Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 176
Filtrar
1.
Circ Res ; 124(8): 1253-1265, 2019 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-30739581

RESUMEN

RATIONALE: Regeneration of denuded or injured endothelium is an important component of vascular injury response. Cell-cell communication between endothelial cells and smooth muscle cells (SMCs) plays a critical role not only in vascular homeostasis but also in disease. We have previously demonstrated that PKCδ (protein kinase C-delta) regulates multiple components of vascular injury response including apoptosis of SMCs and production of chemokines, thus is an attractive candidate for a role in SMC-endothelial cells communication. OBJECTIVE: To test whether PKCδ-mediated paracrine functions of SMCs influence reendothelialization in rodent models of arterial injury. METHODS AND RESULTS: Femoral artery wire injury was performed in SMC-conditional Prkcd knockout mice, and carotid angioplasty was conducted in rats receiving transient Prkcd knockdown or overexpression. SMC-specific knockout of Prkcd impaired reendothelialization, reflected by a smaller Evans blue-excluding area in the knockout compared with the wild-type controls. A similar impediment to reendothelialization was observed in rats with SMC-specific knockdown of Prkcd. In contrast, SMC-specific gene transfer of Prkcd accelerated reendothelialization. In vitro, medium conditioned by AdPKCδ-infected SMCs increased endothelial wound closure without affecting their proliferation. A polymerase chain reaction-based array analysis identified Cxcl1 and Cxcl7 among others as PKCδ-mediated chemokines produced by SMCs. Mechanistically, we postulated that PKCδ regulates Cxcl7 expression through STAT3 (signal transducer and activator of transcription 3) as knockdown of STAT3 abolished Cxcl7 expression. The role of CXCL7 in SMC-endothelial cells communication was demonstrated by blocking CXCL7 or its receptor CXCR2, both significantly inhibited endothelial wound closure. Furthermore, insertion of a Cxcl7 cDNA in the lentiviral vector that carries a Prkcd shRNA overcame the adverse effects of Prkcd knockdown on reendothelialization. CONCLUSIONS: SMCs promote reendothelialization in a PKCδ-dependent paracrine mechanism, likely through CXCL7-mediated recruitment of endothelial cells from uninjured endothelium.


Asunto(s)
Células Endoteliales/metabolismo , Miocitos del Músculo Liso/metabolismo , Comunicación Paracrina/fisiología , Proteína Quinasa C-delta/metabolismo , Regeneración/genética , Lesiones del Sistema Vascular/metabolismo , Animales , Apoptosis/fisiología , Traumatismos de las Arterias Carótidas/genética , Traumatismos de las Arterias Carótidas/metabolismo , Quimiocina CXCL1/biosíntesis , Quimiocinas/biosíntesis , Quimiocinas CXC/antagonistas & inhibidores , Quimiocinas CXC/biosíntesis , Quimiocinas CXC/genética , Arteria Femoral/lesiones , Técnicas de Inactivación de Genes , Ratones , Ratones Transgénicos , Proteína Quinasa C-delta/genética , Receptores de Interleucina-8B/antagonistas & inhibidores , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Lesiones del Sistema Vascular/fisiopatología , Cicatrización de Heridas
2.
Cell Physiol Biochem ; 45(5): 1731-1746, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29495008

RESUMEN

BACKGROUND/AIMS: Breast cancer is a common cause of cancer mortality throughout the world. The cross-talk between cancer cells and interstitial cells exerts significant effects on neoplasia and tumor development and is modulated in part by chemokines. CXC is one of four chemokine families involved in mediating survival, angiogenesis, and immunosensitization by chemoattracting leukocytes, and it incentivizes tumor cell growth, invasion and metastasis in the tumor microenvironment. However, the differential expression profiles and prognostic values of these chemokines remains to be elucidated. METHODS: In this study, we compared transcriptional CXC chemokines and survival data of patients with breast carcinoma (BC) using the ONCOMINE dataset, Kaplan-Meier Plotter, TCGA and cBioPortal. RESULTS: We discovered increased mRNA levels for CXCL8/10/11/16/17, whereas mRNA expression of CXCL1/2/3/4/5/6/7/12/14 was lower in BC patients compared to non-tumor tissues. Kaplan-Meier plots revealed that high mRNA levels of CXCL1/2/3/4/5/6/7/12/14 correlate with relapse-free survival (RFS) in all types of BC patients. Conversely, high CXCL8/10/11 predicted worse RFS in BC patients. Significantly, high transcription levels of CXCL9/12/13/14 conferred an overall survival (OS) advantage in BC patients, while high levels of CXCL8 demonstrated shorter OS in all BC sufferers. CONCLUSIONS: Integrative bioinformatics analysis suggests that CXCL8/12/14 are potential suitable targets for precision therapy in BC patients compared to other CXC chemokines.


Asunto(s)
Quimiocinas CXC/metabolismo , Biología Computacional/métodos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Quimiocina CXCL9/genética , Quimiocina CXCL9/metabolismo , Quimiocinas CXC/antagonistas & inhibidores , Quimiocinas CXC/genética , Bases de Datos Factuales , Supervivencia sin Enfermedad , Femenino , Redes Reguladoras de Genes , Humanos , Interleucina-8/genética , Interleucina-8/metabolismo , Estimación de Kaplan-Meier , Pronóstico , ARN Mensajero/metabolismo , Microambiente Tumoral
3.
J Virol ; 90(7): 3366-84, 2016 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-26764004

RESUMEN

UNLABELLED: Previous studies in our laboratory have identified equine CXCL16 (EqCXCL16) to be a candidate molecule and possible cell entry receptor for equine arteritis virus (EAV). In horses, the CXCL16 gene is located on equine chromosome 11 (ECA11) and encodes a glycosylated, type I transmembrane protein with 247 amino acids. Stable transfection of HEK-293T cells with plasmid DNA carrying EqCXCL16 (HEK-EqCXCL16 cells) increased the proportion of the cell population permissive to EAV infection from <3% to almost 100%. The increase in permissiveness was blocked either by transfection of HEK-EqCXCL16 cells with small interfering RNAs (siRNAs) directed against EqCXCL16 or by pretreatment with guinea pig polyclonal antibody against EqCXCL16 protein (Gp anti-EqCXCL16 pAb). Furthermore, using a virus overlay protein-binding assay (VOPBA) in combination with far-Western blotting, gradient-purified EAV particles were shown to bind directly to the EqCXCL16 protein in vitro. The binding of biotinylated virulent EAV strain Bucyrus at 4°C was significantly higher in HEK-EqCXCL16 cells than nontransfected HEK-293T cells. Finally, the results demonstrated that EAV preferentially infects subpopulations of horse CD14(+) monocytes expressing EqCXCL16 and that infection of these cells is significantly reduced by pretreatment with Gp anti-EqCXCL16 pAb. The collective data from this study provide confirmatory evidence that the transmembrane form of EqCXCL16 likely plays a major role in EAV host cell entry processes, possibly acting as a primary receptor molecule for this virus. IMPORTANCE: Outbreaks of EVA can be a source of significant economic loss for the equine industry from high rates of abortion in pregnant mares, death in young foals, establishment of the carrier state in stallions, and trade restrictions imposed by various countries. Similar to other arteriviruses, EAV primarily targets cells of the monocyte/macrophage lineage, which, when infected, are believed to play a critical role in EVA pathogenesis. To this point, however, the host-specified molecules involved in EAV binding and entry into monocytes/macrophages have not been identified. Identification of the cellular receptors for EAV may provide insights to design antivirals and better prophylactic reagents. In this study, we have demonstrated that EqCXCL16 acts as an EAV entry receptor in EAV-susceptible cells, equine monocytes. These findings represent a significant advance in our understanding of the fundamental mechanisms associated with the entry of EAV into susceptible cells.


Asunto(s)
Quimiocinas CXC/fisiología , Equartevirus/fisiología , Especificidad del Huésped/genética , Receptores Virales/genética , Internalización del Virus , Secuencia de Aminoácidos , Animales , Anticuerpos Antivirales/inmunología , Infecciones por Arterivirus/virología , Secuencia de Bases , Línea Celular , Quimiocinas CXC/antagonistas & inhibidores , Quimiocinas CXC/genética , Cricetinae , Equartevirus/genética , Cobayas , Células HEK293 , Enfermedades de los Caballos/virología , Caballos , Humanos , Interferencia de ARN , ARN Interferente Pequeño/genética , Conejos , Receptores Virales/metabolismo , Análisis de Secuencia de ADN , Acoplamiento Viral
4.
Pulm Pharmacol Ther ; 31: 55-62, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25681618

RESUMEN

Inhalation of agricultural occupational dusts from swine confinement facilities can result in lung inflammation. The innate immune response to organic barn dusts results in production of a number of pro-inflammatory factors in the lungs of barn workers such as cytokines, chemokines, and an influx of neutrophils. Many of these inflammatory factors are influenced by the chemokine CXCL8/IL-8 (KC or MIP-2 in mice). Previously, we have demonstrated that an endotoxin-independent component of swine barn dust extract (SBE) elevates lung chemokines in a protein kinase C (PKC)-dependent manner resulting in the significant formation of lung inflammatory cell infiltrates in a mouse model of SBE injury. In this study we test the ability of a CXCR1/CXCR2 antagonist, CXCL8(3-74)K11R/G31P (G31P) to block many of the features of lung-inflammation in response to challenge with SBE in an established mouse exposure system. Injection of G31P concurrent with SBE nasal instillation over a course of 3 weeks significantly reduced neutrophil accumulation in the lungs of barn dust exposed animals compared to those given SBE alone. There was a similar reduction in pro-inflammatory cytokines and chemokines IL-6, KC, and MIP-2 in SBE plus G31P-treated mice. In addition to excreted products, the receptors ICAM-1, CXCR1, and CXCR2, which all were elevated with SBE exposure, were also decreased with G31P treatment. SBE activation of PKCα and PKCε was reduced as well with G31P treatment. Thus, G31P was found to be highly effective at reducing several features of lung inflammation in mice exposed to barn dust extracts.


Asunto(s)
Quimiocinas CXC/antagonistas & inhibidores , Inflamación/fisiopatología , Interleucina-8/farmacología , Fragmentos de Péptidos/farmacología , Crianza de Animales Domésticos , Animales , Líquido del Lavado Bronquioalveolar/inmunología , Quimiocinas/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Polvo , Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Ratones , Enfermedades Profesionales/fisiopatología , Proteína Quinasa C/metabolismo , Porcinos
5.
Radiother Oncol ; 194: 110194, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38447871

