Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters

Publication year range
1.
Infect Immun ; 88(4)2020 03 23.
Article in English | MEDLINE | ID: mdl-31964750

ABSTRACT

Human genital Chlamydia infection is a major public health concern due to the serious reproductive system complications. Chlamydia binds several receptor tyrosine kinases (RTKs) on host cells, including the epidermal growth factor receptor (EGFR), and activates cellular signaling cascades for host invasion, cytoskeletal remodeling, optimal inclusion development, and induction of pathogenic epithelial-mesenchyme transition (EMT). Chlamydia also upregulates transforming growth factor beta (TGF-ß) expression, whose signaling pathway synergizes with the EGFR cascade, but its role in infectivity, inclusions, and EMT induction is unknown. We hypothesized that the EGFR and TGF-ß signaling pathways cooperate during chlamydial infection for optimal inclusion development and stable EMT induction. The results revealed that Chlamydia upregulated TGF-ß expression as early as 6 h postinfection of epithelial cells and stimulated both the EGFR and TGF-ß signaling pathways. Inhibition of either the EGFR or TGF-ßR1 signaling substantially reduced inclusion development; however, the combined inhibition of both EGFR and TGF-ßR1 signaling reduced inclusions by over 90% and prevented EMT induction. Importantly, EGFR inhibition suppressed TGF-ß expression, and an inhibitory thrombospondin-1 (Tsp1)-based peptide inhibited chlamydia-induced EMT, revealing a major source of active TGF-ß during infection. Finally, TGF-ßR signaling inhibition suppressed the expression of transforming acidic coiled-coil protein-3 (TACC3), which stabilizes EGFR signaling, suggesting reciprocal regulation between TGF-ß and EGFR signaling during chlamydial infection. Thus, RTK-mediated host invasion by chlamydia upregulated TGF-ß expression and signaling, which cooperated with other cellular signaling cascades and cytoskeletal remodeling to support optimal inclusion development and EMT induction. This finding may provide new targets for chlamydial disease biomarkers and prevention.


Subject(s)
Chlamydia Infections/physiopathology , Chlamydia/growth & development , Epithelial Cells/microbiology , ErbB Receptors/metabolism , Host-Pathogen Interactions , Signal Transduction , Transforming Growth Factor beta/metabolism , Animals , Cell Line , Endocytosis , Epithelial-Mesenchymal Transition , Inclusion Bodies/microbiology , Mice , Models, Biological
2.
BMC Genomics ; 20(1): 143, 2019 Feb 18.
Article in English | MEDLINE | ID: mdl-30777008

ABSTRACT

BACKGROUND: Genital C. trachomatis infection may cause pelvic inflammatory disease (PID) that can lead to tubal factor infertility (TFI). Understanding the pathogenesis of chlamydial complications including the pathophysiological processes within the female host genital tract is important in preventing adverse pathology. MicroRNAs regulate several pathophysiological processes of infectious and non-infectious etiologies. In this study, we tested the hypothesis that the miRNA profile of single and repeat genital chlamydial infections will be different and that these differences will be time dependent. Thus, we analyzed and compared differentially expressed mice genital tract miRNAs after single and repeat chlamydia infections using a C. muridarum mouse model. Mice were sacrificed and their genital tract tissues were collected at 1, 2, 4, and 8 weeks after a single and repeat chlamydia infections. Histopathology, and miRNA sequencing were performed. RESULTS: Histopathology presentation showed that the oviduct and uterus of reinfected mice were more inflamed, distended and dilated compared to mice infected once. The miRNAs expression profile was different in the reproductive tissues after a reinfection, with a greater number of miRNAs expressed after reinfection. Also, the number of miRNAs expressed each week after chlamydia infection and reinfection varied, with weeks eight and one having the highest number of differentially expressed miRNAs for chlamydia infection and reinfection respectively. Ten miRNAs; mmu-miR-378b, mmu-miR-204-5p, mmu-miR-151-5p, mmu-miR-142-3p, mmu-miR-128-3p, mmu-miR-335-3p, mmu-miR-195a-3p, mmu-miR-142-5p, mmu-miR-106a-5p and mmu-miR-92a-3p were common in both primary chlamydia infection and reinfection. Pathway analysis showed that, amongst other functions, the differentially regulated miRNAs control pathways involved in cellular and tissue development, disease conditions and toxicity. CONCLUSIONS: This study provides insights into the changes in miRNA expression over time after chlamydia infection and reinfection, as well as the pathways they regulate to determine pathological outcomes. The miRNAs networks generated in our study shows that there are differences in the focus molecules involved in significant biological functions in chlamydia infection and reinfection, implying that chlamydial pathogenesis occurs differently for each type of infection and that this could be important when determining treatments regime and disease outcome. The study underscores the crucial role of host factors in chlamydia pathogenesis.


Subject(s)
Chlamydia Infections/genetics , Chlamydia Infections/microbiology , Chlamydia , Genitalia/microbiology , MicroRNAs/genetics , Transcriptome , Animals , Biopsy , Cell Line , Chlamydia Infections/pathology , Computational Biology/methods , Disease Models, Animal , Female , Gene Expression Profiling , Gene Expression Regulation , Genitalia/pathology , Humans , Immunohistochemistry , Mice
3.
Biochem Biophys Res Commun ; 508(2): 421-429, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30503337

ABSTRACT

The unfolded protein response (UPR) contributes to chlamydial pathogenesis, as a source of lipids and ATP during replication, and for establishing the initial anti-apoptotic state of host cell that ensures successful inclusion development. The molecular mechanism(s) of UPR induction by Chlamydia is unknown. Chlamydia use type III secretion system (T3SS) effector proteins (e.g, the Translocated Actin-Recruiting Phosphoprotein (Tarp) to stimulate host cell's cytoskeletal reorganization that facilitates invasion and inclusion development. We investigated the hypothesis that T3SS effector-mediated assembly of myosin-II complex produces activated non-muscle myosin heavy chain II (NMMHC-II), which then binds the UPR master regulator (BiP) and/or transducers to induce UPR. Our results revealed the interaction of the chlamydial effector proteins (CT228 and Tarp) with components of the myosin II complex and UPR regulator and transducer during infection. These interactions caused the activation and binding of NMMHC-II to BiP and IRE1α leading to UPR induction. In addition, specific inhibitors of myosin light chain kinase, Tarp oligomerization and myosin ATPase significantly reduced UPR activation and Chlamydia replication. Thus, Chlamydia induce UPR through T3SS effector-mediated activation of NMMHC-II components of the myosin complex to facilitate infectivity. The finding provides greater insights into chlamydial pathogenesis with the potential to identify therapeutic targets and formulations.


Subject(s)
Chlamydia muridarum/pathogenicity , Chlamydia trachomatis/pathogenicity , Host Microbial Interactions/physiology , Unfolded Protein Response/physiology , Animals , Chlamydia Infections/etiology , Chlamydia Infections/metabolism , Chlamydia Infections/microbiology , Chlamydia muridarum/metabolism , Chlamydia trachomatis/metabolism , HeLa Cells , Humans , Inclusion Bodies/metabolism , Mice , Myosin Type II/metabolism , Type III Secretion Systems/metabolism
4.
Infect Immun ; 86(1)2018 01.
Article in English | MEDLINE | ID: mdl-29084894

ABSTRACT

The reproductive system complications of genital chlamydial infection include fallopian tube fibrosis and tubal factor infertility. However, the molecular pathogenesis of these complications remains poorly understood. The induction of pathogenic epithelial-mesenchymal transition (EMT) through microRNA (miRNA) dysregulation was recently proposed as the pathogenic basis of chlamydial complications. Focusing on fibrogenesis, we investigated the hypothesis that chlamydia-induced fibrosis is caused by EMT-driven generation of myofibroblasts, the effector cells of fibrosis that produce excessive extracellular matrix (ECM) proteins. The results revealed that the targets of a major category of altered miRNAs during chlamydial infection are key components of the pathophysiological process of fibrogenesis; these target molecules include collagen types I, III, and IV, transforming growth factor ß (TGF-ß), TGF-ß receptor 1 (TGF-ßR1), connective tissue growth factor (CTGF), E-cadherin, SRY-box 7 (SOX7), and NFAT (nuclear factor of activated T cells) kinase dual-specificity tyrosine (Y) phosphorylation-regulated kinase 1a (Dyrk1a). Chlamydial induction of EMT resulted in the generation of α-smooth muscle actin (α-SMA)-positive myofibroblasts that produced ECM proteins, including collagen types I and III and fibronectin. Furthermore, the inhibition of EMT prevented the generation of myofibroblasts and production of ECM proteins during chlamydial infection. These findings may provide useful avenues for targeting EMT or specific components of the EMT pathways as a therapeutic intervention strategy to prevent chlamydia-related complications.


Subject(s)
Chlamydia Infections/complications , Chlamydia Infections/pathology , Chlamydia/pathogenicity , Epithelial-Mesenchymal Transition/physiology , Fibrosis/etiology , Fibrosis/pathology , Actins/metabolism , Animals , Cadherins/metabolism , Cell Line , Chlamydia Infections/microbiology , Collagen/metabolism , Connective Tissue Growth Factor/metabolism , Extracellular Matrix Proteins/metabolism , Fibronectins/metabolism , Fibrosis/microbiology , Mice , MicroRNAs/metabolism , Myofibroblasts/microbiology , Myofibroblasts/pathology , NFATC Transcription Factors/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism , SOXF Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism
5.
J Infect Dis ; 215(3): 456-465, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27932618

ABSTRACT

Chlamydia is an obligate intracellular bacterium that relies on host cells for essential nutrients and adenosine triphosphate (ATP) for a productive infection. Although the unfolded protein response (UPR) plays a major role in certain microbial infectivity, its role in chlamydial pathogenesis is unknown. We hypothesized that Chlamydia induces UPR and exploits it to upregulate host cell uptake and metabolism of glucose, production of ATP, phospholipids, and other molecules required for its replicative development and host survival. Using a combination of biochemical and pathway inhibition assays, we showed that the 3 UPR pathway transducers-protein kinase RNA-activated (PKR)-like ER kinase (PERK), inositol-requiring enzyme-1α (IRE1α), and activating transcription factor-6α (ATF6α)-were activated during Chlamydia infection. The kinase activity of PERK and ribonuclease (RNase) of IRE1α mediated the upregulation of hexokinase II and production of ATP via substrate-level phosphorylation. In addition, the activation of PERK and IRE1α promoted autophagy formation and apoptosis resistance for host survival. Moreover, the activation of IRE1α resulted in the generation of spliced X-box binding protein 1 (sXBP1) and upregulation of lipid production. The vital role of UPR pathways in Chlamydia development and pathogenesis could lead to the identification of potential molecular targets for therapeutics against Chlamydia.


Subject(s)
Chlamydia Infections/microbiology , Chlamydia/pathogenicity , Unfolded Protein Response , Activating Transcription Factor 6/metabolism , Animals , Apoptosis , Cell Survival , Chlamydia Infections/metabolism , Endoribonucleases/metabolism , Enzyme Activation , Female , Mice , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , eIF-2 Kinase/metabolism
6.
BMC Immunol ; 18(1): 27, 2017 05 19.
Article in English | MEDLINE | ID: mdl-28525970

ABSTRACT

BACKGROUND: We have previously reported that interleukin-10 (IL-10) deficient dendritic cells (DCs) are potent antigen presenting cells that induced elevated protective immunity against Chlamydia. To further investigate the molecular and biochemical mechanism underlying the superior immunostimulatory property of IL-10 deficient DCs we performed proteomic analysis on protein profiles from Chlamydia-pulsed wild-type (WT) and IL-10-/- DCs to identify differentially expressed proteins with immunomodulatory properties. RESULTS: The results showed that alpha enolase (ENO1), a metabolic enzyme involved in the last step of glycolysis was significantly upregulated in Chlamydia-pulsed IL-10-/- DCs compared to WT DCs. We further studied the immunoregulatory role of ENO1 in DC function by generating ENO1 knockdown DCs, using lentiviral siRNA technology. We analyzed the effect of the ENO1 knockdown on DC functions after pulsing with Chlamydia. Pyruvate assay, transmission electron microscopy, flow cytometry, confocal microscopy, cytokine, T-cell activation and adoptive transfer assays were also used to study DC function. The results showed that ENO1 knockdown DCs had impaired maturation and activation, with significant decrease in intracellular pyruvate concentration as compared with the Chlamydia-pulsed WT DCs. Adoptive transfer of Chlamydia-pulsed ENO1 knockdown DCs were poorly immunogenic in vitro and in vivo, especially the ability to induce protective immunity against genital chlamydia infection. The marked remodeling of the mitochondrial morphology of Chlamydia-pulsed ENO1 knockdown DCs compared to the Chlamydia-pulsed WT DCs was associated with the dysregulation of translocase of the outer membrane (TOM) 20 and adenine nucleotide translocator (ANT) 1/2/3/4 that regulate mitochondrial permeability. The results suggest that an enhanced glycolysis is required for efficient antigen processing and presentation by DCs to induce a robust immune response. CONCLUSIONS: The upregulation of ENO1 contributes to the superior immunostimulatory function of IL-10 deficient DCs. Our studies indicated that ENO1 deficiency causes the reduced production of pyruvate, which then contributes to a dysfunction in mitochondrial homeostasis that may affect DC survival, maturation and antigen presenting properties. Modulation of ENO1 thus provides a potentially effective strategy to boost DC function and promote immunity against infectious and non-infectious diseases.


Subject(s)
Biomarkers, Tumor/genetics , Chlamydia Infections/immunology , Chlamydia trachomatis/immunology , DNA-Binding Proteins/genetics , Dendritic Cells/physiology , Genitalia/immunology , Phosphopyruvate Hydratase/genetics , Tumor Suppressor Proteins/genetics , Animals , Antigen Presentation , Biomarkers, Tumor/metabolism , Cell Membrane Permeability , Cells, Cultured , DNA-Binding Proteins/metabolism , Dendritic Cells/microbiology , Female , Genitalia/microbiology , Immunity, Innate , Interleukin-10/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Phosphopyruvate Hydratase/metabolism , Proteomics , Pyruvic Acid/metabolism , RNA, Small Interfering/genetics , Tumor Suppressor Proteins/metabolism , Up-Regulation
7.
J Clin Microbiol ; 55(7): 2035-2044, 2017 07.
Article in English | MEDLINE | ID: mdl-28404679

ABSTRACT

Infections of the central nervous system (CNS) are often acute, with significant morbidity and mortality. Routine diagnosis of such infections is limited in developing countries and requires modern equipment in advanced laboratories that may be unavailable to a number of patients in sub-Saharan Africa. We developed a TaqMan array card (TAC) that detects multiple pathogens simultaneously from cerebrospinal fluid. The 21-pathogen CNS multiple-pathogen TAC (CNS-TAC) assay includes two parasites (Balamuthia mandrillaris and Acanthamoeba), six bacterial pathogens (Streptococcus pneumoniae, Haemophilus influenzae, Neisseria meningitidis, Mycoplasma pneumoniae, Mycobacterium tuberculosis, and Bartonella), and 13 viruses (parechovirus, dengue virus, Nipah virus, varicella-zoster virus, mumps virus, measles virus, lyssavirus, herpes simplex viruses 1 and 2, Epstein-Barr virus, enterovirus, cytomegalovirus, and chikungunya virus). The card also includes human RNase P as a nucleic acid extraction control and an internal manufacturer control, GAPDH (glyceraldehyde-3-phosphate dehydrogenase). This CNS-TAC assay can test up to eight samples for all 21 agents within 2.5 h following nucleic acid extraction. The assay was validated for linearity, limit of detection, sensitivity, and specificity by using either live viruses (dengue, mumps, and measles viruses) or nucleic acid material (Nipah and chikungunya viruses). Of 120 samples tested by individual real-time PCR, 35 were positive for eight different targets, whereas the CNS-TAC assay detected 37 positive samples across nine different targets. The CNS-TAC assays showed 85.6% sensitivity and 96.7% specificity. Therefore, the CNS-TAC assay may be useful for outbreak investigation and surveillance of suspected neurological disease.


Subject(s)
Central Nervous System Infections/diagnosis , Microbiological Techniques/methods , Molecular Diagnostic Techniques/methods , Adolescent , Adult , Africa South of the Sahara , Aged , Aged, 80 and over , Amoebozoa/isolation & purification , Bacteria/isolation & purification , Central Nervous System Infections/microbiology , Central Nervous System Infections/parasitology , Central Nervous System Infections/virology , Child , Child, Preschool , Female , Humans , Infant , Male , Middle Aged , Reference Standards , Sensitivity and Specificity , Viruses/isolation & purification , Young Adult
8.
BMC Immunol ; 15: 584, 2014 Dec 31.
Article in English | MEDLINE | ID: mdl-25551828

ABSTRACT

BACKGROUND: We previously showed that the Vibrio cholerae ghost platform (VCG; empty V. cholerae cell envelopes) is an effective delivery system for vaccine antigens promoting the induction of substantial immunity in the absence of external adjuvants. However, the mechanism by which these cell envelopes enhance immunity and stimulate a predominantly Th1 cellular and humoral immune response has not been elucidated. We hypothesized that the immunostimulatory ability of VCG involves dendritic cell (DC) activation. OBJECTIVE: The aims of this study were: a) to investigate the ability of DCs [using mouse bone marrow-derived DCs (BMDCs) as a model system] to take up and internalize VCGs; b) to evaluate the immunomodulatory effect of internalized VCGs on DC activation and maturation and their functional capacity to present chlamydial antigen to naïve and infection-sensitized CD4+ T cells and; c) to evaluate the ability of VCGs to enhance the protective immunity of a chlamydial antigen. RESULTS: VCGs were efficiently internalized by DCs without affecting their viability and modulated DC-mediated immune responses. VCG-pulsed DCs showed increased secretion of proinflammatory cytokines and expression of co-stimulatory molecules associated with DC maturation in response to stimulation with UV-irradiated chlamydial elementary bodies (UV-EBs). Furthermore, this interaction resulted in effective chlamydial antigen presentation to infection-sensitized but not naïve CD4+ T cells and enhancement of protective immunity. CONCLUSIONS: The present study demonstrated that VCGs activate DCs leading to the surface expression of co-stimulatory molecules associated with DC activation and maturation and enhancement of protective immunity induced by a chlamydial antigen. The results indicate that the immunoenhancing activity of VCG for increased T-cell activation against antigens is mediated, at least in part, through DC triggering. Thus, VCGs could be harnessed as immunomodulators to target antigens to DCs for enhancement of protective immunity against microbial infections.


Subject(s)
Antigen Presentation , Antigens, Bacterial , Chlamydia trachomatis , Dendritic Cells/immunology , Th1 Cells/immunology , Vibrio cholerae , Animals , Antigens, Bacterial/chemistry , Antigens, Bacterial/immunology , Chlamydia trachomatis/chemistry , Chlamydia trachomatis/immunology , Female , HeLa Cells , Humans , Lymphocyte Activation , Mice , Vibrio cholerae/chemistry , Vibrio cholerae/immunology
9.
J Infect Dis ; 207(7): 1095-104, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23303804

ABSTRACT

Tubal factor infertility (TFI) represents 36% of female infertility and genital infection by Chlamydia trachomatis (C. trachomatis) is a major cause. Although TFI is associated with host inflammatory responses to bacterial components, the molecular pathogenesis of Chlamydia-induced infertility remains poorly understood. We investigated the hypothesis that activation of specific cysteine proteases, the caspases, during C. trachomatis genital infection causes the disruption of key fertility-promoting molecules required for embryo development and implantation. We analyzed the effect of caspase inhibition on infertility and the integrity of Dicer, a caspase-sensitive, fertility-promoting ribonuclease III enzyme, and key micro-RNAs in the reproductive system. Genital infection with the inflammation- and caspase-inducing, wild-type C. trachomatis serovar L2 led to infertility, but the noninflammation-inducing, plasmid-free strain did not. We confirmed that caspase-mediated apoptotic tissue destruction may contribute to chlamydial pathogenesis. Caspase-1 or -3 deficiency, or local administration of the pan caspase inhibitor, Z-VAD-FMK into normal mice protected against Chlamydia-induced infertility. Finally, the oviducts of infected infertile mice showed evidence of caspase-mediated cleavage inactivation of Dicer and alteration in critical miRNAs that regulate growth, differentiation, and development, including mir-21. These results provide new insight into the molecular pathogenesis of TFI with significant implications for new strategies for treatment and prevention of chlamydial complications.


Subject(s)
Caspase 1/metabolism , Caspase 3/metabolism , Chlamydia trachomatis/pathogenicity , Infertility, Female/microbiology , Infertility, Female/prevention & control , Pregnancy Complications, Infectious/prevention & control , Animals , Apoptosis , Caspase 1/genetics , Caspase 3/genetics , Chlamydia Infections/enzymology , Chlamydia Infections/microbiology , Chlamydia Infections/pathology , Enzyme Activation , Female , HeLa Cells , Humans , Infertility, Female/enzymology , Inflammation/microbiology , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , Pregnancy Complications, Infectious/enzymology , Pregnancy Complications, Infectious/microbiology , Pregnancy Complications, Infectious/pathology
10.
J Clin Microbiol ; 51(4): 1298-300, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23390274

ABSTRACT

Trichomonas vaginalis infections are usually asymptomatic or can result in nonspecific clinical symptoms, which makes laboratory-based detection of this protozoan parasite essential for diagnosis and treatment. We report the development of a battery of highly sensitive and specific PCR assays for detection of T. vaginalis in urine, a noninvasive specimen, and development of a protocol for differentiating among Trichomonas species that commonly infect humans.


Subject(s)
Clinical Laboratory Techniques/methods , Molecular Diagnostic Techniques/methods , Parasitology/methods , Polymerase Chain Reaction/methods , Trichomonas Infections/diagnosis , Trichomonas vaginalis/isolation & purification , Urine/parasitology , Adolescent , Child , Child, Preschool , Female , Humans , Infant , Sensitivity and Specificity , Trichomonas vaginalis/classification , Trichomonas vaginalis/genetics
11.
J Immunol ; 181(6): 4037-42, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18768859

ABSTRACT

We investigated the hypothesis that the enhanced Ag-presenting function of IL-10-deficient dendritic cells (DCs) is related to specific immunoregulatory cytoskeletal molecules expressed when exposed to Ags. We analyzed the role of a prominent cytoskeletal protein, LEK1, in the immunoregulation of DC functions; specifically cytokine secretion, costimulatory molecule expression, and T cell activation against Chlamydia. Targeted knockdown of LEK1 expression using specific antisense oligonucleotides resulted in the rapid maturation of Chlamydia-exposed DCs as measured by FACS analysis of key activation markers (i.e., CD14, CD40, CD54, CD80, CD86, CD197, CD205, and MHC class II). The secretion of mostly Th1 cytokines and chemokines (IL-1a, IL-9, IL-12, MIP-1a, and GM-CSF but not IL-4 and IL-10) was also enhanced by blocking of LEK1. The function of LEK1 in DC regulation involves cytoskeletal changes, since the dynamics of expression of vimentin and actin, key proteins of the cellular cytoskeleton, were altered after exposure of LEK1 knockdown DCs to Chlamydia. Furthermore, targeted inhibition of LEK1 expression resulted in the enhancement of the immunostimulatory capacity of DCs for T cell activation against Chlamydia. Thus, LEK1 knockdown DCs activated immune T cells at least 10-fold over untreated DCs. These results suggest that the effect of IL-10 deficiency is mediated through LEK1-related events that lead to rapid maturation of DCs and acquisition of the capacity to activate an elevated T cell response. Targeted modulation of LEK1 expression provides a novel strategy for augmenting the immunostimulatory function of DCs for inducing an effective immunity against pathogens.


Subject(s)
Chlamydia trachomatis/immunology , Chromosomal Proteins, Non-Histone/physiology , Dendritic Cells/immunology , Dendritic Cells/microbiology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/microbiology , Animals , Biomarkers/analysis , Cell Differentiation/immunology , Cells, Cultured , Chromosomal Proteins, Non-Histone/antagonists & inhibitors , Chromosomal Proteins, Non-Histone/biosynthesis , Chromosomal Proteins, Non-Histone/deficiency , Cytokines/biosynthesis , Cytokines/genetics , Cytokines/metabolism , Cytoskeletal Proteins/antagonists & inhibitors , Cytoskeletal Proteins/biosynthesis , Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/physiology , Dendritic Cells/cytology , Dendritic Cells/metabolism , Female , Interleukin-10/antagonists & inhibitors , Interleukin-10/biosynthesis , Interleukin-10/genetics , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins , Oligonucleotides, Antisense/pharmacology , T-Lymphocyte Subsets/metabolism
12.
Sci Rep ; 10(1): 15389, 2020 09 21.
Article in English | MEDLINE | ID: mdl-32958779

ABSTRACT

Shift work, performed by approximately 21 million Americans, is irregular or unusual work schedule hours occurring after 6:00 pm. Shift work has been shown to disrupt circadian rhythms and is associated with several adverse health outcomes and chronic diseases such as cancer, gastrointestinal and psychiatric diseases and disorders. It is unclear if shift work influences the complications associated with certain infectious agents, such as pelvic inflammatory disease, ectopic pregnancy and tubal factor infertility resulting from genital chlamydial infection. We used an Environmental circadian disruption (ECD) model mimicking circadian disruption occurring during shift work, where mice had a 6-h advance in the normal light/dark cycle (LD) every week for a month. Control group mice were housed under normal 12/12 LD cycle. Our hypothesis was that compared to controls, mice that had their circadian rhythms disrupted in this ECD model will have a higher Chlamydia load, more pathology and decreased fertility rate following Chlamydia infection. Results showed that, compared to controls, mice that had their circadian rhythms disrupted (ECD) had higher Chlamydia loads, more tissue alterations or lesions, and lower fertility rate associated with chlamydial infection. Also, infected ECD mice elicited higher proinflammatory cytokines compared to mice under normal 12/12 LD cycle. These results imply that there might be an association between shift work and the increased likelihood of developing more severe disease from Chlamydia infection.


Subject(s)
Chlamydia Infections/etiology , Circadian Rhythm/physiology , Shift Work Schedule/adverse effects , Animals , Chlamydia/pathogenicity , Chlamydia Infections/metabolism , Chlamydia Infections/pathology , Chlamydia muridarum/pathogenicity , Female , Mice , Mice, Inbred C57BL , Pelvic Inflammatory Disease/etiology , Photoperiod , Pregnancy , Pregnancy, Ectopic/etiology
13.
J Clin Microbiol ; 47(1): 215-6, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19005149

ABSTRACT

We evaluated the performance of the BioStar Chlamydia OIA (optical immunoassay) in adolescent females (n = 261) from an inner city population. With a reference standard of two different nucleic acid amplification tests, the sensitivity and specificity of the BioStar Chlamydia OIA were 59.4 and 98.4%, respectively. Due to its relatively low sensitivity, the BioStar Chlamydia OIA should only be used in conjunction with more sensitive laboratory tests unless laboratory tests are unavailable or timely return for treatment is unlikely.


Subject(s)
Bacteriological Techniques/methods , Chlamydia Infections/diagnosis , Chlamydia trachomatis/immunology , Chlamydia trachomatis/isolation & purification , Adolescent , Female , Humans , Immunoassay , Sensitivity and Specificity , Urban Population , Young Adult
15.
Sci Rep ; 9(1): 11405, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31388084

ABSTRACT

Genital chlamydia infection in women causes complications such as pelvic inflammatory disease and tubal factor infertility, but it is unclear why some women are more susceptible than others. Possible factors, such as time of day of chlamydia infection on chlamydial pathogenesis has not been determined. We hypothesised that infections during the day, will cause increased complications compared to infections at night. Mice placed under normal 12:12 light: dark (LD) cycle were infected intravaginally with Chlamydia muridarum either at zeitgeber time 3, ZT3 and ZT15. Infectivity was monitored by periodic vaginal swabs and chlamydiae isolation. Blood and vaginal washes were collected for host immunologic response assessments. The reproductive tracts of the mice were examined histopathologically, and fertility was determined by embryo enumeration after mating. Mice infected at ZT3 shed significantly more C. muridarum than mice infected at ZT15. This correlated with the increased genital tract pathology observed in mice infected at ZT3. Mice infected at ZT3 were less fertile than mice infected at ZT15. The results suggest that the time of day of infection influences chlamydial pathogenesis, it indicates a possible association between complications from chlamydia infection and host circadian clock, which may lead to a better understanding of chlamydial pathogenesis.


Subject(s)
Chlamydia Infections/immunology , Chlamydia muridarum/pathogenicity , Circadian Clocks/immunology , Pelvic Inflammatory Disease/immunology , Vagina/microbiology , Animals , Chlamydia Infections/blood , Chlamydia Infections/complications , Chlamydia Infections/microbiology , Disease Models, Animal , Female , Host Microbial Interactions/immunology , Humans , Mice , Pelvic Inflammatory Disease/microbiology , Photoperiod , Vagina/immunology , Vagina/pathology
16.
J Clin Microbiol ; 46(1): 355-6, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18032619

ABSTRACT

We genotyped Chlamydia trachomatis strains from 45 women or men living in either a rural indigenous community or in urban heterosexual communities. We found six different C. trachomatis serovars: E (n = 22; 48.9%), F (n = 10; 22.2%), J/Ja (n = 5; 11.1%), D/Da (n = 4; 8.9%), G (n = 3; 6.7%), and K (n = 1; 2.2%). The distribution of C. trachomatis serovars among members of the indigenous rural and the urban Australian communities appears similar to that in other Western countries.


Subject(s)
Chlamydia Infections/microbiology , Chlamydia trachomatis/classification , Chlamydia trachomatis/isolation & purification , Adult , Australia/epidemiology , Bacterial Outer Membrane Proteins/genetics , Bacterial Typing Techniques , Chlamydia Infections/epidemiology , Female , Genotype , Humans , Male , Molecular Epidemiology , Polymerase Chain Reaction/methods , Rural Population , Sequence Analysis, DNA , Urban Population , Urine/microbiology
17.
Expert Rev Vaccines ; 17(3): 217-227, 2018 03.
Article in English | MEDLINE | ID: mdl-29382248

ABSTRACT

INTRODUCTION: There is a persisting global burden and considerable public health challenge by the plethora of ocular, genital and respiratory diseases caused by members of the Gram-negative bacteria of the genus Chlamydia. The major diseases are conjunctivitis and blinding trachoma, non-gonococcal urethritis, cervicitis, pelvic inflammatory disease, ectopic pregnancy, tubal factor infertility, and interstitial pneumonia. The failures in screening and other prevention programs led to the current medical opinion that an efficacious prophylactic vaccine is the best approach to protect humans from chlamydial infections. Unfortunately, there is no human Chlamydia vaccine despite successful veterinary vaccines. A major challenge has been the effective delivery of vaccine antigens to induce safe and effective immune effectors to confer long-term protective immunity. The dawn of the era of biodegradable polymeric nanoparticles and the adjuvanted derivatives may accelerate the realization of the dream of human vaccine in the foreseeable future. AREAS COVERED: This review focuses on the current status of human chlamydial vaccine research, specifically the potential of biodegradable polymeric nanovaccines to provide efficacious Chlamydia vaccines in the near future. EXPERT COMMENTARY: The safety of biodegradable polymeric nanoparticles-based experimental vaccines with or without adjuvants and the array of available chlamydial vaccine candidates would suggest that clinical trials in humans may be imminent. Also, the promising results from vaccine testing in animal models could lead to human vaccines against trachoma and reproductive diseases simultaneously.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Bacterial Vaccines/administration & dosage , Chlamydia Infections/prevention & control , Animals , Bacterial Vaccines/immunology , Chlamydia Infections/immunology , Drug Delivery Systems , Humans , Nanoparticles , Polymers/chemistry
18.
J Microbiol Immunol Infect ; 40(3): 188-200, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17639158

ABSTRACT

BACKGROUND AND PURPOSE: Genital infections due to Chlamydia trachomatis pose a considerable public health challenge worldwide and a vaccine is urgently needed to protect against these infections. We examined whether a vaccine composed of a combination of the major outer membrane protein (MOMP) and porin B protein (PorB) of C. trachomatis would have a protective advantage over a single subunit construct. METHODS: Single and multisubunit vaccines expressing MOMP and PorB were constructed and evaluated in the mouse model of genital infection. Thus, groups of female C57BL/6 mice were immunized intramuscularly with recombinant Vibrio cholerae ghosts (VCG) expressing the vaccine antigens or VCG alone and humoral and cell-mediated immune responses were evaluated. RESULTS: Significant levels of Chlamydia-specific secretory immunoglobulin A and immunoglobulin G2a were detected in vaginal washes and serum of immunized mice. The multisubunit construct induced a significantly higher level of T-helper Type 1 response than the single subunits as measured by the amount of interferon-gamma produced by immune T cells in response to re-stimulation with ultraviolet-irradiated elementary bodies in vitro. Three weeks after the last immunization, animals were challenged intravaginally with 10(7) inclusion-forming units of C. trachomatis serovar D. There was a significant difference in the intensity and duration of vaginal shedding between the vaccine-immunized mice and controls. All the animals immunized with the multisubunit vaccine had completely resolved the infection 2 weeks post-challenge. Higher numbers of embryos were observed in vaccinated animals than in controls, indicating protection against infertility. CONCLUSION: These results underscore the potential, albeit moderate, vaccine advantage of the multisubunit formulation.


Subject(s)
Bacterial Outer Membrane Proteins/immunology , Bacterial Proteins/immunology , Bacterial Vaccines/immunology , Chlamydia Infections/immunology , Chlamydia trachomatis/immunology , Porins/immunology , Vaginal Diseases/immunology , Animals , Chlamydia Infections/microbiology , Chlamydia Infections/prevention & control , Female , Fertility , Immunoglobulin A/blood , Immunoglobulin A/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Mice , Mice, Inbred C57BL , Vaccination , Vaccines, Subunit/immunology , Vaginal Diseases/microbiology , Vaginal Diseases/prevention & control
19.
PLoS One ; 12(12): e0188643, 2017.
Article in English | MEDLINE | ID: mdl-29216217

ABSTRACT

Chlamydia trachomatis is a bacterial agent that causes sexually transmitted infections worldwide. The regulatory functions of dendritic cells (DCs) play a major role in protective immunity against Chlamydia infections. Here, we investigated the role of ASC in DCs metabolism and the regulation of DCs activation and function during Chlamydia infection. Following Chlamydia stimulation, maturation and antigen presenting functions were impaired in ASC-/- DCs compared to wild type (WT) DCs, in addition, ASC deficiency induced a tolerant phenotype in Chlamydia stimulated DCs. Using real-time extracellular flux analysis, we showed that activation in Chlamydia stimulated WT DCs is associated with a metabolic change in which mitochondrial oxidative phosphorylation (OXPHOS) is inhibited and the cells become committed to utilizing glucose through aerobic glycolysis for differentiation and antigen presenting functions. However, in ASC-/- DCs Chlamydia-induced metabolic change was prevented and there was a significant effect on mitochondrial morphology. The mitochondria of Chlamydia stimulated ASC-/- DCs had disrupted cristae compared to the normal narrow pleomorphic cristae found in stimulated WT DCs. In conclusion, our results suggest that Chlamydia-mediated activation of DCs is associated with a metabolic transition in which OXPHOS is inhibited, thereby dedicating the DCs to aerobic glycolysis, while ASC deficiency disrupts DCs function by inhibiting the reprogramming of DCs metabolism within the mitochondria, from glycolysis to electron transport chain.


Subject(s)
Chlamydia Infections/immunology , Chlamydia trachomatis/pathogenicity , Dendritic Cells/immunology , Animals , Cytokines/metabolism , Dendritic Cells/metabolism , Female , Mice , Mice, Inbred C57BL , Oxidative Phosphorylation
20.
Microbes Infect ; 8(3): 604-11, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16527508

ABSTRACT

Chlamydia trachomatis is the leading cause of bacterial sexually transmitted diseases worldwide. Urogenital strains are classified into serotypes and genotypes based on the major outer membrane protein and its gene, ompA, respectively. Studies of the association of serotypes with clinical signs and symptoms have produced conflicting results while no studies have evaluated associations with ompA polymorphisms. We designed a population-based cross-sectional study of 344 men and women with urogenital chlamydial infections (excluding co-pathogen infections) presenting to clinics serving five U.S. cities from 1995 to 1997. Signs, symptoms and sequelae of chlamydial infection (mucopurulent cervicitis, vaginal or urethral discharge; dysuria; lower abdominal pain; abnormal vaginal bleeding; and pelvic inflammatory disease) were analyzed for associations with serotype and ompA polymorphisms. One hundred and fifty-three (44.5%) of 344 patients had symptoms consistent with urogenital chlamydial infection. Gender, reason for visit and city were significant independent predictors of symptom status. Men were 2.2 times more likely than women to report any symptoms (P=0.03) and 2.8 times more likely to report a urethral discharge than women were to report a vaginal discharge in adjusted analyses (P=0.007). Differences in serotype or ompA were not predictive except for an association between serotype F and pelvic inflammatory disease (P=0.046); however, the number of these cases was small. While there was no clinically prognostic value associated with serotype or ompA polymorphism for urogenital chlamydial infections except for serotype F, future studies might utilize multilocus genomic typing to identify chlamydial strains associated with clinical phenotypes.


Subject(s)
Bacterial Outer Membrane Proteins/genetics , Chlamydia Infections/epidemiology , Chlamydia Infections/microbiology , Chlamydia trachomatis/classification , Chlamydia trachomatis/genetics , Polymorphism, Genetic/genetics , Adolescent , Adult , Chlamydia trachomatis/physiology , Cross-Sectional Studies , Female , Humans , Male , Serotyping
SELECTION OF CITATIONS
SEARCH DETAIL