Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Scand J Immunol ; 99(5): e13359, 2024 May.
Article in English | MEDLINE | ID: mdl-38605527

ABSTRACT

Chlamydia trachomatis infection is the leading cause of bacterial urogenital infection and has been demonstrated to drive inflammation and scarring of the reproductive tract. Recent studies have identified key triggers of proinflammatory adaptive immune responses driven by innate leukocytes and epithelia driving immunopathology. Utilizing chimeric mouse models, we investigated the definitive source and role of IL17 and IL17 signalling receptors during early Chlamydia muridarum infection of the female urogenital tract. Bone marrow transplants from wild-type (WT) and IL17A-/- mice to recipients demonstrated equivocal infection kinetics in the reproductive tract, but interestingly, adoptive transfer of IL17A-/- immune cells to WT recipients resulted in no infertility, suggesting a haematopoietic (as opposed to tissue) source of IL17 driving immunopathology. To further delineate the role of IL17 in immunopathology, we infected WT and IL17 receptor A (IL17RA)-/- female mice and observed a significant reduction in immunopathology in IL17RA-/- mice. WT bone marrow transplants to IL17RA-/- recipient mice prevented hydrosalpinx, suggesting signalling through IL17RA drives immunopathology. Furthermore, early chemical inhibition of IL17 signalling significantly reduced hydrosalpinx, suggesting IL17 acts as an innate driver of disease. Early during the infection, IL17 was produced by γδ T cells in the cervico-vagina, but more importantly, by neutrophils at the site of infertility in the oviducts. Taken together, these data suggest innate production of IL17 by haematopoietic leukocytes drives immunopathology in the epithelia during early C. muridarum infection of the female reproductive tract.


Subject(s)
Chlamydia Infections , Chlamydia muridarum , Interleukin-17 , Reproductive Tract Infections , Animals , Female , Mice , Mice, Inbred C57BL , Reproductive Tract Infections/pathology
2.
Scand J Immunol ; 99(1): e13331, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38441219

ABSTRACT

Chlamydia trachomatis infections are an important sexually transmitted infection that can lead to inflammation, scarring and hydrosalpinx/infertility. However, infections are commonly clinically asymptomatic and do not receive treatment. The underlying cause of asymptomatic immunopathology remains unknown. Here, we demonstrate that IgG produced during male infection enhanced the incidence of immunopathology and infertility in females. Human endocervical cells expressing the neonatal Fc Receptor (FcRn) increased translocation of human IgG-opsonized C. trachomatis. Using total IgG purified from infected male mice, we opsonized C. muridarum and then infected female mice, mimicking sexual transmission. Following infection, IgG-opsonized Chlamydia was found to transcytose the epithelial barrier in the uterus, where it was phagocytosed by antigen-presenting cells (APCs) and trafficked to the draining lymph nodes. APCs then expanded both CD4+ and CD8+ T cell populations and caused significantly more infertility in female mice infected with non-opsonized Chlamydia. Enhanced phagocytosis of IgG-opsonized Chlamydia significantly increased pro-inflammatory signalling and T cell proliferation. As IgG is transcytosed by FcRn, we utilized FcRn-/- mice and observed that shedding kinetics of Chlamydia were only affected in FcRn-/- mice infected with IgG-opsonized Chlamydia. Depletion of CD8+ T cells in FcRn-/- mice lead to a significant reduction in the incidence of infertility. Taken together, these data demonstrate that IgG seroconversion during male infection can amplify female immunopathology, dependent on FcRn transcytosis, APC differentiation and enhanced CD8 T cell responses.


Subject(s)
Chlamydia , Infertility , Humans , Female , Male , Animals , Mice , CD8-Positive T-Lymphocytes , Immunoglobulin G , Genitalia
3.
J Immunol ; 208(3): 603-617, 2022 02 01.
Article in English | MEDLINE | ID: mdl-35022277

ABSTRACT

MicroRNAs (miRNAs/miRs) are small, endogenous noncoding RNAs that are important post-transcriptional regulators with clear roles in the development of the immune system and immune responses. Using miRNA microarray profiling, we characterized the expression profile of naive and in vivo generated murine effector antiviral CD8+ T cells. We observed that out of 362 measurable mature miRNAs, 120 were differentially expressed by at least 2-fold in influenza-specific effector CD8+ CTLs compared with naive CD8+ T cells. One miRNA found to be highly downregulated on both strands in effector CTLs was miR-139. Because previous studies have indicated a role for miR-139-mediated regulation of CTL effector responses, we hypothesized that deletion of miR-139 would enhance antiviral CTL responses during influenza virus infection. We generated miR-139-/- mice or overexpressed miR-139 in T cells to assess the functional contribution of miR-139 expression in CD8+ T cell responses. Our study demonstrates that the development of naive T cells and generation or differentiation of effector or memory CD8+ T cell responses to influenza virus infection are not impacted by miR-139 deficiency or overexpression; yet, miR-139-/- CD8+ T cells are outcompeted by wild-type CD8+ T cells in a competition setting and demonstrate reduced responses to Listeria monocytogenes Using an in vitro model of T cell exhaustion, we confirmed that miR-139 expression similarly does not impact the development of T cell exhaustion. We conclude that despite significant downregulation of miR-139 following in vivo and in vitro activation, miR-139 expression is dispensable for influenza-specific CTL responses.


Subject(s)
Influenza A virus/immunology , Listeria monocytogenes/immunology , MicroRNAs/genetics , Orthomyxoviridae Infections/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Down-Regulation/genetics , Female , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/immunology
4.
Biol Reprod ; 108(5): 758-777, 2023 05 10.
Article in English | MEDLINE | ID: mdl-36799886

ABSTRACT

Chlamydia is the most common bacterial sexually transmitted infection worldwide and it is widely acknowledged that controlling the rampant community transmission of this infection requires vaccine development. In this study, for the first time, we elucidate the long-term response to male mouse chlamydial vaccination with chlamydial major outer membrane protein (MOMP) and ISCOMATRIX (IMX) both prophylactically and in a novel therapeutic setting. Vaccination significantly reduced and, in some cases, cleared chlamydial burden from the prostates, epididymides, and testes, which correlates with high IgG and IgA tires in tissues and serum. Important markers of sperm health and fertility were protected including sperm motility and proteins associated with fertility in men. Within splenocytes, expression of IFNγ, TNFα, IL17, IL13, IL10, and TGFß were changed by both infection and vaccination within CD4 and CD8 T cells and regulatory T cells. Within the testicular tissue, phenotypic and concentration changes were observed in macrophages and T cells (resident and transitory). This revealed some pathogenic phenotypes associated with infection and critically that vaccination allows maintenance of testicular homeostasis, likely by preventing significant influx of CD4 T cells and promoting IL10 production. Finally, we demonstrated the testes contained immature (B220+) B cells and mature (CD138+) Chlamydia-specific plasma cells. Thus, through vaccination, we can maintain the healthy function of the testes, which is vital to protection of male fertility.


Subject(s)
Chlamydia Infections , Chlamydia muridarum , Male , Animals , Mice , Chlamydia Infections/prevention & control , Chlamydia Infections/complications , Interleukin-10 , Semen , Sperm Motility , Spermatozoa/pathology , Vaccination , Bacterial Outer Membrane Proteins
5.
Scand J Immunol ; 97(5): e13263, 2023 May.
Article in English | MEDLINE | ID: mdl-36872855

ABSTRACT

Urogenital chlamydial infections continue to increase with over 127 million people affected annually, causing significant economic and public health pressures. While the role of traditional MHCI and II peptide presentation is well defined in chlamydial infections, the role of lipid antigens in immunity remains unclear. Natural killer (NK) T cells are important effector cells that recognize and respond to lipid antigens during infections. Chlamydial infection of antigen-presenting cells facilitates presentation of lipid on the MHCI-like protein, CD1d, which stimulates NKT cells to respond. During urogenital chlamydial infection, wild-type (WT) female mice had significantly greater chlamydial burden than CD1d-/- (NKT-deficient) mice, and had significantly greater incidence and severity of immunopathology in both primary and secondary infections. WT mice had similar vaginal lymphocytic infiltrate, but 59% more oviduct occlusion compared to CD1d-/- mice. Transcriptional array analysis of oviducts day 6 post-infection revealed WT mice had elevated levels of Ifnγ (6-fold), Tnfα (38-fold), Il6 (2.5-fold), Il1ß (3-fold) and Il17a (6-fold) mRNA compared to CD1d-/- mice. In infected females, oviduct tissues had an elevated infiltration of CD4+ -invariant NKT (iNKT) cells, however, iNKT-deficient Jα18-/- mice had no significant differences in hydrosalpinx severity or incidence compared to WT controls. Lipid mass spectrometry of surface-cleaved CD1d in infected macrophages revealed an enhancement of presented lipids and cellular sequestration of sphingomyelin. Taken together, these data suggest an immunopathogenic role for non-invariant NKT cells in urogenital chlamydial infections, facilitated by lipid presentation via CD1d via infected antigen-presenting cells.


Subject(s)
Infertility , Natural Killer T-Cells , Mice , Female , Animals , Antigens, CD1d , Antigen-Presenting Cells , Proteins , Infertility/metabolism , Lipids , Mice, Inbred C57BL
6.
BMC Pediatr ; 22(1): 185, 2022 04 07.
Article in English | MEDLINE | ID: mdl-35392855

ABSTRACT

BACKGROUND: There is limited data on pediatric ventilator-associated events (PedVAE) in the neonatal intensive care unit (NICU) setting, since the CDC mandated state reporting of these events in January 2019. This study sought to describe PedVAE rates and characteristics in the NICU population. METHODS: Single-center case-control study of infants requiring mechanical ventilation in a 39-bed level IV NICU between January 1, 2018 and December 31, 2020. Baseline infant demographic, respiratory support and antibiotic use data was obtained and comparisons were performed between patients with potential PedVAEs and those without events. RESULT: Two hundred and nine infants were mechanically ventilated. Two of the 126 patients ventilated for ≥4 days met CDC criteria for PedVAEs with a total of 3 events, and 32 (25%) received antibiotics with escalation of respiratory support, primarily for tracheitis. CONCLUSION: NICU-specific data on PedVAE is limited. Only 2 infants in the study period met the current CDC criteria for PedVAE with a rate of 0.9 events per 1000 ventilator days. The current CDC PedVAE definition might be inadequate to identify actionable VAEs to inform prevention efforts in the NICU population, and alternate indices could better characterize these events.


Subject(s)
Intensive Care Units, Neonatal , Pneumonia, Ventilator-Associated , Anti-Bacterial Agents , Case-Control Studies , Centers for Disease Control and Prevention, U.S. , Child , Humans , Infant , Infant, Newborn , Pneumonia, Ventilator-Associated/diagnosis , Pneumonia, Ventilator-Associated/epidemiology , Pneumonia, Ventilator-Associated/etiology , Respiration, Artificial/adverse effects , United States , Ventilators, Mechanical
7.
PLoS Pathog ; 15(10): e1008072, 2019 10.
Article in English | MEDLINE | ID: mdl-31603951

ABSTRACT

Respiratory viral infections contribute substantially to global infant losses and disproportionately affect preterm neonates. Using our previously established neonatal murine model of influenza infection, we demonstrate that three-day old mice are exceptionally sensitive to influenza virus infection and exhibit high mortality and viral load. Intranasal pre- and post-treatment of neonatal mice with Lactobacillus rhamnosus GG (LGG), an immune modulator in respiratory viral infection of adult mice and human preterm neonates, considerably improves neonatal mice survival after influenza virus infection. We determine that both live and heat-killed intranasal LGG are equally efficacious in protection of neonates. Early in influenza infection, neonatal transcriptional responses in the lung are delayed compared to adults. These responses increase by 24 hours post-infection, demonstrating a delay in the kinetics of the neonatal anti-viral response. LGG pretreatment improves immune gene transcriptional responses during early infection and specifically upregulates type I IFN pathways. This is critical for protection, as neonatal mice intranasally pre-treated with IFNß before influenza virus infection are also protected. Using transgenic mice, we demonstrate that the protective effect of LGG is mediated through a MyD88-dependent mechanism, specifically via TLR4. LGG can improve both early control of virus and transcriptional responsiveness and could serve as a simple and safe intervention to protect neonates.


Subject(s)
Influenza A virus/physiology , Lacticaseibacillus rhamnosus/growth & development , Lung/immunology , Orthomyxoviridae Infections/prevention & control , Administration, Intranasal , Animals , Animals, Newborn , Disease Models, Animal , Lung/virology , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology
8.
Pediatr Res ; 89(5): 1126-1135, 2021 04.
Article in English | MEDLINE | ID: mdl-32303051

ABSTRACT

BACKGROUND: Endogenous pulmonary stem cells (PSCs) play an important role in lung development and repair; however, little is known about their role in bronchopulmonary dysplasia (BPD). We hypothesize that an endogenous PSC marker stage-specific embryonic antigen-1 (SSEA-1) and its enzyme, α1,3-fucosyltransferase IX (FUT9) play an important role in decreasing inflammation and restoring lung structure in experimental BPD. METHODS: We studied the expression of SSEA-1, and its enzyme FUT9, in wild-type (WT) C57BL/6 mice, in room air and hyperoxia. Effects of intraperitoneal administration of recombinant human FUT9 (rhFUT9) on lung airway and parenchymal inflammation, alveolarization, and apoptosis were evaluated. RESULTS: On hyperoxia exposure, SSEA-1 significantly decreased at postnatal day 14 in hyperoxia-exposed BPD mice, accompanied by a decrease in FUT9. BPD and respiratory distress syndrome (RDS) in human lungs showed decreased expression of SSEA-1 as compared to their term controls. Importantly, intraperitoneal administration of FUT9 in the neonatal BPD mouse model resulted in significant decrease in pulmonary airway (but not lung parenchymal) inflammation, alveolar-capillary leakage, alveolar simplification, and cell death in the hyperoxia-exposed BPD mice. CONCLUSIONS: An important role of endogenous PSC marker SSEA-1 and its enzyme FUT9 is demonstrated, indicating early systemic intervention with FUT9 as a potential therapeutic option for BPD. IMPACT: Administration of rhFUT9, an enzyme of endogenous stem cell marker SSEA-1, reduces pulmonary airway (but not lung parenchymal) inflammation, alveolar-capillary leak and cell death in the BPD mouse model. SSEA-1 is reported for the first time in experimental BPD models, and in human RDS and BPD. rhFUT9 treatment ameliorates hyperoxia-induced lung injury in a developmentally appropriate BPD mouse model. Our results have translational potential as a therapeutic modality for BPD in the developing lung.


Subject(s)
Bronchopulmonary Dysplasia/drug therapy , Fucosyltransferases/therapeutic use , Lewis X Antigen/metabolism , Lung/cytology , Stem Cells/metabolism , Animals , Animals, Newborn , Biomarkers/metabolism , Mice , Mice, Inbred C57BL
9.
Int J Mol Sci ; 22(16)2021 Aug 09.
Article in English | MEDLINE | ID: mdl-34445253

ABSTRACT

Bronchopulmonary dysplasia (BPD) is the most common complication of prematurity and a key contributor to the large health care burden associated with prematurity, longer hospital stays, higher hospital costs, and frequent re-hospitalizations of affected patients through the first year of life and increased resource utilization throughout childhood. This disease is associated with abnormal pulmonary function that may lead to BPD-associated pulmonary hypertension (PH), a major contributor to neonatal mortality and morbidity. In the absence of any definitive treatment options, this life-threatening disease is associated with high resource utilization during and after neonatal intensive care unit (NICU) stay. The goal of this study was to test the safety and efficacy of a small molecule derivative of chitin, AVR-48, as prophylactic therapy for preventing experimental BPD in a mouse model. Two doses of AVR-48 were delivered either intranasally (0.11 mg/kg), intraperitoneally (10 mg/kg), or intravenously (IV) (10 mg/kg) to newborn mouse pups on postnatal day (P)2 and P4. The outcomes were assessed by measuring total inflammatory cells in the broncho-alveolar lavage fluid (BALF), chord length, septal thickness, and radial alveolar counts of the alveoli, Fulton's Index (for PH), cell proliferation and cell death by immunostaining, and markers of inflammation by Western blotting and ELISA. The bioavailability and safety of the drug were assessed by pharmacokinetic and toxicity studies in both neonatal mice and rat pups (P3-P5). Following AVR-48 treatment, alveolar simplification was improved, as evident from chord length, septal thickness, and radial alveolar counts; total inflammatory cells were decreased in the BALF; Fulton's Index was decreased and lung inflammation and cell death were decreased, while angiogenesis and cell proliferation were increased. AVR-48 was found to be safe and the no-observed-adverse-effect level (NOAEL) in rat pups was determined to be 100 mg/kg when delivered via IV dosing with a 20-fold safety margin. With no reported toxicity and with a shorter half-life, AVR-48 is able to reverse the worsening cardiopulmonary phenotype of experimental BPD and BPD-PH, compared to controls, thus positioning it as a future drug candidate.


Subject(s)
Bronchopulmonary Dysplasia , Chitin , Hypertension, Pulmonary , Neovascularization, Physiologic/drug effects , Pulmonary Alveoli , Animals , Animals, Newborn , Bronchopulmonary Dysplasia/drug therapy , Bronchopulmonary Dysplasia/metabolism , Bronchopulmonary Dysplasia/pathology , Chitin/chemistry , Chitin/pharmacology , Disease Models, Animal , Drug Evaluation, Preclinical , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Mice , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , Rats
10.
Biol Reprod ; 102(4): 888-901, 2020 04 15.
Article in English | MEDLINE | ID: mdl-31965142

ABSTRACT

With approximately 131 million new genital tract infections occurring each year, Chlamydia is the most common sexually transmitted bacterial pathogen worldwide. Male and female infections occur at similar rates and both cause serious pathological sequelae. Despite this, the impact of chlamydial infection on male fertility has long been debated, and the effects of paternal chlamydial infection on offspring development are unknown. Using a male mouse chronic infection model, we show that chlamydial infection persists in the testes, adversely affecting the testicular environment. Infection increased leukocyte infiltration, disrupted the blood:testis barrier and reduced spermiogenic cell numbers and seminiferous tubule volume. Sperm from infected mice had decreased motility, increased abnormal morphology, decreased zona-binding capacity, and increased DNA damage. Serum anti-sperm antibodies were also increased. When both acutely and chronically infected male mice were bred with healthy female mice, 16.7% of pups displayed developmental abnormalities. Female offspring of chronically infected sires had smaller reproductive tracts than offspring of noninfected sires. The male pups of infected sires displayed delayed testicular development, with abnormalities in sperm vitality, motility, and sperm-oocyte binding evident at sexual maturity. These data suggest that chronic testicular Chlamydia infection can contribute to male infertility, which may have an intergenerational impact on sperm quality.


Subject(s)
Chlamydia Infections/microbiology , Chlamydia muridarum , Fertility/physiology , Infertility, Male/microbiology , Prenatal Exposure Delayed Effects/microbiology , Testis/microbiology , Animals , Female , Male , Mice , Pregnancy , Sperm Motility/physiology
11.
Immunol Cell Biol ; 97(10): 865-876, 2019 11.
Article in English | MEDLINE | ID: mdl-31348541

ABSTRACT

Chlamydia infection remains the leading sexually-transmitted bacterial infection worldwide, causing damaging sequelae such as tubal scarring, infertility and ectopic pregnancy. As infection is often asymptomatic, prevention via vaccination is the optimal strategy for disease control. Vaccination strategies aimed at preventing bacterial infection have shown some promise, although these strategies often fail to prevent damaging inflammatory pathology when Chlamydia is encountered. Using a murine model of Chlamydia muridarum genital infection, we employed two established independent models to compare immune responses underpinning pathologic development of genital Chlamydia infection. Model one uses antibiotic treatment during infection, with only early treatment preventing pathology. Model two uses a plasmid-cured variant strain of C. muridarum that does not cause pathologic outcomes like the plasmid-containing wild-type counterpart. Using these infection models, contrasted by the development of pathology, we identified an unexpected role for macrophages. We observed that mice showing signs of pathology had greater numbers of activated macrophages present in the oviducts. This may have been due to early differences in macrophage activation and proinflammatory signaling leading to persistent or enhanced infection. These results provide valuable insight into the cellular mechanisms driving pathology in Chlamydia infection and contribute to the design and development of more effective vaccine strategies for protection against the deleterious sequelae of Chlamydia infection of the female reproductive tract.


Subject(s)
Azithromycin/pharmacology , Chlamydia muridarum/physiology , Drug Resistance, Bacterial/drug effects , Fallopian Tubes/pathology , Inflammation/pathology , Macrophages/microbiology , Oviducts/pathology , Animals , Chlamydia Infections/genetics , Chlamydia Infections/immunology , Chlamydia Infections/microbiology , Chlamydia Infections/pathology , Chlamydia muridarum/drug effects , Chronic Disease , Cytokines/metabolism , Fallopian Tubes/drug effects , Female , Gene Expression Regulation/drug effects , Inflammation Mediators/metabolism , Macrophages/drug effects , Macrophages/pathology , Mice, Inbred BALB C , Oviducts/drug effects
12.
Biol Reprod ; 101(4): 748-759, 2019 10 25.
Article in English | MEDLINE | ID: mdl-31373361

ABSTRACT

The incidence of Chlamydia infection, in both females and males, is increasing worldwide. Male infections have been associated clinically with urethritis, epididymitis, and orchitis, believed to be caused by ascending infection, although the impact of infection on male fertility remains controversial. Using a mouse model of male chlamydial infection, we show that all the major testicular cell populations, germ cells, Sertoli cells, Leydig cells, and testicular macrophages can be productively infected. Furthermore, sperm isolated from vas deferens of infected mice also had increased levels of DNA damage as early as 4 weeks post-infection. Bilateral vasectomy, prior to infection, did not affect the chlamydial load recovered from testes at 2, 4, and 8 weeks post-infection, and Chlamydia-infected macrophages were detectable in blood and the testes as soon as 3 days post-infection. Partial depletion of macrophages with clodronate liposomes significantly reduced the testicular chlamydial burden, consistent with a hematogenous route of infection, with Chlamydia transported to the testes in infected macrophages. These data suggest that macrophages serve as Trojan horses, transporting Chlamydia from the penile urethra to the testes within 3 days of infection, bypassing the entire male reproductive tract. In the testes, infected macrophages likely transfer infection to Leydig, Sertoli, and germ cells, causing sperm DNA damage and impaired spermatogenesis.


Subject(s)
Chlamydia Infections/complications , Chlamydia muridarum/physiology , Infertility, Male , Macrophages/microbiology , Testis/microbiology , Urethra/microbiology , Animals , Cells, Cultured , Chlamydia Infections/genetics , Chlamydia Infections/microbiology , Chlamydia Infections/pathology , Chlamydia muridarum/genetics , DNA Damage , Infertility, Male/genetics , Infertility, Male/microbiology , Infertility, Male/pathology , Macrophages/pathology , Male , Mice, Inbred C57BL , Orchitis/complications , Orchitis/microbiology , Orchitis/pathology , Organisms, Genetically Modified , Spermatozoa/metabolism , Spermatozoa/microbiology , Testis/pathology , Urethra/pathology
13.
Hum Reprod ; 34(10): 1891-1898, 2019 10 02.
Article in English | MEDLINE | ID: mdl-31586185

ABSTRACT

STUDY QUESTION: Can Chlamydia be found in the testes of infertile men? SUMMARY ANSWER: Chlamydia can be found in 16.7% of fresh testicular biopsies and 45.3% of fixed testicular biopsies taken from a selection of infertile men. WHAT IS KNOWN ALREADY: Male chlamydial infection has been understudied despite male and female infections occurring at similar rates. This is particularly true of asymptomatic infections, which occur in 50% of cases. Chlamydial infection has also been associated with increased sperm DNA damage and reduced male fertility. STUDY DESIGN, SIZE, DURATION: We collected diagnostic (fixed, n = 100) and therapeutic (fresh, n = 18) human testicular biopsies during sperm recovery procedures from moderately to severely infertile men in a cross-sectional approach to sampling. PARTICIPANTS/MATERIALS, SETTING, METHODS: The diagnostic and therapeutic biopsies were tested for Chlamydia-specific DNA and protein, using real-time PCR and immunohistochemical approaches, respectively. Serum samples matched to the fresh biopsies were also assayed for the presence of Chlamydia-specific antibodies using immunoblotting techniques. MAIN RESULTS AND THE ROLE OF CHANCE: Chlamydial major outer membrane protein was detected in fixed biopsies at a rate of 45.3%. This was confirmed by detection of chlamydial DNA and TC0500 protein (replication marker). C. trachomatis DNA was detected in fresh biopsies at a rate of 16.7%, and the sera from each of these three positive patients contained C. trachomatis-specific antibodies. Overall, C. trachomatis-specific antibodies were detected in 72.2% of the serum samples from the patients providing fresh biopsies, although none of the patients were symptomatic nor had they reported a previous sexually transmitted infection diagnosis including Chlamydia. LIMITATIONS, REASONS FOR CAUTION: No reproductively healthy male testicular biopsies were tested for the presence of Chlamydia DNA or proteins or Chlamydia-specific antibodies due to the unavailability of these samples. WIDER IMPLICATIONS FOR THE FINDINGS: Application of Chlamydia-specific PCR and immunohistochemistry in this human male infertility context of testicular biopsies reveals evidence of a high prevalence of previously unrecognised infection, which may potentially have a pathogenic role in spermatogenic failure. STUDY FUNDING/COMPETING INTEREST(S): Funding for this project was provided by the Australian NHMRC under project grant number APP1062198. We also acknowledge assistance from the Monash IVF Group and Queensland Fertility Group in the collection of fresh biopsies, and the Monash Health and co-author McLachlan (declared equity interest) in retrieval and sectioning of fixed biopsies. E.M. declares an equity interest in the study due to financing of fixed biopsy sectioning. All other authors declare no conflicts of interest. TRIAL REGISTRATION NUMBER: N/A.


Subject(s)
Azoospermia/microbiology , Chlamydia Infections/diagnosis , Chlamydia trachomatis/isolation & purification , Testis/microbiology , Asymptomatic Infections , Azoospermia/diagnosis , Azoospermia/pathology , Azoospermia/therapy , Chlamydia Infections/complications , Chlamydia Infections/microbiology , Chlamydia Infections/pathology , Chlamydia trachomatis/genetics , Cross-Sectional Studies , DNA, Bacterial/isolation & purification , Humans , Male , Sperm Retrieval , Testis/pathology
14.
J Immunol ; 196(6): 2602-13, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26873987

ABSTRACT

Currently, there is little consensus regarding the most appropriate animal model to study acute infection and the virus-specific CD8(+) T cell (CTL) responses in neonates. TCRß high-throughput sequencing in naive CTL of differently aged neonatal mice was performed, which demonstrated differential Vß family gene usage. Using an acute influenza infection model, we examined the TCR repertoire of the CTL response in neonatal and adult mice infected with influenza type A virus. Three-day-old mice mounted a greatly reduced primary NP(366-374)-specific CTL response when compared with 7-d-old and adult mice, whereas secondary CTL responses were normal. Analysis of NP(366-374)-specific CTL TCR repertoire revealed different Vß gene usage and greatly reduced public clonotypes in 3-d-old neonates. This could underlie the impaired CTL response in these neonates. To directly test this, we examined whether controlling the TCR would restore neonatal CTL responses. We performed adoptive transfers of both nontransgenic and TCR-transgenic OVA(257-264)-specific (OT-I) CD8(+) T cells into influenza-infected hosts, which revealed that naive neonatal and adult OT-I cells expand equally well in neonatal and adult hosts. In contrast, nontransgenic neonatal CD8(+) T cells when transferred into adults failed to expand. We further demonstrate that differences in TCR avidity may contribute to decreased expansion of the endogenous neonatal CTL. These studies highlight the rapid evolution of the neonatal TCR repertoire during the first week of life and show that impaired neonatal CTL immunity results from an immature TCR repertoire, rather than intrinsic signaling defects or a suppressive environment.


Subject(s)
Animals, Newborn/immunology , CD8-Positive T-Lymphocytes/immunology , Lymphopoiesis/immunology , Orthomyxoviridae Infections/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Adoptive Transfer , Animals , Cell Separation , Disease Models, Animal , Flow Cytometry , High-Throughput Nucleotide Sequencing , Influenza A virus , Mice , Mice, Inbred C57BL , Mice, Transgenic
15.
J Immunol ; 196(3): 1186-98, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26740110

ABSTRACT

The p110δ isoform of PI3K is known to play an important role in immunity, yet its contribution to CTL responses has not been fully elucidated. Using murine p110δ-deficient CD8(+) T cells, we demonstrated a critical role for the p110δ subunit in the generation of optimal primary and memory CD8(+) T cell responses. This was demonstrated in both acute viral and intracellular bacterial infections in mice. We show that p110δ signaling is required for CD8(+) T cell activation, proliferation and effector cytokine production. We provide evidence that the effects of p110δ signaling are mediated via Akt activation and through the regulation of TCR-activated oxidative phosphorylation and aerobic glycolysis. In light of recent clinical trials that employ drugs targeting p110δ in certain cancers and other diseases, our study suggests caution in using these drugs in patients, as they could potentially increase susceptibility to infectious diseases. These studies therefore reveal a novel and direct role for p110δ signaling in in vivo CD8(+) T cell immunity to microbial pathogens.


Subject(s)
Bacterial Infections/enzymology , CD8-Positive T-Lymphocytes/enzymology , Lymphocyte Activation/immunology , Phosphatidylinositol 3-Kinases/immunology , Virus Diseases/enzymology , Adoptive Transfer , Animals , Bacterial Infections/immunology , CD8-Positive T-Lymphocytes/immunology , Female , Flow Cytometry , Immunologic Memory/immunology , Isoenzymes/immunology , Mice , Mice, Knockout , Signal Transduction/immunology , Virus Diseases/immunology
16.
J Infect Dis ; 215(3): 475-483, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28011914

ABSTRACT

Background: Streptococcus agalactiae can cause urinary tract infection (UTI). The role of the S. agalactiae global virulence regulator, CovR, in UTI pathogenesis is unknown. Methods: We used murine and human bladder uroepithelial cell models of UTI and S. agalactiae mutants in covR and related factors, including ß-hemolysin/cytolysin (ß-h/c), surface-anchored adhesin HvgA, and capsule to study the role of CovR in UTI. Results: We found that covR-deficient serotype III S. agalactiae 874391 was significantly attenuated for colonization in mice and adhesion to uroepithelial cells. Mice infected with covR-deficient S. agalactiae produced less proinflammatory cytokines than those infected with wild-type 874391. Acute cytotoxicity in uroepithelial cells triggered by covR-deficient but not wild-type 874391 was associated with significant caspase 3 activation. Mechanistically, covR mutation significantly altered the expression of several genes in S. agalactiae 874391 that encode key virulence factors, including ß-h/c and HvgA, but not capsule. Subsequent mutational analyses revealed that HvgA and capsule, but not the ß-h/c, exerted significant effects on colonization of the murine urinary tract in vivo. Conclusions: S. agalactiae CovR promotes bladder infection and inflammation, as well as adhesion to and viability of uroepithelial cells. The pathogenesis of S. agalactiae UTI is complex, multifactorial, and influenced by virulence effects of CovR, HvgA, and capsule.


Subject(s)
Bacterial Proteins/physiology , Streptococcus agalactiae/pathogenicity , Urinary Tract Infections/microbiology , Virulence Factors/physiology , Adhesins, Bacterial/physiology , Animals , Bacterial Adhesion , Bacterial Capsules/physiology , Cell Line , Cytokines/metabolism , Cytotoxicity, Immunologic , Female , Gene Expression , Humans , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Urinary Bladder/metabolism , Urothelium/microbiology
17.
Pediatr Res ; 82(1): 133-140, 2017 07.
Article in English | MEDLINE | ID: mdl-28355204

ABSTRACT

BackgroundCD31, expressed by the majority of the neonatal T-cell pool, is involved in modulation of T-cell receptor signaling by increasing the threshold for T-cell activation. Therefore, CD31 could modulate neonatal tolerance and adaptive immune responses.MethodsLymphocytes were harvested from murine neonates at different ages, human late preterm and term cord blood, and adult peripheral blood. Human samples were activated over a 5-day period to simulate acute inflammation. Mice were infected with influenza; lungs and spleens were harvested at days 6 and 9 post infection and analyzed by flow cytometry.ResultsCD31-expressing neonatal murine CD4+ and CD8a+ T cells increase over the first week of life. Upon in vitro stimulation, human infants' CD4+ and CD8a+ T cells shed CD31 faster in comparison with adults. In the context of acute infection, mice infected at 3 days of age have an increased number of naive and activated CD31+ T lymphocytes at the site of infection at days 6 and 9 post infection, as compared with those infected at 7 days of age; however, the opposite is true in the periphery.ConclusionDifferences in trafficking of CD31+ cytotoxic T lymphocytes (CTLs) during acute influenza infection could modulate tolerance and contribute to a dampened adaptive immune response in neonates.


Subject(s)
Orthomyxoviridae Infections/immunology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , T-Lymphocytes/cytology , Animals , Fetal Blood/cytology , Flow Cytometry , Humans , Immune System , Infant, Premature , Lung/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Signal Transduction , Time Factors
18.
J Infect Dis ; 213(4): 659-68, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26324782

ABSTRACT

BACKGROUND: CD14, a coreceptor for several pattern recognition receptors and a widely used monocyte/macrophage marker, plays a key role in host responses to gram-negative bacteria. Despite the central role of CD14 in the inflammatory response to lipopolysaccharide and other microbial products and in the dissemination of bacteria in some infections, the signaling networks controlled by CD14 during urinary tract infection (UTI) are unknown. METHODS: We used uropathogenic Escherichia coli (UPEC) infection of wild-type (WT) C57BL/6 and Cd14(-/-) mice and RNA sequencing to define the CD14-dependent transcriptional signature and the role of CD14 in host defense against UTI in the bladder. RESULTS: UPEC induced the upregulation of Cd14 and the monocyte/macrophage-related genes Emr1/F4/80 and Csf1r/c-fms, which was associated with lower UPEC burdens in WT mice, compared with Cd14(-/-) mice. Exacerbation of infection in Cd14(-/-) mice was associated with the absence of a 491-gene transcriptional signature in the bladder that encompassed multiple host networks not previously associated with this receptor. CD14-dependent pathways included immune cell trafficking, differential cytokine production in macrophages, and interleukin 17 signaling. Depletion of monocytes/macrophages in the bladder by administration of liposomal clodronate led to higher UPEC burdens. CONCLUSIONS: This study identifies new host protective and signaling roles for CD14 in the bladder during UPEC UTI.


Subject(s)
Lipopolysaccharide Receptors/metabolism , Macrophages/immunology , Signal Transduction , Urinary Bladder/immunology , Urinary Tract Infections/immunology , Uropathogenic Escherichia coli/immunology , Animals , Female , Gene Deletion , Gene Expression Profiling , Lipopolysaccharide Receptors/genetics , Mice, Inbred C57BL , Mice, Knockout , Urinary Tract Infections/microbiology
19.
Crit Rev Microbiol ; 42(5): 780-99, 2016 Sep.
Article in English | MEDLINE | ID: mdl-26006172

ABSTRACT

Urinary tract infections (UTIs) are among the most common bacterial infections in humans. Murine models of human UTI are vital experimental tools that have helped to elucidate UTI pathogenesis and advance knowledge of potential treatment and infection prevention strategies. Fundamentally, several variables are inherent in different murine models, and understanding the limitations of these variables provides an opportunity to understand how models may be best applied to research aimed at mimicking human disease. In this review, we discuss variables inherent in murine UTI model studies and how these affect model usage, data analysis and data interpretation. We examine recent studies that have elucidated UTI host-pathogen interactions from the perspective of gene expression, and review new studies of biofilm and UTI preventative approaches. We also consider potential standards for variables inherent in murine UTI models and discuss how these might expand the utility of models for mimicking human disease and uncovering new aspects of pathogenesis.


Subject(s)
Bacterial Infections/microbiology , Bacterial Physiological Phenomena , Disease Models, Animal , Mice , Urinary Tract Infections/microbiology , Animals , Bacteria/genetics , Bacterial Infections/pathology , Humans , Urinary Tract Infections/pathology
20.
J Immunol ; 192(4): 1718-31, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24453257

ABSTRACT

Genital tract carriage of group B streptococcus (GBS) is prevalent among adult women; however, the dynamics of chronic GBS genital tract carriage, including how GBS persists in this immunologically active host niche long term, are not well defined. To our knowledge, in this study, we report the first animal model of chronic GBS genital tract colonization using female mice synchronized into estrus by delivery of 17ß-estradiol prior to intravaginal challenge with wild-type GBS 874391. Cervicovaginal swabs, which were used to measure bacterial persistence, showed that GBS colonized the vaginal mucosa of mice at high numbers (10(6)-10(7) CFU/swab) for at least 90 d. Cellular and histological analyses showed that chronic GBS colonization of the murine genital tract caused significant lymphocyte and PMN cell infiltrates, which were localized to the vaginal mucosal surface. Long-term colonization was independent of regular hormone cycling. Immunological analyses of 23 soluble proteins related to chemotaxis and inflammation showed that the host response to GBS in the genital tract comprised markers of innate immune activation including cytokines such as GM-CSF and TNF-α. A nonhemolytic isogenic mutant of GBS 874391, Δcyle9, was impaired for colonization and was associated with amplified local PMN responses. Induction of DNA neutrophil extracellular traps, which was observed in GBS-infected human PMNs in vitro in a hemolysin-dependent manner, appeared to be part of this response. Overall, this study defines key infection dynamics in a novel murine model of chronic GBS genital tract colonization and establishes previously unknown cellular and soluble defense responses to GBS in the female genital tract.


Subject(s)
Hemolysin Proteins/metabolism , Reproductive Tract Infections/immunology , Streptococcus agalactiae/immunology , Vagina/immunology , Animals , Biomarkers , Cell Movement/immunology , Cytokines/metabolism , Disease Models, Animal , Estradiol/metabolism , Female , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Inflammation/immunology , Mice , Mice, Inbred C57BL , Neutrophils/immunology , Reproductive Tract Infections/microbiology , Streptococcal Infections/immunology , Streptococcal Infections/microbiology , Streptococcus agalactiae/metabolism , Tumor Necrosis Factor-alpha/metabolism , Vagina/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL