Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 34
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Infect Immun ; 91(2): e0039222, 2023 02 16.
Article de Anglais | MEDLINE | ID: mdl-36722979

RÉSUMÉ

Chlamydia trachomatis is the most common cause of infectious blindness and sexually transmitted bacterial infection globally. C. trachomatis contains a conserved chlamydial plasmid with eight coding sequences. Plasmid-cured Chlamydia strains are attenuated and display reduced infectivity in cell culture and in vivo genital infection of female mice. Mutants that do not express the plasmid-encoded proteins Pgp3, a secreted protein with unknown function, or Pgp4, a putative regulator of pgp3 and other chromosomal loci, display an infectivity defect similar to plasmid-deficient strains. Our objective was to determine the combined and individual contributions of Pgp3 and Pgp4 to this phenotype. Deletion of pgp3 and pgp4 resulted in an infectivity defect detected by competition assay in cell culture and in mice. The pgp3 locus was placed under the control of an anhydrotetracycline-inducible promoter to examine the individual contributions of Pgp3 and Pgp4 to infectivity. Expression of pgp3 was induced 100- to 1,000-fold after anhydrotetracycline administration, regardless of the presence or absence of pgp4. However, secreted Pgp3 was not detected when pgp4 was deleted, confirming a role for Pgp4 in Pgp3 secretion. We discovered that expression of pgp3 or pgp4 alone was insufficient to restore normal infectivity, which required expression of both Pgp3 and Pgp4. These results suggest Pgp3 and Pgp4 are both required for infectivity during C. trachomatis infection. Future studies are required to determine the mechanism by which Pgp3 and Pgp4 influence chlamydial infectivity as well as the potential roles of Pgp4-regulated loci.


Sujet(s)
Infections à Chlamydia , Chlamydia trachomatis , Animaux , Femelle , Souris , Antigènes bactériens/génétique , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme , Infections à Chlamydia/microbiologie , Chlamydia trachomatis/génétique , Chlamydia trachomatis/pathogénicité , Plasmides/génétique , Virulence/génétique
2.
Pathogens ; 10(10)2021 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-34684219

RÉSUMÉ

Chlamydia trachomatis (Ct) causes the most prevalent bacterial sexually transmitted disease leading to ectopic pregnancy and infertility. Swine not only have many similarities to humans, but they are also susceptible to Ct. Despite these benefits and the ease of access to primary tissue from this food animal, in vitro research in swine has been underutilized. This study will provide basic understanding of the Ct host-pathogen interactions in porcine oviduct epithelial cells (pOECs)-the counterparts of human Fallopian tube epithelial cells. Using NanoString technology, flow cytometry, and confocal and transmission-electron microscopy, we studied the Ct developmental cycle in pOECs, the cellular immune response, and the expression and location of the tight junction protein claudin-4. We show that Ct productively completes its developmental cycle in pOECs and induces an immune response to Ct similar to human cells: Ct mainly induced the upregulation of interferon regulated genes and T-cell attracting chemokines. Furthermore, Ct infection induced an accumulation of claudin-4 in the Ct inclusion with a coinciding reduction of membrane-bound claudin-4. Downstream effects of the reduced membrane-bound claudin-4 expression could potentially include a reduction in tight-junction expression, impaired epithelial barrier function as well as increased susceptibility to co-infections. Thereby, this study justifies the investigation of the effect of Ct on tight junctions and the mucosal epithelial barrier function. Taken together, this study demonstrates that primary pOECs represent an excellent in vitro model for research into Ct pathogenesis, cell biology and immunity.

3.
J Infect Dis ; 224(12 Suppl 2): S47-S55, 2021 08 16.
Article de Anglais | MEDLINE | ID: mdl-34396406

RÉSUMÉ

Chlamydia trachomatis-genital infection in women can be modeled in mice using Chlamydia muridarum. Using this model, it has been shown that the cytokines tumor necrosis factor (TNF)α and interleukin (IL)-1α lead to irreversible tissue damage in the oviducts. In this study, we investigated the contribution of TNFα on IL-1α synthesis in infected epithelial cells. We show that C muridarum infection enhanced TNFα-induced IL-1α expression and release in a mouse epithelial cell line. In addition to IL-1α, several TNFα-induced inflammatory genes were also highly induced, and infection enhanced TNF-induced cell death. In the mouse model of genital infection, oviducts from mice lacking the TNFα receptor displayed minimal staining for IL-1α compared with wild-type oviducts. Our results suggest TNFα and IL-1α enhance each other's downstream effects resulting in a hyperinflammatory response to chlamydial infection. We propose that biologics targeting TNF-induced IL-1α synthesis could be used to mitigate tissue damage during chlamydial infection.


Sujet(s)
Mort cellulaire , Infections à Chlamydia , Chlamydia muridarum/immunologie , Interleukine-1 alpha , Facteur de nécrose tumorale alpha , Animaux , Infections à Chlamydia/immunologie , Infections à Chlamydia/métabolisme , Cellules épithéliales , Femelle , Interleukine-1 alpha/immunologie , Interleukine-1 alpha/métabolisme , Souris , Souris de lignée C57BL , Facteur de nécrose tumorale alpha/immunologie , Facteur de nécrose tumorale alpha/métabolisme
4.
Infect Immun ; 89(10): e0007221, 2021 09 16.
Article de Anglais | MEDLINE | ID: mdl-34125599

RÉSUMÉ

Genital infections with Chlamydia trachomatis can lead to uterine and oviduct tissue damage in the female reproductive tract. Neutrophils are strongly associated with tissue damage during chlamydial infection, while an adaptive CD4 T cell response is necessary to combat infection. Activation of triggering receptor expressed on myeloid cells-1 (TREM-1) on neutrophils has previously been shown to induce and/or enhance degranulation synergistically with Toll-like receptor (TLR) signaling. Additionally, TREM-1 can promote neutrophil transepithelial migration. In this study, we sought to determine the contribution of TREM-1,3 to immunopathology in the female mouse genital tract during Chlamydia muridarum infection. Relative to control mice, trem1,3-/- mice had no difference in chlamydial burden or duration of lower-genital-tract infection. We also observed a similar incidence of hydrosalpinx 45 days postinfection in trem1,3-/- compared to wild-type (WT) mice. However, compared to WT mice, trem1,3-/- mice developed significantly fewer hydrometra in uterine horns. Early in infection, trem1,3-/- mice displayed a notable decrease in the number of uterine glands containing polymorphonuclear cells and uterine horn lumens had fewer neutrophils, with increased granulocyte colony-stimulating factor (G-CSF). trem1,3-/- mice also had reduced erosion of the luminal epithelium. These data indicate that TREM-1,3 contributes to transepithelial neutrophil migration in the uterus and uterine glands, promoting the occurrence of hydrometra in infected mice.


Sujet(s)
Infections à Chlamydia/immunologie , Chlamydia muridarum/immunologie , Récepteurs immunologiques/immunologie , Récepteur de déclenchement de type-1 exprimé sur les cellules myéloïdes/immunologie , Utérus/immunologie , Immunité acquise/immunologie , Animaux , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/microbiologie , Mouvement cellulaire/immunologie , Infections à Chlamydia/métabolisme , Infections à Chlamydia/microbiologie , Chlamydia trachomatis/immunologie , Modèles animaux de maladie humaine , Épithélium/immunologie , Épithélium/métabolisme , Épithélium/microbiologie , Femelle , Système génital de la femme/immunologie , Système génital de la femme/métabolisme , Système génital de la femme/microbiologie , Souris , Souris de lignée C57BL , Granulocytes neutrophiles/immunologie , Granulocytes neutrophiles/métabolisme , Granulocytes neutrophiles/microbiologie , Oviductes/immunologie , Oviductes/métabolisme , Oviductes/microbiologie , Récepteurs immunologiques/métabolisme , Infections de l'appareil reproducteur/immunologie , Infections de l'appareil reproducteur/métabolisme , Infections de l'appareil reproducteur/microbiologie , Récepteur de déclenchement de type-1 exprimé sur les cellules myéloïdes/métabolisme , Utérus/métabolisme , Utérus/microbiologie
5.
J Immunol ; 205(11): 3037-3049, 2020 12 01.
Article de Anglais | MEDLINE | ID: mdl-33087404

RÉSUMÉ

Chlamydia trachomatis infection of the female genital tract can lead to irreversible fallopian tube scarring. In the mouse model of genital infection using Chlamydia muridarum, IL-1R signaling plays a critical role in oviduct tissue damage. In this study, we investigated the pathologic role of IL-1α, one of the two proinflammatory cytokines that bind to IL-1R. Il1a-/- mice infected with C. muridarum cleared infection at their cervix at the same rate as wild-type (WT) mice, but were significantly protected from end point oviduct damage and fibrosis. The contribution of IL-1α to oviduct pathology was more dramatic than observed in mice deficient for IL-1ß. Although chlamydial burden was similar in WT and Il1a-/- oviduct during peak days of infection, levels of IL-1ß, IL-6, CSF3, and CXCL2 were reduced in Il1a-/- oviduct lysates. During infection, Il1a-/- oviducts and uterine horns exhibited reduced neutrophil infiltration, and this reduction persisted after the infection resolved. The absence of IL-1α did not compromise CD4 T cell recruitment or function during primary or secondary chlamydial infection. IL-1α is expressed predominantly by luminal cells of the genital tract in response to infection, and low levels of expression persisted after the infection cleared. Ab-mediated depletion of IL-1α in WT mice prevented infection-induced oviduct damage, further supporting a key role for IL-1α in oviduct pathology.


Sujet(s)
Infections à Chlamydia/métabolisme , Système génital de la femme/métabolisme , Interleukine-1 alpha/métabolisme , Oviductes/métabolisme , Animaux , Lymphocytes T CD4+/métabolisme , Col de l'utérus/métabolisme , Col de l'utérus/microbiologie , Infections à Chlamydia/microbiologie , Chlamydia muridarum/pathogénicité , Modèles animaux de maladie humaine , Femelle , Système génital de la femme/microbiologie , Interleukine-1 bêta/métabolisme , Souris , Souris de lignée C57BL , Infiltration par les neutrophiles/physiologie , Oviductes/microbiologie , Infections de l'appareil reproducteur/métabolisme , Infections de l'appareil reproducteur/microbiologie
6.
Infect Immun ; 88(9)2020 08 19.
Article de Anglais | MEDLINE | ID: mdl-32601108

RÉSUMÉ

Chlamydia trachomatis infection of the human fallopian tubes can lead to damaging inflammation and scarring, ultimately resulting in infertility. To study the human cellular responses to chlamydial infection, researchers have frequently used transformed cell lines that can have limited translational relevance. We developed a primary human fallopian tube epithelial cell model based on a method previously established for culture of primary human bronchial epithelial cells. After protease digestion and physical dissociation of excised fallopian tubes, epithelial cell precursors were expanded in growth factor-containing medium. Expanded cells were cryopreserved to generate a biobank of cells from multiple donors and cultured at an air-liquid interface. Culture conditions stimulated cellular differentiation into polarized mucin-secreting and multiciliated cells, recapitulating the architecture of human fallopian tube epithelium. The polarized and differentiated cells were infected with a clinical isolate of C. trachomatis, and inclusions containing chlamydial developmental forms were visualized by fluorescence and electron microscopy. Apical secretions from infected cells contained increased amounts of proteins associated with chlamydial growth and replication, including transferrin receptor protein 1, the amino acid transporters SLC3A2 and SLC1A5, and the T-cell chemoattractants CXCL10, CXCL11, and RANTES. Flow cytometry revealed that chlamydial infection induced cell surface expression of T-cell homing and activation proteins, including ICAM-1, VCAM-1, HLA class I and II, and interferon gamma receptor. This human fallopian tube epithelial cell culture model is an important tool with translational potential for studying cellular responses to Chlamydia and other sexually transmitted pathogens.


Sujet(s)
Cellules épithéliales/immunologie , Régulation de l'expression des gènes/immunologie , Interactions hôte-microbes/immunologie , Lymphocytes T/immunologie , Adulte , Système ASC de transport d'acides aminés/génétique , Système ASC de transport d'acides aminés/immunologie , Antigènes CD/génétique , Antigènes CD/immunologie , Marqueurs biologiques/métabolisme , Chimiokine CCL5/génétique , Chimiokine CCL5/immunologie , Chimiokine CXCL10/génétique , Chimiokine CXCL10/immunologie , Chimiokine CXCL11/génétique , Chimiokine CXCL11/immunologie , Infections à Chlamydia/génétique , Infections à Chlamydia/immunologie , Infections à Chlamydia/microbiologie , Chlamydia trachomatis/croissance et développement , Chlamydia trachomatis/immunologie , Cellules épithéliales/microbiologie , Trompes utérines/cytologie , Trompes utérines/chirurgie , Femelle , Chaine lourde de l'antigène CD98/génétique , Chaine lourde de l'antigène CD98/immunologie , Antigènes d'histocompatibilité de classe I/génétique , Antigènes d'histocompatibilité de classe I/immunologie , Antigènes d'histocompatibilité de classe II/génétique , Antigènes d'histocompatibilité de classe II/immunologie , Interactions hôte-microbes/génétique , Humains , Molécule-1 d'adhérence intercellulaire/génétique , Molécule-1 d'adhérence intercellulaire/immunologie , Antigènes mineurs d'histocompatibilité/génétique , Antigènes mineurs d'histocompatibilité/immunologie , Modèles biologiques , Culture de cellules primaires , Récepteur interféron/génétique , Récepteur interféron/immunologie , Récepteurs à la transferrine/génétique , Récepteurs à la transferrine/immunologie , Salpingectomie , Lymphocytes T/microbiologie , Molécule-1 d'adhérence des cellules vasculaires/génétique , Molécule-1 d'adhérence des cellules vasculaires/immunologie ,
7.
Atherosclerosis ; 290: 103-110, 2019 11.
Article de Anglais | MEDLINE | ID: mdl-31604170

RÉSUMÉ

BACKGROUND AND AIMS: Atherosclerosis is a chronic inflammatory disease, and recent studies have shown that infection at remote sites can contribute to the progression of atherosclerosis in hyperlipidemic mouse models. In this report, we tested the hypothesis that genital Chlamydia infection could accelerate the onset and progression of atherosclerosis. METHODS: Apolipoprotein E (Apoe-/-) and LDL receptor knockout (Ldlr-/-) mice on a high-fat diet were infected intra-vaginally with Chlamydia muridarum. Atherosclerotic lesions on the aortic sinuses and in the descending aorta were assessed at 8-weeks post-infection. Systemic, macrophage, and vascular site inflammatory responses were assessed and quantified. RESULTS: Compared to the uninfected groups, infected Apoe-/- and Ldlr-/- mice developed significantly more atherosclerotic lesions in the aortic sinus and in the descending aorta. Increased lesions were associated with higher circulating levels of serum amyloid A-1, IL-1ß, TNF-α, and increased VCAM-1 expression in the aortic sinus, suggesting an association with inflammatory responses observed during C. muridarum infection. Genital infection courses were similar in Apoe-/-, Ldlr-/-, and wild type mice. Further, Apoe-/- mice developed severe uterine pathology with increased dilatations. Apoe-deficiency also augmented cytokine/chemokine response in C. muridarum infected macrophages, suggesting that the difference in macrophage response could have contributed to the genital pathology in Apoe-/- mice. CONCLUSIONS: Overall, these studies demonstrate that genital Chlamydia infection exacerbates atherosclerotic lesions in hyperlipidemic mouse and suggest a novel role for Apoe in full recovery of uterine anatomy after chlamydial infection.


Sujet(s)
Maladies de l'aorte/étiologie , Athérosclérose/étiologie , Infections à Chlamydia/complications , Chlamydia muridarum/pathogénicité , Hyperlipidémies/complications , Infections de l'appareil reproducteur/complications , Utérus/microbiologie , Animaux , Maladies de l'aorte/métabolisme , Maladies de l'aorte/microbiologie , Maladies de l'aorte/anatomopathologie , Athérosclérose/métabolisme , Athérosclérose/microbiologie , Athérosclérose/anatomopathologie , Cellules cultivées , Infections à Chlamydia/microbiologie , Infections à Chlamydia/anatomopathologie , Cytokines/sang , Modèles animaux de maladie humaine , Évolution de la maladie , Femelle , Hyperlipidémies/métabolisme , Médiateurs de l'inflammation/sang , Macrophages/métabolisme , Macrophages/microbiologie , Souris invalidées pour les gènes ApoE , Plaque d'athérosclérose , Récepteurs aux lipoprotéines LDL/déficit , Récepteurs aux lipoprotéines LDL/génétique , Infections de l'appareil reproducteur/microbiologie , Infections de l'appareil reproducteur/anatomopathologie , Facteurs temps , Utérus/anatomopathologie
8.
Infect Immun ; 87(8)2019 08.
Article de Anglais | MEDLINE | ID: mdl-31160363

RÉSUMÉ

It has been shown that caspase-1, but not its upstream activator, ASC, contributes to oviduct pathology during mouse genital Chlamydia muridarum infection. We hypothesized that this dichotomy is due to the inadvertent absence of caspase-11 in previously used caspase-1-deficient mice. To address this, we studied the independent contributions of caspase-1 and -11 during genital Chlamydia infection. Our results show that caspase-11 deficiency was sufficient to recapitulate the effect of the combined absence of both caspase-1 and caspase-11 on oviduct pathology. Further, mice that were deficient for both caspase-1 and -11 but that expressed caspase-11 as a transgene (essentially, caspase-1-deficient mice) had no significant difference in oviduct pathology from control mice. Caspase-11-deficient mice showed reduced dilation in both the oviducts and uterus. To determine the mechanism by which caspase-11-deficient mice developed reduced pathology, the chlamydial burden and immune cell infiltration were determined in the oviducts. In the caspase-11-deficient mice, we observed increased chlamydial burdens in the upper genital tract, which correlated with increased CD4 T cell recruitment, suggesting a contribution of caspase-11 in infection control. Additionally, there were significantly fewer neutrophils in the oviducts of caspase-11-deficient mice, supporting the observed decrease in the incidence of oviduct pathology. Therefore, caspase-11 activation contributes to pathogen control and oviduct disease independently of caspase-1 activation.


Sujet(s)
Caspases/physiologie , Infections à Chlamydia/anatomopathologie , Oviductes/anatomopathologie , Infections de l'appareil reproducteur/anatomopathologie , Animaux , Caspase-1/physiologie , Caspases/génétique , Caspases initiatrices , Femelle , Souris , Souris de lignée C57BL , Infiltration par les neutrophiles
9.
Infect Immun ; 86(7)2018 07.
Article de Anglais | MEDLINE | ID: mdl-29661927

RÉSUMÉ

CD4 T cells and antibody are required for optimal acquired immunity to Chlamydia muridarum genital tract infection, and T cell-mediated gamma interferon (IFN-γ) production is necessary to clear infection in the absence of humoral immunity. However, the role of T cell-independent immune responses during primary infection remains unclear. We investigated this question by inoculating wild-type and immune-deficient mice with C. muridarum CM001, a clonal isolate capable of enhanced extragenital replication. Genital inoculation of wild-type mice resulted in transient dissemination to the lungs and spleen that then was rapidly cleared from these organs. However, CM001 genital infection proved lethal for STAT1-/- and IFNG-/- mice, in which IFN-γ signaling was absent, and for Rag1-/- mice, which lacked T and B cells and in which innate IFN-γ signaling was retained. In contrast, B cell-deficient muMT mice, which can generate a Th1 response, and T cell-deficient mice with intact B cell and innate IFN-γ signaling survived. These data collectively indicate that IFN-γ prevents lethal CM001 dissemination in the absence of T cells and suggests a B cell corequirement. Adoptive transfer of convalescent-phase immune serum but not naive IgM to Rag1-/- mice infected with CM001 significantly increased the survival time, while transfer of naive B cells completely rescued Rag1-/- mice from CM001 lethality. Protection was associated with a significant reduction in the lung chlamydial burden of genitally infected mice. These data reveal an important cooperation between T cell-independent B cell responses and innate IFN-γ in chlamydial host defense and suggest that interactions between T cell-independent antibody and IFN-γ are essential for limiting extragenital dissemination.


Sujet(s)
Lymphocytes B/immunologie , Infections à Chlamydia/immunologie , Chlamydia muridarum , Interféron gamma/immunologie , Infections de l'appareil reproducteur/immunologie , Lymphocytes T/physiologie , Animaux , Infections à Chlamydia/mortalité , Chlamydia muridarum/génétique , Femelle , Protéines à homéodomaine/physiologie , Souris , Souris de lignée C57BL , Plasmides , Infections de l'appareil reproducteur/mortalité
10.
J Immunol ; 200(8): 2941-2956, 2018 04 15.
Article de Anglais | MEDLINE | ID: mdl-29531169

RÉSUMÉ

Sexually transmitted infections with Chlamydia trachomatis and/or Neisseria gonorrhoeae and rates of pelvic inflammatory disease (PID) in women continue to rise, with reinfection being common because of poor adaptive immunity. Diagnosis remains imprecise, and pathogenesis data are derived primarily from monoinfection of mice with C. trachomatis or N. gonorrhoeae By comparing blood mRNA responses of women with C. trachomatis- and/or N. gonorrhoeae-induced PID and histologic endometritis with those from women with C. trachomatis and/or N. gonorrhoeae infection limited to their cervix and asymptomatic uninfected women determined via microarray, we discovered important pathogenic mechanisms in PID and response differences that provide a pathway to biomarker discovery. Women with N. gonorrhoeae- and/or C. trachomatis-induced PID exhibit overexpression of myeloid cell genes and suppression of protein synthesis, mitochondrial oxidative phosphorylation, and T cell-specific genes. Coinfected women exhibited the greatest activation of cell death pathways and suppression of responses essential for adaptive immunity. Women solely infected with C. trachomatis expressed elevated levels of type I and type II IFN genes, and enhanced type I IFN-induced chemokines in cervical secretions were associated with ascension of C. trachomatis to the endometrium. Blood microarrays reveal discrete pathobiological endotypes in women with PID that are driven by pathogen invasion of the upper genital tract.


Sujet(s)
Infections à Chlamydia/immunologie , Gonorrhée/immunologie , Maladie inflammatoire pelvienne/sang , Maladie inflammatoire pelvienne/étiologie , Maladie inflammatoire pelvienne/immunologie , Immunité acquise/immunologie , Adolescent , Adulte , Infections à Chlamydia/complications , Chlamydia trachomatis/immunologie , Co-infection , Femelle , Gonorrhée/complications , Humains , Neisseria gonorrhoeae/immunologie , Jeune adulte
11.
Immunol Cell Biol ; 96(3): 246-256, 2018 03.
Article de Anglais | MEDLINE | ID: mdl-29363185

RÉSUMÉ

Chlamydia muridarum and Chlamydia caviae have equivalent growth rates in mouse epithelial cells but only C. muridarum replicates inside mouse macrophages, while C. caviae does not. Macrophages infected with C. muridarum or C. caviae were used to address the hypothesis that the early signaling pathways initiated during infection depend on the fate of chlamydiae in the host cell. Transmission electron microscopy of C. muridarum-infected macrophages showed intact chlamydial elementary bodies and reticulate bodies 2 h postinfection in compact vacuoles. Conversely, in macrophages infected with C. caviae, chlamydiae were observed in large phagocytic vacuoles. Furthermore, C. caviae infections failed to develop into inclusions or produce viable bacteria. Expression of proinflammatory cytokines TNFα, IL-1ß and MMP13 was similar in C. caviae- or C. muridarum-infected macrophages at 3 h postinfection, indicating that chlamydial survival is not required for initiation of these responses. IL-1ß secretion, dependent on inflammasome activation, occurred in C. caviae-infected macrophages despite no chlamydial growth. Conversely, IFNß mRNA was observed only in C. muridarum- but not in C. caviae-infected macrophages. These data demonstrate that differential signaling events are initiated during a productive versus nonproductive chlamydial infection in a macrophage.


Sujet(s)
Infections à Chlamydia/métabolisme , Infections à Chlamydia/microbiologie , Chlamydia/physiologie , Espace intracellulaire/microbiologie , Macrophages/métabolisme , Macrophages/microbiologie , Transduction du signal , Animaux , Lignée cellulaire , Chlamydia/croissance et développement , Chlamydia/ultrastructure , Infections à Chlamydia/génétique , Infections à Chlamydia/anatomopathologie , Endosomes/métabolisme , Endosomes/ultrastructure , Régulation de l'expression des gènes , Inflammation/génétique , Interleukine-1 bêta , Macrophages/anatomopathologie , Macrophages/ultrastructure , Souris de lignée C57BL , Facteur de transcription NF-kappa B/métabolisme , ARN messager/génétique , ARN messager/métabolisme
12.
J Immunol ; 199(8): 2845-2854, 2017 10 15.
Article de Anglais | MEDLINE | ID: mdl-28855311

RÉSUMÉ

Chlamydia is responsible for millions of new infections annually, and current efforts focus on understanding cellular immunity for targeted vaccine development. The Chlamydia-specific CD4 T cell response is characterized by the production of IFN-γ, and polyfunctional Th1 responses are associated with enhanced protection. A major limitation in studying these responses is the paucity of tools available for detection, quantification, and characterization of polyfunctional Ag-specific T cells. We addressed this problem by developing a TCR-transgenic (Tg) mouse with CD4 T cells that respond to a common Ag in Chlamydia muridarum and Chlamydia trachomatis Using an adoptive-transfer approach, we show that naive Tg CD4 T cells become activated, proliferate, migrate to the infected tissue, and acquire a polyfunctional Th1 phenotype in infected mice. Polyfunctional Tg Th1 effectors demonstrated enhanced IFN-γ production compared with polyclonal cells, protected immune-deficient mice against lethality, mediated bacterial clearance, and orchestrated an anamnestic response. Adoptive transfer of Chlamydia-specific CD4 TCR-Tg T cells with polyfunctional capacity offers a powerful approach for analysis of protective effector and memory responses against chlamydial infection and demonstrates that an effective monoclonal CD4 T cell response may successfully guide subunit vaccination strategies.


Sujet(s)
Vaccins antibactériens/immunologie , Infections à Chlamydia/immunologie , Chlamydia muridarum/immunologie , Chlamydia trachomatis/immunologie , Lymphocytes auxiliaires Th1/immunologie , Animaux , Antigènes bactériens/immunologie , Charge bactérienne , Mouvement cellulaire , Prolifération cellulaire , Cellules cultivées , Réactions croisées , Humains , Mémoire immunologique , Interféron gamma/métabolisme , Activation des lymphocytes , Souris , Souris de lignée C57BL , Souris transgéniques , Récepteur lymphocytaire T antigène, alpha-bêta/génétique , Lymphocytes auxiliaires Th1/microbiologie
13.
J Leukoc Biol ; 101(6): 1373-1381, 2017 06.
Article de Anglais | MEDLINE | ID: mdl-28258153

RÉSUMÉ

Pulmonary tuberculosis (TB) caused by Mycobacterium tuberculosis (Mtb) is a leading cause of global morbidity and mortality. The only licensed TB vaccine, Mycobacterium bovis bacillus Calmette-Guerin (BCG), has variable efficacy in protecting against pulmonary TB. Thus, the development of more effective TB vaccines is critical to control the TB epidemic. Specifically, vaccines delivered through the mucosal route are known to induce Th17 responses and provide superior protection against Mtb infection. However, already tested Th17-inducing mucosal adjuvants, such as heat-labile enterotoxins and cholera toxins, are not considered safe for use in humans. In the current study, we rationally screened adjuvants for their ability to induce Th17-polarizing cytokines in dendritic cells (DCs) and determined whether they could be used in a protective mucosal TB vaccine. Our new studies show that monophosphoryl lipid A (MPL), when used in combination with chitosan, potently induces Th17-polarizing cytokines in DCs and downstream Th17/Th1 mucosal responses and confers significant protection in mice challenged with a clinical Mtb strain. Additionally, we show that both TLRs and the inflammasome pathways are activated in DCs by MPL-chitosan to mediate induction of Th17-polarizing cytokines. Together, our studies put forward the potential of a new, protective mucosal TB vaccine candidate, which incorporates safe adjuvants already approved for use in humans.


Sujet(s)
Vaccin BCG/usage thérapeutique , Muqueuse/immunologie , Mycobacterium tuberculosis/immunologie , Cellules Th17/immunologie , Tuberculose pulmonaire/prévention et contrôle , Administration par voie nasale , Animaux , Cytokines/métabolisme , Cellules dendritiques/effets des médicaments et des substances chimiques , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Souris , Souris de lignée C57BL , Souris knockout , Muqueuse/effets des médicaments et des substances chimiques , Muqueuse/métabolisme , Cellules Th17/effets des médicaments et des substances chimiques , Cellules Th17/métabolisme , Tuberculose pulmonaire/immunologie , Tuberculose pulmonaire/microbiologie
14.
Infect Immun ; 83(12): 4740-9, 2015 Dec.
Article de Anglais | MEDLINE | ID: mdl-26416908

RÉSUMÉ

Interferon (IFN)-inducible guanylate binding proteins (GBPs) mediate cell-autonomous host resistance to bacterial pathogens and promote inflammasome activation. The prevailing model postulates that these two GBP-controlled activities are directly linked through GBP-dependent vacuolar lysis. It was proposed that the rupture of pathogen-containing vacuoles (PVs) by GBPs destroyed the microbial refuge and simultaneously contaminated the host cell cytosol with microbial activators of inflammasomes. Here, we demonstrate that GBP-mediated host resistance and GBP-mediated inflammatory responses can be uncoupled. We show that PVs formed by the rodent pathogen Chlamydia muridarum, so-called inclusions, remain free of GBPs and that C. muridarum is impervious to GBP-mediated restrictions on bacterial growth. Although GBPs neither bind to C. muridarum inclusions nor restrict C. muridarum growth, we find that GBPs promote inflammasome activation in C. muridarum-infected macrophages. We demonstrate that C. muridarum infections induce GBP-dependent pyroptosis through both caspase-11-dependent noncanonical and caspase-1-dependent canonical inflammasomes. Among canonical inflammasomes, we find that C. muridarum and the human pathogen Chlamydia trachomatis activate not only NLRP3 but also AIM2. Our data show that GBPs support fast-kinetics processing and secretion of interleukin-1ß (IL-1ß) and IL-18 by the NLRP3 inflammasome but are dispensable for the secretion of the same cytokines at later times postinfection. Because IFN-γ fails to induce IL-1ß transcription, GBP-dependent fast-kinetics inflammasome activation can drive the preferential processing of constitutively expressed IL-18 in IFN-γ-primed macrophages in the absence of prior Toll-like receptor stimulation. Together, our results reveal that GBPs control the kinetics of inflammasome activation and thereby shape macrophage responses to Chlamydia infections.


Sujet(s)
Infections à Chlamydia/immunologie , Chlamydia muridarum/immunologie , Protéines G/immunologie , Inflammasomes/immunologie , Macrophages/immunologie , Animaux , Protéines de transport/génétique , Protéines de transport/immunologie , Caspases/génétique , Caspases/immunologie , Caspases initiatrices , Infections à Chlamydia/génétique , Infections à Chlamydia/microbiologie , Infections à Chlamydia/anatomopathologie , Chlamydia muridarum/génétique , Chlamydia muridarum/pathogénicité , Chlamydia trachomatis/génétique , Chlamydia trachomatis/immunologie , Chlamydia trachomatis/pathogénicité , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/immunologie , Fibroblastes/immunologie , Fibroblastes/microbiologie , Protéines G/génétique , Régulation de l'expression des gènes , Interactions hôte-pathogène , Corps d'inclusion/immunologie , Corps d'inclusion/microbiologie , Inflammasomes/génétique , Interféron gamma/génétique , Interféron gamma/immunologie , Interleukine-18/génétique , Interleukine-18/immunologie , Interleukine-1 bêta/génétique , Interleukine-1 bêta/immunologie , Macrophages/microbiologie , Souris , Souris de lignée C57BL , Souris knockout , Protéine-3 de la famille des NLR contenant un domaine pyrine , Culture de cellules primaires , Transduction du signal , Vacuoles/immunologie , Vacuoles/microbiologie
15.
J Immunol ; 193(5): 2394-404, 2014 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-25070851

RÉSUMÉ

IFN-ß has been implicated as an effector of oviduct pathology resulting from genital chlamydial infection in the mouse model. In this study, we investigated the role of cytosolic DNA and engagement of DNA sensors in IFN-ß expression during chlamydial infection. We determined that three-prime repair exonuclease-1, a host 3' to 5' exonuclease, reduced IFN-ß expression significantly during chlamydial infection using small interfering RNA and gene knockout fibroblasts, implicating cytosolic DNA as a ligand for this response. The DNA sensor cyclic GMP-AMP synthase (cGAS) has been shown to bind cytosolic DNA to generate cyclic GMP-AMP, which binds to the signaling adaptor stimulator of IFN genes (STING) to induce IFN-ß expression. We determined that cGAS is required for IFN-ß expression during chlamydial infection in multiple cell types. Interestingly, although infected cells deficient for STING or cGAS alone failed to induce IFN-ß, coculture of cells depleted for either STING or cGAS rescued IFN-ß expression. These data demonstrate that cyclic GMP-AMP produced in infected cGAS(+)STING(-) cells can migrate into adjacent cells via gap junctions to function in trans in cGAS(-)STING(+) cells. Furthermore, we observed cGAS localized in punctate regions on the cytosolic side of the chlamydial inclusion membrane in association with STING, indicating that chlamydial DNA is most likely recognized outside the inclusion as infection progresses. These novel findings provide evidence that cGAS-mediated DNA sensing directs IFN-ß expression during Chlamydia trachomatis infection and suggest that effectors from infected cells can directly upregulate IFN-ß expression in adjacent uninfected cells during in vivo infection, contributing to pathogenesis.


Sujet(s)
Infections à Chlamydia/immunologie , Chlamydia trachomatis/immunologie , ADN bactérien/immunologie , Interféron bêta/immunologie , Nucleotidyltransferases/immunologie , Animaux , Infections à Chlamydia/génétique , Infections à Chlamydia/anatomopathologie , Chlamydia trachomatis/génétique , Cytosol/immunologie , ADN bactérien/génétique , Jonctions communicantes/génétique , Jonctions communicantes/immunologie , Régulation de l'expression des gènes/génétique , Régulation de l'expression des gènes/immunologie , Techniques de knock-down de gènes , Cellules HeLa , Humains , Interféron bêta/génétique , Protéines membranaires/génétique , Protéines membranaires/immunologie , Souris , Nucléotides cycliques/génétique , Nucléotides cycliques/immunologie , Nucleotidyltransferases/génétique
16.
Cytokine ; 65(2): 202-9, 2014 Feb.
Article de Anglais | MEDLINE | ID: mdl-24301038

RÉSUMÉ

OBJECTIVE: Our group and others have shown that serial intra-lesional injections of common warts with skin testing reagents such as Candida, mumps and Trichophyton are effective in regressing injected and non-injected warts. Anti-HPV T-cell responses appear to be induced. The goal of this study was to understand the mechanisms of how Candida skin testing reagent enhances immune responses. METHODS: The following immunological features were studied to understand how Candida induces immune responses in healthy subjects: (1) proliferative capacity of T-cells upon exposure to Candida through monocyte-derived human Langerhans cells (LCs) measured using alamarBlue, (2) cytokine (IL-1ß, IL-6, IL-8, IL-10, IL-12p40, IL-23Ap19, IFN-γ, and TNF- expression upon Candida stimulation of LCs by quantitative reverse transcription (qRT)-PCR and cytokine secretion by ELISA, (3) expression of pattern recognition receptors (PRRs) known to associate with Candida albicans (DC-SIGN, dectin-1, dectin-2, galectin-3, mincle, mannose receptor, Toll-like receptors 1, 2, 4, 6, and 9) on LCs by qRT-PCR, (4) role of dectin-1 in IL-12 production by antibody blocking, and (5) induction of Th1, Th2, and/or Th17 responses by intracellular cytokine staining of CD4 cells exposed to Candida pulsed LCs for IFN-γ, IL-4, and IL-17A. RESULTS: T-cell proliferation upon stimulation with Candida-pulsed LCs was significantly higher compared to proliferation in the absence of Candida (p=0.004). The most frequently expressed cytokine in stimulated LCs was IL-12p40 mRNA, and IL-12p40 and IL-12p70 were also detected at protein levels. All other cytokine mRNAs examined were detected in the following order of decreasing frequency: IL23Ap19, IFN-γ, IL-1ß, IL-6, IL-8, and IL-10. LCs expressed all PRRs examined. Anti-dectin-1 inhibited IL-12p40 mRNA production upon Candida stimulation of LCs from some healthy subjects. IFN-γ secretion was increased and IL-4 secretion was decreased in CD4 cells of a few healthy subjects, but IL-17A was essentially unchanged upon Candida treatment. CONCLUSIONS: Proliferation of T-cells in a substantial majority of healthy subjects can be demonstrated with Candida stimulation. We show Th1 promotion and dectin-1 stimulation of LCs as potential mechanisms in some healthy subjects.


Sujet(s)
Candida/composition chimique , Santé , Interleukine-12/métabolisme , Cellules de Langerhans/métabolisme , Lectines de type C/métabolisme , Antigènes CD/métabolisme , Antigènes CD1/métabolisme , Cadhérines/métabolisme , Candida/immunologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Volontaires sains , Humains , Indicateurs et réactifs/pharmacologie , Espace intracellulaire/effets des médicaments et des substances chimiques , Espace intracellulaire/métabolisme , Cellules de Langerhans/effets des médicaments et des substances chimiques , Cellules de Langerhans/immunologie , Lectines de type C/immunologie , Lectines liant le mannose/métabolisme , Récepteurs de reconnaissance de motifs moléculaires/métabolisme , RT-PCR , Tests cutanés , Lymphocytes T/cytologie , Lymphocytes T/effets des médicaments et des substances chimiques
17.
J Immunol ; 191(8): 4269-79, 2013 Oct 15.
Article de Anglais | MEDLINE | ID: mdl-24038087

RÉSUMÉ

Resolution of Chlamydia genital tract infection is delayed in the absence of MyD88. In these studies, we first used bone marrow chimeras to demonstrate a requirement for MyD88 expression by hematopoietic cells in the presence of a wild-type epithelium. Using mixed bone marrow chimeras we then determined that MyD88 expression was specifically required in the adaptive immune compartment. Furthermore, adoptive transfer experiments revealed that CD4(+) T cell expression of MyD88 was necessary for normal resolution of genital tract infection. This requirement was associated with a reduced ability of MyD88(-/-)CD4(+) T cells to accumulate in the draining lymph nodes and genital tract when exposed to the same inflammatory milieu as wild-type CD4(+) T cells. We also demonstrated that the impaired infection control we observed in the absence of MyD88 could not be recapitulated by deficiencies in TLR or IL-1R signaling. In vitro, we detected an increased frequency of apoptotic MyD88(-/-)CD4(+) T cells upon activation in the absence of exogenous ligands for receptors upstream of MyD88. These data reveal an intrinsic requirement for MyD88 in CD4(+) T cells during Chlamydia infection and indicate that the importance of MyD88 extends beyond innate immune responses by directly influencing adaptive immunity.


Sujet(s)
Lymphocytes T CD4+/immunologie , Infections à Chlamydia/immunologie , Chlamydia muridarum/immunologie , Facteur de différenciation myéloïde-88/métabolisme , Infections de l'appareil reproducteur/immunologie , Transfert adoptif , Animaux , Moelle osseuse/immunologie , Lymphocytes T CD4+/métabolisme , Infections à Chlamydia/microbiologie , Femelle , Système génital de la femme/cytologie , Système génital de la femme/immunologie , Système génital de la femme/microbiologie , Noeuds lymphatiques/cytologie , Noeuds lymphatiques/immunologie , Souris , Souris de lignée C57BL , Souris knockout , Facteur de différenciation myéloïde-88/biosynthèse , Facteur de différenciation myéloïde-88/génétique , Récepteurs à l'interleukine-1/métabolisme , Infections de l'appareil reproducteur/microbiologie
18.
J Immunol ; 188(6): 2866-75, 2012 Mar 15.
Article de Anglais | MEDLINE | ID: mdl-22331066

RÉSUMÉ

IL-1ß has been implicated in the development of oviduct pathology during Chlamydia muridarum genital infection in the mouse model. The goal of this study was to characterize the role of IL-1 signaling and the inflammasome-activation pathways during genital chlamydial infection. Compared with control mice, IL-1R-deficient mice displayed delayed clearance and increased chlamydial colonization. Consistent with the role for IL-1 signaling in infection clearance, mice deficient for the IL-1R antagonist cleared infection at a faster rate. Despite increased infection, IL-1R-deficient mice had significantly reduced oviduct pathology, which was associated with decreased numbers of neutrophils, but more macrophages, in the genital tract. IL-1ß secretion is dependent on caspase-1 and apoptosis-associated speck-like protein containing caspase recruitment domain (ASC) inflammasome during in vitro infection of primed macrophages with C. muridarum. To investigate the role of inflammasome components during in vivo genital infection, mice lacking NLRP3, NLRC4, and ASC were tested and found to display no reduction in oviduct pathology compared with control mice. Mice deficient for ASC displayed a prolonged course of infection, which was associated with reduced T cell recruitment and proliferation. Further, ASC-deficient mice displayed normal levels of IL-1ß in genital secretions. However, a significant decrease in caspase-1-dependent IL-18 was observed in both ASC- and NLRP3-deficient mice. These data demonstrate a major role for IL-1 signaling, but a limited role for the inflammasome pathway, in IL-1ß secretion and development of oviduct pathology during genital chlamydial infection. The data also suggest an IL-1-independent role for ASC in adaptive immunity during genital chlamydial infection.


Sujet(s)
Infections à Chlamydia/immunologie , Infections à Chlamydia/anatomopathologie , Interleukine-1/immunologie , Oviductes/anatomopathologie , Transduction du signal/immunologie , Animaux , Protéines régulatrices de l'apoptose , Protéines adaptatrices de signalisation CARD , Caspase-1/immunologie , Caspase-1/métabolisme , Séparation cellulaire , Infections à Chlamydia/métabolisme , Chlamydia muridarum/immunologie , Protéines du cytosquelette/immunologie , Protéines du cytosquelette/métabolisme , Test ELISA , Femelle , Cytométrie en flux , Inflammasomes/immunologie , Inflammasomes/métabolisme , Interleukine-1/métabolisme , Macrophages/immunologie , Souris , Souris knockout , Récepteurs à l'interleukine-1/immunologie , Récepteurs à l'interleukine-1/métabolisme , Lymphocytes T/immunologie
19.
Infect Immun ; 79(10): 3922-33, 2011 Oct.
Article de Anglais | MEDLINE | ID: mdl-21788382

RÉSUMÉ

Mice with the type I interferon (IFN) receptor gene knocked out (IFNAR KO mice) or deficient for alpha/beta IFN (IFN-α/ß) signaling clear chlamydial infection earlier than control mice and develop less oviduct pathology. Initiation of host IFN-ß transcription during an in vitro chlamydial infection requires interferon regulatory transcription factor 3 (IRF3). The goal of the present study was to characterize the influence of IRF3 on chlamydial genital infection and its relationship to IFN-ß expression in the mouse model. IRF3 KO mice were able to resolve infection as well as control mice, overcoming increased chlamydial colonization and tissue burden early during infection. As previously observed for IFNAR KO mice, IRF3 KO mice generated a potent antigen-specific T cell response. However, in contrast to IFNAR KO mice, IRF3 KO mice exhibited unusually severe dilatation and pathology in the uterine horns but normal oviduct pathology after infection. Although IFN-ß expression in vivo was dependent on the presence of IRF3 early in infection (before day 4), the IFN-independent function of IRF3 was likely driving this phenotype. Specifically, early during infection, the number of apoptotic cells and the number of inflammatory cells were significantly less in uterine horns from IRF3 KO mice than in those from control mice, despite an increased chlamydial burden. To delineate the effects of IFN-ß versus IRF3, neutralizing IFN-ß antibody was administered to wild-type (WT) mice during chlamydial infection. IFN-ß depletion in WT mice mimicked that in IFNΑR KO mice but not that in IRF3 KO mice with respect to both chlamydial clearance and reduced oviduct pathology. These data suggest that IRF3 has a role in protection from uterine horn pathology that is independent of its function in IFN-ß expression.


Sujet(s)
Infections à Chlamydia/immunologie , Chlamydia muridarum/immunologie , Maladies de l'appareil génital féminin/immunologie , Facteur-3 de régulation d'interféron/immunologie , Utérus/anatomopathologie , Animaux , Infections à Chlamydia/microbiologie , Chlamydia muridarum/pathogénicité , Cytokines/métabolisme , Modèles animaux de maladie humaine , Femelle , Maladies de l'appareil génital féminin/microbiologie , Facteur-3 de régulation d'interféron/génétique , Facteur-3 de régulation d'interféron/métabolisme , Interféron bêta/génétique , Interféron bêta/métabolisme , Activation des lymphocytes , Souris , Souris de lignée C57BL , Souris knockout , Transduction du signal , Lymphocytes T/immunologie , Utérus/microbiologie
20.
Infect Immun ; 79(5): 1889-97, 2011 May.
Article de Anglais | MEDLINE | ID: mdl-21402767

RÉSUMÉ

Trachoma, the world's leading cause of preventable blindness, is produced by chronic ocular infection with Chlamydia trachomatis, an obligate intracellular bacterium. While many studies have focused on immune mechanisms for trachoma during chronic stages of infection, less research has targeted immune mechanisms in primary ocular infections, events that could impact chronic responses. The goal of this study was to investigate the function of neutrophils during primary chlamydial ocular infection by using the guinea pig model of Chlamydia caviae inclusion conjunctivitis. We hypothesized that neutrophils help modulate the adaptive response and promote host tissue damage. To test these hypotheses, guinea pigs with primary C. caviae ocular infections were depleted of neutrophils by using rabbit antineutrophil antiserum, and immune responses and immunopathology were evaluated during the first 7 days of infection. Results showed that neutrophil depletion dramatically decreased ocular pathology, both clinically and histologically. The adaptive response was also altered, with increased C. caviae-specific IgA titers in tears and serum and decreased numbers of CD4(+) and CD8(+) T cells in infected conjunctivae. Additionally, there were changes in conjunctival chemokines and cytokines, such as increased expression of IgA-promoting interleukin-5 and anti-inflammatory transforming growth factor ß, along with decreased expression of T cell-recruiting CCL5 (RANTES). This study, the first to investigate the role of neutrophils in primary chlamydial ocular infection, indicates a previously unappreciated role for neutrophils in modulating the adaptive response and suggests a prominent role for neutrophils in chlamydia-associated ocular pathology.


Sujet(s)
Immunité acquise/immunologie , Infections à Chlamydia/immunologie , Infections bactériennes de l'oeil/immunologie , Granulocytes neutrophiles/immunologie , Animaux , Séparation cellulaire , Infections à Chlamydia/anatomopathologie , Modèles animaux de maladie humaine , Test ELISA , Infections bactériennes de l'oeil/anatomopathologie , Femelle , Cytométrie en flux , Cochons d'Inde , Lapins , RT-PCR , Trachome/immunologie , Trachome/anatomopathologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...