Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Vascular ; 29(1): 85-91, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32588787

RESUMEN

OBJECTIVES: Somatic mosaicism of PIK3CA gene is currently recognized as the molecular driver of Klippel-Trenaunay syndrome. However, given the limitation of the current technologies, PIK3CA somatic mutations are detected only in a limited proportion of Klippel-Trenaunay syndrome cases and tissue biopsy remains an invasive high risky, sometimes life-threatening, diagnostic procedure. Next generation sequencing liquid biopsy using cell-free DNA has emerged as an innovative non-invasive approach for early detection and monitoring of cancer. This approach, overcoming the space-time profile constraint of tissue biopsies, opens a new scenario also for others diseases caused by somatic mutations. METHODS: In the present study, we performed a comprehensive analysis of seven patients (four females and three males) with Klippel-Trenaunay syndrome. Blood samples from both peripheral and efferent vein from malformation were collected and cell-free DNA was extracted from plasma. Tissue biopsies from vascular lesions were also collected when available. Cell-free DNA libraries were performed using Oncomine™ Pan-Cancer Cell-Free Assay. Ion Proton for sequencing and Ion Reporter Software for analysis were used (Life Technologies, Carlsbad, CA, USA). RESULTS: Cell-free circulating DNA analysis revealed pathogenic mutations in PIK3CA gene in all patients. The mutational load was higher in plasma obtained from the efferent vein at lesional site (0.81%) than in the peripheral vein (0.64%) leading to conclude for a causative role of the identified variants. Tissue analysis, available for one amputated patient, confirmed the presence of the mutation at the malformation site at a high molecular frequency (14-25%), confirming its causative role. CONCLUSIONS: Our data prove for the first time that the cell-free DNA-next generation sequencing-liquid biopsy, which is currently used exclusively in an oncologic setting, is indeed the most effective tool for Klippel-Trenaunay syndrome diagnosis and tailored personalized treatment.


Asunto(s)
Ácidos Nucleicos Libres de Células/genética , Fosfatidilinositol 3-Quinasa Clase I/genética , ADN/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Síndrome de Klippel-Trenaunay-Weber/diagnóstico , Mosaicismo , Mutación , Análisis de Secuencia de ADN , Adulto , Ácidos Nucleicos Libres de Células/sangre , Toma de Decisiones Clínicas , ADN/sangre , Femenino , Marcadores Genéticos , Humanos , Síndrome de Klippel-Trenaunay-Weber/sangre , Síndrome de Klippel-Trenaunay-Weber/genética , Síndrome de Klippel-Trenaunay-Weber/terapia , Biopsia Líquida , Masculino , Persona de Mediana Edad , Fenotipo , Proyectos Piloto , Valor Predictivo de las Pruebas , Pronóstico
2.
J Cell Sci ; 128(5): 863-77, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25588838

RESUMEN

Non-amoeboid cell migration is characterised by dynamic competition among multiple protrusions to establish new adhesion sites at the cell's leading edge. However, the mechanisms that regulate the decision to disassemble or to grow nascent adhesions are not fully understood. Here we show that, in endothelial cells, 3-phosphoinositide-dependent protein kinase 1 (PDK1) promotes focal adhesion (FA) turnover by controlling endocytosis of integrin αvß3 in a PI3K-dependent manner. We demonstrate that PDK1 binds and phosphorylates integrin αvß3. Downregulation of PDK1 increases FA size and slows down their disassembly. This process requires both PDK1 kinase activity and PI3K activation but does not involve Akt. Moreover, PDK1 silencing stabilises FA in membrane protrusions decreasing migration of endothelial cells on vitronectin. These results indicate that modulation of integrin endocytosis by PDK1 hampers endothelial cell adhesion and migration on extracellular matrix, thus unveiling a novel role for this kinase.


Asunto(s)
Movimiento Celular/fisiología , Endocitosis/fisiología , Adhesiones Focales/metabolismo , Integrina alfaVbeta3/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Adhesiones Focales/genética , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Integrina alfaVbeta3/genética , Fosforilación/fisiología , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora
3.
Biochim Biophys Acta ; 1856(2): 178-88, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26238471

RESUMEN

The ability of cells to migrate is essential for different physiological processes including embryonic development, angiogenesis, tissue repair and immune response. In the context of cancer such abilities acquire dramatic implications, as they are exploited by tumor cells to invade neighboring or distant healthy tissues. 3-Phosphoinositide dependent protein kinase-1 (PDK1 or PDPK1) is an ancient serine-threonine kinase belonging to AGC kinase family. An increasing amount of data points at a pivotal role for PDK1 in the regulation of cell migration. PDK1 is a transducer of PI3K signaling and activates multiple downstream effectors, thereby representing an essential hub coordinating signals coming from extracellular cues to the cytoskeletal machinery, the final executor of cell movement. Akt, PAK1, ß3 integrin, ROCK1, MRCKα and PLCγ1 are, according to the literature, the signaling transducers through which PDK1 regulates cell migration. In addition, PDK1 contributes to tumor cell invasion by regulating invadopodia formation and both amoeboid and collective cancer cell invasion. This and other pieces of evidence, such as its reported overexpression across several tumor types, corroborate a PDK1 role tumor aggressiveness. Altogether, these findings indicate the possibility to rationally target PDK1 in human tumors in order to counteract cancer cell dissemination in the organism.


Asunto(s)
Movimiento Celular/fisiología , Citoesqueleto/fisiología , Invasividad Neoplásica/patología , Invasividad Neoplásica/fisiopatología , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/fisiología , Animales , Humanos , Modelos Biológicos , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora
4.
Blood ; 121(21): e129-37, 2013 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-23471306

RESUMEN

The intrinsic complexity of the process of vessel formation limits the efficacy of cellular assays for elucidation of its molecular and pharmacologic mechanisms. We developed an ex vivo three-dimensional (3D) assay of sprouting angiogenesis with arterial explants from human umbilical cords. In this assay, human arterial rings were embedded in basement membrane extract gel, leading to a network of capillarylike structures upon vascular endothelial growth factor (VEGF) A stimulation. The angiogenic outgrowth consisted of endothelial cells, which actively internalized acetylated-low-density lipoprotein, surrounded by pericytes. Computer-assisted quantification of this vascular network demonstrated considerable sensitivity of this assay to several angiogenic inhibitors, including kinase inhibitors and monoclonal antibodies. We also performed targeted gene knockdown on this model by directly infecting explanted umbilical arteries with lentiviruses carrying short-hairpin RNA. Downregulation of VEGFR2 resulted in a significant reduction of the sprouting capability, demonstrating the relevance of human vascular explants for functional genomics studies. Furthermore, a modification of this assay led to development of a 3D model of tumor-driven angiogenesis, in which angiogenic outgrowth was sustained by spheroids of prostate cancer cells in absence of exogenous growth factors. The human arterial ring assay bridges the gap between in vitro endothelial cell and animal model, and is a powerful system for identification of genes and drugs that regulate human angiogenesis.


Asunto(s)
Aorta/citología , Técnicas de Cultivo de Célula/métodos , Neovascularización Patológica/patología , Neovascularización Patológica/fisiopatología , Neoplasias de la Próstata/patología , Arterias Umbilicales/citología , Inhibidores de la Angiogénesis/farmacología , Animales , Línea Celular Tumoral , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Imagenología Tridimensional/métodos , Lentivirus/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/tratamiento farmacológico , Neoplasias de la Próstata/irrigación sanguínea , Transducción Genética/métodos , Arterias Umbilicales/fisiología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
5.
Nat Commun ; 15(1): 7495, 2024 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-39209908

RESUMEN

The breadth and depth at which cancer models are interrogated contribute to the successful clinical translation of drug discovery efforts. In colorectal cancer (CRC), model availability is limited by a dearth of large-scale collections of patient-derived xenografts (PDXs) and paired tumoroids from metastatic disease, where experimental therapies are typically tested. Here we introduce XENTURION, an open-science resource offering a platform of 128 PDX models from patients with metastatic CRC, along with matched PDX-derived tumoroids. Multidimensional omics analyses indicate that tumoroids retain extensive molecular fidelity with parental PDXs. A tumoroid-based trial with the anti-EGFR antibody cetuximab reveals variable sensitivities that are consistent with clinical response biomarkers, mirror tumor growth changes in matched PDXs, and recapitulate EGFR genetic deletion outcomes. Inhibition of adaptive signals upregulated by EGFR blockade increases the magnitude of cetuximab response. These findings illustrate the potential of large living biobanks, providing avenues for molecularly informed preclinical research in oncology.


Asunto(s)
Cetuximab , Neoplasias Colorrectales , Receptores ErbB , Ensayos Antitumor por Modelo de Xenoinjerto , Humanos , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/tratamiento farmacológico , Animales , Cetuximab/uso terapéutico , Cetuximab/farmacología , Receptores ErbB/metabolismo , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Ratones , Femenino , Metástasis de la Neoplasia , Masculino
6.
Biochim Biophys Acta ; 1823(10): 1925-35, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22867989

RESUMEN

Blood vessel formation depends on the highly coordinated actions of a variety of angiogenic regulators. Vascular endothelial growth factor (VEGF) and Angiopoietin-1 (Ang-1) are both potent and essential proangiogenic factors with complementary roles in vascular development and function. Whereas VEGF is required for the formation of the initial vascular plexus, Ang-1 contributes to the stabilization and maturation of growing blood vessels. Here, we provide evidence of a novel microRNA (miRNA)-dependent molecular mechanism of Ang-1 signalling modulation aimed at stabilizing adult vasculature. MiRNAs are short non-coding RNA molecules that post-trascriptionally regulate gene expression by translational suppression or in some instances by cleavage of the respective mRNA target. Our data indicate that endothelial cells of mature vessels express high levels of miR-126, which primarily targets phosphoinositide-3-kinase regulatory subunit 2 (p85ß). Down-regulation of miR-126 and over-expression of p85ß in endothelial cells inhibit the biological functions of Ang-1. Additionally, knockdown of miR-126 in zebrafish resulted in vascular remodelling and maturation defects, reminiscent of the Ang-1 loss-of-function phenotype. Our findings suggest that miR-126-mediated phosphoinositide-3-kinase regulation, not only fine-tunes VEGF-signaling, but it strongly enhances the activities of Ang-1 on vessel stabilization and maturation.


Asunto(s)
Angiopoyetina 1/metabolismo , Vasos Sanguíneos/embriología , Fosfatidilinositol 3-Quinasa Clase Ia/metabolismo , MicroARNs/metabolismo , Morfogénesis , Transducción de Señal/genética , Angiopoyetina 1/genética , Animales , Vasos Sanguíneos/metabolismo , Supervivencia Celular , Regulación hacia Abajo/genética , Embrión no Mamífero/irrigación sanguínea , Embrión no Mamífero/metabolismo , Activación Enzimática , Regulación del Desarrollo de la Expresión Génica , Células HeLa , Células Endoteliales de la Vena Umbilical Humana , Humanos , Técnicas In Vitro , Ratones , MicroARNs/genética , Neovascularización Fisiológica/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo
7.
Mol Oncol ; 17(9): 1699-1725, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-36587372

RESUMEN

Metastasisation occurs through the acquisition of invasive and survival capabilities that allow tumour cells to colonise distant sites. While the role of multicellular aggregates in cancer dissemination is acknowledged, the mechanisms that drive the formation of multiclonal cell aggregates are not fully elucidated. Here, we show that cancer cells of different tissue of origins can perform collective directional migration and can actively form heteroclonal aggregates in 3D, through a proliferation-independent mechanism. Coalescence of distant cell clusters is mediated by subcellular actin-rich protrusions and multicellular outgrowths that extend towards neighbouring aggregates. Coherently, perturbation of cytoskeletal dynamics impairs collective migration while myosin II activation is necessary for multicellular movements. We put forward the hypothesis that cluster attraction is mediated by secreted soluble factors. Such a hypothesis is consistent with the abrogation of aggregation by inhibition of PI3K/AKT/mTOR and MEK/ERK, the chemoattracting activity of conditioned culture media and with a wide screening of secreted proteins. Our results present a novel collective migration model and shed light on the mechanisms of formation of heteroclonal aggregates in cancer.


Asunto(s)
Neoplasias , Fosfatidilinositol 3-Quinasas , Humanos , Movimiento Celular , Actinas/metabolismo
8.
Traffic ; 11(8): 1107-18, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20477989

RESUMEN

The bidirectional communication between integrin alphavbeta3 and vascular endothelial growth factor (VEGF) receptors acts to integrate and coordinate endothelial cell (EC) activity during angiogenesis. However, the molecular mechanisms involved in this signaling crosstalk are only partially revealed. We have found that protein kinase D1 (PKD1) was activated by VEGF-A, but not by other angiogenic factors, and associated with alphavbeta3 integrin. Moreover, knockdown of PKD1 increased endocytosis of alphavbeta3 and reduced its return from endosomes to the plasma membrane leading to accumulation of the integrin in Rab5- and Rab4-positive endosomes. Consistent with this, PKD1 knockdown caused defects in focal complex formation and reduced EC migration in response to VEGF-A. Moreover, knockdown of PKD1 reduced EC motility on vitronectin, whereas migration on collagen I was not PKD1 dependent. These results suggest that PKD1-regulated alphavbeta3 trafficking contributes to the angiogenesis process by integrating VEGF-A signaling with extracellular matrix interactions.


Asunto(s)
Movimiento Celular/fisiología , Células Endoteliales/fisiología , Integrina alfaVbeta3/metabolismo , Proteína Quinasa C/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Extensiones de la Superficie Celular/metabolismo , Células Cultivadas , Endocitosis/fisiología , Endosomas/metabolismo , Células Endoteliales/citología , Adhesiones Focales/metabolismo , Humanos , Proteína Quinasa C/genética , Transporte de Proteínas/fisiología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal/fisiología , Vitronectina/metabolismo
9.
J Cell Biol ; 176(7): 1035-47, 2007 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-17371830

RESUMEN

The serine/threonine protein kinase phosphoinositide-dependent kinase 1 (PDK1) plays a central role in cellular signaling by phosphorylating members of the AGC family of kinases, including PKB/Akt. We now present evidence showing that PDK1 is essential for the motility of vascular endothelial cells (ECs) and that it is involved in the regulation of their chemotaxis. ECs differentiated from mouse embryonic stem cells lacking PDK1 completely lost their ability to migrate in vitro in response to vascular endothelial growth factor-A (VEGF-A). In addition, PDK1(-/-) embryoid bodies exhibit evident developmental and vascular defects that can be attributed to a reduced cell migration. Moreover, the overexpression of PDK1 increased the EC migration induced by VEGF-A. We propose a model of spatial distribution of PDK1 and Akt in which the synthesis of phosphatidylinositol 3,4,5 triphosphate at plasma membrane by activation of phosphoinositide 3-kinase recruits both proteins at the leading edge of the polarized ECs and promotes cell chemotaxis. These findings establish a mechanism for the spatial localization of PDK1 and its substrate Akt to regulate directional migration.


Asunto(s)
Movimiento Celular/fisiología , Células Endoteliales/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas Dependientes de 3-Fosfoinosítido , Animales , Membrana Celular/metabolismo , Polaridad Celular/fisiología , Células Cultivadas , Quimiotaxis/fisiología , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/metabolismo , Células Endoteliales/citología , Humanos , Ratones , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología
10.
Sci Rep ; 10(1): 591, 2020 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-31953410

RESUMEN

The activation of the majority of AGC kinases is regulated by two phosphorylation events on two conserved serine/threonine residues located on the activation loop and on the hydrophobic motif, respectively. In AGC kinase family, phosphomimetic substitutions with aspartate or glutamate, leading to constitutive activation, have frequently occurred at the hydrophobic motif site. On the contrary, phosphomimetic substitutions in the activation loop are absent across the evolution of AGC kinases. This observation is explained by the failure of aspartate and glutamate to mimic phosphorylatable serine/threonine in this regulatory site. By detailed 3D structural simulations of RSK2 and further biochemical evaluation in cells, we show that the phosphomimetic residue on the activation loop fails to form a critical salt bridge with R114, necessary to reorient the αC-helix and to activate the protein. By a phylogenetic analysis, we point at a possible coevolution of a phosphorylatable activation loop and the presence of a conserved positively charged amino acid on the αC-helix. In sum, our analysis leads to the unfeasibility of phosphomimetic substitution in the activation loop of RSK and, at the same time, highlights the peculiar structural role of activation loop phosphorylation.


Asunto(s)
Sustitución de Aminoácidos , Proteínas Quinasas S6 Ribosómicas 90-kDa/química , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Secuencias de Aminoácidos , Activación Enzimática , Evolución Molecular , Células HEK293 , Células HeLa , Humanos , Modelos Moleculares , Simulación de Dinámica Molecular , Imitación Molecular , Fosforilación , Filogenia , Estructura Secundaria de Proteína , Proteínas Quinasas S6 Ribosómicas 90-kDa/genética
11.
Cells ; 8(9)2019 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-31546913

RESUMEN

Vascular physiology relies on the concerted dynamics of several cell types, including pericytes, endothelial, and vascular smooth muscle cells. The interactions between such cell types are inherently dynamic and are not easily described with static, fixed, experimental approaches. Pericytes are mural cells that support vascular development, remodeling, and homeostasis, and are involved in a number of pathological situations including cancer. The dynamic interplay between pericytes and endothelial cells is at the basis of vascular physiology and few experimental tools exist to properly describe and study it. Here we employ a previously developed ex vivo murine aortic explant to study the formation of new blood capillary-like structures close to physiological situation. We develop several mouse models to culture, identify, characterize, and follow simultaneously single endothelial cells and pericytes during angiogenesis. We employ microscopy and image analysis to dissect the interactions between cell types and the process of cellular recruitment on the newly forming vessel. We find that pericytes are recruited on the developing sprout by proliferation, migrate independently from endothelial cells, and can proliferate on the growing capillary. Our results help elucidating several relevant mechanisms of interactions between endothelial cells and pericytes.


Asunto(s)
Células Endoteliales/metabolismo , Neovascularización Fisiológica , Pericitos/metabolismo , Animales , Aorta/citología , Aorta/metabolismo , Células Endoteliales/citología , Ratones , Ratones Transgénicos , Pericitos/citología
12.
Cancer Lett ; 449: 252-262, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30790678

RESUMEN

Prostate cancer (PCa) is one of the most common cancer in men. Although hormone-sensitive PCa responds to androgen-deprivation, there are no effective therapies for castration-resistant PCa. It has been recently suggested that proton pump inhibitors (PPIs) may increase the risk of certain cancers; however, association with PCa remains elusive. Here, we evaluated the tumorigenic activities of PPIs in vitro, in PCa cell lines and epithelial cells from benign prostatic hyperplasia (BPH) and in vivo, in PCa mice xenografts. PPIs increased survival and proliferation, and inhibited apoptosis in LNCaP cells. These effects were attenuated or absent in androgen-insensitive DU-145 and PC3 cells, respectively. Specifically, omeprazole (OME) promoted cell cycle progression, increased c-Myc expression, ErbB2 activity and PSA secretion. Furthermore, OME induced the phosphorylation of MAPK-ERK1/2, PI3K/Akt and GSK-3ß, and blunted the expression and activity of cellular prostatic acid phosphatase. OME also increased survival, proliferation and PSA levels in BPH cells. In vivo, OME promoted tumor growth in mice bearing LNCaP xenografts. Our results indicate that PPIs display tumorigenic activities in PCa cells, suggesting that their long-term administration in patients should be carefully monitored.


Asunto(s)
Fosfatasa Ácida/antagonistas & inhibidores , Proliferación Celular/efectos de los fármacos , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Neoplasias Hormono-Dependientes/enzimología , Omeprazol/toxicidad , Fosfatidilinositol 3-Quinasa/metabolismo , Neoplasias de la Próstata/enzimología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Inhibidores de la Bomba de Protones/toxicidad , Receptor ErbB-2/metabolismo , Fosfatasa Ácida/metabolismo , Animales , Apoptosis/efectos de los fármacos , Humanos , Masculino , Ratones Endogámicos NOD , Ratones SCID , Neoplasias Hormono-Dependientes/patología , Células PC-3 , Fosforilación , Neoplasias de la Próstata/patología , Transducción de Señal
13.
Cell Death Dis ; 9(2): 45, 2018 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-29352118

RESUMEN

Somatic activating mutations within the PIK3CA gene have been recently detected in sporadic lymphatic and venous malformations, and in vascular malformations (VM) associated to overgrowth syndromes, such as CLOVES and Klippel-Trenaunay syndrome. Although VM are often limited to specific tissue areas and can be well treated, in extended or recurrent lesions novel therapeutic approaches are needed. We generated a mouse model of VM by local expression of PIK3CA-activating mutation in endothelial cells. PIK3CA-driven lesions are characterized by large areas of hemorrhage, hyperplastic vessels, infiltrates of inflammatory cells, and elevated endothelial cell density. Such vascular lesions are ameliorated by administration of dual PI3K/mTOR inhibitor, BEZ235, and mTOR inhibitor, Everolimus. Unexpectedly, the expression of PIK3CA-activating mutations in human endothelial cells results in both increased proliferation rates and senescence. Moreover, active forms of PIK3CA strongly promote the angiogenic sprouting. Treatment with PI3K/mTOR inhibitors restores normal endothelial cell proliferation rate and reduces the amount of senescent cells, whereas treatment with Akt inhibitor is less effective. Our findings reveal that PIK3CA mutations have a key role in the pathogenesis of VM and PIK3CA-driven experimental lesions can be effectively treated by PI3K/mTOR inhibitors.


Asunto(s)
Fosfatidilinositol 3-Quinasas/genética , Inhibidores de las Quinasa Fosfoinosítidos-3 , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Malformaciones Vasculares/genética , Animales , Bovinos , Fosfatidilinositol 3-Quinasa Clase I/genética , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Embrión de Mamíferos , Células Endoteliales , Humanos , Ratones , Ratones Transgénicos , Mutación , Fosfatidilinositol 3-Quinasas/metabolismo , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Cordón Umbilical , Malformaciones Vasculares/metabolismo , Malformaciones Vasculares/patología
14.
J Cell Biol ; 217(1): 231-249, 2018 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-29162624

RESUMEN

Extrusion of apoptotic cells from epithelial tissues requires orchestrated morphological rearrangements of the apoptotic cell and its neighbors. However, the connections between the apoptotic cascade and events leading to extrusion are not fully understood. Here, we characterize an apoptotic extrusion apical actin ring (EAAR) that is assembled within the apoptotic cell and drives epithelial extrusion. Caspase-mediated cleavage of myotonic dystrophy kinase-related CDC42-binding kinase-α (MRCKα) triggers a signaling pathway that leads to the assembly of EAAR that pulls actin bundles, resulting in the compaction and removal of the cell body. We provide a detailed portrait of the EAAR including F-actin flow, the contribution of myosin contraction, and actin polymerization at bundles' terminals when the product of MRCKα cleavage is expressed. These results add to our understanding of the mechanisms controlling the process of epithelial extrusion by establishing a causal relationship between the triggering events of apoptosis, the activation of MRCKα, and its subsequent effects on the dynamics of actomyosin cytoskeleton rearrangement.


Asunto(s)
Actomiosina/metabolismo , Apoptosis/fisiología , Caspasas/metabolismo , Células Epiteliales/metabolismo , Proteína Quinasa de Distrofia Miotónica/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Animales , Células CACO-2 , Miosinas Cardíacas/metabolismo , Línea Celular , Perros , Células HEK293 , Células HeLa , Humanos , Células de Riñón Canino Madin Darby , Centro Organizador de los Microtúbulos/fisiología , Cadenas Ligeras de Miosina/metabolismo , Miosinas/metabolismo , Transducción de Señal/fisiología , Quinasas Asociadas a rho/metabolismo
15.
Cancers (Basel) ; 9(3)2017 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-28287465

RESUMEN

Dissecting the cellular signaling that governs the motility of eukaryotic cells is one of the fundamental tasks of modern cell biology, not only because of the large number of physiological processes in which cell migration is crucial, but even more so because of the pathological ones, in particular tumor invasion and metastasis. Cell migration requires the coordination of at least four major processes: polarization of intracellular signaling, regulation of the actin cytoskeleton and membrane extension, focal adhesion and integrin signaling and contractile forces generation and rear retraction. Among the molecular components involved in the regulation of locomotion, the phosphatidylinositol-3-kinase (PI3K) pathway has been shown to exert fundamental role. A pivotal node of such pathway is represented by the serine/threonine kinase 3-phosphoinositide-dependent protein kinase-1 (PDPK1 or PDK1). PDK1, and the majority of its substrates, belong to the AGC family of kinases (related to cAMP-dependent protein kinase 1, cyclic Guanosine monophosphate-dependent protein kinase and protein kinase C), and control a plethora of cellular processes, downstream either to PI3K or to other pathways, such as RAS GTPase-MAPK (mitogen-activated protein kinase). Interestingly, PDK1 has been demonstrated to be crucial for the regulation of each step of cell migration, by activating several proteins such as protein kinase B/Akt (PKB/Akt), myotonic dystrophy-related CDC42-binding kinases alpha (MRCKα), Rho associated coiled-coil containing protein kinase 1 (ROCK1), phospholipase C gamma 1 (PLCγ1) and ß3 integrin. Moreover, PDK1 regulates cancer cell invasion as well, thus representing a possible target to prevent cancer metastasis in human patients. The aim of this review is to summarize the various mechanisms by which PDK1 controls the cell migration process, from cell polarization to actin cytoskeleton and focal adhesion regulation, and finally, to discuss the evidence supporting a role for PDK1 in cancer cell invasion and dissemination.

16.
J Drug Target ; 25(9-10): 891-898, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28817973

RESUMEN

AGMA1, a prevailingly cationic, guanidine-bearing, linear, amphoteric polyamidoamine is an effective siRNA condensing agent. Here two AGMA1 samples of different molecular weight, i.e. AGMA1-5 and AGMA1-10 were evaluated as siRNA condensing agents and transfection promoters. AGMA1-10 formed stable polyplexes with a size lower than 50 nm and positive zeta potential. AGMA1-5 polyplexes were larger, about 100 nm in size. AGMA1-10 polyplexes, but not AGMA1-5 proved to be an effective intracellular siRNA carrier, able to trigger gene silencing in Hela and PC3 cell lines without eliciting cytotoxic effects. AGMA1-10 knocked down AKT-1 expression upon transfection with an AKT-1 specific siRNA. The polyplex entry mechanism was investigated and was mediated by macropinocytosis. In conclusion, AGMA1 has potential as an efficient, non-toxic tool for the intracellular delivery of siRNA and warrants further investigation.


Asunto(s)
Agmatina/análogos & derivados , Técnicas de Transferencia de Gen , Poliaminas/administración & dosificación , ARN Interferente Pequeño/administración & dosificación , Agmatina/administración & dosificación , Agmatina/metabolismo , Citoplasma/efectos de los fármacos , Citoplasma/metabolismo , Técnicas de Transferencia de Gen/normas , Células HeLa , Humanos , Poliaminas/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo
17.
FASEB J ; 19(12): 1713-5, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16037098

RESUMEN

Thrombospondin-1 (TSP-1), a natural inhibitor of angiogenesis, acts directly on endothelial cells (EC) via CD36 to inhibit their migration and morphogenesis induced by basic fibroblast growth factor. Here we show that CD36 triggered by TSP-1 inhibits in vitro angiogenesis stimulated by vascular endothelial growth factor-A (VEGF-A). To demonstrate that the TSP-1 inhibitory signal was mediated by CD36, we transduced CD36 in CD36-deficient endothelial cells. Both TSP-1 and the agonist anti-CD36 mAb SMO, which mimics TSP-1 activity, reduced the VEGF-A165-induced migration and sprouting of CD36-ECs. To address the mechanisms by which CD36 may exert its angiostatic function, we investigated the functional components of the C-terminal cytoplasmic tail by site-directed mutagenesis. Our results indicate that C464, R467, and K469 of CD36 are required for the inhibitory activity of TSP-1. In contrast, point mutation of C466 did not alter TSP-1 ability to inhibit EC migration and sprouting. Moreover, we show that activation of CD36 by TSP-1 down-modulates the VEGF receptor-2 (VEGFR-2) and p38 mitogen-associated protein kinase phosphorylation induced by VEGF-A165, and this effect was specifically abolished by point mutation at C464. These results identify specific amino acids of the C-terminal cytoplasmic tail of CD36 crucial for the in vitro angiostatic activity of TSP-1 and extend our knowledge of regulation of VEGFR-2-mediated biological activities on ECs.


Asunto(s)
Proteínas Angiostáticas/química , Antígenos CD36/biosíntesis , Neovascularización Patológica , Secuencia de Aminoácidos , Apoptosis , Movimiento Celular , Separación Celular , Células Cultivadas , Citoplasma/metabolismo , ADN Complementario/metabolismo , Regulación hacia Abajo , Citometría de Flujo , Humanos , Immunoblotting , Inmunoprecipitación , Integrina beta1/metabolismo , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación , Fosforilación , Mutación Puntual , Estructura Terciaria de Proteína , Retroviridae/genética , Homología de Secuencia de Aminoácido , Trombospondina 1/metabolismo , Trombospondina 1/fisiología , Venas Umbilicales/citología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
18.
Oncotarget ; 7(47): 76577-76589, 2016 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-27391433

RESUMEN

BACKGROUND: Thymidylate synthase (TS), one of the key enzymes for thymidine synthesis, is a target of pemetrexed (PEM), a key agent for the systemic therapy of malignant pleural mesothelioma (MPM) and its overexpression has been correlated to PEM-resistance. In MPM, experimental data report activation of the c-SRC tyrosine kinase suggesting it as a potential target to be further investigated. RESULTS: MPM cell lines showed different sensitivity, being MSTO the most and REN the least sensitive to PEM. REN cells showed high levels of both TS and SRC: dasatinib inhibited SRC activation and suppressed TS protein expression, starting from 100 nM dose, blocking the PEM-induced up regulation of TS protein levels. Dasatinib treatment impaired cells migration, and both sequential and co-administration with PEM significantly increased apoptosis. Dasatinib pretreatment improved sensitivity to PEM, downregulated TS promoter activity and, in association with PEM, modulated the downstream PI3K-Akt-mTOR signaling. Cell lines and Methods: In three MPM cell lines (MPP89, REN and MSTO), the effects of c-SRC inhibition, in correlation with TS expression and PEM sensitivity, were evaluated. PEM and dasatinib, a SRC inhibitor, were administered as single agents, in combination or sequentially. Cell viability, apoptosis and migration, as well as TS expression and SRC activation have been assessed. CONCLUSIONS: These data indicate that dasatinib sensitizes mesothelioma cells to PEM through TS down-regulation.


Asunto(s)
Antineoplásicos/farmacología , Dasatinib/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Pemetrexed/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Interacciones Farmacológicas , Resistencia a Antineoplásicos/genética , Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Pulmonares , Mesotelioma , Mesotelioma Maligno , Fosfatidilinositol 3-Quinasas/metabolismo , Regiones Promotoras Genéticas , Biosíntesis de Proteínas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Timidilato Sintasa/genética , Timidilato Sintasa/metabolismo
19.
Methods Mol Biol ; 1214: 41-7, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25468598

RESUMEN

In vitro assays with endothelial cells (EC) cultured on three-dimensional gel recapitulate several aspects of vascular morphogenesis and pathological angiogenesis. The two most used in vitro assays of vascular morphogenesis are the tube formation on extracellular matrix gel and the sprouting from EC spheroids. Tube formation assay measures the ability of EC, plated on gel derived from reconstituted basement membrane, to form capillary-like structures. Sprouting assay is based on spheroids of EC, embedded in collagen gel and stimulated with angiogenic factors, which originate a complex network of capillary-like structures invading the gel. Both these assays can be exploited for antiangiogenic drug screening and gene function analysis during vascular morphogenesis.


Asunto(s)
Capilares/crecimiento & desarrollo , Técnicas Citológicas/métodos , Células Endoteliales de la Vena Umbilical Humana/citología , Morfogénesis , Capilares/citología , Matriz Extracelular/metabolismo , Humanos , Neovascularización Fisiológica , Esferoides Celulares/citología
20.
Sci Rep ; 5: 10206, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25976978

RESUMEN

Cellular protrusions are highly dynamic structures involved in fundamental processes, including cell migration and invasion. For a cell to migrate, its leading edge must form protrusions, and then adhere or retract. The spatial and temporal coordination of protrusions and retraction is yet to be fully understood. The study of protrusion dynamics mainly relies on live-microscopy often coupled to fluorescent labeling. Here we report the use of an alternative, label-free, quantitative and rapid assay to analyze protrusion dynamics in a cell population based on the real-time recording of cell activity by means of electronic sensors. Cells are seeded on a plate covered with electrodes and their shape changes map into measured impedance variations. Upon growth factor stimulation the impedance increases due to protrusive activity and decreases following retraction. Compared to microscopy-based methods, impedance measurements are suitable to high-throughput studies on different cell lines, growth factors and chemical compounds. We present data indicating that this assay lends itself to dissect the biochemical signaling pathways controlling adhesive protrusions. Indeed, we show that the protrusion phase is sustained by actin polymerization, directly driven by growth factor stimulation. Contraction instead mainly relies on myosin action, pointing at a pivotal role of myosin in lamellipodia retraction.


Asunto(s)
Movimiento Celular/fisiología , Extensiones de la Superficie Celular/fisiología , Impedancia Eléctrica , Seudópodos/fisiología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Línea Celular Tumoral , Cetuximab/farmacología , Factor de Crecimiento Epidérmico/metabolismo , Células HEK293 , Células HeLa , Factor de Crecimiento de Hepatocito/metabolismo , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Miosinas/antagonistas & inhibidores , Tiazolidinas/farmacología , Factor A de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA