Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Arterioscler Thromb Vasc Biol ; 43(5): 684-696, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36924232

RESUMEN

BACKGROUND: Adaptation of fat depots to change in fuel availability is critical for metabolic flexibility and cardiometabolic health. The mechanisms responsible for fat depot-specific lipid sensing and shuttling remain elusive. Adipose tissue microvascular endothelial cells (AT-EC) regulates bidirectional fatty acid fluxes depending on fed or fasted state. How AT-EC sense and adapt to metabolic changes according to AT location remains to be established. METHODS: We combined transcriptional analysis of native human AT-EC together with in vitro approaches in primary human AT-EC and in vivo and ex vivo studies of mice under fed and fasted conditions. RESULTS: Transcriptional large-scale analysis of human AT-EC isolated from gluteofemoral and abdominal subcutaneous AT revealed that the endothelium exhibits a fat depot-specific signature associated with lipid handling and Notch signaling enrichment. We uncovered a functional link between metabolic status and endothelial DLL4 (delta-like canonical notch ligand 4), which decreases with fasting. DLL4 regulates fatty acid uptake through nontranscriptional modulation of macropinocytosis-dependent long chain fatty acid uptake. Importantly, the changes in DLL4 expression, in response to energy transition state, is impaired under obesogenic conditions, an early alteration coinciding with a defect in systemic fatty acid fluxes adaptation and a resistance to weight loss. CONCLUSIONS: DLL4 is a major actor in the adaptive mechanisms of AT-EC to regulate lipid fluxes. It likely contributes to fat depot-dependent metabolism in response to energy transition states. AT-EC alteration with obesity may favor metabolic inflexibility and the development of cardiometabolic disorders.


Asunto(s)
Enfermedades Cardiovasculares , Células Endoteliales , Ratones , Humanos , Animales , Células Endoteliales/metabolismo , Ácidos Grasos/metabolismo , Obesidad/genética , Obesidad/metabolismo , Ayuno , Endotelio/metabolismo , Enfermedades Cardiovasculares/metabolismo , Proteínas de Unión al Calcio/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo
2.
Aging Cell ; 22(3): e13776, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36617688

RESUMEN

Senescence is a key event in the impairment of adipose tissue (AT) function with obesity and aging but the underlying molecular and cellular players remain to be fully defined, particularly with respect to the human AT progenitors. We have found distinct profiles of senescent progenitors based on AT location between stroma from visceral versus subcutaneous AT. In addition to flow cytometry, we characterized the location differences with transcriptomic and proteomic approaches, uncovering the genes and developmental pathways that are underlying replicative senescence. We identified key components to include INBHA as well as SFRP4 and GREM1, antagonists for the WNT and BMP pathways, in the senescence-associated secretory phenotype and NOTCH3 in the senescence-associated intrinsic phenotype. Notch activation in AT progenitors inhibits adipogenesis and promotes myofibrogenesis independently of TGFß. In addition, we demonstrate that NOTCH3 is enriched in the premyofibroblast progenitor subset, which preferentially accumulates in the visceral AT of patients with an early obesity trajectory. Herein, we reveal that NOTCH3 plays a role in the balance of progenitor fate determination preferring myofibrogenesis at the expense of adipogenesis. Progenitor NOTCH3 may constitute a tool to monitor replicative senescence and to limit AT dysfunction in obesity and aging.


Asunto(s)
Senescencia Celular , Proteómica , Humanos , Senescencia Celular/genética , Tejido Adiposo/metabolismo , Envejecimiento/metabolismo , Obesidad/metabolismo
3.
Cells ; 11(22)2022 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-36428969

RESUMEN

Adipose tissue (AT) expansion either through hypertrophy or hyperplasia is determinant in the link between obesity and metabolic alteration. The present study aims to profile the unhealthy subcutaneous and visceral AT (SAT, VAT) expansion in obesity and in the outcomes of bariatric surgery (BS). The repartition of adipocytes according to diameter and the numbers of progenitor subtypes and immune cells of SAT and VAT from 161 obese patients were determined by cell imaging and flow cytometry, respectively. Associations with insulin resistance (IR) prior to BS as well as with the loss of excessive weight (EWL) and IR at 1 and 3 years post-BS were studied; prior to BS, SAT and VAT, unhealthy expansions are characterized by the accumulation of adipogenic progenitors and CD4+ T lymphocytes and by adipocyte hypertrophy and elevated macrophage numbers, respectively. Such SAT stromal profile and VAT adipocyte hypertrophy are associated with adverse BS outcomes. Finally, myofibrogenic progenitors are a common determinant of weight and IR trajectories post-BS; the study suggests that adipogenesis in SAT and adipocyte hypertrophy in VAT are common determinants of metabolic alterations with obesity and of the weight loss and metabolic response to bariatric surgery. The data open up new avenues to better understand and predict individual outcomes in response to changes in energy balance.


Asunto(s)
Cirugía Bariátrica , Resistencia a la Insulina , Humanos , Adipocitos/metabolismo , Obesidad/metabolismo , Resistencia a la Insulina/fisiología , Células del Estroma/metabolismo , Hipertrofia
4.
Biomedicines ; 10(10)2022 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-36289874

RESUMEN

The amount and the distribution of body fat exhibit trajectories that are sex- and human species-specific and both are determinants for health. The enhanced accumulation of fat in the truncal part of the body as a risk factor for cardiovascular and metabolic diseases is well supported by epidemiological studies. In addition, a possible independent protective role of the gluteofemoral fat compartment and of the brown adipose tissue is emerging. The present narrative review summarizes the current knowledge on sexual dimorphism in fat depot amount and repartition and consequences on cardiometabolic and reproductive health. The drivers of the sex differences and fat depot repartition, considered to be the results of complex interactions between sex determination pathways determined by the sex chromosome composition, genetic variability, sex hormones and the environment, are discussed. Finally, the inter- and intra-depot heterogeneity in adipocytes and progenitors, emphasized recently by unbiased large-scale approaches, is highlighted.

5.
J Physiol Biochem ; 78(2): 543-556, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35066863

RESUMEN

Among the dietary amines present in foods and beverages, tyramine has been widely studied since its excessive ingestion can cause catecholamine release and hypertensive crisis. However, tyramine exerts other actions than depleting nerve endings: it activates subtypes of trace amine associated receptors (TAARs) and is oxidized by monoamine oxidases (MAO). Although we have recently described that tyramine is antilipolytic in human adipocytes, no clear evidence has been reported about its effects on glucose transport in the same cell model, while tyramine mimics various insulin-like effects in rodent fat cells, such as activation of glucose transport, lipogenesis, and adipogenesis. Our aim was therefore to characterize the effects of tyramine on glucose transport in human adipocytes. The uptake of the non-metabolizable analogue 2-deoxyglucose (2-DG) was explored in adipocytes from human subcutaneous abdominal adipose tissue obtained from women undergoing reconstructive surgery. Human insulin used as reference agent multiplied by three times the basal 2-DG uptake. Tyramine was ineffective from 0.01 to 10 µM and stimulatory at 100 µM-1 mM, without reaching the maximal effect of insulin. This partial insulin-like effect was not improved by vanadium and was impaired by MAO-A and MAO-B inhibitors. Contrarily to benzylamine, mainly oxidized by semicarbazide-sensitive amine oxidase (SSAO), tyramine activation of glucose transport was not inhibited by semicarbazide. Tyramine effect was not dependent on the Gi-coupled receptor activation but was impaired by antioxidants and reproduced by hydrogen peroxide. In all, the oxidation of high doses of tyramine, already reported to inhibit lipolysis in human fat cells, also partially mimic another effect of insulin in these cells, the glucose uptake activation. Thus, other MAO substrates are potentially able to modulate carbohydrate metabolism.


Asunto(s)
Amina Oxidasa (conteniendo Cobre) , Tiramina , Adipocitos/metabolismo , Femenino , Glucosa/metabolismo , Humanos , Insulina/metabolismo , Monoaminooxidasa/metabolismo , Tiramina/farmacología
6.
Molecules ; 26(13)2021 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-34201708

RESUMEN

Caffeine is a plant alkaloid present in food and beverages consumed worldwide. It has high lipid solubility with recognized actions in the central nervous system and in peripheral tissues, notably the adipose depots. However, the literature is scant regarding caffeine's influence on adipocyte functions other than lipolysis, such as glucose incorporation into lipids (lipogenesis) and amine oxidation. The objective of this study was to explore the direct effects of caffeine and of isobutylmethylxanthine (IBMX) on these adipocyte functions. Glucose transport into fat cells freshly isolated from mice, rats, or humans was monitored by determining [3H]-2-deoxyglucose (2-DG) uptake, while the incorporation of radiolabeled glucose into cell lipids was used as an index of lipogenic activity. Oxidation of benzylamine by primary amine oxidase (PrAO) was inhibited by increasing doses of caffeine in human adipose tissue preparations with an inhibition constant (Ki) in the millimolar range. Caffeine inhibited basal and insulin-stimulated glucose transport as well as lipogenesis in rodent adipose cells. The antilipogenic action of caffeine was also observed in adipocytes from mice genetically invalidated for PrAO activity, indicating that PrAO activity was not required for lipogenesis inhibition. These caffeine inhibitory properties were extended to human adipocytes: relative to basal 2-DG uptake, set at 1.0 ± 0.2 for 6 individuals, 0.1 mM caffeine tended to reduce uptake to 0.83 ± 0.08. Insulin increased uptake by 3.86 ± 1.11 fold when tested alone at 100 nM, and by 3.21 ± 0.80 when combined with caffeine. Our results reinforce the recommendation of caffeine's potential in the treatment or prevention of obesity complications.


Asunto(s)
Adipocitos/efectos de los fármacos , Aminas Biogénicas/metabolismo , Cafeína/farmacología , Glucosa/metabolismo , Lipogénesis/efectos de los fármacos , Monoaminooxidasa/metabolismo , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Bencilaminas/metabolismo , Transporte Biológico/efectos de los fármacos , Desoxiglucosa/metabolismo , Humanos , Insulina/metabolismo , Lipólisis/efectos de los fármacos , Ratones , Ratas , Xantinas/farmacología
7.
J Physiol Biochem ; 75(3): 263-273, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30919256

RESUMEN

Various amino acid (AA) metabolites are used as supplements to facilitate metabolic control and enhance responsiveness of insulin-sensitive tissues. ß-hydroxy-ß-methylbutyrate (HMB) is a leucine metabolite proposed to prevent muscle wasting and to mitigate insulin resistance. Taurine, commonly added to energizing drinks, is a metabolite of methionine and cysteine present in bile juice, and proposed to be involved in lipid digestion and to be pro-lipolytic in adipocytes. N-methyltyramine (NMT) is a phenylalanine metabolite found in orange juices at 0.1-3 ppm while its effects on lipid mobilization remain controversial. Here, the putative lipolytic effects of these AA metabolites were studied and it was tested whether they could enhance insulin antilipolytic response in adipocytes. Release of glycerol and non-esterified fatty acids (NEFAs) was measured after a 2-h incubation of adipocytes obtained from control and diet-induced obese mice or from obese patients. In mouse, none of the tested AA derivatives was lipolytic from 1 µM to 1 mM. These compounds did not improve insulin antilipolytic effect or isoprenaline lipolytic action, except for 1 mM NMT that impaired triacylglycerol breakdown in obese mice. In human adipocytes, HMB and taurine were not lipolytic, while NMT weakly activated glycerol and NEFA release at 1 mM. However, 100 µM NMT impaired isoprenaline-stimulated lipolysis in a manner that was hardly added to insulin antilipolytic effect. Since none of these AA derivatives acutely helped or replaced insulin antilipolytic effect in adipocytes, the present in vitro observations do not support their proposed insulin-sensitizing properties. Moreover, NMT, HMB, and taurine were not notably lipolytic.


Asunto(s)
Adipocitos , Insulina/metabolismo , Lipólisis/efectos de los fármacos , Taurina/farmacología , Tiramina/análogos & derivados , Valeratos/farmacología , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Adulto , Animales , Femenino , Humanos , Resistencia a la Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Obesidad/metabolismo , Tiramina/farmacología
8.
Arterioscler Thromb Vasc Biol ; 38(5): 1134-1146, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29545239

RESUMEN

OBJECTIVE: Adipose tissue (AT) dysfunction associated with obesity or aging is a major cause for lipid redistribution and the progression of cardiometabolic disorders. Our goal is to decipher the contribution of human AT microvascular endothelial cells (ECs) in the maintenance of fatty acid (FA) fluxes and the impact of senescence on their function. APPROACH AND RESULTS: We used freshly isolated primary microvascular ECs from human AT. Our data identified the endothelial FA handling machinery including FATPs (FA transport proteins) FATP1, FATP3, FATP4, and CD36 as well as FABP4 (FA binding protein 4). We showed that PPARγ (peroxisome proliferator-activated receptor gamma) regulates the expression of FATP1, CD36, and FABP4 and is a major regulator of FA uptake in human AT EC (hATEC). We provided evidence that endothelial PPARγ activity is modulated by senescence. Indeed, the positive regulation of FA transport by PPARγ agonist was abolished, whereas the emergence of an inflammatory response was favored in senescent hATEC. This was associated with the retention of nuclear FOXO1 (forkhead box protein O1), whereas nuclear PPARγ translocation was impaired. CONCLUSIONS: These data support the notion that PPARγ is a key regulator of primary hATEC function including FA handling and inflammatory response. However, the outcome of PPARγ activation is modulated by senescence, a phenomenon that may impact the ability of hATEC to properly respond to and handle lipid fluxes. Finally, our work highlights the role of hATEC in the regulation of FA fluxes and reveals that dysfunction of these cells with accelerated aging is likely to participate to AT dysfunction and the redistribution of lipids.


Asunto(s)
Grasa Abdominal/irrigación sanguínea , Senescencia Celular , Células Endoteliales/metabolismo , Ácidos Grasos/metabolismo , Inflamación/metabolismo , Microvasos/metabolismo , PPAR gamma/metabolismo , Transporte Activo de Núcleo Celular , Proliferación Celular , Células Cultivadas , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Células Endoteliales/ultraestructura , Proteínas de Unión a Ácidos Grasos/genética , Proteínas de Unión a Ácidos Grasos/metabolismo , Femenino , Proteína Forkhead Box O1/metabolismo , Humanos , Inflamación/genética , Inflamación/patología , Microvasos/ultraestructura , PPAR gamma/genética , Transducción de Señal
9.
Nat Commun ; 8(1): 1620, 2017 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-29158473

RESUMEN

Endothelial cells transduce mechanical forces from blood flow into intracellular signals required for vascular homeostasis. Here we show that endothelial NOTCH1 is responsive to shear stress, and is necessary for the maintenance of junctional integrity, cell elongation, and suppression of proliferation, phenotypes induced by laminar shear stress. NOTCH1 receptor localizes downstream of flow and canonical NOTCH signaling scales with the magnitude of fluid shear stress. Reduction of NOTCH1 destabilizes cellular junctions and triggers endothelial proliferation. NOTCH1 suppression results in changes in expression of genes involved in the regulation of intracellular calcium and proliferation, and preventing the increase of calcium signaling rescues the cell-cell junctional defects. Furthermore, loss of Notch1 in adult endothelium increases hypercholesterolemia-induced atherosclerosis in the descending aorta. We propose that NOTCH1 is atheroprotective and acts as a mechanosensor in adult arteries, where it integrates responses to laminar shear stress and regulates junctional integrity through modulation of calcium signaling.


Asunto(s)
Arterias/metabolismo , Mecanotransducción Celular , Receptor Notch1/metabolismo , Animales , Arterias/química , Calcio/metabolismo , Células Endoteliales/química , Células Endoteliales/metabolismo , Endotelio Vascular/química , Endotelio Vascular/metabolismo , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor Notch1/genética , Estrés Mecánico
10.
Phytother Res ; 31(8): 1273-1282, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28627722

RESUMEN

The anti-obesity effects of resveratrol shown in rodents are not transposed into an efficient therapy of human obesity. Consequently, the search for molecules mimicking or surpassing resveratrol actions is ongoing. The natural phenolic compound pterostilbene exhibits beneficial health effects and has the capacity to limit fat mass in animal models. In this study, we tested whether pterostilbene modulates triacylglycerol accumulation/breakdown. Prolonged exposure to pterostilbene or resveratrol inhibited adipocyte differentiation in 3T3-F442A preadipocytes. Acute effects on lipolysis, antilipolysis and lipogenesis were determined for pterostilbene in mouse adipocytes, and compared with resveratrol. Pterostilbene was also tested on glycerol release and glucose uptake in subcutaneous human adipocytes. Dose-response analyses did not reveal a clear lipolytic effect in both species. The antilipolytic effect of insulin was improved by pterostilbene at 1-10 µM in mouse fat cells only, while at 1 mM, the phenolic compound was antilipolytic in human fat cells in a manner not additive to insulin. Pterostilbene dose-dependently inhibited glucose incorporation into lipids similarly to resveratrol and caffeine. However, only the former did not inhibit insulin-stimulated glucose uptake. Indeed, pterostilbene abolished the insulin lipogenic effect without inhibiting its antilipolytic action and rapid activation of glucose uptake. Pterostilbene therefore exhibits a unique panel of direct interactions with adipocytes that relies on its reported anti-obesity and antidiabetic properties. Copyright © 2017 John Wiley & Sons, Ltd.


Asunto(s)
Adipocitos/efectos de los fármacos , Lipogénesis/efectos de los fármacos , Lipólisis/efectos de los fármacos , Estilbenos/farmacología , Células 3T3 , Adulto , Animales , Transporte Biológico , Cafeína/farmacología , Diferenciación Celular/efectos de los fármacos , Femenino , Glucosa/metabolismo , Glicerol/metabolismo , Humanos , Insulina/metabolismo , Ratones , Persona de Mediana Edad , Obesidad/metabolismo , Resveratrol
11.
Curr Opin Lipidol ; 27(5): 513-20, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27454451

RESUMEN

PURPOSE OF REVIEW: Notch signaling is an evolutionary conserved pathway critical for cardiovascular development and angiogenesis. More recently, the contribution of Notch signaling to the homeostasis of the adult vasculature has emerged as an important novel paradigm, but much remains to be understood. RECENT FINDINGS: Recent findings shed light on the impact of Notch in vascular and immune responses to microenvironmental signals as well as on the onset of atherosclerosis. In the past year, studies in human and mice explored the role of Notch in the maintenance of a nonactivated endothelium. Novel pieces of evidence suggest that this pathway is sensitive to environmental factors, including inflammatory mediators and diet-derived by-products. SUMMARY: An emerging theme is the ability of Notch to respond to changes in the microenvironment, including glucose and lipid metabolites. In turn, alterations in Notch enable an important link between metabolism and transcriptional changes, thus this receptor appears to function as a metabolic sensor with direct implications to gene expression.


Asunto(s)
Células Endoteliales , Metabolismo de los Lípidos , Receptores Notch/metabolismo , Animales , Células Endoteliales/citología , Células Endoteliales/patología , Humanos , Inflamación/metabolismo , Inflamación/patología , Transducción de Señal
12.
J Exp Med ; 212(12): 2147-63, 2015 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-26552708

RESUMEN

Although much progress has been made in identifying the mechanisms that trigger endothelial activation and inflammatory cell recruitment during atherosclerosis, less is known about the intrinsic pathways that counteract these events. Here we identified NOTCH1 as an antagonist of endothelial cell (EC) activation. NOTCH1 was constitutively expressed by adult arterial endothelium, but levels were significantly reduced by high-fat diet. Furthermore, treatment of human aortic ECs (HAECs) with inflammatory lipids (oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine [Ox-PAPC]) and proinflammatory cytokines (TNF and IL1ß) decreased Notch1 expression and signaling in vitro through a mechanism that requires STAT3 activation. Reduction of NOTCH1 in HAECs by siRNA, in the absence of inflammatory lipids or cytokines, increased inflammatory molecules and binding of monocytes. Conversely, some of the effects mediated by Ox-PAPC were reversed by increased NOTCH1 signaling, suggesting a link between lipid-mediated inflammation and Notch1. Interestingly, reduction of NOTCH1 by Ox-PAPC in HAECs was associated with a genetic variant previously correlated to high-density lipoprotein in a human genome-wide association study. Finally, endothelial Notch1 heterozygous mice showed higher diet-induced atherosclerosis. Based on these findings, we propose that reduction of endothelial NOTCH1 is a predisposing factor in the onset of vascular inflammation and initiation of atherosclerosis.


Asunto(s)
Aterosclerosis/metabolismo , Células Endoteliales/metabolismo , Inflamación/metabolismo , Lípidos/sangre , Receptor Notch1/metabolismo , Adulto , Animales , Aterosclerosis/etiología , Aterosclerosis/genética , Línea Celular Tumoral , Células Cultivadas , Dieta Alta en Grasa/efectos adversos , Células Endoteliales/efectos de los fármacos , Femenino , Humanos , Inflamación/genética , Interleucina-1beta/farmacología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía Confocal , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosfatidilcolinas/farmacología , Interferencia de ARN , Receptor Notch1/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcriptoma/efectos de los fármacos , Transcriptoma/genética , Factor de Necrosis Tumoral alfa/farmacología
13.
Cancer Discov ; 5(2): 112-4, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25656896

RESUMEN

SUMMARY: The signaling specificity conveyed by distinct combination of NOTCH receptors/ligands has remained elusive. In this issue of Cancer Discovery, through the development of ligand-specific NOTCH inhibitors, Kangsamaksin and colleagues uncovered unique signaling outcomes downstream of DLL- and JAG-receptor activation and demonstrated their effects in the suppression of tumor angiogenesis.


Asunto(s)
Fragmentos Fc de Inmunoglobulinas/administración & dosificación , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Neoplasias/irrigación sanguínea , Neoplasias/terapia , Receptor Notch1/administración & dosificación , Receptores Notch/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/administración & dosificación , Animales , Femenino , Humanos
14.
Dev Cell ; 31(6): 707-21, 2014 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-25535917

RESUMEN

Acquisition and maintenance of vascular smooth muscle fate are essential for the morphogenesis and function of the circulatory system. Loss of contractile properties or changes in the identity of vascular smooth muscle cells (vSMCs) can result in structural alterations associated with aneurysms and vascular wall calcification. Here we report that maturation of sclerotome-derived vSMCs depends on a transcriptional switch between mouse embryonic days 13 and 14.5. At this time, Notch/Jag1-mediated repression of sclerotome transcription factors Pax1, Scx, and Sox9 is necessary to fully enable vSMC maturation. Specifically, Notch signaling in vSMCs antagonizes sclerotome and cartilage transcription factors and promotes upregulation of contractile genes. In the absence of the Notch ligand Jag1, vSMCs acquire a chondrocytic transcriptional repertoire that can lead to ossification. Importantly, our findings suggest that sustained Notch signaling is essential throughout vSMC life to maintain contractile function, prevent vSMC reprogramming, and promote vascular wall integrity.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Condrogénesis/fisiología , Regulación del Desarrollo de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/metabolismo , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Factor de Transcripción SOX9/metabolismo , Transporte Activo de Núcleo Celular , Animales , Cartílago/metabolismo , Linaje de la Célula , Condrocitos/citología , Femenino , Proteína Jagged-1 , Ligandos , Masculino , Ratones , Contracción Muscular , Receptores Notch/metabolismo , Análisis de Secuencia de ARN , Proteínas Serrate-Jagged , Transducción de Señal , Factores de Tiempo , Factores de Transcripción/metabolismo
15.
J Exp Med ; 211(3): 499-513, 2014 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-24534191

RESUMEN

Netherton syndrome (NS) is a severe genetic skin disease in which absence of a key protease inhibitor causes congenital exfoliative erythroderma, eczematous-like lesions, and atopic manifestations. Several proteases are overactive in NS, including kallikrein-related peptidase (KLK) 5, KLK7, and elastase-2 (ELA2), which are suggested to be part of a proteolytic cascade initiated by KLK5. To address the role of KLK5 in NS, we have generated a new transgenic murine model expressing human KLK5 in the granular layer of the epidermis (Tg-KLK5). Transgene expression resulted in increased proteolytic activity attributable to KLK5 and its downstream targets KLK7, KLK14, and ELA2. Tg-KLK5 mice developed an exfoliative erythroderma with scaling, growth delay, and hair abnormalities. The skin barrier was defective and the stratum corneum was detached through desmosomal cleavage. Importantly, Tg-KLK5 mice displayed cutaneous and systemic hallmarks of severe inflammation and allergy with pruritus. The skin showed enhanced expression of inflammatory cytokines and chemokines, infiltration of immune cells, and markers of Th2/Th17/Th22 T cell responses. Moreover, serum IgE and Tslp levels were elevated. Our study identifies KLK5 as an important contributor to the NS proteolytic cascade and provides a new and viable model for the evaluation of future targeted therapies for NS or related diseases such as atopic dermatitis.


Asunto(s)
Modelos Animales de Enfermedad , Calicreínas/metabolismo , Síndrome de Netherton/genética , Síndrome de Netherton/patología , Animales , Western Blotting , Quimiocinas/metabolismo , Citocinas/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Calicreínas/genética , Ratones , Ratones Transgénicos , Microscopía Confocal , Microscopía Electrónica de Transmisión , Reacción en Cadena en Tiempo Real de la Polimerasa , Piel/metabolismo , Piel/patología , Estadísticas no Paramétricas , Pérdida Insensible de Agua/fisiología
16.
Sci Signal ; 7(307): ra1, 2014 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-24399295

RESUMEN

Although vascular complications are a hallmark of diabetes, the molecular mechanisms that underlie endothelial dysfunction are unclear. We showed that reactive oxygen species generated from hyperglycemia promoted ligand-independent phosphorylation of vascular endothelial growth factor receptor 2 (VEGFR2). This VEGFR2 signaling occurred within the Golgi compartment and resulted in progressively decreased availability of VEGFR2 at the cell surface. Consequently, the responses of endothelial cells to exogenous VEGF in a mouse model of diabetes were impaired because of a specific deficiency of VEGFR2 at the cell surface, despite a lack of change in transcript abundance. Hyperglycemia-induced phosphorylation of VEGFR2 did not require intrinsic receptor kinase activity and was instead mediated by Src family kinases. The reduced cell surface abundance of VEGFR2 in diabetic mice was reversed by treatment with the antioxidant N-acetyl-L-cysteine, suggesting a causative role for oxidative stress. These findings uncover a mode of ligand-independent VEGFR2 signaling that can progressively lead to continuously muted responses to exogenous VEGF and limit angiogenic events.


Asunto(s)
Vasos Sanguíneos/fisiopatología , Diabetes Mellitus Experimental/fisiopatología , Transducción de Señal/fisiología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Acetilcisteína/farmacología , Animales , Vasos Sanguíneos/metabolismo , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Relación Dosis-Respuesta a Droga , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Endotelio Vascular/fisiopatología , Depuradores de Radicales Libres/farmacología , Glucosa/farmacología , Aparato de Golgi/efectos de los fármacos , Aparato de Golgi/metabolismo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Immunoblotting , Ligandos , Ratones , Ratones Noqueados , Ratones Obesos , Ratones Transgénicos , Fosforilación/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
17.
Nat Commun ; 4: 2609, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24153254

RESUMEN

The functional diversity of the arterial and venous endothelia is regulated through a complex system of signalling pathways and downstream transcription factors. Here we report that the transcription factor Sox17, which is known as a regulator of endoderm and hemopoietic differentiation, is selectively expressed in arteries, and not in veins, in the mouse embryo and in mouse postnatal retina and adult. Endothelial cell-specific inactivation of Sox17 in the mouse embryo is accompanied by a lack of arterial differentiation and vascular remodelling that results in embryo death in utero. In mouse postnatal retina, abrogation of Sox17 expression in endothelial cells leads to strong vascular hypersprouting, loss of arterial identity and large arteriovenous malformations. Mechanistically, Sox17 acts upstream of the Notch system and downstream of the canonical Wnt system. These data introduce Sox17 as a component of the complex signalling network that orchestrates arterial/venous specification.


Asunto(s)
Arterias/metabolismo , Endodermo/metabolismo , Células Endoteliales/metabolismo , Proteínas HMGB/metabolismo , Morfogénesis/genética , Retina/metabolismo , Factores de Transcripción SOXF/metabolismo , Venas/metabolismo , Animales , Arterias/citología , Diferenciación Celular , Proliferación Celular , Embrión de Mamíferos , Endodermo/irrigación sanguínea , Endodermo/citología , Células Endoteliales/citología , Regulación del Desarrollo de la Expresión Génica , Proteínas HMGB/genética , Ratones , Neovascularización Patológica , Receptores Notch/genética , Receptores Notch/metabolismo , Retina/citología , Factores de Transcripción SOXF/genética , Transducción de Señal , Venas/citología , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
18.
Biol Reprod ; 88(4): 91, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23467742

RESUMEN

Leydig cells are the steroidogenic lineage of the mammalian testis that produces testosterone, a key hormone required throughout male fetal and adult life for virilization and spermatogenesis. Both fetal and adult Leydig cells arise from a progenitor population in the testis interstitium but are thought to be lineage-independent of one another. Genetic evidence indicates that Notch signaling is required during fetal life to maintain a balance between differentiated Leydig cells and their progenitors, but the elusive progenitor cell type and ligands involved have not been identified. In this study, we show that the Notch pathway signals through the ligand JAG1 in perivascular interstitial cells during fetal life. In the early postnatal testis, we show that circulating levels of testosterone directly affect Notch signaling, implicating a feedback role for systemic circulating factors in the regulation of progenitor cells. Between Postnatal Days 3 and 21, as fetal Leydig cells disappear from the testis and are replaced by adult Leydig cells, the perivascular population of interstitial cells active for Notch signaling declines, consistent with distinct regulation of adult Leydig progenitors.


Asunto(s)
Células Intersticiales del Testículo/fisiología , Células Madre/fisiología , Testículo/embriología , Testosterona/metabolismo , Animales , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión al Calcio/fisiología , Feto/efectos de los fármacos , Feto/metabolismo , Feto/fisiología , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/fisiología , Proteína Jagged-1 , Células Intersticiales del Testículo/efectos de los fármacos , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores Notch/metabolismo , Receptores Notch/fisiología , Proteínas Serrate-Jagged , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Células Madre/efectos de los fármacos , Testosterona/sangre , Testosterona/farmacología
19.
Development ; 139(23): 4449-60, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23095891

RESUMEN

The Notch signaling pathway is an important contributor to the development and homeostasis of the cardiovascular system. Not surprisingly, mutations in Notch receptors and ligands have been linked to a variety of hereditary diseases that impact both the heart and the vasculature. In particular, mutations in the gene encoding the human Notch ligand jagged 1 result in a multisystem autosomal dominant disorder called Alagille syndrome, which includes tetralogy of Fallot among its more severe cardiac pathologies. Jagged 1 is expressed throughout the developing embryo, particularly in endothelial cells. Here, we demonstrate that endothelial-specific deletion of Jag1 leads to cardiovascular defects in both embryonic and adult mice that are reminiscent of those in Alagille syndrome. Mutant mice display right ventricular hypertrophy, overriding aorta, ventricular septal defects, coronary vessel abnormalities and valve defects. Examination of mid-gestational embryos revealed that the loss of Jag1, similar to the loss of Notch1, disrupts endothelial-to-mesenchymal transition during endocardial cushion formation. Furthermore, adult mutant mice exhibit cardiac valve calcifications associated with abnormal matrix remodeling and induction of bone morphogenesis. This work shows that the endothelium is responsible for the wide spectrum of cardiac phenotypes displayed in Alagille Syndrome and it demonstrates a crucial role for Jag1 in valve morphogenesis.


Asunto(s)
Síndrome de Alagille/genética , Calcinosis/genética , Proteínas de Unión al Calcio/genética , Cardiomiopatías/genética , Cardiopatías Congénitas/genética , Enfermedades de las Válvulas Cardíacas/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Proteínas de la Membrana/genética , Animales , Proteínas de Unión al Calcio/metabolismo , Cardiomiopatías/metabolismo , Anomalías de los Vasos Coronarios/genética , Anomalías de los Vasos Coronarios/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Endotelio/citología , Endotelio/metabolismo , Cardiopatías Congénitas/metabolismo , Defectos del Tabique Interventricular/genética , Defectos del Tabique Interventricular/metabolismo , Enfermedades de las Válvulas Cardíacas/metabolismo , Hipertrofia Ventricular Derecha/genética , Hipertrofia Ventricular Derecha/metabolismo , Proteína Jagged-1 , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Morfogénesis , Técnicas de Cultivo de Órganos , Receptores Notch/genética , Receptores Notch/metabolismo , Proteínas Serrate-Jagged
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...