Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Cancer Immunol Immunother ; 71(7): 1611-1623, 2022 Jul.
Article in English | MEDLINE | ID: mdl-34731283

ABSTRACT

We tested the concept that host preexisting influenza A virus immunity can be redirected to inhibit tumor growth and metastasis through systemic administration of influenza A virus-related peptides to targeted tumors. Mice infected with influenza A virus strain A/Puerto Rico/8/34 (PR8) were used as a model of a host with preexisting viral immunity. The extent to which preexisting influenza A immunity in PR8-immunized mice can be redirected to inhibit tumor growth and metastasis was first examined by ectopic expression of influenza A nucleoprotein (NP) and hemagglutinin (HA) in syngeneic mammary tumor cells via lentiviral transduction. Then, the feasibility of implementing this strategy using a systemic therapy approach was assessed by systemic delivery of major histocompatibility complex class I (MHC-I)-compatible peptides to targeted mammary tumors overexpressing human epidermal growth factor receptor-2 (HER2) in mice using a novel HER2-targeting single-lipid nanoparticle (SLNP). Our results show that preexisting influenza A immunity in PR8-immunized mice could be quickly redirected to syngeneic tumors expressing influenza A NP and HA, leading to strong inhibition of tumor growth and metastasis and improvement of survival compared to the findings in antigen-naïve control mice. MHC-I-compatible peptides could be delivered to targeted mammary tumors in mice using the HER2-targeting SLNP for antigen presentation, which subsequently redirected preexisting influenza A immunity to the tumors to exert antitumor activities. In conclusion, preexisting influenza A immunity can be repurposed for cancer immunotherapy through systemic delivery of influenza A-related peptides to targeted tumors. Further development of the strategy for clinical translation is warranted.


Subject(s)
Influenza A virus , Influenza, Human , Neoplasms , Orthomyxoviridae Infections , Animals , Antibodies, Viral , Humans , Immunotherapy , Influenza, Human/prevention & control , Liposomes , Mice , Mice, Inbred BALB C , Nanoparticles , Neoplasms/therapy , Orthomyxoviridae Infections/prevention & control , Peptides
2.
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
3.
Lab Invest ; 91(10): 1530-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21769086

ABSTRACT

Respiratory dysfunction in adults has been correlated with neonatal Chlamydia trachomatis pneumonia in several studies, but a causal association has not been clearly demonstrated. In this study, we examined radial alveolar counts (RACs) by microscopy, and airway and parenchymal lung function using a small animal ventilator in juvenile (5 weeks age) and adult (8 weeks age) BALB/c mice challenged as neonates with Chlamydia muridarum (C. mur) on day 1 or day 7 after birth, representing saccular (human pre-term neonates) and alveolar (human term neonates) stages of lung development, respectively. Pups challenged with C. mur on either day 1 or 7 after birth demonstrated significantly enhanced airway hyperreactivity and lung compliance, both as juveniles (5 weeks age) and adults (8 weeks age), compared with mock-challenged mice. Moreover, mice challenged neonatally with Chlamydia displayed significantly reduced RACs, suggesting emphysematous changes. Antimicrobial treatment during the neonatal infection induced early bacterial clearance and partially ameliorated the Chlamydia-induced lung dysfunction as adults. These results suggest that neonatal chlamydial pneumonia, especially in pre-term neonates, is a cause of respiratory dysfunction continuing into adulthood, and that antimicrobial administration may be partially effective in preventing the adverse respiratory sequelae in adulthood. The results of our studies also emphasize the importance of prenatal screening and treatment of pregnant women for C. trachomatis in order to prevent the infection of neonates.


Subject(s)
Aging , Animals, Newborn , Chlamydia Infections/pathology , Chlamydia Infections/physiopathology , Pneumonia, Bacterial/pathology , Pneumonia, Bacterial/physiopathology , Respiratory System/pathology , Respiratory System/physiopathology , Animals , Animals, Newborn/growth & development , Anti-Bacterial Agents/administration & dosage , Bronchial Hyperreactivity/etiology , Chlamydia Infections/complications , Chlamydia Infections/drug therapy , Drug Administration Schedule , Erythromycin/administration & dosage , Lung/drug effects , Lung/pathology , Lung/physiopathology , Lung Compliance , Mice , Mice, Inbred BALB C , Pneumonia, Bacterial/complications , Pneumonia, Bacterial/drug therapy , Pulmonary Alveoli/growth & development
4.
Blood Cancer J ; 10(11): 110, 2020 11 02.
Article in English | MEDLINE | ID: mdl-33149123

ABSTRACT

Therapeutically targeting CD138, a define multiple myeloma (MM) antigen, is not yet approved for patients. We here developed and determined the preclinical efficacy of VIS832, a novel therapeutic monoclonal antibody (MoAb) with differentiated CD138 target binding to BB4 that is anti-CD138 MoAb scaffold for indatuximab ravtansine (BT062). VIS832 demonstrated enhanced CD138-binding avidity and significantly improved potency to kill MM cell lines and autologous patient MM cells regardless of resistance to current standard-of-care therapies, via robust antibody-dependent cellular cytotoxicity and phagocytosis mediated by NK and macrophage effector cells, respectively. Specifically, CD38-targeting daratumumab-resistant MM cells were highly susceptible to VIS832 which, unlike daratumumab, spares NK cells. Superior maximal cytolysis of VIS832 vs. daratumumab corresponded to higher CD138 vs. CD38 levels in MM cells. Furthermore, VIS832 acted synergistically with lenalidomide or bortezomib to deplete MM cells. Importantly, VIS832 at a sub-optimal dose inhibited disseminated MM1S tumors in vivo as monotherapy (P < 0.0001), and rapidly eradicated myeloma burden in all mice concomitantly receiving bortezomib, with 100% host survival. Taken together, these data strongly support clinical development of VIS832, alone and in combination, for the therapeutic treatment of MM in relapsed and refractory patients while pointing to its potential therapeutic use earlier in disease intervention.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Bortezomib/pharmacology , Immunoconjugates/pharmacology , Multiple Myeloma/drug therapy , Neoplasm Proteins/antagonists & inhibitors , Syndecan-1/antagonists & inhibitors , Animals , Antineoplastic Agents, Immunological/immunology , Bortezomib/agonists , Cell Line, Tumor , Drug Synergism , Humans , Maytansine/agonists , Maytansine/analogs & derivatives , Maytansine/pharmacology , Mice , Mice, SCID , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Neoplasm Proteins/immunology , Syndecan-1/immunology , Xenograft Model Antitumor Assays
5.
Cancer Lett ; 430: 47-56, 2018 08 28.
Article in English | MEDLINE | ID: mdl-29746929

ABSTRACT

Here, we report that treatment of syngeneic mouse tumors transduced to overexpress human epidermal growth factor receptor-2 (HER2) with the anti-human HER2 antibody trastuzumab upregulated the level of programmed death-ligand 1 (PD-L1), an important negative regulator of T-cell response, in a transgenic mouse model immune-tolerant to human HER2. We further found that trastuzumab alone had no detectable effect on the level of PD-L1 expression in monocultures of HER2-overexpressing human breast cancer cells but upregulated PD-L1 in the same panel of HER2-overexpressing breast cancer cells when they were co-cultured with human peripheral blood mononuclear cells, and the upregulation of PD-L1 could be blocked by an IFNγ-neutralizing antibody. Inhibition of HER2 intrinsic signaling via HER2 expression knockdown or kinase inhibition had variable and cell-context-specific effects on downregulating the PD-L1 level. Analysis of The Cancer Genome Atlas database showed no direct correlation between HER2 and PD-L1 at the messenger RNA level. Trastuzumab-mediated upregulation of PD-L1 through engagement of immune effector cells may function as a potential mechanism of trastuzumab resistance. Our data justify further investigation of the value of adding anti-PD-1 or anti-PD-L1 therapy to trastuzumab-based treatment.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , B7-H1 Antigen/metabolism , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Receptor, ErbB-2/metabolism , Trastuzumab/pharmacology , Animals , Antineoplastic Agents, Immunological/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/genetics , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Culture Techniques , Cell Line, Tumor , Coculture Techniques , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/immunology , Interferon-gamma/metabolism , Leukocytes, Mononuclear , Melanoma, Experimental/drug therapy , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Signal Transduction , Trastuzumab/therapeutic use , Up-Regulation/drug effects
6.
Cancer Lett ; 384: 39-49, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27693630

ABSTRACT

Cetuximab inhibits HIF-1-regulated glycolysis in cancer cells, thereby reversing the Warburg effect and leading to inhibition of cancer cell metabolism. AMP-activated protein kinase (AMPK) is activated after cetuximab treatment, and a sustained AMPK activity is a mechanism contributing to cetuximab resistance. Here, we investigated how acetyl-CoA carboxylase (ACC), a downstream target of AMPK, rewires cancer metabolism in response to cetuximab treatment. We found that introduction of experimental ACC mutants lacking the AMPK phosphorylation sites (ACC1_S79A and ACC2_S212A) into head and neck squamous cell carcinoma (HNSCC) cells protected HNSCC cells from cetuximab-induced growth inhibition. HNSCC cells with acquired cetuximab resistance contained not only high levels of T172-phosphorylated AMPK and S79-phosphorylated ACC1 but also an increased level of total ACC. These findings were corroborated in tumor specimens of HNSCC patients treated with cetuximab. Cetuximab plus TOFA (an allosteric inhibitor of ACC) achieved remarkable growth inhibition of cetuximab-resistant HNSCC xenografts. Our data suggest a novel paradigm in which cetuximab-mediated activation of AMPK and subsequent phosphorylation and inhibition of ACC is followed by a compensatory increase in total ACC, which rewires cancer metabolism from glycolysis-dependent to lipogenesis-dependent.


Subject(s)
Acetyl-CoA Carboxylase/metabolism , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/drug therapy , Cetuximab/pharmacology , Drug Resistance, Neoplasm , Glycolysis/drug effects , Head and Neck Neoplasms/drug therapy , Lipolysis/drug effects , AMP-Activated Protein Kinases/metabolism , Acetyl-CoA Carboxylase/antagonists & inhibitors , Acetyl-CoA Carboxylase/genetics , Adult , Aged , Aged, 80 and over , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Enzyme Inhibitors/pharmacology , Female , Head and Neck Neoplasms/enzymology , Head and Neck Neoplasms/pathology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mice, Nude , Middle Aged , Mutation , Phosphorylation , RNA Interference , Signal Transduction/drug effects , Squamous Cell Carcinoma of Head and Neck , Time Factors , Transfection , Xenograft Model Antitumor Assays
7.
Oncoimmunology ; 5(4): e1100790, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27141382

ABSTRACT

It is increasingly recognized that trastuzumab, an antibody approved for treating human epidermal growth factor receptor 2 (HER2)-overexpressing breast cancer, exerts some of its antitumor effects through enhanced T cell responses. Full activation of CD8+ T cells requires both expression of major histocompatibility complex class I molecules (HLA-ABC) and expression of the T cell costimulatory molecule CD80 or CD86; however, the impact of trastuzumab treatment on the expression of HLA-ABC and CD80 and CD86 has not been investigated in HER2-overexpressing breast cancer cells. In this study, we found that, while there is no direct correlation between the expression of HER2 and HLA-ABC in breast cancer, knockdown of HER2 or inhibition of HER2 kinase by lapatinib downregulated HLA-ABC expression. Trastuzumab had no meaningful effects on HLA-ABC expression in HER2-overexpressing breast cancer cells in monoculture; however, treatment of such cells with trastuzumab in co-culture with human peripheral blood mononuclear cells (PBMC) significantly upregulated not only HLA-ABC expression but also CD86 expression. We showed that this upregulation was mediated by interferon gamma (IFNγ) secreted from the natural killer (NK) cells in PBMC as a result of engagement of NK cells by trastuzumab. We further confirmed this effect of trastuzumab in vivo using a mouse mammary tumor model transduced to overexpress human HER2. Together, our data provide evidence that trastuzumab upregulates expression of HLA-ABC and T cell costimulatory molecules in HER2-overexpressing breast cancer cells in the presence of PBMC, which supports the view that T-cell-mediated immune responses are involved in trastuzumab-mediated antitumor effects.

