Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 938
Filter
1.
Pathol Int ; 74(10): 611-617, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39056535

ABSTRACT

Epstein-Barr virus (EBV) is a major cause of infectious mononucleosis (IM), characterized by fever, fatigue, sore throat, lymphadenopathy, atypical lymphocytosis, and elevated liver enzymes. However, ascites is a rare complication associated with IM. We present a rare case of IM with ascites and peritonitis in a patient who underwent a peritoneal biopsy. A 20-year-old woman presented with fatigue and abdominal distension. Laboratory examination revealed atypical lymphocytes in peripheral blood (54%) and elevated liver enzymes. EBV serological tests revealed a recent primary infection (EBV VCA IgM 1:160). Computed tomography revealed moderate ascites and peritonitis. Adenocarcinoma was suspected based on the ascites' cytology. Considering possible complications of IM and adenocarcinoma, a laparoscopic biopsy was performed. Histological findings of biopsy specimens from the peritoneum, omentum, and fimbria of the fallopian tube demonstrated severe inflammatory cell infiltration and focal aggregation of large EBV-encoded RNA-1 (EBER1)-positive B cells, mimicking EBV-positive polymorphous B-cell lymphoproliferative disorder. Furthermore, intracytoplasmic inclusion bodies of Chlamydia trachomatis were observed by immunohistochemistry. Real-time polymerase chain reaction detected C. trachomatis in cervical secretions. Two months after laparoscopy, ascites decreased, and the diagnosis was IM-associated peritonitis with C. trachomatis infection. IM should be considered as a differential diagnosis in young patients with ascites.


Subject(s)
Chlamydia Infections , Chlamydia trachomatis , Infectious Mononucleosis , Peritonitis , Humans , Female , Infectious Mononucleosis/pathology , Infectious Mononucleosis/complications , Infectious Mononucleosis/diagnosis , Peritonitis/pathology , Peritonitis/microbiology , Peritonitis/diagnosis , Young Adult , Chlamydia trachomatis/isolation & purification , Chlamydia Infections/pathology , Chlamydia Infections/complications , Chlamydia Infections/diagnosis , Herpesvirus 4, Human/isolation & purification , Diagnosis, Differential
2.
J Neuroinflammation ; 21(1): 158, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38879567

ABSTRACT

Respiratory infections are one of the most common causes of illness and morbidity in neonates worldwide. In the acute phase infections are known to cause wide-spread peripheral inflammation. However, the inflammatory consequences to the critical neural control centres for respiration have not been explored. Utilising a well characterised model of neonatal respiratory infection, we investigated acute responses within the medulla oblongata which contains key respiratory regions. Neonatal mice were intranasally inoculated within 24 h of birth, with either Chlamydia muridarum or sham-infected, and tissue collected on postnatal day 15, the peak of peripheral inflammation. A key finding of this study is that, while the periphery appeared to show no sex-specific effects of a neonatal respiratory infection, sex had a significant impact on the inflammatory response of the medulla oblongata. There was a distinct sex-specific response in the medulla coincident with peak of peripheral inflammation, with females demonstrating an upregulation of anti-inflammatory cytokines and males showing very few changes. Microglia also demonstrated sex-specificity with the morphology of females and males differing based upon the nuclei. Astrocytes showed limited changes during the acute response to neonatal infection. These data highlight the strong sex-specific impact of a respiratory infection can have on the medulla in the acute inflammatory phase.


Subject(s)
Animals, Newborn , Chlamydia Infections , Chlamydia muridarum , Animals , Mice , Female , Chlamydia Infections/microbiology , Chlamydia Infections/pathology , Male , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/pathology , Brain Stem/pathology , Neuroinflammatory Diseases/microbiology , Neuroinflammatory Diseases/pathology , Neuroinflammatory Diseases/immunology , Sex Characteristics , Mice, Inbred C57BL , Cytokines/metabolism
3.
PLoS Pathog ; 20(6): e1012303, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38885287

