Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
1.
Cell ; 180(1): 33-49.e22, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31813624

ABSTRACT

Gut-innervating nociceptor sensory neurons respond to noxious stimuli by initiating protective responses including pain and inflammation; however, their role in enteric infections is unclear. Here, we find that nociceptor neurons critically mediate host defense against the bacterial pathogen Salmonella enterica serovar Typhimurium (STm). Dorsal root ganglia nociceptors protect against STm colonization, invasion, and dissemination from the gut. Nociceptors regulate the density of microfold (M) cells in ileum Peyer's patch (PP) follicle-associated epithelia (FAE) to limit entry points for STm invasion. Downstream of M cells, nociceptors maintain levels of segmentous filamentous bacteria (SFB), a gut microbe residing on ileum villi and PP FAE that mediates resistance to STm infection. TRPV1+ nociceptors directly respond to STm by releasing calcitonin gene-related peptide (CGRP), a neuropeptide that modulates M cells and SFB levels to protect against Salmonella infection. These findings reveal a major role for nociceptor neurons in sensing and defending against enteric pathogens.


Subject(s)
Gastrointestinal Microbiome/physiology , Host Microbial Interactions/physiology , Nociceptors/physiology , Animals , Epithelium/metabolism , Female , Ganglia, Spinal/metabolism , Ganglia, Spinal/microbiology , Intestinal Mucosa/microbiology , Male , Mice , Mice, Inbred C57BL , Nociceptors/metabolism , Peyer's Patches/innervation , Peyer's Patches/metabolism , Salmonella Infections/metabolism , Salmonella typhimurium/metabolism , Salmonella typhimurium/pathogenicity , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/physiology
2.
J Immunol ; 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38905023

ABSTRACT

Chlamydia trachomatis is the most common cause of bacterial sexually transmitted infection in both men and women. Immunity to C. trachomatis involves many cell types, but CD4+ T cells play a key role in protecting the host during natural infection. Specifically, IFN-γ production by CD4+ T cells is the main effector responsible for bacterial clearance, yet the exact mechanism by which IFN-γ confers protection is poorly defined. In our efforts to define the specific mechanisms for bacterial clearance, we now show that IFN-γ upregulates expression of MHC class II (MHCII) on nonhematopoietic cells during C. trachomatis infection in vivo. We also find that MHCII expression on epithelial cells of the upper genital tract contributes to the efficient clearance of bacteria mediated by pathogen-specific CD4+ Th1 cells. As we further cataloged the protective mechanisms of C. trachomatis-specific CD4+ T cells, we found that the T cells also express granzyme B (GzmB) when coincubated with infected cells. In addition, during C. trachomatis infection of mice, primed activated-naive CD4+ Th1 cells displayed elevated granzyme transcripts (GzmA, GzmB, GzmM, GzmK, GzmC) compared with memory CD4+ T cells in vivo. Finally, using intracellular cytokine staining and a GzmB-/- mouse strain, we show that C. trachomatis-specific CD4+ Th1 cells express GzmB upon Ag stimulation, and that this correlates with Chlamydia clearance in vivo. Together these results have led us to conclude that Chlamydia-specific CD4+ Th1 cells develop cytotoxic capacity through engagement with nonhematopoietic MHCII, and this correlates to C. trachomatis clearance.

3.
PLoS Pathog ; 18(10): e1010903, 2022 10.
Article in English | MEDLINE | ID: mdl-36265003

ABSTRACT

The Lyme disease bacterial pathogen, Borrelia burgdorferi, establishes a long-term infection inside its mammalian hosts. Despite the continued presence of the bacteria in animal models of disease, inflammation is transitory and resolves spontaneously. T cells with limited effector functions and the inability to become activated by antigen, termed exhausted T cells, are present in many long-term infections. These exhausted T cells mediate a balance between pathogen clearance and preventing tissue damage resulting from excess inflammation. Exhausted T cells express a variety of immunoinhibitory molecules, including the molecule PD-1. Following B. burgdorferi infection, we found that PD-1 and its ligand PD-L1 are significantly upregulated on CD4+ T cells and antigen presenting cell subsets, respectively. Using mice deficient in PD-1, we found that the PD-1/PD-L1 pathway did not impact bacterial clearance but did impact T cell expansion and accumulation in the ankle joint and popliteal lymph nodes without affecting B cell populations or antibody production, suggesting that the PD-1/PD-L1 pathway may play a role in shaping the T cell populations present in affected tissues.


Subject(s)
Borrelia burgdorferi , Lyme Disease , Mice , Animals , Programmed Cell Death 1 Receptor , B7-H1 Antigen , Lyme Disease/microbiology , CD4-Positive T-Lymphocytes , Inflammation , Mammals
4.
Proc Natl Acad Sci U S A ; 115(9): 2216-2221, 2018 02 27.
Article in English | MEDLINE | ID: mdl-29440378

ABSTRACT

Infection with Chlamydia trachomatis drives severe mucosal immunopathology; however, the immune responses that are required for mediating pathology vs. protection are not well understood. Here, we employed a mouse model to identify immune responses required for C. trachomatis-induced upper genital tract pathology and to determine whether these responses are also required for bacterial clearance. In mice as in humans, immunopathology was characterized by extravasation of leukocytes into the upper genital tract that occluded luminal spaces in the uterus and ovaries. Flow cytometry identified these cells as neutrophils at early time points and CD4+ and CD8+ T cells at later time points. To determine what draws these cells to C. trachomatis-infected tissue, we measured the expression of 700 inflammation-related genes in the upper genital tract and found an up-regulation of many chemokines, including a node of interaction between CXCL9/10/11 and their common receptor CXCR3. Either depleting neutrophils or reducing T-cell numbers by CXCR3 blockade was sufficient to significantly ameliorate immunopathology but had no effect on bacterial burden, demonstrating that these responses are necessary for mucosal pathology but dispensable for C. trachomatis clearance. Therapies that specifically target these host responses may therefore prove useful in ameliorating C. trachomatis-induced pathology without exacerbating infection or transmission.


Subject(s)
Chlamydia Infections/pathology , Chlamydia trachomatis/classification , Genitalia, Female/pathology , Animals , Chlamydia Infections/microbiology , Female , Genitalia, Female/microbiology , Mice , Monocytes/physiology , Neutrophils/physiology , T-Lymphocytes
5.
J Biol Chem ; 293(16): 6022-6038, 2018 04 20.
Article in English | MEDLINE | ID: mdl-29496999

ABSTRACT

Germline-encoded receptors recognizing common pathogen-associated molecular patterns are a central element of the innate immune system and play an important role in shaping the host response to infection. Many of the innate immune molecules central to these signaling pathways are evolutionarily conserved. LysMD3 is a novel molecule containing a putative peptidoglycan-binding domain that has orthologs in humans, mice, zebrafish, flies, and worms. We found that the lysin motif (LysM) of LysMD3 is likely related to a previously described peptidoglycan-binding LysM found in bacteria. Mouse LysMD3 is a type II integral membrane protein that co-localizes with GM130+ structures, consistent with localization to the Golgi apparatus. We describe here two lines of mLysMD3-deficient mice for in vivo characterization of mLysMD3 function. We found that mLysMD3-deficient mice were born at Mendelian ratios and had no obvious pathological abnormalities. They also exhibited no obvious immune response deficiencies in a number of models of infection and inflammation. mLysMD3-deficient mice exhibited no signs of intestinal dysbiosis by 16S analysis or alterations in intestinal gene expression by RNA sequencing. We conclude that mLysMD3 contains a LysM with cytoplasmic orientation, but we were unable to define a physiological role for the molecule in vivo.


Subject(s)
Gene Deletion , Animals , Autoantigens/analysis , Bacterial Infections/genetics , Bacterial Infections/immunology , CRISPR-Cas Systems , Female , Immunity, Innate , Inflammation/genetics , Inflammation/immunology , Male , Membrane Proteins/analysis , Mice , Mycoses/genetics , Mycoses/immunology , Phylogeny , Virus Diseases/genetics , Virus Diseases/immunology
6.
Infect Immun ; 87(7)2019 07.
Article in English | MEDLINE | ID: mdl-30988057

ABSTRACT

Antigen-specific CD4+ T cells against Chlamydia are crucial for driving bacterial clearance and mediating protection against reinfection. Although the Chlamydia trachomatis protein Cta1 has been identified to be a dominant murine CD4+ T cell antigen, its level of expression during the bacterial developmental cycle and precise localization within the host cell are unknown. Newly developed tools for Chlamydia genetic manipulation have allowed us to generate a C. trachomatis strain expressing a heterologous CD4+ T cell epitope from ovalbumin (OVA) consisting of OVA residues 323 to 339 (OVA323-339). By tagging proteins expressed in C. trachomatis with OVA323-339, we can begin to understand how protein expression, developmental regulation, and subcellular compartmentalization affect the potential of those proteins to serve as antigens. When OVA323-339 was expressed as a fusion with green fluorescent protein, we found that we were able to elicit an OT-II T cell response in an antigen-dependent manner, but surprisingly, these T cells were unable to reduce bacterial burden in mice. These data suggest that the subcellular localization of antigen, the level of antigen expression, or the timing of expression within the developmental cycle of Chlamydia may play a crucial role in eliciting a protective CD4+ T cell response.


Subject(s)
Antigens, Bacterial/immunology , CD4-Positive T-Lymphocytes/immunology , Chlamydia Infections , Chlamydia trachomatis/immunology , Ovalbumin/metabolism , Animals , Chlamydia Infections/immunology , Chlamydia Infections/microbiology , Disease Models, Animal , Female , Mice , Mice, Inbred C57BL
7.
Infect Immun ; 87(9)2019 09.
Article in English | MEDLINE | ID: mdl-31285254

ABSTRACT

Chlamydia trachomatis is the most commonly reported bacterial sexually transmitted infection in the United States. Modeling infection in animals can be challenging, as mice naturally clear C. trachomatis when it is deposited in the lower genital tract. However, C. trachomatis can productively infect mice when the lower genital tract is bypassed and bacteria are deposited directly into the upper genital tract via transcervical inoculation. Interestingly, the mouse-adapted Chlamydia species C. muridarum can infect mice both by transcervical inoculation and by natural ascension if introduced into the vaginal vault. In this study, we investigated whether the route of infection plays a role in the downstream immune responses to C. muridarum infection. We found that transcervical infection with C. muridarum results in higher bacterial burdens in the upper genital tract at earlier time points, correlating with levels of innate immune cells. When bacterial burdens were equivalent in intravaginally and transcervically infected mice at later time points, we observed substantially higher levels of adaptive immune cells in transcervically infected mice. Our data suggest that different routes of infection with the same organism can elicit different immune responses in the same tissue.


Subject(s)
Chlamydia Infections , Chlamydia muridarum/immunology , Chlamydia trachomatis/immunology , Inflammation/immunology , Reproductive Tract Infections , Animals , Chlamydia Infections/immunology , Chlamydia Infections/microbiology , Disease Models, Animal , Female , Mice , Reproductive Tract Infections/immunology , Reproductive Tract Infections/microbiology
8.
J Cell Sci ; 130(14): 2251-2265, 2017 Jul 15.
Article in English | MEDLINE | ID: mdl-28536105

ABSTRACT

Tether complexes play important roles in endocytic and exocytic trafficking of lipids and proteins. In yeast, the multisubunit transport protein particle (TRAPP) tether regulates endoplasmic reticulum (ER)-to-Golgi and intra-Golgi transport and is also implicated in autophagy. In addition, the TRAPP complex acts as a guanine nucleotide exchange factor (GEF) for Ypt1, which is homologous to human Rab1a and Rab1b. Here, we show that human TRAPPC13 and other TRAPP subunits are critically involved in the survival response to several Golgi-disrupting agents. Loss of TRAPPC13 partially preserves the secretory pathway and viability in response to brefeldin A, in a manner that is dependent on ARF1 and the large GEF GBF1, and concomitant with reduced caspase activation and ER stress marker induction. TRAPPC13 depletion reduces Rab1a and Rab1b activity, impairs autophagy and leads to increased infectivity to the pathogenic bacterium Shigella flexneri in response to brefeldin A. Thus, our results lend support for the existence of a mammalian TRAPPIII complex containing TRAPPC13, which is important for autophagic flux under certain stress conditions.


Subject(s)
Antigens, Neoplasm/metabolism , Golgi Apparatus/metabolism , Vesicular Transport Proteins/metabolism , A549 Cells , ADP-Ribosylation Factor 1/metabolism , Anti-Bacterial Agents/pharmacology , Antigens, Neoplasm/drug effects , Autophagy/physiology , Brefeldin A/pharmacology , Dysentery, Bacillary/drug therapy , Dysentery, Bacillary/metabolism , Gene Knockdown Techniques , Guanine Nucleotide Exchange Factors/metabolism , HEK293 Cells , HT29 Cells , HeLa Cells , Humans , Shigella flexneri/drug effects , Vesicular Transport Proteins/antagonists & inhibitors , Vesicular Transport Proteins/drug effects
9.
J Immunol ; 198(7): 2772-2784, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28258198

ABSTRACT

Vaccine strategies to enhance CD8+ CTL responses remain a current challenge because they should overcome the plasmatic and endosomal membranes for favoring exogenous Ag access to the cytosol of APCs. As a way to avoid this hurdle, sticholysin (St) II, a pore-forming protein from the Caribbean Sea anemone Stichodactyla helianthus, was encapsulated with OVA into liposomes (Lp/OVA/StII) to assess their efficacy to induce a CTL response. OVA-specific CD8+ T cells transferred to mice immunized with Lp/OVA/StII experienced a greater expansion than when the recipients were injected with the vesicles without St, mostly exhibiting a memory phenotype. Consequently, Lp/OVA/StII induced a more potent effector function, as shown by CTLs, in vivo assays. Furthermore, treatment of E.G7-OVA tumor-bearing mice with Lp/OVA/StII significantly reduced tumor growth being more noticeable in the preventive assay. The contribution of CD4+ and CD8+ T cells to CTL and antitumor activity, respectively, was elucidated. Interestingly, the irreversibly inactive variant of the StI mutant StI W111C, encapsulated with OVA into Lp, elicited a similar OVA-specific CTL response to that observed with Lp/OVA/StII or vesicles encapsulating recombinant StI or the reversibly inactive StI W111C dimer. These findings suggest the relative independence between StII pore-forming activity and its immunomodulatory properties. In addition, StII-induced in vitro maturation of dendritic cells might be supporting these properties. These results are the first evidence, to our knowledge, that StII, a pore-forming protein from a marine eukaryotic organism, encapsulated into Lp functions as an adjuvant to induce a robust specific CTL response.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Cancer Vaccines/immunology , Cnidarian Venoms/administration & dosage , Neoplasms, Experimental/pathology , T-Lymphocytes, Cytotoxic/drug effects , Animals , Cnidarian Venoms/immunology , Female , Flow Cytometry , Liposomes/immunology , Mice , Mice, Inbred C57BL , T-Lymphocytes, Cytotoxic/immunology
10.
J Immunol ; 195(4): 1665-75, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26179901

ABSTRACT

The obligate intracellular bacterium Chlamydia trachomatis is the most common cause of bacterial sexually transmitted disease in the United States and the leading cause of preventable blindness worldwide. Transfer of cultured Chlamydia-specific CD8(+) T cells or vaccination with recombinant virus expressing an MHC I-restricted Chlamydia Ag confers protection, yet surprisingly a protective CD8(+) T cell response is not stimulated following natural infection. In this study, we demonstrate that the presence of excess IL-12 and IFN-γ contributes to poor memory CD8(+) T cell development during C. trachomatis infection of mice. IL-12 is required for CD8(+) T cell expansion but drives effector CD8(+) T cells into a short-lived fate, whereas IFN-γ signaling impairs the development of effector memory cells. We show that transient blockade of IL-12 and IFN-γ during priming promotes the development of memory precursor effector CD8(+) T cells and increases the number of memory T cells that participate in the recall protection against subsequent infection. Overall, this study identifies key factors shaping memory development of Chlamydia-specific CD8(+) T cells that will inform future vaccine development against this and other pathogens.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Chlamydia Infections/immunology , Chlamydia Infections/metabolism , Chlamydia trachomatis/immunology , Immunologic Memory , Interferon-gamma/metabolism , Interleukin-12/metabolism , Animals , Chlamydia Infections/genetics , Cytokines/metabolism , Disease Models, Animal , Gene Expression , Immunophenotyping , Inflammation Mediators/metabolism , Interleukin-2/immunology , Interleukin-2/metabolism , Mice , Mice, Transgenic , Mucous Membrane/immunology , Mucous Membrane/metabolism , Mucous Membrane/microbiology , Phenotype , Signal Transduction , Spleen/immunology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism
11.
J Immunol ; 194(5): 2319-29, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25637024

ABSTRACT

Understanding the cellular populations and mechanisms responsible for overcoming immune compartmentalization is valuable for designing vaccination strategies targeting distal mucosae. In this study, we show that the human pathogen Chlamydia trachomatis infects the murine respiratory and genital mucosae and that T cells, but not Abs, elicited through intranasal immunization can protect against a subsequent transcervical challenge. Unlike the genital infection where CD8(+) T cells are primed, yet fail to confer protection, we found that intranasal priming engages both CD4(+) and CD8(+) T cells, allowing for protection against genital infection with C. trachomatis. The protection is largely dependent on IFN-γ secretion by T cells. Moreover, different chemokine receptors are critical for C. trachomatis-specific CD4(+) T cells to home to the lung, rather than the CXCR3- and CCR5-dependent migration observed during genital infection. Overall, this study demonstrates that the cross-mucosa protective immunity against genital C. trachomatis infection following intranasal immunization is not dependent on Ab response but is mediated by not only CD4(+) T cells but also by CD8(+) T cells. This study provides insights for the development of vaccines against mucosal pathogens that threaten reproductive health worldwide.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Chlamydia Infections/prevention & control , Immunity, Cellular , Interferon-gamma/metabolism , Mucous Membrane/immunology , Administration, Intranasal , Animals , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/microbiology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/microbiology , CD8-Positive T-Lymphocytes/pathology , Cell Movement , Chlamydia Infections/immunology , Chlamydia Infections/microbiology , Chlamydia Infections/pathology , Chlamydia trachomatis/immunology , Female , Humans , Immunization , Interferon-gamma/biosynthesis , Lung/immunology , Lung/microbiology , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mucous Membrane/microbiology , Mucous Membrane/pathology , Uterus/immunology , Uterus/microbiology , Uterus/pathology
12.
J Immunol ; 192(3): 1079-90, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24353266

ABSTRACT

Chlamydia trachomatis infection is the most common bacterial sexually transmitted disease in the United States. Repeated infections with C. trachomatis lead to serious sequelae, such as infertility. It is unclear why the adaptive immune system, specifically the CD8(+) T cell response, is unable to protect against subsequent C. trachomatis infections. In this article, we characterize the mucosal CD8(+) T cell response to C. trachomatis in the murine genital tract. We demonstrate that the immunoinhibitory ligand, PD-L1, contributes to the defective CD8(+) T cell response. Deletion or inhibition of PD-L1 restores the CD8(+) T cell response and enhances C. trachomatis clearance.


Subject(s)
B7-H1 Antigen/physiology , CD8-Positive T-Lymphocytes/immunology , Cervix Uteri/immunology , Chlamydia Infections/immunology , Chlamydia trachomatis/immunology , Genital Diseases, Female/immunology , Immunity, Mucosal/immunology , T-Lymphocyte Subsets/immunology , Uterus/immunology , Animals , B7-1 Antigen/immunology , B7-H1 Antigen/deficiency , Cervix Uteri/microbiology , Female , Genital Diseases, Female/microbiology , Hematopoietic Stem Cells/immunology , Immune Tolerance/immunology , Mice , Mice, Inbred C57BL , Mucous Membrane/immunology , Radiation Chimera , Uterus/microbiology
13.
J Immunol ; 192(9): 4284-93, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24659687

ABSTRACT

Chlamydia trachomatis infection is the most common sexually transmitted bacterial infection in the United States and a significant health burden worldwide. Protection from Chlamydia infection in the genital mucosa is dependent on IFN-γ derived from CD4(+) Th1 cells. These CD4(+) T cells must home successfully to the genital tract to exert their effector function and decrease C. trachomatis burden. Although adhesion receptors expressed by CD4(+) T cells in the genital tract have been characterized, the integrin receptor required for Chlamydia-specific CD4(+) T cell-mediated protection has not been explored. In this study, we demonstrate that C. trachomatis infection of the upper genital tract results in recruitment of Chlamydia-specific CD4(+) T cells robustly expressing the integrin α4ß1. Interfering with α4ß1, but not α4ß7, function resulted in defective CD4(+) T cell trafficking to the uterus and high bacterial load. We conclude that integrin α4ß1 is necessary for CD4(+) T cell-mediated protection against C. trachomatis infection in the genital mucosa. By identifying homing molecules required for successful CD4(+) T cell trafficking to C. trachomatis-infected tissues, we will be better equipped to design vaccines that elicit sterilizing, long-lasting immunity without inducing immune pathologies in the upper genital tract.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Chlamydia Infections/immunology , Chlamydia trachomatis/immunology , Integrin alpha4beta1/immunology , Animals , Chemotaxis, Leukocyte/immunology , Female , Flow Cytometry , Genitalia, Female/immunology , Genitalia, Female/metabolism , Genitalia, Female/microbiology , Integrin alpha4beta1/metabolism , Mice , Mice, Inbred C57BL , Mucous Membrane/immunology , Mucous Membrane/metabolism , Polymerase Chain Reaction
14.
Proc Natl Acad Sci U S A ; 109(25): 10059-64, 2012 Jun 19.
Article in English | MEDLINE | ID: mdl-22675117

ABSTRACT

Chlamydia trachomatis is a pathogen responsible for a prevalent sexually transmitted disease. It is also the most common cause of infectious blindness in the developing world. We performed a loss-of-function genetic screen in human haploid cells to identify host factors important in C. trachomatis L2 infection. We identified and confirmed B3GAT3, B4GALT7, and SLC35B2, which encode glucuronosyltransferase I, galactosyltransferase I, and the 3'-phosphoadenosine 5'-phosphosulfate transporter 1, respectively, as important in facilitating Chlamydia infection. Knockout of any of these three genes inhibits Chlamydia attachment. In complementation studies, we found that the introduction of functional copies of these three genes into the null clones restored full susceptibility to Chlamydia infection. The degree of attachment of Chlamydia strongly correlates with the level of sulfation of the host cell, not simply with the amount of heparan sulfate. Thus, other, as-yet unidentified sulfated macromolecules must contribute to infection. These results demonstrate the utility of screens in haploid cells to study interactions of human cells with bacteria. Furthermore, the human null clones generated can be used to investigate the role of heparan sulfate and sulfation in other settings not limited to infectious disease.


Subject(s)
Bacterial Adhesion/physiology , Chlamydia trachomatis/physiology , Sulfates/metabolism , Bacterial Proteins/genetics , Cells, Cultured , Chlamydia trachomatis/genetics , Haploidy , Humans
15.
PLoS Pathog ; 8(8): e1002809, 2012.
Article in English | MEDLINE | ID: mdl-22912573

ABSTRACT

The activation of host cells by interferon gamma (IFNγ) is essential for inhibiting the intracellular replication of most microbial pathogens. Although significant advances have been made in identifying IFNγ-dependent host factors that suppress intracellular bacteria, little is known about how IFNγ enables cells to recognize, or restrict, the growth of pathogens that replicate in the host cytoplasm. The replication of the cytosolic bacterial pathogen Shigella flexneri is significantly inhibited in IFNγ-stimulated cells, however the specific mechanisms that mediate this inhibition have remained elusive. We found that S. flexneri efficiently invades IFNγ-activated mouse embryonic fibroblasts (MEFs) and escapes from the vacuole, suggesting that IFNγ acts by blocking S. flexneri replication in the cytosol. This restriction on cytosolic growth was dependent on interferon regulatory factor 1 (IRF1), an IFNγ-inducible transcription factor capable of inducing IFNγ-mediated cell-autonomous immunity. To identify host factors that restrict S. flexneri growth, we used whole genome microarrays to identify mammalian genes whose expression in S. flexneri-infected cells is controlled by IFNγ and IRF1. Among the genes we identified was the pattern recognition receptor (PRR) retanoic acid-inducible gene I (RIG-I), a cytoplasmic sensor of foreign RNA that had not been previously known to play a role in S. flexneri infection. We found that RIG-I and its downstream signaling adaptor mitochondrial antiviral signaling protein (MAVS)--but not cytosolic Nod-like receptors (NLRs)--are critically important for IFNγ-mediated S. flexneri growth restriction. The recently described RNA polymerase III pathway, which transcribes foreign cytosolic DNA into the RIG-I ligand 5'-triphosphate RNA, appeared to be involved in this restriction. The finding that RIG-I responds to S. flexneri infection during the IFNγ response extends the range of PRRs that are capable of recognizing this bacterium. Additionally, these findings expand our understanding of how IFNγ recognizes, and ultimately restricts, bacterial pathogens within host cells.


Subject(s)
Cytoplasm/immunology , DEAD-box RNA Helicases/immunology , Dysentery, Bacillary/immunology , Immunity, Innate , Interferon-gamma/immunology , Shigella flexneri/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Animals , Cells, Cultured , Cytoplasm/genetics , Cytoplasm/microbiology , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , Dysentery, Bacillary/genetics , Embryo, Mammalian/immunology , Embryo, Mammalian/microbiology , Fibroblasts/immunology , Fibroblasts/microbiology , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/immunology , Interferon-gamma/genetics , Mice , Mice, Knockout , Signal Transduction/genetics , Signal Transduction/immunology
16.
J Immunol ; 189(5): 2441-9, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22855710

ABSTRACT

Chlamydia trachomatis infection is the most common bacterial sexually transmitted disease in the United States. Chlamydia infections that ascend to the upper genital tract can persist, trigger inflammation, and result in serious sequelae such as infertility. However, mouse models in which the vaginal vault is inoculated with C. trachomatis do not recapitulate the course of human disease. These intravaginal infections of the mouse do not ascend efficiently to the upper genital tract, do not cause persistent infection, do not induce significant inflammation, and do not induce significant CD4⁺ T cell infiltration. In this article, we describe a noninvasive transcervical infection model in which we bypass the cervix and directly inoculate C. trachomatis into the uterus. We show that direct C. trachomatis infection of the murine upper genital tract stimulates a robust Chlamydia-specific CD4⁺ T cell response that is both necessary and sufficient to clear infection and provide protection against reinfection.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/microbiology , Chlamydia Infections/immunology , Chlamydia Infections/prevention & control , Chlamydia trachomatis/immunology , Genitalia, Female/immunology , Genitalia, Female/microbiology , Amino Acid Sequence , Animals , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/immunology , CD4-Positive T-Lymphocytes/pathology , Cervix Uteri/immunology , Cervix Uteri/microbiology , Cervix Uteri/pathology , Chlamydia Infections/pathology , Disease Models, Animal , Female , Mice , Mice, 129 Strain , Mice, Congenic , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data
17.
PLoS Pathog ; 7(6): e1001346, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21731484

ABSTRACT

The obligate intracellular pathogen Chlamydia trachomatis is the most common cause of bacterial sexually transmitted diseases in the United States. In women C. trachomatis can establish persistent genital infections that lead to pelvic inflammatory disease and sterility. In contrast to natural infections in humans, experimentally induced infections with C. trachomatis in mice are rapidly cleared. The cytokine interferon-γ (IFNγ) plays a critical role in the clearance of C. trachomatis infections in mice. Because IFNγ induces an antimicrobial defense system in mice but not in humans that is composed of a large family of Immunity Related GTPases (IRGs), we questioned whether mice deficient in IRG immunity would develop persistent infections with C. trachomatis as observed in human patients. We found that IRG-deficient Irgm1/m3((-/-)) mice transiently develop high bacterial burden post intrauterine infection, but subsequently clear the infection more efficiently than wildtype mice. We show that the delayed but highly effective clearance of intrauterine C. trachomatis infections in Irgm1/m3((-/-)) mice is dependent on an exacerbated CD4(+) T cell response. These findings indicate that the absence of the predominant murine innate effector mechanism restricting C. trachomatis growth inside epithelial cells results in a compensatory adaptive immune response, which is at least in part driven by CD4(+) T cells and prevents the establishment of a persistent infection in mice.


Subject(s)
Adaptive Immunity , CD4-Positive T-Lymphocytes/immunology , Chlamydia Infections/immunology , Chlamydia trachomatis/immunology , Hydro-Lyases/deficiency , Animals , CD4-Positive T-Lymphocytes/microbiology , Epithelial Cells/microbiology , Mice , Mice, Knockout
18.
J Exp Med ; 203(2): 383-91, 2006 Feb 20.
Article in English | MEDLINE | ID: mdl-16461341

ABSTRACT

Studies using major histocompatibility complex (MHC)-Ia-deficient mice have shown that MHC-Ib-restricted CD8+ T cells can clear infections caused by intracellular pathogens such as Listeria monocytogenes. M3-restricted CD8+ T cells, which recognize short hydrophobic N-formylated peptides, appear to comprise a substantial portion of the MHC-Ib-restricted T cell response in the mouse model of L. monocytogenes infection. In this study, we isolated formyltransferase (fmt) mutant strains of L. monocytogenes that lacked the ability to add formyl groups to nascent polypeptides. These fmt mutant Listeria strains did not produce antigens that could be recognized by M3-restricted T cells. We showed that immunization of MHC-Ia-deficient mice with fmt mutant Listeria resulted in stimulation of a protective memory response that cleared subsequent challenge with wild-type L. monocytogenes, despite the fact that M3-restricted CD8+ T cells did not proliferate in these mice. These data suggest that M3-restricted T cells are not required for protection against L. monocytogenes and underscore the importance of searching for new antigen-presenting molecules among the large MHC-Ib family of proteins.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/microbiology , Histocompatibility Antigens Class I/genetics , Listeria monocytogenes/immunology , Listeriosis/immunology , Animals , Cell Line, Tumor , Cell Proliferation , Histocompatibility Antigens Class I/physiology , Hydroxymethyl and Formyl Transferases/genetics , Listeria monocytogenes/enzymology , Listeria monocytogenes/genetics , Listeriosis/microbiology , Liver/immunology , Liver/microbiology , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Mutation , Peptides/metabolism , Spleen/immunology , Spleen/microbiology
19.
Infect Immun ; 79(5): 2021-30, 2011 May.
Article in English | MEDLINE | ID: mdl-21357720

ABSTRACT

CD8(+) T lymphocytes often play a primary role in adaptive immunity to cytosolic microbial pathogens. Surprisingly, CD8(+) T cells are not required for protective immunity to the enteric pathogen Shigella flexneri, despite the ability of Shigella to actively secrete proteins into the host cytoplasm, a location from which antigenic peptides are processed for presentation to CD8(+) T cells. To determine why CD8(+) T cells fail to play a role in adaptive immunity to S. flexneri, we investigated whether antigen-specific CD8(+) T cells are primed during infection but are unable to confer protection or, alternatively, whether T cells fail to be primed. To test whether Shigella is capable of stimulating an antigen-specific CD8(+) T-cell response, we created an S. flexneri strain that constitutively secretes a viral CD8(+) T-cell epitope via the Shigella type III secretion system and characterized the CD8(+) T-cell response to this strain both in mice and in cultured cells. Surprisingly, no T cells specific for the viral epitope were stimulated in mice infected with this strain, and cells infected with the recombinant strain were not targeted by epitope-specific T cells. Additionally, we found that the usually robust T-cell response to antigens artificially introduced into the cytoplasm of cultured cells was significantly reduced when the antigen-presenting cell was infected with Shigella. Collectively, these results suggest that antigen-specific CD8(+) T cells are not primed during S. flexneri infection and, as a result, afford little protection to the host during primary or subsequent infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dysentery, Bacillary/immunology , Lymphocyte Activation/immunology , Shigella flexneri/immunology , Animals , Antigens, Bacterial/immunology , Epitopes, T-Lymphocyte/immunology , Immunoblotting , Mice , Mice, Inbred C57BL
20.
PLoS Pathog ; 5(9): e1000573, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19730693

ABSTRACT

All Yersinia species target and bind to phagocytic cells, but uptake and destruction of bacteria are prevented by injection of anti-phagocytic Yop proteins into the host cell. Here we provide evidence that CD8(+) T cells, which canonically eliminate intracellular pathogens, are important for restricting Yersinia, even though bacteria are primarily found in an extracellular locale during the course of disease. In a model of infection with attenuated Y. pseudotuberculosis, mice deficient for CD8(+) T cells were more susceptible to infection than immunocompetent mice. Although exposure to attenuated Y. pseudotuberculosis generated T(H)1-type antibody responses and conferred protection against challenge with fully virulent bacteria, depletion of CD8(+) T cells during challenge severely compromised protective immunity. Strikingly, mice lacking the T cell effector molecule perforin also succumbed to Y. pseudotuberculosis infection. Given that the function of perforin is to kill antigen-presenting cells, we reasoned that cell death marks bacteria-associated host cells for internalization by neighboring phagocytes, thus allowing ingestion and clearance of the attached bacteria. Supportive of this model, cytolytic T cell killing of Y. pseudotuberculosis-associated host cells results in engulfment by neighboring phagocytes of both bacteria and target cells, bypassing anti-phagocytosis. Our findings are consistent with a novel function for cell-mediated immune responses protecting against extracellular pathogens like Yersinia: perforin and CD8(+) T cells are critical for hosts to overcome the anti-phagocytic action of Yops.


Subject(s)
Antigen-Presenting Cells/immunology , CD8-Positive T-Lymphocytes/immunology , Yersinia pseudotuberculosis Infections/immunology , Yersinia pseudotuberculosis/immunology , Animals , Bacterial Vaccines/immunology , Disease Models, Animal , Host-Pathogen Interactions/immunology , Immunoglobulin G/metabolism , Kaplan-Meier Estimate , Liver/immunology , Liver/microbiology , Lymphoid Tissue/immunology , Lymphoid Tissue/microbiology , Mice , Mice, Inbred C57BL , Phagocytosis , Pore Forming Cytotoxic Proteins/immunology , Statistics, Nonparametric , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Helper-Inducer/immunology , Vaccines, Attenuated/immunology , Yersinia pseudotuberculosis/pathogenicity
SELECTION OF CITATIONS
SEARCH DETAIL