Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cancers (Basel) ; 15(18)2023 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-37760615

RESUMEN

Tumour progression relies on interactions with untransformed cells in the tumour microenvironment (TME), including cancer-associated fibroblasts (CAFs), which promote blood supply, tumour progression, and immune evasion. Eph receptor tyrosine kinases are cell guidance receptors that are most active during development but re-emerge in cancer and are recognised drug targets. EphA3 is overexpressed in a wide range of tumour types, and we previously found expression particularly in stromal and vascular tissues of the TME. To investigate its role in the TME, we generated transgenic mice with inducible shRNA-mediated knockdown of EphA3 expression. EphA3 knockdown was confirmed in aortic mesenchymal stem cells (MSCs), which displayed reduced angiogenic capacity. In mice with syngeneic lung tumours, EphA3 knockdown reduced vasculature and CAF/MSC-like cells in tumours, and inhibited tumour growth, which was confirmed also in a melanoma model. Single cell RNA sequencing analysis of multiple human tumour types confirmed EphA3 expression in CAFs, including in breast cancer, where EphA3 was particularly prominent in perivascular- and myofibroblast-like CAFs. Our results thus indicate expression of the cell guidance receptor EphA3 in distinct CAF subpopulations is important in supporting tumour angiogenesis and tumour growth, highlighting its potential as a therapeutic target.

2.
Cancers (Basel) ; 14(13)2022 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-35804938

RESUMEN

ADAM10 is a transmembrane metalloprotease that sheds a variety of cell surface proteins, including receptors and ligands that regulate a range of developmental processes which re-emerge during tumour development. While ADAM10 is ubiquitously expressed, its activity is normally tightly regulated, but becomes deregulated in tumours. We previously reported the generation of a monoclonal antibody, 8C7, which preferentially recognises an active form of ADAM10 in human and mouse tumours. We now report our investigation of the mechanism of this specificity, and the preferential targeting of 8C7 to human tumour cell xenografts in mice. We also report the development of novel 8C7 antibody-drug conjugates that preferentially kill cells displaying the 8C7 epitope, and that can inhibit tumour growth in mice. This study provides the first demonstration that antibody-drug conjugates targeting an active conformer of ADAM10, a widely expressed transmembrane metalloprotease, enable tumour-selective targeting and inhibition.

3.
Cancer Res ; 81(4): 801-805, 2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33177063

RESUMEN

The tumor microenvironment (TME) promotes tumor development via complex intercellular signaling, aiding tumor growth and suppressing immunity. Eph receptors (Eph) and their ephrin ligands control cell interactions during normal development, and reemerge in tumors and the TME, where they are implicated in invasion, metastasis, and angiogenesis. Recent studies also indicate roles for Ephs in suppressing immune responses by controlling tumor interactions with innate and adaptive immune cells within the TME. Accordingly, inhibiting these functions can promote immune response and efficacy of immune checkpoint inhibition. This research highlights Ephs as potential targets to enhance efficacy of immune-based therapies in patients with cancer.


Asunto(s)
Tolerancia Inmunológica/fisiología , Neoplasias/inmunología , Neoplasias/patología , Receptores de la Familia Eph/fisiología , Microambiente Tumoral/inmunología , Animales , Efrinas/metabolismo , Humanos , Inmunidad/genética , Neoplasias/genética , Transducción de Señal/genética , Transducción de Señal/inmunología , Escape del Tumor/genética , Escape del Tumor/inmunología , Microambiente Tumoral/genética
4.
Pharmaceuticals (Basel) ; 13(5)2020 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-32397088

RESUMEN

The Eph subfamily of receptor tyrosine kinases mediate cell-cell communication controlling cell and tissue patterning during development. While generally less active in adult tissues, they often re-emerge in cancers, particularly on undifferentiated or progenitor cells in tumors and the tumor microenvironment, associated with tumor initiation, angiogenesis and metastasis. Eph receptors are thus attractive therapeutic targets, and monoclonal antibodies have been commonly developed and tested for anti-cancer activity in preclinical models, and in some cases in the clinic. This review summarizes 20 years of research on various antibody-based approaches to target Eph receptors in tumors and the tumor microenvironment, including their mode of action, tumor specificity, and efficacy in pre-clinical and clinical testing.

5.
J Exp Med ; 213(9): 1741-57, 2016 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-27503072

RESUMEN

The transmembrane metalloprotease ADAM10 sheds a range of cell surface proteins, including ligands and receptors of the Notch, Eph, and erbB families, thereby activating signaling pathways critical for tumor initiation and maintenance. ADAM10 is thus a promising therapeutic target. Although widely expressed, its activity is normally tightly regulated. We now report prevalence of an active form of ADAM10 in tumors compared with normal tissues, in mouse models and humans, identified by our conformation-specific antibody mAb 8C7. Structure/function experiments indicate mAb 8C7 binds an active conformation dependent on disulfide isomerization and oxidative conditions, common in tumors. Moreover, this active ADAM10 form marks cancer stem-like cells with active Notch signaling, known to mediate chemoresistance. Importantly, specific targeting of active ADAM10 with 8C7 inhibits Notch activity and tumor growth in mouse models, particularly regrowth after chemotherapy. Our results indicate targeted inhibition of active ADAM10 as a potential therapy for ADAM10-dependent tumor development and drug resistance.


Asunto(s)
Proteína ADAM10/fisiología , Neoplasias Experimentales/patología , Células Madre Neoplásicas/patología , Proteína ADAM10/antagonistas & inhibidores , Proteína ADAM10/química , Proteína ADAM17/fisiología , Secuencias de Aminoácidos , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Receptores Notch/fisiología
6.
PLoS One ; 9(11): e112106, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25420155

RESUMEN

Eph and ephrin proteins are essential cell guidance cues that orchestrate cell navigation and control cell-cell interactions during developmental tissue patterning, organogenesis and vasculogenesis. They have been extensively studied in animal models of embryogenesis and adult tissue regeneration, but less is known about their expression and function during human tissue and organ regeneration. We discovered the hypoxia inducible factor (HIF)-1α-controlled expression of EphA3, an Eph family member with critical functions during human tumour progression, in the vascularised tissue of regenerating human endometrium and on isolated human endometrial multipotent mesenchymal stromal cells (eMSCs), but not in other highly vascularised human organs. EphA3 affinity-isolation from human biopsy tissue yielded multipotent CD29+/CD73+/CD90+/CD146+ eMSCs that can be clonally propagated and respond to EphA3 agonists with EphA3 phosphorylation, cell contraction, cell-cell segregation and directed cell migration. EphA3 silencing significantly inhibited the ability of transplanted eMSCs to support neovascularisation in immunocompromised mice. In accord with established roles of Eph receptors in mediating interactions between endothelial and perivascular stromal cells during mouse development, our findings suggest that HIF-1α-controlled expression of EphA3 on human MSCs functions during the hypoxia-initiated early stages of adult blood vessel formation.


Asunto(s)
Células Madre Mesenquimatosas/metabolismo , Células Madre Multipotentes/metabolismo , Neovascularización Fisiológica , Receptor EphA3/genética , Adulto , Animales , Western Blotting , Hipoxia de la Célula , Células Cultivadas , Endometrio/citología , Femenino , Expresión Génica , Xenoinjertos/irrigación sanguínea , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Trasplante de Células Madre Mesenquimatosas/métodos , Ratones Endogámicos BALB C , Ratones Desnudos , Microscopía Fluorescente , Células Madre Multipotentes/trasplante , Interferencia de ARN , Receptor EphA3/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trasplante Heterólogo , Adulto Joven
7.
Growth Factors ; 32(6): 176-89, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25391995

RESUMEN

Eph receptor tyrosine kinases control cell-cell interactions during normal and oncogenic development, and are implicated in a range of processes including angiogenesis, stem cell maintenance and metastasis. They are thus of great interest as targets for cancer therapy. EphA3, originally isolated from leukemic and melanoma cells, is presently one of the most promising therapeutic targets, with multiple tumor-promoting roles in a variety of cancer types. This review focuses on EphA3, its functions in controlling cellular behavior, both in normal and pathological development, and most particularly in cancer.


Asunto(s)
Neoplasias/metabolismo , Receptores de la Familia Eph/metabolismo , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Humanos , Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Receptores de la Familia Eph/antagonistas & inhibidores , Receptores de la Familia Eph/química , Receptores de la Familia Eph/genética
8.
Cancer Res ; 74(16): 4470-81, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-25125683

RESUMEN

Eph receptor tyrosine kinases are critical for cell-cell communication during normal and oncogenic tissue patterning and tumor growth. Somatic mutation profiles of several cancer genomes suggest EphA3 as a tumor suppressor, but its oncogenic expression pattern and role in tumorigenesis remain largely undefined. Here, we report unexpected EphA3 overexpression within the microenvironment of a range of human cancers and mouse tumor xenografts where its activation inhibits tumor growth. EphA3 is found on mouse bone marrow-derived cells with mesenchymal and myeloid phenotypes, and activation of EphA3(+)/CD90(+)/Sca1(+) mesenchymal/stromal cells with an EphA3 agonist leads to cell contraction, cell-cell segregation, and apoptosis. Treatment of mice with an agonistic α-EphA3 antibody inhibits tumor growth by severely disrupting the integrity and function of newly formed tumor stroma and microvasculature. Our data define EphA3 as a novel target for selective ablation of the tumor microenvironment and demonstrate the potential of EphA3 agonists for anticancer therapy.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Proteínas Tirosina Quinasas Receptoras/agonistas , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Receptor EphA3/agonistas , Receptor EphA3/biosíntesis , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Transformación Celular Neoplásica , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Ratones , Ratones Desnudos , Terapia Molecular Dirigida , Proteínas Tirosina Quinasas Receptoras/inmunología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor EphA3/inmunología , Receptor EphA3/metabolismo , Transducción de Señal , Células del Estroma/efectos de los fármacos , Células del Estroma/patología , Microambiente Tumoral/efectos de los fármacos
9.
J Cell Sci ; 125(Pt 24): 6084-93, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23108669

RESUMEN

The ADAM10 transmembrane metalloprotease cleaves a variety of cell surface proteins that are important in disease, including ligands for receptor tyrosine kinases of the erbB and Eph families. ADAM10-mediated cleavage of ephrins, the ligands for Eph receptors, is suggested to control Eph/ephrin-mediated cell-cell adhesion and segregation, important during normal developmental processes, and implicated in tumour neo-angiogenesis and metastasis. We previously identified a substrate-binding pocket in the ADAM10 C domain that binds the EphA/ephrin-A complex thereby regulating ephrin cleavage. We have now generated monoclonal antibodies specifically recognising this region of ADAM10, which inhibit ephrin cleavage and Eph/ephrin-mediated cell function, including ephrin-induced Eph receptor internalisation, phosphorylation and Eph-mediated cell segregation. Our studies confirm the important role of ADAM10 in cell-cell interactions mediated by both A- and B-type Eph receptors, and suggest antibodies against the ADAM10 substrate-recognition pocket as promising therapeutic agents, acting by inhibiting cleavage of ephrins and potentially other ADAM10 substrates.


Asunto(s)
Proteínas ADAM/metabolismo , Anticuerpos Monoclonales/metabolismo , Efrinas/metabolismo , Receptores de la Familia Eph/metabolismo , Proteínas ADAM/genética , Proteínas ADAM/inmunología , Animales , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Sitios de Unión de Anticuerpos , Bovinos , Adhesión Celular , Células HEK293 , Humanos , Ratones , Unión Proteica , Transducción de Señal
10.
J Cell Biol ; 191(6): 1189-203, 2010 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-21135139

RESUMEN

Eph receptors orchestrate cell positioning during normal and oncogenic development. Their function is spatially and temporally controlled by protein tyrosine phosphatases (PTPs), but the underlying mechanisms are unclear and the identity of most regulatory PTPs are unknown. We demonstrate here that PTP1B governs signaling and biological activity of EphA3. Changes in PTP1B expression significantly affect duration and amplitude of EphA3 phosphorylation and biological function, whereas confocal fluorescence lifetime imaging microscopy (FLIM) reveals direct interactions between PTP1B and EphA3 before ligand-stimulated receptor internalization and, subsequently, on endosomes. Moreover, overexpression of wild-type (w/t) PTP1B and the [D-A] substrate-trapping mutant decelerate ephrin-induced EphA3 trafficking in a dose-dependent manner, which reveals its role in controlling EphA3 cell surface concentration. Furthermore, we provide evidence that in areas of Eph/ephrin-mediated cell-cell contacts, the EphA3-PTP1B interaction can occur directly at the plasma membrane. Our studies for the first time provide molecular, mechanistic, and functional insights into the role of PTP1B controlling Eph/ephrin-facilitated cellular interactions.


Asunto(s)
Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Receptor EphA1/metabolismo , Membrana Celular/metabolismo , Células Cultivadas , Humanos , Microscopía Confocal
11.
Am J Pathol ; 160(5): 1555-60, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-12000706

RESUMEN

Tumor development is thought to require both increased proliferation and inhibition of apoptosis. However, the relationship between cell replication and cell death in liver tumorigenesis is complex because both proliferation and apoptosis increase during hepatocarcinogenesis. To investigate the effect of the anti-apoptotic gene Bcl-2 in liver carcinogenesis, we established a line of double transgenic mice that express transforming growth factor-alpha (TGF-alpha), a liver mitogen, and Bcl-2. Double transgenic mice, TGF-alpha and Bcl-2 single transgenics, and wild type received an injection of diethylnitrosamine at 15 days of age. This alkylating agent induces liver carcinogenesis and its effect is greatly enhanced by TGF-alpha. We report that Bcl-2 expression inhibited diethylnitrosamine-induced liver carcinogenesis and counteracted the enhancing effect of TGF-alpha. Bcl-2 delayed the growth of proliferative foci at the early stages of carcinogenesis and inhibited cell proliferation in these foci. The effect of Bcl-2 on liver carcinogenesis is consistent with its reported ability to interfere with cell replication. The data demonstrate that the expression of an anti-apoptotic gene during liver carcinogenesis causes a delay rather than an increase in tumorigenesis.


Asunto(s)
Neoplasias Hepáticas/patología , Proteínas Proto-Oncogénicas c-bcl-2/fisiología , Alquilantes/administración & dosificación , Animales , Animales Recién Nacidos , División Celular/fisiología , Dietilnitrosamina/administración & dosificación , Femenino , Expresión Génica , Genotipo , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas/inducido químicamente , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-bcl-2/genética , Organismos Libres de Patógenos Específicos , Factores de Tiempo , Factor de Crecimiento Transformador alfa/genética , Factor de Crecimiento Transformador alfa/fisiología
12.
Oncogene ; 21(10): 1548-55, 2002 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-11896583

RESUMEN

Bcl-2 is the prototype of a family of genes that prevent apoptosis. However, several reports indicate that Bcl-2 may also act as a cell cycle modulator. In several human tumors, Bcl-2 expression correlates with a more favorable prognosis and lower tumor proliferative activity. We have shown that Bcl-2 expression delays liver tumor development in transgenic mice even when the gene is turned on shortly before the time of tumor development. We hypothesized that Bcl-2 may delay liver tumorigenesis by interfering with hepatocyte proliferation. To test whether Bcl-2 expression may act on hepatocyte replication we studied liver regeneration in Bcl-2 transgenic mice and wild-type littermates. DNA replication was delayed by approximately 8 h in Bcl-2 transgenic mice compared to the timing of the response in wild-type littermates. Cyclin D expression showed no alterations in the regenerating liver of Bcl-2 transgenic mice. In contrast, there was a delay in the expression of p107, cyclin E and in the activity of cyclin E/cdk 2 activity. These results show that Bcl-2 expression delays cell cycle progression in hepatocytes and suggests that it acts at a step involving cyclin E and p107.


Asunto(s)
Hepatocitos/metabolismo , Regeneración Hepática , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Animales , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/metabolismo , ADN/biosíntesis , Replicación del ADN , Hepatectomía , Inmunohistoquímica , Cinética , Masculino , Ratones , Ratones Transgénicos , Proteínas Nucleares/inmunología , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/inmunología , Proteína p107 Similar a la del Retinoblastoma
13.
Oncogene ; 21(1): 140-7, 2002 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-11791184

RESUMEN

In the present study, we examined the effects of over-expression of the potential tumor suppressor gene IGFBP-rP1/mac25 on cell-cycle kinetics in prostate cancer cells. The majority of the high expressing IGFBP-rP1/mac25 cell population was located in the G1 and sub-G0/G1 peaks; synchronizing cells in G2/M with nocodazole demonstrated the high expressing IGFBP-rP1/mac25 clones were delayed in the G1 phase of the cell cycle. Unscheduled expression of cyclin A in the sub-G0/G1 peak occurred in the IGFBP-rP1/mac25 clones. Immunoblots showed decreased cyclin D1 and p21 and increased cyclin E, p16, and p27 in the high expressing IGFBP-rP1/mac25 clones compared to the control cells. Cyclin D1/cdk-4,6 and cyclin E/cdk-2 kinase activities decreased but cyclin A/cdk-2 kinase activity increased for the high expressing IGFBP-rP1/mac25 clones compared to control cells. A pRb immunoprecipitation demonstrated more binding of E2F-1 to pRb in the high expressing IGFBP-rP1/mac25 clones than in control cells. Finally, cell senescence, as assessed by senescence-associated beta-galactosidase, demonstrated significantly more staining in the IGFBP-rP1/mac25 cells than control cells. These results suggest that IGFBP-rP1/mac25 alters the cell cycle kinetics of the M12 prostate cell line by delaying the cells in the G1 phase of the cell cycle. In addition, the appearance of cyclin A in the sub-G0/G1 phase of the cell cycle and the increased kinase activity of cyclin A/cdk-2 in the IGFBP-rP1/mac25 clones suggests that cyclin A is associated with the apoptotic cells.


Asunto(s)
Adenocarcinoma/patología , Apoptosis/genética , Proteínas Portadoras/fisiología , Ciclina A/fisiología , Proteínas de Unión al ADN , Fase G1/genética , Regulación Neoplásica de la Expresión Génica , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina , Proteínas de Neoplasias/fisiología , Neoplasias de la Próstata/patología , Adenocarcinoma/genética , Western Blotting , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/genética , Proteínas de Ciclo Celular/biosíntesis , Proteínas de Ciclo Celular/genética , División Celular , Senescencia Celular , Citoesqueleto/efectos de los fármacos , Factores de Transcripción E2F , Factor de Transcripción E2F1 , Humanos , Sustancias Macromoleculares , Masculino , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Nocodazol/farmacología , Neoplasias de la Próstata/genética , Proteínas Recombinantes de Fusión/fisiología , Proteína de Retinoblastoma/metabolismo , Factores de Transcripción/metabolismo , Transfección , Células Tumorales Cultivadas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...