8.
Article in English | MEDLINE | ID: mdl-23483844

ABSTRACT

The role of TNF-α in chlamydial clearance is uncertain. Antibody-mediated depletion of TNF-α in mice and guinea pigs has been shown not to significantly affect chlamydial clearance, whereas production of TNF-α in addition to IFN-γ from T cells has been shown to correlate with enhanced clearance. The aim of our study is to evaluate the mechanistic role of TNF-α in clearance of primary and secondary chlamydial infection from the genital tract (GT) using C57BL/6 TNF-α deficient (TNF-α(-/-)) and wild type (WT) mice. Chlamydial shedding from the lower GT was evaluated following primary and secondary intravaginal challenge. Also, antibody and antigen specific cytokine responses were analyzed from the infected GT and spleens, and oviduct pathology determined to analyze the role of TNF-α in upper GT pathological sequelae. MHC II(-/-) mice, known to display muted adaptive immune responses and failure to resolve genital chlamydial infections, were used as a negative control. Following both primary and secondary genital chlamydial infection, TNF-α(-/-) mice exhibited elevated granzyme B production, but similar IFN-γ and antibody responses. Importantly, absence of TNF-α did not significantly alter the resolution of infection. However, TNF-α(-/-) mice displayed significantly reduced upper genital tract (UGT) pathology compared to WT mice. This study demonstrates mechanistically that optimal chlamydial clearance following primary and secondary chlamydial genital infection can occur in the complete absence of TNF-α, and considered with the reduction of upper GT pathology in TNF-α(-/-) mice, suggests that targeted induction of anti-chlamydial TNF-α responses by vaccination may be unnecessary, and moreover could be potentially pathogenic.


Subject(s)
Chlamydia Infections/microbiology , Chlamydia muridarum/pathogenicity , Genital Diseases, Female/microbiology , Tumor Necrosis Factor-alpha/metabolism , Animals , Antibody Formation , Bacterial Shedding , Chlamydia Infections/pathology , Chlamydia muridarum/physiology , Cytokines/metabolism , Disease Models, Animal , Fallopian Tubes/microbiology , Fallopian Tubes/pathology , Female , Genital Diseases, Female/pathology , Granzymes/metabolism , Guinea Pigs , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Spleen/microbiology , Vagina/microbiology
9.
PLoS One ; 8(5): e64917, 2013.
Article in English | MEDLINE | ID: mdl-23741420

ABSTRACT

Chlamydia trachomatis is the most common bacterial sexually transmitted disease in the world and specifically in the United States, with the highest incidence in age-groups 14-19 years. In a subset of females, the C. trachomatis genital infection leads to serious pathological sequelae including pelvic inflammatory disease, ectopic pregnancy, and infertility. Chlamydia pneumoniae, another member of the same genus, is a common cause of community acquired respiratory infection with significant number of children aged 5-14 yr displaying sero-conversion. Since these bacteriae share several antigenic determinants, we evaluated whether intranasal immunization with live C. pneumoniae (1×10(6) inclusion forming units; IFU) in 5 week old female C57BL/6 mice would induce cross-species protection against subsequent intravaginal challenge with Chlamydia muridarum (5×10(4) IFU), which causes a similar genital infection and pathology in mice as C. trachomatis in humans. Mice vaccinated intranasally with live C. pneumoniae, but not mock (PBS) immunized animals, displayed high levels of splenic cellular antigen-specific IFN-γ production and serum antibody response against C. muridarum and C. trachomatis. Mice vaccinated with C. pneumoniae displayed a significant reduction in the vaginal C. muridarum shedding as early as day 12 after secondary i.vag. challenge compared to PBS (mock) immunized mice. At day 19 after C. muridarum challenge, 100% of C. pneumoniae vaccinated mice had cleared the infection compared to none (0%) of the mock immunized mice, which cleared the infection by day 27. At day 80 after C. muridarum challenge, C. pneumoniae vaccinated mice displayed a significant reduction in the incidence (50%) and degree of hydrosalpinx compared to mock immunized animals (100%). These results suggest that respiratory C. pneumoniae infection induces accelerated chlamydial clearance and reduction of oviduct pathology following genital C. muridarum challenge, and may have important implications to the C. trachomatis-induced reproductive disease in humans.


Subject(s)
Bacterial Vaccines/immunology , Chlamydia Infections/prevention & control , Chlamydia muridarum/immunology , Chlamydophila pneumoniae/immunology , Administration, Intranasal , Animals , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Bacterial Vaccines/administration & dosage , Chlamydia Infections/immunology , Chlamydia Infections/microbiology , Cross Reactions , Disease Models, Animal , Female , Interferon-gamma/immunology , Interferon-gamma/metabolism , Mice , Oviducts/immunology , Oviducts/pathology , Spleen/immunology , Spleen/metabolism , Vaccination , Vagina/immunology , Vagina/microbiology , Vagina/pathology
10.
PLoS One ; 6(2): e16847, 2011 Feb 09.
Article in English | MEDLINE | ID: mdl-21347387

ABSTRACT

The role of antigen-specific secretory IgA (SIgA) has been studied extensively, whereas there is a limited body of evidence regarding the contribution of non-specific SIgA to innate immune defenses against invading pathogens. In this study, we evaluated the effects of non-specific SIgA against infection with Vibrio cholerae O139 strain MO10 and biofilm formation. Seven day old infant mice deficient in IgA (IgA(-/-) mice) displayed significantly greater intestinal MO10 burden at 24 hr post-challenge when compared to IgA(+/+) pups. Importantly, cross-fostering of IgA(-/-) pups with IgA(+/+) nursing dams reversed the greater susceptibility to MO10 infection, suggesting a role for non-specific SIgA in protection against the infection. Since biofilm formation is associated with virulence of MO10, we further examined the role of human non-specific SIgA on this virulence phenotype of the pathogen. Human non-specific SIgA, in a dose-dependent fashion, significantly reduced the biofilm formation by MO10 without affecting the viability of the bacterium. Such an inhibitory effect was not induced by human serum IgA, IgG, or IgM, suggesting a role for the oligosaccharide-rich secretory component (SC) of SIgA. This was supported by the demonstration that SIgA treated with endoglycosidase H, to cleave the high-mannose containing terminal chitobiose residues, did not induce a reduction in biofilm formation by MO10. Furthermore, the addition of free mannose per se, across a wide dose range, induced significant reduction in MO10 biofilm formation. Collectively, these results suggest that mannose containing oligosaccharides within human non-specific secretory IgA can alter important virulence phenotypes of Vibrio cholerae such as biofilm formation, without affecting viability of the microorganism. Such effects may contribute significantly to innate immune defenses against invading pathogens in vivo in the gastrointestinal tract.


Subject(s)
Biofilms/drug effects , Biofilms/growth & development , Immunoglobulin A, Secretory/chemistry , Mannose , Oligosaccharides/chemistry , Oligosaccharides/pharmacology , Vibrio cholerae/physiology , Animals , Female , Humans , Immunoglobulin A, Secretory/metabolism , Immunoglobulin A, Secretory/pharmacology , Intestinal Mucosa/metabolism , Intestines/drug effects , Intestines/microbiology , Mice , Milk/chemistry , Vibrio cholerae/drug effects
11.
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
12.
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
SELECTION OF CITATIONS
SEARCH DETAIL