Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Article in English | MEDLINE | ID: mdl-30782995

ABSTRACT

We have identified recombinant human cystatins 9 (rCST9) and C (rCSTC) as a combination immunotherapeutic treatment against multidrug-resistant (MDR) New Delhi metallo-ß-lactamase-1 (NDM-1)-producing Klebsiella pneumoniae We evaluated the lasting protection of rCST9/rCSTC treatment against MDR NDM-1 K. pneumoniae pneumonia. Results showed that rCST9/rCSTC treatment modulated endogenous serum biomarkers, cystatins 9 and C and amyloid A, associated with poor patient outcomes and provided prophylactic and long-term protection in a murine model of pneumonia.


Subject(s)
Cystatins/blood , Inflammation/blood , Animals , Cystatin C/metabolism , Immunomodulation/drug effects , Immunotherapy , Klebsiella pneumoniae/drug effects , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Serum Amyloid A Protein/metabolism
2.
Article in English | MEDLINE | ID: mdl-29229643

ABSTRACT

Multidrug-resistant (MDR) bacterial pneumonia can induce dysregulated pulmonary and systemic inflammation leading to morbidity and mortality. Antibiotics to treat MDR pathogens do not function to modulate the extent and intensity of inflammation and can have serious side effects. Here we evaluate the efficacy of two human cysteine proteinase inhibitors, cystatin 9 (CST9) and cystatin C (CSTC), as a novel immunotherapeutic treatment to combat MDR New Delhi metallo-beta-lactamase-1 (NDM-1)-producing Klebsiella pneumoniae Our results showed that mice infected intranasally (i.n.) with a 90% lethal dose (LD90) challenge of NDM-1 K. pneumoniae and then treated with the combination of human recombinant CST9 (rCST9) and rCSTC (rCSTs; 50 pg of each i.n. at 1 h postinfection [p.i.] and/or 500 pg of each intraperitoneally [i.p.] at 3 days p.i.) had significantly improved survival compared to that of infected mice alone or infected mice treated with individual rCSTs (P < 0.05). Results showed that both of our optimal rCST treatment regimens modulated pulmonary and systemic proinflammatory cytokine secretion in the serum, lungs, liver, and spleen in infected mice (P < 0.05). Treatment also significantly decreased the bacterial burden (P < 0.05) while preserving lung integrity, with reduced inflammatory cell accumulation compared to that in infected mice. Further, rCST treatment regimens reduced lipid peroxidation and cell apoptosis in the lungs of infected mice. Additionally, in vitro studies showed that rCSTs (50 or 500 pg of each) directly decreased the viability of NDM-1 K. pneumoniae In conclusion, the data showed that rCST9/rCSTC worked synergistically to modulate host inflammation against MDR NDM-1 K. pneumoniae pneumonia, which significantly improved survival. Therefore, rCST9/rCSTC is a promising therapeutic candidate for the treatment of bacterial pneumonia.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Cystatin C/therapeutic use , Cystatins/therapeutic use , Klebsiella Infections/drug therapy , Klebsiella Infections/metabolism , Klebsiella pneumoniae/drug effects , beta-Lactamases/metabolism , Animals , Drug Resistance, Multiple, Bacterial , Female , Humans , Immunotherapy/methods , Klebsiella Infections/microbiology , Klebsiella pneumoniae/pathogenicity , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests
3.
Immunol Cell Biol ; 95(5): 454-460, 2017 05.
Article in English | MEDLINE | ID: mdl-27990018

ABSTRACT

We have comprehensively demonstrated using the mouse model that intranasal immunization with recombinant chlamydial protease-like activity factor (rCPAF) leads to a significant reduction in bacterial burden, genital tract pathology and preserves fertility following intravaginal genital chlamydial challenge. In the present report, we evaluated the protective efficacy of rCPAF immunization in guinea pigs, a second animal model for genital chlamydial infection. Using a vaccination strategy similar to the mouse model, we intranasally immunized female guinea pigs with rCPAF plus CpG deoxynucleotides (CpG; as an adjuvant), and challenged intravaginally with C. trachomatis serovar D (CT-D). Immunization with rCPAF/CpG significantly reduced vaginal CT-D shedding and induced resolution of infection by day 24, compared with day 33 in CpG alone treated and challenged animals. Immunization induced robust anti-rCPAF serum IgG 2 weeks following the last immunization, and was sustained at a high-level 4 weeks post challenge. Upregulation of antigen-specific IFN-γ gene expression was observed in rCPAF/CpG-vaccinated splenocytes. Importantly, a significant reduction in inflammation in the genital tissue in rCPAF/CpG-immunized guinea pigs compared with CpG-immunized animals was observed. Taken together, this study provides evidence of the protective efficacy of rCPAF as a vaccine candidate in a second animal model of genital chlamydial infection.


Subject(s)
Chlamydia Infections/immunology , Chlamydia Infections/microbiology , Chlamydia trachomatis/physiology , Endopeptidases/immunology , Animals , Chlamydia Infections/genetics , Gene Expression Regulation , Genitalia/microbiology , Genitalia/pathology , Guinea Pigs , Immunization , Immunoglobulin G/immunology , Interferon-gamma/genetics , Interferon-gamma/metabolism , Oligodeoxyribonucleotides/immunology
4.
Proteomics ; 16(9): 1347-60, 2016 05.
Article in English | MEDLINE | ID: mdl-27134121

ABSTRACT

Chlamydia trachomatis is the leading causative agent of bacterial sexually transmitted infections worldwide which can lead to female pelvic inflammatory disease and infertility. A greater understanding of host response during chlamydial infection is essential to design intervention technique to reduce the increasing incidence rate of genital chlamydial infection. In this study, we investigated proteome changes in epithelial cells during C. trachomatis infection by using an isobaric tags for relative and absolute quantitation (iTRAQ) labeling technique coupled with a liquid chromatography-tandem mass spectrometry (LC-MS(3) ) analysis. C. trachomatis (serovar D, MOI 1)-infected HeLa-229 human cervical carcinoma epithelial cells (at 2, 4 and 8 h) showed profound modifications of proteome profile which involved 606 host proteins. MGST1, SUGP2 and ATXN10 were among the top in the list of the differentially upregulated protein. Through pathway analysis, we suggested the involvement of eukaryotic initiation factor 2 (eIF2) and mammalian target of rapamycin (mTOR) in host cells upon C. trachomatis infection. Network analysis underscored the participation of DNA repair mechanism during C. trachomatis infection. In summary, intense modifications of proteome profile in C. trachomatis-infected HeLa-229 cells indicate complex host-pathogen interactions at early phase of chlamydial infection.


Subject(s)
Chlamydia trachomatis/growth & development , Eukaryotic Initiation Factor-2/genetics , Host-Pathogen Interactions , TOR Serine-Threonine Kinases/genetics , Ataxin-10/genetics , Ataxin-10/metabolism , Chlamydia trachomatis/pathogenicity , Chromatography, Liquid , Eukaryotic Initiation Factor-2/metabolism , Gene Expression Profiling , Gene Expression Regulation , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , HeLa Cells , Humans , Metabolic Networks and Pathways/genetics , Proteomics/methods , Serine-Arginine Splicing Factors/genetics , Serine-Arginine Splicing Factors/metabolism , Signal Transduction , Staining and Labeling/methods , TOR Serine-Threonine Kinases/metabolism , Tandem Mass Spectrometry , Time Factors
5.
PLoS Pathog ; 10(10): e1004439, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25340543

ABSTRACT

Francisella tularensis causes the disease tularemia. Human pulmonary exposure to the most virulent form, F. tularensis subsp. tularensis (Ftt), leads to high morbidity and mortality, resulting in this bacterium being classified as a potential biothreat agent. However, a closely-related species, F. novicida, is avirulent in healthy humans. No tularemia vaccine is currently approved for human use. We demonstrate that a single dose vaccine of a live attenuated F. novicida strain (Fn iglD) protects against subsequent pulmonary challenge with Ftt using two different animal models, Fischer 344 rats and cynomolgus macaques (NHP). The Fn iglD vaccine showed protective efficacy in rats, as did a Ftt iglD vaccine, suggesting no disadvantage to utilizing the low human virulent Francisella species to induce protective immunity. Comparison of specific antibody profiles in vaccinated rat and NHP sera by proteome array identified a core set of immunodominant antigens in vaccinated animals. This is the first report of a defined live attenuated vaccine that demonstrates efficacy against pulmonary tularemia in a NHP, and indicates that the low human virulence F. novicida functions as an effective tularemia vaccine platform.


Subject(s)
Bacterial Vaccines/immunology , Francisella tularensis , Immunodominant Epitopes/immunology , Tularemia/immunology , Animals , Macaca fascicularis , Mice , Models, Animal , Rats, Inbred F344 , Tularemia/mortality , Tularemia/prevention & control , Vaccination , Vaccines, Attenuated/immunology
6.
BMC Microbiol ; 16: 45, 2016 Mar 18.
Article in English | MEDLINE | ID: mdl-26987367

ABSTRACT

BACKGROUND: The 7.5 kb cryptic plasmid of Chlamydia trachomatis has been shown to be a virulence factor in animal models, but its significance in humans still remains unknown. The aim of this study was to investigate the prevalence and potential involvement of the C. trachomatis cryptic plasmid in causing various clinical manifestations; including infertility, reproductive tract disintegrity, menstrual disorder, and polycystic ovarian syndrome (PCOS) among genital C. trachomatis-infected patients. RESULTS: A total of 180 female patients of child bearing age (mean 30.9 years old, IQR:27-35) with gynecological complications and subfertility issues, who visited Obstetrics and Gynecology clinics in Kuala Lumpur, Malaysia were recruited for the study. Prevalence of genital chlamydial infection among these patients was alarmingly high at 51.1% (92/180). Of the 92 chlamydia-infected patients, 93.5% (86/92) were infected with plasmid-bearing (+) C. trachomatis while the remaining 6.5% (6/92) were caused by the plasmid-free (-) variant. Our data showed that genital C. trachomatis infection was associated with infertility issues, inflammation in the reproductive tract (mucopurulent cervicitis or endometriosis), irregular menstrual cycles and polycystic ovarian syndrome (PCOS). However, no statistical significance was detected among patients with plasmid (+) versus plasmid (-) C. trachomatis infection. Interestingly, plasmid (+) C. trachomatis was detected in all patients with PCOS, and the plasmid copy numbers were significantly higher among PCOS patients, relative to non-PCOS patients. CONCLUSION: Our findings show a high incidence of C. trachomatis infection among women with infertility or gynecological problems in Malaysia. However, due to the low number of plasmid (-) C. trachomatis cases, a significant role of the plasmid in causing virulence in human requires further investigation of a larger cohort.


Subject(s)
Chlamydia Infections/microbiology , Chlamydia trachomatis/genetics , Chlamydia trachomatis/isolation & purification , Plasmids/genetics , Adult , Chlamydia trachomatis/classification , Chlamydia trachomatis/metabolism , Cohort Studies , Female , Gynecology/statistics & numerical data , Humans , Malaysia/epidemiology , Obstetrics/statistics & numerical data , Plasmids/metabolism , Pregnancy , Prevalence
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.
Cell Immunol ; 295(2): 83-91, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25863744

ABSTRACT

The human pathogen Chlamydia pneumoniae has been implicated in chronic inflammatory diseases including type 2 diabetes. Therefore, we designed a study to evaluate pancreatic beta cells and mast cells during chlamydial infection. Our study revealed that C. pneumoniae infected mast cells significantly (p<0.005) decreased beta cell ATP and insulin production, in contrast to uninfected mast cells co-cultured with beta cells. Infected mast cells exhibited pyknotic nuclei and active caspase-3 and caspase-1 expression. Additionally, ex vivo analyses of tissues collected from C. pneumoniae infected mice showed increased interleukin-1ß production in splenocytes and pancreatic tissues as was observed with in vitro mast cell-beta cell co-cultures during C. pneumoniae infection. Notably, infected mast cells promoted beta cell destruction. Our findings reveal the negative effect of C. pneumoniae on mast cells, and the consequential impact on pancreatic beta cell function and viability.


Subject(s)
Chlamydia Infections/immunology , Chlamydophila pneumoniae/immunology , Diabetes Mellitus, Type 2/microbiology , Insulin-Secreting Cells/microbiology , Mast Cells/microbiology , Animals , Caspase 1/analysis , Caspase 3/analysis , Cell Survival/immunology , Chlamydia Infections/microbiology , Coculture Techniques , Diabetes Mellitus, Type 2/immunology , Flow Cytometry , Insulin-Secreting Cells/immunology , Interleukin-1beta/analysis , Liver/cytology , Mast Cells/immunology , Mice, Inbred C57BL , Mice, Obese , Microscopy, Confocal , Microscopy, Electron, Scanning , Specific Pathogen-Free Organisms , Spleen/cytology
9.
Infect Immun ; 82(9): 3910-8, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25001601

ABSTRACT

Multidrug-resistant Acinetobacter baumannii is among the most prevalent bacterial pathogens associated with trauma-related wound and bloodstream infections. Although septic shock and disseminated intravascular coagulation have been reported following fulminant A. baumannii sepsis, little is known about the protective host immune response to this pathogen. In this study, we examined the role of PTX3, a soluble pattern recognition receptor with reported antimicrobial properties and stored within neutrophil granules. PTX3 production by murine J774a.1 macrophages was assessed following challenge with A. baumannii strains ATCC 19606 and clinical isolates (CI) 77, 78, 79, 80, and 86. Interestingly, only CI strains 79, 80, and 86 induced PTX3 synthesis in murine J774a.1 macrophages, with greatest production observed following CI 79 and 86 challenge. Subsequently, C57BL/6 mice were challenged intraperitoneally with CI 77 and 79 to assess the role of PTX3 in vivo. A. baumannii strain CI 79 exhibited significantly (P < 0.0005) increased mortality, with an approximate 50% lethal dose (LD50) of 10(5) CFU, while an equivalent dose of CI 77 exhibited no mortality. Plasma leukocyte chemokines (KC, MCP-1, and RANTES) and myeloperoxidase activity were also significantly elevated following challenge with CI 79, indicating neutrophil recruitment/activation associated with significant elevation in serum PTX3 levels. Furthermore, 10-fold-greater PTX3 levels were observed in mouse serum 12 h postchallenge, comparing CI 79 to CI 77 (1,561 ng/ml versus 145 ng/ml), with concomitant severe pathology (liver and spleen) and coagulopathy. Together, these results suggest that elevation of PTX3 is associated with fulminant disease during A. baumannii sepsis.


Subject(s)
Acinetobacter baumannii/immunology , C-Reactive Protein/immunology , Nerve Tissue Proteins/immunology , Sepsis/immunology , Shock, Septic/immunology , Acinetobacter Infections/blood , Acinetobacter Infections/immunology , Acinetobacter Infections/microbiology , Animals , Cell Line , Chemokines/blood , Macrophages/immunology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Monocytes/immunology , Monocytes/microbiology , Nerve Tissue Proteins/blood , Neutrophils/immunology , Neutrophils/microbiology , Peroxidase/blood , Sepsis/blood , Sepsis/microbiology , Shock, Septic/blood , Shock, Septic/mortality
10.
J Immunol ; 188(11): 5604-11, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22529298

ABSTRACT

TLR signaling is critical for early host defense against pathogens, but the contributions of mast cell TLR-mediated mechanisms and subsequent effector functions during pulmonary infection are largely unknown. We have previously demonstrated that mast cells, through the production of IL-4, effectively control Francisella tularensis replication. In this study, the highly human virulent strain of F. tularensis SCHU S4 and the live vaccine strain were used to investigate the contribution of mast cell/TLR regulation of Francisella. Mast cells required TLR2 for effective bacterial killing, regulation of the hydrolytic enzyme cathepsin L, and for coordination and trafficking of MHC class II and lysosomal-associated membrane protein 2. Infected TLR2(-/-) mast cells, in contrast to wild-type and TLR4(-/-) cells, lacked detectable IL-4 and displayed increased cell death with a 2-3 log increase of F. tularensis replication, but could be rescued with rIL-4 treatment. Importantly, MHC class II and lysosomal-associated membrane protein 2 localization with labeled F. tularensis in the lungs was greater in wild-type than in TLR2(-/-) mice. These results provide evidence for the important effector contribution of mast cells and TLR2-mediated signaling on early innate processes in the lung following pulmonary F. tularensis infection and provide additional insight into possible mechanisms by which intracellular pathogens modulate respiratory immune defenses.


Subject(s)
Francisella tularensis/growth & development , Francisella tularensis/immunology , Mast Cells/immunology , Mast Cells/metabolism , Signal Transduction/immunology , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 2/physiology , Animals , Cell Death/genetics , Cell Death/immunology , Interleukin-4/deficiency , Mast Cells/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Transport/genetics , Protein Transport/immunology , Signal Transduction/genetics , Toll-Like Receptor 4/physiology , Tularemia/immunology , Tularemia/microbiology , Tularemia/prevention & control
11.
Vaccines (Basel) ; 12(4)2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38675740

ABSTRACT

Multi-drug-resistant (MDR) Acinetobacter baumannii is an opportunistic pathogen associated with hospital-acquired infections. Due to its environmental persistence, virulence, and limited treatment options, this organism causes both increased patient mortality and incurred healthcare costs. Thus, prophylactic vaccination could be ideal for intervention against MDR Acinetobacter infection in susceptible populations. In this study, we employed immunoinformatics to identify peptides containing both putative B- and T-cell epitopes from proteins associated with A. baumannii pathogenesis. A novel Acinetobacter Multi-Epitope Vaccine (AMEV2) was constructed using an A. baumannii thioredoxin A (TrxA) leading protein sequence followed by five identified peptide antigens. Antisera from A. baumannii infected mice demonstrated reactivity to rAMEV2, and subcutaneous immunization of mice with rAMEV2 produced high antibody titer against the construct as well as peptide components. Immunization results in increased frequency of IL-4-secreting splenocytes indicative of a Th2 response. AMEV2-immunized mice were protected against intranasal challenge with a hypervirulent strain of A. baumannii and demonstrated reduced bacterial burden at 48 h. In contrast, all mock vaccinated mice succumbed to infection within 3 days. Results presented here provide insight into the effectiveness of immunoinformatic-based vaccine design and its potential as an effective strategy to combat the rise of MDR pathogens.

12.
J Biol Chem ; 287(44): 37185-94, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-22923614

ABSTRACT

Following pulmonary infection with Francisella tularensis, we observed an unexpected but significant reduction of alkaline phosphatase, an enzyme normally up-regulated following inflammation. However, no reduction was observed in mice infected with a closely related gram-negative pneumonic organism (Klebsiella pneumoniae) suggesting the inhibition may be Francisella-specific. In similar fashion to in vivo observations, addition of Francisella lysate to exogenous alkaline phosphatase (tissue-nonspecific isozyme) was inhibitory. Partial purification and subsequent proteomic analysis indicated the inhibitory factor to be the heat shock protein DnaK. Incubation with increasing amounts of anti-DnaK antibody reduced the inhibitory effect in a dose-dependent manner. Furthermore, DnaK contains an adenosine triphosphate binding domain at its N terminus, and addition of adenosine triphosphate enhances dissociation of DnaK with its target protein, e.g. alkaline phosphatase. Addition of adenosine triphosphate resulted in decreased DnaK co-immunoprecipitated with alkaline phosphatase as well as reduction of Francisella-mediated alkaline phosphatase inhibition further supporting the binding of Francisella DnaK to alkaline phosphatase. Release of DnaK via secretion and/or bacterial cell lysis into the extracellular milieu and inhibition of plasma alkaline phosphatase could promote an orchestrated, inflammatory response advantageous to Francisella.


Subject(s)
Alkaline Phosphatase/blood , Bacteremia/microbiology , Bacterial Proteins/physiology , Francisella/physiology , HSP70 Heat-Shock Proteins/physiology , Tularemia/microbiology , Adenosine Triphosphate/chemistry , Alkaline Phosphatase/antagonists & inhibitors , Animals , Bacterial Load , Bacterial Proteins/chemistry , Bacterial Proteins/isolation & purification , Bacterial Proteins/metabolism , Chaperonin 60/metabolism , Chromatography, DEAE-Cellulose , Female , HSP70 Heat-Shock Proteins/chemistry , HSP70 Heat-Shock Proteins/isolation & purification , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Host-Pathogen Interactions , Mice , Mice, Inbred BALB C , Molecular Weight , Peptide Fragments/chemistry , Protein Binding , Spectrometry, Mass, Electrospray Ionization
13.
J Immunol ; 186(5): 2750-6, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21270409

ABSTRACT

Ab-mediated blockade of the adhesion molecule VLA-4 has been shown to ameliorate disease in human multiple sclerosis patients and experimental autoimmune encephalomyelitis (EAE) animal models. We wanted to determine whether anti-VLA-4 Ab treatment affected the function and persistence of autoreactive T cells in mice with EAE. Unexpectedly, we observed a high level of mortality in anti-VLA-4 mAb (PS/2)-treated mice with actively induced EAE despite decreased disease severity. Investigation of the underlying mechanism showed that injection of PS/2 mAb in combination with pertussis toxin resulted in anaphylaxis and mortality. Furthermore, the data showed that CD4(+) T cells were required for this effect and suggested a role for IL-1ß and TNF-α in the underlying pathology. The results reveal a previously not appreciated deleterious effect of anti-VLA-4 Ab treatment in combination with exposure to pertussis toxin.


Subject(s)
Anaphylaxis/immunology , Anaphylaxis/mortality , Antibodies, Monoclonal/administration & dosage , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/mortality , Integrin alpha4beta1/immunology , Pertussis Toxin/administration & dosage , Anaphylaxis/genetics , Animals , Antibodies, Blocking/administration & dosage , Drug Combinations , Encephalomyelitis, Autoimmune, Experimental/genetics , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Myelin Proteins , Myelin-Associated Glycoprotein/administration & dosage , Myelin-Associated Glycoprotein/immunology , Myelin-Oligodendrocyte Glycoprotein , Survival Analysis , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/genetics
14.
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
15.
Int J Microbiol ; 2022: 6082140, 2022.
Article in English | MEDLINE | ID: mdl-35498397

ABSTRACT

Fetuin-A is an acute phase glycoprotein shown to counter in a regulatory manner proinflammatory cytokine production to maintain homeostasis during inflammation. We report here that in wild-type mice 12 days after Chlamydia muridarum (Cm) intranasal challenge, fetuin-A content in the lungs decreased 46%, while INF-γ increased 44%, consistent with a negative regulatory role of fetuin-A in inflammation. Importantly, the observed increased IFN-γ production was abrogated in fetuin-A-deficient AHSG mice suggesting that IFN-γ induction following Cm infection is fetuin-A dependent. Assessment of expression of genes associated with inflammation revealed fetuin-A-dependent upregulation of TBX21 (a Th1 cell-specific transcription factor) in the lungs of Cm-infected WT mice that correlated with IFN-γ induction. Additionally, the effect of fetuin-A deficiency in mounting an adaptive immune response to Cm infection was demonstrated using a splenocyte recall assay. Although preliminary in nature, these findings are suggestive of fetuin-A involvement following Cm pulmonary infection and underscores the need to investigate further the role of fetuin-A in the immune response and the consequences of its gene deletion.

16.
Front Immunol ; 13: 1070424, 2022.
Article in English | MEDLINE | ID: mdl-36466845

ABSTRACT

Acinetobacter baumannii is a nosocomic opportunistic Gram-negative bacteria known for its extensive drug-resistant phenotype. A. baumannii hospital-acquired infections are major contributors to increased costs and mortality observed during the COVID-19 pandemic. With few effective antimicrobials available for treatment of this pathogen, immune-based therapy becomes an attractive strategy to combat multi-drug resistant Acinetobacter infection. Immunotherapeutics is a field of growing interest with advances in vaccines and monoclonal antibodies providing insight into the protective immune response required to successfully combat this pathogen. This review focuses on current knowledge describing the adaptive immune response to A. baumannii, the importance of antibody-mediated protection, developments in cell-mediated protection, and their respective therapeutic application going forward. With A. baumannii's increasing resistance to most current antimicrobials, elucidating an effective host adaptive immune response is paramount in the guidance of future immunotherapeutic development.


Subject(s)
Acinetobacter baumannii , COVID-19 Drug Treatment , Cross Infection , Humans , Pandemics , Antibodies, Monoclonal
17.
Exp Biol Med (Maywood) ; 247(12): 1055-1060, 2022 06.
Article in English | MEDLINE | ID: mdl-35369776

ABSTRACT

Understanding the immune response to SARS-CoV-2 is important for development of effective diagnostics and vaccines. We report here a broad antibody response to SARS-CoV-2 spike protein receptor binding domain (RBD) in 100 convalescent patient plasma samples. Antibody isotypes IgA, IgM, and IgG exhibited significantly higher anti-RBD titers when compared to SARS-CoV-2 negative controls. IgG subtyping indicated IgG1 and IgG3 to be most abundant. Greater than 90 % of SARS-CoV-2 positive plasma samples tested exhibited significant neutralization capacity using a surrogate virus neutralization assay. Of the IgG subclasses, IgG1 and IgG3 exhibited the highest viral neutralization capacity; whereas, IgG2 and IgG4 viral neutralization was not observed. Comparison of SARS-CoV-2 elicited total IgG binding to emerging variant (alpha, beta, and delta) RBDs indicated decreased binding. Furthermore, neutralization by SARS-CoV-2 convalescent plasma of delta and omicron variant RBDs was significantly decreased suggesting that neutralizing antibodies in convalescent plasma are less effective in inhibiting variants currently in circulation.


Subject(s)
COVID-19 , Immunity, Humoral , Antibodies, Neutralizing , Antibodies, Viral , COVID-19/therapy , Humans , Immunization, Passive , Immunoglobulin A , Immunoglobulin G , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , COVID-19 Serotherapy
18.
Exp Biol Med (Maywood) ; 247(3): 282-288, 2022 02.
Article in English | MEDLINE | ID: mdl-34713732

ABSTRACT

Acinetobacter baumannii is a Gram-negative bacterium responsible for many hospital-acquired infections including ventilator-associated pneumonia and sepsis. We have previously identified A. baumannii thioredoxin A protein (TrxA) as a virulence factor with a multitude of functions including reduction of protein disulfides. TrxA plays an important role in resistance to oxidative stress facilitating host immune evasion in part by alteration of type IV pili and cell surface hydrophobicity. Other virulence factors such as outer membrane vesicles (OMV) shed by bacteria have been shown to mediate bacterial intercellular communication and modulate host immune response. To investigate whether OMVs can be modulated by TrxA, we isolated OMVs from wild type (WT) and TrxA-deficient (ΔtrxA) A. baumannii clinical isolate Ci79 and carried out a functional and proteomic comparison. Despite attenuation of ΔtrxA in a mouse challenge model, pulmonary inoculation of ΔtrxA OMVs resulted in increased lung permeability compared to WT OMVs. Furthermore, ΔtrxA OMVs induced more J774 macrophage-like cell death than WT OMVs. This ΔtrxA OMV-mediated cell death was abrogated when cells were incubated with protease-K-treated OMVs suggesting OMV proteins were responsible for cytotoxicity. We therefore compared WT and mutant OMV proteins using proteomic analysis. We observed that up-regulated and unique ΔtrxA OMV proteins consisted of many membrane bound proteins involved in small molecule transport as well as proteolytic activity. Bacterial OmpA, metalloprotease, and fimbrial protein have been shown to enhance mammalian cell apoptosis through various mechanisms. Differential packaging of these proteins in ΔtrxA OMVs may contribute to the increased cytotoxicity observed in this study.


Subject(s)
Acinetobacter baumannii/pathogenicity , Bacterial Outer Membrane Proteins/metabolism , Bacterial Outer Membrane/pathology , Thioredoxins/metabolism , Acinetobacter Infections/microbiology , Acinetobacter Infections/pathology , Acinetobacter baumannii/isolation & purification , Animals , Bacterial Outer Membrane/metabolism , Extracellular Vesicles/pathology , Host-Pathogen Interactions/physiology , Humans , Lung/microbiology , Lung/pathology , Mice, Inbred C57BL , Thioredoxins/genetics , Virulence Factors/genetics , Virulence Factors/metabolism
19.
Infect Immun ; 79(6): 2356-61, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21464086

ABSTRACT

Francisella tularensis is able to survive and replicate within host macrophages, a trait that is associated with the high virulence of this bacterium. The trpAB genes encode the enzymes required for the final two steps in tryptophan biosynthesis, with TrpB being responsible for the conversion of indole to tryptophan. Consistent with this function, an F. tularensis subsp. novicida trpB mutant is unable to grow in defined medium in the absence of tryptophan. The trpB mutant is also attenuated for virulence in a mouse pulmonary model of tularemia. However, the trpB mutant remains virulent in gamma interferon receptor-deficient (IFN-γR(-/-)) mice, demonstrating that IFN-γ-mediated signaling contributes to clearance of the trpB mutant. IFN-γ limits intracellular survival of the trpB mutant within bone marrow-derived macrophages from wild-type but not IFN-γR(-/-) mice. An F. tularensis subsp. tularensis trpB mutant is also attenuated for virulence in mice and survival within IFN-γ-treated macrophages, indicating that tryptophan prototrophy is also important in a human-virulent F. tularensis subspecies. These results demonstrate that trpB contributes to F. tularensis virulence by enabling intracellular growth under IFN-γ-mediated tryptophan limitation.


Subject(s)
Francisella tularensis/immunology , Interferon-gamma/physiology , Tryptophan/physiology , Tularemia/microbiology , Animals , Francisella tularensis/genetics , Francisella tularensis/physiology , Genes, Bacterial/genetics , Host-Pathogen Interactions/immunology , Macrophages/immunology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Tryptophan/biosynthesis , Tularemia/immunology
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL