Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Blood ; 130(23): 2548-2558, 2017 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-28899852

RESUMEN

Hemostasis in vertebrates involves both a cellular and a protein component. Previous studies in jawless vertebrates (cyclostomes) suggest that the protein response, which involves thrombin-catalyzed conversion of a soluble plasma protein, fibrinogen, into a polymeric fibrin clot, is conserved in all vertebrates. However, similar data are lacking for the cellular response, which in gnathostomes is regulated by von Willebrand factor (VWF), a glycoprotein that mediates the adhesion of platelets to the subendothelial matrix of injured blood vessels. To gain evolutionary insights into the cellular phase of coagulation, we asked whether a functional vwf gene is present in the Atlantic hagfish, Myxine glutinosa We found a single vwf transcript that encodes a simpler protein compared with higher vertebrates, the most striking difference being the absence of an A3 domain, which otherwise binds collagen under high-flow conditions. Immunohistochemical analyses of hagfish tissues and blood revealed Vwf expression in endothelial cells and thrombocytes. Electron microscopic studies of hagfish tissues demonstrated the presence of Weibel-Palade bodies in the endothelium. Hagfish Vwf formed high-molecular-weight multimers in hagfish plasma and in stably transfected CHO cells. In functional assays, botrocetin promoted VWF-dependent thrombocyte aggregation. A search for vwf sequences in the genome of sea squirts, the closest invertebrate relatives of hagfish, failed to reveal evidence of an intact vwf gene. Together, our findings suggest that VWF evolved in the ancestral vertebrate following the divergence of the urochordates some 500 million years ago and that it acquired increasing complexity though sequential insertion of functional modules.


Asunto(s)
Anguila Babosa , Factor de von Willebrand/genética , Factor de von Willebrand/metabolismo , Proteína ADAMTS13/metabolismo , Secuencia de Aminoácidos , Animales , Células CHO , Clonación Molecular , Cricetulus , ADN Complementario , Endotelio Vascular/metabolismo , Evolución Molecular , Expresión Génica , Homeostasis , Humanos , Modelos Moleculares , Agregación Plaquetaria , Conformación Proteica , Dominios Proteicos , Pliegue de Proteína , Multimerización de Proteína , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Proteolisis , Relación Estructura-Actividad , Vertebrados , Cuerpos de Weibel-Palade/metabolismo , Cuerpos de Weibel-Palade/ultraestructura , Factor de von Willebrand/química
2.
Am J Pathol ; 187(10): 2337-2347, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28736316

RESUMEN

Tumors induce their heterogeneous vasculature by secreting vascular endothelial growth factor (VEGF)-A. Anti-VEGF/VEGF receptor (VEGFR) drugs treat cancer, but the underlying mechanisms remain unclear. An adenovirus expressing VEGF-A (Ad-VEGF-A164) replicates the tumor vasculature in mice without tumor cells. Mother vessels (MV) are the first angiogenic vessel type to form in tumors and after Ad-VEGF-A164. Multiday treatments with a VEGF trap reverted MV back to normal microvessels. We now show that, within hours, a single dose of several anti-VEGF drugs collapsed MV to form glomeruloid microvascular proliferations (GMP), accompanied by only modest endothelial cell death. GMP, common in many human cancers but of uncertain origin, served as an intermediary step in MV reversion to normal microvessels. The vasodisruptive drug combretastatin CA4 also targeted MV selectively but acted differently, extensively killing MV endothelium. Antivascular changes were quantified with a novel Evans blue dye assay that measured vascular volumes. As in tumors, Ad-VEGF-A164 strikingly increased endothelial nitric oxide synthase (eNOS) expression. The eNOS inhibitor N(G)-Nitro-l-arginine methyl ester mimicked anti-VEGF/VEGFR drugs, rapidly collapsing MV to GMP. Inhibition of eNOS reduces synthesis of its vasodilatory product, nitric oxide, leading to arterial contraction. Patients and mice receiving anti-VEGF/VEGFR drugs develop hypertension, reflecting systemic arterial contraction. Together, anti-VEGF/VEGFR drugs act in part by inhibiting eNOS, causing vasocontraction, MV collapse to GMP, and subsequent reversion of GMP to normal microvessels, all without extensive vascular killing.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Vasos Sanguíneos/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Adenoviridae/metabolismo , Animales , Bibencilos/farmacología , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Hipertensión/patología , Ratones Endogámicos C57BL , Ratones Desnudos , Microvasos/efectos de los fármacos , Microvasos/patología , Modelos Biológicos , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular , Proteínas Recombinantes de Fusión/farmacología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
3.
Biochem Biophys Res Commun ; 465(3): 338-43, 2015 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-26241677

RESUMEN

Transmembrane-4 L-six family member-1 (TM4SF1) is a small plasma membrane-associated glycoprotein that is highly and selectively expressed on the plasma membranes of tumor cells, cultured endothelial cells, and, in vivo, on tumor-associated endothelium. Immunofluorescence microscopy also demonstrated TM4SF1 in cytoplasm and, tentatively, within nuclei. With monoclonal antibody 8G4, and the finer resolution afforded by immuno-nanogold transmission electron microscopy, we now demonstrate TM4SF1 in uncoated cytoplasmic vesicles, nuclear pores and nucleoplasm. Because of its prominent surface location on tumor cells and tumor-associated endothelium, TM4SF1 has potential as a dual therapeutic target using an antibody drug conjugate (ADC) approach. For ADC to be successful, antibodies reacting with cell surface antigens must be internalized for delivery of associated toxins to intracellular targets. We now report that 8G4 is efficiently taken up into cultured endothelial cells by uncoated vesicles in a dynamin-dependent, clathrin-independent manner. It is then transported along microtubules through the cytoplasm and passes through nuclear pores into the nucleus. These findings validate TM4SF1 as an attractive candidate for cancer therapy with antibody-bound toxins that have the capacity to react with either cytoplasmic or nuclear targets in tumor cells or tumor-associated vascular endothelium.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Antígenos de Superficie/inmunología , Clatrina/inmunología , Dinaminas/inmunología , Células Endoteliales/inmunología , Proteínas de Neoplasias/inmunología , Fracciones Subcelulares/inmunología , Células Cultivadas , Células Endoteliales/ultraestructura , Humanos
4.
Blood ; 121(11): 2074-83, 2013 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-23303825

RESUMEN

Eosinophils release their granule proteins extracellularly through exocytosis, piecemeal degranulation, or cytolytic degranulation. Findings in diverse human eosinophilic diseases of intact extracellular eosinophil granules, either free or clustered, indicate that eosinophil cytolysis occurs in vivo, but the mechanisms and consequences of lytic eosinophil degranulation are poorly understood. We demonstrate that activated human eosinophils can undergo extracellular DNA trap cell death (ETosis) that cytolytically releases free eosinophil granules. Eosinophil ETosis (EETosis), in response to immobilized immunoglobulins (IgG, IgA), cytokines with platelet activating factor, calcium ionophore, or phorbol myristate acetate, develops within 120 minutes in a reduced NADP (NADPH) oxidase-dependent manner. Initially, nuclear lobular formation is lost and some granules are released by budding off from the cell as plasma membrane-enveloped clusters. Following nuclear chromatolysis, plasma membrane lysis liberates DNA that forms weblike extracellular DNA nets and releases free intact granules. EETosis-released eosinophil granules, still retaining eosinophil cationic granule proteins, can be activated to secrete when stimulated with CC chemokine ligand 11 (eotaxin-1). Our results indicate that an active NADPH oxidase-dependent mechanism of cytolytic, nonapoptotic eosinophil death initiates nuclear chromatolysis that eventuates in the release of intact secretion-competent granules and the formation of extracellular DNA nets.


Asunto(s)
Degranulación de la Célula , ADN/metabolismo , Eosinófilos/metabolismo , Eosinófilos/fisiología , Vesículas Secretoras/metabolismo , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Muerte Celular/fisiología , Degranulación de la Célula/efectos de los fármacos , Degranulación de la Célula/fisiología , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Células Cultivadas , Quimiocina CCL11/farmacología , Relación Dosis-Respuesta a Droga , Eosinófilos/efectos de los fármacos , Exocitosis/efectos de los fármacos , Exocitosis/fisiología , Espacio Extracelular/efectos de los fármacos , Espacio Extracelular/genética , Espacio Extracelular/metabolismo , Humanos , Vesículas Secretoras/efectos de los fármacos
5.
J Immunol ; 191(6): 2871-8, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23945142

RESUMEN

Mast cell (MC) activation through the high-affinity IgE receptor FcεRI leads to the release of mediators involved in immediate-type allergic reactions. Although Abs against the tetraspanins CD63 and CD81 inhibit FcεRI-induced MC degranulation, the intrinsic role of these molecules in FcεRI-induced MC activation is unknown. In MCs, CD63 is expressed at the cell surface and in lysosomes (particularly secretory lysosomes that contain allergic mediators). In this study, we investigated the role of CD63 in MC using a CD63 knockout mouse model. CD63-deficiency did not affect in vivo MC numbers and tissue distribution. Bone marrow-derived MC developed normally in the absence of CD63 protein. However, CD63-deficient bone marrow-derived MC showed a significant decrease in FcεRI-mediated degranulation, but not PMA/ionomycin-induced degranulation, as shown by ß-hexosaminidase release assays. The secretion of TNF-α, which is both released from granules and synthesized de novo upon MC activation, was also decreased. IL-6 secretion and production of the lipid mediator leukotriene C4 were unaffected. There were no ultrastructural differences in granule content and morphology, late endosomal/lysosomal marker expression, FcεRI-induced global tyrosine phosphorylation, and Akt phosphorylation. Finally, local reconstitution in genetically MC-deficient Kit(w/w-v) mice was unaffected by the absence of CD63. However, the sites reconstituted with CD63-deficient MC developed significantly attenuated cutaneous anaphylactic reactions. These findings demonstrate that the absence of CD63 results in a significant decrease of MC degranulation, which translates into a reduction of acute allergic reactions in vivo, thus identifying CD63 as an important component of allergic inflammation.


Asunto(s)
Anafilaxia/inmunología , Degranulación de la Célula/inmunología , Mastocitos/inmunología , Tetraspanina 30/inmunología , Traslado Adoptivo , Anafilaxia/metabolismo , Animales , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente Indirecta , Immunoblotting , Inmunoglobulina E/inmunología , Mastocitos/metabolismo , Ratones , Ratones Noqueados , Microscopía Confocal , Microscopía Electrónica de Transmisión , Tetraspanina 30/metabolismo
6.
Angiogenesis ; 17(4): 897-907, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24986520

RESUMEN

Transmembrane-4 L-six family member-1 (TM4SF1) is a small plasma membrane glycoprotein that regulates cell motility and proliferation. TM4SF1 is an attractive cancer target because of its high expression in both tumor cells and on the vascular endothelial cells lining tumor blood vessels. We generated mouse monoclonal antibodies against human TM4SF1 in order to evaluate their therapeutic potential; 13 of the antibodies we generated reacted with extracellular loop-2 (EL2), TM4SF1's larger extracellular, lumen-facing domain. However, none of these antibodies reacted with mouse TM4SF1, likely because the EL2 of mouse TM4SF1 differs significantly from that of its human counterpart. Therefore, to test our antibodies in vivo, we employed an established model of engineered human vessels in which human endothelial colony-forming cells (ECFC) and human mesenchymal stem cells (MSC) are incorporated into Matrigel plugs that are implanted subcutaneously in immunodeficient nude mice. We modified the original protocol by (1) preculturing human ECFC on laminin, fibronectin, and collagen-coated plates, and (2) increasing the ECFC/MSC ratio. These modifications significantly increased the human vascular network in Matrigel implants. Two injections of one of our anti-TM4SF1 EL2 monoclonal antibodies, 8G4, effectively eliminated the human vascular component present in these plugs; they also abrogated human PC3 prostate cancer cells that were incorporated into the ECFC/MSC Matrigel mix. Together, these studies provide a mouse model for assessing tumor xenografts that are supplied by a human vascular network and demonstrate that anti-TM4SF1 antibodies such as 8G4 hold promise for cancer therapy.


Asunto(s)
Antígenos de Superficie/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/química , Línea Celular Tumoral , Perfilación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana , Humanos , Células Madre Mesenquimatosas , Ratones , Ratones Desnudos , Datos de Secuencia Molecular , Trasplante de Neoplasias , Neovascularización Patológica , Ingeniería de Tejidos/métodos , Transcripción Genética
7.
PLoS Pathog ; 8(7): e1002729, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22792061

RESUMEN

Phagocytosis of invading microorganisms by specialized cells such as macrophages and neutrophils is a key component of the innate immune response. These cells capture and engulf pathogens and subsequently destroy them in intracellular vacuoles-the phagosomes. Pathogen phagocytosis and progression and maturation of pathogen-containing phagosomes, a crucial event to acquire microbicidal features, occurs in parallel with accentuated formation of lipid-rich organelles, termed lipid bodies (LBs), or lipid droplets. Experimental and clinical infections with different pathogens such as bacteria, parasites, and viruses induce LB accumulation in cells from the immune system. Within these cells, LBs synthesize and store inflammatory mediators and are considered structural markers of inflammation. In addition to LB accumulation, interaction of these organelles with pathogen-containing phagosomes has increasingly been recognized in response to infections and may have implications in the outcome or survival of the microorganism within host cells. In this review, we summarize our current knowledge on the LB-phagosome interaction within cells from the immune system, with emphasis on macrophages, and discuss the functional meaning of this event during infectious diseases.


Asunto(s)
Infecciones , Macrófagos , Neutrófilos/inmunología , Fagosomas/inmunología , Animales , Bacterias/inmunología , Humanos , Inmunidad Innata , Cuerpos de Inclusión/inmunología , Infecciones/inmunología , Infecciones/microbiología , Infecciones/parasitología , Inflamación/inmunología , Inflamación/microbiología , Inflamación/parasitología , Mediadores de Inflamación , Macrófagos/inmunología , Macrófagos/microbiología , Macrófagos/parasitología , Ratones , Fagocitosis
8.
Blood ; 120(11): 2167-73, 2012 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-22674805

RESUMEN

VEGF induces vascular permeability (VP) in ischemic diseases and cancer, leading to many pathophysiological consequences. The molecular mechanisms by which VEGF acts to induce hyperpermeability are poorly understood and in vivo models that easily facilitate real-time, genetic studies of VP do not exist. In the present study, we report a heat-inducible VEGF transgenic zebrafish (Danio rerio) model through which VP can be monitored in real time. Using this approach with morpholino-mediated gene knock-down and knockout mice, we describe a novel role of phospholipase Cß3 as a negative regulator of VEGF-mediated VP by regulating intracellular Ca2+ release. Our results suggest an important effect of PLCß3 on VP and provide a new model with which to identify genetic regulators of VP crucial to several disease processes.


Asunto(s)
Permeabilidad Capilar , Endotelio Vascular/metabolismo , Fosfolipasa C beta/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Animales Modificados Genéticamente , Señalización del Calcio/efectos de los fármacos , Permeabilidad Capilar/efectos de los fármacos , Células Cultivadas , Embrión no Mamífero , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Respuesta al Choque Térmico , Ensayos Analíticos de Alto Rendimiento , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Ratones , Ratones Noqueados , Morfolinos/farmacología , Fosfolipasa C beta/antagonistas & inhibidores , Fosfolipasa C beta/genética , Regiones Promotoras Genéticas/efectos de los fármacos , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/genética , Pez Cebra , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
9.
J Immunol ; 188(8): 3686-99, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22442443

RESUMEN

Adaptive immunity requires that T cells efficiently scan diverse cell surfaces to identify cognate Ag. However, the basic cellular mechanisms remain unclear. In this study, we investigated this process using vascular endothelial cells, APCs that possess a unique and extremely advantageous, planar morphology. High-resolution imaging revealed that CD4 memory/effector T cells dynamically probe the endothelium by extending submicron-scale, actin-rich "invadosome/podosome-like protrusions" (ILPs). The intimate intercellular contacts enforced by ILPs consistently preceded and supported T cell activation in response to endothelial MHC class II/Ag. The resulting calcium flux stabilized dense arrays of ILPs (each enriched in TCR, protein kinase C-θ, ZAP70, phosphotyrosine, and HS1), forming what we term a podo-synapse. Similar findings were made using CD8 CTLs on endothelium. Furthermore, careful re-examination of both traditional APC models and professional APCs suggests broad relevance for ILPs in facilitating Ag recognition. Together, our results indicate that ILPs function as sensory organelles that serve as actuators of immune surveillance.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Extensiones de la Superficie Celular/inmunología , Células Endoteliales/inmunología , Inmunidad Adaptativa , Animales , Presentación de Antígeno , Células Presentadoras de Antígenos/citología , Antígenos Bacterianos/inmunología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD8-positivos/citología , Células CHO , Calcio/inmunología , Calcio/metabolismo , Señalización del Calcio , Comunicación Celular/inmunología , Extensiones de la Superficie Celular/ultraestructura , Cricetinae , Células Endoteliales/citología , Genes MHC Clase II , Humanos , Memoria Inmunológica , Activación de Linfocitos , Receptores de Antígenos de Linfocitos T/biosíntesis , Receptores de Antígenos de Linfocitos T/inmunología , Migración Transendotelial y Transepitelial , Transfección
10.
Cancer Cell ; 10(2): 159-70, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16904613

RESUMEN

Endothelial cells in growing tumors express activated Akt, which when modeled by transgenic endothelial expression of myrAkt1 was sufficient to recapitulate the abnormal structural and functional features of tumor blood vessels in nontumor tissues. Sustained endothelial Akt activation caused increased blood vessel size and generalized edema from chronic vascular permeability, while acute permeability in response to VEGF-A was unaffected. These changes were reversible, demonstrating an ongoing requirement for Akt signaling for the maintenance of these phenotypes. Furthermore, rapamycin inhibited endothelial Akt signaling, vascular changes from myrAkt1, tumor growth, and tumor vascular permeability. Akt signaling in the tumor vascular stroma was sensitive to rapamycin, suggesting that rapamycin may affect tumor growth in part by acting as a vascular Akt inhibitor.


Asunto(s)
Células Endoteliales/patología , Endotelio Vascular/patología , Neoplasias/irrigación sanguínea , Neovascularización Patológica/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sirolimus/farmacología , Animales , Permeabilidad Capilar , Células Cultivadas , Edema/metabolismo , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Humanos , Ratones , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-akt/genética , Ratas , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/fisiología
11.
Stem Cells ; 30(11): 2472-86, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22887987

RESUMEN

Systemically administered adult mesenchymal stem cells (MSCs), which are being explored in clinical trials to treat inflammatory disease, exhibit the critical ability to extravasate at sites of inflammation. We aimed to characterize the basic cellular processes mediating this extravasation and compare them to those involved in leukocyte transmigration. Using high-resolution confocal and dynamic microscopy, we show that, like leukocytes, human bone marrow-derived MSC preferentially adhere to and migrate across tumor necrosis factor-α-activated endothelium in a vascular cell adhesion molecule-1 (VCAM-1) and G-protein-coupled receptor signaling-dependent manner. As several studies have suggested, we observed that a fraction of MSC was integrated into endothelium. In addition, we observed two modes of transmigration not previously observed for MSC: Paracellular (between endothelial cells) and transcellular (directly through individual endothelial cells) diapedesis through discrete gaps and pores in the endothelial monolayer, in association with VCAM-1-enriched "transmigratory cups". Contrasting leukocytes, MSC transmigration was not preceded by significant lateral migration and occurred on the time scale of hours rather than minutes. Interestingly, rather than lamellipodia and invadosomes, MSC exhibited nonapoptotic membrane blebbing activity that was similar to activities previously described for metastatic tumor and embryonic germ cells. Our studies suggest that low avidity binding between endothelium and MSC may grant a permissive environment for MSC blebbing. MSC blebbing was associated with early stages of transmigration, in which blebs could exert forces on underlying endothelial cells indicating potential functioning in breaching the endothelium. Collectively, our data suggest that MSC transmigrate actively into inflamed tissues via both leukocyte-like and novel mechanisms.


Asunto(s)
Células Endoteliales/fisiología , Células Madre Mesenquimatosas/fisiología , Migración Transendotelial y Transepitelial , Factor de Necrosis Tumoral alfa/fisiología , Animales , Adhesión Celular , Membrana Celular/metabolismo , Micropartículas Derivadas de Células/metabolismo , Células Cultivadas , Técnicas de Cocultivo , Células Endoteliales/inmunología , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Humanos , Leucocitos/fisiología , Células Madre Mesenquimatosas/ultraestructura , Microvasos/citología , Ratas , Molécula 1 de Adhesión Celular Vascular/metabolismo
12.
Cancer Cell ; 7(3): 251-61, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15766663

RESUMEN

Angiogenesis inhibitors, such as TNP-470 and the nontoxic HPMA copolymer-TNP-470 (caplostatin), are emerging as a class of anticancer drugs. We report that TNP-470 and caplostatin inhibit vascular hyperpermeability of tumor blood vessels as well as that induced in mouse skin by different mediators. Treatment with TNP-470 or angiostatin for 3 days was sufficient to reduce permeability of tumor blood vessels, delayed-type hypersensitivity, and pulmonary edema induced by IL-2. TNP-470 also inhibited VPF/VEGF-induced phosphorylation of VEGFR-2, calcium influx, and RhoA activation in endothelial cells. These results identify an activity of TNP-470, that of inhibiting vessel hyperpermeability. This activity likely contributes to TNP-470's antiangiogenic effect and suggests that caplostatin can be used in the treatment of cancer and inflammation.


Asunto(s)
Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/farmacología , Capilares/efectos de los fármacos , Permeabilidad Capilar/efectos de los fármacos , Sesquiterpenos/química , Sesquiterpenos/farmacología , Angiostatinas/farmacología , Animales , Calcio/metabolismo , Capilares/metabolismo , Capilares/ultraestructura , Permeabilidad Capilar/fisiología , Movimiento Celular/efectos de los fármacos , Ciclohexanos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/ultraestructura , Femenino , Hipersensibilidad Tardía , Interleucina-2/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neoplasias/irrigación sanguínea , Neoplasias/metabolismo , Neoplasias/patología , O-(Cloroacetilcarbamoil) Fumagilol , Edema Pulmonar/inducido químicamente , Piel/irrigación sanguínea , Piel/efectos de los fármacos , Piel/patología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
13.
Angiogenesis ; 14(3): 345-54, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21626280

RESUMEN

Transmembrane-4-L-six-family-1 (TM4SF1) is a tetraspanin-like membrane protein that is highly and selectively expressed by cultured endothelial cells (EC) and, in vivo, by EC lining angiogenic tumor blood vessels. TM4SF1 is necessary for the formation of unusually long (up to a 50 µm), thin (~100-300 nm wide), F-actin-poor EC cell projections that we term 'nanopodia'. Immunostaining of nanopodia at both the light and electron microsopic levels localized TM4SF1 in a regularly spaced, banded pattern, forming TM4FS1-enriched domains. Live cell imaging of GFP-transduced HUVEC demonstrated that EC project nanopodia as they migrate and interact with neighboring cells. When TM4SF1 mRNA levels in EC were increased from the normal ~90 mRNA copies/cell to ~400 copies/cell through adenoviral transduction, EC projected more and longer nanopodia from the entire cell circumference but were unable to polarize or migrate effectively. When fibroblasts, which normally express TM4SF1 at ~5 copies/cell, were transduced to express TM4SF1 at EC-like levels, they formed typical TM4SF1-banded nanopodia, and broadened, EC-like lamellipodia. Mass-spectrometry demonstrated that TM4SF1 interacted with myosin-10 and ß-actin, proteins involved in filopodia formation and cell migration. In summary, TM4SF1, like genuine tetraspanins, serves as a molecular organizer that interacts with membrane and cytoskeleton-associated proteins and uniquely initiates the formation of nanopodia and facilitates cell polarization and migration.


Asunto(s)
Antígenos de Superficie/metabolismo , Estructuras de la Membrana Celular/metabolismo , Movimiento Celular/fisiología , Polaridad Celular/fisiología , Células Endoteliales/metabolismo , Fibroblastos/metabolismo , Proteínas de Neoplasias/metabolismo , Seudópodos/metabolismo , Antígenos de Superficie/genética , Estructuras de la Membrana Celular/genética , Células Cultivadas , Citoesqueleto/genética , Citoesqueleto/metabolismo , Células Endoteliales/citología , Fibroblastos/citología , Humanos , Proteínas de Neoplasias/genética , Seudópodos/genética
14.
Blood ; 113(13): 3092-101, 2009 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-19171875

RESUMEN

Eosinophil chemotaxis and survival within tissues are key components in the development of tissue eosinophilia and subsequent effector responses. In this study, we demonstrate a novel mechanism of eosinophil autoregulation affecting migration and survival mediated through Notch signaling. We show for the first time that human blood eosinophils express Notch receptors and Notch ligands, expressions of which are influenced by the presence of eosinophil-activating granulocyte-macrophage colony-stimulating factor (GM-CSF). Evidence of Notch receptor activation and subsequent transcription of the Notch-responsive gene HES1 were observed in GM-CSF-stimulated eosinophils, confirming functionality of eosinophil-expressed Notch-signaling components. Moreover, by inhibiting Notch signaling with gamma-secretase inhibitors or Notch receptor-specific neutralizing antibodies, we demonstrate that autocrine Notch signaling enhances stimulus-mediated actin rearrangement and eosinophil chemokinesis, and impairs eosinophil viability. Taken together, these data suggest autocrine Notch signaling, enhanced in response to tissue- or inflammatory-derived signals, influences eosinophil activity and longevity, which may ultimately contribute to the development of tissue eosinophilia and exacerbation or remediation of eosinophil effector functions.


Asunto(s)
Comunicación Autocrina/fisiología , Diferenciación Celular/fisiología , Eosinófilos/metabolismo , Ligandos , Receptores Notch/metabolismo , Comunicación Autocrina/genética , Comunicación Autocrina/inmunología , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Endopeptidasas , Inhibidores Enzimáticos/farmacología , Eosinófilos/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Proteína Jagged-2 , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Péptido Hidrolasas , Receptores Notch/genética , Proteínas Serrate-Jagged , Proteínas Solubles de Unión al Factor Sensible a la N-Etilmaleimida/antagonistas & inhibidores
15.
Proc Natl Acad Sci U S A ; 105(47): 18478-83, 2008 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-19017810

RESUMEN

Intracellular granules in several types of leukocytes contain preformed proteins whose secretions contribute to immune and inflammatory functions of leukocytes, including eosinophils, cells notably associated with asthma, allergic inflammation, and helminthic infections. Cytokines and chemokines typically elicit extracellular secretion of granule proteins by engaging receptors expressed externally on the plasma membranes of cells, including eosinophils. Eosinophil granules, in addition to being intracellular organelles, are found as intact membrane-bound structures extracellularly in tissue sites of eosinophil-associated diseases. Neither the secretory capacities of cell-free eosinophil granules nor the presence of functional cytokine and chemokine receptors on membranes of leukocyte granules have been recognized. Here, we show that granules of human eosinophils express membrane receptors for a cytokine, IFN-gamma, and G protein-coupled membrane receptors for a chemokine, eotaxin, and that these receptors function by activating signal-transducing pathways within granules to elicit secretion from within granules. Capacities of intracellular granule organelles to function autonomously outside of eosinophils as independent, ligand-responsive, secretion-competent structures constitute a novel postcytolytic mechanism for regulated secretion of eosinophil granule proteins that may contribute to eosinophil-mediated inflammation and immunomodulation.


Asunto(s)
Gránulos Citoplasmáticos/fisiología , Eosinófilos/ultraestructura , Orgánulos/fisiología , Western Blotting , Brefeldino A/farmacología , Citocinas/metabolismo , Eosinófilos/efectos de los fármacos , Citometría de Flujo , Humanos , Interferón gamma/fisiología , Microscopía Electrónica de Transmisión , Transducción de Señal
16.
J Exp Med ; 200(4): 507-17, 2004 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-15314076

RESUMEN

Using mice in which the eGfp gene replaced the first exon of the Il4 gene (G4 mice), we examined production of interleukin (IL)-4 during infection by the intestinal nematode Nippostrongylus brasiliensis (Nb). Nb infection induced green fluorescent protein (GFP)pos cells that were FcepsilonRIpos, CD49bbright, c-kitneg, and Gr1neg. These cells had lobulated nuclei and granules characteristic of basophils. They were found mainly in the liver and lung, to a lesser degree in the spleen, but not in the lymph nodes. Although some liver basophils from naive mice express GFP, Nb infection enhanced GFP expression and increased the number of tissue basophils. Similar basophil GFP expression was found in infected Stat6-/- mice. Basophils did not increase in number in infected Rag2-/- mice; Rag2-/- mice reconstituted with CD4 T cells allowed significant basophil accumulation, indicating that CD4 T cells can direct both tissue migration of basophils and enhanced IL-4 production. IL-4 production was immunoglobulin independent and only partially dependent on IL-3. Thus, infection with a parasite that induces a "Th2-type response" resulted in accumulation of tissue basophils, and these cells, stimulated by a non-FcR cross-linking mechanism, are a principal source of in vivo IL-4 production.


Asunto(s)
Basófilos/inmunología , Regulación de la Expresión Génica/inmunología , Interleucina-4/inmunología , Ratones/parasitología , Células Th2/inmunología , Trichostrongyloidea/inmunología , Animales , Basófilos/fisiología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/fisiología , Cartilla de ADN , Citometría de Flujo , Proteínas Fluorescentes Verdes , Inmunoglobulina E/inmunología , Interleucina-4/sangre , Interleucina-4/genética , Hígado/inmunología , Hígado/ultraestructura , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Pulmón/inmunología , Pulmón/ultraestructura , Ratones/genética , Ratones/inmunología , Ratones Transgénicos , Microscopía Electrónica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
J Exp Med ; 196(11): 1497-506, 2002 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-12461084

RESUMEN

Vascular permeability factor/vascular endothelial growth factor (VPF/VEGF, VEGF-A) is a multifunctional cytokine with important roles in pathological angiogenesis. Using an adenoviral vector engineered to express murine VEGF-A(164), we previously investigated the steps and mechanisms by which this cytokine induced the formation of new blood vessels in adult immunodeficient mice and demonstrated that the newly formed blood vessels closely resembled those found in VEGF-A-expressing tumors. We now report that, in addition to inducing angiogenesis, VEGF-A(164) also induces a strong lymphangiogenic response. This finding was unanticipated because lymphangiogenesis has been thought to be mediated by other members of the VPF/VEGF family, namely, VEGF-C and VEGF-D. The new "giant" lymphatics generated by VEGF-A(164) were structurally and functionally abnormal: greatly enlarged with incompetent valves, sluggish flow, and delayed lymph clearance. They closely resembled the large lymphatics found in lymphangiomas/lymphatic malformations, perhaps implicating VEGF-A in the pathogenesis of these lesions. Whereas the angiogenic response was maintained only as long as VEGF-A was expressed, giant lymphatics, once formed, became VEGF-A independent and persisted indefinitely, long after VEGF-A expression ceased. These findings raise the possibility that similar, abnormal lymphatics develop in other pathologies in which VEGF-A is overexpressed, e.g., malignant tumors and chronic inflammation.


Asunto(s)
Inductores de la Angiogénesis/fisiología , Factores de Crecimiento Endotelial/fisiología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Sistema Linfático/patología , Linfocinas/fisiología , Neovascularización Patológica/etiología , Adenoviridae/genética , Animales , Femenino , Metástasis Linfática , Sistema Linfático/fisiología , Linfoma/etiología , Ratones , Timidina/metabolismo , Factor A de Crecimiento Endotelial Vascular , Receptor 2 de Factores de Crecimiento Endotelial Vascular/fisiología , Factores de Crecimiento Endotelial Vascular
18.
Am J Pathol ; 175(4): 1768-76, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19729487

RESUMEN

Caveolin-1, the signature protein of endothelial cell caveolae, has many important functions in vascular cells. Caveolae are thought to be the transcellular pathway by which plasma proteins cross normal capillary endothelium, but, unexpectedly, cav-1(-/-) mice, which lack caveolae, have increased permeability to plasma albumin. The acute increase in vascular permeability induced by agents such as vascular endothelial growth factor (VEGF)-A occurs through venules, not capillaries, and particularly through the vesiculo-vacuolar organelle (VVO), a unique structure composed of numerous interconnecting vesicles and vacuoles that together span the venular endothelium from lumen to ablumen. Furthermore, the hyperpermeable blood vessels found in pathological angiogenesis, mother vessels, are derived from venules. The present experiments made use of cav-1(-/-) mice to investigate the relationship between caveolae and VVOs and the roles of caveolin-1 in VVO structure in the acute vascular hyperpermeability induced by VEGF-A and in pathological angiogenesis and associated chronic vascular hyperpermeability. We found that VVOs expressed caveolin-1 variably but, in contrast to caveolae, were present in normal numbers and with apparently unaltered structure in cav-1(-/-) mice. Nonetheless, VEGF-A-induced hyperpermeability was strikingly reduced in cav-1(-/-) mice, as was pathological angiogenesis and associated chronic vascular hyperpermeability, whether induced by VEGF-A(164) or by a tumor. Thus, caveolin-1 is not necessary for VVO structure but may have important roles in regulating VVO function in acute vascular hyperpermeability and angiogenesis.


Asunto(s)
Permeabilidad Capilar/fisiología , Caveolina 1/deficiencia , Neovascularización Patológica/fisiopatología , Adenoviridae , Animales , Caveolina 1/metabolismo , Línea Celular Tumoral , Proliferación Celular , Endotelio Vascular/metabolismo , Endotelio Vascular/ultraestructura , Femenino , Ratones , Ratones Endogámicos C57BL , Neoplasias/irrigación sanguínea , Neoplasias/patología , Neovascularización Patológica/metabolismo , Piel/irrigación sanguínea , Piel/patología , Piel/ultraestructura , Vacuolas/metabolismo , Vacuolas/ultraestructura , Factor A de Crecimiento Endotelial Vascular/metabolismo
19.
Semin Thromb Hemost ; 36(3): 321-31, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20490982

RESUMEN

The blood vessels supplying tumors are strikingly heterogeneous and differ from their normal counterparts with respect to organization, structure, and function. Six distinctly different tumor vessel types have been identified, and much has been learned about the steps and mechanisms by which they form. Four of the six vessel types (mother vessels, capillaries, glomeruloid microvascular proliferations, and vascular malformations) develop from preexisting normal venules and capillaries by angiogenesis. The two remaining vessel types (feeder arteries and draining veins) develop from arterio-venogenesis, a parallel, poorly understood process that involves the remodeling of preexisting arteries and veins. All six of these tumor vessel types can be induced to form sequentially in normal mouse tissues by an adenoviral vector expressing vascular endothelial growth factor (VEGF)-A164. Current antiangiogenic cancer therapies directed at VEGF-A or its receptors have been of only limited benefit to cancer patients, perhaps because they target only the endothelial cells of the tumor blood vessel subset that requires exogenous VEGF-A for maintenance. A goal of future work is to identify therapeutic targets on tumor blood vessel endothelial cells that have lost this requirement.


Asunto(s)
Neoplasias/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/patología , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Humanos , Fenotipo , Factor A de Crecimiento Endotelial Vascular/uso terapéutico
20.
Microsc Microanal ; 16(6): 653-60, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20875166

RESUMEN

Mechanisms governing secretion of proteins underlie the biologic activities and functions of human eosinophils, leukocytes of the innate immune system, involved in allergic, inflammatory, and immunoregulatory responses. In response to varied stimuli, eosinophils are recruited from the circulation into inflammatory foci, where they modulate immune responses through the release of granule-derived products. Transmission electron microscopy (TEM) is the only technique that can clearly identify and distinguish between different modes of cell secretion. In this review, we highlight the advances in understanding mechanisms of eosinophil secretion, based on TEM findings, that have been made over the past years and that have provided unprecedented insights into the functional capabilities of these cells.


Asunto(s)
Citocinas/metabolismo , Eosinófilos/metabolismo , Eosinófilos/ultraestructura , Mediadores de Inflamación/metabolismo , Microscopía Electrónica/métodos , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA