Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
Metabolism ; 121: 154804, 2021 08.
Article in English | MEDLINE | ID: mdl-34097917

ABSTRACT

AIMS/HYPOTHESIS: Inflammation has a major role in diabetic kidney disease. We thus investigated the role of the IL-8-CXCR1/2 axis in favoring kidney damage in diabetes. METHODS: Urinary IL-8 levels were measured in 1247 patients of the Joslin Kidney Study in type 2 diabetes (T2D). The expression of IL-8 and of its membrane receptors CXCR1/CXCR2 was quantified in kidney tissues in patients with T2D and in controls. The effect of CXCR1/2 blockade on diabetic kidney disease was evaluated in db/db mice. RESULTS: IL-8 urinary levels were increased in patients with T2D and diabetic kidney disease, with the highest urinary IL-8 levels found in the patients with the largest decline in glomerular filtration rate, with an increased albumin/creatine ratio and the worst renal outcome. Moreover, glomerular IL-8 renal expression was increased in patients with T2D, as compared to controls. High glucose elicits abundant IL-8 secretion in cultured human immortalized podocytes in vitro. Finally, in diabetic db/db mice and in podocytes in vitro, CXCR1/2 blockade mitigated albuminuria, reduced mesangial expansion, decreased podocyte apoptosis and reduced DNA damage. CONCLUSIONS/INTERPRETATION: The IL-8- CXCR1/2 axis may have a role in diabetic kidney disease by inducing podocyte damage. Indeed, targeting the IL-8-CXCR1/2 axis may reduce the burden of diabetic kidney disease.


Subject(s)
Diabetic Nephropathies/genetics , Interleukin-8/physiology , Receptors, CXCR/physiology , Adult , Animals , Case-Control Studies , Cells, Cultured , Cohort Studies , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Humans , Interleukin-8/genetics , Interleukin-8/metabolism , Italy , Kidney/metabolism , Kidney/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Podocytes/metabolism , Podocytes/pathology , Receptors, Interleukin-8A/genetics , Receptors, Interleukin-8A/metabolism , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/metabolism , Signal Transduction/physiology
2.
Med Oncol ; 38(5): 58, 2021 Apr 13.
Article in English | MEDLINE | ID: mdl-33847822

ABSTRACT

C-X-C chemokine receptor 7 (CXCR7), a novel receptor of C-X-C motif chemokine ligand 12 (CXCL12), is associated with the occurrence and metastasis of various malignant tumours. However, the role, function and underlying mechanisms of CXCR7 expression in cervical cancer remain undefined. The expression level of CXCR7 was evaluated in cervical cancer samples by immunohistochemistry and real-time PCR analyses. Western blot analysis was used to examine the expression level of CXCR7 in cervical cancer cell lines. HeLa cells were genetically silenced or pharmacologically inhibited for CXCR7 or CXCR4. Transwell and CCK-8 assays were used to examine cell migration and proliferation. The expression levels of MMP2, MMP9, TIMP-1 and TIMP-2 in HeLa cells were assessed by western blot or real-time PCR. HeLa cells silenced for CXCR7 were subcutaneously injected into nude mice to form tumours. The expression of CXCR7 in nude mice was investigated by immunohistochemical staining. Tumour volumes and weights were measured. The in vivo expression levels of MMP2, MMP9, TIMP-1 and TIMP-2 were determined by western blot analysis and real-time PCR. CXCR7 was overexpressed in cervical cancer tissues and cell lines. CXCL12 was highly expressed in cervical cancer lines. CXCR7 silencing or CCX733 treatment rather than CXCR4 silencing or AMD3100 treatment suppressed the proliferation, migration and invasion of cervical cancer cells stimulated by CXCL12. In a xenograft tumour model, CXCR7 silencing or CCX733 treatment inhibited the volumes and weights of xenograft tumours. In addition, downregulation of CXCR7 decreased the expression levels of MMP2 and MMP9 but increased the expression levels of TIMP-1 and TIMP-2 in vivo. These data support the finding that the downregulation of CXCR7 suppresses the proliferation and metastasis of cervical cancer cells. Inhibition of CXCR7 may be a potential targeted therapy for cervical cancer.


Subject(s)
Carcinoma, Squamous Cell/pathology , Chemokine CXCL12/physiology , Neoplasm Proteins/physiology , Receptors, CXCR/physiology , Signal Transduction/physiology , Uterine Cervical Neoplasms/pathology , Animals , Carcinoma, Squamous Cell/drug therapy , Cell Division , Cell Line , Cell Line, Tumor , Cervix Uteri/cytology , Epithelial Cells/metabolism , Female , Humans , Matrix Metalloproteinases/biosynthesis , Mice , Mice, Nude , Molecular Targeted Therapy , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Proteins/antagonists & inhibitors , RNA Interference , RNA, Small Interfering/genetics , Receptors, CXCR/antagonists & inhibitors , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/genetics , Receptors, CXCR4/physiology , Uterine Cervical Neoplasms/drug therapy , Xenograft Model Antitumor Assays
3.
Osteoarthritis Cartilage ; 29(3): 313-322, 2021 03.
Article in English | MEDLINE | ID: mdl-33253889

ABSTRACT

Stromal cell-derived factor 1 (SDF-1), also known as CXC motif chemokine ligand 12 (CXCL12), is recognized as a homeostatic cytokine with strong chemotactic potency. It plays an important role in physiological and pathological processes, such as the development of multiple tissues and organs, the regulation of cell distribution, and tumour metastasis. SDF-1 has two receptors, CXC chemokine receptor type 4 (CXCR4) and CXC chemokine receptor type 7 (CXCR7). SDF-1 affects the proliferation, survival, differentiation and maturation of chondrocytes by binding to CXCR4 on chondrocytes. Therefore, SDF-1 has been used as an exogenous regulatory target in many studies to explore the mechanism of cartilage development. SDF-1 is also a potential therapeutic target for osteoarthritis (OA) and rheumatoid arthritis (RA), because of its role in pathological initiation and regulation. In addition, SDF-1 shows potent capacity in the repair of cartilage defects by recruiting endogenous stem cells in a cartilage tissue engineering context. To summarize the specific role of SDF-1 on cartilage development and disease, all articles had been screened out in PubMed by May 30, 2020. The search was limited to studies published in English. Search terms included SDF-1; CXCL12; CXCR4; chondrocyte; cartilage; OA; RA, and forty-seven papers were studied. Besides, we reviewed references in the articles we searched to get additional relevant backgrounds. The review aims to conclude the current knowledge regarding the physiological and pathological role of SDF-1 on the cartilage and chondrocyte. More investigations are required to determine methods targeted SDF-1 to cartilage development and interventions to cartilage diseases.


Subject(s)
Arthritis, Rheumatoid/metabolism , Chemokine CXCL12/metabolism , Chondrocytes/physiology , Chondrogenesis/physiology , Osteoarthritis/metabolism , Arthritis, Rheumatoid/physiopathology , Cartilage Diseases/metabolism , Cartilage Diseases/physiopathology , Cell Differentiation/physiology , Cell Proliferation/physiology , Cell Survival , Chemokine CXCL12/physiology , Humans , Osteoarthritis/physiopathology , Receptors, CXCR/physiology , Receptors, CXCR4/physiology
4.
Article in English | MEDLINE | ID: mdl-32816229

ABSTRACT

Chemokine receptors, a diverse group within the seven-transmembrane G protein-coupled receptor superfamily, are frequently overexpressed in malignant tumors. Ligand binding activates multiple downstream signal transduction cascades that drive tumor growth and metastasis, resulting in poor clinical outcome. These receptors are thus considered promising targets for anti-tumor therapy. This article reviews recent studies on the expression and function of CXC chemokine receptors in various tumor microenvironments and recent developments in cancer therapy using CXC chemokine receptor antagonists.


Subject(s)
Neoplasms/pathology , Receptors, CXCR/physiology , Signal Transduction , Tumor Microenvironment , Humans
5.
J Cell Mol Med ; 24(18): 10589-10603, 2020 09.
Article in English | MEDLINE | ID: mdl-32761803

ABSTRACT

Low-energy shock wave (LESW) has been recognized as a promising non-invasive intervention to prevent the organs or tissues against ischaemia reperfusion injury (IRI), whereas its effect on kidney injury is rarely explored. To investigate the protective role of pretreatment with LESW on renal IRI in rats, animals were randomly divided into Sham, LESW, IRI and LESW + IRI groups. At 4, 12, 24 hours and 3 and 7 days after reperfusion, serum samples and renal tissues were harvested for performing the analysis of renal function, histopathology, immunohistochemistry, flow cytometry and Western blot, as well as enzyme-linked immunosorbent assay. Moreover, circulating endothelial progenitor cells (EPCs) were isolated, labelled with fluorescent dye and injected by tail vein. The fluorescent signals of EPCs were detected using fluorescence microscope and in vivo imaging system to track the distribution of injected circulating EPCs. Results showed that pretreatment with LESW could significantly reduce kidney injury biomarkers, tubular damage, and cell apoptosis, and promote cell proliferation and vascularization in IRI kidneys. The renoprotective role of LESW pretreatment would be attributed to the remarkably increased EPCs in the treated kidneys, part of which were recruited from circulation through SDF-1/CXCR7 pathway. In conclusion, pretreatment with LESW could increase the recruitment of circulating EPCs to attenuate and repair renal IRI.


Subject(s)
Endothelial Progenitor Cells/physiology , Extracorporeal Shockwave Therapy , Kidney/blood supply , Reperfusion Injury/prevention & control , Animals , Apoptosis , Cell Movement , Chemokine CXCL12/biosynthesis , Chemokine CXCL12/genetics , Chemokine CXCL12/physiology , Extracorporeal Shockwave Therapy/methods , Fluorescent Dyes/pharmacokinetics , In Situ Nick-End Labeling , Kidney/pathology , Kidney/physiology , Male , Microscopy, Fluorescence , Microvessels/pathology , Random Allocation , Rats , Rats, Sprague-Dawley , Receptors, CXCR/antagonists & inhibitors , Receptors, CXCR/biosynthesis , Receptors, CXCR/genetics , Receptors, CXCR/physiology , Regeneration , Reperfusion Injury/blood , Reperfusion Injury/pathology , Signal Transduction , Time Factors
6.
Mol Pharmacol ; 96(6): 737-752, 2019 12.
Article in English | MEDLINE | ID: mdl-31548340

ABSTRACT

The two G protein-coupled receptors (GPCRs) C-X-C chemokine receptor type 4 (CXCR4) and atypical chemokine receptor 3 (ACKR3) are part of the class A chemokine GPCR family and represent important drug targets for human immunodeficiency virus (HIV) infection, cancer, and inflammation diseases. CXCR4 is one of only three chemokine receptors with a US Food and Drug Administration approved therapeutic agent, the small-molecule modulator AMD3100. In this review, known modulators of the two receptors are discussed in detail. Initially, the structural relationship between receptors and ligands is reviewed on the basis of common structural motifs and available crystal structures. To date, no atypical chemokine receptor has been crystallized, which makes ligand design and predictions for these receptors more difficult. Next, the selectivity, receptor activation, and the resulting ligand-induced signaling output of chemokines and other peptide ligands are reviewed. Binding of pepducins, a class of lipid-peptides whose basis is the internal loop of a GPCR, to CXCR4 is also discussed. Finally, small-molecule modulators of CXCR4 and ACKR3 are reviewed. These modulators have led to the development of radio- and fluorescently labeled tool compounds, enabling the visualization of ligand binding and receptor characterization both in vitro and in vivo. SIGNIFICANCE STATEMENT: To investigate the pharmacological modulation of CXCR4 and ACKR3, significant effort has been focused on the discovery and development of a range of ligands, including small-molecule modulators, pepducins, and synthetic peptides. Imaging tools, such as fluorescent probes, also play a pivotal role in the field of drug discovery. This review aims to provide an overview of the aforementioned modulators that facilitate the study of CXCR4 and ACKR3 receptors.


Subject(s)
Receptors, CXCR4/physiology , Receptors, CXCR/physiology , Amino Acid Sequence , Animals , Benzylamines , Cyclams , Heterocyclic Compounds/metabolism , Heterocyclic Compounds/pharmacology , Humans , Protein Binding/drug effects , Protein Binding/physiology , Protein Structure, Secondary , Protein Structure, Tertiary , Receptors, CXCR/agonists , Receptors, CXCR/antagonists & inhibitors , Receptors, CXCR4/agonists , Receptors, CXCR4/antagonists & inhibitors
7.
PLoS One ; 13(6): e0198789, 2018.
Article in English | MEDLINE | ID: mdl-29920526

ABSTRACT

The CXCR4/CXCL12 axis has been extensively associated with different types of cancer correlating with higher aggressiveness and metastasis. In diffuse large B-cell lymphoma (DLBCL), the expression of the chemokine receptor CXCR4 is involved in the dissemination of malignant B cells and is a marker of poor prognosis. CXCR7 is a chemokine receptor that binds to the same ligand as CXCR4 and regulates de CXCR4-CXCL12 axis. These findings together with the report of CXCR7 prognostic value in several tumor types, led us to evaluate the expression of CXCR7 in diffuse large B-cell lymphoma biopsies. Here, we describe that CXCR7 receptor is an independent prognostic factor that associates with good clinical outcome. Moreover, the expression of CXCR7 associates with increased survival in CXCR4+ but not in CXCR4- DLBCL patients. Thus, the combined immunohistochemical evaluation of both CXCR7 and CXCR4 expression in DLBCL biopsies may improve their prognostic value as single markers. Finally, we show that CXCR7 overexpression in vitro is able to diminish DLBCL cell survival and increase their sensitivity to antitumor drugs. Hence, further studies on the CXCR7 receptor may establish its role in DLBCL and the molecular mechanisms that modulate CXCR4 activity.


Subject(s)
Gene Expression Regulation, Neoplastic , Lymphoma, Large B-Cell, Diffuse/genetics , Neoplasm Proteins/biosynthesis , Receptors, CXCR4/analysis , Receptors, CXCR/biosynthesis , Adult , Aged , Biomarkers, Tumor , Biopsy , Cell Line, Tumor , Chemokine CXCL12/physiology , Drug Resistance, Neoplasm/genetics , Female , Humans , Kaplan-Meier Estimate , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/mortality , Lymphoma, Large B-Cell, Diffuse/pathology , Male , Middle Aged , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Prognosis , Proportional Hazards Models , Receptors, CXCR/genetics , Receptors, CXCR/physiology
8.
Eur Heart J ; 38(25): 1993-2005, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28431006

ABSTRACT

AIMS: Hyperlipidaemia enhances susceptibility to thrombosis, while platelet oxidixed LDL (oxLDL) binding in acute coronary syndrome (ACS) correlates with activation status. This study explores the platelet lipidome in symptomatic coronary artery disease (CAD) patients and the functional consequences of the chemokine CXCL12 and its receptors CXCR-4/-7 on lipid uptake in platelets. METHODS AND RESULTS: Platelet-oxLDL detected by flow cytometry was enhanced (P = 0.04) in CAD patients, moderately correlated with platelet CXCR7 surface expression (ρ = 0.39; P < 0.001), while inversely with CXCR4 (ρ = 0.35; P < 0.001). Platelet-oxLDL was elevated (P = 0.01) in ACS patients with angiographic evidence of intracoronary thrombi. Ex vivo analysis of intracoronary thrombi sections revealed oxLDL deposition in platelet-enriched areas verified by immunofluorescence confocal microscopy. LDL-oxLDL uptake enhanced reactive oxygen species, mitochondrial superoxide generation, intraplatelet LDL to oxLDL conversion, and lipid peroxidation, counteracted by SOD2-mimetic MnTMPyP. Lipidomic analysis revealed enhanced intraplatelet-oxidized phospholipids, cholesteryl esters, sphingomyelin, ceramides, di- and triacylglycerols, acylcarnitines in CAD patients compared with age-matched controls as ascertained by liquid chromatography hyphenated to high-resolution mass spectrometry. LDL-oxLDL induced degranulation, αIIbß3-integrin activation, apoptosis, thrombin generation estimated by calibrated automated thrombinoscopy, and shape change verified by live imaging using scanning ion conductance microscopy. Further, LDL-oxLDL enhanced thrombus formation ex vivo and in vivo in mice (ferric chloride-induced carotid artery injury). LDL-oxLDL enhanced platelet CXCL12 release, differentially regulated CXCR4-CXCR7 surface exposure, while CXCL12 prompted LDL-oxLDL uptake and synergistically augmented the LDL-oxLDL-induced pro-oxidative, thrombogenic impact on platelet function. CONCLUSION: An altered platelet lipidome might be associated with thrombotic disposition in CAD, a mechanism potentially regulated by CXCL12-CXCR4-CXCR7 axis.


Subject(s)
Blood Platelets/metabolism , Coronary Artery Disease/etiology , Lipoproteins, LDL/metabolism , Acute Coronary Syndrome/etiology , Acute Coronary Syndrome/metabolism , Aged , Case-Control Studies , Chemokine CXCL12/metabolism , Chemokine CXCL12/physiology , Coronary Artery Disease/metabolism , Coronary Thrombosis/etiology , Coronary Thrombosis/metabolism , Female , Humans , Lipid Metabolism/physiology , Lipid Peroxidation/physiology , Male , Middle Aged , Reactive Oxygen Species/metabolism , Receptors, CXCR/metabolism , Receptors, CXCR/physiology , Receptors, CXCR4/metabolism , Receptors, CXCR4/physiology
9.
Arch Gynecol Obstet ; 295(4): 987-995, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28239742

ABSTRACT

PURPOSE: Stroma-derived factor-1 (SDF-1) and its receptor C-X-C chemokine receptor-4 (CXCR4) are involved in human endometrial carcinoma (EC) progression. CXCR7 is another important receptor of SDF-1 and has a higher affinity with SDF-1 compared with that of CXCR4. This paper aims to study the effects of the SDF-1/CXCR7 axis on the growth and invasion ability of EC cells. METHODS: CXCR7 expression was evaluated by quantitative RT-PCR, immunohistochemistry, immunocytochemistry and Western blotting in EC cell lines and 30 cases of primary EC tissue from patients. EC cell line proliferation and migration were assessed following knockdown of CXCR7 by MTT and transwell assays. RESULTS: The results showed that CXCR7 was highly expressed at both mRNA and protein levels in the EC cells and tissue. siCXCR7 effectively silenced CXCR7 in Ishikawa and AN3CA cells. Treatment with 17ß-oestradiol (17ß-E2) significantly increased the levels of CXCR7 and SDF-1 in Con, siCon and siCXCR7 treated Ishikawa. siCXCR7 persistently inhibited CXCR7 expression, even in cells treated with 17ß-E2. Moreover, in vitro functional analyses, silencing CXCR7 resulted in decreased proliferation in Ishikawa and AN3CA cells. Treatment with 17ß-E2 and SDF-1 significantly promoted the growth and migration in siCon treated Ishikawa and AN3CA. Interestingly, in response to 17ß-E2 and SDF-1 stimulation, siCXCR7 continuously inhibited the growth and invasion of Ishikawa and AN3CA cells. CONCLUSION: Our results indicate that SDF-1/CXCR7 plays a positive role in the proliferation and invasion of EC cells. CXCR7 inhibition treatment may provide a promising strategy for anti-tumour therapy for EC.


Subject(s)
Cell Proliferation/genetics , Chemokine CXCL12/physiology , Endometrial Neoplasms/pathology , Endothelial Cells/pathology , Neoplasm Invasiveness/genetics , Receptors, CXCR/physiology , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Chemokine CXCL12/genetics , Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Estradiol/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunohistochemistry , RNA, Messenger/metabolism , Receptors, CXCR/genetics , Signal Transduction/drug effects , Signal Transduction/genetics
10.
PLoS One ; 11(1): e0146503, 2016.
Article in English | MEDLINE | ID: mdl-26741980

ABSTRACT

Differentiation of oligodendroglial precursor cells (OPCs), a crucial prerequisite for central nervous system (CNS) remyelination in diseases such as Multiple Sclerosis (MS), is modulated by a multitude of extrinsic and intrinsic factors. In a previous study we revealed that the chemokine CXCL12 stimulates rodent OPC differentiation via activation of its receptor CXCR7. We could now demonstrate that CXCR7 is also expressed on NogoA- and Nkx2.2-positive oligodendroglial cells in human MS brains and that stimulation of cultured primary fetal human OPCs with CXCL12 promotes their differentiation as measured by surface marker expression and morphologic complexity. Pharmacological inhibition of CXCR7 effectively blocks these CXCL12-dependent effects. Our findings therefore suggest that a specific activation of CXCR7 could provide a means to promote oligodendroglial differentiation facilitating endogenous remyelination activities.


Subject(s)
Oligodendroglia/physiology , Receptors, CXCR/physiology , Adult , Aged , Case-Control Studies , Cell Differentiation , Cell Shape , Cells, Cultured , Chemokine CXCL12/physiology , Female , Homeobox Protein Nkx-2.2 , Homeodomain Proteins , Humans , Male , Middle Aged , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Nuclear Proteins , Retrospective Studies , Transcription Factors , Young Adult
11.
Oncogene ; 35(16): 2123-32, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-26212008

ABSTRACT

In the tumor microenvironment, chemokine system has a critical role in tumorigenesis and metastasis. The acquisition of stem-like properties by cancer cells is involved in metastasis and drug resistance, which are pivotal problems that result in poor outcomes in patients with lung cancer. Patients with advanced lung cancer present high plasma levels of transforming growth factor-ß1 (TGFß1), which correlate with poor prognostic features. Therefore, TGFß1 may be important in the tumor microenvironment, where chemokines are widely expressed. However, the role of chemokines in TGFß1-induced tumor progression still remains unclear. In our study, TGFß1 upregulated CXC chemokine receptor expression, migration, invasion, epithelial-mesenchymal transition (EMT) and cancer stem cell (CSC) formation in lung adenocarcinoma. We found that CXCR7 was the most upregulated chemokine receptor induced by TGFß1. CXCR7 knockdown resulted in reduction of migration, invasion and EMT induced by TGFß1, whereas CXCR4 knockdown did not reverse TGFß1-promoted EMT. CXCR7 silencing significantly decreased cancer sphere-forming capacity, stem-like properties, chemoresistance and TGFß1-induced CSC tumor initiation in vivo. In clinical samples, high TGFß1 and CXCR7 expression was significantly associated with the late stages of lung adenocarcinoma. Moreover, TGFß1 and CXCR7 coexpression was positively correlated with the CSC marker, CD44, which is associated with lymph node metastasis. Besides, patients with high expression of both CXCR7 and TGFß1 presented a significantly worse survival rate. These results suggest that the TGFß1-CXCR7 axis may be a prognostic marker and may provide novel targets for combinational therapies to be used in the treatment of advanced lung cancer in the future.


Subject(s)
Adenocarcinoma/pathology , Epithelial-Mesenchymal Transition/physiology , Lung Neoplasms/pathology , Receptors, CXCR/physiology , Transforming Growth Factor beta1/physiology , Adenocarcinoma/metabolism , Cell Line, Tumor , Humans , Lung Neoplasms/metabolism , Neoplastic Stem Cells/pathology , Receptors, CXCR/genetics , Receptors, CXCR4/genetics , Receptors, CXCR4/physiology , Signal Transduction , Smad Proteins/metabolism
12.
Oncogene ; 35(13): 1716-24, 2016 Mar 31.
Article in English | MEDLINE | ID: mdl-26119946

ABSTRACT

Atypical chemokine receptor CXCR7 (ACKR3) functions as a scavenger receptor for chemokine CXCL12, a molecule that promotes multiple steps in tumor growth and metastasis in breast cancer and multiple other malignancies. Although normal vascular endothelium expresses low levels of CXCR7, marked upregulation of CXCR7 occurs in tumor vasculature in breast cancer and other tumors. To investigate effects of endothelial CXCR7 in breast cancer, we conditionally deleted this receptor from vascular endothelium of adult mice, generating CXCR7(ΔEND/ΔEND) animals. CXCR7(ΔEND/ΔEND) mice appeared phenotypically normal, although these animals exhibited a modest 35±3% increase in plasma CXCL12 as compared with control. Using two different syngeneic, orthotopic tumor implant models of breast cancer, we discovered that CXCR7(ΔEND/ΔEND) mice had significantly greater local recurrence of cancer following resection, elevated numbers of circulating tumor cells and more spontaneous metastases. CXCR7(ΔEND/ΔEND) mice also showed greater experimental metastases following intracardiac injection of cancer cells. These results establish that endothelial CXCR7 limits breast cancer metastasis at multiple steps in the metastatic cascade, advancing understanding of CXCL12 pathways in tumor environments and informing ongoing drug development targeting CXCR7 in cancer.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Endothelium, Vascular/metabolism , Receptors, CXCR/physiology , Animals , Cell Line, Tumor , Female , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Metastasis , Receptors, CXCR/genetics , Receptors, CXCR/metabolism , Tumor Microenvironment/genetics
13.
Stem Cells Transl Med ; 4(10): 1122-30, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26253714

ABSTRACT

UNLABELLED: Remyelination is an important repair process after ischemic stroke-induced white matter injury. It often fails because of the insufficient recruitment of oligodendrocyte progenitor cells (OPCs) to the demyelinated site or the inefficient differentiation of OPCs to oligodendrocytes. We investigated whether CXCL12 gene therapy promoted remyelination after middle cerebral artery occlusion in adult mice. The results showed that CXCL12 gene therapy at 1 week after ischemia could protect myelin sheath integrity in the perifocal region, increase the number of platelet-derived growth factor receptor-α (PDGFRα)-positive and PDGFRα/bromodeoxyuridine-double positive OPCs in the subventricular zone, and further enhance their migration to the ischemic lesion area. Coadministration of AMD3100, the antagonist for CXCL12 receptor CXCR4, eliminated the beneficial effect of CXCL12 on myelin sheath integrity and negatively influenced OPC proliferation and migration. At 5 weeks after ischemia, CXCR4 was found on the PDGFRα- and/or neuron/glia type 2 (NG2)-positive OPCs but not on the myelin basic protein-positive mature myelin sheaths, and CXCR7 was only expressed on the mature myelin sheath in the ischemic mouse brain. Our data indicated that CXCL12 gene therapy effectively protected white matter and promoted its repair after ischemic injury. The treatment at 1 week after ischemia is effective, suggesting that this strategy has a longer therapeutic time window than the treatments currently available. SIGNIFICANCE: This study has demonstrated for the first time that CXCL12 gene therapy significantly ameliorates brain ischemia-induced white matter injury and promotes oligodendrocyte progenitor cell proliferation in the subventricular zone and migration to the perifocal area in the ischemic mouse brain. Additional data showed that CXCR4 receptor plays an important role during the proliferation and migration of oligodendrocyte progenitor cells, and CXCR7 might play a role during maturation. In contrast to many experimental studies that provide treatment before ischemic insult, CXCL12 gene therapy was performed 1 week after brain ischemia, which significantly prolonged the therapeutic time window of brain ischemia.


Subject(s)
Brain Ischemia/therapy , Chemokine CXCL12/genetics , Genetic Therapy , White Matter/pathology , Acute Disease , Animals , Biomarkers/analysis , Brain Ischemia/pathology , Cell Division , Cell Movement , Dependovirus/genetics , Gene Expression Profiling , Genetic Vectors/genetics , Genetic Vectors/therapeutic use , Male , Mice , Mice, Inbred ICR , Myelin Sheath/physiology , Oligodendroglia/cytology , Receptor, Platelet-Derived Growth Factor alpha/analysis , Receptors, CXCR/physiology , Receptors, CXCR4/physiology
14.
Dev Cell ; 30(5): 490-1, 2014 Sep 08.
Article in English | MEDLINE | ID: mdl-25203203

ABSTRACT

Decoy receptors have ligand binding capacity but, in contrast to cognate receptors, do not initiate typical downstream signaling cascades. In this issue of Developmental Cell, Klein and colleagues (2014) demonstrate that CXCR7 acts as a decoy receptor for adrenomedullin, a peptide hormone with key roles in cardiovascular development.


Subject(s)
Adrenomedullin/metabolism , Gene Expression Regulation, Developmental , Heart/embryology , Lymphatic Vessels/embryology , Receptors, CXCR/physiology , Animals , Female , Humans , Male
15.
Dev Cell ; 30(5): 528-40, 2014 Sep 08.
Article in English | MEDLINE | ID: mdl-25203207

ABSTRACT

Atypical 7-transmembrane receptors, often called decoy receptors, act promiscuously as molecular sinks to regulate ligand bioavailability and consequently temper the signaling of canonical G protein-coupled receptor (GPCR) pathways. Loss of mammalian CXCR7, the most recently described decoy receptor, results in postnatal lethality due to aberrant cardiac development and myocyte hyperplasia. Here, we provide the molecular underpinning for this proliferative phenotype by demonstrating that the dosage and signaling of adrenomedullin (Adm, gene; AM, protein)-a mitogenic peptide hormone required for normal cardiovascular development-is tightly controlled by CXCR7. To this end, Cxcr7(-/-) mice exhibit gain-of-function cardiac and lymphatic vascular phenotypes that can be reversed upon genetic depletion of adrenomedullin ligand. In addition to identifying a biological ligand accountable for the phenotypes of Cxcr7(-/-) mice, these results reveal a previously underappreciated role for decoy receptors as molecular rheostats in controlling the timing and extent of GPCR-mediated cardiac and vascular development.


Subject(s)
Adrenomedullin/metabolism , Gene Expression Regulation, Developmental , Heart/embryology , Lymphatic Vessels/embryology , Receptors, CXCR/physiology , Animals , Cell Movement , Cell Proliferation , Female , HEK293 Cells , Humans , Ligands , Male , Mice , Mice, Knockout , Muscle Cells/cytology , Oligonucleotide Array Sequence Analysis , Phenotype , Receptors, CXCR/genetics , Signal Transduction
16.
Arthritis Rheumatol ; 66(11): 3001-12, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25132679

ABSTRACT

OBJECTIVE: The chemokine receptor CXCR6 is highly expressed on lymphocytes isolated from the synovium of patients with rheumatoid arthritis, psoriatic arthritis, or juvenile idiopathic arthritis, suggesting that CXCR6 regulates immune cell activation or infiltration into arthritic joints. This study was undertaken to examine the role of CXCR6 in T cell activation and arthritis development. METHODS: A collagen-induced arthritis model was used to examine arthritis development in wild-type and CXCR6(-/-) mice. CXCR6 expression, lymphocyte accumulation, and intracellular cytokine production were examined by flow cytometry. Collagen-specific antibodies were measured in the serum. Collagen-specific recall responses were examined in vitro via proliferation and cytokine release assays. T cell homing to inflamed joints was examined using competitive adoptive transfer of dye-labeled lymphocytes from wild-type and CXCR6(-/-) mice. RESULTS: The numbers of CXCR6+ T cells were increased in the paws and draining lymph nodes of arthritic mice. The incidence of arthritis, disease severity, extent of T cell accumulation, and levels of collagen-specific IgG2a antibodies were significantly reduced in CXCR6(-/-) mice compared to wild-type mice. T cells from wild-type mice exhibited Th1 (interferon-γ [IFNγ]) polarization in the inguinal lymph nodes following immunization. At disease peak, this shifted to a Th17 (interleukin-17A [IL-17A]) response in the popliteal lymph nodes. T cells in CXCR6(-/-) mice exhibited impaired cytokine polarization, resulting in a decreased frequency and number of IL-17A- and IFNγ-producing cells. Recruitment of activated CXCR6(-/-) mouse T cells to the inflamed paws was impaired compared to recruitment of wild-type mouse T cells. CONCLUSION: These experiments demonstrate that CXCR6 plays important roles in the pathogenesis of arthritis through its effects on both T cell cytokine polarization and homing of T cells to inflamed joints.


Subject(s)
Arthritis, Experimental/pathology , Arthritis, Experimental/physiopathology , Cell Movement/physiology , Cytokines/metabolism , Receptors, CXCR/physiology , T-Lymphocytes/pathology , Animals , Arthritis, Experimental/metabolism , Disease Models, Animal , Female , Immunoglobulin G/metabolism , In Vitro Techniques , Incidence , Interferon-gamma/metabolism , Interleukin-17/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, CXCR/deficiency , Receptors, CXCR/genetics , Receptors, CXCR6 , Severity of Illness Index , Up-Regulation/physiology
17.
Development ; 141(16): 3188-96, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25063456

ABSTRACT

Collective migration of cells in the zebrafish posterior lateral line primordium (PLLp) along a path defined by Cxcl12a expression depends on Cxcr4b receptors in leading cells and on Cxcr7b in trailing cells. Cxcr7b-mediated degradation of Cxcl12a by trailing cells generates a local gradient of Cxcl12a that guides PLLp migration. Agent-based computer models were built to explore how a polarized response to Cxcl12a, mediated by Cxcr4b in leading cells and prevented by Cxcr7b in trailing cells, determines unidirectional migration of the PLLp. These chemokine signaling-based models effectively recapitulate many behaviors of the PLLp and provide potential explanations for the characteristic behaviors that emerge when the PLLp is severed by laser to generate leading and trailing fragments. As predicted by our models, the bilateral stretching of the leading fragment is lost when chemokine signaling is blocked in the PLLp. However, movement of the trailing fragment toward the leading cells, which was also thought to be chemokine dependent, persists. This suggested that a chemokine-independent mechanism, not accounted for in our models, is responsible for this behavior. Further investigation of trailing cell behavior shows that their movement toward leading cells depends on FGF signaling and it can be re-oriented by exogenous FGF sources. Together, our observations reveal the simple yet elegant manner in which leading and trailing cells coordinate migration; while leading cells steer PLLp migration by following chemokine cues, cells further back play follow-the-leader as they migrate toward FGFs produced by leading cells.


Subject(s)
Gene Expression Regulation, Developmental , Lateral Line System/embryology , Zebrafish/embryology , Animals , Animals, Genetically Modified , Cell Communication , Cell Movement , Chemokine CXCL12/physiology , Chemokines/metabolism , Computer Simulation , Fibroblast Growth Factors/metabolism , Receptors, CXCR/physiology , Receptors, CXCR4/physiology , Signal Transduction , Zebrafish Proteins/physiology
18.
Mol Med ; 20: 435-47, 2014 Oct 13.
Article in English | MEDLINE | ID: mdl-25032954

ABSTRACT

Chemokine (C-X-C motif) receptor (CXCR) 4 and atypical chemokine receptor (ACKR) 3 ligands have been reported to modulate cardiovascular function in various disease models. The underlying mechanisms, however, remain unknown. Thus, it was the aim of the present study to determine how pharmacological modulation of CXCR4 and ACKR3 regulate cardiovascular function. In vivo administration of TC14012, a CXCR4 antagonist and ACKR3 agonist, caused cardiovascular collapse in normal animals. During the cardiovascular stress response to hemorrhagic shock, ubiquitin, a CXCR4 agonist, stabilized blood pressure, whereas coactivation of CXCR4 and ACKR3 with CXC chemokine ligand 12 (CXCL12), or blockade of CXCR4 with AMD3100 showed opposite effects. While CXCR4 and ACKR3 ligands did not affect myocardial function, they selectively altered vascular reactivity upon α1-adrenergic receptor (AR) activation in pressure myography experiments. CXCR4 activation with ubiquitin enhanced α1-AR-mediated vasoconstriction, whereas ACKR3 activation with various natural and synthetic ligands antagonized α1-AR-mediated vasoconstriction. The opposing effects of CXCR4 and ACKR3 activation by CXCL12 could be dissected pharmacologically. CXCR4 and ACKR3 ligands did not affect vasoconstriction upon activation of voltage-operated Ca(2+) channels or endothelin receptors. Effects of CXCR4 and ACKR3 agonists on vascular α1-AR responsiveness were independent of the endothelium. These findings suggest that CXCR4 and ACKR3 modulate α1-AR reactivity in vascular smooth muscle and regulate hemodynamics in normal and pathological conditions. Our observations point toward CXCR4 and ACKR3 as new pharmacological targets to control vasoreactivity and blood pressure.


Subject(s)
Receptors, Adrenergic, alpha-1/physiology , Receptors, CXCR4/physiology , Receptors, CXCR/physiology , Adrenergic Agonists/pharmacology , Animals , Benzylamines , Blood Pressure/drug effects , Blood Pressure/physiology , Chemokine CXCL12/pharmacology , Cyclams , Heterocyclic Compounds/pharmacology , In Vitro Techniques , Ligands , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/physiology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Oligopeptides/pharmacology , Phenylephrine/pharmacology , Rats, Inbred Lew , Receptors, CXCR/agonists , Receptors, CXCR4/agonists , Receptors, CXCR4/antagonists & inhibitors , Shock, Hemorrhagic/physiopathology , Ubiquitin/pharmacology , Vasoconstriction/drug effects , Ventricular Function, Left/drug effects
19.
J Infect Dis ; 210(9): 1508-16, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-24823625

ABSTRACT

It is well established that immunization with attenuated malaria sporozoites induces CD8(+) T cells that eliminate parasite-infected hepatocytes. Liver memory CD8(+) T cells induced by immunization with parasites undergo a unique differentiation program and have enhanced expression of CXCR6. Following immunization with malaria parasites, CXCR6-deficient memory CD8(+) T cells recovered from the liver display altered cell-surface expression markers as compared to their wild-type counterparts, but they exhibit normal cytokine secretion and expression of cytotoxic mediators on a per-cell basis. Most importantly, CXCR6-deficient CD8(+) T cells migrate to the liver normally after immunization with Plasmodium sporozoites or vaccinia virus, but a few weeks later their numbers severely decrease in this organ, losing their capacity to inhibit malaria parasite development in the liver. These studies are the first to show that CXCR6 is critical for the development and maintenance of protective memory CD8(+) T cells in the liver.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/physiology , Liver Diseases, Parasitic/immunology , Receptors, CXCR/physiology , Adoptive Transfer , Animals , Female , Flow Cytometry , Malaria/immunology , Malaria/parasitology , Male , Mice, Inbred C57BL , Mice, Transgenic , Plasmodium berghei/immunology , Receptors, CXCR6
20.
J Surg Res ; 191(2): 379-88, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24814201

ABSTRACT

BACKGROUND: Although papillary thyroid carcinoma (PTC) has favorable prognosis, it is prone to cervical lymph node metastasis. Chemokine receptors play a role in metastasis of tumor cells, and accumulating evidence suggests an important role for the chemokine receptor CXCR7 in cancer development. We previously demonstrated high expression of CXCR7 protein in PTC tissue. In this study, we further evaluated the role of CXCR7 in PTC. METHODS: The expression of CXCR7 messenger RNA and protein in 79 cases of PTC and peritumoral tissues was detected by real-time quantitative polymerase chain reaction and Western blot. The association between CXCR7 expression and clinicopathologic characteristics in PTC was analyzed. Stable CXCR7 overexpression and knockdown PTC cells were constructed and used to examine proliferation, cell cycle, apoptosis and invasion of PTC cells by 3-(4,5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide, propidium iodide staining, 7-amino-actinomycin D staining, and invasion assay. We examined cell cycle regulatory protein levels by Western blot. RESULTS: CXCR7 messenger RNA and protein levels were markedly increased in PTC and correlated with tumor progression. CXCR7 could regulate proliferation, cell cycle, apoptosis, invasion, and the expression of cell cycle regulatory proteins involved in the S-G2 phase transition. Knockdown of CXCR7 in PTC cells suppressed cell proliferation and invasion, decreased expression of cyclin A, CDK2 and PCNA, increased expression of p21 and p57, induced S phase arrest, and promoted apoptosis. CONCLUSIONS: CXCR7 plays an important role in regulating growth and metastasis ability of PTC cell and provides a potential target for therapeutic interventions in PTC.


Subject(s)
Carcinoma/pathology , Receptors, CXCR/physiology , Thyroid Neoplasms/pathology , Adult , Aged , Apoptosis , Carcinoma, Papillary , Cell Line, Tumor , Cell Proliferation , Chemokine CXCL12/physiology , Disease Progression , Female , Humans , Male , Middle Aged , Neoplasm Invasiveness , RNA, Messenger/analysis , Receptors, CXCR/genetics , Thyroid Cancer, Papillary
SELECTION OF CITATIONS
SEARCH DETAIL
...