Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
EMBO Mol Med ; 15(11): e18144, 2023 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-37791581

RESUMEN

Glioblastoma (GBM) remains the most malignant primary brain tumor, with a median survival rarely exceeding 2 years. Tumor heterogeneity and an immunosuppressive microenvironment are key factors contributing to the poor response rates of current therapeutic approaches. GBM-associated macrophages (GAMs) often exhibit immunosuppressive features that promote tumor progression. However, their dynamic interactions with GBM tumor cells remain poorly understood. Here, we used patient-derived GBM stem cell cultures and combined single-cell RNA sequencing of GAM-GBM co-cultures and real-time in vivo monitoring of GAM-GBM interactions in orthotopic zebrafish xenograft models to provide insight into the cellular, molecular, and spatial heterogeneity. Our analyses revealed substantial heterogeneity across GBM patients in GBM-induced GAM polarization and the ability to attract and activate GAMs-features that correlated with patient survival. Differential gene expression analysis, immunohistochemistry on original tumor samples, and knock-out experiments in zebrafish subsequently identified LGALS1 as a primary regulator of immunosuppression. Overall, our work highlights that GAM-GBM interactions can be studied in a clinically relevant way using co-cultures and avatar models, while offering new opportunities to identify promising immune-modulating targets.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Animales , Humanos , Glioblastoma/patología , Pez Cebra , Galectina 1/genética , Galectina 1/metabolismo , Galectina 1/uso terapéutico , Línea Celular Tumoral , Macrófagos/metabolismo , Neoplasias Encefálicas/patología , Microambiente Tumoral/genética
2.
Int J Mol Sci ; 23(19)2022 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-36232721

RESUMEN

During vascular development, endothelial cAMP-dependent protein kinase A (PKA) regulates angiogenesis by controlling the number of tip cells, and PKA inhibition leads to excessive angiogenesis. Whether this role of endothelial PKA is restricted to embryonic and neonatal development or is also required for vascular homeostasis later on is unknown. Here, we show that perinatal (postnatal days P1-P3) of later (P28-P32) inhibition of endothelial PKA using dominant-negative PKA expressed under the control of endothelial-specific Cdh5-CreERT2 recombinase (dnPKAiEC mice) leads to severe subcutaneous edema, hypoalbuminemia, hypoglycemia and premature death. These changes were accompanied by the local hypersprouting of blood vessels in fat pads and the secondary enlargement of subcutaneous lymphatic vessels. Most noticeably, endothelial PKA inhibition caused a dramatic disorganization of the liver vasculature. Hepatic changes correlated with decreased gluconeogenesis, while liver albumin production seems to be unaffected and hypoalbuminemia is rather a result of increased leakage into the interstitium. Interestingly, the expression of dnPKA only in lymphatics using Prox1-CreERT2 produced no phenotype. Likewise, the mosaic expression in only endothelial subpopulations using Vegfr3-CreERT2 was insufficient to induce edema or hypoglycemia. Increased expression of the tip cell marker ESM1 indicated that the inhibition of PKA induced an angiogenic response in the liver, although tissue derived pro- and anti-angiogenic factors were unchanged. These data indicate that endothelial PKA is a gatekeeper of endothelial cell activation not only in development but also in adult homeostasis, preventing the aberrant reactivation of the angiogenic program.


Asunto(s)
Vasos Sanguíneos , Subunidades Catalíticas de Proteína Quinasa Dependientes de AMP Cíclico , Células Endoteliales , Hígado , Albúminas , Animales , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/fisiología , AMP Cíclico , Subunidades Catalíticas de Proteína Quinasa Dependientes de AMP Cíclico/metabolismo , Subunidades Catalíticas de Proteína Quinasa Dependientes de AMP Cíclico/fisiología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Homeostasis , Hipoalbuminemia , Hipoglucemia , Hígado/metabolismo , Hígado/fisiología , Ratones , Recombinasas
4.
Elife ; 82019 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-31580256

RESUMEN

The cAMP-dependent protein kinase A (PKA) regulates various cellular functions in health and disease. In endothelial cells PKA activity promotes vessel maturation and limits tip cell formation. Here, we used a chemical genetic screen to identify endothelial-specific direct substrates of PKA in human umbilical vein endothelial cells (HUVEC) that may mediate these effects. Amongst several candidates, we identified ATG16L1, a regulator of autophagy, as novel target of PKA. Biochemical validation, mass spectrometry and peptide spot arrays revealed that PKA phosphorylates ATG16L1α at Ser268 and ATG16L1ß at Ser269, driving phosphorylation-dependent degradation of ATG16L1 protein. Reducing PKA activity increased ATG16L1 protein levels and endothelial autophagy. Mouse in vivo genetics and pharmacological experiments demonstrated that autophagy inhibition partially rescues vascular hypersprouting caused by PKA deficiency. Together these results indicate that endothelial PKA activity mediates a critical switch from active sprouting to quiescence in part through phosphorylation of ATG16L1, which in turn reduces endothelial autophagy.


Asunto(s)
Proteínas Relacionadas con la Autofagia/metabolismo , Autofagia , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Células Endoteliales/enzimología , Neovascularización Fisiológica , Procesamiento Proteico-Postraduccional , Animales , Línea Celular , Humanos , Ratones , Fosforilación
5.
Methods Mol Biol ; 1862: 227-243, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30315471

RESUMEN

We describe here a method for generating mouse orthotopic gliomas in order to follow their progression over time by multi-photon laser scanning microscopy. After craniotomy of the parietal bone, glioma cells are implanted in the brain cortex and a glass window is cemented atop, allowing chronical imaging of the tumor. The expression of different fluorescent proteins in tumor cells and in specific cell types of a number of currently available transgenic mouse strains allows obtaining multicolor 3D images of the tumor over time. This technique is suitable both to evaluate the effect of pharmacological treatments and to unravel basic mechanisms of tumor-host interactions.


Asunto(s)
Neoplasias Encefálicas/diagnóstico por imagen , Glioma/diagnóstico por imagen , Microscopía Intravital/métodos , Animales , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Neoplasias Encefálicas/patología , Técnicas de Cultivo de Célula/instrumentación , Técnicas de Cultivo de Célula/métodos , Línea Celular Tumoral/trasplante , Craneotomía , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Glioma/patología , Humanos , Imagenología Tridimensional/instrumentación , Imagenología Tridimensional/métodos , Microscopía Intravital/instrumentación , Proteínas Luminiscentes/química , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Microscopía Confocal/instrumentación , Microscopía Confocal/métodos , Microscopía de Fluorescencia por Excitación Multifotónica/instrumentación , Microscopía de Fluorescencia por Excitación Multifotónica/métodos , Ensayos Antitumor por Modelo de Xenoinjerto/instrumentación , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
6.
EMBO Mol Med ; 9(12): 1629-1645, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29038312

RESUMEN

Glioma growth and progression are characterized by abundant development of blood vessels that are highly aberrant and poorly functional, with detrimental consequences for drug delivery efficacy. The mechanisms driving this vessel dysmorphia during tumor progression are poorly understood. Using longitudinal intravital imaging in a mouse glioma model, we identify that dynamic sprouting and functional morphogenesis of a highly branched vessel network characterize the initial tumor growth, dramatically changing to vessel expansion, leakage, and loss of branching complexity in the later stages. This vascular phenotype transition was accompanied by recruitment of predominantly pro-inflammatory M1-like macrophages in the early stages, followed by in situ repolarization to M2-like macrophages, which produced VEGF-A and relocate to perivascular areas. A similar enrichment and perivascular accumulation of M2 versus M1 macrophages correlated with vessel dilation and malignancy in human glioma samples of different WHO malignancy grade. Targeting macrophages using anti-CSF1 treatment restored normal blood vessel patterning and function. Combination treatment with chemotherapy showed survival benefit, suggesting that targeting macrophages as the key driver of blood vessel dysmorphia in glioma progression presents opportunities to improve efficacy of chemotherapeutic agents. We propose that vessel dysfunction is not simply a general feature of tumor vessel formation, but rather an emergent property resulting from a dynamic and functional reorganization of the tumor stroma and its angiogenic influences.


Asunto(s)
Vasos Sanguíneos/patología , Neoplasias Encefálicas/patología , Glioma/patología , Animales , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos Alquilantes/uso terapéutico , Vasos Sanguíneos/anomalías , Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/mortalidad , Línea Celular Tumoral , Dacarbazina/análogos & derivados , Dacarbazina/uso terapéutico , Modelos Animales de Enfermedad , Femenino , Glioma/irrigación sanguínea , Glioma/tratamiento farmacológico , Glioma/mortalidad , Humanos , Factor Estimulante de Colonias de Macrófagos/inmunología , Macrófagos/citología , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neovascularización Patológica/patología , Fenotipo , Proteínas Proto-Oncogénicas c-sis/genética , Temozolomida , Factor A de Crecimiento Endotelial Vascular/metabolismo
7.
Cell Stem Cell ; 21(5): 679-693.e6, 2017 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-29033351

RESUMEN

To identify the cells at the origin of melanoma, we combined single-cell lineage-tracing and transcriptomics approaches with time-lapse imaging. A mouse model that recapitulates key histopathological features of human melanomagenesis was created by inducing a BRafV600E-driven melanomagenic program in tail interfollicular melanocytes. Most targeted mature, melanin-producing melanocytes expanded clonally within the epidermis before losing their differentiated features through transcriptional reprogramming and eventually invading the dermis. Tumors did not form within interscales, which contain both mature and dormant amelanotic melanocytes. The hair follicle bulge, which contains melanocyte stem cells, was also refractory to melanomagenesis. These studies identify varying tumor susceptibilities within the melanocytic lineage, highlighting pigment-producing cells as the melanoma cell of origin, and indicate that regional variation in tumor predisposition is dictated by microenvironmental cues rather than intrinsic differences in cellular origin. Critically, this work provides in vivo evidence that differentiated somatic cells can be reprogrammed into cancer initiating cells.


Asunto(s)
Desdiferenciación Celular , Melanocitos/patología , Melanoma/genética , Mutación/genética , Proteínas Proto-Oncogénicas B-raf/genética , Neoplasias Cutáneas/genética , Pigmentación de la Piel , Animales , Biomarcadores/metabolismo , Carcinogénesis/metabolismo , Carcinogénesis/patología , Proliferación Celular , Dermis/patología , Folículo Piloso/patología , Humanos , Melanocitos/metabolismo , Melanoma/patología , Ratones , Invasividad Neoplásica , Neoplasias Cutáneas/patología , Nicho de Células Madre , Cola (estructura animal) , Transcriptoma/genética , Melanoma Cutáneo Maligno
8.
Development ; 143(19): 3582-3590, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27702786

RESUMEN

cAMP-dependent protein kinase A (PKA) is a ubiquitously expressed serine/threonine kinase that regulates a variety of cellular functions. Here, we demonstrate that endothelial PKA activity is essential for vascular development, specifically regulating the transition from sprouting to stabilization of nascent vessels. Inhibition of endothelial PKA by endothelial cell-specific expression of dominant-negative PKA in mice led to perturbed vascular development, hemorrhage and embryonic lethality at mid-gestation. During perinatal retinal angiogenesis, inhibition of PKA resulted in hypersprouting as a result of increased numbers of tip cells. In zebrafish, cell autonomous PKA inhibition also increased and sustained endothelial cell motility, driving cells to become tip cells. Although these effects of PKA inhibition were highly reminiscent of Notch inhibition effects, our data demonstrate that PKA and Notch independently regulate tip and stalk cell formation and behavior.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Neovascularización Fisiológica/fisiología , Receptores Notch/metabolismo , Retina/citología , Retina/metabolismo , Animales , Movimiento Celular/genética , Movimiento Celular/fisiología , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Ratones , Ratones Mutantes , Neovascularización Fisiológica/genética , Reacción en Cadena de la Polimerasa , Transducción de Señal/genética , Transducción de Señal/fisiología , Pez Cebra
9.
Elife ; 52016 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-27074663

RESUMEN

Formation of a regularly branched blood vessel network is crucial in development and physiology. Here we show that the expression of the Notch ligand Dll4 fluctuates in individual endothelial cells within sprouting vessels in the mouse retina in vivo and in correlation with dynamic cell movement in mouse embryonic stem cell-derived sprouting assays. We also find that sprout elongation and branching associates with a highly differential phase pattern of Dll4 between endothelial cells. Stimulation with pathologically high levels of Vegf, or overexpression of Dll4, leads to Notch dependent synchronization of Dll4 fluctuations within clusters, both in vitro and in vivo. Our results demonstrate that the Vegf-Dll4/Notch feedback system normally operates to generate heterogeneity between endothelial cells driving branching, whilst synchronization drives vessel expansion. We propose that this sensitive phase transition in the behaviour of the Vegf-Dll4/Notch feedback loop underlies the morphogen function of Vegfa in vascular patterning.


Asunto(s)
Neoplasias Encefálicas/genética , Células Endoteliales/metabolismo , Glioblastoma/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Neovascularización Patológica/genética , Receptores Notch/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Proteínas de Unión al Calcio , Movimiento Celular/efectos de los fármacos , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Retroalimentación Fisiológica , Regulación de la Expresión Génica , Genes Reporteros , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Ratones , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/efectos de los fármacos , Células Madre Embrionarias de Ratones/metabolismo , Trasplante de Neoplasias , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Neovascularización Fisiológica/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Notch/genética , Retina/citología , Retina/metabolismo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/farmacología
10.
Dev Cell ; 32(1): 123-32, 2015 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-25584798

RESUMEN

During blood vessel formation, endothelial cells (ECs) establish cell-cell junctions and rearrange to form multicellular tubes. Here, we show that during lumen formation, the actin nucleator and elongation factor, formin-like 3 (fmnl3), localizes to EC junctions, where filamentous actin (F-actin) cables assemble. Fluorescent actin reporters and fluorescence recovery after photobleaching experiments in zebrafish embryos identified a pool of dynamic F-actin with high turnover at EC junctions in vessels. Knockdown of fmnl3 expression, chemical inhibition of formin function, and expression of dominant-negative fmnl3 revealed that formin activity maintains a stable F-actin content at EC junctions by continual polymerization of F-actin cables. Reduced actin polymerization leads to destabilized endothelial junctions and consequently to failure in blood vessel lumenization and lumen instability. Our findings highlight the importance of formin activity in blood vessel morphogenesis.


Asunto(s)
Actinas/metabolismo , Embrión no Mamífero/metabolismo , Endotelio Vascular/metabolismo , Proteínas de la Membrana/metabolismo , Neovascularización Fisiológica , Oligonucleótidos Antisentido/farmacología , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Citoesqueleto de Actina/metabolismo , Uniones Adherentes/fisiología , Animales , Embrión no Mamífero/citología , Endotelio Vascular/citología , Forminas , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Morfolinos/farmacología , Polimerizacion , Transducción de Señal , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo , Proteínas de Pez Cebra/antagonistas & inhibidores , Proteínas de Pez Cebra/genética
11.
J Vis Exp ; (86)2014 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-24798209

RESUMEN

Glioblastoma multiforme (GBM) is the most aggressive form of brain tumors with no curative treatments available to date. Murine models of this pathology rely on the injection of a suspension of glioma cells into the brain parenchyma following incision of the dura-mater. Whereas the cells have to be injected superficially to be accessible to intravital two-photon microscopy, superficial injections fail to recapitulate the physiopathological conditions. Indeed, escaping through the injection tract most tumor cells reach the extra-dural space where they expand abnormally fast in absence of mechanical constraints from the parenchyma. Our improvements consist not only in focally implanting a glioma spheroid rather than injecting a suspension of glioma cells in the superficial layers of the cerebral cortex but also in clogging the injection site by a cross-linked dextran gel hemi-bead that is glued to the surrounding parenchyma and sealed to dura-mater with cyanoacrylate. Altogether these measures enforce the physiological expansion and infiltration of the tumor cells inside the brain parenchyma. Craniotomy was finally closed with a glass window cemented to the skull to allow chronic imaging over weeks in absence of scar tissue development. Taking advantage of fluorescent transgenic animals grafted with fluorescent tumor cells we have shown that the dynamics of interactions occurring between glioma cells, neurons (e.g. Thy1-CFP mice) and vasculature (highlighted by an intravenous injection of a fluorescent dye) can be visualized by intravital two-photon microscopy during the progression of the disease. The possibility to image a tumor at microscopic resolution in a minimally compromised cerebral environment represents an improvement of current GBM animal models which should benefit the field of neuro-oncology and drug testing.


Asunto(s)
Neoplasias Encefálicas/patología , Glioblastoma/patología , Microscopía de Fluorescencia por Excitación Multifotónica/métodos , Animales , Procesos de Crecimiento Celular/fisiología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Ratones
12.
Nat Cell Biol ; 16(4): 309-21, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24658686

RESUMEN

Endothelial cells show surprising cell rearrangement behaviour during angiogenic sprouting; however, the underlying mechanisms and functional importance remain unclear. By combining computational modelling with experimentation, we identify that Notch/VEGFR-regulated differential dynamics of VE-cadherin junctions drive functional endothelial cell rearrangements during sprouting. We propose that continual flux in Notch signalling levels in individual cells results in differential VE-cadherin turnover and junctional-cortex protrusions, which powers differential cell movement. In cultured endothelial cells, Notch signalling quantitatively reduced junctional VE-cadherin mobility. In simulations, only differential adhesion dynamics generated long-range position changes, required for tip cell competition and stalk cell intercalation. Simulation and quantitative image analysis on VE-cadherin junctional patterning in vivo identified that differential VE-cadherin mobility is lost under pathological high VEGF conditions, in retinopathy and tumour vessels. Our results provide a mechanistic concept for how cells rearrange during normal sprouting and how rearrangement switches to generate abnormal vessels in pathologies.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Células Endoteliales/patología , Neovascularización Patológica/patología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Células Cultivadas , Simulación por Computador , Retinopatía Diabética/metabolismo , Retinopatía Diabética/patología , Células Endoteliales/metabolismo , Femenino , Humanos , Procesamiento de Imagen Asistido por Computador , Uniones Intercelulares/patología , Masculino , Ratones , Ratones Transgénicos , Neovascularización Patológica/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
13.
Development ; 140(19): 4031-40, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24046319

RESUMEN

Actin filaments are instrumental in driving processes such as migration, cytokinesis and endocytosis and provide cells with mechanical support. During angiogenesis, actin-rich filopodia protrusions have been proposed to drive endothelial tip cell functions by translating guidance cues into directional migration and mediating new contacts during anastomosis. To investigate the structural organisation, dynamics and functional importance of F-actin in endothelial cells (ECs) during angiogenesis in vivo, we generated a transgenic zebrafish line expressing Lifeact-EGFP in ECs. Live imaging identifies dynamic and transient F-actin-based structures, such as filopodia, contractile ring and cell cortex, and more persistent F-actin-based structures, such as cell junctions. For functional analysis, we used low concentrations of Latrunculin B that preferentially inhibited F-actin polymerisation in filopodia. In the absence of filopodia, ECs continued to migrate, albeit at reduced velocity. Detailed morphological analysis reveals that ECs generate lamellipodia that are sufficient to drive EC migration when filopodia formation is inhibited. Vessel guidance continues unperturbed during intersegmental vessel development in the absence of filopodia. Additionally, hypersprouting induced by loss of Dll4 and attraction of aberrant vessels towards ectopic sources of Vegfa165 can occur in the absence of endothelial filopodia protrusion. These results reveal that the induction of tip cells and the integration of endothelial guidance cues do not require filopodia. Anastomosis, however, shows regional variations in filopodia requirement, suggesting that ECs might rely on different protrusive structures depending on the nature of the environment or of angiogenic cues.


Asunto(s)
Células Endoteliales/citología , Seudópodos/metabolismo , Seudópodos/fisiología , Actinas/metabolismo , Animales , Compuestos Bicíclicos Heterocíclicos con Puentes/metabolismo , Células Endoteliales/metabolismo , Neovascularización Fisiológica/fisiología , Tiazolidinas/metabolismo , Pez Cebra , Proteínas de Pez Cebra/metabolismo
14.
PLoS One ; 8(9): e72655, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24069154

RESUMEN

The spatiotemporal and longitudinal monitoring of cellular processes occurring in tumors is critical for oncological research. We focused on glioblastoma multiforme (GBM), an untreatable highly vascularized brain tumor whose progression is thought to critically depend on the oxygen and metabolites supplied by blood vessels. We optimized protocols for orthotopic GBM grafting in mice that were able to recapitulate the biophysical constraints normally governing tumor progression and were suitable for intravital multiphoton microscopy. We repeatedly imaged tumor cells and blood vessels during GBM development. We established methods for quantitative correlative analyses of dynamic imaging data over wide fields in order to cover the entire tumor. We searched whether correlations existed between blood vessel density, tumor cell density and proliferation in control tumors. Extensive vascular remodeling and the formation of new vessels accompanied U87 tumor cell growth, but no strong correlation was found between local cell density and the extent of local blood vessel density irrespective of the tumor area or time points. The technique moreover proves useful for comparative analysis of mice subjected either to Bevacizumab anti-angiogenic treatment that targets VEGF or to AMD3100, an antagonist of CXCR4 receptor. Bevacizumab treatment massively reduced tumoral vessel densities but only transiently reduced U87 tumor growth rate. Again, there was no correlation between local blood vessel density and local cell density. Moreover, Bev applied only prior to tumor implantation inhibited tumor growth to the same extent as post-grafting treatment. AMD3100 achieved a potent inhibition of tumor growth without significant reduction in blood vessel density. These results indicate that in the brain, in this model, tumor growth can be sustained without an increase in blood vessel density and suggest that GBM growth is rather governed by stromal properties.


Asunto(s)
Glioblastoma/irrigación sanguínea , Glioblastoma/patología , Animales , Anticuerpos Monoclonales Humanizados/uso terapéutico , Bencilaminas , Bevacizumab , Línea Celular Tumoral , Ciclamas , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Compuestos Heterocíclicos/uso terapéutico , Humanos , Masculino , Ratones , Ratones Desnudos , Receptores CXCR4/antagonistas & inhibidores , Receptores CXCR4/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
15.
J Clin Invest ; 120(7): 2516-27, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20530873

RESUMEN

Tumor cell resistance to ionizing radiation and chemotherapy is a major obstacle in cancer therapy. One factor contributing to this is integrin-mediated adhesion to ECM. The adapter protein particularly interesting new cysteine-histidine-rich 1 (PINCH1) is recruited to integrin adhesion sites and promotes cell survival, but the mechanisms underlying this effect are not well understood. Here we have shown that PINCH1 is expressed at elevated levels in human tumors of diverse origins relative to normal tissue. Furthermore, PINCH1 promoted cell survival upon treatment with ionizing radiation in vitro and in vivo by perpetuating Akt1 phosphorylation and activity. Mechanistically, PINCH1 was found to directly bind to protein phosphatase 1alpha (PP1alpha) - an Akt1-regulating protein - and inhibit PP1alpha activity, resulting in increased Akt1 phosphorylation and enhanced radioresistance. Thus, our data suggest that targeting signaling molecules such as PINCH1 that function downstream of focal adhesions (the complexes that mediate tumor cell adhesion to ECM) may overcome radio- and chemoresistance, providing new therapeutic approaches for cancer.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Neoplasias/radioterapia , Proteína Fosfatasa 1/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Tolerancia a Radiación/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales , Adhesión Celular/fisiología , Supervivencia Celular , Cisteína/metabolismo , Matriz Extracelular/metabolismo , Matriz Extracelular/fisiología , Adhesiones Focales/metabolismo , Adhesiones Focales/fisiología , Histidina/metabolismo , Humanos , Integrinas/metabolismo , Proteínas con Dominio LIM , Proteínas de la Membrana , Neoplasias/metabolismo , Fosforilación
16.
J Cell Sci ; 122(Pt 11): 1800-11, 2009 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-19435803

RESUMEN

Integrin-linked kinase (ILK) and cytoplasmic adaptors of the PINCH and parvin families form a ternary complex, termed IPP, that localizes to integrin adhesions. We show here that deletion of the genes encoding ILK or PINCH1 similarly blocks maturation of focal adhesions to tensin-rich and phosphotyrosine-poor fibrillar adhesions (FBs) by downregulating expression or recruitment of tensin and destabilizing alpha5beta1-integrin-cytoskeleton linkages. As IPP components are interdependent for integrin targeting and protein stability, functional dissection of the complex was achieved by fusing ILK, PINCH, parvin or their individual motifs to the cytoplasmic tail of beta3 integrin, normally excluded from FBs. Using this novel gain-of-function approach, we demonstrated that expression of the C-terminal kinase domain of ILK can restore tensin recruitment and prompt focal-adhesion maturation in IPP-null cells. Debilitating mutations in the paxillin- or ATP-binding sites of ILK, together with alpha-parvin silencing, revealed a determinant role for ILK-parvin association, but not for direct paxillin binding, in this function. We propose a model in which the C-terminal domain of ILK promotes integrin sorting by reinforcing alpha5beta1-integrin-actin linkage and controls force transmission by targeting tensin to maturing adhesions.


Asunto(s)
Actinina/metabolismo , Proteínas de Unión al ADN/metabolismo , Adhesiones Focales/metabolismo , Proteínas de Microfilamentos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Actinina/genética , Actinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Células Cultivadas , Citoesqueleto/metabolismo , Proteínas de Unión al ADN/genética , Fibroblastos/citología , Fibroblastos/metabolismo , Silenciador del Gen , Integrina alfa5beta1/genética , Integrina alfa5beta1/metabolismo , Integrina beta3/genética , Integrina beta3/metabolismo , Proteínas con Dominio LIM , Proteínas de la Membrana , Ratones , Proteínas de Microfilamentos/genética , Complejos Multiproteicos/metabolismo , Paxillin/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Tensinas , Vinculina/metabolismo
17.
Acta Oncol ; 47(8): 1502-12, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18607838

RESUMEN

BACKGROUND: The gene encoding cortactin, CTTN (locus 11q13), an actin-binding substrate of Src kinases, is frequently amplified in breast and head and neck squamous cell carcinomas (HNSCC) and cortactin overexpression is thought to contribute in a significant way to the invasive phenotype of these tumors. Elevated Epidermal Growth Factor receptor (EGFR) expression is also commonly observed in HNSCC and has been associated with poor prognosis and resistance to cytotoxic agents, including ionizing radiation. It has been suggested that cortactin overexpression may increase EGFR levels in these tumors by affecting receptor downregulation, however we recently found by multivariate analysis, that cortactin expression status remained an independent prognostic factor for local recurrence, disease-free survival, and overall survival. MATERIAL AND METHODS: To examine the potential link between cortactin overexpression and EGFR status, we compared cortactin and EGFR levels in a series of tumor lines derived from HNSCC. RNAi-mediated silencing was performed in cortactin overexpressing cells and in vivo tumoral potential with respect to cortactin and EGFR status was analyzed. RESULTS AND DISCUSSION: Cortactin and EGFR levels were not strictly coupled in these lines and cortactin depletion did not decrease steady state receptor levels, although it did affect the epithelial to mesenchymal phenotypic conversion of cells. These results, together with clinical findings point to the existence of an EGFR-independent role of cortactin in HNSCC that may have important implications regarding the design of targeted therapies to combat tumor spread.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Cortactina/metabolismo , Receptores ErbB/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Animales , Southern Blotting , Western Blotting , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Células Cultivadas , Cortactina/genética , Receptores ErbB/genética , Femenino , Técnica del Anticuerpo Fluorescente , Amplificación de Genes , Neoplasias de Cabeza y Cuello/genética , Humanos , Técnicas para Inmunoenzimas , Riñón/metabolismo , Riñón/patología , Ratones , Ratones Desnudos , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/farmacología
18.
Genomics ; 90(2): 285-9, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17543498

RESUMEN

An essential component of microtubules, alpha-tubulin is also a multigene family in many species. An orthology-based nomenclature for this gene family has previously been difficult to assign due to incomplete genome builds and the high degree of sequence similarity between members of this family. Using the current genome builds, sequence analysis of human, mouse, and rat alpha-tubulin genes has enabled an updated nomenclature to be generated. This revised nomenclature provides a unified language for the discussion of these genes in mammalian species; it has been approved by the gene nomenclature committees of the three species and is supported by researchers in the field.


Asunto(s)
Ratones/genética , Familia de Multigenes , Ratas/genética , Terminología como Asunto , Tubulina (Proteína)/genética , Animales , ADN Complementario/metabolismo , Humanos , Filogenia
19.
J Cell Sci ; 118(Pt 24): 5899-910, 2005 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-16317048

RESUMEN

PINCH2 belongs, together with PINCH1, to a new family of focal adhesion proteins, the members of which are composed of five LIM domains. PINCH1 and PINCH2 interact, through their first LIM domain, with the integrin-linked kinase and thereby link integrins with several signal transduction pathways. Despite their high similarity, it has been shown that PINCH1 and PINCH2 could exert distinct functions during cell spreading and cell survival. To investigate the function of PINCH2 in vivo, we deleted PINCH2 in mouse using the loxP/Cre system. In contrast to the PINCH1-deficient mice, which die at the peri-implantation stage, PINCH2-null mice are viable, fertile and show no overt phenotype. Histological analysis of tissues that express high levels of PINCH2 such as bladder and kidney revealed no apparent abnormalities, but showed a significant upregulation of PINCH1, suggesting that the two PINCH proteins may have, at least in part, overlapping function in vivo. To further test this possibility, we established PINCH1-null mouse embryonic fibroblasts, which express neither PINCH1 nor PINCH2. We found that in fibroblasts with a PINCH1/2-null background, PINCH2 is able to rescue the spreading and adhesion defects of mutant fibroblasts to the same extent as PINCH1. Furthermore, we show that the LIM1 domain only of either PINCH1 or PINCH2 can prevent ILK degradation despite their failure to localize to focal adhesions. Altogether these results suggest that PINCH1 and PINCH2 share overlapping functions and operate dependently and independently of their subcellular localization.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Adhesiones Focales/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Adhesión Celular/genética , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Adhesiones Focales/genética , Riñón/citología , Riñón/metabolismo , Proteínas con Dominio LIM , Proteínas de la Membrana , Ratones , Ratones Mutantes , Especificidad de Órganos/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Terciaria de Proteína/genética , Regulación hacia Arriba , Vejiga Urinaria/citología , Vejiga Urinaria/metabolismo
20.
J Cell Sci ; 118(Pt 13): 2913-21, 2005 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-15976450

RESUMEN

PINCH1 is composed of 5 LIM domains, binds integrin-linked kinase (ILK) and locates to integrin-mediated adhesion sites. In order to investigate PINCH1 function we generated mice and embryonic stem (ES) cell-derived embryoid bodies (EBs) lacking the PINCH1 gene. Similar to mice lacking beta1 integrin or Ilk, loss of PINCH1 arrested development at the peri-implantation stage. In contrast to beta1 integrin or Ilk mutants, however, disruption of the PINCH1 gene produced implantation chambers with visible cell clumps even at embryonic day 9.5. In order to define the phenotype leading to the peri-implantation lethality we made PINCH1-null EBs and found similar but also additional defects not observed in beta1 integrin or Ilk mutant EBs. The similarities included abnormal epiblast polarity, impaired cavitation and detachment of endoderm and epiblast from basement membranes. Additional defects, which were not observed in beta1 integrin- or ILK-deficient mice or EBs, included abnormal cell-cell adhesion of endoderm and epiblast as well as the presence of apoptotic cells in the endodermal cell layer. Although ILK and PINCH1 were shown to be involved in the phosphorylation of serine-473 of PKB/Akt, immunostaining with specific antibodies revealed no apparent alteration of PKB/Akt phosphorylation in PINCH1-deficient EBs. Altogether these data demonstrate an important role of PINCH1 for integrin function, actin organization, cell-cell adhesion and endodermal cell survival during the implanting of mouse embryos.


Asunto(s)
Adhesión Celular/fisiología , Polaridad Celular/fisiología , Supervivencia Celular/fisiología , Proteínas de Unión al ADN/fisiología , Implantación del Embrión/fisiología , Matriz Extracelular/fisiología , Actinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Apoptosis/fisiología , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Matriz Extracelular/ultraestructura , Proteínas con Dominio LIM , Proteínas de la Membrana , Ratones , Datos de Secuencia Molecular , Uniones Estrechas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...