51
|
Glück R. Intranasal immunization against influenza. JOURNAL OF AEROSOL MEDICINE : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY FOR AEROSOLS IN MEDICINE 2002; 15:221-8. [PMID: 12184872 DOI: 10.1089/089426802320282347] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Nasalflu is a novel influenza subunit vaccine, which is administered by the intranasal route using a spray device. Nasalflu is based on the virosomal concept which is registered in the EU as Epaxal Berna, a vaccine against Hepatitis A, and Inflexal Berna V, a subunit influenza vaccine. The virosome is a carrier system which delivers antigens to cells and is able to induce both B- and T-cell immunity. When virosomal vaccines are given parenterally, an immune response is elicited fast and sufficiently.
Collapse
Affiliation(s)
- Reinhard Glück
- Berna Biotech Ltd., Rehhagstrasse 79, CH-3018 Bern, Switzerland.
| |
Collapse
|
52
|
Nagai T, Kiyohara H, Munakata K, Shirahata T, Sunazuka T, Harigaya Y, Yamada H. Pinellic acid from the tuber of Pinellia ternata Breitenbach as an effective oral adjuvant for nasal influenza vaccine. Int Immunopharmacol 2002; 2:1183-93. [PMID: 12349955 DOI: 10.1016/s1567-5769(02)00086-3] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
This study describes the isolation, purification, characterization, and adjuvant activity of an orally active adjuvant substance from the tuber of Pinellia ternata, as an active herbal component of the traditional Japanese herbal (Kampo) medicine, Sho-seiryu-to (SST, Chinese name: Xiao-Qing-Long-Tang), which has been reported to show oral adjuvant activity for nasally administered influenza HA vaccine [Int. J. Immunopharmacol. 16 (1994) 605]. The active compound was identified as 9S, 12S, 13S-trihydroxy-10E-octadecenoic acid using infrared spectra, proton magnetic resonance, mass spectrometry, and circular dichroism, and named pinellic acid. Oral administration of pinellic acid (1 microg) to BALB/c mice given primary and secondary intranasal inoculations of influenza HA vaccine (1 microg) enhanced antiviral IgA antibody (Ab) titers 5.2- and 2.5-fold in nasal and bronchoalveolar washes, respectively, and antiviral IgG Ab titers 3-fold in bronchoalveolar wash and serum. Intranasal administration of pinellic acid (1 microg) with influenza HA vaccine (1 microg) slightly enhanced antiviral IgG Ab titers in bronchoalveolar wash and serum but not antiviral IgA Ab titers in nasal and bronchoalveolar washes. Pinellic acid showed no hemolytic activity. The results of this study suggest that pinellic acid may provide a safe and potent oral adjuvant for nasal influenza HA vaccine.
Collapse
Affiliation(s)
- Takayuki Nagai
- Oriental Medicine Research Center, The Kitasato Institute, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
53
|
Abstract
Dental caries is one of the most common infectious diseases. Of the oral bacteria, mutans streptococci, such as Streptococcus mutans and S. sobrinus, are considered to be causative agents of dental caries in humans. There have been numerous studies of the immunology of mutans streptococci. To control dental caries, dental caries vaccines have been produced using various cell-surface antigens of these organisms. Progress in recombinant DNA technology and peptide synthesis has been applied to the development of recombinant and synthetic peptide vaccines to control dental caries. Significant protective effects against dental caries have been shown in experimental animals, such as mice, rats and monkeys, which have been subcutaneously, orally, or intranasally immunized with these antigens. Only a few studies, however, have examined the efficacy of dental caries vaccines in humans. Recently, local passive immunization using murine monoclonal antibodies, transgenic plant antibodies, egg-yolk antibodies, and bovine milk antibodies to antigens of mutans streptococci have been used to control the colonization of the organisms and the induction of dental caries in human. Such immunization procedures may be a safer approach for controlling human dental caries than active immunization.
Collapse
Affiliation(s)
- Toshihiko Koga
- Department of Preventive Dentistry, Kyushu University Faculty of Dental Science, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | | | | | | |
Collapse
|
54
|
Asahi Y, Yoshikawa T, Watanabe I, Iwasaki T, Hasegawa H, Sato Y, Shimada SI, Nanno M, Matsuoka Y, Ohwaki M, Iwakura Y, Suzuki Y, Aizawa C, Sata T, Kurata T, Tamura SI. Protection against influenza virus infection in polymeric Ig receptor knockout mice immunized intranasally with adjuvant-combined vaccines. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:2930-8. [PMID: 11884464 DOI: 10.4049/jimmunol.168.6.2930] [Citation(s) in RCA: 158] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The role of secretory IgA in conferring cross-protective immunity was examined in polymeric (p)IgR knockout (KO) mice immunized intranasally with different inactivated vaccines prepared from A/PR/8/34 (H1N1), A/Yamagata/120/86 (H1N1), A/Beijing/262/95 (H1N1), and B/Ibaraki/2/85 viruses and infected with the A/PR/8/34 virus in the upper respiratory tract (RT)-restricting volume. In wild-type mice, immunization with A/PR/8/34 or its variant (A/Yamagata/120/86 and A/Beijing/262/95) vaccines conferred complete protection or partial cross-protection against infection, while the B-type virus vaccine failed to provide protection. The protection or cross-protection was accompanied by an increase in the nasal A/PR/8/34 hemagglutinin-reactive IgA concentration, which was estimated to be >30 times the serum IgA concentration and much higher than the nasal IgG concentration. In contrast, the blockade of transepithelial transport of dimeric IgA in pIgR-KO mice reduced the degree of protection or cross-protection, in parallel with the marked increase in serum IgA concentration and the decrease in nasal IgA concentration (about 20 and 0.3 times those in wild-type mice, respectively). The degree of the reduction of protection or cross-protection was moderately reversed by the low but non-negligible level of nasal IgA, transudates from the accumulated serum IgA. These results, together with the absence of the IgA-dependent cross-protection in the lower RT and the unaltered level of nasal or serum IgG in wild-type and pIgR-KO mice, confirm that the actively secreted IgA plays an important role in cross-protection against variant virus infection in the upper RT, which cannot be substituted by serum IgG.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Administration, Intranasal
- Animals
- Antibodies, Viral/biosynthesis
- Antibodies, Viral/chemistry
- Cholera Toxin/administration & dosage
- Cholera Toxin/immunology
- Female
- Hemagglutinin Glycoproteins, Influenza Virus/administration & dosage
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Immunization, Secondary
- Immunoglobulin A/biosynthesis
- Immunoglobulin A/chemistry
- Immunoglobulin G/biosynthesis
- Immunoglobulin G/chemistry
- Influenza A virus/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/immunology
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Mice, Transgenic
- Orthomyxoviridae Infections/genetics
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/prevention & control
- Receptors, Polymeric Immunoglobulin/deficiency
- Receptors, Polymeric Immunoglobulin/genetics
- Respiratory Tract Infections/immunology
- Respiratory Tract Infections/prevention & control
- Vaccines, Combined/administration & dosage
- Vaccines, Combined/immunology
Collapse
Affiliation(s)
- Yasuko Asahi
- Department of Pathology, National Institute of Infectious Diseases, Yakult Central Institute for Microbiological Research, and Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
55
|
Zhang Y, Pacheco S, Acuna CL, Switzer KC, Wang Y, Gilmore X, Harriman GR, Mbawuike IN. Immunoglobulin A-deficient mice exhibit altered T helper 1-type immune responses but retain mucosal immunity to influenza virus. Immunology 2002; 105:286-94. [PMID: 11918690 PMCID: PMC1782659 DOI: 10.1046/j.0019-2805.2001.01368.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
We have previously demonstrated that immunoglobulin A (IgA)(-/-) knockout (KO) mice exhibit levels of susceptibility to influenza virus infection that are similar to those of their normal IgA(+/+) littermates. To understand the mechanism of this apparent mucosal immunity without IgA, immunoglobulin isotype and T helper 1 (Th1)-type [interferon-gamma (IFN-gamma)] and Th2-type [interleukin (IL)-4, IL-5)] cytokine responses to influenza vaccine were evaluated. Intranasal immunization with influenza virus subunit vaccine plus cholera toxin/cholera toxin B subunit (CT/CTB) induced significant influenza virus-specific immunoglobulin G (IgG) antibody in the serum and nasal passages of both IgA(-/-) and IgA(+/+) mice, while IgA antibodies were induced only in IgA(+/+) mice. IgA KO mice exhibited an IgG1 subclass haemagglutinin (HA)-specific response but no detectable IgG2a and IgG2b responses. In contrast, IgA(+/+) mice exhibited significant IgG1 as well as IgG2a responses. This indicates a predominant Th2-type response in IgA KO mice compared to normal mice. Following stimulation with influenza virus in vitro, splenic lymphocytes from immunized IgA(-/-) mice produced significantly lower levels of IFN-gamma than IgA(+/+) mice (P < 0.001), but elaborated similar levels of IL-4 and IL-5. This was true at both protein and mRNA levels. Immunized mice were challenged intranasally with a small inoculum of influenza virus to allow deposition of virus in the nasal mucosal passages. Compared to non-immunized mice, immunized IgA(-/-) and IgA(+/+) mice exhibited significant, but similar levels of reduction in virus titres in the nose and lung. These results demonstrate that in addition to IgA deficiency, IgA gene deletion also resulted in down-regulated Th1-type immune responses and confirm our previous data that IgA antibody is not indispensable for the prevention of influenza virus infection.
Collapse
Affiliation(s)
- Yongxin Zhang
- Influenza Research Center, Respiratory Pathogens Research Unit, Department of Molecular Virology, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
56
|
Maeyama J, Isaka M, Yasuda Y, Matano K, Kozuka S, Taniguchi T, Ohkuma K, Tochikubo K, Goto N. Cytokine responses to recombinant cholera toxin B subunit produced by Bacillus brevis as a mucosal adjuvant. Microbiol Immunol 2001; 45:111-7. [PMID: 11293476 DOI: 10.1111/j.1348-0421.2001.tb01276.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
We attempted to clarify the mechanism of the mucosal adjuvanticity of recombinant cholera toxin B subunit (rCTB), which is inherently uncontaminated with the holotoxin produced by Bacillus brevis and has a powerful mucosal adjuvant activity, on cytokine responses compared with that of cholera toxin (CT). rCTB had no ability to stimulate cyclic AMP formation in mouse peritoneal macrophages (Mphi). Cytokine production by non-immunized Mphi cultured with rCTB or CT and by the spleen cells of mice co-immunized intranasally with ovalbumin (OVA) and rCTB or CT was examined. rCTB alone did not induce interleukin (IL)-1alpha/beta or IL-6 production by Mphi, but combination of rCTB with lipopolysaccharide (LPS) enhanced both IL-1alpha/beta production. Conversely, CT plus LPS suppressed IL-1alpha/beta production more than LPS alone. Both rCTB and CT suppressed IL-12 secretion induced by interferon gamma (IFN gamma) plus LPS. IL-2, IL-4, IL-5, and IL-10 were secreted by mouse spleen cells restimulated with OVA after intranasal co-administration of OVA together with rCTB, and in response to CT, the same cytokines were secreted. The different effect of rCTB on Mphi from that of CT may mean a difference between the mechanisms of rCTB and CT during the early stage of an immune response.
Collapse
Affiliation(s)
- J Maeyama
- Department of Safety Research on Biologics, National Institute of Infectious Diseases, Musashimurayama, Tokyo, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
57
|
Nagai T, Suzuki Y, Kiyohara H, Susa E, Kato T, Nagamine T, Hagiwara Y, Tamura S, Yabe T, Aizawa C, Yamada H. Onjisaponins, from the root of Polygala tenuifolia Willdenow, as effective adjuvants for nasal influenza and diphtheria-pertussis-tetanus vaccines. Vaccine 2001; 19:4824-34. [PMID: 11535335 DOI: 10.1016/s0264-410x(01)00215-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Active substances from hot water extracts from 267 different Chinese and Japanese medicinal herbs were screened for mucosal adjuvant activity with influenza HA vaccine in mice. The extract from the root of Polygala tenuifolia was found to contain potent mucosal adjuvant activity. The active substances were purified and identified as onjisaponins A, E, F, and G. When each onjisaponin (10 microg) was intranasally (i.n.) inoculated with influenza vaccine (10 microg) in mice, serum hemagglutination-inhibiting (HI) antibody titers increased 3-14 times over control mice administered vaccine alone after 4 weeks. When each onjisaponin (10 microg) was i.n. inoculated with the vaccine (10 microg) followed by i.n. vaccination of the vaccine alone after 3 weeks, serum HI antibody titers increased 27-50 fold over those mice given i.n. vaccinations without onjisaponins. These same conditions also significantly increased nasal anti-influenza virus IgA antibody titers. Two inoculations with onjisaponin F (1 microg) and influenza HA vaccine (1 microg) at 3 weeks intervals, significantly increased serum HI antibody and nasal anti-influenza virus IgA and IgG antibody titers after only 1 week over mice given HA vaccine alone after the secondary vaccination. Intranasal vaccination with onjisaponin F inhibited proliferation of mouse adapted influenza virus A/PR/8/34 in bronchoalveolar lavages of infected mice. Separate intranasal vaccinations with onjisaponins A, E, F, and G (10 microg) each and diphtheria-pertussis-tetanus (DPT) vaccine (10 microg) of mice followed by i.n. vaccination with DPT vaccine alone after 4 weeks showed significant increases in serum IgG and nasal IgA antibody titers after 2 weeks following secondary vaccination over mice vaccinated with DPT vaccine alone. All onjisaponins showed little hemolytic activity at concentrations up to 100 microg/ml. The results of this study suggest that onjisaponins may provide safe and potent adjuvants for intranasal inoculation of influenza HA and DPT vaccines.
Collapse
Affiliation(s)
- T Nagai
- Oriental Medicine Research Center, The Kitasato Institute, 5-9-1 Shirokane, Minato-ku, 108-8642, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
58
|
Asanuma H, Hirokawa K, Uchiyama M, Suzuki Y, Aizawa C, Kurata T, Sata T, Tamura S. Immune responses and protection in different strains of aged mice immunized intranasally with an adjuvant-combined influenza vaccine. Vaccine 2001; 19:3981-9. [PMID: 11427274 DOI: 10.1016/s0264-410x(01)00129-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Immune responses and protection against influenza virus infection were compared between young (2 months) and aged (18 months) BALB/c, C3H and C57BL/6 (B6) mice after intranasal vaccination. The mice were immunized with 2.5 microg protein of A/PR/8/34 (PR8) (H1N1) virus vaccine containing a cholera toxin adjuvant. In both the young and aged BALB/c mice, high levels of PR8-specific antibody-forming cell (AFC) responses were induced in the nasal-associated lymphoid tissue (NALT) 7 days after immunization. Nasal wash IgA and serum IgG antibody (Ab) responses to the PR8 haemagglutinin (HA) 4 weeks after immunization were slightly higher in the young mice than in the aged mice. The young mice showed complete protection against challenge infection, while the aged mice showed only a partial protection. In the C3H mice, NALT-AFC, and IgA and IgG Ab responses were higher in the young mice than those in the aged mice in parallel with the more efficient protection in the young mice than in the aged mice. Both the young and aged B6 mice showed no NALT-AFC responses, scarce IgA and IgG Ab responses and no protection. In the BALB/c mice, IgG1 and IgG2a levels were significantly lower in the aged mice. On the other hand, in the C3H mice, only IgG2a level was significantly lower in the aged mice. Similar results were obtained in terms of immune responses and protection between the young and aged mice of three different strains of mice after intra-nasal immunization with 0.1 microg of PR8 vaccine containing the adjuvant, two-times at 4-week intervals. In the B6 mice, the immune response was improved by immunization with a higher dose of the adjuvant-combined vaccine. These results suggest that local Ab responses, as well as systemic Ab responses, are downregulated in aged mice, although the degree of the downregulation of immune responses differs from strain to strain.
Collapse
Affiliation(s)
- H Asanuma
- Department of Pathology, National Institute of Infectious Diseases, Toyama 1-23-1, Shinjuku-ku, Tokyo 162-8640, Japan
| | | | | | | | | | | | | | | |
Collapse
|
59
|
McCluskie MJ, Weeratna RD, Clements JD, Davis HL. Mucosal immunization of mice using CpG DNA and/or mutants of the heat-labile enterotoxin of Escherichia coli as adjuvants. Vaccine 2001; 19:3759-68. [PMID: 11395211 DOI: 10.1016/s0264-410x(01)00088-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Cholera toxin (CT) and the Escherichia coli heat-labile enterotoxin (LT) are potent mucosal adjuvants in animals associated, at least in part, with their ability to induce cAMP. While toxicity generally precludes their use in humans, a number of different subunit or genetically detoxified mutants of CT and LT have been developed. Another type of adjuvant that has been shown to be effective at mucosal surfaces comprises synthetic oligodeoxynucleotides (ODN) containing immunostimulatory CpG motifs (CpG ODN). We have previously demonstrated a synergy between CpG ODN and native toxins after intranasal (IN) administration to mice, and herein have examined whether this synergy is linked to the cAMP activity. The adjuvanticity of CpG ODN was evaluated with IN and oral delivery of tetanus toxoid or the hepatitis B surface antigen, relative to and in combination with native LT holotoxin (LTh), three active site mutants (LTS61F, LTA69G, LTE112K), a protease site mutant (LTR192G), and the B subunit of LT (LTB). At an equivalent dose, the adjuvants could generally be divided into two groups: one that included CpG ODN, LTh, LTR192G, and LTA69G which acted as strong adjuvants; and the second which comprised LTB, LTS61F, and LTE112K, which produced significantly weaker immune responses. When CpG ODN was co-administered with bacterial toxin-derivatives, in most cases, no synergy between CpG and the LT derivatives was found for strength of the humoral response. Nevertheless, for both routes and antigens, CpG ODN combined with any LT derivative induced a more Type 1-like response than LT derivative alone. These results suggest that while the synergy seen previously with native toxins may have been due in part to inherent cAMP activity, it may have also depended on the particular antigen used and the route of immunization.
Collapse
Affiliation(s)
- M J McCluskie
- Coley Pharmaceutical Canada, 725 Parkdale Avenue, Ottawa, Canada K1Y 4E9.
| | | | | | | |
Collapse
|
60
|
Haan L, Verweij WR, Holtrop M, Brands R, van Scharrenburg GJ, Palache AM, Agsteribbe E, Wilschut J. Nasal or intramuscular immunization of mice with influenza subunit antigen and the B subunit of Escherichia coli heat-labile toxin induces IgA- or IgG-mediated protective mucosal immunity. Vaccine 2001; 19:2898-907. [PMID: 11282201 DOI: 10.1016/s0264-410x(00)00556-9] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Local mucosal IgA antibodies play a central role in protection of the respiratory tract against influenza virus infection. Therefore, new-generation influenza vaccines should aim at stimulating not only systemic, but also local antibody responses. Previously, we demonstrated that the recombinant B subunit of the Escherichia coli heat-labile toxin (LTB) is a potent adjuvant towards nasally administered influenza subunit antigen. Here, we investigated the protection conferred by LTB-supplemented influenza subunit antigen given intranasally (i.n.) or intramuscularly (i.m.) to mice. Both i.n. and i.m. immunization with subunit antigen and LTB completely protected the animals against viral infection. Protection upon i.n. immunization was associated with the induction of antigen-specific serum IgG and mucosal IgA, whereas protection upon i.m. immunization correlated with strong serum and mucosal IgG, but not IgA responses. We conclude that LTB-supplemented influenza subunit antigen, given either i.n. or i.m, induces protective antibody-mediated mucosal immunity and thus represents a promising novel flu vaccine candidate.
Collapse
Affiliation(s)
- L Haan
- Department of Medical Microbiology, Molecular Virology Section, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
61
|
Hagiwar Y, Tsuji T, Iwasaki T, Kadowaki S, Asanuma H, Chen Z, Komase K, Suzuki Y, Aizawa C, Kurata T, Tamura S. Effectiveness and safety of mutant Escherichia coli heat-labile enterotoxin (LT H44A) as an adjuvant for nasal influenza vaccine. Vaccine 2001; 19:2071-9. [PMID: 11228379 DOI: 10.1016/s0264-410x(00)00414-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The effectiveness and safety of mutant Escherichia coli heat-labile enterotoxin, LT H44A (His to Arg substitution at position 44 from the N-terminus of the A1 fragment of the A subunit) as an adjuvant for nasal influenza vaccine were examined. (1) When 0.2 microg of LT H44A, together with 0.2 microg of influenza A/PR/8/34 virus (PR8, H1N1) vaccine, was administered intranasally into BALB/c mice (twice, 4 weeks apart), anti-PR8 hemagglutinin (HA) IgA and IgG antibody (Ab) responses were induced at levels that were sufficient to provide either complete protection against infection with a small volume of PR8 virus suspension or partial protection against infection with a lethal dose of the suspension. The dose of the mutant LT and vaccine used here (0.2 microg/ 20 g doses mouse) corresponded to the estimated dose per person, i.e. 0.1 mg/10 kg body weight. (2) Using these vaccination conditions, no additional total IgE Ab responses were induced. (3) The mutant was confirmed to be less toxic than the native LT when the toxicity was analyzed either using Y1 adrenal cells in vitro (1/483 EC(50)) or by an ileal loop test. (4) One hundred micrograms of the mutant, administered intranasally or intraperitoneally into guinea-pigs (Heartley strain, 0.3-0.4 kg), caused no body-weight changes 7 days after administration, although 100 microg of the native LT administered intraperitoneally caused death in all guinea-pigs due to diarrhea within 2 days. The intranasal administration of 100 microg of the mutant resulted in almost no pathological changes in the nasal mucosa 3 days after administration. These results suggest that LT H44A, which can be produced in high yields in an E. coli culture (about 5 mg/l), could be used as one of the effective and safe adjuvants for nasal influenza vaccine in humans.
Collapse
Affiliation(s)
- Y Hagiwar
- Department of Pathology, National Institute of Infectious Diseases, Toyama 1-23-1, Shinjuku-ku, 162-8640, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
62
|
Hagiwara Y, Iwasaki T, Asanuma H, Sato Y, Sata T, Aizawa C, Kurata T, Tamura S. Effects of intranasal administration of cholera toxin (or Escherichia coli heat-labile enterotoxin) B subunits supplemented with a trace amount of the holotoxin on the brain. Vaccine 2001; 19:1652-60. [PMID: 11166888 DOI: 10.1016/s0264-410x(00)00412-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Effects of intranasal administration of cholera toxin (CT) [or Escherichia coli heat-labile enterotoxin (LT)] B subunits supplemented with a trace amount of the holotoxin, CTB* or LTB*, on the brain were examined in BALB/c mice by comparing with those of the intracerebral injection. Intracerebral injection of CTB* at doses more than 10 microg/mouse caused significant body weight loss and dose-dependent death within 7 days, with localization of conjugates of horseradish peroxidase with CTB (HRP-CTB) in the ventricular system and in the perineural space of olfactory nerves of the nasal mucosa 3 h after injection. Intracerebral injection of CTB* at doses less than 3 microg/mouse (or LTB* at doses less than 22.7 microg/mouse) did not cause any significant body weight loss for 7 days, with localization of HRP-CTB in the brain but not in the nasal mucosa. On the other hand, intranasal administration of 10 microg of CTB* caused localization of HRP-CTB in the nasal mucosa but not in the brain 3 h after administration and caused body weight loss even after 30 administrations. Neither any histological changes of brain tissues nor marked changes in serum biochemical parameters were found in mice after the 30 administrations of CTB* or LTB*. These results suggest that 0.1 microg of CTB* or LTB*, which is known to be close to the minimal effective dose as an adjuvant for nasal influenza vaccine in mice and corresponds to 100 microg per person, can be used as a safe nasal adjuvant without adversely affecting the brain.
Collapse
Affiliation(s)
- Y Hagiwara
- Department of Pathology, National Institute of Infectious Diseases, Toyama 1-23-1, Shinjuku-ku, 162-8640, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
63
|
McCluskie MJ, Weeratna RD, Davis HL. Intranasal Immunization of Mice with CpG DNA Induces Strong Systemic and Mucosal Responses That Are Influenced by Other Mucosal Adjuvants and Antigen Distribution. Mol Med 2000. [DOI: 10.1007/bf03401824] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
|
64
|
Abstract
Since the initial discovery of H. pylori by Marshall and Warren 17 years ago, much progress has been made in treating this infection. However, as we enter the millennium, H. pylori infection continues to be one of the most common infections of mankind. In addition, eradication of H. pylori still requires multiple antimicrobial agents. A better understanding of the host immune response to H. pylori infection should allow investigators to develop immunotherapies to prevent the acquisition of infection and eradicate existing chronic H. pylori infection.
Collapse
Affiliation(s)
- T G Blanchard
- Department of Pediatrics, Case Western Reserve University School of Medicine, Rainbow Babies and Children's Hospital, Cleveland, Ohio, USA
| | | |
Collapse
|
65
|
Matsuo K, Yoshikawa T, Asanuma H, Iwasaki T, Hagiwara Y, Chen Z, Kadowaki SE, Tsujimoto H, Kurata T, Tamura SI. Induction of innate immunity by nasal influenza vaccine administered in combination with an adjuvant (cholera toxin). Vaccine 2000; 18:2713-22. [PMID: 10781859 DOI: 10.1016/s0264-410x(00)00055-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Inactivated influenza vaccine was administered intranasally to BALB/c mice together with an adjuvant (cholera toxin B subunit [CTB] supplemented with a trace amount of the whole toxin, CTB*) and its ability to induce innate immunity and confer protection against influenza was examined. Nasal wash virus titres 3 days after inoculation of homologous viruses were measured as an index of the ability of the vaccine to confer protection in mice immunized with either CTB*-combined vaccine or CTB* alone 1-21 days previously. The results were as follows. (1) Partial but significant reduction of the nasal-wash virus titres (prevention) was detected beginning 3 days after the vaccination, that is, 2 days earlier than the appearance of both virus-specific antibody-forming cells (AFCs) in the nasal-associated lymphoid tissue (NALT) and virus-specific IgA antibody responses in the nasal washes of mice immunized with the CTB*-combined vaccine. (2) The protection, detected on day 3 and peaking on day 5 but lost by day 21, was also conferred in mice immunized with CTB* alone. (3) The non-specific prevention was detected at doses of more than 0.3 microg of CTB*/mouse. (4) The nonspecific protection beginning 3 days after the immunization involved the enhanced expression of cytokine mRNAs (IL-15 and IL-18), considered responsible for natural killer (NK) cell activation, by the non-T cell populations in the NALT. (5) Normal NALT cells, when cultured in vitro with CTB*, secreted IL-1beta within a few hours in culture. These results demonstrate that the CTB*-combined vaccine, when given intranasally into mice, can confer nonspecific protection against influenza beginning 3 days after the vaccination and that CTB* also possessed this ability to confer protection non-specifically and temporarily by inducing the secretion of IL-1beta, one of the most important cytokines that initiates both innate and adaptive immunity, and also NK cell activity.
Collapse
Affiliation(s)
- K Matsuo
- Department of Pathology, National Institute of Infectious Diseases, Toyama 1-23-1, Shinjuku-ku, Tokyo, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
66
|
Goto N, Maeyama J, Yasuda Y, Isaka M, Matano K, Kozuka S, Taniguchi T, Miura Y, Ohkuma K, Tochikubo K. Safety evaluation of recombinant cholera toxin B subunit produced by Bacillus brevis as a mucosal adjuvant. Vaccine 2000; 18:2164-71. [PMID: 10715532 DOI: 10.1016/s0264-410x(99)00337-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
Mucosal immune responses are known to play important roles in the establishment of protective immunity to microbial infections through mucosa. We examined the toxic effects of recombinant cholera toxin B subunit (rCTB) secreted by Gram-positive bacterium Bacillus brevis as a mucosal adjuvant. Incubation of guinea-pig peritoneal macrophages with cholera toxin (CT) or aluminium hydroxide gel (Al-gel) released a significantly higher activity of lactate dehydrogenase than did commercial natural CTB (CTB) or rCTB. Intraintestinal or intramuscular administration of CT, CTB or Al-gel caused severe histopathological reactions. CT also caused infiltration of neutrophils and irregular arrangement or partial loss of the respiratory epithelium. In addition, CT and CTB elicited vascular permeability-increasing effects. rCTB elicited no toxic effects to macrophages and no vascular permeability-increasing effects. Moreover, it is noticeable that no distinct local histopathological reactions were observed in the nasal cavity, the small-intestinal loop or the muscle given rCTB. These results suggest that, from a safety standpoint, rCTB is a useful candidate as mucosal vaccine adjuvant.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/toxicity
- Administration, Intranasal
- Aluminum Hydroxide
- Animals
- Bacillus/metabolism
- Capillary Leak Syndrome/etiology
- Capillary Permeability/drug effects
- Cells, Cultured
- Cholera Toxin/administration & dosage
- Cholera Toxin/genetics
- Cholera Toxin/immunology
- Cholera Toxin/toxicity
- Female
- Guinea Pigs
- Inflammation/etiology
- Injections
- Injections, Intramuscular
- Intestinal Mucosa/immunology
- Intestinal Mucosa/pathology
- Intestine, Small/pathology
- L-Lactate Dehydrogenase/metabolism
- Macrophages, Peritoneal/immunology
- Macrophages, Peritoneal/metabolism
- Mice
- Mice, Inbred BALB C
- Muscle, Skeletal/pathology
- Nasal Cavity/pathology
- Nasal Mucosa/immunology
- Nasal Mucosa/pathology
- Peptide Fragments/administration & dosage
- Peptide Fragments/genetics
- Peptide Fragments/immunology
- Peptide Fragments/toxicity
- Rabbits
- Recombinant Fusion Proteins/administration & dosage
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- Recombinant Fusion Proteins/toxicity
- Safety
Collapse
Affiliation(s)
- N Goto
- Department of Safety Research on Biologics, National Institute of Infectious Diseases, Gakuen, Musashimurayama, Tokyo, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Matsuo K, Iwasaki T, Asanuma H, Yoshikawa T, Chen Z, Tsujimoto H, Kurata T, Tamura SS. Cytokine mRNAs in the nasal-associated lymphoid tissue during influenza virus infection and nasal vaccination. Vaccine 2000; 18:1344-50. [PMID: 10618531 DOI: 10.1016/s0264-410x(99)00401-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Intranasal immunization with a current inactivated influenza vaccine together with an adjuvant (cholera toxin B subunit supplemented with a trace amount of whole toxin, CTB*) was confirmed in BALB/c mice to mimic influenza virus (A/PR/8/34, H1N1) infection with respect to mucosal IgA antibody responses, in which IgA antibody-forming cell responses in the nasal-associated lymphoid tissue (NALT) were involved with a peak around 7 days after infection or vaccination. Next, the expression of various cytokine mRNAs in the NALT was compared in mice either infected with viruses or immunized with CTB*-combined vaccine, to examine Th cell and cytokine regulation of mucosal IgA antibody responses. In infected mice, strong IL-2, weak IL-4, strong IL-6 and strong IFN-gamma mRNA expressions were induced during early days of infection; especially, IFN-gamma mRNA was expressed by both CD4(+) and CD8(+) T cells around 7 days after infection. In mice given CTB*-combined vaccine, weak IL-2, strong IL-4, strong IL-6 and weak IFN-gamma mRNA expressions were induced during early days of vaccination; especially, IL-4 mRNA was expressed by CD4(+) T cells. Thus, IL-6 mRNAs were expressed strongly in both infected and vaccinated mice. The IFN-gamma-rich cytokine mRNA profiles in the infected mice were reflected upon serum IgG2a-rich Ab responses, while the IL-4-rich profiles in the vaccinated mice were reflected upon the IgG1-rich Ab responses. Thus, influenza virus infection and CTB*-combined nasal vaccine induced Th1 dominant and Th2 dominant cytokine profiles, respectively, while the similarity of mucosal IgA antibody responses between infection and vaccination could be explained by the appearance of IL-6 mRNAs.
Collapse
Affiliation(s)
- K Matsuo
- Department of Pathology, National Institute of Infectious Diseases, Toyama 1-23-1, Shinjuku-ku, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
68
|
Rappuoli R, Pizza M, Douce G, Dougan G. Structure and mucosal adjuvanticity of cholera and Escherichia coli heat-labile enterotoxins. IMMUNOLOGY TODAY 1999; 20:493-500. [PMID: 10529776 DOI: 10.1016/s0167-5699(99)01523-6] [Citation(s) in RCA: 223] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- R Rappuoli
- IRIS, Chiron S.p.A., Via Fiorentina 1, 53100 Siena, Italy.
| | | | | | | |
Collapse
|
69
|
Cheng E, Cárdenas-Freytag L, Clements JD. The role of cAMP in mucosal adjuvanticity of Escherichia coli heat-labile enterotoxin (LT). Vaccine 1999; 18:38-49. [PMID: 10501233 DOI: 10.1016/s0264-410x(99)00168-1] [Citation(s) in RCA: 75] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Heat-labile enterotoxin (LT) produced by enterotoxigenic Escherichia coli (ETEC) and cholera toxin (CT) produced by Vibrio cholerae have been shown to function as potent mucosal adjuvants. A number of studies have examined the effects of different mutations at either the active site or the protease site of LT and CT and the influence of those mutations on toxicity and adjuvanticity. However, different observations reported by various groups using a variety of animal models with different antigens or different routes of immunization have provided contradictory findings and evoked many questions regarding the underlying mechanisms of mucosal adjuvanticity of LT and CT. In this study, the role of cAMP in mucosal adjuvanticity was examined by comparing three LT active site mutants (S61F, A69G, E112K), a protease site mutant (R192G) and recombinant LT-B for toxicity, cAMP activity and mucosal adjuvanticity using tetanus toxoid (TT) as a model antigen. While all mutants examined showed reduced toxicity, the effects of each mutation on its ability to function as an adjuvant varied. Following intranasal immunization, native LT as well as protease and active site mutants of LT induced serum anti-TT IgG and their responses were virtually indistinguishable from one another. In addition, LT-B was also able to enhance production of serum anti-TT IgG, though at a level significantly lower than that achieved by native LT and mutants. Following oral immunization, the best serum anti-TT IgG responses were obtained with native LT and mutants that retained the ability to induce accumulation of cAMP. Despite the nearly identical serum anti-TT IgG responses following intranasal immunization, there was a strong correlation between the ability to induce accumulation of cAMP in cultured Caco-2 cells and the ability to elicit production of antigen-specific Th1 or Th2 cytokines.
Collapse
Affiliation(s)
- E Cheng
- Department of Microbiology and Immunology, Tulane University Medical Center, New Orleans, LA 70112-2699, USA
| | | | | |
Collapse
|
70
|
De Haan L, Holtrop M, Verweij WR, Agsteribbe E, Wilschut J. Mucosal immunogenicity and adjuvant activity of the recombinant A subunit of the Escherichia coli heat-labile enterotoxin. Immunology 1999; 97:706-13. [PMID: 10457227 PMCID: PMC2326879 DOI: 10.1046/j.1365-2567.1999.00817.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The Escherichia coli heat-labile enterotoxin (LT) is an exceptionally effective mucosal immunogen and mucosal immunoadjuvant towards coadministered antigens. Although, in general, the molecular basis of these properties is poorly understood, both the toxic ADP-ribosylation activity of the LTA subunit and the cellular toxin receptor, ganglioside, GM1-binding properties of the LTB-pentamer have been suggested to be involved. In recent studies we found that GM1-binding is not essential for the adjuvanticity of LT, suggesting an important role for the LTA subunit in immune stimulation. We now describe the immunomodulatory properties of recombinant LTA molecules with or without ADP-ribosylation activity, LTA(His)10 and LTA-E112K(His)10, respectively. These molecules were expressed as fusion proteins with an N-terminal His-tag to allow simple purification on nickel-chelate columns. Their immunogenic and immunoadjuvant properties were assessed upon intranasal administration to mice, and antigen-specific serum immunoglobulin-isotype and -subtype responses and mucosal secretory immunoglobulin A (IgA) responses were monitored using enzyme-linked immunosorbent assay. With respect to immunogenicity, both LTA(His)10 and LTA-E112K(His)10 failed to induce antibody responses. On the other hand, immunization with both LT and the non-toxic LT-E112K mutant not only induced brisk LTB-specific, but also LTA-specific serum and mucosal antibody responses. Therefore, we conclude that linkage of LTA to the LTB pentamer is essential for the induction of LTA-specific responses. With respect to adjuvanticity, both LTA(His)10 and LTA-E112K(His)10 were found to stimulate serum and mucosal antibody responses towards coadministered influenza subunit antigen. Remarkably, responses obtained with LTA(His)10 were comparable in both magnitude and serum immunoglobulin isotype and subtype distributions to those observed after coimmunization with LT, LT-E112K, or recombinant LTB. We conclude that LTA, by itself, can act as a potent adjuvant for intranasally administered antigens in a fashion independent of ADP-ribosylation activity and association with the LTB pentamer.
Collapse
Affiliation(s)
- L De Haan
- Department of Physiological Chemistry, Groningen Utrecht Institute for Drug Exploration (GUIDE), University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands
| | | | | | | | | |
Collapse
|
71
|
Ekström J, Hu KF, Bengtsson KL, Morein B. Iscom and iscom-matrix enhance by intranasal route the IgA responses to OVA and rCTB in local and remote mucosal secretions. Vaccine 1999; 17:2690-701. [PMID: 10418920 DOI: 10.1016/s0264-410x(99)00052-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Iscoms, with rCTB incorporated via the GM1 receptor, enhanced in mice the mucosal immunogenicity of rCTB as antigen after intranasal (i.n.) administration both by inducing IgA response in the remote intestinal tract mucosa and by a 100-fold increase of the specific IgA locally in the lungs. Iscom-matrix as a separate entity mixed with rCTB enhanced the rCTB-IgA response similarly. While OVA in iscoms induced high mucosal IgA responses, iscom-matrix co-administered with OVA induced low or no mucosal IgA response to OVA. A synergism between iscoms and rCTB could only be seen as an adjuvant targeting effect enhancing the IgA response to OVA in the remote genital tract mucosa. In serum, the immunomodulatory effect of iscoms after i.n. administration was seen as an enhanced serum IgG2a response.
Collapse
Affiliation(s)
- J Ekström
- Department of Virology, National Veterinary Institute, Biomedical Centre, Uppsala, Sweden
| | | | | | | |
Collapse
|
72
|
Tanaka Y, Nakano H, Ishikawa F, Yoshida M, Gyotoku Y, Kakiuchi T. Cholera toxin increases intracellular pH in B lymphoma cells and decreases their antigen-presenting ability. Eur J Immunol 1999; 29:1561-70. [PMID: 10359110 DOI: 10.1002/(sici)1521-4141(199905)29:05<1561::aid-immu1561>3.0.co;2-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Cholera toxin (CT) can function as a potent adjuvant in the mucosal immune response. However, we have found that treatment of A20-HL murine B lymphoma cells with CT severely inhibits the presentation of ovalbumin (OVA) to cells of the T cell clone 42-6A specific for OVA(323-339)/I-Ad, whereas it does not affect the presentation of OVA(323-339) peptide. CT treatment did not affect the expression of B7-1, B7-2, ICAM-1, LFA-1 or MHC class II on, or the internalization of OVA into A20-HL cells. In CT-treated A20-HL cells, degradation of OVA was decreased, and intracellular pH was raised to a level approximately equivalent to that in CH3NH2-treated cells. Treatment with CH3NH2 is known to raise the pH in endocytic structures and thus inhibits antigen processing. Treatment of A20-HL cells with dibutyryl-cAMP similarly increased intracellular pH. The increase in intracellular pH following CT treatment was inhibited by a cAMP inhibitor, 2',3'-dideoxyadenosine. These results strongly suggest that CT treatment of A20-HL cells inhibits their antigen-presenting cell function by triggering the cAMP cascade, increasing intracellular pH, and reducing the degradation of OVA.
Collapse
Affiliation(s)
- Y Tanaka
- Department of Immunology, Toho University School of Medicine, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
73
|
Ramachandra L, Chu RS, Askew D, Noss EH, Canaday DH, Potter NS, Johnsen A, Krieg AM, Nedrud JG, Boom WH, Harding CV. Phagocytic antigen processing and effects of microbial products on antigen processing and T-cell responses. Immunol Rev 1999; 168:217-39. [PMID: 10399077 DOI: 10.1111/j.1600-065x.1999.tb01295.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Processing of exogenous antigens and microbes involves contributions by multiple different endocytic and phagocytic compartments. During the processing of soluble antigens, different endocytic compartments have been demonstrated to use distinct antigen-processing mechanisms and to process distinct sets of antigenic epitopes. Processing of particulate and microbial antigens involves phagocytosis and functions contributed by phagocytic compartments. Recent data from our laboratory demonstrate that phagosomes containing antigen-conjugated latex beads are fully competent class II MHC (MHC-II) antigen-processing organelles, which generate peptide:MHC-II complexes. In addition, phagocytosed antigen enters an alternate class I MHC (MHC-I) processing pathway that results in loading of peptides derived from exogenous antigens onto MHC-I molecules, in contrast to the cytosolic antigen source utilized by the conventional MHC-I antigen-processing pathway. Antigen processing and other immune response mechanisms may be activated or inhibited by microbial components to the benefit of either the host or the pathogen. For example, antigen processing and T-cell responses (e.g. Th1 vs Th2 differentiation) are modulated by multiple distinct microbial components, including lipopolysaccharide, cholera toxin, heat labile enterotoxin of Escherichia coli, DNA containing CpG motifs (found in prokaryotic and invertebrate DNA but not mammalian DNA) and components of Mycobacterium tuberculosis.
Collapse
Affiliation(s)
- L Ramachandra
- Institute of Pathology, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
74
|
Blanchard TG, Czinn SJ, Nedrud JG. Host response and vaccine development to Helicobacter pylori infection. Curr Top Microbiol Immunol 1999; 241:181-213. [PMID: 10087662 DOI: 10.1007/978-3-642-60013-5_10] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Studies in both humans and animals demonstrate that H. pylori is capable of illiciting an innate response that in part is regulated by the genetic makeup of the host. These innate responses includes stimulating immune effector mechanisms at the cellular and biochemical level resulting in the influx of neutrophils into the lamina propria and have even been shown to modify gastric acid secretion. The availability of good animal models of chronic Helicobacter infection has also allowed investigators to begin to examine how the adaptive host immune response prevents and/or exacerbates Helicobacter-induced gastroduodenal disease. The experimental H. felis/mouse model has been utilized by a number of laboratories to investigate mechanisms of host defense against chronic Helicobacter infection. This model and the more recently developed H. pylori rodent model has not only allowed investigators to confirm the feasibility of immunotherapy to prevent and/or cure Helicobacter infection but also to begin to examine how the host immune response prevents and/or exacerbates Helicobacter-induced gastroduodenal disease. Based on these studies a hypothesis is emerging that suggests that protection and/or cure from Helicobacter infection is mediated primarily by an upregulated cellular immune response which may act via an antibody independent mechanism. Paradoxically, following natural infection with H. pylori, a component of the cellular immune response also promotes chronic gastric inflammation without clearance of the organism. The recent development of reliable and reproducible H. pylori/rodent models of disease and the availability of numerous inbred strains, transgenic and knockout animals, will allow investigators to continue to explore the role the host cellular and humoral immune response plays in promoting or preventing this infection.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Administration, Oral
- Animals
- Antibodies, Bacterial/immunology
- Antibody Specificity
- Bacterial Vaccines/administration & dosage
- Bacterial Vaccines/immunology
- Clinical Trials as Topic
- Cytokines/metabolism
- Disease Models, Animal
- Gastric Acid/metabolism
- Gastric Mucosa/immunology
- Gastrointestinal Diseases/immunology
- Gastrointestinal Diseases/microbiology
- Gastrointestinal Diseases/therapy
- Genetic Predisposition to Disease
- Helicobacter Infections/immunology
- Helicobacter Infections/therapy
- Helicobacter pylori/immunology
- Humans
- Immunity, Active
- Immunity, Cellular
- Immunity, Innate
- Immunity, Mucosal/immunology
- Mice
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- T G Blanchard
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | | |
Collapse
|
75
|
Yanagita M, Hiroi T, Kitagaki N, Hamada S, Ito HO, Shimauchi H, Murakami S, Okada H, Kiyono H. Nasopharyngeal-Associated Lymphoreticular Tissue (NALT) Immunity: Fimbriae-Specific Th1 and Th2 Cell-Regulated IgA Responses for the Inhibition of Bacterial Attachment to Epithelial Cells and Subsequent Inflammatory Cytokine Production. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.6.3559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
To investigate the antibacterial activity of mucosal Th1 and Th2 immune responses induced nasally and orally, mice were immunized with mucosal vaccine containing fimbrial protein of Porphyromonas gingivalis, a causative agent for a destructive chronic inflammation in the periodontium, and cholera toxin (CT) as mucosal adjuvant. Nasal vaccine containing low doses of fimbriae (10 μg) and CT (1 μg) induced Ag-specific Th1/Th2-type response in CD4+ T cells in mucosal effector tissues, including nasal passage and submandibular glands, which accounted for the generation of Ag-specific IgA-producing cells. In contrast, oral immunization required higher amounts of fimbriae and CT for the induction of Ag-specific IgA responses. Fimbriae-specific IgA mAbs generated from submandibular glands of nasally immunized mice inhibited P. gingivalis attachment to and reduced subsequent inflammatory cytokine production from epithelial cells. These findings suggest that nasal vaccination is an effective immunization regimen for the induction of Ag-specific Th1 and Th2 cell-driven IgA immune responses that possess the ability to inhibit bacterial attachment to epithelial cells and subsequent inflammatory cytokine production.
Collapse
Affiliation(s)
- Manabu Yanagita
- *Department of Mucosal Immunology, Research Institute for Microbial Diseases, and Departments of
- †Periodontology and Endodontology, and
| | - Takachika Hiroi
- *Department of Mucosal Immunology, Research Institute for Microbial Diseases, and Departments of
| | - Noriko Kitagaki
- *Department of Mucosal Immunology, Research Institute for Microbial Diseases, and Departments of
| | - Shigeyuki Hamada
- ‡Oral Microbiology, Faculty of Dentistry, Osaka University, Osaka, Japan; and
| | - Hiro-o Ito
- §Department of Preventive Dentistry, Kagoshima University Dental School, Kagoshima, Japan
| | | | | | | | - Hiroshi Kiyono
- *Department of Mucosal Immunology, Research Institute for Microbial Diseases, and Departments of
| |
Collapse
|
76
|
Mbawuike IN, Pacheco S, Acuna CL, Switzer KC, Zhang Y, Harriman GR. Mucosal Immunity to Influenza Without IgA: An IgA Knockout Mouse Model. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.5.2530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
IgA knockout mice (IgA−/−) were generated by gene targeting and were used to determine the role of IgA in protection against mucosal infection by influenza and the value of immunization for preferential induction of secretory IgA. Aerosol challenge of naive IgA−/− mice and their wild-type IgA+/+ littermates with sublethal and lethal doses of influenza virus resulted in similar levels of pulmonary virus infection and mortality. Intranasal and i.p. immunization with influenza vaccine plus cholera toxin/cholera toxin B induced significant mucosal and serum influenza hemagglutinin-specific IgA Abs in IgA+/+ (but not IgA−/−) mice as well as IgG and IgM Abs in both IgA−/− and IgA+/+ mice; both exhibited similar levels of pulmonary and nasal virus replication and mortality following a lethal influenza virus challenge. Monoclonal anti-hemagglutinin IgG1, IgG2a, IgM, and polymeric IgA Abs were equally effective in preventing influenza virus infection in IgA−/− mice. These results indicate that IgA is not required for prevention of influenza virus infection and disease. Indeed, while mucosal immunization for selective induction of IgA against influenza may constitute a useful approach for control of influenza and other respiratory viral infections, strategies that stimulate other Igs in addition may be more desirable.
Collapse
Affiliation(s)
- Innocent N. Mbawuike
- *Departments of Microbiology and Immunology, Influenza Research Center, Respiratory Pathogens Research Unit, and Departments of
| | - Susan Pacheco
- †Medicine and
- ‡Pediatrics, Baylor College of Medicine, Houston, TX 77030
| | - Catherine L. Acuna
- *Departments of Microbiology and Immunology, Influenza Research Center, Respiratory Pathogens Research Unit, and Departments of
| | - Kirsten C. Switzer
- *Departments of Microbiology and Immunology, Influenza Research Center, Respiratory Pathogens Research Unit, and Departments of
| | - Yongxin Zhang
- *Departments of Microbiology and Immunology, Influenza Research Center, Respiratory Pathogens Research Unit, and Departments of
| | | |
Collapse
|
77
|
Verweij WR, de Haan L, Holtrop M, Agsteribbe E, Brands R, van Scharrenburg GJ, Wilschut J. Mucosal immunoadjuvant activity of recombinant Escherichia coli heat-labile enterotoxin and its B subunit: induction of systemic IgG and secretory IgA responses in mice by intranasal immunization with influenza virus surface antigen. Vaccine 1998; 16:2069-76. [PMID: 9796066 DOI: 10.1016/s0264-410x(98)00076-0] [Citation(s) in RCA: 72] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
The Escherichia coli heat-labile enterotoxin (LT) is a very potent mucosal immunogen. LT also has strong adjuvant activity towards coadministered unrelated antigens and is therefore of potential interest for development of mucosal vaccines. However, despite the great demand for such mucosal vaccines, the use of LT holotoxin as an adjuvant is essentially precluded by its toxicity. LT is composed of an A subunit, carrying the toxic ADP-ribosylation activity, and a pentamer of identical B subunits, which mediates binding to ganglioside GM1, the cellular receptor for the toxin. In this paper, we demonstrate that recombinant enzymatically inactive variants of LT, including the LTB pentamer by itself, retain the immunoadjuvant activity of LT holotoxin in a murine influenza model. Mice were immunized intranasally (i.n.) with influenza virus subunit antigen, consisting mostly of the isolated surface glycoprotein hemagglutinin (HA), supplemented with either recombinant LTB (rLTB), a nontoxic LT mutant (E112K, with a Glu112-->Lys substitution in the A subunit), or LT holotoxin, and the induction of systemic IgG and local S-IgA responses was evaluated by direct enzyme-linked immunosorbent assay (ELISA). Immunization with subunit antigen alone resulted in a poor systemic IgG response and no detectable S-IgA. However, supplementation of the antigen with E112K or rLTB resulted in a substantial stimulation of the serum IgG level and in induction of a strong S-IgA response in the nasal cavity. The adjuvant activity of E112K or rLTB under these conditions was essentially the same as that of the LT holotoxin. The present results demonstrate that nontoxic variants of LT, rLTB in particular, represent promising immunoadjuvants for potential application in an i.n. influenza virus subunit vaccine. Nontoxic LT variants may also be used in i.n. vaccine formulations directed against other mucosal pathogens. In this respect, it is of interest that LT(B)-stimulated antibody responses after i.n. immunization were also observed at distant mucosal sites, including the urogenital system. This, in principle, opens the possibility to develop i.n. vaccines against sexually transmitted infectious diseases.
Collapse
Affiliation(s)
- W R Verweij
- Department of Physiological Chemistry, Groningen-Utrecht Institute for Drug Exploration (GUIDE), University of Groningen, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
78
|
Matousek MP, Nedrud JG, Cieplak W, Harding CV. Inhibition of class II major histocompatibility complex antigen processing by Escherichia coli heat-labile enterotoxin requires an enzymatically active A subunit. Infect Immun 1998; 66:3480-4. [PMID: 9632629 PMCID: PMC108376 DOI: 10.1128/iai.66.7.3480-3484.1998] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Escherichia coli heat-labile enterotoxin (LT) and cholera toxin (CT) were found to inhibit intracellular antigen processing. Processing was not inhibited by mutant LT with attenuated ADP-ribosyltransferase activity, CT B or LT B subunit, which enhanced presentation of preexisting cell surface peptide-class II major histocompatibility complex complexes. Inhibition of antigen processing correlated with A subunit ADP-ribosyltransferase activity.
Collapse
Affiliation(s)
- M P Matousek
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | | | | | | |
Collapse
|
79
|
de Haan L, Verweij WR, Feil IK, Holtrop M, Hol WG, Agsteribbe E, Wilschut J. Role of GM1 binding in the mucosal immunogenicity and adjuvant activity of the Escherichia coli heat-labile enterotoxin and its B subunit. Immunology 1998; 94:424-30. [PMID: 9767427 PMCID: PMC1364263 DOI: 10.1046/j.1365-2567.1998.00535.x] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Escherichia coli (E. coli) heat-labile toxin (LT) is a potent mucosal immunogen and immunoadjuvant towards co-administered antigens. LT is composed of one copy of the A subunit, which has ADP-ribosylation activity, and a homopentamer of B subunits, which has affinity for the toxin receptor, the ganglioside GM1. Both the ADP-ribosylation activity of LTA and GM1 binding of LTB have been proposed to be involved in immune stimulation. We investigated the roles of these activities in the immunogenicity of recombinant LT or LTB upon intranasal immunization of mice using LT/LTB mutants, lacking either ADP-ribosylation activity, GM1-binding affinity, or both. Likewise, the adjuvant properties of these LT/LTB variants towards influenza virus subunit antigen were investigated. With respect to the immunogenicity of LT and LTB, we found that GM1-binding activity is essential for effective induction of anti-LTB antibodies. On the other hand, an LT mutant lacking ADP-ribosylation activity retained the immunogenic properties of the native toxin, indicating that ADP ribosylation is not critically involved. Whereas adjuvanticity of LTB was found to be directly related to GM1-binding activity, adjuvanticity of LT was found to be independent of GM1-binding affinity. Moreover, a mutant lacking both GM1-binding and ADP-ribosylation activity, also retained adjuvanticity. These results demonstrate that neither ADP-ribosylation activity nor GM1 binding are essential for adjuvanticity of LT, and suggest an ADP-ribosylation-independent adjuvant effect of the A subunit.
Collapse
Affiliation(s)
- L de Haan
- Department of Physiological Chemistry, Groningen Utrecht Institute for Drug Exploration (GUIDE), University of Groningen, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
80
|
de Haan L, Verweij W, Agsteribbe E, Wilschut J. The role of ADP-ribosylation and G(M1)-binding activity in the mucosal immunogenicity and adjuvanticity of the Escherichia coli heat-labile enterotoxin and Vibrio cholerae cholera toxin. Immunol Cell Biol 1998; 76:270-9. [PMID: 9682971 DOI: 10.1046/j.1440-1711.1998.00745.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The mucosal route of vaccination has attracted a great deal of attention recently. Not only is mucosal application of vaccines, for example, orally or intranasally, particularly convenient, it also offers the possibility to induce locally produced and secreted S-IgA antibodies in addition to systemic IgG antibodies. These IgA antibodies are known to play a key role in protection against pathogens that invade the host through mucosal surfaces. Induction of such responses is not readily achieved by currently used vaccination strategies, which generally involve intramuscular or subcutaneous injection with inactivated pathogens or antigens thereof. For the induction of a mucosal immune response, the vaccine needs to be applied locally. However, local vaccination with non-replicating antigens is usually ineffective and may result in tolerance unless a mucosal immunoadjuvant is included. The most potent mucosal immunoadjuvants known to date are probably cholera toxin (CT) and the closely related Escherichia coli heat-labile enterotoxin (LT). Although CT and LT have become standard adjuvants for experimental mucosal vaccines, the intrinsic toxicity has thus far precluded their use as adjuvants for human vaccine formulations. In the present review, the mucosal immunogenic and adjuvant properties of LT and CT are described, with special emphasis on the functional role of the individual subunits on their immune-stimulatory properties.
Collapse
Affiliation(s)
- L de Haan
- Department of Physiological Chemistry, Groningen Utrecht Institute for Drug Exploration, University of Groningen, The Netherlands
| | | | | | | |
Collapse
|
81
|
Blanchard TG, Lycke N, Czinn SJ, Nedrud JG. Recombinant cholera toxin B subunit is not an effective mucosal adjuvant for oral immunization of mice against Helicobacter felis. Immunol Suppl 1998; 94:22-7. [PMID: 9708182 PMCID: PMC1364326 DOI: 10.1046/j.1365-2567.1998.00482.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Cholera toxin is a potent oral mucosal adjuvant for enteric immunization. Several studies suggest that commercial cholera toxin B subunit (cCTB; purified from holotoxin) may be an effective non-toxic alternative for oral immunization. The present study was performed, using an infectious disease model, to determine if the oral mucosal adjuvanticity of CTB is dependent on contaminating holotoxin. Mice were orally immunized with Helicobacter felis sonicate and either cholera holotoxin, cCTB or recombinant cholera toxin B subunit (rCTB). Serum immunoglobulin G (IgG) and intestinal immunoglobulin A (IgA) antibody responses were determined and the mice were challenged with live H. felis to determine the degree of protective immunity induced. All orally immunized mice responded with serum IgG antibody titres regardless of the adjuvant used. However, only mice immunized with either holotoxin or the cCTB responded with an intestinal mucosal IgA response. Consistent with the production of mucosal antibodies, mice immunized with either holotoxin or cCTB as adjuvants were protected from challenge while mice receiving H. felis sonicate and rCTB all became infected. cCTB induced the accumulation of cAMP in mouse thymocytes at a level equal to 0.1% of that induced by holotoxin, whereas rCTB was devoid of any activity. These results indicate that CTB possesses no intrinsic mucosal adjuvant activity when administered orally. Therefore, when used as an oral adjuvant, CTB should also include small, non-toxic doses of cholera toxin.
Collapse
Affiliation(s)
- T G Blanchard
- Institute of Pathology, Case Western Reserve University, Cleveland, OH 44106-4943, USA
| | | | | | | |
Collapse
|
82
|
MESH Headings
- AIDS Vaccines
- Adjuvants, Immunologic
- Administration, Intranasal
- Administration, Oral
- Animals
- Bacterial Vaccines
- China
- Europe
- Genome
- Glycoconjugates/immunology
- Haplorhini
- History, 18th Century
- History, 19th Century
- History, 20th Century
- History, Ancient
- Immunologic Memory
- Immunotherapy, Active
- Peptides/immunology
- Plants, Genetically Modified
- Replicon
- SAIDS Vaccines
- Species Specificity
- Vaccination/history
- Vaccination/methods
- Vaccines/history
- Vaccines, Attenuated
- Vaccines, Combined
- Vaccines, DNA
- Vaccines, Synthetic
- Viral Vaccines
Collapse
Affiliation(s)
- M A Liu
- Chiron Vaccines Research, Meryville, California 94608, USA
| |
Collapse
|
83
|
Giuliani MM, Del Giudice G, Giannelli V, Dougan G, Douce G, Rappuoli R, Pizza M. Mucosal adjuvanticity and immunogenicity of LTR72, a novel mutant of Escherichia coli heat-labile enterotoxin with partial knockout of ADP-ribosyltransferase activity. J Exp Med 1998; 187:1123-32. [PMID: 9529328 PMCID: PMC2212201 DOI: 10.1084/jem.187.7.1123] [Citation(s) in RCA: 206] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/1998] [Indexed: 02/01/2023] Open
Abstract
Heat-labile Escherichia coli enterotoxin (LT) has the innate property of being a strong mucosal immunogen and adjuvant. In the attempt to reduce toxicity and maintain the useful immunological properties, several LT mutants have been produced. Some of these are promising mucosal adjuvants. However, so far, only those that were still toxic maintained full adjuvanticity. In this paper we describe a novel LT mutant with greatly reduced toxicity that maintains most of the adjuvanticity. The new mutant (LTR72), that contains a substitution Ala --> Arg in position 72 of the A subunit, showed only 0.6% of the LT enzymatic activity, was 100,000-fold less toxic than wild-type LT in Y1 cells in vitro, and was at least 20 times less effective than wild-type LT in the rabbit ileal loop assay in vivo. At a dose of 1 microg, LTR72 exhibited a mucosal adjuvanticity, similar to that observed with wild-type LT, better than that induced by the nontoxic, enzymatically inactive LTK63 mutant, and much greater than that of the recombinant B subunit. This trend was consistent for both the amounts and kinetics of the antibody induced, and priming of antigen-specific T lymphocytes. The data suggest that the innate high adjuvanticity of LT derives from the independent contribution of the nontoxic AB complex and the enzymatic activity. LTR72 optimizes the use of both properties: the enzymatic activity for which traces are enough, and the nontoxic AB complex, the effect of which is dose dependent. In fact, in dose-response experiments in mice, 20 microg of LTR72 were a stronger mucosal adjuvant than wild-type LT. This suggests that LTR72 may be an excellent candidate to be tested in clinical trials.
Collapse
|
84
|
Wu HY, Russell MW. Induction of mucosal and systemic immune responses by intranasal immunization using recombinant cholera toxin B subunit as an adjuvant. Vaccine 1998; 16:286-92. [PMID: 9607044 DOI: 10.1016/s0264-410x(97)00168-0] [Citation(s) in RCA: 95] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Intranasal (i.n.) immunization with Streptococcus mutans surface protein AgI/II mixed with cholera toxin B subunit (CTB) containing a trace amount of cholera toxin (CT) induces strong immune responses in mucosal and systemic sites, but whether pure CTB alone has an adjuvant effect has been questioned. To determine the adjuvant effect of recombinant (r) CTB, mice were immunized with 10 micrograms of AgI/II either mixed with or conjugated to 5 micrograms of rCTB, and antibody responses in saliva, nasal wash, gut wash, vaginal wash, and serum were assayed by ELISA. The results showed that AgI/II either mixed with or conjugated to rCTB could induce both mucosal IgA and systemic IgG antibodies to higher levels than in mice similarly immunized with AgI/II alone. Some responses, especially serum IgG antibodies, were enhanced by adding 5 micrograms CT to the immunogen, whereas overall mice immunized with AgI/II mixed with CTB contaminated with CT tended to generate the strongest mucosal IgA and serum IgG responses to AgI/II. However, rCTB used as an adjuvant induced lower antibody responses against itself than CTB intentionally or inadvertently mixed with CT. These results show that rCTB can serve as an adjuvant for protein immunogens administered by the i.n. route.
Collapse
Affiliation(s)
- H Y Wu
- Department of Microbiology, University of Alabama at Birmingham 35294-2170, USA.
| | | |
Collapse
|
85
|
Tochikubo K, Isaka M, Yasuda Y, Kozuka S, Matano K, Miura Y, Taniguchi T. Recombinant cholera toxin B subunit acts as an adjuvant for the mucosal and systemic responses of mice to mucosally co-administered bovine serum albumin. Vaccine 1998; 16:150-5. [PMID: 9607023 DOI: 10.1016/s0264-410x(97)00194-1] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
We examined the mucosal adjuvant activity of recombinant cholera toxin B subunit (rCTB) produced by Bacillus brevis carrying pNU212-CTB by intranasal or oral co-administration of bovine serum albumin (BSA). Intranasal administration stimulated a high level of BSA-specific serum IgG antibody response and BSA-specific IgA antibody responses in the nasal and pulmonary lavages. Oral administration induced a moderate level of BSA-specific serum IgG antibody and a low level of BSA-specific IgA antibody in the large intestinal washes. These results show that CTB alone can act as an intranasal or oral delivery carrier; it also has strong adjuvant properties for stimulating serum IgG and mucosal IgA immune responses to unrelated, non-coupled antigens after intranasal or oral co-immunization.
Collapse
Affiliation(s)
- K Tochikubo
- Department of Microbiology, Nagoya City University, Medical School, Japan
| | | | | | | | | | | | | |
Collapse
|
86
|
Komase K, Tamura S, Matsuo K, Watanabe K, Hattori N, Odaka A, Suzuki Y, Kurata T, Aizawa C. Mutants of Escherichia coli heat-labile enterotoxin as an adjuvant for nasal influenza vaccine. Vaccine 1998; 16:248-54. [PMID: 9607038 DOI: 10.1016/s0264-410x(97)00176-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The effectiveness and safety of known mutants of Escherichia coli heat-labile enterotoxin (LT) as an adjuvant for nasal influenza vaccine were examined. Six mutants, called LT7K (Arg to Lys), LT61F (Ser to Phe), LT112K (Glu to Lys), LT118E (Gly to Glu), LT146E (Arg to Glu) and LT192G (Arg to Gly) were constructed by the replacement of one amino acid at one position of the A1 subunit to another using site-directed mutagenesis. All mutants were confirmed to be less toxic than wild-type LT when analyzed using Y-1 adrenal cells in vitro. When influenza vaccine was administered intranasally with LT7K and LT192G, BALB/c mice developed high levels of serum and local antibodies to the HA molecules. The adjuvant activity of these mutant LTs corresponded to that of wild-type LT when 1 microgram of these mutant LTs (or wild-type LT) was coadministered with the vaccine. From the point of view of safety, LT7K was considered to be the most potent mucosal adjuvant and was examined in more detail. The adjuvant activity of the mutant was lowered more rapidly with a decrease in dose than was that of wild-type LT. The low level of adjuvant of a relatively small amount of LT7K was heightened by adding LTB to the mutant LT. These results suggest that LT7K supplemented with LTB can be used as a less toxic, effective adjuvant for nasal influenza vaccine.
Collapse
Affiliation(s)
- K Komase
- Center for Basic Research, Kitasato Institute, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
87
|
van den Akker F, Pizza M, Rappuoli R, Hol WG. Crystal structure of a non-toxic mutant of heat-labile enterotoxin, which is a potent mucosal adjuvant. Protein Sci 1997; 6:2650-4. [PMID: 9416617 PMCID: PMC2143616 DOI: 10.1002/pro.5560061220] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Two closely related bacterial toxins, heat-labile enterotoxin (LT-I) and cholera toxin (CT), not only invoke a toxic activity that affects many victims worldwide but also contain a beneficial mucosal adjuvant activity that significantly enhances the potency of vaccines in general. For the purpose of vaccine design it is most interesting that the undesirable toxic activity of these toxins can be eliminated by the single-site mutation Ser63Lys in the A subunit while the mucosal adjuvant activity is still present. The crystal structure of the Ser63Lys mutant of LT-I is determined at 2.0 A resolution. Its structure appears to be essentially the same as the wild-type LT-I structure. The substitution Ser63Lys was designed, based on the wild-type LT-I crystal structure, to decrease toxicity by interfering with NAD binding and/or catalysis. In the mutant crystal structure, the newly introduced lysine side chain is indeed positioned such that it could potentially obstruct the productive binding mode of the substrate NAD while at the same time its positive charge could possibly interfere with the critical function of nearby charged groups in the active site of LT-I. The fact that the Ser63Lys mutant of LT-I does not disrupt the wild-type LT-I structure makes the non-toxic mutant potentially suitable, from a structural point of view, to be used as a vaccine to prevent enterotoxigenic E. coli infections. The structural similarity of mutant and wild-type toxin might also be the reason why the inactive Ser63Lys variant retains its adjuvant activity.
Collapse
Affiliation(s)
- F van den Akker
- Department of Biochemistry, University of Washington, Seattle 98195-7420, USA
| | | | | | | |
Collapse
|
88
|
Tamura S, Yajima A, Hatori E, Tamura S, Asanuma H, Suzuki Y, Aizawa C, Kurata T. Effects of frequent intranasal administration of adjuvant-combined influenza vaccine on the protection against virus infection. Vaccine 1997; 15:1784-90. [PMID: 9364684 DOI: 10.1016/s0264-410x(97)00113-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
In previous papers, we have shown that Escherichia coli heat-labile enterotoxin B subunit, supplemented with a trace amount of the holotoxin (LTB*) could be used as a potent adjuvant for a nasal influenza HA (haemagglutinin) vaccine in humans. The present study was designed to determine whether the effectiveness of a combined LTB*-HA vaccine could be limited by preexisting immunity to LTB and how many times the adjuvant-combined vaccine could be administered intranasally without reducing its protective efficacy in BALB/c, C3H and B10 mice. The magnitude of both nasal and serum Ab responses to HA vaccine was correlated with the degree of protection against virus infection. Higher doses of LTB*-combined vaccine were required for inducing high enough levels of anti-HA Ab responses to provide complete protection in low responder mice. Repeated pretreatments with LTB* alone (more than six times), which provided high levels of preexisting Abs to LTB, inhibited the induction of anti-HA Ab responses and reduced the protective efficacy of the adjuvant-combined vaccine. However, the LTB*-combined vaccine could be given repeatedly (about ten times) to mice without reducing the effectiveness of the adjuvant-combined vaccine. These results suggest that the LTB*-combined nasal influenza vaccine can be given to humans once every few years when an epidemic of influenza may occur.
Collapse
Affiliation(s)
- S Tamura
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
89
|
Mestecky J, Moldoveanu Z, Michalek S, Morrow C, Compans R, Schafer D, Russell M. Current options for vaccine delivery systems by mucosal routes. J Control Release 1997. [DOI: 10.1016/s0168-3659(97)00036-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
90
|
Douce G, Fontana M, Pizza M, Rappuoli R, Dougan G. Intranasal immunogenicity and adjuvanticity of site-directed mutant derivatives of cholera toxin. Infect Immun 1997; 65:2821-8. [PMID: 9199455 PMCID: PMC175397 DOI: 10.1128/iai.65.7.2821-2828.1997] [Citation(s) in RCA: 128] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Genetically modified derivatives of cholera toxin (CT), harboring a single amino acid substitution in and around the NAD binding cleft of the A subunit, were isolated following site-directed mutagenesis of the ctxA gene. Two mutants of CT, designated CTS106 (with a proline-to-serine change at position 106) and CTK63 (with a serine-to-lysine change at position 63), were found to have substantially reduced ADP-ribosyltransferase activity and toxicity; CTK63 was completely nontoxic in all assays, whereas CTS106 was 10(4) times less toxic than wild-type CT. The mucosal adjuvanticity and immunogenicity of derivatives of CT were assessed by intranasal immunization of mice, with either ovalbumin or fragment C of tetanus toxin as a bystander antigen. Mice immunized with wild-type CT produced both local (immunoglobulin A in mucosal washes) and systemic immune responses to both CT and bystander antigens. CTS106 showed good local and systemic responses to bystander proteins and to itself. Interestingly, mice immunized with the nontoxic derivative of CT, CTK63, generated weak immune responses to the bystander antigens which were similar to those achieved when CT B subunit was used as an adjuvant. In parallel experiments, an equivalent nontoxic mutant of the Escherichia coli heat-labile enterotoxin, LTK63 (with a serine-to-lysine change at position 63), was tested (9). In contrast to CTK63, LTK63 was found to be more immunogenic and a better intranasal adjuvant than recombinant heat-labile enterotoxin B subunit or CTK63. This information, together with data on immunoglobulin subclass responses, suggests that although highly homologous, CT and heat-labile enterotoxin should not be considered biologically identical in terms of their ability to act as intranasal adjuvants.
Collapse
Affiliation(s)
- G Douce
- Department of Biochemistry, Imperial College of Science, Technology and Medicine, London, United Kingdom
| | | | | | | | | |
Collapse
|
91
|
Olsen CW, McGregor MW, Dybdahl-Sissoko N, Schram BR, Nelson KM, Lunn DP, Macklin MD, Swain WF, Hinshaw VS. Immunogenicity and efficacy of baculovirus-expressed and DNA-based equine influenza virus hemagglutinin vaccines in mice. Vaccine 1997; 15:1149-56. [PMID: 9269061 DOI: 10.1016/s0264-410x(96)00309-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Two fundamentally different approaches to vaccination of BALB/c mice with the hemagglutinin (HA) of A/Equine/Kentucky/1/81 (H3N8) (Eq/KY) were evaluated, that is, administration of HA protein vs administration of HA-encoding DNA. Each vaccine was tested for its immunogenicity and ability to provide protection from homologous virus challenge. HA protein was synthesized in vitro by infection of Sf21 insect cells with a recombinant baculovirus. Intranasal administration of this vaccine induced virus-specific antibodies, as measured by enzyme-linked immunosorbent assay (ELISA), but did not induce virus neutralizing (VN) antibodies. This route of administration provided partial protection from virus challenge, but interestingly, this protection was completely abrogated, rather than enhanced, by co-administration of 10 micrograms of cholera holotoxin. As a second approach, mice were directly vaccinated in vivo by Accell gene gun delivery of plasmid DNA encoding the Eq/KY HA gene. This approach induced VN antibodies as well as virus-specific ELISA antibodies. When two doses of DNA vaccine were administered 3 weeks apart, mice were not protected from challenge, although they cleared the infection more rapidly than control mice. However, when the second DNA vaccination was delayed until 9 weeks after the first, 9 out of 10 vaccinated mice were completely protected. These results indicate that the time between initial and booster DNA vaccinations may be an important variable in determining DNA vaccination efficacy.
Collapse
Affiliation(s)
- C W Olsen
- Department of Pathobiological Science, School of Veterinary Medicine, University of Wisconsin-Madison 53706, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Snider DP, Underdown BJ, McDermott MR. Intranasal antigen targeting to MHC class II molecules primes local IgA and serum IgG antibody responses in mice. Immunol Suppl 1997; 90:323-9. [PMID: 9155636 PMCID: PMC1456616 DOI: 10.1111/j.1365-2567.1997.00323.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Covalent conjugates of hen egg lysozyme (HEL) and anti-major histocompatibility complex (MHC) class II monoclonal antibodies (mAb) were used to immunize mice intranasally. Three weeks after intranasal (IN) priming, mice responded rapidly to IN challenge with a mixture of HEL and cholera toxin (CT), by producing large titres of anti-HEL IgA and IgG1 antibody in serum, and IgA antibody in nasal secretions. No secondary response to HEL plus CT occurred in mice that received no priming or mice primed with HEL alone. The secondary serum IgG antibody response was dominated by the IgG1 subclass. HEL combined with CT adjuvant worked much better than HEL alone in producing the secondary response. Control conjugates, containing an IgG isotype-matched mAb without specificity for mouse tissues, provided poor priming. Mice expressing MHC class II molecules, to which the anti-MHC II mAb could not bind, produced a weak antibody response compared with those that expressed the appropriate. MHC class II molecule. Our results demonstrate that immunotargeting to MHC class II molecules via the IN route allows priming of the local IgA and circulating IgG antibody, and indicate that this technique is a feasible approach for delivery of subunit vaccines in the upper respiratory tract.
Collapse
Affiliation(s)
- D P Snider
- Department of Pathology, McMaster University, Hamilton, Ontario, Canada
| | | | | |
Collapse
|
93
|
Toida N, Hajishengallis G, Wu HY, Russell MW. Oral immunization with the saliva-binding region of Streptococcus mutans AgI/II genetically coupled to the cholera toxin B subunit elicits T-helper-cell responses in gut-associated lymphoid tissues. Infect Immun 1997; 65:909-15. [PMID: 9038296 PMCID: PMC175068 DOI: 10.1128/iai.65.3.909-915.1997] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Mice immunized intragastrically (i.g.) with a genetically constructed chimeric protein consisting of the saliva-binding region (SBR) of Streptococcus mutans AgI/II coupled to cholera toxin (CT) A2 and B subunits (CTA2/B) develop serum immunoglobulin G (IgG) and mucosal IgA antibody responses against AgI/II that are enhanced by the coadministration of CT as an adjuvant. To investigate the development of antigen-specific T cells in the gut-associated lymphoid tissues, mice were immunized i.g. with SBR, SBR-CTA2/B, or SBR-CTA2/B plus CT. AgI/II-specific T cells in Peyer's patches (PP), mesenteric lymph nodes (MLN), and spleen were assayed by lymphoproliferation and flow cytometry for the expression of T-cell surface markers, and cytokine mRNA expression was evaluated by reverse transcription-PCR. T-cell responses were consistent with antibody responses but were detectable after the first immunization. Proliferative responses of PP and MLN cells upon stimulation with AgI/II in vitro were low and delayed in mice given SBR alone, and these cells displayed a mixed type 1 and 2 (or Th0) pattern of cytokine expression. Immunization with SBR-CTA2/B resulted in greater AgI/II-specific proliferative responses in PP cells and an increase in the proportion of CD4+ T cells. Coadministration of CT with SBR-CTA2/B led to greater proliferative responses especially in the MLN cells, which then showed an increase in CD4+ cells. Immunization with SBR-CTA2/B (with or without CT) skewed the cytokine expression pattern in PP and MLN cells toward Th2. The results indicate that T helper cells were induced in gut-associated lymphoid tissues by i.g. immunization with SBR-CTA2/B, concomitantly with and prior to the appearance of circulating and mucosal antibodies.
Collapse
Affiliation(s)
- N Toida
- Department of Microbiology, University of Alabama at Birmingham, 35294, USA
| | | | | | | |
Collapse
|
94
|
Tamura S, Hatori E, Tsuruhara T, Aizawa C, Kurata T. Suppression of delayed-type hypersensitivity and IgE antibody responses to ovalbumin by intranasal administration of Escherichia coli heat-labile enterotoxin B subunit-conjugated ovalbumin. Vaccine 1997; 15:225-9. [PMID: 9066042 DOI: 10.1016/s0264-410x(96)00135-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Oral administration of a small amount of antigen conjugated cholera toxin B subunit is known to induce tolerance to the antigen. In the present experiments, whether nasal administration of allergen conjugated to Escherichia coli heat-labile toxin B subunit (LTB) induced tolerance was examined in BALB/c mice. A single administration of a small amount of LTB-coupled ovalbumin (OVA) suppressed the induction of delayed-type hypersensitivity and IgE antibody responses to OVA which was administered parenterally after nasal administration of LTB-coupled OVA. The antigen-specific suppression was abrogated by the addition of the holotoxin to LTB-coupled OVA. The suppression, induced by nasal administration with a small amount of allergen conjugated to a mucosa-binding molecule, may be applicable for preventing the development of allergy.
Collapse
Affiliation(s)
- S Tamura
- Department of Pathology, National Institute of Health, Tokyo, Japan
| | | | | | | | | |
Collapse
|
95
|
Partidos CD, Pizza M, Rappuoli R, Steward MW. The adjuvant effect of a non-toxic mutant of heat-labile enterotoxin of Escherichia coli for the induction of measles virus-specific CTL responses after intranasal co-immunization with a synthetic peptide. Immunol Suppl 1996; 89:483-7. [PMID: 9014810 PMCID: PMC1456583 DOI: 10.1046/j.1365-2567.1996.d01-790.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The intranasal route has been shown to be effective for immunization. However, immunization via this route may require the use of potent and safe adjuvant. The construction of non-toxic mutants of heat labile enterotoxin of Escherichia coli (LT), which is a potent mucosal adjuvant, is a major breakthrough for the development of mucosal vaccines. In this study we have assessed the ability of an LT mutant (LTK63) to act as an adjuvant following intranasal co-immunization with a peptide corresponding to a measles virus cytotoxic T lymphocyte (CTL) epitope. LTK63 was more effective at potentiating the in vivo induction of peptide-specific and measles virus-specific CTL responses than was administration of the peptide in saline. A concentration of 10 micrograms/dose of LTK63 was found to be the most effective in potentiating the in vivo priming of peptide-specific and measles virus-specific CTL responses. These findings highlight the potential of the non-toxic mutant of LT as a safe mucosal adjuvant for use in humans.
Collapse
Affiliation(s)
- C D Partidos
- Department of Clinical Sciences, London School of Hygiene and Tropical Medicine, University of London, UK
| | | | | | | |
Collapse
|
96
|
Kozuka S, Yasuda Y, Tochikubo K. Expression and secretion of the S2 subunit of pertussis toxin in Bacillus brevis. Vaccine 1996; 14:1707-11. [PMID: 9032903 DOI: 10.1016/s0264-410x(96)00082-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
We have been trying to develop a mass production system for each of the subunits (S2, S3, S4, S5) of the pertussis toxin B oligomer (PTB) by using a Bacillus brevis-pNU212 system. In consequence a moderately efficient expression-secretion system for S2 was constructed by fusing the mature S2 gene from Bordetella pertussis Tohama with the signal-peptide coding region of pNU212 and by introducing the plasmid pNU212-S2 into B. brevis HPD31 by electroporation. The clone producing S2 secreted about 70 mg of recombinant S2 (rS2) per liter of PY-erythromycin medium after 5 days incubation at 37 degrees C. The rS2 purified by an ammonium sulfate fractionation at 30-50% saturation and sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) was identical to the native S2 in respect to the molecular weight determined by SDS-PAGE and the amino terminal amino acid sequence. The nucleotide sequence of the S2 gene in B. pertussis Tohama inserted into pNU212 was identical with that of the S2 gene in other virulent B. pertussis strains except that an adenine at position 52 of the latter was replaced by a guanine in the former, causing an amino acid substitution (glycine in the former for serine in the latter) at position 18.
Collapse
Affiliation(s)
- S Kozuka
- Department of Microbiology, Nagoya City University, Medical School, Japan
| | | | | |
Collapse
|
97
|
Husband AJ, Bao S, McClure SJ, Emery DL, Ramsay AJ. Antigen delivery strategies for mucosal vaccines. Int J Parasitol 1996; 26:825-34. [PMID: 8923132 DOI: 10.1016/s0020-7519(96)80050-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Progress towards effective vaccines to control internal parasites, especially those affecting mucosal compartments, has been inhibited by the combined problems of the antigenic complexity of parasites and the lack of understanding of the host response. However, the accumulation of information regarding regulation of mucosal immunity has enabled a reappraisal of vaccination options to provide appropriate mucosal effector responses. The pivotal role of T cell influences, and in particular the contribution of cytokine signals, has been clearly established from in vitro studies, but data emerging from our laboratories provide evidence for these effects in vivo. We have demonstrated the role of T cells in determining the outcome of an intestinal response and propose a role for local Th2 cytokine production in this regard. To support this proposition, the distribution of cytokine mRNA has been determined by in situ hybridisation techniques in normal and parasitised animals. Further, we have shown that in the absence of Th2 cytokines (using gene knockout animals) mucosal responses are grossly deficient; we have also shown that this defect can be overcome by vector-directed gene therapy. These studies have indicated that new mucosal immunisation opportunities exist by combining traditional immunisation approaches with strategies to upregulate local cytokine production. However, the success of these new strategies will depend on selection of highly immunogenic subunit antigens, coupled with techniques for cytokine manipulation and delivery with appropriate adjuvant/vehicle formulations. This paper reviews delivery technologies available to chaperone labile antigenic and genetic material to appropriate sites for mucosal stimulation after systemic or oral administration.
Collapse
Affiliation(s)
- A J Husband
- Department of Veterinary Pathology, University of Sydney, NSW, Australia.
| | | | | | | | | |
Collapse
|
98
|
Takamatsu-Matsushita N, Yamaguchi N, Kawasaki M, Yamashita Y, Takehara T, Koga T. Immunogenicity of Actinobacillus actinomycetemcomitans serotype b-specific polysaccharide-protein conjugate. ORAL MICROBIOLOGY AND IMMUNOLOGY 1996; 11:220-5. [PMID: 9002873 DOI: 10.1111/j.1399-302x.1996.tb00173.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
To enhance the immunogenicity of capsular-like serotype b-specific polysaccharide antigen (SPA) of Actinobacillus actinomycetemcomitans, the purified antigen was coupled with bovine serum albumin via an adipic acid hydrazide functional group. The conjugate (SPA-bovine serum albumin) or the native SPA was administered subcutaneously or intranasally to BALB/c mice. Neither subcutaneous immunization with SPA emulsified in Freund adjuvant nor intranasal immunization with SPA and cholera toxin B subunit elicited any antibody responses to the polysaccharide antigen. High serum immunoglobulin M (IgM) and IgG responses to SPA were induced by subcutaneous immunization with SPA-bovine serum albumin in Freund adjuvant. Serum and salivary IgA responses to SPA in addition to IgM and IgG responses were induced by intranasal immunization with the conjugate and cholera toxin B subunit. To investigate the functional activity of SPA-specific antibodies, the opsonic activity of sera from BALB/c mice immunized with the conjugate was assessed by chemiluminescence assay using human polymorphonuclear leukocytes. Murine antisera efficiently opsonized A. actinomycetemcomitans serotype b in the assay, suggesting that antibodies to SPA of the organism might serve as a protective role.
Collapse
|
99
|
de Haan L, Holtrop M, Verweij WR, Agsteribbe E, Wilschut J. Mucosal immunogenicity of the Escherichia coli heat-labile enterotoxin: role of the A subunit. Vaccine 1996; 14:260-6. [PMID: 8744549 DOI: 10.1016/0264-410x(95)00235-s] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The Escherichia coli heat-labile enterotoxin (LT) is a potent mucosal immunogen, inducing high secretory as well as systemic antibody responses upon oral or intranasal (i.n.) administration. In addition, LT has the capacity to act as an adjuvant in antibody responses against coadministered other antigens. To investigate the role of the individual subunits of LT in the mucosal immunogenicity and adjuvanticity of LT, the LT holotoxin and the non-toxic B subunit (LTB) were cloned separately and purified from overproducing E. coli cultures. Mice were immunized i.n. with the recombinant LT, LTB and combinations of the two and the induction of LTB-specific serum IgG and IgA as well as mucosal S-IgA was monitored. Intranasal administration of 2 micrograms LTB by itself induced a moderate systemic and a low mucosal antibody response, the latter being restricted to the site of immunization. However, addition of a trace amount (50 ng) of LT holotoxin to LTB strongly stimulated both serum antibody and mucosal S-IgA responses to LTB: the antibody levels induced by 2 micrograms LTB supplemented with 50 ng LT were similar to those seen after immunization with 2.9 micrograms of the LT holotoxin alone (representing an amount of 2 micrograms LTB). Furthermore, immunization with LT-supplemented LTB or with LT holotoxin alone, but not immunization with LTB alone, induced an S-IgA response in distant mucosal tissues including the lung, intestine and the urogenital system. Nicking of the LTA chain with trypsin did not enhance the immunogenicity of LT. These results indicate that, although the LTA chain plays an important role in the mucosal immunogenicity of LT including priming of the common mucosal immune system, extremely low amounts of the LT holotoxin suffice for the induction of high antibody responses to LTB, the trace LT and LTB acting in a synergistic fashion.
Collapse
Affiliation(s)
- L de Haan
- Department of Physiological Chemistry Groningen Institute for Drug Studies (GIDS), University of Groningen, The Netherlands
| | | | | | | | | |
Collapse
|
100
|
Partidos CD, Vohra P, Steward MW. Induction of measles virus-specific cytotoxic T-cell responses after intranasal immunization with synthetic peptides. Immunol Suppl 1996; 87:179-85. [PMID: 8698377 PMCID: PMC1384271 DOI: 10.1046/j.1365-2567.1996.462527.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
We have investigated the structural requirements for the induction of cytotoxic T-cell responses (CTL) in vivo after intranasal immunization with an immunodominant CTL epitope from the nucleoprotein of measles virus (MV). For the induction of CTL responses, covalent linkage of the CTL epitope to a helper T-cell epitope was required and the orientation of the epitopes influenced the immunogenicity of the CTL epitope. The presence of two copies as compared with one copy of a T-helper epitope, rendered the CTL epitope more immunogenic and resulted in the in vivo induction of MV-specific CTLs without the need for an adjuvant. The role of CTL responses to this epitope in protection after intranasal administration was evaluated in a mouse model against challenge with a neuroadapted strain of MV. Although a decreased mortality in the peptide immunized compared with that in unimmunized mice was observed, the protection achieved was not significant. These findings highlight the importance of the rational design of synthetic immunogens for the induction of CTL responses and the potential of the intranasal route for immunization.
Collapse
Affiliation(s)
- C D Partidos
- Department of Clinical Sciences, London School of Hygiene and Tropical Medicine, UK
| | | | | |
Collapse
|