Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Mucosal Immunol ; 9(6): 1571-1583, 2016 11.
Article in English | MEDLINE | ID: mdl-27007679

ABSTRACT

Depot-medroxyprogesterone acetate (DMPA) is a hormonal contraceptive especially popular in areas with high prevalence of HIV and other sexually transmitted infections (STI). Although observational studies identify DMPA as an important STI risk factor, mechanisms underlying this connection are undefined. Levonorgestrel (LNG) is another progestin used for hormonal contraception, but its effect on STI susceptibility is much less explored. Using a mouse model of genital herpes simplex virus type 2 (HSV-2) infection, we herein found that DMPA and LNG similarly reduced genital expression of the desmosomal cadherin desmoglein-1α (DSG1α), enhanced access of inflammatory cells to genital tissue by increasing mucosal epithelial permeability, and increased susceptibility to viral infection. Additional studies with uninfected mice revealed that DMPA-mediated increases in mucosal permeability promoted tissue inflammation by facilitating endogenous vaginal microbiota invasion. Conversely, concomitant treatment of mice with DMPA and intravaginal estrogen restored mucosal barrier function and prevented HSV-2 infection. Evaluating ectocervical biopsy tissue from women before and 1 month after initiating DMPA remarkably revealed that inflammation and barrier protection were altered by treatment identically to changes seen in progestin-treated mice. Together, our work reveals DMPA and LNG diminish the genital mucosal barrier; a first-line defense against all STI, but may offer foundation for new contraceptive strategies less compromising of barrier protection.


Subject(s)
Contraceptive Agents, Female/pharmacology , Disease Susceptibility , Herpes Genitalis/virology , Herpesvirus 2, Human/drug effects , Levonorgestrel/pharmacology , Medroxyprogesterone Acetate/pharmacology , Mucous Membrane/drug effects , Mucous Membrane/virology , Animals , Disease Models, Animal , Estrogens/pharmacology , Female , Mice , Mucous Membrane/metabolism , Permeability/drug effects , Vagina/drug effects , Vagina/immunology , Vagina/metabolism , Vagina/virology
2.
Mucosal Immunol ; 8(4): 735-45, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25563500

ABSTRACT

Induction of mucosal immunoglobulin-A (IgA) capable of providing a first line of defense against bacterial and viral pathogens remains a major goal of needle-free vaccines given via mucosal routes. Innate immune cells are known to play a central role in induction of IgA responses by mucosal vaccines, but the relative contribution of myeloid cell subsets to these responses has not firmly been established. Using an in vivo model of sublingual vaccination with Bacillus anthracis edema toxin (EdTx) as adjuvant, we examined the role of myeloid cell subsets for mucosal secretory IgA responses. Sublingual immunization of wild-type mice resulted in a transient increase of neutrophils in sublingual tissues and cervical lymph nodes. These mice later developed Ag-specific serum IgG responses, but not serum or mucosal IgA. Interestingly, EdTx failed to increase neutrophils in sublingual tissues and cervical lymph nodes of IKKß(ΔMye) mice, and these mice developed IgA responses. Partial depletion of neutrophils before immunization of wild-type mice allowed the development of both mucosal and serum IgA responses. Finally, co-culture of B cells with neutrophils from either wild-type or IKKß(ΔMye) mice suppressed secretion of IgA, but not IgM or IgG. These results identify a new role for neutrophils as negative regulators of IgA responses.


Subject(s)
Immunity, Mucosal , Immunoglobulin A, Secretory/immunology , Mucous Membrane/immunology , Neutrophils/immunology , Administration, Sublingual , Animals , Antibody Formation , Antigens, Bacterial/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Bacterial Toxins/immunology , I-kappa B Kinase/deficiency , I-kappa B Kinase/metabolism , Immunization , Leukocyte Count , Lymph Nodes/immunology , Mice , Mice, Transgenic , Mucous Membrane/metabolism , Myeloid Cells/immunology , Myeloid Cells/metabolism , Neutrophil Infiltration/immunology , Neutrophils/metabolism , Signal Transduction
3.
Mucosal Immunol ; 7(2): 257-67, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23839064

