Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Kidney Int ; 99(5): 1088-1101, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33359500

RESUMEN

Chronic kidney disease (CKD) promotes development of cardiac abnormalities and is highly prevalent in patients with heart failure, particularly in those with preserved ejection fraction. CKD is associated with endothelial dysfunction, however, whether CKD can induce impairment of endothelium-to-cardiomyocyte crosstalk leading to impairment of cardiomyocyte function is not known. The sodium-glucose co-transporter 2 inhibitor, empagliflozin, reduced cardiovascular events in diabetic patients with or without CKD, suggesting its potential as a new treatment for heart failure with preserved ejection fraction. We hypothesized that uremic serum from patients with CKD would impair endothelial control of cardiomyocyte relaxation and contraction, and that empagliflozin would protect against this effect. Using a co-culture system of human cardiac microvascular endothelial cells with adult rat ventricular cardiomyocytes to measure cardiomyocyte relaxation and contraction, we showed that serum from patients with CKD impaired endothelial enhancement of cardiomyocyte function which was rescued by empagliflozin. Exposure to uremic serum reduced human cardiac microvascular endothelial cell nitric oxide bioavailability, and increased mitochondrial reactive oxygen species and 3-nitrotyrosine levels, indicating nitric oxide scavenging by reactive oxygen species. Empagliflozin attenuated uremic serum-induced generation of endothelial mitochondrial reactive oxygen species, leading to restoration of nitric oxide production and endothelium-mediated enhancement of nitric oxide levels in cardiomyocytes, an effect largely independent of sodium-hydrogen exchanger-1. Thus, empagliflozin restores the beneficial effect of cardiac microvascular endothelial cells on cardiomyocyte function by reducing mitochondrial oxidative damage, leading to reduced reactive oxygen species accumulation and increased endothelial nitric oxide bioavailability.


Asunto(s)
Miocitos Cardíacos , Insuficiencia Renal Crónica , Animales , Compuestos de Bencidrilo , Células Endoteliales , Endotelio , Endotelio Vascular , Glucósidos , Humanos , Óxido Nítrico , Ratas , Insuficiencia Renal Crónica/tratamiento farmacológico
2.
J Cell Physiol ; 234(11): 20520-20532, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31016754

RESUMEN

Fracture repair is characterized by cytokine production and hypoxia. To better predict cytokine modulation of mesenchymal stem cell (MSC)-aided bone healing, we investigated whether interleukin 4 (IL-4), IL-6, and their combination, affect osteogenic differentiation, vascular endothelial growth factor (VEGF) production, and/or mammalian target of rapamycin complex 1 (mTORC1) activation by MSCs under normoxia or hypoxia. Human adipose stem cells (hASCs) were cultured with IL-4, IL-6, or their combination for 3 days under normoxia (20% O 2 ) or hypoxia (1% O 2 ), followed by 11 days without cytokines under normoxia or hypoxia. Hypoxia did not alter IL-4 or IL-6-modulated gene or protein expression by hASCs. IL-4 alone decreased runt-related transcription factor 2 (RUNX2) and collagen type 1 (COL1) gene expression, alkaline phosphatase (ALP) activity, and VEGF protein production by hASCs under normoxia and hypoxia, and decreased mineralization of hASCs under hypoxia. In contrast, IL-6 increased mineralization of hASCs under normoxia, and enhanced RUNX2 gene expression under normoxia and hypoxia. Neither IL-4 nor IL-6 affected phosphorylation of the mTORC1 effector protein P70S6K. IL-4 combined with IL-6 diminished the inhibitory effect of IL-4 on ALP activity, bone nodule formation, and VEGF production, and decreased RUNX2 and COL1 expression, similar to IL-4 alone, under normoxia and hypoxia. In conclusion, IL-4 alone, but not in combination with IL-6, inhibits osteogenic differentiation and angiogenic stimulation potential of hASCs under normoxia and hypoxia, likely through pathways other than mTORC1. These results indicate that cytokines may differentially affect bone healing and regeneration when applied in isolation or in combination.


Asunto(s)
Tejido Adiposo/citología , Diferenciación Celular/efectos de los fármacos , Interleucina-4/farmacología , Interleucina-6/farmacología , Osteogénesis/efectos de los fármacos , Células Madre/efectos de los fármacos , Células Madre/fisiología , Adulto , Desarrollo Óseo/efectos de los fármacos , Diferenciación Celular/fisiología , Proliferación Celular , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Persona de Mediana Edad , Osteogénesis/fisiología , Oxígeno , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
3.
Cell Physiol Biochem ; 53(5): 865-886, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31724838

RESUMEN

BACKGROUND/AIMS: Heart failure is characterized by chronic low-grade vascular inflammation, which in itself can lead to endothelial dysfunction. Clinical trials showed reductions in heart failure-related hospitalizations of type 2 diabetic patients using sodium glucose co-transporter 2 inhibitors (SGLT2i's). Whether and how SGLT2i's directly affect the endothelium under inflammatory conditions is not completely understood. The aim of the study was to investigate whether the SGLT2i Empagliflozin (EMPA) and Dapagliflozin (DAPA) reduce tumor necrosis factor α (TNFα) induced endothelial inflammation in vitro. METHODS: Human coronary arterial endothelial cells (HCAECs) and human umbilical vein endothelial cells (HUVECs) were (pre-)incubated with 1 µM EMPA or DAPA and subsequently exposed to 10 ng/ml TNFα. ROS and NO were measured using live cell imaging. Target proteins were either determined by infrared western blotting or fluorescence activated cell sorting (FACS). The connection between Cav-1 and eNOS was determined by co-immunoprecipitation. RESULTS: Nitric oxide (NO) bioavailability was reduced by TNFα and both EMPA and DAPA restored NO levels in TNFα-stimulated HCAECs. Intracellular ROS was increased by TNFα, and this increase was completely abolished by EMPA and DAPA in HCAECs by means of live cell imaging. eNOS signaling was significantly disturbed after 24 h when cells were exposed to TNFα for 24h, yet the presence of both SGLT2is did not prevent this disruption. TNFα-induced enhanced permeability at t=24h was unaffected in HUVECs by EMPA. Similarly, adhesion molecule expression (VCAM-1 and ICAM-1) was elevated after 4h TNFα (1.5-5.5 fold increase of VCAM-1 and 4-12 fold increase of ICAM-1) but were unaffected by EMPA and DAPA in both cell types. Although we detected expression of SGLT2 protein levels, the fact that we could not silence this expression by means of siRNA and the mRNA levels of SGLT2 were not detectable in HCAECs, suggests aspecificity or our SGLT2 antibody and absence of SGLT2 in our cells. CONCLUSION: These data suggest that EMPA and DAPA rather restore NO bioavailability by inhibiting ROS generation than by affecting eNOS expression or signaling, barrier function and adhesion molecules expression in TNFα-induced endothelial cells. Furthermore, the observed effects cannot be ascribed to the inhibition of SGLT2 in endothelial cells.


Asunto(s)
Compuestos de Bencidrilo/farmacología , Regulación hacia Abajo/efectos de los fármacos , Glucósidos/farmacología , Óxido Nítrico/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Vasos Coronarios/citología , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Permeabilidad/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transportador 2 de Sodio-Glucosa/genética , Transportador 2 de Sodio-Glucosa/metabolismo , Molécula 1 de Adhesión Celular Vascular
4.
PLoS Comput Biol ; 14(7): e1006239, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29979675

RESUMEN

In experimental assays of angiogenesis in three-dimensional fibrin matrices, a temporary scaffold formed during wound healing, the type and composition of fibrin impacts the level of sprouting. More sprouts form on high molecular weight (HMW) than on low molecular weight (LMW) fibrin. It is unclear what mechanisms regulate the number and the positions of the vascular-like structures in cell cultures. To address this question, we propose a mechanistic simulation model of endothelial cell migration and fibrin proteolysis by the plasmin system. The model is a hybrid, cell-based and continuum, computational model based on the cellular Potts model and sets of partial-differential equations. Based on the model results, we propose that a positive feedback mechanism between uPAR, plasmin and transforming growth factor ß1 (TGFß1) selects cells in the monolayer for matrix invasion. Invading cells releases TGFß1 from the extracellular matrix through plasmin-mediated fibrin degradation. The activated TGFß1 further stimulates fibrin degradation and keeps proteolysis active as the sprout invades the fibrin matrix. The binding capacity for TGFß1 of LMW is reduced relative to that of HMW. This leads to reduced activation of proteolysis and, consequently, reduced cell ingrowth in LMW fibrin compared to HMW fibrin. Thus our model predicts that endothelial cells in LMW fibrin matrices compared to HMW matrices show reduced sprouting due to a lower bio-availability of TGFß1.


Asunto(s)
Simulación por Computador , Fibrinógeno/metabolismo , Fibrinolisina/metabolismo , Neovascularización Fisiológica/fisiología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Disponibilidad Biológica , Movimiento Celular , Células Cultivadas , Células Endoteliales/citología , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Matriz Extracelular/metabolismo , Fibrina/química , Fibrina/metabolismo , Fibrinólisis , Humanos , Técnicas In Vitro , Peso Molecular , Proteolisis , Reproducibilidad de los Resultados
5.
Ann Vasc Surg ; 57: 210-219, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30684630

RESUMEN

BACKGROUND: In abdominal aortic aneurysm (AAA), pathophysiology deterioration of the medial aortic layer plays a critical role. Key players in vessel wall degeneration are reactive oxygen species (ROS), smooth muscle cell apoptosis, and extracellular matrix degeneration by matrix metalloproteinase-9 (MMP-9). Lipocalin-2, also neutrophil gelatinase-associated lipocalin (NGAL), is suggested to be involved in these degenerative processes in other cardiovascular diseases. We aimed to further investigate the role of NGAL in AAA development and rupture. METHODS: In this observational study, aneurysm tissue and blood of ruptured (n = 13) AAA patients were investigated versus nonruptured (n = 26) patients. Nondilated aortas (n = 5) from deceased patients and venous blood from healthy volunteers (n = 10) served as controls. NGAL concentrations in tissue and blood were measured by enzyme-linked immunosorbent assay and immunofluorescence microscopy. Nitrotyrosine (marker of ROS), MMP-9, and caspase-3 (marker of apoptosis) in aneurysm tissue were measured by immunofluorescence microscopy. AAA expansion rates were calculated retrospectively. RESULTS: NGAL (in µg/mL) blood concentration in ruptured AAA was 46 (range 22-122) vs. 26 (range 6-55) in nonruptured AAA (P < 0.01) and 14 (range 12-22) in controls (P < 0.01). In the aneurysm wall of ruptured AAA, NGAL concentration was 4.7 (range 1.4-25) vs. 4.4 (range 0.2-14) in nonruptured AAA (not significant) and 1.8 (range 1.2-2.7) in nondilated aortas (P = 0.04). In the medial layer, NGAL correlated positively with nitrotyrosine (Rs = 0.80, P < 0.01), MMP-9 (Rs = 0.56, P = 0.02), and caspase-3 (Rs = 0.75, P = 0.01). NGAL did not correlate to AAA expansion rate in blood or tissue (P = 0.34 and P = 0.95, respectively). CONCLUSIONS: This study demonstrates that NGAL blood concentration is higher in ruptured AAA patients than in nonruptured AAA. NGAL expression in the AAA wall is also higher than in nondilated aorta. Furthermore, its expression is associated with factors of vessel wall deterioration. Based on our study results, we could not determine NGAL as a biomarker for AAA growth or rupture. However, our findings do support a potential role of NGAL in the development of AAA.


Asunto(s)
Aorta Abdominal/química , Aneurisma de la Aorta Abdominal/sangre , Rotura de la Aorta/sangre , Lipocalina 2/sangre , Adulto , Anciano , Anciano de 80 o más Años , Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/patología , Rotura de la Aorta/patología , Apoptosis , Biomarcadores/sangre , Caspasa 3/análisis , Dilatación Patológica , Progresión de la Enfermedad , Femenino , Humanos , Masculino , Metaloproteinasa 9 de la Matriz/análisis , Persona de Mediana Edad , Estrés Oxidativo , Estudios Retrospectivos , Tirosina/análogos & derivados , Tirosina/análisis , Regulación hacia Arriba , Remodelación Vascular
6.
J Cell Physiol ; 233(10): 6714-6722, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29345319

RESUMEN

Abnormal cutaneous wound healing can lead to formation of fibrotic hypertrophic scars. Although several clinical risk factors have been described, the cross-talk between different cell types resulting in hypertrophic scar formation is still poorly understood. The aim of this in vitro study was to investigate whether endothelial cells (EC) may play a role in skin fibrosis, for example, hypertrophic scar formation after full-thickness skin trauma. Using a collagen/elastin matrix, we developed an in vitro fibrosis model to study the interaction between EC and dermal fibroblasts or adipose tissue-derived mesenchymal stromal cells (ASC). Tissue equivalents containing dermal fibroblasts and EC displayed a normal phenotype. In contrast, tissue equivalents containing ASC and EC displayed a fibrotic phenotype indicated by contraction of the matrix, higher gene expression of ACTA2, COL1A, COL3A, and less secretion of follistatin. The contraction was in part mediated via the TGF-ß pathway, as both inhibition of the ALK4/5/7 receptors and the addition of recombinant follistatin resulted in decreased matrix contraction (75 ± 11% and 24 ± 8%, respectively). In conclusion, our study shows that EC may play a critical role in fibrotic events, as seen in hypertrophic scars, by stimulating ASC-mediated matrix contraction via regulation of fibrosis-related proteins.


Asunto(s)
Cicatriz Hipertrófica/genética , Células Endoteliales/metabolismo , Fibrosis/genética , Células Madre Mesenquimatosas/metabolismo , Actinas/genética , Receptores de Activinas Tipo I/genética , Quinasa de Linfoma Anaplásico/genética , Línea Celular , Movimiento Celular/genética , Cicatriz Hipertrófica/patología , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis/patología , Folistatina/farmacología , Humanos , Células Madre Mesenquimatosas/patología , Receptor Tipo I de Factor de Crecimiento Transformador beta/genética , Piel/lesiones , Piel/metabolismo , Piel/patología , Cicatrización de Heridas/genética
7.
Angiogenesis ; 21(3): 425-532, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29766399

RESUMEN

The formation of new blood vessels, or angiogenesis, is a complex process that plays important roles in growth and development, tissue and organ regeneration, as well as numerous pathological conditions. Angiogenesis undergoes multiple discrete steps that can be individually evaluated and quantified by a large number of bioassays. These independent assessments hold advantages but also have limitations. This article describes in vivo, ex vivo, and in vitro bioassays that are available for the evaluation of angiogenesis and highlights critical aspects that are relevant for their execution and proper interpretation. As such, this collaborative work is the first edition of consensus guidelines on angiogenesis bioassays to serve for current and future reference.


Asunto(s)
Bioensayo/métodos , Neoplasias , Neovascularización Patológica , Animales , Bioensayo/instrumentación , Guías como Asunto , Humanos , Ratones , Neoplasias/irrigación sanguínea , Neoplasias/metabolismo , Neoplasias/patología , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología
8.
Clin Sci (Lond) ; 132(22): 2437-2449, 2018 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-30389858

RESUMEN

MicroRNAs (miRNAs), small non-coding RNAs, have emerged as important, epigenetic regulators of endothelial function. Metabolic disturbances such as diabetes alter miRNA expression. In adults, the miRNA transcriptome as well as endothelial function differ between the sexes. Here, we hypothesized that metabolic disturbances associated with gestational diabetes (GDM) alter miRNA signatures in feto-placental endothelial cells (fpEC), dependent on fetal sex. We isolated human primary fpEC after normal and GDM-complicated pregnancies with male and female neonates and screened for differential miRNA expression using next-generation miRNA sequencing. To test for miRNAs commonly regulated in fpEC of female and male progeny, data were stratified for fetal sex and maternal body mass index (BMI). Analyses were also performed separately for female and male fpEC, again accounting for maternal BMI as covariate. Potential biological pathways regulated by the altered set of miRNAs were determined using mirPath software. Maternal GDM altered 26 miRNA signatures when male and female fpEC were analyzed together. Separate analysis of male versus female fpEC revealed 22 GDM affected miRNAs in the females and only 4 in the males, without overlap. Biological functions potentially modulated by the affected miRNAs related to 'Protein Processing in Endoplasmic Reticulum' and 'Proteoglycans in Cancer'. Maternal GDM alters miRNA signatures in fpEC, and biological functions affected by these miRNAs relate to well-known adverse functional consequences of diabetes on endothelium. GDM effects were highly dependent on fetal sex with miRNA signatures in female fpEC being more susceptible to metabolic derangements of GDM than miRNAs in male fpEC.


Asunto(s)
Diabetes Gestacional/genética , Células Endoteliales/metabolismo , MicroARNs/genética , Placenta/irrigación sanguínea , Circulación Placentaria , Caracteres Sexuales , Transcriptoma , Adulto , Estudios de Casos y Controles , Células Cultivadas , Diabetes Gestacional/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Masculino , MicroARNs/metabolismo , Embarazo , Factores de Riesgo , Análisis para Determinación del Sexo , Factores Sexuales , Adulto Joven
9.
Int J Mol Sci ; 19(12)2018 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-30486375

RESUMEN

In pulmonary arterial hypertension (PAH), lung-angioproliferation leads to increased pulmonary vascular resistance, while simultaneous myocardial microvessel loss contributes to right ventricular (RV) failure. Endothelial colony forming cells (ECFC) are highly proliferative, angiogenic cells that may contribute to either pulmonary vascular obstruction or to RV microvascular adaptation. We hypothesize ECFC phenotypes (outgrowth, proliferation, tube formation) are related to markers of disease severity in a prospective cohort-study of 33 PAH and 30 healthy subjects. ECFC were transplanted in pulmonary trunk banded rats with RV failure. The presence of ECFC outgrowth in PAH patients was associated with low RV ejection fraction, low central venous saturation and a shorter time to clinical worsening (5.4 months (0.6⁻29.2) vs. 36.5 months (7.4⁻63.4), p = 0.032). Functionally, PAH ECFC had higher proliferative rates compared to control in vitro, although inter-patient variability was high. ECFC proliferation was inversely related to RV end diastolic volume (R² = 0.39, p = 0.018), but not pulmonary vascular resistance. Tube formation-ability was similar among donors. Normal and highly proliferative PAH ECFC were transplanted in pulmonary trunk banded rats. While no effect on hemodynamic measurements was observed, RV vascular density was restored. In conclusion, we found that ECFC outgrowth associates with high clinical severity in PAH, suggesting recruitment. Transplantation of highly proliferative ECFC restored myocardial vascular density in pulmonary trunk banded rats, while RV functional improvements were not observed.


Asunto(s)
Biomarcadores , Células Progenitoras Endoteliales/metabolismo , Hipertensión Pulmonar/diagnóstico , Hipertensión Pulmonar/metabolismo , Arteria Pulmonar/fisiopatología , Adulto , Anciano , Animales , Proliferación Celular , Modelos Animales de Enfermedad , Células Progenitoras Endoteliales/trasplante , Femenino , Hemodinámica , Humanos , Hipertensión Pulmonar/mortalidad , Hipertensión Pulmonar/fisiopatología , Masculino , Persona de Mediana Edad , Neovascularización Fisiológica , Pronóstico , Ratas , Índice de Severidad de la Enfermedad , Trasplante de Células Madre , Resistencia Vascular , Disfunción Ventricular Derecha
10.
Angiogenesis ; 20(1): 39-54, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27699500

RESUMEN

During prolonged hypoxic conditions, endothelial cells change their gene expression to adjust to the low oxygen environment. This process is mainly regulated by the hypoxia-inducible factors, HIF-1α and HIF-2α. Although endothelial cells do not form sprouts during prolonged hypoxic culturing, silencing of HIF-2α partially restores sprout formation. The present study identifies novel HIF-2α-target genes that may regulate endothelial sprouting during prolonged hypoxia. The gene expression profile of primary human microvascular endothelial cells (hMVECs) that were cultured at 20 % oxygen was compared to hMVECs that were cultured at 1 % oxygen for 14 days by using genome-wide RNA-sequencing. The differentially regulated genes in hypoxia were compared to the genes that were differentially regulated upon silencing of HIF-2α in hypoxia. Surprisingly, KEGG pathway analysis showed that metabolic pathways were enriched within genes upregulated in response to hypoxia and enriched within genes downregulated upon HIF-2α silencing. Moreover, 51 HIF-2α-regulated genes were screened for their role in endothelial sprouting in hypoxia, of which four genes ARRDC3, MME, PPARG and RALGPS2 directly influenced endothelial sprouting during prolonged hypoxic culturing. The manipulation of specific downstream targets of HIF-2α provides a new, but to be further evaluated, perspective for restoring reduced neovascularization in several pathological conditions, such as diabetic ulcers or other chronic wounds, for improvement of vascularization of implanted tissue-engineered scaffolds.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Células Endoteliales/metabolismo , Regulación de la Expresión Génica , Microvasos/citología , Neovascularización Fisiológica/genética , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/genética , Células Endoteliales/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Humanos , Oxígeno/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transfección
11.
Int J Mol Sci ; 18(9)2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28862673

RESUMEN

Chronic hypoxia is associated with muscle wasting and decreased oxidative capacity. By contrast, training under hypoxia may enhance hypertrophy and increase oxidative capacity as well as oxygen transport to the mitochondria, by increasing myoglobin (Mb) expression. The latter may be a feasible strategy to prevent atrophy under hypoxia and enhance an eventual hypertrophic response to anabolic stimulation. Mb expression may be further enhanced by lipid supplementation. We investigated individual and combined effects of hypoxia, insulin-like growth factor (IGF)-1 and lipids, in mouse skeletal muscle C2C12 myotubes. Differentiated C2C12 myotubes were cultured for 24 h under 20%, 5% and 2% oxygen with or without IGF-1 and/or lipid treatment. In culture under 20% oxygen, IGF-1 induced 51% hypertrophy. Hypertrophy was only 32% under 5% and abrogated under 2% oxygen. This was not explained by changes in expression of genes involved in contractile protein synthesis or degradation, suggesting a reduced rate of translation rather than of transcription. Myoglobin mRNA expression increased by 75% under 5% O2 but decreased by 50% upon IGF-1 treatment under 20% O2, compared to control. Inhibition of mammalian target of rapamycin (mTOR) activation using rapamycin restored Mb mRNA expression to control levels. Lipid supplementation had no effect on Mb gene expression. Thus, IGF-1-induced anabolic signaling can be a strategy to improve muscle size under mild hypoxia, but lowers Mb gene expression.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina/genética , Atrofia Muscular/genética , Mioglobina/genética , Animales , Regulación de la Expresión Génica/genética , Humanos , Hipoxia/genética , Hipoxia/patología , Ratones , Mitocondrias/genética , Mitocondrias/metabolismo , Contracción Muscular/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Atrofia Muscular/patología , Factores Reguladores Miogénicos , Mioglobina/metabolismo , Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Succinato Deshidrogenasa/genética , Serina-Treonina Quinasas TOR/genética , Congéneres de la Testosterona/metabolismo
12.
Angiogenesis ; 19(3): 325-38, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27043316

RESUMEN

Endothelial colony-forming cells (ECFC) are grown from circulating CD34(+) progenitors present in adult peripheral blood, but during in vitro expansion part of the cells lose CD34. To evaluate whether the regulation of CD34 characterizes the angiogenic phenotypical features of PB-ECFCs, we investigated the properties of CD34(+) and CD34(-) ECFCs with respect to their ability to form capillary-like tubes in 3D fibrin matrices, tip-cell gene expression, and barrier integrity. Selection of CD34(+) and CD34(-) ECFCs from subcultured ECFCs was accomplished by magnetic sorting (FACS: CD34(+): 95 % pos; CD34(-): 99 % neg). Both fractions proliferated at same rate, while CD34(+) ECFCs exhibited higher tube-forming capacity and tip-cell gene expression than CD3(4-) cells. However, during cell culture CD34(-) cells re-expressed CD34. Cell-seeding density, cell-cell contact formation, and serum supplements modulated CD34 expression. CD34 expression in ECFCs was strongly suppressed by newborn calf serum. Stimulation with FGF-2, VEGF, or HGF prepared in medium supplemented with 3 % albumin did not change CD34 mRNA or surface expression. Silencing of CD34 with siRNA resulted in strengthening of cell-cell contacts and increased barrier function of ECFC monolayers as measured by ECIS. Furthermore, CD34 siRNA reduced tube formation by ECFC, but did not affect tip-cell gene expression. These findings demonstrate that CD34(+) and CD34(-) cells are different phenotypes of similar cells and that CD34 (1) can be regulated in ECFC; (2) is positively involved in capillary-like sprout formation; (3) is associated but not causally related to tip-cell gene expression; and (4) can affect endothelial barrier function.


Asunto(s)
Células Madre Adultas/citología , Células Madre Adultas/inmunología , Antígenos CD34/metabolismo , Células Endoteliales/citología , Células Endoteliales/inmunología , Células Madre Adultas/metabolismo , Animales , Antígenos CD34/genética , Vasos Sanguíneos/citología , Vasos Sanguíneos/crecimiento & desarrollo , Vasos Sanguíneos/inmunología , Bovinos , Recuento de Células , Proliferación Celular , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Medios de Cultivo , Células Endoteliales/metabolismo , Expresión Génica , Humanos , Neovascularización Fisiológica , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética
13.
Am J Physiol Heart Circ Physiol ; 309(10): H1667-78, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26432845

RESUMEN

Circulating angiogenic cells (CACs) are monocyte-derived cells with endothelial characteristics, which contribute to both angiogenesis and arteriogenesis in a paracrine way. Interferon-ß (IFN-ß) is known to inhibit these divergent processes in animals and patients. We hypothesized that IFN-ß might act by affecting the differentiation and function of CACs. CACs were cultured from peripheral blood mononuclear cells and phenotypically characterized by surface expression of monocytic and endothelial markers. IFN-ß significantly reduced the number of CACs by 18-64%. Apoptosis was not induced by IFN-ß, neither in mononuclear cells during differentiation, nor after maturation to CACs. Rather, IFN-ß impaired adhesion to, and spreading on, fibronectin, which was dependent on α5ß1 (VLA-5)-integrin. IFN-ß affected the function of VLA-5 in mature CACs, leading to rounding and detachment of cells, by induction of calpain 1 activity. Cell rounding and detachment was completely reversed by inhibition of calpain 1 activity in mature CACs. During in vitro capillary formation, CAC addition and calpain 1 inhibition enhanced sprouting of endothelial cells to a comparable extent, but were not sufficient to rescue tube formation in the presence of IFN-ß. We show that the IFN-ß-induced reduction of the numbers of in vitro differentiated CACs is based on activation of calpain 1, resulting in an attenuated adhesion to extracellular matrix proteins via VLA-5. In vivo, this could lead to inhibition of vessel formation due to reduction of the locally recruited CAC numbers and their paracrine angiogenic factors.


Asunto(s)
Calpaína/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Interferón beta/farmacología , Leucocitos Mononucleares/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Apoptosis/efectos de los fármacos , Calpaína/metabolismo , Células Cultivadas , Células Endoteliales/metabolismo , Fibronectinas , Humanos , Técnicas In Vitro , Integrina alfa5beta1/efectos de los fármacos , Integrina alfa5beta1/metabolismo , Leucocitos Mononucleares/metabolismo , Neovascularización Fisiológica/fisiología
14.
J Pathol ; 234(3): 375-85, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25043127

RESUMEN

Angiogenesis is essential during development and in pathological conditions such as chronic inflammation and cancer progression. Inhibition of angiogenesis by targeting vascular endothelial growth factor (VEGF) blocks disease progression, but most patients eventually develop resistance which may result from compensatory signalling pathways. In endothelial cells (ECs), expression of the pro-angiogenic chemokine CXCL12 is regulated by non-canonical nuclear factor (NF)-κB signalling. Here, we report that NF-κB-inducing kinase (NIK) and subsequent non-canonical NF-κB signalling regulate both inflammation-induced and tumour-associated angiogenesis. NIK is highly expressed in endothelial cells (ECs) in tumour tissues and inflamed rheumatoid arthritis synovial tissue. Furthermore, non-canonical NF-κB signalling in human microvascular ECs significantly enhanced vascular tube formation, which was completely blocked by siRNA targeting NIK. Interestingly, Nik(-/-) mice exhibited normal angiogenesis during development and unaltered TNFα- or VEGF-induced angiogenic responses, whereas angiogenesis induced by non-canonical NF-κB stimuli was significantly reduced. In addition, angiogenesis in experimental arthritis and a murine tumour model was severely impaired in these mice. These studies provide evidence for a role of non-canonical NF-κB signalling in pathological angiogenesis, and identify NIK as a potential therapeutic target in chronic inflammatory diseases and tumour neoangiogenesis.


Asunto(s)
Inflamación/metabolismo , Neoplasias Experimentales/metabolismo , Neovascularización Patológica/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/fisiología , Animales , Células Endoteliales/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Inflamación/patología , Ratones , Ratones Noqueados , Neoplasias Experimentales/patología , Quinasa de Factor Nuclear kappa B
15.
J Mol Cell Cardiol ; 67: 94-102, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24389343

RESUMEN

Acute myocardial infarction (AMI) is accompanied by increased expression of Toll like receptors (TLR)-2 and TLR4 on circulating monocytes. In animal models, blocking TLR2/4 signaling reduces inflammatory cell influx and infarct size. The clinical consequences of TLR activation during AMI in humans are unknown, including its role in long-term cardiac functional outcome Therefore, we analyzed gene expression in whole blood samples from 28 patients with an acute ST elevation myocardial infarction (STEMI), enrolled in the EXenatide trial for AMI patients (EXAMI), both at admission and after 4-month follow-up, by whole genome expression profiling and real-time PCR. Cardiac function was determined by cardiac magnetic resonance (CMR) imaging at baseline and after 4-month follow-up. TLR pathway activation was shown by increased expression of TLR4 and its downstream genes, including IL-18R1, IL-18R2, IL-8, MMP9, HIF1A, and NFKBIA. In contrast, expression of the classical TLR-induced genes, TNF, was reduced. Bioinformatics analysis and in vitro experiments explained this noncanonical TLR response by identification of a pivotal role for HIF-1α. The extent of TLR activation and IL-18R1/2 expression in circulating cells preceded massive troponin-T release and correlated with the CMR-measured ischemic area (R=0.48, p=0.01). In conclusion, we identified a novel HIF-1-dependent noncanonical TLR activation pathway in circulating leukocytes leading to enhanced IL-18R expression which correlated with the magnitude of the ischemic area. This knowledge may contribute to our mechanistic understanding of the involvement of the innate immune system during STEMI and may yield diagnostic and prognostic value for patients with myocardial infarction.


Asunto(s)
Interleucina-18/metabolismo , Infarto del Miocardio/fisiopatología , Receptor Toll-Like 4/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Interleucina-18/sangre , Interleucina-18/genética , Leucocitos/metabolismo , Persona de Mediana Edad , Receptor Toll-Like 4/sangre , Receptor Toll-Like 4/genética , Regulación hacia Arriba
16.
J Vasc Res ; 51(3): 231-8, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25116857

RESUMEN

Metallothioneins (MTs) are small cysteine-rich proteins which are involved in e.g. metal homeostasis, metal detoxification and protection against oxidative stress. In addition, several MTs have been shown to regulate expression of proangiogenic growth factors like vascular endothelial growth factor. Detailed information about the expression and regulation of specific MT isoforms in endothelial cells (EC) is limited. We therefore performed extensive mRNA expression profiling of all known human MTs in EC. We found that the basal endothelial expression is restricted to MT1E, MT1X, MT2A, and MT3. Physiological activation of EC by exposure to serum increased the expression of MT1E and MT2A and induced the expression of MT1M. Furthermore, exposure to zinc or copper induced the expression of most MT1 isoforms, while hypoxia specifically increased the expression of MT1E, MT1M, MT1X, and MT3. Finally, knockdown of the dominant MT isoform in EC, i.e. MT2A, resulted in decreased proliferation and sprouting as well as in increased migration of human umbilical vein EC. Together, these findings provide a link between MTs and angiogenesis.


Asunto(s)
Células Endoteliales/metabolismo , Metalotioneína/biosíntesis , Metalotioneína/fisiología , Movimiento Celular/efectos de los fármacos , Cobre/farmacología , Células Endoteliales/efectos de los fármacos , Regulación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana , Humanos , Isoformas de Proteínas/metabolismo , ARN Mensajero/metabolismo , Zinc/farmacología
17.
Int J Mol Sci ; 15(11): 19791-815, 2014 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-25365172

RESUMEN

In patients with chronic wounds, autologous tissue repair is often not sufficient to heal the wound. These patients might benefit from regenerative medicine or the implantation of a tissue-engineered scaffold. Both wound healing and tissue engineering is dependent on the formation of a microvascular network. This process is highly regulated by hypoxia and the transcription factors hypoxia-inducible factors-1α (HIF-1α) and -2α (HIF-2α). Even though much is known about the function of HIF-1α in wound healing, knowledge about the function of HIF-2α in wound healing is lacking. This review focuses on the function of HIF-1α and HIF-2α in microvascular network formation, wound healing, and therapy strategies.


Asunto(s)
Hipoxia , Medicina Regenerativa , Transducción de Señal , Ingeniería de Tejidos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/química , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Humanos , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Neovascularización Fisiológica , Cicatrización de Heridas
18.
Int J Cancer ; 131(4): E484-93, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21953673

RESUMEN

Vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitors are effective agents in the treatment of metastatic renal cell cancer (mRCC). We here investigated whether inhibition of VEGFR signalin by sunitinib causes changes in plasma proteins associated with tumor endothelium. Forty-three patients with mRCC received sunitinib 50 mg/day in a 4-weeks on 2-weeks off schedule. Sequential plasma samples were obtained before treatment (C1D1), on C1D14, on C1D28, and on C2D1 before start of cycle 2. Plasma levels were assessed for VEGF, soluble vascular cell adhesion molecule-1 (sVCAM-1), soluble intercellular cell adhesion molecule-1 (sICAM-1), von Willebrand factor (vWF), circulating angiopoietin-2 (Ang-2) and soluble Tie-2 (sTie-2). Total tumor burden was calculated at baseline and at first evaluation. Progression-free survival (PFS) and overall survival (OS) were determined. Tumor burden was positively associated with baseline circulating Ang-2 [Spearman's rho (ρ) = 0.378, p = 0.028] and vWF (ρ = 0.417, p = 0.008). During sunitinib treatment, circulating Ang-2 and sTie-2 significantly decreased (p < 0.001 for both), plasma levels of sVCAM-1 and VEGF significantly increased (p = 0.022 and p < 0.001), whereas those of sICAM-1 and vWF remained stable. These protein changes had recovered on C2D1. The reduction in circulating Ang-2 levels on C1D28 was positively correlated with the percentage decrease in tumor burden (ρ = 0.605; p = 0.002). Baseline protein levels and subsequent changes were not associated with PFS or OS. In conclusion, sunitinib-induced changes in Ang-2, sTie-2, sVCAM-1 and VEGF are related to the administration schedule, while reduction in Ang-2 is also associated with decrease in tumor burden.


Asunto(s)
Antineoplásicos/farmacología , Endotelio Vascular/efectos de los fármacos , Indoles/farmacología , Neoplasias Renales/tratamiento farmacológico , Pirroles/farmacología , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/uso terapéutico , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Indoles/uso terapéutico , Neoplasias Renales/patología , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Pirroles/uso terapéutico , Sunitinib
19.
Angiogenesis ; 12(3): 275-85, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19449108

RESUMEN

The fibrinous exudate of a wound or tumor stroma facilitates angiogenesis. We studied the involvement of RGD-binding integrins during tube formation in human plasma-derived fibrin clots and human purified fibrin matrices. Capillary-like tube formation by human microvascular endothelial cells in a 3D plasma-derived fibrinous matrix was induced by FGF-2 and TNF-alpha and depended largely on cell-bound u-PA and plasmin activities. While tube formation was minimally affected by the addition of either the alphavbeta3-integrin inhibiting mAb LM609 or the alpha5-integrin inhibiting mAb IIA1, the general RGD-antagonist echistatin completely inhibited this process. Remarkably, when alphavbeta3- and alpha5beta1-integrins were inhibited simultaneously, tube formation was reduced by 78%. It was accompanied by a 44% reduction of u-PA antigen accumulation and 41% less production of fibrin degradation products. alphavbeta5-integrin-blocking antibodies further enhanced the inhibition by mAb LM609 and mAb IIA1 to 94%, but had no effect by themselves. alphav-specific cRGD only inhibited angiogenesis when alpha5beta1-integrin was simultaneously blocked. Endostatin mimicked the effect of alpha5beta1-integrin and inhibited tube formation only in the presence of LM609 or cRGD (73 and 80%, respectively). Comparable results were obtained when purified fibrin matrices were used instead of the plasma-derived fibrinous matrices. These data show that blocking of tube formation in a fibrinous exudate requires the simultaneous inhibition of alphavbeta3- and alpha5beta1-integrins. This may bear impact on attempts to influence angiogenesis in a fibrinous environment.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Capilares/efectos de los fármacos , Fibrina/farmacología , Integrina alfa5beta1/antagonistas & inhibidores , Integrina alfaVbeta3/antagonistas & inhibidores , Neovascularización Fisiológica/efectos de los fármacos , Inhibidores de la Angiogénesis/farmacología , Anticuerpos Monoclonales/administración & dosificación , Capilares/crecimiento & desarrollo , Capilares/metabolismo , Técnicas de Cultivo de Célula , Células Cultivadas , Combinación de Medicamentos , Evaluación Preclínica de Medicamentos , Endostatinas/farmacología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Matriz Extracelular/química , Fibrina/química , Humanos , Integrina alfa5beta1/inmunología , Integrina alfa5beta1/fisiología , Integrina alfaVbeta3/inmunología , Integrina alfaVbeta3/fisiología , Oligopéptidos/metabolismo , Unión Proteica , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
20.
Cardiovasc Res ; 78(2): 203-12, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18079100

RESUMEN

Sprouting angiogenesis is an invasive process that involves proteolytic activities required for the degradation of the endothelial basement membrane, cell migration with removal of obstructing matrix proteins, and generation of space in the matrix to allow endothelial cells to form a proper lumen. In the last decade it has become clear that besides these matrix-degrading properties, proteases exert additional, more subtle functions that play a key role in angiogenesis. These functions are discussed with specific emphasis on membrane type-1 matrix metalloproteinase (MT1-MMP), other MMPs, and the related ADAMs (a disintegrin and metalloproteinase domain). Proteases modulate the balance between pro- and anti-angiogenic factors by activation and modification of growth factors and chemokines, ectodomain shedding with accompanied receptor activation, shedding of cytokines from membrane-bound precursors, and generation of (matrix) protein fragments that inhibit or activate angiogenesis. Furthermore, they participate in the recruitment of leukocytes and progenitor cells, which contribute to the onset and progression of angiogenesis. Proteases facilitate the mobilization of progenitor cells in the bone marrow as well as the entry of these cells and leukocytes into the angiogenic area. The interaction between pericytes and the newly formed endothelial tubes is accompanied by silencing of MMP activities. Better understanding of the various activities of proteases may be helpful in developing more specific inhibitors that could result in tailor-made modification of proteolytic activities in disease.


Asunto(s)
Movimiento Celular , Células Endoteliales/metabolismo , Matriz Extracelular/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Neovascularización Fisiológica , Transducción de Señal , Proteínas ADAM/metabolismo , Proteínas Angiogénicas/metabolismo , Proteínas Angiostáticas/metabolismo , Animales , Células de la Médula Ósea/metabolismo , Capilares/metabolismo , Células Endoteliales/enzimología , Humanos , Metaloproteinasas de la Matriz Asociadas a la Membrana/metabolismo , Pericitos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA