Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Am J Pathol ; 191(12): 2203-2218, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34428425

RESUMEN

Bladder cancer invasion depends on mammalian target of rapamycin complex 2 (mTORC2) activity, although the downstream mTORC2 effectors that mediate this effect have not been fully defined. One potential downstream effector is the arginine derivative nitric oxide (NO). This study identified a stage-associated increase in the expression of the NO-generating enzymes endothelial NO synthase (eNOS) and inducible NOS (iNOS) in human bladder cancer. Reduction of NOS activity by pharmacologic inhibition or silencing of NOS enzymes reduced cancer cell invasion, with similar effects observed using the NO scavenger cobinamide. By contrast, enhanced invasion was seen with the NO donor Deta-NONOate and an analog of the downstream NO second messenger cGMP. Next, NOS expression was evaluated in invadopodia, which are cellular protrusions that form the invasive tips of cancer cells. Invadopodia were enriched in both iNOS protein and mTORC2 activity, and invadopodia formation was increased by Deta-NONOate and decreased by cobinamide and ablation of mTORC2 activity. Additionally, mTORC2 increased expression of iNOS. Using a zebrafish model, injection of iNOS- or rictor-silenced cells reduced the frequency of bladder cancer cell metastasis in zebrafish. These results indicate that mTORC2 can mediate bladder cancer cell invasion through increased iNOS expression, resulting in increased NO and cGMP production in invadopodia and further propagation of invadopodia formation.


Asunto(s)
Diana Mecanicista del Complejo 2 de la Rapamicina/fisiología , Óxido Nítrico/metabolismo , Podosomas/metabolismo , Neoplasias de la Vejiga Urinaria/patología , Animales , Carcinoma de Células Transicionales/genética , Carcinoma de Células Transicionales/metabolismo , Carcinoma de Células Transicionales/patología , Embrión no Mamífero , Humanos , Invasividad Neoplásica , Metástasis de la Neoplasia , Podosomas/genética , Podosomas/patología , Células Tumorales Cultivadas , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/metabolismo , Pez Cebra/embriología
2.
Exp Cell Res ; 363(2): 271-282, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29355494

RESUMEN

Extracellular matrix (ECM) serves as a reservoir for biologically active factors, such as growth factors and proteases that influence the tumor cell behavior. ADAMTS-1 (a disintegrin and metalloprotease with thrombospondin motifs) is a secreted protease that has the ability to modify the ECM during physiological and pathological processes. Here, we analyzed the role played by ADAMTS-1 regulating HGF and TGF-ß1 activities in the high-grade fibrosarcoma cell line (HT1080). We generated HT1080 and HEK293T cells overexpressing ADAMTS-1. HT1080 cells overexpressing ADAMTS-1 (HT1080-MPA) exhibited a significant decrease in cell proliferation and migration velocity, both in presence of HGF. We obtained similar results with ADAMTS-1-enriched conditioned medium from other cell type. However, ADAMTS-1 overexpression failed to affect TGF-ß1 activity associated with HT1080 cell proliferation and migration velocity. Immunoblotting showed that ADAMTS-1 overexpression disturbs c-Met activation upon HGF stimulation. Downstream ERK1/2 and FAK signaling pathways are also influenced by this protease. Additionally, ADAMTS-1 decreased the size of the fibrosarcospheres, both under normal conditions and in the presence of HGF. Likewise, in presence of HGF, ADAMTS-1 overexpression in HT1080 disrupted microtumors formation in vivo. These microtumors, including individual cells, presented characteristics of non-invasive lesions (rounded morphology). Our results suggest that ADAMTS-1 is involved in regulating HGF-related functions on fibrosarcoma cells. This protease may then represent an endogenous mechanism in controlling the bioavailability of different growth factors that have a direct influence on tumor cell behavior.


Asunto(s)
Proteína ADAMTS1/metabolismo , Proliferación Celular/efectos de los fármacos , Factor de Crecimiento de Hepatocito/farmacología , Proteínas Proto-Oncogénicas c-met/metabolismo , Línea Celular Tumoral , Movimiento Celular/fisiología , Matriz Extracelular/metabolismo , Fibrosarcoma/tratamiento farmacológico , Fibrosarcoma/patología , Células HEK293 , Humanos , Proteína Quinasa 3 Activada por Mitógenos/metabolismo
3.
J Cell Sci ; 126(Pt 4): 904-13, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23321642

RESUMEN

Breast cancer and melanoma cells commonly metastasize to the brain using homing mechanisms that are poorly understood. Cancer patients with brain metastases display poor prognosis and survival due to the lack of effective therapeutics and treatment strategies. Recent work using intravital microscopy and preclinical animal models indicates that metastatic cells colonize the brain, specifically in close contact with the existing brain vasculature. However, it is not known how contact with the vascular niche promotes microtumor formation. Here, we investigate the role of connexins in mediating early events in brain colonization using transparent zebrafish and chicken embryo models of brain metastasis. We provide evidence that breast cancer and melanoma cells utilize connexin gap junction proteins (Cx43, Cx26) to initiate brain metastatic lesion formation in association with the vasculature. RNAi depletion of connexins or pharmacological blocking of connexin-mediated cell-cell communication with carbenoxolone inhibited brain colonization by blocking tumor cell extravasation and blood vessel co-option. Activation of the metastatic gene twist in breast cancer cells increased Cx43 protein expression and gap junction communication, leading to increased extravasation, blood vessel co-option and brain colonization. Conversely, inhibiting twist activity reduced Cx43-mediated gap junction coupling and brain colonization. Database analyses of patient histories revealed increased expression of Cx26 and Cx43 in primary melanoma and breast cancer tumors, respectively, which correlated with increased cancer recurrence and metastasis. Together, our data indicate that Cx43 and Cx26 mediate cancer cell metastasis to the brain and suggest that connexins might be exploited therapeutically to benefit cancer patients with metastatic disease.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/secundario , Neoplasias de la Mama/complicaciones , Neoplasias de la Mama/metabolismo , Conexinas/metabolismo , Melanoma/complicaciones , Melanoma/metabolismo , Animales , Neoplasias Encefálicas/genética , Neoplasias de la Mama/genética , Embrión de Pollo , Conexina 26 , Conexina 43/genética , Conexina 43/metabolismo , Conexinas/genética , Femenino , Humanos , Melanoma/genética , Ratones , Ratones Desnudos , Metástasis de la Neoplasia/genética , Interferencia de ARN
4.
J Biol Chem ; 288(1): 123-31, 2013 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-23105102

RESUMEN

Pseudopodium-enriched atypical kinase 1 (PEAK1) is a recently described tyrosine kinase that associates with the actin cytoskeleton and focal adhesion (FA) in migrating cells. PEAK1 is known to promote cell migration, but the responsible mechanisms remain unclear. Here, we show that PEAK1 controls FA assembly and disassembly in a dynamic pathway controlled by PEAK1 phosphorylation at Tyr-665. Knockdown of endogenous PEAK1 inhibits random cell migration. In PEAK1-deficient cells, FA lifetimes are decreased, FA assembly times are shortened, and FA disassembly times are extended. Phosphorylation of Tyr-665 in PEAK1 is essential for normal PEAK1 localization and its function in the regulation of FAs; however, constitutive phosphorylation of PEAK1 Tyr-665 is also disruptive of its function, indicating a requirement for precise spatiotemporal regulation of PEAK1. Src family kinases are required for normal PEAK1 localization and function. Finally, we provide evidence that PEAK1 promotes cancer cell invasion through Matrigel by a mechanism that requires dynamic regulation of Tyr-665 phosphorylation.


Asunto(s)
Adhesiones Focales/química , Regulación de la Expresión Génica , Proteínas Tirosina Quinasas/química , Tirosina/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Colágeno/química , Combinación de Medicamentos , Humanos , Laminina/química , Paxillin/metabolismo , Fosforilación , Proteoglicanos/química , Factores de Tiempo , Familia-src Quinasas/metabolismo
5.
Proc Natl Acad Sci U S A ; 107(24): 10920-5, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20534451

RESUMEN

Regulation of the actin-myosin cytoskeleton plays a central role in cell migration and cancer progression. Here, we report the discovery of a cytoskeleton-associated kinase, pseudopodium-enriched atypical kinase 1 (PEAK1). PEAK1 is a 190-kDa nonreceptor tyrosine kinase that localizes to actin filaments and focal adhesions. PEAK1 undergoes Src-induced tyrosine phosphorylation, regulates the p130Cas-Crk-paxillin and Erk signaling pathways, and operates downstream of integrin and epidermal growth factor receptors (EGFR) to control cell spreading, migration, and proliferation. Perturbation of PEAK1 levels in cancer cells alters anchorage-independent growth and tumor progression in mice. Notably, primary and metastatic samples from colon cancer patients display amplified PEAK1 levels in 81% of the cases. Our findings indicate that PEAK1 is an important cytoskeletal regulatory kinase and possible target for anticancer therapy.


Asunto(s)
Citoesqueleto/metabolismo , Neoplasias/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Seudópodos/metabolismo , Actinas/metabolismo , Animales , Secuencia de Bases , Línea Celular , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Neoplasias del Colon/metabolismo , Biología Computacional , Cartilla de ADN/genética , Femenino , Adhesiones Focales/metabolismo , Humanos , Técnicas In Vitro , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/secundario , Ratones , Ratones Desnudos , Neoplasias/etiología , Neoplasias Pancreáticas/metabolismo , Fosfotirosina/metabolismo , Proteínas Tirosina Quinasas/genética , Proteómica , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Familia-src Quinasas/metabolismo
6.
Proc Natl Acad Sci U S A ; 107(9): 4299-304, 2010 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-20154271

RESUMEN

Kinases are known to regulate fundamental processes in cancer including tumor proliferation, metastasis, neovascularization, and chemoresistance. Accordingly, kinase inhibitors have been a major focus of drug development, and several kinase inhibitors are now approved for various cancer indications. Typically, kinase inhibitors are selected via high-throughput screening using catalytic kinase domains at low ATP concentration, and this process often yields ATP mimetics that lack specificity and/or function poorly in cells where ATP levels are high. Molecules targeting the allosteric site in the inactive kinase conformation (type II inhibitors) provide an alternative for developing selective inhibitors that are physiologically active. By applying a rational design approach using a constrained amino-triazole scaffold predicted to stabilize kinases in the inactive state, we generated a series of selective type II inhibitors of PDGFRbeta and B-RAF, important targets for pericyte recruitment and endothelial cell survival, respectively. These molecules were designed in silico and screened for antivascular activity in both cell-based models and a Tg(fli1-EGFP) zebrafish embryogenesis model. Dual inhibition of PDGFRbeta and B-RAF cellular signaling demonstrated synergistic antiangiogenic activity in both zebrafish and murine models of angiogenesis, and a combination of previously characterized PDGFRbeta and RAF inhibitors validated the synergy. Our lead compound was selected as an orally active molecule with favorable pharmacokinetic properties which demonstrated target inhibition in vivo leading to suppression of murine orthotopic tumors in both the kidney and pancreas.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Carcinoma de Células Renales/patología , División Celular/efectos de los fármacos , Neoplasias Renales/patología , Neovascularización Patológica , Neoplasias Pancreáticas/patología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Administración Oral , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/metabolismo , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Pez Cebra
7.
J Biol Chem ; 286(20): 18190-201, 2011 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-21454597

RESUMEN

Brain development and spinal cord regeneration require neurite sprouting and growth cone navigation in response to extension and collapsing factors present in the extracellular environment. These external guidance cues control neurite growth cone extension and retraction processes through intracellular protein phosphorylation of numerous cytoskeletal, adhesion, and polarity complex signaling proteins. However, the complex kinase/substrate signaling networks that mediate neuritogenesis have not been investigated. Here, we compare the neurite phosphoproteome under growth and retraction conditions using neurite purification methodology combined with mass spectrometry. More than 4000 non-redundant phosphorylation sites from 1883 proteins have been annotated and mapped to signaling pathways that control kinase/phosphatase networks, cytoskeleton remodeling, and axon/dendrite specification. Comprehensive informatics and functional studies revealed a compartmentalized ERK activation/deactivation cytoskeletal switch that governs neurite growth and retraction, respectively. Our findings provide the first system-wide analysis of the phosphoprotein signaling networks that enable neurite growth and retraction and reveal an important molecular switch that governs neuritogenesis.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas del Tejido Nervioso/metabolismo , Neuritas/metabolismo , Fosfoproteínas/metabolismo , Línea Celular , Quinasas MAP Reguladas por Señal Extracelular/genética , Humanos , Proteínas del Tejido Nervioso/genética , Fosfoproteínas/genética
8.
Blood ; 116(25): 5773-83, 2010 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-20826718

RESUMEN

Angiogenesis is controlled by signals that stimulate motility in endothelial cells at the tips of vascular sprouts while maintaining cell-cell adhesion in the stalks of angiogenic sprouts. We show here that Gs-linked G protein-coupled receptor activation of cAMP-dependent protein kinase (PKA) plays an important role in regulating the switch between endothelial cell adhesion and migration by activating C-terminal Src kinase, leading to inhibition of pp60Src. Activated PKA blocks pp60Src-dependent vascular endot helial-cadherin phosphorylation, thereby stimulating cell-cell adhesion while suppressing endothelial cell polarization, motility, angiogenesis, and vascular permeability. Similar to the actions of Notch and Dll4, PKA activation blocks sprouting in newly forming embryonic blood vessels, while PKA inhibition promotes excessive sprouting in these vessels. These findings demonstrate that G protein-coupled receptors and PKA regulate vascular sprouting during angiogenesis by controlling endothelial cell migration and cell-cell adhesion through their actions on pp60Src.


Asunto(s)
Adhesión Celular/fisiología , Movimiento Celular/fisiología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Neovascularización Fisiológica , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Transducción de Señal , Western Blotting , Proteína Tirosina Quinasa CSK , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Humanos , Inmunoprecipitación , Fosforilación , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas pp60(c-src)/antagonistas & inhibidores , Proteínas Proto-Oncogénicas pp60(c-src)/genética , Venas Umbilicales/citología , Venas Umbilicales/metabolismo , Familia-src Quinasas
9.
Nature ; 440(7087): 1069-72, 2006 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-16547516

RESUMEN

Rho family GTPases regulate the actin and adhesion dynamics that control cell migration. Current models postulate that Rac promotes membrane protrusion at the leading edge and that RhoA regulates contractility in the cell body. However, there is evidence that RhoA also regulates membrane protrusion. Here we use a fluorescent biosensor, based on a novel design preserving reversible membrane interactions, to visualize the spatiotemporal dynamics of RhoA activity during cell migration. In randomly migrating cells, RhoA activity is concentrated in a sharp band directly at the edge of protrusions. It is observed sporadically in retracting tails, and is low in the cell body. RhoA activity is also associated with peripheral ruffles and pinocytic vesicles, but not with dorsal ruffles induced by platelet-derived growth factor (PDGF). In contrast to randomly migrating cells, PDGF-induced membrane protrusions have low RhoA activity, potentially because PDGF strongly activates Rac, which has previously been shown to antagonize RhoA activity. Our data therefore show that different extracellular cues induce distinct patterns of RhoA signalling during membrane protrusion.


Asunto(s)
Movimiento Celular , Extensiones de la Superficie Celular/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Células 3T3 , Animales , Técnicas Biosensibles , Extensiones de la Superficie Celular/efectos de los fármacos , Fibroblastos , Ratones , Pinocitosis , Factor de Crecimiento Derivado de Plaquetas/farmacología , Factores de Tiempo , Proteínas de Unión al GTP rac/metabolismo
10.
Nat Biomed Eng ; 6(7): 882-897, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-34931077

RESUMEN

Targeting the delivery of therapeutics specifically to diseased tissue enhances their efficacy and decreases their side effects. Here we show that mesenchymal stromal cells with their nuclei removed by density-gradient centrifugation following the genetic modification of the cells for their display of chemoattractant receptors and endothelial-cell-binding molecules are effective vehicles for the targeted delivery of therapeutics. The enucleated cells neither proliferate nor permanently engraft in the host, yet retain the organelles for energy and protein production, undergo integrin-regulated adhesion to inflamed endothelial cells, and actively home to chemokine gradients established by diseased tissues. In mouse models of acute inflammation and of pancreatitis, systemically administered enucleated cells expressing two types of chemokine receptor and an endothelial adhesion molecule enhanced the delivery of an anti-inflammatory cytokine to diseased tissue (with respect to unmodified stromal cells and to exosomes derived from bone-marrow-derived stromal cells), attenuating inflammation and ameliorating disease pathology. Enucleated cells retain most of the cells' functionality, yet acquire the cargo-carrying characteristics of cell-free delivery systems, and hence represent a versatile delivery vehicle and therapeutic system.


Asunto(s)
Sistemas de Liberación de Medicamentos , Células Madre Mesenquimatosas , Animales , Quimiocinas/metabolismo , Citocinas/metabolismo , Células Endoteliales/metabolismo , Humanos , Inflamación/metabolismo , Ratones
11.
Proteomics ; 11(10): 2019-26, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21500348

RESUMEN

In this study, we evaluated a concatenated low pH (pH 3) and high pH (pH 10) reversed-phase liquid chromatography strategy as a first dimension for two-dimensional liquid chromatography tandem mass spectrometry ("shotgun") proteomic analysis of trypsin-digested human MCF10A cell sample. Compared with the more traditional strong cation exchange method, the use of concatenated high pH reversed-phase liquid chromatography as a first-dimension fractionation strategy resulted in 1.8- and 1.6-fold increases in the number of peptide and protein identifications (with two or more unique peptides), respectively. In addition to broader identifications, advantages of the concatenated high pH fractionation approach include improved protein sequence coverage, simplified sample processing, and reduced sample losses. The results demonstrate that the concatenated high pH reversed-phased strategy is an attractive alternative to strong cation exchange for two-dimensional shotgun proteomic analysis.


Asunto(s)
Mama/química , Cromatografía de Fase Inversa/métodos , Fragmentos de Péptidos/aislamiento & purificación , Mapeo Peptídico/métodos , Proteoma/química , Acetonitrilos/química , Mama/citología , Mama/metabolismo , Línea Celular , Análisis por Conglomerados , Células Epiteliales/química , Células Epiteliales/citología , Células Epiteliales/metabolismo , Formiatos/química , Humanos , Concentración de Iones de Hidrógeno , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Proteoma/metabolismo , Proteómica/métodos , Espectrometría de Masas en Tándem/métodos , Tripsina/metabolismo , Urea/química
12.
Circ Res ; 104(8): 952-60, 2009 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-19265037

RESUMEN

Lipid accumulation in arteries induces vascular inflammation and atherosclerosis, the major cause of heart attack and stroke in humans. Extreme hyperlipidemia induced in mice and rabbits enables modeling many aspects of human atherosclerosis, but microscopic examination of plaques is possible only postmortem. Here we report that feeding adult zebrafish (Danio rerio) a high-cholesterol diet (HCD) resulted in hypercholesterolemia, remarkable lipoprotein oxidation, and fatty streak formation in the arteries. Feeding an HCD supplemented with a fluorescent cholesteryl ester to optically transparent fli1:EGFP zebrafish larvae in which endothelial cells express green fluorescent protein (GFP), and using confocal microscopy enabled monitoring vascular lipid accumulation and the endothelial cell layer disorganization and thickening in a live animal. The HCD feeding also increased leakage of a fluorescent dextran from the blood vessels. Administering ezetimibe significantly diminished the HCD-induced endothelial cell layer thickening and improved its barrier function. Feeding HCD to lyz:DsRed2 larvae in which macrophages and granulocytes express DsRed resulted in the accumulation of fluorescent myeloid cells in the vascular wall. Using a fluorogenic substrate for phospholipase A(2) (PLA(2)), we observed an increased vascular PLA(2) activity in live HCD-fed larvae compared to control larvae. Furthermore, by transplanting genetically modified murine cells into HCD-fed larvae, we demonstrated that toll-like receptor-4 was required for efficient in vivo lipid uptake by macrophages. These results suggest that the novel zebrafish model is suitable for studying temporal characteristics of certain inflammatory processes of early atherogenesis and the in vivo function of vascular cells.


Asunto(s)
Aterosclerosis/metabolismo , Endotelio Vascular/metabolismo , Hipercolesterolemia/metabolismo , Metabolismo de los Lípidos , Lipoproteínas/metabolismo , Macrófagos/metabolismo , Pez Cebra/metabolismo , Factores de Edad , Envejecimiento/metabolismo , Animales , Animales Modificados Genéticamente , Anticolesterolemiantes/farmacología , Aterosclerosis/etiología , Aterosclerosis/patología , Azetidinas/farmacología , Línea Celular , Colesterol en la Dieta/administración & dosificación , Modelos Animales de Enfermedad , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Ezetimiba , Femenino , Proteínas Fluorescentes Verdes/genética , Humanos , Hipercolesterolemia/etiología , Hipercolesterolemia/patología , Larva/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Lipoproteínas/sangre , Proteínas Luminiscentes/genética , Macrófagos/trasplante , Masculino , Ratones , Microscopía Confocal , Oxidación-Reducción , Permeabilidad , Fosfolipasas A2/metabolismo , Factores de Tiempo , Receptor Toll-Like 4/metabolismo , Pez Cebra/embriología , Pez Cebra/genética
13.
Proc Natl Acad Sci U S A ; 105(7): 2313-8, 2008 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-18268346

RESUMEN

Screening for novel anticancer drugs in chemical libraries isolated from marine organisms, we identified the lipopeptide somocystinamide A (ScA) as a pluripotent inhibitor of angiogenesis and tumor cell proliferation. The antiproliferative activity was largely attributable to induction of programmed cell death. Sensitivity to ScA was significantly increased among cells expressing caspase 8, whereas siRNA knockdown of caspase 8 increased survival after exposure to ScA. ScA rapidly and efficiently partitioned into liposomes while retaining full antiproliferative activity. Consistent with the induction of apoptosis via the lipid compartment, we noted accumulation and aggregation of ceramide in treated cells and subsequent colocalization with caspase 8. Angiogenic endothelial cells were extremely sensitive to ScA. Picomolar concentrations of ScA disrupted proliferation and endothelial tubule formation in vitro. Systemic treatment of zebrafish or local treatment of the chick chorioallantoic membrane with ScA resulted in dose-dependent inhibition of angiogenesis, whereas topical treatment blocked tumor growth among caspase-8-expressing tumors. Together, the results reveal an unexpected mechanism of action for this unique lipopeptide and suggest future development of this and similar agents as antiangiogenesis and anticancer drugs.


Asunto(s)
Apoptosis/efectos de los fármacos , Caspasa 8/metabolismo , Disulfuros/farmacología , Lipoproteínas/farmacología , Inhibidores de la Angiogénesis/farmacología , Animales , Animales Modificados Genéticamente , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Pollos , Disulfuros/química , Embrión no Mamífero/irrigación sanguínea , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/embriología , Humanos , Estructura Molecular , Océanos y Mares , Fosfolípidos/metabolismo , Sensibilidad y Especificidad
14.
Proc Natl Acad Sci U S A ; 105(6): 1931-6, 2008 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-18245386

RESUMEN

Neurite extension and growth cone navigation are guided by extracellular cues that control cytoskeletal rearrangements. However, understanding the complex signaling mechanisms that mediate neuritogenesis has been limited by the inability to biochemically separate the neurite and soma for spatial proteomic and bioinformatic analyses. Here, we apply global proteome profiling in combination with a neurite purification methodology for comparative analysis of the soma and neurite proteomes of neuroblastoma cells. The spatial relationship of 4,855 proteins were mapped, revealing networks of signaling proteins that control integrins, the actin cytoskeleton, and axonal guidance in the extending neurite. Bioinformatics and functional analyses revealed a spatially compartmentalized Rac/Cdc42 signaling network that operates in conjunction with multiple guanine-nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs) to control neurite formation. Interestingly, RNA interference experiments revealed that the different GEFs and GAPs regulate specialized functions during neurite formation, including neurite growth and retraction kinetics, cytoskeletal organization, and cell polarity. Our findings provide insight into the spatial organization of signaling networks that enable neuritogenesis and provide a comprehensive system-wide profile of proteins that mediate this process, including those that control Rac and Cdc42 signaling.


Asunto(s)
Neuritas , Proteoma , Proteína de Unión al GTP cdc42/fisiología , Proteínas de Unión al GTP rac/fisiología , Línea Celular Tumoral , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Interferencia de ARN , Transducción de Señal , Proteínas de Unión al GTP rho/metabolismo
15.
Front Oncol ; 11: 697626, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34422650

RESUMEN

Angiogenesis is the formation of new vessels from pre-existing vasculature. The heparan sulfate chains from endothelial cell proteoglycans interact with the major angiogenic factors, regulating blood vessels´ formation. Since the FDA´s first approval, anti-angiogenic therapy has shown tumor progression inhibition and increased patient survival. Previous work in our group has selected an HS-binding peptide using a phage display system. Therefore, we investigated the effect of the selected peptide in angiogenesis and tumor progression. The HS-binding peptide showed a higher affinity for heparin N-sulfated. The HS-binding peptide was able to inhibit the proliferation of human endothelial umbilical cord cells (HUVEC) by modulation of FGF-2. It was verified a significant decrease in the tube formation of human endothelial cells and capillary formation of mice aorta treated with HS-binding peptide. HS-binding peptide also inhibited the formation of sub-intestinal blood vessels in zebrafish embryos. Additionally, in zebrafish embryos, the tumor size decreased after treatment with HS-binding peptide.

16.
J Cell Biol ; 156(4): 725-36, 2002 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-11839772

RESUMEN

Initiation of cell migration requires morphological polarization with formation of a dominant leading pseudopodium and rear compartment. A molecular understanding of this process has been limited, due to the inability to biochemically separate the leading pseudopodium from the rear of the cell. Here we examine the spatio-temporal localization and activation of cytoskeletal-associated signals in purified pseudopodia directed to undergo growth or retraction. Pseudopodia growth requires assembly of a p130Crk-associated substrate (CAS)/c-CrkII (Crk) scaffold, which facilitates translocation and activation of Rac1. Interestingly, Rac1 activation then serves as a positive-feedback loop to maintain CAS/Crk coupling and pseudopodia extension. Conversely, disassembly of this molecular scaffold is critical for export and down regulation of Rac1 activity and induction of pseudopodia retraction. Surprisingly, the uncoupling of Crk from CAS during pseudopodium retraction is independent of changes in focal adhesion kinase activity and CAS tyrosine phosphorylation. These findings establish CAS/Crk as an essential scaffold for Rac1-mediated pseudopodia growth and retraction, and illustrate spatio-temporal segregation of cytoskeletal signals during cell polarization.


Asunto(s)
Fosfoproteínas/metabolismo , Proteínas Quinasas/metabolismo , Proteínas , Proteínas Proto-Oncogénicas/metabolismo , Seudópodos/metabolismo , Ubiquitina-Proteína Ligasas , Proteína de Unión al GTP rac1/metabolismo , Células 3T3 , Animales , Células COS , Polaridad Celular , Factores Quimiotácticos/farmacología , Quimiotaxis/fisiología , Chlorocebus aethiops , Proteína Sustrato Asociada a CrK , Activación Enzimática , Lisofosfolípidos/farmacología , Ratones , Proteínas Proto-Oncogénicas c-cbl , Proteínas Proto-Oncogénicas c-crk , Seudópodos/efectos de los fármacos , Seudópodos/fisiología , Proteína p130 Similar a la del Retinoblastoma , Proteína de Unión al GTP cdc42/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Proteínas de Unión al GTP rho/metabolismo
17.
J Cell Biol ; 165(3): 421-32, 2004 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-15138294

RESUMEN

Large-scale proteomic and functional analysis of isolated pseudopodia revealed the Lim, actin, and SH3 domain protein (Lasp-1) as a novel protein necessary for cell migration, but not adhesion to, the extracellular matrix (ECM). Lasp-1 is a ubiquitously expressed actin-binding protein with a unique domain configuration containing SH3 and LIM domains, and is overexpressed in 8-12% of human breast cancers. We find that stimulation of nonmotile and quiescent cells with growth factors or ECM proteins facilitates Lasp-1 relocalization from the cell periphery to the leading edge of the pseudopodium, where it associates with nascent focal complexes and areas of actin polymerization. Interestingly, although Lasp-1 dynamics in migratory cells occur independently of c-Abl kinase activity and tyrosine phosphorylation, c-Abl activation by apoptotic agents specifically promotes phosphorylation of Lasp-1 at tyrosine 171, which is associated with the loss of Lasp-1 localization to focal adhesions and induction of cell death. Thus, Lasp-1 is a dynamic focal adhesion protein necessary for cell migration and survival in response to growth factors and ECM proteins.


Asunto(s)
Movimiento Celular/genética , Adhesiones Focales/metabolismo , Proteínas de Homeodominio/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Seudópodos/metabolismo , Actinas/biosíntesis , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Carcinoma/genética , Carcinoma/metabolismo , Supervivencia Celular/genética , Proteínas del Citoesqueleto , Proteínas de la Matriz Extracelular/metabolismo , Proteínas de la Matriz Extracelular/farmacología , Quinasa 1 de Adhesión Focal , Proteína-Tirosina Quinasas de Adhesión Focal , Adhesiones Focales/genética , Sustancias de Crecimiento/metabolismo , Sustancias de Crecimiento/farmacología , Proteínas de Homeodominio/genética , Proteínas con Dominio LIM , Ratones , Células 3T3 NIH , Metástasis de la Neoplasia/genética , Proteínas de Neoplasias/genética , Fosforilación , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/genética , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas c-abl/metabolismo , Seudópodos/genética
18.
Sci STKE ; 2007(400): pl4, 2007 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-17712138

RESUMEN

Directional cell migration requires the formation of a dominant pseudopodium in the direction toward which the cell migrates. When a migratory cell is stimulated with a chemoattractant or extracellular matrix (ECM) gradient, it responds with localized amplification of signals on the side facing the gradient. The signals mediate reorganization of the actin-myosin cytoskeleton, leading to morphological polarization of the cell and pseudopodium extension. To identify these signals, we developed an approach to biochemically isolate the pseudopodium from the cell body using 3.0-micrometer porous filters for large-scale quantitative proteomic and phosphoproteomic analysis. Here, we detail the methodology for pseudopodium purification and proteomic analysis. This model system should be widely applicable for the analysis of the pseudopodium proteome from various migratory cell lines, including primary and cancer cell lines stimulated with a diverse array of chemoattractants, ECM proteins, or both.


Asunto(s)
Proteoma/análisis , Proteómica/métodos , Seudópodos/metabolismo , Animales , Western Blotting , Células COS , Movimiento Celular/efectos de los fármacos , Polaridad Celular/efectos de los fármacos , Factores Quimiotácticos/farmacología , Quimiotaxis/efectos de los fármacos , Chlorocebus aethiops , Cromatografía Liquida , Citoesqueleto/metabolismo , Electroforesis en Gel de Poliacrilamida , Ratones , Microscopía Fluorescente , Células 3T3 NIH , Seudópodos/efectos de los fármacos , Seudópodos/fisiología , Transducción de Señal/efectos de los fármacos , Espectrometría de Masas en Tándem , Tripsina/metabolismo
19.
Mol Biol Cell ; 16(1): 84-96, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15525681

RESUMEN

Although it is known that the spatial coordination of Rac and Rho activity is essential for cell migration, the molecular mechanisms regulating these GTPases during migration are unknown. We found that the expression of constitutively activated R-Ras (38V) blocked membrane protrusion and random migration. In contrast, expression of dominant negative R-Ras (41A) enhanced migrational persistence and membrane protrusion. Endogenous R-Ras is necessary for cell migration, as cells that were transfected with siRNA for R-Ras did not migrate. Expression of R-Ras (38V) decreased Rac activity and increased Rho activity around the entire cell periphery, whereas expression of dominant negative R-Ras (41A) showed the converse, suggesting that R-Ras can spatially activate Rho and inactivate Rac. Consistent with this role, endogenous R-Ras localized and was preferentially activated at the leading edge of migratory cells in response to adhesion. The effects of R-Ras on cell migration are mediated by PI3-Kinase, as an effector mutant that uncouples PI3-Kinase binding from R-Ras (38V) rescued migration. From these data, we hypothesize that R-Ras plays a key role in cell migration by locally regulating the switch from Rac to Rho activity after membrane protrusion and adhesion.


Asunto(s)
Membrana Celular/metabolismo , GTP Fosfohidrolasas/fisiología , Proteínas de Unión al GTP rac/metabolismo , Proteínas ras/fisiología , Proteínas de Unión al GTP rho/metabolismo , Adhesión Celular , Línea Celular Tumoral , Movimiento Celular , Colágeno/metabolismo , Relación Dosis-Respuesta a Droga , Genes Dominantes , Humanos , Microscopía Fluorescente , Microscopía por Video , Modelos Biológicos , Mutación , Fosfatidilinositol 3-Quinasas/metabolismo , ARN Interferente Pequeño/metabolismo , Factores de Tiempo , Transfección
20.
Cancer Res ; 78(6): 1444-1456, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29321164

RESUMEN

There remains intense interest in tractable approaches to target or silence the KRAS oncoprotein as a rational therapeutic strategy to attack pancreatic ductal adenocarcinoma (PDAC) and other cancers that overexpress it. Here we provide evidence that accumulation of the KRAS oncoprotein is controlled by a self-regulating feed-forward regulatory loop that utilizes a unique hypusinated isoform of the translation elongation factor eIF5A and the tyrosine kinase PEAK1. Oncogenic activation of KRAS increased eIF5A-PEAK1 translational signaling, which in turn facilitated increased KRAS protein synthesis. Mechanistic investigations show that this feed-forward positive regulatory pathway was controlled by oncogenic KRAS-driven metabolic demands, operated independently of canonical mTOR signaling, and did not involve new KRAS gene transcription. Perturbing eIF5A-PEAK1 signaling, by genetic or pharmacologic strategies or by blocking glutamine synthesis, was sufficient to inhibit expression of KRAS, eIF5A, and PEAK1, to attenuate cancer cell growth and migration, and to block tumor formation in established preclinical mouse models of PDAC. Levels of KRAS, eIF5A, and PEAK1 protein increased during cancer progression with the highest levels of expression observed in metastatic cell populations. Combinatorial targeting of eIF5A hypusination and the RAS-ERK signaling pathway cooperated to attenuate KRAS expression and its downstream signaling along with cell growth in vitro and tumor formation in vivo Collectively, our findings highlight a new mechanistic strategy to attenuate KRAS expression as a therapeutic strategy to target PDAC and other human cancers driven by KRAS activation.Significance: These findings highlight a new mechanistic strategy to attenuate KRAS expression as a therapeutic strategy to target human cancers driven by KRAS activation. Cancer Res; 78(6); 1444-56. ©2018 AACR.


Asunto(s)
Carcinoma Ductal Pancreático/metabolismo , Neoplasias Pancreáticas/metabolismo , Factores de Iniciación de Péptidos/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Retroalimentación Fisiológica , Femenino , GTP Fosfohidrolasas/metabolismo , Glutamina/metabolismo , Humanos , Lisina/análogos & derivados , Lisina/metabolismo , Proteínas de la Membrana/metabolismo , Ratones Desnudos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Factores de Iniciación de Péptidos/genética , Proteínas Tirosina Quinasas/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas de Unión al ARN/genética , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto , Factor 5A Eucariótico de Iniciación de Traducción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA