Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros











Intervalo de año de publicación
1.
J Neuroinflammation ; 20(1): 258, 2023 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-37946211

RESUMEN

BACKGROUND: Although peripheral nerves have an intrinsic self-repair capacity following damage, functional recovery is limited in patients. It is a well-established fact that macrophages accumulate at the site of injury. Numerous studies indicate that the phenotypic shift from M1 macrophage to M2 macrophage plays a crucial role in the process of axon regeneration. This polarity change is observed exclusively in peripheral macrophages but not in microglia and CNS macrophages. However, the molecular basis of axonal regeneration by M2 macrophage is not yet fully understood. Herein, we aimed to identify the M2 macrophage-derived axon regeneration factor. METHODS: We established a peripheral nerve injury model by transection of the inferior alveolar nerve (IANX) in Sprague-Dawley rats. Transcriptome analysis was performed on the injured nerve. Recovery from sensory deficits in the mandibular region and histological reconnection of IAN after IANX were assessed in rats with macrophage depletion by clodronate. We investigated the effects of adoptive transfer of M2 macrophages or M2-derived cathepsin S (CTSS) on the sensory deficit. CTSS initiating signaling was explored by western blot analysis in IANX rats and immunohistochemistry in co-culture of primary fibroblasts and Schwann cells (SCs). RESULTS: Transcriptome analysis revealed that CTSS, a macrophage-selective lysosomal protease, was upregulated in the IAN after its injury. Spontaneous but partial recovery from a sensory deficit in the mandibular region after IANX was abrogated by macrophage ablation at the injured site. In addition, a robust induction of c-Jun, a marker of the repair-supportive phenotype of SCs, after IANX was abolished by macrophage ablation. As in transcriptome analysis, CTSS was upregulated at the injured IAN than in the intact IAN. Endogenous recovery from hypoesthesia was facilitated by supplementation of CTSS but delayed by pharmacological inhibition or genetic silencing of CTSS at the injured site. Adoptive transfer of M2-polarized macrophages at this site facilitated sensory recovery dependent on CTSS in macrophages. Post-IANX, CTSS caused the cleavage of Ephrin-B2 in fibroblasts, which, in turn, bound EphB2 in SCs. CTSS-induced Ephrin-B2 cleavage was also observed in human sensory nerves. Inhibition of CTSS-induced Ephrin-B2 signaling suppressed c-Jun induction in SCs and sensory recovery. CONCLUSIONS: These results suggest that M2 macrophage-derived CTSS contributes to axon regeneration by activating SCs via Ephrin-B2 shedding from fibroblasts.


Asunto(s)
Axones , Traumatismos de los Nervios Periféricos , Animales , Humanos , Ratas , Axones/patología , Catepsinas/metabolismo , Catepsinas/farmacología , Efrina-B2/metabolismo , Efrina-B2/farmacología , Fibroblastos/metabolismo , Macrófagos/metabolismo , Regeneración Nerviosa , Traumatismos de los Nervios Periféricos/metabolismo , Nervios Periféricos/patología , Ratas Sprague-Dawley , Células de Schwann/metabolismo
2.
Arch Oral Biol ; 87: 62-71, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29272761

RESUMEN

OBJECTIVE: To investigate the role of the EphrinB2 signaling pathway in the osteogenesis/odontogenesis of human dental pulp stem cells (DPSCs). DESIGN: The endogenous expression levels of EphrinB2 and its cognate receptors EphB2 and EphB4 in DPSCs were analyzed by qRT-PCR and Western blotting after 7, 14 and 21 days of osteogenic/odontogenic induction culture. Additionally, the phosphorylation of EphrinB2, EphB4 and ERK1/2 proteins at early time-points following osteogenic induction, were also investigated by Western blots. Subsequently, we investigated whether supplementation of recombinant EphrinB2-Fc within the induction milieu can enhance the osteogenic/odontogenic differentiation of DPSCs. RESULTS: Endogenous gene and protein expression levels of EphrinB2, EphB2 and EphB4 were upregulated in induced versus non-induced DPSCs, over 21 days of osteogenic/odontogenic induction. Western blots showed increase in phosphorylated EphrinB2, EphB4 and ERK1/2 proteins at early time-points following osteogenic induction. Preliminary investigation of a concentration range (0, 0.5, 1 and 2 µg/ml) of recombinant EphrinB2-Fc within osteogenic induction media, showed that 0.5 µg/ml was optimal for enhancing the osteogenic/odontogenic differentiation of DPSCs over a culture duration of 14 days. Subsequently, more comprehensive qRT-PCR analysis with 0.5 µg/ml EphrinB2-Fc revealed significant upregulation of several key osteogenic marker genes in treated versus untreated DPSCs after 21 days of osteogenic/odontogenic induction. By 7 days of osteogenic induction, DPSCs treated with 0.5 µg/ml EphrinB2-Fc exhibited significantly more calcium mineralization (Alizarin red S staining) and alkaline phosphatase activity than the untreated control. CONCLUSIONS: EphrinB2 signaling plays a key role in the osteogenic/odontogenic differentiation of DPSCs.


Asunto(s)
Diferenciación Celular/fisiología , Pulpa Dental/citología , Efrina-B2/farmacología , Transducción de Señal/fisiología , Western Blotting , Efrina-B2/metabolismo , Humanos , Odontogénesis/fisiología , Osteogénesis/fisiología , Fosforilación , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor EphB2/metabolismo , Receptor EphB2/farmacología , Receptor EphB4/metabolismo , Receptor EphB4/farmacología , Regulación hacia Arriba
3.
J Vasc Surg ; 65(1): 179-189, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26817610

RESUMEN

OBJECTIVE: Vein graft adaptation is characterized by loss of expression of the tyrosine kinase receptor Eph-B4, the embryonic determinant of venous identity, without increased expression of its ligand ephrin-B2, the embryonic determinant of arterial identity. Endothelial nitric oxide synthase (eNOS) is an important mediator of vessel remodeling. We hypothesized that the mechanism of action of Eph-B4 during vein graft adaptation might be through regulation of downstream eNOS activity. METHODS: Mouse lung endothelial cells were stimulated with ephrin-B2/Fc, without and with preclustering, without and with the eNOS inhibitor Nω-nitro-l-arginine methyl ester hydrochloride or the Eph-B4 inhibitor NVP-BHG712, and assessed by Western blot and immunofluorescence for eNOS and Eph-B4 phosphorylation. Nitric oxide (NO) production was assessed using an NO-specific chemiluminescence analyzer. Cell migration was assessed using a Transwell assay. Human and mouse vein graft specimens were examined for eNOS activity by Western blot, and vessel remodeling was assessed in vein grafts in wild-type or eNOS knockout mice. RESULTS: Ephrin-B2/Fc stimulated both Eph-B4 and eNOS phosphorylation in a bimodal temporal distribution (n = 4; P < .05), with preclustered ephrin-B2/Fc causing prolonged peak Eph-B4 and eNOS phosphorylation as well as altered subcellular localization (n = 4; P < .05). Ephrin-B2/Fc increased NO release (n = 3; P < .01) as well as increased endothelial cell migration (n = 6; P < .05) in an eNOS-dependent fashion. Both human and mouse vein grafts showed increased eNOS phosphorylation compared with normal veins (n = 3; P < .05). Vein grafts from eNOS knockout mice showed less dilation and less wall thickening compared with wild-type vein grafts (n = 7; P < .05). CONCLUSIONS: eNOS is a mediator of vein graft adaptation to the arterial environment. Eph-B4 stimulates eNOS phosphorylation in vitro and may mediate vein graft adaptation by regulation of eNOS activity in vivo.


Asunto(s)
Óxido Nítrico Sintasa de Tipo III/metabolismo , Receptor EphB4/metabolismo , Vena Safena/trasplante , Remodelación Vascular , Vena Cava Inferior/trasplante , Adaptación Fisiológica , Animales , Movimiento Celular , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Efrina-B2/farmacología , Genotipo , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo III/deficiencia , Óxido Nítrico Sintasa de Tipo III/genética , Fenotipo , Fosforilación , Vena Safena/enzimología , Vena Safena/patología , Transducción de Señal , Factores de Tiempo , Vena Cava Inferior/efectos de los fármacos , Vena Cava Inferior/enzimología , Vena Cava Inferior/patología
4.
Ann Neurol ; 80(3): 388-400, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27399303

RESUMEN

OBJECTIVE: To demonstrate that ephrin-B2 (the ligand of EphB2 receptor) antagonizes the pathogenic effects of patients' N-methyl-D-aspartate receptor (NMDAR) antibodies on memory and synaptic plasticity. METHODS: One hundred twenty-two C57BL/6J mice infused with cerebrospinal fluid (CSF) from patients with anti-NMDAR encephalitis or controls, with or without ephrin-B2, were investigated. CSF was infused through ventricular catheters connected to subcutaneous osmotic pumps over 14 days. Memory, behavioral tasks, locomotor activity, presence of human antibodies specifically bound to hippocampal NMDAR, and antibody effects on the density of cell-surface and synaptic NMDAR and EphB2 were examined at different time points using reported techniques. Short- and long-term synaptic plasticity were determined in acute brain sections; the Schaffer collateral pathway was stimulated and the field excitatory postsynaptic potentials were recorded in the CA1 region of the hippocampus. RESULTS: Mice infused with patients' CSF, but not control CSF, developed progressive memory deficit and depressive-like behavior along with deposits of NMDAR antibodies in the hippocampus. These findings were associated with a decrease of the density of cell-surface and synaptic NMDAR and EphB2, and marked impairment of long-term synaptic plasticity without altering short-term plasticity. Administration of ephrin-B2 prevented the pathogenic effects of the antibodies in all the investigated paradigms assessing memory, depressive-like behavior, density of cell-surface and synaptic NMDAR and EphB2, and long-term synaptic plasticity. INTERPRETATION: Administration of ephrin-B2 prevents the pathogenic effects of anti-NMDAR encephalitis antibodies on memory and behavior, levels of cell-surface NMDAR, and synaptic plasticity. These findings reveal a strategy beyond immunotherapy to antagonize patients' antibody effects. Ann Neurol 2016;80:388-400.


Asunto(s)
Encefalitis Antirreceptor N-Metil-D-Aspartato/tratamiento farmacológico , Anticuerpos/efectos de los fármacos , Región CA1 Hipocampal/efectos de los fármacos , Depresión/prevención & control , Efrina-B2/farmacología , Trastornos de la Memoria/prevención & control , Plasticidad Neuronal/efectos de los fármacos , Animales , Encefalitis Antirreceptor N-Metil-D-Aspartato/líquido cefalorraquídeo , Encefalitis Antirreceptor N-Metil-D-Aspartato/inmunología , Anticuerpos/inmunología , Conducta Animal , Región CA1 Hipocampal/inmunología , Depresión/etiología , Depresión/inmunología , Modelos Animales de Enfermedad , Humanos , Masculino , Trastornos de la Memoria/etiología , Trastornos de la Memoria/inmunología , Ratones , Ratones Endogámicos C57BL , Plasticidad Neuronal/inmunología , Receptor EphB2
5.
J Vasc Surg ; 63(3): 795-804, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25446283

RESUMEN

BACKGROUND: Vein bypass is an essential therapy for patients with advanced peripheral and coronary artery disease despite development of neointimal hyperplasia. We have shown that stimulation of the receptor tyrosine kinase ephrin type-B receptor 4 (Eph-B4) with its ligand ephrin-B2 prevents neointimal hyperplasia in murine vein grafts. This study determines whether Eph-B4 in adult human veins is capable of phosphorylation and activation of downstream signaling pathways, as well as functional to release nitric oxide (NO) and prevent neointimal hyperplasia in vitro. METHODS: Discarded human saphenous veins were taken from the operating room and placed in organ culture without or with ephrin-B2/Fc (2 µg/mL) for 14 days, and the neointima/media ratio was measured in matched veins. Primary human umbilical vein endothelial cells were treated with ephrin-B2/Fc (2 µg/mL) and examined with quantitative polymerase chain reaction, Western blot, immunoassays, and for release of NO. Ephrin-B2/Fc (2 µg/mL) was placed on the adventitia of saphenous veins treated with arterial shear stress for 24 hours in a bioreactor and activated Eph-B4 examined with immunofluorescence. RESULTS: The baseline intima/media ratio in saphenous vein rings was 0.456 ± 0.097, which increased to 0.726 ± 0.142 in untreated veins after 14 days in organ culture but only to 0.630 ± 0.132 in veins treated with ephrin-B2/Fc (n = 19, P = .017). Ephrin-B2/Fc stimulated Akt, endothelial NO synthase and caveolin-1 phosphorylation, and NO release (P = .007) from human umbilical vein endothelial cells (n = 6). Ephrin-B2/Fc delivered to the adventitia stimulated endothelial Eph-B4 phosphorylation after 24 hours of arterial stress in a bioreactor (n = 3). CONCLUSIONS: Eph-B4 is present and functional in adult human saphenous veins, with intact downstream signaling pathways capable of NO release and prevention of neointimal hyperplasia in vitro. Adventitial delivery of ephrin-B2/Fc activates endothelial Eph-B4 in saphenous veins treated with arterial shear stress in vitro. These results suggest that stimulation of Eph-B4 function may be a candidate strategy for translation to human clinical trials designed to inhibit venous neointimal hyperplasia.


Asunto(s)
Efrina-B2/farmacología , Fragmentos Fc de Inmunoglobulinas/farmacología , Neointima , Receptor EphB4/agonistas , Vena Safena/efectos de los fármacos , Reactores Biológicos , Caveolina 1/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Activación Enzimática , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Hiperplasia , Mecanotransducción Celular/efectos de los fármacos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosforilación , Cultivo Primario de Células , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor EphB4/genética , Receptor EphB4/metabolismo , Vena Safena/metabolismo , Vena Safena/patología , Estrés Mecánico , Técnicas de Cultivo de Tejidos/instrumentación
6.
J Cell Physiol ; 230(12): 2961-72, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25893671

RESUMEN

The mammalian secondary palate forms from shelves of epithelia-covered mesenchyme that meet at midline and fuse. The midline epithelial seam (MES) is thought to degrade by apoptosis, epithelial-to-mesenchymal transition (EMT), or both. Failure to degrade the MES blocks fusion and causes cleft palate. It was previously thought that transforming growth factor ß3 (Tgfß3) is required to initiate fusion. Members of the Eph tyrosine kinase receptor family and their membrane-bound ephrin ligands are expressed on the MES. We demonstrated that treatment of mouse palates with recombinant EphB2/Fc to activate ephrin reverse signaling (where the ephrin acts as a receptor and transduces signals from its cytodomain) was sufficient to cause mouse palatal fusion when Tgfß3 signaling was blocked by an antibody against Tgfß3 or by an inhibitor of the TgfßrI serine/threonine receptor kinase. Cultured palatal epithelial cells traded their expression of epithelial cell markers for that of mesenchymal cells and became motile after treatment with EphB2/Fc. They concurrently increased their expression of the EMT-associated transcription factors Snail, Sip1, and Twist1. EphB2/Fc did not cause apoptosis in these cells. These data reveal that ephrin reverse signaling directs palatal fusion in mammals through a mechanism that involves EMT but not apoptosis and activates a gene expression program not previously associated with ephrin reverse signaling.


Asunto(s)
Desarrollo Óseo/efectos de los fármacos , Efrina-B2/farmacología , Efrinas/metabolismo , Células Epiteliales/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Hueso Paladar/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta3/metabolismo , Animales , Movimiento Celular , Células Cultivadas , Células Epiteliales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Ratones , Morfogénesis , Hueso Paladar/embriología , Hueso Paladar/metabolismo , Proteínas Recombinantes/farmacología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta3/antagonistas & inhibidores
7.
Cell Physiol Biochem ; 35(1): 104-15, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25547331

RESUMEN

UNLABELLED: BACKGROUND/ AIMS: The knowledge of the molecular network that governs fetal lung branching is an essential step towards the discovery of novel therapeutic targets against pulmonary pathologies. Lung consists of two highly branched systems: airways and vasculature. Ephrins and its receptors, Eph, have been implicated in cardiovascular development, angiogenesis and vascular remodeling. This study aims to clarify the role of these factors during lung morphogenesis. METHODS: Ephrins-B1, -B2 and receptor EphB4 expression pattern was assessed in fetal rat lungs between 15.5 and 21.5 days post-conception, by immunohistochemistry. Fetal rat lungs were harvested at 13.5 dpc, cultured during 4 days and treated with increasing doses of ephrins-B1 and -B2 and the activity of key signaling pathways was assessed. RESULTS: Ephrin-B1 presents mesenchymal expression, whereas ephrin-B2 and its receptor EphB4 were expressed by the epithelium. Both ephrins stimulated pulmonary branching. Moreover, while ephrin-B1 did not affect the pathways studied, ephrin-B2 supplementation decreased activity of JNK, ERK and STAT. This study characterizes the expression pattern of ephrins-B1, -B2 and EphB4 receptor throughout rat lung development. CONCLUSION: Our data highlight a possible role of ephrins as molecular stimulators of lung morphogenesis. Moreover, it supports the idea that classical vascular factors might play a role as airway growth promoters.


Asunto(s)
Efrina-B1/metabolismo , Efrina-B2/metabolismo , Pulmón/crecimiento & desarrollo , Animales , Células Cultivadas , Desarrollo Embrionario , Efrina-B1/genética , Efrina-B1/farmacología , Efrina-B2/genética , Efrina-B2/farmacología , Epitelio/metabolismo , Femenino , Feto/metabolismo , Feto/patología , Técnicas In Vitro , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Pulmón/efectos de los fármacos , Pulmón/patología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Morfogénesis , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptor EphB4/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos
8.
Dis Markers ; 35(6): 933-8, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24427781

RESUMEN

BACKGROUND: EphB4 receptor tyrosine kinase is of diagnostic and therapeutic value due to its overexpression in breast tumors. Dual functions of tumor promotion and suppression have been reported for this receptor based on presence or absence of its ligand. To elucidate such discrepancy, we aimed to determine the effect of time- and dose-dependent stimulation of EphB4 on viability and invasion of breast cancer cells via recombinant ephrinB2-Fc. METHODS: Cells were seeded into multiwell plates and were stimulated by various concentrations of preclustered ephrinB2-Fc. Cell viability was measured on days 3 and 6 following treatment using alamar-blue when cells were in different states of confluence. RESULTS: Stimulation of cells with ephrinB2 did not pose any significant effect on cell viability before reaching confluence, while inhibition of cell growth was detected after 6 days when cells were in postconfluent state following a dose-dependent manner. EphrinB2 treatment did not affect tubular formation and invasion on matrigel. CONCLUSION: This study showed that EphB4 can differentially inhibit cells at post confluent state and that presence of ligand manifests growth-inhibitory properties of EphB4 receptor. It is concluded that growth inhibition has occurred possibly due to long treatment with ligand, a process which leads to receptor downregulation.


Asunto(s)
Efrina-B2/farmacología , Receptor EphB4/metabolismo , Neoplasias de la Mama , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular , Supervivencia Celular/efectos de los fármacos , Activación Enzimática , Femenino , Humanos , Fosforilación , Procesamiento Proteico-Postraduccional , Receptor EphB4/agonistas
9.
Dev Dyn ; 241(12): 1901-10, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23129351

RESUMEN

INTRODUCTION: Ephrin-B2 on osteoclasts was reported to promote bone formation as part of homeostasis by activating the EphB4 tyrosine kinase receptor on osteoblasts. Little is known about the role of ephrin-B signaling to EphBs in developmental bone formation. RESULTS: We observed expression of an ephrin-B2 LacZ chimeric allele in the periosteum, sutural bone fronts, and dura mater of embryonic and neonatal mice. Expression in the adult skull was confined to sutures, but was heavily upregulated at sites of bone injury. Culture of embryonic calvariae with soluble recombinant ephrin-B2/Fc doubled their bone content without altering suture width or overall skull morphology. Ephrin-B2/Fc also stimulated osteoblast marker gene expression in cultured MC3T3 preosteoblastic cells without the need for type 1 collagen-induced differentiation. EphB4 was absent in embryonic and adult skulls. However, EphB1 and EphB2, both physiological receptors for ephrin-Bs, were expressed at sites of osteogenesis, and EphB1 knockout mice displayed a reduction in calvarial bone content compared to controls. CONCLUSIONS: These data support a role for ephrin-B2 in the development and healing of bone through activation of osteoblast-specific gene expression. EphB1 and EphB2 are likely candidates receptors for the ephrin-B2 in bone.


Asunto(s)
Efrina-B2/metabolismo , Osteoblastos/metabolismo , Osteogénesis/fisiología , Cráneo/embriología , Animales , Antígenos de Diferenciación/biosíntesis , Antígenos de Diferenciación/genética , Línea Celular , Efrina-B2/genética , Efrina-B2/farmacología , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/farmacología , Ratones , Ratones Noqueados , Técnicas de Cultivo de Órganos , Osteoblastos/citología , Osteogénesis/efectos de los fármacos , Receptor EphB1/genética , Receptor EphB1/metabolismo , Receptor EphB2/genética , Receptor EphB2/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología , Cráneo/citología
10.
Eur J Immunol ; 42(6): 1562-72, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22622783

RESUMEN

Bidirectional signals via Eph receptors/ephrins have been recognized as major forms of contact-dependent cell communications such as cell attraction and repulsion. T cells express EphBs, and their ligands, the ephrin-Bs, have been known as costimulatory molecules for T-cell proliferation. Recently, another remarkable feature of ephrin-As has emerged in the form of a concentration-dependent transition from promotion to inhibition in axon growth. Here we examined whether this modification plays a role in ephrin-B costimulation in murine primary T cells. Low doses of ephrin-B1 and ephrin-B2 costimulated T-cell proliferation induced by anti-CD3, but high concentrations strongly inhibited it. In contrast, ephrin-B3 showed a steadily increasing stimulatory effect. This modulation was virtually preserved in T cells from mice simultaneously lacking four genes, EphB1, EphB2, EphB3, and EphB6. High concentrations of ephrin-B1/B2, but not ephrin-B3, inhibited the anti-CD3-induced phosphorylation of Lck and its downstream signals such as Erk and Akt. Additionally, high doses of any ephrin-Bs could phosphorylate EphB4. However, only ephrin-B1/B2 but not ephrin-B3 recruited SHP1, a phosphatase to suppress the phosphorylation of Lck. These data suggest that EphB4 signaling could engage in negative feedback to TCR signals. T-cell activation may be finely adjusted by the combination and concentration of ephrin-Bs expressed in the immunological microenvironment.


Asunto(s)
Efrina-B1/farmacología , Efrina-B2/farmacología , Activación de Linfocitos/efectos de los fármacos , Linfocitos T/inmunología , Animales , Relación Dosis-Respuesta a Droga , Retroalimentación Fisiológica , Ratones , Ratones Endogámicos C57BL , Proteína Tirosina Fosfatasa no Receptora Tipo 6/fisiología , Receptor EphB3/farmacología , Receptor EphB4/farmacología , Receptores de Antígenos de Linfocitos T/fisiología , Transducción de Señal
11.
Blood ; 115(12): 2412-9, 2010 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-20061560

RESUMEN

Eph receptors and their ephrin ligands are involved in normal hematopoietic development and tumorigenesis. Using methylated CpG island amplification/DNA promoter microarray, we identified several EPH receptor and EPHRIN genes as potential hypermethylation targets in acute lymphoblastic leukemia (ALL). We subsequently studied the DNA methylation status of the Eph/ephrin family by bisulfite pyrosequencing. Hypermethylation of EPHA2, -A4, -A5, -A6, -A7, -A10, EPHB1, -B2, -B3, -B4, EFNA1, -A3, -A5, and EFNB1 and -B2 genes was detected in leukemia cell lines and primary ALL bone marrow samples. Expression analysis of EPHB4, EFNB2, and EFNA5 genes demonstrated that DNA methylation was associated with gene silencing. We cloned the promoter region of EPHB4 and demonstrated that promoter hypermethylation can result in EPHB4 transcriptional silencing. Restoration of EPHB4 expression by lentiviral transduction resulted in reduced proliferation and apoptotic cell death in Raji cells in which EPHB4 is methylated and silenced. Finally, we demonstrated that phosphorylated Akt is down-regulated in Raji cells transduced with EPHB4. These results suggest that epigenetic silencing by hypermethylation of EPH/EPHRIN family genes contributes to ALL pathogenesis and that EPHB4 can function as a tumor suppressor in ALL.


Asunto(s)
Metilación de ADN , Efrina-B2/genética , Epigénesis Genética , Regulación Leucémica de la Expresión Génica , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Receptor EphB4/genética , Apoptosis/fisiología , División Celular/fisiología , Efrina-B2/metabolismo , Efrina-B2/farmacología , Efrinas/genética , Efrinas/metabolismo , Efrinas/farmacología , Silenciador del Gen , Genes Supresores de Tumor , Humanos , Células Jurkat , Familia de Multigenes/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos , Leucemia-Linfoma Linfoblástico de Células Precursoras/mortalidad , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Prevalencia , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor EphB4/metabolismo , Receptores de la Familia Eph/genética , Receptores de la Familia Eph/metabolismo , Análisis de Supervivencia , Transducción Genética
12.
Int J Oncol ; 34(2): 321-7, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19148465

RESUMEN

The ephB4-ephrinB2 system plays an important role in the interaction of tumor cells with endothelial cells (ECs). To assess the role of ephB4 in the in vivo growth of head and neck squamous cell carcinoma (HNSCC), we used ephrinB2-Fc, a fusion protein consisting of the extracellular domain of ephrin-B2 and the Fc portion of human IgG1, as the soluble ligand for ephB4. EphrinB2-Fc injection into HNSCC xenografted mice significantly suppressed xenograft growth, accompanied by a decrease in vessel cross-sectional area, but there was no change in vessel number. EphrinB2-Fc injection also induced the formation of mature blood vessels rich in alpha-smooth muscle actin positive pericytes in the xenograft tissue. In vitro assays revealed that ephrinB2-Fc inhibited the proliferation of human umbilical vein ECs (HUVECs) but not tumor cells. Furthermore, real-time quantitative RT-PCR showed that ephrinB2-Fc down-regulated matrix metalloproteinase-2 mRNA expression in HUVECs and vascular endothelial growth factor-A in tumor cells. These data suggest that treatment with ephrinB2-Fc, the soluble ligand of ephB4, inhibited the growth of HNSCC through vessel maturation/stabilization, preventing leakiness and endothelial sprout formation.


Asunto(s)
Carcinoma de Células Escamosas/patología , División Celular/efectos de los fármacos , Efrina-B2/farmacología , Neoplasias de Cabeza y Cuello/patología , Animales , Apoptosis/efectos de los fármacos , Carcinoma de Células Escamosas/irrigación sanguínea , Cartilla de ADN , Neoplasias de Cabeza y Cuello/irrigación sanguínea , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neovascularización Patológica/patología , Reacción en Cadena de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trasplante Heterólogo
13.
J Clin Invest ; 117(6): 1527-37, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17510705

RESUMEN

Endothelial progenitor cell (EPC) transplantation has beneficial effects for therapeutic neovascularization; however, only a small proportion of injected cells home to the lesion and incorporate into the neocapillaries. Consequently, this type of cell therapy requires substantial improvement to be of clinical value. Erythropoietin-producing human hepatocellular carcinoma (Eph) receptors and their ephrin ligands are key regulators of vascular development. We postulated that activation of the EphB4/ephrin-B2 system may enhance EPC proangiogenic potential. In this report, we demonstrate in a nude mouse model of hind limb ischemia that EphB4 activation with an ephrin-B2-Fc chimeric protein increases the angiogenic potential of human EPCs. This effect was abolished by EphB4 siRNA, confirming that it is mediated by EphB4. EphB4 activation enhanced P selectin glycoprotein ligand-1 (PSGL-1) expression and EPC adhesion. Inhibition of PSGL-1 by siRNA reversed the proangiogenic and adhesive effects of EphB4 activation. Moreover, neutralizing antibodies to E selectin and P selectin blocked ephrin-B2-Fc-stimulated EPC adhesion properties. Thus, activation of EphB4 enhances EPC proangiogenic capacity through induction of PSGL-1 expression and adhesion to E selectin and P selectin. Therefore, activation of EphB4 is an innovative and potentially valuable therapeutic strategy for improving the recruitment of EPCs to sites of neovascularization and thereby the efficiency of cell-based proangiogenic therapy.


Asunto(s)
Células Endoteliales/metabolismo , Células Madre Fetales/metabolismo , Glicoproteínas de Membrana/metabolismo , Neovascularización Fisiológica , Receptor EphB4/metabolismo , Animales , Secuencia de Bases , Adhesión Celular , Células Cultivadas , Cartilla de ADN/genética , Selectina E/metabolismo , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Efrina-B2/metabolismo , Efrina-B2/farmacología , Sangre Fetal/citología , Células Madre Fetales/citología , Células Madre Fetales/efectos de los fármacos , Miembro Posterior/irrigación sanguínea , Humanos , Técnicas In Vitro , Isquemia/metabolismo , Isquemia/patología , Isquemia/terapia , Masculino , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/genética , Ratones , Ratones Desnudos , Neovascularización Fisiológica/efectos de los fármacos , Selectina-P/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , Receptor EphB4/antagonistas & inhibidores , Receptor EphB4/genética
14.
Biochemistry ; 46(7): 1799-810, 2007 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-17249693

RESUMEN

Azurin is a member of a family of metalloproteins called cupredoxins. Although previously thought to be involved in electron transfer, azurin has recently been shown to preferentially enter cancer cells than normal cells and induce apoptosis in such cells. Azurin also demonstrates structural similarity to a ligand known as ephrinB2, which binds its cognate receptor tyrosine kinase EphB2 to initiate cell signaling. Eph/ephrin signaling is known to be involved in cancer progression. We now demonstrate that azurin binds to the EphB2-Fc receptor with high affinity. We have localized a C-terminal domain of azurin (Azu 96-113) that exhibits structural similarity to ephrinB2 at the G-H loop region known to be involved in receptor binding. A synthetic peptide (Azu 96-113) as well as a GST fusion derivative GST-Azu 88-113 interferes with the growth of various human cancer cells. In a prostate cancer cell line DU145 lacking functional EphB2, azurin or its GST-fusion derivatives had little cytotoxic effect. However, in DU145 cells expressing functional EphB2, azurin and GST-Azu 88-113 demonstrated significant cytotoxicity, whereas ephrinB2 promoted cell growth. Azurin inhibited the ephrinB2-mediated autophosphorlyation of the EphB2 tyrosine residue, thus interfering in upstream cell signaling and contributing to cancer cell growth inhibition.


Asunto(s)
Azurina/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Receptor EphB2/metabolismo , Tirosina/metabolismo , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología , Azurina/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cobre , Efrina-B2/metabolismo , Efrina-B2/farmacología , Humanos , Modelos Moleculares , Mutación , Péptidos/síntesis química , Péptidos/química , Péptidos/farmacología , Fosforilación , Estructura Terciaria de Proteína , Receptor EphB2/química
15.
Cell Metab ; 4(2): 111-21, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16890539

RESUMEN

Bone homeostasis requires a delicate balance between the activities of bone-resorbing osteoclasts and bone-forming osteoblasts. Various molecules coordinate osteoclast function with that of osteoblasts; however, molecules that mediate osteoclast-osteoblast interactions by simultaneous signal transduction in both cell types have not yet been identified. Here we show that osteoclasts express the NFATc1 target gene Efnb2 (encoding ephrinB2), while osteoblasts express the receptor EphB4, along with other ephrin-Eph family members. Using gain- and loss-of-function experiments, we demonstrate that reverse signaling through ephrinB2 into osteoclast precursors suppresses osteoclast differentiation by inhibiting the osteoclastogenic c-Fos-NFATc1 cascade. In addition, forward signaling through EphB4 into osteoblasts enhances osteogenic differentiation, and overexpression of EphB4 in osteoblasts increases bone mass in transgenic mice. These data demonstrate that ephrin-Eph bidirectional signaling links two major molecular mechanisms for cell differentiation--one in osteoclasts and the other in osteoblasts--thereby maintaining bone homeostasis.


Asunto(s)
Huesos/fisiología , Efrina-B2/fisiología , Homeostasis , Receptor EphB4/metabolismo , Transducción de Señal , Animales , Secuencia de Bases , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Efrina-B2/genética , Efrina-B2/farmacología , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Factores de Transcripción NFATC/antagonistas & inhibidores , Factores de Transcripción NFATC/metabolismo , Osteoblastos/citología , Osteoclastos/citología , Proteínas Proto-Oncogénicas c-fos/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptor EphB4/genética , Regulación hacia Arriba
16.
Nat Cell Biol ; 8(8): 815-25, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16862147

RESUMEN

Recent evidence supports a role for EphB receptor tyrosine kinases as tumour suppressors in colorectal and prostate cancer. However, it is unclear how these receptors inhibit cancer cell tumorigenicity - an activity that is highly unusual for a family of receptor tyrosine kinases. Here, we report that the EphB4 receptor can behave as a tumour suppressor in a mouse xenograft model of breast cancer when stimulated by its ligand, ephrin-B2. In breast cancer cells, EphB4 activates an antioncogenic pathway involving Abl family tyrosine kinases and the Crk adaptor protein. This Abl-Crk pathway inhibits breast cancer cell viability and proliferation in addition to motility and invasion, and also downregulates the pro-invasive matrix metalloprotease, MMP-2. Consistent with these effects, EphB4 and the Abl-Crk pathway are constitutively active in non-transformed mammary epithelial cells. These findings identify a novel Eph receptor signalling pathway with tumour-suppressor activity and predict that therapeutic intervention to activate EphB4 signalling will inhibit tumour progression.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas Proto-Oncogénicas c-abl/metabolismo , Proteínas Proto-Oncogénicas c-crk/metabolismo , Receptor EphB4/fisiología , Transducción de Señal/fisiología , Animales , Benzamidas , Neoplasias de la Mama/patología , Neoplasias de la Mama/prevención & control , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Efrina-B2/genética , Efrina-B2/metabolismo , Efrina-B2/farmacología , Femenino , Humanos , Mesilato de Imatinib , Fragmentos Fc de Inmunoglobulinas/genética , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Neoplasias Mamarias Experimentales/prevención & control , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Receptor EphB4/agonistas , Receptor EphB4/metabolismo , Proteínas Recombinantes de Fusión/farmacología
17.
J Cell Sci ; 119(Pt 11): 2299-309, 2006 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-16723736

RESUMEN

Eph receptor tyrosine kinases are key players during the development of the embryonic vasculature; however, their role and regulation in adult angiogenesis remain to be defined. Caveolae are flask-shaped invaginations of the cell membrane; their major structural protein, caveolin-1, has been shown to regulate signaling molecules localized in these micro-domains. The interaction of caveolin-1 with several of these proteins is mediated by the binding of its scaffolding domain to a region containing hydrophobic amino acids within these proteins. The presence of such a motif within the EphB1 kinase domain prompted us to investigate the caveolar localization and regulation of EphB1 by caveolin-1. We report that EphB1 receptors are localized in caveolae, and directly interact with caveolin-1 upon ligand stimulation. This interaction, as well as EphB1-mediated activation of extracellular-signal-regulated kinase (ERK), was abrogated by overexpression of a caveolin-1 mutant lacking a functional scaffolding domain. Interaction between Ephs and caveolin-1 is not restricted to the B-subclass of receptors, since we show that EphA2 also interacts with caveolin-1. Furthermore, we demonstrate that the caveolin-binding motif within the kinase domain of EphB1 is primordial for its correct membrane targeting. Taken together, our findings establish caveolin-1 as an important regulator of downstream signaling and membrane targeting of EphB1.


Asunto(s)
Caveolina 1/metabolismo , Membrana Celular/metabolismo , Receptor EphB1/metabolismo , Transducción de Señal/fisiología , Animales , Células CHO , Células COS , Caveolina 1/efectos de los fármacos , Caveolina 1/genética , Línea Celular Tumoral , Chlorocebus aethiops , Colesterol/farmacología , Cricetinae , Efrina-B2/farmacología , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Fosforilación , Transporte de Proteínas/fisiología , Receptor EphA2/metabolismo
18.
Cell ; 124(1): 161-73, 2006 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-16413489

RESUMEN

New blood vessels are initially formed through the assembly or sprouting of endothelial cells, but the recruitment of supporting pericytes and vascular smooth muscle cells (mural cells) ensures the formation of a mature and stable vascular network. Defective mural-cell coverage is associated with the poorly organized and leaky vasculature seen in tumors or other human diseases. Here we report that mural cells require ephrin-B2, a ligand for Eph receptor tyrosine kinases, for normal association with small-diameter blood vessels (microvessels). Tissue-specific mutant mice display perinatal lethality; vascular defects in skin, lung, gastrointestinal tract, and kidney glomeruli; and abnormal migration of smooth muscle cells to lymphatic capillaries. Cultured ephrin-B2-deficient smooth muscle cells are defective in spreading, focal-adhesion formation, and polarized migration and show increased motility. Our results indicate that the role of ephrin-B2 and EphB receptors in these processes involves Crk-p130(CAS) signaling and suggest that ephrin-B2 has some cell-cell-contact-independent functions.


Asunto(s)
Vasos Sanguíneos/crecimiento & desarrollo , Movimiento Celular/efectos de los fármacos , Efrina-B2/fisiología , Animales , Vasos Sanguíneos/citología , Vasos Sanguíneos/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Efrina-B2/genética , Efrina-B2/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Modelos Biológicos , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Neovascularización Fisiológica/fisiología , Fenotipo , Transducción de Señal/fisiología
19.
J Neurosci ; 24(8): 1976-86, 2004 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-14985440

RESUMEN

During development, EphB proteins serve as axon guidance molecules for retinal ganglion cell axon pathfinding toward the optic nerve head and in midbrain targets. To better understand the mechanisms by which EphB proteins influence retinal growth cone behavior, we investigated how axon responses to EphB were modulated by laminin and L1, two guidance molecules that retinal axons encounter during in vivo pathfinding. Unlike EphB stimulation in the presence of laminin, which triggers typical growth cone collapse, growth cones co-stimulated by L1 did not respond to EphB. Moreover, EphB exposure in the presence of both laminin and L1 resulted in a novel growth cone inhibition manifested as a pause in axon elongation with maintenance of normal growth cone morphology and filopodial activity. Pauses were not associated with loss of growth cone actin but were accompanied by a redistribution of the microtubule cytoskeleton with increased numbers of microtubules extending into filopodia and to the peripheral edge of the growth cone. This phenomenon was accompanied by reduced levels of the growth cone microtubule destabilizing protein SCG10. Antibody blockade of SCG10 function in growth cones resulted in both changes in microtubule distribution and pause responses mirroring those elicited by EphB in the presence of laminin and L1. These results demonstrate that retinal growth cone responsiveness to EphB is regulated by co-impinging signals from other axon guidance molecules. Furthermore, the results are consistent with EphB-mediated axon guidance mechanisms that involve the SCG10-mediated regulation of the growth cone microtubule cytoskeleton.


Asunto(s)
Efrina-B2/farmacología , Conos de Crecimiento/efectos de los fármacos , Laminina/farmacología , Microtúbulos/efectos de los fármacos , Molécula L1 de Adhesión de Célula Nerviosa/farmacología , Animales , Anticuerpos/farmacología , Axones/efectos de los fármacos , Axones/fisiología , Proteínas de Unión al Calcio , Células Cultivadas , Interacciones Farmacológicas , Efrina-B2/genética , Conos de Crecimiento/fisiología , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Péptidos y Proteínas de Señalización Intracelular , Proteínas de la Membrana , Ratones , Microtúbulos/metabolismo , Factores de Crecimiento Nervioso/antagonistas & inhibidores , Factores de Crecimiento Nervioso/metabolismo , Molécula L1 de Adhesión de Célula Nerviosa/genética , Neuronas/efectos de los fármacos , Neuronas/fisiología , Neuronas/ultraestructura , Fragmentos de Péptidos/farmacología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología , Retina/citología , Estatmina
20.
Biochem Biophys Res Commun ; 313(1): 80-8, 2004 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-14672701

RESUMEN

Signaling between the ligand ephrinB2 and the respective receptors of the EphB class is known to play a vital role during vascular morphogenesis and angiogenesis. The relative contribution of each EphB receptor type present on endothelial cells to these processes remains to be determined. It has been shown that ephrinB2-EphB receptor signal transduction leads to a repulsive migratory behavior of endothelial cells. It remained unclear whether this anti-migratory effect can be mediated by EphB4 signaling alone or whether other EphB receptors are necessary. It also remained unclear whether the kinase activity of EphB4 is pivotal to its action. To answer these questions, we developed a cellular migration system solely dependent on ephrinB2-EphB4 signaling. Using this system, we could show that EphB4 activation leads to the inhibition of cell migration. Furthermore we identified PP2, a known inhibitor of kinases of the Src family, and PD 153035, a known inhibitor of EGF receptor kinase, as inhibitors of EphB4 kinase activity. Using PP2, the inhibition of cell migration by ephrinB2 could be relieved, demonstrating that the kinase function of EphB4 is of prominent importance in this process. These results show that EphB4 activation is not only accompanying ephrinB2 induced repulsive behavior of cells, but is capable of directly mediating this effect.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Efrina-B2/metabolismo , Efrina-B2/farmacología , Receptor EphB4/metabolismo , Animales , Células CHO , Células Cultivadas , Cricetinae , Endotelio Vascular/citología , Inhibidores Enzimáticos/farmacología , Efrina-B2/antagonistas & inhibidores , Receptores ErbB/antagonistas & inhibidores , Humanos , Microcirculación , Fosforilación , Pirimidinas/farmacología , Quinazolinas/farmacología , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal , Factores de Crecimiento Endotelial Vascular/farmacología , Familia-src Quinasas/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA