Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Nat Commun ; 13(1): 2760, 2022 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-35589749

RESUMEN

Autophagy has vasculoprotective roles, but whether and how it regulates lymphatic endothelial cells (LEC) homeostasis and lymphangiogenesis is unknown. Here, we show that genetic deficiency of autophagy in LEC impairs responses to VEGF-C and injury-driven corneal lymphangiogenesis. Autophagy loss in LEC compromises the expression of main effectors of LEC identity, like VEGFR3, affects mitochondrial dynamics and causes an accumulation of lipid droplets (LDs) in vitro and in vivo. When lipophagy is impaired, mitochondrial ATP production, fatty acid oxidation, acetyl-CoA/CoA ratio and expression of lymphangiogenic PROX1 target genes are dwindled. Enforcing mitochondria fusion by silencing dynamin-related-protein 1 (DRP1) in autophagy-deficient LEC fails to restore LDs turnover and lymphatic gene expression, whereas supplementing the fatty acid precursor acetate rescues VEGFR3 levels and signaling, and lymphangiogenesis in LEC-Atg5-/- mice. Our findings reveal that lipophagy in LEC by supporting FAO, preserves a mitochondrial-PROX1 gene expression circuit that safeguards LEC responsiveness to lymphangiogenic mediators and lymphangiogenesis.


Asunto(s)
Linfangiogénesis , Vasos Linfáticos , Animales , Autofagia/genética , Células Endoteliales/metabolismo , Ácidos Grasos/metabolismo , Gotas Lipídicas/metabolismo , Linfangiogénesis/genética , Vasos Linfáticos/metabolismo , Ratones , Mitocondrias , Factores de Transcripción/metabolismo
2.
Cell Metab ; 28(6): 881-894.e13, 2018 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-30146488

RESUMEN

Little is known about the metabolism of quiescent endothelial cells (QECs). Nonetheless, when dysfunctional, QECs contribute to multiple diseases. Previously, we demonstrated that proliferating endothelial cells (PECs) use fatty acid ß-oxidation (FAO) for de novo dNTP synthesis. We report now that QECs are not hypometabolic, but upregulate FAO >3-fold higher than PECs, not to support biomass or energy production but to sustain the tricarboxylic acid cycle for redox homeostasis through NADPH regeneration. Hence, endothelial loss of FAO-controlling CPT1A in CPT1AΔEC mice promotes EC dysfunction (leukocyte infiltration, barrier disruption) by increasing endothelial oxidative stress, rendering CPT1AΔEC mice more susceptible to LPS and inflammatory bowel disease. Mechanistically, Notch1 orchestrates the use of FAO for redox balance in QECs. Supplementation of acetate (metabolized to acetyl-coenzyme A) restores endothelial quiescence and counters oxidative stress-mediated EC dysfunction in CPT1AΔEC mice, offering therapeutic opportunities. Thus, QECs use FAO for vasculoprotection against oxidative stress-prone exposure.


Asunto(s)
Carnitina O-Palmitoiltransferasa/metabolismo , Metabolismo Energético , Ácidos Grasos/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , NADP/metabolismo , Receptor Notch1/metabolismo , Animales , Proliferación Celular , Células HEK293 , Homeostasis , Humanos , Ratones , Ratones Endogámicos C57BL , Oxidación-Reducción , Estrés Oxidativo
3.
Cell Metab ; 28(4): 573-587.e13, 2018 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-30017355

RESUMEN

The role of phosphoglycerate dehydrogenase (PHGDH), a key enzyme of the serine synthesis pathway (SSP), in endothelial cells (ECs) remains poorly characterized. We report that mouse neonates with EC-specific PHGDH deficiency suffer lethal vascular defects within days of gene inactivation, due to reduced EC proliferation and survival. In addition to nucleotide synthesis impairment, PHGDH knockdown (PHGDHKD) caused oxidative stress, due not only to decreased glutathione and NADPH synthesis but also to mitochondrial dysfunction. Electron transport chain (ETC) enzyme activities were compromised upon PHGDHKD because of insufficient heme production due to cellular serine depletion, not observed in other cell types. As a result of heme depletion, elevated reactive oxygen species levels caused EC demise. Supplementation of hemin in PHGDHKD ECs restored ETC function and rescued the apoptosis and angiogenesis defects. These data argue that ECs die upon PHGDH inhibition, even without external serine deprivation, illustrating an unusual importance of serine synthesis for ECs.


Asunto(s)
Células Endoteliales/metabolismo , Hemo/metabolismo , Fosfoglicerato-Deshidrogenasa/genética , Fosfoglicerato-Deshidrogenasa/metabolismo , Serina/metabolismo , Apoptosis , Errores Innatos del Metabolismo de los Carbohidratos/metabolismo , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Suplementos Dietéticos , Técnicas de Silenciamiento del Gen , Hemina/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Microcefalia/metabolismo , Mitocondrias/metabolismo , Mitofagia , Neovascularización Fisiológica , Estrés Oxidativo , Fosfoglicerato-Deshidrogenasa/deficiencia , Biosíntesis de Proteínas , Trastornos Psicomotores/metabolismo , Purinas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Convulsiones/metabolismo
4.
EMBO J ; 36(16): 2334-2352, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28659375

RESUMEN

Endothelial cell (EC) metabolism is emerging as a regulator of angiogenesis, but the precise role of glutamine metabolism in ECs is unknown. Here, we show that depriving ECs of glutamine or inhibiting glutaminase 1 (GLS1) caused vessel sprouting defects due to impaired proliferation and migration, and reduced pathological ocular angiogenesis. Inhibition of glutamine metabolism in ECs did not cause energy distress, but impaired tricarboxylic acid (TCA) cycle anaplerosis, macromolecule production, and redox homeostasis. Only the combination of TCA cycle replenishment plus asparagine supplementation restored the metabolic aberrations and proliferation defect caused by glutamine deprivation. Mechanistically, glutamine provided nitrogen for asparagine synthesis to sustain cellular homeostasis. While ECs can take up asparagine, silencing asparagine synthetase (ASNS, which converts glutamine-derived nitrogen and aspartate to asparagine) impaired EC sprouting even in the presence of glutamine and asparagine. Asparagine further proved crucial in glutamine-deprived ECs to restore protein synthesis, suppress ER stress, and reactivate mTOR signaling. These findings reveal a novel link between endothelial glutamine and asparagine metabolism in vessel sprouting.


Asunto(s)
Asparagina/metabolismo , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Glutamina/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Medios de Cultivo/química , Células Endoteliales/metabolismo , Glutaminasa/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Redes y Vías Metabólicas , Neovascularización Patológica
5.
Nature ; 542(7639): 49-54, 2017 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-28024299

RESUMEN

Lymphatic vessels are lined by lymphatic endothelial cells (LECs), and are critical for health. However, the role of metabolism in lymphatic development has not yet been elucidated. Here we report that in transgenic mouse models, LEC-specific loss of CPT1A, a rate-controlling enzyme in fatty acid ß-oxidation, impairs lymphatic development. LECs use fatty acid ß-oxidation to proliferate and for epigenetic regulation of lymphatic marker expression during LEC differentiation. Mechanistically, the transcription factor PROX1 upregulates CPT1A expression, which increases acetyl coenzyme A production dependent on fatty acid ß-oxidation. Acetyl coenzyme A is used by the histone acetyltransferase p300 to acetylate histones at lymphangiogenic genes. PROX1-p300 interaction facilitates preferential histone acetylation at PROX1-target genes. Through this metabolism-dependent mechanism, PROX1 mediates epigenetic changes that promote lymphangiogenesis. Notably, blockade of CPT1 enzymes inhibits injury-induced lymphangiogenesis, and replenishing acetyl coenzyme A by supplementing acetate rescues this process in vivo.


Asunto(s)
Ácidos Grasos/química , Ácidos Grasos/metabolismo , Linfangiogénesis , Vasos Linfáticos/citología , Vasos Linfáticos/metabolismo , Acetatos/farmacología , Acetilcoenzima A/metabolismo , Acetilación/efectos de los fármacos , Animales , Carnitina O-Palmitoiltransferasa/antagonistas & inhibidores , Carnitina O-Palmitoiltransferasa/genética , Carnitina O-Palmitoiltransferasa/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Epigénesis Genética , Femenino , Histonas/metabolismo , Proteínas de Homeodominio/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Linfangiogénesis/efectos de los fármacos , Linfangiogénesis/genética , Vasos Linfáticos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Oxidación-Reducción/efectos de los fármacos , Biosíntesis de Proteínas , Transcripción Genética , Proteínas Supresoras de Tumor/metabolismo , Arterias Umbilicales/citología , Regulación hacia Arriba
6.
Cell Metab ; 23(2): 265-79, 2016 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-26863487

RESUMEN

Cell-based therapy is a promising strategy in regenerative medicine, but the poor survival rate of the implanted cells remains a major challenge and limits clinical translation. We preconditioned periosteal cells to the hypoxic and ischemic environment of the bone defect site by deleting prolyl hydroxylase domain-containing protein 2 (PHD2), resulting in hypoxia-inducible factor 1 alpha (HIF-1α) stabilization. This strategy increased postimplantation cell survival and improved bone regeneration. The enhanced cell viability was angiogenesis independent but relied on combined changes in glutamine and glycogen metabolism. HIF-1α stabilization stimulated glutaminase-mediated glutathione synthesis, maintaining redox homeostasis at baseline and during oxidative or nutrient stress. Simultaneously, HIF-1α signaling increased glycogen storage, preventing an energy deficit during nutrient or oxygen deprivation. Pharmacological inhibition of PHD2 recapitulated the adaptations in glutamine and glycogen metabolism and, consequently, the beneficial effects on cell survival. Thus, targeting cellular metabolism is an appealing strategy for bone regeneration and cell-based therapy in general.


Asunto(s)
Metabolismo Energético , Glutamina/metabolismo , Glucógeno/metabolismo , Homeostasis , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Osteocitos/trasplante , Animales , Regeneración Ósea , Respiración de la Célula , Supervivencia Celular , Eliminación de Gen , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Glutaminasa/metabolismo , Ratones , Neovascularización Fisiológica , Osteocitos/metabolismo , Oxidación-Reducción , Estrés Oxidativo , Periostio/patología , Especies Reactivas de Oxígeno/metabolismo
7.
Expert Opin Ther Targets ; 18(11): 1339-54, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25297943

RESUMEN

INTRODUCTION: Placental growth factor (PLGF) belongs to the VEGF family, which among the three VEGF receptors binds exclusively to VEGFR1, present on various cell types. Isoform PLGF-2 also binds the neuropilin co-receptors. PLGF is dispensable for development and health but has a prominent role in pathology including cancer. This has triggered the question whether PLGF targeting might offer an alternative to current antiangiogenesis therapy, which encounters problems of refractoriness and acquired resistance. AREAS COVERED: This article reviews the available literature on the characteristics of PLGF, its role(s) in cancer and the findings on PLGF inhibition in preclinical models with attention to as yet unresolved questions and summarizes data from initial clinical trials. EXPERT OPINION: Preclinical studies show that inhibition of PLGF, either by genetic inhibition or by pharmacological blockade using distinct independently generated anti-PLGF antibodies, slows down tumor growth and metastasis and even induces regression of pre-existing medulloblastoma, the most frequent brain cancer in children. These promising preclinical findings, together with the acceptable safety profile of anti-PLGF administration in Phase I clinical trials, have attracted attention to PLGF as a potential target for therapy.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias/tratamiento farmacológico , Proteínas Gestacionales/antagonistas & inhibidores , Inhibidores de la Angiogénesis/farmacología , Animales , Antineoplásicos/efectos adversos , Niño , Ensayos Clínicos como Asunto , Diseño de Fármacos , Evaluación Preclínica de Medicamentos , Resistencia a Antineoplásicos , Humanos , Terapia Molecular Dirigida , Neoplasias/patología , Factor de Crecimiento Placentario , Proteínas Gestacionales/genética , Proteínas Gestacionales/metabolismo
8.
J Cell Mol Med ; 15(5): 1122-35, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-20629994

RESUMEN

Artemisinins are plant products with a wide range of medicinal applications. Most prominently, artesunate is a well tolerated and effective drug for treating malaria, but is also active against several protozoal and schistosomal infections, and additionally exhibits anti-angiogenic, anti-tumorigenic and anti-viral properties. The array of activities of the artemisinins, and the recent emergence of malaria resistance to artesunate, prompted us to synthesize and evaluate several novel artemisinin-like derivatives. Sixteen distinct derivatives were therefore synthesized and the in vitro cytotoxic effects of each were tested with different cell lines. The in vivo anti-angiogenic properties were evaluated using a zebrafish embryo model. We herein report the identification of several novel artemisinin-like compounds that are easily synthesized, stable at room temperature, may overcome drug-resistance pathways and are more active in vitro and in vivo than the commonly used artesunate. These promising findings raise the hopes of identifying safer and more effective strategies to treat a range of infections and cancer.


Asunto(s)
Inhibidores de la Angiogénesis/síntesis química , Inhibidores de la Angiogénesis/farmacología , Artemisininas/química , Artemisininas/farmacología , Animales , Artemisia annua/química , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Resistencia a Medicamentos , Citometría de Flujo , Extractos Vegetales/química , Relación Estructura-Actividad , Porcinos , Pez Cebra/embriología
10.
Heart Rhythm ; 7(12): 1862-9, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20728579

RESUMEN

BACKGROUND: Patients with long QT syndrome (LQTS) are at increased risk not only for ventricular arrhythmias but also for atrial pathology including atrial fibrillation (AF). Some patients with "lone" AF carry Na(+)-channel mutations. OBJECTIVE: The purpose of this study was to determine the mechanisms underlying atrial pathology in LQTS. METHODS: In mice with a heterozygous knock-in long QT syndrome type 3 (LQT3) mutant of the cardiac Na(+) channel (ΔKPQ-SCN5A) and wild-type (WT) littermates, atrial size, function, and electrophysiologic parameters were measured in intact Langendorff-perfused hearts, and histologic analysis was performed. RESULTS: Atrial action potential duration, effective refractory period, cycle length, and PQ interval were prolonged in ΔKPQ-SCN5A hearts (all P < .05). Flecainide (1 µM) reversed atrial action potential duration prolongation and induced postrepolarization refractoriness (P < .05). Arrhythmias were infrequent during regular rapid atrial rate in both WT and ΔKPQ-SCN5A but were inducible in 15 (38%) of 40 ΔKPQ-SCN5A and 8 (29%) of 28 WT mice upon extrastimulation. Pacing protocols generating rapid alterations in rate provoked atrial extrasystoles and arrhythmias in 6 (66%) of 9 ΔKPQ-SCN5A but in 0 (0%) of 6 WT mice (P < .05). Atrial diameter was increased by nearly 10% in ΔKPQ-SCN5A mice > 5 months old without increase in fibrotic tissue. CONCLUSION: Murine hearts bearing an LQT3 mutation show abnormalities in atrial electrophysiology and subtle changes in atrial dimension, including an atrial arrhythmogenic phenotype on provocation. These results support clinical data suggesting that LQTS mutations can cause atrial pathology and arrhythmogenesis and indicate that murine sodium channel LQTS models may be useful for exploring underlying mechanisms.


Asunto(s)
Atrios Cardíacos/fisiopatología , Activación del Canal Iónico/genética , Síndrome de QT Prolongado/genética , Síndrome de QT Prolongado/fisiopatología , Canales de Sodio/genética , Potenciales de Acción/fisiología , Animales , Antiarrítmicos/farmacología , Fibrilación Atrial/genética , Fibrilación Atrial/fisiopatología , Ecocardiografía Doppler , Técnicas Electrofisiológicas Cardíacas , Femenino , Fibrosis , Flecainida/farmacología , Técnicas de Sustitución del Gen , Atrios Cardíacos/patología , Técnicas In Vitro , Masculino , Ratones , Ratones Transgénicos , Mutación , Canal de Sodio Activado por Voltaje NAV1.5 , Recuperación de la Función/genética
12.
J Clin Invest ; 113(2): 188-99, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14722611

RESUMEN

VEGF is crucial for metaphyseal bone vascularization. In contrast, the angiogenic factors required for vascularization of epiphyseal cartilage are unknown, although this represents a developmentally and clinically important aspect of bone growth. The VEGF gene is alternatively transcribed into VEGF(120), VEGF(164), and VEGF(188) isoforms that differ in matrix association and receptor binding. Their role in bone development was studied in mice expressing single isoforms. Here we report that expression of only VEGF(164) or only VEGF(188) (in VEGF(188/188) mice) was sufficient for metaphyseal development. VEGF(188/188) mice, however, showed dwarfism, disrupted development of growth plates and secondary ossification centers, and knee joint dysplasia. This phenotype was at least partly due to impaired vascularization surrounding the epiphysis, resulting in ectopically increased hypoxia and massive chondrocyte apoptosis in the interior of the epiphyseal cartilage. In addition to the vascular defect, we provide in vitro evidence that the VEGF(188) isoform alone is also insufficient to regulate chondrocyte proliferation and survival responses to hypoxia. Consistent herewith, chondrocytes in or close to the hypoxic zone in VEGF(188/188) mice showed increased proliferation and decreased differentiation. These findings indicate that the insoluble VEGF(188) isoform is insufficient for establishing epiphyseal vascularization and regulating cartilage development during endochondral bone formation.


Asunto(s)
Condrocitos/citología , Epífisis/irrigación sanguínea , Epífisis/metabolismo , Factor A de Crecimiento Endotelial Vascular/química , Factor A de Crecimiento Endotelial Vascular/genética , Proteínas de Xenopus , Angiografía , Animales , Desarrollo Óseo , Bromodesoxiuridina/farmacología , Cartílago/citología , Cartílago/patología , Diferenciación Celular , División Celular , Supervivencia Celular , Condrocitos/metabolismo , ADN Complementario/metabolismo , Hipoxia , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Ratones , Modelos Biológicos , Mutagénesis , Neovascularización Fisiológica , Proteínas del Tejido Nervioso/metabolismo , Fenotipo , Unión Proteica , Isoformas de Proteínas , ARN Mensajero/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Recombinación Genética , Ribonucleoproteínas/metabolismo
13.
Cancer Res ; 63(7): 1500-7, 2003 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-12670896

RESUMEN

Hyperthermia (HT) associated with radiotherapy or chemotherapy is a promising method for cancer treatment, although the molecular mechanisms of this process are not well understood. HT exhibits various antitumor effects, including damage of tumor vasculature. Here, we investigate the effect of HT on in vitro and in vivo angiogenesis. We show that heat treatment of endothelial cells (ECs) affect their differentiation into capillary-like structures in two models of in vitro angiogenesis. Furthermore, the formation of new vessels promoted by angiogenic inducers in the chick embryo chorioallantoic membrane assay is impaired after heat treatment. These effects cannot be explained by direct cytotoxicity but are dependent on modulation of angiogenesis-involved genes. Gene expression profile of ECs subjected to heat shock demonstrates that plasminogen activator inhibitor 1 (PAI-1), a protein involved in the control of extracellular matrix degradation, is specifically up-regulated. The use of anti-PAI-1-neutralizing antibodies reverts the effect of HT on the in vitro EC morphogenesis and in vivo vessel formation. Moreover, microvessel outgrowth from PAI-1(-/-) aortic rings was not affected by HT compared with aortic rings from PAI-1(+/+) mice. Heat treatment of murine mammary adenocarcinomas results in inhibition of tumor growth, associated with a reduction of microvessel number and an increase of PAI-1 expression. These results indicate that heat-mediated PAI-1 induction is an important pathway by which HT exerts its antitumor activity and may represent a rationale for a combined cancer therapy based on HT associated with antiangiogenic molecules.


Asunto(s)
Endotelio Vascular/citología , Hipertermia Inducida/métodos , Neovascularización Fisiológica/fisiología , Inhibidor 1 de Activador Plasminogénico/fisiología , Alantoides/irrigación sanguínea , Animales , Anticuerpos/inmunología , Anticuerpos/farmacología , Carcinoma Pulmonar de Lewis/irrigación sanguínea , Carcinoma Pulmonar de Lewis/terapia , División Celular/fisiología , Supervivencia Celular/fisiología , Embrión de Pollo , Corion/irrigación sanguínea , Endotelio Vascular/crecimiento & desarrollo , Humanos , Técnicas In Vitro , Neoplasias Mamarias Experimentales/irrigación sanguínea , Neoplasias Mamarias Experimentales/terapia , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/terapia , Inhibidor 1 de Activador Plasminogénico/biosíntesis , Inhibidor 1 de Activador Plasminogénico/genética , Inhibidor 1 de Activador Plasminogénico/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA