Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 244
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 20(8): 992-1003, 2019 08.
Article in English | MEDLINE | ID: mdl-31263279

ABSTRACT

Here we identify a group 2 innate lymphoid cell (ILC2) subpopulation that can convert into interleukin-17 (IL-17)-producing NKp44- ILC3-like cells. c-Kit and CCR6 define this ILC2 subpopulation that exhibits ILC3 features, including RORγt, enabling the conversion into IL-17-producing cells in response to IL-1ß and IL-23. We also report a role for transforming growth factor-ß in promoting the conversion of c-Kit- ILC2s into RORγt-expressing cells by inducing the upregulation of IL23R, CCR6 and KIT messenger RNA in these cells. This switch was dependent on RORγt and the downregulation of GATA-3. IL-4 was able to reverse this event, supporting a role for this cytokine in maintaining ILC2 identity. Notably, this plasticity has physiological relevance because a subset of RORγt+ ILC2s express the skin-homing receptor CCR10, and the frequencies of IL-17-producing ILC3s are increased at the expense of ILC2s within the lesional skin of patients with psoriasis.


Subject(s)
Interleukin-17/immunology , Lymphocytes/immunology , Psoriasis/pathology , Skin/pathology , Cells, Cultured , Humans , Interleukin-1beta/immunology , Interleukin-23 Subunit p19/immunology , Interleukin-4/immunology , Lymphocytes/cytology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Psoriasis/immunology , Receptors, CCR10/metabolism , Skin/immunology , Transforming Growth Factor beta/metabolism
2.
Nat Immunol ; 17(1): 48-56, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26523865

ABSTRACT

Innate lymphoid cells (ILCs) 'preferentially' localize into barrier tissues, where they function in tissue protection but can also contribute to inflammatory diseases. The mechanisms that regulate the establishment of ILCs in barrier tissues are poorly understood. Here we found that under steady-state conditions, ILCs in skin-draining lymph nodes (sLNs) were continuously activated to acquire regulatory properties and high expression of the chemokine receptor CCR10 for localization into the skin. CCR10(+) ILCs promoted the homeostasis of skin-resident T cells and, reciprocally, their establishment in the skin required T cell-regulated homeostatic environments. CD207(+) dendritic cells expressing the transcription factor Foxn1 were required for the proper generation of CCR10(+) ILCs. These observations reveal mechanisms that underlie the specific programming and priming of skin-homing CCR10(+) ILCs in the sLNs.


Subject(s)
Homeostasis/immunology , Lymph Nodes/immunology , Lymphocytes/immunology , Receptors, CCR10/immunology , Skin/immunology , Adoptive Transfer , Animals , Flow Cytometry , Immunity, Innate/immunology , Lymph Nodes/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout
3.
J Immunol ; 211(1): 118-129, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37222480

ABSTRACT

Four major mucosal-associated chemokines, CCL25, CCL28, CXCL14, and CXCL17, play an important role in protecting mucosal surfaces from infectious pathogens. However, their role in protection against genital herpes remains to be fully explored. The CCL28 is a chemoattractant for the CCR10 receptor-expressing immune cells and is produced homeostatically in the human vaginal mucosa (VM). In this study, we investigated the role of the CCL28/CCR10 chemokine axis in mobilizing protective antiviral B and T cell subsets into the VM site of herpes infection. We report a significant increase in the frequencies of HSV-specific memory CCR10+CD44+CD8+ T cells, expressing high levels of CCR10, in herpes-infected asymptomatic (ASYMP) women compared with symptomatic women. Similarly, a significant increase in the CCL28 chemokine (a ligand of CCR10), was detected in the VM of herpes-infected ASYMP C57BL/6 mice, associated with the mobilization of high frequencies of HSV-specific effector memory CCR10+CD44+CD62L-CD8+ TEM cells and memory CCR10+B220+CD27+ B cells in the VM of HSV-infected ASYMP mice. Inversely, compared with wild-type C57BL/6 mice, the CCL28 knockout (CCL28-/-) mice (1) appeared to be more susceptible to intravaginal infection and reinfection with HSV type 2, and (2) exhibited a significant decrease in the frequencies of HSV-specific effector memory CCR10+CD44+CD62L-CD8+ TEM cells and of memory CD27+B220+ B cells in the infected VM. These findings suggest a critical role of the CCL28/CCR10 chemokine axis in the mobilization of antiviral memory B and T cells within the VM to protect against genital herpes infection and disease.


Subject(s)
Herpes Genitalis , Humans , Female , Mice , Animals , Antiviral Agents/metabolism , Mice, Inbred C57BL , CD8-Positive T-Lymphocytes , Herpesvirus 2, Human , Mucous Membrane , Antiviral Restriction Factors , Receptors, CCR10/metabolism , Chemokines, CC/metabolism , Hyaluronan Receptors/metabolism
4.
Am J Pathol ; 192(2): 332-343, 2022 02.
Article in English | MEDLINE | ID: mdl-35144761

ABSTRACT

Dry eye disease (DED) features the inflammatory response of the ocular surface. Pro-inflammatory T helper 17 (Th17) cells are important for the pathogenesis of DED. In the present study a mouse DED model was used to discover two Th17 subsets in draining lymph nodes and conjunctivae based on the expression of IL-17 receptor E (IL-17RE) and CCR10: IL-17RElowCCR10- Th17 and IL-17REhighCCR10+ Th17. IL-17REhighCCR10+ Th17 expressed more retinoic acid-related orphan receptor gamma t but fewer T-box-expressed-in-T-cells than IL-17RElowCCR10- Th17. In addition, the former expressed higher IL-17A, IL-21, and IL-22 but fewer IFN-γ than the latter. Further analysis showed that IL-17REhighCCR10+ Th17 did not express IFN-γ in vivo, whereas IL-17RElowCCR10- Th17 contained IFN-γ-expressing Th17/Th1 cells. Moreover, IL-17REhighCCR10+ Th17 possessed more phosphorylated p38 mitogen-activated protein kinase (MAPK) and Jnk than IL-17RElowCCR10- Th17, suggesting higher activation of MAPK signaling in IL-17REhighCCR10+ Th17. In vitro treatment with IL-17C effectively maintained IL-17A expression in Th17 cells through p38 MAPK rather than Jnk MAPK. Furthermore, the adoptive transfer of the two Th17 subpopulations indicated their equivalent pathogenicity in DED. Interestingly, IL-17REhighCCR10+ Th17 cells were able to phenotypically polarize to IL-17RElowCCR10- Th17 cells in vivo. In conclusion, the current study revealed novel Th17 subsets with differential phenotypes, functions, and signaling status in DED, thus deepening the understanding of Th17 pathogenicity, and exhibited Th17 heterogeneity in DED.


Subject(s)
Dry Eye Syndromes/immunology , MAP Kinase Signaling System/immunology , Receptors, CCR10/immunology , Receptors, Interleukin-17/immunology , Th17 Cells/immunology , Animals , Disease Models, Animal , Dry Eye Syndromes/genetics , Dry Eye Syndromes/pathology , MAP Kinase Signaling System/genetics , Mice , Mice, Transgenic , Receptors, CCR10/genetics , Receptors, Interleukin-17/genetics , Th17 Cells/pathology
5.
Immunity ; 41(4): 592-604, 2014 Oct 16.
Article in English | MEDLINE | ID: mdl-25308333

ABSTRACT

Lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) is a pattern-recognition receptor for a variety of endogenous and exogenous ligands. However, LOX-1 function in the host immune response is not fully understood. Here, we report that LOX-1 expressed on dendritic cells (DCs) and B cells promotes humoral responses. On B cells LOX-1 signaling upregulated CCR7, promoting cellular migration toward lymphoid tissues. LOX-1 signaling on DCs licensed the cells to promote B cell differentiation into class-switched plasmablasts and led to downregulation of chemokine receptor CXCR5 and upregulation of chemokine receptor CCR10 on plasmablasts, enabling their exit from germinal centers and migration toward local mucosa and skin. Finally, we found that targeting influenza hemagglutinin 1 (HA1) subunit to LOX-1 elicited HA1-specific protective antibody responses in rhesus macaques. Thus, LOX-1 expressed on B cells and DC cells has complementary functions to promote humoral immune responses.


Subject(s)
B-Lymphocytes/immunology , Dendritic Cells/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Immunoglobulin Class Switching/immunology , Scavenger Receptors, Class E/immunology , Animals , Antibody Formation/immunology , Cell Differentiation/immunology , Cell Movement/immunology , Germinal Center/cytology , Humans , Immunoglobulin A/biosynthesis , Immunoglobulin G/biosynthesis , Immunoglobulin M/biosynthesis , Lymphocyte Activation/immunology , Macaca mulatta , Male , Mucous Membrane/cytology , Receptors, CCR10/biosynthesis , Receptors, CCR7/biosynthesis , Receptors, CXCR5/biosynthesis , Scavenger Receptors, Class E/biosynthesis , Signal Transduction/immunology , Skin/cytology
6.
Eur J Immunol ; 51(12): 3194-3201, 2021 12.
Article in English | MEDLINE | ID: mdl-34564853

ABSTRACT

Accelerate lung repair in SARS-CoV-2 pneumonia is essential for pandemic handling. Innate lymphoid cells (ILCs) are likely players, given their role in mucosal protection and tissue homeostasis. We studied ILC subpopulations at two time points in a cohort of patients admitted in the hospital due to SARS-CoV-2 infection. COVID-19 patients with moderate/severe respiratory failure featured profound depletion of circulating ILCs at hospital admission, in agreement with overall lymphocyte depletion. However, ILCs recovered in direct correlation with lung function improvement as measured by oxygenation index and in negative association with inflammatory and lung/endothelial damage markers like RAGE. While both ILC1 and ILC2 expanded, ILC2 showed the most striking phenotype changes, with CCR10 upregulation in strong correlation with these parameters. Overall, CCR10+ ILC2 emerge as relevant contributors to SARS-CoV-2 pneumonia recovery.


Subject(s)
Biomarkers/metabolism , COVID-19/immunology , Lung/pathology , Lymphocytes/immunology , Pneumonia, Viral/immunology , Receptors, CCR10/metabolism , SARS-CoV-2/physiology , Adult , Aged , Antigens, Neoplasm/metabolism , Cell Proliferation , Cytokines/metabolism , Female , Humans , Immunity, Innate , Male , Middle Aged , Mitogen-Activated Protein Kinases/metabolism , Recovery of Function , Th2 Cells/immunology , Up-Regulation
7.
Expert Rev Mol Med ; 24: e37, 2022 09 26.
Article in English | MEDLINE | ID: mdl-36155126

ABSTRACT

Cancer is now one of the major causes of death across the globe. The imbalance of cytokine and chemokine secretion has been reported to be involved in cancer development. Meanwhile, CC chemokines have received considerable interest in cancer research. CCR10, as the latest identified CC chemokine receptor (CCR), has been implicated in the recruitment and infiltration of immune cells, especially lymphocytes, into epithelia such as skin via ligation to two ligands, CCL27 and CCL28. Other than homoeostatic function, several mechanisms have been shown to dysregulate CCR10/CCL27-CCL28 expression in the tumour microenvironment. As such, these receptors and ligands mediate T-cell trafficking in the tumour microenvironment. Depending on the types of lymphocytes recruited, CCR10/CCL27-CCL28 interaction has been shown to play conflicting roles in cancer development. If they were T helper and cytotoxic T cells and natural killer cells, the role of this axis would be tumour-suppressive. In contrast, if CCR10/CCL27-CCL28 recruited regulatory T cells, cancer-associated fibroblasts or myeloid-derived suppressor cells, it would lead to tumour progression. In addition to the trafficking of lymphocytes and immune cells, CCR10 also leads to the migration of tumour cells or endothelial cells (called angiogenesis and lymphangiogenesis) to promote tumour metastasis. Furthermore, CCR10 signalling triggers tumour-promoting signalling such as PI3K/AKT and mitogen-activated protein kinase/extracellular signal-regulated kinase, resulting in tumour cell growth. Since CCR10/CCL27-CCL28 is dysregulated in the tumour tissues, it is suggested that analysis and measurement of them might predict tumour development. Finally, it is hoped using therapeutic approaches based on this axis might increase our knowledge to overcome tumour progression.


Subject(s)
Neoplasms , Receptors, CCR10 , Humans , Endothelial Cells/metabolism , Ligands , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Chemokines, CC/metabolism , Receptors, CCR , Neoplasms/etiology , Neoplasms/genetics , Extracellular Signal-Regulated MAP Kinases , Tumor Microenvironment/genetics , Chemokine CCL27
8.
J Allergy Clin Immunol ; 148(5): 1281-1292, 2021 11.
Article in English | MEDLINE | ID: mdl-33932468

ABSTRACT

BACKGROUND: In previous human skin single-cell data, inflammatory cells constituted only a small fraction of the overall cell population, such that functional subsets were difficult to ascertain. OBJECTIVE: Our aims were to overcome the aforesaid limitation by applying single-cell transcriptomics to emigrating cells from skin and elucidate ex vivo gene expression profiles of pathogenic versus regulatory immune cell subsets in the skin of individuals with psoriasis. METHODS: We harvested emigrating cells from human psoriasis skin after incubation in culture medium without enzyme digestion or cell sorting and analyzed cells with single-cell RNA sequencing and flow cytometry simultaneously. RESULTS: Unsupervised clustering of harvested cells from psoriasis skin and control skin identified natural killer cells, T-cell subsets, dendritic cell subsets, melanocytes, and keratinocytes in different layers. Comparison between psoriasis cells and control cells within each cluster revealed that (1) cutaneous type 17 T cells display highly differing transcriptome profiles depending on IL-17A versus IL-17F expression and IFN-γ versus IL-10 expression; (2) semimature dendritic cells are regulatory dendritic cells with high IL-10 expression, but a subset of semimature dendritic cells expresses IL-23A and IL-36G in psoriasis; and (3) CCL27-CCR10 interaction is potentially impaired in psoriasis because of decreased CCL27 expression in basal keratinocytes. CONCLUSION: We propose that single-cell transcriptomics applied to emigrating cells from human skin provides an innovative study platform to compare gene expression profiles of heterogenous immune cells in various inflammatory skin diseases.


Subject(s)
Dendritic Cells/immunology , Psoriasis/immunology , Skin/immunology , Th17 Cells/immunology , Cell Movement , Cell Separation , Cells, Cultured , Chemokine CCL27/metabolism , Cytokines/metabolism , Flow Cytometry , Gene Expression Profiling , Humans , Receptors, CCR10/metabolism , Sequence Analysis, RNA , Single-Cell Analysis , Transcriptome
9.
Beijing Da Xue Xue Bao Yi Xue Ban ; 54(6): 1074-1078, 2022 Dec 18.
Article in Zh | MEDLINE | ID: mdl-36533335

ABSTRACT

OBJECTIVE: To examine the expression of chemokine receptor CCR10 on monocytes/macrophages in the joints of patients with rheumatoid arthritis (RA), and to investigate the role of chemokine CCL28 and its receptor CCR10 in the migration of RA monocytes and its mechanism. METHODS: The expression of CCR10 in synovial tissues from 8 RA patients, 4 osteoarthritis (OA) patients, and 4 normal controls was analyzed by immunohistochemistry, and cell staining was scored on a 0-5 scales. Flow cytometry was used to measure the percentage of CCR10 positive cells in CD14+ monocytes from peripheral blood of 26 RA patients and 20 healthy controls, as well as from synovial fluid of 15 RA patients. The chemotactic migration of monocytes from RA patients and healthy controls in response to CCL28 was evaluated using an in vitro Transwell system. Western blotting was conducted to assess phosphorylation of the extracellular signal-regulated kinase (ERK) and protein kinase B (Akt) pathways in RA monocytes upon CCL28 treatment. RESULTS: CCR10 was predominantly expressed in RA synovial lining cells and sublining macrophages, endothelial cells, and lymphocytes. CCR10 expression was significantly increased on lining cells and sublining macrophages in RA synovial tissue compared with OA and normal synovial tissue (both P < 0.01). The patients with RA had markedly elevated expression of CCR10 on peripheral blood CD14+ monocytes compared with the healthy controls [(15.6±3.0)% vs. (7.7±3.8)%, P < 0.01]. CCR10 expression on synovial fluid monocytes from the RA patients was (32.0±15.0)%, which was significantly higher than that on RA peripheral blood monocytes (P < 0.01). In vitro, CCL28 caused significant migration of CD14+ monocytes from peripheral blood of the RA patients and the healthy controls at concentrations ranging from 10-100 µg/L (all P < 0.01). The presence of neutralizing antibody to CCR10 greatly suppressed CCL28-driven chemotaxis of RA monocytes (P < 0.01). Stimulation of RA monocytes with CCL28 induced a remarkable increase in phosphorylation of ERK and Akt (both P < 0.05). ERK inhibitor (U0126) and phosphatidylinositol 3-kinase (PI3K) inhibitor (LY294002) strongly reduced the migration of RA monocytes in response to CCL28 (both P < 0.01). CONCLUSION: RA patients had increased CCR10 expression on peripheral blood, synovial fluid, and synovial tissue monocytes/macrophages. CCL28 ligation to CCR10 promoted RA monocyte migration through activation of the ERK and PI3K/Akt signaling pathways. The CCL28-CCR10 pathway could participate in monocyte recruitment into RA joints, thereby contributing to synovial inflammation and bone destruction.


Subject(s)
Arthritis, Rheumatoid , Osteoarthritis , Humans , Monocytes/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Endothelial Cells/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Synovial Membrane , Chemokines, CC/metabolism , Synovial Fluid , Receptors, CCR10/metabolism
10.
Nat Immunol ; 10(8): 864-71, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19578368

ABSTRACT

Interleukin 22 (IL-22) is a member of the IL-10 cytokine family that is involved in inflammatory and wound healing processes. Originally considered a T helper type 1 (T(H)1)-associated cytokine, IL-22 has since been shown to be produced mainly by IL-17-producing helper T cells (T(H)-17 cells). Here we describe a previously uncharacterized IL-22-producing human helper T cell population that coexpressed the chemokine receptor CCR6 and the skin-homing receptors CCR4 and CCR10. These cells were distinct from both T(H)-17 cells and T(H)1 cells. Downregulation of either the aryl hydrocarbon receptor (AHR) or the transcription factor RORC by RNA-mediated interference affected IL-22 production, whereas IL-17 production was affected only by downregulation of RORC by RNA-mediated interference. AHR agonists substantially altered the balance of IL-22- versus IL-17-producing cells. This subset of IL-22-producing cells may be important in skin homeostasis and pathology.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Interleukins/biosynthesis , T-Lymphocytes, Helper-Inducer/metabolism , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation , Down-Regulation , Humans , Immunologic Memory , Interferon-gamma/biosynthesis , Interleukin-13/biosynthesis , Interleukin-17/biosynthesis , Lymphocyte Activation , Nuclear Receptor Subfamily 1, Group F, Member 3 , Receptors, Aryl Hydrocarbon/agonists , Receptors, Aryl Hydrocarbon/physiology , Receptors, CCR10/biosynthesis , Receptors, CCR4/biosynthesis , Receptors, CCR6/biosynthesis , Receptors, Retinoic Acid/physiology , Receptors, Thyroid Hormone/physiology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology , Interleukin-22
11.
Nat Immunol ; 10(8): 857-63, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19578369

ABSTRACT

Interleukin 22 (IL-22) is a cytokine produced by the T(H)-17 lineage of helper T cells and NK-22 subset of natural killer cells that acts on epithelial cells and keratinocytes and has been linked to skin homeostasis and inflammation. Here we characterize a population of human skin-homing memory CD4(+) T cells that expressed the chemokine receptors CCR10, CCR6 and CCR4 and produced IL-22 but neither IL-17 nor interferon-gamma (IFN-gamma). Clones isolated from this population produced IL-22 only and had low or undetectable expression of the T(H)-17 and T helper type 1 (T(H)1) transcription factors RORgammat and T-bet. The differentiation of T cells producing only IL-22 was efficiently induced in naive T cells by plasmacytoid dendritic cells in an IL-6- and tumor necrosis factor-dependent way. Our findings delineate a previously unknown subset of human CD4(+) effector T cells dedicated to skin pathophysiology.


Subject(s)
Interleukin-17/biosynthesis , Interleukins/biosynthesis , Skin/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation , Humans , Immunologic Memory , In Vitro Techniques , Nuclear Receptor Subfamily 1, Group F, Member 3 , Receptors, CCR10/metabolism , Receptors, CCR4/metabolism , Receptors, CCR6/metabolism , Receptors, Retinoic Acid/metabolism , Receptors, Thyroid Hormone/metabolism , T-Box Domain Proteins/metabolism , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology , Interleukin-22
12.
FASEB J ; 34(4): 5838-5850, 2020 04.
Article in English | MEDLINE | ID: mdl-32124475

ABSTRACT

Chemokines and their receptors play important roles in vascular homeostasis, development, and angiogenesis. Little is known regarding the molecular signaling mechanisms activated by CCL28 chemokine via its primary receptor CCR10 in endothelial cells (ECs). Here, we test the hypothesis that CCL28/CCR10 signaling plays an important role in regulating skin wound angiogenesis through endothelial nitric oxide synthase (eNOS)-dependent Src, PI3K, and MAPK signaling. We observed nitric oxide (NO) production in human primary ECs stimulated with exogenous CCL28, which also induced direct binding of CCR10 and eNOS resulting in inhibition of eNOS activity. Knockdown of CCR10 with siRNA lead to reduced eNOS expression and tube formation suggesting the involvement of CCR10 in EC angiogenesis. Based on this interaction, we engineered a myristoylated 7 amino acid CCR10-binding domain (Myr-CBD7) peptide and showed that this can block eNOS interaction with CCR10, but not with calmodulin, resulting in upregulation of eNOS activity. Importantly, topical administration of Myr-CBD7 peptide on mouse dermal wounds not only blocked CCR10-eNOS interaction, but also enhanced expression of eNOS, CD31, and IL-4 with reduction of CCL28 and IL-6 levels associated with improved wound healing. These results point to a potential therapeutic strategy to upregulate NO bioavailability, enhance angiogenesis, and improve wound healing by disrupting CCL28-activated CCR10-eNOS interaction.


Subject(s)
Chemokines, CC/metabolism , Neovascularization, Physiologic , Nitric Oxide Synthase Type III/metabolism , Receptors, CCR10/metabolism , Skin/physiopathology , Wound Healing , Animals , Chemokines, CC/genetics , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type III/genetics , Receptors, CCR10/genetics , Skin/injuries
13.
Int J Mol Sci ; 22(8)2021 Apr 19.
Article in English | MEDLINE | ID: mdl-33921794

ABSTRACT

Chemokines interact with chemokine receptors in a promiscuous network, such that each receptor can be activated by multiple chemokines. Moreover, different chemokines have been reported to preferentially activate different signalling pathways via the same receptor, a phenomenon known as biased agonism. The human CC chemokine receptors (CCRs) CCR4, CCR7 and CCR10 play important roles in T cell trafficking and have been reported to display biased agonism. To systematically characterize these effects, we analysed G protein- and ß-arrestin-mediated signal transduction resulting from stimulation of these receptors by each of their cognate chemokine ligands within the same cellular background. Although the chemokines did not elicit ligand-biased agonism, the three receptors exhibited different arrays of signaling outcomes. Stimulation of CCR4 by either CC chemokine ligand 17 (CCL17) or CCL22 induced ß-arrestin recruitment but not G protein-mediated signaling, suggesting that CCR4 has the potential to act as a scavenger receptor. At CCR7, both CCL19 and CCL21 stimulated G protein signaling and ß-arrestin recruitment, with CCL19 consistently displaying higher potency. At CCR10, CCL27 and CCL28(4-108) stimulated both G protein signaling and ß-arrestin recruitment, whereas CCL28(1-108) was inactive, suggesting that CCL28(4-108) is the biologically relevant form of this chemokine. These comparisons emphasize the intrinsic abilities of different receptors to couple with different downstream signaling pathways. Comparison of these results with previous studies indicates that differential agonism at these receptors may be highly dependent on the cellular context.


Subject(s)
Chemokines/metabolism , Receptors, CCR10/metabolism , Receptors, CCR4/metabolism , Receptors, CCR7/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Receptors, CCR/genetics , Receptors, CCR/metabolism , Receptors, CCR10/genetics , Receptors, CCR4/genetics , Receptors, CCR7/genetics , Signal Transduction/genetics , Signal Transduction/physiology
14.
Nat Immunol ; 9(11): 1253-60, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18836452

ABSTRACT

Dendritic cells (DCs) are 'professional' antigen-presenting cells that are key in the regulation of immune responses. Here we characterize a unique subset of tolerogenic DCs that expressed the chemokine receptor CCR9 and migrated to the CCR9 ligand CCL25, a chemokine linked to the homing of T cells and DCs to the gut. CCR9(+) DCs were of the plasmacytoid DC (pDC) lineage, had an immature phenotype and rapidly downregulated CCR9 in response to maturation-inducing pDC-restricted Toll-like receptor ligands. CCR9(+) pDCs were potent inducers of regulatory T cell function and suppressed antigen-specific immune responses both in vitro and in vivo, including inhibiting acute graft-versus-host disease induced by allogeneic CD4(+) donor T cells in irradiated recipients. Our results identify a highly immunosuppressive population of pDCs present in lymphoid tissues.


Subject(s)
Dendritic Cells/immunology , Graft vs Host Disease/immunology , Receptors, CCR10/immunology , Self Tolerance , Transplantation Tolerance , Animals , Biomarkers/metabolism , Cell Differentiation , Cell Line, Tumor , Cell Movement , Chemokines, CC/metabolism , Dendritic Cells/metabolism , Down-Regulation , Intestines/immunology , Leukocyte Common Antigens/metabolism , Ligands , Mice , Mice, Congenic , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptors, CCR10/metabolism , T-Lymphocytes, Regulatory/immunology , Toll-Like Receptors/metabolism , Chemokine Receptor D6
15.
Allergy ; 74(5): 933-943, 2019 05.
Article in English | MEDLINE | ID: mdl-30475388

ABSTRACT

BACKGROUND: We previously showed that patients with severe allergic asthma have high numbers of circulating ILC2s expressing CCR10. METHOD: Herein, CCR10+ ILC2s were further analyzed in the blood of healthy individuals or patients with allergic and non-allergic asthma. Characteristics of human CCR10+ and CCR10- ILC2s were assessed by flow cytometry as well as single-cell multiplex RT-qPCR. The role of CCR10+ ILC2s in asthma pathophysiology was studied in allergen-treated mice. RESULTS: When compared to healthy controls, CCR10+ ILC2s are enriched in the blood of both allergic and non-allergic severe asthmatic patients, and these cells are recruited to the lungs. Plasma concentrations of the CCR10 ligand CCL27 are significantly increased in severe asthmatics when compared to non-asthmatic patients. CCR10+ ILC2s secrete little TH 2 cytokines, but exhibit ILC1-like properties, including a capacity to produce IFN-γ. Also, single-cell analysis reveals that the CCR10+ ILC2 subset is enriched in cells expressing amphiregulin. CCR10+ ILC2 depletion, as well as blocking of IFN-γ activity, exacerbates airway hyperreactivity in allergen-challenged mice, providing evidence for a protective role of these cells in allergic inflammation. CONCLUSIONS: Frequencies of circulating CCR10+ ILC2s and CCL27 plasma concentrations represent candidate markers of asthma severity. The characterization of CCR10+ ILC2s in human samples and in mouse asthma models suggests that these cells downregulate allergic inflammation through IFN-γ production.


Subject(s)
Asthma/immunology , Asthma/metabolism , Immunity, Innate , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Receptors, CCR10/metabolism , Allergens/immunology , Animals , Asthma/diagnosis , Asthma/physiopathology , Biomarkers , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility , Humans , Interferon-gamma/biosynthesis , Lymphocyte Count , Lymphocyte Subsets/drug effects , Mice , Severity of Illness Index
16.
J Immunol ; 199(4): 1393-1404, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28674178

ABSTRACT

The engulfment of apoptotic polymorphonuclear cells (PMN) during the resolution of inflammation leads to macrophage reprogramming culminating in reduced proinflammatory and increased anti-inflammatory mediator secretion. The atypical chemokine receptor D6/ACKR2 is expressed on apoptotic PMN and plays an important role in regulating macrophage properties during and after engulfment. In this study, we found that the inflammatory chemokine CCL5 is mostly retained (75%) during the resolution of zymosan A peritonitis in mice. Moreover, this chemokine is secreted by resolution-phase macrophages (2.5 ng/ml) and promotes their reprogramming in vivo in D6+/+ mice (2-fold increase in IL-10/IL-12 ratio) but not their D6-/- counterparts. In addition, CCL5 enhanced macrophage reprogramming ex vivo exclusively when bound to D6+/+ apoptotic PMN. Signaling through p38MAPK and JNK in reprogrammed macrophages was enhanced by CCL5-bound apoptotic PMN (3.6-4 fold) in a D6-dependent manner, and was essential for reprogramming. Thus, CCL5 exerts a novel proresolving role on macrophages when acting in concert with apoptotic PMN-expressed D6.


Subject(s)
Apoptosis , Chemokine CCL5/immunology , Chemokine CCL5/metabolism , Macrophages/physiology , Neutrophils/immunology , Peritonitis/immunology , Receptors, CCR10/metabolism , Animals , Chemokine CCL5/pharmacology , Gene Expression Regulation , Inflammation/metabolism , Macrophages/immunology , Mice , Peritonitis/chemically induced , Protein Binding , Receptors, CCR10/genetics , Receptors, CCR10/immunology , Zymosan/administration & dosage , Chemokine Receptor D6
17.
J Immunol ; 199(7): 2451-2459, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28855310

ABSTRACT

Infection or inflammation of the skin recruits effector CD8+ T cells that enter the epidermis and form populations of long-lived tissue-resident memory T (TRM) cells. These skin TRM cells migrate within the constrained epidermal environment by extending multiple dynamic dendritic projections and squeezing between keratinocytes to survey the tissue for pathogens. In this study, we examined the signals required for this distinctive mode of T cell migration by inhibiting key cytoskeletal components and performing intravital two-photon microscopy to visualize TRM cell behavior. We found that TRM cell motility and dendrite formation required an intact actomyosin cytoskeleton and the Rho-associated coiled-coil containing kinases. We also identified an essential role for microtubules for maintaining skin TRM cell shape and cellular integrity. We reveal a role for pertussis toxin-sensitive signaling for TRM cell dendritic morphology and migration that is independent of CXCR3 or CXCR6, or the skin-selective chemokine receptors CCR10 and CCR8. However, we found that CXCR6 and CCR10 expression by CD8+ T cells was required for the optimal formation of memory T cell populations, in particular TRM cell populations in the skin.


Subject(s)
CD8-Positive T-Lymphocytes/physiology , Cell Movement , Epidermis/immunology , Immunologic Memory , Receptors, Chemokine/metabolism , Skin/immunology , Actomyosin/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/physiology , Epidermal Cells , Intravital Microscopy/methods , Mice , Microtubules/metabolism , Pertussis Toxin/metabolism , Receptors, CCR10/genetics , Receptors, CCR10/metabolism , Receptors, CCR8/metabolism , Receptors, CXCR/genetics , Receptors, CXCR/metabolism , Receptors, CXCR3/metabolism , Receptors, CXCR6 , Receptors, Chemokine/genetics , Signal Transduction , Skin/anatomy & histology , Skin/cytology , rho-Associated Kinases/metabolism
18.
Acta Derm Venereol ; 99(9): 809-812, 2019 Jul 01.
Article in English | MEDLINE | ID: mdl-31045236

ABSTRACT

Tumor cells in cutaneous T-cell lymphoma express limited numbers of chemokine receptors. We investigated the expression patterns of CXCR3, CCR3, CCR4 and CCR10 in mycosis fungoides, Sézary syndrome, lym-phomatoid papulosis and anaplastic large cell lymphoma in 121 skin biopsy samples. CXCR3 was expressed in 86% of mycosis fungoides cases but in no anaplastic large cell lymphoma cases. CCR3 was expressed in 73% of cases of CD30+ lymphoproliferative disorders such as lymphomatoid papulosis and anaplastic large cell lymphoma. Mycosis fungoides/Sézary syndrome patients with high CCR3 or CCR4 expression had a poorer survival prognosis than mycosis fungoides/Sézary syndrome patients whose tumor cells did not express these receptors. CCR10 was expressed in 50% of mycosis fungoides/Sézary syndrome cases and in 13% of cases with CD30+ lym-phoproliferative disorders. These results suggest that differential patterns of CXCR3, CCR3, CCR4 and CCR10 expression are useful for the diagnosis of cutaneous T-cell lymphoma. Moreover, expression of CCR3 or CCR4 suggests a poor prognosis in mycosis fungoides/Sézary syndrome.


Subject(s)
Biomarkers, Tumor/analysis , Mycosis Fungoides/immunology , Receptors, CCR3/analysis , Receptors, CCR4/analysis , Sezary Syndrome/immunology , Skin Neoplasms/immunology , Humans , Mycosis Fungoides/mortality , Mycosis Fungoides/pathology , Prognosis , Receptors, CCR10/analysis , Receptors, CXCR3/analysis , Sezary Syndrome/mortality , Sezary Syndrome/pathology , Skin Neoplasms/mortality , Skin Neoplasms/pathology , Up-Regulation
19.
J Virol ; 91(19)2017 10 01.
Article in English | MEDLINE | ID: mdl-28701399

ABSTRACT

Herpes simplex virus (HSV) infection is restricted to epithelial cells and neurons and is controlled by CD8 T cells. These cells both traffic to epithelial sites of recurrent lytic infection and to ganglia and persist at the dermal-epidermal junction for up to 12 weeks after lesion resolution. We previously showed that cutaneous lymphocyte-associated antigen (CLA), a functional E-selectin ligand (ESL), is selectively expressed on circulating HSV-2-specific CD8 T cells. CLA/ESL mediates adhesion of T cells to inflamed vascular endothelium. Later stages in T-cell homing involve chemokines (Ch) and lymphocyte chemokine receptors (ChR) for vascular wall arrest and diapedesis. Several candidate ChR have been implicated in skin homing. We measured cell surface ChR on HSV-specific human peripheral blood CD8 T cells and extended our studies to HSV-1. We observed preferential cell surface expression of CCR10 and CXCR3 by HSV-specific CD8 T cells compared to CD8 T cells specific for control viruses, Epstein-Barr virus (EBV) and cytomegalovirus (CMV), and compared to bulk memory CD8 T cells. CXCR3 ligand mRNA levels were selectively increased in skin biopsy specimens from persons with recurrent HSV-2, while the mRNA levels of the CCR10 ligand CCL27 were equivalent in lesion and control skin. Our data are consistent with a model in which CCL27 drives baseline recruitment of HSV-specific CD8 T cells expressing CCR10, while interferon-responsive CXCR3 ligands recruit additional cells in response to virus-driven inflammation.IMPORTANCE HSV-2 causes very localized recurrent infections in the skin and genital mucosa. Virus-specific CD8 T cells home to the site of recurrent infection and participate in viral clearance. The exit of T cells from the blood involves the use of chemokine receptors on the T-cell surface and chemokines that are present in infected tissue. In this study, circulating HSV-2-specific CD8 T cells were identified using specific fluorescent tetramer reagents, and their expression of several candidate skin-homing-associated chemokine receptors was measured using flow cytometry. We found that two chemokine receptors, CXCR3 and CCR10, are upregulated on HSV-specific CD8 T cells in blood. The chemokines corresponding to these receptors are also expressed in infected tissues. Vaccine strategies to prime CD8 T cells to home to HSV lesions should elicit these chemokine receptors if possible to increase the homing of vaccine-primed cells to sites of infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Chemokine CCL27/immunology , Herpes Simplex/immunology , Herpesvirus 2, Human/immunology , Lymphocyte Activation/immunology , Receptors, CCR10/immunology , Receptors, CXCR3/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , CD8-Positive T-Lymphocytes/metabolism , Chemokine CCL27/genetics , Cytomegalovirus/immunology , Female , Flow Cytometry , Herpes Simplex/virology , Herpesvirus 4, Human/immunology , Humans , Immunologic Memory/immunology , Male , Membrane Glycoproteins/immunology , RNA, Messenger/genetics , Receptors, CCR10/biosynthesis , Receptors, CCR10/genetics , Receptors, CXCR3/biosynthesis , Receptors, CXCR3/genetics , Skin/virology
20.
Immunity ; 30(1): 120-9, 2009 Jan 16.
Article in English | MEDLINE | ID: mdl-19144318

ABSTRACT

Mucosal immunoglobulin A (IgA) secreted by local plasma cells (PCs) is a critical component of mucosal immunity. Although IgA class switching can occur at mucosal sites, high-affinity PCs are optimally generated in germinal centers (GCs) in a T cell-dependent fashion. However, how CD4(+) helper T cells induce mucosal-homing IgA-PCs remains unclear. Here, we show that transforming growth factor beta1 (TGFbeta1) and interleukin 21 (IL-21), produced by follicular helper T cells (Tfh), synergized to generate abundant IgA-plasmablasts (PBs). In the presence of IL-21, TGFbeta1 promoted naive B cell proliferation and differentiation and overrode IL-21-induced IgG class switching in favor of IgA. Furthermore, TGFbeta1 and IL-21 downregulated CXCR5 while upregulating CCR10 on plasmablasts, enabling their exit from GCs and migration toward local mucosa. This was supported by the presence of CCR10(+)IgA(+)PBs in tonsil GCs. These findings show that Tfh contribute to mucosal IgA. Thus, mucosal vaccines should aim to induce robust Tfh responses.


Subject(s)
Chemotaxis, Leukocyte , Immunoglobulin A/metabolism , Mucous Membrane/immunology , Plasma Cells/immunology , T-Lymphocytes/immunology , B-Lymphocytes/immunology , Cell Differentiation , Cell Proliferation , Child , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin A/classification , Interleukin-2/genetics , Interleukin-2/metabolism , Interleukins/physiology , Polymerase Chain Reaction , Receptors, CCR10/metabolism , Receptors, CXCR5/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Reference Standards , Transforming Growth Factor beta1/physiology , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL