Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Extra Corpor Technol ; 56(2): 37-44, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38888546

ABSTRACT

INTRODUCTION: Cardiopulmonary bypass is an essential component of cardiothoracic surgeries. However, significant complications such as systemic inflammatory response syndrome (SIRS) resulting from cardiopulmonary bypass (CPB) are a common occurrence due to contact between circulating blood and foreign surfaces that leads to platelet activation. It is suggested that different available CPB circuit coatings can potentially reduce platelet activation. However, there have been no published evidence-based reports confirming these claims. In addition, there is no well-established protocol for studying platelet activation biomarkers during CPB in vitro in a laboratory setting. METHODS: CPB was simulated in the laboratory using bovine blood in two different types of coated CPB circuits: Trillium® Biosurface by Medtronic, and XcoatingTM Surface by Terumo. Fresh bovine blood samples were collected and circulated through the CPB circuit following the standard protocol used in the operation rooms. Blood samples were then collected at 5 min, 30 min, and 55 min during the circulation. Blood plasmas were separated and subjected to enzyme-linked immunosorbent assay to measure most established platelet activation markers P-selectin, Platelet Factor 4 (PF4), Glycoprotein IIb/IIIa (GPIIb/IIIa), and ß-thromboglobulin (ß-TG) at different time points. RESULTS: The biomarker values at 30 min and 55 min were compared to the base values at 5 min for each type of CPB circuit. The results of the means from all measured biomarkers showed data measurements that indicated no significant variability within each coating. All collected data points fell within ±2 SD of the means, which was considered acceptable variations across technical replicates.  Conclusion: In this study, we were able to establish an in vitro protocol in the laboratory setting that is precise and reliable with minimum intra-variability. This established protocol will allow for future studies in which different coated CPB circuits can be compared for their effectiveness in blocking platelet activation during the CPB.


Subject(s)
Biomarkers , Cardiopulmonary Bypass , Coated Materials, Biocompatible , Platelet Activation , Cardiopulmonary Bypass/instrumentation , Cardiopulmonary Bypass/adverse effects , Platelet Activation/physiology , Animals , Biomarkers/blood , Cattle , Materials Testing/methods
2.
Pathog Dis ; 822024 Feb 07.
Article in English | MEDLINE | ID: mdl-38684476

ABSTRACT

We have demonstrated previously that TNF-α-producing CD8+ T cells mediate chlamydial pathogenesis, likely in an antigen (Ag)-specific fashion. Here we hypothesize that inhibition of Ag-specific CD8+ T cell response after immunization and/or challenge would correlate with protection against oviduct pathology induced by a protective vaccine regimen. Intranasal (i.n.) live chlamydial elementary body (EB), intramuscular (i.m.) live EB, or i.n. irrelevant antigen, bovine serum albumin (BSA), immunized animals induced near-total protection, 50% protection, or no protection, respectively against oviduct pathology following i.vag. C. muridarum challenge. In these models, we evaluated Ag-specific CD8+ T cell cytokine response at various time-periods after immunization or challenge. The results show protective efficacy of vaccine regimens correlated with reduction of Ag-specific CD8+ T cell TNF-α responses following i.vag. chlamydial challenge, not after immunization. Depletion of CD4+ T cells abrogated, whereas adoptive transfer of Ag-specific CD4+ T cells induced the significant reduction of Ag-specific CD8+ T cell TNF-α response after chlamydial challenge. In conclusion, protective anti-chlamydial vaccine regimens induce Ag-specific CD4+ T cell response that mediate early inhibition of pathogenic CD8+ T cell response following challenge and may serve as a predictive biomarker of protection against Chlamydia -induced chronic pathologies.


Subject(s)
Bacterial Vaccines , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Chlamydia Infections , Animals , CD8-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Chlamydia Infections/immunology , Chlamydia Infections/prevention & control , Bacterial Vaccines/immunology , Bacterial Vaccines/administration & dosage , Female , Mice , Disease Models, Animal , Tumor Necrosis Factor-alpha/metabolism , Chlamydia muridarum/immunology
3.
J Am Heart Assoc ; 11(23): e027598, 2022 12 06.
Article in English | MEDLINE | ID: mdl-36453629

ABSTRACT

Background Stiffer aortas are associated with a faster rate of aortic root (AoR) dilation and higher risk of aortic dissection in patients with Marfan syndrome. We have previously shown that mild aerobic exercise reduces aortic stiffness and rate of AoR dilation in a Marfan mouse model. In this study, we investigated if these results could be translated to pediatric patients with Marfan syndrome. Methods and Results We enrolled 24 patients with Marfan syndrome aged 8 to 19 years to participate in a 6-month physical activity intervention, excluding those with ventricular dysfunction or prior history of aortic surgery. We instructed patients to take 10 000 steps per day, tracked by an activity tracker. At baseline and 6 months, we measured AoR dimension, arterial stiffness, endothelial function, physical activity indices, inflammatory biomarkers, and coping scores. Controls consisted of 15 age-matched patients with Marfan syndrome. Twenty-four patients with Marfan syndrome (median age, 14.4 years [interquartile range {IQR}, 12.2-16.8], 14 male patients) were enrolled. Baseline assessment demonstrated that the majority of these patients were sedentary and had abnormal arterial health. Twenty-two patients completed the intervention and took an average of 7709±2177 steps per day (median, 7627 [IQR, 6344-9671]). Patients wore their Garmin trackers at a median of 92.8% (IQR, 84%-97%) of their intervention days. AoR Z score in the intervention group had a significantly lower rate of change per year compared with the controls (rate of change, -0.24 versus +0.008; P=0.01). Conclusions In this clinical intervention in pediatric patients with Marfan syndrome, we demonstrated that a simple physical activity intervention was feasible in this population and has the potential to decrease the AoR dilation rate. REGISTRATION URL: https://www.clinicaltrials.gov; Unique identifier: NCT03567460.


Subject(s)
Marfan Syndrome , Male , Animals , Mice , Marfan Syndrome/complications , Aorta, Thoracic , Health Status , Exercise
4.
Immunol Cell Biol ; 97(1): 85-91, 2019 01.
Article in English | MEDLINE | ID: mdl-30051926

ABSTRACT

We have shown previously that intranasal vaccination with recombinant chlamydial protease-like activity factor (rCPAF: antigen) and interleukin-12 (IL-12) as an adjuvant induces robust protection against pathological consequences of female genital tract infection with Chlamydia muridarum, a closely related species and a rodent model for the human pathogen Chlamydia trachomatis. Another related species Chlamydia pneumoniae, a human respiratory pathogen, has been associated with exacerbation of atherosclerotic pathology. CPAF is highly conserved among Chlamydia spp. leading us to hypothesize that immunization with rCPAF with IL-12 will protect against high-fat diet (HFD) and C. pneumoniae-induced acceleration of atherosclerosis. rCPAF ± IL-12 immunization induced robust splenic antigen (Ag)-specific IFN-γ and TNF-α production and significantly elevated serum total anti-CPAF Ab, IgG2c, and IgG1 antibody levels compared to mock or IL-12 alone groups. The addition of IL-12 to rCPAF significantly elevated splenic Ag-specific IFN-γ production and IgG2c/IgG1 anti-CPAF antibody ratio. Following intranasal C. pneumoniae challenge and HFD feeding, rCPAF ± IL-12-immunized mice displayed significantly enhanced splenic IFN-γ, not TNF-α, response on days 6 and 9 after challenge, and significantly reduced lung chlamydial burden on day 9 post-challenge compared to mock- or IL-12-immunized mice. Importantly, rCPAF ± IL-12-immunized mice displayed significantly reduced atherosclerotic pathology in the aortas after C. pneumoniae challenge. Serum cholesterol levels were comparable between the groups suggesting that the observed differences in pathology were due to protective immunity against the infection. Together, these results confirm and extend our previous observations that CPAF is a promising candidate antigen for a multisubunit vaccine regimen to protect against Chlamydia-induced pathologies, including atherosclerosis.


Subject(s)
Atherosclerosis/immunology , Chlamydophila Infections/prevention & control , Chlamydophila pneumoniae/immunology , Endopeptidases/administration & dosage , Interleukin-12/administration & dosage , Recombinant Proteins/administration & dosage , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Animals , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/immunology , Atherosclerosis/etiology , Atherosclerosis/prevention & control , Chlamydophila Infections/complications , Endopeptidases/genetics , Endopeptidases/immunology , Immunogenicity, Vaccine , Interleukin-12/immunology , Mice , Recombinant Proteins/immunology
5.
Curr Top Microbiol Immunol ; 412: 183-215, 2018.
Article in English | MEDLINE | ID: mdl-27370346

ABSTRACT

Chlamydial infections lead to a number of clinically relevant diseases and induce significant morbidity in human populations. It is generally understood that certain components of the host immune response to infection also mediate such disease pathologies. A clear understanding of pathogenic mechanisms will enable us to devise better preventive and/or intervention strategies to mitigate the morbidity caused by these infections. Over the years, numerous studies have been conducted to explore the immunopathogenic mechanisms of Chlamydia-induced diseases of the eye, reproductive tract, respiratory tract, and cardiovascular systems. In this article, we provide an overview of the diseases caused by Chlamydia, animal models used to study disease pathology, and a historical context to the efforts to understand chlamydial pathogenesis. Furthermore, we discuss recent findings regarding pathogenesis, with an emphasis on the role of the adaptive immune response in the development of chlamydial disease sequelae. Finally, we summarize the key insights obtained from studies of chlamydial pathogenesis and avenues that remain to be explored in order to inform the next steps of vaccine development against chlamydial infections.


Subject(s)
Chlamydia Infections/immunology , Chlamydia Infections/microbiology , Chlamydia trachomatis/immunology , Chlamydia trachomatis/pathogenicity , Adaptive Immunity , Animals , Disease Models, Animal , Humans
6.
Immunol Cell Biol ; 94(2): 208-12, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26323581

ABSTRACT

Genital chlamydial infections lead to severe upper reproductive tract pathology in a subset of untreated women. We demonstrated previously that tumor necrosis factor (TNF)-α-producing CD8(+) T cells contribute significantly to chlamydial upper genital tract pathology in female mice. In addition, we observed that minimal chlamydial oviduct pathology develops in OT-1 transgenic (OT-1) mice, wherein the CD8(+) T-cell repertoire is restricted to recognition of the ovalbumin peptide Ova(257-264), suggesting that non-Chlamydia-specific CD8(+) T cells may not be responsible for chlamydial pathogenesis. In the current study, we evaluated whether antigen-specific CD8(+) T cells mediate chlamydial pathology. Groups of wild-type (WT) C57BL/6J, OT-1 mice, and OT-1 mice replete with WT CD8(+) T cells (1 × 10(6) cells per mouse intravenously) were infected intravaginally with C. muridarum (5 × 10(4) IFU/mouse). Serum total anti-Chlamydia antibody and total splenic anti-Chlamydia interferon (IFN)-γ and TNF-α responses were comparable among the three groups of animals. However, Chlamydia-specific IFN-γ and TNF-α production from purified splenic CD8(+) T cells of OT-1 mice was minimal, whereas responses in OT-1 mice replete with WT CD8(+) T cells were comparable to those in WT animals. Vaginal chlamydial clearance was comparable between the three groups of mice. Importantly, the incidence and severity of oviduct and uterine horn pathology was significantly reduced in OT-1 mice but reverted to WT levels in OT-1 mice replete with WT CD8(+) T cells. Collectively, these results demonstrate that Chlamydia-specific CD8(+) T cells contribute significantly to upper genital tract pathology.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Chlamydia Infections/immunology , Chlamydia muridarum/immunology , Genitalia, Female/immunology , Animals , Antibodies, Bacterial/blood , CD8-Positive T-Lymphocytes/microbiology , Cells, Cultured , Female , Genitalia, Female/microbiology , Interferon-gamma/metabolism , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , Mice, Transgenic , Ovalbumin/immunology , Peptide Fragments/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Tumor Necrosis Factor-alpha/metabolism
7.
Immunology ; 145(4): 534-42, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25865776

ABSTRACT

The hallmark of chlamydial infection is the development of upper genital pathology in the form of hydrosalpinx and oviduct and/or tubal dilatation. Although molecular events leading to genital tissue presentation and cellular architectural remodelling are unclear, early-stage host immune responses are believed to contribute to these long-term sequelae. Recently, we reported the contribution of selected infection-associated microRNAs (miRs) in the generation of host immunity at early-stage infection (day 6 after intravaginal Chlamydia muridarum challenge in C57BL/6 mice). In this report, we describe the contribution of an infection-associated microRNA, i.e. miR-214, to host immunity. Chlamydia muridarum infection in the C57BL/6 mouse genital tract significantly down-regulated miR-214 while up-regulating intracellular adhesion molecule 1 (ICAM1) gene expression. These in vivo observations were confirmed by establishing direct regulation of ICAM-1 by miR-214 in ex vivo genital cell cultures in the presence of miR-214 mimic and inhibitor. Because, ICAM-1 contributes to recruitment of neutrophils following infection, we also demonstrated that alteration of ICAM1 by miR-214 in interleukin-17A-deficient (IL-17A(-/-) ) mice correlated with reduction of neutrophils infiltrating genital tissue at day 6 after challenge. Additionally, these early-stage events resulted in significantly decreased genital pathology in IL-17A(-/-) mice compared with C57BL/6 mice. This report provides evidence for early-stage regulation of ICAM1 by microRNAs, resulting in reduction of genital pathology associated with chlamydial infection.


Subject(s)
Chlamydia Infections/immunology , Chlamydia muridarum/immunology , Down-Regulation/immunology , Intercellular Adhesion Molecule-1/immunology , MicroRNAs/immunology , Reproductive Tract Infections/immunology , Up-Regulation/immunology , Animals , Chlamydia Infections/genetics , Chlamydia Infections/pathology , Chlamydia muridarum/genetics , Interleukin-17/genetics , Interleukin-17/immunology , Male , Mice , Mice, Knockout , MicroRNAs/genetics , Neutrophil Infiltration/genetics , Neutrophil Infiltration/immunology , Neutrophils/immunology , Neutrophils/pathology , Reproductive Tract Infections/genetics , Reproductive Tract Infections/pathology
8.
J Infect Dis ; 211(12): 2014-22, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25552370

ABSTRACT

BACKGROUND: We demonstrated previously that tumor necrosis factor α (TNF-α)-producing Chlamydia-specific CD8(+) T cells cause oviduct pathological sequelae. METHODS: In the current study, we used wild-type C57BL/6J (WT) mice with a deficiency in genes encoding TNF receptor superfamily member 1a (TNFR1; TNFR1 knockout [KO] mice), TNF receptor superfamily member 1b (TNFR2; TNFR2 KO mice), and both TNFR1 and TNFR2 (TNFR1/2 double KO [DKO] mice) and mix-match adoptive transfers of CD8(+) T cells to study chlamydial pathogenesis. RESULTS: TNFR1 KO, TNFR2 KO, and TNFR1/2 DKO mice displayed comparable clearance of primary or secondary genital Chlamydia muridarum infection but significantly reduced oviduct pathology, compared with WT animals. The Chlamydia-specific total cellular cytokine response in splenic and draining lymph nodes and the antibody response in serum were comparable between the WT and KO animals. However, CD8(+) T cells from TNFR2 KO mice displayed significantly reduced activation (CD11a expression and cytokine production), compared with TNFR1 KO or WT animals. Repletion of TNFR2 KO mice with WT CD8(+) T cells but not with TNFR2 KO CD8(+) T cells and repletion of TNFR1 KO mice with either WT or TNFR1 KO CD8(+) T cells restored oviduct pathology to WT levels in both KO groups. CONCLUSIONS: Collectively, these results demonstrate that TNFR2-bearing CD8(+) T cells and TNFR1-bearing non-CD8(+) T cells contribute significantly to oviduct pathology following genital chlamydial infection.


Subject(s)
CD8-Positive T-Lymphocytes/chemistry , Chlamydia Infections/pathology , Receptors, Tumor Necrosis Factor, Type II/analysis , Receptors, Tumor Necrosis Factor, Type I/analysis , Reproductive Tract Infections/pathology , Animals , Female , Mice, Inbred C57BL , Mice, Knockout
9.
Vaccines (Basel) ; 2(3): 515-36, 2014 Jul 02.
Article in English | MEDLINE | ID: mdl-26344743

ABSTRACT

Conventional vaccine strategies have been highly efficacious for several decades in reducing mortality and morbidity due to infectious diseases. The bane of conventional vaccines, such as those that include whole organisms or large proteins, appear to be the inclusion of unnecessary antigenic load that, not only contributes little to the protective immune response, but complicates the situation by inducing allergenic and/or reactogenic responses. Peptide vaccines are an attractive alternative strategy that relies on usage of short peptide fragments to engineer the induction of highly targeted immune responses, consequently avoiding allergenic and/or reactogenic sequences. Conversely, peptide vaccines used in isolation are often weakly immunogenic and require particulate carriers for delivery and adjuvanting. In this article, we discuss the specific advantages and considerations in targeted induction of immune responses by peptide vaccines and progresses in the development of such vaccines against various diseases. Additionally, we also discuss the development of particulate carrier strategies and the inherent challenges with regard to safety when combining such technologies with peptide vaccines.

10.
Vaccine ; 31(48): 5722-8, 2013 Nov 19.
Article in English | MEDLINE | ID: mdl-24096029

ABSTRACT

Vaccination with recombinant chlamydial protease-like activity factor (rCPAF) has been shown to provide robust protection against genital Chlamydia infection. Adoptive transfer of IFN-γ competent CPAF-specific CD4⁺ T cells was sufficient to induce early resolution of chlamydial infection and reduction of subsequent pathology in recipient IFN-γ-deficient mice indicating the importance of IFN-γ secreting CD4⁺ T cells in host defense against Chlamydia. In this study, we identify CD4⁺ T cell reactive CPAF epitopes and characterize the activation of epitope-specific CD4⁺ T cells following antigen immunization or Chlamydia challenge. Using the HLA-DR4 (HLA-DRB1*0401) transgenic mouse for screening overlapping peptides that induced T cell IFN-γ production, we identified at least 5 CPAF T cell epitopes presented by the HLA-DR4 complex. Immunization of HLA-DR4 transgenic mice with a rCPAFep fusion protein containing these 5 epitopes induced a robust cell-mediated immune response and significantly accelerated the resolution of genital and pulmonary Chlamydia infection. rCPAFep vaccination induced CPAF-specific CD4⁺ T cells in the spleen were detected using HLA-DR4/CPAF-epitope tetramers. Additionally, CPAF-specific CD4⁺ clones could be detected in the mouse spleen following Chlamydia muridarum and a human Chlamydia trachomatis strain challenge using these novel tetramers. These results provide the first direct evidence that a novel CPAF epitope vaccine can provide protection and that HLA-DR4/CPAF-epitope tetramers can detect CPAF epitope-specific CD4⁺ T cells in HLA-DR4 mice following C. muridarum or C. trachomatis infection. Such tetramers could be a useful tool for monitoring CD4⁺ T cells in immunity to Chlamydia infection and in developing epitope-based human vaccines using the murine model.


Subject(s)
Bacterial Vaccines/immunology , Chlamydia Infections/prevention & control , Epitopes, T-Lymphocyte/immunology , HLA-DR4 Antigen/immunology , Animals , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , CD4-Positive T-Lymphocytes/immunology , Chlamydia Infections/immunology , Chlamydia muridarum/immunology , Chlamydia trachomatis/immunology , Epitopes, T-Lymphocyte/genetics , Female , HLA-DR4 Antigen/genetics , Humans , Interferon-gamma/metabolism , Mice , Mice, Transgenic , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/prevention & control , Reproductive Tract Infections/immunology
11.
Infect Immun ; 80(6): 2177-85, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22493083

ABSTRACT

A licensed vaccine against Francisella tularensis is currently not available. Two Francisella tularensis subsp. novicida (herein referred to by its earlier name, Francisella novicida) attenuated strains, the ΔiglB and ΔfopC strains, have previously been evaluated as potential vaccine candidates against pneumonic tularemia in experimental animals. F. novicida ΔiglB, a Francisella pathogenicity island (FPI) mutant, is deficient in phagosomal escape and intracellular growth, whereas F. novicida ΔfopC, lacking the outer membrane lipoprotein FopC, which is required for evasion of gamma interferon (IFN-γ)-mediated signaling, is able to escape and replicate in the cytosol. To dissect the difference in protective immune mechanisms conferred by these two vaccine strains, we examined the efficacy of the F. novicida ΔiglB and ΔfopC mutants against pulmonary live-vaccine-strain (LVS) challenge and found that both strains provided comparable protection in wild-type, major histocompatibility complex class I (MHC I) knockout, and MHC II knockout mice. However, F. novicida ΔfopC-vaccinated but not F. novicida ΔiglB-vaccinated perforin-deficient mice were more susceptible and exhibited greater bacterial burdens than similarly vaccinated wild-type mice. Moreover, perforin produced by natural killer (NK) cells and release of granzyme contributed to inhibition of LVS replication within macrophages. This NK cell-mediated LVS inhibition was enhanced with anti-F. novicida ΔfopC immune serum, suggesting antibody-dependent cell-mediated cytotoxicity (ADCC) in F. novicida ΔfopC-mediated protection. Overall, this study provides additional immunological insight into the basis for protection conferred by live attenuated F. novicida strains with different phenotypes and supports further investigation of this organism as a vaccine platform for tularemia.


Subject(s)
Bacterial Vaccines , Francisella tularensis/immunology , Granzymes/metabolism , Perforin/metabolism , Tularemia/prevention & control , Animals , Bacterial Proteins/metabolism , CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/physiology , Cells, Cultured , Coculture Techniques , Gene Expression Regulation , Genes, MHC Class I/genetics , Genes, MHC Class I/physiology , Genes, MHC Class II/genetics , Genes, MHC Class II/physiology , Granzymes/genetics , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Perforin/genetics , Tularemia/immunology , Vaccination , Vaccines, Attenuated
12.
Infect Immun ; 79(7): 2928-35, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21536799

ABSTRACT

The immunopathogenesis of Chlamydia trachomatis-induced oviduct pathological sequelae is not well understood. Mice genetically deficient in perforin (perforin(-/-) mice) or tumor necrosis factor alpha (TNF-α) production (TNF-α(-/-) mice) displayed comparable vaginal chlamydial clearance rates but significantly reduced oviduct pathology (hydrosalpinx) compared to that of wild-type mice. Since both perforin and TNF-α are effector mechanisms of CD8(+) T cells, we evaluated the role of CD8(+) T cells during genital Chlamydia muridarum infection and oviduct sequelae. Following vaginal chlamydial challenge, (i) mice deficient in TAP I (and therefore the major histocompatibility complex [MHC] I pathway and CD8(+) T cells), (ii) wild-type mice depleted of CD8(+) T cells, and (iii) mice genetically deficient in CD8 (CD8(-/-) mice) all displayed similar levels of vaginal chlamydial clearance but significantly reduced hydrosalpinx, compared to those of wild-type C57BL/6 mice, suggesting a role for CD8(+) T cells in chlamydial pathogenesis. Repletion of CD8(-/-) mice with wild-type or perforin(-/-), but not TNF-α(-/-), CD8(+) T cells at the time of challenge restored hydrosalpinx to levels observed in wild-type C57BL/6 mice, suggesting that TNF-α production from CD8(+) T cells is important for pathogenesis. Additionally, repletion of TNF-α(-/-) mice with TNF-α(+/+) CD8(+) T cells significantly enhanced the incidence of hydrosalpinx and oviduct dilatation compared to those of TNF-α(-/-) mice but not to the levels found in wild-type mice, suggesting that TNF-α production from CD8(+) T cells and non-CD8(+) cells cooperates to induce optimal oviduct pathology following genital chlamydial infection. These results provide compelling new evidence supporting the contribution of CD8(+) T cells and TNF-α production to Chlamydia-induced reproductive tract sequelae.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Chlamydia Infections/immunology , Chlamydia muridarum , Fallopian Tubes/pathology , Genital Diseases, Female/immunology , Tumor Necrosis Factor-alpha/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , Chlamydia Infections/microbiology , Chlamydia Infections/pathology , Fallopian Tubes/microbiology , Female , Genital Diseases, Female/microbiology , Genital Diseases, Female/pathology , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Perforin/biosynthesis , Perforin/genetics , Tumor Necrosis Factor-alpha/genetics , Vagina/microbiology
13.
Vaccine ; 29(14): 2519-22, 2011 Mar 21.
Article in English | MEDLINE | ID: mdl-21300093

ABSTRACT

We previously have shown the efficacy of recombinant (r) chlamydial protease-like activity factor (CPAF) vaccination against hydrosalpinx development following primary genital chlamydial challenge. In this study, we evaluated further the protection induced by rCPAF vaccination against infertility. Following primary challenge, fertility levels were not significantly different between the mock- and CPAF-vaccinated and Chlamydia alone challenged mice. However, following secondary genital chlamydial challenge, mock (PBS) immunized mice displayed a significant reduction of fertility compared to age-matched naïve mice, while mice vaccinated intranasally with rCPAF+CpG displayed significant prevention of infertility. These results suggest that hydrosalpinx may be a reliable indicator of impending infertility, and that rCPAF is a promising candidate to prevent infertility resulting from repeated genital chlamydial infections.


Subject(s)
Bacterial Vaccines/immunology , Chlamydia Infections/immunology , Chlamydia Infections/prevention & control , Endopeptidases/immunology , Infertility, Female/prevention & control , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Animals , Bacterial Vaccines/administration & dosage , Chlamydia Infections/complications , Endopeptidases/administration & dosage , Female , Mice , Mice, Inbred BALB C , Oligodeoxyribonucleotides/administration & dosage , Vaccination/methods
14.
Front Immunol ; 2: 73, 2011.
Article in English | MEDLINE | ID: mdl-22566862

ABSTRACT

We have shown that immunization with soluble recombinant chlamydial protease-like activity factor (rCPAF) and a T helper 1 type adjuvant can induce significantly enhanced bacterial clearance and protection against Chlamydia-induced pathological sequelae in the genital tract. In this study, we investigated the use of bone marrow derived dendritic cells (BMDCs) pulsed ex vivo with rCPAF + CpG in an adoptive subcutaneous immunization for the ability to induce protective immunity against genital chlamydial infection. We found that BMDCs pulsed with rCPAF + CpG efficiently up-regulated the expression of activation markers CD86, CD80, CD40, and major histocompatibility complex class II (MHC II), and secreted interleukin-12, but not IL-10 and IL-4. Mice adoptively immunized with rCPAF + CpG-pulsed BMDCs or UV-EB + CpG-pulsed BMDCs produced elevated levels of antigen-specific IFN-γ and enhanced IgG1 and IgG2a antibodies. Moreover, mice immunized with rCPAF + CpG-pulsed BMDCs or UV-EB + CpG-pulsed BMDCs exhibited significantly reduced genital Chlamydia shedding, accelerated resolution of infection, and reduced oviduct pathology when compared to infected mock-immunized animals. These results suggest that adoptive subcutaneous immunization with ex vivo rCPAF-pulsed BMDCs is an effective approach, comparable to that induced by UV-EB-BMDCs, for inducing robust anti-Chlamydia immunity.

15.
Infect Immun ; 78(9): 3942-9, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20605976

ABSTRACT

We have previously demonstrated the efficacy of recombinant chlamydial protease-like activity factor (rCPAF; a secreted chlamydial protein) in inducing antigen-specific CD4+ T cell/gamma interferon (IFN-gamma)-mediated but not antibody-mediated chlamydial clearance and reduction of upper genital tract (UGT) pathological sequelae. Since chlamydial integral antigens may induce neutralizing antibody protection, we further evaluated induction of protective immunity using a combination of rCPAF and UV-inactivated chlamydial elementary bodies (UV-EB) against vaginal chlamydial challenge in comparison to immunization with the individual components or live EB. The rCPAF-UV-EB immunization induced a significantly enhanced anti-UV-EB cellular and antibody response and a reduced anti-CPAF cellular and antibody response, compared to immunization with the respective individual components. Moreover, vaccination with UV-EB and rCPAF-UV-EB induced serum antibodies that neutralized chlamydial infectivity. The rCPAF-UV-EB immunization resulted in a significant reduction of vaginal chlamydial shedding and induced earlier bacterial clearance than vaccination of mice with the individual components. Importantly, the UGT sequelae were significantly reduced in mice immunized with rCPAF or rCPAF-UV-EB, but not in those immunized with UV-EB alone, and approached the levels of protection induced by live EB. These results collectively suggest that a combination of neutralizing antibodies induced by integral chlamydial antigens and cell-mediated responses induced by secreted proteins such as CPAF induces optimal protective immunity against genital chlamydial infections.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Bacterial Vaccines/immunology , Chlamydia Infections/prevention & control , Chlamydia muridarum/immunology , Genital Diseases, Female/prevention & control , Animals , Antibodies, Bacterial/blood , Cricetinae , Female , Immunization , Mice , Mice, Inbred BALB C , Recombinant Fusion Proteins/immunology , Vaccines, Synthetic/immunology , Vagina/microbiology
16.
Vaccine ; 28(11): 2323-9, 2010 Mar 08.
Article in English | MEDLINE | ID: mdl-20056182

ABSTRACT

We have shown previously that vaccination with recombinant chlamydial protease-like activity factor (rCPAF) plus interleukin-12 as an adjuvant induces robust protective immunity against primary genital Chlamydia muridarum challenge in mice. Since CPAF is a protease, we compared the effects of enzymatically active and inactive (heat denatured) rCPAF to determine whether proteolytic activity is expendable for the induction of protective immunity against chlamydial challenge. Active, but not inactive, rCPAF immunization induced high levels of anti-active CPAF antibody, whereas both induced robust splenic CPAF-specific IFN-gamma production. Vaccination with active or inactive rCPAF induced enhanced vaginal chlamydial clearance as early as day 6 with complete resolution of infection by day 18, compared to day 30 in mock-vaccinated and challenged animals. Importantly, significant and comparable reductions in oviduct pathology were observed in active and inactive rCPAF-vaccinated mice compared to mock-vaccinated animals. Thus, rCPAF induced anti-chlamydial immunity is largely independent of enzymatic activity and secondary or higher order protein conformation.


Subject(s)
Bacterial Vaccines/immunology , Chlamydia Infections/prevention & control , Endopeptidases/immunology , Animals , Antibodies, Bacterial/blood , Chlamydia Infections/immunology , Chlamydia muridarum/immunology , Female , Genitalia, Female/immunology , Hot Temperature , Interferon-gamma/metabolism , Lymphocytes/immunology , Lymphogranuloma Venereum/prevention & control , Mice , Mice, Inbred BALB C , Protein Denaturation , Spleen/immunology , Vaccines, Inactivated/immunology , Vaccines, Synthetic/immunology
17.
FEMS Immunol Med Microbiol ; 55(2): 271-9, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19281569

ABSTRACT

Mice deficient in B cells (micromT mice) were used to evaluate the role of antibody in enhanced chlamydial clearance and reduction of pathology afforded by vaccination with recombinant chlamydial protease-like activity factor (rCPAF). Enhanced, but comparable, chlamydial clearance was observed in micromT and wild-type (WT) mice after rCPAF+CpG vaccination. Chlamydia-induced pathology was present in mock-immunized animals, but at significantly greater levels in micromT than WT mice, whereas vaccinated micromT and WT mice exhibited similar reductions in pathology. Thus, antibodies may play a role in protection against chlamydial pathology after primary infection, but were largely dispensable in rCPAF+CpG-induced chlamydial clearance and reduction in pathology.


Subject(s)
Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Bacterial Vaccines/immunology , Chlamydia Infections/prevention & control , Chlamydia muridarum/immunology , Genital Diseases, Female/prevention & control , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Animals , Chlamydia Infections/immunology , Chlamydia Infections/microbiology , Chlamydia Infections/pathology , Colony Count, Microbial , Female , Genital Diseases, Female/immunology , Genital Diseases, Female/microbiology , Genital Diseases, Female/pathology , Mice , Oligodeoxyribonucleotides/administration & dosage , Oligodeoxyribonucleotides/pharmacology , Vaccines, Synthetic/immunology
18.
J Immunol ; 180(5): 3375-82, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18292563

ABSTRACT

Chlamydia has been shown to evade host-specific IFN-gamma-mediated bacterial killing; however, IFN-gamma-deficient mice exhibit suboptimal late phase vaginal Chlamydia muridarum clearance, greater dissemination, and oviduct pathology. These findings introduce constraints in understanding results from murine chlamydial vaccination studies in context of potential implications to humans. In this study, we used mice deficient in either IFN-gamma or the IFN-gamma receptor for intranasal vaccination with a defined secreted chlamydial Ag, chlamydial protease-like activity factor (CPAF), plus CpG and examined the role of IFN-gamma derived from adoptively transferred Ag-specific CD4+ T cells in protective immunity against genital C. muridarum infection. We found that early Ag-specific IFN-gamma induction and CD4+ T cell infiltration correlates with the onset of genital chlamydial clearance. Adoptively transferred IFN-gamma competent CPAF-specific CD4+ T cells failed to enhance the resolution of genital chlamydial infection within recipient IFN-gamma receptor-deficient mice. Conversely, IFN-gamma production from adoptively transferred CPAF-specific CD4+ T cells was sufficient in IFN-gamma-deficient mice to induce early resolution of infection and reduction of subsequent pathology. These results provide the first direct evidence that enhanced anti-C. muridarum protective immunity induced by Ag-specific CD4+ T cells is dependent upon IFN-gamma signaling and that such cells produce sufficient IFN-gamma to mediate the protective effects. Additionally, MHC class II pathway was sufficient for induction of robust protective anti-C. muridarum immunity. Thus, targeting soluble candidate Ags via MHC class II to CD4+ T cells may be a viable vaccine strategy to induce optimal IFN-gamma production for effective protective immunity against human genital chlamydial infection.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Vaccines/immunology , CD4-Positive T-Lymphocytes/transplantation , Chlamydia Infections/prevention & control , Chlamydia muridarum/immunology , Epitopes, T-Lymphocyte/immunology , Interferon-gamma/biosynthesis , Vaginal Diseases/prevention & control , Administration, Intranasal , Adoptive Transfer , Animals , Antigen Presentation/genetics , Antigens, Bacterial/administration & dosage , Bacterial Vaccines/administration & dosage , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Chlamydia Infections/immunology , Chlamydia Infections/microbiology , Endopeptidases/administration & dosage , Endopeptidases/immunology , Epitopes, T-Lymphocyte/administration & dosage , Female , Interferon-gamma/deficiency , Interferon-gamma/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Oligodeoxyribonucleotides/administration & dosage , Oligodeoxyribonucleotides/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Vaginal Diseases/immunology , Vaginal Diseases/microbiology , beta 2-Microglobulin/deficiency , beta 2-Microglobulin/genetics
19.
Clin Vaccine Immunol ; 14(12): 1537-44, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17942608

ABSTRACT

An important consideration for antichlamydial vaccine development is the induction of cross-serovar protection, since multiple serovars (D to L) of Chlamydia trachomatis cause genital infections. We have shown previously that vaccination with C. trachomatis-derived recombinant chlamydial protease-like activity factor (rCPAF) induced significant earlier resolution of Chlamydia muridarum infection and reduced oviduct pathology. However, the vaccinated mice continued to shed chlamydiae for up to 2 weeks after challenge. In this study, C. trachomatis serovar D recombinant proteins, such as recombinant major outer membrane protein (rMOMP), recombinant inclusion membrane protein A (rIncA), and rCPAF were administered intranasally, individually or in combinations, with murine interleukin-12 (IL-12) as an adjuvant, and cross-species immunity against intravaginal C. muridarum infection was examined. Immunization with rCPAF plus IL-12 (rCPAF+IL-12), compared to immunization with rIncA+IL-12 or rMOMP+IL-12, induced the greatest antigen-specific gamma interferon production from purified CD4(+) T cells and concurrently enhanced serum antibody production. All (100%) the animals vaccinated with rCPAF+IL-12 alone or in any combination completely resolved the infection by day 18 after challenge compared to animals vaccinated with rIncA+IL-12 (50%), rMOMP+IL-12 (33%), or phosphate-buffered saline (mock vaccinated; 0%). Moreover, oviduct pathology in mice vaccinated by any regimen that included rCPAF, but not rMOMP+IL-12 or rIncA+IL-12 alone, was markedly reduced compared to mock-immunized animals. The addition of rMOMP and/or rIncA did not significantly enhance the rCPAF+IL-12-induced effect on bacterial clearance or oviduct pathology. These results suggest a greater conservation of protective linear antigenic epitopes within CPAF than MOMP or IncA across the examined serovars and the need to identify other highly conserved antigens for use with rCPAF in a multisubunit recombinant vaccine.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Vaccines/immunology , Chlamydia Infections/prevention & control , Genital Diseases, Female/immunology , Vaccination , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Animals , Antigens, Bacterial/administration & dosage , Bacterial Outer Membrane Proteins/administration & dosage , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/immunology , Bacterial Outer Membrane Proteins/isolation & purification , Bacterial Proteins/administration & dosage , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bacterial Proteins/isolation & purification , Bacterial Vaccines/administration & dosage , Chlamydia Infections/immunology , Chlamydia muridarum/classification , Chlamydia muridarum/immunology , Cross Reactions/immunology , Drug Therapy, Combination , Endopeptidases/administration & dosage , Endopeptidases/chemistry , Endopeptidases/genetics , Endopeptidases/immunology , Endopeptidases/isolation & purification , Epitopes , Female , Genital Diseases, Female/microbiology , HeLa Cells , Humans , Interleukin-12/administration & dosage , Interleukin-12/genetics , Interleukin-12/immunology , Mice , Mice, Inbred BALB C , Oviducts/pathology , Phosphoproteins/administration & dosage , Phosphoproteins/chemistry , Phosphoproteins/genetics , Phosphoproteins/immunology , Phosphoproteins/isolation & purification , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...