Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 286
Filter
1.
FASEB J ; 35(10): e21917, 2021 10.
Article in English | MEDLINE | ID: mdl-34533865

ABSTRACT

The resolution of inflammation facilitates proper wound healing and limits tissue repair short of exaggerated fibrotic scarring. The atypical chemokine receptor (ACKR)2/D6 scavenges inflammatory chemokines, while IFN-ß is a recently unveiled pro-resolving cytokine. Both effector molecules limit acute inflammatory episodes and promote their resolution in various organs. Here, we found fibrotic skin lesions from ACKR2-/- mice presented increased epidermal and dermal thickening, atrophy of the subcutaneous adipose tissue, augmented disorientation of collagen deposition, and enhanced deformation and loss of hair follicles compared to WT counterparts. In addition, affected skin sections from ACKR2-/- mice contained reduced levels of the pro-resolving mediators IFN-ß and IL-10, but increased levels of the pro-inflammatory chemokines CCL2 and 3, the pro-fibrotic cytokine TGF-ß, and the immune-stimulating cytokine IL-12. Notably, treatment with exogenous IFN-ß rescued, at least in part, all the pro-fibrotic outcomes and lesion size in ACKR2-/- mice and promoted expression of the pro-resolving enzyme 12/15-lipoxygenase (LO) in both ACKR2-/- and WT mice. Moreover, Ifnb-/- mice displayed enhanced pro-fibrotic indices upon exposure to bleomycin. These findings suggest ACKR2 is an important mediator in limiting inflammatory skin fibrosis and acts via IFN-ß production to promote the resolution of inflammation and minimize tissue scaring.


Subject(s)
Alopecia/metabolism , Fibrosis/metabolism , Interferon-beta/metabolism , Receptors, Chemokine/metabolism , Skin/metabolism , Alopecia/genetics , Alopecia/pathology , Animals , Collagen/metabolism , Fibrosis/genetics , Fibrosis/pathology , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Interferon-beta/deficiency , Interferon-beta/genetics , Interleukin-10/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Phenotype , Receptors, Chemokine/deficiency , Receptors, Chemokine/genetics , Skin/pathology
2.
Histochem Cell Biol ; 156(2): 133-146, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33993340

ABSTRACT

Immunosuppressive drugs such as cyclosporine A (CSA) can disrupt thymic structure and functions, ultimately inducing syngeneic/autologous graft-versus-host disease together with involuted medullas. To elucidate the effects of CSA on the thymus more precisely, we analyzed the effects of CSA on the thymus and T cell system using rats. In addition to confirming the phenomena already reported, we newly found that the proportion of recent thymic emigrants also greatly decreased, suggesting impaired supply. Immunohistologically, the medullary thymic epithelial cells (mTECs) presented with a relative decrease in the subset with a competent phenotype and downregulation of class II major histocompatibility complex molecules. In control rats, thymic dendritic cells (DCs) comprised two subsets, XCR1+SIRP1α-CD4- and XCR1-SIRP1α+CD4+. The former had a tendency to selectively localize in the previously-reported epithelium-containing areas of the rat medullas, and the number was significantly reduced by CSA treatment. The epithelium-free areas, another unique domains in the rat medullas, contained significantly more Foxp3+ thymic Tregs. With CSA treatment, the epithelium-free areas presented strong involution, and the number and distribution of Tregs in the medulla were greatly reduced. These results suggest that CSA inhibits the production of single-positive thymocytes, including Tregs, and disturbs the microenvironment of the thymic medulla, with a decrease of the competent mTECs and disorganization of epithelium-free areas and DC subsets, leading to a generation of autoreactive T cells with selective medullary involution.


Subject(s)
Cyclosporine/pharmacology , Epithelial Cells/drug effects , Forkhead Transcription Factors/analysis , Immunosuppressive Agents/pharmacology , T-Lymphocytes, Regulatory/drug effects , Thymus Gland/drug effects , Animals , Cyclosporine/administration & dosage , Dendritic Cells/drug effects , Dendritic Cells/pathology , Epithelial Cells/pathology , Forkhead Transcription Factors/metabolism , Immunohistochemistry , Immunosuppressive Agents/administration & dosage , Injections, Subcutaneous , Male , Optical Imaging , Rats , Rats, Inbred Lew , Receptors, Chemokine/analysis , Receptors, Chemokine/deficiency , Receptors, Chemokine/metabolism , T-Lymphocytes, Regulatory/pathology , Thymocytes/drug effects , Thymocytes/pathology , Thymus Gland/pathology
3.
J Neurosci ; 40(36): 6991-7007, 2020 09 02.
Article in English | MEDLINE | ID: mdl-32801154

ABSTRACT

Pathologic features of Alzheimer's disease (AD) include accumulation of amyloid ß (Aß) and hyperphosphorylated tau protein. We have shown previously that the chemokine-like receptor 1 (CMKLR1) is a functional receptor for Aß, and CMKLR1 contributes to the uptake of Aß. However, it is unclear whether CMKLR1 ameliorates or aggravates the process of AD. Here, we show that deletion of the gene coding for CMKLR1 significantly increased Aß deposits in brains of both male and female amyloid ß precursor protein/presenilin-1 mice. However, it markedly decreased the mortality of these mice. Behavioral studies found that CMKLR1 deficiency improved cognitive impairment of male and female amyloid ß precursor protein/presenilin-1 mice and intracerebroventricular-streptozotocin injection AD mice. We further explored the effect of CMKLR1 on tau pathology. We found that CMKLR1 deficiency or inhibition attenuated the hyperphosphorylation of tau in brains of AD mice in vivo and in the neuronal cells in vitro The expression of CMKLR1 on the neurons affected tau phosphorylation by participating in tau seeding. Together, these results uncover a novel mechanism of CMKLR1 in the pathologic process of AD and suggest that inhibiting the promotion effect of CMKLR1 on tau seeding may provide a new strategy for the treatment of AD.SIGNIFICANCE STATEMENT Evidence suggests that inflammation is involved in the pathologic progression of AD. The chemokine-like receptor 1 (CMKLR1), belonging to the family of GPCRs, is able to bind and uptake amyloid ß. We show here, for the first time, that, although CMKLR1 deficiency increased amyloid ß deposits in AD mice, it reduced the mortality and improved the cognitive deficits of AD mice. We furthermore show that CMKLR1 deficiency or inhibition attenuated tau hyperphosphorylation in brains of AD model mice in vivo and in neuronal cells in vitro Finally, we first discovered that the expression of CMKLR1 on neurons affected tau phosphorylation by participating in tau seeding. These findings suggest that inhibition of CMKLR1 may provide a new strategy for the treatment of AD.


Subject(s)
Alzheimer Disease/metabolism , Cognition , Receptors, Chemokine/genetics , tau Proteins/metabolism , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Female , Gene Deletion , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Neurons/physiology , Phosphorylation , Presenilin-1/genetics , Presenilin-1/metabolism , Receptors, Chemokine/deficiency , Receptors, Chemokine/metabolism
4.
Development ; 147(12)2020 06 17.
Article in English | MEDLINE | ID: mdl-32467242

ABSTRACT

Macrophages are key regulators of developmental processes, including those involved in mammary gland development. We have previously demonstrated that the atypical chemokine receptor ACKR2 contributes to the control of ductal epithelial branching in the developing mammary gland by regulating macrophage dynamics. ACKR2 is a chemokine-scavenging receptor that mediates its effects through collaboration with inflammatory chemokine receptors (iCCRs). Here, we reveal reciprocal regulation of branching morphogenesis in the mammary gland, whereby stromal ACKR2 modulates levels of the shared ligand CCL7 to control the movement of a key population of CCR1-expressing macrophages to the ductal epithelium. In addition, oestrogen, which is essential for ductal elongation during puberty, upregulates CCR1 expression on macrophages. The age at which girls develop breasts is decreasing, which raises the risk of diseases including breast cancer. This study presents a previously unknown mechanism controlling the rate of mammary gland development during puberty and highlights potential therapeutic targets.


Subject(s)
Macrophages/metabolism , Mammary Glands, Animal/growth & development , Receptors, Chemokine/metabolism , Animals , Chemokine CCL3/deficiency , Chemokine CCL3/genetics , Chemokine CCL3/metabolism , Chemokine CCL5/deficiency , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , Epithelium/metabolism , Estradiol/pharmacology , Female , Lectins, C-Type/metabolism , Macrophages/cytology , Mammary Glands, Animal/metabolism , Mannose Receptor , Mannose-Binding Lectins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Morphogenesis , Receptors, CCR1/deficiency , Receptors, CCR1/genetics , Receptors, CCR1/metabolism , Receptors, Cell Surface/metabolism , Receptors, Chemokine/deficiency , Receptors, Chemokine/genetics , Up-Regulation/drug effects
5.
Am J Physiol Endocrinol Metab ; 318(3): E371-E380, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31910029

ABSTRACT

Excess androgen-induced obesity has become a public health problem, and its prevalence has increased substantially in recent years. Chemokine-like receptor 1 (CMKLR1), a receptor of chemerin secreted by adipose tissue, is linked to adipocyte differentiation, adipose tissue development, and obesity. However, the effect of CMKLR1 signaling on androgen-mediated adiposity in vivo remains unclear. Using CMKLR1-knockout mice, we constructed an androgen-excess female mouse model through 5α-dihydrotestosterone (DHT) treatment and an androgen-deficient male mouse model by orchidectomy (ORX). For mechanism investigation, we used 2-(α-Naphthoyl) ethyltrimethylammonium iodide (α-NETA), an antagonist of CMKLR1, to suppress CMKLR1 in vivo and wortmannin, a PI3K signaling antagonist, to treat brown adipose tissue (BAT) explant cultures in vitro. Furthermore, we used histological examination and quantitative PCR, as well as Western blot analysis, glucose tolerance tests, and biochemical analysis of serum, to describe the phenotypes and the changes in gene expression. We demonstrated that excess androgen in the female mice resulted in larger cells in the white adipose tissue (WAT) and the BAT, whereas androgen deprivation in the male mice induced a reduction in cell size. Both of these adipocyte size effects could be attenuated in the CMKLR1-knockout mice. CMKLR1 deficiency influenced the effect of androgen treatment on adipose tissue by regulating the mRNA expression of the androgen receptor (AR) and adipocyte markers (such as Fabp4 and Cidea). Moreover, suppression of CMKLR1 by α-NETA could also reduce the extent of the adipocyte cell enlargement caused by DHT. Furthermore, we found that DHT could reduce the levels of phosphorylated ERK (pERK) in the BAT, while CMKLR1 inactivation inhibited this effect, which had been induced by DHT, through the PI3K signaling pathway. These findings reveal an antiobesity role of CMKLR1 deficiency in regulating lipid accumulation, highlighting the scientific importance for the further development of small-molecule CMKLR1 antagonists as fundamental research tools and/or as potential drugs for use in the treatment of adiposity.


Subject(s)
Androgens/pharmacology , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Receptors, Chemokine/deficiency , Adipocytes/metabolism , Adipocytes/ultrastructure , Adipose Tissue, Brown/drug effects , Androgens/deficiency , Animals , Body Weight , Cell Size , Dihydrotestosterone/pharmacology , Female , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Naphthalenes/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Quaternary Ammonium Compounds/pharmacology , Receptors, Chemokine/antagonists & inhibitors , Receptors, Chemokine/genetics , Wortmannin/pharmacology
6.
Life Sci Alliance ; 2(6)2019 12.
Article in English | MEDLINE | ID: mdl-31792059

ABSTRACT

CX3CR1, one of the highest expressed genes in microglia in mice and humans, is implicated in numerous microglial functions. However, the molecular mechanisms underlying Cx3cr1 signaling are not well understood. Here, we analyzed transcriptomes of Cx3cr1-deficient microglia under varying conditions by RNA-sequencing (RNA-seq). In 2-mo-old mice, Cx3cr1 deletion resulted in the down-regulation of a subset of immune-related genes, without substantial epigenetic changes in markers of active chromatin. Surprisingly, Cx3cr1-deficient microglia from young mice exhibited a transcriptome consistent with that of aged Cx3cr1-sufficient animals, suggesting a premature aging transcriptomic signature. Immunohistochemical analysis of microglia in young and aged mice revealed that loss of Cx3cr1 modulates microglial morphology in a comparable fashion. Our results suggest that CX3CR1 may regulate microglial function in part by modulating the expression levels of a subset of inflammatory genes during chronological aging, making Cx3cr1-deficient mice useful for studying aged microglia.


Subject(s)
Aging, Premature/genetics , CX3C Chemokine Receptor 1/deficiency , Microglia/metabolism , Animals , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , Disease Models, Animal , Female , Gene Deletion , Genetic Profile , Inflammation/genetics , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neurons/metabolism , Receptors, Chemokine/deficiency , Signal Transduction , Transcriptome
7.
PLoS Biol ; 17(5): e3000287, 2019 05.
Article in English | MEDLINE | ID: mdl-31141500

ABSTRACT

Atypical chemokine receptor 2 (ACKR2) is a chemokine-scavenging receptor. ACKR2-/-embryos display a reduction in size of a novel, to our knowledge, embryonic skin macrophage population referred to as 'intermediate' cells. CC chemokine receptor 2 (CCR2)-/-embryos display an identical phenotype, indicating that these cells require CCR2 to enable them to populate embryonic skin. Further analysis revealed that ACKR2-/-embryos have higher circulating concentrations of the CCR2 ligand, CC ligand 2 (CCL2); thus, ACKR2 regulates intraembryonic CCL2 levels. We show that ACKR2 is strongly expressed by trophoblasts and that it blocks movement of inflammatory chemokines, such as CCL2, from the maternal decidua into the embryonic circulation. We propose that trophoblastic ACKR2 is responsible for ensuring chemokine compartmentalisation on the maternal decidua, without which chemokines enter the embryonic circulation, disrupting gradients essential for directed intraembryonic cell migration. Overall, therefore, we describe a novel, to our knowledge, molecular mechanism whereby maternal decidual chemokines can function in a compartmentalised fashion without interfering with intraembryonic leukocyte migration. These data suggest similar functions for other atypical chemokine receptors in the placenta and indicate that defects in such receptors may have unanticipated developmental consequences.


Subject(s)
Chemokines/metabolism , Mammals/metabolism , Placenta/metabolism , Animals , Cell Movement , Decidua/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Female , Macrophages/metabolism , Mice, Inbred C57BL , Monocytes/metabolism , Pregnancy , Receptors, Chemokine/deficiency , Receptors, Chemokine/metabolism , Skin/embryology , Skin/metabolism , Transcription, Genetic , Yolk Sac/metabolism
8.
Cardiovasc Res ; 115(10): 1557-1566, 2019 08 01.
Article in English | MEDLINE | ID: mdl-30597013

ABSTRACT

AIMS: Vascular calcification, a marker of increased cardiovascular risk, is an active process orchestrated by smooth muscle cells. Observational studies indicate that omega-3 fatty acids protect against vascular calcification, but the mechanisms are unknown. The G-protein coupled receptor ChemR23 transduces the resolution of inflammation induced by the omega-3-derived lipid mediator resolvin E1. ChemR23 also contributes to osteoblastic differentiation of stem cells and bone formation, but its role in vascular calcification is unknown. The aim of this study was to establish the role of ChemR23 in smooth muscle cell fate and calcification. METHODS AND RESULTS: Gene expression analysis in epigastric arteries derived from patients with chronic kidney disease and vascular calcification revealed that ChemR23 mRNA levels predicted a synthetic smooth muscle cell phenotype. Genetic deletion of ChemR23 in mice prevented smooth muscle cell de-differentiation. ChemR23-deficient smooth muscle cells maintained a non-synthetic phenotype and exhibited resistance to phosphate-induced calcification. Moreover, ChemR23-deficient mice were protected against vitamin D3-induced vascular calcification. Resolvin E1 inhibited smooth muscle cell calcification through ChemR23. Introduction of the Caenorhabditis elegans Fat1 transgene, leading to an endogenous omega-3 fatty acid synthesis and hence increased substrate for resolvin E1 formation, significantly diminished the differences in phosphate-induced calcification between ChemR23+/+ and ChemR23-/- mice. CONCLUSION: This study identifies ChemR23 as a previously unrecognized determinant of synthetic and osteoblastic smooth muscle cell phenotype, favouring phosphate-induced vascular calcification. This effect may be of particular importance in the absence of ChemR23 ligands, such as resolvin E1, which acts as a calcification inhibitor under hyperphosphatic conditions.


Subject(s)
Adaptation, Physiological , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Osteogenesis , Phosphates/metabolism , Receptors, Chemokine/metabolism , Vascular Calcification/metabolism , Adaptation, Physiological/drug effects , Adult , Aged , Animals , Cadherins/genetics , Cadherins/metabolism , Cholecalciferol , Disease Models, Animal , Eicosapentaenoic Acid/analogs & derivatives , Eicosapentaenoic Acid/pharmacology , Female , HEK293 Cells , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Osteogenesis/drug effects , Rats , Receptors, Chemokine/deficiency , Receptors, Chemokine/drug effects , Receptors, Chemokine/genetics , Signal Transduction , Vascular Calcification/chemically induced , Vascular Calcification/pathology , Vascular Calcification/prevention & control
9.
Kidney Int ; 93(4): 826-841, 2018 04.
Article in English | MEDLINE | ID: mdl-29395335

ABSTRACT

The atypical chemokine receptor 2 (ACKR2), also named D6, regulates local levels of inflammatory chemokines by internalization and degradation. To explore potential anti-inflammatory functions of ACKR2 in glomerulonephritis, we induced autologous nephrotoxic nephritis in C57/BL6 wild-type and Ackr2-deficient mice. Renal ACKR2 expression increased and localized to interstitial lymphatic endothelium during nephritis. At two weeks Ackr2-/-mice developed increased albuminuria and urea levels compared to wild-type mice. Histological analysis revealed increased structural damage in the glomerular and tubulointerstitial compartments within Ackr2-/- kidneys. This correlated with excessive renal leukocyte infiltration of CD4+ T cells and mononuclear phagocytes with increased numbers in the tubulointerstitium but not glomeruli in knockout mice. Expression of inflammatory mediators and especially markers of fibrotic tissue remodeling were increased along with higher levels of ACKR2 inflammatory chemokine ligands like CCL2 in nephritic Ackr2-/- kidneys. In vitro, Ackr2 deficiency in TNF-stimulated tubulointerstitial tissue but not glomeruli increased chemokine levels. These results are in line with ACKR2 expression in interstitial lymphatic endothelial cells, which also assures efflux of activated leukocytes into regional lymph nodes. Consistently, nephritic Ackr2-/- mice showed reduced adaptive cellular immune responses indicated by decreased regional T-cell activation. However, this did not prevent aggravated injury in the kidneys of Ackr2-/- mice with nephrotoxic nephritis due to simultaneously increased tubulointerstitial chemokine levels, leukocyte infiltration and fibrosis. Thus, ACKR2 is important in limiting renal inflammation and fibrotic remodeling in progressive nephrotoxic nephritis. Hence, ACKR2 may be a potential target for therapeutic interventions in immune complex glomerulonephritis.


Subject(s)
Glomerulonephritis/prevention & control , Immune Complex Diseases/prevention & control , Kidney Glomerulus/metabolism , Kidney Tubules/metabolism , Receptors, Chemokine/metabolism , Adaptive Immunity , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Chemotaxis, Leukocyte , Disease Models, Animal , Disease Progression , Fibrosis , Glomerulonephritis/immunology , Glomerulonephritis/metabolism , Glomerulonephritis/pathology , Immune Complex Diseases/immunology , Immune Complex Diseases/metabolism , Immune Complex Diseases/pathology , Inflammation Mediators/metabolism , Kidney Glomerulus/immunology , Kidney Glomerulus/pathology , Kidney Tubules/immunology , Kidney Tubules/pathology , Lymphocyte Activation , Male , Mice, Inbred C57BL , Mice, Knockout , Mononuclear Phagocyte System/immunology , Mononuclear Phagocyte System/metabolism , Receptors, Chemokine/deficiency , Receptors, Chemokine/genetics , Signal Transduction
10.
J Neurosci ; 37(44): 10541-10553, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28951447

ABSTRACT

Brief monocular deprivation (MD) shifts ocular dominance and reduces the density of thalamic synapses in layer 4 of the mouse primary visual cortex (V1). We found that microglial lysosome content is also increased as a result of MD. Previous studies have shown that the microglial fractalkine receptor CX3CR1 is involved in synaptic development and hippocampal plasticity. We therefore tested the hypothesis that neuron-to-microglial communication via CX3CR1 is an essential component of visual cortical development and plasticity in male mice. Our data show that CX3CR1 is not required for normal development of V1 responses to visual stimulation, multiple forms of experience-dependent plasticity, or the synapse loss that accompanies MD in layer 4. By ruling out an essential role for fractalkine signaling, our study narrows the search for understanding how microglia respond to active synapse modification in the visual cortex.SIGNIFICANCE STATEMENT Microglia in the visual cortex respond to monocular deprivation with increased lysosome content, but signaling through the fractalkine receptor CX3CR1 is not an essential component in the mechanisms of visual cortical development or experience-dependent synaptic plasticity.


Subject(s)
Evoked Potentials, Visual/physiology , Microglia/metabolism , Neuronal Plasticity/physiology , Receptors, Chemokine/deficiency , Visual Cortex/growth & development , Visual Cortex/metabolism , Animals , CX3C Chemokine Receptor 1 , Cell Communication/physiology , Geniculate Bodies/growth & development , Geniculate Bodies/metabolism , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Organ Culture Techniques , Vision, Monocular/physiology
11.
PLoS One ; 12(8): e0182643, 2017.
Article in English | MEDLINE | ID: mdl-28800592

ABSTRACT

Studies on inflammatory disorders elucidated the pivotal role of the CX3CL1/CX3CR1 axis with respect to the pathophysiology and diseases progression. Coxsackievirus B3 (CVB3)-induced myocarditis is associated with severe cardiac inflammation, which may progress to heart failure. We therefore investigated the influence of CX3CR1 ablation in the model of acute myocarditis, which was induced by inoculation with 5x105 plaque forming units of CVB3 (Nancy strain) in either CX3CR1-/- or C57BL6/j (WT) mice. Seven days after infection, myocardial inflammation, remodeling, and titin expression and phosphorylation were examined by immunohistochemistry, real-time PCR and Pro-Q diamond stain. Cardiac function was assessed by tip catheter. Compared to WT CVB3 mice, CX3CR1-/- CVB3 mice exhibited enhanced left ventricular expression of inflammatory cytokines and chemokines, which was associated with an increase of immune cell infiltration/presence. This shift towards a pro-inflammatory immune response further resulted in increased cardiac fibrosis and cardiomyocyte apoptosis, which was reflected by an impaired cardiac function in CX3CR1-/- CVB3 compared to WT CVB3 mice. These findings demonstrate a cardioprotective role of CX3CR1 in CVB3-infected mice and indicate the relevance of the CX3CL1/CX3CR1 system in CVB3-induced myocarditis.


Subject(s)
Chemokine CX3CL1/immunology , Coxsackievirus Infections/genetics , Enterovirus B, Human/pathogenicity , Host-Pathogen Interactions/immunology , Myocarditis/genetics , Receptors, Chemokine/immunology , Animals , Apoptosis , CX3C Chemokine Receptor 1 , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Chemokine CX3CL1/genetics , Coxsackievirus Infections/immunology , Coxsackievirus Infections/pathology , Coxsackievirus Infections/virology , Disease Models, Animal , Enterovirus B, Human/growth & development , Gene Expression Regulation , Heart Function Tests , Humans , Interleukins/genetics , Interleukins/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocarditis/immunology , Myocarditis/pathology , Myocarditis/virology , Myocytes, Cardiac/immunology , Myocytes, Cardiac/pathology , Phosphorylation , Protein Kinases/genetics , Protein Kinases/immunology , Receptors, Chemokine/deficiency , Receptors, Chemokine/genetics
12.
Blood ; 130(10): 1223-1234, 2017 09 07.
Article in English | MEDLINE | ID: mdl-28743719

ABSTRACT

CCRL2 is a 7-transmembrane domain receptor that shares structural and functional similarities with the family of atypical chemokine receptors (ACKRs). CCRL2 is upregulated by inflammatory signals and, unlike other ACKRs, it is not a chemoattractant-scavenging receptor, does not activate ß-arrestins, and is widely expressed by many leukocyte subsets. Therefore, the biological role of CCRL2 in immunity is still unclear. We report that CCRL2-deficient mice have a defect in neutrophil recruitment and are protected in 2 models of inflammatory arthritis. In vitro, CCRL2 was found to constitutively form homodimers and heterodimers with CXCR2, a main neutrophil chemotactic receptor. By heterodimerization, CCRL2 could regulate membrane expression and promote CXCR2 functions, including the activation of ß2-integrins. Therefore, upregulation of CCRL2 observed under inflammatory conditions is functional to finely tune CXCR2-mediated neutrophil recruitment at sites of inflammation.


Subject(s)
Arthritis/metabolism , Arthritis/pathology , Neutrophils/pathology , Receptors, Chemokine/metabolism , Receptors, Interleukin-8B/metabolism , Animals , Arthritis/complications , CD18 Antigens/metabolism , Cell Survival , Disease Models, Animal , Inflammation/complications , Inflammation/pathology , Mice, Knockout , Neutrophil Infiltration , Protein Conformation , Protein Multimerization , Receptors, CCR , Receptors, Chemokine/chemistry , Receptors, Chemokine/deficiency , Receptors, Interleukin-8B/chemistry , Signal Transduction
13.
Circ Res ; 120(11): 1789-1799, 2017 May 26.
Article in English | MEDLINE | ID: mdl-28302649

ABSTRACT

RATIONALE: Nonclassical mouse monocyte (CX3CR1high, Ly-6Clow) patrolling along the vessels of the microcirculation is critical for endothelial homeostasis and inflammation. Because of technical challenges, it is currently not established how patrolling occurs in large arteries. OBJECTIVE: This study was undertaken to elucidate the molecular, migratory, and functional phenotypes of patrolling monocytes in the high shear and pulsatile environment of large arteries in healthy, hyperlipidemic, and atherosclerotic conditions. METHODS AND RESULTS: Applying a new method for stable, long-term 2-photon intravital microscopy of unrestrained large arteries in live CX3CR1-GFP (green fluorescent protein) mice, we show that nonclassical monocytes patrol inside healthy carotid arteries at a velocity of 36 µm/min, 3× faster than in microvessels. The tracks are less straight but lead preferentially downstream. The number of patrolling monocytes is increased 9-fold by feeding wild-type mice a Western diet or by applying topical TLR7/8 (Toll-like receptor) agonists. A similar increase is seen in CX3CR1+/GFP/apoE-/- mice on chow diet, with a further 2- to 3-fold increase on Western diet (22-fold over healthy). In plaque conditions, monocytes are readily captured onto the endothelium from free flow. Stable patrolling is unaffected in CX3CR1-deficient mice and involves the contribution of LFA-1 (lymphocyte-associated antigen 1) and α4 integrins. The endothelial damage in atherosclerotic carotid arteries was assessed by electron microscopy and correlates with the number of intraluminal patrollers. Abolishing patrolling monocytes in Nr4a1-/- apoE-/- mice leads to pronounced endothelial apoptosis. CONCLUSIONS: Arterial patrolling is a prominent new feature of nonclassical monocytes with unique molecular and kinetic properties. It is highly upregulated in hyperlipidemia and atherosclerosis in a CX3CR1-independent fashion and plays a potential role in endothelial protection.


Subject(s)
Arteries/metabolism , Atherosclerosis/metabolism , Diet, Western/adverse effects , Endothelium, Vascular/metabolism , Monocytes/metabolism , Receptors, Chemokine/deficiency , Animals , Arteries/pathology , Atherosclerosis/pathology , CX3C Chemokine Receptor 1 , Endothelium, Vascular/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout
14.
J Leukoc Biol ; 101(5): 1143-1153, 2017 05.
Article in English | MEDLINE | ID: mdl-28148720

ABSTRACT

Precursors of dendritic cells (pre-DCs) arise in the bone marrow (BM), egress to the blood, and finally migrate to peripheral tissue, where they differentiate to conventional dendritic cells (cDCs). Upon their activation, antigen-bearing cDCs migrate from peripheral tissue to regional lymph nodes (LNs) in a manner dependent on the chemokine receptor, CCR7. To maintain immune homeostasis, these departing cDCs must be replenished by new cDCs that develop from pre-DCs, but the molecular signals that direct pre-DC trafficking from the BM to the blood and peripheral tissues remain poorly understood. In the present study, we found that pre-DCs express the chemokine receptors CXCR4, CCR2, and CX3CR1, and that each of these receptors has a distinct role in pre-DC trafficking. Flow cytometric analysis of pre-DCs lacking CXCR4 revealed that this receptor is required for the retention of pre-DCs in the BM. Analyses of mice lacking CCR2 or CX3CR1, or both, revealed that they promote pre-DC migration to the lung at steady state. CCR2, but not CX3CR1, was required for pre-DC migration to the inflamed lung. Thus, these multiple chemokine receptors cooperate in a step-wise fashion to coordinate the trafficking of pre-DCs from the BM to the circulation and peripheral tissues.


Subject(s)
Bone Marrow Cells/immunology , Dendritic Cells/immunology , Lung/immunology , Pneumonia/immunology , Receptors, CCR2/immunology , Receptors, CXCR4/immunology , Receptors, Chemokine/immunology , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/pathology , CX3C Chemokine Receptor 1 , Cell Differentiation , Cell Movement/drug effects , Dendritic Cells/drug effects , Dendritic Cells/pathology , Gene Expression Regulation , Lipopolysaccharides , Lung/drug effects , Lung/pathology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymph Nodes/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumonia/chemically induced , Pneumonia/genetics , Pneumonia/pathology , Receptors, CCR2/deficiency , Receptors, CCR2/genetics , Receptors, CCR7/genetics , Receptors, CCR7/immunology , Receptors, CXCR4/deficiency , Receptors, CXCR4/genetics , Receptors, Chemokine/deficiency , Receptors, Chemokine/genetics , Signal Transduction
15.
BMC Neurosci ; 18(1): 11, 2017 01 06.
Article in English | MEDLINE | ID: mdl-28061814

ABSTRACT

BACKGROUND: The fractalkine/CX3C chemokine receptor 1 (CX3CR1) pathway has been identified to play an essential role in the chemotaxis of microglia, leukocyte trafficking and microglia/macrophage recruitment. It has also been shown to be important in the regulation of the inflammatory response in the early phase after experimental stroke. The present study was performed to investigate if CX3CR1 deficiency affects microglia during the first 14 days with consequences for tissue damage after experimental stroke. RESULTS: CX3CR1 deficiency significantly increased the number of intersections of GFP positive microglia in the proximal peri-infarct area at 2, 7 and 14 days following tMCAO compared to heterozygous and wildtype littermates. In addition, the length of microglial branches increased until day 7 in CX3CR1 knockout mice while the presence of a functional CX3CR1 allele resulted in a gradual reduction of their length following tMCAO. After stroke, wildtype, heterozygous and CX3CR1 deficient mice did not show differences in the composite neuroscore and assessment of infarct volumes from CX3CR1 wildtype, heterozygous and deficient mice revealed no differences between the genotypes 7 and 14 days after stroke. CONCLUSION: Results demonstrate that CX3CR1 deficiency affects the morphology of GFP positive microglia located in the proximal peri-infarct region during the first 14 days after tMCAO. Our data also indicate that CX3CR1 deficiency does not affect definite infarct volumes. Modulation of the CX3CR1 pathway may have implication for microglia function contributing to mechanisms of tissue reorganization in the post-ischemic brain.


Subject(s)
Microglia/metabolism , Microglia/pathology , Receptors, Chemokine/deficiency , Stroke/metabolism , Stroke/pathology , Animals , Brain/metabolism , Brain/pathology , CX3C Chemokine Receptor 1 , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunohistochemistry , Male , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/metabolism , Receptors, Chemokine/genetics , Recovery of Function/physiology , Time Factors
16.
Int J Colorectal Dis ; 32(3): 315-324, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27942903

ABSTRACT

PURPOSE: Inflammatory conditions like inflammatory bowel diseases (IBD) are characterized by increased immune cell infiltration. The chemokine ligand CX3CL1 and its receptor CX3CR1 have been shown to be involved in leukocyte adhesion, transendothelial recruitment, and chemotaxis. Therefore, the objective of this study was to describe CX3CL1-CX3CR1-mediated signaling in the induction of immune cell recruitment during experimental murine colitis. METHODS: Acute colitis was induced by dextran sodium sulfate (DSS), and sepsis was induced by injection of lipopolysaccharide (LPS). Serum concentrations of CX3CR1 and CX3CL1 were measured by ELISA. Wild-type and CX3CR1-/- mice were challenged with DSS, and on day 6, intravital microscopy was performed to monitor colonic leukocyte and platelet recruitment. Intestinal inflammation was assessed by disease activity, histopathology, and neutrophil infiltration. RESULTS: CX3CR1 was upregulated in DSS colitis and LPS-induced sepsis. CX3CR1-/- mice were protected from disease severity and intestinal injury in DSS colitis, and CX3CR1 deficiency resulted in reduced rolling of leukocytes and platelets. CONCLUSIONS: In the present study, we provide evidence for a crucial role of CX3CL1-CX3CR1 in experimental colitis, in particular for intestinal leukocyte recruitment during murine colitis. Our findings suggest that CX3CR1 blockade represents a potential therapeutic strategy for treatment of IBD.


Subject(s)
Chemokine CX3CL1/metabolism , Colitis/chemically induced , Colitis/genetics , Down-Regulation , Leukocytes/pathology , Receptors, Chemokine/genetics , Animals , Blood Platelets/pathology , CX3C Chemokine Receptor 1 , Cell Adhesion , Colitis/metabolism , Colitis/pathology , Colon/blood supply , Colon/pathology , Dextran Sulfate , Disease Susceptibility , Inflammation/metabolism , Inflammation/pathology , Leukocyte Rolling , Leukocytes/metabolism , Male , Mice, Inbred C57BL , Microvessels/pathology , Neutrophil Infiltration , Receptors, Chemokine/deficiency , Receptors, Chemokine/metabolism
17.
J Neuroinflammation ; 13(1): 196, 2016 08 25.
Article in English | MEDLINE | ID: mdl-27562117

ABSTRACT

BACKGROUND: Alphaviruses can cause fatal encephalitis in humans. Natural infections occur via the bite of infected mosquitos, but aerosol transmissibility makes some of these viruses potential bioterrorism agents. Central nervous system (CNS) host responses contribute to alphavirus pathogenesis in experimental models and are logical therapeutic targets. We investigated whether reactive oxygen species (ROS) generated by nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (Nox) activity within the CNS contributes to fatal alphavirus encephalitis in mice. METHODS: Infected animals were treated systemically with the angiotensin receptor-blocking drug, telmisartan, given its ability to cross the blood-brain barrier, selectively block type-1 angiotensin receptors (AT1R), and inhibit Nox-derived ROS production in vascular smooth muscle and other extraneural tissues. Clinical, virological, biochemical, and histopathological outcomes were followed over time. RESULTS: The importance of the angiotensin II (Ang II)/AT1R axis in disease pathogenesis was confirmed by demonstrating increased Ang II levels in the CNS following infection, enhanced disease survival when CNS Ang II production was suppressed, increased AT1R expression on microglia and tissue-infiltrating myeloid cells, and enhanced disease survival in AT1R-deficient mice compared to wild-type (WT) controls. Systemic administration of telmisartan protected WT mice from lethal encephalitis caused by two different alphaviruses in a dose-dependent manner without altering virus replication or exerting any anti-inflammatory effects in the CNS. Infection triggered up-regulation of multiple Nox subunits in the CNS, while drug treatment inhibited local Nox activity, ROS production, and oxidative neuronal damage. Telmisartan proved ineffective in Nox-deficient mice, demonstrating that this enzyme is its main target in this experimental setting. CONCLUSIONS: Nox-derived ROS, likely arising from CNS myeloid cells triggered by AT1R signaling, are pathogenic during fatal alphavirus encephalitis in mice. Systemically administered telmisartan at non-hypotensive doses targets Nox activity in the CNS to exert a neuroprotective effect. Disruption of this pathway may have broader implications for the treatment of related infections as well as for other CNS diseases driven by oxidative injury.


Subject(s)
Central Nervous System/pathology , Encephalomyelitis, Equine/pathology , Myeloid Cells/metabolism , Receptor, Angiotensin, Type 1/metabolism , Signal Transduction/physiology , ATPases Associated with Diverse Cellular Activities , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Benzimidazoles/pharmacology , Benzoates/pharmacology , CX3C Chemokine Receptor 1 , Central Nervous System/drug effects , Central Nervous System/virology , DNA Helicases/genetics , DNA Helicases/metabolism , Disease Models, Animal , Encephalomyelitis, Equine/drug therapy , Encephalomyelitis, Equine/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Membrane Transport Proteins/deficiency , Membrane Transport Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cells/classification , Myeloid Cells/ultrastructure , Myeloid Cells/virology , Neurons/pathology , Neurons/ultrastructure , Oxygen Consumption/drug effects , Reactive Oxygen Species/metabolism , Receptors, Chemokine/deficiency , Receptors, Chemokine/genetics , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Telmisartan
18.
Biochim Biophys Acta ; 1861(11): 1614-1622, 2016 11.
Article in English | MEDLINE | ID: mdl-27417459

ABSTRACT

Brown adipose tissue (BAT) plays essential role in metabolic- and thermoregulation and displays morphological and functional plasticity in response to environmental and metabolic challenges. BAT is a heterogeneous tissue containing adipocytes and various immune-related cells, however, their interaction in regulation of BAT function is not fully elucidated. Fractalkine is a chemokine synthesized by adipocytes, which recruits fractalkine receptor (CX3CR1)-expressing leukocytes into the adipose tissue. Using transgenic mice, in which the fractalkine receptor, Cx3cr1 gene was replaced by Gfp, we evaluated whether deficiency in fractalkine signaling affects BAT remodeling and function in high-fat-diet - induced obesity. Homo- and heterozygote male CX3CR1-GFP mice were fed with normal or fat enriched (FatED) diet for 10weeks. Interscapular BAT was collected for molecular biological analysis. Heterozygous animals in which fractalkine signaling remains intact, gain more weight during FatED than CX3CR1 deficient gfp/gfp homozygotes. FatED in controls resulted in macrophage recruitment to the BAT with increased expression of proinflammatory mediators (Il1a, b, Tnfa and Ccl2). Local BAT inflammation was accompanied by increased expression of lipogenic enzymes and resulted in BAT "whitening". By contrast, fractalkine receptor deficiency prevented accumulation of tissue macrophages, selectively attenuated the expression of Tnfa, Il1a and Ccl2, increased BAT expression of lipolytic enzymes (Atgl, Hsl and Mgtl) and upregulated genes involved thermo-metabolism (Ucp1, Pparg Pgc1a) in response to FatED. These results highlight the importance of fractalkine-CX3CR1 interaction in recruitment of macrophages into the BAT of obese mice which might contribute to local tissue inflammation, adipose tissue remodeling and regulation of metabolic-related genes.


Subject(s)
Adipose Tissue, Brown/metabolism , Gene Expression Regulation , Macrophages/metabolism , Obesity/genetics , Obesity/metabolism , Receptors, Chemokine/metabolism , Adipose Tissue, White/metabolism , Animals , Biomarkers/metabolism , Body Weight , CX3C Chemokine Receptor 1 , Chemokine CX3CL1/deficiency , Chemokine CX3CL1/metabolism , Cold Temperature , Diet, High-Fat , Green Fluorescent Proteins/metabolism , Inflammation Mediators/metabolism , Lipogenesis/genetics , Lipolysis/genetics , Male , Mice, Inbred C57BL , Organ Size , Receptors, Chemokine/deficiency , Thermogenesis/genetics , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
19.
Am J Respir Cell Mol Biol ; 55(5): 657-666, 2016 11.
Article in English | MEDLINE | ID: mdl-27285858

ABSTRACT

LPS-induced lung injury in the mouse is one of the most robust experimental models used for studies of acute lung injury (ALI) and acute respiratory distress syndrome in humans. Prior clinical and experimental studies support an important role for complement activation, particularly production of C5a, in the pathophysiology of human ALI/acute respiratory distress syndrome. In the mouse model, however, the precise role of C5a and its receptors is unclear. C5L2, an enigmatic second receptor for C5a, has been characterized, and results have generated substantial debate regarding its in vivo function. Our previous work with human neutrophils revealed a unique role for C5L2 in negatively modulating C5a-C5a receptor (C5aR)-mediated cellular activation, in which antibody-mediated blockade of C5L2 resulted in augmented C5a-C5aR responses. Here, we demonstrate that C5L2-/- mice (BALB/c background) administered intranasal LPS exhibit significantly more airway edema and hemorrhage than do wild-type animals. Bronchoalveolar lavage fluid and lung homogenates have significantly more neutrophils and myeloperoxidase activity, as well as proinflammatory cytokines and chemokines. When a blocking antibody against the C5aR was administered before LPS administration, the increased neutrophilic infiltration and cytokine levels were reversed. Thus, our data show not only that C5a contributes significantly to LPS-induced ALI in the mouse, but also that C5L2 plays an important antiinflammatory role in this model through actions resulting at least in part from negative modulation of C5a receptor activation.


Subject(s)
Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Receptors, Chemokine/metabolism , Acute Lung Injury/genetics , Acute Lung Injury/physiopathology , Administration, Intranasal , Animals , Bronchoalveolar Lavage Fluid , Chemokines/genetics , Chemokines/metabolism , Edema/complications , Edema/pathology , Edema/physiopathology , Gene Expression Regulation , Hemorrhage/complications , Hemorrhage/pathology , Hemorrhage/physiopathology , Inflammation Mediators/metabolism , Lipopolysaccharides/administration & dosage , Lung/pathology , Mice, Inbred BALB C , Neutrophils/metabolism , Peroxidase/metabolism , Receptor, Anaphylatoxin C5a , Receptors, Chemokine/antagonists & inhibitors , Receptors, Chemokine/deficiency , Respiratory Mechanics , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism
20.
Sci Rep ; 6: 23505, 2016 Mar 23.
Article in English | MEDLINE | ID: mdl-27005831

ABSTRACT

Intestinal immune homeostasis requires dynamic crosstalk between innate and adaptive immune cells. Dendritic cells (DCs) exist as multiple phenotypically and functionally distinct sub-populations within tissues, where they initiate immune responses and promote homeostasis. In the gut, there exists a minor DC subset defined as CD103(+)CD11b(-) that also expresses the chemokine receptor XCR1. In other tissues, XCR1(+) DCs cross-present antigen and contribute to immunity against viruses and cancer, however the roles of XCR1(+) DCs and XCR1 in the intestine are unknown. We showed that mice lacking XCR1(+) DCs are specifically deficient in intraepithelial and lamina propria (LP) T cell populations, with remaining T cells exhibiting an atypical phenotype and being prone to death, and are also more susceptible to chemically-induced colitis. Mice deficient in either XCR1 or its ligand, XCL1, similarly possess diminished intestinal T cell populations, and an accumulation of XCR1(+) DCs in the gut. Combined with transcriptome and surface marker expression analysis, these observations lead us to hypothesise that T cell-derived XCL1 facilitates intestinal XCR1(+) DC activation and migration, and that XCR1(+) DCs in turn provide support for T cell survival and function. Thus XCR1(+) DCs and the XCR1/XCL1 chemokine axis have previously-unappreciated roles in intestinal immune homeostasis.


Subject(s)
Chemokines, C/metabolism , Dendritic Cells/physiology , Intestines/immunology , Receptors, Chemokine/metabolism , T-Lymphocytes/cytology , Animals , Cell Movement , Cell Proliferation , Cell Survival , Cells, Cultured , Chemokines, C/deficiency , Cross-Priming , Dendritic Cells/immunology , Gene Expression Profiling/methods , Gene Expression Regulation , Homeostasis , Intestines/cytology , Mice , Receptors, Chemokine/deficiency , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...