colonized with S. mutans PM14 showed a decreased in B. pertussis lung colony count (P = 0.03) on day 3 compared with control mice colonized by the parent S. mutans 834.
Collapse
Affiliation(s)
- P P Mallaley
- Department of Microbiology and Immunology, Faculty of Medicine, Halifax, Canada
| | | | | | | | | |
Collapse
|
23
|
Aminian M, Sivam S, Lee CW, Halperin SA, Lee SF. Expression and purification of a trivalent pertussis toxin-diphtheria toxin-tetanus toxin fusion protein in Escherichia coli. Protein Expr Purif 2006; 51:170-8. [PMID: 16950635 DOI: 10.1016/j.pep.2006.07.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2006] [Revised: 07/17/2006] [Accepted: 07/24/2006] [Indexed: 10/24/2022]
Abstract
Pertussis toxoid, diphtheria toxoid, and tetanus toxoid are key components of diphtheria-tetanus-acellular pertussis vaccines. The efficacy of the vaccines is well documented, however, the vaccines are expensive partly because the antigens are derived from three different bacteria. In this study, a fusion protein (PDT) composed of the immunoprotective S1 fragment of pertussis toxin, the full-length non-toxic diphtheria toxin, and fragment C of tetanus toxin was constructed via genetic means. The correct fusion was verified by restriction endonuclease analysis and Western immunoblotting. Escherichia coli carrying the recombinant plasmid (pCoPDT) produced a 161kDa protein that was recognized by antibodies specific to the three toxins. The expression of the PDT protein was inducible by isopropyl-beta-d-thio-galactoside but the total amount of protein produced was relatively low. Attempts to improve the protein yield by expression in an E. coli strain (Rosetta-gami 2) that could alleviate rare-codon usage bias and by supplementation of the growth media with amino acids deemed to be a limiting factor in translation were not successful. The PDT protein remained in the insoluble fraction when the recombinant E. coli was grown at 37 degrees C but the protein became soluble when the bacteria were grown at 22 degrees C. The PDT protein was isolated via affinity chromatography on a NiCAM column. The protein was associated with five other proteins via disulfide bonds and non-covalent interactions. Following treatment with beta-mercaptoethanol, the PDT fusion was purified to homogeneity by preparative polyacrylamide gel electrophoresis with a yield of 45 microg/L of culture. Antisera generated against the purified PDT protein recognized the native toxins indicating that some, if not all, of the native epitopes were conserved.
Collapse
Affiliation(s)
- Mahdi Aminian
- Department of Microbiology and Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada B3H 3J5
| | | | | | | | | |
Collapse
|
24
|
Knight JB, Huang YY, Halperin SA, Anderson R, Morris A, Macmillan A, Jones T, Burt DS, Van Nest G, Lee SF. Immunogenicity and protective efficacy of a recombinant filamentous haemagglutinin from Bordetella pertussis. Clin Exp Immunol 2006; 144:543-51. [PMID: 16734625 PMCID: PMC1941966 DOI: 10.1111/j.1365-2249.2006.03097.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Bordetella pertussis is the causative agent of whooping cough, a major childhood pathogen; acellular vaccines consisting of purified B. pertussis antigens such as filamentous haemagglutinin (FHA) are commonly used to prevent pertussis. Despite the importance of FHA in B. pertussis pathogenesis and its inclusion in most acellular vaccines, the functional importance of individual domains in the induction of protective immunity is largely unknown. In this study, we have purified a recombinant FHA protein from Escherichia coli consisting of a 42 kDa maltose binding domain of E. coli and the 43 kDa type I immunodominant domain of FHA. The fusion protein (Mal85) was purified from E. coli cell lysates via affinity chromatography with an amylose column. Mal85 was then delivered to BALB/c mice intranasally encapsulated in liposomes, formulated with Protollin(TM) or in conjunction with an immunostimulatory CpG oligonucleotide. Mice were also vaccinated intraperitoneally with alum-adsorbed Mal85. Sera from all treatment groups showed strong IgG responses to Mal85 and recognized native FHA. Specific salivary IgA was induced in mice vaccinated with Mal85 in liposomes, Protollin(TM) and delivered with CpG. Vaccination with Mal85 encapsulated in liposomes or formulated with Protollin(TM) provided protection against aerosol challenge with B. pertussis in BALB/c mice. These data indicate that the type I domain of FHA is a protective antigen in mice and may serve as a candidate for inclusion in new acellular pertussis vaccines.
Collapse
MESH Headings
- Adhesins, Bacterial/immunology
- Adhesins, Bacterial/isolation & purification
- Adjuvants, Immunologic
- Animals
- Antibodies, Bacterial/biosynthesis
- Antigens, Bacterial/immunology
- Antigens, Bacterial/isolation & purification
- Bordetella pertussis/immunology
- CpG Islands/immunology
- Cysteine Endopeptidases
- Drug Combinations
- Female
- Hemagglutinins/immunology
- Hemagglutinins/isolation & purification
- Immunization/methods
- Immunoglobulin A, Secretory/biosynthesis
- Immunoglobulin G/biosynthesis
- Lipopolysaccharides
- Liposomes
- Mice
- Mice, Inbred BALB C
- Pertussis Vaccine/immunology
- Saliva/immunology
- Vaccines, Synthetic/immunology
- Virulence Factors, Bordetella/immunology
- Virulence Factors, Bordetella/isolation & purification
- Whooping Cough/prevention & control
Collapse
Affiliation(s)
- J B Knight
- Department of Microbiology and Immunology, Faculty of Medicine, Dalhousie University and the IWK Health Centre, Halifax, NS, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Tinker JK, Erbe JL, Holmes RK. Characterization of fluorescent chimeras of cholera toxin and Escherichia coli heat-labile enterotoxins produced by use of the twin arginine translocation system. Infect Immun 2005; 73:3627-35. [PMID: 15908392 PMCID: PMC1111858 DOI: 10.1128/iai.73.6.3627-3635.2005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cholera toxin (CT) is an AB(5) toxin responsible for the profuse secretory diarrhea resulting from Vibrio cholerae infection. CT consists of a pentameric, receptor-binding B subunit (CTB) and a monomeric A subunit (CTA) that has latent enzymatic activity. In addition to its enterotoxicity, CT has potent mucosal adjuvant activity and can also function as a carrier molecule with many potential applications in cell biology. In earlier studies, the toxic CTA(1) domain was replaced by several other antigenic protein domains to produce holotoxin-like chimeras for use as potential mucosal vaccines. In the present study we utilized the twin arginine translocation (tat) system to produce fluorescent CT chimeras, as well as fluorescent chimeras of Escherichia coli heat-labile toxins LTI and LTIIb. Fusion proteins containing either green fluorescent protein (GFP) or monomeric red fluorescent protein (mRFP) and the A(2) domain of CT, LTI, or LTIIb were transported to the periplasm of E. coli by the tat system, and the corresponding B polypeptides of CT, LTI, and LTIIb were transported to the periplasm by the sec system. The fluorescent fusion proteins were shown to assemble spontaneously and efficiently with the corresponding B polypeptides in the periplasm to form chimeric holotoxin-like molecules, and these chimeras bound to and entered cultured cells in a manner similar to native CT, LTI, or LTIIb. The GFP and mRFP derivatives of CT, LT, and LTIIb developed here are useful tools for studies on the cell biology of trafficking of the CT/LT family of bacterial enterotoxins. In addition, these constructs provide proof in principle for the development of novel chimeric CT-like or LT-like vaccine candidates containing CTA(2) fusion proteins that cannot be delivered to the periplasm of E. coli by use of the sec secretion pathway.
Collapse
Affiliation(s)
- Juliette K Tinker
- Department of Microbiology, Mail stop 8333, University of Colorado Health Sciences Center at Fitzsimmons, P.O. Box 6511, Aurora, CO 80045, USA
| | | | | |
Collapse
|
26
|
Arêas APM, Oliveira MLS, Miyaji EN, Leite LCC, Ho PL. Intradermal immunization of mice with cholera toxin B-pneumococcal surface protein A fusion protein is protective against intraperitoneal challenge with Streptococcus pneumoniae. Infect Immun 2005; 73:3810-3. [PMID: 15908419 PMCID: PMC1111874 DOI: 10.1128/iai.73.6.3810-3813.2005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
27
|
Lee CW, Lee SF, Halperin SA. Expression and immunogenicity of a recombinant diphtheria toxin fragment A in Streptococcus gordonii. Appl Environ Microbiol 2004; 70:4569-74. [PMID: 15294787 PMCID: PMC492408 DOI: 10.1128/aem.70.8.4569-4574.2004] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A nontoxic mutant diphtheria toxin fragment A (DTA) was genetically fused in single, double, or triple copy to the major surface protein antigen P1 (SpaP) and surface expressed in Streptococcus gordonii DL-1. The expression was verified by Western immunoblotting. Mouse antisera raised against the recombinant S. gordonii recognized the native diphtheria toxinm suggesting the recombinant DTA was immunogenic. When given intranasally to mice with cholera toxin subunit B as the adjuvant, the recombinant S. gordonii expressing double copies of DTA (SpaP-DTA(2)) induced a mucosal immunoglobulin A response and a weak systemic immunoglobulin G response. S. gordonii SpaP-DTA(2) was able to orally colonize BALB/c mice for a 15-week period and elicited a mucosal response, but a serum immunoglobulin G response was not apparent. The antisera failed to neutralize diphtheria toxin cytotoxicity in a Vero cell assay.
Collapse
Affiliation(s)
- Chiang W Lee
- Department of Applied Oral Sciences, Faculty of Dentistry, Dalhousie University, Halifax, Nova Scotia B3H 3J5, Canada
| | | | | |
Collapse
|
28
|
Kamachi K, Arakawa Y. Expression of a C terminally truncated form of pertussis toxin S1 subunit effectively induces protection against pertussis toxin following DNA-based immunization. Infect Immun 2004; 72:4293-6. [PMID: 15213178 PMCID: PMC427423 DOI: 10.1128/iai.72.7.4293-4296.2004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Four plasmids encoding different C terminally and N terminally truncated pertussis toxin S1 subunits of Bordetella pertussis were constructed and tested for inducibility of protection against pertussis toxin in mice after DNA-based immunization. The region encoding an N-terminal 180-amino-acid fragment of the S1 subunit had the most potent ability to induce protective immunity.
Collapse
Affiliation(s)
- Kazunari Kamachi
- Department of Bacterial Pathogenesis and Infection Control, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashimurayma-shi, Tokyo 208-0011, Japan.
| | | |
Collapse
|
29
|
Cuadros C, Lopez-Hernandez FJ, Dominguez AL, McClelland M, Lustgarten J. Flagellin fusion proteins as adjuvants or vaccines induce specific immune responses. Infect Immun 2004; 72:2810-6. [PMID: 15102791 PMCID: PMC387897 DOI: 10.1128/iai.72.5.2810-2816.2004] [Citation(s) in RCA: 138] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Vaccination is the most efficient prophylaxis against a variety of infectious diseases. New vaccination strategies rely on the incorporation of effective adjuvants, which stimulate the innate immune response and, in turn, activate the adaptive immune response. It is well established that flagellin induces inflammatory responses through the activation of antigen-presenting cells (APCs). In order to evaluate whether flagellin can serve as a carrier for the development of adjuvants or vaccines, we prepared a flagellin-enhanced green fluorescent protein (EGFP) fusion protein. Our results demonstrate that a flagellin-EGFP fusion protein is capable of stimulating APCs, resulting in the maturation of these cells and secretion of proinflammatory cytokines. Furthermore, APCs pulsed with the flagellin-EGFP fusion protein effectively process and present EGFP antigens. More importantly, animals immunized with the flagellin-EGFP fusion protein developed specific anti-EGFP T-cell responses. In contrast, recombinant EGFP was not able to stimulate APCs, nor did it induce a T-cell response. Thus, recombinant-flagellin fusion proteins may be suitable carriers as adjuvants or vaccines for the development of new vaccination strategies to induce and boost immune responses against infectious diseases and cancer.
Collapse
Affiliation(s)
- Camilo Cuadros
- Sidney Kimmel Cancer Center, San Diego, California 92121, USA
| | | | | | | | | |
Collapse
|