1
|
Lee JS, Yoon S, Han SJ, Kim ED, Kim J, Shin HS, Seo KY. Eyedrop vaccination: an immunization route with promises for effective responses to pandemics. Expert Rev Vaccines 2021; 21:91-101. [PMID: 34788181 DOI: 10.1080/14760584.2022.2008246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Mucosal vaccines have several advantages over parenteral vaccines. They induce both systemic and mucosal antigen-specific immune responses, allow easy administration, and bypass the need for trained medical personnel. AREAS COVERED Eye mucosa is a novel route of mucosal vaccine administration. Eyedrop vaccination induces systemic and mucosal immune responses similar to other forms of mucosal vaccines such as oral and intranasal vaccines. EXPERT OPINION Eyedrop vaccines are free of serious adverse side effects like the infiltration of CNS by pathogens. Studies over the years have shown promising results for eye drop vaccines against infectious agents like the influenza virus, Salmonella typhi, and Escherichia coli in animal models. Such efficacy and safety of eyedrop vaccination enable the application of eyedrop vaccines against other infectious diseases as well as chronic diseases. In this review of published literature, we examine the mechanism, efficacy, and safety of eyedrop vaccines and contemplate their role in times of a pandemic.
Collapse
Affiliation(s)
- Jihei Sara Lee
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Sangchul Yoon
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul, South Korea.,Department of Medical Humanities and Social Sciences, Yonsei University College of Medicine, Seoul, South Korea
| | - Soo Jung Han
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Eun-Do Kim
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul, South Korea.,Brain Korea 21 Plus Project for Medical Science, Yonsei University, Seoul, Republic of Korea
| | - Jiyeon Kim
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul, South Korea.,Brain Korea 21 Plus Project for Medical Science, Yonsei University, Seoul, Republic of Korea
| | - Hae-Sol Shin
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul, South Korea.,Korea Mouse Sensory Phenotyping Center (Kmspc), Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kyoung Yul Seo
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul, South Korea.,Korea Mouse Sensory Phenotyping Center (Kmspc), Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
2
|
Knapp MPA, Johnson TA, Ritter MK, Rainer RO, Fiester SE, Grier JT, Connell TD, Arce S. Immunomodulatory regulation by heat-labile enterotoxins and potential therapeutic applications. Expert Rev Vaccines 2021; 20:975-987. [PMID: 34148503 DOI: 10.1080/14760584.2021.1945449] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Introduction: Heat-labile enterotoxins (HLTs) and their cognate ganglioside receptors have been extensively studied because of their therapeutic potential. Gangliosides play arole in modulating effector cells of the immune system, and HLTs provide a novel means for stimulating ganglioside-mediated responses in immunocompetent cells.Areas covered: To evaluate the mechanisms of HLT adjuvanticity, a systemic literature review was performed using relevant keyword searches of the PubMed database, accessing literature published as recently as late 2020. Since HLTs bind to specific ganglioside receptors on immunocytes, they can act as regulators via stimulation or tapering of immune responses from associated signal transduction events. Binding of HLTs to gangliosides can increase proliferation of T-cells, increase cytokine release, augment mucosal/systemic antibody responses, and increase the effectiveness of antigen presenting cells. Subunit components also independently stimulate certain immune responses. Mutant forms of HLTs have potent immunomodulatory effects without the toxicity associated with holotoxins.Expert opinion: HLTs have been the subject of abundant research exploring their use as vaccine adjuvants, in the treatment of autoimmune conditions, in cancer therapy, and for weight loss, proving that these molecules are promising tools in the field of immunotherapy.
Collapse
Affiliation(s)
- Mary-Peyton A Knapp
- University of South Carolina School of Medicine Greenville, Department of Biomedical Sciences, Greenville, SC, USA
| | - Taylor A Johnson
- University of South Carolina School of Medicine Greenville, Department of Biomedical Sciences, Greenville, SC, USA
| | - Madison K Ritter
- University of South Carolina School of Medicine Greenville, Department of Biomedical Sciences, Greenville, SC, USA
| | - Robert O Rainer
- University of South Carolina School of Medicine Greenville, Department of Biomedical Sciences, Greenville, SC, USA.,Prisma Health, Department of Pathology, Greenville, SC, USA
| | - Steven E Fiester
- University of South Carolina School of Medicine Greenville, Department of Biomedical Sciences, Greenville, SC, USA.,Prisma Health, Department of Pathology, Greenville, SC, USA
| | - Jennifer T Grier
- University of South Carolina School of Medicine Greenville, Department of Biomedical Sciences, Greenville, SC, USA
| | - Terry D Connell
- University of Buffalo, Jacobs School of Medicine and Biomedical Sciences and the Witebsky Center of Microbial Pathogenesis and Immunology, Buffalo, NY, USA
| | - Sergio Arce
- University of South Carolina School of Medicine Greenville, Department of Biomedical Sciences, Greenville, SC, USA.,Prisma Health, Cancer Institute, Greenville, SC, USA
| |
Collapse
|
3
|
Royal JM, Reeves MA, Matoba N. Repeated Oral Administration of a KDEL-tagged Recombinant Cholera Toxin B Subunit Effectively Mitigates DSS Colitis Despite a Robust Immunogenic Response. Toxins (Basel) 2019; 11:E678. [PMID: 31756977 PMCID: PMC6950078 DOI: 10.3390/toxins11120678] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 11/18/2019] [Accepted: 11/19/2019] [Indexed: 12/22/2022] Open
Abstract
Cholera toxin B subunit (CTB), a non-toxic homopentameric component of Vibrio cholerae holotoxin, is an oral cholera vaccine antigen that induces an anti-toxin antibody response. Recently, we demonstrated that a recombinant CTB variant with a Lys-Asp-Glu-Leu (KDEL) endoplasmic reticulum retention motif (CTB-KDEL) exhibits colon mucosal healing effects that have therapeutic implications for inflammatory bowel disease (IBD). Herein, we investigated the feasibility of CTB-KDEL for the treatment of chronic colitis. We found that weekly oral administration of CTB-KDEL, dosed before or after the onset of chronic colitis, induced by repeated dextran sodium sulfate (DSS) exposure, could significantly reduce disease activity index scores, intestinal permeability, inflammation, and histological signs of chronicity. To address the consequences of immunogenicity, mice (C57BL/6 or C3H/HeJ strains) were pre-exposed to CTB-KDEL then subjected to DSS colitis and CTB-KDEL treatment. While the pre-dosing of CTB-KDEL elicited high-titer anti-drug antibodies (ADAs) of the immunoglobin A (IgA) isotype in the intestine of C57BL/6 mice, the therapeutic effects of CTB-KDEL were similar to those observed in C3H/HeJ mice, which showed minimal ADAs under the same experimental conditions. Thus, the immunogenicity of CTB-KDEL does not seem to impede the protein's mucosal healing efficacy. These results support the development of CTB-KDEL for IBD therapy.
Collapse
Affiliation(s)
- Joshua M. Royal
- James Graham Brown Cancer Center, Center for Predictive Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA;
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA;
| | - Micaela A. Reeves
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA;
| | - Nobuyuki Matoba
- James Graham Brown Cancer Center, Center for Predictive Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA;
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA;
| |
Collapse
|
4
|
ADP-ribosylating enterotoxins as vaccine adjuvants. Curr Opin Pharmacol 2018; 41:42-51. [PMID: 29702466 DOI: 10.1016/j.coph.2018.03.015] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 03/30/2018] [Indexed: 01/18/2023]
Abstract
Most infections are caused by pathogens that access the body at mucosal sites. Hence, development of mucosal vaccines to prevent local infection or invasion of pathogens appears highly warranted, especially since only mucosal immunization will stimulate strong local IgA responses and tissue resident memory CD4 and CD8 T cells. The most significant obstacle to developing such vaccines is the lack of approved adjuvants that can effectively and safely enhance relevant mucosal and systemic immune responses. The most potent mucosal adjuvants known today are the adenosine diphosphate (ADP)-ribosylating bacterial enterotoxins cholera toxin (CT) and Escherichia coli heat-labile toxins (LTs). Unfortunately, these molecules are also very toxic, which precludes their clinical use. However, much effort has been devoted to developing derivatives of these enterotoxins with low or no toxicity and retained adjuvant activity. Although it is fair to say that we know more about how these toxins affect the immune system than ever before, we still lack a detailed understanding of how and why these toxins are effective adjuvants. In the present review, we provide a state-of-the-art overview of the mechanism of action of the holotoxins and the strategies used for improving the toxin-based adjuvants.
Collapse
|
5
|
Marinova S, Nenkov P, Markova R, Nikolaeva S, Kostadinova R, Mitov I, Vretenarska M. Cellular and Humoral Systemic and Mucosal Immune Responses Stimulated by an Oral Polybacterial Immunomodulator in Patients with Chronic Urinary Tract Infections. Int J Immunopathol Pharmacol 2016; 18:457-473. [PMID: 16164822 DOI: 10.1177/039463200501800306] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
An oral polybacterial immunomodulator Urostim (U), composed of killed cells and their lysates from E. coli expressing type 1 and P-pili, E. coli Re mutant, P. mirabilis, K. pneumoniae and E. faecalis was created for immunoprophylaxis and immunotherapy of urinary tract infections (UTIs). In experimental animal models, the stimulating effect of U on lymphocyte functional activity, macrophage phagocytosis and antibody producing cells, was established. In this study the immuno-modulating effects of U on the proliferating capacity and ultrastructural morphologic changes of lymphocytes, cytokine production and specific systemic humoral and mucosal immune responses in patients with UTIs have been evaluated. Patients enrolled in the study, received orally 50 mg U daily for a period of three months. On days 0,30 and 90 a quantitative analysis was performed on lymphoproliferative responses to polyclonal mitogens, IL-2 and the specific antigen U, the production of specific serum and saliva IgA, IgM and IgG antibodies to all components of U and the concentration of pro-inflammatory cytokines. There was significant improvement of non-specific and specific lymphoproliferative responses on days 30 and 90 after the onset of treatment with U, confirmed by electron-microscopic studies. The highest concentrations of serum proinflammatory cytokines TNF-α, IL-1β, and IL-6 were registered at baseline followed by a decrease until the end of the observation period. This finding correlates with the gradual decrease of immune activation as measured by the spontaneous lymphocyte proliferation. Data from the production of specific antibacterial antibodies in serum and saliva show two types of reactions. The first type was registered in patients with low pre-treatment levels in whom the concentration of specific antibodies increased on days 30 and 90. The second type of reaction was observed in patients with high pre-treatment levels, which dropped on day 30 and were usually followed by an increase at the end of the study. These results provide evidence for the immuno-modulating effect of U. Our data show that the oral administration of the polybacterial immunomodulator Urostim stimulates adequate cellular and humoral systemic and mucosal immune responses in patients with chronic UTIs.
Collapse
Affiliation(s)
- S Marinova
- National Center of Infectious and Parasitic Diseases (NCIPD), Sofia, Bulgaria
| | | | | | | | | | | | | |
Collapse
|
6
|
The case for plant-made veterinary immunotherapeutics. Biotechnol Adv 2016; 34:597-604. [PMID: 26875776 DOI: 10.1016/j.biotechadv.2016.02.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 01/14/2016] [Accepted: 02/11/2016] [Indexed: 12/11/2022]
Abstract
The excessive use of antibiotics in food animal production has contributed to resistance in pathogenic bacteria, thereby triggering regulations and consumer demands to limit their use. Alternatives for disease control are therefore required that are cost-effective and compatible with intensive production. While vaccines are widely used and effective, they are available against a minority of animal diseases, and development of novel vaccines and other immunotherapeutics is therefore needed. Production of such proteins recombinantly in plants can provide products that are effective and safe, can be orally administered with minimal processing, and are easily scalable with a relatively low capital investment. The present report thus advocates the use of plants for producing vaccines and antibodies to protect farm animals from diseases that have thus far been managed with antibiotics; and highlights recent advances in product efficacy, competitiveness, and regulatory approval.
Collapse
|
7
|
Li H, Zhang J, He Y, Li B, Chen L, Huang W, Zou Q, Wu C. Intranasal immunization with an epitope-based vaccine results in earlier protection, but not better protective efficacy, against Helicobacter pylori compared to subcutaneous immunization. Immunol Res 2015; 62:368-76. [DOI: 10.1007/s12026-015-8666-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
8
|
Qi Y, Kang H, Zheng X, Wang H, Gao Y, Yang S, Xia X. Incorporation of membrane-anchored flagellin or Escherichia coli heat-labile enterotoxin B subunit enhances the immunogenicity of rabies virus-like particles in mice and dogs. Front Microbiol 2015; 6:169. [PMID: 25784906 PMCID: PMC4347500 DOI: 10.3389/fmicb.2015.00169] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 02/13/2015] [Indexed: 01/20/2023] Open
Abstract
Rabies remains an important worldwide public health threat, so safe, effective, and affordable vaccines are still being sought. Virus-like particle-based vaccines targeting various viral pathogens have been successfully produced, licensed, and commercialized. Here, we designed and constructed two chimeric rabies virus-like particles (cRVLPs) containing rabies virus (RABV) glycoprotein (G), matrix (M) protein, and membrane-anchored flagellin (EVLP-F) or Escherichia coli heat-labile enterotoxin B subunit (EVLP-L) as molecular adjuvants to enhance the immune response against rabies. The immunogenicity and potential of cRVLPs as novel rabies vaccine were evaluated by intramuscular vaccination in mouse and dog models. Mouse studies demonstrated that both EVLP-F and EVLP-L induced faster and larger virus-neutralizing antibodies (VNAs) responses and elicited greater numbers of CD4+ and CD8+ T cells secreting IFN-γ or IL-4 compared with a standard rabies VLP (sRVLP) containing only G and M. Moreover, cRVLPs recruited and/or activated more B cells and dendritic cells in inguinal lymph nodes. EVLP-F induced a strong, specific IgG2a response but not an IgG1 response, suggesting the activation of Th1 class immunity; in contrast, Th2 class immunity was observed with EVLP-L. The significantly enhanced humoral and cellular immune responses induced by cRVLPs provided complete protection against lethal challenge with RABV. Most importantly, dogs vaccinated with EVLP-F or EVLP-L exhibited increased VNA titers in sera and enhanced IFN-γ and IL-4 secretion from peripheral blood mononuclear cells. Taken together, these results illustrate that when incorporated into sRVLP, membrane-anchored flagellin, and heat-labile enterotoxin B subunit possess strong adjuvant activity. EVLP-F and EVLP-L induce significantly enhanced RABV-specific humoral and cellular immune responses in both mouse and dog. Therefore, these cRVLPs may be developed as safe and more efficacious rabies vaccine candidate for animals.
Collapse
Affiliation(s)
- Yinglin Qi
- College of Veterinary Medicine, Jilin University Changchun, China ; Institute of Military Veterinary Medicine, Academy of Military Medical Science Changchun, China
| | - Hongtao Kang
- Institute of Military Veterinary Medicine, Academy of Military Medical Science Changchun, China ; College of Veterinary Medicine, South China Agricultural University Guangzhou, China
| | - Xuexing Zheng
- Institute of Military Veterinary Medicine, Academy of Military Medical Science Changchun, China
| | - Hualei Wang
- Institute of Military Veterinary Medicine, Academy of Military Medical Science Changchun, China
| | - Yuwei Gao
- Institute of Military Veterinary Medicine, Academy of Military Medical Science Changchun, China
| | - Songtao Yang
- College of Veterinary Medicine, Jilin University Changchun, China ; Institute of Military Veterinary Medicine, Academy of Military Medical Science Changchun, China
| | - Xianzhu Xia
- College of Veterinary Medicine, Jilin University Changchun, China ; Institute of Military Veterinary Medicine, Academy of Military Medical Science Changchun, China
| |
Collapse
|
9
|
Azegami T, Yuki Y, Kiyono H. Challenges in mucosal vaccines for the control of infectious diseases. Int Immunol 2014; 26:517-28. [DOI: 10.1093/intimm/dxu063] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
10
|
Alejo DM, Moraes MP, Liao X, Dias CC, Tulman ER, Diaz-San Segundo F, Rood D, Grubman MJ, Silbart LK. An adenovirus vectored mucosal adjuvant augments protection of mice immunized intranasally with an adenovirus-vectored foot-and-mouth disease virus subunit vaccine. Vaccine 2013; 31:2302-9. [PMID: 23499593 DOI: 10.1016/j.vaccine.2013.02.060] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 02/08/2013] [Accepted: 02/27/2013] [Indexed: 11/30/2022]
Abstract
Foot-and-mouth disease virus (FMDV) is a highly contagious pathogen that causes severe morbidity and economic losses to the livestock industry in many countries. The oral and respiratory mucosae are the main ports of entry of FMDV, so the stimulation of local immunity in these tissues may help prevent initial infection and viral spread. E. coli heat-labile enterotoxin (LT) has been described as one of the few molecules that have adjuvant activity at mucosal surfaces. The objective of this study was to evaluate the efficacy of replication-defective adenovirus 5 (Ad5) vectors encoding either of two LT-based mucosal adjuvants, LTB or LTR72. These vectored adjuvants were delivered intranasally to mice concurrent with an Ad5-FMDV vaccine (Ad5-A24) to assess their ability to augment mucosal and systemic humoral immune responses to Ad5-A24 and protection against FMDV. Mice receiving Ad5-A24 plus Ad5-LTR72 had higher levels of mucosal and systemic neutralizing antibodies than those receiving Ad5-A24 alone or Ad5-A24 plus Ad5-LTB. The vaccine plus Ad5-LTR72 group also demonstrated 100% survival after intradermal challenge with a lethal dose of homologous FMDV serotype A24. These results suggest that Ad5-LTR72 could be used as an important tool to enhance mucosal and systemic immunity against FMDV and potentially other pathogens with a common route of entry.
Collapse
Affiliation(s)
- Diana M Alejo
- Department of Animal Science, University of Connecticut, Storrs, CT 06269, United States
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Bellot P, Tiels P, Melkebeek V, Devriendt B, Goddeeris B, Cox E. Maltose-binding protein is a potential carrier for oral immunizations. Vet Immunol Immunopathol 2013; 152:101-8. [DOI: 10.1016/j.vetimm.2012.09.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
12
|
Nochi T, Yuki Y, Takahashi H, Sawada SI, Mejima M, Kohda T, Harada N, Kong IG, Sato A, Kataoka N, Tokuhara D, Kurokawa S, Takahashi Y, Tsukada H, Kozaki S, Akiyoshi K, Kiyono H. Nanogel antigenic protein-delivery system for adjuvant-free intranasal vaccines. NATURE MATERIALS 2010; 9:572-578. [PMID: 20562880 DOI: 10.1038/nmat2784] [Citation(s) in RCA: 342] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2009] [Accepted: 05/10/2010] [Indexed: 05/28/2023]
Abstract
Nanotechnology is an innovative method of freely controlling nanometre-sized materials. Recent outbreaks of mucosal infectious diseases have increased the demands for development of mucosal vaccines because they induce both systemic and mucosal antigen-specific immune responses. Here we developed an intranasal vaccine-delivery system with a nanometre-sized hydrogel ('nanogel') consisting of a cationic type of cholesteryl-group-bearing pullulan (cCHP). A non-toxic subunit fragment of Clostridium botulinum type-A neurotoxin BoHc/A administered intranasally with cCHP nanogel (cCHP-BoHc/A) continuously adhered to the nasal epithelium and was effectively taken up by mucosal dendritic cells after its release from the cCHP nanogel. Vigorous botulinum-neurotoxin-A-neutralizing serum IgG and secretory IgA antibody responses were induced without co-administration of mucosal adjuvant. Importantly, intranasally administered cCHP-BoHc/A did not accumulate in the olfactory bulbs or brain. Moreover, intranasally immunized tetanus toxoid with cCHP nanogel induced strong tetanus-toxoid-specific systemic and mucosal immune responses. These results indicate that cCHP nanogel can be used as a universal protein-based antigen-delivery vehicle for adjuvant-free intranasal vaccination.
Collapse
Affiliation(s)
- Tomonori Nochi
- Division of Mucosal Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Liang S, Hajishengallis G. Heat-Labile Enterotoxins as Adjuvants or Anti-Inflammatory Agents. Immunol Invest 2010; 39:449-67. [DOI: 10.3109/08820130903563998] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Shuang Liang
- University of Louisville School of Dentistry, Oral Health and Systemic Disease, Louisville, KY, USA
| | - George Hajishengallis
- University of Louisville School of Dentistry, Oral Health and Systemic Disease, Louisville, KY, USA
- University of Louisville School of Medicine, Department of Microbiology and Immunology, Louisville, KY, USA
| |
Collapse
|
14
|
Concomitant enterotoxigenic Escherichia coli infection induces increased immune responses to Vibrio cholerae O1 antigens in patients with cholera in Bangladesh. Infect Immun 2010; 78:2117-24. [PMID: 20176796 DOI: 10.1128/iai.01426-09] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Vibrio cholerae O1 and enterotoxigenic Escherichia coli (ETEC) are major bacterial pathogens that cause dehydrating disease requiring hospitalization of children and adults. The cholera toxin (CT) produced by V. cholerae O1 and the heat-labile toxin (LT) and/or heat-stable toxin (ST) of ETEC are responsible for secretory diarrhea. We have observed that about 13% of hospitalized diarrheal patients are concomitantly infected with V. cholerae O1 and ETEC. In order to understand the outcome of such dual infections on the clinical and immunological responses in cholera patients, we studied patients infected with V. cholerae O1 (group VC; n = 25), those infected with both V. cholerae O1 and ETEC (group VCET; n = 25), and those infected with ETEC only (group ET; n = 25). The VCET group showed more severe dehydration and had a higher intake of intravenous fluid and more vomiting than the ETEC group (P = 0.01 to 0.003). The VCET patients showed higher vibriocidal responses and increased antibody titers to cholera toxin and lipopolysaccharide (LPS) in plasma than did the V. cholerae O1 patients (P = 0.02 to <0.001). All responses in the V. cholerae O1 and in the VCET groups were more robust than those seen in the group infected with ETEC only (P = 0.01 to <0.001). We thus show that concomitant colonization with ETEC induces immune responses to V. cholerae antigens that are more robust than those seen with V. cholerae O1 infection alone. It is possible that LT or other factors expressed by ETEC may play a role as a mucosal adjuvant in enhancing the immune responses to V. cholerae O1.
Collapse
|
15
|
Molina DM, Pal S, Kayala MA, Teng A, Kim PJ, Baldi P, Felgner PL, Liang X, de la Maza LM. Identification of immunodominant antigens of Chlamydia trachomatis using proteome microarrays. Vaccine 2009; 28:3014-24. [PMID: 20044059 DOI: 10.1016/j.vaccine.2009.12.020] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2009] [Revised: 12/02/2009] [Accepted: 12/14/2009] [Indexed: 12/26/2022]
Abstract
Chlamydia trachomatis is the most common bacterial sexually transmitted pathogen in the world. In order to control this infection there is an urgent need to formulate a vaccine. Identification of protective antigens is required to implement a subunit vaccine. To identify potential antigen vaccine candidates, three strains of mice, BALB/c, C3H/HeN and C57BL/6, were inoculated with live and inactivated C. trachomatis mouse pneumonitis (MoPn) by different routes of immunization. Using a protein microarray, serum samples collected after immunization were tested for the presence of antibodies against specific chlamydial antigens. A total of 225 open reading frames (ORF) of the C. trachomatis genome were cloned, expressed, and printed in the microarray. Using this protein microarray, a total of seven C. trachomatis dominant antigens were identified (TC0052, TC0189, TC0582, TC0660, TC0726, TC0816 and, TC0828) as recognized by IgG antibodies from all three strains of animals after immunization. In addition, the microarray was probed to determine if the antibody response exhibited a Th1 or Th2 bias. Animals immunized with live organisms mounted a predominant Th1 response against most of the chlamydial antigens while mice immunized with inactivated Chlamydia mounted a Th2-biased response. In conclusion, using a high throughput protein microarray we have identified a set of novel proteins that can be tested for their ability to protect against a chlamydial infection.
Collapse
Affiliation(s)
- Douglas M Molina
- ImmPORT Therapeutics, 1 Technology Drive, Suite E309, Irvine, CA 92618, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Kataoka K, Fujihashi K. Dendritic cell-targeting DNA-based mucosal adjuvants for the development of mucosal vaccines. Expert Rev Vaccines 2009; 8:1183-93. [PMID: 19722892 DOI: 10.1586/erv.09.80] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
In order to establish effective mucosal immunity against various mucosal pathogens, vaccines must be delivered via the mucosal route and contain effective adjuvant(s). Since mucosal adjuvants can simply mix with the antigen, it is relatively easy to adapt them for different types of vaccine development. Even in simple admixture vaccines, the adjuvant itself must be prepared without any complications. Thus, CpG oligodeoxynucleotides or plasmids encoding certain cDNA(s) would be potent mucosal adjuvant candidates when compared with other substances that can be used as mucosal adjuvants. The strategy of a DNA-based mucosal adjuvant facilitates the targeting of mucosal dendritic cells, and thus is an effective and safe approach. It would also provide great flexibility for the development of effective vaccines for various mucosal pathogens.
Collapse
Affiliation(s)
- Kosuke Kataoka
- Department of Preventive Dentistry, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan.
| | | |
Collapse
|
17
|
Ishii M, Kojima N. Mucosal adjuvant activity of oligomannose-coated liposomes for nasal immunization. Glycoconj J 2009; 27:115-23. [PMID: 19816665 DOI: 10.1007/s10719-009-9263-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2009] [Revised: 09/23/2009] [Accepted: 09/24/2009] [Indexed: 10/20/2022]
Abstract
In the present study, we investigated the effectiveness of liposomes coated with a neoglycolipid consisting of mannotriose and dipalmitoylphosphatidylcholine (Man3-DPPE) as an adjuvant for induction of mucosal immunity. Immunization of BALB/c mice with ovalbumin (OVA)-encapsulated Man3-DPPE-coated liposomes (oligomannose-coated liposomes; OMLs) by a nasal route produced high levels of OVA-specific IgG and IgA antibodies in serum of immunized mice 1 week after the last nasal immunization, whereas no significant serum antibody responses were observed in mice that received OVA in uncoated liposomes or OVA alone. Seven weeks after the last nasal immunization, nasal challenge with an excess amount of OVA in mice that had received OVA/OMLs led to an anamnestic response to the antigen that resulted in 5- to 10-fold increases of antigen-specific serum IgG and IgA antibodies. Only mice immunized nasally with OML/OVA secreted antigen-specific secretory IgA in nasal washes and produced interferon-gamma secreting cells in nasopharyngeal-associated lymphoreticular tissue. Taken together, these results show that nasal administration of OMLs induces mucosal and systemic immunity that are specific for the entrapped antigen in the liposomes. Thus, liposomes coated with synthetic neoglycolipids might be useful as adjuvants for induction of mucosal immunity.
