Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Med Sci ; 18(2): 482-493, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33390817

RESUMEN

Focused ultrasound (FUS) is used to locally and transiently induce blood-brain barrier (BBB) permeability, allowing targeted drug delivery to the brain. The purpose of the current study is to evaluate the potential of Vasculotide to accelerate the recovery of the BBB following FUS disruption in the TgCRND8 mouse model of amyloidosis, characteristic of Alzheimer's disease (AD). Accelerating the restoration of the BBB post-FUS would represent an additional safety procedure, which could be beneficial for clinical applications. Methods: TgCRND8 mice and their non-transgenic littermates were treated with Vasculotide (250 ng, intraperitoneal) every 48 hours for 3 months. BBB permeability was induced using FUS, in presence of intravenously injected microbubbles, in TgCRND8 and non-transgenic mice, and confirmed at time 0 by MRI enhancement using the contrast agent gadolinium. BBB closure was assessed at 6, 12 and 20 hours by MRI. In a separate cohort of animals, BBB closure was assessed at 24-hours post-FUS using Evans blue injected intravenously and followed by histological evaluation. Results: Chronic Vasculotide administration significantly reduces the ultra-harmonic threshold required for FUS-induced BBB permeability in the TgCRND8 mice. In addition, Vasculotide treatment led to a faster restoration of the BBB following FUS in TgCRND8 mice. BBB closure after FUS is not significantly different between TgCRND8 and non-transgenic mice. BBB permeability was assessed by gadolinium up to 20-hours post-FUS, demonstrating 87% closure in Vasculotide treated TgCRND8 mice, as opposed to 52% in PBS treated TgCRND8 mice, 58% in PBS treated non-transgenic mice, and 74% in Vasculotide treated non-transgenic mice. In both TgCRND8 mice and non-transgenic littermates the BBB was impermeable to Evans blue dye at 24-hours post-FUS. Conclusion: Vasculotide reduces the pressure required for microbubble ultra-harmonic onset for FUS-induced BBB permeability and it accelerates BBB restoration in a mouse model of amyloidosis, suggesting its potential clinical utility to promote vascular health, plasticity and repair in AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Barrera Hematoencefálica/efectos de los fármacos , Permeabilidad Capilar/efectos de los fármacos , Fragmentos de Péptidos/administración & dosificación , Ondas Ultrasónicas/efectos adversos , Enfermedad de Alzheimer/diagnóstico , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Animales , Barrera Hematoencefálica/diagnóstico por imagen , Barrera Hematoencefálica/patología , Barrera Hematoencefálica/efectos de la radiación , Permeabilidad Capilar/efectos de la radiación , Medios de Contraste/administración & dosificación , Modelos Animales de Enfermedad , Femenino , Humanos , Inyecciones Intraperitoneales , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Transgénicos , Microburbujas
2.
Proc Natl Acad Sci U S A ; 113(9): 2472-7, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26884170

RESUMEN

Ligands of the endothelial-enriched tunica interna endothelial cell kinase 2 (Tie2) are markedly imbalanced in severe infections associated with vascular leakage, yet regulation of the receptor itself has been understudied in this context. Here, we show that TIE2 gene expression may constitute a novel vascular barrier control mechanism in diverse infections. Tie2 expression declined rapidly in wide-ranging models of leak-associated infections, including anthrax, influenza, malaria, and sepsis. Forced Tie2 suppression sufficed to attenuate barrier function and sensitize endothelium to permeability mediators. Rapid reduction of pulmonary Tie2 in otherwise healthy animals attenuated downstream kinase signaling to the barrier effector vascular endothelial (VE)-cadherin and induced vascular leakage. Compared with wild-type littermates, mice possessing one allele of Tie2 suffered more severe vascular leakage and higher mortality in two different sepsis models. Common genetic variants that influence TIE2 expression were then sought in the HapMap3 cohort. Remarkably, each of the three strongest predicted cis-acting SNPs in HapMap3 was also associated with the risk of acute respiratory distress syndrome (ARDS) in an intensive care unit cohort of 1,614 subjects. The haplotype associated with the highest TIE2 expression conferred a 28% reduction in the risk of ARDS independent of other major clinical variables, including disease severity. In contrast, the most common haplotype was associated with both the lowest TIE2 expression and 31% higher ARDS risk. Together, the results implicate common genetic variation at the TIE2 locus as a determinant of vascular leak-related clinical outcomes from common infections, suggesting new tools to identify individuals at unusual risk for deleterious complications of infection.


Asunto(s)
Permeabilidad Capilar , Infecciones/fisiopatología , Receptor TIE-2/genética , Animales , Endotelio Vascular/fisiopatología , Ratones
3.
Crit Care ; 21(1): 274, 2017 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-29132435

RESUMEN

BACKGROUND: Community-acquired pneumonia (CAP) is a significant cause of morbidity and mortality worldwide. Despite effective antimicrobial therapy, CAP can induce pulmonary endothelial hyperpermeability resulting in life-threatening lung failure due to an exaggerated host-pathogen interaction. Treatment of acute lung injury is mainly supportive because key elements of inflammation-induced barrier disruption remain undetermined. Angiopoietin-1 (Ang-1)-mediated Tie2 activation reduces, and the Ang-1 antagonist Ang-2 increases, inflammation and endothelial permeability in sepsis. Vasculotide (VT) is a polyethylene glycol-clustered Tie2-binding peptide that mimics the actions of Ang-1. The aim of our study was to experimentally test whether VT is capable of diminishing pneumonia-induced lung injury. METHODS: VT binding and phosphorylation of Tie2 were analyzed using tryptophan fluorescence spectroscopy and phospho-Tie-2 enzyme-linked immunosorbent assay. Human and murine lung endothelial cells were investigated by immunofluorescence staining and electric cell-substrate impedance sensing. Pulmonary hyperpermeability was quantified in VT-pretreated, isolated, perfused, and ventilated mouse lungs stimulated with the pneumococcal exotoxin pneumolysin (PLY). Furthermore, Streptococcus pneumoniae-infected mice were therapeutically treated with VT. RESULTS: VT showed dose-dependent binding and phosphorylation of Tie2. Pretreatment with VT protected lung endothelial cell monolayers from PLY-induced disruption. In isolated mouse lungs, VT decreased PLY-induced pulmonary permeability. Likewise, therapeutic treatment with VT of S. pneumoniae-infected mice significantly reduced pneumonia-induced hyperpermeability. However, effects by VT on the pulmonary or systemic inflammatory response were not observed. CONCLUSIONS: VT promoted pulmonary endothelial stability and reduced lung permeability in different models of pneumococcal pneumonia. Thus, VT may provide a novel therapeutic perspective for reduction of permeability in pneumococcal pneumonia-induced lung injury.


Asunto(s)
Permeabilidad Capilar/efectos de los fármacos , Pulmón/efectos de los fármacos , Fragmentos de Péptidos/farmacocinética , Animales , Infecciones Comunitarias Adquiridas/tratamiento farmacológico , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Femenino , Ratones , Ratones Endogámicos C57BL , Fragmentos de Péptidos/uso terapéutico , Neumonía Neumocócica/tratamiento farmacológico , Espectrometría de Fluorescencia/métodos , Estadísticas no Paramétricas , Streptococcus pneumoniae/metabolismo , Streptococcus pneumoniae/patogenicidad
4.
Proc Natl Acad Sci U S A ; 109(18): 6993-8, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22509029

RESUMEN

Vascular smooth muscle cells (VSMC) have been suggested to arise from various developmental sources during embryogenesis, depending on the vascular bed. However, evidence also points to a common subpopulation of vascular progenitor cells predisposed to VSMC fate in the embryo. In the present study, we use binary transgenic reporter mice to identify a Tie1(+)CD31(dim)vascular endothelial (VE)-cadherin(-)CD45(-) precursor that gives rise to VSMC in vivo in all vascular beds examined. This precursor does not represent a mature endothelial cell, because a VE-cadherin promoter-driven reporter shows no expression in VSMC during murine development. Blockade of Notch signaling in the Tie1(+) precursor cell, but not the VE-cadherin(+) endothelial cell, decreases VSMC investment of developing arteries, leading to localized hemorrhage in the embryo at the time of vascular maturation. However, Notch signaling is not required in the Tie1(+) precursor after establishment of a stable artery. Thus, Notch activity is required in the differentiation of a Tie1(+) local precursor to VSMC in a spatiotemporal fashion across all vascular beds.


Asunto(s)
Diferenciación Celular/fisiología , Mioblastos del Músculo Liso/citología , Mioblastos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Neovascularización Fisiológica , Receptores Notch/metabolismo , Animales , Antígenos CD/genética , Arterias/embriología , Arterias/crecimiento & desarrollo , Arterias/metabolismo , Secuencia de Bases , Cadherinas/deficiencia , Cadherinas/genética , Diferenciación Celular/genética , Cartilla de ADN/genética , Femenino , Antígenos Comunes de Leucocito/deficiencia , Antígenos Comunes de Leucocito/genética , Ratones , Ratones Transgénicos , Neovascularización Fisiológica/genética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Embarazo , Receptor TIE-1/metabolismo , Receptores Notch/antagonistas & inhibidores , Transducción de Señal
5.
Stem Cells ; 31(2): 293-304, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23135963

RESUMEN

The clinical application of hematopoietic progenitor cell-based therapies for the treatment of hematological diseases is hindered by current protocols, which are cumbersome and have limited efficacy to augment the progenitor cell pool. We report that inhibition of T-cell protein tyrosine phosphatase (TC-PTP), an enzyme involved in the regulation of cytokine signaling, through gene knockout results in a ninefold increase in the number of hematopoietic progenitors in murine bone marrow (BM). This effect could be reproduced using a short (48 hours) treatment with a pharmacological inhibitor of TC-PTP in murine BM, as well as in human BM, peripheral blood, and cord blood. We also demonstrate that the ex vivo use of TC-PTP inhibitor only provides a temporary effect on stem cells and did not alter their capacity to reconstitute all hematopoietic components in vivo. We establish that one of the mechanisms whereby inhibition of TC-PTP mediates its effects involves the interleukin-18 (IL-18) signaling pathway, leading to increased production of IL-12 and interferon-gamma by progenitor cells. Together, our results reveal a previously unrecognized role for IL-18 in contributing to the augmentation of the stem cell pool and provide a novel and simple method to rapidly expand progenitor cells from a variety of sources using a pharmacological compound.


Asunto(s)
Células de la Médula Ósea/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Células Madre Hematopoyéticas/efectos de los fármacos , Interleucina-18/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 2/antagonistas & inhibidores , Tiazolidinas/farmacología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Recuento de Células , Sangre Fetal/citología , Sangre Fetal/efectos de los fármacos , Sangre Fetal/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Movilización de Célula Madre Hematopoyética , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Interleucina-12/biosíntesis , Interleucina-12/inmunología , Interleucina-18/inmunología , Interleucina-18/farmacología , Ratones , Ratones Transgénicos , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 2/metabolismo , ARN Interferente Pequeño/genética , Transducción de Señal/efectos de los fármacos
6.
BMC Cancer ; 14: 614, 2014 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-25159192

RESUMEN

BACKGROUND: Most cancer patients are treated with radiotherapy, but the treatment can also damage the surrounding normal tissue. Acute skin damage from cancer radiotherapy diminishes patients' quality of life, yet effective biological interventions for this damage are lacking. Protecting microvascular endothelial cells from irradiation-induced perturbations is emerging as a targeted damage-reduction strategy. Since Angiopoetin-1 signaling through the Tie2 receptor on endothelial cells opposes microvascular perturbations in other disease contexts, we used a preclinical Angiopoietin-1 mimic called Vasculotide to investigate its effect on skin radiation toxicity using a preclinical model. METHODS: Athymic mice were treated intraperitoneally with saline or Vasculotide and their flank skin was irradiated with a single large dose of ionizing radiation. Acute cutaneous damage and wound healing were evaluated by clinical skin grading, histology and immunostaining. Diffuse reflectance optical spectroscopy, myeloperoxidase-dependent bioluminescence imaging of neutrophils and a serum cytokine array were used to assess inflammation. Microvascular endothelial cell response to radiation was tested with in vitro clonogenic and Matrigel tubule formation assays. Tumour xenograft growth delay experiments were also performed. Appreciable differences between treatment groups were assessed mainly using parametric and non-parametric statistical tests comparing areas under curves, followed by post-hoc comparisons. RESULTS: In vivo, different schedules of Vasculotide treatment reduced the size of the irradiation-induced wound. Although skin damage scores remained similar on individual days, Vasculotide administered post irradiation resulted in less skin damage overall. Vasculotide alleviated irradiation-induced inflammation in the form of reduced levels of oxygenated hemoglobin, myeloperoxidase bioluminescence and chemokine MIP-2. Surprisingly, Vasculotide-treated animals also had higher microvascular endothelial cell density in wound granulation tissue. In vitro, Vasculotide enhanced the survival and function of irradiated endothelial cells. CONCLUSIONS: Vasculotide administration reduces acute skin radiation damage in mice, and may do so by affecting several biological processes. This radiation protection approach may have clinical impact for cancer radiotherapy patients by reducing the severity of their acute skin radiation damage.


Asunto(s)
Angiopoyetina 1/química , Materiales Biomiméticos/administración & dosificación , Péptidos/administración & dosificación , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Piel/efectos de los fármacos , Piel/patología , Cicatrización de Heridas/efectos de los fármacos , Animales , Materiales Biomiméticos/uso terapéutico , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Citocinas/sangre , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/efectos de la radiación , Humanos , Ratones , Ratones Desnudos , Neovascularización Fisiológica/efectos de los fármacos , Péptidos/uso terapéutico , Traumatismos Experimentales por Radiación/patología , Radiación Ionizante
7.
J Surg Res ; 187(1): 43-52, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24176205

RESUMEN

BACKGROUND: South Asian ethnicity is an independent risk factor for mortality after coronary artery bypass. We tested the hypothesis that this risk results from a greater inflammatory response to cardiopulmonary bypass (CPB). METHODS: This was a single-site prospective cohort study. We compared the inflammatory response to CPB in 20 Caucasians and 17 South Asians undergoing isolated coronary artery bypass grafting surgery. RESULTS: Plasma levels of proinflammatory cytokines (interleukin [IL]-6, IL-8, IL-12, interferon gamma, and tumor necrosis factor) and anti-inflammatory mediators (IL-10 and soluble TNF receptor I) were measured. The Toll-like receptor (TLR) signaling pathway was examined in peripheral blood monocytes by flow cytometry, measuring surface expression of TLR2, TLR4, and coreceptor CD14 and activation of downstream messenger molecules (interleukin-1 receptor-associated kinase 4, nuclear factor kappa from B cells (NF-κB), c-Jun amino-terminal kinase, p38 mitogen-activated protein kinase, and Protein Kinase B). South Asians had persistently higher plasma levels of IL-6 and exhibited increased TLR signaling through the p38 mitogen-activated protein kinase and Protein Kinase B pathways in inflammatory monocytes after CPB. This increased inflammatory response was paralleled clinically by a higher sequential organ failure assessment score (5.1 ± 1.4 versus 1.5 ± 1.6, P = 0.027) and prolonged cardiovascular system failure (23.5% versus 0%) 48 h after CPB. CONCLUSIONS: South Asians develop an exacerbated systemic inflammatory response after CPB, which may contribute to the higher morbidity and mortality associated with coronary artery bypass in this population. These patients may benefit from targeted anti-inflammatory therapies designed to mitigate the adverse consequences resulting from this response.


Asunto(s)
Puente Cardiopulmonar/efectos adversos , Síndrome de Respuesta Inflamatoria Sistémica/inmunología , Receptor Toll-Like 2/inmunología , Receptor Toll-Like 4/inmunología , Anciano , Asia Sudoriental , Pueblo Asiatico/estadística & datos numéricos , Biomarcadores/metabolismo , Puente Cardiopulmonar/mortalidad , Puente Cardiopulmonar/estadística & datos numéricos , Sistema Cardiovascular/inmunología , Citocinas/sangre , Citocinas/inmunología , Femenino , Humanos , Inflamación/etnología , Inflamación/mortalidad , Masculino , Persona de Mediana Edad , Monocitos/inmunología , Monocitos/metabolismo , Estudios Prospectivos , Transducción de Señal/inmunología , Síndrome de Respuesta Inflamatoria Sistémica/etnología , Síndrome de Respuesta Inflamatoria Sistémica/mortalidad , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo , Población Blanca/estadística & datos numéricos
8.
Dev Biol ; 367(1): 40-54, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22546694

RESUMEN

There is a reciprocal interaction between pancreatic islet cells and vascular endothelial cells (EC) in which EC-derived signals promote islet cell differentiation and islet development while islet cell-derived angiogenic factors promote EC recruitment and extensive islet vascularization. To examine the role of angiogenic factors in the coordinated development of islets and their associated vessels, we used a "tet-on" inducible system (mice expressing rat insulin promoter-reverse tetracycline activator transgene and a tet-operon-angiogenic factor transgene) to increase the ß cell production of vascular endothelial growth factor-A (VEGF-A), angiopoietin-1 (Ang1), or angiopoietin-2 (Ang2) during islet cell differentiation and islet development. In VEGF-A overexpressing embryos, ECs began to accumulate around epithelial tubes residing in the central region of the developing pancreas (associated with endocrine cells) as early as embryonic day 12.5 (E12.5) and increased dramatically by E16.5. While α and ß cells formed islet cell clusters in control embryos at E16.5, the increased EC population perturbed endocrine cell differentiation and islet cell clustering in VEGF-A overexpressing embryos. With continued overexpression of VEGF-A, α and ß cells became scattered, remained adjacent to ductal structures, and never coalesced into islets, resulting in a reduction in ß cell proliferation and ß cell mass at postnatal day 1. A similar impact on islet morphology was observed when VEGF-A was overexpressed in ß cells during the postnatal period. In contrast, increased expression of Ang1 or Ang2 in ß cells in developing or adult islets did not alter islet differentiation, development, or morphology, but altered islet EC ultrastructure. These data indicate that (1) increased EC number does not promote, but actually impairs ß cell proliferation and islet formation; (2) the level of VEGF-A production by islet endocrine cells is critical for islet vascularization during development and postnatally; (3) angiopoietin-Tie2 signaling in endothelial cells does not have a crucial role in the development or maintenance of islet vascularization.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/citología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Angiopoyetina 1/metabolismo , Angiopoyetina 2/metabolismo , Animales , Recuento de Células , Células Endoteliales/metabolismo , Islotes Pancreáticos/irrigación sanguínea , Islotes Pancreáticos/metabolismo , Ratones
9.
Blood ; 118(18): 5050-9, 2011 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-21868579

RESUMEN

In human inflammatory diseases, we identified endothelial angiopoietin-2 (Ang-2) expression to be strongly associated with inflammations mediated by myeloid cells but not lymphocytes. To identify the underlying mechanism, we made use of a transgenic mouse model with inducible endothelial cell-specific expression of Ang-2. In this model, in the absence of inflammatory stimuli, long-term expression of Ang-2 led to a time-dependent accumulation of myeloid cells in numerous organs, suggesting that Ang-2 is sufficient to recruit myeloid cells. In models of acute inflammation, such as delayed-type hypersensitivity and peritonitis, Ang-2 transgenic animals showed an increased responsiveness. Intravital fluorescence video microscopy revealed augmented cell adhesion as an underlying event. Consequently, we demonstrated that Ang-2 is able to induce strong monocyte adhesion under shear in vitro, which could be blocked by antibodies to ß2-integrin. Taken together, our results describe Ang-2 as a novel, endothelial-derived regulator of myeloid cell infiltration that modulates ß2-integrin-mediated adhesion in a paracrine manner.


Asunto(s)
Angiopoyetina 2/fisiología , Antígenos CD18/fisiología , Movimiento Celular/genética , Células Mieloides/fisiología , Adulto , Angiopoyetina 2/genética , Angiopoyetina 2/metabolismo , Animales , Antígenos CD18/genética , Antígenos CD18/metabolismo , Adhesión Celular/genética , Células Cultivadas , Predisposición Genética a la Enfermedad , Humanos , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Ratones , Ratones Transgénicos , Monocitos/metabolismo , Monocitos/fisiología , Células Mieloides/metabolismo , Células Progenitoras Mieloides/metabolismo , Células Progenitoras Mieloides/fisiología , Transducción de Señal/genética , Transducción de Señal/fisiología
10.
Cancer Cell ; 7(1): 101-11, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15652753

RESUMEN

Development of antiangiogenic therapies would be significantly facilitated by quantitative surrogate pharmacodynamic markers. Circulating peripheral blood endothelial cells (CECs) and/or their putative progenitor subset (CEPs) have been proposed but not yet fully validated for this purpose. Herein, we provide such validation by showing a striking correlation between highly genetically heterogeneous bFGF- or VEGF-induced angiogenesis and intrinsic CEC or CEP levels measured by flow cytometry, among eight different inbred mouse strains. Moreover, studies using genetically altered mice showed that levels of these cells are affected by regulators of angiogenesis, including VEGF, Tie-2, and thrombospondin-1. Finally, treatment with a targeted VEGFR-2 antibody caused a dose-dependent reduction in viable CEPs that precisely paralleled its previously and empirically determined antitumor activity.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Biomarcadores , Células Endoteliales/fisiología , Neovascularización Patológica , Células Madre/fisiología , Inhibidores de la Angiogénesis/metabolismo , Animales , Bioensayo/métodos , Supervivencia Celular , Relación Dosis-Respuesta a Droga , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Citometría de Flujo , Humanos , Masculino , Ratones , Ratones Endogámicos , Ratones Transgénicos , Fenotipo , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Células Madre/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
11.
Blood ; 116(3): 428-36, 2010 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-20445019

RESUMEN

The activation of Fli-1, an Ets transcription factor, is the critical genetic event in Friend murine leukemia virus (F-MuLV)-induced erythroleukemia. Fli-1 overexpression leads to erythropoietin-dependent erythroblast proliferation, enhanced survival, and inhibition of terminal differentiation, through activation of the Ras pathway. However, the mechanism by which Fli-1 activates this signal transduction pathway has yet to be identified. Down-regulation of the Src homology 2 (SH2) domain-containing inositol-5-phosphatase-1 (SHIP-1) is associated with erythropoietin-stimulated erythroleukemic cells and correlates with increased proliferation of transformed cells. In this study, we have shown that F-MuLV-infected SHIP-1 knockout mice display accelerated erythroleukemia progression. In addition, RNA interference (RNAi)-mediated suppression of SHIP-1 in erythroleukemia cells activates the phosphatidylinositol 3-kinase (PI 3-K) and extracellular signal-regulated kinase/mitogen-activated protein kinase (ERK/MAPK) pathways, blocks erythroid differentiation, accelerates erythropoietin-induced proliferation, and leads to PI 3-K-dependent Fli-1 up-regulation. Chromatin immunoprecipitation and luciferase assays confirmed that Fli-1 binds directly to an Ets DNA binding site within the SHIP-1 promoter and suppresses SHIP-1 transcription. These data provide evidence to suggest that SHIP-1 is a direct Fli-1 target, SHIP-1 and Fli-1 regulate each other in a negative feedback loop, and the suppression of SHIP-1 by Fli-1 plays an important role in the transformation of erythroid progenitors by F-MuLV.


Asunto(s)
Leucemia Eritroblástica Aguda/etiología , Monoéster Fosfórico Hidrolasas/metabolismo , Proteína Proto-Oncogénica c-fli-1/metabolismo , Animales , Secuencia de Bases , Sitios de Unión/genética , Línea Celular , ADN/genética , ADN/metabolismo , Cartilla de ADN/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Retroalimentación Fisiológica , Virus de la Leucemia Murina de Friend/patogenicidad , Humanos , Inositol Polifosfato 5-Fosfatasas , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/virología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Modelos Biológicos , Datos de Secuencia Molecular , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas , Monoéster Fosfórico Hidrolasas/antagonistas & inhibidores , Monoéster Fosfórico Hidrolasas/deficiencia , Monoéster Fosfórico Hidrolasas/genética , Fosforilación , Regiones Promotoras Genéticas , Proteína Proto-Oncogénica c-fli-1/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/genética
12.
Acta Neuropathol ; 124(6): 763-75, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23143192

RESUMEN

The cellular and molecular mechanisms of tumor angiogenesis and its prospects for anti-angiogenic cancer therapy are major issues in almost all current concepts of both cancer biology and targeted cancer therapy. Currently, (1) sprouting angiogenesis, (2) vascular co-option, (3) vascular intussusception, (4) vasculogenic mimicry, (5) bone marrow-derived vasculogenesis, (6) cancer stem-like cell-derived vasculogenesis and (7) myeloid cell-driven angiogenesis are all considered to contribute to tumor angiogenesis. Many of these processes have been described in developmental angiogenesis; however, the relative contribution and relevance of these in human brain cancer remain unclear. Preclinical tumor models support a role for sprouting angiogenesis, vascular co-option and myeloid cell-derived angiogenesis in glioma vascularization, whereas a role for the other four mechanisms remains controversial and rather enigmatic. The anti-angiogenesis drug Avastin (Bevacizumab), which targets VEGF, has become one of the most popular cancer drugs in the world. Anti-angiogenic therapy may lead to vascular normalization and as such facilitate conventional cytotoxic chemotherapy. However, preclinical and clinical studies suggest that anti-VEGF therapy using bevacizumab may also lead to a pro-migratory phenotype in therapy resistant glioblastomas and thus actively promote tumor invasion and recurrent tumor growth. This review focusses on (1) mechanisms of tumor angiogenesis in human malignant glioma that are of particular relevance for targeted therapy and (2) controversial issues in tumor angiogenesis such as cancer stem-like cell-derived vasculogenesis and bone-marrow-derived vasculogenesis.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Glioma/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/patología , Glioma/irrigación sanguínea , Glioma/patología , Humanos , Transducción de Señal/fisiología
13.
Crit Care ; 15(5): R261, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22040774

RESUMEN

INTRODUCTION: Angiopoietin-1 (Angpt1), the natural agonist ligand for the endothelial Tie2 receptor, is a non-redundant endothelial survival and vascular stabilization factor that reduces endothelial permeability and inhibits leukocyte-endothelium interactions. Here we evaluate the efficacy of a novel polyethylene glycol (PEG)-clustered Tie2 agonist peptide, Vasculotide (VT), to protect against vascular leakage and mortality in a murine model of polymicrobial abdominal sepsis. METHODS: Polymicrobial abdominal sepsis in C57BL6 mice was induced by cecal-ligation-and-puncture (CLP). Mice were treated with different dosages of VT or equal volume of phosphate-buffered saline (PBS). Sham-operated animals served as time-matched controls. RESULTS: Systemic administration of VT induced long-lasting Tie2 activation in vivo. VT protected against sepsis-induced endothelial barrier dysfunction, as evidenced by attenuation of vascular leakage and leukocyte transmigration into the peritoneal cavity. Histological analysis revealed that VT treatment ameliorated leukocyte infiltration in kidneys of septic mice, probably due to reduced endothelial adhesion molecule expression. VT-driven effects were associated with significantly improved organ function and reduced circulating cytokine levels. The endothelial-specific action of VT was supported by additional in vitro studies showing no effect of VT on either cytokine release from isolated peritoneal macrophages, or migratory capacity of isolated neutrophils. Finally, administration of VT pre-CLP (Hazard Ratio 0.39 [95% Confidence interval 0.19-0.81] P < 0.001) and post-CLP reduced mortality in septic mice (HR 0.22 [95% CI 0.06-0.83] P < 0.05). CONCLUSIONS: We provide proof of principle in support of the efficacious use of PEGylated VT, a drug-like Tie2 receptor agonist, to counteract microvascular endothelial barrier dysfunction and reduce mortality in a clinically relevant murine sepsis model. Further studies are needed to pave the road for clinical application of this therapeutic concept.


Asunto(s)
Permeabilidad Capilar/fisiología , Endotelio Vascular/metabolismo , Proteínas Tirosina Quinasas Receptoras/agonistas , Proteínas Tirosina Quinasas Receptoras/fisiología , Sepsis/mortalidad , Sepsis/prevención & control , Abdomen/patología , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Péptidos/uso terapéutico , Cavidad Peritoneal/patología , Polietilenglicoles/uso terapéutico , Proteínas Tirosina Quinasas Receptoras/síntesis química , Receptor TIE-2 , Sepsis/metabolismo
14.
Proteomics ; 10(8): 1658-72, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20186751

RESUMEN

Differential protein profiling by 2-D PAGE is generally useful in biomarker discovery, proteome analysis and routine sample preparation prior to analysis by MS. The goal of this study was to compare 2-D PAGE-resolved protein profile of lymphatic endothelial cells to those of venous, and arterial endothelial cells isolated from lymphatic and blood vessels of bovine mesentery (bm). Three 2-D PAGE electrophoretograms were produced for each of the three cell types and quantitatively analyzed. Protein identification by LC-MS/MS was performed to identify 39 proteins found to be present at statistically significantly different levels in the three cell types (p<0.05). Most of the 39 proteins have not been previously reported in EC proteomic studies of 2-D PAGE electrophoretograms. Three proteins, HSPA1B (HSP70 family member), HSPB1 (HSP27 family member), and UBE2D3 (a member of E2 ubiquitin-conjugating enzymes) found to be at highest levels in bm arterial endothelial cells, bm venous endothelial cells, and bm lymphatic endothelial cells, respectively, were validated by immunoblotting with appropriate antibodies. The lack of substantial overlap between our results and those of other groups' comparative studies are discussed. Functional implications of differences in levels of various proteins identified in the three cell types are also discussed.


Asunto(s)
Arterias/química , Células Endoteliales/química , Vasos Linfáticos/química , Mesenterio/química , Proteoma/análisis , Venas/química , Animales , Bovinos , Células Cultivadas , Proteómica
15.
BMC Dev Biol ; 10: 72, 2010 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-20584329

RESUMEN

BACKGROUND: In vivo studies demonstrate that the Prox1 transcription factor plays a critical role in the development of the early lymphatic system. Upon Prox1 expression, early lymphatic endothelial cells differentiate from the cardinal vein and begin to express lymphatic markers such as VEGFR-3, LYVE-1 and Podoplanin. Subsequent in vitro studies have found that differentiated vascular endothelial cells can be reprogrammed by Prox1 to express a lymphatic gene profile, suggesting that Prox1 can initiate the expression of a unique gene signature during lymphangiogenesis. While the in vitro data suggest that gene reprogramming occurs upon Prox1 expression, it is not clear if this is a direct result of Prox1 in vascular endothelial cells in vivo. RESULTS: Overexpression of Prox1 in vascular endothelial cells during embryonic development results in the reprogramming of genes to that of a more lymphatic signature. Consequent to this overexpression, embryos suffer from gross edema that results in embryonic lethality at E13.5. Furthermore, hemorrhaging and anemia is apparent along with clear defects in lymph sac development. Alterations in junctional proteins resulting in an increase in vascular permeability upon Prox1 overexpression may contribute to the complications found during embryonic development. CONCLUSION: We present a novel mouse model that addresses the importance of Prox1 in early embryonic lymphangiogenesis. It is clear that there needs to be a measured pattern of expression of Prox1 during embryonic development. Furthermore, Prox1 reprograms vascular endothelial cells in vivo by creating a molecular signature to that of a lymphatic endothelial cell.


Asunto(s)
Embrión de Mamíferos/metabolismo , Células Endoteliales/metabolismo , Proteínas de Homeodominio/metabolismo , Linfangiogénesis , Proteínas Supresoras de Tumor/metabolismo , Animales , Diferenciación Celular , Embrión de Mamíferos/citología , Perfilación de la Expresión Génica , Ratones , Receptor TIE-1/genética
16.
Cell Commun Signal ; 8: 30, 2010 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-20973951

RESUMEN

BACKGROUND: Growth factor receptor bound (Grb) proteins 7, 10 and 14 are a family of structurally related multi-domain adaptor proteins involved in a variety of biological processes. Grb7, 10 and 14 are known to become serine and/or threonine phosphorylated in response to growth factor (GF) stimulation. Grb7 and 10 have also been shown to become tyrosine phosphorylated under certain conditions. Under experimental conditions Grb7 is tyrosine phosphorylated by the Tie2/Tie-2/Tek angiogenic receptor tyrosine kinase (RTK). Furthermore, Grb14 has also been shown to interact with Tie2, however tyrosine phosphorylation of this Grb family member has yet to be reported. RESULTS: Here we report for the first time tyrosine phosphorylation of Grb14. This phosphorylation requires a kinase competent Tie2 as well as intact tyrosines 1100 and 1106 (Y1100 and Y1106) on the receptor. Furthermore, a complete SH2 domain on Grb14 is required for Grb14 tyrosine phosphorylation by Tie2. Grb14 was also able to become tyrosine phosphorylated in primary endothelial cells when treated with a soluble and potent variant of the Tie2 ligand, cartilage oligomeric matrix protein (COMP) Ang1. CONCLUSION: Our results show that Grb14, like its family members Grb7 and Grb10, is able to be tyrosine phosphorylated. Furthermore, our data indicate a role for Grb14 in endothelial signaling downstream of the Tie2 receptor.

17.
BMC Neurol ; 10: 114, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-21078165

RESUMEN

BACKGROUND: The Blood Brain Barrier (BBB) maintains the homeostasis of central nervous system by preventing the free passage of macromolecules from the systemic circulation into the brain. This normal physiological function of the BBB presents a challenge for delivery of therapeutic compounds into the brain. Recent studies have shown that the application of focused ultrasound together with ultrasound contrast agent (microbubbles) temporarily increases the permeability of the BBB. This effect is associated with breakdown of tight junctions, the structures that regulate the paracellular permeability of the endothelial cell layer. The influence of this ultrasound effect on the activation of intracellular signaling proteins is currently not well understood. Therefore, the aim of this study was to investigate the activation of cell survival signaling molecules in response to ultrasound-mediated BBB opening; METHODS: The BBB was disrupted in two four-spot lines (1-1.5 mm spacing) along the right hemisphere of rat brain with ultrasound beams (0.3 MPa, 120 s, 10 ms bursts, repetition frequency = 1 Hz) in the presence Definity microbubbles. Contrast-enhanced MRI images were acquired to assess the extent of BBB opening upon which the animals were sacrificed and the brains removed and processed for biochemical and immunohistochemical analyses; RESULTS: Immunoblotting of sonicated brain lysates resolved by SDS-PAGE demonstrated an increase in phosphorylation of Akt and its downstream signaling molecule, GSK3ß, while the phosphorylation of MAPK remained unchanged. The elevated levels of pAkt and pGSK3ß are still evident after 24 hours post-sonication, a time point where the integrity of the BBB is known to be re-established. Furthermore, immunofluoresence staining localized this increase in pAkt and pGSK3ß levels to neuronal cells flanking the region of the disrupted BBB; CONCLUSIONS: Our data demonstrates that ultrasound-mediated BBB disruption causes an activation of the Akt signaling pathway in neuronal cells surrounding the disrupted BBB.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Microburbujas , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Análisis de Varianza , Animales , Barrera Hematoencefálica/diagnóstico por imagen , Western Blotting , Medios de Contraste/metabolismo , Técnica del Anticuerpo Fluorescente , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Inmunoprecipitación , Imagen por Resonancia Magnética , Masculino , Fosforilación , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Sonicación , Factores de Tiempo , Ultrasonografía
18.
Biochem J ; 423(3): 375-80, 2009 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-19689429

RESUMEN

Tie2 [where 'Tie' is an acronym from tyrosine kinase with Ig and EGF (epidermal growth factor) homology domains] is a receptor tyrosine kinase expressed predominantly on the surface of endothelial cells. Activated by its ligands, the angiopoietins, Tie2 initiates signalling pathways that modulate vascular stability and angiogenesis. Deletion of either the Tie2 or Ang1 (angiopoietin-1) gene in mice results in lethal vascular defects, signifying their importance in vascular development. The mechanism employed by the Tie2 signalling machinery to attenuate or cause receptor trafficking is not well defined. Stimulation of Tie2-expressing cells with Ang1 results in its ubiquitylation, suggesting that this may provide the necessary signal for receptor turnover. Using a candidate molecule approach, we demonstrate that Tie2 co-immunoprecipitates with c-Cbl in an Ang1-dependent manner and its ubiquitylation can be inhibited by the dominant-interfering molecule v-Cbl (a viral form of c-Cbl that contains only the tyrosine kinase-binding domain region). Inhibition of the Tie2-Cbl interaction by overexpression of v-Cbl blocks ligand-induced Tie2 internalization and degradation. In summary, our results illustrate that c-Cbl interacts with the Tie2 signalling complex in a stimulation-dependent manner, and that this interaction is required for Tie2 ubiquitylation, internalization and degradation.


Asunto(s)
Angiopoyetina 1/metabolismo , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Receptor TIE-2/metabolismo , Transducción de Señal/fisiología , Ubiquitinación/fisiología , Angiopoyetina 1/genética , Angiopoyetina 1/farmacología , Línea Celular , Humanos , Proteína Oncogénica v-cbl/genética , Proteína Oncogénica v-cbl/metabolismo , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/fisiología , Proteínas Proto-Oncogénicas c-cbl/genética , Receptor TIE-2/genética , Transducción de Señal/efectos de los fármacos , Ubiquitinación/efectos de los fármacos
19.
Histochem Cell Biol ; 132(2): 225-37, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19424712

RESUMEN

The tyrosine kinase receptor Tie2 is expressed on endothelial cells, and together with its ligand angiopoietin-1 (Ang1), is important for angiogenesis and vascular stability. Upon activation by Ang1, Tie2 is rapidly internalized and degraded, a mechanism most likely necessary to attenuate receptor activity. Using immunogold electron microscopy, we show that on the surface of endothelial cells, Tie2 is arranged in variably sized clusters containing dimers and higher order oligomers. Clusters of Tie2 were expressed on the apical and basolateral plasma membranes, and on the tips of microvilli. Upon activation by Ang1, Tie2 co-localized with the clathrin heavy chain at the apical and basolateral plasma membranes and within endothelial cells indicating that Tie2 internalizes through clathrin-coated pits. Inhibiting cellular endocytosis by depleting cellular potassium or by acidifying the cytosol blocked the internalization of Tie2 in response to Ang1. Our results suggest that one pathway mediating the internalization of Tie2 in response to Ang1 is through clathrin-coated pits.


Asunto(s)
Angiopoyetina 1/metabolismo , Invaginaciones Cubiertas de la Membrana Celular/enzimología , Endotelio Vascular/enzimología , Receptor TIE-2/metabolismo , Angiopoyetina 1/agonistas , Células Cultivadas , Invaginaciones Cubiertas de la Membrana Celular/ultraestructura , Endotelio Vascular/ultraestructura , Humanos
20.
Am J Pathol ; 173(6): 1891-901, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18988807

RESUMEN

Vascular endothelial growth factor-C (VEGF-C) is the quintessential lymphangiogenic growth factor that is required for the development of the lymphatic system and is capable of stimulating lymphangiogenesis in adults by activating its receptor, VEGFR-3. Although VEGF-C is a major candidate molecule for the development of prolymphangiogenic therapy for defective lymphatic vessels in lymphedema, the stability of lymph vessels generated by exogenous VEGF-C administration is not currently known. We studied VEGF-C-stimulated lymphangiogenesis in inducible transgenic mouse models in which growth factor expression can be spatially and temporally controlled without side effects, such as inflammation. VEGF-C induction in adult mouse skin for 1 to 2 weeks caused robust lymphatic hyperplasia that persisted for at least 6 months. VEGF-C induced lymphangiogenesis in numerous tissues and organs when expressed in the vascular endothelium in either neonates or adult mice. Very few or no effects were observed in either blood vessels or collecting lymph vessels. Additionally, VEGF-C stimulated lymphangiogenesis in embryos after the onset of lymphatic vessel development. Strikingly, a strong angiogenic effect was observed after VEGF-C induction in vascular endothelium at any point before embryonic day 16.5. Our results indicate that blood vessels can undergo VEGF-C-induced angiogenesis even after down-regulation of VEGFR-3 in embryos; however, transient VEGF-C expression in adults can induce long-lasting lymphatic hyperplasia with no obvious side effects on the blood vasculature.


Asunto(s)
Embrión de Mamíferos , Hiperplasia/patología , Linfangiogénesis/fisiología , Vasos Linfáticos , Transgenes , Factor C de Crecimiento Endotelial Vascular/metabolismo , Animales , Biomarcadores/metabolismo , Embrión de Mamíferos/anatomía & histología , Embrión de Mamíferos/fisiología , Células Endoteliales/citología , Células Endoteliales/metabolismo , Femenino , Glicoproteínas/metabolismo , Humanos , Hiperplasia/metabolismo , Vasos Linfáticos/metabolismo , Vasos Linfáticos/patología , Proteínas de Transporte de Membrana , Ratones , Ratones Transgénicos , Neovascularización Fisiológica , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Embarazo , Tirosina/metabolismo , Factor C de Crecimiento Endotelial Vascular/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA