Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Mol Biol Rep ; 47(4): 2561-2572, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32133604

RESUMEN

PURPOSE: We have previously identified insulin-like growth factor 2 (IGF2) and insulin-like growth factor 1 receptor (IGF1R) as essential proteins for tip cell maintenance and sprouting angiogenesis. In this study, we aim to identify other IGF family members involved in endothelial sprouting angiogenesis. METHODS: Effects on sprouting were analyzed in human umbilical vein endothelial cells (HUVECs) using the spheroid-based sprouting model, and were quantified as mean number of sprouts per spheroid and average sprout length. RNA silencing technology was used to knockdown gene expression. Recombinant forms of the ligands (IGF1 and IGF2, insulin) and the IGF-binding proteins (IGFBP) 3 and 4 were used to induce excess effects. Effects on the tip cell phenotype were analyzed by measuring the fraction of CD34+ tip cells using flow cytometry and immunohistochemistry in a 3D angiogenesis model. Experiments were performed in the presence and absence of serum. RESULTS: Knockdown of IGF2 inhibited sprouting in HUVECs, in particular when cultured in the absence of serum, suggesting that components in serum influence the signaling of IGF2 in angiogenesis in vitro. We then determined the effects of IGFBP3 and IGFBP4, which are both present in serum, on IGF2-IGF1R signaling in sprouting angiogenesis in the absence of serum: knockdown of IGFBP3 significantly reduced sprouting angiogenesis, whereas knockdown of IGFBP4 resulted in increased sprouting angiogenesis in both flow cytometry analysis and immunohistochemical analysis of the 3D angiogenesis model. Other IGF family members except INSR did not affect IGF2-IGF1R signaling. CONCLUSIONS: Serum components and IGF binding proteins regulate IGF2 effects on sprouting angiogenesis. Whereas IGFBP3 acts as co-factor for IGF2-IGF1R binding, IGFBP4 inhibits IGF2 signaling.


Asunto(s)
Inductores de la Angiogénesis/metabolismo , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Proteína 4 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Línea Celular Tumoral , Técnicas de Silenciamiento del Gen , Células Endoteliales de la Vena Umbilical Humana , Humanos , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/fisiología , Proteína 4 de Unión a Factor de Crecimiento Similar a la Insulina/fisiología , Factor I del Crecimiento Similar a la Insulina , Factor II del Crecimiento Similar a la Insulina , Morfogénesis , Neovascularización Patológica/metabolismo , Organoides/metabolismo , Receptor IGF Tipo 1 , Receptor IGF Tipo 2 , Transducción de Señal
2.
Br J Cancer ; 121(2): 139-149, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31235865

RESUMEN

BACKGROUND: Crenolanib is a tyrosine kinase inhibitor targeting PDGFR-α, PDGFR-ß and Fms related tyrosine kinase-3 (FLT3) that is currently evaluated in several clinical trials. Although platelet-derived growth factor receptor (PDGFR) signalling pathway is believed to play an important role in angiogenesis and maintenance of functional vasculature, we here demonstrate a direct angiostatic activity of crenolanib independently of PDGFR signalling. METHODS: The activity of crenolanib on cell viability, migration, sprouting, apoptosis and mitosis was assessed in endothelial cells, tumour cells and fibroblasts. Alterations in cell morphology were determined by immunofluorescence experiments. Flow-cytometry analysis and mRNA expression profiles were used to investigate cell differentiation. In vivo efficacy was investigated in human ovarian carcinoma implanted on the chicken chorioallantoic membrane (CAM). RESULTS: Crenolanib was found to inhibit endothelial cell viability, migration and sprout length, and induced apoptosis independently of PDGFR expression. Treated cells  showed altered actin arrangement and nuclear aberrations. Mitosis was affected at several levels including mitosis entry and centrosome clustering. Crenolanib suppressed human ovarian carcinoma tumour growth and angiogenesis in the CAM model. CONCLUSIONS: The PDGFR/FLT3 inhibitor crenolanib targets angiogenesis and inhibits tumour growth in vivo unrelated to PDGFR expression. Based on our findings, we suggest a broad mechanism of action of crenolanib.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antineoplásicos/farmacología , Bencimidazoles/farmacología , Moduladores de la Mitosis/farmacología , Piperidinas/farmacología , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Tirosina Quinasa 3 Similar a fms/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Pollos , Femenino , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Neoplasias Ováricas/irrigación sanguínea , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Receptores del Factor de Crecimiento Derivado de Plaquetas/análisis , Receptores del Factor de Crecimiento Derivado de Plaquetas/fisiología
3.
Angiogenesis ; 21(4): 823-836, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29951828

RESUMEN

Tip cells, the leading cells of angiogenic sprouts, were identified in cultures of human umbilical vein endothelial cells (HUVECs) by using CD34 as a marker. Here, we show that tip cells are also present in primary human microvascular endothelial cells (hMVECs), a more relevant endothelial cell type for angiogenesis. By means of flow cytometry, immunocytochemistry, and qPCR, it is shown that endothelial cell cultures contain a dynamic population of CD34+ cells with many hallmarks of tip cells, including filopodia-like extensions, elevated mRNA levels of known tip cell genes, and responsiveness to stimulation with VEGF and inhibition by DLL4. Furthermore, we demonstrate that our in vitro tip cell model can be exploited to investigate cellular and molecular mechanisms in tip cells and to discover novel targets for anti-angiogenesis therapy in patients. Small interfering RNA (siRNA) was used to knockdown gene expression of the known tip cell genes angiopoietin 2 (ANGPT2) and tyrosine kinase with immunoglobulin-like and EGF-like domains 1 (TIE1), which resulted in similar effects on tip cells and sprouting as compared to inhibition of tip cells in vivo. Finally, we identified two novel tip cell-specific genes in CD34+ tip cells in vitro: insulin-like growth factor 2 (IGF2) and IGF-1-receptor (IGF1R). Knockdown of these genes resulted in a significant decrease in the fraction of tip cells and in the extent of sprouting in vitro and in vivo. In conclusion, this study shows that by using our in vitro tip cell model, two novel essential tip cells genes are identified.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/metabolismo , Factor II del Crecimiento Similar a la Insulina/metabolismo , Microvasos/metabolismo , Receptores de Somatomedina/metabolismo , Angiopoyetina 2/genética , Angiopoyetina 2/metabolismo , Animales , Embrión de Pollo , Técnicas de Silenciamiento del Gen , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Factor II del Crecimiento Similar a la Insulina/genética , Microvasos/citología , Receptor IGF Tipo 1 , Receptor TIE-1/genética , Receptor TIE-1/metabolismo , Receptores de Somatomedina/genética , Pez Cebra
4.
Curr Opin Lipidol ; 27(2): 181-6, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26959705

RESUMEN

PURPOSE OF REVIEW: This article summarizes the current evidence to support a role of sulfatase 2 (SULF2) in triglyceride-rich lipoprotein (TRL) metabolism and angiogenesis. RECENT FINDINGS: Heparan sulfate proteoglycans (HSPG) are involved in the hepatic clearance of TRLs in mice and in humans. Different genetically modified mouse models have been instrumental to provide evidence that syndecan1, the core protein of HSPG, but also the degree of sulfation of the heparin sulfate chain, attached to syndecan 1, is important for hepatic TRL metabolism. Studies in humans demonstrate the regulating role of SULF2 in the hepatic uptake of TRL by HSPG and demonstrate the importance of 6-O-sulfation, modulated by SULF2, for HSPG function. The role of SULF2 in angiogenesis is illustrated by increased SULF2 mRNA expression in the stalk cells of angiogenic vascular sprouts that use fatty acids derived from TRL as a source for biomass production. Interestingly, SULF2 also interferes with HSPG-vascular endothelial growth factor binding, which impacts upon the angiogenic properties of stalk cells. SUMMARY: SULF2 is a multifaceted protein involved in TRL homeostasis and angiogenesis. Future investigations should focus on the potential benefits of targeting SULF2 in atherosclerosis and angiogenesis.


Asunto(s)
Lipoproteínas/metabolismo , Neovascularización Fisiológica , Sulfotransferasas/fisiología , Animales , Aterosclerosis/enzimología , Heparitina Sulfato/metabolismo , Humanos , Metabolismo de los Lípidos , Sulfatasas , Factor A de Crecimiento Endotelial Vascular/fisiología
5.
Angiogenesis ; 18(3): 233-44, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25824484

RESUMEN

Drug combinations can improve angiostatic cancer treatment efficacy and enable the reduction of side effects and drug resistance. Combining drugs is non-trivial due to the high number of possibilities. We applied a feedback system control (FSC) technique with a population-based stochastic search algorithm to navigate through the large parametric space of nine angiostatic drugs at four concentrations to identify optimal low-dose drug combinations. This implied an iterative approach of in vitro testing of endothelial cell viability and algorithm-based analysis. The optimal synergistic drug combination, containing erlotinib, BEZ-235 and RAPTA-C, was reached in a small number of iterations. Final drug combinations showed enhanced endothelial cell specificity and synergistically inhibited proliferation (p < 0.001), but not migration of endothelial cells, and forced enhanced numbers of endothelial cells to undergo apoptosis (p < 0.01). Successful translation of this drug combination was achieved in two preclinical in vivo tumor models. Tumor growth was inhibited synergistically and significantly (p < 0.05 and p < 0.01, respectively) using reduced drug doses as compared to optimal single-drug concentrations. At the applied conditions, single-drug monotherapies had no or negligible activity in these models. We suggest that FSC can be used for rapid identification of effective, reduced dose, multi-drug combinations for the treatment of cancer and other diseases.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica , Neoplasias/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Algoritmos , Animales , Apoptosis , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Movimiento Celular , Supervivencia Celular , Pollos , Membrana Corioalantoides/metabolismo , Cimenos , Ensayos de Selección de Medicamentos Antitumorales , Células Endoteliales/citología , Retroalimentación , Femenino , Humanos , Imidazoles/administración & dosificación , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Compuestos Organometálicos/administración & dosificación , Neoplasias Ováricas/patología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Quinolinas/administración & dosificación , Procesos Estocásticos
6.
Arterioscler Thromb Vasc Biol ; 32(8): 1790-8, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22723443

RESUMEN

OBJECTIVE: The purpose of this study was to fine map previously identified quantitative trait loci affecting atherosclerosis in mice using association analysis. METHODS AND RESULTS: We recently showed that high-resolution association analysis using common inbred strains of mice is feasible if corrected for population structure. To use this approach for atherosclerosis, which requires a sensitizing mutation, we bred human apolipoprotein B-100 transgenic mice with 22 different inbred strains to produce F1 heterozygotes. Mice carrying the dominant transgene were tested for association with high-density single nucleotide polymorphism maps. Here, we focus on high-resolution mapping of the previously described atherosclerosis 30 locus on chromosome 1. Compared with the previous linkage analysis, association improved the resolution of the atherosclerosis 30 locus by more than an order of magnitude. Using expression quantitative trait locus analysis, we identified one of the genes in the region, desmin, as a strong candidate. CONCLUSIONS: Our high-resolution mapping approach accurately identifies and fine maps known atherosclerosis quantitative trait loci. These results suggest that high-resolution genome-wide association analysis for atherosclerosis is feasible in mice.


Asunto(s)
Arteriosclerosis/genética , Mapeo Cromosómico , Sitios de Carácter Cuantitativo , Animales , Arteriosclerosis/etiología , Femenino , Lipoproteínas HDL/sangre , Masculino , Ratones , Ratones Endogámicos , Polimorfismo de Nucleótido Simple , Factores de Riesgo
7.
BMJ Case Rep ; 16(5)2023 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-37221000

RESUMEN

Infectious scleritis is a rare disease entity with potentially devastating visual sequelae. Here we present the clinical history, work-up and aetiology of an unusual case of infectious scleritis.


Asunto(s)
Escleritis , Humanos , Progresión de la Enfermedad , Moraxella , Enfermedades Raras
8.
Artículo en Inglés | MEDLINE | ID: mdl-36728009

RESUMEN

PURPOSE: To describe a case of bilateral diffuse melanocytic proliferation (BDUMP) that was mistaken for nivolumab-induced Vogt-Koyanagi-Harada-like syndrome. METHODS: We present the case of a 58-year-old Caucasian male with metastatic renal clear cell carcinoma for which he was palliatively treated with intravenous nivolumab immunotherapy. Patient developed subacute onset of blurry vision and grey spots in the visual fields of both eyes, macular subretinal fluid, thickening of the RPE and swollen optic nerve heads. Differential diagnosis included nivolumab-induced Vogt-Koyanagi-Harada-disease-like syndrome (nVKH) and patient was initially treated with steroids, which gave no improvement. Investigation showed the development of dark-grey patches in the peripheral retina of both eyes and cataract, which prompted re-evaluation of the diagnosis, deeming BDUMP most likely. The patient was successfully treated with plasmapheresis. DISCUSSION: The initial presentation of the case was incorrectly diagnosed as nVKH. Upon repeat studies of the patients' symptoms and imaging, we concluded we had missed signs of BDUMP. CONCLUSION: The diagnosis BDUMP was missed in the first evaluation. We present this case to discuss the similarities and differences between this disease and nVKH. More importantly, we want to highlight that re-evaluation of the diagnosis upon worsening of a disease was key in this unusual case.

9.
Cells ; 10(4)2021 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-33923753

RESUMEN

During angiogenesis, vascular endothelial growth factor A (VEGFA) regulates endothelial cell (EC) survival, tip cell formation, and stalk cell proliferation via VEGF receptor 2 (VEGFR2). VEGFR2 can interact with VEGFR2 co-receptors such as heparan sulfate proteoglycans (HSPGs) and neuropilin 2 (NRP2), but the exact roles of these co-receptors, or of sulfatase 2 (SULF2), an enzyme that removes sulfate groups from HSPGs and inhibits HSPG-mediated uptake of very low density lipoprotein (VLDL), in angiogenesis and tip cell biology are unknown. In the present study, we investigated whether the modulation of binding of VEGFA to VEGFR2 by knockdown of SULF2 or NRP2 affects sprouting angiogenesis, tip cell formation, proliferation of non-tip cells, and EC survival, or uptake of VLDL. To this end, we employed VEGFA splice variant 121, which lacks an HSPG binding domain, and VEGFA splice variant 165, which does have this domain, in in vitro models of angiogenic tip cells and vascular sprouting. We conclude that VEGFA165 and VEGFA121 have similar inducing effects on tip cells and sprouting in vitro, and that the binding of VEGFA165 to HSPGs in the extracellular matrix does not seem to play a role, as knockdown of SULF2 did not alter these effects. Co-binding of NRP2 appears to regulate VEGFA-VEGFR2-induced sprout initiation, but not tip cell formation. Finally, as the addition of VLDL increased sprout formation but not tip cell formation, and as VLDL uptake was limited to non-tip cells, our findings suggest that VLDL plays a role in sprout formation by providing biomass for stalk cell proliferation.


Asunto(s)
Heparitina Sulfato/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Neovascularización Fisiológica , Neuropilina-2/metabolismo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo , Apoptosis , Humanos , Lipoproteínas VLDL/metabolismo , Sulfatasas/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
10.
PLoS One ; 11(11): e0159478, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27828952

RESUMEN

Angiogenesis involves the formation of new blood vessels by sprouting or splitting of existing blood vessels. During sprouting, a highly motile type of endothelial cell, called the tip cell, migrates from the blood vessels followed by stalk cells, an endothelial cell type that forms the body of the sprout. To get more insight into how tip cells contribute to angiogenesis, we extended an existing computational model of vascular network formation based on the cellular Potts model with tip and stalk differentiation, without making a priori assumptions about the differences between tip cells and stalk cells. To predict potential differences, we looked for parameter values that make tip cells (a) move to the sprout tip, and (b) change the morphology of the angiogenic networks. The screening predicted that if tip cells respond less effectively to an endothelial chemoattractant than stalk cells, they move to the tips of the sprouts, which impacts the morphology of the networks. A comparison of this model prediction with genes expressed differentially in tip and stalk cells revealed that the endothelial chemoattractant Apelin and its receptor APJ may match the model prediction. To test the model prediction we inhibited Apelin signaling in our model and in an in vitro model of angiogenic sprouting, and found that in both cases inhibition of Apelin or of its receptor APJ reduces sprouting. Based on the prediction of the computational model, we propose that the differential expression of Apelin and APJ yields a "self-generated" gradient mechanisms that accelerates the extension of the sprout.


Asunto(s)
Vasos Sanguíneos/fisiología , Biología Computacional/métodos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Neovascularización Fisiológica/fisiología , Transducción de Señal/fisiología , Algoritmos , Animales , Apelina , Receptores de Apelina , Vasos Sanguíneos/citología , Vasos Sanguíneos/metabolismo , Movimiento Celular/fisiología , Quimiotaxis/fisiología , Simulación por Computador , Células Endoteliales/citología , Células Endoteliales/fisiología , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Modelos Biológicos , Interferencia de ARN , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
12.
PLoS One ; 11(6): e0157902, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27352134

RESUMEN

The sialomucins CD34 and podocalyxin (PODXL) are anti-adhesive molecules expressed at the luminal membrane of endothelial cells of small blood vessels and facilitate vascular lumen formation in the developing mouse aorta. CD34 transcript and protein levels are increased during human angiogenesis, its expression is particularly enriched on endothelial tip cell filopodia and CD34 is a marker for tip cells in vitro. Here, we investigated whether CD34 merely marks endothelial tip cells or has a functional role in tip cells and angiogenesis. We assessed that silencing CD34 in human microvascular endothelial cells has little effect on endothelial cell migration or invasion, but has a significant effect on vascular-endothelial growth factor-induced angiogenic sprouting activity in vitro. In vivo, the absence of CD34 reduced the density of filopodia on retinal endothelial tip cells in neonatal mice, but did not influence the overall architecture of the retinal vascular network. In oxygen-induced retinopathy, Cd34-/- mice showed normal intra-retinal regenerative angiogenesis but the number of pathological epi-retinal neovascular tufts were reduced. We conclude that CD34 is not essential for developmental vascularization in the retina, but its expression promotes the formation of pathological, invasive vessels during neovascularization.


Asunto(s)
Antígenos CD34/metabolismo , Neovascularización Patológica/metabolismo , Retinopatía de la Prematuridad/metabolismo , Animales , Antígenos CD34/genética , Línea Celular , Células Cultivadas , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Oxígeno/toxicidad , Vasos Retinianos/metabolismo , Retinopatía de la Prematuridad/etiología , Retinopatía de la Prematuridad/patología , Sialoglicoproteínas/genética , Sialoglicoproteínas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA