Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Immunol ; 210(11): 1700-1716, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37093875

ABSTRACT

Th17 cells have been investigated in mice primarily for their contributions to autoimmune diseases. However, the pathways of differentiation of Th17 and related Th cells (type 17 cells) and the structure of the type 17 memory population in humans are not well understood; such understanding is critical for manipulating these cells in vivo. By exploiting differences in levels of surface CCR6, we found that human type 17 memory cells, including individual T cell clonotypes, form an elongated continuum of type 17 character along which cells can be driven by increasing RORγt. This continuum includes cells preserved within the memory pool with potentials that reflect the early preferential activation of multiple over single lineages. The phenotypes and epigenomes of CCR6+ cells are stable across cell divisions under noninflammatory conditions. Nonetheless, activation in polarizing and nonpolarizing conditions can yield additional functionalities, revealing, respectively, both environmentally induced and imprinted mechanisms that contribute differentially across the type 17 continuum to yield the unusual plasticity ascribed to type 17 cells.


Subject(s)
Autoimmune Diseases , Th17 Cells , Humans , Cell Differentiation , Phenotype , Receptors, CCR6/genetics , Th1 Cells/metabolism
2.
Nat Immunol ; 13(8): 744-52, 2012 Jun 24.
Article in English | MEDLINE | ID: mdl-22729248

ABSTRACT

Langerhans cells (LCs) are epidermal dendritic cells with incompletely understood origins that associate with hair follicles for unknown reasons. Here we show that in response to external stress, mouse hair follicles recruited Gr-1(hi) monocyte-derived precursors of LCs whose epidermal entry was dependent on the chemokine receptors CCR2 and CCR6, whereas the chemokine receptor CCR8 inhibited the recruitment of LCs. Distinct hair-follicle regions had differences in their expression of ligands for CCR2 and CCR6. The isthmus expressed the chemokine CCL2; the infundibulum expressed the chemokine CCL20; and keratinocytes in the bulge produced the chemokine CCL8, which is the ligand for CCR8. Thus, distinct hair-follicle keratinocyte subpopulations promoted or inhibited repopulation with LCs via differences in chemokine production, a feature also noted in humans. Pre-LCs failed to enter hairless skin in mice or humans, which establishes hair follicles as portals for LCs.


Subject(s)
Chemokines/biosynthesis , Hair Follicle/immunology , Langerhans Cells/physiology , Stress, Physiological , Alopecia , Animals , Cell Movement , Chemokine CCL20/biosynthesis , Chemokine CCL8/biosynthesis , Chemokines/metabolism , Hair Follicle/metabolism , Humans , Keratinocytes/metabolism , Langerhans Cells/immunology , Mice , Mice, Hairless , Receptors, CCR2/metabolism , Receptors, CCR6/metabolism , Receptors, CCR8/metabolism , Skin/immunology
3.
J Immunol ; 203(12): 3179-3189, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31740485

ABSTRACT

In mice, the ability of naive T (TN) cells to mount an effector response correlates with TCR sensitivity for self-derived Ags, which can be quantified indirectly by measuring surface expression levels of CD5. Equivalent findings have not been reported previously in humans. We identified two discrete subsets of human CD8+ TN cells, defined by the absence or presence of the chemokine receptor CXCR3. The more abundant CXCR3+ TN cell subset displayed an effector-like transcriptional profile and expressed TCRs with physicochemical characteristics indicative of enhanced interactions with peptide-HLA class I Ags. Moreover, CXCR3+ TN cells frequently produced IL-2 and TNF in response to nonspecific activation directly ex vivo and differentiated readily into Ag-specific effector cells in vitro. Comparative analyses further revealed that human CXCR3+ TN cells were transcriptionally equivalent to murine CXCR3+ TN cells, which expressed high levels of CD5. These findings provide support for the notion that effector differentiation is shaped by heterogeneity in the preimmune repertoire of human CD8+ T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Receptors, CXCR3/metabolism , Adult , Age Factors , Aged , Animals , Biomarkers , Cells, Cultured , Female , Humans , Immunologic Memory , Immunophenotyping , Lymphocyte Activation/immunology , Male , Mice , Middle Aged , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Young Adult
4.
J Immunol ; 194(9): 4350-61, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25833398

ABSTRACT

Th17 cells, which express the chemokine receptor CCR6, are implicated in many immune-mediated disorders, such as psoriasis and multiple sclerosis. We found that expression levels of CCR6 on human effector/memory CD4(+) T cells reflect a continuum of Th17 differentiation. By evaluating the transcriptome in cells with increasing CCR6, we detected progressive upregulation of ZBTB16, which encodes the broad complex, tramtrack, bric-à-brac-zinc finger transcription factor promyelocytic leukemia zinc finger protein (PLZF). Using chromatin immunoprecipitation for modified histones, p300, and PLZF, we identified enhancer-like sites at -9/-10 and -13/-14 kb from the upstream transcription start site of CCR6 that bind PLZF in CCR6(+) cells. For Th cells from adult blood, both in the CCR6(+) memory population and in naive cells activated ex vivo, knockdown of ZBTB16 downregulated CCR6 and other Th17-associated genes. ZBTB16 and RORC (which encodes the "master regulator" RORγt) cross-regulate each other, and PLZF binds at the RORC promoter in CCR6(+) cells. In naive Th cells from cord blood, ZBTB16 expression was confined to CD161(+) cells, which are Th17 cell precursors. ZBTB16 was not expressed in mouse Th17 cells, and Th17 cells could be made from luxoid mice, which harbor an inactivating mutation in Zbtb16. These studies demonstrate a role for PLZF as an activator of transcription important both for Th17 differentiation and the maintenance of the Th17 phenotype in human cells, expand the role of PLZF as a critical regulator in the human adaptive immune system, and identify a novel, essential element in a regulatory network that is of significant therapeutic interest.


Subject(s)
Kruppel-Like Transcription Factors/metabolism , Phenotype , Receptors, CCR6/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Enhancer Elements, Genetic , Gene Expression , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Immunologic Memory , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Transgenic , NK Cell Lectin-Like Receptor Subfamily B/metabolism , Promyelocytic Leukemia Zinc Finger Protein , Protein Binding , Receptors, CCR6/genetics , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Th17 Cells/cytology
5.
Pharmacol Rev ; 66(1): 1-79, 2014.
Article in English | MEDLINE | ID: mdl-24218476

ABSTRACT

Sixteen years ago, the Nomenclature Committee of the International Union of Pharmacology approved a system for naming human seven-transmembrane (7TM) G protein-coupled chemokine receptors, the large family of leukocyte chemoattractant receptors that regulates immune system development and function, in large part by mediating leukocyte trafficking. This was announced in Pharmacological Reviews in a major overview of the first decade of research in this field [Murphy PM, Baggiolini M, Charo IF, Hébert CA, Horuk R, Matsushima K, Miller LH, Oppenheim JJ, and Power CA (2000) Pharmacol Rev 52:145-176]. Since then, several new receptors have been discovered, and major advances have been made for the others in many areas, including structural biology, signal transduction mechanisms, biology, and pharmacology. New and diverse roles have been identified in infection, immunity, inflammation, development, cancer, and other areas. The first two drugs acting at chemokine receptors have been approved by the U.S. Food and Drug Administration (FDA), maraviroc targeting CCR5 in human immunodeficiency virus (HIV)/AIDS, and plerixafor targeting CXCR4 for stem cell mobilization for transplantation in cancer, and other candidates are now undergoing pivotal clinical trials for diverse disease indications. In addition, a subfamily of atypical chemokine receptors has emerged that may signal through arrestins instead of G proteins to act as chemokine scavengers, and many microbial and invertebrate G protein-coupled chemokine receptors and soluble chemokine-binding proteins have been described. Here, we review this extended family of chemokine receptors and chemokine-binding proteins at the basic, translational, and clinical levels, including an update on drug development. We also introduce a new nomenclature for atypical chemokine receptors with the stem ACKR (atypical chemokine receptor) approved by the Nomenclature Committee of the International Union of Pharmacology and the Human Genome Nomenclature Committee.


Subject(s)
Receptors, Chemokine , Animals , Arthropod Proteins/genetics , Arthropod Proteins/metabolism , Humans , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Receptors, Chemokine/classification , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Terminology as Topic , Ticks , Viral Proteins/genetics , Viral Proteins/metabolism
6.
J Immunol ; 192(4): 1778-86, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24442439

ABSTRACT

It was reported that host defense against pulmonary Klebsiella pneumoniae infection requires IL-22, which was proposed to be of T cell origin. Supporting a role for IL-22, we found that Il22(-/-) mice had decreased survival compared with wild-type mice after intratracheal infection with K. pneumoniae. Surprisingly, however, Rag2(-/-) mice did not differ from wild-type mice in survival or levels of IL-22 in the lungs postinfection with K. pneumoniae. In contrast, K. pneumoniae-infected Rag2(-/-)Il2rg(-/-) mice failed to produce IL-22. These data suggested a possible role for NK cells or other innate lymphoid cells in host defense and production of IL-22. Unlike NK cell-like innate lymphoid cells that produce IL-22 and display a surface phenotype of NK1.1(-)NKp46(+)CCR6(+), lung NK cells showed the conventional phenotype, NK1.1(+)NKp46(+)CCR6(-). Mice depleted of NK cells using anti-asialo GM1 showed decreased survival and higher lung bacterial counts, as well as increased dissemination of K. pneumoniae to blood and liver, compared with control-treated mice. NK cell depletion also led to decreased production of IL-22 in the lung. Within 1 d postinfection, although there was no increase in the number of lung NK cells, a subset of lung NK cells became competent to produce IL-22, and such cells were found in both wild-type and Rag2(-/-) mice. Our data suggest that, during pulmonary infection of mice with K. pneumoniae, conventional NK cells are required for optimal host defense, which includes the production of IL-22.


Subject(s)
Interleukins/metabolism , Killer Cells, Natural/immunology , Klebsiella Infections/immunology , Pneumonia/immunology , Animals , Antigens, Ly/metabolism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Female , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Interleukins/biosynthesis , Interleukins/genetics , Klebsiella Infections/mortality , Klebsiella pneumoniae/immunology , Lung/cytology , Lung/immunology , Lymphocyte Depletion , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NK Cell Lectin-Like Receptor Subfamily B/metabolism , Natural Cytotoxicity Triggering Receptor 1/metabolism , Pneumonia/mortality , Receptors, CCR6/metabolism , Survival , Interleukin-22
7.
Mol Pharmacol ; 88(5): 894-910, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26316539

ABSTRACT

CXCR6, the receptor for CXCL16, is expressed on multiple cell types and can be a coreceptor for human immunodeficiency virus 1. Except for CXCR6, all human chemokine receptors contain the D(3.49)R(3.50)Y(3.51) sequence, and all but two contain A(3.53) at the cytoplasmic terminus of the third transmembrane helix (H3C), a region within class A G protein-coupled receptors that contacts G proteins. In CXCR6, H3C contains D(3.49)R(3.50)F(3.51)I(3.52)V(3.53) at positions 126-130. We investigated the importance and interdependence of the canonical D126 and the noncanonical F128 and V130 in CXCR6 by mutating D126 to Y, F128 to Y, and V130 to A singly and in combination. For comparison, we mutated the analogous positions D142, Y144, and A146 to Y, F, and V, respectively, in CCR6, a related receptor containing the canonical sequences. Mutants were analyzed in both human embryonic kidney 293T and Jurkat E6-1 cells. Our data show that for CXCR6 and/or CCR6, mutations in H3C can affect both receptor signaling and chemokine binding; noncanonical H3C sequences are functionally linked, with dual changes mitigating the effects of single mutations; mutations in H3C that compromise receptor activity show selective defects in the use of individual Gi/o proteins; and the effects of mutations in H3C on receptor function and selectivity in Gi/o protein use can be cell-type specific. Our findings indicate that the ability of CXCR6 to make promiscuous use of the available Gi/o proteins is exquisitely dependent on sequences within the H3C and suggest that the native sequence allows for preservation of this function across different cellular environments.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Receptors, Chemokine/chemistry , Receptors, Virus/chemistry , Amino Acid Motifs , Amino Acid Sequence , Cells, Cultured , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , HEK293 Cells , Humans , Jurkat Cells , Models, Molecular , Mutagenesis , Receptors, CXCR6 , Receptors, Chemokine/physiology , Receptors, Virus/physiology , Structure-Activity Relationship
8.
Circ Res ; 109(4): 374-81, 2011 Aug 05.
Article in English | MEDLINE | ID: mdl-21680896

ABSTRACT

RATIONALE: The chemokine receptor Ccr6 is a G-protein-coupled receptor expressed on various types of leukocytes identified in mouse atherosclerotic lesions. Recent evidence suggests that both CCR6 and its ligand CCL20 are also present in human atheroma; however, their functional roles in atherogenesis remain undefined. OBJECTIVE: Our objective was to delineate the role of Ccr6 in atherogenesis in the apolipoprotein E-deficient (ApoE(-/-)) mouse model of atherosclerosis. METHODS AND RESULTS: Both Ccr6 and Ccl20 are expressed in atherosclerotic aorta from ApoE(-/-) mice. Aortic lesion area in Ccr6(-/-)ApoE(-/-) mice was ∼40% and ∼30% smaller than in Ccr6(+/+)ApoE(-/-) mice at 16 and 24 weeks of age, respectively. Transplantation of bone marrow from Ccr6(-/-) mice into ApoE(-/-) mice resulted in ∼40% less atherosclerotic lesion area than for bone marrow from Ccr6(+/+) mice; lesions in Ccr6(-/-)ApoE(-/-) mice had 44% less macrophage content than lesions in Ccr6(+/+)ApoE(-/-) mice. Ccr6 was expressed on a subset of primary mouse monocytes. Accordingly, Ccl20 induced chemotaxis of primary monocytes from wild-type but not Ccr6(-/-) mice; moreover, Ccl20 induced monocytosis in ApoE(-/-) mice in vivo. Consistent with this, we observed 30% fewer monocytes in circulating blood of Ccr6(-/-)ApoE(-/-) mice, mainly because of fewer CD11b(+)Ly6C(high) inflammatory monocytes. CONCLUSIONS: Ccr6 promotes atherosclerosis in ApoE-deficient mice, which may be due in part to Ccr6 support of normal monocyte levels in blood, as well as direct Ccr6-dependent monocyte migration.


Subject(s)
Aorta/immunology , Aortic Diseases/prevention & control , Apolipoproteins E/deficiency , Atherosclerosis/prevention & control , Gene Deletion , Receptors, CCR6/deficiency , Animals , Antigens, Ly/blood , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/immunology , Aortic Diseases/pathology , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , Bone Marrow Transplantation , CD11b Antigen/blood , Cell Line , Chemokine CCL20/metabolism , Chemotaxis, Leukocyte , Disease Models, Animal , Female , Leukocyte Count , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Receptors, CCR6/genetics , Time Factors
9.
bioRxiv ; 2023 Feb 13.
Article in English | MEDLINE | ID: mdl-36789428

ABSTRACT

Pro-inflammatory T cells co-express multiple chemokine receptors, but the distinct functions of individual receptors on these cells are largely unknown. Human Th17 cells uniformly express the chemokine receptor CCR6, and we discovered that the subgroup of CD4+CCR6+ cells that co-express CCR2 possess a pathogenic Th17 signature, can produce inflammatory cytokines independent of TCR activation, and are unusually efficient at transendothelial migration (TEM). The ligand for CCR6, CCL20, was capable of binding to activated endothelial cells (ECs) and inducing firm arrest of CCR6+CCR2+ cells under conditions of flow - but CCR6 could not mediate TEM. By contrast, CCL2 and other ligands for CCR2, despite being secreted from both luminal and basal sides of ECs, failed to bind to the EC surfaces - and CCR2 could not mediate arrest. Nonetheless, CCR2 was required for TEM. To understand if CCR2's inability to mediate arrest was due solely to an absence of EC-bound ligands, we generated a CCL2-CXCL9 chimeric chemokine that could bind to the EC surface. Although display of CCL2 on the ECs did indeed lead to CCR2-mediated arrest of CCR6+CCR2+ cells, activating CCR2 with surface-bound CCL2 blocked TEM. We conclude that mediating arrest and TEM are mutually exclusive activities of chemokine receptors and/or their ligands that depend, respectively, on chemokines that bind to the EC luminal surfaces versus non-binding chemokines that form transendothelial gradients under conditions of flow. Our findings provide fundamental insights into mechanisms of lymphocyte extravasation and may lead to novel strategies to block or enhance their migration into tissue.

10.
bioRxiv ; 2023 Jan 06.
Article in English | MEDLINE | ID: mdl-36789418

ABSTRACT

Th17 cells have been investigated in mice primarily for their contributions to autoimmune diseases. However, the pathways of differentiation of Th17 and related (type 17) cells and the structure of the type 17 memory population in humans are not well understood; such understanding is critical for manipulating these cells in vivo . By exploiting differences in levels of surface CCR6, we found that human type 17 memory cells, including individual T cell clonotypes, form an elongated continuum of type 17 character along which cells can be driven by increasing RORγt. This continuum includes cells preserved within the memory pool with potentials that reflect the early preferential activation of multiple over single lineages. The CCR6 + cells' phenotypes and epigenomes are stable across cell divisions under homeostatic conditions. Nonetheless, activation in polarizing and non-polarizing conditions can yield additional functionalities, revealing, respectively, both environmentally induced and imprinted mechanisms that contribute differentially across the continuum to yield the unusual plasticity ascribed to type 17 cells.

11.
J Immunol ; 185(6): 3295-304, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20720211

ABSTRACT

Expression of CCR6 and its ligand, CCL20, are increased in the colon of humans with inflammatory bowel diseases and mice with experimental colitis; however, their role in disease pathogenesis remains obscure. In this study, we demonstrate a role for CCR6 on regulatory T (Treg) cells in the T cell-transfer model of colitis. Rag2(-/-) mice given Ccr6(-/-)CD4(+)CD45RB(high) T cells had more severe colitis with increased IFN-gamma-producing T cells, compared with the mice given wild-type cells. Although an equivalent frequency of induced/acquired Treg (iTreg) cells was observed in mesenteric lymph nodes and colon from both groups, the suppressive capacity of Ccr6(-/-) iTreg cells was impaired. Cotransfer studies of wild-type or Ccr6(-/-) Treg cells with CD4(+)CD45RB(high) T cells also showed a defect in suppression by Ccr6(-/-) Treg cells. CCR6(+) Treg cells were characterized as Ag-activated and IL-10-producing in the steady-state and preferentially migrated to the colon during inflammation. Thus, we conclude that CCR6 expression on Treg cells was required for the full function of Treg cell-mediated suppression in the T cell-transfer model of colitis. CCR6 may contribute to the regulation of colitis by directing its function in Ag-specific, IL-10-producing iTreg cells to the inflamed colon.


Subject(s)
Colitis/immunology , Colitis/pathology , Immunophenotyping , Interleukin-10/biosynthesis , Receptors, CCR6/genetics , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Adoptive Transfer , Animals , Cell Movement/genetics , Cell Movement/immunology , Colitis/genetics , Colitis/prevention & control , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Female , Humans , Inflammation Mediators/physiology , Interferon-gamma/biosynthesis , Interleukin-10/genetics , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, CCR6/deficiency , Receptors, CCR6/physiology , Resting Phase, Cell Cycle/genetics , Resting Phase, Cell Cycle/immunology , Severity of Illness Index , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/transplantation , T-Lymphocytes, Regulatory/metabolism
12.
J Immunol ; 184(7): 3450-60, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20194716

ABSTRACT

Donor Ag-reactive CD4 and CD8 T cell production of IFN-gamma is a principal effector mechanism promoting tissue injury during allograft rejection. The CXCR3-binding chemokines CXCL9 and CXCL10 recruit donor-reactive T cells to the allograft, but their role during the priming of donor-reactive T cells to effector function is unknown. Using a murine model of MHC-mismatched cardiac transplantation, we investigated the influence of CXCL9 and CXCL10 during donor-reactive T cell priming. In allograft recipient spleens, CXCL9 and CXCL10 were expressed as early as 24 h posttransplant and increased with similar kinetics, concurrently with CXCR3 expression on T cells. CXCL9, but not CXCL10, expression required NK cell production of IFN-gamma. The absence of CXCL9 in donor allografts, recipients, or both significantly decreased the frequency of donor-reactive CD8 T cells producing IFN-gamma and increased the frequency of donor-reactive CD8 T cells producing IL-17A. In contrast, the absence of CXCL10 increased the frequency of IFN-gamma-producing CD8 T cells in a CXCL9-dependent manner. These data provide novel evidence that donor-reactive CD8 T cells use the CXCR3 chemokine axis as a costimulation pathway during priming to allografts where CXCL9 promotes the development of IFN-gamma-producing CD8 T cells, and CXCL10 antagonizes this skewing.


Subject(s)
Chemokine CXCL10/immunology , Chemokine CXCL9/immunology , Graft Rejection/immunology , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Separation , Chemokine CXCL10/metabolism , Chemokine CXCL9/metabolism , Flow Cytometry , Graft Survival/immunology , Heart Transplantation/immunology , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Mice , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Homologous
13.
J Immunol ; 185(11): 6646-63, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20980630

ABSTRACT

Because T cells act primarily through short-distance interactions, homing receptors can identify colocalizing cells that serve common functions. Expression patterns for multiple chemokine receptors on CD4(+) T cells from human blood suggested a hierarchy of receptors that are induced and accumulate during effector/memory cell differentiation. We characterized CD4(+)CD45RO(+) T cells based on expression of two of these receptors, CCR5 and CCR2, the principal subsets being CCR5(-)CCR2(-) (∼70%), CCR5(+)CCR2(-) (∼25%), and CCR5(+)CCR2(+) (∼5%). Relationships among expression of CCR5 and CCR2 and CD62L, and the subsets' proliferation histories, suggested a pathway of progressive effector/memory differentiation from the CCR5(-)CCR2(-) to CCR5(+)CCR2(-) to CCR5(+)CCR2(+) cells. Sensitivity and rapidity of TCR-mediated activation, TCR signaling, and effector cytokine production by the subsets were consistent with such a pathway. The subsets also showed increasing responsiveness to IL-7, and the CCR5(+)CCR2(+) cells were CD127(bright) and invariably showed the greatest response to tetanus toxoid. CCR5(+)CCR2(+) cells also expressed the largest repertoire of chemokine receptors and migrated to the greatest number of chemokines. By contrast, the CCR5(+)CCR2(-) cells had the greatest percentages of regulatory T cells, activated/cycling cells, and CMV-reactive cells, and were most susceptible to apoptosis. Our results indicate that increasing memory cell differentiation can be uncoupled from susceptibility to death, and is associated with an increase in chemokine responsiveness, suggesting that vaccination (or infection) can produce a stable population of effector-capable memory cells that are highly enriched in the CCR5(+)CCR2(+) subset and ideally equipped for rapid recall responses in tissue.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Immunologic Memory , Receptors, CCR2/physiology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , CD4-Positive T-Lymphocytes/cytology , Cell Differentiation/immunology , Cells, Cultured , Humans , Immunophenotyping , Receptors, CCR2/biosynthesis , Receptors, CCR5/biosynthesis , Receptors, CCR5/immunology , Receptors, CCR5/metabolism , Resting Phase, Cell Cycle/immunology , T-Lymphocyte Subsets/cytology
14.
J Invest Dermatol ; 142(5): 1381-1390.e11, 2022 05.
Article in English | MEDLINE | ID: mdl-34808237

ABSTRACT

Bile acids (BAs), produced in the liver and further transformed in the gut, are cholesterol-derived molecules involved in essential physiological processes. Recent studies suggest that BAs regulate T helper 17 cell function, but the underlying mechanism of this action and their therapeutic value in disease models remains unclear. Using an IL-23 minicircle DNA-based murine model of psoriasiform dermatitis, we showed that oral administration of secondary BAs, including lithocholic acid (LCA), deoxycholic acid, and 3-oxoLCA, significantly improved psoriasiform dermatitis without inducing apparent hepatotoxicity. Of the BAs tested, LCA possessed the greatest potency in treating psoriasiform dermatitis. Intravenous administration of LCA at a much lower dosage (compared with oral treatment) showed a comparable antipsoriatic effect and markedly suppressed the IL-17A response. Ex vivo experiments revealed that LCA reduced IL-17A production in IL-23-stimulated murine T cells in the absence of BA receptors TGR5 or FXR. Strikingly, BAs inhibited CCL20 expression in keratinocytes, which led to reduced migration of CCR6-expressing Jurkat cells cultured in the conditioned medium of stimulated keratinocytes. Thus, BAs improve psoriasiform dermatitis with minimal toxicity via direct inhibition of IL-17A production and blockade of CCL20-mediated trafficking, supporting the potential use of BAs in psoriasis.


Subject(s)
Eczema , Psoriasis , Animals , Bile Acids and Salts/therapeutic use , Chemokine CCL20 , Humans , Interleukin-17/metabolism , Interleukin-23 , Mice , Psoriasis/drug therapy , Psoriasis/metabolism , Receptors, CCR6
15.
J Biol Chem ; 285(46): 35537-50, 2010 Nov 12.
Article in English | MEDLINE | ID: mdl-20829352

ABSTRACT

Because T cell differentiation leads to an expanded repertoire of chemokine receptors, a subgroup of G protein-coupled receptors, we hypothesized that the repertoire of G proteins might be altered in parallel. We analyzed the abundance of mRNA and/or protein of six G protein α-subunits in human CD4(+) and CD8(+) T cell subsets from blood. Although most G protein α-subunits were similarly expressed in all subsets, the abundance of Gα(o), a protein not previously described in hematopoietic cells, was much higher in memory versus naive cells. Consistent with these data, activation of naive CD4(+) T cells in vitro significantly increased the abundance of Gα(o) in cells stimulated under nonpolarizing or T(H)17 (but not T(H)1 or T(H)2)-polarizing conditions. In functional studies, the use of a chimeric G protein α-subunit, Gα(qo5), demonstrated that chemokine receptors could couple to Gα(o)-containing G proteins. We also found that Gα(i1), another α-subunit not described previously in leukocytes, was expressed in naive T cells but virtually absent from memory subsets. Corresponding to their patterns of expression, siRNA-mediated knockdown of Gα(o) in memory (but not naive) and Gα(i1) in naive (but not memory) CD4(+) T cells inhibited chemokine-dependent migration. Moreover, although even in Gα(o)- and Gα(i1)-expressing cells mRNAs of these α-subunits were much less abundant than Gα(i2) or Gα(i3), knockdown of any of these subunits impaired chemokine receptor-mediated migration similarly. Together, our data reveal a change in the repertoire of Gα(i/o) subunits during T cell differentiation and suggest functional equivalence among Gα(i/o) subunits irrespective of their relative abundance.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation , GTP-Binding Protein alpha Subunits/metabolism , Blotting, Western , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Cells, Cultured , Chemokine CXCL12/metabolism , Chemotaxis/genetics , Female , Fetal Blood/cytology , GTP-Binding Protein alpha Subunit, Gi2/genetics , GTP-Binding Protein alpha Subunit, Gi2/metabolism , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Gene Expression Profiling , HEK293 Cells , Humans , Infant, Newborn , Jurkat Cells , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Pregnancy , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, CCR6/genetics , Receptors, CCR6/metabolism , Receptors, CXCR6 , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Receptors, Virus/genetics , Receptors, Virus/metabolism , Reverse Transcriptase Polymerase Chain Reaction
16.
Eur J Immunol ; 40(11): 3183-3197, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20963786

ABSTRACT

Although regulation of CXCR3 and CCR4 is related to Th1 and Th2 differentiation, respectively, many CXCR3(+) and CCR4(+) cells do not express IFN-γ and/or IL-4, suggesting that the chemokine receptor genes might be inducible by mechanisms that are lineage-independent. We investigated the regulation of CXCR3 versus IFNG, and CCR4 versus IL4 in human CD4(+) T cells by analyzing modifications of histone H3. In naïve cord-blood cells, under nonpolarizing conditions not inducing IL4, CCR4 was induced to high levels without many of the activation-associated changes in promoter histone H3 found for both IL4 and CCR4 in Th2 cells. Importantly, CCR4 expression was stable in Th2 cells, but fell in nonpolarized cells after the cells were rested; this decline could be reversed by increasing histone acetylation using sodium butyrate. Patterns of histone H3 modifications in CXCR3(+) CCR4(-) and CXCR3(-) CCR4(+) CD4(+) T-cell subsets from adult blood matched those in cells cultured under polarizing conditions in vitro. Our data show that high-level lineage-independent induction of CCR4 can occur following T-cell activation without accessibility-associated changes in histone H3, but that without such changes expression is transient rather than persistent.


Subject(s)
Gene Expression Regulation/immunology , Histones/immunology , Lymphocyte Activation/immunology , Receptors, CCR4/immunology , Th2 Cells/immunology , Acetylation/drug effects , Butyrates/pharmacology , Cells, Cultured , Gene Expression Regulation/drug effects , Histones/metabolism , Humans , Interleukin-4/immunology , Interleukin-4/metabolism , Lymphocyte Activation/drug effects , Methylation/drug effects , Receptors, CCR4/biosynthesis , Receptors, CXCR3/biosynthesis , Receptors, CXCR3/immunology , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/metabolism
17.
PLoS Pathog ; 5(12): e1000703, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20041174

ABSTRACT

We sought to define protective mechanisms of immunity to Staphylococcus aureus and Candida albicans bloodstream infections in mice immunized with the recombinant N-terminus of Als3p (rAls3p-N) vaccine plus aluminum hydroxide (Al(OH(3)) adjuvant, or adjuvant controls. Deficiency of IFN-gamma but not IL-17A enhanced susceptibility of control mice to both infections. However, vaccine-induced protective immunity against both infections required CD4+ T-cell-derived IFN-gamma and IL-17A, and functional phagocytic effectors. Vaccination primed Th1, Th17, and Th1/17 lymphocytes, which produced pro-inflammatory cytokines that enhanced phagocytic killing of both organisms. Vaccinated, infected mice had increased IFN-gamma, IL-17, and KC, increased neutrophil influx, and decreased organism burden in tissues. In summary, rAls3p-N vaccination induced a Th1/Th17 response, resulting in recruitment and activation of phagocytes at sites of infection, and more effective clearance of S. aureus and C. albicans from tissues. Thus, vaccine-mediated adaptive immunity can protect against both infections by targeting microbes for destruction by innate effectors.


Subject(s)
Adaptive Immunity , Candidiasis/immunology , Fungal Proteins/immunology , Fungal Vaccines/immunology , Staphylococcal Infections/immunology , T-Lymphocyte Subsets/immunology , Adjuvants, Immunologic/pharmacology , Adoptive Transfer , Aluminum Hydroxide/immunology , Animals , Candida albicans/immunology , Candidiasis/prevention & control , Female , Interferon-gamma , Interleukin-17/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Staphylococcal Infections/prevention & control , Staphylococcus aureus/immunology , Th1 Cells/immunology , Vaccines/immunology
18.
Am J Pathol ; 174(6): 2172-81, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19389928

ABSTRACT

Chemokines, including monokine induced by interferon-gamma (Mig/CXCL9), are produced both in allografts and during the direct T-cell infiltration that mediates graft rejection. Neither the specific production nor contribution of allograft donor versus recipient Mig in allograft rejection is currently known. C57BL/6 mice with a targeted deletion in the Mig gene were used as both skin allograft donors and recipients in a class II major histocompatibility complex-mismatched graft model to test the requirement for donor- versus recipient-derived Mig for acute rejection. B6.Mig(-/-) allografts had a 10-day prolonged survival in B6.H-2(bm12) recipients when compared with wild-type C57BL/6 allograft donors, and B6.H-2(bm12) skin allografts had a 5-day prolonged survival in B6.Mig(-/-) versus wild-type recipients. Transplantation of B6.Mig(-/-) skin grafts onto B6.H-2(bm12).Mig(-/-) recipients resulted in further prolonged allograft survival with more than 30% of the grafts surviving longer than 60 days. Prolonged allograft survival was also associated with delayed cellular infiltration into grafts but not with altered T-cell proliferative responses to donor stimulators. Immunohistochemical staining of allograft sections indicated that Mig is produced by both donor- and recipient-derived sources, but Mig from each of these sources appeared in different areas of the allograft tissue. These results therefore demonstrate the synergy of donor- and recipient-derived Mig in promoting T-cell infiltration into allografts.


Subject(s)
Chemokine CXCL9/biosynthesis , Graft Rejection/immunology , Skin Transplantation/immunology , Transplantation, Homologous/immunology , Animals , Chemokine CXCL9/genetics , Chemokine CXCL9/immunology , Chemotaxis, Leukocyte/immunology , Flow Cytometry , Graft Survival/immunology , Histocompatibility Antigens Class II/genetics , Immunohistochemistry , Interferon-gamma/immunology , Lymphocyte Culture Test, Mixed , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Monokines/biosynthesis , Monokines/genetics , Monokines/immunology , T-Lymphocytes/immunology
19.
Methods Mol Biol ; 2098: 179-188, 2020.
Article in English | MEDLINE | ID: mdl-31792823

ABSTRACT

Human MAIT cells show little expression of the selectin CD62L and the chemokine receptor CCR7, which are important for entering lymph nodes, and high expression of selectin ligands and chemokine receptors that mediate trafficking into inflamed tissue. Extravasation of leukocytes into tissue requires sequential steps including rolling, firm arrest, crawling, and transendothelial migration, and can be modeled using endothelial cell monolayers in flow chambers that approximate the sheer stress found in post-capillary venules. Using MAIT cells purified from elutriated lymphocytes by fluorescence-activated cell sorting, we have used flow chambers to demonstrate roles for individual chemokine receptors in specific steps required for extravasation. These methods provide a general way to study the molecular mechanisms underlying MAIT cell trafficking from blood into tissue.


Subject(s)
Biological Assay , Cell Movement/immunology , Mucosal-Associated Invariant T Cells/immunology , Mucosal-Associated Invariant T Cells/metabolism , Biological Assay/methods , Biomarkers , Cell Movement/genetics , Flow Cytometry , Humans , Immunophenotyping , Mucosal-Associated Invariant T Cells/enzymology
20.
Bioorg Med Chem ; 17(4): 1486-93, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19188071

ABSTRACT

CXCR4 is a chemokine receptor which has been shown to be exploited by various tumors for increased survival, invasion, and homing to target organs. We developed a one step radiosynthesis for labeling the CXCR4-specific antagonist AMD3100 with Cu-64 to produce (64)Cu-AMD3100 with a specific activity of 11.28Ci/ micromol (417GBq/ micromol) at the end of radiosynthesis. Incorporation of Cu(II) ion into AMD3100 did not change its ability to inhibit cellular migration in response to the (only) CXCR4 ligand, SDF-1/CXCL12. (64)Cu-AMD3100 binding affinity to CXCR4 was found to be 62.7 microM. Biodistribution of (64)Cu-AMD3100 showed accumulation in CXCR4-expressing organs and tissues, a renal clearance pathway, and an anomalous specific accumulation in the liver. We conclude that (64)Cu-AMD3100 exhibits promise as a potential PET imaging agent for visualization of CXCR4-positive tumors and metastases and might be used to guide and monitor anti-CXCR4 tumor therapy.


Subject(s)
Copper Radioisotopes/chemistry , Heterocyclic Compounds/chemistry , Radiopharmaceuticals/chemical synthesis , Receptors, CXCR4/analysis , Animals , Benzylamines , Calcium Signaling/drug effects , Cell Line, Tumor , Cyclams , Diagnostic Imaging/methods , Heterocyclic Compounds/metabolism , Heterocyclic Compounds/pharmacology , Humans , Isotope Labeling/methods , Jurkat Cells , Mice , Mice, Inbred C57BL , Positron-Emission Tomography/methods , Protein Binding , Radiopharmaceuticals/metabolism , Radiopharmaceuticals/pharmacology , Receptors, CXCR4/antagonists & inhibitors , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL