Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Vaccine ; 33(15): 1865-72, 2015 Apr 08.
Article in English | MEDLINE | ID: mdl-25698486

ABSTRACT

Chlamydia abortus (C. abortus) is the causative agent of ovine enzootic abortion (OEA) and poses a zoonotic risk to pregnant women. Current live attenuated 1B vaccines are efficacious but cause disease in vaccinated animals and inactivated vaccines are only marginally protective. We tested the ability of a new C. abortus subunit vaccine candidate based on the conserved and immunogenic polymorphic membrane protein D (Pmp18D) formulated in CpG1826+FL (Fms-like tyrosine kinase 3 Ligand; Flt3L) or Vibrio cholerae ghosts (VCG) to induce innate and cross protective immunity against genital C. abortus infection. We found that delivery of rPmp18D with VCG was more effective than with CpG+FL in up-regulating the expression of molecules critically involved in T cell activation and differentiation, including MHC II, CD40, CD80, and CD86, activation of TLRs and NLRP3 inflammasome engagement, and secretion of IL-1ß and TNF-α but not IL-10 and IL-4. rVCG-Pmp18D-immunized mice elicited more robust antigen-specific IFN-γ, IgA and IgG2c antibody responses compared to CpG+FL-delivered rPmp18D. Based on the number of mice with positive vaginal cultures, length of vaginal shedding, and number of inclusion forming units recovered following challenge with the heterologous C. abortus strain B577, vaccine delivery with VCG induced superior protective immunity than delivery with a combination of CpG1826 and FL, a nasal DC-targeting adjuvant. These results demonstrate that the ability of VCG to enhance protective immunity against genital C. abortus infection is superior to that of CpG+FL adjuvants.


Subject(s)
Bacterial Vaccines/immunology , Chlamydia Infections/prevention & control , Chlamydia/immunology , Cross Protection , Vibrio cholerae/immunology , Adjuvants, Immunologic , Administration, Intranasal , Animals , Antibodies, Bacterial/immunology , Chlamydia Infections/immunology , Chlamydia Infections/microbiology , Dendritic Cells/immunology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Immunity, Innate , Mice , Mice, Inbred C57BL , Oligodeoxyribonucleotides/immunology , T-Lymphocytes/immunology , Toll-Like Receptors/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Vagina/microbiology , Vibrio cholerae/isolation & purification
2.
Vaccine ; 29(21): 3802-10, 2011 May 12.
Article in English | MEDLINE | ID: mdl-21421002

ABSTRACT

The need to simultaneously target infections with epidemiological overlap in the population with a single vaccine provides the basis for developing combination vaccines. Vibrio cholerae ghosts (rVCG) offer an attractive approach for developing vaccines against a number of human and animal pathogens. In this study, we constructed a multisubunit vaccine candidate co-expressing the serovar D-derived Porin B and polymorphic membrane protein-D proteins of Chlamydia trachomatis and evaluated its ability to simultaneously induce broad-based chlamydial immunity and elicit a vibriocidal antibody response to the Vibrio carrier envelope. Intramuscular (IM) immunization with the vaccine candidate elicited high levels of antigen-specific genital mucosal and systemic Th1 cell-mediated and humoral immune responses against heterologous serovars and strains, including serovars E-H and L. Also, in addition to the multisubunit vaccine, the single subunit constructs conferred significant cross protection against the heterologous mouse strain, Chlamydia muridarum. Furthermore, all mice immunized with rVCG vaccine constructs responded with a significant rise in vibriocidal antibody titer, the surrogate marker for protection in cholera. These findings demonstrate the ability of the multisubunit vaccine to induce cross protective chlamydial as well as vibriocidal immunity and establish the possibility of developing a broadly efficacious Chlamydia-cholera combination vaccine.


Subject(s)
Antibodies, Bacterial/blood , Bacterial Proteins/immunology , Bacterial Vaccines/administration & dosage , Chlamydia Infections/prevention & control , Cholera/prevention & control , Drug Evaluation , Porins/immunology , Animals , Antibody Formation , Bacterial Vaccines/immunology , Chlamydia Infections/immunology , Chlamydia trachomatis/immunology , Cholera/immunology , Cross Protection , Cytokines/immunology , Female , Genetic Vectors , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Vaccines, Combined/administration & dosage , Vaccines, Combined/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Vibrio cholerae/immunology
3.
Vaccine ; 29(7): 1472-80, 2011 Feb 04.
Article in English | MEDLINE | ID: mdl-21184858

ABSTRACT

We tested the hypothesis that intramuscular immunization with a multisubunit chlamydial vaccine candidate will induce long lasting immune responses in mice. Accordingly, groups of female C57BL/6 mice were immunized intramuscularly with Vibrio cholerae ghosts (VCG) expressing the Poring B and polymorphic membrane protein-D proteins of Chlamydia trachomatis or a control antigen. Humoral and cell-mediated immune responses were evaluated following immunization and after live chlamydial infection. Immunization induced an anamnestic response characterized by chlamydial-specific IgG2a and IgA antibodies in sera and vaginal lavage as well as specific genital and splenic T cell responses. The results also revealed that the local mucosal and systemic cellular and humoral immune effectors induced in mice following immunization with the vaccine candidate are long lasting. Vaccinated mice cleared intravaginal challenge with 10(5) chlamydial inclusion forming units within 12 days compared to control mice, which shed up to 2 × 10(3) IFUs at this time point. Moreover, rechallenge of mice 98 days after resolution of the primary infection resulted in the recall and retention of a relatively high frequency of chlamydial-specific Th1 cells and IgG2a in the genital mucosa. These results provide the first evidence that a VCG-based multisubunit chlamydial vaccine is capable of effectively stimulating anamnestic systemic and mucosal immune responses in mice. The data support further vaccine evaluation and testing for induction of long-term protective immunity.


Subject(s)
Bacterial Proteins/immunology , Bacterial Vaccines/immunology , Chlamydia Infections/prevention & control , Immunologic Memory , Membrane Proteins/immunology , Porins/immunology , Animals , Antibodies, Bacterial/blood , Cell Proliferation , Chlamydia Infections/immunology , Chlamydia trachomatis/immunology , Female , Immunity, Cellular , Immunity, Humoral , Immunoglobulin A/blood , Immunoglobulin A/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Injections, Intramuscular , Mice , Mice, Inbred C57BL , Spleen/immunology , Th1 Cells/immunology , Vagina/immunology , Vibrio cholerae/immunology , Virus Shedding
4.
Drugs Today (Barc) ; 45 Suppl B: 51-9, 2009 Nov.
Article in English | MEDLINE | ID: mdl-20011695

ABSTRACT

The integrated approach to the study of female genital tract infection (GTI) with Chlamydia trachomatis is a conceptual framework through which a consistent and comprehensive evidence-based understanding of C. trachomatis GTI could evolve. One application of this approach has been to identify candidate genes that may play a role in the course and severity of C. trachomatis GTI in women, using human clinical and genetic data together with results obtained in the female mouse model to guide the selection process This model has been proven robust enough: i) to identify stable phenotypic differences in the course and outcome of GTI among commonly used immunocompetent inbred mouse stains that are used in the construction of gene knockout (KO) and transgenic mice; as well as ii) to serve as a platform in which to assess the influence of genetic differences among human genital tract isolates of C. trachomatis as well as between this biovar and the mouse biovar, Chlamydia muridarum. This review presents a summary of published and unpublished results from 25 years of studies in immunodeficient and gene-deficient KO mice that both inform our present understanding of the immunogenetics of C. trachomatis GTI and serve to guide candidate gene selection.


Subject(s)
Chlamydia Infections/genetics , Chlamydia trachomatis , Disease Models, Animal , Genital Diseases, Female/genetics , Animals , Chlamydia Infections/etiology , Chlamydia Infections/immunology , Female , Genital Diseases, Female/etiology , Genital Diseases, Female/immunology , Mice , Mice, Knockout
5.
West Indian med. j ; 56(6): 487-490, Dec. 2007. tab
Article in English | LILACS | ID: lil-507260

ABSTRACT

Mixed lymphocyte responses assays were conducted at 25.0 and 250.0 microg/mL of the crude ethanolic extract of Boehmeria jamaicensis Urb (coded as BJE) using peripheral lymphocytes obtained from individuals suffering from the common cold after four days of infection and from healthy individuals (without the common cold infection). At a concentration of 25 ug/mL, gamma interferon (IFN-gamma) was increased by 24.03 fold and interleukin 4 (IL-4) by 1.71 fold for the cells obtained from individuals with the common cold (Group A). The extract suppressed IFN-gamma by 8.3% while IL-4 was stimulated by 9.90 fold from peripheral lymphocytes obtained from healthy individuals (Group B). Gamma interferon was suppressed at 250 microg/mL while IL-4 was elevated by 1.86 fold for cells obtained from individuals suffering from the common cold (Group A). In conclusion, BJE could have implications for the treatment of the common cold.


Ensayos de reacci¨®n linfocitaria mixta fueron realizados a 25.0 y 250.0 ¦Ìg/mL de extracto etan¨®lico crudo de Boehmeria jamaicensis Urb (codificado como BJE), usando linfocitos perif¨¦ricos obtenidos de individuos con catarro com¨²n luego de cuatro d¨ªas de infecci¨®n, y de individuos sanos (sin la infecci¨®n del catarro com¨²n). Se hall¨® que el interfer¨®n-gamma (IFN-¦Ã) aument¨® en 24.03 veces, y la interleucina 4 (IL-4) en 1.71 veces para las c¨¦lulas obtenidas de individuos con catarro com¨²n, a 25¦Ìg/mL. El extracto inhibi¨® IFN-¦Ã en un 8.3 % en tanto que el IL-4 fue estimulado en 9.90 veces a partirde los linfocitos perif¨¦ricos obtenidos de individuos sanos. El gamma-interfer¨®n fue inhibido a 250 ¦Ìg/mL, mientras que la IL-4 se elev¨® en 1.86 veces para las c¨¦lulas obtenidas de individuos que sufren de catarro com¨²n.


Subject(s)
Humans , Boehmeria , Phytotherapy/methods , Interferon-gamma/immunology , /immunology , Common Cold/immunology , Common Cold/therapy , Sinusitis/immunology , Sinusitis/therapy , Sinusitis/microbiology
6.
West Indian Med J ; 56(6): 487-90, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18646490

ABSTRACT

Mixed lymphocyte responses assays were conducted at 25.0 and 250.0 microg/mL of the crude ethanolic extract of Boehmeria jamaicensis Urb (coded as BJE) using peripheral lymphocytes obtained from individuals suffering from the common cold after four days of infection and from healthy individuals (without the common cold infection). At a concentration of 25 ug/mL, gamma interferon (IFN-gamma) was increased by 24.03 fold and interleukin 4 (IL-4) by 1.71 fold for the cells obtained from individuals with the common cold (Group A). The extract suppressed IFN-gamma by 8.3% while IL-4 was stimulated by 9.90 fold from peripheral lymphocytes obtained from healthy individuals (Group B). Gamma interferon was suppressed at 250 microg/mL while IL-4 was elevated by 1.86 fold for cells obtained from individuals suffering from the common cold (Group A). In conclusion, BJE could have implications for the treatment of the common cold.


Subject(s)
Bacterial Infections/immunology , Bacterial Infections/therapy , Boehmeria , Common Cold/immunology , Common Cold/therapy , Interferon-gamma/immunology , Interleukin-4/immunology , Phytotherapy/methods , Sinusitis/immunology , Sinusitis/therapy , Bacterial Infections/complications , Humans , Sinusitis/microbiology
7.
Infect Immun ; 69(3): 1832-40, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11179361

ABSTRACT

Immunization(s) fostering the induction of genital mucosa-targeted immune effectors is the goal of vaccines against sexually transmitted diseases. However, it is uncertain whether vaccine administration should be based on the current assumptions about the common mucosal immune system. We investigated the relationship between mucosal sites of infection, infection-induced inflammation, and immune-mediated bacterial clearance in mice using the epitheliotropic pathogen Chlamydia trachomatis. Chlamydial infection of the conjunctival, pulmonary, or genital mucosae stimulated significant changes in tissue architecture with dramatic up-regulation of the vascular addressin, VCAM, a vigorous mixed-cell inflammatory response with an influx of alpha4beta1+ T cells, and clearance of bacteria within 30 days. Conversely, intestinal mucosa infection was physiologically inapparent, with no change in expression of the local MAdCAM addressin, no VCAM induction, no histologically detectable inflammation, and no tissue pathology. Microbial clearance was complete within 60 days in the small intestine but bacterial titers remained at high levels for at least 8 months in the large intestine. These findings are compatible with the notion that VCAM plays a functional role in recruiting cells to inflammatory foci, and its absence from the intestinal mucosa contributes to immunologic homeostasis at that site. Also, expression of type 1 T cell-mediated immunity to intracellular Chlamydia may exhibit tissue-specific variation, with the rate and possibly the mechanism(s) of clearance differing between enteric and nonenteric mucosae. The implications of these data for the common mucosal immune system and the delivery of vaccines against mucosal pathogens are discussed.


Subject(s)
Chlamydia Infections/immunology , Chlamydia trachomatis/immunology , Mucous Membrane/immunology , Animals , Antigens, Surface/isolation & purification , B-Lymphocytes/immunology , Cell Adhesion Molecules/isolation & purification , Conjunctiva/immunology , Epithelial Cells/immunology , Female , Inflammation , Integrins/isolation & purification , Intestinal Mucosa/immunology , Membrane Proteins , Mice , Mice, Inbred C57BL , Respiratory Mucosa/immunology , T-Lymphocytes/immunology , Urogenital System/immunology
8.
Cell Mol Biol (Noisy-le-grand) ; 47(6): 981-6, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11785664

ABSTRACT

An essential component of the HIV-1 life cycle involves insertion in the genome of an infected cell. The site of HIV-1 integration has the potential to disrupt a gene and perturb a normal cellular function. To begin to address whether disease pathogenesis may correlate with the site of insertion, flanking cellular sequences at these HIV integrated regions were directly amplified from peripheral blood mononuclear cells DNA from a broad range of infected individuals using an inverse polymerase chain reaction strategy. Amplified flanking regions were sequenced and examined for similarity to the nucleic acid database. In this group of analyzed samples, the HIV-1 provirus was inserted within non-coding regions throughout the genome of the infected host, in which 7/14 sites were positioned in close proximity to different Alu repetitive elements while 2/14 sites were located within intron sequences. Insertions were also detected at sites without a specific gene designation but not within short tandem repetitive sequences, telomeres or centromeric repeat regions. Altogether, it is expected that this approach will yield new information on sites of integration by HIV-1 that may be associated with the pathogenic manifestations of disease progression.


Subject(s)
Blood/virology , DNA, Viral/analysis , HIV Infections/virology , HIV-1/genetics , Lymphocytes/virology , Virus Integration , Base Sequence , Chromosomes, Human, Pair 22 , Chromosomes, Human, Pair 6 , HIV-1/isolation & purification , Humans , Introns , Molecular Sequence Data , Proviruses/genetics , Proviruses/isolation & purification , Sequence Alignment , Sequence Analysis, DNA
9.
Infect Immun ; 68(12): 6798-806, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11083798

ABSTRACT

The significance of delivery systems in modern vaccine design strategies is underscored by the fact that a promising vaccine formulation may fail in vivo due to an inappropriate delivery method. We evaluated the immunogenicity and efficacy of a candidate vaccine comprising the major outer membrane protein (MOMP) of Chlamydia trachomatis delivered with the lipophilic immune response-stimulating complexes (ISCOMs) as a vehicle with adjuvant properties, in a murine model of chlamydial genital infection. Immunocompetent BALB/c mice were immunized intranasally (IN) or intramuscularly (IM) with MOMP, MOMP-ISCOMs, and live or heat-inactivated C. trachomatis serovar D. The level of local genital mucosal Th1 response was measured by assaying for antigen-specific Th1 cell induction and recruitment into the genital mucosa at different times after immunization. Immunization with MOMP-ISCOMs by the IM route induced the greatest and fastest local genital mucosal Th1 response, first detectable 2 weeks after exposure. Among the other routes and regimens tested, only IN immunization with MOMP-ISCOMs induced detectable and statistically significant levels of local genital mucosal Th1 response during the 8-week test period (P < 0.001). In addition, when T cells from immunized mice were adoptively transferred into syngeneic naive animals and challenged intravaginally with Chlamydia, recipients of IM immunization of MOMP-ISCOMs cleared their infection within 1 week and were resistant to reinfection. Animals that received IN immunization of MOMP-ISCOMs were partially protected, shedding fewer chlamydiae than did control mice. Altogether, the results suggested that IM delivery of MOMP-ISCOMs may be a suitable vaccine regimen potentially capable of inducing protective mucosal immunity against C. trachomatis infection.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Bacterial Outer Membrane Proteins/immunology , Bacterial Vaccines/immunology , Chlamydia Infections/prevention & control , Chlamydia trachomatis/immunology , Genital Diseases, Female/prevention & control , ISCOMs/administration & dosage , Porins , Adoptive Transfer , Animals , Bacterial Outer Membrane Proteins/administration & dosage , Female , Humans , Immunization , Mice , Th1 Cells/immunology
10.
J Immunol ; 164(8): 4212-9, 2000 Apr 15.
Article in English | MEDLINE | ID: mdl-10754317

ABSTRACT

A new paradigm for designing vaccines against certain microbial pathogens, including Chlamydia trachomatis, is based on the induction of local mucosal Th1 response. IL-10 is an anti-inflammatory cytokine that exerts negative immunoregulatory influence on Th1 response. This study investigated whether biochemical modulation of endogenous IL-10 expression at the level of APCs is a practical strategy for enhancing the specific Th1 response against pathogens controlled by Th1 immunity. The results revealed that the high resistance of genetically engineered IL-10-/- (IL-10KO) mice to genital chlamydial infection is a function of the predilection of their APCs to rapidly and preferentially activate a high Th1 response. Thus, in microbiological analysis, IL-10KO mice suffered a shorter duration of infection, less microbial burden, and limited ascending infection than immunocompetent wild-type mice. Also, IL-10KO were resistant to reinfection after 8 wk of the primary infection. Cellular and molecular immunologic evaluation indicated that IL-10KO mice induced greater frequency of chlamydial-specific Th1 response following C. trachomatis infection. Moreover, IL-10KO APCs or antisense IL-10 oligonucleotide-treated wild-type APCs were potent activators of Th1 response from naive or immune T cells. Furthermore, both Ag-pulsed dendritic cells from IL-10KO mice and IL-10 antisense-treated dendritic cells from wild-type mice were efficient cellular vaccines in adoptive immunotherapeutic vaccination against genital chlamydial infection. These findings may furnish a novel immunotherapeutic strategy for boosting the Th1 response against T cell-controlled pathogens and tumors, using IL-10-deficient APCs as vaccine delivery agents.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antigen Presentation/genetics , Bacterial Vaccines/immunology , Dendritic Cells/metabolism , Gene Expression Regulation/immunology , Interleukin-10/antagonists & inhibitors , Interleukin-10/genetics , Lymphocyte Activation/genetics , Th1 Cells/immunology , Adjuvants, Immunologic/genetics , Adjuvants, Immunologic/therapeutic use , Animals , Antigen Presentation/drug effects , Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , Bacterial Vaccines/genetics , Bacterial Vaccines/therapeutic use , Chlamydia Infections/genetics , Chlamydia Infections/immunology , Chlamydia Infections/prevention & control , Dendritic Cells/drug effects , Dendritic Cells/immunology , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Female , Gene Expression Regulation/drug effects , Genetic Predisposition to Disease , Humans , Immunotherapy, Adoptive , Interleukin-10/biosynthesis , Interleukin-10/therapeutic use , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotides, Antisense/therapeutic use , Th1 Cells/drug effects , Th1 Cells/metabolism , Uterine Cervicitis/genetics , Uterine Cervicitis/immunology , Uterine Cervicitis/microbiology , Uterine Cervicitis/prevention & control , Vaginosis, Bacterial/genetics , Vaginosis, Bacterial/immunology , Vaginosis, Bacterial/prevention & control
11.
Immunology ; 98(4): 510-9, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10594682

ABSTRACT

Recent studies in animal models of genital chlamydial disease revealed that early recruitment of dendritic cells and specific T helper type-1 (Th1) cells into the genital mucosae is crucial for reducing the severity of the acute phase of a cervico-vaginal infection and arresting ascending disease. These immune effectors are therefore important for preventing major complications of genital chlamydial infection. Other in vitro studies showed that intercellular adhesion molecule-1 (ICAM-1) plays a role in the antichlamydial action of specific CD4+ and CD8+ T cells. In the present study, we investigated the clinicopathological consequences of ICAM-1 deficiency during chlamydial genital infection in ICAM-1 knockout (ICAM-1KO) mice, and analysed the cellular and molecular immunological bases for any observed pathology or complication. Following a primary genital infection of female ICAM-l-/- and ICAM-1+/+ mice, the intensity of the disease during the first 3 weeks (as assessed by shedding of chlamydiae in the genital tract) was significantly greater in ICAM-1KO mice than in ICAM-1+/+ mice (P < 0.0001), although both ICAM-l-/- and ICAM-1+/+ mice subsequently cleared the primary infection. There was greater ascending disease during the initial stage of the infection, and a higher incidence of tubal disease (hydrosalpinx formation) after multiple infections in ICAM-l-/- mice. Analysis of the cellular and molecular bases for the increased acute and ascending disease in ICAM-l-/- mice revealed that the high affinity of ICAM-1 for leucocyte function antigen type-1 is a property that promotes rapid activation of specific Th1 cells, as well as their early recruitment into the genital mucosa. Moreover, ICAM-1 was more important for naive T-cell activation than primed Th1 cells, although its absence delayed or suppressed immune T-cell activation by at least 50%. Taken together, these results indicated that ICAM-1 is crucial for rapid T-cell activation, early recruitment and control of genitally acquired Chlamydia trachomatis.


Subject(s)
Chlamydia Infections/immunology , Chlamydia trachomatis , Genital Diseases, Female/immunology , Intercellular Adhesion Molecule-1/metabolism , Lymphocyte Activation , Th1 Cells/immunology , Acute Disease , Analysis of Variance , Animals , Antigen Presentation , Chlamydia Infections/pathology , Fallopian Tube Diseases/immunology , Fallopian Tube Diseases/pathology , Fallopian Tubes/pathology , Female , Flow Cytometry , Genital Diseases, Female/pathology , Intercellular Adhesion Molecule-1/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Spleen/immunology , Time Factors
12.
Microbiol Immunol ; 42(9): 617-25, 1998.
Article in English | MEDLINE | ID: mdl-9802562

ABSTRACT

The antimicrobial activity of T cell-derived cytokines, especially interferon (IFN)-gamma, against intracellular pathogens, such as Chlamydia trachomatis, involves the induction of 3 major biochemical processes: tryptophan catabolism, nitric oxide (NO) induction and intracellular iron (Fe) deprivation. Since the epithelial cell is the natural target of chlamydial infection, the presence of these antimicrobial systems in the cell would suggest that they may be involved in T cell control of intracellular multiplication of Chlamydia. However, the controversy over whether these 3 antimicrobial processes are present in both mice and humans has precluded the assessment of the relative contribution of each of the 3 mechanisms to chlamydial inhibition in the same epithelial cell from either mice or humans. In the present study, we identified a Chlamydia-susceptible human epithelial cell line, RT4, that possesses the 3 antimicrobial systems, and we examined the role of nitric oxide (NO) induction, and deprivation of tryptophan or Fe in cytokine-induced inhibition of chlamydiae. It was found that the 3 antimicrobial systems contributed to cytokine-mediated inhibition of the intracellular growth of Chlamydia. NO induction accounted for approximately 20% of the growth inhibition; tryptophan catabolism contributed approximately 30%; iron deprivation was least effective; but the combination of the 3 systems accounted for greater than 60% of the inhibition observed. These results indicate that immune control of chlamydial growth in human epithelial cells may involve multiple mechanisms that include NO induction, tryptophan catabolism and Fe deprivation.


Subject(s)
Cell Communication/immunology , Chlamydia trachomatis/growth & development , Iron/administration & dosage , Nitric Oxide/biosynthesis , Tryptophan/metabolism , Cell Line/drug effects , Cell Line/microbiology , Cytokines/pharmacology , Epithelial Cells/microbiology , Fluorescent Antibody Technique, Direct , HT29 Cells/drug effects , HT29 Cells/microbiology , Humans , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Receptors, Transferrin/analysis , T-Lymphocytes/immunology
13.
Infect Immun ; 66(9): 4030-5, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9712743

ABSTRACT

The induction of local T helper type 1 (Th1)-mediated cellular immunity is crucial for resistance of mice to genital infection by the obligate intracellular bacterium Chlamydia trachomatis. We tested the hypothesis that the route of immunization that elicits relatively high numbers of chlamydia-specific, gamma interferon (IFN-gamma)-secreting T lymphocytes (ISTLs) in the genital tract would induce optimal protective immunity against reinfection. Female BALB/c mice were infected intravaginally (i.v.), intranasally (i.n.), orally (p.o.), or subcutaneously (s.c.) with C. trachomatis. At days 7, 14, 21, and 28 postinfection, T cells isolated from the genital tract tissues were restimulated with chlamydial antigen in vitro, and the amounts of IFN-gamma induced were measured by a sandwiched enzyme-linked immunosorbent assay method. At day 7 postinfection, i.n.- and i.v.-immunized mice had high levels of chlamydia-specific ISTLs in their genital tracts (203.58 +/- 68.1 and 225.5 +/- 12.1 pg/ml, respectively). However, there were no detectable ISTLs in the genital tracts of p.o.- or s.c.-infected mice. When preinfected mice were challenged i.v. 70 days later, animals preexposed by the i.n. route were highly resistant to reinfection, with greatly reduced chlamydial burden, and suffered an attenuated infection that resolved by day 6 postchallenge. Animals preexposed by the i.v. route were modestly protected, whereas p.o. and s.c. groups were indistinguishable in this regard from control mice. The resistance of i.n.-immunized mice (and to some extent the i.v.-exposed mice) to reinfection was associated with early appearance (within 24 h) of high levels of genital ISTLs compared with mice preinfected by other routes. Furthermore, although i.n. and i.v.-immunized mice had comparable levels of chlamydia-specific immunoglobulin A (IgA) antibodies in their vaginal washes, the levels of IgG2a were four- sixfold higher in i.n.-immunized mice than in any of the other groups. The results suggested that immunization routes that foster rapid induction of vigorous genital mucosal cell-mediated immune (CMI) effectors (e.g., IFN-gamma), the CMI-associated humoral effector, IgG2a, and to some extent secretory IgA produce protective immunity against chlamydial genital infection. Therefore, i.n. immunization is a potential delivery route of choice in the development of a vaccine against Chlamydia.


Subject(s)
Chlamydia Infections/immunology , Chlamydia trachomatis/immunology , Genital Diseases, Female/immunology , Interferon-gamma/metabolism , T-Lymphocytes/immunology , Administration, Intranasal , Administration, Oral , Animals , Antibodies, Bacterial/analysis , Cells, Cultured , Chlamydia Infections/physiopathology , Disease Models, Animal , Female , Genital Diseases, Female/physiopathology , HeLa Cells , Humans , Immunity, Mucosal , Immunoglobulin A, Secretory/analysis , Immunoglobulin G/analysis , Injections, Intravenous , Injections, Subcutaneous , Mice , Mice, Inbred BALB C , T-Lymphocytes/metabolism , T-Lymphocytes/microbiology , Vagina/immunology
14.
Infect Immun ; 66(4): 1282-6, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9529043

ABSTRACT

Type 1 CD4+-T-cell-mediated immunity is crucial for the resolution of chlamydial infection of the murine female genital tract. Previous studies demonstrating a correlation between CD4+-T-cell-mediated inhibition of chlamydial growth and gamma interferon (IFN-gamma)-mediated induction of nitric oxide synthase suggested a potential role for the nitric oxide (NO) effector pathway in the clearance of Chlamydia from genital epithelial cells by the immune system. To clarify the role of this pathway, the growth levels of Chlamydia trachomatis organisms in normal (iNOS+/+) mice and in genetically engineered mice lacking the inducible nitric oxide synthase (iNOS) gene (iNOS-/- mice) were compared. There was no significant difference in the course of genital chlamydial infections in iNOS+/+ and iNOS-/- mice as determined by recovery of Chlamydia organisms shed from genital epithelial cells. Dissemination of Chlamydia to the spleen and lungs occurred to a greater extent in iNOS-/- than in iNOS+/+ mice, which correlated with a marginal increase in the susceptibility of macrophages from iNOS-/- mice to chlamydial infection in vitro. However, infections were rapidly cleared from all affected tissues, with no clinical signs of disease. The finding of minimal dissemination in iNOS-/- mice suggested that activation of the iNOS effector pathway was not the primary target of IFN-gamma during CD4+-T-cell-mediated control of chlamydial growth in macrophages because previous reports demonstrated extensive and often fatal dissemination of Chlamydia in mice lacking IFN-gamma. In summary, these results indicate that the iNOS effector pathway is not required for elimination of Chlamydia from epithelial cells lining the female genital tract of mice although it may contribute to the control of dissemination of C. trachomatis by infected macrophages.


Subject(s)
Chlamydia Infections/immunology , Chlamydia trachomatis , Genital Diseases, Female/immunology , Nitric Oxide Synthase/physiology , Animals , Female , Interferon-gamma/physiology , Macrophages, Peritoneal/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II
15.
Endocrinology ; 138(7): 3047-55, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9202251

ABSTRACT

Osteoclast development requires cell-to-cell contact between hematopoietic osteoclast progenitors and bone marrow stromal/osteoblastic support cells. Based on this, we hypothesized that osteopontin, an adhesion protein produced by osteoclasts and osteoblasts, plays a role in osteoclastogenesis. Using in situ hybridization, we demonstrate that cells expressing the osteopontin messenger RNA (mRNA) appear after 3 days of culturing murine bone marrow cells. The number of these cells increases thereafter, reaching a peak on day 5. In the same cultures, cells expressing alkaline phosphatase (AP) or tartrate resistant acid phosphatase (TRAP), phenotypic markers for osteoblastic and osteoclast-like cells, respectively, appeared subsequent to the appearance of the osteopontin-positive cells. By means of a combination of in situ hybridization and histostaining, it was shown that the osteopontin mRNA was localized in 30-50% of the AP-positive or the TRAP-positive, as well as in nonspecific esterase (NSE)-positive, cells. The number of cells expressing both the osteopontin mRNA and either one of the three phenotypic markers was significantly increased in bone marrow cultures from estrogen-deficient mice, as compared with controls. Conversely, the number of all three populations of double positive cells was decreased in cultures treated with a specific antimouse rabbit osteopontin antibody or an RGD peptide. These findings indicate that osteopontin is expressed during the early stages of the differentiation of osteoclast and osteoblast progenitors in the bone marrow and that its cell adhesion properties are required for osteoclastogenesis.


Subject(s)
Bone Marrow Cells , Osteoblasts/cytology , Osteoclasts/cytology , Ovary/physiology , Sialoglycoproteins/physiology , Stem Cells/metabolism , Animals , Antibodies/metabolism , Calcitriol/pharmacology , Cell Communication , Cell Differentiation , Female , In Situ Hybridization , Mice , Oligopeptides/pharmacology , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteopontin , Ovariectomy , RNA, Messenger/metabolism , Rabbits , Sialoglycoproteins/genetics , Sialoglycoproteins/immunology
16.
Biochem Biophys Res Commun ; 232(3): 595-601, 1997 Mar 27.
Article in English | MEDLINE | ID: mdl-9126319

ABSTRACT

It was previously shown that murine T cell clones could inhibit the intracellular growth of the mouse strain of Chlamydia trachomatis by cytokine-mediated induction of the inducible nitric oxide synthase (iNOS) system in epithelial cells, an effect enhanced by direct epithelial-T cell interaction via specific adhesion molecules. These findings and other recent reports showing that human mucosal epithelial cells secrete nitric oxide (NO) via iNOS expression would suggest that mucosal epithelial-derived NO may be involved in mucosal defense against Chlamydia and other pathogens that infect epithelial cells. As an initial approach to investigating whether NO contributes to chlamydial control in humans, the present studies evaluated the susceptibility of human isolates of C. trachomatis to NO delivered by chemical donors or via induction of the epithelial iNOS system by a cytokine-secreting T cell clone. It was found that a chlamydial-specific, cytokine-secreting, murine T lymphocyte clone (clone 2.14-0) could inhibit the intraepithelial growth of human strains of Chlamydia trachomatis (serovar E and H, and Lymphogranuloma venerum type L2) via the iNOS pathway when the clone was co-cultured with chlamydial-infected epithelial cells. Furthermore, treatment of infected epithelial cells with 50 microM of the NO donor, S-nitroso-L-glutathione, resulted in significant inhibition (approximately 70%) of chlamydial multiplication, while the NO scavenger, myoglobin plus ascorbate, could reverse the effect, demonstrating that NO could directly inhibit human strains of Chlamydia. The results are consistent with the hypothesis that the IFN-gamma-inducible iNOS pathway can contribute to chlamydial control in humans.


Subject(s)
Chlamydia trachomatis/drug effects , Chlamydia trachomatis/immunology , Nitric Oxide/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Cell Line , Chlamydia Infections/immunology , Chlamydia Infections/metabolism , Chlamydia Infections/prevention & control , Chlamydia trachomatis/growth & development , Epithelium/immunology , Epithelium/metabolism , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
17.
J Leukoc Biol ; 59(5): 656-62, 1996 May.
Article in English | MEDLINE | ID: mdl-8656050

ABSTRACT

T cell-mediated immunity against Chlamydia in mice is mediated at least in part by T cell-derived interferon-gamma (IFN-gamma) induction of the nitric oxide synthase (iNOS) system in infected epithelial cells. Although IFN-gamma alone could stimulate nitric oxide (NO) production from epithelial cells and inhibit the intracellular growth of Chlamydia, the effectiveness was less than when infected epithelial cells were co-cultured with IFN-gamma-producing T cell clones. In co-cultures containing T cells and infected epithelial cells, additional NO produced by activated T cells could augment chlamydial killing; however, T cell-derived NO was insufficient to account for the total NO present in the co-culture and therefore could not explain the dramatic increase in chlamydial inhibition under those conditions. To determine whether direct cell-to-cell interaction involving adhesion molecules was involved in increased NO induction, the ability of neutralizing monoclonal antibodies directed against intercellular adhesion molecule type 1 (ICAM-1) and leukocyte function antigen-1 (LFA-1) to suppress NO production and lower intracellular chlamydial inhibition was investigated. It was found that monoclonal antibodies against ICAM-1/LFA-1 could significantly reduce the capacity of a protective CD4+ type 1 (Thl) clone (clone 2.14-0) to inhibit the intracellular growth of the C. trachomatis agent of mouse pneumonitis (MoPn). The suppression of the anti-chlamydial action of the clone by antibodies correlated with approximately 50% decrease in NO production. Also, paraformaldehyde-fixed clone 2.14-0 could enhance NO induction and chlamydial inhibition mediated by IFN-gamma, and this effect could be reversed by anti-ICAM-1/LFA-1 antibodies. The results indicated that epithelial-T cell interaction via adhesion molecules enhances NO production and increased chlamydial inhibition by IFN-gamma-secreting T cells.


Subject(s)
Chlamydia/immunology , Integrins/physiology , Nitric Oxide/biosynthesis , T-Lymphocytes/physiology , Animals , Cell Communication , Cells, Cultured , Coculture Techniques , Epithelial Cells , Female , Interferon-gamma/pharmacology , Lymphocyte Function-Associated Antigen-1/physiology , Mice , Mice, Inbred BALB C
18.
Immunology ; 88(1): 1-5, 1996 May.
Article in English | MEDLINE | ID: mdl-8707333

ABSTRACT

T-cell-mediated immunity is crucial for the control of Chlamydia in mice. Recent evidence from studies in an in vitro model of the mucosal epithelium, the polarized epithelial-lymphocyte co-culture (PELC) system, indicated that protective murine T cells mediated intracellular inhibition of the Chlamydia trachomatis agent of mouse pneumonitis (MoPn) at least partly by activating the interferon-gamma (IFN-gamma)-inducible nitric oxide synthase (iNOS) pathway. To investigate whether nitric oxide played a role in controlling chlamydial infection in vivo, the protective capacity of a chlamydial-specific T-cell clone (clone 2.14-0) was analysed in mice in the presence of a specific inhibitor of iNOS. The results revealed that the ability of this clone to clear Chlamydia in vivo is in part mediated by induction of nitric oxide (NO) production. The L-arginine analogue and iNOS inhibitor, NG-monomethyl-L-arginine monoacetate (MLA), increased the chlamydial burden in infected mice and inhibited the ability of clone 2.14-0 to clear genital MoPn infection in vivo. The results are consistent with the working hypothesis that the IFN-gamma-inducible iNOS pathway is involved in the control of Chlamydia by T lymphocytes in mice.


Subject(s)
Chlamydia Infections/immunology , Chlamydia trachomatis , Nitric Oxide/physiology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Arginine/agonists , Arginine/analogs & derivatives , Arginine/pharmacology , Clone Cells , Immunity, Cellular , Mice , Mice, Inbred BALB C , Nitric Oxide/urine , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/physiology , omega-N-Methylarginine
19.
Immunology ; 87(1): 1-8, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8666420

ABSTRACT

T-cell mediated immunity (CMI) is crucial for protection against genital chlamydial infection in mice. To define the underlying molecular mechanism for this protection, several T-cell clones generated against the Chlamydia trachomatis agent of mouse pneumonitis (MoPn) were analysed in an in vitro model of the mucosal epithelium, the polarized epithelial-lymphocyte co-culture (PELC) system, for immunobiological functions that correlated with chlamydial inhibition. The six clones analysed were classified as protective or non-protective on the basis of their ability to cure genital chlamydial infection in syngeneic mice. The results revealed a direct relationship between the ability of a clone to protect in vivo and to inhibit the multiplication of MoPn in vitro. Also, the protective ability of a clone correlated with its capacity to elaborate relatively high levels of interferon-gamma (IFN-gamma) and to induce nitric oxide (NO) production. Moreover, neutralizing anti-IFN-gamma antibodies used alone at 50 micrograms/ml or in combination with anti-tumour necrosis-factor (TNF-alpha), and the L-arginine analogue and NO synthase inhibitor, NG-monomethyl-L-arginine monoacetate (MLA), could significantly suppress the ability of protective clones to inhibit MoPn in epithelial cells. The results suggested that the IFN-gamma-inducible NO synthease pathway is important for chlamydial control in mice. Furthermore, IFN-gamma could stimulate infected murine epithelial cells (line TM3) to secrete NO, resulting in inhibition of MoPn growth. However, the degree of MoPn inhibition obtained with IFN-gamma alone was less than that observed when T cells were co-cultured with infected epithelial cells. T-cell-derived NO could partly explain the enhanced chlamydial inhibition when T cells were co-cultured with infected epithelial cells. These results are consistent with the hypothesis that, besides T-cell-derived IFN-gamma, other factors associated with lymphoepithelial interactions are likely to contribute an important role in chlamydial control by T cells in mice.


Subject(s)
Chlamydia Infections/immunology , Chlamydia trachomatis/immunology , Nitric Oxide/immunology , T-Lymphocytes/immunology , Animals , Cell Culture Techniques , Clone Cells/immunology , Cytokines/immunology , Epithelium/immunology , Female , Immunity, Cellular , Interferon-gamma/metabolism , Mice , Mice, Inbred BALB C , Pneumonia, Bacterial/immunology
20.
Int J Radiat Biol ; 67(5): 557-64, 1995 May.
Article in English | MEDLINE | ID: mdl-7775831

ABSTRACT

A chlamydial-specific T cell clone, capable of inhibiting the growth of infectious Chlamydia in vivo and in vitro, was employed to investigate the effect of gamma-irradiation on the ability of effector T cells to control infections. Clone 2.14-0 (CD4+), specific for the Chlamydia trachomatis biovar agent of mouse pneumonitis (MoPn), was irradiated with varying doses (0, 2.5, 5.0, 10.0, 20.0 and 40.0 Gy) and its biological functions and ability to inhibit the intraepithelial growth of MoPn were assessed. The results revealed that although gamma-irradiation drastically reduced the proliferative response of the clone to antigen, it did not affect the phenotype and the effector function of inhibiting chlamydial growth. The preservation of anti-chlamydial function after gamma-irradiation correlated with the retention of IFN-gamma and TNF-alpha secretion in response to antigenic stimulation. We conclude that the biologic functions of T cells requiring proliferation and differentiation are more likely to be adversely affected by gamma-irradiation on the short-term, but the effector functions, possibly associated with cytokines and cytolysis, may be preserved among persisting effector T cells in an irradiated individual.


Subject(s)
Chlamydia Infections/immunology , Immunity, Cellular/radiation effects , T-Lymphocytes/radiation effects , Animals , Antigens, Bacterial/immunology , Antigens, CD/metabolism , Cell Line , Chlamydia trachomatis , Clone Cells , Cytokines/metabolism , Dose-Response Relationship, Radiation , Female , Gamma Rays , In Vitro Techniques , Lymphocyte Activation/radiation effects , Mice , Mice, Nude , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...