Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
Nat Immunol ; 12(10): 949-58, 2011 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-21909092

RESUMEN

The transcription factor RORγt is required for the development of several innate lymphoid populations, such as lymphoid tissue-inducer cells (LTi cells) and cells that secrete interleukin 17 (IL-17) or IL-22. The progenitor cells as well as the developmental stages that lead to the emergence of RORγt(+) innate lymphoid cells (ILCs) remain undefined. Here we identify the chemokine receptor CXCR6 as an additional marker of the development of ILCs and show that common lymphoid progenitors lost B cell and T cell potential as they successively acquired expression of the integrin α(4)ß(7) and CXCR6. Whereas fetal RORγt(+) cells matured in the fetal liver environment, adult bone marrow-derived RORγt(+) ILCs matured outside the bone marrow, in a Notch2-dependent manner. Therefore, fetal and adult environments influence the differentiation of RORγt(+) cells differently.


Asunto(s)
Feto/inmunología , Linfocitos/inmunología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/fisiología , Receptor Notch2/fisiología , Transducción de Señal , Animales , Diferenciación Celular , Células Cultivadas , Proteínas de Unión al ADN/fisiología , Inmunidad Innata , Integrinas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores CXCR/fisiología , Receptores CXCR6
2.
Artículo en Inglés | MEDLINE | ID: mdl-32816229

RESUMEN

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.


Asunto(s)
Neoplasias/patología , Receptores CXCR/fisiología , Transducción de Señal , Microambiente Tumoral , Humanos
3.
Osteoarthritis Cartilage ; 29(3): 313-322, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33253889

RESUMEN

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.


Asunto(s)
Artritis Reumatoide/metabolismo , Quimiocina CXCL12/metabolismo , Condrocitos/fisiología , Condrogénesis/fisiología , Osteoartritis/metabolismo , Artritis Reumatoide/fisiopatología , Enfermedades de los Cartílagos/metabolismo , Enfermedades de los Cartílagos/fisiopatología , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Supervivencia Celular , Quimiocina CXCL12/fisiología , Humanos , Osteoartritis/fisiopatología , Receptores CXCR/fisiología , Receptores CXCR4/fisiología
4.
J Cell Mol Med ; 24(18): 10589-10603, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32761803

RESUMEN

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.


Asunto(s)
Células Progenitoras Endoteliales/fisiología , Tratamiento con Ondas de Choque Extracorpóreas , Riñón/irrigación sanguínea , Daño por Reperfusión/prevención & control , Animales , Apoptosis , Movimiento Celular , Quimiocina CXCL12/biosíntesis , Quimiocina CXCL12/genética , Quimiocina CXCL12/fisiología , Tratamiento con Ondas de Choque Extracorpóreas/métodos , Colorantes Fluorescentes/farmacocinética , Etiquetado Corte-Fin in Situ , Riñón/patología , Riñón/fisiología , Masculino , Microscopía Fluorescente , Microvasos/patología , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Receptores CXCR/antagonistas & inhibidores , Receptores CXCR/biosíntesis , Receptores CXCR/genética , Receptores CXCR/fisiología , Regeneración , Daño por Reperfusión/sangre , Daño por Reperfusión/patología , Transducción de Señal , Factores de Tiempo
5.
Mol Pharmacol ; 96(6): 737-752, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31548340

RESUMEN

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.


Asunto(s)
Receptores CXCR4/fisiología , Receptores CXCR/fisiología , Secuencia de Aminoácidos , Animales , Bencilaminas , Ciclamas , Compuestos Heterocíclicos/metabolismo , Compuestos Heterocíclicos/farmacología , Humanos , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Receptores CXCR/agonistas , Receptores CXCR/antagonistas & inhibidores , Receptores CXCR4/agonistas , Receptores CXCR4/antagonistas & inhibidores
6.
Eur Heart J ; 38(25): 1993-2005, 2017 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-28431006

RESUMEN

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.


Asunto(s)
Plaquetas/metabolismo , Enfermedad de la Arteria Coronaria/etiología , Lipoproteínas LDL/metabolismo , Síndrome Coronario Agudo/etiología , Síndrome Coronario Agudo/metabolismo , Anciano , Estudios de Casos y Controles , Quimiocina CXCL12/metabolismo , Quimiocina CXCL12/fisiología , Enfermedad de la Arteria Coronaria/metabolismo , Trombosis Coronaria/etiología , Trombosis Coronaria/metabolismo , Femenino , Humanos , Metabolismo de los Lípidos/fisiología , Peroxidación de Lípido/fisiología , Masculino , Persona de Mediana Edad , Especies Reactivas de Oxígeno/metabolismo , Receptores CXCR/metabolismo , Receptores CXCR/fisiología , Receptores CXCR4/metabolismo , Receptores CXCR4/fisiología
7.
Development ; 141(16): 3188-96, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25063456

RESUMEN

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.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Sistema de la Línea Lateral/embriología , Pez Cebra/embriología , Animales , Animales Modificados Genéticamente , Comunicación Celular , Movimiento Celular , Quimiocina CXCL12/fisiología , Quimiocinas/metabolismo , Simulación por Computador , Factores de Crecimiento de Fibroblastos/metabolismo , Receptores CXCR/fisiología , Receptores CXCR4/fisiología , Transducción de Señal , Proteínas de Pez Cebra/fisiología
8.
Arch Gynecol Obstet ; 295(4): 987-995, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28239742

RESUMEN

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.


Asunto(s)
Proliferación Celular/genética , Quimiocina CXCL12/fisiología , Neoplasias Endometriales/patología , Células Endoteliales/patología , Invasividad Neoplásica/genética , Receptores CXCR/fisiología , Western Blotting , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quimiocina CXCL12/genética , Neoplasias Endometriales/genética , Neoplasias Endometriales/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Estradiol/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Inmunohistoquímica , ARN Mensajero/metabolismo , Receptores CXCR/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
9.
Hepatology ; 59(5): 1850-63, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24002871

RESUMEN

UNLABELLED: The MYC oncogene is overexpressed in hepatocellular carcinoma (HCC) and has been associated with widespread microRNA (miRNA) repression; however, the underlying mechanisms are largely unknown. Here, we report that the c-Myc oncogenic transcription factor physically interacts with enhancer of zeste homolog 2 (EZH2), a core enzymatic unit of polycomb repressive complex 2 (PRC2). Furthermore, miR-101, an important tumor-suppressive miRNA in human hepatocarcinomas, is epigenetically repressed by PRC2 complex in a c-Myc-mediated manner. miR-101, in turn, inhibits the expression of two subunits of PRC2 (EZH2 and EED), thus creating a double-negative feedback loop that regulates the process of hepatocarcinogenesis. Restoration of miR-101 expression suppresses multiple malignant phenotypes of HCC cells by coordinate repression of a cohort of oncogenes, including STMN1, JUNB, and CXCR7, and further increases expression of endogenous miR-101 by inhibition of PRC2 activation. In addition, co-overexpression of c-Myc and EZH2 in HCC samples was closely associated with lower expression of miR-101 (P < 0.0001) and poorer prognosis of HCC patients (P < 0.01). CONCLUSIONS: c-Myc collaborates with EZH2-containing PRC2 complex in silencing tumor-suppressive miRNAs during hepatocarcinogenesis and provides promising therapeutic candidates for human HCC.


Asunto(s)
Carcinoma Hepatocelular/genética , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Neoplasias Hepáticas/genética , MicroARNs/fisiología , Proteínas Proto-Oncogénicas c-myc/fisiología , Animales , Carcinoma Hepatocelular/mortalidad , Carcinoma Hepatocelular/patología , Metilación de ADN , Proteína Potenciadora del Homólogo Zeste 2 , Humanos , Neoplasias Hepáticas/mortalidad , Neoplasias Hepáticas/patología , Ratones , Ratones Endogámicos BALB C , MicroARNs/antagonistas & inhibidores , Complejo Represivo Polycomb 2/metabolismo , Complejo Represivo Polycomb 2/fisiología , Receptores CXCR/fisiología
10.
J Infect Dis ; 210(9): 1508-16, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24823625

RESUMEN

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.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica/fisiología , Parasitosis Hepáticas/inmunología , Receptores CXCR/fisiología , Traslado Adoptivo , Animales , Femenino , Citometría de Flujo , Malaria/inmunología , Malaria/parasitología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Plasmodium berghei/inmunología , Receptores CXCR6
11.
J Neurosci ; 33(44): 17527-37, 2013 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-24174685

RESUMEN

Gonadotropin-releasing hormone (GnRH) neurons are neuroendocrine cells, located in the hypothalamus, that play an essential role in mammalian reproduction. These neurons originate in the nasal placode and migrate during embryonic development, in association with olfactory/vomeronasal nerves, first in the nose, then through the cribriform plate to enter the forebrain, before settling in the hypothalamus. One of the molecules required for their early migration in the nose is the chemokine CXCL12, which is expressed in the embryonic nasal mesenchyme in an increasing ventral to dorsal gradient, presumably guiding GnRH neurons toward the forebrain. Mice lacking CXCR4, the receptor for CXCL12, exhibit defective GnRH cell movement and a significant reduction in their number, suggesting that CXCL12/CXCR4 signaling is important in the migration and survival of these neurons. Here, we investigated the role of the more recently identified second CXCL12 receptor, CXCR7, in GnRH neuron development. We demonstrate that CXCR7 is expressed along the migratory path of GnRH neurons in the nasal cavity and, although not expressed by GnRH neurons, it affects their migration as indicated by the ectopic accumulation of these cells in the nasal compartment in CXCR7(-/-) mice. Absence of CXCR7 caused abnormal accumulation of CXCL12-RFP at CXCR4-positive sites in the nasal area of CXCL12-RFP-transgenic mice and excessive CXCL12-dependent intracellular clustering of CXCR4 in GnRH neurons, suggesting internalization. These findings imply that CXCR7 regulates CXCL12 availability by acting as a scavenger along the migratory path of GnRH neurons and, thus, influences the migration of these cells in a noncell-autonomous manner.


Asunto(s)
Movimiento Celular/fisiología , Quimiocina CXCL12/genética , Hormona Liberadora de Gonadotropina/fisiología , Neuronas/citología , Neuronas/fisiología , Receptores CXCR/genética , Receptores CXCR/fisiología , Animales , Quimiocina CXCL12/biosíntesis , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Embarazo , Receptores CXCR/deficiencia , Receptores CXCR4/deficiencia , Receptores CXCR4/genética
12.
Mol Med ; 20: 435-47, 2014 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-25032954

RESUMEN

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.


Asunto(s)
Receptores Adrenérgicos alfa 1/fisiología , Receptores CXCR4/fisiología , Receptores CXCR/fisiología , Agonistas Adrenérgicos/farmacología , Animales , Bencilaminas , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Quimiocina CXCL12/farmacología , Ciclamas , Compuestos Heterocíclicos/farmacología , Técnicas In Vitro , Ligandos , Masculino , Arterias Mesentéricas/efectos de los fármacos , Arterias Mesentéricas/fisiología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Oligopéptidos/farmacología , Fenilefrina/farmacología , Ratas Endogámicas Lew , Receptores CXCR/agonistas , Receptores CXCR4/agonistas , Receptores CXCR4/antagonistas & inhibidores , Choque Hemorrágico/fisiopatología , Ubiquitina/farmacología , Vasoconstricción/efectos de los fármacos , Función Ventricular Izquierda/efectos de los fármacos
13.
J Surg Res ; 191(2): 379-88, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24814201

RESUMEN

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.


Asunto(s)
Carcinoma/patología , Receptores CXCR/fisiología , Neoplasias de la Tiroides/patología , Adulto , Anciano , Apoptosis , Carcinoma Papilar , Línea Celular Tumoral , Proliferación Celular , Quimiocina CXCL12/fisiología , Progresión de la Enfermedad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Invasividad Neoplásica , ARN Mensajero/análisis , Receptores CXCR/genética , Cáncer Papilar Tiroideo
14.
Arthritis Rheum ; 65(7): 1736-46, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23633118

RESUMEN

OBJECTIVE: To examine the possibility that CXCL16 recruits endothelial cells (ECs) to developing neovasculature in rheumatoid arthritis (RA) synovium. METHODS: We utilized the RA synovial tissue SCID mouse chimera system to examine human microvascular EC (HMVEC) and human endothelial progenitor cell (EPC) recruitment into engrafted human synovium that was injected intragraft with CXCL16-immunodepleted RA synovial fluid (SF). CXCR6-deficient and wild-type (WT) C57BL/6 mice were primed to develop K/BxN serum-induced arthritis and evaluated for angiogenesis. HMVECs and EPCs from human cord blood were also examined for CXCR6 expression, by immunofluorescence and assessment of CXCL16 signaling activity. RESULTS: CXCR6 was prominently expressed on human EPCs and HMVECs, and its expression on HMVECs could be up-regulated by interleukin-1ß. SCID mice injected with CXCL16-depleted RA SF exhibited a significant reduction in EPC recruitment. In experiments using the K/BxN serum-induced inflammatory arthritis model, CXCR6(-/-) mice showed profound reductions in hemoglobin levels, which correlated with reductions in monocyte and T cell recruitment to arthritic joint tissue compared to that observed in WT mice. Additionally, HMVECs and EPCs responded to CXCL16 stimulation, but exhibited unique signal transduction pathways and homing properties. CONCLUSION: These results indicate that CXCL16 and its receptor CXCR6 may be a central ligand/receptor pair that is closely associated with EPC recruitment and blood vessel formation in the RA joint.


Asunto(s)
Artritis Experimental/metabolismo , Artritis Reumatoide/metabolismo , Quimiocina CXCL6/fisiología , Quimiocinas CXC/fisiología , Células Endoteliales/fisiología , Neovascularización Patológica/metabolismo , Receptores CXCR/fisiología , Receptores de Quimiocina/fisiología , Receptores Depuradores/fisiología , Receptores Virales/fisiología , Animales , Artritis Experimental/fisiopatología , Artritis Reumatoide/fisiopatología , Quimiocina CXCL16 , Quimiotaxis/fisiología , Células Endoteliales/metabolismo , Humanos , Interleucina-1beta/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Ratones Transgénicos , Neovascularización Patológica/fisiopatología , Receptores CXCR/efectos de los fármacos , Receptores CXCR/genética , Receptores CXCR6 , Receptores de Quimiocina/metabolismo , Receptores Virales/metabolismo , Transducción de Señal/fisiología , Células Madre/fisiología , Membrana Sinovial/metabolismo
15.
Biochem Biophys Res Commun ; 441(3): 675-80, 2013 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-24184476

RESUMEN

Numerous studies have reported that CXCR4 and CXCR7 play an essential, but differential role in stromal cell-derived factor-1 (SDF-1)-inducing cell chemotaxis, viability and paracrine actions of BMSCs. Adipose tissue-derived mesenchymal stem cells (ADSCs) have been suggested to be potential seed cells for clinical application instead of bone marrow derived stroma cell (BMSCs). However, the function of SDF-1/CXCR4 and SDF-1/CXCR7 in ADSCs is not well understood. This study was designed to analyze the effect of SDF-1/CXCR4 and SDF-1/CXCR7 axis on ADSCs biological behaviors in vitro. Using Flow cytometry and Western blot methods, we found for the first time that CXCR4/CXCR7 expression was increased after treatment with SDF-1 in ADSCs. SDF-1 promoted ADSCs paracrine, proliferation and migration abilities. CXCR4 or CXCR7 antibody suppressed ADSCs paracrine action induced by SDF-1. The migration of ADSCs can be abolished by CXCR4 antibody, while the proliferation of ADSCs was only downregulated by CXCR7 antibody. Our study indicated that the angiogenesis of ADSCs is, at least partly, mediated by SDF-1/CXCR4 and SDF-1/CXCR7 axis. However, only binding of SDF-1/CXCR7 was required for proliferation of ADSCs, and CXCR7 was required for migration of ADSCs induced by SDF-1. Our studies provide evidence that the activation of either axis may be helpful to improve the effectiveness of ADSCs-based stem cell therapy.


Asunto(s)
Tejido Adiposo/citología , Movimiento Celular , Quimiocina CXCL12/fisiología , Células Madre Mesenquimatosas/fisiología , Neovascularización Fisiológica , Receptores CXCR4/fisiología , Receptores CXCR/fisiología , Anticuerpos , Proliferación Celular , Quimiocina CXCL12/antagonistas & inhibidores , Humanos , Comunicación Paracrina , Receptores CXCR/antagonistas & inhibidores , Receptores CXCR4/antagonistas & inhibidores , Regulación hacia Arriba
16.
Stem Cells ; 30(11): 2571-83, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22987307

RESUMEN

Chemokine CXCL12 is widely expressed in the central nervous system and essential for the proper functioning of human neural progenitor cells (hNPCs). Although CXCL12 is known to function through its receptor CXCR4, recent data have suggested that CXCL12 binds to chemokine receptor CXCR7 with higher affinity than to CXCR4. However, little is known about the function of CXCR7 in hNPCs. Using a primary hNPC culture system, we demonstrated that CXCL12 promotes hNPC survival in the events of camptothecin-induced apoptosis or growth factor deprivation, and that this effect requires both CXCR7 and CXCR4. Through fluorescence-activated cell sorting analysis and immunocytochemistry, we determined that CXCR7 is mainly localized in the early endosome, while CXCR4 is more broadly expressed at the cell surface and on both early and recycling endosomes. Furthermore, we found that endocytosis is required for the prosurvival function of CXCL12. Using dual-color total internal reflection fluorescence microscopy and immunoprecipitation, we demonstrated that CXCR7 quickly trafficks to plasma membrane in mediating CXCL12 endocytosis and colocalizes with CXCR4 after CXCL12 treatment. Investigating the molecular mechanisms, we found that ERK1/2 endocytotic signaling pathway is essential for hNPC survival upon apoptotic challenges. Consistent with these findings, a significantly higher number of apoptotic NPCs were found in the developing brain of CXCR7 knockout mice. In conclusion, CXCL12 protects hNPCs from apoptotic challenges through CXCR7- and CXCR4-mediated endocytotic signaling. Since survival of hNPCs is important for neurogenesis, CXCR7 may become a new therapeutic target to properly regulate critical processes of brain development.


Asunto(s)
Quimiocina CXCL12/fisiología , Endocitosis , Células-Madre Neurales/fisiología , Receptores CXCR4/metabolismo , Receptores CXCR/metabolismo , Animales , Apoptosis/efectos de los fármacos , Camptotecina/farmacología , Supervivencia Celular , Células Cultivadas , Cerebro/embriología , Cerebro/metabolismo , Endosomas/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Péptidos y Proteínas de Señalización Intercelular/fisiología , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Noqueados , Células-Madre Neurales/metabolismo , Unión Proteica , ARN Interferente Pequeño/genética , Receptores CXCR/genética , Receptores CXCR/fisiología , Receptores CXCR4/genética , Receptores CXCR4/fisiología
17.
Cell Mol Neurobiol ; 33(1): 111-8, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22940879

RESUMEN

We previously demonstrated that in astrocytes, SDF-1/CXCL12 exclusively signals through CXCR7 despite the additional presence of the alternate SDF-1/CXCL12 receptor, CXCR4. In addition, we provided evidence that astrocytic CXCR7-signalling involves a G protein-dependent mechanism. This is insofar remarkable as in all other cell types studied to date, CXCR7 either acts as a scavenger chemokine receptor, a modulator of CXCR4, or a non-classical chemokine receptor, signalling through ß-arrestin. To begin to unravel the molecular framework impinging the selective function of CXCR7 on a given cell type, we have now analysed the role of G protein-coupled receptor kinases (Grks) in astrocytic CXCR7 signalling. We demonstrate that Grk2 mediates signalling of SDF-1/CXCL12-bound CXCR7 as suggested by the finding that SDF-1/CXCL12-induced activation of Erk1/2 and Akt is abrogated following RNAi-mediated inhibition of Grk2, but not of Grk3, Grk5, or Grk6. We further unravel that Grk2 additionally controls signalling of SDF-1/CXCL12-bound CXCR7 in astrocytes by mediating internalization and subsequent silencing of CXCR7. Finally, we demonstrate that Grk2 is likewise expressed by microglial cells and Schwann cells, cell types in which CXCR7 does not act as a classical chemokine receptor. In conclusion, our findings establish that Grk2 tightly controls CXCR7 signalling in astrocytes, but does not imprint the cell type-specific function of this chemokine receptor.


Asunto(s)
Astrocitos/metabolismo , Quinasa 2 del Receptor Acoplado a Proteína-G/fisiología , Receptores CXCR/metabolismo , Transducción de Señal/fisiología , Animales , Animales Recién Nacidos , Astrocitos/citología , Astrocitos/fisiología , Células Cultivadas , Microglía/química , Microglía/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores CXCR/fisiología , Células de Schwann/química , Células de Schwann/metabolismo
18.
J Inherit Metab Dis ; 36(3): 455-66, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23344887

RESUMEN

This study investigates glio-vascular interactions in human fetal brain at midgestation, specifically examining the expression and immunolocalization of the CXCL12/CXCR4/CXCR7 ligand-receptor axis and its possible role in the vascular patterning of the developing brain. At midgestation, the telencephalic vesicles are characterized by well developed radial glia cells (RGCs), the first differentiated astrocytes and a basic vascular network mainly built of radial vessels. RGCs have been recognized to contribute to cerebral cortex neuro-vascular architecture and have also been demonstrated to act as a significant source of neural cells (Rakic, Brain Res 33:471-476, 1971; Malatesta et al, Development 127:5253-5263, 2000). According to our hypothesis CXCL12, a potent migration and differentiation chemokine released by RGCs, may act as a linking factor coordinating neuroblast migration with vessel growth and patterning through the activation of different ligand/receptor axes. The obtained results support this hypothesis showing that together with CXCR4/CXCR7-reactive neuroblasts, which migrate in close association with CXCL12 RGCs, layer-specific subsets of CXCL12 RGCs and astrocytes specifically contact the microvessel wall. Moreover, the CXCL12/CXCR4/CXCR7 system appears to be directly involved in microvessel growth, its members being differentially expressed in angiogenically activated microvessels and vascular sprouts.


Asunto(s)
Encéfalo/irrigación sanguínea , Encéfalo/embriología , Comunicación Celular/fisiología , Quimiocina CXCL12/fisiología , Receptores CXCR4/fisiología , Receptores CXCR/fisiología , Vasos Sanguíneos/embriología , Vasos Sanguíneos/crecimiento & desarrollo , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/fisiología , Encéfalo/metabolismo , Encéfalo/patología , Quimiocina CXCL12/metabolismo , Feto/metabolismo , Feto/patología , Edad Gestacional , Humanos , Inmunohistoquímica , Ligandos , Neovascularización Fisiológica/fisiología , Neuroglía/metabolismo , Neuroglía/fisiología , Neuronas/metabolismo , Neuronas/fisiología , Receptores CXCR/metabolismo , Receptores CXCR4/metabolismo , Transducción de Señal/fisiología
19.
Ophthalmic Res ; 50(1): 6-12, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23549077

RESUMEN

BACKGROUND/AIMS: Stromal cell-derived factor-1 (SDF-1) has been shown to mediate a broad range of biological processes via CXCR4, once regarded as its only receptor. CXCR7 is a recently identified receptor for SDF-1. This study aimed to investigate whether the CXCR7/CXCR4/SDF-1 axis is involved in choroidal neovascularization (CNV) formation in an in vitro hypoxic model. METHODS: CXCR7 siRNA and/or CXCR4 siRNA was transfected into a hypoxic model of the choroid-retinal endothelial RF/6A cell line. CCK-8 analysis, transwell migration analysis, annexin V-FITC and propidium iodide staining, and Matrigel tube formation analysis were performed to investigate the role of CXCR4 and CXCR7 in SDF-1-induced proliferation, migration, survival and tube formation of RF/6A cells. RESULTS: CXCR4, but not CXCR7, mediates SDF-1-induced RF/6A cell migration and proliferation under hypoxic conditions, whereas CXCR7 was exclusively involved in RF/6A cell survival. In addition, CXCR7 and CXCR4 acted together to regulate RF/6A cell tube formation. CONCLUSION: The CXCR7/CXCR4/SDF-1 axis plays an important role in the formation of CNV, and may become a novel target for the treatment of CNV-associated diseases.


Asunto(s)
Quimiocina CXCL12/fisiología , Neovascularización Coroidal/patología , Células Endoteliales/fisiología , Receptores CXCR/fisiología , Análisis de Varianza , Animales , Western Blotting , Movimiento Celular/fisiología , Proliferación Celular , Supervivencia Celular/fisiología , Células Cultivadas , Coroides/citología , Modelos Animales de Enfermedad , Hipoxia/metabolismo , Macaca mulatta , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores CXCR4/fisiología , Retina/citología
20.
Proc Natl Acad Sci U S A ; 107(2): 628-32, 2010 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-20018651

RESUMEN

Ubiquitously expressed seven-transmembrane receptors (7TMRs) classically signal through heterotrimeric G proteins and are commonly referred to as G protein-coupled receptors. It is now recognized that 7TMRs also signal through beta-arrestins, which act as versatile adapters controlling receptor signaling, desensitization, and trafficking. Most endogenous receptors appear to signal in a balanced fashion using both beta-arrestin and G protein-mediated pathways. Some 7TMRs are thought to be nonsignaling "decoys" because of their inability to activate typical G protein signaling pathways; it has been proposed that these receptors act to scavenge ligands or function as coreceptors. Here we demonstrate that ligand binding to the decoy receptor CXCR7 does not result in activation of signaling pathways typical of G proteins but does activate MAP kinases through beta-arrestins in transiently transfected cells. Furthermore, we observe that vascular smooth muscle cells that endogenously express CXCR7 migrate to its ligand interferon-inducible T-cell alpha chemoattractant (ITAC), an effect that is significantly attenuated by treatment with either a CXCR7 antagonist or beta-arrestin depletion by siRNA. This example of an endogenous "beta-arrestin-biased" 7TMR that signals through beta-arrestin in the absence of G protein activation demonstrates that some 7TMRs encoded in the genome have evolved to signal through beta-arrestin exclusively and suggests that other receptors that are currently thought to be orphans or decoys may also signal through such nonclassical pathways.


Asunto(s)
Arrestinas/fisiología , Receptores CXCR/fisiología , Linfocitos T/fisiología , Animales , Línea Celular , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/fisiología , Humanos , Ratones , Músculo Liso Vascular/fisiología , Fosforilación , ARN Mensajero/genética , Ratas , Receptores CXCR/genética , Receptores de Quimiocina/genética , Transducción de Señal/fisiología , Linfocitos T/citología , beta-Arrestinas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA