Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Allergy ; 68(8): 1021-8, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23909913

ABSTRACT

BACKGROUND: Modified vaccinia virus Ankara (MVA)-encoding antigens are considered as safe vaccine candidates for various infectious diseases in humans. Here, we investigated the immune-modulating properties of MVA-encoding ovalbumin (MVA-OVA) on the allergen-specific immune response. METHODS: The immune-modulating properties of MVA-OVA were investigated using GM-CSF-differentiated BMDCs from C57BL/6 mice. OVA expression upon MVA-OVA infection of BMDCs was monitored. Activation and maturation markers on viable MVA-OVA-infected mDCs were analyzed by flow cytometry. Secretion of INF-γ, IL-2, and IL-10 was determined in a co-culture of BMDCs infected with wtMVA or MVA-OVA and OVA-specific OT-I CD8(+) and OT-II CD4(+ ) T cells. BALB/c mice were vaccinated with wtMVA, MVA-OVA, or PBS, sensitized to OVA/alum and challenged with a diet containing chicken egg white. OVA-specific IgE, IgG1, and IgG2a and cytokine secretion from mesenteric lymph node (MLN) cells were analyzed. Body weight, body temperature, food uptake, intestinal inflammation, and health condition of mice were monitored. RESULTS: Infection with wtMVA and MVA-OVA induced comparable activation of mDCs. MVA-OVA-infected BMDCs expressed OVA and induced enhanced IFN-γ and IL-2 secretion from OVA-specific CD8(+ ) T cells in comparison with OVA, wtMVA, or OVA plus wtMVA. Prophylactic vaccination with MVA-OVA significantly repressed OVA-specific IgE, whereas OVA-specific IgG2a was induced. MVA-OVA vaccination suppressed TH 2 cytokine production in MLN cells and prevented the onset of allergic symptoms and inflammation in a mouse model of OVA-induced intestinal allergy. CONCLUSION: Modified vaccinia virus Ankara-ovalbumin (MVA-OVA) vaccination induces a strong OVA-specific TH 1- immune response, likely mediated by the induction of IFN-γ and IgG2a. Finally, MVA-based vaccines need to be evaluated for their therapeutic potential in established allergy models.


Subject(s)
Allergens/immunology , Food Hypersensitivity/immunology , Food Hypersensitivity/prevention & control , Immunotherapy, Adoptive/methods , Intestinal Mucosa/immunology , Vaccinia virus/immunology , Viral Vaccines/immunology , Allergens/genetics , Allergens/therapeutic use , Animals , Bone Marrow Transplantation/methods , Cells, Cultured , Coculture Techniques , Dendritic Cells/immunology , Dendritic Cells/transplantation , Dendritic Cells/virology , Disease Models, Animal , Food Hypersensitivity/genetics , Inflammation/immunology , Inflammation/prevention & control , Inflammation/virology , Intestinal Mucosa/pathology , Intestinal Mucosa/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Ovalbumin/genetics , Ovalbumin/immunology , Ovalbumin/therapeutic use , Th1 Cells/immunology , Th1 Cells/metabolism , Th1 Cells/virology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/therapeutic use , Vaccinia/genetics , Vaccinia/immunology , Vaccinia/pathology , Vaccinia virus/genetics , Viral Vaccines/genetics , Viral Vaccines/therapeutic use
2.
Scand J Immunol ; 78(1): 61-8, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23672535

ABSTRACT

ß-Glucans, glucose polymers that are the main constituents of the outer cell walls of micro-organisms such as fungi and yeast, are known to play an immunostimulatory role. We prepared ß-glucan (ß-(1-3),(1-6)-D-glucan) from an edible cultured fungus through fermentation techniques using a strain of Aureobasidium pullulans ADK-34. The purity of this ß-glucan preparation (AP-FBG) was demonstrated to be high through various instrumental analyses. We then examined the effects of AP-FBG on intestinal immune systems. We prepared Peyer's patch (PP) cells and measured interleukin (IL)-5, IL-6, and IgA production in culture media with AP-FBG. We found that both cytokines and IgA increased; furthermore, IL-6 secreted by PP dendritic cells (PPDCs) cultured in the presence of AP-FBG significantly increased. We tested IgA production after oral administration of AP-FBG for 2 weeks and found that AP-FBG tended to promote the production of IgA in the small intestine. Interestingly, we observed a significant increase in IgA production in the small intestines of mice treated with cyclophosphamide (CY; an immunosuppressant) after oral administration of AP-FBG diet compared with CY-treated and control diet mice. Production of IL-6 and IgA by PP cells and IL-6 production by PPDCs in AP-FBG-fed and CY-treated mice also increased. These results demonstrate that AP-FBG has the ability to activate PPDC and induce IL-6 production and IgA secretion in PP cells. These abilities were more clearly expressed when AP-FBG was orally administered in a CY-induced immunosuppressed condition. Therefore, AP-FBG may be a useful ingredient for preparing functional foods with immunomodulatory activities.


Subject(s)
Adjuvants, Immunologic/pharmacology , Ascomycota/chemistry , Intestines/immunology , beta-Glucans/pharmacology , Animals , Cyclophosphamide/pharmacology , Cytokines/biosynthesis , Female , Immunoglobulin A/biosynthesis , Male , Mice , Mice, Inbred BALB C , beta-Glucans/analysis
3.
Mol Genet Metab ; 103(1): 98-100, 2011 May.
Article in English | MEDLINE | ID: mdl-21320791

ABSTRACT

Animal and human studies of enzyme replacement therapy for Pompe disease have indicated that antibodies generated against the infused recombinant human acid α-glucosidase (rhGAA) can negatively impact therapeutic outcome. In this study, we show that oral administration of rhGAA into mice can reduce the titer of anti-rhGAA antibody following immunization with rhGAA. Oral administration of rhGAA is safe and antigen specific, it offers advantages over other immunosuppressive drugs.


Subject(s)
Antibodies/blood , Antibodies/immunology , Enzyme Replacement Therapy , Glycogen Storage Disease Type II/immunology , Glycogen Storage Disease Type II/therapy , alpha-Glucosidases/immunology , Administration, Oral , Animals , Antibody Formation , Disease Models, Animal , Humans , Immunoglobulin E/blood , Immunoglobulin G/blood , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Recombinant Proteins/administration & dosage
4.
Clin Exp Immunol ; 140(2): 249-57, 2005 May.
Article in English | MEDLINE | ID: mdl-15807848

ABSTRACT

Migration of intraepithelial lymphocytes (IELs) into intestinal epithelium is not yet well understood. We established an IEL-cell line from ovalbumin (OVA) 23-3 transgenic (Tg) mice and investigated the effect of antigen stimulation on the dynamic process of IEL migration into small intestinal mucosa. The cell line was a T cell receptor (TCR) alphabeta(+) CD4(+) CD8(-) phenotype, expressing alphaEbeta7 integrin in 90% of cells. Under intravital microscopy, the lined IELs adhered selectively to the microvessels of the intestinal villus tip of the Tg mice. The accumulation of IELs was significantly inhibited by an antibody against beta7-integrin and MAdCAM-1. When IELs were stimulated with OVA, the accumulation was attenuated compared to that of resting cells, with decreased expression of alphaEbeta7 integrin. In Tg mice fed with OVA, the number of IELs which migrated in the villus mucosa was significantly smaller than in the non-fed controls. The preferential migratory capacity of IELs to villus mucosa may be altered by specific antigen stimulations.


Subject(s)
Intestinal Mucosa/immunology , Intestine, Small/immunology , T-Lymphocyte Subsets/immunology , Animals , Antigens/immunology , Antigens, Surface/metabolism , Cell Adhesion/immunology , Cell Adhesion Molecules/immunology , Cell Line , Cell Movement/immunology , Immunity, Mucosal , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Ovalbumin/immunology
5.
Clin Exp Allergy ; 32(4): 563-70, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11972603

ABSTRACT

BACKGROUND: Our previous study using allergen-sensitized murine splenocyte cultures has shown that Lactobacillus casei strain Shirota (LcS), a lactic acid bacterium widely used as a starter for fermented milk products, suppresses IgE production through promoting a dominant Th1-type response mediated by IL-12 induction. OBJECTIVE: We tried to evaluate the ability of LcS to suppress both IgE response and allergic reactions in vivo using a food allergy model with ovalbumin-specific T cell receptor transgenic (OVA-TCR-Tg) mice. METHODS: The ability of heat-killed LcS to induce IL-12 in serum was tested. OVA-TCR-Tg mice were fed a diet containing OVA for 4 weeks and injected with LcS intraperitoneally three times in the first week of this period. Cytokine and antibody secretion by splenocytes, and serum IgE and IgG1 responses were examined. The inhibitory effect of LcS on systemic anaphylaxis induced by intravenous challenge of OVA-fed OVA-TCR-Tg mice with OVA was also tested. RESULTS: Intraperitoneal injection of LcS induced an IL-12 response in the serum of OVA-TCR-Tg mice. In the food allergy model, LcS administration skewed the pattern of cytokine production by splenocytes toward Th1 dominance, and suppressed IgE and IgG1 secretion by splenocytes. The ability of LcS to modulate cytokine production was blocked by anti-IL-12 antibody treatment. LcS also inhibited serum OVA-specific IgE and IgG1 responses and diminished systemic anaphylaxis. CONCLUSION: LcS administration suppresses IgE and IgG1 responses and systemic allergic reactions in a food allergy model, suggesting a possible use of this lactic acid bacterium in preventing food allergy.


Subject(s)
Anaphylaxis/prevention & control , Food Hypersensitivity/immunology , Immunoglobulin E/blood , Immunoglobulin G/blood , Lacticaseibacillus casei , Animals , Antibodies/pharmacology , Cells, Cultured , Cytokines/biosynthesis , Genes, T-Cell Receptor , Immunoglobulin E/biosynthesis , Immunoglobulin G/biosynthesis , Interleukin-12/antagonists & inhibitors , Interleukin-12/blood , Interleukin-12/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Ovalbumin/immunology , Peptide Fragments/immunology , Spleen/cytology , Spleen/immunology , Th1 Cells/immunology
6.
Int Immunol ; 13(9): 1165-74, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11526097

ABSTRACT

The recirculation of lymphocytes through the intestinal mucosa is important for specific immune defense, but the origin and differentiation of intraepithelial lymphocytes (IEL) are not fully understood. The present study therefore used intravital microscopy to investigate the migration of IEL to the villus mucosa and Peyer's patches of the small intestine. IEL were separated from inverted murine small intestine and mesenteric lymph node (MLN) T cells were also isolated. The adhesion of fluorescence-labeled lymphocytes to postcapillary venules (PCV) of Peyer's patches and arcade microvessels of small intestinal villi was observed after injection. In some experiments, the effect of antibodies against adhesion molecules on cell kinetics were investigated. IEL time-dependently accumulated in villus microvessels of the small intestine, whereas few MLN cells did. Few IEL adhered to the PCV of Peyer's patches. IEL were shown to express alpha(E)beta(7)-integrin but not L-selectin. The accumulation of IEL in villus archade was significantly inhibited by antibody against beta(7)-integrin or mucosal addressin cell adhesion molecules (MAdCAM)-1, but not by alpha(E)-integrin. The combined blocking of beta(7)-integrin and MAdCAM-1 further attenuated the sticking of IEL in this area, although it did not entirely block the IEL adherence. The adherence of CD4(+) or TCRalphabeta IEL to villus microvessels was significantly greater than that of CD4(-) or TCRgammadelta IEL. It was demonstrated in situ for the first time that IEL adhered selectively to the villus microvessels of the small intestine partly via beta(7) and MAdCAM-1.


Subject(s)
Integrin alpha Chains , Integrin beta Chains , Intestinal Mucosa/immunology , Intestine, Small/immunology , Lymphocytes/immunology , Microcirculation/immunology , Animals , Antigens, CD , CD4 Antigens , Cell Adhesion , Cell Adhesion Molecules , Cell Differentiation , Cell Movement , Female , Immunoglobulins , Integrins , Intestinal Mucosa/cytology , Intestine, Small/blood supply , Intestine, Small/cytology , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymphocyte Subsets/cytology , Lymphocyte Subsets/immunology , Lymphocytes/cytology , Male , Mesentery/cytology , Mesentery/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microcirculation/cytology , Mucoproteins , Peyer's Patches/blood supply , Peyer's Patches/cytology , Peyer's Patches/immunology , Receptors, Antigen, T-Cell, alpha-beta , Receptors, Lymphocyte Homing
7.
Biosci Biotechnol Biochem ; 65(5): 1170-4, 2001 May.
Article in English | MEDLINE | ID: mdl-11440133

ABSTRACT

Oral administration of a protein without adjuvant brings about oral tolerance (systemic hyporesponsiveness) to that protein by mechanisms such as antigen-induced apoptosis. We monitored the number and apoptosis induction of CD4+ T cells in antigen-specific T cell receptor transgenic mice fed the antigen ovalbumin to identify where events leading to oral tolerance occurred. The antigen was distributed throughout the body, causing apoptosis and a decrease in cell number of CD4+ T cells in most of the lymphoid system: the spleen, peripheral lymph nodes, and the thymus which was not previously reported to be affected. Although apoptosis was induced in the Peyer's patches, the cell number did not change. Unexpectedly, T cells in the mesenteric lymph nodes did not undergo apoptosis; instead, they were more numerous as compared to that in the case of control animals not administered the antigen. The results suggested that the orally administered antigen activated the intestinal immune system, while it induced immune tolerance in other sites.


Subject(s)
Antigens/immunology , Apoptosis , CD4-Positive T-Lymphocytes/cytology , Cell Division/immunology , Administration, Oral , Animals , Antigens/administration & dosage , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation , Mice , Mice, Transgenic
8.
Cytotechnology ; 36(1-3): 145-53, 2001 Jul.
Article in English | MEDLINE | ID: mdl-19003325

ABSTRACT

The response of splenic CD4 T cells from ovalbumin (OVA)-specific T cell receptor (TCR) transgenic mice after long-term feeding of a diet containing this antigen was examined. These CD4 T cells exhibited a decreased response to OVA peptide stimulation, in terms of proliferation, interleukin-2 secretion, and CD40 ligand expression, compared to those from mice fed a control diet lacking OVA, demonstrating that oral tolerance of T cells had been induced through oral intake of the antigen. We investigated the intracellular signaling pathways, which were Ca/CN cascade and Ras/MAPK cascade, of these tolerant CD4 T cells using phorbol-12-myristate-13-acetate (PMA) and ionomycin, which are known to directly stimulate these pathways. In contrast to the decreased response to TCR stimulation by OVA peptide, it was shown that the response of splenic CD4 T cells to these reagents in the state of oral tolerance was stronger. These results suggest that splenic CD4 T cells in the state of oral tolerance have an impairment in signaling, in which signals are not transmitted from the TCR to downstream signaling pathways, and have impairments in the vicinity of TCR.

9.
Biosci Biotechnol Biochem ; 64(7): 1459-65, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10945264

ABSTRACT

We have investigated the effects of dietary nucleotides on intraepithelial lymphocytes (IEL) and intestinal epithelial cells (IEC) in weanling mice. The proportion of T-cell receptor (TCR) gammadelta+ IEL in BALB/c mice fed a diet supplemented with nucleotides (NT(+) diet) was significantly higher than that in mice fed the nucleotide-free diet, while the proportion of TCR alphabeta+ IEL in NT(+) diet-fed mice was significantly decreased. The change of the TCR alphabeta+/TCR gammadelta+ ratio was mainly observed in a CD8 alphaalpha+ subset of IEL. IEC from NT(+) diet-fed mice produced a higher level of IL-7, which is important in the development of TCR gammadelta+ IEL, than those from control diet-fed mice. The expression levels of IL-7 and IL-2 receptors on IEL were not different between the two dietary groups. Our findings suggest that the increased population of a TCR gammadelta+ IEL subset by feeding nucleotides may be caused by the increased production of IL-7 by IEC.


Subject(s)
Cytidine Monophosphate/metabolism , Dietary Supplements , Guanosine Monophosphate/metabolism , Inosine Monophosphate/metabolism , Interleukin-7/biosynthesis , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocyte Subsets/metabolism , Uridine Monophosphate/metabolism , Animals , Epithelial Cells/metabolism , Female , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Lymphocyte Count , Mice , Mice, Inbred BALB C , Receptors, Interleukin-2/biosynthesis , Receptors, Interleukin-7/biosynthesis , T-Lymphocyte Subsets/classification , T-Lymphocyte Subsets/cytology
10.
Int Arch Allergy Immunol ; 122(1): 33-41, 2000 May.
Article in English | MEDLINE | ID: mdl-10859467

ABSTRACT

BACKGROUND: It has been reported that dietary nucleotides enhance T helper cell activities. In this study, we have determined the effects of dietary nucleotides on antigen-specific Th1 and Th2 responses and IgE responses. METHODS: Ovalbumin (OVA)-specific T cell receptor (TCR) transgenic (OVA-TCR Tg) mice, 3 weeks old, were fed a nucleotide-free diet (NT(-) diet) or the NT(-) diet supplemented with dietary nucleotides (NT(+) diet) for 4 weeks. Cytokine production by spleen cells and macrophages obtained from these mice was measured in vitro. BALB/c mice, 3 weeks old, immunized intraperitoneally with OVA adsorbed onto alum, were fed the NT(-) diet or the NT(+) diet for 4 weeks. Serum levels of antigen-specific antibodies in the BALB/c mice were determined by ELISA. RESULTS: The level of production of antigen-specific interferon-gamma by spleen cells was significantly higher in the OVA-TCR Tg mice fed the NT(+) diet than in the control mice. The levels of secretion of bioactive IL-12 by spleen cells and peritoneal macrophages were also significantly increased in the NT(+) diet group. The serum OVA-specific IgE level was significantly decreased in BALB/c mice fed the NT(+) diet compared with those fed the NT(-) diet. CONCLUSION: These results show that dietary nucleotides up-regulate the antigen-specific Th1 immune response through the enhancement of IL-12 production and suppress the antigen-specific IgE response.


Subject(s)
Adjuvants, Immunologic , Antibody Specificity , Diet , Immunoglobulin E/immunology , Nucleotides/immunology , Th1 Cells/immunology , Animals , Antigens/immunology , Female , Interferon-gamma/analysis , Interleukin-12/analysis , Interleukin-4/analysis , Macrophages, Peritoneal/immunology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Ovalbumin/immunology , Receptors, Antigen, T-Cell/genetics , Spleen/immunology
11.
J Allergy Clin Immunol ; 105(4): 788-95, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10756231

ABSTRACT

BACKGROUND: The mechanism by which orally ingested allergens elicit an IgE response remains unclear because there are few animal models available for investigation of this response. OBJECTIVE: We tried to develop a murine model suitable for investigation of the IgE response to orally ingested allergens, which would allow us to identify T cells that could promote IgE production. METHODS: Ovalbumin (OVA)-specific T-cell receptor transgenic mice were fed a diet containing OVA, and both the serum antibody response and cytokine production by splenocytes were examined. RESULTS: Oral administration of OVA to transgenic mice led to an increase in the levels of both antigen-specific IgE and total IgE in the sera. Subsequent intravenous challenge of OVA-fed transgenic mice with OVA resulted in anaphylactic shock. Analysis of cytokine production by splenocytes revealed that high IL-4-producing T cells appeared in the spleen 1 week after the start of feeding the OVA diet. T cells from these mice were found to promote IgE secretion by BALB/c B cells in vitro. This helper activity and the levels of IL-4 secretion were diminished after long-term feeding. These findings suggest the possibility that the orally ingested antigen elicited a response by a subpopulation of T cells that produce high levels of T(H2)-type cytokines and that promote IgE secretion, and these same T cells were tolerized by the orally ingested antigen. CONCLUSION: This experimental model with transgenic mice may be a useful tool for further studies of the cellular and molecular mechanisms of the T-cell and IgE responses to orally ingested antigens.


Subject(s)
Antigens/administration & dosage , Immunoglobulin E/blood , Administration, Oral , Allergens/immunology , Anaphylaxis/chemically induced , Animals , Cytokines/biosynthesis , Cytokines/metabolism , Epitopes , Female , Injections, Intraperitoneal , Injections, Intravenous , Mice , Mice, Inbred BALB C , Mice, Transgenic , Models, Immunological , Ovalbumin/administration & dosage , Ovalbumin/immunology , Peyer's Patches/cytology , Peyer's Patches/metabolism , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Spleen/cytology , Spleen/metabolism , T-Lymphocytes, Helper-Inducer/physiology
12.
FEBS Lett ; 465(1): 28-33, 2000 Jan 07.
Article in English | MEDLINE | ID: mdl-10620701

ABSTRACT

Naive CD4(+) T cells differentiate into two types of helper T cells showing an interferon-gamma-predominant (Th1) or an interleukin-4-predominant (Th2) cytokine secretion profile after repeated antigenic stimulation. Their differentiation can be influenced by slight differences in the interaction between the T cell receptor (TCR) and its ligand at the time of primary activation. However, the primary response of freshly isolated naive CD4(+) T cells to altered TCR ligands is still unclear. Here, we investigated the primary response of splenic naive CD4(+) T cells derived from transgenic mice expressing TCR specific for residues 323-339 of ovalbumin (OVA323-339) bound to I-A(d) molecules. Naive CD4(+) T cells secreted either Th1- or Th2-type cytokines immediately after stimulation with OVA323-339 or its single amino acid-substituted analogs. Helper activity for antibody secretion by co-cultured resting B cells was also found in the primary response, accompanied by either low-level Th2-type cytokine secretion or no apparent cytokine secretion. Our results clearly indicate that dichotomy of the Th1/Th2 cytokine secretion profile can be elicited upon primary activation of naive CD4(+) T cells. We also demonstrate that the helper activity of naive CD4(+) T cells for antibody production does not correspond to the amounts of the relevant cytokines secreted.


Subject(s)
Antibodies/immunology , Antigens/pharmacology , CD4-Positive T-Lymphocytes/immunology , Amino Acid Sequence , Animals , Antigens/chemistry , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Coculture Techniques , Histocompatibility Antigens Class II/chemistry , Histocompatibility Antigens Class II/pharmacology , Interleukin-2/analysis , Interleukin-4/analysis , Lymphocyte Activation , Mice , Mice, Transgenic , Molecular Sequence Data , Th1 Cells/immunology , Th2 Cells/immunology , Tumor Necrosis Factor-alpha/analysis
13.
Cytotechnology ; 33(1-3): 237-45, 2000 Jul.
Article in English | MEDLINE | ID: mdl-19002831

ABSTRACT

Although oral administration of a soluble proteinantigen can induce various immune responses, theeffect of the dosage of oral antigen on thepredominance of Th2-type cytokine and antibodyresponses has not been well clarified yet. In thepresent study, we fed T cell receptor (TCR) transgenic(tg) mice various amounts of chicken ovalbumin (0.1,5, and 250 mg) and examined the resulting immuneresponses to this antigen. In these TCR tg mice, theresponses of antigen-specific T cells were greatlyamplified concomitantly with significantantigen-specific cytokine secretion. We found that ahigh dose (250 mg) of antigen significantlyupregulated the serum antigen-specific IgG1 and IgAantibody responses in mice later intraperitoneallyinjected with antigen plus adjuvant. The miceadministered the same oral dose but not immunizedshowed upregulation of Th2-type IL-4 and IL-5secretion and downregulation of Th1-type IL-2 andIFN-gamma. This enhancement of Th2-type cytokineand antibody responses was more marked when largerdoses of antigen orally administered. These resultsdemonstrated that antigen feeding induces thedevelopment of T cells secreting Th2-type cytokines ina dose-dependent manner and that these T cells have ahelper function for the production of antibodies ofthe Th2-type isotypes. This experimental system shouldbe useful to screen foods and other substances thatcan modulate Th2-type responses relating to allergy.

15.
Clin Immunol Immunopathol ; 88(3): 277-86, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9743615

ABSTRACT

Modulation of CD8(+) T-cell responses specific for an exogenous antigen by epitope variants would be advantageous to develop a novel means of antigen-specific immune regulation. We have analyzed CD8(+) T-cell responses to single amino acid-substituted variants of a peptide corresponding to residues 142-149 (p142-149; LAYFYPEL) of alphas1-casein, a major milk allergen, which is a dominant determinant restricted by H-2Kb. An analog peptide L142I with a substitution of Ile for Leu at the nonanchor N-terminal residue induced more IFN-gamma secretion than p142-149 from specific CD8(+) T cells. Furthermore, L142I could prime CD8(+) T cells more efficiently in vivo, and these L142I-primed cells secreted more IFN-gamma than p142-149-primed CD8(+) T cells upon stimulation with p142-149 in vitro. These findings are mainly explained by the greater ability of L142I to form stable Kb-peptide complexes. These findings indicate that appropriate analog peptides may be useful as efficient inducers of CD8(+) T cells which recognize the parent peptide and secrete IFN-gamma, a potent inhibitor of Th2-dependent events, including IgE production.


Subject(s)
CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Caseins/immunology , Caseins/pharmacology , Epitopes/immunology , Interferon-gamma/biosynthesis , Oligopeptides/immunology , Oligopeptides/pharmacology , Peptide Fragments/immunology , Peptide Fragments/pharmacology , Amino Acid Sequence , Animals , Caseins/metabolism , Cattle , Epitopes/metabolism , Female , H-2 Antigens/immunology , H-2 Antigens/metabolism , Interferon-gamma/immunology , Interferon-gamma/metabolism , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Oligopeptides/chemical synthesis , Peptide Fragments/metabolism , Sequence Homology, Amino Acid , Stimulation, Chemical
16.
Clin Immunol Immunopathol ; 88(1): 70-9, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9683552

ABSTRACT

Collagen-induced arthritis (CIA) is an autoimmune animal model for some types of human rheumatoid arthritis (RA). We have evaluated the effectiveness of intranasal administration of antigen in inhibiting CIA in DBA/1 mice. The intranasal administration of heat-denatured or trypsin-digested bovine type II collagen (CII) before immunization with CII strongly delayed the onset of CIA, whereas administration of native CII did not do so. The mice administered denatured or digested CII possessed much lower titers of anti-CII IgG2a than the control mice, whereas titers of anti-CII IgG1 and IgG2b were unchanged or slightly decreased. Responding to CII and peptides containing immunodominant T cell determinants, lymph node cells from mice administered denatured CII produced less IFN-gamma. These results suggest that intranasal administration of antigen downregulated preferentially Th1-type responses, whereas an enhanced Th2-type response was not observed. We demonstrate that the methods shown here are a possible treatment for rheumatoid arthritis.


Subject(s)
Arthritis/prevention & control , Collagen/administration & dosage , Administration, Intranasal , Amino Acid Sequence , Animals , Arthritis/etiology , Arthritis/immunology , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/therapy , Autoantibodies/blood , Autoimmune Diseases/etiology , Autoimmune Diseases/immunology , Autoimmune Diseases/prevention & control , Cattle , Collagen/chemistry , Collagen/immunology , Disease Models, Animal , Female , Humans , Immunodominant Epitopes/genetics , Interferon-gamma/biosynthesis , Interferon-gamma/metabolism , Mice , Mice, Inbred DBA , Molecular Sequence Data , Peptide Fragments/administration & dosage , Peptide Fragments/chemistry , Peptide Fragments/immunology , Protein Denaturation , Th1 Cells/immunology , Th2 Cells/immunology
17.
Clin Immunol Immunopathol ; 87(3): 282-91, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9646838

ABSTRACT

The participation of each lymphocyte compartment in the induction of oral tolerance for antibody response was investigated by means of a new cell-transfer experimental system, using severe combined immunodeficiency (SCID) mice. Various lymphocyte compartments from BALB/c mice were transferred into SCID mice and these mice were evaluated for oral tolerance induction. First, whole splenocytes from BALB/c mice were transferred into SCID mice and these mice were orally administered bovine alpha s1-casein. The specific antibody response in these mice after subsequent immunization with antigen was greatly reduced compared to controls which were not fed the antigen, and it was demonstrated that oral tolerance was induced in SCID mice bearing donor splenocytes. Oral tolerance was induced in SCID mice that were reconstituted with only T cells, revealing that B cells were not required for the induction of oral tolerance. Further, oral tolerance was induced in SCID mice reconstituted with CD8-depleted splenocytes but not in mice reconstituted with only CD8+ T cells. These results demonstrate that oral tolerance could be induced in SCID mice bearing normal splenocytes and that interaction of CD4+ T cells with antigen-presenting cells other than B cells are responsible for the induction of oral tolerance. Our experimental system may be useful for investigations with human lymphocytes.


Subject(s)
Antigen-Presenting Cells/immunology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immune Tolerance/immunology , Immunotherapy, Adoptive , Administration, Oral , Animals , Antibodies/blood , Antibody Formation/drug effects , Antibody Formation/immunology , Antigens/administration & dosage , Antigens/immunology , Caseins/administration & dosage , Caseins/immunology , Cattle , Female , Lymph Nodes/cytology , Lymph Nodes/immunology , Mice , Mice, Inbred BALB C , Mice, SCID , Spleen/cytology , Spleen/immunology
18.
J Allergy Clin Immunol ; 101(5): 660-71, 1998 May.
Article in English | MEDLINE | ID: mdl-9600504

ABSTRACT

In an effort to clarify the etiology of milk allergy from the standpoint of allergen-specific immune reactions, we investigated the determinants of IgE, IgG4, and T cells specific for bovine alpha(s)1-casein from the same individual patients by using its synthetic peptides and cyanogen bromide-digested fragments. Alpha(s)1-casein is a major allergen in cow's milk, and its unique conformation enabled us to investigate the determinants of antibodies without consideration about missing the reactivities because of conformational changes. Nine patients were selected as subjects from among 129 milk-sensitive infants screened by ELISA to assess the anti-alpha(s)1-casein IgE levels in their sera. By using ELISA for epitope mapping, a C-terminal region of alpha(s)1-casein was identified as a common binding site for IgE from all of these patients, whereas those for anti-alpha(s)1-casein IgG4 were located in multiple regions of alpha(s)1-casein. We determined the specificities of seven alpha(s)1-casein-specific T-cell lines established from peripheral blood mononuclear cells of two of the patients. These T cells have been shown to secrete IL-4. All of the T-cell lines had different specificities to alpha(s)1-casein. However, a common amino acid residue use was found among the determinants of various T-cell lines from each patient. The results suggest that patients allergic to cow's milk have characteristic B cells recognizing a limited region of alpha(s)1-casein and secreting alpha(s)1-casein-specific IgE. These B cells may interact particularly with T cells recognizing determinants with a common structure.


Subject(s)
B-Lymphocytes/immunology , Caseins/immunology , Immunoglobulin E/immunology , Immunoglobulin G/immunology , Milk Hypersensitivity/immunology , T-Lymphocytes, Cytotoxic/immunology , Amino Acid Sequence , Animals , Caseins/chemical synthesis , Caseins/metabolism , Cattle , Cyanogen Bromide/metabolism , Epitope Mapping , Epitopes, T-Lymphocyte/immunology , Humans , Molecular Sequence Data , Peptides/immunology
19.
Int Arch Allergy Immunol ; 115(4): 278-87, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9566350

ABSTRACT

BACKGROUND: Lactobacillus casei is a nonpathogenic gram-positive bacterium widely used in dairy products and has been shown to enhance the cellular immunity of the host. METHODS: To examine the inhibitory effect of L. casei on IgE production, splenocytes obtained from ovalbumin (OVA)-primed BALB/c mice were restimulated in vitro with the same antigen in the presence of heat-killed L. casei. The effect of this bacterium on T helper (Th) phenotype development was also examined with naive T cells from OVA-specific T cell receptor-transgenic mice. RESULTS: L. casei induced IFN-gamma, but inhibited IL-4 and IL-5 secretion, and markedly suppressed total and antigen-specific IgE secretion by OVA-stimulated splenocytes. The inhibitory effect of L. casei on IgE, IL-4, and IL-5 production was partially abrogated by addition of neutralizing antibody to IFN-gamma. Augmented IL-12 production was also observed in the cell cultures containing L. casei, and anti-IL-12 monoclonal antibody completely restored the IgE, IL-4, and IL-5 production to the control levels. The IL-12 augmentation by L. casei was macrophage-dependent. The Th cell development assay showed the ability of L. casei to induce Th1 development preferentially. This effect was also completely blocked by anti-IL-12 antibody. CONCLUSIONS: This is the first demonstration that a nonpathogenic microorganism, L. casei, can inhibit antigen-induced IgE production through induction of IL-12 secretion by macrophages. The findings suggest a potential use of this organism in preventing IgE-mediated allergy.


Subject(s)
Cytokines/biosynthesis , Immunoglobulin E/biosynthesis , Lacticaseibacillus casei/immunology , Spleen/immunology , Allergens/immunology , Animals , Cell Line , Cytokines/immunology , Female , Interferon-gamma/biosynthesis , Interleukin-12/biosynthesis , Interleukin-4/biosynthesis , Interleukin-5/biosynthesis , Macrophages/immunology , Mice , Mice, Inbred BALB C , Ovalbumin , Spleen/metabolism
20.
FEBS Lett ; 423(2): 138-42, 1998 Feb 20.
Article in English | MEDLINE | ID: mdl-9512346

ABSTRACT

The CD8+ T cell clone 5F1 produces interleukin 10 (IL-10) and interferon gamma(IFN-gamma) in response to stimulation with a peptide corresponding to region 142-149 of bovine alpha(s1)-casein (p142-149). Ninety analog peptides derived from p142-149 with single amino acid substitutions of putative T cell receptor contact residues were prepared to examine whether production of IL-10 and IFN-gamma by 5F1 can be altered by stimulation with these peptides. We found that some peptides triggered only IL-10 production whereas others induced production of IFN-gamma alone or both of these cytokines. Peptides inducing IFN-gamma production triggered both cytotoxicity and a proliferative response, whereas peptides inducing production of IL-10 but not IFN-gamma triggered neither of these responses. Our results clearly demonstrate that the signaling pathway required for IL-10 production in CD8+ T cells differs from that required for IFN-gamma production. The distinct cellular signals for IL-10 production appear to be independent of those for cytotoxicity and the proliferative response of CD8+ T cells.


Subject(s)
CD8-Positive T-Lymphocytes/physiology , Interleukin-10/biosynthesis , Peptides/pharmacology , Signal Transduction/drug effects , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Caseins/immunology , Caseins/isolation & purification , Cattle , Female , Interferon-gamma/biosynthesis , Mice , Mice, Inbred C57BL , Peptides/immunology , Point Mutation , T-Lymphocyte Subsets
SELECTION OF CITATIONS
SEARCH DETAIL
...