Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Nat Immunol ; 20(5): 602-612, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30886418

RESUMEN

Despite intense interest in antiviral T cell priming, the routes by which virions move in lymph nodes (LNs) are imperfectly understood. Current models fail to explain how virus-infected cells rapidly appear within the LN interior after viral infection. To better understand virion trafficking in the LN, we determined the locations of virions and infected cells after administration to mice of vaccinia virus or Zika virus. Notably, many rapidly infected cells in the LN interior were adjacent to LN conduits. Through the use of confocal and electron microscopy, we clearly visualized virions within conduits. Functionally, CD8+ T cells rapidly and preferentially associated with vaccinia virus-infected cells in the LN paracortex, which led to T cell activation in the LN interior. These results reveal that it is possible for even large virions to flow through LN conduits and infect dendritic cells within the T cell zone to prime CD8+ T cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Ganglios Linfáticos/inmunología , Activación de Linfocitos/inmunología , Virión/inmunología , Animales , Linfocitos T CD8-positivos/virología , Femenino , Ganglios Linfáticos/ultraestructura , Ganglios Linfáticos/virología , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía Confocal , Microscopía Electrónica de Transmisión , Virus Vaccinia/inmunología , Virus Vaccinia/fisiología , Virión/fisiología , Virión/ultraestructura , Virosis/inmunología , Virosis/virología , Virus Zika/inmunología , Virus Zika/fisiología
2.
Cell ; 144(3): 402-13, 2011 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-21295700

RESUMEN

The functions of caveolae, the characteristic plasma membrane invaginations, remain debated. Their abundance in cells experiencing mechanical stress led us to investigate their role in membrane-mediated mechanical response. Acute mechanical stress induced by osmotic swelling or by uniaxial stretching results in a rapid disappearance of caveolae, in a reduced caveolin/Cavin1 interaction, and in an increase of free caveolins at the plasma membrane. Tether-pulling force measurements in cells and in plasma membrane spheres demonstrate that caveola flattening and disassembly is the primary actin- and ATP-independent cell response that buffers membrane tension surges during mechanical stress. Conversely, stress release leads to complete caveola reassembly in an actin- and ATP-dependent process. The absence of a functional caveola reservoir in myotubes from muscular dystrophic patients enhanced membrane fragility under mechanical stress. Our findings support a new role for caveolae as a physiological membrane reservoir that quickly accommodates sudden and acute mechanical stresses.


Asunto(s)
Caveolas/fisiología , Células Endoteliales/citología , Células Musculares/fisiología , Actinas/fisiología , Adenosina Trifosfato/fisiología , Animales , Caveolas/ultraestructura , Línea Celular , Células Endoteliales/fisiología , Humanos , Ratones , Células Musculares/citología , Estrés Mecánico
3.
Int J Mol Sci ; 24(6)2023 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-36982602

RESUMEN

Cholesterol is a key component of all mammalian cell membranes. Disruptions in cholesterol metabolism have been observed in the context of various diseases, including neurodegenerative disorders such as Alzheimer's disease (AD). The genetic and pharmacological blockade of acyl-CoA:cholesterol acyltransferase 1/sterol O-acyltransferase 1 (ACAT1/SOAT1), a cholesterol storage enzyme found on the endoplasmic reticulum (ER) and enriched at the mitochondria-associated ER membrane (MAM), has been shown to reduce amyloid pathology and rescue cognitive deficits in mouse models of AD. Additionally, blocking ACAT1/SOAT1 activity stimulates autophagy and lysosomal biogenesis; however, the exact molecular connection between the ACAT1/SOAT1 blockade and these observed benefits remain unknown. Here, using biochemical fractionation techniques, we observe cholesterol accumulation at the MAM which leads to ACAT1/SOAT1 enrichment in this domain. MAM proteomics data suggests that ACAT1/SOAT1 inhibition strengthens the ER-mitochondria connection. Confocal and electron microscopy confirms that ACAT1/SOAT1 inhibition increases the number of ER-mitochondria contact sites and strengthens this connection by shortening the distance between these two organelles. This work demonstrates how directly manipulating local cholesterol levels at the MAM can alter inter-organellar contact sites and suggests that cholesterol buildup at the MAM is the impetus behind the therapeutic benefits of ACAT1/SOAT1 inhibition.


Asunto(s)
Enfermedad de Alzheimer , Colesterol , Animales , Ratones , Enfermedad de Alzheimer/metabolismo , Colesterol/metabolismo , Retículo Endoplásmico/metabolismo , Mamíferos/metabolismo , Mitocondrias/metabolismo , Esteroles/metabolismo , Acetil-CoA C-Aciltransferasa/metabolismo , Esterol O-Aciltransferasa/metabolismo
4.
Genes Chromosomes Cancer ; 60(2): 116-122, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33159395

RESUMEN

To our knowledge, we describe the first mesenchymal tumor with a novel GLI1-FOXO4 fusion gene. This well-circumscribed kidney tumor displayed variably myxoid and epithelioid histologic features with a focally nodular growth pattern. The tumor cells showed bland, round to ovoid nuclei, with no overt high-grade features. The tumor showed focal immunopositivity for smooth muscle actin and Melan-A, which raised the possibility of a relationship with a perivascular epithelioid cell tumor. The clinical and morphologic features appear distinct from other reported neoplasms harboring GLI1 or FOXO4 gene rearrangements. The patient underwent radical nephrectomy and is without evidence of disease during a relatively short clinical follow-up period. However, the features of this tumor likely warrant long-term follow-up to monitor for the possibility of a late recurrence or metastasis. In addition to reporting this novel fusion-positive tumor, we also provide a brief review of GLI1 and FOXO4 gene functions in both normal and neoplastic contexts.


Asunto(s)
Proteínas de Ciclo Celular/genética , Factores de Transcripción Forkhead/genética , Neoplasias Renales/genética , Mesenquimoma/genética , Proteínas de Fusión Oncogénica/genética , Proteína con Dedos de Zinc GLI1/genética , Humanos , Neoplasias Renales/patología , Masculino , Mesenquimoma/patología , Persona de Mediana Edad
5.
J Cell Mol Med ; 23(2): 920-933, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30394679

RESUMEN

Endothelial diaphragms are subcellular structures critical for mammalian survival with poorly understood biogenesis. Plasmalemma vesicle associated protein (PLVAP) is the only known diaphragm component and is necessary for diaphragm formation. Very little is known about PLVAP regulation. Phorbol esters (PMA) are known to induce de novo PLVAP expression and diaphragm formation. We show that this induction relies on the de novo production of soluble factors that will act in an autocrine manner to induce PLVAP transcription and protein expression. We identified vascular endothelial growth factor-A (VEGF-A) signalling through VEGFR2 as a necessary but not sufficient downstream event as VEGF-A inhibition with antibodies and siRNA or pharmacological inhibition of VEGFR2 only partially inhibit PLVAP upregulation. In terms of downstream pathways, inhibition of MEK1/Erk1/2 MAP kinase blocked PLVAP upregulation, whereas inhibition of p38 and JNK MAP kinases or PI3K and Akt had no effect on PMA-induced PLVAP expression. In conclusion, we show that VEGF-A along with other secreted proteins act synergistically to up-regulate PLVAP in MEK1/Erk1/2 dependent manner, bringing us one step further into understanding the genesis of the essential structures that are endothelial diaphragms.


Asunto(s)
Comunicación Autocrina/genética , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , MAP Quinasa Quinasa 1/genética , Proteínas de la Membrana/genética , Acetato de Tetradecanoilforbol/farmacología , Factor A de Crecimiento Endotelial Vascular/genética , Antracenos/farmacología , Axitinib/farmacología , Butadienos/farmacología , Flavonoides/farmacología , Regulación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Imidazoles/farmacología , Indazoles/farmacología , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 4/antagonistas & inhibidores , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/metabolismo , Proteínas de la Membrana/agonistas , Proteínas de la Membrana/metabolismo , Nitrilos/farmacología , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Piridinas/farmacología , Pirimidinas/farmacología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Sulfonamidas/farmacología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
6.
Arterioscler Thromb Vasc Biol ; 38(2): 353-362, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29284606

RESUMEN

OBJECTIVE: The assembly of a functional vascular system requires a coordinated and dynamic transition from activation to maturation. High vascular endothelial growth factor activity promotes activation, including junction destabilization and cell motility. Maturation involves junctional stabilization and formation of a functional endothelial barrier. The identity and mechanism of action of prostabilization signals are still mostly unknown. Bone morphogenetic protein receptors and their ligands have important functions during embryonic vessel assembly and maturation. Previous work has suggested a role for growth differentiation factor 6 (GDF6; bone morphogenetic protein 13) in vascular integrity although GDF6's mechanism of action was not clear. Therefore, we sought to further explore the requirement for GDF6 in vascular stabilization. APPROACH AND RESULTS: We investigated the role of GDF6 in promoting endothelial vascular integrity in vivo in zebrafish and in cultured human umbilical vein endothelial cells in vitro. We report that GDF6 promotes vascular integrity by counteracting vascular endothelial growth factor activity. GDF6-deficient endothelium has increased vascular endothelial growth factor signaling, increased vascular endothelial-cadherin Y658 phosphorylation, vascular endothelial-cadherin delocalization from cell-cell interfaces, and weakened endothelial cell adherence junctions that become prone to vascular leak. CONCLUSIONS: Our results suggest that GDF6 promotes vascular stabilization by restraining vascular endothelial growth factor signaling. Understanding how GDF6 affects vascular integrity may help to provide insights into hemorrhage and associated vascular pathologies in humans.


Asunto(s)
Permeabilidad Capilar , Embrión no Mamífero/irrigación sanguínea , Células Endoteliales/metabolismo , Factor 6 de Diferenciación de Crecimiento/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Antígenos CD/genética , Antígenos CD/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Células Cultivadas , Regulación del Desarrollo de la Expresión Génica , Factor 6 de Diferenciación de Crecimiento/genética , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Neovascularización Fisiológica , Fosforilación , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/genética
7.
Proc Natl Acad Sci U S A ; 113(47): 13408-13413, 2016 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-27810956

RESUMEN

As a key component of the vertebrate neuroendocrine system, the pituitary gland relies on the progressive and coordinated development of distinct hormone-producing cell types and an invading vascular network. The molecular mechanisms that drive formation of the pituitary vasculature, which is necessary for regulated synthesis and secretion of hormones that maintain homeostasis, metabolism, and endocrine function, remain poorly understood. Here, we report that expression of integrin ß1 in embryonic pituitary epithelial cells is required for angiogenesis in the developing mouse pituitary gland. Deletion of pituitary epithelial integrin ß1 before the onset of angiogenesis resulted in failure of invading endothelial cells to recruit pericytes efficiently, whereas deletion later in embryogenesis led to decreased vascular density and lumen formation. In both cases, lack of epithelial integrin ß1 was associated with a complete absence of vasculature in the pituitary gland at birth. Within pituitary epithelial cells, integrin ß1 directs a large transcriptional program that includes components of the extracellular matrix and associated signaling factors that are linked to the observed non-cell-autonomous effects on angiogenesis. We conclude that epithelial integrin ß1 functions as a critical and canonical regulator of developmental angiogenesis in the pituitary gland, thus providing insight into the long-standing systems biology conundrum of how vascular invasion is coordinated with tissue development.


Asunto(s)
Desarrollo Embrionario , Células Epiteliales/metabolismo , Integrina beta1/metabolismo , Neovascularización Fisiológica , Hipófisis/citología , Hipófisis/embriología , Animales , Animales Recién Nacidos , Recuento de Células , Diferenciación Celular , Desarrollo Embrionario/genética , Células Endoteliales/citología , Células Endoteliales/metabolismo , Matriz Extracelular/metabolismo , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Marcación de Gen , Integrasas/metabolismo , Ratones , Neovascularización Fisiológica/genética , Factores de Transcripción Paired Box/metabolismo , Pericitos/citología , Pericitos/metabolismo , Fenotipo , Hipófisis/metabolismo , Análisis de Secuencia de ARN , Factores de Tiempo , Factor C de Crecimiento Endotelial Vascular/metabolismo
8.
J Am Soc Nephrol ; 29(4): 1097-1107, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29237738

RESUMEN

Urinary concentrating ability is central to mammalian water balance and depends on a medullary osmotic gradient generated by a countercurrent multiplication mechanism. Medullary hyperosmolarity is protected from washout by countercurrent exchange and efficient removal of interstitial fluid resorbed from the loop of Henle and collecting ducts. In most tissues, lymphatic vessels drain excess interstitial fluid back to the venous circulation. However, the renal medulla is devoid of classic lymphatics. Studies have suggested that the fenestrated ascending vasa recta (AVRs) drain the interstitial fluid in this location, but this function has not been conclusively shown. We report that late gestational deletion of the angiopoietin receptor endothelial tyrosine kinase 2 (Tie2) or both angiopoietin-1 and angiopoietin-2 prevents AVR formation in mice. The absence of AVR associated with rapid accumulation of fluid and cysts in the medullary interstitium, loss of medullary vascular bundles, and decreased urine concentrating ability. In transgenic reporter mice with normal angiopoietin-Tie2 signaling, medullary AVR exhibited an unusual hybrid endothelial phenotype, expressing lymphatic markers (prospero homeobox protein 1 and vascular endothelial growth factor receptor 3) as well as blood endothelial markers (CD34, endomucin, platelet endothelial cell adhesion molecule 1, and plasmalemmal vesicle-associated protein). Taken together, our data redefine the AVRs as Tie2 signaling-dependent specialized hybrid vessels and provide genetic evidence of the critical role of AVR in the countercurrent exchange mechanism and the structural integrity of the renal medulla.


Asunto(s)
Angiopoyetina 1/fisiología , Angiopoyetina 2/fisiología , Líquido Extracelular/metabolismo , Capacidad de Concentración Renal/fisiología , Médula Renal/irrigación sanguínea , Receptor TIE-2/fisiología , Angiopoyetina 1/deficiencia , Angiopoyetina 1/genética , Angiopoyetina 2/deficiencia , Angiopoyetina 2/genética , Animales , Tipificación del Cuerpo , Linaje de la Célula , Endotelio Vascular , Genes Reporteros , Edad Gestacional , Proteínas de Homeodominio/análisis , Enfermedades Renales Quísticas/genética , Médula Renal/embriología , Médula Renal/fisiología , Ratones , Ratones Noqueados , Ratones Transgénicos , Miofibroblastos/patología , Ósmosis , Receptor TIE-2/deficiencia , Receptor TIE-2/genética , Circulación Renal , Transducción de Señal , Proteínas Supresoras de Tumor/análisis , Receptor 3 de Factores de Crecimiento Endotelial Vascular/análisis
9.
Circ Res ; 118(1): 48-55, 2016 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-26602865

RESUMEN

RATIONALE: Caveolin-1 (Cav-1) negatively regulates endothelial nitric oxide (NO) synthase-derived NO production, and this has been mapped to several residues on Cav-1, including F92. Herein, we reasoned that endothelial expression of an F92ACav-1 transgene would let us decipher the mechanisms and relationships between caveolae structure and intracellular signaling. OBJECTIVE: This study was designed to separate caveolae formation from its downstream signaling effects. METHODS AND RESULTS: An endothelial-specific doxycycline-regulated mouse model for the expression of Cav-1-F92A was developed. Blood pressure by telemetry and nitric oxide bioavailability by electron paramagnetic resonance and phosphorylation of vasodilator-stimulated phosphoprotein were determined. Caveolae integrity in the presence of Cav-1-F92A was measured by stabilization of caveolin-2, sucrose gradient, and electron microscopy. Histological analysis of heart and lung, echocardiography, and signaling were performed. CONCLUSIONS: This study shows that mutant Cav-1-F92A forms caveolae structures similar to WT but leads to increases in NO bioavailability in vivo, thereby demonstrating that caveolae formation and downstream signaling events occur through independent mechanisms.


Asunto(s)
Caveolas/metabolismo , Caveolina 1/biosíntesis , Caveolina 1/genética , Líquido Intracelular/metabolismo , Transducción de Señal/fisiología , Animales , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Caveolas/efectos de los fármacos , Doxiciclina/farmacología , Humanos , Líquido Intracelular/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Óxido Nítrico/metabolismo , Transducción de Señal/efectos de los fármacos , Desacopladores/farmacología
10.
J Immunol ; 197(10): 3970-3981, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27742829

RESUMEN

Plasmalemma vesicle-associated protein (Plvap) is an endothelial protein with roles in endothelial diaphragm formation and maintenance of basal vascular permeability. At the same time, Plvap has roles in immunity by facilitating leukocyte diapedesis at inflammatory sites and controlling peripheral lymph node morphogenesis and the entry of soluble Ags into lymph node conduits. Based on its postulated role in diapedesis, we have investigated the role of Plvap in hematopoiesis and show that deletion of Plvap results in a dramatic decrease of IgM+IgDlo B cells in both the spleen and the peritoneal cavity. Tissue-specific deletion of Plvap demonstrates that the defect is B cell extrinsic, because B cell and pan-hematopoietic Plvap deletion has no effect on IgM+IgDlo B cell numbers. Endothelial-specific deletion of Plvap in the embryo or at adult stage recapitulates the full Plvap knockout phenotype, whereas endothelial-specific reconstitution of Plvap under the Chd5 promoter rescues the IgM+IgDlo B cell phenotype. Taken together, these results show that Plvap expression in endothelial cells is important in the maintenance of IgM+ B cells in the spleen and peritoneal cavity.


Asunto(s)
Linfocitos B/metabolismo , Proteínas Portadoras/metabolismo , Homeostasis , Proteínas de la Membrana/metabolismo , Células Precursoras de Linfocitos B/metabolismo , Bazo/citología , Animales , Linfocitos B/patología , Permeabilidad Capilar , Proteínas Portadoras/genética , ADN Helicasas/genética , Células Endoteliales/química , Células Endoteliales/metabolismo , Regulación de la Expresión Génica , Inmunoglobulina D/genética , Inmunoglobulina D/metabolismo , Inmunoglobulina M/genética , Inmunoglobulina M/metabolismo , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Cavidad Peritoneal/citología , Fenotipo , Bazo/inmunología , Migración Transendotelial y Transepitelial/inmunología
11.
BMC Cancer ; 13: 211, 2013 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-23621985

RESUMEN

BACKGROUND: New pharmacologic targets are urgently needed to treat or prevent lung cancer, the most common cause of cancer death for men and women. This study identified one such target. This is the canonical Wnt signaling pathway, which is deregulated in cancers, including those lacking adenomatous polyposis coli or ß-catenin mutations. Two poly-ADP-ribose polymerase (PARP) enzymes regulate canonical Wnt activity: tankyrase (TNKS) 1 and TNKS2. These enzymes poly-ADP-ribosylate (PARsylate) and destabilize axin, a key component of the ß-catenin phosphorylation complex. METHODS: This study used comprehensive gene profiles to uncover deregulation of the Wnt pathway in murine transgenic and human lung cancers, relative to normal lung. Antineoplastic consequences of genetic and pharmacologic targeting of TNKS in murine and human lung cancer cell lines were explored, and validated in vivo in mice by implantation of murine transgenic lung cancer cells engineered with reduced TNKS expression relative to controls. RESULTS: Microarray analyses comparing Wnt pathway members in malignant versus normal tissues of a murine transgenic cyclin E lung cancer model revealed deregulation of Wnt pathway components, including TNKS1 and TNKS2. Real-time PCR assays independently confirmed these results in paired normal-malignant murine and human lung tissues. Individual treatments of a panel of human and murine lung cancer cell lines with the TNKS inhibitors XAV939 and IWR-1 dose-dependently repressed cell growth and increased cellular axin 1 and tankyrase levels. These inhibitors also repressed expression of a Wnt-responsive luciferase construct, implicating the Wnt pathway in conferring these antineoplastic effects. Individual or combined knockdown of TNKS1 and TNKS2 with siRNAs or shRNAs reduced lung cancer cell growth, stabilized axin, and repressed tumor formation in murine xenograft and syngeneic lung cancer models. CONCLUSIONS: Findings reported here uncovered deregulation of specific components of the Wnt pathway in both human and murine lung cancer models. Repressing TNKS activity through either genetic or pharmacological approaches antagonized canonical Wnt signaling, reduced murine and human lung cancer cell line growth, and decreased tumor formation in mouse models. Taken together, these findings implicate the use of TNKS inhibitors to target the Wnt pathway to combat lung cancer.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Tanquirasas/genética , Vía de Señalización Wnt/genética , Análisis de Varianza , Animales , Proteína Axina/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Femenino , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Compuestos Heterocíclicos con 3 Anillos/farmacología , Compuestos Heterocíclicos con 3 Anillos/uso terapéutico , Humanos , Imidas/farmacología , Imidas/uso terapéutico , Pulmón/enzimología , Neoplasias Pulmonares/enzimología , Ratones , Análisis por Micromatrices , Quinolinas/farmacología , Quinolinas/uso terapéutico , Reacción en Cadena en Tiempo Real de la Polimerasa , Tanquirasas/antagonistas & inhibidores , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/metabolismo
12.
J Cell Mol Med ; 16(11): 2690-700, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22568538

RESUMEN

PV1 is an endothelial-specific protein with structural roles in the formation of diaphragms in endothelial cells of normal vessels. PV1 is also highly expressed on endothelial cells of many solid tumours. On the basis of in vitro data, PV1 is thought to actively participate in angiogenesis. To test whether or not PV1 has a function in tumour angiogenesis and in tumour growth in vivo, we have treated pancreatic tumour-bearing mice by single-dose intratumoural delivery of lentiviruses encoding for two different shRNAs targeting murine PV1. We find that PV1 down-regulation by shRNAs inhibits the growth of established tumours derived from two different human pancreatic adenocarcinoma cell lines (AsPC-1 and BxPC-3). The effect observed is because of down-regulation of PV1 in the tumour endothelial cells of host origin, PV1 being specifically expressed in tumour vascular endothelial cells and not in cancer or other stromal cells. There are no differences in vascular density of tumours treated or not with PV1 shRNA, and gain and loss of function of PV1 in endothelial cells does not modify either their proliferation or migration, suggesting that tumour angiogenesis is not impaired. Together, our data argue that down-regulation of PV1 in tumour endothelial cells results in the inhibition of tumour growth via a mechanism different from inhibiting angiogenesis.


Asunto(s)
Adenocarcinoma/genética , Adenocarcinoma/patología , Proteínas Portadoras/genética , Proteínas de la Membrana/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Adenocarcinoma/irrigación sanguínea , Animales , Secuencia de Bases , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Regulación hacia Abajo , Ensayos de Selección de Medicamentos Antitumorales , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Lentivirus/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Ratones Desnudos , Datos de Secuencia Molecular , Neovascularización Patológica/genética , Neoplasias Pancreáticas/irrigación sanguínea , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/farmacología , Células del Estroma/metabolismo , Células del Estroma/patología
13.
Cancer Immunol Immunother ; 61(12): 2273-82, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22684520

RESUMEN

Mast cells have emerged as critical intermediaries in the regulation of peripheral tolerance. Their presence in many precancerous lesions and tumors is associated with a poor prognosis, suggesting mast cells may promote an immunosuppressive tumor microenvironment and impede the development of protective anti-tumor immunity. The studies presented herein investigate how mast cells influence tumor-specific T cell responses. Male MB49 tumor cells, expressing HY antigens, induce anti-tumor IFN-γ(+) T cell responses in female mice. However, normal female mice cannot control progressive MB49 tumor growth. In contrast, mast cell-deficient c-Kit(Wsh) (W(sh)) female mice controlled tumor growth and exhibited enhanced survival. The role of mast cells in curtailing the development of protective immunity was shown by increased mortality in mast cell-reconstituted W(sh) mice with tumors. Confirmation of enhanced immunity in female W(sh) mice was provided by (1) higher frequency of tumor-specific IFN-γ(+) CD8(+) T cells in tumor-draining lymph nodes compared with WT females and (2) significantly increased ratios of intratumoral CD4(+) and CD8(+) T effector cells relative to tumor cells in W(sh) mice compared to WT. These studies are the first to reveal that mast cells impair both regional adaptive immune responses and responses within the tumor microenvironment to diminish protective anti-tumor immunity.


Asunto(s)
Mastocitos/inmunología , Neoplasias/inmunología , Neoplasias/patología , Microambiente Tumoral/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Femenino , Antígeno H-Y/inmunología , Inmunidad/inmunología , Interferón gamma/inmunología , Masculino , Ratones , Neoplasias/irrigación sanguínea , Neovascularización Patológica/inmunología , Neovascularización Patológica/patología
14.
FASEB J ; 25(11): 3838-48, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21795505

RESUMEN

Caveolin-1 plays a crucial role in atherosclerosis, which is mainly attributed to its effects on low-density-lipoprotein (LDL) transcytosis. However, caveolin-1 has also been implicated in the regulation of inflammation. We investigated the effects of caveolin-1 deficiency in atherosclerosis with its accompanying changes in plaque- and lymphoid-related immunology and inflammation. Cav1(-/-)Apoe(-/-) mice exhibited a 15-fold reduction in plaque size with plaques containing fewer macrophages, T cells, and neutrophils. Intravital microscopy revealed 83% less leukocyte adhesion to the vessel wall in Cav1(-/-)Apoe(-/-) mice, which could be attributed to reduced endothelial chemokine ligand-2 (CCL-2/MCP-1) and vascular cell adhesion molecule-1 (VCAM-1) expression. Caveolin-1 deficiency resulted in a 57% increase in regulatory T cells and a 4% decrease in CD4(+) effector T cells in lymphoid organs. Bone marrow transplantations revealed that Cav1(-/-)Apoe(-/-) mice receiving Cav1(+/+)Apoe(-/-) or Cav1(-/-)Apoe(-/-) bone marrow presented 4- to 4.5-fold smaller plaques with no additional phenotypic changes. In contrast, atherosclerosis was not affected in Cav1(+/+) Apoe(-/-) recipients receiving Cav1(-/-)Apoe(-/-) or Cav1(+/+) Apoe(-/-) bone marrow. However, the presence of Cav1(-/-) Apoe(-/-) bone marrow was associated with an anti-inflammatory T-cell profile. Our study reveals that nonhematopoietic caveolin-1 determines plaque size, whereas hematopoietic caveolin-1 regulates lymphoid immune-modulation. However, both are required for phenotypic modulation of plaques.


Asunto(s)
Aterosclerosis/inmunología , Caveolina 1/deficiencia , Animales , Apolipoproteínas E/deficiencia , Arterias/inmunología , Aterosclerosis/prevención & control , Femenino , Inflamación/prevención & control , Leucocitos/inmunología , Metabolismo de los Lípidos , Masculino , Ratones , Placa Aterosclerótica/patología , Linfocitos T Reguladores/inmunología , Molécula 1 de Adhesión Celular Vascular/metabolismo
15.
PLoS One ; 17(6): e0268986, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35675336

RESUMEN

Tyrosine-protein kinase receptor Tie2, also known as Tunica interna Endothelial cell Kinase or TEK plays a prominent role in endothelial responses to angiogenic and inflammatory stimuli. Here we generated a novel inducible Tie2 knockout mouse model, which targets mature (micro)vascular endothelium, enabling the study of the organ-specific contribution of Tie2 to these responses. Mice with floxed Tie2 exon 9 alleles (Tie2floxed/floxed) were crossed with end-SCL-Cre-ERT transgenic mice, generating offspring in which Tie2 exon 9 is deleted in the endothelial compartment upon tamoxifen-induced activation of Cre-recombinase (Tie2ΔE9). Successful deletion of Tie2 exon 9 in kidney, lung, heart, aorta, and liver, was accompanied by a heterogeneous, organ-dependent reduction in Tie2 mRNA and protein expression. Microvascular compartment-specific reduction in Tie2 mRNA and protein occurred in arterioles of all studied organs, in renal glomeruli, and in lung capillaries. In kidney, lung, and heart, reduced Tie2 expression was accompanied by a reduction in Tie1 mRNA expression. The heterogeneous, organ- and microvascular compartment-dependent knockout pattern of Tie2 in the Tie2floxed/floxed;end-SCL-Cre-ERT mouse model suggests that future studies using similar knockout strategies should include a meticulous analysis of the knockout extent of the gene of interest, prior to studying its role in pathological conditions, so that proper conclusions can be drawn.


Asunto(s)
Células Endoteliales , Tamoxifeno , Animales , Células Endoteliales/metabolismo , Integrasas , Ratones , Ratones Noqueados , Ratones Transgénicos , ARN Mensajero/metabolismo , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Tamoxifeno/metabolismo , Tamoxifeno/farmacología
16.
J Clin Invest ; 118(10): 3355-66, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18776942

RESUMEN

The integrity of the endothelial monolayer is essential to blood vessel homeostasis and active regulation of endothelial permeability. The FGF system plays important roles in a wide variety of physiologic and pathologic conditions; however, its role in the adult vasculature has not been defined. To assess the role of the FGF system in the adult endothelial monolayer, we disrupted FGF signaling in bovine aortic endothelial cells and human saphenous vein endothelial cells in vitro and in adult mouse and rat endothelial cells in vivo using soluble FGF traps or a dominant inhibitor of all FGF receptors. The inhibition of FGF signaling using these approaches resulted in dissociation of the VE-cadherin/p120-catenin complex and disassembly of adherens and tight junctions, which progressed to loss of endothelial cells, severe impairment of the endothelial barrier function, and finally, disintegration of the vasculature. Thus, FGF signaling plays a key role in the maintenance of vascular integrity.


Asunto(s)
Vasos Sanguíneos/metabolismo , Endotelio Vascular/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal , Uniones Adherentes/metabolismo , Animales , Cadherinas/metabolismo , Permeabilidad Capilar/fisiología , Bovinos , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/ultraestructura , Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Regulación de la Expresión Génica , Humanos , Masculino , Ratones , Ratas , Factores de Tiempo
17.
Biochem Biophys Res Commun ; 412(2): 220-5, 2011 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-21827737

RESUMEN

Plasmalemmal vesicle associated protein (Plvap/PV1) is a structural protein required for the formation of the stomatal diaphragms of caveolae. Caveolae are plasma membrane invaginations that were implicated in SV40 virus entry in primate cells. Here we show that de novo Plvap/PV1 expression in CV-1 green monkey epithelial cells significantly reduces the ability of SV40 virus to establish productive infection, when cells are incubated with low concentrations of the virus. However, in presence of high viral titers PV1 has no effect on SV40 virus infectivity. Mechanistically, PV1 expression does not reduce the cell surface expression of known SV40 receptors such as GM1 ganglioside and MHC class I proteins. Furthermore, PV1 does not reduce the binding of virus-like particles made by SV40 VP1 protein to the CV-1 cell surface and does not impact their internalization when cells are incubated with either high or low VLP concentrations. These results suggest that PV1 protein is able to block SV40 infectivity at low but not at high viral concentration either by interfering with the infective internalization pathway at the cell surface or at a post internalization step.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de la Membrana/metabolismo , Infecciones por Polyomavirus/virología , Virus 40 de los Simios/patogenicidad , Infecciones Tumorales por Virus/virología , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/genética , Línea Celular , Chlorocebus aethiops , Células HeLa , Humanos , Proteínas de la Membrana/genética , Datos de Secuencia Molecular , Infecciones por Polyomavirus/metabolismo , Infecciones Tumorales por Virus/metabolismo
18.
Nat Med ; 10(3): 310-5, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14770178

RESUMEN

The TAT protein transduction domain (PTD) has been used to deliver a wide variety of biologically active cargo for the treatment of multiple preclinical disease models, including cancer and stroke. However, the mechanism of transduction remains unknown. Because of the TAT PTD's strong cell-surface binding, early assumptions regarding cellular uptake suggested a direct penetration mechanism across the lipid bilayer by a temperature- and energy-independent process. Here we show, using a transducible TAT-Cre recombinase reporter assay on live cells, that after an initial ionic cell-surface interaction, TAT-fusion proteins are rapidly internalized by lipid raft-dependent macropinocytosis. Transduction was independent of interleukin-2 receptor/raft-, caveolar- and clathrin-mediated endocytosis and phagocytosis. Using this information, we developed a transducible, pH-sensitive, fusogenic dTAT-HA2 peptide that markedly enhanced TAT-Cre escape from macropinosomes. Taken together, these observations provide a scientific basis for the development of new, biologically active, transducible therapeutic molecules.


Asunto(s)
Productos del Gen tat/metabolismo , Microdominios de Membrana/metabolismo , Péptidos/metabolismo , Pinocitosis/fisiología , Proteínas Virales de Fusión/metabolismo , Amilorida/metabolismo , Animales , Transporte Biológico/fisiología , Caveolina 1 , Caveolinas/metabolismo , Línea Celular , Clatrina/metabolismo , Citocalasina D/metabolismo , Endosomas/metabolismo , Productos del Gen tat/genética , Humanos , Ratones , Inhibidores de la Síntesis del Ácido Nucleico/metabolismo , Péptidos/genética , Linfocitos T/fisiología , Transducción Genética , Proteínas Virales de Fusión/genética
19.
Semin Thromb Hemost ; 36(3): 236-45, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20490976

RESUMEN

Vascular endothelium lines the entire cardiovascular system where it performs a series of vital functions by its control of microvascular permeability, vessel wall tone, coagulation and anticoagulation cascades, lipid homeostasis, inflammation, angiogenesis, and vasculogenesis. The vertebrate endothelial cells display a remarkable heterogeneity in terms of morphology, molecular makeup, and functional output. This heterogeneity was documented very early by electron microscopy studies that established morphologically recognizable endothelial phenotypes in vascular beds of different organs and, moreover, within the different vascular segments of each organ. This review discusses endothelial heterogeneity from a morphological standpoint and the latest developments in our understanding of the components, structure, and function of the endothelial specific organelles that form the hallmark of these phenotypes.


Asunto(s)
Endotelio Vascular/crecimiento & desarrollo , Animales , Proteínas Portadoras/fisiología , Diferenciación Celular , Endotelio Vascular/ultraestructura , Humanos , Proteínas de la Membrana/fisiología , Fenotipo
20.
Mol Neuropsychiatry ; 5(Suppl 1): 60-71, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32399470

RESUMEN

PTEN is a lipid and protein phosphatase that regulates cell growth and survival. Mutations to PTEN are highly penetrant for autism spectrum disorder (ASD). Here, we briefly review the evidence linking PTEN mutations to ASD and the mouse models that have been used to study the role of PTEN in neurodevelopment. We then focus on the cellular phenotypes associated with PTEN loss in neurons, highlighting the role PTEN plays in neuronal proliferation, migration, survival, morphology, and plasticity.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA