Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 110(2): E141-50, 2013 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-23236128

RESUMEN

During development, the hematopoietic lineage transits through hemogenic endothelium, but the signaling pathways effecting this transition are incompletely characterized. Although the Hedgehog (Hh) pathway is hypothesized to play a role in patterning blood formation, early embryonic lethality of mice lacking Hh signaling precludes such analysis. To determine a role for Hh signaling in patterning of hemogenic endothelium, we assessed the effect of altered Hh signaling in differentiating mouse ES cells, cultured mouse embryos, and developing zebrafish embryos. In differentiating mouse ES cells and mouse yolk sac cultures, addition of Indian Hh ligand increased hematopoietic progenitors, whereas chemical inhibition of Hh signaling reduced hematopoietic progenitors without affecting primitive streak mesoderm formation. In the setting of Hh inhibition, induction of either Notch signaling or overexpression of Stem cell leukemia (Scl)/T-cell acute lymphocytic leukemia protein 1 rescued hemogenic vascular-endothelial cadherin(+) cells and hematopoietic progenitor formation. Together, our results reveal that Scl overexpression is sufficient to rescue the developmental defects caused by blocking the Hh and Notch pathways, and inform our understanding of the embryonic endothelial-to-hematopoietic transition.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/fisiología , Endotelio/fisiología , Proteínas Hedgehog/metabolismo , Células Madre Hematopoyéticas/citología , Proteínas Proto-Oncogénicas/metabolismo , Receptores Notch/metabolismo , Transducción de Señal/fisiología , Animales , Ensayo de Unidades Formadoras de Colonias , Embrión de Mamíferos , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Células Madre Hematopoyéticas/fisiología , Hibridación in Situ , Ratones , Ratones Endogámicos C57BL , Proteína 1 de la Leucemia Linfocítica T Aguda , Pez Cebra
2.
J Cell Sci ; 126(Pt 24): 5541-52, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-24046447

RESUMEN

Transient disruption of endothelial adherens junctions and cytoskeletal remodeling are responsible for increases in vascular permeability induced by inflammatory stimuli and vascular endothelial growth factor (VEGF). Nitric oxide (NO) produced by endothelial NO synthase (eNOS) is crucial for VEGF-induced changes in permeability in vivo; however, the molecular mechanism by which endogenous NO modulates endothelial permeability is not clear. Here, we show that the lack of eNOS reduces VEGF-induced permeability, an effect mediated by enhanced activation of the Rac GTPase and stabilization of cortical actin. The loss of NO increased the recruitment of the Rac guanine-nucleotide-exchange factor (GEF) TIAM1 to adherens junctions and VE-cadherin (also known as cadherin 5), and reduced Rho activation and stress fiber formation. In addition, NO deficiency reduced VEGF-induced VE-cadherin phosphorylation and impaired the localization, but not the activation, of c-Src to cell junctions. The physiological role of eNOS activation is clear given that VEGF-, histamine- and inflammation-induced vascular permeability is reduced in mice bearing a non-phosphorylatable knock-in mutation of the key eNOS phosphorylation site S1176. Thus, NO is crucial for Rho GTPase-dependent regulation of cytoskeletal architecture leading to reversible changes in vascular permeability.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Endotelio Vascular/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico/fisiología , Uniones Adherentes/metabolismo , Animales , Proteína Tirosina Quinasa CSK , Permeabilidad Capilar , Células Cultivadas , Células Endoteliales/enzimología , Endotelio Vascular/citología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo III/genética , Fosforilación , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Fibras de Estrés/metabolismo , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T , Factor A de Crecimiento Endotelial Vascular/fisiología , Familia-src Quinasas/metabolismo
3.
Am J Pathol ; 182(5): 1900-9, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23499551

RESUMEN

Cancer cells are often characterized by high proliferation rates, a consequence of increased mitotic signaling coupled with unchecked cellular growth. We recently demonstrated that vascular endothelial cells unexpectedly express ferlins, a family of muscle-specific proteins capable of regulating the fusion of lipid patches to the plasma membrane, and that these highly regulated membrane fusion events are essential to endothelial cell proliferation and homeostasis. Here, we show that human and mouse breast cancer cell lines also express myoferlin at various levels, and that the processes of transformation, epithelial-mesenchymal transition, and metastasis do not appear to have any effect on myoferlin expression in vitro. In vivo, we observed that solid mouse and human carcinoma tissues also express high levels of myoferlin protein. Loss-of-function studies performed in mice revealed that myoferlin gene knockdown can attenuate cancer cell proliferation in vitro and decrease tumor burden, and that accelerated tumor cell growth appears to rely on intact myoferlin-dependent membrane repair and signaling under exponential growth conditions. To our knowledge, these data provide the first evidence of myoferlin expression in solid human and mouse tumors. We have thus identified a novel membrane repair process that likely helps sustain the high growth rates characteristic of tumors, and we suggest that interfering with normal myoferlin expression and/or membrane repair and remodeling may provide therapeutically relevant antiproliferative effects.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Carcinogénesis/metabolismo , Carcinogénesis/patología , Carcinoma/metabolismo , Carcinoma/patología , Membrana Celular/patología , Proteínas de la Membrana/metabolismo , Proteínas Musculares/metabolismo , Anciano , Animales , Carcinoma/irrigación sanguínea , Línea Celular Tumoral , Membrana Celular/metabolismo , Proliferación Celular , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Ratones
4.
Blood ; 120(16): 3371-81, 2012 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-22936663

RESUMEN

Angiogenic sprouting requires that cell-cell contacts be maintained during migration of endothelial cells. Angiopoietin-1 (Ang-1) and vascular endothelial growth factor act oppositely on endothelial cell junctions. We found that Ang-1 promotes collective and directional migration and, in contrast to VEGF, induces the formation of a complex formed of atypical protein kinase C (PKC)-ζ and ß-catenin at cell-cell junctions and at the leading edge of migrating endothelial cells. This complex brings Par3, Par6, and adherens junction proteins at the front of migrating cells to locally activate Rac1 in response to Ang-1. The colocalization of PKCζ and ß-catenin at leading edge along with PKCζ-dependent stabilization of cell-cell contacts promotes directed and collective endothelial cell migration. Consistent with these results, down-regulation of PKCζ in endothelial cells alters Ang-1-induced sprouting in vitro and knockdown in developing zebrafish results in intersegmental vessel defects caused by a perturbed directionality of tip cells and by loss of cell contacts between tip and stalk cells. These results reveal that PKCζ and ß-catenin function in a complex at adherens junctions and at the leading edge of migrating endothelial cells to modulate collective and directional migration during angiogenesis.


Asunto(s)
Angiopoyetina 1/farmacología , Movimiento Celular/fisiología , Endotelio Vascular/metabolismo , Neovascularización Fisiológica/fisiología , Proteína Quinasa C/metabolismo , beta Catenina/metabolismo , Uniones Adherentes/metabolismo , Animales , Animales Modificados Genéticamente , Aorta/citología , Aorta/metabolismo , Células COS , Bovinos , Movimiento Celular/efectos de los fármacos , Polaridad Celular , Células Cultivadas , Chlorocebus aethiops , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Endotelio Vascular/citología , Técnica del Anticuerpo Fluorescente , Uniones Intercelulares/metabolismo , Microinyecciones , Cicatrización de Heridas , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo
5.
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
6.
J Exp Med ; 204(10): 2373-82, 2007 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17893196

RESUMEN

Caveolin-1 (Cav-1) is the principal structural component of caveolae organelles in smooth muscle cells, adipocytes, fibroblasts, epithelial cells, and endothelial cells (ECs). Cav-1-deficient (Cav-1 knockout [KO]) mice are viable and show increases of nitric oxide (NO) production in vasculature, cardiomyopathy, and pulmonary dysfunction. In this study, we generated EC-specific Cav-1-reconstituted (Cav-1 RC) mice and reexamined vascular, cardiac, and pulmonary phenotypes. Cav-1 KO pulmonary arteries had decreased smooth muscle contractility and increased endothelial NO synthase activation and hypotension; the latter two effects were rescued completely in Cav-1 RC mice. Cav-1 KO mice exhibited myocardial hypertrophy, pulmonary hypertension, and alveolar cell hyperproliferation caused by constitutive activation of p42/44 mitogen-activated protein kinase and Akt. Interestingly, in Cav-1 RC mice, cardiac hypertrophy and pulmonary hypertension were completely rescued, whereas alveolar hyperplasia was partially recovered because of the lack of rescue of Cav-1 in bronchiolar epithelial cells. These results provide clear physiological evidence supporting the important role of cell type-specific Cav-1 expression governing multiple phenotypes in the vasculature, heart, and lung.


Asunto(s)
Vasos Sanguíneos/metabolismo , Caveolina 1/metabolismo , Regulación de la Expresión Génica , Cardiopatías Congénitas/metabolismo , Enfermedades Pulmonares/metabolismo , Animales , Vasos Sanguíneos/patología , Peso Corporal , Caveolina 1/deficiencia , Caveolina 1/genética , Colágeno/biosíntesis , Endotelio/metabolismo , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/patología , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/patología , Enfermedades Pulmonares/genética , Enfermedades Pulmonares/patología , Ratones , Ratones Noqueados , Contracción Miocárdica , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fenotipo , Transducción de Señal
8.
Mol Ther Methods Clin Dev ; 31: 101135, 2023 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-38027064

RESUMEN

Immunotherapy of acute myeloid leukemia (AML) has been challenging because the lack of tumor-specific antigens results in "on-target, off-tumor" toxicity. To unlock the full potential of AML therapies, we used CRISPR-Cas9 to genetically ablate the myeloid protein CD33 from healthy donor hematopoietic stem and progenitor cells (HSPCs), creating tremtelectogene empogeditemcel (trem-cel). Trem-cel is a HSPC transplant product designed to provide a reconstituted hematopoietic compartment that is resistant to anti-CD33 drug cytotoxicity. Here, we describe preclinical studies and process development of clinical-scale manufacturing of trem-cel. Preclinical data showed proof-of-concept with loss of CD33 surface protein and no impact on myeloid cell differentiation or function. At clinical scale, trem-cel could be manufactured reproducibly, routinely achieving >70% CD33 editing with no effect on cell viability, differentiation, and function. Trem-cel pharmacology studies using mouse xenograft models showed long-term engraftment, multilineage differentiation, and persistence of gene editing. Toxicology assessment revealed no adverse findings, and no significant or reproducible off-target editing events. Importantly, CD33-knockout myeloid cells were resistant to the CD33-targeted agent gemtuzumab ozogamicin in vitro and in vivo. These studies supported the initiation of the first-in-human, multicenter clinical trial evaluating the safety and efficacy of trem-cel in patients with AML (NCT04849910).

9.
Cancer Cell ; 6(6): 529-31, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15607955

RESUMEN

Antiangiogenic therapy for solid tumors clearly destroys tumor vasculature and reduces tumor growth. As an unexpected bonus, drugs that neutralize VEGF signaling generate a "normalization window" for tumor vasculature. This occurs via the recruitment of pericytes to the tumor vasculature, an effect associated with the transient stabilization of vessels and improved oxygen delivery to hypoxic zones. The normalization process is mediated by angiopoietin-1 and matrix metalloproteinases and creates a window of opportunity for improved sensitivity to ionizing radiation and the delivery of chemotherapeutic drugs.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Inhibidores de la Angiogénesis/uso terapéutico , Angiopoyetina 1/metabolismo , Animales , Membrana Basal/efectos de los fármacos , Membrana Basal/metabolismo , Membrana Basal/efectos de la radiación , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/patología , Vasos Sanguíneos/efectos de la radiación , Movimiento Celular/efectos de los fármacos , Movimiento Celular/efectos de la radiación , Colagenasas/metabolismo , Terapia Combinada/métodos , Rayos gamma/uso terapéutico , Glioma/tratamiento farmacológico , Glioma/metabolismo , Glioma/radioterapia , Humanos , Ratones , Modelos Biológicos , Neoplasias/irrigación sanguínea , Neoplasias/radioterapia , Neovascularización Patológica/metabolismo , Neovascularización Patológica/radioterapia , Pericitos/efectos de los fármacos , Pericitos/efectos de la radiación , Receptor TIE-2/antagonistas & inhibidores , Factores de Tiempo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Cancer Cell ; 4(1): 31-9, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12892711

RESUMEN

Tumor vasculature is hyperpermeable to macromolecules compared to normal vasculature; however, the relationship between tumor hyperpermeability and tumor progression is poorly understood. Here we show that a cell-permeable peptide derived from caveolin-1, termed cavtratin, reduces microvascular hyperpermeability and delays tumor progression in mice. These antipermeability and antitumor actions of cavtratin occur in the absence of direct cytostatic or antiangiogenic effects. Cavtratin blocks microvascular permeability by inhibiting endothelial nitric oxide synthase (eNOS), as the antipermeability and antitumor actions of cavtratin are markedly diminished in eNOS knockout mice. Our results support the concepts that hyperpermeability of tumor blood vessels contributes to tumor progression and that blockade of eNOS may be exploited as a novel target for antitumor therapy.


Asunto(s)
Permeabilidad Capilar , Carcinoma Hepatocelular/prevención & control , Carcinoma Pulmonar de Lewis/prevención & control , Caveolinas/uso terapéutico , Neovascularización Fisiológica/fisiología , Fragmentos de Péptidos/uso terapéutico , Animales , Carcinoma Hepatocelular/irrigación sanguínea , Carcinoma Hepatocelular/patología , Carcinoma Pulmonar de Lewis/irrigación sanguínea , Carcinoma Pulmonar de Lewis/patología , Caveolina 1 , Progresión de la Enfermedad , Endotelio Vascular/citología , Inhibidores Enzimáticos/farmacología , Neoplasias Hepáticas Experimentales/irrigación sanguínea , Neoplasias Hepáticas Experimentales/patología , Neoplasias Hepáticas Experimentales/prevención & control , Neoplasias Pulmonares/irrigación sanguínea , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II , Óxido Nítrico Sintasa de Tipo III , Factor A de Crecimiento Endotelial Vascular/fisiología
11.
Mol Cell Proteomics ; 9(10): 2109-24, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20585024

RESUMEN

Caveolae are organelles abundant in the plasma membrane of many specialized cells including endothelial cells (ECs), epithelial cells, and adipocytes, and in these cells, caveolin-1 (Cav-1) is the major coat protein essential for the formation of caveolae. To identify proteins that require Cav-1 for stable incorporation into membrane raft domains, a quantitative proteomics analysis using isobaric tagging for relative and absolute quantification was performed on rafts isolated from wild-type and Cav-1-deficient mice. In three independent experiments, 117 proteins were consistently identified in membrane rafts with the largest differences in the levels of Cav-2 and in the caveola regulatory proteins Cavin-1 and Cavin-2. Because the lung is highly enriched in ECs, we validated and characterized the role of the newly described protein Cavin-1 in several cardiovascular tissues and in ECs. Cavin-1 was highly expressed in ECs lining blood vessels and in cultured ECs. Knockdown of Cavin-1 reduced the levels of Cav-1 and -2 and weakly influenced the formation of high molecular weight oligomers containing Cav-1 and -2. Cavin-1 silencing enhanced basal nitric oxide release from ECs but blocked proangiogenic phenotypes such as EC proliferation, migration, and morphogenesis in vitro. Thus, these data support an important role of Cavin-1 as a regulator of caveola function in ECs.


Asunto(s)
Caveolina 1/metabolismo , ADN Polimerasa I/metabolismo , Proteómica , Animales , Secuencia de Bases , Western Blotting , Caveolina 1/genética , Línea Celular , Movimiento Celular , Proliferación Celular , Cromatografía por Intercambio Iónico , Silenciador del Gen , Humanos , Espectrometría de Masas , Ratones , Ratones Noqueados , Microscopía Fluorescente , Óxido Nítrico/metabolismo , ARN Interferente Pequeño
12.
Haematologica ; 96(12): 1753-60, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21933862

RESUMEN

BACKGROUND: Controversy persists regarding the role of Notch signaling in myelopoiesis. We have used genetic approaches, employing two Notch zebrafish mutants deadly seven (DES) and beamter (BEA) with disrupted function of notch1a and deltaC, respectively, and Notch1a morphants to analyze the development of leukocyte populations in embryonic and mature fish. DESIGN AND METHODS: Myelomonocytes were quantified in early embryos by in situ hybridization using a myeloper-oxidase (mpx) probe. Morpholinos were used to knock down expression of Notch1a or DeltaC. Wound healing assays and/or flow cytometry were used to quantify myelomonocytes in 5-day post-fertilization (dpf) Notch mutants (BEA and DES), morphants or pu.1:GFP, mpx:GFP and fms:RFP transgenic embryos. Flow cytometry was performed on 2-3 month old mutant fish. RESULTS: The number of mpx(+) cells in embryos was reduced at 48 hpf (but not at 26 hpf) in DES compared to WT. At 5 dpf this was reflected by a reduction in the number of myelomonocytic cells found at the wound site in mutants and in Notch1a morphants. This was due to a reduced number of myelomonocytes developing rather than a deficit in the migratory ability since transient inhibition of Notch signaling using DAPT had no effect. The early deficit in myelopoiesis was maintained into later life, 2-3 month old BEA and DES fish having a decreased proportion of myelomonocytes in both the hematopoietic organ (kidney marrow) and the periphery (coelomic cavity). CONCLUSIONS: Our results indicate that defects in Notch signaling affect definitive hematopoiesis, altering myelopoiesis from the early stages of development into the adult.


Asunto(s)
Embrión no Mamífero/embriología , Proteínas de Homeodominio/metabolismo , Mielopoyesis/fisiología , Proteínas del Tejido Nervioso/metabolismo , Receptor Notch1/metabolismo , Transducción de Señal/fisiología , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Animales , Embrión no Mamífero/citología , Proteínas de Homeodominio/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/genética , Organismos Modificados Genéticamente/embriología , Organismos Modificados Genéticamente/genética , Receptor Notch1/genética , Pez Cebra/genética , Proteínas de Pez Cebra/genética
13.
J Cell Biol ; 174(3): 369-77, 2006 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-16864653

RESUMEN

Lipid modifications mediate the subcellular localization and biological activity of many proteins, including endothelial nitric oxide synthase (eNOS). This enzyme resides on the cytoplasmic aspect of the Golgi apparatus and in caveolae and is dually acylated by both N-myristoylation and S-palmitoylation. Palmitoylation-deficient mutants of eNOS release less nitric oxide (NO). We identify enzymes that palmitoylate eNOS in vivo. Transfection of human embryonic kidney 293 cells with the complementary DNA (cDNA) for eNOS and 23 cDNA clones encoding the Asp-His-His-Cys motif (DHHC) palmitoyl transferase family members showed that five clones (2, 3, 7, 8, and 21) enhanced incorporation of [3H]-palmitate into eNOS. Human endothelial cells express all five of these enzymes, which colocalize with eNOS in the Golgi and plasma membrane and interact with eNOS. Importantly, inhibition of DHHC-21 palmitoyl transferase, but not DHHC-3, in human endothelial cells reduces eNOS palmitoylation, eNOS targeting, and stimulated NO production. Collectively, our data describe five new Golgi-targeted DHHC enzymes in human endothelial cells and suggest a regulatory role of DHHC-21 in governing eNOS localization and function.


Asunto(s)
Aciltransferasas/metabolismo , Aparato de Golgi/enzimología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Ácido Palmítico/metabolismo , Acilación , Secuencias de Aminoácidos , Animales , Células COS , Membrana Celular/metabolismo , Células Cultivadas , Chlorocebus aethiops , Células Endoteliales/citología , Células Endoteliales/enzimología , Humanos , Inmunoprecipitación , Óxido Nítrico/metabolismo , Unión Proteica , Transporte de Proteínas
14.
J Pediatr Endocrinol Metab ; 24(9-10): 771-3, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22145473

RESUMEN

Pituitary abscess is a rare but potentially life-threatening infectious process. Diagnosis is challenging as symptoms are non-specific and signs of infection may be absent. We report the case of a previously healthy 17-year-old male who presented with worsening headaches, polyuria, polydipsia and no clinical signs of infection. On evaluation, he was found to have hypopituitarism with diabetes insipidus, hypothyroidism and adrenal insufficiency. An imaging study revealed a pituitary mass. He underwent transsphenoidal biopsy to rule out tumor. The abscess was drained transsphenoidally and he was treated with parental antibiotics. Magnetic resonance imaging one year later revealed a normal pituitary without any evidence of abscess or mass. He continues to require thyroid, adrenal and anti-diuretic hormone replacements. As with any pituitary lesion, prompt complete hypothalamic pituitary evaluation is essential to avoid potentially life-threatening consequences.


Asunto(s)
Absceso Encefálico/patología , Diabetes Insípida/patología , Hipopituitarismo/patología , Imagen por Resonancia Magnética , Enfermedades de la Hipófisis/patología , Adolescente , Absceso Encefálico/complicaciones , Absceso Encefálico/terapia , Diabetes Insípida/etiología , Drenaje , Humanos , Hipopituitarismo/etiología , Masculino , Enfermedades de la Hipófisis/etiología
15.
Proc Natl Acad Sci U S A ; 105(50): 20009-14, 2008 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-19060214

RESUMEN

Although COX-dependent production of prostaglandins (PGs) is known to be crucial for tumor angiogenesis and growth, the role of PGD(2) remains virtually unknown. Here we show that PGD(2) receptor (DP) deficiency enhances tumor progression accompanied by abnormal vascular expansion. In tumors, angiogenic endothelial cells highly express DP receptor, and its deficiency accelerates vascular leakage and angiogenesis. Administration of a synthetic DP agonist, BW245C, markedly suppresses tumor growth as well as tumor hyperpermeability in WT mice, but not in DP-deficient mice. In a corneal angiogenesis assay and a modified Miles assay, host DP deficiency potentiates angiogenesis and vascular hyperpermeability under COX-2-active situation, whereas exogenous administration of BW245C strongly inhibits both angiogenic properties in WT mice. In an in vitro assay, BW245C does not affect endothelial migration and tube formation, processes that are necessary for angiogenesis; however, it strongly improves endothelial barrier function via an increase in intracellular cAMP production. Our results identify PGD(2)/DP receptor as a new regulator of tumor vascular permeability, indicating DP agonism may be exploited as a potential therapy for the treatment of cancer.


Asunto(s)
Permeabilidad Capilar , Endotelio Vascular/metabolismo , Neoplasias/irrigación sanguínea , Neovascularización Patológica/metabolismo , Receptores Inmunológicos/metabolismo , Receptores de Prostaglandina/metabolismo , Animales , Permeabilidad Capilar/efectos de los fármacos , Movimiento Celular , Proliferación Celular , Neovascularización de la Córnea/inducido químicamente , AMP Cíclico/metabolismo , Endotelio Vascular/patología , Hidantoínas/farmacología , Interleucina-1beta/farmacología , Ratones , Ratones Mutantes , Receptores Inmunológicos/agonistas , Receptores Inmunológicos/genética , Receptores de Prostaglandina/agonistas , Receptores de Prostaglandina/genética , Factor A de Crecimiento Endotelial Vascular/farmacología
16.
J Clin Invest ; 116(5): 1284-91, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16670769

RESUMEN

Caveolae in endothelial cells have been implicated as plasma membrane microdomains that sense or transduce hemodynamic changes into biochemical signals that regulate vascular function. Therefore we compared long- and short-term flow-mediated mechanotransduction in vessels from WT mice, caveolin-1 knockout (Cav-1 KO) mice, and Cav-1 KO mice reconstituted with a transgene expressing Cav-1 specifically in endothelial cells (Cav-1 RC mice). Arterial remodeling during chronic changes in flow and shear stress were initially examined in these mice. Ligation of the left external carotid for 14 days to lower blood flow in the common carotid artery reduced the lumen diameter of carotid arteries from WT and Cav-1 RC mice. In Cav-1 KO mice, the decrease in blood flow did not reduce the lumen diameter but paradoxically increased wall thickness and cellular proliferation. In addition, in isolated pressurized carotid arteries, flow-mediated dilation was markedly reduced in Cav-1 KO arteries compared with those of WT mice. This impairment in response to flow was rescued by reconstituting Cav-1 into the endothelium. In conclusion, these results showed that endothelial Cav-1 and caveolae are necessary for both rapid and long-term mechanotransduction in intact blood vessels.


Asunto(s)
Caveolas/metabolismo , Caveolina 1/fisiología , Endotelio Vascular/patología , Animales , Arterias Carótidas/patología , Caveolina 1/genética , Células Endoteliales , Endotelio Vascular/metabolismo , Ratones , Ratones Noqueados , Modelos Biológicos , Transducción de Señal , Estrés Mecánico
17.
Arterioscler Thromb Vasc Biol ; 28(1): 105-11, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17975117

RESUMEN

OBJECTIVE: Heat-shock protein 90 (Hsp90) coordinates the regulation of diverse signaling proteins. We try to develop a new tool to explore the regulatory functions of Hsp90 in endothelial cells (ECs) instead of the existing chemical approaches. METHODS AND RESULTS: We designed a dominant-negative Hsp90 construct by site-direct mutagenesis of residue Asp-88 to Asn (D88N-Hsp90) based on the structure of the ATP/ADP-binding site. Recombinant wild-type Hsp90 protein binds ATP-Sepharose beads in manner inhibited by ATP or 17-AAG, a specific inhibitor for Hsp90, however the binding activity of D88N-Hsp90 was markedly reduced and the inhibitory effects of ATP or 17-AAG were negligible. The dimerization between endogenous Hsp90alpha and exogenous HA-Hsp90beta was confirmed by immunoprecipitation, however the association between eNOS and D88N-Hsp90 was less than WT-Hsp90. Furthermore, adenoviral transduction of bovine aortic ECs with D88N-Hsp90 suppressed VEGF-induced phosphorylation of Akt, eNOS, and NO release and the inhibitory effect was blocked by okadaic acid. Moreover, D88N-Hsp90 abolished VEGF-stimulated Rac activation and suppressed VEGF-induced stress fiber formation. Transduction with D88N-Hsp90 decreased growth medium mediated migration of wild-type ECs, but not Akt1(-/-) ECs suggesting that Akt is key target of Hsp90. CONCLUSIONS: Our data demonstrate that dominant-negative Hsp90 modulates endothelial cell mobility mainly through PP2A-mediated dephosphorylation of Akt and Rac activation.


Asunto(s)
Movimiento Celular/fisiología , Células Endoteliales/enzimología , Proteínas HSP90 de Choque Térmico/fisiología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico/metabolismo , Factor A de Crecimiento Endotelial Vascular/fisiología , Adenoviridae , Animales , Bovinos , Células Cultivadas , Pulmón/citología , Ratones , Mutagénesis Sitio-Dirigida , Proteínas Proto-Oncogénicas c-akt/fisiología , Transducción de Señal
18.
Cancer Res ; 67(6): 2849-56, 2007 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-17363608

RESUMEN

Caveolin-1 (Cav-1) is a major structural protein that is essential to the formation of the organelle, caveolae. Cav-1 knockout (KO) mice were observed to be completely devoid of caveolae yet they exhibit a hyperpermeable vasculature. Given the nature of the hyperpermeable Cav-1 KO endothelium, we sought to investigate if tumors grown in Cav-1 KO mice would be leaky and grow faster. Indeed, Lewis lung carcinoma cells implanted into Cav-1 KO mice had increased tumor vascular permeability, measured by Evans blue extravasation and fibrinogen deposition compared with tumors implanted into wild-type (WT) mice. Cav-1 KO mice also had significantly higher tumor growth rates, attributable to increased tumor angiogenesis and decreased tumor cell death. Furthermore, administration of an antipermeability peptide, cavtratin, was able to correct the tumor hyperpermeability as well as attenuate the increased tumor growth. Mechanistically, endothelial cells isolated from Cav-1 KO mice exhibited increased tyrosine phosphorylation on vascular endothelial growth factor (VEGF) receptor-2 (VEGFR-2) and decreased association with the adherens junction protein, VE-cadherin. Thus, the loss of Cav-1 increases tumor permeability and growth and that may relate to enhanced VEGF signaling due to lack of Cav-1 inhibition of VEGFR-2 or decreased VE-cadherin mediated VEGFR-2 phosphorylation.


Asunto(s)
Carcinoma Pulmonar de Lewis/irrigación sanguínea , Caveolina 1/deficiencia , Uniones Adherentes/metabolismo , Secuencia de Aminoácidos , Animales , Apoptosis/fisiología , Permeabilidad Capilar , Carcinoma Pulmonar de Lewis/patología , Caveolina 1/genética , Procesos de Crecimiento Celular/fisiología , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Fosforilación , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
20.
Circ Res ; 95(2): 204-9, 2004 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-15192027

RESUMEN

Functional interactions between the initial and collecting lymphatics, as well as the molecular players involved, remain elusive. In this study, we assessed the influence of nitric oxide (NO) on lymphatic fluid velocity and flow, using a mouse tail model that permits intravital microscopy and microlymphangiography. We found that NO synthase (NOS) inhibition decreased lymphatic fluid velocity in the initial lymphatics, without any effect on their morphology. Using the same model, we found a similar effect in eNOS-/- mice and in mice treated with a selective endothelial NOS (eNOS) inhibitor. Next, we uncoupled the superficial initial lymphatics from the deeper collecting lymphatics by ligating the latter and found that lymphatic fluid velocity in NOS-inhibited mice became equal to that in control animals. Surprisingly, lymphatic fluid velocity was significantly increased after ligating the collecting lymphatics, and there was a concomitant increase in injection rate and mean lymphatic vessel diameter. Our results provide the first in vivo evidence that eNOS affects function of the whole microlymphatic system and that it is regulated via the collecting lymphatics.


Asunto(s)
Sistema Linfático/fisiología , Óxido Nítrico Sintasa/fisiología , Animales , Presión Sanguínea/efectos de los fármacos , Cruzamientos Genéticos , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/farmacología , Femenino , Bombas de Infusión Implantables , Ligadura , Sistema Linfático/enzimología , Linfografía/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Óxido Nítrico/fisiología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/deficiencia , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa de Tipo II , Óxido Nítrico Sintasa de Tipo III , Reología , Cola (estructura animal) , omega-N-Metilarginina/administración & dosificación , omega-N-Metilarginina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA