101
|
Markova R, Marinova S, Petrunov B, Cvetanov J, Nenkov P, Radinov A, Tchorbadjiiska L, Konstantinova D. Stimulating effect of an oral polybacterial immunomodulator on the proliferative activity of guinea pig lymphocytes. INTERNATIONAL JOURNAL OF IMMUNOPHARMACOLOGY 1997; 19:205-14. [PMID: 9373771 DOI: 10.1016/s0192-0561(97)00023-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
A preparation for the prophylaxis and treatment of inflammations of oral mucosa and parodont Dentavax (D) was investigated in guinea pigs. Animals were given orally D for 5 consecutive days and a month later the procedure was repeated. On day 3, 10, 21, and 28 after immunization and reimmunization lymphoproliferative responses to PHA, rIL-2, LPS and D were measured by the radiometric blast transformation assay in peripheral blood, spleen, mesenteric lymph nodes (MLN) and Peyer's patches (PP). The percentage of cells entering S and G2/M-phases of cell cycle was assessed by the flow cytometric DNA analysis. A correlation in proliferative activity of cells after in vitro stimulation with PHA and LPS has been established by both methods. Peak values of lymphocyte stimulation were found on day 10, especially after the second administration of D in all organs tested, mainly in MLNs and spleen. Electron-microscopic studies demonstrated an extensive development of the endoplasmatic reticulum in plasmatic cells from spleen, PPs, mesenteric, bronchial and inguinal lymph nodes. The results obtained may be considered a proof of the immunostimulating effect of Dentavax.
Collapse
Affiliation(s)
- R Markova
- National Centre of Infectious and Parasitic Diseases, Sofia, Bulgaria
| | | | | | | | | | | | | | | |
Collapse
|
102
|
Caley IJ, Betts MR, Irlbeck DM, Davis NL, Swanstrom R, Frelinger JA, Johnston RE. Humoral, mucosal, and cellular immunity in response to a human immunodeficiency virus type 1 immunogen expressed by a Venezuelan equine encephalitis virus vaccine vector. J Virol 1997; 71:3031-8. [PMID: 9060663 PMCID: PMC191432 DOI: 10.1128/jvi.71.4.3031-3038.1997] [Citation(s) in RCA: 104] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
A molecularly cloned attenuated strain of Venezuelan equine encephalitis virus (VEE) has been genetically configured as a replication-competent vaccine vector for the expression of heterologous viral proteins (N. L. Davis, K. W. Brown, and R. E. Johnston, J. Virol. 70:3781-3787, 1996). The matrix/capsid (MA/CA) coding domain of human immunodeficiency virus type 1 (HIV-1) was cloned into the VEE vector to determine the ability of a VEE vector to stimulate an anti-HIV immune response in mice. The VEE-MA/CA vector replicated rapidly in the cytoplasm of baby hamster kidney (BHK) cells and expressed large quantities of antigenically identifiable MA/CA protein. When injected subcutaneously into BALB/c mice, the vector invaded and replicated in the draining lymphoid tissues, expressing HIV-1 MA/CA at a site of potent immune activity. Anti-MA/CA immunoglobulin G (IgG) and IgA antibodies were present in serum of all immunized mice, and titers increased after a second booster inoculation. IgA antibodies specific for MA/CA were detected in vaginal washes of mice that received two subcutaneous immunizations. Cytotoxic T-lymphocyte responses specific for MA/CA were detected following immunization with the MA/CA-expressing VEE vector. These findings demonstrate the ability of a VEE-based vaccine vector system to stimulate a comprehensive humoral and cellular immune response. The multifaceted nature of this response makes VEE an attractive vaccine for immunization against virus infections such as HIV-1, for which the correlates of protective immunity remain unclear, but may include multiple components of the immune system.
Collapse
MESH Headings
- AIDS Vaccines/immunology
- Animals
- Antibody Formation
- Capsid/genetics
- Capsid/immunology
- Cell Line
- Cricetinae
- Cytotoxicity Tests, Immunologic
- Encephalitis Virus, Venezuelan Equine/genetics
- Encephalitis Virus, Venezuelan Equine/immunology
- Female
- Gene Products, gag/genetics
- Gene Products, gag/immunology
- Genetic Vectors/immunology
- HIV Antibodies/blood
- HIV-1/immunology
- Humans
- Immunity, Cellular
- Immunity, Mucosal
- Lymphoid Tissue/immunology
- Mice
- Mice, Inbred BALB C
- T-Lymphocytes, Cytotoxic/immunology
- Vaccination
- Vaccines, Synthetic/immunology
- Vagina/immunology
- Vagina/pathology
- Viral Matrix Proteins/genetics
- Viral Matrix Proteins/immunology
Collapse
Affiliation(s)
- I J Caley
- Department of Microbiology, School of Medicine, University of North Carolina, Chapel Hill 27599, USA
| | | | | | | | | | | | | |
Collapse
|
103
|
Melhus A, Hermansson A, Forsgren A, Prellner K. A resolved pneumococcal infection protects against nontypeable Haemophilus influenzae: an evaluation of different routes of whole cell immunization in protection against experimental acute otitis media. Int J Pediatr Otorhinolaryngol 1997; 39:119-31. [PMID: 9104620 DOI: 10.1016/s0165-5876(96)01474-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
A conferred cross-protection between Haemophilus influenzae type b (Hib) and nontypeable H. influenzae (NTHi) was demonstrated in a previous study of experimental recurrent otitis media. To explore cross-protection further, and to compare oral administration of whole cells with two more conventional routes for vaccination against acute otitis media (AOM), a total number of 79 rats were immunized perorally, subcutaneously and intrabullarly with H. influenzae or pneumococci and thereafter challenged in the middle ear with NTHi or Hib 4 or 9 weeks later. Otomicroscopic changes, bacterial cultures, and serum IgG antibody levels were monitored. The study demonstrated that while peroral administration did not elicit any protection, a resolved pneumococcal AOM could reduce the susceptibility to reinfection with NTHi. In the latter case no cross-reacting antibodies were detected, but the protective rate was 50% or more, and it was comparable with that found after subcutaneous or intrabullar immunization with homologous NTHi or Hib strains. The results suggest that the protection of the rat middle ear mucosa may involve unspecific responses.
Collapse
Affiliation(s)
- A Melhus
- Department of Medical Microbiology, Lund University, Malmö General Hospital, Sweden
| | | | | | | |
Collapse
|
104
|
Marinaro M, Boyaka PN, Finkelman FD, Kiyono H, Jackson RJ, Jirillo E, McGhee JR. Oral but not parenteral interleukin (IL)-12 redirects T helper 2 (Th2)-type responses to an oral vaccine without altering mucosal IgA responses. J Exp Med 1997; 185:415-27. [PMID: 9053442 PMCID: PMC2196042 DOI: 10.1084/jem.185.3.415] [Citation(s) in RCA: 108] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/1996] [Revised: 10/16/1996] [Indexed: 02/03/2023] Open
Abstract
Our past studies have shown that the mucosal adjuvant cholera toxin (CT) induces T helper type 2 (Th2) responses with systemic IgG1, IgE and mucosal secretory IgA (S-IgA) antibodies (Abs). In this study, recombinant murine IL-12 (rmIL-12) was given either parenterally or orally to mice orally immunized with tetanus toxoid (TT) and CT to determine whether this cytokine could redirect the CT-induced Th2-type responses and what effect this shift would have on S-IgA Ab responses. Intraperitoneal administration of rmIL-12 shifted TT-specific responses toward Th1-type and resulted in CD4+ T cells producing IFN-gamma and IL-2 with markedly reduced levels of Th2-type cytokines. This cytokine profile was accompanied by increased delayed-type hypersensitivity (DTH) and shifts in serum IgG1 to IgG2a and IgG3 anti-TT Ab responses. Further, serum IgE and S-IgA Ab responses were markedly reduced by parenteral IL-12. When IL-12 complexed to liposomes was given orally both shifts to IgG2a and IgG3 and low IgE Abs again occurred concomitant with enhanced serum IFN-gamma and DTH responses. Interestingly, oral rmIL-12 did not result in significant levels of serum IL-12 nor altered S-IgA Ab responses and resulted in higher levels of some Th2-type cytokines both in vitro and in vivo when compared with parenteral IL-12. Our results show that the shifts in systemic immune responses with intact S-IgA Abs which occur after oral delivery of IL-12-liposomes are due to cytokine effects in the Peyer's patches and suggest new strategies for the targeted manipulation of Th1- and Th2-type responses to mucosal vaccines.
Collapse
Affiliation(s)
- M Marinaro
- Department of Microbiology, University of Alabama at Birmingham 35294-2170, USA
| | | | | | | | | | | | | |
Collapse
|
105
|
Stankiewicz M, Cabaj W, Pernthaner A, Hadas E. Immunisation of sheep by drug-abbreviated infections of Ostertagia circumcincta and Trichostrongylus colubriformis against field challenge of gastro-intestinal nematodes. Vet Parasitol 1996; 67:121-32. [PMID: 9011021 DOI: 10.1016/s0304-4017(96)01028-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
A very high level of protection was achieved against homologous (up to 97%) and heterologous (up to 87%) infections in 12-month-old Romney sheep immunised with oxfendazole-abbreviated infections of Ostertagia circumcincta and Trichostrongylus colubriformis. No significant protection occurred following ivermectin-abbreviated infections. None of the immunised sheep showed an increase in antibody level against excretory-secretory antigen of T. colubriformis infective larvae. The immunisation procedures did not cause a decrease in wool production, or liveweight gains compared with non-immunised controls.
Collapse
Affiliation(s)
- M Stankiewicz
- AgResearch Wallaceville Animal Research Centre, Upper Hutt, New Zealand.
| | | | | | | |
Collapse
|
106
|
Lowell GH, Colleton C, Frost D, Kaminski RW, Hughes M, Hatch J, Hooper C, Estep J, Pitt L, Topper M, Hunt RE, Baker W, Baze WB. Immunogenicity and efficacy against lethal aerosol staphylococcal enterotoxin B challenge in monkeys by intramuscular and respiratory delivery of proteosome-toxoid vaccines. Infect Immun 1996; 64:4686-93. [PMID: 8890226 PMCID: PMC174432 DOI: 10.1128/iai.64.11.4686-4693.1996] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Staphylococcal enterotoxin B (SEB), a primary cause of food poisoning, is also a superantigen that can cause toxic shock after traumatic or surgical staphylococcal wound [correction of would] infections or viral influenza-associated staphylococcal superinfections or when aerosolized for use as a potential biologic warfare threat agent. Intranasal or intramuscular (i.m.) immunization with formalinized SEB toxoid formulated with meningococcal outer membrane protein proteosomes has previously been shown to be immunogenic and protective against lethal respiratory or parenteral SEB challenge in murine models of SEB intoxication. Here, it is demonstrated that immunization of nonhuman primates with the proteosome-SEB toxoid vaccine is safe, immunogenic, and protective against lethal aerosol challenge with 15 50% lethal doses of SEB. Monkeys (10 per group) were primed i.m. and given booster injections by either the i.m. or intratracheal route without adverse side effects. Anamnestic anti-SEB serum immunoglobulin G (IgG) responses were elicited in all monkeys, but strong IgA responses in sera and bronchial secretions were elicited both pre- and post-SEB challenge only in monkeys given booster injections intratracheally. The proteosome-SEB toxoid vaccine was efficacious by both routes in protecting 100% of monkeys against severe symptomatology and death from aerosolized-SEB intoxication. These data confirm the safety, immunogenicity, and efficacy in monkeys of parenteral and respiratory vaccination with the proteosome-SEB toxoid, thereby supporting clinical trials of this vaccine in humans. The safety and enhancement of both bronchial and systemic IgA and IgG responses by the proteosome vaccine delivered by a respiratory route are also encouraging for the development of mucosally delivered proteosome vaccines to protect against SEB and other toxic or infectious respiratory pathogens.
Collapse
Affiliation(s)
- G H Lowell
- Division of Pathology, Walter Reed Army Institute of Research, Washington, D.C. 20307, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
107
|
Abstract
Conventionally the efficiency of an adjuvant is measured by the capacity to induce enhanced antibody serum titres and cell mediated immunity (CMI) to a given antigen. Nowadays the capacity of an adjuvant is also measured by the quality as well as the magnitude of the induced immune response, guided by the protective immune response required. Quality includes isotype and IgG subclass responses, T-helper cell responses characterized by the cytokine profile and cytotoxic T cells (CTL). In the early phase of immunization some adjuvants influence the antigen administration and uptake by a so-called depot effect exemplified by aluminium hydroxide gel and oil adjuvants, which possibly is not as desired as alledged. A modern depot is exerted by slow release formulations continuously releasing the antigen over a period of time or by pulses at intervals aiming at 'single injection' vaccine. Great efforts are made to formulate efficient delivery formulations targeting the antigens from the site of administration, to draining lymph nodes or distant lymphatic tissue or to mucosal surfaces by parenteral or mucosal administrations. Nowadays, non-replicating carriers besides replicating vaccines are formulated to induce mucosal immune responses encompassing secretory IgA and CMI. Efforts to evoke immune responses on mucosal membranes distant from the site of administration have resulted mostly in little success. For a long time it was considered that CTL under the restriction of MHC Class I only could be evoked by replicating viruses or intracellular parasites. However, novel adjuvant delivery systems readily induce CTL by delivering the antigen to the APC resulting in intracellular transport to the cytosol for the MHC Class I presentation system, as well as to the endosomal pathway for the MHC Class II presentation.
Collapse
Affiliation(s)
- B Morein
- Swedish University of Agricultural Sciences, Uppsala, Sweden
| | | | | | | |
Collapse
|
108
|
Li X, Fox JG, Erdman SE, Lipman NS. Intestinal plasmacytosis in rabbits: a histologic and ultrastructural study. Vet Pathol 1996; 33:721-4. [PMID: 8952036 DOI: 10.1177/030098589603300616] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
A retrospective study revealed intestinal plasmacytosis in 53 of 102 rabbits used in various experimental studies and as controls. The breeds affected included New Zealand white (n = 46), Dutch-belted (n = 6), and Watanabe (n = 1) rabbits. Sex predisposition was not found in any breed. The mean (+/-SD) ages were 3.1 +/- 1.4 years for New Zealand white rabbits, 1.3 +/- 1.1 for Dutch-belted rabbits, and 2 years for the Watanabe rabbit. The severity increased with animal age. The incidence was higher (P < 0.05) in rabbits used in antibody production and cholesterol studies. The lesions were characterized by multifocal to diffuse infiltration of well-differentiated plasma cells in the intestinal mucosa. Electron microscopic examination revealed typical plasma cell morphology of the infiltrating cells. Small intestine and cecum were the major sites affected. In severe cases, colon, rectum, trachea, esophagus, mesenteric lymph node, and spleen were also involved.
Collapse
Affiliation(s)
- X Li
- Comparative Medicine & ARC, University of Chicago, IL 60637, USA
| | | | | | | |
Collapse
|
109
|
Marinaro M, Boyaka PN, Jackson RJ, Finkelman FD, Kiyono H, McGhee JR. Interleukin-12 alters helper T-cell subsets and antibody profiles induced by the mucosal adjuvant cholera toxin. Ann N Y Acad Sci 1996; 795:361-5. [PMID: 8958955 DOI: 10.1111/j.1749-6632.1996.tb52693.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
We have shown that systemic administration of rmIL-12 could trigger Th1-type responses to a protein antigen delivered orally with CT as mucosal adjuvant. The most striking finding was that IL-12 could retain its regulatory effects when orally administered and could redirect the immune response to the oral vaccine toward a Th1-type. However, regulation by orally administered IL-12 differed from parenteral treatment with IL-12 since only the latter treatment affected mucosal S-IgA responses. These findings have important implications for the development of mucosal vaccines that induce the desired immune response.
Collapse
Affiliation(s)
- M Marinaro
- Department of Microbiology, University of Alabama at Birmingham Medical Center 35294-2170, USA
| | | | | | | | | | | |
Collapse
|
110
|
Gorczynski RM, Cohen Z, Levy G, Fu XM. A role for gamma(delta)TCR+ cells in regulation of rejection of small intestinal allografts in rats. Transplantation 1996; 62:844-51. [PMID: 8824487 DOI: 10.1097/00007890-199609270-00024] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Lewis (LEW) rats received (Lewis x Brown Norway)F1 (LBNF1) small intestinal allografts (SIT) with graft venous drainage to either the portal vein (pv) or inferior vena cava (iv), along with immunization (pv or iv) with irradiated LBNF1 spleen cells. As reported earlier, in rats with pv drained grafts there was an increase in gammadeltaTCR+ cells infiltrating the Peyer's patches (PP) and mesenteric lymph node (MLN) compared with iv drained grafts. After restimulation in culture with irradiated LBNF1 spleen stimulator cells these PP and MLN cells from SIT rats with pv graft drainage were a prominent source of TGFbeta, IL-4, and IL-10. When subpopulations of cells from PP preparations were analyzed, an enriched (<2%betaTCR+) gammadeltaTCR+ population from SIT rats with pv graft drainage, but not iv drainage, was detected that suppressed in vitro type-1 cytokine production (IL-2, IFNgamma) from alphabetaTCR+ (<2%gammadeltaTCR+) cells derived from the MLN or peripheral lymph nodes (PLN) of these same animals. On adoptive transfer to naive LEW rats simultaneously receiving LBNF1 SIT, gammadeltaTCR+ enriched PP cells from these primary donors (pv immunized, SIT rats with pv graft drainage) produced prolonged graft/ animal survival compared with PP cells obtained from primary donors that had iv drained grafts. In addition, simultaneous infusion of anti-gammadeltaTCR monoclonal antibody into SIT rats with pv graft drainage blocked the graft enhancement normally seen in these animals. These data are consistent with an important role for type-2 cytokine producing gammadeltaTCR+ cells in the regulation of graft rejection in this model.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- Graft Rejection/immunology
- Graft Rejection/pathology
- Graft Survival
- Intestine, Small/blood supply
- Intestine, Small/immunology
- Intestine, Small/transplantation
- Lymph Nodes/immunology
- Lymph Nodes/pathology
- Lymphokines/metabolism
- Peyer's Patches/immunology
- Peyer's Patches/pathology
- Portal Vein
- Rats
- Rats, Inbred BN
- Rats, Inbred Lew
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- Th2 Cells/immunology
- Th2 Cells/metabolism
- Transplantation, Homologous
- Vena Cava, Inferior
Collapse
Affiliation(s)
- R M Gorczynski
- Multi-Organ Transplant Program, The Toronto Hospital, Ontario, Canada
| | | | | | | |
Collapse
|
111
|
Okahashi N, Yamamoto M, Vancott JL, Chatfield SN, Roberts M, Bluethmann H, Hiroi T, Kiyono H, McGhee JR. Oral immunization of interleukin-4 (IL-4) knockout mice with a recombinant Salmonella strain or cholera toxin reveals that CD4+ Th2 cells producing IL-6 and IL-10 are associated with mucosal immunoglobulin A responses. Infect Immun 1996; 64:1516-25. [PMID: 8613355 PMCID: PMC173956 DOI: 10.1128/iai.64.5.1516-1525.1996] [Citation(s) in RCA: 149] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Mucosal immunoglobulin A (IgA) responses are often associated with Th2-type cells and derived cytokines, and interleukin-4 (IL-4) knockout (IL-4-/-) mice with impaired Th2 cells respond poorly to oral antigens. However, we have noted that IL-4-/- mice have normal mucosal IgA levels, which led us to query whether different oral delivery systems could elicit mucosal immunity. Two oral regimens were used: (i) a live recombinant Salmonella strain which expresses fragment C (ToxC) of tetanus toxin, and (ii) soluble tetanus toxoid (TT) with cholera toxin (CT) as an adjuvant. Oral immunization of IL-4-/- mice with recombinant Salmonella vaccine expressing ToxC induced brisk mucosal IgA and serum IgG (mainly IgG2a) anti-TT antibody responses. TT-specific CD4+ T cells from spleen or Peyer's patches produced gamma interferon, indicative of Th1 responses; however, IL-6 and IL-10 were also seen. Oral immunization of IL-4-/- mice with TT and CT induced weak mucosal IgA to TT; however, brisk IgA anti-CT-B responses and CT-B-specific CD4+ T cells producing IL-6 and IL-10 were also noted. These results show that although IL-4-dependent antibody responses are impaired, mucosal IgA responses are induced in IL-4-/- mice. These result suggest that certain cytokines, i.e., IL-6 and IL-10 from Th2-type cells, play an important compensatory role in the induction and regulation of mucosal IgA responses.
Collapse
Affiliation(s)
- N Okahashi
- Mucosal Immunization Research Group, University of Alabama, Birmingham Medical Center 35294, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
112
|
Zhang T, Stanley SL. Oral immunization with an attenuated vaccine strain of Salmonella typhimurium expressing the serine-rich Entamoeba histolytica protein induces an antiamebic immune response and protects gerbils from amebic liver abscess. Infect Immun 1996; 64:1526-31. [PMID: 8613356 PMCID: PMC173957 DOI: 10.1128/iai.64.5.1526-1531.1996] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Attenuated salmonellae represent attractive candidates for the delivery of foreign antigens by oral vaccination. In this report, we describe the high-level expression of a recombinant fusion protein containing the serine-rich Entamoeba histolytica protein (SREHP), a protective antigen derived from virulent amebae, and a bacterially derived maltose-binding protein (MBP) in an attenuated strain of Salmonella typhimurium. Mice and gerbils immunized with S. typhimurium expressing SREHP-MBP produced mucosal immunoglobulin A antiamebic antibodies and serum immunoglobulin G antiamebic antibodies. Gerbils vaccinated with S typhimurium SREHP-MBP were protected against amebic liver abscess, the most common extraintestinal complication of amebiasis. Our findings indicate that the induction of mucosal and immune responses to the amebic SREHP antigen is dependent on the level of SREHP-MBP expression in S. typhimurium and establish that oral vaccination with SREHP can produce protective immunity to invasive amebiasis.
Collapse
MESH Headings
- Administration, Oral
- Animals
- Antibodies, Protozoan/biosynthesis
- Antibodies, Protozoan/blood
- Antigens, Protozoan/administration & dosage
- Antigens, Protozoan/genetics
- Bacterial Proteins/genetics
- Bacterial Proteins/immunology
- Carrier Proteins/genetics
- Carrier Proteins/immunology
- Entamoeba histolytica/genetics
- Entamoeba histolytica/immunology
- Female
- Gerbillinae
- Immunoglobulin A/biosynthesis
- Immunoglobulin G/blood
- Liver Abscess, Amebic/immunology
- Liver Abscess, Amebic/prevention & control
- Maltose-Binding Proteins
- Membrane Proteins/genetics
- Membrane Proteins/immunology
- Mice
- Mice, Inbred BALB C
- Mucous Membrane/immunology
- Protozoan Proteins/genetics
- Protozoan Proteins/immunology
- Protozoan Vaccines/administration & dosage
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- Salmonella typhimurium/genetics
- Salmonella typhimurium/immunology
- Vaccines, Attenuated/administration & dosage
- Vaccines, Synthetic/administration & dosage
Collapse
Affiliation(s)
- T Zhang
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | |
Collapse
|
113
|
Autenrieth IB, Kempf V, Sprinz T, Preger S, Schnell A. Defense mechanisms in Peyer's patches and mesenteric lymph nodes against Yersinia enterocolitica involve integrins and cytokines. Infect Immun 1996; 64:1357-68. [PMID: 8606101 PMCID: PMC173926 DOI: 10.1128/iai.64.4.1357-1368.1996] [Citation(s) in RCA: 77] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Adhesion molecules and cytokines are involved in regulation of cellular host responses in infection processes. In this study the roles of the integrins Mac-1 and VLA-4, as well as those of the cytokines tumor necrosis factor alpha (TNF-alpha) and gamma interferon (IFN-gamma), in defense mechanisms against Yersinia enterocolitica in Peyer's patches (PP) and mesenteric lymph nodes (MLN) were investigated by blocking these molecules with antibodies in vivo prior to orogastric Yersinia infection. Intestinal Yersinia infection caused abscesses composed of polymorphonuclear (Mac-1+ VLA-4+ Pgp-1+ ICAM-1-) and mononuclear (Mac-1+ VLA-4+ Pgp-1+ ICAM-inhibited phagocytosis of yersiniae by macrophages, (ii) reduced Yersinia-specific proliferation and IFN-gamma production of T cells from PP and MLN, and (iii) caused increased bacterial growth in PP and MLN followed by profound tissue destruction. Neutralization of TNF-alpha or IFN-gamma had comparable effects, suggesting that cell-mediated host responses including activated macrophages are required for control of yersiniae in intestinal tissues. The number of Mac-1+ cells in PP and MLN increased after yersinia infection, and recruitment of these cells was not blocked by administration of anticytokine or anti-integrin antibodies. While anti-VLA-4, -TNF-alpha, or -IFN-gamma antibody treatment caused an increased dissemination of yersiniae from PP to the spleen systemic dissemination was reduced by anti-Mac-1 antibodies. The results of this study suggest that the cytokines IFN-gamma and TNF-alpha as well as the integrins Mac-1 and VLA-4 are involved in protective cellular host defense mechanisms in PP and MLN against Y. enterocolitica, the latter probably being involved in both cell-cell and cell-pathogen interactions.
Collapse
Affiliation(s)
- I B Autenrieth
- Institut für Hygiene and Mikrobiologie der Universität Würzburg, Germany
| | | | | | | | | |
Collapse
|
114
|
Takahashi I, Kiyono H, Jackson RJ, Fujihashi K, Staats HF, Hamada S, Clements JD, Bost KL, McGhee JR. Epitope maps of the Escherichia coli heat-labile toxin B subunit for development of a synthetic oral vaccine. Infect Immun 1996; 64:1290-8. [PMID: 8606092 PMCID: PMC173917 DOI: 10.1128/iai.64.4.1290-1298.1996] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Linear B- and T-cell epitopes spanning all 103 amino acids of the Escherichia coli heat-labile toxin B subunit (LT-B) were assessed in mice orally immunized with native LT or with recombinant Salmonella enteritidis expressing LT-B. Oral administration of native LT induced mucosal immunoglobulin A (IgA) antibodies reactive with an epitope at residues 85 to 91, while IgA induced by recombinant Salmonella LT-B reacted with an epitope at residues 36 to 44. Serum IgG anti-LT-B antibodies from mice orally immunized with either LT or with recombinant Salmonella LT-B were directed to both epitopes. A single T-cell epitope spanning residues 34 to 42 was identified by T-cell proliferative and cytokine responses. When a 20-mer peptide (residues 26 to 45) with B- and T-cell epitopes was given orally to BALB/c (H-2(d)) and B10 congenic (I-A(d), I-A(b), and I-A(k)) mice, significant fecal IgA and serum IgG anti-LT-B antibodies were induced. The peptide also induced LT-B-specific T-cell proliferative responses in these mice. Orally administered LT-B peptide (residues 26 to 45) induced a cytokine profile indicative of both T helper 1- and 2-type cells. The remarkable immunogenicity of this 20-mer peptide makes it a candidate for a vaccine to protect against enterotoxigenic E. coli.
Collapse
Affiliation(s)
- I Takahashi
- Departments of Microbiology and Oral Biology, University of Alabama at Birmingham Medical Center, 35294, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
115
|
Di Tommaso A, Saletti G, Pizza M, Rappuoli R, Dougan G, Abrignani S, Douce G, De Magistris MT. Induction of antigen-specific antibodies in vaginal secretions by using a nontoxic mutant of heat-labile enterotoxin as a mucosal adjuvant. Infect Immun 1996; 64:974-9. [PMID: 8641809 PMCID: PMC173865 DOI: 10.1128/iai.64.3.974-979.1996] [Citation(s) in RCA: 173] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Immunization of the female reproductive tract is important for protection against sexually transmitted diseases and other pathogens of the reproductive tract. However, intravaginal immunization with soluble antigens generally does not induce high levels of secretory immunoglobulin A (IgA). We recently developed safe mucosal adjuvants by genetically detoxifying Escherichia coli heat-labile enterotoxin, a molecule with a strong mucosal adjuvant activity, and here we describe the use of the nontoxic mutant LTK63 to induce a response in the mouse vagina against ovalbumin (Ova). We compared intravaginal and intranasal routes of immunization for induction of systemic and vaginal responses against LTK63 and Ova. We found that LTK63 is a potent mucosal immunogen when given by either the intravaginal or intranasal route. It induces a strong systemic antibody response and IgG and long-lasting IgA in the vagina. The appearance of vaginal IgA is delayed in the intranasally immunized mice, but the levels of vaginal anti-LTK63 IgA after repeated immunizations are higher in the intranasally immunized mice than in the intravaginally immunized mice. LTK63 also acts as a mucosal adjuvant, inducing a serum response against Ova, when given by both the intravaginal and intranasal routes. However, vaginal IgA against Ova is stimulated more efficiently when LTK63 and antigen are given intranasally. In conclusion, our results demonstrate that LTK63 can be used as a mucosal adjuvant to induce antigen-specific antibodies in vaginal secretions and show that the intranasal route of immunization is the most effective for this purpose.
Collapse
|
116
|
Gorczynski RM, Chen Z, Hoang Y, Rossi-Bergman B. A subset of gamma delta T-cell receptor-positive cells produce T-helper type-2 cytokines and regulate mouse skin graft rejection following portal venous pretransplant preimmunization. Immunology 1996; 87:381-9. [PMID: 8778022 PMCID: PMC1384105 DOI: 10.1046/j.1365-2567.1996.481554.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
C3H/HeJ mice received B10.BR skin grafts following portal or lateral tail vein infusion of irradiated B10.BR spleen cells. Thereafter mice were injected with anti-alpha beta or anti-gamma delta T-cell receptor (TCR) monoclonal antibody (mAb). Anti-gamma delta TCR mAb abolished the increased graft survival afforded by portal venous (p.v.) immunization, and reversed the bias towards expression of mRNA for type-2 cytokines [interleukin-4 (IL-4), IL-10] seen in lymphoid tissue of p.v.-immunized mice. When gamma delta TCR+ and alpha beta TCR+ cells were isolated from the intestinal epithelial compartment (IEL), liver or Peyer's Patch (PP) of p.v.-immunized mice, the gamma delta TCR+ cells were found to be enriched in cells producing type-2 cytokines on rechallenge with irradiated B10.BR cells in vitro. gamma delta TCR+ cells from p.v.-immunized mice were further expanded in vitro with anti-CD3 and cytokines (combined IL-2 and IL-4). Following expansion these cells were capable of adoptively transferring increased B10.BR skin graft survival to naive mice, and continued to show a bias in type-2 cytokine synthesis after allostimulation in vitro. When gamma delta TCR chain expression was assessed in cells taken from p.v.-immunized mice, or in cells expanded in culture, our data suggest that p.v. immunization leads to oligoclonal, not polyclonal, expansion of those gamma delta TCR+ cells involved in inhibition of graft rejection.
Collapse
Affiliation(s)
- R M Gorczynski
- Department of Surgery, University of Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
117
|
Brandtzaeg P. The human intestinal immune system: basic cellular and humoral mechanisms. BAILLIERE'S CLINICAL RHEUMATOLOGY 1996; 10:1-24. [PMID: 8674142 DOI: 10.1016/s0950-3579(96)80003-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Adaptive immune protection of mucous membranes is provided mainly by secretory IgA (SIgA) antibodies. This first-line defence is accomplished through an ingenious cooperation between the mucosal B-cell system and the epithelial glycoprotein called secretory component (SC). This is quantitatively the most important receptor of the immune system because it is responsible for external transport of locally produced polymeric IgA (pIgA), which is the major humoral mediator substance of the whole immune system. Transmembrane SC belongs to the Ig supergene family and functions as a general pIg receptor, also translocating pentameric IgM externally to form secretory IgM. The B-cells responsible for local pIg production are initially stimulated in lymphoepithelial structures, particularly the Peyer's patches in the distal small intestine, from which they migrate as memory cells to exocrine tissues all over the body. Mucous membranes are thus furnished with secretory antibodies in an integrated way, ensuring a variety of specificities at every secretory site. There is currently great interest in exploiting this integrated or "common' mucosal immune system for oral vaccination against pathogenic infectious agents and also to induce therapeutic peripheral tolerance to ameliorate T-cell-mediated autoimmune diseases. Much remains to be learnt about antigen uptake and processing necessary to elicit stimulatory or suppressive mucosal immune responses, and how normal homeostasis is maintained in the intestinal mucosa. Considerable information has accumulated about various types of immune deviation that may lead to local or extraintestinal hypersensitivity reactions against luminal antigen, but the crucial mechanisms remain obscure.
Collapse
Affiliation(s)
- P Brandtzaeg
- Institute of Pathology, Medical Faculty, University of Oslo, Norway
| |
Collapse
|
118
|
Avtushenko SS, Sorokin EM, Zoschenkova NY, Zacharova NG, Naichin AN. Clinical and immunological characteristics of the emulsion form of inactivated influenza vaccine delivered by oral immunization. J Biotechnol 1996; 44:21-8. [PMID: 8717382 DOI: 10.1016/0168-1656(95)00105-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Prophylaxis of human respiratory diseases caused by influenza viruses is actually a problem of infectious pathology because of their wide prevalence. In our investigations, safety, reactogenicity and immunological activity of the orally administered emulsion-inactivated influenza vaccine prepared from influenza virus strains of types A(H1N1), A(H3N2) and B have been studied. Clinical studies of the emulsion-inactivated influenza vaccine on volunteers has shown its safety and nonreactogenicity. The orally administered vaccine did not cause weak, middle or strong general or local reactions including clinical, biochemical, haemotological and immunological reactions. The emulsion-inactivated vaccine has high immunological activity and induces reliable increases in the level of secretory immunoglobulin A to influenza viruses A and B in protective titers in nasal secretions and saliva of volunteers after one oral administration. The obtained results indicate the expediency of further investigation and improvement of inactivated influenza vaccine for oral administration.
Collapse
MESH Headings
- Administration, Oral
- Adolescent
- Adult
- Antibodies, Viral/biosynthesis
- Antibodies, Viral/blood
- Antibody Formation
- Emulsions
- Humans
- Immunoglobulin A, Secretory/analysis
- Immunoglobulin A, Secretory/biosynthesis
- Influenza A virus/immunology
- Influenza B virus/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/adverse effects
- Influenza Vaccines/immunology
- Influenza, Human/epidemiology
- Influenza, Human/immunology
- Influenza, Human/prevention & control
- Prevalence
- Safety
- Saliva/immunology
- Vaccines, Inactivated/administration & dosage
- Vaccines, Inactivated/adverse effects
- Vaccines, Inactivated/immunology
Collapse
Affiliation(s)
- S S Avtushenko
- Research Institute of Highly Pure Biopreparations, St. Petersburg, Russia
| | | | | | | | | |
Collapse
|
119
|
Affiliation(s)
- P C Doherty
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
120
|
Torres JF, Lyerly DM, Hill JE, Monath TP. Evaluation of formalin-inactivated Clostridium difficile vaccines administered by parenteral and mucosal routes of immunization in hamsters. Infect Immun 1995; 63:4619-27. [PMID: 7591115 PMCID: PMC173664 DOI: 10.1128/iai.63.12.4619-4627.1995] [Citation(s) in RCA: 89] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Clostridium difficile produces toxins that cause inflammation, necrosis, and fluid in the intestine and is the most important cause of nosocomial antibiotic-associated diarrhea and colitis. We evaluated C. difficile antigens as vaccines to protect against systemic and intestinal disease in a hamster model of clindamycin colitis. Formalin-inactivated culture filtrates from a highly toxigenic strain were administered by mucosal routes (intranasal, intragastric, and rectal) with cholera toxin as a mucosal adjuvant. A preparation of culture filtrate and killed whole cells was also tested rectally. The toxoid was also tested parenterally (subcutaneously and intraperitoneally) and by a combination of three intranasal immunizations followed by a combined intranasal-intraperitoneal boost. Serum antibodies against toxins A and B and whole-cell antigen were measured by enzyme-linked immunosorbent assay, neutralization of cytotoxic activity, and bacterial agglutination. The two rectal immunization regimens induced low antibody responses and protected only 20% of hamsters against death and 0% against diarrhea. The intragastric regimen induced high antibody responses but low protection, 40% against death and 0% against diarrhea. Hamsters immunized by the intranasal, intraperitoneal, and subcutaneous routes were 100% protected against death and partially protected (40, 40, and 20%, respectively) against diarrhea. Among the latter groups, intraperitoneally immunized animals had the highest serum anticytotoxic activity and the highest agglutinating antibody responses. Hamsters immunized intranasally and revaccinated intraperitoneally were 100% protected against both death and diarrhea. Protection against death and diarrhea correlated with antibody responses to all antigens tested. The results indicate that optimal protection against C. difficile disease can be achieved with combined parenteral and mucosal immunization.
Collapse
Affiliation(s)
- J F Torres
- OraVax, Inc., Cambridge, Massachusetts 02139, USA
| | | | | | | |
Collapse
|
121
|
Karem KL, Chatfield S, Kuklin N, Rouse BT. Differential induction of carrier antigen-specific immunity by Salmonella typhimurium live-vaccine strains after single mucosal or intravenous immunization of BALB/c mice. Infect Immun 1995; 63:4557-63. [PMID: 7591105 PMCID: PMC173654 DOI: 10.1128/iai.63.12.4557-4563.1995] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
In this study, we constructed strain KR21 (chi 4550 delta cya delta crp delta asd/pYA292asd(+)-toxC+) and compared it with BRD847 (aroA aroD/pnirB-toxC) for the ability to induce humoral and cellular immunity after a single oral or intravenous immunization in 3- to 4-week-old BALB/c mice. ToxC-specific serum immunoglobulin G (IgG) was detectable in animals orally immunized with either BRD847 or KR21. However, after intravenous immunization, IgG was detected only in BRD847-immunized animals. Measurement of immunoglobin types IgG1 and IgG2a suggests that a Th1 cellular response is prominent after immunizations with either system. ToxC-specific IgA was detected in fecal and vaginal samples of animals immunized orally and intravenously with BRD847, while those immunized with KR21 failed to show fecal or vaginal IgA responses. Delayed-type hypersensitivity was used as a measure of induction of T-cell responses in vivo. Mice immunized either orally or intravenously with BRD847 showed significant ear swelling responses after ToxC injections, while KR21-immunized animals failed to show a cellular response. These data indicate that the aroA aroD/pnirB system holds greater potential for inducing global immunity after a single dose when directly compared with the balanced lethal system (delta cya delta crp delta asd/pYA292asd+).
Collapse
MESH Headings
- Administration, Oral
- Animals
- Antigens, Bacterial/genetics
- Antigens, Bacterial/immunology
- Bacterial Vaccines/administration & dosage
- Bacterial Vaccines/immunology
- Female
- Hypersensitivity, Delayed
- Immunization
- Immunoglobulin A, Secretory/blood
- Immunoglobulin G/blood
- Immunoglobulin G/classification
- Injections, Intravenous
- Mice
- Mice, Inbred BALB C
- Plasmids
- Salmonella typhimurium/genetics
- Salmonella typhimurium/immunology
- Tetanus Toxin/genetics
- Vaccines, Attenuated/administration & dosage
- Vaccines, Attenuated/immunology
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- K L Karem
- Department of Microbiology, University of Tennessee College of Veterinary Medicine, Knoxville 37996, USA
| | | | | | | |
Collapse
|
122
|
Muster T, Ferko B, Klima A, Purtscher M, Trkola A, Schulz P, Grassauer A, Engelhardt OG, García-Sástre A, Palese P. Mucosal model of immunization against human immunodeficiency virus type 1 with a chimeric influenza virus. J Virol 1995; 69:6678-86. [PMID: 7474077 PMCID: PMC189577 DOI: 10.1128/jvi.69.11.6678-6686.1995] [Citation(s) in RCA: 93] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Previously, we constructed a chimeric influenza virus that expresses the highly conserved amino acid sequence ELDKWA of gp41 of human immunodeficiency virus type 1 (HIV-1). Antisera elicited in mice by infection with this chimeric virus showed neutralizing activity against distantly related HIV-1 isolates (T. Muster, R. Guinea, A. Trkola, M. Purtscher, A. Klima, F. Steindl, P. Palese, and H. Katinger, J. Virol. 68:4031-4034, 1994). In the present study, we demonstrated that intranasal immunizations with this chimeric virus are also able to induce a humoral immune response at the mucosal level. The immunized mice had ELDKWA-specific immunoglobulins A in respiratory, intestinal, and vaginal secretions. Sustained levels of these secretory immunoglobulins A were detectable for more than 1 year after immunization. The results show that influenza virus can be used to efficiently induce secretory antibodies against antigens from foreign pathogens. Since long-lasting mucosal immunity in the genital and intestinal tracts might be essential for protective immunity against HIV-1, influenza virus appears to be a promising vector for HIV-1-derived immunogens.
Collapse
Affiliation(s)
- T Muster
- Institut für Angewandte Mikrobiologie, Universität für Bodenkultur, Vienna, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
123
|
Horowitz NS, Staats HF, Palker TJ. Effect of bismuth salts on systemic and mucosal immune responses to orally administered cholera toxin. IMMUNOPHARMACOLOGY 1995; 31:31-41. [PMID: 8655289 DOI: 10.1016/0162-3109(95)00031-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
While the antimicrobial and antisecretory effects of bismuth salts are well documented, little is known regarding their effects on immune responses to enterotoxins such as that of V. cholerae or to orally administered vaccine antigens. To evaluate the effects of Pepto Bismol (PB) on the induction of systemic and mucosal immune responses to cholera toxin (CT), C57BL/6 mice were orally administered 10 micrograms CT and PB, or mice were pretreated with PB 30 min prior to CT administration. When co-administered with CT, PB attenuated serum IgG1, IgG2a, IgG2b and IgG3 anti-CT responses in a dose-dependent manner and also reduced levels of circulating anti-CT IgA and total serum IgE. Similarly, anti-CT intestinal IgA responses were also decreased. However, when administered 30 min prior to CT, PB had little to no effect on serum or intestinal anti-CT immunoglobulin responses. Administration of bismuth subsalicylate (BSS), the active component of PB, or sodium salicylate did not reduce immune responses to CT, suggesting that the combination of BSS plus other constituents contained within PB contributed to the decreased immune response to CT. Moreover, bismuth subgallate alone inhibited antibody responses to CT. Our data are consistent with the hypothesis that, when administered orally with CT, PB and bismuth subgallate create a physical barrier to antigen uptake.
Collapse
Affiliation(s)
- N S Horowitz
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | | | | |
Collapse
|
124
|
Maloy KJ, Donachie AM, Mowat AM. Induction of Th1 and Th2 CD4+ T cell responses by oral or parenteral immunization with ISCOMS. Eur J Immunol 1995; 25:2835-41. [PMID: 7589080 DOI: 10.1002/eji.1830251019] [Citation(s) in RCA: 71] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
We examined the ability of oral or parenteral immunization with immune stimulating complexes containing ovalbumin (ISCOMS-OVA) to prime T cell proliferative and cytokine responses. A single subcutaneous immunization with ISCOMS-OVA primed potent antigen-specific proliferative responses in the draining popliteal lymph node, which were entirely dependent on the presence of CD4+ T cells. CD8+ T cells did not proliferate in vitro even in the presence of the appropriate peptide epitope and exogenous interleukin (IL)-2. Primed popliteal lymph node cells produced IL-2, IL-5 and interferon (IFN)-gamma, but not IL-4 when restimulated with OVA in vitro. Serum antigen-specific IgG1 and IgG2a antibody responses were also primed by subcutaneous immunization with ISCOMS-OVA, confirming the stimulation of both Th1 and Th2 cells in vivo. Spleen cells from subcutaneously primed mice produced a similar pattern of cytokines, indicating that disseminated priming had occurred. Oral immunization with ISCOMS-OVA also primed local antigen-specific proliferative responses in the mesenteric lymph node and primed an identical pattern of systemic cytokine responses in the spleen. The ability of ISCOMS to prime both Th1 and Th2 CD4+ T cell responses may be central to their potent adjuvant activities and confirm the potential of ISCOMS as future oral vaccine vectors.
Collapse
Affiliation(s)
- K J Maloy
- Department of Immunology, University of Glasgow, Western Infirmary, Scotland
| | | | | |
Collapse
|
125
|
Ferrero RL, Thiberge JM, Kansau I, Wuscher N, Huerre M, Labigne A. The GroES homolog of Helicobacter pylori confers protective immunity against mucosal infection in mice. Proc Natl Acad Sci U S A 1995; 92:6499-503. [PMID: 7604021 PMCID: PMC41545 DOI: 10.1073/pnas.92.14.6499] [Citation(s) in RCA: 222] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Helicobacter pylori is an important etiologic agent of gastroduodenal disease. In common with other organisms, H. pylori bacteria express heat shock proteins that share homologies with the GroES-GroEL class of proteins from Escherichia coli. We have assessed the heat shock proteins of H. pylori as potential protective antigens in a murine model of gastric Helicobacter infection. Orogastric immunization of mice with recombinant H. pylori GroES- and GroEL-like proteins protected 80% (n = 20) and 70% (n = 10) of animals, respectively, from a challenge dose of 10(4) Helicobacter felis bacteria (compared to control mice, P = 0.0042 and P = 0.0904, respectively). All mice (n = 19) that were immunized with a dual antigen preparation, consisting of H. pylori GroES-like protein and the B subunit of H. pylori urease, were protected against infection. This represented a level of protection equivalent to that provided by a sonicated Helicobacter extract (P = 0.955). Antibodies directed against the recombinant H. pylori antigens were predominantly of the IgG1 class, suggesting that a type 2 T-helper cell response was involved in protection. This work reports a protein belonging to the GroES class of heat shock proteins that was shown to induce protective immunity. In conclusion, GroES-like and urease B-subunit proteins have been identified as potential components of a future H. pylori subunit vaccine.
Collapse
Affiliation(s)
- R L Ferrero
- Unité des Entérobactéries, Institut National de la Santé et de la Recherche Médicle U389, Paris, France
| | | | | | | | | | | |
Collapse
|
126
|
Fedorka-Cray PJ, Kelley LC, Stabel TJ, Gray JT, Laufer JA. Alternate routes of invasion may affect pathogenesis of Salmonella typhimurium in swine. Infect Immun 1995; 63:2658-64. [PMID: 7790082 PMCID: PMC173356 DOI: 10.1128/iai.63.7.2658-2664.1995] [Citation(s) in RCA: 131] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Transmission of Salmonella typhimurium in swine is traditionally believed to occur by the fecal-oral route, with invasion through the intestinal wall and Peyer's patches. However, involvement of the upper respiratory tract may be equally important. An esophagotomy was performed on 6- to 8-week-old pigs. Esophagotomized pigs were challenged intranasally with 10(9) CFU of S. typhimurium cells and necropsied at 3, 6, 12, and 18 h postinoculation (p.i.). By 3 h p.i., S. typhimurium was recovered from cecum, colon, head, and thoracic tissues and from the middle ileum involving a large number of Peyer's patches. The ileocolic lymph nodes and ileocolic junction were not positive for S. typhimurium until 6 and 12 h p.i., respectively. Additional pigs were inoculated transthoracically with 10(9) CFU of S. typhimurium and necropsied at 3 and 18 h p.i. By 3 h p.i., all tissues were positive for S. typhimurium. Tonsil explants seeded with 10(9) CFU of S. typhimurium indicated that within 6 h p.i., S. typhimurium was located within the tonsilar crypts. These data show that after intranasal inoculation, S. typhimurium rapidly appears in the gut tissues and suggest that the tonsils and lung may be important sites for invasion and dissemination of Salmonella species.
Collapse
Affiliation(s)
- P J Fedorka-Cray
- USDA-Agricultural Research Service-National Animal Disease Center, Ames, Iowa 50010, USA
| | | | | | | | | |
Collapse
|
127
|
Bukawa H, Sekigawa K, Hamajima K, Fukushima J, Yamada Y, Kiyono H, Okuda K. Neutralization of HIV-1 by secretory IgA induced by oral immunization with a new macromolecular multicomponent peptide vaccine candidate. Nat Med 1995; 1:681-5. [PMID: 7585151 DOI: 10.1038/nm0795-681] [Citation(s) in RCA: 103] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Control of pandemic infection of human immunodeficiency virus type 1 (HIV-1) requires some means of developing mucosal immunity against HIV-1 because sexual transmission of the virus occurs mainly through the mucosal tissues. However, there is no evidence as yet that the secretory immunoglobulin A (IgA) antibody induced by immunization with antigens in experimental animals can neutralize HIV-1. We demonstrate here that oral immunization with a new macromolecular peptide antigen and cholera toxin (CT) induces a high titre (1:2") of gut-associated and secretory IgA antibody to HIV-1. Using three different neutralizing assays, we clearly demonstrate that this secretory IgA antibody is able to neutralize HIV-1IIIB, HIV-1SF2 and HIV-1MN. Our new approach may prove to be important in the development of a mucosal vaccine that will provide protection of mucosal surfaces against HIV-1.
Collapse
Affiliation(s)
- H Bukawa
- Department of Oral and Maxillofacial Surgery, Yokohama City University School of Medicine, Japan
| | | | | | | | | | | | | |
Collapse
|
128
|
Abstract
Adaptive immunological protection of mucous membranes is provided mainly by secretory IgA antibodies. Such "first line" defence is accomplished through a unique cooperation between the mucosal B-cell system and the secretory component (SC) expressed basolaterally on glandular epithelial cells. This transmembrane glycoprotein is quantitatively the most important receptor of the immune system because it is responsible for external transport of locally produced polymeric IgA (pIgA), which is the major product of humoral immunity. Transmembrane SC belongs to the Ig supergene family and functions as a general pIg receptor, also mediating the external translocation of pentameric IgM to form secretory IgM. The B cells responsible for local pIg production are initially stimulated in lymphoepithelial structures, particularly the Peyer's patches in the distal small intestine, from which they migrate as memory cells to exocrine tissues all over the body. Mucous membranes are thus furnished with secretory antibodies in an integrated way, ensuring a variety of specificities at every secretory site. There is currently great interest in exploiting this integrated or "common" mucosal immune system for oral vaccination against pathogenic infectious agents. However, much remains to be learned about mechanisms for antigen uptake and processing necessary to elicit mucosal immunity as well as the molecular biology and cytokine regulation of SC-dependent pIg transport. Moreover, evidence is emerging for the existence of subcompartmentalization in the mucosal immune system, particularly a dichotomy in cellular migration between the gut and the upper airway, which may complicate the design of efficient local vaccines.
Collapse
Affiliation(s)
- P Brandtzaeg
- Laboratory for Immunohistochemistry and Immunopathology (LIIPAT), University of Oslo, National Hospital, Norway
| |
Collapse
|