RESUMEN

High precision, image-guided radiotherapy (RT) has increased the therapeutic ratio, enabling higher tumor and lower normal tissue doses, leading to improved patient outcomes. Nevertheless, some patients remain at risk of developing serious side effects.In many clinical situations, the radiation tolerance of normal tissues close to the target volume limits the dose that can safely be delivered and thus the potential for tumor control and cure. This is particularly so in patients being re-treated for tumor progression or a second primary tumor within a previous irradiated volume, scenarios that are becoming more frequent in clinical practice.Various normal tissue 'radioprotective' drugs with the potential to reduce side effects have been studied previously. Unfortunately, most have failed to impact clinical practice because of lack of therapeutic efficacy, concern about concurrent tumor protection or excessive drug-related toxicity. This review highlights the evidence indicating that targeting the CXCL12/CXCR4 pathway can mitigate acute and late RT-induced injury and reduce treatment side effects in a manner that overcomes these previous translational challenges. Pre-clinical studies involving a broad range of normal tissues commonly affected in clinical practice, including skin, lung, the gastrointestinal tract and brain, have shown that CXCL12 signalling is upregulated by RT and attracts CXCR4-expressing inflammatory cells that exacerbate acute tissue injury and late fibrosis. These studies also provide convincing evidence that inhibition of CXCL12/CXCR4 signalling during or after RT can reduce or prevent RT side effects, warranting further evaluation in clinical studies. Greater dialogue with the pharmaceutical industry is needed to prioritize the development and availability of CXCL12/CXCR4 inhibitors for future RT studies.


Asunto(s)
Quimiocina CXCL12 , Neoplasias , Traumatismos por Radiación , Protectores contra Radiación , Transducción de Señal , Animales , Humanos , Quimiocina CXCL12/metabolismo , Neoplasias/radioterapia , Traumatismos por Radiación/prevención & control , Tolerancia a Radiación/efectos de los fármacos , Protectores contra Radiación/farmacología , Protectores contra Radiación/uso terapéutico , Radioterapia Guiada por Imagen/métodos , Receptores CXCR4/antagonistas & inhibidores , Receptores CXCR4/metabolismo , Transducción de Señal/efectos de los fármacos , Quimiocinas CXC/antagonistas & inhibidores
6.
Endocr J ; 60(2): 137-47, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23037942

RESUMEN

Muscle contractile activity functions as a potent stimulus for acute interleukin (IL)-6 expression in working skeletal muscles. Recently, we established an "in vitro contraction model" using highly-developed contractile C2C12 myotubes by applying electric pulse stimulation (EPS). Herein, we characterize the effects of EPS-evoked contraction on IL-6 expression in contractile C2C12 myotubes. Both secretion and mRNA expression of IL-6 were significantly up-regulated by EPS in a frequency-dependent manner in contracting myotubes during a 24-h period, and the response was blunted by cyclosporine A, a calcineurin inhibitor. Longer time (~12h) was required for the induction of IL-6 after the initiation of EPS as compared to that of other contraction-inducible CXC chemokines such as CXCL1/KC, which were induced in less than 3 hours. Furthermore, these acute inducible CXC chemokines exhibited no autocrine effect on IL-6 expression. Importantly, contraction-dependent IL-6 up-regulation was markedly suppressed in the presence of high levels of glucose along with increased glycogen accumulations. Experimental manipulation of intracellular glycogen contents by modulating available glucose or pyruvate during a certain EPS period further established the suppressive effect of glycogen accumulations on contraction-induced IL-6 up-regulation, which appeared to be independent of calcineurin activity. We also document that EPS-evoked contractile activity improved insulin-responsiveness in terms of intracellular glycogen accumulations. Taken together, these data provide important insights into the regulation of IL-6 expression in response to contractile activity of muscle cells, which is difficult to examine using in vivo experimental techniques. Our present results thus expand the usefulness of our "in vitro contraction model".


Asunto(s)
Glucógeno/metabolismo , Resistencia a la Insulina , Interleucina-6/metabolismo , Contracción Muscular , Fibras Musculares Esqueléticas/metabolismo , Regulación hacia Arriba , Animales , Calcineurina/metabolismo , Inhibidores de la Calcineurina , Señalización del Calcio/efectos de los fármacos , Línea Celular , Quimiocinas CXC/antagonistas & inhibidores , Quimiocinas CXC/metabolismo , Estimulación Eléctrica , Glucosa/metabolismo , Hiperglucemia/metabolismo , Hipoglucemiantes/metabolismo , Hipoglucemiantes/farmacología , Inmunosupresores/farmacología , Insulina/metabolismo , Insulina/farmacología , Interleucina-6/genética , Cinética , Ratones , Contracción Muscular/efectos de los fármacos , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/inmunología , ARN Mensajero/metabolismo , Regulación hacia Arriba/efectos de los fármacos
7.
Am J Physiol Cell Physiol ; 302(4): C644-51, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22173866

RESUMEN

Suicidal death of erythrocytes, or eryptosis, is characterized by cell shrinkage and cell membrane scrambling leading to phosphatidylserine exposure at the cell surface. Eryptosis is triggered by increase of cytosolic Ca2+ activity, which may result from treatment with the Ca2+ ionophore ionomycin or from energy depletion by removal of glucose. The present study tested the hypothesis that phosphatidylserine exposure at the erythrocyte surface fosters adherence to endothelial cells of the vascular wall under flow conditions at arterial shear rates and that binding of eryptotic cells to endothelial cells is mediated by the transmembrane CXC chemokine ligand 16 (CXCL16). To this end, human erythrocytes were exposed to energy depletion (for 48 h) or treated with the Ca2+ ionophore ionomycin (1 µM for 30 min). Phosphatidylserine exposure was quantified utilizing annexin-V binding, cell volume was estimated from forward scatter in FACS analysis, and erythrocyte adhesion to human vascular endothelial cells (HUVEC) was determined in a flow chamber model. As a result, both, ionomycin and glucose depletion, triggered eryptosis and enhanced the percentage of erythrocytes adhering to HUVEC under flow conditions at arterial shear rates. The adhesion was significantly blunted in the presence of erythrocyte phosphatidylserine-coating annexin-V (5 µl/ml), of a neutralizing antibody against endothelial CXCL16 (4 µg/ml), and following silencing of endothelial CXCL16 with small interfering RNA. The present observations demonstrate that eryptotic erythrocytes adhere to endothelial cells of the vascular wall in part by interaction of phosphatidylserine exposed at the erythrocyte surface with endothelial CXCL16.


Asunto(s)
Calcio/metabolismo , Membrana Celular/metabolismo , Quimiocinas CXC/metabolismo , Endotelio Vascular/metabolismo , Eritrocitos/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Fosfatidilserinas/metabolismo , Receptores Depuradores/metabolismo , Anexina A5/metabolismo , Anticuerpos/metabolismo , Ionóforos de Calcio/metabolismo , Adhesión Celular , Muerte Celular/fisiología , Tamaño de la Célula , Quimiocina CXCL16 , Quimiocinas CXC/antagonistas & inhibidores , Quimiocinas CXC/genética , Citosol/metabolismo , Endotelio Vascular/citología , Recuento de Eritrocitos , Eritrocitos/citología , Silenciador del Gen , Glucosa/deficiencia , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Ionomicina/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptores Depuradores/antagonistas & inhibidores , Receptores Depuradores/genética , Reología
8.
Am J Physiol Cell Physiol ; 303(9): C991-9, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22954799

RESUMEN

Eryptosis, the suicidal erythrocyte death, leads to cell shrinkage and cell membrane scrambling with phosphatidylserine exposure at the cell surface. Eryptotic erythrocytes adhere to the vascular wall by binding of phosphatidylserine to the CXC chemokine ligand 16 (CXCL16). Stimulators of eryptosis include increased cytosolic Ca(2+) activity, energy depletion, and activation of ceramide-producing sphingomyelinase. The present study explored whether sphingomyelinase triggers erythrocyte adhesion to endothelial cells. To this end, human erythrocytes were exposed for 6 h to bacterial sphingomyelinase (1-10 mU/ml) and phosphatidylserine exposure was estimated from fluorescent annexin-V-binding, cell volume from forward scatter in FACS-analysis, erythrocyte adhesion to human umbilical vein endothelial cells (HUVEC) from trapping of labeled erythrocytes in a flow chamber under flow conditions at arterial shear rates, and CXCL16 protein abundance utilizing Western blotting and FACS analysis of fluorescent antibody binding. As a result, sphingomyelinase (≥1 mU/ml) triggered cell shrinkage, phosphatidylserine exposure and erythrocyte adhesion to HUVEC, effects blunted by Ca(2+) removal. Adhesion was significantly blunted by phosphatidylserine-coating annexin-V (5 µl/ml), following addition of neutralizing antibodies against endothelial CXCL16 (4 µg/ml) and following silencing of the CXCL16 gene with small interfering RNA. Pretreatment of HUVEC with sphingomyelinase upregulated CXCL16 protein abundance. Six hours pretreatment of HUVEC with sphingomyelinase (10 mU/ml) or C6-ceramide (50 µM) augmented erythrocyte adhesion following a 30-min treatment with Ca(2+) ionophore ionomycin (1 µM) or following energy depletion by 48-h glucose removal. Thus exposure to sphingomyelinase or C6-ceramide triggers eryptosis followed by phosphatidylserine- and CXCL16-sensitive adhesion of eryptotic erythrocytes to HUVEC.


Asunto(s)
Apoptosis/efectos de los fármacos , Eritrocitos/efectos de los fármacos , Esfingomielina Fosfodiesterasa/farmacología , Anexina A5/fisiología , Anticuerpos Neutralizantes/farmacología , Apoptosis/fisiología , Calcio/farmacología , Ionóforos de Calcio/farmacología , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Tamaño de la Célula , Células Cultivadas , Ceramidas/farmacología , Quimiocina CXCL16 , Quimiocinas CXC/antagonistas & inhibidores , Quimiocinas CXC/genética , Quimiocinas CXC/fisiología , Eritrocitos/fisiología , Silenciador del Gen , Glucosa/farmacología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Ionomicina/farmacología , Fosfatidilserinas/fisiología , Receptores Depuradores/antagonistas & inhibidores , Receptores Depuradores/genética , Receptores Depuradores/fisiología
10.
J Immunol ; 182(1): 507-14, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19109182

RESUMEN

The skin is constantly exposed to commensal microflora and pathogenic microbes. The stratum corneum of the outermost skin layer employs distinct tools such as harsh growth conditions and numerous antimicrobial peptides (AMPs) to discriminate between beneficial cutaneous microflora and harmful bacteria. How the skin deals with microbes that have gained access to the live part of the skin as a result of microinjuries is ill defined. In this study, we report that the chemokine CXCL14 is a broad-spectrum AMP with killing activity for cutaneous gram-positive bacteria and Candida albicans as well as the gram-negative enterobacterium Escherichia coli. Based on two separate bacteria-killing assays, CXCL14 compares favorably with other tested AMPs, including human beta-defensin and the chemokine CCL20. Increased salt concentrations and skin-typical pH conditions did not abrogate its AMP function. This novel AMP is highly abundant in the epidermis and dermis of healthy human skin but is down-modulated under conditions of inflammation and disease. We propose that CXCL14 fights bacteria at the earliest stage of infection, well before the establishment of inflammation, and thus fulfills a unique role in antimicrobial immunity.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/fisiología , Quimiocinas CXC/fisiología , Enfermedades Cutáneas Infecciosas/inmunología , Enfermedades Cutáneas Infecciosas/terapia , Animales , Péptidos Catiónicos Antimicrobianos/aislamiento & purificación , Candidiasis/inmunología , Candidiasis/microbiología , Candidiasis/terapia , Línea Celular Transformada , Línea Celular Tumoral , Sistema Libre de Células/inmunología , Sistema Libre de Células/microbiología , Quimiocinas CXC/antagonistas & inhibidores , Quimiocinas CXC/biosíntesis , Quimiocinas CXC/genética , Relación Dosis-Respuesta Inmunológica , Infecciones por Bacterias Gramnegativas/inmunología , Infecciones por Bacterias Gramnegativas/microbiología , Infecciones por Bacterias Gramnegativas/terapia , Infecciones por Bacterias Grampositivas/inmunología , Infecciones por Bacterias Grampositivas/microbiología , Infecciones por Bacterias Grampositivas/terapia , Humanos , Mediadores de Inflamación/aislamiento & purificación , Mediadores de Inflamación/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Enfermedades Cutáneas Infecciosas/microbiología , Enfermedades Cutáneas Infecciosas/patología
11.
Int Immunopharmacol ; 97: 107681, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33932697

RESUMEN

Cancer is a leading cause of death worldwide and imposes a substantial financial burden. Therefore, it is essential to develop cost-effective approaches to inhibit tumor growth and development. The imbalance of cytokines and chemokines play an important role among different mechanisms involved in cancer development. One of the strongly conserved chemokines that is constitutively expressed in skin epithelia is the chemokine CXCL14. As a member of the CXC subfamily of chemokines, CXCL14 is responsible for the infiltration of immune cells, maturation of dendritic cells, upregulation of major histocompatibility complex (MHC)-I expression, and cell mobilization. Moreover, dysregulation of CXCL14 in several cancers has been identified by several studies. Depending on the type or origin of the tumor and components of the tumor microenvironment, CXCL14 plays a conflicting role in cancer. Although fibroblast-derived CXCL14 has a tumor-supportive role, epithelial-derived CXCL14 mainly inhibits tumor progression. Hence, this review will elucidate what is known on the mechanisms of CXCL14 and its therapeutic approaches in tumor treatment. CXCL14 is a promising approach for cancer immunotherapy.


Asunto(s)
Antineoplásicos/farmacología , Biomarcadores de Tumor/metabolismo , Quimiocinas CXC/metabolismo , Neoplasias/inmunología , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/agonistas , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/antagonistas & inhibidores , Quimiocinas CXC/agonistas , Quimiocinas CXC/análisis , Quimiocinas CXC/antagonistas & inhibidores , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/inmunología , Fibroblastos/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Neoplasias/diagnóstico , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología , Regulación hacia Arriba
12.
J Exp Med ; 192(3): 313-24, 2000 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-10934220

RESUMEN

We report that stromal cell-derived factor (SDF)-1 has the remarkable capacity to induce sustained signaling through CXC chemokine receptor 4 (CXCR4). In contrast to other chemokines, such as monocyte chemotactic protein 1 (CC chemokine receptor 2 [CCR2]), macrophage inflammatory protein 1beta (CCR5), liver and activation-regulated chemokine (LARC [CCR6]), Epstein-Barr virus-induced molecule 1 ligand chemokine (ELC [CCR7]), and IP10 (CXCR3), SDF-1 stimulates the prolonged activation of protein kinase B and extracellular signal-regulated kinase (ERK)-2. Activation of protein kinase B is reversed by displacement of SDF-1 from CXCR4 or inhibition of phosphatidylinositol 3-kinase. Although increasing concentrations of SDF-1 enhance CXCR4 internalization, kinase activation is prolonged. In addition, restimulation yields >60% of initial protein kinase B activity, indicating that the remaining receptors are not desensitized. Furthermore, activation is prolonged by inhibiting SDF-1 degradation. The sustained activation of cell survival and mitogenic pathways may account for the unique role of SDF-1 and CXCR4 in embryogenesis and lymphopoiesis.


Asunto(s)
Quimiocinas CXC/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas/metabolismo , Receptores CXCR4/metabolismo , Transducción de Señal , Linfocitos T/metabolismo , Androstadienos/farmacología , Animales , Células Cultivadas , Quimiocina CXCL12 , Quimiocinas CXC/antagonistas & inhibidores , Quimiocinas CXC/farmacología , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Humanos , Ratones , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt , Linfocitos T/citología , Factores de Tiempo , Wortmanina
13.
Biochem Biophys Res Commun ; 391(1): 609-14, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19932682

RESUMEN

CXCL16 is a chemokine that is expressed in both transmembrane and secreted isoforms. Both variants have been implicated in atherosclerosis. Increased CXCL16 expression on the surface of human aortic smooth muscle cells induced by interferon gamma (IFNgamma) signaling results in enhanced oxidized low density lipoprotein uptake and enhanced recruitment of pro-inflammatory cells. Docosahexaenoic acid (DHA), an omega-3 fatty acid, is known to inhibit IFNgamma signaling in inflammatory cells. Therefore, we have investigated the effects of DHA treatment on the ability of IFNgamma to induce CXCL16 expression in human aortic smooth muscle cells. We observed that DHA treatment significantly reduced IFNgamma-induced CXCL16 expression. As a result, the pro-atherosclerotic functions of CXCL16 were also inhibited. Furthermore, IFNgamma-induced STAT1 phosphorylation was inhibited by DHA, suggesting a potential mechanism. In conclusion, our data suggest inhibition of IFNgamma signaling as one of the mechanisms behind the beneficial effects of DHA during atherosclerosis. These findings may prove to be important in other disease fields that identify IFNgamma as a regulator.


Asunto(s)
Aorta/efectos de los fármacos , Quimiocinas CXC/antagonistas & inhibidores , Ácidos Docosahexaenoicos/farmacología , Interferón gamma/antagonistas & inhibidores , Miocitos del Músculo Liso/efectos de los fármacos , Receptores Depuradores/antagonistas & inhibidores , Aorta/metabolismo , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/metabolismo , Células Cultivadas , Quimiocina CXCL16 , Quimiocinas CXC/biosíntesis , Quimiotaxis/efectos de los fármacos , Ácidos Docosahexaenoicos/uso terapéutico , Regulación hacia Abajo , Humanos , Interferón gamma/metabolismo , Interferón gamma/farmacología , Lipoproteínas LDL/metabolismo , Miocitos del Músculo Liso/metabolismo , Fosforilación , Receptores Depuradores/biosíntesis , Factor de Transcripción STAT1/metabolismo
14.
Circ Res ; 102(2): 218-25, 2008 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17991878

RESUMEN

Obese individuals often have low plasma adiponectin and concomitant chronic inflammation with a predisposition to metabolic and cardiovascular diseases. The present study reports a novel antiinflammatory action of adiponectin in human monocyte-derived macrophages (MPhi) suppressing T-lymphocyte accumulation in atherogenesis. RNA profiling of lipopolysaccharide-stimulated human MPhi identified CXC chemokine ligands (CXCLs), such as IP-10 (interferon [IFN]-inducible protein 10) (CXCL10), I-TAC (IFN-inducible T-cell alpha chemoattractant) (CXCL11), and Mig (monokine induced by IFN-gamma) (CXCL9), T-lymphocyte chemoattractants associated with atherogenesis, among the top 14 transcripts suppressed by adiponectin. Real-time quantitative RT-PCR and ELISA verified that adiponectin inhibited expression of these chemokines at both the mRNA and protein levels in a concentration-dependent manner. Adiponectin reduced the release by lipopolysaccharide-stimulated MPhi of chemoattractant activity for CXC chemokine receptor 3-transfected (receptor for IP-10, Mig, and I-TAC) lymphocytes. Adiponectin decreased lipopolysaccharide-inducible IP-10 promoter activity in promoter-transfected THP-1 MPhi but did not change IP-10 mRNA stability. In lipopolysaccharide-stimulated MPhi, reduction of IFN-beta by adiponectin preceded inhibition of IP-10 mRNA expression. Immunoblot and chromatin immunoprecipitation analyses demonstrated that adiponectin attenuated activation of the transcription factor IFN regulatory factor 3, involved in the MyD88-independent pathway of Toll-like receptor 4 signaling, and subsequent IFN regulatory factor 3 binding to IFN-beta promoter. In vivo studies further demonstrated that apolipoprotein E/adiponectin double-deficient (apoE-/-APN-/-) mice had increased plasma IP-10 levels, accelerated T-lymphocyte accumulation in atheromata, and augmented atherogenesis compared with apoE single-deficient (apoE-/-APN+/+) mice. This study establishes that low levels of adiponectin associated with obesity, the metabolic syndrome, and diabetes favor T-lymphocyte recruitment and contribute to adaptive immune response during atherogenesis.


Asunto(s)
Adiponectina/farmacología , Arteriosclerosis/inmunología , Quimiocinas CXC/antagonistas & inhibidores , Quimiotaxis de Leucocito/efectos de los fármacos , Receptores CXCR3/metabolismo , Linfocitos T/efectos de los fármacos , Células Cultivadas , Diabetes Mellitus , Relación Dosis-Respuesta a Droga , Humanos , Inmunidad/efectos de los fármacos , Macrófagos , Síndrome Metabólico , Obesidad , Receptores CXCR3/genética , Transfección
15.
J Surg Res ; 162(2): 264-73, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19691980

RESUMEN

BACKGROUND: Neutrophil sequestration plays an important role in mediating local and remote organ injury induced by ischemia and reperfusion (I/R). The Glu-Leu-Arg (ELR)-CXC subfamily of chemokines, all CXCR1 or CXCR2 ligands, are primary agonists for such neutrophil recruitment. Herein, we assessed the effects of a combined CXCR1/CXCR2 antagonist, CXCL8((3-72))K11R/G31P (G31P), on neutrophilic local (gut) and distant organ injury and outcomes after superior mesenteric artery I/R in rats. METHODS: Male Sprague-Dawley rats (n=6-10) were subjected to either sham treatment or superior mesenteric artery ischemia for 1h; all animals received either saline or G31P (500 mug/kg, s.c.) and were euthanized for assessment after either 2 or 5h of arterial reperfusion. Survival and gut pathology, and pulmonary neutrophils were assessed directly, while bronchoalveolar lavage (BAL) fluid total protein levels and red blood cell (RBC) numbers were determined by protein assay and direct counting. Expression of inflammatory mediators in the lung and jejunum was measured by quantitative RT-PCR, colorimetric or gel zymography assays. RESULTS: Sham treatment animals suffered no discernible gut or pulmonary pathology. At 2 and 5h after reperfusion, the survival levels of the saline-treated I/R injury animals were 80% and 50%, respectively, while all G31P-treated animals survived. I/R injury led to substantial villous pathology within the jejunum, and G31P significantly reduced these pathology scores as well as neutrophil infiltration of the jejunal lamina propria and lung parenchyma, and vascular leakage into the airways (BAL protein). The tissue injury increased expression of myeloperoxidase and matrix metalloproteinase (MMP)-2 and MMP-9 in the gut tissues, but G31P treatment did not significantly affect this response. Intestinal I/R increased expression of IL-1, IL-6, GRO, and MIP-2 in the ischemic jejunum and the lung tissues, but here too G31P treatment had no palliative effects on these responses. CONCLUSION: These results suggest that full-spectrum ELR-CXC chemokine antagonism has significant protective effects against I/R-induced local and remote organ injury.


Asunto(s)
Quimiocinas CXC/antagonistas & inhibidores , Intestinos/irrigación sanguínea , Isquemia/patología , Isquemia/prevención & control , Arteria Mesentérica Superior/patología , Receptores de Superficie Celular/antagonistas & inhibidores , Daño por Reperfusión/prevención & control , Animales , Líquido del Lavado Bronquioalveolar , Quimiocinas CXC/genética , Interleucina-1beta/genética , Interleucina-6/genética , Yeyuno/enzimología , Yeyuno/fisiopatología , Pulmón/enzimología , Pulmón/fisiopatología , Masculino , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Superficie Celular/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
Nature ; 425(6955): 307-11, 2003 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-13679920

RESUMEN

Organ-specific metastasis is governed, in part, by interactions between chemokine receptors on cancer cells and matching chemokines in target organs. For example, malignant breast cancer cells express the chemokine receptor CXCR4 and commonly metastasize to organs that are an abundant source of the CXCR4-specific ligand stromal cell-derived factor-1alpha (ref. 1). It is still uncertain how an evolving tumour cell is reprogrammed to express CXCR4, thus implementing the tendency to metastasize to specific organs. Here we show that the von Hippel-Lindau tumour suppressor protein pVHL negatively regulates CXCR4 expression owing to its capacity to target hypoxia-inducible factor (HIF) for degradation under normoxic conditions. This process is suppressed under hypoxic conditions, resulting in HIF-dependent CXCR4 activation. An analysis of clear cell renal carcinoma that manifests mutation of the VHL gene in most cases revealed an association of strong CXCR4 expression with poor tumour-specific survival. These results suggest a mechanism for CXCR4 activation during tumour cell evolution and imply that VHL inactivation acquired by incipient tumour cells early in tumorigenesis confers not only a selective survival advantage but also the tendency to home to selected organs.


Asunto(s)
Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Ligasas/metabolismo , Receptores CXCR4/metabolismo , Factores de Transcripción , Proteínas Supresoras de Tumor , Ubiquitina-Proteína Ligasas , Adenocarcinoma de Células Claras/genética , Adenocarcinoma de Células Claras/metabolismo , Adenocarcinoma de Células Claras/patología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular , Quimiocina CXCL12 , Quimiocinas CXC/antagonistas & inhibidores , Quimiocinas CXC/metabolismo , Quimiotaxis , Clonación Molecular , Proteínas de Unión al ADN/metabolismo , Humanos , Hipoxia/metabolismo , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Metástasis de la Neoplasia , Proteínas Nucleares/metabolismo , Especificidad de Órganos , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores CXCR4/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau
17.
J Clin Invest ; 116(10): 2593-6, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17016556

RESUMEN

HSCs differ during ontogeny in some important parameters, including anatomic site of residence and cell cycling characteristics. In this issue of the JCI, Bowie et al. show that postnatal HSCs as well as fetal liver HSCs in mice are active in the cell cycle at much higher rates than that of adult HSCs; however, this increased frequency of cycling abruptly ceases 4 weeks after birth (see the related article beginning on page 2808). The cycling postnatal HSCs expressed high levels of CXC chemokine ligand 12 (CXCL12, also known as stromal cell-derived factor 1 [SDF-1]), a chemokine previously implicated in stem cell trafficking to the marrow cavity and shown to be expressed by cells within the hematopoietic microenvironment. These cells also possessed an engraftment defect impeding reconstitution in irradiated recipient mice, which was reversible by pretransplant administration of antagonists of the CXCL12 receptor, CXCR4. Such agents are currently clinically available, suggesting that this approach could be used to improve stem cell transplantation and engraftment.


Asunto(s)
Células Madre Hematopoyéticas/citología , Sistema Hematopoyético/citología , Adulto , Animales , Células de la Médula Ósea/citología , Ciclo Celular/fisiología , Proliferación Celular , Quimiocina CXCL12 , Quimiocinas CXC/antagonistas & inhibidores , Quimiocinas CXC/genética , Niño , Hematopoyesis/fisiología , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Sistema Hematopoyético/embriología , Sistema Hematopoyético/crecimiento & desarrollo , Humanos , Hígado/citología , Hígado/embriología , Ratones , Modelos Biológicos
18.
J Clin Invest ; 116(10): 2808-16, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17016561

RESUMEN

The regulation of HSC proliferation and engraftment of the BM is an important but poorly understood process, particularly during ontogeny. Here we show that in mice, all HSCs are cycling until 3 weeks after birth. Then, within 1 week, most became quiescent. Prior to 4 weeks of age, the proliferating HSCs with long-term multilineage repopulating activity displayed an engraftment defect when transiting S/G2/M. During these cell cycle phases, their expression of CXC chemokine ligand 12 (CXCL12; also referred to as stromal cell-derived factor 1 [SDF-1]) transiently increased. The defective engrafting activity of HSCs in S/G2/M was reversed when cells were allowed to progress into G1 prior to injection or when the hosts (but not the cells) were pretreated with a CXCL12 antagonist. Interestingly, the enhancing effect of CXCL12 antagonist pretreatment was exclusive to transplants of long-term multilineage repopulating HSCs in S/G2/M. These results demonstrate what we believe to be a new HSC regulatory checkpoint during development. They also suggest an ability of HSCs to express CXCL12 in a fashion that changes with cell cycle progression and is associated with a defective engraftment that can be overcome by in vivo administration of a CXCL12 antagonist.


Asunto(s)
Proliferación Celular , Células Madre Hematopoyéticas/citología , Sistema Hematopoyético/citología , Animales , Animales Recién Nacidos , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Ciclo Celular/efectos de los fármacos , Ciclo Celular/fisiología , Quimiocina CXCL12 , Quimiocinas CXC/antagonistas & inhibidores , Quimiocinas CXC/genética , Fluorouracilo/farmacología , Expresión Génica/genética , Supervivencia de Injerto/efectos de los fármacos , Supervivencia de Injerto/fisiología , Hematopoyesis/efectos de los fármacos , Hematopoyesis/fisiología , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Sistema Hematopoyético/embriología , Sistema Hematopoyético/crecimiento & desarrollo , Receptores de Hialuranos/genética , Integrina alfa4/genética , Hígado/citología , Hígado/efectos de los fármacos , Hígado/embriología , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-kit/genética , Receptores CXCR4/genética , Timidina/farmacología , Molécula 1 de Adhesión Celular Vascular/genética
19.
Eur Rev Med Pharmacol Sci ; 23(9): 3821-3828, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-31115009

RESUMEN

OBJECTIVE: Emerging evidence has indicated that microRNAs (miRNAs) play crucial roles in regulating cancer carcinogenesis; however, its role in oral squamous cell carcinoma (OSCC) remains largely unknown. Our work was aimed to investigate the role of miR-4513 in regulating OSCC cells behaviors. MATERIALS AND METHODS: MiR-4513 expression in OSCC cells was analyzed by quantitative Real Time-Polymerase Chain Reaction (qRT-PCR). Cell proliferation, migration, invasion, and apoptosis were analyzed by the cell counting kit-8 (CCK-8) assay, wound-healing assay, transwell invasion assay, and flow cytometry, respectively. The connections of miR-4513 and CXC ligand 17 (CXCL17) were analyzed by luciferase reporter assay and Western blot assay. RESULTS: MiR-4513 expression was found elevated in the OSCC cell lines. The downregulation of miR-4513 expression inhibits cell proliferation, migration, invasion and, at the same time, promotes apoptosis. Furthermore, we validated CXCL17 as a direct target of miR-4513. Knocking down the expression of CXCL17, inhibited the effects of miR-4513 on OSCC cell behaviors. CONCLUSIONS: Our results suggested the oncogenic role of miR-4513 in OSCC, and therefore it might be used as a target for the OSCC treatment.


Asunto(s)
Apoptosis , Proliferación Celular , Quimiocinas CXC/metabolismo , MicroARNs/metabolismo , Antagomirs/metabolismo , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Movimiento Celular , Quimiocinas CXC/antagonistas & inhibidores , Quimiocinas CXC/genética , Humanos , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Neoplasias de la Boca/genética , Neoplasias de la Boca/patología , Mutagénesis , Interferencia de ARN , ARN Interferente Pequeño/metabolismo
20.
Biochem Biophys Res Commun ; 370(2): 311-6, 2008 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-18373975

RESUMEN

In this study, we analyzed the regulation and functional role of CXCL16 in human mesangial cells (hMCs). We can show, that CXCL16 is constitutively expressed in hMCs and is further up-regulated by cytokine mix (IFNgamma, TNFalpha, and IL1beta). The constitutive release of CXCL16 from hMCs was rapidly induced by the stimulation with cytokines. We identified ADAM10 and ADAM17 as being responsible for the cytokine-induced shedding of CXCL16. Notably, targeting ADAM10 and ADAM17 in hMCs decreased the chemotaxis of T-Jurkat cells, whereas the inhibition of CXCL16 had no significant influence. This suggests that both proteases are important players in the recruitment of immune cells into the glomerulus, but other substrates than CXCL16 are involved in this process. Finally, we could show that the inhibition of CXCL16, ADAM10, and ADAM17 led to a strong reduction of cell proliferation and migration of hMCs. This finding could be important to develop novel diagnostic and therapeutic strategies to treat mesangial proliferative kidney diseases.


Asunto(s)
Proteínas ADAM/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Quimiocinas CXC/metabolismo , Citocinas/metabolismo , Proteínas de la Membrana/metabolismo , Células Mesangiales/fisiología , Receptores Depuradores/metabolismo , Proteínas ADAM/antagonistas & inhibidores , Proteínas ADAM/genética , Proteína ADAM10 , Proteína ADAM17 , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/genética , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CXCL16 , Quimiocinas CXC/antagonistas & inhibidores , Quimiocinas CXC/genética , Quimiotaxis/efectos de los fármacos , Citocinas/farmacología , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Células Jurkat , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Células Mesangiales/efectos de los fármacos , Células Mesangiales/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Receptores Depuradores/antagonistas & inhibidores , Receptores Depuradores/genética , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA