Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 245
Filter
1.
Cell Death Dis ; 15(6): 425, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38890311

ABSTRACT

Neutrophil reverse migration (rM) is a recently identified phenomenon in which neutrophils migrate away from the inflammatory site back into the vasculature following initial infiltration, which involved in the resolution of loci inflammatory response or dissemination of inflammation. Present study was aimed to explore the mechanisms in neutrophil rM. By scRNA-seq on the white blood cells in acute lung injury model, we found rM-ed neutrophils exhibited increased gene expression of C-C motif chemokine receptor-like 2 (Ccrl2), an atypical chemokine receptor. Furthermore, an air pouch model was established to directly track rM-ed neutrophils in vivo. Air pouches were generated by 3 ml filtered sterile air injected subcutaneously for 3 days, and then LPS (2 mg/kg) was injected into the pouches to mimic the inflammatory state. For the rM-ed neutrophil tracking system, cell tracker CMFDA were injected into the air pouch to stain the inflammatory loci cells, and after 6 h, stained cells in blood were regarded as the rM-ed neutrophil. Based on this tracking system, we confirmed that rM-ed neutrophils showed increased CCRL2. We also found that the concentrations of the CCRL2 ligand chemerin in plasma was increased in the late stage. Neutralizing chemerin decreased the rM-ed neutrophil ratio in the blood. These results suggest that circulating chemerin attracts neutrophils to leave inflammatory sites by interacting with CCRL2, which might involve in the dissemination of inflammation.


Subject(s)
Cell Movement , Chemokines , Intercellular Signaling Peptides and Proteins , Neutrophils , Neutrophils/metabolism , Chemokines/metabolism , Animals , Mice , Intercellular Signaling Peptides and Proteins/metabolism , Mice, Inbred C57BL , Male , Humans , Receptors, CCR/metabolism , Inflammation/pathology , Inflammation/metabolism , Acute Lung Injury/metabolism , Acute Lung Injury/pathology
2.
Am J Physiol Cell Physiol ; 326(5): C1320-C1333, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38497114

ABSTRACT

Intramuscular fat (IMF) refers to the lipid stored in skeletal muscle tissue. The number and size of intramuscular adipocytes are the primary factors that regulate IMF content. Intramuscular adipocytes can be derived from either in situ or ectopic migration. In this study, it was discovered that the regulation of IMF levels is achieved through the chemokine (C-C motif) ligand 5 (CCL5)/chemokine (C-C motif) receptor 5 (CCR5) pathway by modulating adipocyte migration. In coculture experiments, C2C12 myotubes were more effective in promoting the migration of 3T3-L1 preadipocytes than C2C12 myoblasts, along with increasing CCL5. Correspondingly, overexpressing the CCR5, one of the receptors of CCL5, in 3T3-L1 preadipocytes facilitated their migration. Conversely, the application of the CCL5/CCR5 inhibitor, MARAVIROC (MVC), reduced this migration. In vivo, transplanted experiments of subcutaneous adipose tissue (SCAT) from transgenic mice expressing green fluorescent protein (GFP) provided evidence that injecting recombinant CCL5 (rCCL5) into skeletal muscle promotes the migration of subcutaneous adipocytes to the skeletal muscle. The level of CCL5 in skeletal muscle increased with obesity. Blocking the CCL5/CCR5 axis by MVC inhibited IMF deposition, whereas elevated skeletal muscle CCL5 promoted IMF deposition in obese mice. These results establish a link between the IMF and the CCL5/CCR5 pathway, which could have a potential application for modulating IMF through adipocyte migration.NEW & NOTEWORTHY C2C12 myotubes attract 3T3-L1 preadipocyte migration regulated by the chemokine (C-C motif) ligand 5 (CCL5)/ chemokine (C-C motif) receptor 5 (CCR5) axis. High levels of skeletal muscle-specific CCL5 promote the migration of subcutaneous adipocytes to skeletal muscle and induce the intramuscular fat (IMF) content.


Subject(s)
Adipocytes , Chemokine CCL5 , Myokines , Obesity , Animals , Mice , Chemokine CCL5/genetics , Chemokine CCL5/pharmacology , Ligands , Mice, Obese , Muscle, Skeletal/metabolism , Receptors, CCR/metabolism , Adipocytes/metabolism , Obesity/genetics , Obesity/metabolism , Obesity/pathology
3.
Nat Commun ; 14(1): 5152, 2023 08 24.
Article in English | MEDLINE | ID: mdl-37620389

ABSTRACT

Intestinal intraepithelial lymphocytes (IELs) reside in the gut epithelial layer, where they help in maintaining intestinal homeostasis. Peripheral CD4+ T cells can develop into CD4+CD8αα+ IELs upon arrival at the gut epithelium via the lamina propria (LP). Although this specific differentiation of T cells is well established, the mechanisms preventing it from occurring in the LP remain unclear. Here, we show that chemokine receptor 9 (CCR9) expression is low in epithelial CD4+CD8αα+ IELs, but CCR9 deficiency results in CD4+CD8αα+ over-differentiation in both the epithelium and the LP. Single-cell RNA sequencing shows an enriched precursor cell cluster for CD4+CD8αα+ IELs in Ccr9-/- mice. CD4+ T cells isolated from the epithelium of Ccr9-/- mice also display increased expression of Cbfß2, and the genomic occupancy modification of Cbfß2 expression reveals its important function in CD4+CD8αα+ differentiation. These results implicate a link between CCR9 downregulation and Cbfb2 splicing upregulation to enhance CD4+CD8αα+ IEL differentiation.


Subject(s)
Intraepithelial Lymphocytes , Receptors, CCR , Animals , Mice , Cell Differentiation , Down-Regulation , Epithelium , Up-Regulation , Receptors, CCR/metabolism
4.
Front Immunol ; 14: 1159856, 2023.
Article in English | MEDLINE | ID: mdl-37122736

ABSTRACT

Background: Idiopathic pulmonary fibrosis (IPF) is a chronic progressive interstitial lung disease with limited therapeutic options. Recent studies have demonstrated that chemokines play a vital role in IPF pathogenesis. In the present study, we explored whether the gene signature associated with chemokines could be used as a reliable biological marker for patients with IPF. Methods: Chemokine-related differentially expressed genes (CR-DEGs) in IPF and control lung tissue samples were identified using data from the Gene Expression Omnibus database. A chemokine-related signature of the diagnostic model was established using the LASSO-Cox regression. In addition, unsupervised cluster analysis was conducted using consensus-clustering algorithms. The CIBERSORT algorithm was used to calculate immune cell infiltration across patient subgroups. Finally, we established a mouse model of bleomycin-induced pulmonary fibrosis and a model of fibroblasts treated with TGFß1. Expression levels of chemokine-related signature genes were determined using real-time quantitative polymerase chain reaction (RT-qPCR). Results: We established a chemokine-related eleven-gene signature of a diagnostic model consisting of CXCL2, CCRL2, ARRB1, XCL1, GRK5, PPBP, CCL19, CCL13, CCL11, CXCL6, and CXCL13, which could easily distinguish between IPF patients and controls. Additionally, we identified two subtypes of IPF samples based on chemokine-related gene expression. Pulmonary function parameters and stromal scores were significantly higher in subtype 1 than in subtype 2. Several immune cell types, especially plasma cells and macrophages, differ significantly between the two subtypes. RT-qPCR results showed that the expression levels of Cxcl2 and Ccl2 increased considerably in bleomycin-induced mice. Meanwhile, Arrb1, Ccrl2, Grk5, and Ppbp expression was significantly reduced. Furthermore, multiple chemokine-related genes were altered in TGFß1 or TNFα-induced fibroblast cells. Conclusions: A novel chemokine-related eleven-signature of diagnostic model was developed. These genes are potential biomarkers of IPF and may play essential roles in its pathogenesis.


Subject(s)
Idiopathic Pulmonary Fibrosis , Mice , Animals , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/diagnosis , Lung/pathology , Chemokines/genetics , Chemokines/metabolism , Biomarkers , Bleomycin , Receptors, CCR/metabolism
5.
PLoS One ; 18(1): e0280590, 2023.
Article in English | MEDLINE | ID: mdl-36662882

ABSTRACT

C-C motif chemokine receptor-like 2 (CCRL2) is a non-signaling 7 transmembrane receptor that binds chemotactic ligands to shape leukocyte recruitment to sites of inflammation. However, there is a lack of consensus on the ligands that directly bind CCRL2 or their functional impact. Studies with CCRL2 knockout mice have demonstrated that neutrophils have impaired degranulation and migration in response to CXCL8, where the underlying molecular mechanism is proposed to be due to the formation of CCRL2 heterodimers with the chemokine receptor CXCR2. Herein, we characterized the ligands that bind directly to CCRL2 and interrogated the impact of CCRL2 neutralization on CXCL8 signaling in neutrophils using pharmacological antibody tools. Using flow cytometry and Surface Plasmon Resonance microscopy (SPRm) cell binding experiments, we confirmed that chemerin, but not previously reported C-C chemokines, binds CCRL2. Furthermore, we identified human and mouse CCRL2 antibodies that neutralized chemerin binding to CCRL2. Unexpectedly, we found that neutralization of CCRL2 with these antibodies did not attenuate CXCL8-induced human neutrophil degranulation nor CXCL8-induced murine neutrophil recruitment to the peritoneum. Based on the observed differences in modulating CCRL2 function with neutralizing antibodies compared to the reported CCRL2 deficient murine models, we hypothesize that the ligand binding function of CCRL2 is dispensable for CXCL8 signaling in neutrophils. Finally, extensive profiling of CCRL2 expression on peripheral blood leukocytes revealed monocytes, dendritic cells (DC), and subpopulations of natural killer T (NKT) cells as additional targets, highlighting potential roles for CCRL2 in human cell types beyond neutrophils that warrants future investigation.


Subject(s)
Neutrophil Activation , Receptors, CCR , Humans , Animals , Mice , Receptors, CCR/metabolism , Antibodies, Neutralizing/pharmacology , Ligands , Signal Transduction , Interleukin-8 , Neutrophils/metabolism
6.
J Immunol ; 209(12): 2281-2286, 2022 12 15.
Article in English | MEDLINE | ID: mdl-36469843

ABSTRACT

CD8+ T lymphocytes infiltrate the brain during congenital CMV infection and promote viral clearance. However, the mechanisms by which CD8+ T cells are recruited to the brain remain unclear. Using a mouse model of congenital CMV, we found a gut-homing chemokine receptor (CCR9) was preferentially expressed in CD8+ T cells localized in the brain postinfection. In the absence of CCR9 or CCL25 (CCR9's ligand) expression, CD8+ T cells failed to migrate to key sites of infection in the brain and protect the host from severe forms of disease. Interestingly, we found that expression of CCR9 on CD8+ T cells was also responsible for spatial temporal positioning of T cells in the brain. Collectively, our data demonstrate that the CMV-infected brain uses a similar mechanism for CD8+ T cell homing as the small intestine.


Subject(s)
Cytomegalovirus Infections , Receptors, CCR , Humans , Receptors, CCR/metabolism , CD8-Positive T-Lymphocytes/metabolism , Intestine, Small/metabolism , Cytomegalovirus Infections/metabolism , Brain/metabolism
7.
Diabetes Res Clin Pract ; 191: 110077, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36089102

ABSTRACT

AIMS: Following ST-segment elevation myocardial infarction (STEMI), recruitment and activation of monocytes [classical (CD14++CD16-CCR2++), intermediate (CD14++CD16+CCR2+), non-classical (CD14LowCD16++CCR2Low)] are needed for myocardial wound healing. Monocyte surface receptor CC chemokine receptor type 2 (CCR2) is responsible for monocyte chemotaxis to sites of inflammation and the lipopolysaccharide (LPS)-binding protein co-receptor, CD14, is involved in pro-inflammatory monocyte activation. The purpose of this investigation was to determine the effects of ex-vivo LPS activation on monocyte subset CD14 and CCR2 expression in post-STEMI individuals with normal and elevated random blood glucose. METHODS: Post-STEMI subjects were identified as normal random glucose (NG, <98 mg/dL, n = 13) or impaired random glucose (IG, ≥98 mg/dL, n = 26) and monocytes were analyzed for non-activated and LPS-activated (1 µg/mL for 4 h) CCR2 and CD14 expression. RESULTS: Non-activated intermediate monocytes from IG showed decreased CD14 expression when compared to NG, which was maintained following LPS-activation. The NG group showed a larger absolute reduction in classical CCR2 expression, leading to a significant difference between NG and IG following LPS-activation. CONCLUSION: Results suggest a heightened response to pro-inflammatory activation in IG following STEMI, which may impair or delay post-STEMI myocardial healing, and thus increase the incidence of chronic heart failure. NIH 1R34HL121402.


Subject(s)
Hyperglycemia , Lipopolysaccharide Receptors/immunology , ST Elevation Myocardial Infarction , Blood Glucose/metabolism , Humans , Hyperglycemia/metabolism , Lipopolysaccharides/pharmacology , Monocytes/metabolism , Receptors, CCR/metabolism , Receptors, CCR2/metabolism , Receptors, IgG/metabolism
8.
Front Immunol ; 13: 825635, 2022.
Article in English | MEDLINE | ID: mdl-35967322

ABSTRACT

Relapsed or refractory T acute lymphoblastic leukemia (T-ALL) still carries poor prognosis. Aiming to improve outcomes, the therapeutic potential of an anti-CCR9 monoclonal antibody (mAb 92R), targeting the human chemokine-receptor CCR9 is analyzed on orthotopic xenotransplants. 92R mAb treatment of mice carrying human CCR9+ T-ALL cell lines or primary T cell leukemias inhibits tumor growth and increases survival. The therapeutic effects of 92R are specific and synergize with chemotherapeutic agents increasing survival. Furthermore, 92R decreases size of non-hematopoietic tumors with a forced CCR9 expression and of solid tumors generated by the pancreatic adenocarcinoma cell line AsPC-1. In addition, a humanized version of 92R mAb (Srb1) is also able to inhibit growth of CCR9+ T-ALL tumor cells in vivo, increasing survival 2.66-fold. Finally, 92R mAb prevents liver accumulation of infiltrates and reduces tumor cell numbers in already formed infiltrates. Thus, the humanized version of 92R mAb (Srb1), displays therapeutic potential for CCR9+ tumor treatment and might represent one of the first therapeutic antibodies for precision medicine on T-ALL patients.


Subject(s)
Adenocarcinoma , Pancreatic Neoplasms , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Animals , Heterografts , Humans , Mice , Receptors, CCR/metabolism
9.
JCI Insight ; 7(17)2022 09 08.
Article in English | MEDLINE | ID: mdl-35943802

ABSTRACT

Plasmacytoid dendritic cells (pDCs) perform dual proinflammatory and immunosuppressive roles. We recently reported the potential of pDC therapy for treatment of intractable acute liver failure. However, establishment of efficient methods to deliver pDCs to the liver is essential for future clinical therapeutic applications. The present study demonstrates a higher abundance of liver and peripheral blood pDCs in mice lacking C-C motif chemokine receptor 9 (CCR9), a pDC gut-homing receptor, than in WT mice. Adoptive transfer of Ccr9-/- pDCs resulted in a higher efficiency of migration to the liver than WT pDCs did, while WT pDCs migrated efficiently to the original target organ, the small intestine. Further, Ccr9-/- pDCs consistently migrated efficiently to livers with concanavalin A-induced inflammation, and exerted a more effective immunosuppressive effect, resulting in better protection against acute liver inflammation than that demonstrated by WT pDCs. These findings highlight the therapeutic potential of the manipulation of the CCR9 axis as an approach to improve migration of immunosuppressive pDCs to the liver in order to exploit their beneficial effects in acute liver disease.


Subject(s)
Chemokines , Dendritic Cells , Receptors, CCR/metabolism , Adoptive Transfer , Animals , Inflammation , Liver , Mice
10.
Mucosal Immunol ; 15(5): 882-895, 2022 05.
Article in English | MEDLINE | ID: mdl-35778600

ABSTRACT

The chemokine receptor CCR9 equips T cells with the ability to respond to CCL25, a chemokine that is highly expressed in the thymus and the small intestine (SI). Notably, CCR9 is mostly expressed on CD8 but not on CD4 lineage T cells, thus imposing distinct tissue tropism on CD4 and CD8 T cells. The molecular basis and the consequences for such a dichotomy, however, have not been fully examined and explained. Here, we demonstrate that the forced expression of CCR9 interferes with the tissue trafficking and differentiation of CD4 T cells in SI intraepithelial tissues. While CCR9 overexpression did not alter CD4 T cell generation in the thymus, the forced expression of CCR9 was detrimental for the proper tissue distribution of CD4 T cells in the periphery, and strikingly also for their terminal differentiation in the gut epithelium. Specifically, the differentiation of SI epithelial CD4 T cells into immunoregulatory CD4+CD8αα+ T cells was impaired by overexpression of CCR9 and conversely increased by the genetic deletion of CCR9. Collectively, our results reveal a previously unappreciated role for CCR9 in the tissue homeostasis and effector function of CD4 T cells in the gut.


Subject(s)
Intraepithelial Lymphocytes , Receptors, CCR , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation , Intestines , Intraepithelial Lymphocytes/metabolism , Receptors, CCR/genetics , Receptors, CCR/metabolism
11.
Sci Rep ; 12(1): 8989, 2022 05 29.
Article in English | MEDLINE | ID: mdl-35644891

ABSTRACT

Understanding of the distribution of chemerin and its receptors, Chemokine-like Receptor 1 (CMKLR1), G Protein-coupled Receptor 1 (GPR1) and Chemokine (C-C motif) receptor-like 2 (CCRL2), in the egg and the embryonic annexes is currently lacking, and their role during embryogenesis remains unknown. By immunoblot using monoclonal anti-chicken antibodies and Enzyme Linked Immunosorbent Assays (ELISA), we found that chemerin is expressed 10 times higher in albumen eggs than in blood plasma, and it is also abundant in the perivitelline membrane but undetectable in yolk. Chicken chemerin can inhibit bacterial growth. By Reverse Transcription-quantitative Polymerisation Chain Reaction (RT-qPCR), western-blot, and immunofluorescence, we show that chemerin is locally produced by the oviduct magnum that participates in albumen formation. Using cultures of magnum explants, we demonstrate that progesterone (P4) and oestradiol (E2) treatment increases chemerin secretion into cultured media and expression in magnum. Chemerin and its three receptors are present in amniotic and Chorio Allantoic Membranes (CAM). Only CMKLR1 expression decreased from embryonic day (ED) 7 to ED11 and remained low until ED18. Chemerin concentrations strongly increased in amniotic fluid at D14 when egg albumen crossed the amniotic membrane. In ovo injections of neutralising chemerin and CMKLR1 antibodies (0.01, 0.1 and 1 µg) increased embryo mortality, which occurred mainly at ED12-13, in a dose-dependent manner. Chemerin treatment increased primary CAM viability. Finally, chemerin and CMKLR1 inhibition within the CAM led to a decrease in blood vessel development and associated angiogenic gene expression. Our results show an important function of the chemerin system during embryo development in chickens, suggesting the potential use of this adipokine as a predictive marker for egg fertility or hatchability.


Subject(s)
Chemokines , Chickens , Embryonic Development , Oviducts , Albumins , Animals , Chemokines/metabolism , Chickens/metabolism , Female , Oviducts/metabolism , Ovum , Receptors, CCR/metabolism , Receptors, Chemokine/metabolism , Receptors, G-Protein-Coupled/metabolism
12.
Proteins ; 90(9): 1714-1720, 2022 09.
Article in English | MEDLINE | ID: mdl-35437825

ABSTRACT

Chemokine (C-C motif) receptor-like 2 (CCRL2), is a seven transmembrane receptor closely related to the chemokine receptors CCR1, CCR2, CCR3, and CCR5. Nevertheless, CCRL2 is unable to activate conventional G-protein dependent signaling and to induce cell directional migration. The only commonly accepted CCRL2 ligand is the nonchemokine chemotactic protein chemerin (RARRES2). The chemerin binding to CCLR2 does induce leukocyte chemotaxis, yet, genetic targeting of CCRL2 was shown to modulate the inflammatory response in different experimental models. This mechanism was shown to be crucial for lung dendritic cell migration, neutrophil recruitment, and Natural Killer cell-dependent immune surveillance in lung cancer. To gain more insight in the interactions involved in the CCRL2-chemerin, the binding complexes were generated by protein-protein docking, then submitted to accelerated molecular dynamics. The obtained trajectories were inspected by principal component analyses followed by kernel density estimation to identify the ligand-receptor regions most frequently involved in the binding. To conclude, the reported analyses led to the identification of the putative hot-spot residues involved in CCRL2-chemerin binding.


Subject(s)
Intercellular Signaling Peptides and Proteins , Molecular Dynamics Simulation , Chemokines/genetics , Chemokines/metabolism , Ligands , Receptors, CCR/genetics , Receptors, CCR/metabolism
13.
Neonatology ; 119(3): 320-326, 2022.
Article in English | MEDLINE | ID: mdl-35279661

ABSTRACT

BACKGROUND: Circulating CCR9+ IL-17+ regulatory T (Treg) cells and intestinal barrier biomarkers, such as trefoil factor 3 (TFF3), intestinal-fatty acid binding protein (I-FABP), and Zonulin, are associated with gastrointestinal inflammatory diseases. So far, it is still difficult for clinicians to predict early necrotizing enterocolitis (NEC). STUDY DESIGN: This study included 13 patients with stage I NEC-like presentation (early NEC), 24 patients with one stage of II or III NEC (confirmed NEC), and 80 non-NEC and nonsepsis preterm infants (control group). Another 16 patients experienced at least two stages. We used flow cytometry to measure the frequency of CCR9+ IL-17+ Treg cells and enzyme-linked immunosorbent assay to measure TFF3, I-FABP, and Zonulin levels in the peripheral blood. RESULTS: The demographic and clinical characteristics of patients were comparable. Compared with controls, CCR9+ IL-17+ Treg cells were markedly increased in early NEC and slightly increased in confirmed NEC; in contrast, plasma TFF3, I-FABP, and Zonulin concentrations were notably elevated in confirmed NEC and slightly elevated or not elevated in early NEC. Moreover, for patients who experienced at least two stages, dynamic monitoring of the above indicators also verified this. CONCLUSIONS: This study suggested that an elevated frequency of circulating CCR9+ IL-17+ Treg cells could be a predictor of intestinal inflammation in patients with early NEC.


Subject(s)
Enterocolitis, Necrotizing , Fetal Diseases , Infant, Newborn, Diseases , Infant, Premature, Diseases , Interleukin-17/metabolism , Receptors, CCR/metabolism , Biomarkers , Enterocolitis, Necrotizing/diagnosis , Enterocolitis, Necrotizing/metabolism , Female , Humans , Infant , Infant, Newborn , Infant, Premature , T-Lymphocytes, Regulatory/metabolism
14.
Front Immunol ; 13: 812899, 2022.
Article in English | MEDLINE | ID: mdl-35185906

ABSTRACT

Organized intestinal mucosal immune response appears to be restricted to tetrapods. In teleost fish, there is no evidence for the existence of a particular intestinal region that facilitates the interaction of antigen-presenting cells (APCs) and T cells, such as secondary lymphoid organs. Indeed, despite their importance in the defense against pathogens, the location and manner of APC-T cell interaction within the fish gut is unknown. Here, using non-invasive live imaging of newly developed transgenic reporter lines, we addressed the spatial organization and behavior of APCs and T cells in the intestine of medaka fish both during homeostasis and inflammation. We report that Ccr9a+ T cells are recruited to a band in the lamina propria next to the muscularis mucosa in which Ccl25-expressing cells are present. Ccr9a+ T cells contact APCs for several minutes, in a process mediated by connexin 43. This type of interaction was observed in homeostasis and inflammation, with the interaction being longer and more frequent during inflammation. Thus, our results demonstrate that the mucosal immune response in the intestine of medaka is organized and endowed with a specific region with specialized microenvironment and function.


Subject(s)
Antigen-Presenting Cells/immunology , Intestinal Mucosa/immunology , T-Lymphocytes/immunology , Animals , Chemokines, CC/metabolism , Oryzias/immunology , Receptors, CCR/metabolism
15.
Cell Rep ; 38(5): 110334, 2022 02 01.
Article in English | MEDLINE | ID: mdl-35108538

ABSTRACT

T cell migration via afferent lymphatics to draining lymph nodes (dLNs) depends on expression of CCR7 in T cells and CCL21 in the lymphatic vasculature. Once T cells have entered lymphatic capillaries, they slowly migrate into contracting collecting vessels. Here, lymph flow picks up, inducing T cell detachment and rapid transport to the dLNs. We find that the atypical chemokine receptor 4 (ACKR4), which binds and internalizes CCL19 and CCL21, is induced by lymph flow in endothelial cells lining lymphatic collectors, enabling them to scavenge these chemokines. In the absence of ACKR4, migration of T cells to dLNs in TPA-induced inflammation is significantly reduced. While entry into capillaries is not impaired, T cells accumulate in the ACKR4-deficient dermal collecting vessel segments. Overall, our findings identify an ACKR4-mediated mechanism by which lymphatic collectors facilitate the detachment of lymph-borne T cells in inflammation and their transition from crawling to free-flow toward the dLNs.


Subject(s)
Inflammation/metabolism , Receptors, CCR7/metabolism , Receptors, CCR/metabolism , T-Lymphocytes/metabolism , Animals , Cell Movement/physiology , Dendritic Cells/metabolism , Endothelial Cells/metabolism , Humans , Lymph Nodes/metabolism , Lymphatic Vessels/metabolism , Mice , Skin/metabolism
16.
Angew Chem Int Ed Engl ; 61(12): e202116782, 2022 03 14.
Article in English | MEDLINE | ID: mdl-34936714

ABSTRACT

A conserved intracellular allosteric binding site (IABS) has recently been identified at several G protein-coupled receptors (GPCRs). Starting from vercirnon, an intracellular C-C chemokine receptor type 9 (CCR9) antagonist and previous phase III clinical candidate for the treatment of Crohn's disease, we developed a chemical biology toolbox targeting the IABS of CCR9. We first synthesized a fluorescent ligand enabling equilibrium and kinetic binding studies via NanoBRET as well as fluorescence microscopy. Applying this molecular tool in a membrane-based setup and in living cells, we discovered a 4-aminopyrimidine analogue as a new intracellular CCR9 antagonist with improved affinity. To chemically induce CCR9 degradation, we then developed the first PROTAC targeting the IABS of GPCRs. In a proof-of-principle study, we succeeded in showing that our CCR9-PROTAC is able to reduce CCR9 levels, thereby offering an unprecedented approach to modulate GPCR activity.


Subject(s)
Receptors, CCR , Receptors, G-Protein-Coupled , Allosteric Site , Ligands , Receptors, CCR/metabolism , Receptors, G-Protein-Coupled/metabolism
17.
Mol Immunol ; 140: 267-275, 2021 12.
Article in English | MEDLINE | ID: mdl-34808497

ABSTRACT

BACKGROUND: Leukemia is a group of hematopoietic malignancies characterized by the accumulation and infiltration of abnormal hematopoietic stem cells or early progenitor cells. T cell acute lymphoblastic leukemia (T-ALL) is a hematologic malignancy occurring in 15 % of pediatric and 25 % of adult ALL cases. Infiltration and metastasis of leukemic cells to specific organs are consequences of disease relapse and dismal prognosis. Long non-coding RNAs (lncRNAs) have been identified to function in the migration, invasion and infiltration of tumors by regulating gene expression. Our previous studies showed that CC chemokine receptor 9 (CCR9), which specifically bind to CC chemokine ligand 25 (CCL25), promotes T-ALL infiltration. METHODS: Bioinformatic methods were used to screen LINC00853 in gene expression omnibus (GEO) datasets. RT-qPCR, western bolt and flow cytometry were applied to detect the expression of LINC00853 and CCR9. Transwell and martrigel-transwell were employed to assess the cells migration and invasion abilities. Fluorescence microscope was applied to observed the green fluorescence protein positive (GFP+) cells. Lentivirus and adenovirus were packed to construct nc-blank, sh-LINC00853-blank and sh-LINC00853-rescue jurkat cell lines. RESULTS: In this study, we found out the negative correlation of LINC00853 and CCR9 expression. LINC00853 was downregulated while CCR9 was upregulated in GEO datasets, T-ALL cell lines and clinical samples. Moreover, LINC00853 suppressed jurkat cells migration and invasion in vitro and restrained infiltration in liver, spleen, kidney, lung, brain, ovary of nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice. CONCLUSIONS: These findings indicate that LINC00853 restrains T-ALL cell invasion and infiltration by regulating CCR9/CCL25.


Subject(s)
Chemokines, CC/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , RNA, Long Noncoding/metabolism , Receptors, CCR/metabolism , Body Weight , Cell Line, Tumor , Cell Movement , Databases, Genetic , Gene Expression Regulation, Leukemic , Humans , Neoplasm Invasiveness , Organ Specificity , RNA, Long Noncoding/genetics
18.
Nat Commun ; 12(1): 6760, 2021 11 19.
Article in English | MEDLINE | ID: mdl-34799557

ABSTRACT

Common genetic polymorphisms associated with COVID-19 illness can be utilized for discovering molecular pathways and cell types driving disease pathogenesis. Given the importance of immune cells in the pathogenesis of COVID-19 illness, here we assessed the effects of COVID-19-risk variants on gene expression in a wide range of immune cell types. Transcriptome-wide association study and colocalization analysis revealed putative causal genes and the specific immune cell types where gene expression is most influenced by COVID-19-risk variants. Notable examples include OAS1 in non-classical monocytes, DTX1 in B cells, IL10RB in NK cells, CXCR6 in follicular helper T cells, CCR9 in regulatory T cells and ARL17A in TH2 cells. By analysis of transposase accessible chromatin and H3K27ac-based chromatin-interaction maps of immune cell types, we prioritized potentially functional COVID-19-risk variants. Our study highlights the potential of COVID-19 genetic risk variants to impact the function of diverse immune cell types and influence severe disease manifestations.


Subject(s)
COVID-19/genetics , COVID-19/immunology , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Humans , Receptors, CCR/genetics , Receptors, CCR/metabolism , Risk Factors , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism
19.
Front Immunol ; 12: 721453, 2021.
Article in English | MEDLINE | ID: mdl-34539657

ABSTRACT

Previous studies have evaluated the roles of T and B cells in the pathogenesis of Sjögren's syndrome (SS); however, their relationships with age-dependent and metabolic abnormalities remain unclear. We examined the impacts of changes associated with aging or metabolic abnormalities on populations of T and B cells and SS disease severity. We detected increased populations of IL-17-producing T and B cells, which regulate inflammation, in the salivary glands of NOD/ShiLtJ mice. Inflammation-induced human submandibular gland cell death, determined based on p-MLKL and RIPK3 expression levels, was significantly increased by IL-17 treatment. Among IL-17-expressing cells in the salivary gland, peripheral blood, and spleen, the α4ß7 (gut-homing integrin)-negative population was significantly increased in aged NOD/ShiLtJ mice. The α4ß7-positive population markedly increased in the intestines of aged NOD/ShiLtJ mice following retinoic acid (RA) treatment. A significant increase in α4ß7-negative IL-17-expressing cells in salivary glands may be involved in the onset and progression of SS. These results suggest the potential therapeutic utility of RA in SS treatment.


Subject(s)
Interleukin-17/metabolism , Receptors, CCR/metabolism , Receptors, Lymphocyte Homing/metabolism , Sjogren's Syndrome/etiology , Sjogren's Syndrome/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Animals , Biomarkers , Blood Glucose , Cell Death , Cell Self Renewal , Disease Models, Animal , Disease Susceptibility , Interleukin-17/blood , Mice , Salivary Glands/immunology , Salivary Glands/metabolism , Salivary Glands/pathology , Sjogren's Syndrome/pathology , Stem Cells/cytology , Stem Cells/metabolism
20.
EBioMedicine ; 71: 103570, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34500304

ABSTRACT

BACKGROUND: HIV infection promotes the expansion of immunosuppressive regulatory T-cells (Tregs), contributing to immune dysfunction, tissue fibrosis and disease progression. Early antiretroviral treatment (ART) upon HIV infection improves CD4 count and decreases immune activation. However, Treg dynamics and their epigenetic regulation following early ART initiation remain understudied. METHODS: Treg subsets were characterized by flow cytometry in 103 individuals, including untreated HIV-infected participants in acute and chronic phases, ART-treated in early infection, elite controllers (ECs), immunological controllers (ICs), and HIV-uninfected controls. The methylation status of six regulatory regions of the foxp3 gene was assessed using MiSeq technology. FINDINGS: Total Treg frequency increased overtime during HIV infection, which was normalized in early ART recipients. Tregs in untreated individuals expressed higher levels of activation and immunosuppressive markers (CD39, and LAP(TGF-ß1)), which remained unchanged following early ART. Expression of gut migration markers (CCR9, Integrin-ß7) by Tregs was elevated during untreated HIV infection, while they declined with the duration of ART but not upon early ART initiation. Notably, gut-homing Tregs expressing LAP(TGF-ß1) and CD39 remained higher despite early treatment. Additionally, the increase in LAP(TGF-ß1)+ Tregs overtime were consistent with higher demethylation of conserved non-coding sequence (CNS)-1 in the foxp3 gene. Remarkably, LAP(TGF-ß1)-expressing Tregs in ECs were significantly higher than in uninfected subjects, while the markers of Treg activation and gut migration were not different. INTERPRETATION: Early ART initiation was unable to control the levels of immunosuppressive Treg subsets and their gut migration potential, which could ultimately contribute to gut tissue fibrosis and HIV disease progression. FUNDING: This study was funded by the Canadian Institutes of Health Research (CIHR, grant MOP 142294) and in part by the AIDS and Infectious Diseases Network of the Réseau SIDA et maladies infectieuses du Fonds de recherche du Québec-Santé (FRQ-S).


Subject(s)
Anti-HIV Agents/pharmacology , DNA Methylation , Epigenesis, Genetic , HIV Infections/genetics , T-Lymphocytes, Regulatory/drug effects , Adult , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/therapeutic use , Antigens, CD/genetics , Antigens, CD/metabolism , Apyrase/genetics , Apyrase/metabolism , Female , Forkhead Transcription Factors/genetics , HIV Infections/drug therapy , HIV Infections/immunology , Humans , Integrin beta Chains/genetics , Integrin beta Chains/metabolism , Male , Receptors, CCR/genetics , Receptors, CCR/metabolism , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...