ABSTRACT

Chlamydia trachomatis is a clinically important bacterium that infects epithelial cells of the genitourinary and respiratory tracts and the eye. These differentiated cells are in a quiescent growth state and have a surface organelle called a primary cilium, but the standard Chlamydia cell culture infection model uses cycling cells that lack primary cilia. To investigate if these differences are relevant, we performed infections with host cells that have a primary cilium. We found that C. trachomatis caused progressive loss of the primary cilium that was prevented by disrupting Aurora A (AurA), HDAC6 or calmodulin, which are components of the cellular cilia disassembly pathway. Stabilization of the primary cilium by targeting this pathway caused a large reduction in infectious progeny although there were no changes in chlamydial inclusion growth, chlamydial replication or the ultrastructural appearance of dividing and infectious forms (RBs and EBs, respectively). Thus, the presence of a primary cilium interfered with the production of infectious EBs at a late step in the developmental cycle. C. trachomatis infection also induced quiescent cells to re-enter the cell cycle, as detected by EdU incorporation in S-phase, and Chlamydia-induced cilia disassembly was necessary for cell cycle re-entry. This study therefore describes a novel host-pathogen interaction in which the primary cilium limits a productive Chlamydia infection, and the bacterium counteracts this host cell defense by activating the cellular cilia disassembly pathway.


Subject(s)
Chlamydia Infections , Chlamydia trachomatis , Cilia , Chlamydia trachomatis/physiology , Cilia/microbiology , Cilia/metabolism , Chlamydia Infections/microbiology , Chlamydia Infections/metabolism , Chlamydia Infections/pathology , Humans , Epithelial Cells/microbiology , Epithelial Cells/metabolism
4.
Comp Med ; 74(2): 121-129, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38561234

ABSTRACT

Chlamydia muridarum (Cm), an intracellular bacterium of historical importance, was recently rediscovered as moderately prevalent in research mouse colonies. Cm was first reported as a causative agent of severe pneumonia in mice about 80 y ago, and while it has been used experimentally to model Chlamydia trachomatis infection of humans, there have been no further reports of clinical disease associated with natural infection. We observed clinical disease and pathology in 2 genetically engi- neered mouse (GEM) strains, Il12rb2 KO and STAT1 KO, with impaired interferon-γ signaling and Th1 CD4+ T cell responses in a colony of various GEM strains known to be colonized with and shedding Cm. Clinical signs included poor condition, hunched posture, and poor fecundity. Histopathology revealed disseminated Cm with lesions in pulmonary, gastrointestinal, and urogenital tissues. The presence of Cm was confirmed using both immunohistochemistry for Cm major outer membrane protein-1 antigen and in situ hybridization using a target probe directed against select regions of Cm strain Nigg. Cm was also found in association with a urothelial papilloma in one mouse. These cases provide additional support for excluding Cm from research mouse colonies.


Subject(s)
Chlamydia Infections , Chlamydia muridarum , Mice, Knockout , STAT1 Transcription Factor , Animals , Chlamydia Infections/pathology , Chlamydia Infections/veterinary , Chlamydia Infections/microbiology , Mice , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Female , Receptors, Interleukin-12/deficiency , Receptors, Interleukin-12/genetics , Male , Lung Diseases/microbiology , Lung Diseases/pathology , Lung Diseases/veterinary
6.
Vet Pathol ; 61(4): 621-632, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38240274

ABSTRACT

Chlamydiosis is one of the main causes of the progressive decline of koala populations in eastern Australia. While histologic, immunologic, and molecular studies have provided insights into the basic function of the koala immune system, the in situ immune cell signatures during chlamydial infection of the reproductive tract in koalas have not been investigated. Thirty-two female koalas and 47 males presented to wildlife hospitals with clinical signs suggestive of Chlamydia infection were euthanized with the entire reproductive tract collected for histology; immunohistochemistry (IHC) for T-cell (CD3ε, CD4, and CD8α), B-cell (CD79b), and human leukocyte antigen (HLA)-DR markers; and quantitative real-time polymerase chain reaction (rtPCR) for Chlamydia pecorum. T-cells, B-cells, and HLA-DR-positive cells were observed in both the lower and upper reproductive tracts of male and female koalas with a statistically significant associations between the degree of the inflammatory reaction; the number of CD3, CD4, CD79b, and HLA-DR positive cells; and the PCR load. CD4-positive cells were negatively associated with the severity of the gross lesions. The distribution of immune cells was also variable according to the location within the genital tract in both male and female koalas. These preliminary results represent a step forward towards further exploring mechanisms behind chlamydial infection immunopathogenesis, thus providing valuable information about the immune response and infectious diseases in free-ranging koalas.


Subject(s)
Chlamydia Infections , Chlamydia , Immunohistochemistry , Phascolarctidae , Animals , Phascolarctidae/microbiology , Female , Chlamydia Infections/veterinary , Chlamydia Infections/immunology , Chlamydia Infections/pathology , Chlamydia Infections/microbiology , Male , Immunohistochemistry/veterinary , Chlamydia/immunology , Reproductive Tract Infections/veterinary , Reproductive Tract Infections/microbiology , Reproductive Tract Infections/pathology , Reproductive Tract Infections/immunology , B-Lymphocytes/immunology , B-Lymphocytes/pathology , HLA-DR Antigens/metabolism , Australia , T-Lymphocytes/immunology
7.
Int J Biol Macromol ; 258(Pt 1): 128723, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38101679

ABSTRACT

The Pgp3 subunit vaccine elicits immune protection against Chlamydia trachomatis infection, but additional adjuvants are still required to enhance its immunoprotective efficacy. Flagellin can selectively stimulate immunity and act as an adjuvant. In this research, the FliC-Pgp3 recombinant was successfully expressed and purified. Tri-immunization with the FliC-Pgp3 vaccine in Balb/C mice induced rapid and persistent germinal center B-cell response and Tfh differentiation, promoting a significantly higher IgG antibody titer compared to the Pgp3 group. FliC-Pgp3 immunization primarily induced Th1-type cellular immunity, leading to higher levels of IFN-γ, TNF-α, and IL-2 secreted by CD4+ T cells than in Pgp3-vaccinated mice. Chlamydia muridarum challenge results showed that FliC-Pgp3-vaccinated mice exhibited more rapid clearance of Chlamydia muridarum colonization in the lower genital tract, ensuring a lower hydrosalpinx rate and cumulative score. Histological analysis showed reduced dilation and inflammatory infiltration in the oviduct and uterine horn of FliC-Pgp3-vaccinated mice compared to the PBS and Pgp3 control. Importantly, tri-immunization with FliC-Pgp3 effectively activated CD4+ T cells and dendritic cells, as confirmed by the adoptive transfer, resulting in better immune protection in recipient mice. In summary, the novel FliC-Pgp3 chimeric is hoped to be a novel vaccine with improved immunoprotection against Chlamydia muridarum.


Subject(s)
Chlamydia Infections , Chlamydia muridarum , Animals , Mice , Bacterial Proteins , Antigens, Bacterial , Chlamydia Infections/pathology , Chlamydia Infections/prevention & control , Immunization , Vaccines, Synthetic , Adjuvants, Immunologic
8.
Pathog Dis ; 812023 01 17.
Article in English | MEDLINE | ID: mdl-37480234

ABSTRACT

Mechanism of Chlamydia trachomatis causing tubal ectopic pregnancy (EP) is not well understood. Tetraspanins (tspans), activin-A, and inhibin-A might play a role in the development of pathological conditions leading to EP. The study aimed to elucidate the expression of tspans, activin-A, and inhibin-A with a role of associated cytokines in C. trachomatis-associated EP and analyze interacting partners of DEGs, with an expression of a few important interacting genes. Fallopian tissue and serum were collected from 100 EP (Group I) and 100 controls (Group II) from SJH, New Delhi, India. Detection of C. trachomatis was done by polymerase chain reaction (PCR) and IgG antibodies were detected by enzyme-linked immunosorbent assay. Expression of tspans, activin-A, inhibin-A, and cytokines was analyzed by real time (RT)-PCR and their interacting genes were assessed by STRING. Expression of few disease-associated interacting genes was studied by RT-PCR. A total of 29% (Group I) were C. trachomatis positive. Tspans and activin-A were significantly upregulated, while inhibin-A was significantly downregulated in Group Ia. ITGA1, TLR-2, ITGB2, and Smad-3 were a few interacting genes. Expression of ITGA1, TLR-2, and Smad-3 was significantly upregulated in C. trachomatis-positive EP. Results suggested dysregulated tspans, activin-A, and inhibin-A might play a role in C. trachomatis-infected tubal EP.


Subject(s)
Chlamydia Infections , Pregnancy, Ectopic , Pregnancy , Humans , Female , Pregnancy, Ectopic/etiology , Pregnancy, Ectopic/metabolism , Pregnancy, Ectopic/pathology , Chlamydia trachomatis/genetics , Toll-Like Receptor 2/genetics , Chlamydia Infections/pathology , Activins/genetics , Real-Time Polymerase Chain Reaction , Cytokines/genetics
9.
Front Cell Infect Microbiol ; 13: 1208302, 2023.
Article in English | MEDLINE | ID: mdl-37265500

ABSTRACT

Ocular, genital, and anogenital infection by the obligate intracellular pathogen Chlamydia trachomatis have been consistently associated with scar-forming sequelae. In cases of chronic or repeated infection of the female genital tract, infection-associated fibrosis of the fallopian tubes can result in ectopic pregnancy or infertility. In light of this urgent concern to public health, the underlying mechanism of C. trachomatis-associated scarring is a topic of ongoing study. Fibrosis is understood to be an outcome of persistent injury and/or dysregulated wound healing, in which an aberrantly activated myofibroblast population mediates hypertrophic remodeling of the basement membrane via deposition of collagens and other components of the extracellular matrix, as well as induction of epithelial cell proliferation via growth factor signaling. Initial study of infection-associated immune cell recruitment and pro-inflammatory signaling have suggested the cellular paradigm of chlamydial pathogenesis, wherein inflammation-associated tissue damage and fibrosis are the indirect result of an immune response to the pathogen initiated by host epithelial cells. However, recent work has revealed more direct routes by which C. trachomatis may induce scarring, such as infection-associated induction of growth factor signaling and pro-fibrotic remodeling of the extracellular matrix. Additionally, C. trachomatis infection has been shown to induce an epithelial-to-mesenchymal transition in host epithelial cells, prompting transdifferentiation into a myofibroblast-like phenotype. In this review, we summarize the field's current understanding of Chlamydia-associated fibrosis, reviewing key new findings and identifying opportunities for further research.


Subject(s)
Chlamydia Infections , Cicatrix , Humans , Pregnancy , Female , Chlamydia Infections/pathology , Epithelial Cells/pathology , Chlamydia trachomatis , Fibrosis
10.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 47(9): 1275-1280, 2022 Sep 28.
Article in English, Chinese | MEDLINE | ID: mdl-36411712

ABSTRACT

Chlamydia trachomatis ( CT ) genital tract infection is insidious, and patients often have no conscious symptoms.Delayed treatment after infection can lead to serious complications. Chlamydia muridarum ( CM ) genital tract infection in female mice can simulate CT genital tract infection in women, which is an ideal model to investigate the pathogenesis of CT . CM plasmid protein pGP3, chromosomal protein TC0237/TC0668, CM -specific CD8 + T cells, TNF-α, and IL-13 can induce genital tract inflammation, CD4 + T cells are responsible for CM clearance. However, tubal inflammation persists after genital tract CM is removed. Genital tract CM can spread spontaneously in vivo and colonize the gastrointestinal (GI) tract, but the GI tract CM cannot reverse spread to the genital tract. The survival time and number of CM transmitted from genital tract to GI tract are positively correlated with the long-term lesion of oviduct, while the CM inoculated directly into the GI tract has no pathogenicity in both the genital and GI tract. The double attack pattern of Chlamydia -induced genital tract inflammatory lesions is as follows: CM infection of oviduct epithelial cells initiates the process of oviduct repair as the first attack. After genital CM spreads to the GI tract, activated chlamydia-specific CD8 + T cells are recruited to the genital tract and secreted pro-fibrotic cytokines such as TNF-α and IL-13. This process is called the second attack which transform tubal repair initiated by the first attack into long-term tubal fibrosis/hydrosalpinx. Elucidating the pathogenic mechanism of Chlamydia infection can provide new ideas for the development of Chlamydia vaccine, which is expected to solve the problems of infertility caused by repeated CT infection in women.


Subject(s)
Chlamydia Infections , Chlamydia muridarum , Reproductive Tract Infections , Female , Mice , Animals , Interleukin-13 , CD8-Positive T-Lymphocytes/pathology , Tumor Necrosis Factor-alpha , Chlamydia Infections/pathology , Genitalia/pathology , Gastrointestinal Tract/pathology , Inflammation
11.
Vet Pathol ; 59(6): 983-996, 2022 11.
Article in English | MEDLINE | ID: mdl-36062911

ABSTRACT

This report describes the fetoplacental pathology of Chlamydia psittaci-associated abortion, premature birth, and neonatal loss in 46 of 442 equine abortion investigations between 2015 and 2019. Seven abortions, 26 premature births, and 13 neonatal deaths with positive C. psittaci polymerase chain reaction (PCR) were evaluated. In 83% of cases (38/46), C. psittaci infection was considered as the primary cause of loss based on quantitative PCR (qPCR) confirmation, pathological findings, and exclusion of other causes, and was supported by Chlamydia spp immunolabeling in fetoplacental lesions. Lymphohistiocytic placentitis with vasculitis (36/38) affected the amnion, umbilical cord, and chorioallantois at the umbilical vessel insertion and/or cervical pole. Lymphohistiocytic chorionitis in the subvillous stroma extended to the allantois mostly without villous destruction. Lymphohistiocytic amnionitis and funisitis occurred at the amniotic cord attachment. Lymphohistiocytic hepatitis was observed in 19/38 cases and pneumonia was identified in 26 cases. Chlamydia spp immunolabeled in placenta, lung, liver, or splenic tissue in the cases that were tested (14/38). C. psittaci infection was not the cause of loss in 2 cases with other diseases and of uncertain significance in 6 cases with no conclusive cause of loss. immunohistochemistry (IHC) was negative for 6 of these cases (6/8). The highest Chlamydia load was detected in pooled placental tissues by qPCR. qPCR and IHC had 83% congruence at a qPCR cut-off of 1 gene copy. IHC limits of detection corresponded to infections with 2 × 102 gene copies identified by qPCR. This study confirms the etiological role of C. psittaci as a cause of naturally occurring equine reproductive loss.


Subject(s)
Chlamydia Infections , Chlamydia , Chlamydophila psittaci , Chorioamnionitis , Horse Diseases , Premature Birth , Abortion, Veterinary/pathology , Animals , Chlamydia Infections/complications , Chlamydia Infections/pathology , Chlamydia Infections/veterinary , Chlamydophila psittaci/genetics , Chorioamnionitis/pathology , Chorioamnionitis/veterinary , Female , Horse Diseases/pathology , Horses , Placenta/pathology , Pregnancy , Premature Birth/pathology , Premature Birth/veterinary
12.
Microb Pathog ; 165: 105468, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35257805

ABSTRACT

Chlamydia trachomatis is an established risk factor for ectopic pregnancy (EP) in fallopian tube (FT). Matrix metalloproteinases (MMPs) have potential role in disease pathogenesis, however, dysregulation of extracellular matrix by MMPs/TIMPs (tissue inhibitors of MMPs) in infection-associated EP remains unknown. The aim was to study the expression of MMP-2, -9, -14/TIMP-1, -2, -3 in C. trachomatis-positive tubal EP patients. The study comprised of 100 tubal EP (Group I) and 100 tubal ligation patients (Group II; controls) enrolled from Department of Obstetrics and Gynaecology, VMMC and Safdarjung hospital, New Delhi (India) for collection of FT. Detection of C. trachomatis MOMP was done by PCR while quantitative expression of MMPs/TIMPs was studied by real-time PCR. Data was statistically evaluated by Graphpad prism. Overall, C. trachomatis was found in 18/100 tubal EP patients. After ruling out Neisseria gonnorhoeae and Mycoplasma genitalium, Group I was divided into Group Ia (C. trachomatis DNA-positive) and Group Ib (C. trachomatis DNA-negative; internal controls). Significant upregulation of MMP-2, -9, -14 and downregulated TIMP-1, -2, -3 were found in Group Ia versus controls (Groups Ib/II) (p < 0.05). Fold-change in MMP was significantly higher in Group Ia versus controls ('p' < 0.05). Maximum 5.5-fold upregulation was found in MMP-2. It is apparent by molecular analysis that differential expression of MMPs/TIMPs, particularly enhanced MMP-2 leads to tubal EP in C. trachomatis DNA-positive women.


Subject(s)
Chlamydia Infections , Pregnancy, Ectopic , Chlamydia Infections/pathology , Chlamydia trachomatis/genetics , Fallopian Tubes/pathology , Female , Humans , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Pregnancy , Pregnancy, Ectopic/metabolism , Pregnancy, Ectopic/pathology , Tissue Inhibitor of Metalloproteinase-1/metabolism
13.
Sci Rep ; 11(1): 23856, 2021 12 13.
Article in English | MEDLINE | ID: mdl-34903753

ABSTRACT

Chlamydia gallinacea is one of the new Chlamydia species, encountered predominantly in birds and occasionally in cattle, and its dissemination, pathogenicity and zoonotic potential have not yet been fully elucidated. Until now, no case of clinical infection has been described in poultry, but the number of studies is limited. This study was conducted to evaluate the course of infection and the impact on production parameters in chicken broilers inoculated with the strain 15-56/1 isolated from a Polish flock. The presence of C. gallinacea was confirmed in oropharyngeal and cloacal swabs by real-time PCR from the fifth day post inoculation (dpi). Pathogen DNA was also detected in many internal organs of inoculated chickens. All infected animals remained asymptomatic during the entire experimental period, although statistical analyses showed that broilers in the experimental group exhibited significantly lower body weight gains and feed conversion ratios than animals in the control group. These data indicate that subclinical C. gallinacea infection in broilers may lead to financial losses for poultry farmers.


Subject(s)
Bird Diseases/pathology , Chickens/microbiology , Chlamydia Infections/pathology , Chlamydia/pathogenicity , Animals , Bird Diseases/microbiology , Chickens/growth & development , Chlamydia Infections/microbiology , Weight Loss
14.
FEBS Lett ; 595(24): 2995-3005, 2021 12.
Article in English | MEDLINE | ID: mdl-34741525

ABSTRACT

Chlamydia trachomatis, an obligate intracellular bacterium with limited metabolic capabilities, possesses the futalosine pathway for menaquinone biosynthesis. Futalosine pathway enzymes have promise as narrow-spectrum antibiotic targets, but the activity and essentiality of chlamydial menaquinone biosynthesis have yet to be established. In this work, menaquinone-7 (MK-7) was identified as a C. trachomatis-produced quinone through liquid chromatography-tandem mass spectrometry. An immunofluorescence-based assay revealed that treatment of C. trachomatis-infected HeLa cells with the futalosine pathway inhibitor docosahexaenoic acid (DHA) reduced inclusion number, inclusion size, and infectious progeny. Supplementation with MK-7 nanoparticles rescued the effect of DHA on inclusion number, indicating that the futalosine pathway is a target of DHA in this system. These results open the door for menaquinone biosynthesis inhibitors to be pursued in antichlamydial development.


Subject(s)
Biosynthetic Pathways , Chlamydia Infections/pathology , Chlamydia trachomatis/physiology , Nucleosides/biosynthesis , Vitamin K 2/analogs & derivatives , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Automation , Biosynthetic Pathways/drug effects , Chlamydia Infections/microbiology , Docosahexaenoic Acids/pharmacology , HeLa Cells , Humans , Inclusion Bodies/drug effects , Inclusion Bodies/metabolism , Nanoparticles/chemistry , Nucleosides/chemistry , Vitamin K 2/chemistry , Vitamin K 2/metabolism
15.
J Infect Dis ; 224(12 Suppl 2): S121-S127, 2021 08 16.
Article in English | MEDLINE | ID: mdl-34396414

ABSTRACT

Epidemiologic, clinical, molecular and translational research findings support an interrelationship between Chlamydia trachomatis, pelvic inflammatory disease (PID), and epithelial ovarian cancer (EOC). Overall, the link between C. trachomatis, PID, and EOC seems to be relatively weak, although nondifferential misclassification bias may have attenuated the results. The predominant tubal origin of EOC and the role of chronic inflammation in tumorigenesis suggest that the association is biologically plausible. Thus, C. trachomatis and PID may represent potential risk factors or risk markers for EOC. However, many steps in this chain of events are still poorly understood and need to be addressed in future studies. Research gaps include time of exposure in relation to the long-term consequences and lag time to EOC. Data of differential risk for EOC between chlamydial and nonchlamydial PID is also needed. Another major research gap has been the absence of high-performance biomarkers for C. trachomatis, PID, and EOC, as well as EOC precursors. Biomarkers for C. trachomatis and PID leading to increased risk of EOC should be developed. If the association is confirmed, C. trachomatis and PID prevention efforts may play a role in reducing the burden of EOC.


Subject(s)
Carcinoma, Ovarian Epithelial , Chlamydia Infections/complications , Chlamydia Infections/pathology , Chlamydia trachomatis , Ovarian Neoplasms , Pelvic Inflammatory Disease/microbiology , Biomarkers , Carcinoma, Ovarian Epithelial/epidemiology , Carcinoma, Ovarian Epithelial/etiology , Carcinoma, Ovarian Epithelial/virology , Chlamydia Infections/epidemiology , Female , Humans , Ovarian Neoplasms/epidemiology , Ovarian Neoplasms/etiology , Ovarian Neoplasms/virology , Pelvic Inflammatory Disease/epidemiology
16.
PLoS One ; 16(5): e0251113, 2021.
Article in English | MEDLINE | ID: mdl-33970945

ABSTRACT

BACKGROUND: The United States is experiencing a surge in Chlamydia trachomatis (CT) infections representing a critical need to improve sexually transmitted infection (STI) screening and treatment programs. To understand where patients with STIs seek healthcare, we evaluated the relationship between CT infections and the place where individuals report usually receiving healthcare. METHODS: Our study used a nationally representative sample from the National Health and Nutrition Examination Survey (NHANES) from 1999 to 2016. The study population is adult patients, aged 18 to 39 years in whom a urine CT screen was obtained. Logistic regression models were used to determine if location of usual healthcare was predictive of a positive urine CT screen result. Models were adjusted for known confounders including age, gender, race/ethnicity, education, and insurance status. RESULTS: In this nationally representative sample (n = 19,275; weighted n = 85.8 million), 1.9% of individuals had a positive urine CT result. Participants reported usually going to the doctor's office (70.3%), "no place" (24.8%), Emergency Department (ED) (3.3%), or "other" place (1.7%) for healthcare. In adjusted models, the predicted probability of having a positive urine CT result is higher (4.9% vs 3.2%, p = 0.022; OR = 1.58) among those that reported the ED as their usual place for healthcare compared to those that reported going to a doctor's office or clinic. CONCLUSIONS: Individuals having a positive urine CT screen are associated with using the ED as a usual source for healthcare. Understanding this association has the potential to improve STI clinical and policy interventions as the ED may be a critical site in combatting the record high rates of STIs.


Subject(s)
Chlamydia Infections/epidemiology , Chlamydia Infections/therapy , Chlamydia trachomatis/isolation & purification , Sexually Transmitted Diseases/epidemiology , Sexually Transmitted Diseases/therapy , Adolescent , Adult , Chlamydia Infections/microbiology , Chlamydia Infections/pathology , Databases, Factual , Delivery of Health Care/methods , Emergency Service, Hospital , Female , Humans , Male , Nutrition Surveys/methods , Prevalence , Retrospective Studies , Sexually Transmitted Diseases/microbiology , Sexually Transmitted Diseases/pathology , United States/epidemiology , Young Adult
17.
Pathog Dis ; 79(4)2021 03 31.
Article in English | MEDLINE | ID: mdl-33693620

ABSTRACT

Chlamydia trachomatis is the most commonly reported sexually transmitted infection in the United States. The high prevalence of infection and lack of a vaccine indicate a critical knowledge gap surrounding the host's response to infection and how to effectively generate protective immunity. The immune response to C. trachomatis is complex, with cells of the adaptive immune system playing a crucial role in bacterial clearance. Here, we discuss the CD4+ and CD8+ T cell response to Chlamydia, the importance of antigen specificity and the role of memory T cells during the recall response. Ultimately, a deeper understanding of protective immune responses is necessary to develop a vaccine that prevents the inflammatory diseases associated with Chlamydia infection.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Chlamydia Infections/immunology , Chlamydia trachomatis/pathogenicity , Immune Evasion , Adaptive Immunity , Animals , Bacterial Load , CD4-Positive T-Lymphocytes/microbiology , CD8-Positive T-Lymphocytes/microbiology , Chlamydia Infections/complications , Chlamydia Infections/microbiology , Chlamydia Infections/pathology , Chlamydia muridarum/growth & development , Chlamydia muridarum/immunology , Chlamydia muridarum/pathogenicity , Chlamydia trachomatis/growth & development , Chlamydia trachomatis/immunology , Genitalia/immunology , Genitalia/microbiology , Genitalia/pathology , Humans , Immunity, Innate , Immunologic Memory , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukins/biosynthesis , Interleukins/immunology , Mice
18.
PLoS Pathog ; 17(2): e1009295, 2021 02.
Article in English | MEDLINE | ID: mdl-33635920

ABSTRACT

To date, no reports have linked the multifunctional protein, staphylococcal nuclease domain-containing protein 1 (SND1), to host defense against intracellular infections. In this study, we investigated the role and mechanisms of SND1, by using SND1 knockout (SND1-/-) mice, in host defense against the lung infection of Chlamydia muridarum, an obligate intracellular bacterium. Our data showed that SND1-/- mice exhibited significantly greater body weight loss, higher organism growth, and more severe pathological changes compared with wild-type mice following the infection. Further analysis showed significantly reduced Chlamydia-specific Th1/17 immune responses in SND1-/- mice after infection. Interestingly, the dendritic cells (DCs) isolated from SND1-/- mice showed lower costimulatory molecules expression and IL-12 production, but higher IL-10 production compared with those from wild-type control mice. In the DC-T cell co-culture system, DCs isolated from SND1-/- infected mice showed significantly reduced ability to promote Chlamydia-specific IFN-γ producing Th1 cells but enhanced capacity to induce CD4+T cells into Foxp3+ Treg cells. Adoptive transfer of DCs isolated from SND1-/- mice, unlike those from wild-type control mice, failed to protect the recipients against challenge infection. These findings provide in vivo evidence that SND1 plays an important role in host defense against intracellular bacterial infection, and suggest that SND1 can promote Th1/17 immunity and inhibit the expansion of Treg cells through modulation of the function of DCs.


Subject(s)
Chlamydia Infections/immunology , Chlamydia muridarum/immunology , Dendritic Cells/immunology , Endonucleases/physiology , Lung/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Animals , Chlamydia Infections/metabolism , Chlamydia Infections/microbiology , Chlamydia Infections/pathology , Dendritic Cells/metabolism , Dendritic Cells/microbiology , Female , Immunity, Cellular/immunology , Lung/metabolism , Lung/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout
19.
Pathog Dis ; 79(4)2021 03 31.
Article in English | MEDLINE | ID: mdl-33538819

ABSTRACT

Chlamydiae are pathogenic intracellular bacteria that cause a wide variety of diseases throughout the globe, affecting the eye, lung, coronary arteries and female genital tract. Rather than by direct cellular toxicity, Chlamydia infection generally causes pathology by inducing fibrosis and scarring that is largely mediated by host inflammation. While a robust immune response is required for clearance of the infection, certain elements of that immune response may also damage infected tissue, leading to, in the case of female genital infection, disease sequelae such as pelvic inflammatory disease, infertility and ectopic pregnancy. It has become increasingly clear that the components of the immune system that destroy bacteria and those that cause pathology only partially overlap. In the ongoing quest for a vaccine that prevents Chlamydia-induced disease, it is important to target mechanisms that can achieve protective immunity while preventing mechanisms that damage tissue. This review focuses on mouse models of genital Chlamydia infection and synthesizes recent studies to generate a comprehensive model for immunity in the murine female genital tract, clarifying the respective contributions of various branches of innate and adaptive immunity to both host protection and pathogenic genital scarring.


Subject(s)
Chlamydia Infections/immunology , Chlamydia trachomatis/pathogenicity , Cicatrix/immunology , Host-Pathogen Interactions/immunology , Interferon-gamma/immunology , Interleukins/immunology , Adaptive Immunity , Animals , Bacterial Load , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/microbiology , Chlamydia Infections/complications , Chlamydia Infections/microbiology , Chlamydia Infections/pathology , Chlamydia muridarum/growth & development , Chlamydia muridarum/immunology , Chlamydia muridarum/pathogenicity , Chlamydia trachomatis/growth & development , Chlamydia trachomatis/immunology , Cicatrix/complications , Cicatrix/microbiology , Cicatrix/pathology , Disease Models, Animal , Female , Genitalia/immunology , Genitalia/microbiology , Genitalia/pathology , Humans , Immunity, Innate , Interferon-gamma/biosynthesis , Interleukins/biosynthesis , Mice , Pregnancy
20.
J Immunol ; 206(6): 1251-1265, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33504621

ABSTRACT

Recent studies have identified semaphorin 3E (Sema3E) as a novel mediator of immune responses. However, its function in immunity to infection has yet to be investigated. Using a mouse model of chlamydial lung infection, we show that Sema3E plays a significant role in the host immune response to the infection. We found that Sema3E is induced in the lung after chlamydial infection, and Sema3E deficiency has a detrimental impact on disease course, dendritic cell (DC) function, and T cell responses. Specifically, we found that Sema3E knockout (KO) mice exhibited higher bacterial burden, severe body weight loss, and pathological changes after Chlamydia muridarum lung infection compared with wild-type (WT) mice. The severity of disease in Sema3E KO mice was correlated with reduced Th1/Th17 cytokine responses, increased Th2 response, altered Ab response, and a higher number of regulatory CD4 T cells. Moreover, DCs isolated from Sema3E KO mice showed lower surface expression of costimulatory molecules and production of IL-12, but higher expression of PD-L1, PD-L2, and IL-10 production. Functional DC-T cell coculture studies revealed that DCs from infected Sema3E KO mice failed to induce Th1 and Th17 cell responses compared with DCs from infected WT mice. Upon adoptive transfer, mice receiving DCs from Sema3E KO mice, unlike those receiving DCs from WT mice, were not protected against challenge infection. In conclusion, our data evidenced that Sema3E acts as a critical factor for protective immunity against intracellular bacterial infection by modulating DC functions and T cell subsets.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Chlamydia Infections/immunology , Dendritic Cells/immunology , Semaphorins/metabolism , T-Lymphocyte Subsets/immunology , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/metabolism , Chlamydia Infections/microbiology , Chlamydia Infections/pathology , Chlamydia muridarum/immunology , Coculture Techniques , Dendritic Cells/transplantation , Disease Models, Animal , Humans , Lung/immunology , Lung/microbiology , Lung/pathology , Mice , Mice, Knockout , Semaphorins/genetics , Severity of Illness Index , T-Lymphocyte Subsets/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL