Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 267
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 17(9): 1075-83, 2016 09.
Article in English | MEDLINE | ID: mdl-27400149

ABSTRACT

Adhesion and migration of T cells are controlled by chemokines and by adhesion molecules, especially integrins, and have critical roles in the normal physiological function of T lymphocytes. Using an RNA-mediated interference screen, we identified the WNK1 kinase as a regulator of both integrin-mediated adhesion and T cell migration. We found that WNK1 is a negative regulator of integrin-mediated adhesion, whereas it acts as a positive regulator of migration via the kinases OXSR1 and STK39 and the ion co-transporter SLC12A2. WNK1-deficient T cells home less efficiently to lymphoid organs and migrate more slowly through them. Our results reveal that a pathway previously known only to regulate salt homeostasis in the kidney functions to balance T cell adhesion and migration.


Subject(s)
Cell Adhesion/genetics , Cell Movement/genetics , Minor Histocompatibility Antigens/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Lymphocyte Homing/metabolism , T-Lymphocytes/physiology , Animals , Cells, Cultured , Homeostasis , Ion Transport , Kidney/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Minor Histocompatibility Antigens/genetics , Protein Serine-Threonine Kinases/genetics , RNA Interference , Receptors, Lymphocyte Homing/genetics , Solute Carrier Family 12, Member 2/metabolism , WNK Lysine-Deficient Protein Kinase 1
2.
Nat Immunol ; 12(2): 178-85, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21217760

ABSTRACT

Type 2 helper T cells (T(H)2) are critically involved in allergies and asthma. Here we demonstrate that extracellular matrix protein-1 (ECM1) is highly and selectively expressed in T(H)2 cells. ECM1 deficiency caused impaired T(H)2 responses and reduced allergic airway inflammation in vivo. Functional analysis demonstrated that although the T(H)2 polarization of ECM1-deficient cells was unimpaired, these cells had a defect in migration and were retained in peripheral lymphoid organs. This was associated with reduced expression of KLF2 and S1P(1). We also found that ECM1 could directly bind the interleukin-2 (IL-2) receptor to inhibit IL-2 signaling and activate S1P(1) expression. Our data identify a previously unknown function of ECM1 in regulating T(H)2 cell migration through control of KLF2 and S1P(1) expression.


Subject(s)
Extracellular Matrix Proteins/metabolism , Hypersensitivity/immunology , Nerve Tissue Proteins/metabolism , Receptors, Lymphocyte Homing/metabolism , Th2 Cells/metabolism , Adoptive Transfer , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Movement/genetics , Cell Movement/immunology , Cells, Cultured , Disease Models, Animal , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/immunology , Gene Expression Regulation/immunology , Humans , Lymph Nodes/pathology , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/immunology , Receptors, Lymphocyte Homing/genetics , Receptors, Lymphocyte Homing/immunology , Signal Transduction/immunology , Th2 Cells/immunology , Th2 Cells/pathology , Transgenes/genetics
3.
J Immunol ; 207(7): 1763-1775, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34470859

ABSTRACT

Regulatory T cells (Tregs) reside in nonlymphoid tissues where they carry out unique functions. The molecular mechanisms responsible for Treg accumulation and maintenance in these tissues are relatively unknown. Using an unbiased discovery approach, we identified LAYN (layilin), a C-type lectin-like receptor, to be preferentially and highly expressed on a subset of activated Tregs in healthy and diseased human skin. Expression of layilin on Tregs was induced by TCR-mediated activation in the presence of IL-2 or TGF-ß. Mice with a conditional deletion of layilin in Tregs had reduced accumulation of these cells in tumors. However, these animals somewhat paradoxically had enhanced immune regulation in the tumor microenvironment, resulting in increased tumor growth. Mechanistically, layilin expression on Tregs had a minimal effect on their activation and suppressive capacity in vitro. However, expression of this molecule resulted in a cumulative anchoring effect on Treg dynamic motility in vivo. Taken together, our results suggest a model whereby layilin facilitates Treg adhesion in skin and, in doing so, limits their suppressive capacity. These findings uncover a unique mechanism whereby reduced Treg motility acts to limit immune regulation in nonlymphoid organs and may help guide strategies to exploit this phenomenon for therapeutic benefit.


Subject(s)
Carrier Proteins/metabolism , Membrane Glycoproteins/metabolism , Receptors, Lymphocyte Homing/metabolism , Skin/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Carrier Proteins/genetics , Cell Movement , Cells, Cultured , Humans , Immune Tolerance , Lymphocyte Activation , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Immunological , Organ Specificity , Receptors, Lymphocyte Homing/genetics , Transforming Growth Factor beta/metabolism
4.
Exp Dermatol ; 30(1): 84-91, 2021 01.
Article in English | MEDLINE | ID: mdl-33010063

ABSTRACT

Recently, increasing attention has been paid to senescence-associated secretory phenotype (SASP), a phenomenon that senescent cells secrete molecules such as inflammatory cytokines and matrix metalloproteinases (MMPs), due to its noxious effects on the surrounding tissue. Senescent cells in the blood and liver are known to be properly depleted by macrophages. In the dermis, accumulation of senescent cells has been reported and is thought to be involved with skin ageing. In this study, to elucidate the clearance mechanism of senescent cells in the dermis, we focused on macrophage functions. Our co-culture experiments of senescent fibroblasts and macrophages revealed a two-step clearance mechanism: first, TNF-α secreted from macrophages induces apoptosis in senescent fibroblasts, and then, dead cells are phagocytosed by macrophages. Furthermore, it was suggested that SASP factors suppress both of the two steps of the senescent cell clearance by macrophages. From these findings, normally senescent cells in the dermis are thought to be removed by macrophages, but when senescent cells are excessively accumulated owing to oxidative stress, ultraviolet (UV) ray or other reasons, SASP was suggested to suppress the macrophage-dependent clearance functions and thereby cause further accumulation of senescent cells.


Subject(s)
Fibroblasts/physiology , Macrophages/physiology , Senescence-Associated Secretory Phenotype , Adult , Aged , Antigens, CD/genetics , Antigens, Differentiation, Myelomonocytic/genetics , Apoptosis/drug effects , Cell Adhesion Molecules, Neuronal/genetics , Cell Line , Cell Polarity , Cell Survival/drug effects , Coculture Techniques , Culture Media, Conditioned/pharmacology , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Dermis/cytology , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression/drug effects , Humans , Immunohistochemistry , Infliximab/pharmacology , Male , Phagocytosis , RNA/metabolism , Receptors, CCR7/genetics , Receptors, Cell Surface/genetics , Receptors, Lymphocyte Homing/genetics , S100 Calcium-Binding Protein A4/metabolism , Tumor Necrosis Factor Inhibitors/pharmacology , Young Adult
5.
Br J Cancer ; 123(4): 501-509, 2020 08.
Article in English | MEDLINE | ID: mdl-32595212

ABSTRACT

Clever-1 (also known as Stabilin-1 and FEEL-1) is a scavenger receptor expressed on lymphatic endothelial cells, sinusoidal endothelial cells and immunosuppressive monocytes and macrophages. Its role in cancer growth and spread first became evident in Stab1-/- knockout mice, which have smaller primary tumours and metastases. Subsequent studies in mice and humans have shown that immunotherapeutic blockade of Clever-1 can activate T-cell responses, and that this response is mainly mediated by a phenotypic change in macrophages and monocytes from immunosuppressive to pro-inflammatory following Clever-1 inhibition. Analyses of human cancer cohorts have revealed marked associations between the number of Clever-1-positive macrophages and patient outcome. As hardly any reports to date have addressed the role of Clever-1 in immunotherapy resistance and T-cell dysfunction, we performed data mining using several published cancer cohorts, and observed a remarkable correlation between Clever-1 positivity and resistance to immune checkpoint therapies. This result provides impetus and potential for the ongoing clinical trial targeting Clever-1 in solid tumours, which has so far shown a shift towards immune activation when a particular epitope of Clever-1 is blocked.


Subject(s)
Cell Adhesion Molecules, Neuronal/genetics , Drug Resistance, Neoplasm , Neoplasms/genetics , Receptors, Lymphocyte Homing/genetics , Animals , Cell Adhesion Molecules, Neuronal/metabolism , Disease Progression , Humans , Immunotherapy , Macrophages/metabolism , Mice , Mice, Knockout , Neoplasms/drug therapy , Neoplasms/immunology , Receptors, Lymphocyte Homing/metabolism , T-Lymphocytes/metabolism
6.
Eur J Immunol ; 47(2): 244-250, 2017 02.
Article in English | MEDLINE | ID: mdl-27861803

ABSTRACT

The salivary glands (SGs) of virus-immune mice contain substantial numbers of tissue-resident memory CD8+ T cells (TRM cells) that can provide immunity to local infections. Integrins regulate entry of activated T cells into nonlymphoid tissues but the molecules that mediate migration of virus-specific CD8+ T cells to the SGs have not yet been defined. Here, we found that polyinosinic-polycytidylic acid (poly(I:C)) strongly promoted the accumulation of P14 TCR-transgenic CD8+ TRM cells in SGs in an α4 ß1 integrin-dependent manner. After infection with lymphocytic choriomeningitis virus, accumulation of P14 TRM cells in SGs and intestine but not in kidney was also α4 integrin dependent. Blockade of α4 ß7 by monoclonal antibodies (mAbs) inhibited lymphocytic choriomeningitis virus-induced accumulation of P14 TRM cells in the intestine but not in SGs. In conclusion, our data reveal that α4 ß1 integrin mediates CD8+ TRM accumulation in SGs and that poly(I:C) can be used to direct activated CD8+ T cells to this organ.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Integrin alpha4beta1/metabolism , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Receptors, Lymphocyte Homing/metabolism , Salivary Glands/immunology , T-Lymphocyte Subsets/immunology , Adoptive Transfer , Animals , Cell Movement/genetics , Cells, Cultured , Immunologic Memory , Integrin alpha4beta1/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Poly I-C/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, CXCR3/genetics , Receptors, CXCR3/metabolism , Receptors, Lymphocyte Homing/genetics
7.
J Immunol ; 196(1): 115-23, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26608916

ABSTRACT

In this study, we analyzed the putative functions of stabilin-1 in blood monocytes. Microarray analysis revealed downregulation of several proinflammatory genes in the stabilin-1(high) monocytes when compared with stabilin-1(low) monocytes. When cocultured with stabilin-1(high) monocytes, IFN-γ synthesis by T cells was diminished in Ag-recall assays. Knockdown of stabilin-1 in monocytes increased the synthesis of several proinflammatory molecules, including TNF-α, and supported high IFN-γ and low IL-4 and IL-5 production by T cells in Ag-specific stimulation assays. Anti-stabilin-1 Ab treatment also led to increased IFN-γ synthesis in the recall assays. In clinical settings, the expression of stabilin-1 was diminished on blood monocytes and tissue macrophages under proinflammatory conditions. These data define stabilin-1 as a new immunosuppressive molecule and suggest that stabilin-1(high) monocytes may dampen proinflammatory reactions in vivo.


Subject(s)
Cell Adhesion Molecules, Neuronal/genetics , Inflammation/immunology , Lymphocyte Activation/immunology , Monocytes/immunology , Receptors, Lymphocyte Homing/genetics , Th1 Cells/immunology , Base Sequence , Cells, Cultured , Down-Regulation , Humans , Interferon-gamma/biosynthesis , Interleukin-4/biosynthesis , Interleukin-5/biosynthesis , Macrophages/immunology , Oligonucleotide Array Sequence Analysis , RNA Interference , RNA, Small Interfering , Sequence Analysis, RNA , Tumor Necrosis Factor-alpha/biosynthesis
8.
Invest New Drugs ; 35(5): 566-575, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28631095

ABSTRACT

The availability of effective medication for the treatment of refractory or recurrent neuroblastoma remains limited. This study sought to investigate the effects of increased decorin (DCN) expression on the intratumoral uptake of nab-paclitaxel as a potential novel approach to NB. Correlation between the clinical characteristics of neuroblastoma and the expression of DCN, secreted protein acidic and rich in cysteine (SPARC) and stabilin-1 was evaluated. The anticancer effect of recombinant adeno-associated virus-DCN (rAAV-DCN) was assessed in vivo and in vitro. And the effect of rAAV-DCN on the intratumoral uptake of paclitaxel was also studied in neuroblastoma-grafted nude mice. Overall, 12.5%, 17.7%, and 71.9% of the tumors stained positive for DCN, SPARC and stabilin-1 respectively and correlated to age, stage and N-MYC status in 96 children and adolescents with neuroblastoma. Transfected neuroblastoma cells stably expressed DCN, with in vivo and in vitro studies demonstrating rAAV-DCN sensitized the anticancer effect of nab-paclitaxel. Systemic rAAV-DCN in neuroblastoma-grafted nude mice inhibited stabilin-1, up-regulated SPARC, and increased the intratumoral uptake of paclitaxel. Macrophage depletion or anti-stabilin-1 monoclonal antibody increased the intratumoral uptake of nab-paclitaxel and its anticancer effects to a degree comparable to that achieved by systemic rAAV-DCN. The systemic administration of rAAV-DCN up-regulates DCN in neuroblastoma and accelerates the intratumoral uptake of nab-paclitaxel by inhibiting stabilin-1 mediated SPARC degradation.


Subject(s)
Albumins/pharmacology , Cell Adhesion Molecules, Neuronal/genetics , Decorin/genetics , Dependovirus/genetics , Neuroblastoma/drug therapy , Neuroblastoma/genetics , Paclitaxel/pharmacology , Receptors, Lymphocyte Homing/genetics , Up-Regulation/genetics , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Child , Child, Preschool , Female , Humans , Infant , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Mosquito Vectors/genetics , Transfection/methods , Up-Regulation/drug effects
9.
J Immunol ; 195(12): 5787-94, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26561545

ABSTRACT

IL-1R-associated kinase (IRAK) 1 is an important component of the IL-1R and TLR signaling pathways, which influence Th cell differentiation. In this study, we show that IRAK1 promotes Th17 development by mediating IL-1ß-induced upregulation of IL-23R and subsequent STAT3 phosphorylation, thus enabling sustained IL-17 production. Moreover, we show that IRAK1 signaling fosters Th1 differentiation by mediating T-bet induction and counteracts regulatory T cell generation. Cotransfer experiments revealed that Irak1-deficient CD4(+) T cells have a cell-intrinsic defect in generating Th1 and Th17 cells under inflammatory conditions in spleen, mesenteric lymph nodes, and colon tissue. Furthermore, IRAK1 expression in T cells was shown to be essential for T cell accumulation in the inflamed intestine and mesenteric lymph nodes. Transcriptome analysis ex vivo revealed that IRAK1 promotes T cell activation and induction of gut-homing molecules in a cell-intrinsic manner. Accordingly, Irak1-deficient T cells failed to upregulate surface expression of α4ß7 integrin after transfer into Rag1(-/-) mice, and their ability to induce colitis was greatly impaired. Lack of IRAK1 in recipient mice provided additional protection from colitis. Therefore, IRAK1 plays an important role in intestinal inflammation by mediating T cell activation, differentiation, and accumulation in the gut. Thus, IRAK1 is a promising novel target for therapy of inflammatory bowel diseases.


Subject(s)
Colitis/immunology , Inflammatory Bowel Diseases/immunology , Interleukin-1 Receptor-Associated Kinases/metabolism , Mice, Inbred C57BL/metabolism , Receptors, Lymphocyte Homing/metabolism , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Adoptive Transfer , Animals , Cell Differentiation , Cell Movement/genetics , Cytokines/immunology , Cytokines/metabolism , Inflammation/immunology , Inflammatory Bowel Diseases/drug therapy , Integrins/metabolism , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-17/immunology , Intestines/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL/genetics , Mice, Knockout , Receptors, Lymphocyte Homing/genetics , Signal Transduction/immunology , T-Lymphocytes, Regulatory/transplantation , Th17 Cells/transplantation
10.
J Allergy Clin Immunol ; 133(6): 1676-85.e5, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24373354

ABSTRACT

BACKGROUND: We previously reported abnormalities in circulating B cells in patients with chronic granulomatous disease (CGD) and those with HIV infection. Gastrointestinal complications are common to both diseases and likely involve perturbation of immune cells, including plasma cells (PCs). IgA is the most abundant immunoglobulin in the human body, with roles in protection and maintenance of intestinal homeostasis. IgA is produced primarily by PCs residing in mucosal tissues that are also thought to circulate in the blood. OBJECTIVE: We sought to characterize and compare PCs in patients with infectious (HIV) and noninfectious (CGD and Crohn disease) diseases that have been associated with intestinal inflammation. METHODS: Phenotypic and transcriptional analyses were performed on cells isolated from the blood and colon. RESULTS: IgA-secreting CCR10-expressing PCs predominated in the guts of healthy subjects, whereas in patients with HIV, CGD, and Crohn disease, there was a significant increase in the proportion of IgG-secreting PCs. Where intestinal inflammation was present, IgG-secreting PCs expressed reduced levels of CCR10 and increased levels of CXCR4. The intensity of CXCR4 expression correlated with the frequency of IgG-expressing PCs and the frequency of CXCR4(+)/IgG(+) PCs was associated with the severity of intestinal inflammatory disease yet distinct from PCs and plasmablasts circulating in the blood. CONCLUSIONS: These findings suggest that regardless of the underlying disease, the presence of CXCR4(+)/IgG(+) PCs in the gut is a strong yet localized indicator of intestinal inflammation. Furthermore, our findings suggest that CXCR4(+)/IgG(+) PCs might play a role in immune cell homeostasis during inflammatory processes of the gut.


Subject(s)
Gastroenteritis/immunology , Gastroenteritis/metabolism , Immunoglobulin G/metabolism , Plasma Cells/immunology , Plasma Cells/metabolism , Receptors, CXCR4/metabolism , Adult , Biopsy , Crohn Disease/immunology , Crohn Disease/metabolism , Female , Gastroenteritis/genetics , Granulomatous Disease, Chronic/immunology , Granulomatous Disease, Chronic/metabolism , HIV Infections/immunology , HIV Infections/metabolism , Humans , Immunoglobulin Isotypes/immunology , Immunoglobulin Isotypes/metabolism , Immunophenotyping , Male , Middle Aged , Mucous Membrane/immunology , Mucous Membrane/metabolism , Receptors, Lymphocyte Homing/genetics , Receptors, Lymphocyte Homing/metabolism , Young Adult
11.
Int J Obes (Lond) ; 38(1): 120-5, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23670221

ABSTRACT

OBJECTIVE: We hypothesized that genes within recently identified loci associated with waist-hip ratio (WHR) exhibit fat depot-specific mRNA expression, which correlates with obesity-related traits. METHODS: Adipose tissue (AT) mRNA expression of 6 genes (TBX15/WARS2, STAB1, PIGC, ZNRF3 and GRB14) within these loci showing coincident cis-expression quantitative trait loci was measured in 222 paired samples of human visceral (vis) and subcutaneous (sc) AT. The relationship of mRNA expression levels with obesity-related quantitative traits was assessed by Pearson's correlation analyses. Multivariate linear relationships were assessed by generalized linear regression models. RESULTS: Whereas only PIGC, ZNFR3 and STAB1 mRNA expression in sc AT correlated nominally with WHR (P<0.05, adjusted for age and sex), mRNA expression of all studied genes in at least one of the fat depots correlated significantly with vis and/or sc fat area (P ranging from 0.05 to 4.0 × 10(6), adjusted for age and sex). Consistently, the transcript levels of WARS, PIGC and GRB14 were nominally associated with body mass index (BMI) (P ranging from 0.02 to 9.2 × 10(5), adjusted for age and sex). Moreover, independent of sex, obesity and diabetes status, differential expression between vis and sc AT was observed for all tested genes (P<0.01). Finally, the rs10195252 T-allele was nominally associated with increased GRB14 sc mRNA expression (P=0.025 after adjusting for age, sex and BMI). CONCLUSIONS: Our data including the inter-depot variability of mRNA expression suggests that genes within the WHR-associated loci might be involved in the regulation of fat distribution.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adipose Tissue/metabolism , Body Composition , Body Fat Distribution , Cell Adhesion Molecules, Neuronal/metabolism , Hexosyltransferases/metabolism , Membrane Proteins/metabolism , Obesity/metabolism , Receptors, Lymphocyte Homing/metabolism , Subcutaneous Fat/metabolism , T-Box Domain Proteins/metabolism , Tryptophan-tRNA Ligase/metabolism , Ubiquitin-Protein Ligases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adult , Body Mass Index , Cell Adhesion Molecules, Neuronal/genetics , Female , Genotype , Hexosyltransferases/genetics , Humans , Male , Membrane Proteins/genetics , Middle Aged , Phenotype , Polymorphism, Single Nucleotide , RNA, Messenger/metabolism , Receptors, Lymphocyte Homing/genetics , T-Box Domain Proteins/genetics , Tryptophan-tRNA Ligase/genetics , Ubiquitin-Protein Ligases/genetics , Waist-Hip Ratio
12.
J Biol Chem ; 287(25): 20774-83, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22547069

ABSTRACT

As one of the most widely used drugs worldwide, heparin is an essential anticoagulant required for surgery, dialysis, treatment of thrombosis, cancer, and general circulatory management. Stabilin-2 is a scavenger clearance receptor with high expression in the sinusoidal endothelium of liver. It is believed that Stabilin-2 is the primary receptor for the clearance of unfractionated and low molecular weight heparins in the liver. Here, we identify the modifications and length of the heparin polymer that are required for binding and endocytosis by both human Stabilin receptors: Stabilin-2 and its homolog Stabilin-1 (also found in liver endothelium). Using enzymatically synthesized (35)S-labeled heparan sulfate oligomers, we identified that sulfation of the 3-OH position of N-sulfated glucosamine (GlcNS) is the most beneficial modification for binding and endocytosis via both Stabilin receptors. In addition, our data suggest that a decasaccharide is the minimal size for binding to the Stabilin receptors. These findings define the physical parameters of the heparin structure required for efficient clearance from blood circulation. These results will also aid in the design of synthetic heparins with desired clearance rates.


Subject(s)
Anticoagulants , Cell Adhesion Molecules, Neuronal/metabolism , Heparin , Receptors, Lymphocyte Homing/metabolism , Animals , Anticoagulants/chemical synthesis , Anticoagulants/chemistry , Anticoagulants/metabolism , Anticoagulants/pharmacology , Carbohydrate Conformation , Cell Adhesion Molecules, Neuronal/genetics , Cell Line , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Heparin/chemical synthesis , Heparin/chemistry , Heparin/metabolism , Heparin/pharmacology , Humans , Liver/cytology , Liver/metabolism , Mice , Mice, Inbred BALB C , Receptors, Lymphocyte Homing/genetics
13.
J Immunol ; 187(1): 141-50, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21646294

ABSTRACT

Gut-associated dendritic cells (DC) synthesize all-trans retinoic acid, which is required for inducing gut-tropic lymphocytes. Gut-associated DC from MyD88(-/-) mice, which lack most TLR signals, expressed low levels of retinal dehydrogenases (critical enzymes for all-trans retinoic acid biosynthesis) and were significantly impaired in their ability to induce gut-homing T cells. Pretreatment of extraintestinal DC with a TLR1/2 agonist was sufficient to induce retinal dehydrogenases and to confer these DC with the capacity to induce gut-homing lymphocytes via a mechanism dependent on MyD88 and JNK/MAPK. Moreover, gut-associated DC from TLR2(-/-) mice, or from mice in which JNK was pharmacologically blocked, were impaired in their education to imprint gut-homing T cells, which correlated with a decreased induction of gut-tropic T cells in TLR2(-/-) mice upon immunization. Thus, MyD88-dependent TLR2 signals are necessary and sufficient to educate DC with gut-specific imprinting properties and contribute in vivo to the generation of gut-tropic T cells.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Genomic Imprinting/immunology , Intestinal Mucosa/immunology , Myeloid Differentiation Factor 88/physiology , Signal Transduction/immunology , Toll-Like Receptor 1/physiology , Toll-Like Receptor 2/physiology , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Line, Tumor , Coculture Techniques , Dendritic Cells/cytology , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , Radiation Chimera , Receptors, Lymphocyte Homing/deficiency , Receptors, Lymphocyte Homing/genetics , Receptors, Lymphocyte Homing/physiology , Signal Transduction/genetics
14.
J Immunol ; 186(6): 3364-72, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21296980

ABSTRACT

The chemokine receptor CCR7 represents an important determinant for circulating lymphocytes to enter lymph nodes (LN) via high endothelial venules. High endothelial venules also represent the major site of entry for plasmacytoid dendritic cells (pDC). In the steady-state, murine pDC have been suggested to home to LN engaging the chemokine receptors CXCR3, CXCR4, and CCR5, whereas responsiveness to CCR7 ligands is thought to be acquired only upon activation. In this study, we show that already resting pDC express minute amounts of CCR7 that suffice to trigger migration to CCL19/CCL21 in vitro. Upon activation with TLR ligands, CCR7 levels on pDC are strongly increased. Notably, CCR7-deficient mice display substantially reduced pDC counts in LN but not in bone marrow and spleen. Adoptive cell transfer experiments revealed that under both steady-state as well as inflammatory conditions, the homing of CCR7-deficient pDC is severely impaired, indicating that the reduced cell counts of naive pDC observed in CCR7(-/-) mice reflect an intrinsic homing defect of pDC. Together, these observations provide strong evidence that similar to naive lymphocytes, nonstimulated pDC exploit CCR7 to gain entry into LN. This adds to the repertoire of chemokine receptors permitting them to enter diverse tissues.


Subject(s)
Cell Movement/immunology , Dendritic Cells/immunology , Inflammation Mediators/physiology , Lymph Nodes/cytology , Lymph Nodes/immunology , Receptors, CCR7/physiology , Resting Phase, Cell Cycle/immunology , Adoptive Transfer , Animals , Cell Movement/genetics , Dendritic Cells/pathology , Dendritic Cells/transplantation , Inflammation Mediators/metabolism , Lymph Nodes/pathology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Receptors, CCR7/biosynthesis , Receptors, CCR7/deficiency , Receptors, Lymphocyte Homing/deficiency , Receptors, Lymphocyte Homing/genetics , Receptors, Lymphocyte Homing/physiology , Resting Phase, Cell Cycle/genetics
15.
Eur J Immunol ; 41(7): 2052-63, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21480214

ABSTRACT

Stabilin-1/common lymphatic endothelial and vascular endothelial receptor-1 (CLEVER-1) is a multidomain protein present in lymphatic and vascular endothelial cells and type 2 immunosuppressive macrophages. In adults, stabilin-1/CLEVER-1 is a scavenging receptor and an adhesion molecule, but much less is known about its role during development. Here, we studied the expression and functions of macrophage stabilin-1/CLEVER-1 in human placenta and during human ontogeny. Using newly generated mAbs, we found that stabilin-1/CLEVER-1 is expressed on virtually all macrophages in term placenta, both in the decidua and in the placental villi. Placental stabilin-1/CLEVER-1 was involved in the scavenging of Ac-LDL (acetylated low density lipoprotein) and in the uptake of fluorescently labeled model antigen OVA. siRNA-mediated suppression of stabilin-1/CLEVER-1 altered the cytokine profile produced by placental macrophages. Stabilin-1/CLEVER-1 on placental macrophages mediated their adhesion to placental vessels and supported their transmigration through vascular endothelium. Finally, we found that stabilin-1/CLEVER-1 is induced very early in fetal macrophages, high endothelial venules, and lymphatic vessels in multiple lymphatic organs. Together, these data suggest that macrophage stabilin-1/CLEVER-1 can potentially regulate leukocyte migration and scavenging during the development of the placenta and fetus.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Cell Adhesion Molecules , Cell Adhesion , Macrophages/cytology , Macrophages/metabolism , Placenta/cytology , Receptors, Lymphocyte Homing/metabolism , Antibodies, Monoclonal , Cell Adhesion Molecules, Neuronal/genetics , Cell Movement , Cells, Cultured , Cytokines/biosynthesis , Endothelial Cells/metabolism , Female , Fetal Development , Flow Cytometry , Humans , Immunoblotting , Infant , Leukocytes/physiology , Lipoproteins, LDL/metabolism , Placenta/blood supply , Pregnancy , RNA Interference , RNA, Small Interfering , Receptors, Lymphocyte Homing/genetics , Transendothelial and Transepithelial Migration
16.
J Immunol ; 184(10): 5519-26, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20400707

ABSTRACT

Th17 cells are major effector T cells in the intestine, but the regulation of their tissue tropism within the gut is poorly understood. We investigated the roles of vitamin A and retinoic acid in generation of inflammatory Th17 cells with distinct tissue tropisms within the intestine. We found that Th17 cells with distinct tissue tropisms and pathogenic activities are generated depending on the available concentration of retinoic acid (RA). In contrast to the widespread perception that RA would suppress the generation of Th17 cells, we provide evidence that RA is actually required for generation of Th17 cells with specific tissue tropisms within the gut. Th17 cells induced at suboptimal serum concentrations of RA migrated and induced moderate inflammation mainly in the large intestine, whereas the Th17 cells induced with optimal levels of exogenous RA (approximately 10 nM) migrated to the small intestine and induced more severe inflammation. The Th17 cells, induced in the presence or absence of RA, differentially expressed the trafficking receptors CCR9 and alpha4beta7. CCR9 is required for Th17 cell migration to the small intestine, whereas alpha4beta7 is required for the migration of Th17 cells throughout the whole intestine. Our results identified RA as a major signal that regulates the generation of gut Th17 cells with distinct capacities in migration and inflammatory activities. The results indicate also that specific gut tropism of Th17 cells is determined by the combination of trafficking receptors regulated by the RA signal.


Subject(s)
Chemotaxis, Leukocyte/immunology , Interleukin-17/biosynthesis , Intestine, Small/immunology , Intestine, Small/pathology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/pathology , Tretinoin/physiology , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Chemotaxis, Leukocyte/genetics , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Interleukin-17/physiology , Intestine, Small/metabolism , Lymphocyte Count , Mice , Mice, Inbred AKR , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Lymphocyte Homing/biosynthesis , Receptors, Lymphocyte Homing/genetics , Receptors, Lymphocyte Homing/physiology , Severity of Illness Index , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Helper-Inducer/transplantation
17.
Biochim Biophys Acta ; 1803(5): 553-63, 2010 May.
Article in English | MEDLINE | ID: mdl-20226821

ABSTRACT

The scavenger receptor FEEL-1/stabilin-1 is known as the marker of alternatively activated macrophage and sinusoidal endothelial cell. FEEL-1/stabilin-1 is a multifunctional transmembrane glycoprotein that is implicated in bacterial infection, diabetes, atherosclerosis, wound healing, and innate immunity. In the current study, we have identified the phox-homology domain containing protein SNX17 as a novel interaction partner of FEEL-1/stabilin-1 in endothelial cells. SNX17 directly interacts with FEEL-1/stabilin-1 and regulates its trafficking. Studies using the cytoplasmic domain of truncated or mutant FEEL-1/stabilin-1 suggest that the NPxF motif of the FEEL-1/stabilin-1 cytoplasmic tail is required for its interaction with SNX17. By transfecting cells with small interfering RNA targeting SNX17, total cellular FEEL-1/stabilin-1 expression and FEEL-1/stabilin-1-mediated ligand uptake were significantly decreased due to the enhancement of FEEL-1/stabilin-1 protein degradation. Our results identify SNX17 as a novel interaction partner of FEEL-1/stabilin-1 in endothelial cells.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Cell Membrane/metabolism , Endothelium, Vascular/metabolism , Receptors, Lymphocyte Homing/metabolism , Umbilical Veins/metabolism , Vesicular Transport Proteins/metabolism , Amino Acid Sequence , Cell Adhesion Molecules, Neuronal/genetics , Cells, Cultured , Endothelium, Vascular/cytology , Gene Library , Humans , Molecular Sequence Data , Protein Structure, Tertiary , RNA, Small Interfering/pharmacology , Receptors, Lymphocyte Homing/genetics , Sequence Homology, Amino Acid , Sorting Nexins , Two-Hybrid System Techniques , Umbilical Veins/cytology , Vesicular Transport Proteins/antagonists & inhibitors , Vesicular Transport Proteins/genetics
18.
J Cell Biochem ; 112(4): 1003-8, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21308731

ABSTRACT

SPARC (osteonectin/BM-40), a secreted matricellular protein that promotes cellular deadhesion and motility in wound healing, carcinogenesis, and inflammation, binds to the scavenger receptor stabilin-1 in alternatively activated macrophages and undergoes endocytosis and clearance from the extracellular space. Both SPARC and stabilin-1 are expressed by endothelial cells during inflammation, but their interaction in this context is unknown. We have identified a binding site on SPARC for stabilin-1 by a solid-state peptide array coupled with a modified enzyme-linked immunosorbent assay. A monoclonal antibody that recognizes the identified binding site was also characterized that could be an inhibitor for the SPARC-stabilin-1 interaction in macrophages or endothelial cells.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Epitopes/metabolism , Osteonectin/metabolism , Receptors, Lymphocyte Homing/metabolism , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Binding Sites/genetics , Cell Adhesion Molecules, Neuronal/genetics , Cell Line , Enzyme-Linked Immunosorbent Assay , Epitopes/chemistry , Epitopes/immunology , Humans , Models, Molecular , Osteonectin/chemistry , Osteonectin/genetics , Protein Array Analysis , Protein Binding , Protein Structure, Tertiary , Receptors, Lymphocyte Homing/genetics , Receptors, Scavenger/genetics , Receptors, Scavenger/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Spodoptera
19.
Eur J Immunol ; 40(9): 2539-48, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20690178

ABSTRACT

Treg are endowed with immunosuppressive activities and have been proposed as promising targets for the therapy of autoimmune diseases. As the suppressive capacity of Treg depends on their migration into the affected tissues, we tested here whether modulation of Treg homing would enhance their capacity to suppress inflammation in mouse models of inflammatory bowel disease. Retinoic acid (RA) was used to induce the gut-specific homing receptor alpha(4)beta(7) efficiently and, to some extent, the chemokine receptor CCR9 on in vitro expanded Treg. Upon transfer, RA-treated Treg were indeed more potent suppressors in an acute, small intestinal inflammation model, compared with Treg stimulated without RA. By contrast, the efficacy of Treg to resolve an established, chronic inflammation of the colon in the transfer colitis model was not affected by RA-treatment. In the latter model, a rapid loss of RA-induced alpha(4)beta(7) expression and de novo induction of alpha(4)beta(7) on previously negative cells was observed on transferred Treg, which implies that Treg acquire gut-seeking properties in vivo under inflammatory and/or lymphopenic conditions. Together, our data show that the induction of appropriate homing properties prior to transfer increases the protective potential of adoptively transferred Treg in acute, but not in chronic, inflammatory disorders of the gut.


Subject(s)
Colitis/immunology , Integrins/biosynthesis , Intestine, Small/immunology , Receptors, CCR/biosynthesis , Receptors, Lymphocyte Homing/biosynthesis , T-Lymphocytes, Regulatory/metabolism , Acute Disease , Adoptive Transfer , Animals , Cells, Cultured , Chronic Disease , Colitis/pathology , Disease Models, Animal , Homeodomain Proteins/genetics , Humans , Immunosuppression Therapy , Inflammation , Inflammatory Bowel Diseases/immunology , Integrins/genetics , Integrins/immunology , Intestine, Small/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, CCR/genetics , Receptors, CCR/immunology , Receptors, Lymphocyte Homing/genetics , Receptors, Lymphocyte Homing/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Tretinoin/pharmacology
20.
Curr Opin Cell Biol ; 6(5): 726-33, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7530464

ABSTRACT

Hyaluronate (HA) is an abundant component of extracellular matrix that is believed to be crucial in many cellular processes, including tissue remodeling, the creation of cell-free spaces, inflammation and tumorigenesis. Although several well characterized proteins within the extracellular matrix associate with HA, it is now clear that cells can also bind and respond to HA directly, via cell-surface HA-binding proteins. The cDNAs coding for two families of such proteins, CD44 and RHAMM, have been cloned and characterized. These proteins have been implicated in a number of physiological processes, including cell migration, lymphocyte activation and tumor progression. Although many of these processes depend on an association with HA, some are apparently HA-independent, suggesting that other ligands for these receptors may be involved.


Subject(s)
Carrier Proteins/physiology , Hyaluronic Acid/physiology , Receptors, Cell Surface/physiology , Receptors, Lymphocyte Homing/physiology , Animals , Carrier Proteins/genetics , Carrier Proteins/immunology , Cell Adhesion/physiology , Cell Movement/physiology , Genetic Variation , Growth/physiology , Humans , Hyaluronan Receptors , Lymphocyte Activation , Neoplasm Metastasis , Neoplasms/physiopathology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Receptors, Lymphocyte Homing/genetics , Receptors, Lymphocyte Homing/immunology
SELECTION OF CITATIONS
SEARCH DETAIL