Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 22(17)2021 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-34502339

RESUMEN

Motor neuron disease (MND) comprises a group of fatal neurodegenerative diseases with no effective cure. As progressive motor neuron cell death is one of pathological characteristics of MND, molecules which protect these cells are attractive therapeutic targets. Accumulating evidence indicates that EphA4 activation is involved in MND pathogenesis, and inhibition of EphA4 improves functional outcomes. However, the underlying mechanism of EphA4's function in MND is unclear. In this review, we first present results to demonstrate that EphA4 signalling acts directly on motor neurons to cause cell death. We then review the three most likely mechanisms underlying this effect.


Asunto(s)
Muerte Celular , Enfermedad de la Neurona Motora/patología , Neuronas Motoras/patología , Receptor EphA4/metabolismo , Animales , Humanos , Enfermedad de la Neurona Motora/metabolismo , Neuronas Motoras/metabolismo , Transducción de Señal
2.
Cereb Cortex ; 29(10): 4381-4397, 2019 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-30590507

RESUMEN

The hippocampal dentate gyrus (DG) is a major region of the adult rodent brain in which neurogenesis occurs throughout life. The EphA4 receptor, which regulates neurogenesis and boundary formation in the developing brain, is also expressed in the adult DG, but whether it regulates adult hippocampal neurogenesis is not known. Here, we show that, in the adult mouse brain, EphA4 inhibits hippocampal precursor cell proliferation but does not affect precursor differentiation or survival. Genetic deletion or pharmacological inhibition of EphA4 significantly increased hippocampal precursor proliferation in vivo and in vitro, by blocking EphA4 forward signaling. EphA4 was expressed by mature hippocampal DG neurons but not neural precursor cells, and an EphA4 antagonist, EphA4-Fc, did not activate clonal cultures of precursors until they were co-cultured with non-precursor cells, indicating an indirect effect of EphA4 on the regulation of precursor activity. Supplementation with d-serine blocked the increased precursor proliferation induced by EphA4 inhibition, whereas blocking the interaction between d-serine and N-methyl-d-aspartate receptors (NMDARs) promoted precursor activity, even at the clonal level. Collectively, these findings demonstrate that EphA4 indirectly regulates adult hippocampal precursor proliferation and thus plays a role in neurogenesis via d-serine-regulated NMDAR signaling.


Asunto(s)
Giro Dentado/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis , Receptor EphA4/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Supervivencia Celular , Femenino , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor EphA4/genética , Transducción de Señal
3.
Acta Neuropathol ; 138(6): 1033-1052, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31463571

RESUMEN

Glioblastomas (GBMs) are malignant central nervous system (CNS) neoplasms with a very poor prognosis. They display cellular hierarchies containing self-renewing tumourigenic glioma stem cells (GSCs) in a complex heterogeneous microenvironment. One proposed GSC niche is the extracellular matrix (ECM)-rich perivascular bed of the tumour. Here, we report that the ECM binding dystroglycan (DG) receptor is expressed and functionally glycosylated on GSCs residing in the perivascular niche. Glycosylated αDG is highly expressed and functional on the most aggressive mesenchymal-like (MES-like) GBM tumour compartment. Furthermore, we found that DG acts to maintain an MES-like state via tight control of MAPK activation. Antibody-based blockade of αDG induces robust ERK-mediated differentiation leading to reduced GSC potential. DG was shown to be required for tumour initiation in MES-like GBM, with constitutive loss significantly delaying or preventing tumourigenic potential in-vivo. These findings reveal a central role of the DG receptor, not only as a structural element, but also as a critical factor promoting MES-like GBM and the maintenance of GSCs residing in the perivascular niche.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Distroglicanos/metabolismo , Glioma/metabolismo , Células Madre Neoplásicas/metabolismo , Microambiente Tumoral/fisiología , Animales , Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/cirugía , Transformación Celular Neoplásica , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Glioma/irrigación sanguínea , Glioma/cirugía , Humanos , Ratones Endogámicos NOD , Ratones SCID , Trasplante de Neoplasias
4.
Cancer Immunol Immunother ; 66(9): 1217-1228, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28501939

RESUMEN

We have reported that calcitonin receptor (CTR) is widely expressed in biopsies from the lethal brain tumour glioblastoma by malignant glioma and brain tumour-initiating cells (glioma stem cells) using anti-human CTR antibodies. A monoclonal antibody against an epitope within the extracellular domain of CTR was raised (mAb2C4) and chemically conjugated to either plant ribosome-inactivating proteins (RIPs) dianthin-30 or gelonin, or the drug monomethyl auristatin E (MMAE), and purified. In the high-grade glioma cell line (HGG, representing glioma stem cells) SB2b, in the presence of the triterpene glycoside SO1861, the EC50 for mAb2C4:dianthin was 10.0 pM and for mAb2C4:MMAE [antibody drug conjugate (ADC)] 2.5 nM, 250-fold less potent. With the cell line U87MG, in the presence of SO1861, the EC50 for mAb2C4:dianthin was 20 pM, mAb2C4:gelonin, 20 pM, compared to the ADC (6.3 nM), which is >300 less potent. Several other HGG cell lines that express CTR were tested and the efficacies of mAb2C4:RIP (dianthin or gelonin) were similar. Co-administration of the enhancer SO1861 purified from plants enhances lysosomal escape. Enhancement with SO1861 increased potency of the immunotoxin (>3 log values) compared to the ADC (1 log). The uptake of antibody was demonstrated with the fluorescent conjugate mAb2C4:Alexa Fluor 568, and the release of dianthin-30:Alexa Fluor488 into the cytosol following addition of SO1861 supports our model. These data demonstrate that the immunotoxins are highly potent and that CTR is an effective target expressed by a large proportion of HGG cell lines representative of glioma stem cells and isolated from individual patients.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Oligopéptidos/farmacología , Receptores de Calcitonina/antagonistas & inhibidores , Proteínas Inactivadoras de Ribosomas Tipo 1/farmacología , Anticuerpos Monoclonales/inmunología , Línea Celular Tumoral , Humanos , Receptores de Calcitonina/inmunología , Células Tumorales Cultivadas
5.
J Neurosci ; 34(8): 2921-30, 2014 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-24553933

RESUMEN

Epigenetic mechanisms are essential in regulating neural progenitor cell self-renewal, with the chromatin-modifying protein Enhancer of zeste homolog 2 (EZH2) emerging as a central player in promoting progenitor cell self-renewal during cortical development. Despite this, how Ezh2 is itself regulated remains unclear. Here, we demonstrate that the transcription factor nuclear factor IB (NFIB) plays a key role in this process. Nfib(-/-) mice exhibit an increased number of proliferative ventricular zone cells that express progenitor cell markers and upregulation of EZH2 expression within the neocortex and hippocampus. NFIB binds to the Ezh2 promoter and overexpression of NFIB represses Ezh2 transcription. Finally, key downstream targets of EZH2-mediated epigenetic repression are misregulated in Nfib(-/-) mice. Collectively, these results suggest that the downregulation of Ezh2 transcription by NFIB is an important component of the process of neural progenitor cell differentiation during cortical development.


Asunto(s)
Corteza Cerebral/crecimiento & desarrollo , Epigénesis Genética/fisiología , Factores de Transcripción NFI/genética , Factores de Transcripción NFI/fisiología , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/fisiología , Animales , Recuento de Células , Corteza Cerebral/citología , Corteza Cerebral/fisiología , Ensayo de Cambio de Movilidad Electroforética , Proteína Potenciadora del Homólogo Zeste 2 , Femenino , Hipocampo/citología , Hipocampo/crecimiento & desarrollo , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Análisis por Micromatrices , Mutación/genética , Mutación/fisiología , Células-Madre Neurales/fisiología , Cultivo Primario de Células , Regiones Promotoras Genéticas/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
6.
Mol Imaging ; 14: 385-99, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26218510

RESUMEN

Noninvasive imaging is a critical technology for diagnosis, classification, and subsequent treatment planning for patients with glioblastoma. It has been shown that the EphA2 receptor tyrosine kinase (RTK) is overexpressed in a number of tumors, including glioblastoma. Expression levels of Eph RTKs have been linked to tumor progression, metastatic spread, and poor patient prognosis. As EphA2 is expressed at low levels in normal neural tissues, this protein represents an attractive imaging target for delineation of tumor infiltration, providing an improved platform for image-guided therapy. In this study, EphA2-4B3, a monoclonal antibody specific to human EphA2, was labeled with 64Cu through conjugation to the chelator 1,4,7-triazacyclononane-1,4,7-triacetic acid (NOTA). The resulting complex was used as a positron emission tomography (PET) tracer for the acquisition of high-resolution longitudinal PET/magnetic resonance images. EphA2-4B3-NOTA-64Cu images were qualitatively and quantitatively compared to the current clinical standards of [18F]FDOPA and gadolinium (Gd) contrast-enhanced MRI. We show that EphA2-4B3-NOTA-64Cu effectively delineates tumor boundaries in three different mouse models of glioblastoma. Tumor to brain contrast is significantly higher in EphA2-4B3-NOTA-64Cu images than in [18F]FDOPA images and Gd contrast-enhanced MRI. Furthermore, we show that nonspecific uptake in the liver and spleen can be effectively blocked by a dose of nonspecific (isotype control) IgG.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Imagen por Resonancia Magnética/métodos , Tomografía de Emisión de Positrones/métodos , Receptor EphA2/metabolismo , Animales , Anticuerpos Monoclonales/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Medios de Contraste , Glioblastoma/patología , Compuestos Heterocíclicos/metabolismo , Compuestos Heterocíclicos con 1 Anillo , Humanos , Masculino , Ratones , Persona de Mediana Edad , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Growth Factors ; 32(6): 254-64, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25410964

RESUMEN

The Eph genes are the largest sub-family of receptor tyrosine kinases; however, it is most likely the least understood and the arena for many conflicting reports. In this tribute to Prof. Martin Lackmann and Prof. Tony Pawson, we utilized The Cancer Genome Atlas resources to shed new light on the understanding of this family. We found that mutation and expression analysis define two clusters of co-expressed Eph family genes that relate to aggressive phenotypes across multiple cancer types. Analysis of signal transduction pathways using reverse-phase protein arrays revealed a network of interactions, which associates cluster-specific Eph genes with epithelial-mesenchymal transition, metabolism, DNA-damage repair and apoptosis. Our findings support the role of the Eph family in modulating cancer progression and reveal distinct patterns of Eph expression, which correlate with disease outcome. These observations provide further rationale for seeking cancer therapies, which target the Eph/ephrin system.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias/diagnóstico , Receptores de la Familia Eph/metabolismo , Animales , Biomarcadores de Tumor/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Receptores de la Familia Eph/genética
8.
Tumour Biol ; 35(2): 1459-65, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24068568

RESUMEN

Reduced levels of specific microRNA in cancer are frequently reported and associated with attenuated cancer genes and associated pathways. We previously reported a loss of miR-124a in glioblastoma (GBM) patient specimens; however, the upstream causes of this loss are largely unknown. Loss of miR-124a has been attributed to hypermethylation while other studies have shown miR-124a to be regulated by the repressor-element-1-silencing transcription factor (REST, also known as neuron-restrictive silencing factor). This current study looked at both epigenetic and transcription factor regulation as potential mechanisms resulting in the loss of miR-124a expression in GBM patient specimens and cell lines. Hypermethylation of miR-124a was observed in 82 % of GBM patient specimens (n = 56). In vitro miR-124a expression levels also increased after treatment of several patient-derived cell lines with 5-aza-2'-deoxycytidine. Additionally, we also demonstrated a positive interaction between REST activity and miR-124a using a luciferase-binding assay and we correlated the reciprocal expression of REST and miR-124a in our clinical cohort. This result indicates that miR-124a expression may also be modulated through the upstream targeting of REST. Preclinical studies involving inhibitors of REST and treatment with demethylating agents with the intent to increase miR-124a levels could be interesting.


Asunto(s)
Neoplasias Encefálicas/genética , Glioblastoma/genética , MicroARNs/genética , Proteínas Represoras/genética , Anciano , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular , Metilación de ADN/genética , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Glioblastoma/patología , Humanos , Persona de Mediana Edad
10.
Sci Transl Med ; 16(744): eadg5768, 2024 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-38657024

RESUMEN

Sepsis is a life-threatening disease caused by a dysregulated host response to infection, resulting in 11 million deaths globally each year. Vascular endothelial cell dysfunction results in the loss of endothelial barrier integrity, which contributes to sepsis-induced multiple organ failure and mortality. Erythropoietin-producing hepatocellular carcinoma (Eph) receptors and their ephrin ligands play a key role in vascular endothelial barrier disruption but are currently not a therapeutic target in sepsis. Using a cecal ligation and puncture (CLP) mouse model of sepsis, we showed that prophylactic or therapeutic treatment of mice with EphA4-Fc, a decoy receptor and pan-ephrin inhibitor, resulted in improved survival and a reduction in vascular leak, lung injury, and endothelial cell dysfunction. EphA2-/- mice also exhibited reduced mortality and pathology after CLP compared with wild-type mice. Proteomics of plasma samples from mice with sepsis after CLP revealed dysregulation of a number of Eph/ephrins, including EphA2/ephrin A1. Administration of EphA4-Fc to cultured human endothelial cells pretreated with TNF-α or ephrin-A1 prevented loss of endothelial junction proteins, specifically VE-cadherin, with maintenance of endothelial barrier integrity. In children admitted to hospital with fever and suspected infection, we observed that changes in EphA2/ephrin A1 in serum samples correlated with endothelial and organ dysfunction. Targeting Eph/ephrin signaling may be a potential therapeutic strategy to reduce sepsis-induced endothelial dysfunction and mortality.


Asunto(s)
Células Endoteliales , Efrinas , Sepsis , Transducción de Señal , Animales , Sepsis/complicaciones , Sepsis/metabolismo , Sepsis/patología , Humanos , Células Endoteliales/metabolismo , Ratones , Efrinas/metabolismo , Ratones Endogámicos C57BL , Receptores de la Familia Eph/metabolismo , Ciego/patología , Masculino , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Modelos Animales de Enfermedad
11.
Am J Pathol ; 181(5): 1493-503, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23021982

RESUMEN

The Eph/ephrin receptor-ligand system plays an important role in embryogenesis and adult life, principally by influencing cell behavior through signaling pathways, resulting in modification of the cell cytoskeleton and cell adhesion. There are 10 EphA receptors, and six EphB receptors, distinguished on sequence difference and binding preferences, that interact with the six glycosylphosphatidylinositol-linked ephrin-A ligands and the three transmembrane ephrin-B ligands, respectively. The Eph/ephrin proteins, originally described as developmental regulators that are expressed at low levels postembryonically, are re-expressed after injury to the optic nerve, spinal cord, and brain in fish, amphibians, rodents, and humans. In rodent spinal cord injury, the up-regulation of EphA4 prevents recovery by inhibiting axons from crossing the injury site. Eph/ephrin proteins may be partly responsible for the phenotypic changes to the vascular endothelium in inflammation, which allows fluid and inflammatory cells to pass from the vascular space into the interstitial tissues. Specifically, EphA2/ephrin-A1 signaling in the lung may be responsible for pulmonary inflammation in acute lung injury. A role in T-cell maturation and chronic inflammation (heart failure, inflammatory bowel disease, and rheumatoid arthritis) is also reported. Although there remains much to learn about Eph/ephrin signaling in human disease, and specifically in injury and inflammation, this area of research raises the exciting prospect that novel therapies will be developed that precisely target these pathways.


Asunto(s)
Efrinas/metabolismo , Inflamación/metabolismo , Receptores de la Familia Eph/metabolismo , Transducción de Señal , Heridas y Lesiones/metabolismo , Animales , Humanos , Ligandos
12.
J Biol Chem ; 286(32): 28181-91, 2011 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-21680732

RESUMEN

The giant cadherin FAT1 is one of four vertebrate orthologues of the Drosophila tumor suppressor fat. It engages in several functions, including cell polarity and migration, and in Hippo signaling during development. Homozygous deletions in oral cancer suggest that FAT1 may play a tumor suppressor role, although overexpression of FAT1 has been reported in some other cancers. Here we show using Northern blotting that human melanoma cell lines variably but universally express FAT1 and less commonly FAT2, FAT3, and FAT4. Both normal melanocytes and keratinocytes also express comparable FAT1 mRNA relative to melanoma cells. Analysis of the protein processing of FAT1 in keratinocytes revealed that, like Drosophila FAT, human FAT1 is cleaved into a non-covalent heterodimer before achieving cell surface expression. The use of inhibitors also established that such cleavage requires the proprotein convertase furin. However, in melanoma cells, the non-cleaved proform of FAT1 is also expressed at the cell surface together with the furin-cleaved heterodimer. Moreover, furin-independent processing generates a potentially functional proteolytic product in melanoma cells, a persistent 65-kDa membrane-bound cytoplasmic fragment no longer in association with the extracellular fragment. In vitro localization studies of FAT1 showed that melanoma cells display high levels of cytosolic FAT1 protein, whereas keratinocytes, despite comparable FAT1 expression levels, exhibited mainly cell-cell junctional staining. Such differences in protein distribution appear to reconcile with the different protein products generated by dual FAT1 processing. We suggest that the uncleaved FAT1 could promote altered signaling, and the novel products of alternate processing provide a dominant negative function in melanoma.


Asunto(s)
Cadherinas/metabolismo , Furina/metabolismo , Regulación Neoplásica de la Expresión Génica , Uniones Intercelulares/metabolismo , Melanoma/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Cadherinas/genética , Línea Celular Tumoral , Drosophila melanogaster , Furina/genética , Humanos , Uniones Intercelulares/genética , Uniones Intercelulares/patología , Queratinocitos/metabolismo , Queratinocitos/patología , Melanocitos/metabolismo , Melanocitos/patología , Melanoma/genética , Melanoma/patología , Proteínas Supresoras de Tumor/genética
13.
Biochim Biophys Acta ; 1803(11): 1298-307, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20637247

RESUMEN

CD36/FAT is a transmembrane glycoprotein that functions in the cellular uptake of long-chain fatty acids and also as a scavenger receptor. As such it plays an important role in lipid homeostasis and, pathophysiologically, in the progression of type 2 diabetes and atherosclerosis. CD36 expression is tightly regulated at the levels of both transcription and translation. Here we show that its expression and location are also regulated post-translationally, by palmitoylation. Although palmitoylation of CD36 was not required for receptor maturation and cell surface expression, inhibition of palmitoylation either pharmacologically with cerulenin or by mutation of the relevant cysteines delayed processing at the ER and trafficking through the secretory pathway. The absence of palmitoylation also reduced the half life of the CD36 protein. Additionally, the CD36 palmitoylation mutant did not incorporate efficiently into lipid rafts, a site known to be required for its function of fatty acid uptake, and this reduced the efficiency of uptake of oxidized low density lipoprotein. These findings provide an added level of sophistication where translocation of CD36 to the plasma membrane may be physiologically regulated by palmitoylation.


Asunto(s)
Antígenos CD36/metabolismo , Membrana Celular/metabolismo , Retículo Endoplásmico/metabolismo , Acilación , Alanina/genética , Alanina/metabolismo , Animales , Antígenos CD36/genética , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Cisteína/genética , Cisteína/metabolismo , Citometría de Flujo , Aparato de Golgi/metabolismo , Humanos , Immunoblotting , Lipoproteínas LDL/metabolismo , Lipoilación , Microdominios de Membrana/metabolismo , Microscopía Fluorescente , Mutación , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Procesamiento Postranscripcional del ARN
14.
Cancers (Basel) ; 13(15)2021 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-34359759

RESUMEN

We recently characterised the NUP98-HOXD13 (NHD13) mouse as a model of T-cell pre-leukaemia, featuring thymocytes that can engraft in recipient animals and progress to T-cell acute lymphoblastic leukaemia (T-ALL). However, loss of this engraftment ability by deletion of Lyl1 did not result in any loss of leukemogenesis activity. In the present study, we observe that NHD13 thymocytes overexpress EPHA3, and we characterise thymocyte behaviour in NHD13 mice with deletion of EphA3, which show a markedly reduced incidence of T-ALL. Deletion of EphA3 from the NHD13 mice does not prevent the abnormal accumulation or transplantation ability of these thymocytes. However, upon transplantation, these cells are unable to block the normal progression of recipient wild type (WT) progenitor cells through the normal developmental pathway. This is in contrast to the EphA3+/+ NHD13 thymocytes, which block the progression of incoming WT progenitors past the DN1 stage. Therefore, EphA3 is not critical for classical self-renewal, but is essential for mediating an interaction between the abnormally self-renewing cells and healthy progenitors-an interaction that results in a failure of the healthy cells to differentiate normally. We speculate that this may orchestrate a loss of healthy cell competition, which in itself has been demonstrated to be oncogenic, and that this may explain the decrease in T-ALL incidence in the absence of EphA3. We suggest that pre-leukaemic self-renewal in this model is a complex interplay of cell-intrinsic and -extrinsic factors, and that multiple redundant pathways to leukaemogenesis are active.

15.
Int J Cancer ; 126(9): 2003-11, 2010 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-20039322

RESUMEN

Eph receptors and their ephrin ligands constitute the largest subfamily of receptor tyrosine kinases and are components of the cell signaling pathways involved during development. Eph and ephrin overexpression have been documented in a variety of human cancers including gastrointestinal malignancies and in particular colorectal malignancies. EphB and ephrin B proteins have been implicated in the homeostasis of the gastrointestinal tract where EphB2- and EphB3-ephrin B signaling regulates cell sorting in the mature epithelium. These proteins are also reported to be upregulated in colon carcinomas. The EphA/ephrin A system has also been implicated in epithelial tissue structure and function. More recently, EphA receptors and their corresponding ligands have been implicated in numerous malignancies. Of these, EphA2 in particular has been intensively investigated and has been proposed as a therapeutic target. An interesting observation emerging from these studies is the role for Ephs and ephrins in critical aspects of cell adhesion, migration and positioning, and a crucial role in tumor progression and metastasis. However, the underlying role of Ephs and ephrins in these processes has generally been studied on individual Eph or ephrin genes. Given the multiplicity of Eph expression on gut epithelial cells, a more global approach is needed to define the precise role of Eph-ephrin interaction in malignant transformation. Here, we will review the recent advances on the role of Eph-ephrin signaling in colorectal malignancies.


Asunto(s)
Neoplasias Colorrectales/etiología , Efrinas/fisiología , Receptor EphA1/fisiología , Adhesión Celular , Movimiento Celular , Efrinas/análisis , Humanos , Ligandos , Receptor EphA1/análisis , Transducción de Señal
16.
Blood ; 112(3): 721-32, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18385452

RESUMEN

Signaling by Eph receptors and cell-surface ephrin ligands modulates adhesive cell properties and thereby coordinates cell movement and positioning in normal and oncogenic development. While cell contact-dependent Eph activation frequently leads to cell-cell repulsion, also the diametrically opposite response, cell-cell adhesion, is a probable outcome. However, the molecular principles regulating such disparate functions have remained controversial. We have examined cell-biologic mechanisms underlying this switch by analyzing ephrin-A5-induced cell-morphologic changes of EphA3-positive LK63 pre-B acute lymphoblastic leukemia cells. Their exposure to ephrin-A5 surfaces leads to a rapid conversion from a suspended/nonpolarized to an adherent/polarized cell type, a transition that relies on EphA3 functions operating in the absence of Eph-kinase signaling. Cell morphology change and adhesion of LK63 cells are effectively attenuated by endogenous protein tyrosine phosphatase (PTP) activity, whereby PTP inhibition and productive EphA3-phosphotyrosine signaling reverse the phenotype to nonadherent cells with a condensed cytoskeleton. Our findings suggest that Eph-associated PTP activities not only control receptor phosphorylation levels, but as a result switch the response to ephrin contact from repulsion to adhesion, which may play a role in the pathology of hematopoietic tumors.


Asunto(s)
Adhesión Celular , Efrinas/fisiología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Proteínas Tirosina Fosfatasas/metabolismo , Receptores de la Familia Eph/fisiología , Línea Celular , Línea Celular Tumoral , Polaridad Celular , Forma de la Célula , Efrina-A5/fisiología , Humanos , Fosforilación , Receptor EphA3/fisiología , Transducción de Señal
17.
Future Oncol ; 6(1): 165-76, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20021216

RESUMEN

Ephrins are cell membrane-associated signaling proteins bound by transmembrane Eph receptors on juxtaposed cells. Eph-ephrin interactions result in bidirectional signaling within both receptor- and ligand-bearing cells, with diverse consequences for cell morphology and behavior. Such interactions are especially important during early vertebrate development, and growing evidence has revealed equally important roles in adult-tissue homeostasis. As for the Eph receptors, abnormal expression of ephrins is associated with disease, especially cancer. The ephrins have received less attention than the Ephs in the literature, owing, in part, to their later discovery and that they are fewer in number. Here, we attempt to redress this imbalance and provide an 'ephrin-centric' discussion of the expression and function of ephrins in cancer.


Asunto(s)
Efrinas/metabolismo , Neoplasias/metabolismo , Receptores de la Familia Eph/metabolismo , Transducción de Señal/fisiología , Animales , Humanos
18.
ACS Cent Sci ; 6(5): 727-738, 2020 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-32490189

RESUMEN

Increasing accumulation and retention of nanomedicines within tumor tissue is a significant challenge, particularly in the case of brain tumors where access to the tumor through the vasculature is restricted by the blood-brain barrier (BBB). This makes the application of nanomedicines in neuro-oncology often considered unfeasible, with efficacy limited to regions of significant disease progression and compromised BBB. However, little is understood about how the evolving tumor-brain physiology during disease progression affects the permeability and retention of designer nanomedicines. We report here the development of a modular nanomedicine platform that, when used in conjunction with a unique model of how tumorigenesis affects BBB integrity, allows investigation of how nanomaterial properties affect uptake and retention in brain tissue. By combining different in vivo longitudinal imaging techniques (including positron emission tomography and magnetic resonance imaging), we have evaluated the retention of nanomedicines with predefined physicochemical properties (size and surface functionality) and established a relationship between structure and tissue accumulation as a function of a new parameter that measures BBB leakiness; this offers significant advancements in our ability to relate tumor accumulation of nanomedicines to more physiologically relevant parameters. Our data show that accumulation of nanomedicines in brain tumor tissue is better correlated with the leakiness of the BBB than actual tumor volume. This was evaluated by establishing brain tumors using a spontaneous and endogenously derived glioblastoma model providing a unique opportunity to assess these parameters individually and compare the results across multiple mice. We also quantitatively demonstrate that smaller nanomedicines (20 nm) can indeed cross the BBB and accumulate in tumors at earlier stages of the disease than larger analogues, therefore opening the possibility of developing patient-specific nanoparticle treatment interventions in earlier stages of the disease. Importantly, these results provide a more predictive approach for designing efficacious personalized nanomedicines based on a particular patient's condition.

19.
Cells ; 9(2)2020 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-31973233

RESUMEN

Glioblastoma (GBM) is a treatment-refractory central nervous system (CNS) tumour, and better therapies to treat this aggressive disease are urgently needed. Primary GBM models that represent the true disease state are essential to better understand disease biology and for accurate preclinical therapy assessment. We have previously presented a comprehensive transcriptome characterisation of a panel (n = 12) of primary GBM models (Q-Cell). We have now generated a systematic, quantitative, and deep proteome abundance atlas of the Q-Cell models grown in 3D culture, representing 6167 human proteins. A recent study has highlighted the degree of functional heterogeneity that coexists within individual GBM tumours, describing four cellular states (MES-like, NPC-like, OPC-like and AC-like). We performed comparative proteomic analysis, confirming a good representation of each of the four cell-states across the 13 models examined. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis identified upregulation of a number of GBM-associated cancer pathway proteins. Bioinformatics analysis, using the OncoKB database, identified a number of functional actionable targets that were either uniquely or ubiquitously expressed across the panel. This study provides an in-depth proteomic analysis of the GBM Q-Cell resource, which should prove a valuable functional dataset for future biological and preclinical investigations.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Glioblastoma/metabolismo , Glioblastoma/patología , Proteómica , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Ontología de Genes , Glioblastoma/genética , Humanos , Proteínas de Neoplasias/metabolismo , Proteoma/metabolismo
20.
Nat Commun ; 11(1): 1343, 2020 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-32165640

RESUMEN

Enlarged vestibular aqueduct (EVA) is one of the most commonly identified inner ear malformations in hearing loss patients including Pendred syndrome. While biallelic mutations of the SLC26A4 gene, encoding pendrin, causes non-syndromic hearing loss with EVA or Pendred syndrome, a considerable number of patients appear to carry mono-allelic mutation. This suggests faulty pendrin regulatory machinery results in hearing loss. Here we identify EPHA2 as another causative gene of Pendred syndrome with SLC26A4. EphA2 forms a protein complex with pendrin controlling pendrin localization, which is disrupted in some pathogenic forms of pendrin. Moreover, point mutations leading to amino acid substitution in the EPHA2 gene are identified from patients bearing mono-allelic mutation of SLC26A4. Ephrin-B2 binds to EphA2 triggering internalization with pendrin inducing EphA2 autophosphorylation weakly. The identified EphA2 mutants attenuate ephrin-B2- but not ephrin-A1-induced EphA2 internalization with pendrin. Our results uncover an unexpected role of the Eph/ephrin system in epithelial function.


Asunto(s)
Efrina-A2/genética , Bocio Nodular/genética , Pérdida Auditiva Sensorineural/genética , Transportadores de Sulfato/genética , Secuencia de Aminoácidos , Animales , Efrina-A1/genética , Efrina-A1/metabolismo , Efrina-A2/química , Efrina-A2/metabolismo , Efrina-B2/genética , Efrina-B2/metabolismo , Bocio Nodular/metabolismo , Pérdida Auditiva Sensorineural/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación Puntual , Unión Proteica , Receptor EphA2 , Transportadores de Sulfato/química , Transportadores de Sulfato/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA