Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Microbiol Spectr ; 10(4): e0161722, 2022 08 31.
Article in English | MEDLINE | ID: mdl-35876584

ABSTRACT

The tumor necrosis factor (TNF)-related apoptosis-inducing ligand receptor (TRAIL-R) suppresses inflammation and could therefore affect the course of Chlamydia infections and their long-term sequelae. Wild-type (WT) and TRAIL-R-/- C57BL/6 mice were inoculated vaginally with Chlamydia muridarum; the course of the infection was followed with vaginal cultures and the presence of hydrosalpinx determined. To evaluate the role of TRAIL-R following a secondary infection, the mice were vaginally reinfected. WT and TRAIL-R-/- male mice were also infected and reinfected in the respiratory tract, and the course of the diseases and the infections were followed. Following the primary and secondary vaginal infection, no significant differences in vaginal shedding or hydrosalpinx formation were observed between the WT and TRAIL-R-/- mice. The WT and TRAIL-R-/- mice mounted antibody responses in serum and vaginal washes that were not significantly different. After the primary and secondary intranasal infections of the male mice, changes in body weight were determined, and no significant differences were observed between the WT and TRAIL-R-/- mice. Ten days after the primary and the secondary infections, the weight of the lungs and number of C. muridarum inclusion forming units (IFU) were determined. The lungs of the WT mice weighed less compared with the TRAIL-R-/- mice following a primary infection but not after a secondary infection. No differences in the number of C. muridarum IFU in the lungs were observed between the two groups of mice. In conclusion, despite playing a role in inflammation cell-signaling pathways in vitro, TRAIL-R does not appear to play a major role in the susceptibility, clinical outcomes, or long-term sequelae of C. muridarum infections in vivo. IMPORTANCE TNF-related apoptosis-inducing ligand receptor (TRAIL-R) is involved in suppressing inflammatory responses. Bacterial pathogens such as Chlamydia spp. elicit inflammatory responses in humans following genital, ocular, and respiratory infections. The inflammatory responses are important to control the spread of Chlamydia. However, in certain instances, these inflammatory responses can produce long-term sequelae, including fibrosis. Fibrosis, or scarring, in the genital tract, eye, and respiratory system results in functional deficiencies, including infertility, blindness, and chronic obstructive lung disease, respectively. The goal of this study was to determine if mice deficient in TRAIL-R infected in the genital and respiratory tracts with Chlamydia spp. suffer more or less severe infections, infertility, or lung diseases than wild-type mice. Our results show no differences between the immune responses, infection severity, and long-term sequelae between TRAIL-R knockout and wild-type animals following a genital or a respiratory infection with Chlamydia.


Subject(s)
Chlamydia Infections , Chlamydia muridarum , Coinfection , Infertility , Reproductive Tract Infections , Respiratory Tract Infections , Animals , Chlamydia muridarum/physiology , Female , Fibrosis , Humans , Infertility/pathology , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Reproductive Tract Infections/pathology , Vagina/microbiology
2.
Sci Rep ; 10(1): 21994, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33319815

ABSTRACT

IL-2Rα, in part, comprises the high affinity receptor for IL-2, a cytokine important in immune proliferation, activation, and regulation. IL-2Rα deficient mice (IL-2Rα-KO) develop systemic autoimmune disease and die from severe anemia between 18 and 80 days of age. These mice develop kinetically distinct autoimmune progression, with approximately a quarter dying by 21 days of age and half dying after 30 days. This research aims to define immune parameters and cytokine signaling that distinguish cohorts of IL-2Rα-KO mice that develop early- versus late-stage autoimmune disease. To investigate these differences, we evaluated complete blood counts (CBC), antibody binding of RBCs, T cell numbers and activation, hematopoietic progenitor changes, and signaling kinetics, during autoimmune hemolytic anemia (AIHA) and bone marrow failure. We identified several alterations that, when combined, correlate to disease kinetics. Early onset disease correlates with anti-RBC antibodies, lower hematocrit, and reduced IL-7 signaling. CD8 regulatory T cells (Tregs) have enhanced apoptosis in early disease. Further, early and late end stage disease, while largely similar, had several differences suggesting distinct mechanisms drive autoimmune disease kinetics. Therefore, IL-2Rα-KO disease pathology rates, driven by T cell signaling, promote effector T cell activation and expansion and Treg dysfunction.


Subject(s)
Interleukin-2 Receptor alpha Subunit/deficiency , Signal Transduction , T-Lymphocytes, Regulatory/immunology , Animals , Apoptosis , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Erythrocytes/metabolism , Immunologic Memory , Interleukin-2/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Kinetics , Lymphocyte Activation/immunology , Mice, Inbred BALB C , Mice, Knockout , Thymus Gland/growth & development
3.
PLoS One ; 15(3): e0230718, 2020.
Article in English | MEDLINE | ID: mdl-32210474

ABSTRACT

Chlamydia trachomatis is the most common bacterial sexually-transmitted infection and the major cause of preventable blindness worldwide. The asymptomatic nature of many infections along with uncontrolled inflammation leads to irreversible damage in the upper genital tract and the tarsal conjunctivae, with the major complications of infertility and chronic pelvic pain, and blindness, respectively. Inflammation must, therefore, be tightly regulated to avoid an unrestrained immune response. The genetic factors that regulate inflammation through Toll-like receptor (TLR) signaling pathways during C. trachomatis infection have not been fully characterized. SIGIRR (also known as IL-1R8 or TIR8) can regulate inflammation in response to various pathogens and diseases. However, nothing is known about its role during C. trachomatis infection. Expression of the pro-inflammatory chemokine, IL-8, was measured in epithelial cells infected with C. trachomatis. The effect of SIGIRR was determined by depleting SIGIRR or over-expressing SIGIRR in the epithelial cells before infection. Our results indicate that, in the absence of SIGIRR, epithelial cells induce higher levels of the pro-inflammatory chemokine, IL-8, in response to C. trachomatis infection. In addition, SIGIRR associates with MyD88 in both infected and uninfected infected cells. Collectively, our data demonstrate that SIGIRR functions as a negative regulator of the immune response to C. trachomatis infection. This finding provides insights into the immuno-pathogenesis of C. trachomatis that can be used to treat and identify individuals at risk of uncontrolled inflammation during infection.


Subject(s)
Chlamydia trachomatis/physiology , Receptors, Interleukin-1/metabolism , Toll-Like Receptors/metabolism , Chlamydia trachomatis/immunology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Gene Expression Regulation/immunology , Gene Silencing , HeLa Cells , Humans , Interleukin-8/genetics , Myeloid Differentiation Factor 88/metabolism , RNA, Messenger/genetics , Receptors, Interleukin-1/deficiency , Receptors, Interleukin-1/genetics
5.
J Immunol ; 201(1): 31-40, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29743314

ABSTRACT

CD8 T cells can play both a protective and pathogenic role in inflammation and autoimmune development. Recent studies have highlighted the ability of CD8 T cells to function as T follicular helper (Tfh) cells in the germinal center in the context of infection. However, whether this phenomenon occurs in autoimmunity and contributes to autoimmune pathogenesis is largely unexplored. In this study, we show that CD8 T cells acquire a CD4 Tfh profile in the absence of functional regulatory T cells in both the IL-2-deficient and scurfy mouse models. Depletion of CD8 T cells mitigates autoimmune pathogenesis in IL-2-deficient mice. CD8 T cells express the B cell follicle-localizing chemokine receptor CXCR5, a principal Tfh transcription factor Bcl6, and the Tfh effector cytokine IL-21. CD8 T cells localize to the B cell follicle, express B cell costimulatory proteins, and promote B cell differentiation and Ab isotype class switching. These data reveal a novel contribution of autoreactive CD8 T cells to autoimmune disease, in part, through CD4 follicular-like differentiation and functionality.


Subject(s)
Anemia, Hemolytic, Autoimmune/immunology , Anemia, Hemolytic, Autoimmune/pathology , CD8-Positive T-Lymphocytes/immunology , Immunoglobulin Class Switching/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Autoimmunity/immunology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Differentiation/immunology , Erythrocytes/immunology , Female , Interleukin-2/genetics , Interleukins/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Proto-Oncogene Proteins c-bcl-6/metabolism , Receptors, CXCR5/metabolism
6.
J Autoimmun ; 75: 58-67, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27453063

ABSTRACT

Bone marrow (BM) failure syndrome encompasses a group of disorders characterized by BM stem cell dysfunction, resulting in varying degrees of hypoplasia and blood pancytopenia, and in many patients is autoimmune and inflammatory in nature. The important role of T helper 1 (Th1) polarized CD4+ T cells in driving BM failure has been clearly established in several models. However, animal model data demonstrating a functional role for CD8+ T cells in BM dysfunction is largely lacking and our objective was to test the hypothesis that CD8+ T cells play a non-redundant role in driving BM failure. Clinical evidence implicates a detrimental role for CD8+ T cells in BM failure and a beneficial role for Foxp3+ regulatory T cells (Tregs) in maintaining immune tolerance in the BM. We demonstrate that IL-2-deficient mice, which have a deficit in functional Tregs, develop spontaneous BM failure. Furthermore, we demonstrate a critical role for CD8+ T cells in the development of BM failure, which is dependent on the cytokine, IFNγ. CD8+ T cells promote hematopoietic stem cell dysfunction and depletion of myeloid lineage progenitor cells, resulting in anemia. Adoptive transfer experiments demonstrate that CD8+ T cells dramatically expedite disease progression and promote CD4+ T cell accumulation in the BM. Thus, BM dysregulation in IL-2-deficient mice is mediated by a Th1 and IFNγ-producing CD8+ T cell (Tc1) response.


Subject(s)
Autoimmunity/immunology , Bone Marrow Cells/immunology , Bone Marrow/immunology , CD8-Positive T-Lymphocytes/immunology , Hematopoietic Stem Cells/immunology , Adoptive Transfer , Anemia/genetics , Anemia/immunology , Anemia/metabolism , Animals , Bone Marrow/metabolism , Bone Marrow/pathology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation , Flow Cytometry , Forkhead Transcription Factors , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-2/deficiency , Interleukin-2/genetics , Interleukin-2/immunology , Mice, Inbred BALB C , Mice, Knockout , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
7.
PLoS One ; 9(4): e93939, 2014.
Article in English | MEDLINE | ID: mdl-24695582

ABSTRACT

The immune system eliminates Chlamydia trachomatis infection through inflammation. However, uncontrolled inflammation can enhance pathology. In mice, TNF-related apoptosis-inducing ligand receptor (TRAIL-R), known for its effects on apoptosis, also regulates inflammation. In humans, the four homologues of TRAIL-R had never been investigated for effects on inflammation. Here, we examined whether TRAIL-R regulates inflammation during chlamydial infection. We examined TRAIL-R1 single nucleotide polymorphisms (SNPs) in an Ecuadorian cohort with and without C. trachomatis infections. There was a highly significant association for the TRAIL+626 homozygous mutant GG for infection vs no infection in this population. To confirm the results observed in the human population, primary lung fibroblasts and bone marrow-derived macrophages (BMDMs) were isolated from wildtype (WT) and TRAIL-R-deficient mice, and TRAIL-R1 levels in human cervical epithelial cells were depleted by RNA interference. Infection of BMDMs and primary lung fibroblasts with C. trachomatis strain L2, or the murine pathogen C. muridarum, led to higher levels of MIP2 mRNA expression or IL-1ß secretion from TRAIL-R-deficient cells than WT cells. Similarly, depletion of TRAIL-R1 expression in human epithelial cells resulted in a higher level of IL-8 mRNA expression and protein secretion during C. trachomatis infection. We conclude that human TRAIL-R1 SNPs and murine TRAIL-R modulate the innate immune response against chlamydial infection. This is the first evidence that human TRAIL-R1 is a negative regulator of inflammation and plays a role in modulating Chlamydia pathogenesis.


Subject(s)
Chlamydia Infections/genetics , Genetic Predisposition to Disease , Inflammation/genetics , Polymorphism, Single Nucleotide , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Adolescent , Adult , Animals , Chlamydia Infections/immunology , Chlamydia Infections/metabolism , Chlamydia trachomatis , Female , Fibroblasts/metabolism , Genotype , Humans , Immunity, Innate/genetics , Inflammation/immunology , Inflammation/metabolism , Lung/immunology , Lung/metabolism , Macrophages/metabolism , Mice , Mice, Knockout , Middle Aged , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Young Adult
8.
Cell Microbiol ; 15(5): 779-94, 2013 May.
Article in English | MEDLINE | ID: mdl-23163821

ABSTRACT

Chlamydia pneumoniae is responsible for a high prevalence of respiratory infections worldwide and has been implicated in atherosclerosis. Inflammation is regulated by transcription factor (TF) networks. Yet, the core TF network triggered by chlamydiae remains largely unknown. Primary human coronary artery endothelial cells were mock-infected or infected with C. pneumoniae to generate human transcriptome data throughout the chlamydial developmental cycle. Using systems network analysis, the predominant TF network involved receptor, binding and adhesion and immune response complexes. Cells transfected with interfering RNA against activator protein-1 (AP-1) members FOS, FOSB, JUN and JUNB had significantly decreased expression and protein levels of inflammatory mediators interleukin (IL)6, IL8, CD38 and tumour necrosis factor compared with controls. These mediators have been shown to be associated with C. pneumoniae disease. Expression of AP-1 components was regulated by MAPK3K8, a MAPK pathway component. Additionally, knock-down of JUN and FOS showed significantly decreased expression of Toll-like receptor (TLR)3 during infection, implicating JUN and FOS in TLR3 regulation. TLR3 stimulation led to elevated IL8. These findings suggest that C. pneumoniae initiates signalling via TLR3 and MAPK that activate AP-1, a known immune activator in other bacteria not previously shown for chlamydiae, triggering inflammation linked to C. pneumoniae disease.


Subject(s)
Atherosclerosis/metabolism , Chlamydophila pneumoniae/metabolism , Inflammation/metabolism , Transcription Factor AP-1/metabolism , Atherosclerosis/microbiology , Atherosclerosis/physiopathology , Chlamydophila pneumoniae/genetics , Chlamydophila pneumoniae/pathogenicity , Coronary Vessels/metabolism , Coronary Vessels/microbiology , Coronary Vessels/pathology , Endothelial Cells , Gene Expression Profiling , Gene Expression Regulation , Humans , Inflammation/genetics , Interleukin-8/metabolism , Pneumonia, Bacterial/metabolism , Pneumonia, Bacterial/microbiology , Pneumonia, Bacterial/physiopathology , Toll-Like Receptor 3/metabolism , Transcription Factor AP-1/genetics
9.
PLoS One ; 7(1): e30542, 2012.
Article in English | MEDLINE | ID: mdl-22276210

ABSTRACT

BACKGROUND: Long-term hematopoietic stem cells (LT-HSCs) migrate from the fetal liver (FL) to the fetal bone marrow (FBM) during development. Various adhesion and chemotactic receptor genes have been implicated in the migration of adult LT-HSCs. However, their role in the migration of fetal LT-HSCs is not clearly understood due, in part, to the rare number of these cells in fetal tissues, which preclude classical gene expression analysis. The aim of this study is to characterize the expression of migration related genes in fetal LT-HSC across different anatomical locations during development. METHODOLOGY/PRINCIPAL FINDINGS: We isolated fetal LT-HSC from different developmental stages, as well as different anatomical locations, and performed single-cell multiplex RT-qPCR and flow cytometry analysis of eight molecules involved in adult LT-HSC migration. Our results show that the gene expression of the chemokine receptor Cxcr4 in LT-HSC varies across developmental microenvironments and times, while the cadherin Cdh2 (Ncad) and the calcium receptor Casr show higher gene expression and variability only in FBM at 17.5 days post coitum (dpc). The cadherin Cdh5 (Vecad) maintains high expression variability only during fetal development, while the integrin subunit Itga5 (α5) increases its variability after 14.5 dpc. The integrin subunits Itga4 (α4) and Itgal (Lfa1), as well as the selectin ligand Selplg (Psgl1), did not show differences in their expression in single LT-HSCs irrespective of the developmental times or anatomical microenvironments studied. CONCLUSIONS/SIGNIFICANCE: Our data demonstrate that the expression pattern of phenotypically identical, single LT-HSCs fluctuates as a function of developmental stage and anatomical microenvironment. This is the first exhaustive gene expression comparison of migration-related molecules in fetal tissues across developmental times, enhancing the understanding of LT-HSC migration fate decisions during development.


Subject(s)
Fetus/metabolism , Hematopoietic Stem Cells/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Bone Marrow/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Movement/physiology , Female , Flow Cytometry , Hematopoietic Stem Cells/cytology , Liver/metabolism , Lymphocyte Function-Associated Antigen-1/genetics , Lymphocyte Function-Associated Antigen-1/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Multiplex Polymerase Chain Reaction , Pregnancy , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Receptors, Calcium-Sensing/genetics , Receptors, Calcium-Sensing/metabolism
10.
Carcinogenesis ; 28(1): 60-70, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16774933

ABSTRACT

Non-Hodgkin's lymphoma (NHL) is a group of malignancies with heterogeneous genetic and epigenetic alterations. Discovery of molecular markers that better define NHL should improve diagnosis, prognosis and understanding of the biology. We developed a CpG island DNA microarray for discovery of aberrant methylation targets in cancer, and now apply this method to examine NHL cell lines and primary tumors. This methylation profiling revealed differential patterns in six cell lines originating from different subtypes of NHL. We identified 30 hypermethylated genes in these cell lines and independently confirmed 10 of them. Methylation of 6 of these genes was then further examined in 75 primary NHL specimens composed of four subtypes representing different stages of maturation. Each gene (DLC-1, PCDHGB7, CYP27B1, EFNA5, CCND1 and RARbeta2) was frequently hypermethylated in these NHLs (87, 78, 61, 53, 40 and 38%, respectively), but not in benign follicular hyperplasia. Although some genes such as DLC-1 and PCDHGB7 were methylated in the vast majority of NHLs, others were differentially methylated in specific subtypes. The methylation of the candidate tumor suppressor gene DLC-1 was detected in a high proportion of primary tumor and plasma DNA samples by using quantitative methylation-specific PCR analysis. This promoter hypermethylation inversely correlated with DLC-1 gene expression in primary NHL samples. Thus, this CpG island microarray is a powerful discovery tool to identify novel methylated genes for further studies of their relevant molecular pathways in NHLs and identification of potential epigenetic biomarkers of disease.


Subject(s)
Biomarkers, Tumor/genetics , DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Lymphoma, Non-Hodgkin/genetics , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Blotting, Western , CpG Islands , GTPase-Activating Proteins , Gene Expression Profiling , Genome, Human , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Lymphoma, Non-Hodgkin/metabolism , Lymphoma, Non-Hodgkin/pathology , Microarray Analysis , Middle Aged , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...