Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
J Immunol ; 212(3): 369-374, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38117750

ABSTRACT

NKp44 is a human receptor originally found on activated NK cells, group 1 and group 3 innate lymphoid cells that binds dimers of platelet-derived growth factor D (PDGF-DD). NKp44 is also expressed on tissue plasmacytoid dendritic cells (PDCs), but NKp44-PDGF-DD interaction on PDCs remains unstudied. Engagement of NKp44 with PDGF-DD in vitro enhanced PDC secretion of IFN-α, TNF, and IL-6 in response to the TLR9 ligand CpG-ODN, but not TLR7/8 ligands. In tissues, PDCs were found in close contact with PDGF-DD-expressing cells in the high endothelial venules and epithelium of tonsils, melanomas, and skin lesions infected with Molluscum contagiosum. Recombinant PDGF-DD enhanced the serum IFN-α response to systemic HSV-1 infection in a humanized mouse model. We conclude that NKp44 integrates with TLR9 signaling to enhance PDC cytokine production. These findings may have bearings for immune responses to TLR9-based adjuvants, therapy for tumors expressing PDGF-DD, and infections with DNA viruses that induce PDGF-DD expression to enhance viral spread.


Subject(s)
Immunity, Innate , Toll-Like Receptor 9 , Animals , Mice , Humans , Toll-Like Receptor 9/metabolism , Interferon-alpha/metabolism , Dendritic Cells , Killer Cells, Natural
2.
Cell Rep ; 42(10): 113140, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37768824

ABSTRACT

Dietary fiber strongly impacts the microbiota. Here, we show that a low-fiber diet changes the small intestinal (SI) microbiota and impairs SI Th17, TCRαß+CD8αß+ and TCRαß+CD8αα+ intraepithelial T cell development. We restore T cell development with dietary fiber supplementation, but this defect becomes persistent over generations with constant low-fiber diets. Offspring of low-fiber diet-fed mice have reduced SI T cells even after receiving a fiber-rich diet due to loss of bacteria important for T cell development. In these mice, only a microbiota transplant from a fiber-rich diet-fed mouse and a fiber-rich diet can restore T cell development. Low-fiber diets reduce segmented filamentous bacteria (SFB) abundance, impairing its vertical transmission. SFB colonization and a fiber-rich diet partially restore T cell development. Finally, we observe that low-fiber diet-induced T cell defects render mice more susceptible to Citrobacter rodentium infection. Together, these results demonstrate the importance of fiber to microbiota vertical transmission and host immune system development.


Subject(s)
Gastrointestinal Microbiome , Intraepithelial Lymphocytes , Microbiota , Mice , Animals , Intestine, Small/microbiology , Receptors, Antigen, T-Cell, alpha-beta , Intestinal Mucosa/microbiology , Dietary Fiber , Mice, Inbred C57BL
3.
Cell ; 186(9): 1846-1862.e26, 2023 04 27.
Article in English | MEDLINE | ID: mdl-37028428

ABSTRACT

The use of probiotics by cancer patients is increasing, including among those undergoing immune checkpoint inhibitor (ICI) treatment. Here, we elucidate a critical microbial-host crosstalk between probiotic-released aryl hydrocarbon receptor (AhR) agonist indole-3-aldehyde (I3A) and CD8 T cells within the tumor microenvironment that potently enhances antitumor immunity and facilitates ICI in preclinical melanoma. Our study reveals that probiotic Lactobacillus reuteri (Lr) translocates to, colonizes, and persists within melanoma, where via its released dietary tryptophan catabolite I3A, it locally promotes interferon-γ-producing CD8 T cells, thereby bolstering ICI. Moreover, Lr-secreted I3A was both necessary and sufficient to drive antitumor immunity, and loss of AhR signaling within CD8 T cells abrogated Lr's antitumor effects. Further, a tryptophan-enriched diet potentiated both Lr- and ICI-induced antitumor immunity, dependent on CD8 T cell AhR signaling. Finally, we provide evidence for a potential role of I3A in promoting ICI efficacy and survival in advanced melanoma patients.


Subject(s)
Limosilactobacillus reuteri , Melanoma , Tumor Microenvironment , Humans , Diet , Immune Checkpoint Inhibitors , Limosilactobacillus reuteri/metabolism , Melanoma/therapy , Tryptophan/metabolism , CD8-Positive T-Lymphocytes/immunology , Receptors, Aryl Hydrocarbon/agonists
4.
Trends Immunol ; 43(11): 858-860, 2022 11.
Article in English | MEDLINE | ID: mdl-36243620

ABSTRACT

CD4+ intraepithelial lymphocytes (CD4IEL) are tissue-resident T cells with cytotoxic and regulatory properties; together with peripheral regulatory T cells, they control intestinal inflammation. Recently, Bousbaine and colleagues identified a microbiota-derived conserved antigen that induces CD4IEL differentiation and promotes their regulatory function, attenuating the severity of murine colitis.


Subject(s)
Intraepithelial Lymphocytes , Microbiota , Humans , Mice , Animals , CD4-Positive T-Lymphocytes , T-Lymphocytes, Regulatory , Antigens , Intestinal Mucosa
5.
Cell Host Microbe ; 30(1): 10-12, 2022 01 12.
Article in English | MEDLINE | ID: mdl-35026131

ABSTRACT

In this issue of Cell Host & Microbe, Alexander et al. show that the enzyme cardiac glycoside reductase 2 (cgr2), which is produced by Eggerthella lenta, metabolizes RORγT inhibitors, resulting in an increased Th17 response and more severe inflammation in colitis models. The effect of cgr2 can be neutralized by a diet rich in arginine.


Subject(s)
Colitis , Nuclear Receptor Subfamily 1, Group F, Member 3 , Humans , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Oxidoreductases , Th17 Cells
6.
Proc Natl Acad Sci U S A ; 118(36)2021 09 07.
Article in English | MEDLINE | ID: mdl-34462359

ABSTRACT

Oral infection with Toxoplasma gondii results in dysbiosis and enteritis, both of which revert to normal during chronic infection. However, whether infection leaves a lasting impact on mucosal responses remains uncertain. Here we examined the effect of the chemical irritant dextran sodium sulfate (DSS) on intestinal damage and wound healing in chronically infected mice. Our findings indicate that prior infection with T. gondii exacerbates damage to the colon caused by DSS and impairs wound healing by suppressing stem cell regeneration of the epithelium. Enhanced tissue damage was attributable to inflammatory monocytes that emerge preactivated from bone marrow, migrate to the intestine, and release inflammatory mediators, including nitric oxide. Tissue damage was reversed by neutralization of inflammatory monocytes or nitric oxide, revealing a causal mechanism for tissue damage. Our findings suggest that chronic infection with T. gondii enhances monocyte activation to increase inflammation associated with a secondary environmental insult.


Subject(s)
Colitis/complications , Toxoplasmosis/complications , Animals , Chronic Disease , Disease Susceptibility , Gastrointestinal Microbiome , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Monocytes/pathology , Regeneration , Stem Cells/pathology
7.
bioRxiv ; 2021 May 13.
Article in English | MEDLINE | ID: mdl-34013278

ABSTRACT

Infection with SARS-CoV-2 has caused a pandemic of unprecedented dimensions. SARS-CoV-2 infects airway and lung cells causing viral pneumonia. The importance of type I interferon (IFN) production for the control of SARS-CoV-2 infection is highlighted by the increased severity of COVID-19 in patients with inborn errors of type I IFN response or auto-antibodies against IFN-α. Plasmacytoid dendritic cells (pDCs) are a unique immune cell population specialized in recognizing and controlling viral infections through the production of high concentrations of type I IFN. In this study, we isolated pDCs from healthy donors and showed that pDCs are able to recognize SARS-CoV-2 and rapidly produce large amounts of type I IFN. Sensing of SARS-CoV-2 by pDCs was independent of viral replication since pDCs were also able to recognize UV-inactivated SARS-CoV-2 and produce type I IFN. Transcriptional profiling of SARS-CoV-2 and UV-SARS-CoV-2 stimulated pDCs also showed a rapid type I and III IFN response as well as induction of several chemokines, and the induction of apoptosis in pDCs. Moreover, we modeled SARS-CoV-2 infection in the lung using primary human airway epithelial cells (pHAEs) and showed that co-culture of pDCs with SARS-CoV-2 infected pHAEs induces an antiviral response and upregulation of antigen presentation in pHAE cells. Importantly, the presence of pDCs in the co-culture results in control of SARS-CoV-2 replication in pHAEs. Our study identifies pDCs as one of the key cells that can recognize SARS-CoV-2 infection, produce type I and III IFN and control viral replication in infected cells. IMPORTANCE: Type I interferons (IFNs) are a major part of the innate immune defense against viral infections. The importance of type I interferon (IFN) production for the control of SARS-CoV-2 infection is highlighted by the increased severity of COVID-19 in patients with defects in the type I IFN response. Interestingly, many cells are not able to produce type I IFN after being infected with SARS-CoV-2 and cannot control viral infection. In this study we show that plasmacytoid dendritic cells are able to recognize SARS-CoV-2 and produce type I IFN, and that pDCs are able to help control viral infection in SARS-CoV-2 infected airway epithelial cells.

9.
J Immunol ; 204(8): 2257-2268, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32169845

ABSTRACT

Plasmacytoid dendritic cells (pDCs) produce abundant type I IFNs (IFN-I) in response to viral nucleic acids. Generation of pDCs from bone marrow dendritic cell (DC) progenitors and their maintenance is driven by the transcription factor E2-2 and inhibited by its repressor Id2. In this study, we find that mouse pDCs selectively express the receptor for LIF that signals through STAT3. Stimulation of pDCs with LIF inhibited IFN-I, TNF, and IL-6 responses to CpG and induced expression of the STAT3 targets SOCS3 and Bcl3, which inhibit IFN-I and NF-κB signaling. Moreover, although STAT3 has been also reported to induce E2-2, LIF paradoxically induced its repressor Id2. A late-stage bone marrow DC progenitor expressed low amounts of LIFR and developed into pDCs less efficiently after being exposed to LIF, consistent with the induction of Id2. Conversely, pDC development and serum IFN-I responses to lymphocytic choriomeningitis virus infection were augmented in newly generated mice lacking LIFR in either CD11c+ or hematopoietic cells. Thus, an LIF-driven STAT3 pathway induces SOCS3, Bcl3, and Id2, which render pDCs and late DC progenitors refractory to physiological stimuli controlling pDC functions and development. This pathway can be potentially exploited to prevent inappropriate secretion of IFN-I in autoimmune diseases or promote IFN-I secretion during viral infections.


Subject(s)
Dendritic Cells/cytology , Dendritic Cells/immunology , Leukemia Inhibitory Factor/metabolism , Animals , Interferon Type I/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , STAT3 Transcription Factor/immunology , Signal Transduction/immunology
10.
Nutrition ; 73: 110693, 2020 05.
Article in English | MEDLINE | ID: mdl-32065881

ABSTRACT

Hepatic encephalopathy is a cerebral alteration mainly caused by hepatic insufficiency or portosystemic shunt. It ranges from minimal neurologic manifestations to coma in its overt stage. The multifactorial pathophysiology of hepatic encephalopathy has rendered numerous treatment alternatives, among which the modification of dysbiosis and hyperammonemia are currently considered to be effective, not only clinically, but also regarding cost. Recent work has developed knowledge of probiotics in different clinical settings including the relationship of the gut microenvironment to liver cirrhosis. The aim of this review was to analyze the results of clinical trials on the effect of different probiotics on hepatic encephalopathy.


Subject(s)
Hepatic Encephalopathy , Microbiota , Probiotics , Dysbiosis/therapy , Hepatic Encephalopathy/therapy , Humans , Liver Cirrhosis/therapy
11.
Front Immunol ; 10: 1423, 2019.
Article in English | MEDLINE | ID: mdl-31312200

ABSTRACT

The gastrointestinal tract hosts the largest collection of commensal microbes in the body. Infections at this site can cause significant perturbations in the microbiota, known as dysbiosis, that facilitate the expansion of pathobionts, and can elicit inappropriate immune responses that impair the intestinal barrier function. Dysbiosis typically occurs during intestinal infection with Toxoplasma gondii. Host resistance to T. gondii depends on a potent Th1 response. In addition, a Th17 response is also elicited. How Th17 cells contribute to the host response to T. gondii remains unclear. Here we show that class I-restricted T cell-associated molecule (CRTAM) expression on T cells is required for an optimal IL-17 production during T. gondii infection. Moreover, that the lack of IL-17, results in increased immunopathology caused by an impaired antimicrobial peptide production and bacterial translocation from the intestinal lumen to the mesenteric lymph nodes and spleen.


Subject(s)
Dysbiosis/parasitology , Gastrointestinal Microbiome/immunology , Immunoglobulins/metabolism , Interleukin-17/metabolism , Th17 Cells/immunology , Toxoplasma/immunology , Toxoplasmosis, Animal/immunology , Animals , Cell Differentiation/genetics , Dysbiosis/immunology , Female , Immunoglobulins/genetics , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Toxoplasmosis, Animal/parasitology , alpha-Defensins/metabolism , beta-Defensins/metabolism
12.
Semin Immunopathol ; 40(4): 371-377, 2018 07.
Article in English | MEDLINE | ID: mdl-29951906

ABSTRACT

The intestinal immune system is challenged daily with the task of recognizing and eliminating pathogens while simultaneously tolerating dietary and commensal antigens. All components must effectively coordinate to differentiate a continual barrage of environmental cues and mount appropriate responses dependent on the nature of the stimuli encountered. Playing a pivotal role, the aryl hydrocarbon receptor (AHR) is a chemical sensor that detects both dietary and microbial cues and is important for development, maintenance, and function of several types of intestinal immune cells, particularly innate lymphoid cells (ILCs) and T cells. In this review, we will highlight recent advances in our knowledge of the role of AHR signaling in ILCs, T cells, B cells, and dendritic cells.


Subject(s)
Immunity, Innate , Peyer's Patches/cytology , Peyer's Patches/physiology , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Gene Expression Regulation , Humans , Intestines/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Ligands , Lymphocytes/immunology , Lymphocytes/metabolism , Macrophages/immunology , Macrophages/metabolism , Protein Binding , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
13.
Curr Opin Immunol ; 50: 112-116, 2018 02.
Article in English | MEDLINE | ID: mdl-29452963

ABSTRACT

The immune system of the intestinal tract has the challenging task of recognizing and eliminating intestinal pathogens while maintaining tolerance to dietary and commensal antigens; therefore, it must be able to sense environmental cues within the intestine and mount suitable responses dictated by their pathogenic or nonpathogenic nature. The aryl hydrocarbon receptor (AHR) was originally characterized as a chemical sensor of the environmental pollutant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) [12]. More recently, AHR has emerged as a major chemical sensor expressed in many intestinal immune cells that enables them to distinguish nutritional and microbial cues and is, therefore, important for development, maintenance and function of the intestinal immune system. In this review, we will highlight recent advances in our knowledge of the role of AHR signaling in intestinal innate lymphoid cells (ILC), T cells and B cells.


Subject(s)
Mucosal-Associated Invariant T Cells/immunology , Mucosal-Associated Invariant T Cells/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Differentiation/immunology , Humans , Immunity, Innate , Ligands , Mucosal-Associated Invariant T Cells/cytology , Signal Transduction
14.
Science ; 357(6353): 806-810, 2017 08 25.
Article in English | MEDLINE | ID: mdl-28775213

ABSTRACT

The small intestine contains CD4+CD8αα+ double-positive intraepithelial lymphocytes (DP IELs), which originate from intestinal CD4+ T cells through down-regulation of the transcription factor Thpok and have regulatory functions. DP IELs are absent in germ-free mice, which suggests that their differentiation depends on microbial factors. We found that DP IEL numbers in mice varied in different vivaria, correlating with the presence of Lactobacillus reuteri This species induced DP IELs in germ-free mice and conventionally-raised mice lacking these cells. L. reuteri did not shape the DP-IEL-TCR (TCR, T cell receptor) repertoire but generated indole derivatives of tryptophan that activated the aryl-hydrocarbon receptor in CD4+ T cells, allowing Thpok down-regulation and differentiation into DP IELs. Thus, L. reuteri, together with a tryptophan-rich diet, can reprogram intraepithelial CD4+ T cells into immunoregulatory T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Gastrointestinal Microbiome/immunology , Intestine, Small/immunology , Intestine, Small/microbiology , Limosilactobacillus reuteri/immunology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Down-Regulation , Germ-Free Life , Indoles/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Mice , Mice, Inbred C57BL , Receptors, Aryl Hydrocarbon/metabolism , Transcription Factors/metabolism , Tryptophan/metabolism
15.
Nat Immunol ; 18(9): 995-1003, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28759002

ABSTRACT

Among the features that distinguish type 1 innate lymphoid cells (ILC1s) from natural killer (NK) cells is a gene signature indicative of 'imprinting' by cytokines of the TGF-ß family. We studied mice in which ILC1s and NK cells lacked SMAD4, a signal transducer that facilitates the canonical signaling pathway common to all cytokines of the TGF-ß family. While SMAD4 deficiency did not affect ILC1 differentiation, NK cells unexpectedly acquired an ILC1-like gene signature and were unable to control tumor metastasis or viral infection. Mechanistically, SMAD4 restrained non-canonical TGF-ß signaling mediated by the cytokine receptor TGFßR1 in NK cells. NK cells from a SMAD4-deficient person affected by polyposis were also hyper-responsive to TGF-ß. These results identify SMAD4 as a previously unknown regulator that restricts non-canonical TGF-ß signaling in NK cells.


Subject(s)
Killer Cells, Natural/cytology , Lymphopoiesis/genetics , Smad4 Protein/genetics , Transforming Growth Factor beta/immunology , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/immunology , Animals , Case-Control Studies , Cell Differentiation , Gene Expression Profiling , Humans , Immunity, Innate/immunology , Immunoblotting , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Lymphocytes/cytology , Melanoma, Experimental/immunology , Mice , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/immunology , Smad4 Protein/immunology
16.
PLoS Pathog ; 13(2): e1006195, 2017 02.
Article in English | MEDLINE | ID: mdl-28158275

ABSTRACT

Coronaviruses are of veterinary and medical importance and include highly pathogenic zoonotic viruses, such as SARS-CoV and MERS-CoV. They are known to efficiently evade early innate immune responses, manifesting in almost negligible expression of type-I interferons (IFN-I). This evasion strategy suggests an evolutionary conserved viral function that has evolved to prevent RNA-based sensing of infection in vertebrate hosts. Here we show that the coronavirus endonuclease (EndoU) activity is key to prevent early induction of double-stranded RNA (dsRNA) host cell responses. Replication of EndoU-deficient coronaviruses is greatly attenuated in vivo and severely restricted in primary cells even during the early phase of the infection. In macrophages we found immediate induction of IFN-I expression and RNase L-mediated breakdown of ribosomal RNA. Accordingly, EndoU-deficient viruses can retain replication only in cells that are deficient in IFN-I expression or sensing, and in cells lacking both RNase L and PKR. Collectively our results demonstrate that the coronavirus EndoU efficiently prevents simultaneous activation of host cell dsRNA sensors, such as Mda5, OAS and PKR. The localization of the EndoU activity at the site of viral RNA synthesis-within the replicase complex-suggests that coronaviruses have evolved a viral RNA decay pathway to evade early innate and intrinsic antiviral host cell responses.


Subject(s)
Coronaviridae/enzymology , Coronavirus Infections/immunology , Endonucleases/immunology , Immune Evasion/physiology , Viral Proteins/immunology , Animals , Coronaviridae/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate/immunology , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction
17.
Cell Metab ; 24(1): 158-66, 2016 07 12.
Article in English | MEDLINE | ID: mdl-27374498

ABSTRACT

Remodeling of the tricarboxylic acid (TCA) cycle is a metabolic adaptation accompanying inflammatory macrophage activation. During this process, endogenous metabolites can adopt regulatory roles that govern specific aspects of inflammatory response, as recently shown for succinate, which regulates the pro-inflammatory IL-1ß-HIF-1α axis. Itaconate is one of the most highly induced metabolites in activated macrophages, yet its functional significance remains unknown. Here, we show that itaconate modulates macrophage metabolism and effector functions by inhibiting succinate dehydrogenase-mediated oxidation of succinate. Through this action, itaconate exerts anti-inflammatory effects when administered in vitro and in vivo during macrophage activation and ischemia-reperfusion injury. Using newly generated Irg1(-/-) mice, which lack the ability to produce itaconate, we show that endogenous itaconate regulates succinate levels and function, mitochondrial respiration, and inflammatory cytokine production during macrophage activation. These studies highlight itaconate as a major physiological regulator of the global metabolic rewiring and effector functions of inflammatory macrophages.


Subject(s)
Inflammation/enzymology , Inflammation/pathology , Macrophages/metabolism , Succinate Dehydrogenase/antagonists & inhibitors , Succinates/pharmacology , Animals , Cell Respiration/drug effects , Female , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/enzymology , Macrophages/pathology , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Reperfusion Injury/enzymology , Reperfusion Injury/pathology , Succinate Dehydrogenase/metabolism , Succinic Acid/metabolism
18.
Science ; 352(6293): 1515-6, 2016 Jun 24.
Article in English | MEDLINE | ID: mdl-27339967

Subject(s)
Animals
19.
Immunity ; 44(5): 1127-39, 2016 05 17.
Article in English | MEDLINE | ID: mdl-27156386

ABSTRACT

The signals guiding differentiation of innate lymphoid cells (ILCs) within tissues are not well understood. Salivary gland (SG) ILCs as well as liver and intestinal intraepithelial ILC1 have markers that denote tissue residency and transforming growth factor-ß (TGF-ß) imprinting. We deleted Tgfbr2 in cells expressing the ILC and NK marker NKp46 and found that SG ILCs were reduced in number. They lost distinct tissue markers, such as CD49a, and the effector molecules TRAIL and CD73. Expression of the transcription factor Eomes, which promotes NK cell differentiation, was elevated. Conversely, Eomes deletion in NKp46(+) cells enhanced TGF-ß-imprinting of SG ILCs. Thus, TGF-ß induces SG ILC differentiation by suppressing Eomes. TGF-ß acted through a JNK-dependent, Smad4-independent pathway. Transcriptome analysis demonstrated that SG ILCs had characteristic of both NK cells and ILC1. Finally, TGF-ß imprinting of SG ILCs was synchronized with SG development, highlighting the impact of tissue microenvironment on ILC development.


Subject(s)
Cell Differentiation , Killer Cells, Natural/physiology , Lymphocytes/physiology , Salivary Glands/immunology , Transforming Growth Factor beta/metabolism , Animals , Antigens, Ly/metabolism , Cellular Microenvironment , Gene Expression Profiling , Immunity, Innate , MAP Kinase Kinase 4/metabolism , Mice , Mice, Knockout , Natural Cytotoxicity Triggering Receptor 1/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Smad4 Protein/genetics , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism
20.
J Exp Med ; 211(4): 623-33, 2014 Apr 07.
Article in English | MEDLINE | ID: mdl-24687959

ABSTRACT

Retention of lymphocytes in the intestinal mucosa requires specialized chemokine receptors and adhesion molecules. We find that both CD4(+)CD8(+) and CD4(+) T cells in the intestinal epithelium, as well as CD8(+) T cells in the intestinal mucosa and mesenteric lymph nodes, express the cell adhesion molecule class I-restricted T cell-associated molecule (Crtam) upon activation, whereas the ligand of Crtam, cell adhesion molecule 1 (Cadm1), is expressed on gut CD103(+)DCs. Lack of Crtam-Cadm1 interactions in Crtam(-/-) and Cadm1(-/-) mice results in loss of CD4(+)CD8(+) T cells, which arise from mucosal CD4(+) T cells that acquire a CD8 lineage expression profile. After acute oral infection with Toxoplasma gondii, both WT and Crtam(-/-) mice mounted a robust TH1 response, but markedly fewer TH17 cells were present in the intestinal mucosa of Crtam(-/-) mice. The almost exclusive TH1 response in Crtam(-/-) mice resulted in more efficient control of intestinal T. gondii infection. Thus, Crtam-Cadm1 interactions have a major impact on the residency and maintenance of CD4(+)CD8(+) T cells in the gut mucosa in the steady state. During pathogenic infection, Crtam-Cadm1 interactions regulate the dynamic equilibrium between newly formed CD4(+) T cells and their retention in the gut, thereby shaping representation of disparate CD4(+) T cell subsets and the overall quality of the CD4(+) T cell response.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/parasitology , Immunoglobulins/metabolism , Th17 Cells/immunology , Toxoplasma/physiology , Animals , Cell Adhesion Molecule-1 , Cell Adhesion Molecules/metabolism , Cell Polarity , Cytokines/biosynthesis , Dendritic Cells/metabolism , Gastrointestinal Tract/pathology , Immunoglobulins/deficiency , Interleukin-17/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/parasitology , Intestinal Mucosa/pathology , Ligands , Lymphocyte Count , Mice , Mice, Inbred C57BL , Neutralization Tests , Toxoplasmosis, Animal/immunology , Toxoplasmosis, Animal/parasitology
SELECTION OF CITATIONS
SEARCH DETAIL
...