Collapse
Affiliation(s)
- Mariko Ishii
- Institute of Glycoscience and Department of Applied Biochemistry, Tokai University, Hiratsuka, Kanagawa, 259-1292, Japan
| | | |
Collapse
|
18
|
Chen CG, Lu YT, Lin M, Savelyeva N, Stevenson FK, Zhu D. Amplification of immune responses against a DNA-delivered idiotypic lymphoma antigen by fusion to the B subunit of E. coli heat labile toxin. Vaccine 2009; 27:4289-96. [DOI: 10.1016/j.vaccine.2009.05.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2009] [Revised: 04/27/2009] [Accepted: 05/10/2009] [Indexed: 10/20/2022]
|
19
|
Donaldson DS, Williams NA. Bacterial toxins as immunomodulators. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2009; 666:1-18. [PMID: 20054971 DOI: 10.1007/978-1-4419-1601-3_1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Bacterial toxins are the causative agent at pathology in a variety of diseases. Although not always the primary target of these toxins, many have been shown to have potent immunomodulatory effects, for example, inducing immune responses to co-administered antigens and suppressing activation of immune cells. These abilities of bacterial toxins can be harnessed and used in a therapeutic manner, such as in vaccination or the treatment of autoimmune diseases. Furthermore, the ability of toxins to gain entry to cells can be used in novel bacterial toxin based immuno-therapies in order to deliver antigens into MHC Class I processing pathways. Whether the immunomodulatory properties of these toxins arose in order to enhance bacterial survival within hosts, to aid spread within the population or is pure serendipity, it is interesting to think that these same toxins potentially hold the key to preventing or treating human disease.
Collapse
Affiliation(s)
- David S Donaldson
- Department of Cellular and Molecular Medicine, School of Medicine Sciences, University of Bristol, Bristol, UK
| | | |
Collapse
|
20
|
|
21
|
Transcytosis of F4 fimbriae by villous and dome epithelia in F4-receptor positive pigs supports importance of receptor-dependent endocytosis in oral immunization strategies. Vet Immunol Immunopathol 2008; 124:29-40. [DOI: 10.1016/j.vetimm.2006.10.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2006] [Revised: 09/26/2006] [Accepted: 10/11/2006] [Indexed: 11/24/2022]
|
22
|
Brynjolfsson SF, Bjarnarson SP, Mori E, Del Giudice G, Jonsdottir I. Neonatal immune response and serum bactericidal activity induced by a meningococcal conjugate vaccine is enhanced by LT-K63 and CpG2006. Vaccine 2008; 26:4557-62. [PMID: 18597905 DOI: 10.1016/j.vaccine.2008.05.083] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2008] [Revised: 05/13/2008] [Accepted: 05/29/2008] [Indexed: 11/20/2022]
Abstract
Neonates have a poorly developed immune system. Therefore it is important to develop vaccination strategies that induce protective immunity and immunological memory against pathogens early in life. The immunogenicity of a meningococcal serogroup C polysaccharide conjugate (MenC-CRM(197)) was assessed in neonatal mice, and effects of LT-K63 and CpG2006 and immunisation routes were compared. Neonatal mice were primed subcutaneously (s.c.) or intranasally (i.n.) with MenC-CRM(197) with or without LT-K63 or CpG2006 and re-immunised 16 and 30 days later by the same route and formulation. Antibody levels were measured and generation of immunological memory assessed by affinity maturation and kinetics of the Ab response. Serum bactericidal activity (SBA) was measured to evaluate protective efficacy. The second and third dose of MenC-CRM(197) mixed with either LT-K63 or CpG2006 induced a rapid increase in MenC-specific IgG antibodies, to levels higher than elicited by MenC-CRM(197) alone (P<0.01) and in unimmunised mice (P<0.001), indicating efficient generation of memory by priming through both s.c. and i.n. routes. SBA was detected after three s.c. immunisations with MenC-CRM(197) s.c. alone. However, only two doses of MenC-CRM(197)+LT-K63 or MenC-CRM(197)+CpG2006 were needed to induce SBA levels>16. LT-K63 and CpG2006 enhanced neonatal antibody responses, affinity maturation, immunological memory to the conjugate MenC-CRM(197) and protective immunity. These results encourage the development of neonatal vaccination strategies to induce protective immunity and immunological memory against meningococcal disease.
Collapse
|
23
|
Actinobacillus pleuropneumoniaevaccines: from bacterins to new insights into vaccination strategies. Anim Health Res Rev 2008; 9:25-45. [DOI: 10.1017/s1466252307001338] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
AbstractWith the growing emergence of antibiotic resistance and rising consumer demands concerning food safety, vaccination to prevent bacterial infections is of increasing relevance.Actinobacillus pleuropneumoniaeis the etiological agent of porcine pleuropneumonia, a respiratory disease leading to severe economic losses in the swine industry. Despite all the research and trials that were performed withA. pleuropneumoniaevaccination in the past, a safe vaccine that offers complete protection against all serotypes has yet not reached the market. However, recent advances made in the identification of new potential vaccine candidates and in the targeting of specific immune responses, give encouraging vaccination perspectives. Here, we review past and current knowledge onA. pleuropneumoniaevaccines as well as the newly available genomic tools and vaccination strategies that could be useful in the design of an efficient vaccine againstA. pleuropneumoniaeinfection.
Collapse
|
24
|
Guerrero GG, Moreno-Fierros L. Carrier potential properties of Bacillus thuringiensis Cry1A toxins for a diphtheria toxin epitope. Scand J Immunol 2007; 66:610-8. [PMID: 17949406 DOI: 10.1111/j.1365-3083.2007.01992.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The N-terminal half or toxic fragment of Bacillus thuringiensis Cry proteins is comprised of three structural domains. In a previous paper, we showed that this region plays an important role in the immunogenicity of the B. thuringiensis Cry proteins. Due to this ability and along with their stability it is worthy of investigating whether this region has carrier potential. To approach this, an eight amino acid hydrophobic motif in alpha-helix 7 of wild-type (WT) Cry1A toxins was exchanged for a diphtheria toxin epitope (DTB). The resultant recombinant toxins were tested for their ability to induce specific anti-Cry and anti-diphtheria toxin antibodies in mice after intraperitoneal and nasal immunization. We found that recombinant Cry1A toxins retained their ability to induce serum and mucosal anti-Cry Ab as well as IgG subclasses, although with a varied magnitude. By the systemic route, the effect of the amino acid substitution in the ratio of the IgG1/IgG2a Ab, leading in some sites toward IgG1 or IgG2a is more evident. Interestingly, mice produced specific anti-DTB IgG, and IgA after intranasal immunization. Together, our results support and show the immunogenic properties of the WT Cry1A toxins as well as its carrier potential for a DTB.
Collapse
Affiliation(s)
- G G Guerrero
- Lab de Inmunidad de Mucosas, UBIMED, FES-IZTACALA, UNAM, Tlanepantla Edo de México, Mexico.
| | | |
Collapse
|
25
|
Alcón VL, Baca-Estrada M, Vega-López MA, Willson P, Babiuk LA, Kumar P, Foldvari M. Intranasal immunization using biphasic lipid vesicles as delivery systems for OmlA bacterial protein antigen and CpG oligonucleotides adjuvant in a mouse model. J Pharm Pharmacol 2006; 57:955-62. [PMID: 16102250 DOI: 10.1211/0022357056695] [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] [Indexed: 10/31/2022]
Abstract
The nasal mucosa is an important arm of the mucosal system since it is often the first point of contact for inhaled antigens. The ineffectiveness of the simple delivery of soluble antigens to mucosal membranes for immunization has stimulated extensive studies in appropriate delivery systems and adjuvants. We have evaluated biphasic lipid vesicles as a novel intranasal (i.n.) delivery system (designated as vaccine targeting adjuvant, VTA) containing bacterial antigens and CpG oligodeoxynucleotides (ODNs). Results show that administration of antigen and CpG ODNs in biphasic lipid vesicles resulted in greater induction of IgA levels in serum (P< 0.05) and mucosal antibody responses such as IgA in nasal secretions and lung (P< 0.01) after immunization with a combined subcutaneous (s.c.)/i.n. as compared to s.c./s.c. approach. Based on antibody responses, VTA formulations were found to be suitable as delivery systems for antigens and CpG ODNs by the intranasal route, resulting in a Th2-type of immune response, characterized by IgG1 and IL-4 production at the systemic level.
Collapse
Affiliation(s)
- V L Alcón
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, Canada
| | | | | | | | | | | | | |
Collapse
|
26
|
Tammiruusu A, Penttilä T, Lahesmaa R, Sarvas M, Puolakkainen M, Vuola JM. Intranasal administration of chlamydial outer protein N (CopN) induces protection against pulmonary Chlamydia pneumoniae infection in a mouse model. Vaccine 2006; 25:283-90. [PMID: 16949182 DOI: 10.1016/j.vaccine.2006.07.043] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2006] [Revised: 06/16/2006] [Accepted: 07/20/2006] [Indexed: 11/24/2022]
Abstract
Chlamydia pneumoniae is an intracellular pathogen that grows inside a vacuole, referred to as an inclusion. C. pneumoniae possess a type III secretion system (TTSS), which allows them to secrete effector molecules into the inclusion membrane and to the host cell cytosol. Proteins such as chlamydial outer protein N (CopN) that associate with the inclusion membrane are potential targets for the host's MHC-dependent antigen presentation, thereby representing ideal antigen candidates for T cell-based vaccination. The results of this study showed that intranasal immunization of BALB/c mice with heat-aggregated CopN protein and an Escherichia coli heat-labile toxin (LT) induced a strong immune response, detected as antigen-specific antibody production, lymphocyte proliferation and IFN-gamma production. Furthermore, the immunization induced statistically significant protection against intranasal C. pneumoniae challenge, the level of which correlated with the magnitude of CopN-specific lymphocyte proliferation. Both heat-aggregation of the antigen and the presence of LT adjuvant were required for maximal protective effect.
Collapse
Affiliation(s)
- A Tammiruusu
- Department of Viral Diseases and Immunology, Infection Pathogenesis Laboratory, National Public Health Institute, Mannerheimintie 166, 00300 Helsinki, Finland.
| | | | | | | | | | | |
Collapse
|
27
|
Giacalone MJ, Sabbadini RA, Chambers AL, Pillai S, McGuire KL. Immune responses elicited by bacterial minicells capable of simultaneous DNA and protein antigen delivery. Vaccine 2006; 24:6009-17. [PMID: 16806602 PMCID: PMC7125846 DOI: 10.1016/j.vaccine.2006.04.063] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2006] [Revised: 04/14/2006] [Accepted: 04/20/2006] [Indexed: 11/22/2022]
Abstract
Recent events surrounding emerging infectious diseases, bioterrorism and increasing multidrug antibiotic resistance in bacteria have drastically increased current needs for effective vaccines. Many years of study have shown that live, attenuated pathogens are often more effective at delivering heterologous protein or DNA to induce protective immune responses. However, these vaccine carriers have inherent safety concerns that have limited their development and their use in many patient populations. Studies using nonliving delivery mechanisms have shown that providing both protein antigen and DNA encoding the antigen to an individual induces an improved, more protective immune response but rarely, if ever, are both delivered simultaneously. Here, non-replicating bacterial minicells derived from a commensal E. coli strain are shown to effectively induce antigen-specific immune responses after simultaneous protein and DNA delivery. These data demonstrate the potential use of achromosomal bacterial minicells as a vaccine carrier.
Collapse
Affiliation(s)
| | - Roger A. Sabbadini
- The Molecular Biology Institute, San Diego State University, San Diego, CA, USA
- Department of Biology, Center For Microbial Sciences, San Diego State University, San Diego, CA, USA
| | - Amy L. Chambers
- The Molecular Biology Institute, San Diego State University, San Diego, CA, USA
| | - Sabitha Pillai
- The Molecular Biology Institute, San Diego State University, San Diego, CA, USA
| | - Kathleen L. McGuire
- The Molecular Biology Institute, San Diego State University, San Diego, CA, USA
- Department of Biology, Center For Microbial Sciences, San Diego State University, San Diego, CA, USA
- Corresponding author at: Department of Biology, San Diego State University, 5500 Campanile Drive, San Diego, CA 92182-4614, USA. Tel.: +1 619 594 7191; fax: +1 619 594 5676.
| |
Collapse
|
28
|
Duverger A, Jackson RJ, van Ginkel FW, Fischer R, Tafaro A, Leppla SH, Fujihashi K, Kiyono H, McGhee JR, Boyaka PN. Bacillus anthracis edema toxin acts as an adjuvant for mucosal immune responses to nasally administered vaccine antigens. THE JOURNAL OF IMMUNOLOGY 2006; 176:1776-83. [PMID: 16424208 DOI: 10.4049/jimmunol.176.3.1776] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Anthrax edema toxin (EdTx) is an AB-type toxin that binds to anthrax toxin receptors on target cells via the binding subunit, protective Ag (PA). Edema factor, the enzymatic A subunit of EdTx, is an adenylate cyclase. We found that nasal delivery of EdTx enhanced systemic immunity to nasally coadministered OVA and resulted in high OVA-specific plasma IgA and IgG (mainly IgG1 and IgG2b). The edema factor also enhanced immunity to the binding PA subunit itself and promoted high levels of plasma IgG and IgA responses as well as neutralizing PA Abs. Mice given OVA and EdTx also exhibited both PA- and OVA-specific IgA and IgG Ab responses in saliva as well as IgA Ab responses in vaginal washes. EdTx as adjuvant triggered OVA- and PA-specific + T cells which secreted IFN-gamma and selected Th2-type cytokines. The EdTx up-regulated costimulatory molecule expression by APCs but was less effective than cholera toxin for inducing IL-6 responses either by APCs in vitro or in nasal washes in vivo. Finally, nasally administered EdTx did not target CNS tissues and did not induce IL-1 mRNA responses in the nasopharyngeal-associated lymphoepithelial tissue or in the olfactory bulb epithelium. Thus, EdTx derivatives could represent an alternative to the ganglioside-binding enterotoxin adjuvants and provide new tools for inducing protective immunity to PA-based anthrax vaccines.
Collapse
Affiliation(s)
- Alexandra Duverger
- Department of Microbiology, University of Alabama at Birmingham, AL 35294, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Byrd W, Cassels FJ. The encapsulation of enterotoxigenic Escherichia coli colonization factor CS3 in biodegradable microspheres enhances the murine antibody response following intranasal administration. Microbiology (Reading) 2006; 152:779-786. [PMID: 16514157 DOI: 10.1099/mic.0.28667-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The aim of this study was to measure serum and mucosal antibody responses following intranasal administration of biodegradable poly(dl-lactide-co-glycolide) (PLGA) microspheres loaded with the CS3 colonization factor isolated from enterotoxigenic Escherichia coli (ETEC). The response was compared against that measured in mice similarly administered the native CS3 antigen and in mice co-administered, along with the CS3 antigen, a known mucosal adjuvant, the R192G mutant heat-labile enterotoxin (mLT). The integrity of the CS3 antigen released from the microspheres was maintained as determined by SDS-PAGE and immunoblotting. Native CS3 induced serum and mucosal (bronchoalveolar, small intestinal and faecal) IgG and IgA responses. The co-administration of the mLT mucosal adjuvant significantly enhanced (P<0·001) serum and mucosal antibody responses to the CS3 protein. Likewise, the CS3-loaded PLGA microspheres induced significantly greater (P<0·001) serum and mucosal antibody responses than native CS3, as well as inducing antibody responses superior to those of the CS3 plus mLT formulation. Following administration of CS3 plus mLT, the mice became distressed (loss of activity, increased huddling, ruffled fur), a situation not seen following administration of the CS3-loaded PLGA microspheres. The results in this trial show that the CS3-loaded PLGA microspheres when administered intranasally to mice caused no observable distress to the mice and significantly (P<0·001) enhanced the immunogenicity of the CS3 protein.
Collapse
Affiliation(s)
- Wyatt Byrd
- 18929 Fountain Hills Drive, Germantown, MD 20874, USA
- Department of Enteric Infections, Walter Reed Army Institute of Research, Silver Spring, MD 20910-7500, USA
| | - Frederick J Cassels
- Department of Enteric Infections, Walter Reed Army Institute of Research, Silver Spring, MD 20910-7500, USA
| |
Collapse
|
30
|
Pickering RJ, Smith SD, Strugnell RA, Wesselingh SL, Webster DE. Crude saponins improve the immune response to an oral plant-made measles vaccine. Vaccine 2006; 24:144-50. [PMID: 16154244 DOI: 10.1016/j.vaccine.2005.07.097] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2005] [Accepted: 07/28/2005] [Indexed: 11/22/2022]
Abstract
Millions of people live in areas where infectious diseases, such as measles, are endemic and resources are scarce. Heat-stable vaccines that are delivered orally will greatly enhance vaccination programs in these areas. A stumbling block in the development of oral vaccines is the availability of safe and effective mucosal adjuvants, especially for use with subunit vaccines. The experiments presented here examine the ability of CTB/CT, LT(R192G) and crude Quillaja saponin extracts to stimulate MV-specific immune responses in mice, following oral immunisation with plant-made measles virus hemagglutinin (MV-H) protein. LT(R192G) and crude saponin extracts both functioned as potent mucosal adjuvants when ad-mixed with plant-made MV-H protein, and were more effective than CTB/CT. MV-H protein supplemented with saponin extract induced the strongest MV-specific responses, in the greatest number of mice. Crude saponins are routinely used by the food and beverage industry at concentrations greater than those required for adjuvanticity, and as such, they have a better safety profile than bacterial enterotoxins. This study demonstrates their potential as adjuvants for use with oral plant-made vaccines.
Collapse
Affiliation(s)
- R J Pickering
- The Macfarlane Burnet Institute for Medical Research and Public Health, P.O. Box 2284, Melbourne, Vic. 3001, Australia
| | | | | | | | | |
Collapse
|
31
|
van Ginkel FW, Jackson RJ, Yoshino N, Hagiwara Y, Metzger DJ, Connell TD, Vu HL, Martin M, Fujihashi K, McGhee JR. Enterotoxin-based mucosal adjuvants alter antigen trafficking and induce inflammatory responses in the nasal tract. Infect Immun 2005; 73:6892-902. [PMID: 16177369 PMCID: PMC1230900 DOI: 10.1128/iai.73.10.6892-6902.2005] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The safety of nasal vaccines containing enterotoxin-based mucosal adjuvants has not been studied in detail. Previous studies have indicated that native cholera toxin (nCT) can alter antigen trafficking when applied nasally. In this study, we determined the enterotoxin-based variables that alter antigen trafficking. To measure the influence of enterotoxin-based mucosal adjuvants on antigen trafficking in the nasal tract, native and mutant enterotoxins were coadministered with radiolabeled tetanus toxoid (TT). The nCT and heat-labile enterotoxin type 1 (LTh-1) redirected TT into the olfactory neuroepithelium (ON/E). Antigen redirection occurred mainly across the nasal epithelium without subsequent transport along olfactory neurons into the olfactory bulbs (OB). Thus, no significant accumulation of the vaccine antigen TT was observed in the OB when coadministered with nCT. In contrast, neither mutant CT nor mutant LTh-1, which lack ADP-ribosyltransferase activity, redirected TT antigen into the ON/E. Thus, ADP-ribosyltransferase activity was essential for antigen trafficking across the olfactory epithelium. Accumulation of TT in the ON/E was also due to B-subunit binding to GM1 gangliosides, as was demonstrated (i) by redirection of TT by LTh-1 in a dose-dependent manner, (ii) by ganglioside inhibition of the antigen redirection by LTh-1 and nCT, and (iii) by the use of LT-IIb, a toxin that binds to gangliosides other than GM1. Redirection of TT into the ON/E coincided with elevated production of interleukin 6 (IL-6) but not IL-1beta or tumor necrosis factor alpha in the nasal mucosa. Thus, redirection of TT is dependent on ADP-ribosyltransferase activity and GM1 binding and is associated with production of the inflammatory cytokine IL-6.
Collapse
Affiliation(s)
- Frederik W van Ginkel
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, AL 36849, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Alcon V, Baca-Estrada M, Vega-Lopez M, Willson P, Babiuk LA, Kumar P, Hecker R, Foldvari M. Mucosal delivery of bacterial antigens and CpG oligonucleotides formulated in biphasic lipid vesicles in pigs. AAPS JOURNAL 2005; 7:E566-71. [PMID: 16353934 PMCID: PMC2751259 DOI: 10.1208/aapsj070357] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The ineffectiveness of simple delivery of soluble antigens to mucosal membranes for immunization has stimulated extensive studies of strategies for appropriate delivery systems and adjuvants. Biphasic lipid vesicles are formulations suitable for the delivery of proteins, peptides, and oligo/polynucleotides. The purpose of these studies was to investigate the ability of biphasic lipid vesicles (as vaccine-targeting adjuvants) containing a bacterial antigen and unmethylated oligonucleotides containing CGdinucleotides - CpG motifs (CpG ODNs) to induce systemic and mucosal immune responses in pigs. Results showed that while the protein, either alone or with CpG ODNs, did not induce mucosal immune responses, administration of antigen and CpG ODNs in biphasic lipid vesicles resulted in induction of both systemic and local antibody responses after immunization using a combined mucosal/systemic approach.
Collapse
Affiliation(s)
- Valeria Alcon
- />Department of Pharmaceutics/Pharmacy, College of Pharmacy and Nutrition, University of Saskatchewan, 110 Science Place, S7N 5C9 Saskatoon, Saskatchewan Canada
- />Vaccine and Infectious Disease Organization, Saskatoon, SK Canada
| | | | - Marco Vega-Lopez
- />Vaccine and Infectious Disease Organization, Saskatoon, SK Canada
| | - Philip Willson
- />Vaccine and Infectious Disease Organization, Saskatoon, SK Canada
| | - Lorne A. Babiuk
- />Vaccine and Infectious Disease Organization, Saskatoon, SK Canada
| | - Praveen Kumar
- />PharmaDerm Laboratories Ltd, Innovation Place Research Park, Saskatoon, SK Canada
| | | | - Marianna Foldvari
- />Department of Pharmaceutics/Pharmacy, College of Pharmacy and Nutrition, University of Saskatchewan, 110 Science Place, S7N 5C9 Saskatoon, Saskatchewan Canada
| |
Collapse
|
33
|
Ohmura M, Yamamoto M, Tomiyama-Miyaji C, Yuki Y, Takeda Y, Kiyono H. Nontoxic Shiga toxin derivatives from Escherichia coli possess adjuvant activity for the augmentation of antigen-specific immune responses via dendritic cell activation. Infect Immun 2005; 73:4088-97. [PMID: 15972497 PMCID: PMC1168555 DOI: 10.1128/iai.73.7.4088-4097.2005] [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: 11/20/2022] Open
Abstract
Shiga toxin (Stx) derivatives, such as the Stx1 B subunit (StxB1), which mediates toxin binding to the membrane, and mutant Stx1 (mStx1), which is a nontoxic doubly mutated Stx1 harboring amino acid substitutions in the A subunit, possess adjuvant activity via the activation of dendritic cells (DCs). Our results showed that StxB1 and mStx1, but not native Stx1 (nStx1), resulted in enhanced expression of CD86, CD40, and major histocompatibility complex (MHC) class II molecules and, to some extent, also enhanced the expression of CD80 on bone marrow-derived DCs. StxB1-treated DCs exhibited an increase in tumor necrosis factor alpha and interleukin-12 (IL-12) production, a stimulation of DO11.10 T-cell proliferation, and the production of both Th1 and Th2 cytokines, including gamma interferon (IFN-gamma), IL-4, IL-5, IL-6, and IL-10. When mice were given StxB1 subcutaneously, the levels of CD80, CD86, and CD40, as well as MHC class II expression by splenic DCs, were enhanced. The subcutaneous immunization of mice with ovalbumin (OVA) plus mStx1 or StxB1 induced high titers of OVA-specific immunoglobulin M (IgM), IgG1, and IgG2a in serum. OVA-specific CD4+ T cells isolated from mice immunized with OVA plus mStx1 or StxB1 produced IFN-gamma, IL-4, IL-5, IL-6, and IL-10, indicating that mStx1 and StxB1 elicit both Th1- and Th2-type responses. Importantly, mice immunized subcutaneously with tetanus toxoid plus mStx1 or StxB1 were protected from a lethal challenge with tetanus toxin. These results suggest that nontoxic Stx derivatives, including both StxB1 and mStx1, could be effective adjuvants for the induction of mixed Th-type CD4+ T-cell-mediated antigen-specific antibody responses via the activation of DCs.
Collapse
Affiliation(s)
- Mari Ohmura
- Laboratory for Infectious Immunity, RIKEN Research Center for Allergy and Immunology, Yokohama City, Kanagawa, Japan
| | | | | | | | | | | |
Collapse
|
34
|
Abstract
Induction of immune responses following oral immunization is frequently dependent upon the co-administration of appropriate adjuvants that can initiate and support the transition from innate to adaptive immunity. The three bacterial products with the greatest potential to function as mucosal adjuvants are the ADP-ribosylating enterotoxins (cholera toxin and the heat-labile enterotoxin of Escherichia coli), synthetic oligodeoxynucleotides containing unmethylated CpG dinucleotides (CpG ODN), and monophosphoryl lipid A (MPL). The mechanism of adjuvanticity of the ADP-ribosylating enterotoxins is the subject of considerable debate. Our own view is that adjuvanticity is an outcome and not an event. It is likely that these molecules exert their adjuvant function by interacting with a variety of cell types, including epithelial cells, dendritic cells, macrophages, and possibly B- and T-lymphocytes. The adjuvant activities of CpG and MPL are due to several different effects they have on innate and adaptive immune responses and both MPL and CpG act through MyD88-dependent and -independent pathways. This presentation will summarize the probable mechanisms of action of these diverse mucosal adjuvants and discuss potential synergy between these molecules for use in conjunction with plant-derived vaccines.
Collapse
Affiliation(s)
- L C Freytag
- Department of Microbiology and Immunology, Tulane University Health Sciences Center, New Orleans, LA 70112, USA
| | | |
Collapse
|
35
|
Abstract
Developing efficient adjuvants for human vaccines, in order to elicit broad and sustained immune responses at systemic or mucosal levels, remains a challenge for the vaccine industry. Conventional approaches in the past have been largely empirical and partially successful. Selection was based on the balance between toxicity and adjuvanticity, first in an animal model, and then in clinical trials. The advent of improved biochemical techniques has allowed for the purification or construction of new and well characterised adjuvants. In addition, recent advances in our understanding of the immune system, most particularly with respect to early proinflammatory signals, have led to the identification of new biological targets for vaccine adjuvants. In particular, one can now choose adjuvants able to selectively induce T helper (Th)-1 and/or Th2 responses, according to the vaccine target and the desired immune response. As our knowledge of the cell types and cytokines interacting in the immune responses increases, so does our understanding of the mode of action of adjuvants, as well as the way in which they produce adverse effects.
Collapse
Affiliation(s)
- Bruno Guy
- Research Department, Aventis Pasteur, Marcy l'Etoile, France.
| | | |
Collapse
|
36
|
|
37
|
|
38
|
Lehner T, Bergmeier LA. Mucosal Infection and Immune Responses to Simian Immunodeficiency Virus. Mucosal Immunol 2005. [DOI: 10.1016/b978-012491543-5/50070-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
39
|
Fujihashi K, McGhee JR. Th1/Th2/Th3 Cells for Regulation of Mucosal Immunity, Tolerance, and Inflammation. Mucosal Immunol 2005. [DOI: 10.1016/b978-012491543-5/50032-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
40
|
Ohmura-Hoshino M, Yamamoto M, Yuki Y, Takeda Y, Kiyono H. Non-toxic Stx derivatives from Escherichia coli possess adjuvant activity for mucosal immunity. Vaccine 2004; 22:3751-61. [PMID: 15315856 DOI: 10.1016/j.vaccine.2004.03.034] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2003] [Revised: 02/09/2004] [Accepted: 03/02/2004] [Indexed: 10/26/2022]
Abstract
Both B subunit of Shiga toxin 1 (Stx1-B), which mediates the binding of toxin to the membrane, and mutant Stx1 (mStx1), which is a non-toxic double-mutated Stx1 harboring double amino acid substitutions in the A subunit, possess potent mucosal adjuvant activity. Nasal immunization of mice with ovalbumin (OVA) plus the Stx1-B or mStx1 induced OVA-specific serum IgG and mucosal IgA responses. IgG subclass analysis revealed that mStx1 and Stx1-B as mucosal adjuvants supported Ag-specific IgG1 followed by IgG2b Abs. The co-administration of either mStx1 or Stx1-B with OVA enhanced the production of IL-4, IL-5, IL-6 and IL-10 with low IFN-gamma, by OVA-specific CD4+ T cells. To better elucidate the mechanisms underlying mStx1's and Stx1-B's adjuvant activity, we next sought to examine whether or not dendritic cells (DC) residing in the nasopharyngeal-associated lymphoreticular tissue (NALT) were activated by nasal administration of Stx1-B or mStx1. We found that mice nasally administered with Stx1-B or mStx1 showed an up-regulation in the expression of CD80, CD86 and especially CD40 on NALT DCs. Taken together, these results suggest that non-toxic Stx derivatives could be effective mucosal adjuvants for the induction of Th2-type, CD4+ T cell mediated, antigen-specific mucosal IgA and systemic IgG Ab responses, and that they likely owe their adjuvant activity to the up-regulation of co-stimulatory molecules including CD80, CD86 and CD40 on NALT DCs.
Collapse
Affiliation(s)
- Mari Ohmura-Hoshino
- Department of Microbiology and Immunology, Division of Mucosal Immunology, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | | | | | | | | |
Collapse
|
41
|
Kataoka K, McGhee JR, Kobayashi R, Fujihashi K, Shizukuishi S, Fujihashi K. Nasal Flt3 ligand cDNA elicits CD11c+CD8+ dendritic cells for enhanced mucosal immunity. THE JOURNAL OF IMMUNOLOGY 2004; 172:3612-9. [PMID: 15004163 DOI: 10.4049/jimmunol.172.6.3612] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Nasal immunization is an effective way to induce both mucosal and systemic immune responses. In this study, we assessed a cDNA vector for Flt3 ligand (FL) for its potential to enhance mucosal immunity or tolerance. Interestingly, tolerance was avoided and elevated levels of OVA-specific Ab responses were induced in nasal washes, fecal extracts, and saliva as well as in plasma when compared with mice given nasal OVA plus DNA plasmid without the FL gene. In addition, significant levels of OVA-specific CD4+ T cell proliferative responses and OVA-induced IL-4 and IL-2 production were noted in spleen and cervical lymph nodes. Further, marked increases in FL protein occurred in the nasal lamina propria and submandibular glands and the frequencies of CD11c+CD8+ dendritic cells (DCs) significantly increased in the mucosal tissues. Moreover, these DCs expressed high levels of CD40, CD80, CD86, and MHC class II molecules. Nasal delivery of plasmid FL with OVA resulted in FL expression in both mucosal inductive and effector sites and resulted in expanded activated lymphoid DCs. Thus, nasal plasmid FL prevents mucosal tolerance and enhances active immunity when given by a mucosal route.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/genetics
- Administration, Intranasal
- Animals
- CD11c Antigen/biosynthesis
- CD4-Positive T-Lymphocytes/cytology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD8 Antigens/biosynthesis
- Cell Division/genetics
- Cell Division/immunology
- Cytokines/biosynthesis
- DNA, Complementary/administration & dosage
- DNA, Complementary/immunology
- Dendritic Cells/cytology
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Epitopes, T-Lymphocyte/immunology
- Female
- Genetic Vectors
- Immunity, Mucosal/genetics
- Immunoglobulin A/biosynthesis
- Ligands
- Membrane Proteins/biosynthesis
- Membrane Proteins/genetics
- Membrane Proteins/physiology
- Mice
- Mice, Inbred C57BL
- Nasal Mucosa/immunology
- Nasal Mucosa/metabolism
- Ovalbumin/administration & dosage
- Ovalbumin/immunology
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/immunology
Collapse
Affiliation(s)
- Kosuke Kataoka
- Department of Oral Biology, Immunobiology Vaccine Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | | | |
Collapse
|
42
|
Mauriello EMF, Duc LH, Isticato R, Cangiano G, Hong HA, De Felice M, Ricca E, Cutting SM. Display of heterologous antigens on the Bacillus subtilis spore coat using CotC as a fusion partner. Vaccine 2004; 22:1177-87. [PMID: 15003646 DOI: 10.1016/j.vaccine.2003.09.031] [Citation(s) in RCA: 121] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2003] [Revised: 09/17/2003] [Accepted: 09/24/2003] [Indexed: 11/16/2022]
Abstract
We report the use of CotC, a major component of the Bacillus subtilis spore coat, as a fusion partner for the expression of two heterologous antigens on the spore coat. Recombinant spores expressing tetanus toxin fragment C (TTFC) of Clostridium tetani or the B subunit of the heat-labile toxin of Escherichia coli (LTB) were used for oral dosing and shown to generate specific systemic and mucosal immune responses in a murine model. This report, expanding the previously described expression of TTFC on the spore surface by fusion to CotB [J Bacteriol 183 (2001) 6294] and its use for oral vaccination [Infect Immun 71 (2003) 2810] shows that different antigens can be successfully presented on the spore coat and supports the use of the spore as an efficient vehicle for mucosal immunisation.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antibodies, Bacterial/analysis
- Antibodies, Bacterial/biosynthesis
- Antigens, Bacterial/chemistry
- Antigens, Bacterial/immunology
- Bacillus subtilis/chemistry
- Bacillus subtilis/immunology
- Bacterial Toxins/immunology
- Blotting, Western
- Chromosomes, Bacterial/immunology
- Cyclohexanones/chemistry
- Cyclohexanones/immunology
- Enterotoxins/immunology
- Enzyme-Linked Immunosorbent Assay
- Escherichia coli Proteins
- Female
- Genes, Bacterial/genetics
- Immunity, Mucosal/immunology
- Immunization
- Mice
- Mice, Inbred C57BL
- Neutralization Tests
- Peptide Fragments/immunology
- Recombinant Fusion Proteins/immunology
- Spores, Bacterial/chemistry
- Spores, Bacterial/immunology
- Tetanus/immunology
- Tetanus Toxin/immunology
- Transformation, Bacterial
- Vaccines, Synthetic/chemistry
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- Emilia M F Mauriello
- Dipartimento di Fisiologia Generale ed Ambientale, Sezione di Microbiologia, Università Federico II, Napoli, Italy
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Wagner B, Hufnagl K, Radauer C, Wagner S, Baier K, Scheiner O, Wiedermann U, Breiteneder H. Expression of the B subunit of the heat-labile enterotoxin of Escherichia coli in tobacco mosaic virus-infected Nicotiana benthamiana plants and its characterization as mucosal immunogen and adjuvant. J Immunol Methods 2004; 287:203-15. [PMID: 15099768 DOI: 10.1016/j.jim.2004.02.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2003] [Revised: 01/26/2004] [Accepted: 02/02/2004] [Indexed: 11/20/2022]
Abstract
We have produced biologically active recombinant (r) LTB, the nontoxic B subunit of heat-labile toxin (LT) of Escherichia coli in tobacco mosaic virus (TMV)-infected Nicotiana benthamiana plants. We amplified the LTB encoding sequence with its leader and introduced a hexahistidyl tag and an endoplasmic reticulum retention signal. The resulting product was ligated into a TMV-based plant viral expression vector that was used for the generation of recombinant viral RNA. Eighty-nine percent of N. benthamiana plants inoculated with the recombinant viral RNA were systemically infected as determined by anti-TMV enzyme-linked immunosorbent assay (ELISA) experiments. The rLTB monomer was identified by LT-specific as well as by histidyl-tag-specific immunoblots. rLTB from plant extracts of TMV-infected N. benthamiana leaves was purified to give 75 microg rLTB pentamers per gram fresh plant material and was capable of binding G(M)1 ganglioside. The immunogenicity of the plant-produced rLTB was tested in mice and showed that intranasal application of rLTB (15 microg per mouse) induced LTB-specific IgG1 antibodies. To prove its adjuvanticity, rLTB was intranasally co-administered with the Hevea latex allergen Hev b 3, leading to allergen-specific IgG1 and IgG2a antibody production. The fact that intranasal application of rLTB and Hev b 3 prior to systemic challenge with the allergen enhanced the Th2 responses at the humoral and cellular level indicated that rLTB promoted immune responses that were naturally induced by the antigen/allergen. In conclusion, these results indicate that the plant viral expression system is suitable for the rapid large-scale production of biologically active LTB with strong mucosal adjuvant capacity.
Collapse
Affiliation(s)
- Birgit Wagner
- Department of Pathophysiology, Medical University of Vienna, General Hospital Vienna EBO 3.Q, Waehringer Guertel 18-20, A-1090 Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Gaboriau-Routhiau V, Raibaud P, Dubuquoy C, Moreau MC. Colonization of gnotobiotic mice with human gut microflora at birth protects against Escherichia coli heat-labile enterotoxin-mediated abrogation of oral tolerance. Pediatr Res 2003; 54:739-46. [PMID: 12904595 DOI: 10.1203/01.pdr.0000086902.52137.c9] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Previous work has shown that the indigenous gut microflora in mice plays a protective role against Escherichia coli heat-labile enterotoxin (LT)-mediated abrogation of oral tolerance to an unrelated co-ingested protein. To assess potential protection by human gut microflora, we studied the effect of human gut microflora in a murine model. Oral tolerance was studied in adult gnotobiotic mice (i.e. ex-germ-free mice) colonized with the entire human fecal microflora and orally administered once with LT and ovalbumin. Systemic suppression of IgG, IgG1, IgG2a, and IgE antibody responses was assessed by ELISA. Both specific IgG subclasses and IgE hyporesponsiveness was induced in LT + ovalbumin-fed gnotobiotic mice, indicating that the human gut microflora can protect against the LT-mediated abrogation of oral tolerance. However, as confirmed with mouse gut microflora, this protective effect only occurs when the gut microflora is associated from birth on. Colonization of germ-free mice with a single bacterial strain, E. coli, predominant in the human and mouse gut microflora in the neonatal period, showed that this strain alone did not induce protection. These results supported the hypothesis that the natural establishment of the gut microflora in neonates crucially influenced resistance to LT-mediated abrogation of oral tolerance by reinforcing suppression of both T helper type 1- and T helper type 2-controlled responses, and suggested that sequential bacterial colonization of the gut rather than a single bacterial species may be involved in this phenomenon.
Collapse
|
45
|
Mlcková P, Cechová D, Marusková L, Chalupná P, Novotná O, Prokesová L. Role of T cells in the adjuvant effect of Bacillus firmus on the immune system of mice: intranasal and intratracheal immunization study with ovalbumin. Folia Microbiol (Praha) 2003; 48:427-34. [PMID: 12879759 DOI: 10.1007/bf02931379] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Functions of T cells were determined after intranasal and intratracheal immunization of mice with ovalbumin (Ova) and Bacillus firmus (Bf), a Gram-positive nonpathogenic bacterium of the external environment, or delipidated Bf (dBf) as adjuvants, with the aim to elucidate the mechanism of support of Ova-specific antibody production caused by Bf that had been observed in an identical experiment. Neither Bf nor dBf in a mixture with Ova stimulated Ova-specific T-cell response tested as antigen-specific blast transformation. By contrast, a mild polyclonal stimulation was observed in splenocytes from mice given dBf. In vitro incubation of splenocytes with 100 micrograms (but not 10 micrograms) of Bf or dBf led to a highly significant inhibition of proliferation below the control level in all groups of animals. Supernatants of splenocyte cultures were further tested for cytokine production. IL-10 and IFN-gamma were released after in vitro challenge with dBf and in some cases also with Bf. Analysis of sera demonstrated that administration of Ova + adjuvant brought about an increase in anti-Ova IgG1, IgG2a and IgG2b whereas treatment with Ova alone caused a rise in IgG1 only. The role of Bf or dBf in the enhancement of antigen-specific antibody production could be in influencing macrophages and inducing cytokine milieu composed of IL-10, IFN-gamma and other factors that leads to a bystander stimulation of specifically activated Ova-B cell receptor (Ova-BCR)-bearing cells.
Collapse
Affiliation(s)
- P Mlcková
- Institute of Immunology and Microbiology, 1st Faculty of Medicine, Charles University, 128 00 Prague, Czechia
| | | | | | | | | | | |
Collapse
|
46
|
Marinaro M, Riccomi A, Rappuoli R, Pizza M, Fiorelli V, Tripiciano A, Cafaro A, Ensoli B, De Magistris MT. Mucosal delivery of the human immunodeficiency virus-1 Tat protein in mice elicits systemic neutralizing antibodies, cytotoxic T lymphocytes and mucosal IgA. Vaccine 2003; 21:3972-81. [PMID: 12922133 DOI: 10.1016/s0264-410x(03)00295-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Human immunodeficiency virus (HIV)-1 Tat protein induces protection in non-human primates upon systemic vaccination. In view of the design of mucosal vaccines against HIV-1 we studied the immune response to native Tat (aa 1-86) in mice following intranasal delivery of the protein with two mucosal adjuvants, Escherichia coli heat-labile enterotoxin (LT) and LT-R72, a non-toxic mutant of LT. Immunization with Tat and the two adjuvants induced in BALB/c but not in C57BL/6 mice high and persistent levels of serum IgG and secretory IgA in vaginal and intestinal fluids. Mice sera neutralized Tat and recognized two epitopes mapping in the regions 1-20 and 46-60. Furthermore, their splenocytes proliferated and secreted IFN-gamma and IL-6 in response to Tat. Finally, CTLs were also elicited and they recognized an epitope localized within aa 11-40 of Tat.
Collapse
Affiliation(s)
- Mariarosaria Marinaro
- Laboratory of Bacteriology and Medical Mycology, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
Control of the HIV pandemic requires an effective vaccine. The difficulties in developing a preventive vaccine are generally believed to be due to the rapid rate of mutation of HIV that escapes cytotoxic lymphocytes (CTL) and the problems in induction of neutralising antibodies to wild strains of HIV. These difficulties should re-orientate vaccine strategy into four somewhat neglected areas of immunisation. Innate immunity, with its rapid protective response to infection that is independent of memory and relies on an optimal mucosal adjuvant. Targeting the genital and rectal mucosa, with the associated lymph nodes, as an immune response has to be elicited directly on encountering HIV during sexual intercourse. Stimulating a broadly based adaptive immune response that enhances the memory CD4(+) and CD8(+) T cells and B cells, induces maturation of dendritic cells and results in Th1 polarised immunity. Taking advantage of "experiments of nature", by utilising host antigens, as manifested by protection against HIV infection in homozygous Delta32 CCR5 individuals and in allo-immunity.
Collapse
Affiliation(s)
- Thomas Lehner
- Peter Gorer Department of Immunobiology, Guy's, King's and St. Thomas Hospital Medical School, London, UK.
| |
Collapse
|
48
|
Affiliation(s)
- Thomas Lehner
- Guy's, King's & St. Thomas' Medical School, Guy's Hospital, London SE1 9RT, England.
| | | |
Collapse
|
49
|
Peppoloni S, Ruggiero P, Contorni M, Morandi M, Pizza M, Rappuoli R, Podda A, Del Giudice G. Mutants of the Escherichia coli heat-labile enterotoxin as safe and strong adjuvants for intranasal delivery of vaccines. Expert Rev Vaccines 2003; 2:285-93. [PMID: 12899578 DOI: 10.1586/14760584.2.2.285] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Cholera toxin and Escherichia coli heat-labile enterotoxin are powerful mucosal adjuvants but their high toxicity hampers their use in humans. Site-directed mutagenesis has allowed the generation of several cholera toxin and E. coli heat-labile enterotoxin mutants with abolished or strongly reduced toxicity that still retain strong mucosal adjuvanticity. Among them, LTK63 (Ser to Lys substitution at position 63 in the A subunit) is completely nontoxic and LTR72 (Ala to Arg at position 72) retains a very low residual enzymatic activity. Both of them have been shown to be safe and effective in enhancing the immunogenicity of intranasally coadministered vaccines, also resulting in protective responses in several animal models. Clinical grade preparations of these mutants have now been produced, tested in animals and proven to be totally safe. Indeed, they did not induce any inflammatory event in the respiratory tract nor, more importantly, in the olfactory bulbs and in the meninges. The fully nontoxic LTK63 mutant has now been successfully tested in human volunteers with a trivalent subunit influenza vaccine.
Collapse
Affiliation(s)
- Samuele Peppoloni
- IRIS Research Center, Department of Hygiene, Microbiology and Biostatistical Sciences, University of Modena and Reggio Emilia, Italy
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Marinaro M, Fasano A, De Magistris MT. Zonula occludens toxin acts as an adjuvant through different mucosal routes and induces protective immune responses. Infect Immun 2003; 71:1897-902. [PMID: 12654806 PMCID: PMC152047 DOI: 10.1128/iai.71.4.1897-1902.2003] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Zonula occludens toxin (Zot) is produced by Vibrio cholerae and has the ability to increase mucosal permeability by reversibly affecting the structure of tight junctions. Because of this property, Zot is a promising tool for mucosal drug and antigen (Ag) delivery. Here we show that Zot acts as a mucosal adjuvant to induce long-lasting and protective immune responses upon mucosal immunization of mice. Indeed, the intranasal delivery of ovalbumin with two different recombinant forms of Zot in BALB/c mice resulted in high Ag-specific serum immunoglobulin G titers that were maintained over the course of a year. Moreover, His-Zot induced humoral and cell-mediated responses to tetanus toxoid in C57BL/6 mice and protected the mice against a systemic challenge with tetanus toxin. In addition, we found that Zot also acts as an adjuvant through the intrarectal route and that it has very low immunogenicity compared to the adjuvant Escherichia coli heat-labile enterotoxin. Finally, by using an octapeptide representing the putative binding site of Zot and of its endogenous analogue zonulin, we provide evidence that Zot may bind a mucosal receptor on nasal mucosa and may mimic an endogenous regulator of tight junctions to deliver Ags in the submucosa. In conclusion, Zot is a novel and effective mucosal adjuvant that may be useful for the development of mucosal vaccines.
Collapse
Affiliation(s)
- Mariarosaria Marinaro
- Laboratory of Bacteriology and Medical Mycology, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | | | | |
Collapse
|