Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Infect Immun ; 91(2): e0039222, 2023 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-36722979

RESUMEN

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.


Asunto(s)
Infecciones por Chlamydia , Chlamydia trachomatis , Animales , Femenino , Ratones , Antígenos Bacterianos/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Infecciones por Chlamydia/microbiología , Chlamydia trachomatis/genética , Chlamydia trachomatis/patogenicidad , Plásmidos/genética , Virulencia/genética
2.
Caries Res ; 56(1): 36-46, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34749377

RESUMEN

Identification of the association between Early Childhood Caries (ECC) and Iron Deficiency Anaemia (IDA) will aid paediatricians and paediatric dentists to enhance health promotion measures to reduce the related morbidity in children. This systematic review aims to determine an evidence-based association between ECC and IDA. A systematic search was carried out from MEDLINE via PubMed, EMBASE, LILACS, Cochrane Oral Health Group's Specialized Register, CINAHL via EBSCO, Web of Science, and Scopus up to May 2020. Hand searching and grey literature screening were also conducted. Cross-sectional, case-control, and cohort studies in English language which assessed the association was included. Two reviewers independently assessed the study quality and extracted the outcome data. A total of 1,434 studies were identified. Fourteen studies qualified for qualitative review and 7 of them for a meta-analysis. In comparison with children not affected by ECC, those affected had an increased likelihood of IDA (OR = 6.07 [3.61, 10.21]). The meta-analysis showed no statistical difference when comparing blood parameters (Hb, MCV, and serum ferritin) in children with and without ECC. This systematic review demonstrates an association between ECC and increased odds of IDA rather than it being the cause for IDA. Further longitudinal studies with robust methodology are required to determine an evidence-based association.


Asunto(s)
Anemia Ferropénica , Caries Dental , Deficiencias de Hierro , Anemia Ferropénica/complicaciones , Anemia Ferropénica/prevención & control , Niño , Preescolar , Estudios Transversales , Caries Dental/epidemiología , Caries Dental/etiología , Susceptibilidad a Caries Dentarias , Humanos
3.
Pathogens ; 10(10)2021 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-34684219

RESUMEN

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.

4.
J Infect Dis ; 224(12 Suppl 2): S47-S55, 2021 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-34396406

RESUMEN

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.


Asunto(s)
Muerte Celular , Infecciones por Chlamydia , Chlamydia muridarum/inmunología , Interleucina-1alfa , Factor de Necrosis Tumoral alfa , Animales , Infecciones por Chlamydia/inmunología , Infecciones por Chlamydia/metabolismo , Células Epiteliales , Femenino , Interleucina-1alfa/inmunología , Interleucina-1alfa/metabolismo , Ratones , Ratones Endogámicos C57BL , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
5.
Infect Immun ; 89(10): e0007221, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34125599

RESUMEN

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.


Asunto(s)
Infecciones por Chlamydia/inmunología , Chlamydia muridarum/inmunología , Receptores Inmunológicos/inmunología , Receptor Activador Expresado en Células Mieloides 1/inmunología , Útero/inmunología , Inmunidad Adaptativa/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/microbiología , Movimiento Celular/inmunología , Infecciones por Chlamydia/metabolismo , Infecciones por Chlamydia/microbiología , Chlamydia trachomatis/inmunología , Modelos Animales de Enfermedad , Epitelio/inmunología , Epitelio/metabolismo , Epitelio/microbiología , Femenino , Genitales Femeninos/inmunología , Genitales Femeninos/metabolismo , Genitales Femeninos/microbiología , Ratones , Ratones Endogámicos C57BL , Neutrófilos/inmunología , Neutrófilos/metabolismo , Neutrófilos/microbiología , Oviductos/inmunología , Oviductos/metabolismo , Oviductos/microbiología , Receptores Inmunológicos/metabolismo , Infecciones del Sistema Genital/inmunología , Infecciones del Sistema Genital/metabolismo , Infecciones del Sistema Genital/microbiología , Receptor Activador Expresado en Células Mieloides 1/metabolismo , Útero/metabolismo , Útero/microbiología
6.
J Immunol ; 205(11): 3037-3049, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33087404

RESUMEN

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.


Asunto(s)
Infecciones por Chlamydia/metabolismo , Genitales Femeninos/metabolismo , Interleucina-1alfa/metabolismo , Oviductos/metabolismo , Animales , Linfocitos T CD4-Positivos/metabolismo , Cuello del Útero/metabolismo , Cuello del Útero/microbiología , Infecciones por Chlamydia/microbiología , Chlamydia muridarum/patogenicidad , Modelos Animales de Enfermedad , Femenino , Genitales Femeninos/microbiología , Interleucina-1beta/metabolismo , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila/fisiología , Oviductos/microbiología , Infecciones del Sistema Genital/metabolismo , Infecciones del Sistema Genital/microbiología
7.
Infect Immun ; 88(9)2020 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-32601108

RESUMEN

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.


Asunto(s)
Células Epiteliales/inmunología , Regulación de la Expresión Génica/inmunología , Interacciones Microbiota-Huesped/inmunología , Linfocitos T/inmunología , Adulto , Sistema de Transporte de Aminoácidos ASC/genética , Sistema de Transporte de Aminoácidos ASC/inmunología , Antígenos CD/genética , Antígenos CD/inmunología , Biomarcadores/metabolismo , Quimiocina CCL5/genética , Quimiocina CCL5/inmunología , Quimiocina CXCL10/genética , Quimiocina CXCL10/inmunología , Quimiocina CXCL11/genética , Quimiocina CXCL11/inmunología , Infecciones por Chlamydia/genética , Infecciones por Chlamydia/inmunología , Infecciones por Chlamydia/microbiología , Chlamydia trachomatis/crecimiento & desarrollo , Chlamydia trachomatis/inmunología , Células Epiteliales/microbiología , Trompas Uterinas/citología , Trompas Uterinas/cirugía , Femenino , Cadena Pesada de la Proteína-1 Reguladora de Fusión/genética , Cadena Pesada de la Proteína-1 Reguladora de Fusión/inmunología , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Interacciones Microbiota-Huesped/genética , Humanos , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/inmunología , Antígenos de Histocompatibilidad Menor/genética , Antígenos de Histocompatibilidad Menor/inmunología , Modelos Biológicos , Cultivo Primario de Células , Receptores de Interferón/genética , Receptores de Interferón/inmunología , Receptores de Transferrina/genética , Receptores de Transferrina/inmunología , Salpingectomía , Linfocitos T/microbiología , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/inmunología , Receptor de Interferón gamma
8.
Atherosclerosis ; 290: 103-110, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31604170

RESUMEN

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.


Asunto(s)
Enfermedades de la Aorta/etiología , Aterosclerosis/etiología , Infecciones por Chlamydia/complicaciones , Chlamydia muridarum/patogenicidad , Hiperlipidemias/complicaciones , Infecciones del Sistema Genital/complicaciones , Útero/microbiología , Animales , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/microbiología , Enfermedades de la Aorta/patología , Aterosclerosis/metabolismo , Aterosclerosis/microbiología , Aterosclerosis/patología , Células Cultivadas , Infecciones por Chlamydia/microbiología , Infecciones por Chlamydia/patología , Citocinas/sangre , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Hiperlipidemias/metabolismo , Mediadores de Inflamación/sangre , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones Noqueados para ApoE , Placa Aterosclerótica , Receptores de LDL/deficiencia , Receptores de LDL/genética , Infecciones del Sistema Genital/microbiología , Infecciones del Sistema Genital/patología , Factores de Tiempo , Útero/patología
9.
Infect Immun ; 87(8)2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31160363

RESUMEN

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.


Asunto(s)
Caspasas/fisiología , Infecciones por Chlamydia/patología , Oviductos/patología , Infecciones del Sistema Genital/patología , Animales , Caspasa 1/fisiología , Caspasas/genética , Caspasas Iniciadoras , Femenino , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila
10.
Infect Immun ; 86(7)2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29661927

RESUMEN

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.


Asunto(s)
Linfocitos B/inmunología , Infecciones por Chlamydia/inmunología , Chlamydia muridarum , Interferón gamma/inmunología , Infecciones del Sistema Genital/inmunología , Linfocitos T/fisiología , Animales , Infecciones por Chlamydia/mortalidad , Chlamydia muridarum/genética , Femenino , Proteínas de Homeodominio/fisiología , Ratones , Ratones Endogámicos C57BL , Plásmidos , Infecciones del Sistema Genital/mortalidad
11.
J Immunol ; 200(8): 2941-2956, 2018 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-29531169

RESUMEN

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.


Asunto(s)
Infecciones por Chlamydia/inmunología , Gonorrea/inmunología , Enfermedad Inflamatoria Pélvica/sangre , Enfermedad Inflamatoria Pélvica/etiología , Enfermedad Inflamatoria Pélvica/inmunología , Inmunidad Adaptativa/inmunología , Adolescente , Adulto , Infecciones por Chlamydia/complicaciones , Chlamydia trachomatis/inmunología , Coinfección , Femenino , Gonorrea/complicaciones , Humanos , Neisseria gonorrhoeae/inmunología , Adulto Joven
12.
Immunol Cell Biol ; 96(3): 246-256, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29363185

RESUMEN

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.


Asunto(s)
Infecciones por Chlamydia/metabolismo , Infecciones por Chlamydia/microbiología , Chlamydia/fisiología , Espacio Intracelular/microbiología , Macrófagos/metabolismo , Macrófagos/microbiología , Transducción de Señal , Animales , Línea Celular , Chlamydia/crecimiento & desarrollo , Chlamydia/ultraestructura , Infecciones por Chlamydia/genética , Infecciones por Chlamydia/patología , Endosomas/metabolismo , Endosomas/ultraestructura , Regulación de la Expresión Génica , Inflamación/genética , Interleucina-1beta , Macrófagos/patología , Macrófagos/ultraestructura , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
13.
J Immunol ; 199(8): 2845-2854, 2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28855311

RESUMEN

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.


Asunto(s)
Vacunas Bacterianas/inmunología , Infecciones por Chlamydia/inmunología , Chlamydia muridarum/inmunología , Chlamydia trachomatis/inmunología , Células TH1/inmunología , Animales , Antígenos Bacterianos/inmunología , Carga Bacteriana , Movimiento Celular , Proliferación Celular , Células Cultivadas , Reacciones Cruzadas , Humanos , Memoria Inmunológica , Interferón gamma/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Células TH1/microbiología
14.
J Leukoc Biol ; 101(6): 1373-1381, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28258153

RESUMEN

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.


Asunto(s)
Vacuna BCG/uso terapéutico , Membrana Mucosa/inmunología , Mycobacterium tuberculosis/inmunología , Células Th17/inmunología , Tuberculosis Pulmonar/prevención & control , Administración Intranasal , Animales , Citocinas/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Membrana Mucosa/efectos de los fármacos , Membrana Mucosa/metabolismo , Células Th17/efectos de los fármacos , Células Th17/metabolismo , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/microbiología
15.
Infect Immun ; 83(12): 4740-9, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26416908

RESUMEN

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.


Asunto(s)
Infecciones por Chlamydia/inmunología , Chlamydia muridarum/inmunología , Proteínas de Unión al GTP/inmunología , Inflamasomas/inmunología , Macrófagos/inmunología , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Caspasas/genética , Caspasas/inmunología , Caspasas Iniciadoras , Infecciones por Chlamydia/genética , Infecciones por Chlamydia/microbiología , Infecciones por Chlamydia/patología , Chlamydia muridarum/genética , Chlamydia muridarum/patogenicidad , Chlamydia trachomatis/genética , Chlamydia trachomatis/inmunología , Chlamydia trachomatis/patogenicidad , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Fibroblastos/inmunología , Fibroblastos/microbiología , Proteínas de Unión al GTP/genética , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno , Cuerpos de Inclusión/inmunología , Cuerpos de Inclusión/microbiología , Inflamasomas/genética , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-18/genética , Interleucina-18/inmunología , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Cultivo Primario de Células , Transducción de Señal , Vacuolas/inmunología , Vacuolas/microbiología
16.
J Immunol ; 193(5): 2394-404, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25070851

RESUMEN

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.


Asunto(s)
Infecciones por Chlamydia/inmunología , Chlamydia trachomatis/inmunología , ADN Bacteriano/inmunología , Interferón beta/inmunología , Nucleotidiltransferasas/inmunología , Animales , Infecciones por Chlamydia/genética , Infecciones por Chlamydia/patología , Chlamydia trachomatis/genética , Citosol/inmunología , ADN Bacteriano/genética , Uniones Comunicantes/genética , Uniones Comunicantes/inmunología , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Interferón beta/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Nucleótidos Cíclicos/genética , Nucleótidos Cíclicos/inmunología , Nucleotidiltransferasas/genética
17.
PLoS Pathog ; 10(5): e1004099, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24831696

RESUMEN

Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), infects one third of the world's population. Among these infections, clinical isolates belonging to the W-Beijing appear to be emerging, representing about 50% of Mtb isolates in East Asia, and about 13% of all Mtb isolates worldwide. In animal models, infection with W-Beijing strain, Mtb HN878, is considered "hypervirulent" as it results in increased mortality and causes exacerbated immunopathology in infected animals. We had previously shown the Interleukin (IL) -17 pathway is dispensable for primary immunity against infection with the lab adapted Mtb H37Rv strain. However, it is not known whether IL-17 has any role to play in protective immunity against infection with clinical Mtb isolates. We report here that lab adapted Mtb strains, such as H37Rv, or less virulent Mtb clinical isolates, such as Mtb CDC1551, do not require IL-17 for protective immunity against infection while infection with Mtb HN878 requires IL-17 for early protective immunity. Unexpectedly, Mtb HN878 induces robust production of IL-1ß through a TLR-2-dependent mechanism, which supports potent IL-17 responses. We also show that the role for IL-17 in mediating protective immunity against Mtb HN878 is through IL-17 Receptor signaling in non-hematopoietic cells, mediating the induction of the chemokine, CXCL-13, which is required for localization of T cells within lung lymphoid follicles. Correct T cell localization within lymphoid follicles in the lung is required for maximal macrophage activation and Mtb control. Since IL-17 has a critical role in vaccine-induced immunity against TB, our results have far reaching implications for the design of vaccines and therapies to prevent and treat emerging Mtb strains. In addition, our data changes the existing paradigm that IL-17 is dispensable for primary immunity against Mtb infection, and instead suggests a differential role for IL-17 in early protective immunity against emerging Mtb strains.


Asunto(s)
Inmunidad Innata/genética , Interleucina-17/fisiología , Mycobacterium tuberculosis/inmunología , Animales , Células Cultivadas , Enfermedades Transmisibles Emergentes/genética , Enfermedades Transmisibles Emergentes/inmunología , Citoprotección/genética , Citoprotección/inmunología , Femenino , Interleucina-17/genética , Interleucina-1beta/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mycobacterium tuberculosis/patogenicidad , Receptores Tipo I de Interleucina-1/genética , Receptor Toll-Like 2/fisiología , Tuberculosis/genética , Tuberculosis/inmunología
18.
Cytokine ; 65(2): 202-9, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24301038

RESUMEN

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.


Asunto(s)
Candida/química , Salud , Interleucina-12/metabolismo , Células de Langerhans/metabolismo , Lectinas Tipo C/metabolismo , Antígenos CD/metabolismo , Antígenos CD1/metabolismo , Cadherinas/metabolismo , Candida/inmunología , Proliferación Celular/efectos de los fármacos , Voluntarios Sanos , Humanos , Indicadores y Reactivos/farmacología , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Células de Langerhans/efectos de los fármacos , Células de Langerhans/inmunología , Lectinas Tipo C/inmunología , Lectinas de Unión a Manosa/metabolismo , Receptores de Reconocimiento de Patrones/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Pruebas Cutáneas , Linfocitos T/citología , Linfocitos T/efectos de los fármacos
19.
Vaccine ; 31(49): 5806-13, 2013 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-24135577

RESUMEN

A vaccine adjuvant that can effectively promote cell-mediated immunity is currently not available. Because of the ability of a Candida skin test reagent injection to induce common wart regression, our group is using it as a novel adjuvant in a clinical trial of a peptide-based human papillomavirus therapeutic vaccine. The goal of this current study was to investigate the mechanisms of how Candida enhances the vaccine immune responses. Maturation effects on Langerhans cells, capacity to proliferate T-cells, expression of cytokines and pattern recognition receptors by Langerhans cells, and ability to induce Th1, Th2, and Th17 responses were investigated in healthy subjects. The vaccine, human papillomavirus peptides with Candida, demonstrated partial maturation effects on Langerhans cells indicated by significantly up-regulated CD40 (p=0.00007) and CD80 (p<0.00001) levels, and showed T-cell proliferative capacity (p<0.00001) when presented by Langerhans cells in vitro. Interestingly, the maturation effects were due to the peptides while Candida was responsible for the T-cell proliferation. The cytokine profile (IL-1ß, IL-6, IL-8, IL-10, IL-12p40, IL-23Ap19, IFN-γ and TNF-α) of Langerhans cells treated with the vaccine or Candida alone showed that IL-12p40 mRNA was most frequently induced, and IL-12p70 protein was detected in the supernatants. The presence of pattern recognition receptors 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) were demonstrated in all subjects. On the other hand, the induction of Th1 response demonstrated by IFN-γ secretion by CD4 cells stimulated with the vaccine or Candida pulsed Langerhans cells was demonstrated only in one subject. In summary, the Langerhans cell maturation effects of the vaccine were due to the peptides while the T-cell proliferative capacity was derived from Candida, and the most frequently induced cytokine was IL-12.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Candida/inmunología , Inmunidad Celular , Vacunas contra Papillomavirus/inmunología , Linfocitos T CD4-Positivos/inmunología , Proliferación Celular , Células Cultivadas , Citocinas/inmunología , Humanos , Células de Langerhans/inmunología
20.
J Immunol ; 191(8): 4269-79, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24038087

RESUMEN

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.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Infecciones por Chlamydia/inmunología , Chlamydia muridarum/inmunología , Factor 88 de Diferenciación Mieloide/metabolismo , Infecciones del Sistema Genital/inmunología , Traslado Adoptivo , Animales , Médula Ósea/inmunología , Linfocitos T CD4-Positivos/metabolismo , Infecciones por Chlamydia/microbiología , Femenino , Genitales Femeninos/citología , Genitales Femeninos/inmunología , Genitales Femeninos/microbiología , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/biosíntesis , Factor 88 de Diferenciación Mieloide/genética , Receptores de Interleucina-1/metabolismo , Infecciones del Sistema Genital/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...