Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Mucosal Immunol ; 11(3): 681-692, 2018 05.
Article in English | MEDLINE | ID: mdl-29346347

ABSTRACT

Antibody responses induced at mucosal and nonmucosal sites demonstrate a significant level of autonomy. Here, we demonstrate a key role for mucosal interferon regulatory factor-4 (IRF4)-dependent CD103+CD11b+ (DP), classical dendritic cells (cDCs) in the induction of T-dependent immunoglobulin G (IgG) and immunoglobulin A (IgA) responses in the mesenteric lymph node (MLN) following systemic immunization with soluble flagellin (sFliC). In contrast, IRF8-dependent CD103+CD11b- (SP) are not required for these responses. The lack of this response correlated with a complete absence of sFliC-specific plasma cells in the MLN, small intestinal lamina propria, and surprisingly also the bone marrow (BM). Many sFliC-specific plasma cells accumulating in the BM of immunized wild-type mice expressed α4ß7+, suggesting a mucosal origin. Collectively, these results suggest that mucosal DP cDC contribute to the generation of the sFliC-specific plasma cell pool in the BM and thus serve as a bridge linking the mucosal and systemic immune system.


Subject(s)
Dendritic Cells/immunology , Interferon Regulatory Factors/metabolism , Lymph Nodes/immunology , Mucous Membrane/immunology , Plasma Cells/immunology , Animals , Antigens, CD/metabolism , CD11b Antigen/metabolism , Cells, Cultured , Flagellin/immunology , Immunity, Humoral , Immunoglobulin A/metabolism , Immunoglobulin Class Switching , Immunoglobulin G/metabolism , Integrin alpha Chains/metabolism , Integrin alpha4/metabolism , Integrin beta Chains/metabolism , Interferon Regulatory Factors/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout
2.
Parasite Immunol ; 39(10)2017 Oct.
Article in English | MEDLINE | ID: mdl-28802050

ABSTRACT

Low- and high-dose infections with the murine large intestinal nematode Trichuris muris are associated with induction of adaptive Th1 and Th2 responses, respectively, in mesenteric lymph nodes (MLN). Classical dendritic cells (cDC) accumulate in the large intestinal mucosa and MLN upon T. muris infection, yet their role in driving adaptive responses to infection remains largely unknown. We performed low- and high-dose T. muris infections of mice deficient in defined cDC subsets to investigate their role in induction of adaptive immune responses. Mice lacking IRF4-dependent cDC failed to clear a high-dose infection and displayed impaired Th2 responses. Conversely, mice lacking IRF8-dependent cDC cleared a low-dose infection and displayed an impaired Th1 response while increased production of Th2 cytokines. Finally, mice lacking both IRF4- and IRF8-dependent cDC were able to generate a Th2 response and clear a low-dose infection. Collectively, these results suggest that IRF4- and IRF8-dependent cDC act antagonistically during T. muris infection, and demonstrate that intestinal Th2 responses can be generated towards T. muris in the absence of IRF4-dependent cDC.


Subject(s)
Dendritic Cells/immunology , Interferon Regulatory Factors/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Trichuriasis/immunology , Trichuris/immunology , Animals , Cytokines/biosynthesis , Intestinal Mucosa/immunology , Lymph Nodes/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Trichuriasis/parasitology
3.
Mucosal Immunol ; 10(4): 845-864, 2017 07.
Article in English | MEDLINE | ID: mdl-28378807

ABSTRACT

The intestinal lamina propria (LP) contains a diverse array of mononuclear phagocyte (MNP) subsets, including conventional dendritic cells (cDC), monocytes and tissue-resident macrophages (mφ) that collectively play an essential role in mucosal homeostasis, infection and inflammation. In the current review we discuss the function of intestinal cDC and monocyte-derived MNP, highlighting how these subsets play several non-redundant roles in the regulation of intestinal immune responses. While much remains to be learnt, recent findings also underline how the various populations of MNP adapt to deal with the challenges specific to their environment. Understanding these processes should help target individual subsets for 'fine tuning' immunological responses within the intestine, a process that may be of relevance both for the treatment of inflammatory bowel disease (IBD) and for optimized vaccine design.


Subject(s)
Immunotherapy/methods , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/immunology , Leukocytes, Mononuclear/immunology , Phagocytes/immunology , Vaccines/immunology , Animals , Humans , Immunity, Mucosal , Immunomodulation , Inflammatory Bowel Diseases/therapy
4.
Mucosal Immunol ; 9(1): 171-82, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26080709

ABSTRACT

Disruption of the homeostatic balance of intestinal dendritic cells (DCs) and macrophages (MQs) may contribute to inflammatory bowel disease. We characterized DC and MQ populations, including their ability to produce retinoic acid, in clinical material encompassing Crohn's ileitis, Crohn's colitis and ulcerative colitis (UC) as well as mesenteric lymph nodes (MLNs) draining these sites. Increased CD14(+)DR(int) MQs characterized inflamed intestinal mucosa while total CD141(+) or CD1c(+) DCs numbers were unchanged. However, CD103(+) DCs, including CD141(+)CD103(+) and CD1c(+)CD103(+) DCs, were reduced in inflamed intestine. In MLNs, two CD14(-) DC populations were identified: CD11c(int)HLADR(hi) and CD11c(hi)HLADR(int) cells. A marked increase of CD11c(hi)HLADR(int) DC, particularly DR(int)CD1c(+) DCs, characterized MLNs draining inflamed intestine. The fraction of DC and MQ populations expressing aldehyde dehydrogenase (ALDH) activity, reflecting retinoic acid synthesis, in UC colon, both in active disease and remission, were reduced compared to controls and inflamed Crohn's colon. In contrast, no difference in the frequency of ALDH(+) cells among blood precursors was detected between UC patients and non-inflamed controls. This suggests that ALDH activity in myeloid cells in the colon of UC patients, regardless of whether the disease is active or in remission, is influenced by the intestinal environment.


Subject(s)
Aldehyde Dehydrogenase/immunology , Colitis, Ulcerative/immunology , Colon/immunology , Crohn Disease/immunology , Dendritic Cells/immunology , Macrophages/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Aldehyde Dehydrogenase/genetics , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, CD1/genetics , Antigens, CD1/immunology , Antigens, Surface/genetics , Antigens, Surface/immunology , CD11c Antigen/genetics , CD11c Antigen/immunology , Case-Control Studies , Colitis, Ulcerative/genetics , Colitis, Ulcerative/pathology , Colon/pathology , Crohn Disease/genetics , Crohn Disease/pathology , Dendritic Cells/pathology , Female , Gene Expression Regulation , Glycoproteins/genetics , Glycoproteins/immunology , HLA-DR Antigens/genetics , HLA-DR Antigens/immunology , Humans , Integrin alpha Chains/genetics , Integrin alpha Chains/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Lipopolysaccharide Receptors/genetics , Lipopolysaccharide Receptors/immunology , Lymph Nodes/immunology , Lymph Nodes/pathology , Macrophages/pathology , Male , Middle Aged , Severity of Illness Index , Signal Transduction , Thrombomodulin
5.
Mucosal Immunol ; 8(3): 545-58, 2015 May.
Article in English | MEDLINE | ID: mdl-25269704

ABSTRACT

Mucosal tissues contain large numbers of memory CD4(+) T cells that, through T-cell receptor-dependent interactions with antigen-presenting cells, are believed to have a key role in barrier defense and maintenance of tissue integrity. Here we identify a major subset of memory CD4(+) T cells at barrier surfaces that coexpress interleukin-18 receptor alpha (IL-18Rα) and death receptor-3 (DR3), and display innate lymphocyte functionality. The cytokines IL-15 or the DR3 ligand tumor necrosis factor (TNF)-like cytokine 1A (TL1a) induced memory IL-18Rα(+)DR3(+)CD4(+) T cells to produce interferon-γ, TNF-α, IL-6, IL-5, IL-13, granulocyte-macrophage colony-stimulating factor (GM-CSF), and IL-22 in the presence of IL-12/IL-18. TL1a synergized with IL-15 to enhance this response, while suppressing IL-15-induced IL-10 production. TL1a- and IL-15-mediated cytokine induction required the presence of IL-18, whereas induction of IL-5, IL-13, GM-CSF, and IL-22 was IL-12 independent. IL-18Rα(+)DR3(+)CD4(+) T cells with similar functionality were present in human skin, nasal polyps, and, in particular, the intestine, where in chronic inflammation they localized with IL-18-producing cells in lymphoid aggregates. Collectively, these results suggest that human memory IL-18Rα(+)DR3(+) CD4(+) T cells may contribute to antigen-independent innate responses at barrier surfaces.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Crohn Disease/immunology , Immunity, Innate , Nasal Polyps/immunology , Receptors, Interleukin-18/immunology , Receptors, Tumor Necrosis Factor, Member 25/immunology , CD4-Positive T-Lymphocytes/pathology , Crohn Disease/genetics , Crohn Disease/pathology , Epithelial Cells/immunology , Epithelial Cells/pathology , Gene Expression Regulation , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Humans , Immunity, Mucosal , Immunologic Memory , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-13/genetics , Interleukin-13/immunology , Interleukin-15/genetics , Interleukin-15/immunology , Interleukin-5/genetics , Interleukin-5/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Interleukins/genetics , Interleukins/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Nasal Polyps/genetics , Nasal Polyps/pathology , Primary Cell Culture , Receptors, Interleukin-18/genetics , Receptors, Tumor Necrosis Factor, Member 25/genetics , Signal Transduction , Skin/cytology , Skin/immunology , Tumor Necrosis Factor Ligand Superfamily Member 15/genetics , Tumor Necrosis Factor Ligand Superfamily Member 15/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Interleukin-22
6.
Mucosal Immunol ; 8(1): 38-48, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24850430

ABSTRACT

Cross-presentation of cellular antigens is crucial for priming CD8(+) T cells, and generating immunity to intracellular pathogens--particularly viruses. It is unclear which intestinal phagocytes perform this function in vivo. To address this, we examined dendritic cells (DCs) from the intestinal lymph of IFABP-tOVA 232-4 mice, which express ovalbumin in small intestinal epithelial cells (IECs). Among lymph DCs (LDCs) only CD103(+) CD11b(-) CD8α(+) DCs cross-present IEC-derived ovalbumin to CD8(+) OT-I T cells. Similarly, in the mesenteric lymph nodes (MLNs), cross-presentation of IEC-ovalbumin was limited to the CD11c(+) MHCII(hi) CD8α(+) migratory DCs, but absent from all other subsets, including the resident CD8α(hi) DCs. Crucially, delivery of purified CD8α(+) LDCs, but not other LDC subsets, into the MLN subcapsular lymphatic sinus induced proliferation of ovalbumin-specific, gut-tropic CD8(+) T cells in vivo. Finally, in 232-4 mice treated with R848, CD8α(+) LDCs were uniquely able to cross-prime interferon γ-producing CD8(+) T cells and drive their migration to the intestine. Our results clearly demonstrate that migrating CD8α(+) intestinal DCs are indispensable for cross-presentation of cellular antigens and, in conditions of inflammation, for the initial differentiation of effector CD8(+) T cells. They may therefore represent an important target for the development of antiviral vaccinations.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Ovalbumin/metabolism , Animals , Antigens/immunology , CD8 Antigens/metabolism , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cells, Cultured , Cross-Priming/drug effects , Cross-Priming/genetics , Imidazoles/administration & dosage , Imidazoles/pharmacology , Interferon-gamma/metabolism , Intestinal Mucosa/immunology , Lymph/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Membrane Glycoproteins/agonists , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Ovalbumin/genetics , Ovalbumin/immunology , Toll-Like Receptor 7/agonists
7.
Mucosal Immunol ; 8(2): 327-39, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25138666

ABSTRACT

The identification of intestinal macrophages (mφs) and dendritic cells (DCs) is a matter of intense debate. Although CD103(+) mononuclear phagocytes (MPs) appear to be genuine DCs, the nature and origins of CD103(-) MPs remain controversial. We show here that intestinal CD103(-)CD11b(+) MPs can be separated clearly into DCs and mφs based on phenotype, gene profile, and kinetics. CD64(-)CD103(-)CD11b(+) MPs are classical DCs, being derived from Flt3 ligand-dependent, DC-committed precursors, not Ly6C(hi) monocytes. Surprisingly, a significant proportion of these CD103(-)CD11b(+) DCs express CCR2 and there is a selective decrease in CD103(-)CD11b(+) DCs in mice lacking this chemokine receptor. CCR2(+)CD103(-) DCs are present in both the murine and human intestine, drive interleukin (IL)-17a production by T cells in vitro, and show constitutive expression of IL-12/IL-23p40. These data highlight the heterogeneity of intestinal DCs and reveal a bona fide population of CCR2(+) DCs that is involved in priming mucosal T helper type 17 (Th17) responses.


Subject(s)
Cell Differentiation , Dendritic Cells/cytology , Dendritic Cells/immunology , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , T-Lymphocyte Subsets/metabolism , Th17 Cells/metabolism , Animals , Antigens, CD/metabolism , Dendritic Cells/metabolism , Humans , Immunophenotyping , Integrin alpha Chains/metabolism , Interferon Regulatory Factors/metabolism , Interleukin-12/metabolism , Interleukin-17/biosynthesis , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Transgenic , Monocytes/immunology , Monocytes/metabolism , Phagocytes/immunology , Phagocytes/metabolism , Phenotype , Receptors, CCR2/metabolism , T-Lymphocyte Subsets/immunology , Th17 Cells/immunology
8.
Mucosal Immunol ; 6(3): 498-510, 2013 May.
Article in English | MEDLINE | ID: mdl-22990622

ABSTRACT

Macrophages (mφ) are essential for intestinal homeostasis and the pathology of inflammatory bowel disease (IBD), but it is unclear whether discrete mφ populations carry out these distinct functions or if resident mφ change during inflammation. We show here that most resident mφ in resting mouse colon express very high levels of CX3CR1, are avidly phagocytic and MHCII(hi), but are resistant to Toll-like receptor (TLR) stimulation, produce interleukin 10 constitutively, and express CD163 and CD206. A smaller population of CX3CR1(int) cells is present in resting colon and it expands during experimental colitis. Ly6C(hi)CCR2(+) monocytes can give rise to all mφ subsets in both healthy and inflamed colon and we show that the CX3CR1(int) pool represents a continuum in which newly arrived, recently divided monocytes develop into resident CX3CR1(hi) mφ. This process is arrested during experimental colitis, resulting in the accumulation of TLR-responsive pro-inflammatory mφ. Phenotypic analysis of human intestinal mφ indicates that analogous processes occur in the normal and Crohn's disease ileum. These studies show for the first time that resident and inflammatory mφ in the intestine represent alternative differentiation outcomes of the same precursor and targeting these events could offer routes for therapeutic intervention in IBD.


Subject(s)
Colitis/immunology , Colon/immunology , Inflammatory Bowel Diseases/immunology , Macrophages/immunology , Monocytes/immunology , Animals , Antigens, Ly/metabolism , CX3C Chemokine Receptor 1 , Cell Differentiation , Cell Movement , Cell Proliferation , Cells, Cultured , Colitis/chemically induced , Histocompatibility Antigens Class II/metabolism , Humans , Inflammation/pathology , Interleukin-10/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism
9.
J Intern Med ; 272(5): 411-29, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22946654

ABSTRACT

Crohn's disease and ulcerative colitis are chronic inflammatory disorders of the gastrointestinal tract and are collectively referred to as inflammatory bowel disease (IBD). IBD is a major cause of lifetime morbidity, has a severe impact on quality of life of patients (equivalent to that of rheumatoid arthritis, asthma, migraine or diabetes) and constitutes a substantial economic burden to the healthcare system. The introduction of anti-tumour necrosis factor (TNF) antibodies has dramatically improved the treatment of IBD, but approximately one-third of patients are nonresponders and another 30-50% will eventually lose the therapeutic effect or become intolerant to these antibodies. Thus, there is an urgent and unmet need for new therapies. The aetiologies of the different forms of IBD have not been fully elucidated but there is strong evidence implicating T cells and T-cell migration to the gut in initiating and perpetuating the intestinal inflammatory process and tissue destruction. In recent years, progress in basic science has shed light on the mechanisms regulating T-cell migration to the gut and new therapeutics targeting these pathways have been developed. It is interesting that some of the factors directing the localization of T cells to the gut have been shown to be relatively organ specific, potentially enabling new T-cell-targeted treatments to demonstrate improved safety whilst preserving therapeutic efficacy. Here, fundamental aspects of the gut immune system, the generation of tissue-tropic effector T cells and the mechanisms of T-cell trafficking to the gut mucosa will be reviewed. In addition, the role of these processes in IBD and how they have been exploited for the development of novel therapies for IBD will be discussed.


Subject(s)
Biological Products/therapeutic use , Cell Movement/immunology , Inflammatory Bowel Diseases/immunology , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Biological Products/immunology , Humans , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/physiopathology , Intestinal Mucosa/immunology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/therapeutic use
10.
Mucosal Immunol ; 5(2): 150-60, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22166938

ABSTRACT

The αE integrin chain CD103 identifies a subset of migratory dendritic cells (DCs) in the gut, lung, and skin. To gain further understanding of the function of CD103(+) DCs in regulating adaptive immunity in vivo, we coupled ovalbumin (OVA) to the CD103 antibody M290 (M290.OVA). Intraperitoneal injection of M290.OVA induced OVA-specific CD8(+) and CD4(+) T-cell proliferation in lymph nodes (LNs) of wild-type but not CD103(-/-) mice, or in mice depleted of CD11c(+) cells. In the absence of maturation stimuli, systemic antigen targeting to CD103(+) DCs led to tolerance of CD8(+) T cells, whereas coadministration of adjuvant induced cytotoxic T-lymphocyte (CTL) immunity and antibody production. Mucosal intratracheal application of M290.OVA also induced T-cell proliferation in mediastinal LNs, yet the functional outcome was tolerance that inhibited subsequent development of allergic airway inflammation and immunoglobulin E (IgE) responses to inhaled OVA. These findings identify antigen targeting to CD103(+) DCs as a potential strategy to regulate immune responses in nonlymphoid mucosal tissues.


Subject(s)
Antigens, CD/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Dendritic Cells/metabolism , Integrin alpha Chains/metabolism , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Antibody Formation , Antigens/genetics , Antigens/immunology , Antigens/metabolism , Antigens, CD/genetics , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Proliferation , Cytotoxicity, Immunologic , Dendritic Cells/immunology , Dendritic Cells/pathology , Drug Administration Routes , Humans , Immune Tolerance , Immunization , Immunomodulation , Integrin alpha Chains/genetics , Mice , Mice, Knockout , Ovalbumin/genetics , Ovalbumin/immunology , Ovalbumin/metabolism , Protein Engineering , Recombinant Fusion Proteins/genetics
11.
Mucosal Immunol ; 4(4): 438-47, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21289617

ABSTRACT

Small intestinal lamina propria (SI-LP) CD103(+) dendritic cells (DCs) are imprinted with an ability to metabolize vitamin A (retinol), a property underlying their enhanced capacity to induce the gut-homing receptors CC chemokine receptor-9 and α4ß7 on responding T cells. In this study, we demonstrate that imprinting of CD103(+) DCs is itself critically dependent on vitamin A and occurs locally within the small intestine (SI). The major vitamin A metabolite retinoic acid (RA) induced retinol-metabolizing activity in DCs both in vitro and in vivo, suggesting a direct role for RA in this process. Consistent with this, SI-LP CD103(+) DCs constitutively received RA signals in vivo at significantly higher levels than did colonic CD103(+) DCs. Remarkably, SI CD103(+) DCs remained imprinted in mice depleted of dietary but not of systemic retinol. We found that bile contained high levels of retinol, induced RA receptor-dependent retinol-metabolizing activity in bone marrow-derived DCs, and imprinted these cells with the ability to generate gut-tropic T cells. Taken together, these results suggest a novel and unexpected role for bile in SI-LP CD103(+) DC imprinting.


Subject(s)
Bile/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Intestinal Mucosa/metabolism , Intestines/immunology , Retinoids/metabolism , T-Lymphocytes/immunology , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , Bile/chemistry , Bile/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Dendritic Cells/cytology , Diet , Integrin alpha Chains/immunology , Integrin alpha Chains/metabolism , Lymph Nodes/immunology , Lymph Nodes/metabolism , Mice , Mice, Inbred C57BL , Receptors, CCR/metabolism , Retinoids/immunology , Signal Transduction/immunology , T-Lymphocytes/cytology , Vitamin A/analysis
12.
Mucosal Immunol ; 3(1): 40-8, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19812544

ABSTRACT

Intestinal-derived chemokines have a central role in orchestrating immune cell influx into the normal and inflamed intestine. Here, we identify the chemokine CCL6 as one of the most abundant chemokines constitutively expressed by both murine small intestinal and colonic epithelial cells. CCL6 protein localized to crypt epithelial cells, was detected in the gut lumen and reached high concentrations at the mucosal surface. Its expression was further enhanced in the small intestine following in vivo administration of LPS or after stimulation of the small intestinal epithelial cell line, mIC(c12), with IFNgamma, IL-4 or TNFalpha. Recombinant- and intestinal-derived CCL6 bound to a subset of the intestinal microflora and displayed antibacterial activity. Finally, the human homologs to CCL6, CCL14 and CCL15 were also constitutively expressed at high levels in human intestinal epithelium, were further enhanced in inflammatory bowel disease and displayed similar antibacterial activity. These findings identify a novel role for constitutively expressed, epithelial-derived chemokines as antimicrobial peptides in the intestinal mucosa.


Subject(s)
Anti-Bacterial Agents/biosynthesis , Chemokines, CC/biosynthesis , Epithelial Cells/metabolism , Inflammatory Bowel Diseases/immunology , Macrophage Inflammatory Proteins/biosynthesis , Animals , Bacterial Adhesion/drug effects , Bacterial Adhesion/immunology , Chemokines, CC/genetics , Cytokines/pharmacology , Epithelial Cells/pathology , HT29 Cells , Humans , Immunization , Intestinal Mucosa/pathology , Lipopolysaccharides/administration & dosage , Macrophage Inflammatory Proteins/genetics , Mice , Mice, Inbred C57BL
13.
Mucosal Immunol ; 1(1): 38-48, 2008 Jan.
Article in English | MEDLINE | ID: mdl-19079159

ABSTRACT

Recent studies have highlighted a central role for intestinal dendritic cells (DCs) and vitamin A metabolite retinoic acid (RA) in the generation of alpha4beta7(+) CCR9(+)"gut tropic" effector T cells. Here, using RA-responsive element reporter mice, we demonstrate that both splenic and mesenteric lymph node (MLN) DCs enhanced retinoic acid receptor (RAR) signaling in CD8(+) T cells; however, only a subset of MLN DCs, expressing the integrin alpha-chain CD103, induced an early RAR signal that is required for efficient CCR9 induction. MLN-primed CD8(+) T cells also received enhanced RAR-dependent signals compared with splenic-primed CD8(+) T cells in vivo. Further DC-mediated induction of gut homing receptors was inhibited at a high antigen dose without influencing RAR signaling events, and resulted in less efficient CD8(+) T-cell entry into the small intestinal mucosa. These results highlight a complex interplay between antigen dose and DC subset-induced RAR signaling events in the generation of tissue tropic effector T-cell subsets.


Subject(s)
Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Intestinal Mucosa/immunology , Receptors, Retinoic Acid/immunology , Signal Transduction/immunology , Animals , Antigens, CD/immunology , Dose-Response Relationship, Immunologic , Integrin alpha Chains/immunology , Intestine, Small/immunology , Mice , Mice, Transgenic , Organ Specificity/immunology , Receptors, CCR/immunology , Spleen/cytology , Spleen/immunology
14.
Clin Exp Immunol ; 141(2): 279-87, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15996192

ABSTRACT

Organ-specific lymphocyte homing is dependent on the expression of tissue-specific homing receptors and selected chemokine receptors. During the effector phase of an immune response, IgA and IgG antibody-secreting cells (ASC) are differently distributed in the body. Still, B cell expression of L-selectin and the mucosal homing receptor integrin alpha4beta7 is not related to the isotype produced, but only to the site of antigen encounter. In this study, we examined if differences in chemokine responsiveness between IgA+ and IgG+ B cells could explain their different tissue localization. Circulating CD19+ B cells were isolated and their expression of IgA, IgG, and selected chemokine receptors was determined by flow cytometry. Few Ig+ cells expressed CCR2, CCR3, or CCR9, and there was no difference in the expression of these receptors between IgA+ and IgG+ cells. In contrast, CCR4, CCR5, and CXCR3 was expressed on significantly more IgG+ than IgA+ cells. The function of chemokine receptors on memory B cells and ASC was then tested in the transwell system. IgG+ memory cells migrated to a higher extent than IgA+ cells towards the CXCR3 ligand CXCL11/I-TAC, while there was only a small migration towards the CCR4 ligand CCL17/TARC and the CCR9 ligand CCL25/TECK. ASC migrated poorly to all chemokines tested. In conclusion, this study shows that IgG+ and IgA+ memory B cells have a differential expression of the Th1 associated chemokine receptor CXCR3, as well as of CCR4 and CCR5. In contrast, none of the studied chemokine receptors was preferentially expressed by IgA+ cells.


Subject(s)
B-Lymphocyte Subsets/immunology , Immunoglobulin A/blood , Immunoglobulin G/blood , Receptors, Chemokine/blood , Antibody-Producing Cells/immunology , Cells, Cultured , Chemokine CCL17 , Chemokine CXCL11 , Chemokines, CC/immunology , Chemokines, CXC/immunology , Chemotaxis, Leukocyte/immunology , Dose-Response Relationship, Immunologic , Gastric Mucosa/immunology , Humans , Immunologic Memory/immunology , Integrins/blood
15.
J Exp Med ; 192(5): 761-8, 2000 Sep 04.
Article in English | MEDLINE | ID: mdl-10974041

ABSTRACT

The immune system has evolved specialized cellular and molecular mechanisms for targeting and regulating immune responses at epithelial surfaces. Here we show that small intestinal intraepithelial lymphocytes and lamina propria lymphocytes migrate to thymus-expressed chemokine (TECK). This attraction is mediated by CC chemokine receptor (CCR)9, a chemoattractant receptor expressed at high levels by essentially all CD4(+) and CD8(+) T lymphocytes in the small intestine. Only a small subset of lymphocytes in the colon are CCR9(+), and lymphocytes from other tissues including tonsils, lung, inflamed liver, normal or inflamed skin, inflamed synovium and synovial fluid, breast milk, and seminal fluid are universally CCR9(-). TECK expression is also restricted to the small intestine: immunohistochemistry reveals that intense anti-TECK reactivity characterizes crypt epithelium in the jejunum and ileum, but not in other epithelia of the digestive tract (including stomach and colon), skin, lung, or salivary gland. These results imply a restricted role for lymphocyte CCR9 and its ligand TECK in the small intestine, and provide the first evidence for distinctive mechanisms of lymphocyte recruitment that may permit functional specialization of immune responses in different segments of the gastrointestinal tract. Selective expression of chemokines by differentiated epithelium may represent an important mechanism for targeting and specialization of immune responses.


Subject(s)
Chemokines, CC/analysis , Intestinal Mucosa/immunology , Intestine, Small/immunology , Receptors, Chemokine/analysis , Animals , Chemokines, CC/physiology , Humans , Mice , Organ Specificity , Receptors, CCR , Receptors, Chemokine/physiology , T-Lymphocytes/chemistry
16.
Curr Opin Cell Biol ; 12(5): 563-8, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10978890

ABSTRACT

The epithelia are the avascular layers of cells that cover the environment-exposed surfaces of the body. It appears that T cells localize to selected sites in or adjacent to epithelia via the selective expression of adhesion molecules and chemokine receptors on T cells. These bind to counter-receptors and to chemokines expressed by epithelial cells. Recently, there has been an advance in our understanding of the interaction of the alpha(Ebeta7) integrin with its epithelial cell ligand, E-cadherin. In addition, a new adhesion molecule has been identified on non-intestinal epithelial cells, termed lymphocyte-endothelial-epithelial-cell adhesion molecule (LEEP-CAM). Finally, there have been advances in our understanding of the role of skin- or gut-epithelia-derived chemokines in regulating activated T cell homing to these sites.


Subject(s)
Cell Communication , Epithelial Cells/metabolism , T-Lymphocytes/metabolism , Animals , Cadherins/metabolism , Cell Adhesion Molecules/metabolism , Chemokines/metabolism , Epithelial Cells/cytology , Humans , Integrins/metabolism , Membrane Glycoproteins/metabolism , Protein Binding , T-Lymphocytes/cytology
17.
Curr Biol ; 10(6): 325-8, 2000 Mar 23.
Article in English | MEDLINE | ID: mdl-10744978

ABSTRACT

HIV particles that use the chemokine receptor CXCR4 as a coreceptor for entry into cells (X4-HIV) inefficiently transmit infection across mucosal surfaces [1], despite their presence in seminal fluid and mucosal secretions from infected individuals [2] [3] [4]. In addition, although intestinal lymphocytes are susceptible to infection with either X4-HIV particles or particles that use the chemokine receptor CCR5 for viral entry (R5-HIV) during ex vivo culture [5], only systemic inoculation of R5-chimeric simian-HIV (S-HIV) results in a rapid loss of CD4(+) intestinal lymphocytes in macaques [6]. The mechanisms underlying the inefficient capacity of X4-HIV to transmit infection across mucosal surfaces and to infect intestinal lymphocytes in vivo have remained elusive. The CCR5 ligands RANTES, MIP-1alpha and MIP-1beta suppress infection by R5-HIV-1 particles via induction of CCR5 internalization, and individuals whose peripheral blood lymphocytes produce high levels of these chemokines are relatively resistant to infection [7] [8] [9]. Here, we show that the CXCR4 ligand stromal derived factor-1 (SDF-1) is constitutively expressed by mucosal epithelial cells at sites of HIV transmission and propagation. Furthermore, CXCR4 is selectively downmodulated on intestinal lymphocytes within the setting of prominent SDF-1 expression. We postulate that mucosally derived SDF-1 continuously downmodulates CXCR4 on resident HIV target cells, thereby reducing the transmission and propagation of X4-HIV at mucosal sites. Moreover, such a mechanism could contribute to the delayed emergence of X4 isolates, which predominantly occurs during the later stages of the HIV infection.


Subject(s)
Chemokines, CXC/physiology , HIV/growth & development , Intestinal Mucosa/metabolism , Chemokine CXCL12 , Chemokines, CXC/biosynthesis , Humans , Receptors, CCR5/metabolism , Receptors, CXCR4/biosynthesis , Receptors, CXCR4/genetics
18.
Eur J Immunol ; 30(3): 819-26, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10741397

ABSTRACT

To determine which chemokine receptors might be involved in T lymphocyte localization to the intestinal mucosa, we examined receptor expression on human intestinal lamina propria lymphocytes (LPL), intraepithelial lymphocytes (IEL) and CD45RO+beta7hi gut homing peripheral blood lymphocytes (PBL). Virtually all LPL and IEL expressed CXCR3 and CCR5, receptors that have been associated with Th1(Tc1)/Th0 lymphocytes, while CCR3 and CCR4, receptors associated with Th2 (Tc2)lymphocytes, CCR7, CXCR1 and CXCR2 were not expressed. CXCR3 and CCR5 receptors were functional, as LPL and IEL migrated to their respective ligands I-TAC and RANTES. In addition, most alphaEbeta7- LPL and IEL expressed high levels of CCR2. While the majority of CD45RO(-)beta7hi PBL also expressed CXCR3 and CCR5, a proportion of these cells were CXCR3- and/or CCR5- and some expressed CCR4 and/or CCR7, indicating that lymphocytes recruited to the intestinal mucosa represent a subset of these cells. In summary, our results show that LPL and IEL within the normal intestine express a specific and similar array of chemokine receptors whose ligands are constitutively expressed in the intestinal mucosa and whose expression is up-regulated during intestinal inflammation. These results support the view that CXCR3, CCR5 and CCR2 may play an important role in lymphocyte localization within the intestinal mucosa.


Subject(s)
Integrin beta Chains , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Lymphocytes/immunology , Receptors, Chemokine/metabolism , Animals , Antibodies, Monoclonal , Chemotaxis, Leukocyte , Epithelial Cells/cytology , Epithelial Cells/immunology , Humans , In Vitro Techniques , Inflammation/immunology , Inflammation/pathology , Integrins/metabolism , Leukocyte Common Antigens/metabolism , Mice , Receptors, CCR2 , Receptors, CCR5/metabolism , Receptors, CXCR3 , T-Lymphocytes/immunology
19.
J Exp Med ; 190(9): 1241-56, 1999 Nov 01.
Article in English | MEDLINE | ID: mdl-10544196

ABSTRACT

TECK (thymus-expressed chemokine), a recently described CC chemokine expressed in thymus and small intestine, was found to mediate chemotaxis of human G protein-coupled receptor GPR-9-6/L1.2 transfectants. This activity was blocked by anti-GPR-9-6 monoclonal antibody (mAb) 3C3. GPR-9-6 is expressed on a subset of memory alpha4beta7(high) intestinal trafficking CD4 and CD8 lymphocytes. In addition, all intestinal lamina propria and intraepithelial lymphocytes express GPR-9-6. In contrast, GPR-9-6 is not displayed on cutaneous lymphocyte antigen-positive (CLA(+)) memory CD4 and CD8 lymphocytes, which traffic to skin inflammatory sites, or on other systemic alpha4beta7(-)CLA(-) memory CD4/CD8 lymphocytes. The majority of thymocytes also express GPR-9-6, but natural killer cells, monocytes, eosinophils, basophils, and neutrophils are GPR-9-6 negative. Transcripts of GPR-9-6 and TECK are present in both small intestine and thymus. Importantly, the expression profile of GPR-9-6 correlates with migration to TECK of blood T lymphocytes and thymocytes. As migration of these cells is blocked by anti-GPR-9-6 mAb 3C3, we conclude that GPR-9-6 is the principal chemokine receptor for TECK. In agreement with the nomenclature rules for chemokine receptors, we propose the designation CCR-9 for GPR-9-6. The selective expression of TECK and GPR-9-6 in thymus and small intestine implies a dual role for GPR-9-6/CCR-9, both in T cell development and the mucosal immune response.


Subject(s)
Chemokines, CC/pharmacology , Chemotaxis/immunology , Intestinal Mucosa/immunology , Receptors, Chemokine/immunology , T-Lymphocytes/metabolism , Thymus Gland/immunology , Antibodies, Monoclonal , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Calcium , Cell Line , Chemokines, CC/genetics , Flow Cytometry , Gene Expression Regulation/immunology , Humans , Lymphocyte Activation/immunology , Lymphocyte Subsets/immunology , RNA, Messenger/immunology , Receptors, CCR , Receptors, Chemokine/genetics , Receptors, Lymphocyte Homing/immunology , Transfection
20.
J Immunol ; 162(11): 6641-9, 1999 Jun 01.
Article in English | MEDLINE | ID: mdl-10352281

ABSTRACT

The mucosal lymphocyte integrin alpha E(CD103)beta 7 is thought to be important for intraepithelial lymphocyte (IEL) localization or function. We cloned the murine integrin gene encoding alpha E, localized it to chromosome 11, and generated integrin alpha E-deficient mice. In alpha E-/- mice, intestinal and vaginal IEL numbers were reduced, consistent with the known binding of alpha E beta 7 to E-cadherin expressed on epithelial cells. However, it was surprising that lamina propria T lymphocyte numbers were diminished, as E-cadherin is not expressed in the lamina propria. In contrast, peribronchial, intrapulmonary, Peyer's patch, and splenic T lymphocyte numbers were not reduced in alpha E-deficient mice. Thus, alpha E beta 7 was important for generating or maintaining the gut and vaginal T lymphocytes located diffusely within the epithelium or lamina propria but not for generating the gut-associated organized lymphoid tissues. Finally, the impact of alpha E deficiency upon intestinal IEL numbers was greater at 3-4 wk of life than in younger animals, and affected the TCR alpha beta+ CD8+ T cells more than the gamma delta T cells or the TCR alpha beta+ CD4+CD8- population. These findings suggest that alpha E beta 7 is involved in the expansion/recruitment of TCR alpha beta+ CD8+ IEL following microbial colonization. Integrin alpha E-deficient mice will provide an important tool for studying the role of alpha E beta 7 and of alpha E beta 7-expressing mucosal T lymphocytes in vivo.


Subject(s)
Antigens, CD/genetics , Integrin alpha Chains , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Lymphopenia/genetics , Lymphopenia/immunology , T-Lymphocyte Subsets/immunology , Animals , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Chromosome Mapping , Cloning, Molecular , Crosses, Genetic , Epithelial Cells/immunology , Epithelial Cells/pathology , Female , Hyaluronan Receptors/biosynthesis , Intestinal Mucosa/metabolism , Lymphocyte Count , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Muridae , Peyer's Patches/cytology , Peyer's Patches/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Spleen/cytology , Spleen/pathology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , Vagina/immunology , Vagina/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...