ABSTRACT

Regulation of allergic responses by intestinal epithelial cells (IECs) remains poorly understood. Using a model of oral allergen sensitization in the presence of cholera toxin as adjuvant and mice with cell-specific deletion of inhibitor-κB kinase (IKKß) in IECs (IKKß(ΔIEC)), we addressed the contribution of IECs to allergic sensitization to ingested antigens and allergic manifestations at distant mucosal site of the airways. Cholera toxin induced higher pro-inflammatory responses and altered the profile of the gut microbiota in IKKß(ΔIEC) mice. Antigen-specific immunoglobulin E (IgE) responses were unaltered in IKKß(ΔIEC) mice, but their IgA antibodies (Abs), T helper type 1 (Th1) and Th17 responses were enhanced. Upon nasal antigen challenge, these mice developed lower levels of allergic lung inflammation, which correlated with higher levels of IgA Abs in the airways. The IKKß(ΔIEC) mice also recruited a higher number of gut-sensitized T cells in the airways after nasal antigen challenge and developed airway hyper-responsiveness, which were suppressed by treatment with anti-interleukin-17A. Fecal microbiota transplant during allergic sensitization reduced Th17 responses in IKKß(ΔIEC) mice, but did not affect IgA Ab responses. In summary, we show that IKKß in IECs shapes the gut microbiota and immune responses to ingested antigens and influences allergic responses in the airways via regulation of IgA Ab responses.


Subject(s)
Allergens/immunology , I-kappa B Kinase/metabolism , Immunoglobulin A/immunology , Inflammation/immunology , Inflammation/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Adjuvants, Immunologic , Allergens/administration & dosage , Animals , Antibody Specificity/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cholera Toxin/immunology , Dysbiosis/immunology , Gene Deletion , I-kappa B Kinase/genetics , Immunity, Innate/genetics , Immunity, Innate/immunology , Immunization , Interleukin-17/biosynthesis , Intestinal Mucosa/pathology , Mice , Respiratory System/immunology , Respiratory System/metabolism , Respiratory System/pathology , Signal Transduction
4.
Biochim Biophys Acta ; 1833(12): 3218-3227, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24080087

ABSTRACT

The deregulation of B cell differentiation has been shown to contribute to autoimmune disorders, hematological cancers, and aging. We provide evidence that the retinoic acid-producing enzyme aldehyde dehydrogenase 1a1 (Aldh1a1) is an oncogene suppressor in specific splenic IgG1(+)/CD19(-) and IgG1(+)/CD19(+) B cell populations. Aldh1a1 regulated transcription factors during B cell differentiation in a sequential manner: 1) retinoic acid receptor alpha (Rara) in IgG1(+)/CD19(-) and 2) zinc finger protein Zfp423 and peroxisome proliferator-activated receptor gamma (Pparg) in IgG1(+)/CD19(+) splenocytes. In Aldh1a1(-/-) mice, splenic IgG1(+)/CD19(-) and IgG1(+)/CD19(+) B cells acquired expression of proto-oncogenic genes c-Fos, c-Jun, and Hoxa10 that resulted in splenomegaly. Human multiple myeloma B cell lines also lack Aldh1a1 expression; however, ectopic Aldh1a1 expression rescued Rara and Znf423 expressions in these cells. Our data highlight a mechanism by which an enzyme involved in vitamin A metabolism can improve B cell resistance to oncogenesis.


Subject(s)
Aldehyde Dehydrogenase/metabolism , B-Lymphocytes/metabolism , Genes, Tumor Suppressor , Aldehyde Dehydrogenase/deficiency , Aldehyde Dehydrogenase 1 Family , Animals , Antigens, CD19/metabolism , B-Lymphocytes/drug effects , B-Lymphocytes/pathology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Models, Biological , Multiple Myeloma/genetics , Multiple Myeloma/immunology , Multiple Myeloma/pathology , PPAR gamma/metabolism , Response Elements/genetics , Retinal Dehydrogenase , Spleen/drug effects , Spleen/pathology , Splenomegaly/immunology , Splenomegaly/pathology , Transcription Factors/metabolism , Tretinoin/pharmacology , Vitamin A/metabolism
5.
Acta Odontol Scand ; 59(5): 301-8, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11680650

ABSTRACT

Induction of mucosal immunity by oral immunization with protein antigen alone is difficult: potent mucosal adjuvants, vectors, or other special delivery systems are required. Cholera toxin (CT) has been shown to be an effective adjuvant for the development of mucosal vaccines and, when given with vaccine, induces both mucosal and systemic immune responses via a Th2 cell-dependent pathway. However, and in addition to potential type-I hypersensitivity, a major concern for use of mucosal adjuvants such as CT is that this molecule is not suitable for use in humans because of its inherent toxicity. When we examined the potential toxicity of CT for the central nervous system, both CT and CT-B accumulated in the olfactory nerves/epithelium and olfactory bulbs of mice when given by the nasal route. The development of effective mucosal vaccines for the elderly is also an important issue; however, only limited information is available. When mucosal adjuvanticity of CT was evaluated in aged mice, an early immune dysregulation was evident in the mucosal immune system. The present review discusses these potential problems for effective mucosal vaccine development. Tolerance represents the most common and important response of the host to environmental antigens, including food and commensal bacterial components, for the maintenance of an appropriate immunological homeostasis. We have examined whether Peyer patches could play a more important role for the maintenance of oral tolerance. Using Peyer patch-null mice, we found that mice lacking this gut-associated lymphoid tissue retained their capability to produce secretory IgA antibodies but did not develop normal oral tolerance to protein antigens.


Subject(s)
Immune Tolerance , Immunity, Mucosal , Immunoglobulin A, Secretory/immunology , Mouth Mucosa/immunology , Administration, Inhalation , Aging/immunology , Animals , Central Nervous System/drug effects , Cholera Toxin/immunology , Cholera Toxin/toxicity , Humans , Mice , Peyer's Patches/immunology , Vaccines, Conjugate/administration & dosage , Vaccines, Conjugate/immunology , Vaccines, Conjugate/toxicity
6.
Adv Drug Deliv Rev ; 51(1-3): 71-9, 2001 Sep 23.
Article in English | MEDLINE | ID: mdl-11516780

ABSTRACT

Safe nasal vaccines capable of promoting both mucosal and systemic immunity are needed for effective protection against bacterial and viral pathogens. While parenteral cytokine treatment could lead to unwanted toxicity, the nasal delivery route results in low but biologically active serum cytokine levels. Interleukin (IL)-6, IL-1 and IL-12, which promote either Th2- or Th1-type responses, respectively, also enhance systemic immunity to co-administered antigens. The chemoattractants lymphotactin (Lptn), RANTES and defensins also exerted adjuvant activity for systemic immunity when nasally administered with antigens. However, each cytokine or innate factor promoted a distinct pattern of T helper cell responses and corresponding IgG subclass response. Interleukin-12, IL-1, and the chemokines Lptn and RANTES promote mucosal immunity. In contrast, nasal IL-6 and defensins failed to induce mucosal S-IgA Ab responses, suggesting that mechanisms more complex than T cell activation and chemotaxis are required for the development of mucosal immunity after nasal delivery of cytokines.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Cytokines/administration & dosage , Nasal Mucosa/immunology , Vaccines/administration & dosage , Administration, Intranasal , Animals , Humans , Immunity, Mucosal , Th1 Cells/immunology , Th2 Cells/immunology
7.
J Immunol ; 166(4): 2283-90, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11160283

ABSTRACT

The highly purified saponin derivative, QS-21, from the Quillaja saponaria Molina tree has been proved to be safe for parenteral administration and represents a potential alternative to bacterial enterotoxin derivatives as a mucosal adjuvant. Here we report that p.o. administration of QS-21 with the vaccine protein tetanus toxoid elicited strong serum IgM and IgG Ab responses, which were only slightly enhanced by further oral immunization. The IgG Ab subclass responses were predominantly IgG1 followed by IgG2b for the 50-microg p.o. dose of QS-21, whereas the 250-microg p.o. dose also induced IgG2a and IgG3 Abs. Low oral QS-21 doses induced transient IgE Ab responses 7 days after the primary immunization, whereas no IgE Ab responses were seen in mice given the higher QS-21 dose. Further, low but not high p.o. QS-21 doses triggered Ag-specific secretory IgA (S-IgA) Ab responses. Th cell responses showed higher IFN-gamma (Th1-type) and lower IL-5, IL-6, and IL-10 (Th2-type) secretion after the high QS-21 p.o. dose than after low doses. Interestingly, the mucosal adjuvant activity of low oral QS-21 doses was diminished in IL-4(-/-) mice, suggesting a role for this cytokine in the initiation of mucosal immunity by oral QS-21. In summary, our results show that oral QS-21 enhances immunity to coadministered Ag and that different doses of QS-21 lead to distinct patterns of cytokine and serum Ab responses. We also show that an early IL-4 response is required for the induction of mucosal immunity by oral QS-21 as adjuvant.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Interleukin-4/physiology , Saponins/administration & dosage , Adjuvants, Immunologic/therapeutic use , Administration, Oral , Animals , Antibodies, Bacterial/biosynthesis , Antibodies, Bacterial/blood , Dose-Response Relationship, Immunologic , Drug Administration Schedule , Immunity, Active , Immunity, Mucosal , Immunoglobulin A, Secretory/biosynthesis , Immunoglobulin G/biosynthesis , Immunoglobulin G/blood , Immunoglobulin M/biosynthesis , Immunoglobulin M/blood , Injections, Subcutaneous , Interleukin-4/deficiency , Interleukin-4/genetics , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Tetanus Toxoid/administration & dosage
8.
J Immunol ; 166(1): 162-9, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11123289

ABSTRACT

RANTES is produced by lymphoid and epithelial cells of the mucosa in response to various external stimuli and is chemotactic for lymphocytes. The role of RANTES in adaptive mucosal immunity has not been studied. To better elucidate the role of this chemokine, we have characterized the effects of RANTES on mucosal and systemic immune responses to nasally coadministered OVA. RANTES enhanced Ag-specific serum Ab responses, inducing predominately anti-OVA IgG2a and IgG3 followed by IgG1 and IgG2b subclass Ab responses. RANTES also increased Ag-specific Ab titers in mucosal secretions and these Ab responses were associated with increased numbers of Ab-forming cells, derived from mucosal and systemic compartments. Splenic and mucosally derived CD4(+) T cells of RANTES-treated mice displayed higher Ag-specific proliferative responses and IFN-gamma, IL-2, IL-5, and IL-6 production than control groups receiving OVA alone. In vitro, RANTES up-regulated the expression of CD28, CD40 ligand, and IL-12R by Ag-activated primary T cells from DO11.10 (OVA-specific TCR-transgenic) mice and by resting T cells in a dose-dependent fashion. These studies suggest that RANTES can enhance mucosal and systemic humoral Ab responses through help provided by Th1- and select Th2-type cytokines as well as through the induction of costimulatory molecule and cytokine receptor expression on T lymphocytes. These effects could serve as a link between the initial innate signals of the host and the adaptive immune system.


Subject(s)
Adjuvants, Immunologic/physiology , Chemokine CCL5/physiology , Epitopes/immunology , Immunity, Mucosal/immunology , Ovalbumin/immunology , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Animals , Antibody-Producing Cells/immunology , Antibody-Producing Cells/metabolism , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Chemokine CCL5/administration & dosage , Cytokines/biosynthesis , Epitopes/administration & dosage , Epitopes, T-Lymphocyte/immunology , Female , Immunoglobulin A/biosynthesis , Immunoglobulin G/biosynthesis , Immunoglobulin G/blood , Immunoglobulin M/biosynthesis , Immunoglobulin M/blood , Interleukin-12 , Interphase/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Nasal Mucosa/immunology , Nasal Mucosa/metabolism , Ovalbumin/administration & dosage , Receptors, Interleukin/biosynthesis , Receptors, Interleukin-12 , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
9.
Cell Mol Biol (Noisy-le-grand) ; 47(7): 1115-20, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11838959

ABSTRACT

Oral immunization with the recombinant Salmonella typhimurium strain (BRD 847) expressing the C fragment of tetanus toxin (TT) induces brisk Ag-specific mucosal S-IgA and serum Ab responses characterized by strong IgG2a Abs to the encoded antigen. We have constructed an attenuated Salmonella typhimurium (aroA- aroD-) strain that expresses chicken egg albumin (OVA) to further elucidate the role of Salmonella-induced Th1 cell phenotype on mucosal cell-mediated immunity (CMI). Peyer's patches and spleen lymphocytes from mice that received the oral Salmonella-OVA vaccine showed dramatic increases in the percent cell lysis of the H-2b restricted EG7.OVA tumor cell line. These results indicate that a single dose of rSalmonella vaccine antigen vector is required to illicit systemic and mucosal Th1-type responses and CTLs. These results also support the existence of a highly regulated relationship between specific cell-mediated immunity and a branch of the humoral immune system, i.e. mucosal IgA responses.


Subject(s)
Immunity, Mucosal/immunology , Salmonella typhimurium/immunology , Th1 Cells/immunology , Animals , Cell Division , Cytokines/metabolism , Female , Immunoglobulin G/immunology , Mice , Mice, Inbred C57BL , Ovalbumin/genetics , Ovalbumin/immunology , Peyer's Patches/immunology , Salmonella typhimurium/genetics , Spleen/immunology , Th1 Cells/cytology , Th1 Cells/metabolism , Th2 Cells/cytology , Th2 Cells/immunology , Th2 Cells/metabolism
10.
Am J Pathol ; 157(6): 2023-35, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11106575

ABSTRACT

Subepithelial and intraepithelial lymphocytes of human adenoids and tonsils were characterized and directly compared to determine the potential contribution of these tissues to mucosal and systemic immune responses. The distribution of T and B cell subsets, cytokine patterns, and antibody (Ab) isotype profiles were similar for adenoids and tonsils. Both tissues contained predominantly B cells ( approximately 65%), approximately 5% macrophages, and 30% CD3(+) T cells. The T cells were primarily of the CD4(+) subset ( approximately 80%). Tonsillar intraepithelial lymphocytes were also enriched in B cells. The analysis of dispersed cells revealed a higher frequency of cells secreting IgG than IgA and the predominant Ig subclass profiles were IgG1 > IgG3 and IgA1 > IgA2, respectively. In situ analysis also revealed higher numbers of IgG- than IgA-positive cells. These IgG-positive cells were present in the epithelium and in the subepithelial zones of both tonsils and adenoids. Mitogen-triggered T cells from tonsils and adenoids produced both Th1- and Th2-type cytokines, clearly exhibiting their pluripotentiality for support of cell-mediated and Ab responses. Interestingly, antigen-specific T cells produced interferon-gamma and lower levels of interleukin-5. These results suggest that adenoids and tonsils of the nasopharyngeal-associated lymphoreticular tissues represent a distinct component of the mucosal-associated lymphoreticular tissues with features of both systemic and mucosal compartments.


Subject(s)
Adenoids/physiology , B-Lymphocytes/physiology , Nasopharynx/physiology , Palatine Tonsil/physiology , T-Lymphocytes/physiology , Adenoids/cytology , Adenoids/immunology , Adolescent , Antibodies/analysis , Antibody Formation , Antigens/immunology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Division/drug effects , Cell Division/physiology , Child , Cytokines/biosynthesis , Epithelial Cells/physiology , Humans , Immunoglobulin A/analysis , Immunoglobulin G/analysis , Immunoglobulin Isotypes/analysis , Mitogens/pharmacology , Monocytes/cytology , Palatine Tonsil/cytology , Palatine Tonsil/immunology , Phytohemagglutinins/pharmacology , T-Lymphocyte Subsets/cytology , T-Lymphocytes/immunology
11.
J Clin Invest ; 105(3): 377-86, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10675364

ABSTRACT

The CFTR Cl(-) channel controls salt and water transport across epithelial tissues. Previously, we showed that CFTR-mediated Cl(-) currents in the Xenopus oocyte expression system are inhibited by syntaxin 1A, a component of the membrane trafficking machinery. This negative modulation of CFTR function can be reversed by soluble syntaxin 1A peptides and by the syntaxin 1A binding protein, Munc-18. In the present study, we determined whether syntaxin 1A is expressed in native epithelial tissues that normally express CFTR and whether it modulates CFTR currents in these tissues. Using immunoblotting and immunofluorescence, we observed syntaxin 1A in native gut and airway epithelial tissues and showed that epithelial cells from these tissues express syntaxin 1A at >10-fold molar excess over CFTR. Syntaxin 1A is seen near the apical cell surfaces of human bronchial airway epithelium. Reagents that disrupt the CFTR-syntaxin 1A interaction, including soluble syntaxin 1A cytosolic domain and recombinant Munc-18, augmented cAMP-dependent CFTR Cl(-) currents by more than 2- to 4-fold in mouse tracheal epithelial cells and cells derived from human nasal polyps, but these reagents did not affect CaMK II-activated Cl(-) currents in these cells.


Subject(s)
Antigens, Surface/biosynthesis , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Epithelial Cells/metabolism , Nerve Tissue Proteins/biosynthesis , Respiratory System/metabolism , Animals , Cells, Cultured , Chloride Channels/metabolism , Humans , Ion Transport , Mice , Syntaxin 1 , Xenopus
12.
Am J Trop Med Hyg ; 60(4 Suppl): 35-45, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10344675

ABSTRACT

Vaccines able to induce both secretory IgA for protection of mucosal surfaces and systemic immunity to pathogens invading the host are of great interest in the war against infectious diseases. Mucosal vaccines trigger immune cells in mucosal inductive sites and thus can induce immunity in both the mucosal and systemic compartments. This review presents a critical survey of adjuvants and delivery systems currently being tested for mucosal immunization. A better understanding of cellular and molecular factors involved in the regulation of mucosal immunity will help in the design of safer mucosal vaccines to elicit the appropriate protective immune response to a given pathogen.


Subject(s)
Immunity, Mucosal , Immunoglobulin A, Secretory/biosynthesis , Intestines/immunology , Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Administration, Oral , Animals , Cytokines/immunology , Epithelial Cells/immunology , Humans , Interleukin-12/immunology , Lymphocytes/immunology , Vaccines/administration & dosage
13.
J Immunol ; 162(4): 1959-65, 1999 Feb 15.
Article in English | MEDLINE | ID: mdl-9973465

ABSTRACT

Lymphotactin (Lptn) is a C chemokine produced predominantly by NK and CD8-positive (CD8+) T cells including gammadelta TCR-positive (TCR+) intraepithelial lymphocytes. Lptn is chemotactic for NK and T cells and likely plays an important role in maintaining the integrity of the epithelium and in mucosal immune responses. In this study, we characterized the immune responses to OVA given intranasally with Lptn to mice. This regimen enhanced OVA-specific serum Ab responses and Ab titers in mucosal secretions. Lptn also enhanced OVA-specific Ab-forming cells in mucosal and systemic compartments. CD4-positive (CD4+) T cells isolated from mucosal compartments and spleens of mice intranasally immunized with OVA plus Lptn displayed higher OVA-specific proliferative responses and greater synthesis of IFN-gamma, IL-2, IL-4, IL-5, IL-6, and IL-10 than did CD4+ T cells from mice given OVA without Lptn. These studies indicate that Lptn has adjuvant properties and suggest that Lptn present in the mucosa has the potential to enhance mucosal and systemic Ab responses through help provided by Th1- and Th2-type cells to link the initial innate signals of the mucosa with the acquired immune system.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Chemokines, C , Lymphokines/administration & dosage , Lymphokines/immunology , Nasal Mucosa/immunology , Sialoglycoproteins/administration & dosage , Sialoglycoproteins/immunology , Adjuvants, Immunologic/physiology , Administration, Intranasal , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Dose-Response Relationship, Immunologic , Epitopes, T-Lymphocyte/analysis , Female , Immunity, Mucosal , Immunoglobulin A/biosynthesis , Immunoglobulin G/biosynthesis , Lymphokines/physiology , Mice , Mice, Inbred C57BL , Nasal Mucosa/metabolism , Ovalbumin/administration & dosage , Ovalbumin/immunology , Sialoglycoproteins/physiology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism
14.
Proc Natl Acad Sci U S A ; 96(2): 651-6, 1999 Jan 19.
Article in English | MEDLINE | ID: mdl-9892688

ABSTRACT

Human neutrophil peptide (HNP) defensins were studied to determine their potential effects on adaptive mucosal immunity. Intranasal delivery of HNPs plus ovalbumin (OVA) enhanced OVA-specific serum IgG antibody (Ab) responses. However, OVA-specific IgA Abs were not induced in mucosal secretions or in serum. CD4(+) T cells of intranasally immunized mice displayed higher OVA-specific proliferative responses and elevated production of interferon gamma, interleukin (IL) 5, IL-6, and IL-10 when compared with control groups receiving OVA alone. In vitro, HNPs also enhanced both proliferative responses and T helper (Th) cytokine secretion profiles of CD3epsilon-stimulated spleen- and Peyer's patch-derived naive CD4(+) T cells. HNPs modulated the expression of costimulatory molecules by lipopolysaccharide- or CD3epsilon-stimulated splenic and Peyer's patch B or T cell populations, respectively. These studies show that defensins enhance systemic IgG, but not IgA, Ab responses through help provided by CD4(+) Th1- and Th2-type cytokines and foster B and T cell interactions to link innate immunity with the adaptive immune system.


Subject(s)
CD3 Complex , Immunity , Neutrophils/immunology , Proteins/immunology , Administration, Intranasal , Animals , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Cytokines/immunology , Defensins , Female , Humans , Immunoglobulin G/blood , Kinetics , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred Strains , Nasal Mucosa/immunology , Ovalbumin/immunology , Proteins/pharmacology , Receptors, Antigen, T-Cell/immunology , Spleen/immunology , Th1 Cells/immunology , Th2 Cells/immunology
15.
J Immunol ; 162(1): 114-21, 1999 Jan 01.
Article in English | MEDLINE | ID: mdl-9886376

ABSTRACT

We have investigated the effects of IL-12 and cholera toxin (CT) on the immune response to tetanus toxoid (TT) given by intranasal or oral routes. CT inhibited IL-12-induced IFN-gamma secretion both in vivo and in vitro. Intranasal administration of IL-12 to mice nasally immunized with the combined vaccine of TT and CT resulted in increased TT-specific IgG2a and IgG3 Abs, while IgG1 and IgE Ab responses were markedly reduced. This shift of the CT-induced immune response toward Th1 type was associated with TT-specific CD4+ T cells secreting IFN-gamma and reduced levels of Th2-type cytokines (i.e., IL-4, IL-5, IL-6, and IL-10). In contrast, intranasal IL-12 enhanced the CT-induced serum IgG1 and IgE Ab responses in mice given the combined vaccine orally. IFN-gamma secretion by TT-specific CD4+ T cells was also enhanced; however, Th2-type cytokine responses were predominant. Mucosal secretory IgA responses to oral or nasal vaccines were not affected by intranasal IL-12. Thus, intranasal IL-12 delivery influences Th cell subset development in mucosal inductive sites that are dependent on the route of vaccine delivery.


Subject(s)
Cholera Toxin/immunology , Interleukin-12/administration & dosage , Nasal Mucosa/immunology , Tetanus Toxoid/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Administration, Intranasal , Administration, Oral , Animals , Antibodies, Bacterial/biosynthesis , CD4 Antigens/immunology , Cells, Cultured , Cholera Toxin/administration & dosage , Cytokines/metabolism , Epitopes, T-Lymphocyte/immunology , Interferon-gamma/biosynthesis , Interleukin-12/immunology , Liposomes/administration & dosage , Liposomes/immunology , Mice , Mice, Inbred C57BL , Nasal Mucosa/metabolism , Spleen/cytology , Spleen/immunology , Spleen/metabolism , Tetanus Toxoid/administration & dosage , Th1 Cells/metabolism , Th2 Cells/metabolism , Vaccines, Combined/administration & dosage , Vaccines, Combined/immunology
16.
J Immunol ; 162(1): 122-8, 1999 Jan 01.
Article in English | MEDLINE | ID: mdl-9886377

ABSTRACT

We addressed the effects of two cytokines, IL-6 and IL-12, derived from APCs, for the development of mucosal IgA Ab responses following their nasal delivery with the protein vaccine tetanus toxoid (TT). Mice treated nasally with IL-6 and TT showed higher TT-specific serum IgG (mainly IgG1 and IgG2b) Ab responses than did control mice, but exhibited no IgE and negligible secretory IgA (S-IgA) Ab responses. In contrast, IL-12 administered nasally with TT not only induced sharp increases in TT-specific serum IgG (mainly IgG1 and IgG2b) and IgA, but also elevated mucosal S-IgA Ab responses. Coadministration of IL-6 and IL-12 with TT did not enhance the mucosal or serum Ab responses over those seen with IL-12 alone. TT-specific CD4+ T cells from mice given TT with IL-6 or IL-12 produced higher levels of IFN-gamma, IL-6, and IL-10 than did those from control mice, but only negligible levels of IL-4 and IL-5. In summary, both intranasal IL-6 and IL-12 induced serum Abs that protected mice from systemic challenge with TT, whereas only IL-12 induced mucosal S-IgA Ab responses. The significance of IL-12-induced Th1-type responses for regulation of both mucosal and systemic immunity is discussed.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Interleukin-12/administration & dosage , Interleukin-12/immunology , Nasal Mucosa/immunology , Adjuvants, Immunologic/therapeutic use , Administration, Intranasal , Animals , Antibodies, Bacterial/biosynthesis , CD4-Positive T-Lymphocytes/metabolism , Cytokines/biosynthesis , Immunity, Mucosal/immunology , Immunoglobulin A, Secretory/biosynthesis , Interleukin-12/therapeutic use , Interleukin-6/administration & dosage , Mice , Mice, Inbred C57BL , Tetanus/immunology , Tetanus/prevention & control , Tetanus Toxin/administration & dosage , Tetanus Toxoid/administration & dosage , Tetanus Toxoid/immunology
17.
Immunol Res ; 20(3): 207-17, 1999.
Article in English | MEDLINE | ID: mdl-10741861

ABSTRACT

Recent strategies for understanding the mechanisms underlying mucosal immune responses and subsequent development of mucosal vaccines for induction of targeted immunity now include cytokines and molecules of innate immunity. These studies have shown that cytokines influencing the development of T helper (Th) cells differentially affect the outcome of mucosal vs. systemic immune responses to mucosal vaccines. Serum antigen-specific antibody (Ab) responses were enhanced when either IL-6 or IL-12 was mucosally administered with a protein antigen, while only IL-12 induced antigen-specific mucosal IgA Ab responses. Mucosal IL-6 and IL-12 also affected the type of Th cell responses induced by CD4+ T cells from mice that received IL-12 secreted larger amounts of IFN-gamma and IL-6 when compared with mice nasally treated with IL-6. Discrepancies in the ability to enhance mucosal or systemic immune responses were also observed when human neutrophil peptide (HNP) defensins or lymphotactin were nasally coadministered with protein antigens. Only lymphotactin promoted mucosal secretory IgA (S-IgA) Ab responses while both lymphotactin and defensins enhanced systemic immunity to mucosally co-administered protein antigens. Mixed antigen-specific Th1 -and Th2-type CD4+ T cell responses were induced in the systemic compartment by both lymphotactin and the mixture of HNP-1, HNP-2, and HNP-3 defensins. However, HNPs failed to significantly enhance cytokine secretion by mucosally derived, antigen-specific CD4+ T cells relative to those isolated from the systemic compartment. In summary, these studies clearly show that IL-12 and lymphotactin are able to trigger S-IgA Ab responses and provide new avenues for the design of safe and targeted mucosal vaccines.


Subject(s)
Adjuvants, Immunologic/pharmacology , Chemokines, C , Immunity, Mucosal/drug effects , Immunization , Interleukin-12/pharmacology , alpha-Defensins , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Defensins , Humans , Immunity, Cellular/drug effects , Immunoglobulin A, Secretory/analysis , Interleukin-12/administration & dosage , Interleukin-12/immunology , Interleukin-6/administration & dosage , Interleukin-6/immunology , Interleukin-6/pharmacology , Lymphokines/administration & dosage , Nasal Mucosa/drug effects , Nasal Mucosa/immunology , Proteins/administration & dosage , Sialoglycoproteins/administration & dosage , Tetanus Toxoid/administration & dosage , Th1 Cells/drug effects , Th1 Cells/immunology , Th2 Cells/drug effects , Th2 Cells/immunology , Vaccines/administration & dosage
18.
Antimicrob Agents Chemother ; 42(10): 2607-11, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9756763

ABSTRACT

The mechanism of intestinal secretion of the difluorinated quinolone sparfloxacin was investigated with the epithelial cell line Caco-2 and was compared to that of the P-glycoprotein (P-gp) substrate vinblastine. The P-gp inhibitors verapamil and progesterone significantly increased the epithelial cell accumulation of both vinblastine and sparfloxacin. This increase is likely to result from an inhibition of drug secretion since both vinblastine uptake and sparfloxacin uptake are known to proceed through a passive transmembrane diffusion. The unidirectional fluxes across cell monlayers grown on permeable filters indicated that a net secretion of sparfloxacin and vinblastine occurred across Caco-2 cells. These secretions were significantly inhibited by the MDR-reversing agent verapamil. We conclude that the P-gp is likely to be involved in the intestinal elimination of the difluorinated quinolone sparfloxacin.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Anti-Infective Agents/metabolism , Fluoroquinolones , Intestinal Mucosa/metabolism , Quinolones/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Animals , Caco-2 Cells , Humans , Rats , Vinblastine/pharmacokinetics
19.
Gastroenterology ; 115(4): 866-73, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9753489

ABSTRACT

BACKGROUND & AIMS: Intraepithelial lymphocytes (IELs) are located between epithelial cells that are thought to display unique features and functions at the small intestinal villus tip and crypt levels. We have addressed whether the spatial differences in the intestinal epithelium extend to IELs and subsequent cross-talk between IELs and epithelial cells. METHODS: IELs were isolated from villus tip and crypt portions of mouse small intestine and then compared for spontaneous cytokine production and responsiveness to interleukin (IL)-2 and/or IL-7. RESULTS: No difference was observed between number of beta IELs in villus tips and crypts, whereas a trend toward increased frequencies of IELs bearing the gamma delta form of T-cell receptor was noted in villus tips. Interestingly, the number of beta IELs producing interferon gamma and IL-5 was significantly reduced in the cells from crypts compared with villus tips. Furthermore, villus tip beta IELs exhibited higher responses to stimulation signals provided by IL-2 and/or IL-7 than their crypt counterpart. Such functional differences were not observed with gamma delta IELs from the two intestinal sites. CONCLUSIONS: Distinct molecular cross-talk between IELs and epithelial cells occurs in intestinal villus tips and crypts.


Subject(s)
Epithelial Cells/cytology , Intestine, Small/cytology , Lymphocytes/cytology , Animals , Cell Cycle/physiology , Cytokines/genetics , Cytokines/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Interleukin-2/pharmacology , Interleukin-7/pharmacology , Intestine, Small/drug effects , Intestine, Small/metabolism , Lymphocytes/drug effects , Lymphocytes/metabolism , Mice , Mice, Inbred C3H , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocytes/metabolism
20.
J Virol ; 72(5): 4534-6, 1998 May.
Article in English | MEDLINE | ID: mdl-9557755

ABSTRACT

We have investigated the mechanisms involved in the clearance of viral infection at the epithelium level by analyzing the activity of influenza virus-specific cytotoxic T lymphocytes (CTL) against virus-infected CMT-93 intestinal epithelial cells. Epithelial cells infected with live influenza virus effectively present viral antigens and were lysed by both homotypic and heterotypic influenza virus-specific CD8+ T cells. These results shed new light on the control of viral infection through the elimination of virus-infected epithelial cells by virus-specific CTL and demonstrate that CMT-93 cells furnish an appropriate model for in vitro evaluation of CTL activity against virus-infected epithelial cells.


Subject(s)
Antigens, Viral/immunology , Epithelial Cells/immunology , Influenza A virus/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Epithelial Cells/virology , Guinea Pigs , Humans , Mice , Mice, Inbred C57BL , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...