Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
Cell ; 163(1): 160-73, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26406376

ABSTRACT

Focal adhesion kinase (FAK) promotes anti-tumor immune evasion. Specifically, the kinase activity of nuclear-targeted FAK in squamous cell carcinoma (SCC) cells drives exhaustion of CD8(+) TĀ cells and recruitment of regulatory TĀ cells (Tregs) in the tumor microenvironment by regulating chemokine/cytokine and ligand-receptor networks, including via transcription of Ccl5, which is crucial. These changes inhibit antigen-primed cytotoxic CD8(+) TĀ cell activity, permitting growth of FAK-expressing tumors. Mechanistically, nuclear FAK is associated with chromatin and exists in complex with transcription factors and their upstream regulators that control Ccl5 expression. Furthermore, FAK's immuno-modulatory nuclear activities may be specific to cancerous squamous epithelial cells, as normal keratinocytes do not have nuclear FAK. Finally, we show that a small-molecule FAK kinase inhibitor, VS-4718, which is currently in clinical development, also drives depletion of Tregs and promotes a CD8(+) TĀ cell-mediated anti-tumor response. Therefore, FAK inhibitors may trigger immune-mediated tumor regression, providing previously unrecognized therapeutic opportunities.


Subject(s)
Carcinoma, Squamous Cell/immunology , Chemokine CCL5/genetics , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Skin Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Escape , Aminopyridines/administration & dosage , Animals , Carcinoma, Squamous Cell/metabolism , Chemokine CCL5/immunology , Disease Models, Animal , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Focal Adhesion Protein-Tyrosine Kinases/genetics , Humans , Keratinocytes/metabolism , Mice , Mice, Nude , Skin Neoplasms/metabolism , Transcription, Genetic
2.
Nat Immunol ; 15(7): 623-30, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24813163

ABSTRACT

Afferent lymph-borne dendritic cells essentially rely on the chemokine receptor CCR7 for their transition from the subcapsular lymph node sinus into the parenchyma, a migratory step driven by putative gradients of CCR7 ligands. We found that lymph node fringes indeed contained physiological gradients of the chemokine CCL21, which depended on the expression of CCRL1, the atypical receptor for the CCR7 ligands CCL19 and CCL21. Lymphatic endothelial cells lining the ceiling of the subcapsular sinus, but not those lining the floor, expressed CCRL1, which scavenged chemokines from the sinus lumen. This created chemokine gradients across the sinus floor and enabled the emigration of dendritic cells. In vitro live imaging revealed that spatially confined expression of CCRL1 was necessary and sufficient for the creation of functional chemokine gradients.


Subject(s)
Chemokine CCL21/physiology , Lymph Nodes/immunology , Receptors, CCR/physiology , Animals , Cell Movement , Dendritic Cells/physiology , Mice , Mice, Inbred C57BL
3.
Proc Natl Acad Sci U S A ; 116(51): 25828-25838, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31772019

ABSTRACT

Proinflammatory responses induced by Toll-like receptors (TLRs) are dependent on the activation of the NF-ƄĀøB and mitogen-activated protein kinase (MAPK) pathways, which coordinate the transcription and synthesis of proinflammatory cytokines. We demonstrate that BCL-3, a nuclear IƄĀøB protein that regulates NF-ƄĀøB, also controls TLR-induced MAPK activity by regulating the stability of the TPL-2 kinase. TPL-2 is essential for MAPK activation by TLR ligands, and the rapid proteasomal degradation of active TPL-2 is a critical mechanism limiting TLR-induced MAPK activity. We reveal that TPL-2 is a nucleocytoplasmic shuttling protein and identify the nucleus as the primary site for TPL-2 degradation. BCL-3 interacts with TPL-2 and promotes its degradation by promoting its nuclear localization. As a consequence, Bcl3-/- macrophages have increased TPL-2 stability following TLR stimulation, leading to increased MAPK activity and MAPK-dependent responses. Moreover, BCL-3-mediated regulation of TPL-2 stability sets the MAPK activation threshold and determines the amount of TLR ligand required to initiate the production of inflammatory cytokines. Thus, the nucleus is a key site in the regulation of TLR-induced MAPK activity. BCL-3 links control of the MAPK and NF-ƄĀøB pathways in the nucleus, and BCL-3-mediated TPL-2 regulation impacts on the cellular decision to initiate proinflammatory cytokine production in response to TLR activation.


Subject(s)
B-Cell Lymphoma 3 Protein/metabolism , Cell Nucleus/metabolism , I-kappa B Proteins/metabolism , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System/physiology , Proto-Oncogene Proteins/metabolism , Toll-Like Receptors/metabolism , Animals , B-Cell Lymphoma 3 Protein/genetics , Cytokines/metabolism , Gene Expression Regulation , HEK293 Cells , Humans , Macrophages/metabolism , Mice , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Nuclear Proteins/metabolism , RAW 264.7 Cells
4.
Nat Immunol ; 10(1): 101-8, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19060902

ABSTRACT

The Duffy antigen receptor for chemokines (DARC) belongs to a family of 'silent' heptahelical chemokine receptors that do not couple to G proteins and fail to transmit measurable intracellular signals. DARC binds most inflammatory chemokines and is prominently expressed on venular endothelial cells, where its function has remained contentious. Here we show that DARC, like other silent receptors, internalized chemokines but did not effectively scavenge them. Instead, DARC mediated chemokine transcytosis, which led to apical retention of intact chemokines and more leukocyte migration across monolayers expressing DARC. Mice overexpressing DARC on blood vessel endothelium had enhanced chemokine-induced leukocyte extravasation and contact-hypersensitivity reactions. Thus, interactions of chemokines with DARC support their activity on apposing leukocytes in vitro and in vivo.


Subject(s)
Cell Movement , Chemokines/metabolism , Duffy Blood-Group System/metabolism , Leukocytes/immunology , Receptors, Cell Surface/metabolism , Animals , Cells, Cultured , Dogs , Duffy Blood-Group System/genetics , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Humans , Mice , Protein Transport , Receptors, Cell Surface/genetics
5.
Immunol Invest ; 49(3): 264-286, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31429329

ABSTRACT

Homeostatic leukocyte trafficking into and within the female reproductive tract (FRT) contributes to fertility and reproductive health. It is unclear how this process is regulated in the anatomically distinct reproductive tissues, or whether the genes involved are affected by cyclical changes in reproductive hormones. In tissues such as skin and intestine, mouse studies have defined evolutionarily conserved molecular mechanisms for tissue-specific homing, interstitial positioning, and leukocyte egress. Chemokine family members are invariably involved, with the chemokine expression profile of a tissue regulating leukocyte content. Reproductive tissues (ovary, vagina, cervix, uterine horn) of 8Ā week old virgin female C57BL/6 mice (n =Ā 20) were collected, and expression of mRNA for leukocyte markers and chemokines conducted by qPCR. Lymphocytic and myeloid cell populations within the uterus, cervix, bone marrow and PALN from virgin C57BL/6 mice were determined by flow cytometric analysis. Variation in leukocyte content between reproductive tissues is evident, with the uterus and cervix containing complex mixtures of lymphocytes and myeloid cells. Twenty-six chemokine genes are expressed in the FRT, many by several component tissues, some preferentially by one. Most striking are Xcl1 and Ccl28, which are restricted to the uterus. Ccl20 and genes encoding CXCR2 ligands are primarily transcribed in cervix and vagina. Ovary shows the lowest expression of most chemokine genes, with the notable exception of Ccl21 and Ccl27. We also identify eight chemokines in the vagina whose expression fluctuates substantially across the oestrous cycle. These data reveal complex chemokine networks within the FRT, and provide a framework for future studies of homeostatic leukocyte trafficking into and within these tissues.Abbreviations: BM: bone marrow; DC: dendritic cell; DN: double negative; FRT: female reproductive tract; FSC: forward scatter; NK: natural killer; PALN: para-aortic lymph node; SSC: side scatter; Tregs: regulatory T cells.


Subject(s)
Chemokines/genetics , Genitalia, Female/metabolism , Animals , Estrous Cycle/immunology , Female , Gene Expression Profiling , Genitalia, Female/cytology , Leukocytes/metabolism , Mice , Mice, Inbred C57BL , Myeloid Cells/metabolism , Organ Specificity/immunology
6.
J Immunol ; 201(1): 215-229, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29760193

ABSTRACT

Atypical chemokine receptors (ACKRs) are expressed by discrete populations of stromal cells at specific anatomical locations where they control leukocyte migration by scavenging or transporting chemokines. ACKR4 is an atypical receptor for CCL19, CCL21, and CCL25. In skin, ACKR4 plays indispensable roles in regulating CCR7-dependent APC migration, and there is a paucity of migratory APCs in the skin-draining lymph nodes of Ackr4-deficient mice under steady-state and inflammatory conditions. This is caused by loss of ACKR4-mediated CCL19/21 scavenging by keratinocytes and lymphatic endothelial cells. In contrast, we show in this study that Ackr4 deficiency does not affect dendritic cell abundance in the small intestine and mesenteric lymph nodes, at steady state or after R848-induced mobilization. Moreover, Ackr4 expression is largely restricted to mesenchymal cells in the intestine, where it identifies a previously uncharacterized population of fibroblasts residing exclusively in the submucosa. Compared with related Ackr4- mesenchymal cells, these Ackr4+ fibroblasts have elevated expression of genes encoding endothelial cell regulators and lie in close proximity to submucosal blood and lymphatic vessels. We also provide evidence that Ackr4+ fibroblasts form physical interactions with lymphatic endothelial cells, and engage in molecular interactions with these cells via the VEGFD/VEGFR3 and CCL21/ACKR4 pathways. Thus, intestinal submucosal fibroblasts in mice are a distinct population of intestinal mesenchymal cells that can be identified by their expression of Ackr4 and have transcriptional and anatomical properties that strongly suggest roles in endothelial cell regulation.


Subject(s)
Endothelial Cells/metabolism , Fibroblasts/metabolism , Intestinal Mucosa/metabolism , Receptors, CCR/metabolism , Animals , Cell Movement/physiology , Chemokine CCL21/metabolism , Colitis/chemically induced , Colitis/pathology , Dendritic Cells/cytology , Dextran Sulfate/toxicity , Female , Intestinal Mucosa/cytology , Leukocytes/physiology , Mesoderm/cytology , Mesoderm/metabolism , Mice, Inbred C57BL , Mice, Knockout , Receptors, CCR/genetics , Vascular Endothelial Growth Factor D/metabolism , Vascular Endothelial Growth Factor Receptor-3/metabolism
7.
J Immunol ; 201(8): 2510-2519, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30158126

ABSTRACT

Chemokines have been shown to be essential players in a range of cancer contexts. In this study, we demonstrate that mice deficient in the atypical chemokine receptor Ackr2 display impaired development of metastasis in vivo in both cell line and spontaneous models. Further analysis reveals that this relates to increased expression of the chemokine receptor CCR2, specifically by KLRG1+ NK cells from the Ackr2-/- mice. This leads to increased recruitment of KLRG1+ NK cells to CCL2-expressing tumors and enhanced tumor killing. Together, these data indicate that Ackr2 limits the expression of CCR2 on NK cells and restricts their tumoricidal activity. Our data have important implications for our understanding of the roles for chemokines in the metastatic process and highlight Ackr2 and CCR2 as potentially manipulable therapeutic targets in metastasis.


Subject(s)
Killer Cells, Natural/immunology , Neoplasms, Experimental/immunology , Receptors, Chemokine/metabolism , Animals , Carcinoma, Lewis Lung , Cell Movement , Chemokine CCL2/metabolism , Cytotoxicity, Immunologic , Lectins, C-Type , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Metastasis , Receptors, CCR2/metabolism , Receptors, Chemokine/genetics , Receptors, Immunologic/metabolism
8.
Pharmacol Res ; 145: 104258, 2019 07.
Article in English | MEDLINE | ID: mdl-31063806

ABSTRACT

Greater understanding of tumour immunobiology has led to a new era of cancer treatment in which immuno-oncology (IO) therapies are used to boost anti-cancer immune responses. Prominent among these therapies are immune checkpoint inhibitors (ICIs), antibody-based drugs that can unleash the power of tumour-specific CD8 + T-cells. ICIs targeting the Programmed cell death protein 1 (PD-1) cell surface receptor or its ligand PD-L1 are particularly effective, with clinical studies reporting powerful and durable therapeutic impact against many cancer types, including melanoma and non-small cell lung cancer. ICIs have the potential to transform the landscape of cancer treatment, and their development was recognised by the award of the 2018 Nobel Prize in Physiology or Medicine to James Allison and Tasuku Honjo. However, the proportion of patients responding to anti-PD-1/PD-L1 monotherapy can be low. The next major challenge involves understanding and overcoming the innate and acquired resistance that prevents most patients from responding to PD-1/PD-L1 blockade. In this review, we outline the physiological function of PD-1 and its exploitation by developing tumours. We give an overview of current FDA-approved drugs targeting PD-1 or PD-L1 and summarise clinical progress so far. We then discuss key mechanisms thought to underpin resistance to PD-1/PD-L1 blockade, describing biomarkers that could allow patient responses to be predicted before treatment, and tracked once treatment has started. We also present clinical and pre-clinical combination therapies that have been developed to overcome resistance and which have the potential to substantially extend the therapeutic reach of these revolutionary drugs.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , Drug Resistance, Neoplasm/drug effects , Neoplasms/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , B7-H1 Antigen/immunology , Humans , Immune Evasion/drug effects , Neoplasms/immunology , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes/immunology
9.
Pharmacol Res ; 148: 104442, 2019 10.
Article in English | MEDLINE | ID: mdl-31491469

ABSTRACT

Inflammatory bowel disease (IBD) is a debilitating chronic inflammatory disease of the gastrointestinal (GI) tract. It affects more than 3.5 million people in the western world and places a huge financial burden on healthcare systems. IBD is highly heterogeneous; disease severity and outcomes in IBD are highly variable, and patients may experience episodes of relapse and remission. However, treatment often follows a step-up model whereby the patients start with anti-inflammatory agents (corticosteroids or immunosuppressants) and step-up to monoclonal anti-tumour necrosis factor-α (TNFα) antibodies and then other biologics if the initial drugs cannot control disease. Unfortunately, many patients do not respond to the costly biologics, and thus often still require gut-resective surgery, which decreases quality of life. In order to decrease rates of surgery and ineffective treatments, it is important to identify markers that accurately predict disease progression and treatment responses, to inform decisions about the best choice of therapeutics. Here we examine molecular approaches to patient stratification that aim to increase the effectiveness of treatments and potentially reduce healthcare costs. In the future, it may become possible to stratify patients based on their suitability for specific molecular-targeted therapeutic agents, and eventually use molecular stratification for personalised medicine in IBD.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Inflammatory Bowel Diseases/drug therapy , Biomarkers/metabolism , Disease Progression , Humans , Inflammatory Bowel Diseases/metabolism , Tumor Necrosis Factor-alpha/metabolism
10.
J Immunol ; 199(7): 2291-2304, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28807994

ABSTRACT

The chemokine receptor CCR7 drives leukocyte migration into and within lymph nodes (LNs). It is activated by chemokines CCL19 and CCL21, which are scavenged by the atypical chemokine receptor ACKR4. CCR7-dependent navigation is determined by the distribution of extracellular CCL19 and CCL21, which form concentration gradients at specific microanatomical locations. The mechanisms underpinning the establishment and regulation of these gradients are poorly understood. In this article, we have incorporated multiple biochemical processes describing the CCL19-CCL21-CCR7-ACKR4 network into our model of LN fluid flow to establish a computational model to investigate intranodal chemokine gradients. Importantly, the model recapitulates CCL21 gradients observed experimentally in B cell follicles and interfollicular regions, building confidence in its ability to accurately predict intranodal chemokine distribution. Parameter variation analysis indicates that the directionality of these gradients is robust, but their magnitude is sensitive to these key parameters: chemokine production, diffusivity, matrix binding site availability, and CCR7 abundance. The model indicates that lymph flow shapes intranodal CCL21 gradients, and that CCL19 is functionally important at the boundary between B cell follicles and the T cell area. It also predicts that ACKR4 in LNs prevents CCL19/CCL21 accumulation in efferent lymph, but does not control intranodal gradients. Instead, it attributes the disrupted interfollicular CCL21 gradients observed in Ackr4-deficient LNs to ACKR4 loss upstream. Our novel approach has therefore generated new testable hypotheses and alternative interpretations of experimental data. Moreover, it acts as a framework to investigate gradients at other locations, including those that cannot be visualized experimentally or involve other chemokines.


Subject(s)
Cell Movement , Chemokine CCL19/metabolism , Computer Simulation , Lymph Nodes/physiology , Receptors, CCR/metabolism , Animals , B-Lymphocytes/immunology , Chemokine CCL19/genetics , Chemokine CCL19/immunology , Dendritic Cells/immunology , Humans , Lymph Nodes/immunology , Mice , Receptors, CCR/deficiency , Receptors, CCR/genetics , Receptors, CCR/immunology , Receptors, CCR7/immunology , T-Lymphocytes/immunology
11.
EMBO J ; 33(21): 2564-80, 2014 Nov 03.
Article in English | MEDLINE | ID: mdl-25271254

ABSTRACT

Macrophages regulate lymphatic vasculature development; however, the molecular mechanisms regulating their recruitment to developing, and adult, lymphatic vascular sites are not known. Here, we report that resting mice deficient for the inflammatory chemokine-scavenging receptor, ACKR2, display increased lymphatic vessel density in a range of tissues under resting and regenerating conditions. This appears not to alter dendritic cell migration to draining lymph nodes but is associated with enhanced fluid drainage from peripheral tissues and thus with a hypotensive phenotype. Examination of embryonic skin revealed that this lymphatic vessel density phenotype is developmentally established. Further studies indicated that macrophages and the inflammatory CC-chemokine CCL2, which is scavenged by ACKR2, are associated with this phenotype. Accordingly, mice deficient for the CCL2 signalling receptor, CCR2, displayed a reciprocal phenotype of reduced lymphatic vessel density. Further examination revealed that proximity of pro-lymphangiogenic macrophages to developing lymphatic vessel surfaces is increased in ACKR2-deficient mice and reduced in CCR2-deficient mice. Therefore, these receptors regulate vessel density by reciprocally modulating pro-lymphangiogenic macrophage recruitment, and proximity, to developing, resting and regenerating lymphatic vessels.


Subject(s)
Embryo, Mammalian/embryology , Lymphangiogenesis/physiology , Lymphatic Vessels/embryology , Macrophages/metabolism , Receptors, CCR2/metabolism , Receptors, Chemokine/metabolism , Animals , Embryo, Mammalian/cytology , Lymph Nodes/cytology , Lymph Nodes/embryology , Lymphatic Vessels/cytology , Macrophages/cytology , Mice , Mice, Knockout , Receptors, CCR2/genetics , Receptors, Chemokine/genetics , Skin/cytology , Skin/embryology
12.
J Immunol ; 196(8): 3341-53, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26976955

ABSTRACT

Dermal dendritic cells and epidermal Langerhans cells are APCs that migrate from skin to draining lymph nodes (LN) to drive peripheral tolerance and adaptive immunity. Their migration requires the chemokine receptor CCR7, which directs egress from the skin via dermal lymphatic vessels and extravasation into the LN parenchyma from lymph in the subcapsular sinus. CCR7 is activated by two chemokines: CCL19 and CCL21. CCL21 alone is sufficient for the migration of APCs from skin to LN. CCL19 and CCL21 also bind atypical chemokine receptor (ACKR) 4. ACKR4-mediated CCL21 scavenging by lymphatic endothelial cells lining the subcapsular sinus ceiling stabilizes interfollicular CCL21 gradients that direct lymph-borne CCR7(+)APCs into the parenchyma of mouse LN. In this study, we show that ACKR4 also aids APC egress from mouse skin under steady-state and inflammatory conditions. ACKR4 plays a particularly prominent role during cutaneous inflammation when it facilitates Langerhans cell egress from skin and enables the accumulation of dermal dendritic cells in skin-draining LN. Stromal cells in mouse skin, predominantly keratinocytes and a subset of dermal lymphatic endothelial cells, express ACKR4 and are capable of ACKR4-dependent chemokine scavenging in situ. ACKR4-mediated scavenging of dermal-derived CCL19, rather than CCL21, is critical during inflammation, because the aberrant trafficking of skin-derived APCs inAckr4-deficient mice is completely rescued by genetic deletion ofCcl19 Thus, ACKR4 on stromal cells aids the egress of APCs from mouse skin, and, during inflammation, facilitates CCR7-dependent cell trafficking by scavenging CCL19.


Subject(s)
Chemokine CCL19/metabolism , Dendritic Cells/immunology , Receptors, CCR7/metabolism , Receptors, CCR/metabolism , Skin/pathology , Animals , Cell Movement/immunology , Chemokine CCL19/genetics , Chemokine CCL21/metabolism , Endothelial Cells/metabolism , Inflammation/immunology , Inflammation/pathology , Keratinocytes/metabolism , Lymph Nodes/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Transport/immunology , Receptors, CCR/genetics , Skin/immunology , Stromal Cells/immunology
13.
Eur J Immunol ; 45(2): 574-83, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25521433

ABSTRACT

Thymus colonisation and thymocyte positioning are regulated by interactions between CCR7 and CCR9, and their respective ligands, CCL19/CCL21 and CCL25. The ligands of CCR7 and CCR9 also interact with the atypical receptor CCRL1 (also known as ACKR4), which is expressed in the thymus and has recently been reported to play an important role in normal αƟT-cell development. Here, we show that CCRL1 is expressed within the thymic cortex, predominantly by MHC-II(low) CD40(-) cortical thymic epithelial cells and at the subcapsular zone by a population of podoplanin(+) thymic epithelial cells in mice. Interestingly, CCRL1 is also expressed by stromal cells which surround the pericytes of vessels at the corticomedullary junction, the site for progenitor cell entry and mature thymocyte egress from the thymus. We show that CCRL1 suppresses thymocyte progenitor entry into the thymus, however, the thymus size and cellularity are the same in adult WT and CCRL1(-/-) mice. Moreover, CCRL1(-/-) mice have no major perturbations in T-cell populations at different stages of thymic differentiation and development, and have a similar rate of thymocyte migration into the blood. Collectively, our findings argue against a major role for CCRL1 in normal thymus development and function.


Subject(s)
Epithelial Cells/metabolism , Lymphopoiesis/genetics , Receptors, CCR/genetics , Stromal Cells/metabolism , Thymocytes/metabolism , Thymus Gland/metabolism , Animals , CD40 Antigens/deficiency , CD40 Antigens/genetics , CD40 Antigens/immunology , Cell Differentiation , Cell Movement , Cellular Microenvironment , Epithelial Cells/cytology , Epithelial Cells/immunology , Female , Gene Expression Regulation, Developmental/immunology , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Knockout , Pericytes/cytology , Pericytes/immunology , Receptors, CCR/deficiency , Receptors, CCR/immunology , Receptors, CCR7/genetics , Receptors, CCR7/immunology , Signal Transduction , Stem Cells/cytology , Stem Cells/immunology , Stromal Cells/cytology , Stromal Cells/immunology , Thymocytes/cytology , Thymocytes/immunology , Thymus Gland/cytology , Thymus Gland/growth & development , Thymus Gland/immunology
14.
Trends Immunol ; 34(1): 7-12, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22921835

ABSTRACT

The chemokine-scavenging receptor, D6, is reported to regulate resolution of inflammatory responses. However, recent data also point to an unanticipated role for D6 in coordinating innate and adaptive immune responses. Here, we propose that D6 is essential for preventing inflammatory leukocyte association with lymphatic vasculature. In the absence of D6, inappropriate inflammatory leukocyte accumulation around lymphatic endothelium congests the lymphatic system, impairing fluid and cellular flow from inflamed sites to lymph nodes and reducing efficiency of antigen presentation. Thus, the inability of D6-deficient mice to resolve inflammation may be a byproduct of impaired fluid drainage from inflamed sites and thus we provide a model unifying D6 function in innate and adaptive immune responses.


Subject(s)
Immune System/immunology , Receptors, CCR10/immunology , Adaptive Immunity , Animals , Chemokines/immunology , Humans , Immunity, Innate , Models, Immunological , Chemokine Receptor D6
15.
J Pathol ; 237(1): 85-97, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25950520

ABSTRACT

Pancreatitis is a significant clinical problem and the lack of effective therapeutic options means that treatment is often palliative rather than curative. A deeper understanding of the pathogenesis of both acute and chronic pancreatitis is necessary to develop new therapies. Pathological changes in pancreatitis are dependent on innate immune cell recruitment to the site of initial tissue damage, and on the coordination of downstream inflammatory pathways. The chemokine receptor CXCR2 drives neutrophil recruitment during inflammation, and to investigate its role in pancreatic inflammation, we induced acute and chronic pancreatitis in wild-type and Cxcr2(-/-) mice. Strikingly, Cxcr2(-/-) mice were strongly protected from tissue damage in models of acute pancreatitis, and this could be recapitulated by neutrophil depletion or by the specific deletion of Cxcr2 from myeloid cells. The pancreata of Cxcr2(-/-) mice were also substantially protected from damage during chronic pancreatitis. Neutrophil depletion was less effective in this model, suggesting that CXCR2 on non-neutrophils contributes to the development of chronic pancreatitis. Importantly, pharmacological inhibition of CXCR2 in wild-type mice replicated the protection seen in Cxcr2(-/-) mice in acute and chronic models of pancreatitis. Moreover, acute pancreatic inflammation was reversible by inhibition of CXCR2. Thus, CXCR2 is critically involved in the development of acute and chronic pancreatitis in mice, and its inhibition or loss protects against pancreatic damage. CXCR2 may therefore be a viable therapeutic target in the treatment of pancreatitis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Pancreas/drug effects , Pancreatitis, Chronic/prevention & control , Pancreatitis/prevention & control , Peptides/pharmacology , Receptors, Interleukin-8B/antagonists & inhibitors , Acute Disease , Animals , Ceruletide , Cytoprotection , Disease Models, Animal , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration/drug effects , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/metabolism , Pancreas/immunology , Pancreas/metabolism , Pancreas/pathology , Pancreatitis/chemically induced , Pancreatitis/genetics , Pancreatitis/immunology , Pancreatitis/metabolism , Pancreatitis/pathology , Pancreatitis, Chronic/chemically induced , Pancreatitis, Chronic/genetics , Pancreatitis, Chronic/immunology , Pancreatitis, Chronic/metabolism , Pancreatitis, Chronic/pathology , Receptors, Interleukin-8B/deficiency , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/immunology , Signal Transduction/drug effects , Time Factors
16.
J Immunol ; 193(10): 5218-28, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25297873

ABSTRACT

Inflammatory chemokines produced in the placenta can direct the migration of placental leukocytes using chemokine receptors that decorate the surface of these cells. Fetal trophoblasts can also express receptors for inflammatory chemokines, and they are one of the few cell types that express atypical chemokine receptor 2 (ACKR2), previously known as D6. ACKR2 binds many inflammatory CC chemokines but cannot stimulate cell migration or activate signaling pathways used by conventional chemokine receptors. Existing evidence suggests that ACKR2 is a specialized chemokine scavenger, but its function in primary human trophoblasts has not been explored. In mice, ACKR2 is thought to be dispensable for the reproductive success of unchallenged females that have conceived naturally, but it can suppress inflammation-induced abortion and aid the survival of implanted allogeneic embryos. In this article, we demonstrate that cultured primary human trophoblasts express ACKR2 far more strongly than genes encoding conventional receptors for inflammatory CC chemokines. Moreover, these cells are capable of the rapid internalization and efficient scavenging of extracellular chemokine, and this is mediated by ACKR2. We also report that in unchallenged DBA/1j mice, Ackr2 deficiency increases the incidence of stillbirth and neonatal death, leads to structural defects in the placenta, and can decrease fetal weight. Loss of Ackr2 specifically from fetal cells makes a key contribution to the placental defects. Thus, primary human trophoblasts use ACKR2 to scavenge chemokines, and ACKR2 deficiency can cause abnormal placental structure and reduced neonatal survival.


Subject(s)
Gene Expression Regulation, Developmental , Placenta/metabolism , Receptors, Chemokine/genetics , Trophoblasts/metabolism , Animals , Animals, Newborn , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Chemokine CCL26 , Chemokines, CC/genetics , Chemokines, CC/immunology , Female , Fetal Development , Fetus , Humans , Infant , Longevity , Mice , Mice, Inbred DBA , Mice, Knockout , Placenta/immunology , Placenta/pathology , Pregnancy , Primary Cell Culture , Receptors, Chemokine/deficiency , Receptors, Chemokine/immunology , Signal Transduction , Trophoblasts/immunology , Trophoblasts/pathology
17.
J Immunol ; 193(1): 400-11, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24890717

ABSTRACT

Chemokine-directed leukocyte migration is crucial for effective immune and inflammatory responses. Conventional chemokine receptors (cCKRs) directly control cell movement; atypical chemokine receptors (ACKRs) regulate coexpressed cCKRs; and both cCKRs and ACKRs internalize chemokines to limit their abundance in vivo, a process referred to as scavenging. A leukocyte's migratory and chemokine-scavenging potential is determined by which cCKRs and ACKRs it expresses, and by the ligand specificity, signaling properties, and chemokine internalization capacity of these receptors. Most chemokines can bind at least one cCKR and one ACKR. CCL2 can bind to CCR2 (a cCKR) and two ACKRs (ACKR1 and ACKR2). In this study, by using fluorescent CCL2 uptake to label cells bearing functional CCL2 receptors, we have defined the expression profile, scavenging activity, and ligand specificity of CCL2 receptors on mouse leukocytes. We show that qualitative and quantitative differences in the expression of CCR2 and ACKR2 endow individual leukocyte subsets with distinctive CCL2 receptor profiles and CCL2-scavenging capacities. We reveal that some cells, including plasmacytoid dendritic cells, can express both CCR2 and ACKR2; that Ly6C(high) monocytes have particularly strong CCL2-scavenging potential in vitro and in vivo; and that CCR2 is a much more effective CCL2 scavenger than ACKR2. We confirm the unique, overlapping, ligand specificities of CCR2 and ACKR2 and, unexpectedly, find that cell context influences the interaction of CCL7 and CCL12 with CCR2. Fluorescent chemokine uptake assays were instrumental in providing these novel insights into CCL2 receptor biology, and the sensitivity, specificity, and versatility of these assays are discussed.


Subject(s)
Chemokine CCL2/immunology , Dendritic Cells/immunology , Monocytes/immunology , Plasma Cells/immunology , Receptors, Chemokine/immunology , Animals , Chemokine CCL2/genetics , Chemokine CCL7/genetics , Chemokine CCL7/immunology , Dendritic Cells/cytology , Mice , Mice, Knockout , Monocyte Chemoattractant Proteins/genetics , Monocyte Chemoattractant Proteins/immunology , Monocytes/cytology , Plasma Cells/cytology , Receptors, Chemokine/genetics
18.
J Immunol ; 192(12): 6120-6130, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24850722

ABSTRACT

Cell therapy regimens are frequently compromised by low-efficiency cell homing to therapeutic niches. Improvements in this regard would enhance effectiveness of clinically applicable cell therapy. The major regulators of tissue-specific cellular migration are chemokines, and therefore selection of therapeutic cellular populations for appropriate chemokine receptor expression would enhance tissue-homing competence. A number of practical considerations preclude the use of Abs in this context, and alternative approaches are required. In this study, we demonstrate that appropriately labeled chemokines are at least as effective in detecting their cognate receptors as commercially available Abs. We also demonstrate the utility of biotinylated chemokines as cell-sorting reagents. Specifically, we demonstrate, in the context of CCR7 (essential for lymph node homing of leukocytes), the ability of biotinylated CCL19 with magnetic bead sorting to enrich for CCR7-expressing cells. The sorted cells demonstrate improved CCR7 responsiveness and lymph node-homing capability, and the sorting is effective for both T cells and dendritic cells. Importantly, the ability of chemokines to detect CCR7, and sort for CCR7 positivity, crosses species being effective on murine and human cells. This novel approach to cell sorting is therefore inexpensive, versatile, and applicable to numerous cell therapy contexts. We propose that this represents a significant technological advance with important therapeutic implications.


Subject(s)
Chemokine CCL19/chemistry , Flow Cytometry/methods , Receptors, CCR7/chemistry , Animals , Chemokine CCL19/immunology , Female , Humans , Male , Mice , Receptors, CCR7/immunology
19.
J Biol Chem ; 289(18): 12330-42, 2014 May 02.
Article in English | MEDLINE | ID: mdl-24644289

ABSTRACT

The atypical chemokine receptor, ACKR2 is a pivotal regulator of chemokine-driven inflammatory responses and works by binding, internalizing, and degrading inflammatory CC-chemokines. ACKR2 displays promiscuity of ligand binding and is capable of interacting with up to 14 different inflammatory CC-chemokines. Despite its prominent biological role, little is known about the structure/function relationship within ACKR2, which regulates ligand binding. Here we demonstrate that a conserved tyrosine motif at the N terminus of ACKR2 is essential for ligand binding, internalization, and scavenging. In addition we demonstrate that sulfation of this motif contributes to ligand internalization. Furthermore, a peptide derived from this region is capable of binding inflammatory chemokines and inhibits their interaction with their cognate signaling receptors. Importantly, the peptide is only active in the sulfated form, further confirming the importance of the sulfated tyrosines for function. Finally, we demonstrate that the bacterial protease, staphopain A, can cleave the N terminus of ACKR2 and suppress its ligand internalization activity. Overall, these results shed new light on the nature of the structural motifs in ACKR2 that are responsible for ligand binding. The study also highlights ACKR2-derived N-terminal peptides as being of potential therapeutic significance.


Subject(s)
Amino Acid Motifs , Chemokines/metabolism , Receptors, Chemokine/metabolism , Tyrosine/metabolism , Amino Acid Sequence , Animals , Binding Sites/genetics , Blotting, Western , CHO Cells , Cell Line, Tumor , Cells, Cultured , Cricetinae , Cricetulus , Cysteine Endopeptidases/metabolism , Endocytosis/genetics , HEK293 Cells , Humans , Ligands , Molecular Sequence Data , Mutation , Peptides/metabolism , Protein Binding , RNA Interference , Receptors, Chemokine/chemistry , Receptors, Chemokine/genetics , Sequence Homology, Amino Acid , Sulfates/metabolism , Tyrosine/genetics
20.
Curr Opin Rheumatol ; 27(2): 204-11, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25603038

ABSTRACT

PURPOSE OF REVIEW: To provide an update of past failures, future prospects and key challenges facing the therapeutic targeting of chemokines and their receptors in rheumatoid arthritis. RECENT FINDINGS: Clinical trials in rheumatoid arthritis have been undertaken with small molecule antagonists or neutralizing antibodies targeting CCR1, CCR5 and CXCL10. Some encouraging results have emerged. Laboratory and clinical research has identified CCL19, CXCL13 and CXCL12, and their receptors, as potential future targets. Developments in our appreciation of posttranslational chemokine modification highlight the complexity of chemokine networks operating in inflamed tissues, and the substantial gaps in existing knowledge. SUMMARY: Despite previous disappointments, there are still reasons to be optimistic that drugs targeting chemokines and their receptors could be developed for the treatment of rheumatoid arthritis. However, a deeper understanding of the chemokine networks at work in inflamed joints is a necessary prerequisite.


Subject(s)
Antirheumatic Agents/pharmacology , Arthritis, Rheumatoid/drug therapy , Cell Migration Inhibition/drug effects , Leukocytes/drug effects , Molecular Targeted Therapy/methods , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/immunology , Chemokines/antagonists & inhibitors , Chemokines/immunology , Humans , Molecular Targeted Therapy/trends , Receptors, Chemokine/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL