Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Vis ; 30: 17-35, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38586604

RESUMEN

Purpose: Diabetic macular edema (DME) is a sight-threatening complication of diabetes. Consequently, studying the proteome of DME may provide novel insights into underlying molecular mechanisms. Methods: In this study, aqueous humor samples from eyes with treatment-naïve clinically significant DME (n = 13) and age-matched controls (n = 11) were compared with label-free liquid chromatography-tandem mass spectrometry. Additional aqueous humor samples from eyes with treatment-naïve DME (n = 15) and controls (n = 8) were obtained for validation by enzyme-linked immunosorbent assay (ELISA). Best-corrected visual acuity (BCVA) was evaluated, and the severity of DME was measured as central subfield thickness (CST) employing optical coherence tomography. Control samples were obtained before cataract surgery. Significantly changed proteins were identified using a permutation-based calculation, with a false discovery rate of 0.05. A human donor eye with DME and a control eye were used for immunofluorescence. Results: A total of 101 proteins were differentially expressed in the DME. Regulated proteins were involved in complement activation, glycolysis, extracellular matrix interaction, and cholesterol metabolism. The highest-fold change was observed for the fibrinogen alpha chain (fold change = 17.8). Complement components C2, C5, and C8, fibronectin, and hepatocyte growth factor-like protein were increased in DME and correlated with best-corrected visual acuity (BCVA). Ceruloplasmin and complement component C8 correlated with central subfield thickness (CST). Hemopexin, plasma kallikrein, monocyte differentiation antigen CD14 (CD14), and lipopolysaccharide-binding protein (LBP) were upregulated in the DME. LBP was correlated with vascular endothelial growth factor. The increased level of LBP in DME was confirmed using ELISA. The proteins involved in desmosomal integrity, including desmocollin-1 and desmoglein-1, were downregulated in DME and correlated negatively with CST. Immunofluorescence confirmed the extravasation of fibrinogen at the retinal level in the DME. Conclusion: Elevated levels of pro-inflammatory proteins, including the complement components LBP and CD14, were observed in DME. DME was associated with the loss of basal membrane proteins, compromised desmosomal integrity, and perturbation of glycolysis.


Asunto(s)
Diabetes Mellitus , Retinopatía Diabética , Edema Macular , Humanos , Edema Macular/tratamiento farmacológico , Retinopatía Diabética/complicaciones , Proteoma/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Humor Acuoso/metabolismo , Tomografía de Coherencia Óptica , Fibrinógeno/metabolismo , Inyecciones Intravítreas , Inhibidores de la Angiogénesis/uso terapéutico , Diabetes Mellitus/metabolismo
2.
Diabetologia ; 66(8): 1557-1575, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37351595

RESUMEN

AIMS/HYPOTHESIS: Type 2 diabetes is associated with increased risk of cognitive decline although the pathogenic basis for this remains obscure. Deciphering diabetes-linked molecular mechanisms in cells of the cerebral cortex could uncover novel therapeutic targets. METHODS: Single-cell transcriptomic sequencing (scRNA-seq) was conducted on the cerebral cortex in a mouse model of type 2 diabetes (db/db mice) and in non-diabetic control mice in order to identify gene expression changes in distinct cell subpopulations and alterations in cell type composition. Immunohistochemistry and metabolic assessment were used to validate the findings from scRNA-seq and to investigate whether these cell-specific dysfunctions impact the neurovascular unit (NVU). Furthermore, the behavioural and cognitive alterations related to these dysfunctions in db/db mice were assessed via Morris water maze and novel object discrimination tests. Finally, results were validated in post-mortem sections and protein isolates from individuals with type 2 diabetes. RESULTS: Compared with non-diabetic control mice, the db/db mice demonstrated disrupted brain function as revealed by losses in episodic and spatial memory and this occurred concomitantly with dysfunctional NVU, neuronal circuitry and cerebral atrophy. scRNA-seq of db/db mouse cerebral cortex revealed cell population changes in neurons, glia and microglia linked to functional regulatory disruption including neuronal maturation and altered metabolism. These changes were validated through immunohistochemistry and protein expression analysis not just in the db/db mouse cerebral cortex but also in post-mortem sections and protein isolates from individuals with type 2 diabetes (74.3 ± 5.5 years) compared with non-diabetic control individuals (87.0 ± 8.5 years). Furthermore, metabolic and synaptic gene disruptions were evident in cortical NVU cell populations and associated with a decrease in vascular density. CONCLUSIONS/INTERPRETATION: Taken together, our data reveal disruption in the cellular and molecular architecture of the cerebral cortex induced by diabetes, which can explain, at least in part, the basis for progressive cognitive decline in individuals with type 2 diabetes. DATA AVAILABILITY: The single-cell sequencing data that supports this study are available at GEO accession GSE217665 ( https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE217665 ).


Asunto(s)
Disfunción Cognitiva , Diabetes Mellitus Tipo 2 , Ratones , Animales , Diabetes Mellitus Tipo 2/complicaciones , Disfunción Cognitiva/tratamiento farmacológico , Corteza Cerebral/metabolismo , Modelos Animales de Enfermedad
3.
FASEB J ; 33(4): 5320-5333, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30698992

RESUMEN

Insight into the molecular and cellular processes in blood-retinal barrier (BRB) development, including the contribution of paracellular and transcellular pathways, is still incomplete but may help to understand the inverse process of BRB loss in pathologic eye conditions. In this comprehensive observational study, we describe in detail the formation of the BRB at the molecular level in physiologic conditions, using mice from postnatal day (P)3 to P25. Our data indicate that immature blood vessels already have tight junctions at P5, before the formation of a functional BRB. Expression of the endothelial cell-specific protein plasmalemma vesicle-associated protein (PLVAP), which is known to be involved in transcellular transport and associated with BRB permeability, decreased during development and was absent when a functional barrier was formed. Moreover, we show that PLVAP deficiency causes a transient delay in retinal vascular development and changes in mRNA expression levels of endothelial permeability pathway proteins.-Van der Wijk, A.-E., Wisniewska-Kruk, J., Vogels, I. M. C., van Veen, H. A., Ip, W. F., van der Wel, N. N., van Noorden, C. J. F., Schlingemann, R. O., Klaassen, I. Expression patterns of endothelial permeability pathways in the development of the blood-retinal barrier in mice.


Asunto(s)
Barrera Hematorretinal/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de la Membrana/genética , Animales , Barrera Hematorretinal/embriología , Barrera Hematorretinal/ultraestructura , Western Blotting , Exones/genética , Genotipo , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Ratones Mutantes , Microscopía Electrónica de Transmisión , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transcriptoma
4.
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
5.
Ophthalmologica ; 243(6): 426-435, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32623429

RESUMEN

PURPOSE: We hypothesised that cleaning the internal limiting membrane (ILM) with a flexible nitinol loop following diabetic vitrectomy without peeling may reduce the common occurrence of postoperative epiretinal membrane (ERM) formation. METHODS: Consecutive patients undergoing vitrectomy for proliferative diabetic retinopathy by one surgeon from 2015 to 2019 were studied and divided into 2 cohorts: the control group underwent standard surgery, and the ILM clean group underwent additional cleaning of the macular retina using a flexible nitinol loop after vitrectomy. Masked comparison of ERM on optical coherence tomography was performed at 3 months, and visual acuity (VA) was measured until 12 months postoperatively. RESULTS: Baseline demographics, clinical features, and protein levels were similar between cohorts. The ILM clean group (n = 56) had fewer clinically significant ERM than the control group (n = 50; 4 vs. 20%; p = 0.01), and a significantly lower proportion of the ILM clean group required revision surgery (2 vs. 14%; p = 0.02). VA in the ILM clean group was significantly better than in the control group at 3 months (0.35 vs. 0.50 logMAR; p = 0.02) but not at 12 months (0.34 vs. 0.43 logMAR; p = 0.17). CONCLUSION: ILM cleaning with a flexible nitinol loop following diabetic vitrectomy resulted in significant reduction in ERM formation and reduced necessity for revision surgery. There was significant improvement in VA at 3 months but not over a longer follow-up.


Asunto(s)
Diabetes Mellitus , Retinopatía Diabética , Membrana Epirretinal , Vitrectomía , Membrana Basal , Retinopatía Diabética/terapia , Membrana Epirretinal/cirugía , Humanos , Retina , Estudios Retrospectivos , Tomografía de Coherencia Óptica
6.
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
7.
Diabetologia ; 61(11): 2371-2385, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30151615

RESUMEN

AIMS/HYPOTHESIS: The immunomodulatory capacity of adipose tissue-derived stromal cells (ASCs) is relevant for next-generation cell therapies that aim to reverse tissue dysfunction such as that caused by diabetes. Pericyte dropout from retinal capillaries underlies diabetic retinopathy and the subsequent aberrant angiogenesis. METHODS: We investigated the pericytic function of ASCs after intravitreal injection of ASCs in mice with retinopathy of prematurity as a model for clinical diabetic retinopathy. In addition, ASCs influence their environment by paracrine signalling. For this, we assessed the immunomodulatory capacity of conditioned medium from cultured ASCs (ASC-Cme) on high glucose (HG)-stimulated bovine retinal endothelial cells (BRECs). RESULTS: ASCs augmented and stabilised retinal angiogenesis and co-localised with capillaries at a pericyte-specific position. This indicates that cultured ASCs exert juxtacrine signalling in retinal microvessels. ASC-Cme alleviated HG-induced oxidative stress and its subsequent upregulation of downstream targets in an NF-κB dependent fashion in cultured BRECs. Functionally, monocyte adhesion to the monolayers of activated BRECs was also decreased by treatment with ASC-Cme and correlated with a decline in expression of adhesion-related genes such as SELE, ICAM1 and VCAM1. CONCLUSIONS/INTERPRETATION: The ability of ASC-Cme to immunomodulate HG-challenged BRECs is related to the length of time for which ASCs were preconditioned in HG medium. Conditioned medium from ASCs that had been chronically exposed to HG medium was able to normalise the HG-challenged BRECs to normal glucose levels. In contrast, conditioned medium from ASCs that had been exposed to HG medium for a shorter time did not have this effect. Our results show that the manner of HG preconditioning of ASCs dictates their immunoregulatory properties and thus the potential outcome of treatment of diabetic retinopathy.


Asunto(s)
Tejido Adiposo/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Glucosa/farmacología , Pericitos/citología , Pericitos/efectos de los fármacos , Células del Estroma/citología , Animales , Bovinos , Adhesión Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Retinopatía Diabética/metabolismo , Selectina E/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Estrés Oxidativo/efectos de los fármacos , Retina/citología , Transducción de Señal/efectos de los fármacos , Molécula 1 de Adhesión Celular Vascular/metabolismo , Cicatrización de Heridas/efectos de los fármacos
8.
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
9.
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
10.
Ophthalmic Res ; 59(2): 98-109, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29073609

RESUMEN

INTRODUCTION: Curcumin has multiple biological effects including the modulation of protein homeostasis by the ubiquitin-proteasome system. The purpose of this study was to assess the in vitro cytotoxic and oxidative effects of nano-curcumin and standard curcumin and characterize their effects on proteasome regulation in retinal pigment epithelial (RPE) cells. METHODS: Viability, cell cycle progression, and reactive oxygen species (ROS) production were determined after treatment with nano-curcumin or curcumin. Subsequently, the effects of nano-curcumin and curcumin on proteasome activity and the gene and protein expression of proteasome subunits PA28α, α7, ß5, and ß5i were assessed. RESULTS: Nano-curcumin (5-100 µM) did not show significant cytotoxicity or anti-oxidative effects against H2O2-induced oxidative stress, whereas curcumin (≥10 µM) was cytotoxic and a potent inducer of ROS production. Both nano-curcumin and curcumin induced changes in proteasome-mediated proteolytic activity characterized by increased activity of the proteasome subunits ß2 and ß5i/ß1 and reduced activity of ß5/ß1i. Likewise, nano-curcumin and curcumin affected mRNA and protein levels of household and immunoproteasome subunits. CONCLUSIONS: Nano-curcumin is less toxic to RPE cells and less prone to induce ROS production than curcumin. Both nano-curcumin and curcumin increase proteasome-mediated proteolytic activity. These results suggest that nano-curcumin may be regarded as a proteasome-modulating agent of limited cytotoxicity for RPE cells.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Curcumina/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Epitelio Pigmentado de la Retina/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Humanos , Estrés Oxidativo/efectos de los fármacos , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Pigmentos Retinianos/metabolismo
11.
Am J Pathol ; 186(4): 1044-54, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26878208

RESUMEN

Loss of blood-retinal barrier (BRB) properties induced by vascular endothelial growth factor (VEGF) and other factors is an important cause of diabetic macular edema. Previously, we found that the presence of plasmalemma vesicle-associated protein (PLVAP) in retinal capillaries associates with loss of BRB properties and correlates with increased vascular permeability in diabetic macular edema. In this study, we investigated whether absence of PLVAP protects the BRB from VEGF-induced permeability. We used lentiviral-delivered shRNA or siRNA to inhibit PLVAP expression. The barrier properties of in vitro BRB models were assessed by measuring transendothelial electrical resistance, permeability of differently sized tracers, and the presence of endothelial junction complexes. The effect of VEGF on caveolae formation was studied in human retinal explants. BRB loss in vivo was studied in the mouse oxygen-induced retinopathy model. The inhibition of PLVAP expression resulted in decreased VEGF-induced BRB permeability of fluorescent tracers, both in vivo and in vitro. PLVAP inhibition attenuated transendothelial electrical resistance reduction induced by VEGF in BRB models in vitro and significantly increased transendothelial electrical resistance of the nonbarrier human umbilical vein endothelial cells. Furthermore, PLVAP knockdown prevented VEGF-induced caveolae formation in retinal explants but did not rescue VEGF-induced alterations in endothelial junction complexes. In conclusion, PLVAP is an essential cofactor in VEGF-induced BRB permeability and may become an interesting novel target for diabetic macular edema therapy.


Asunto(s)
Barrera Hematorretinal/metabolismo , Permeabilidad Capilar/fisiología , Retinopatía Diabética/metabolismo , Retina/metabolismo , Vasos Retinianos/metabolismo , Vasos Retinianos/ultraestructura , Animales , Permeabilidad Capilar/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Edema Macular/metabolismo , Edema Macular/patología , Ratones , Ratas , Factor A de Crecimiento Endotelial Vascular/metabolismo
12.
Exp Eye Res ; 133: 37-48, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25819453

RESUMEN

Connective tissue growth factor (CTGF, CCN2) contributes to fibrotic responses in diabetic retinopathy, both before clinical manifestations occur in the pre-clinical stage of diabetic retinopathy (PCDR) and in proliferative diabetic retinopathy (PDR), the late clinical stage of the disease. CTGF is a secreted protein that modulates the actions of many growth factors and extracellular matrix (ECM) proteins, leading to tissue reorganization, such as ECM formation and remodeling, basal lamina (BL) thickening, pericyte apoptosis, angiogenesis, wound healing and fibrosis. In PCDR, CTGF contributes to thickening of the retinal capillary BL and is involved in loss of pericytes. In this stage, CTGF expression is induced by advanced glycation end products, and by growth factors such as vascular endothelial growth factor (VEGF) and transforming growth factor (TGF)-ß. In PDR, the switch from neovascularization to a fibrotic phase - the angio-fibrotic switch - in PDR is driven by CTGF, in a critical balance with vascular endothelial growth factor (VEGF). We discuss here the roles of CTGF in the pathogenesis of DR in relation to ECM remodeling and wound healing mechanisms, and explore whether CTGF may be a potential novel therapeutic target in the clinical management of early as well as late stages of DR.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/fisiología , Retinopatía Diabética/metabolismo , Vitreorretinopatía Proliferativa/metabolismo , Proliferación Celular , Retinopatía Diabética/patología , Progresión de la Enfermedad , Humanos , Vitreorretinopatía Proliferativa/patología
13.
Exp Eye Res ; 122: 123-31, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24703908

RESUMEN

The molecular mechanisms of vascular leakage in diabetic macular edema and proliferative retinopathy are poorly understood, mainly due to the lack of reliable in vivo models. The Akimba (Ins2(Akita)VEGF(+/-)) mouse model combines retinal neovascularization with hyperglycemia, and in contrast to other models, displays the majority of signs of advanced clinical diabetic retinopathy (DR). To study the molecular mechanism that underlies the breakdown of the blood-retinal barrier (BRB) in diabetic macular edema and proliferative diabetic retinopathy, we investigated the retinal vasculature of Akimba and its parental mice Kimba (trVEGF029) and Akita (Ins2(Akita)). Quantitative PCR, immunohistochemistry and fluorescein angiography were used to characterize the retinal vasculature with special reference to the inner BRB. Correlations between the degree of fluorescein leakage and retinal gene expression were tested by calculating the Spearman's correlation coefficient. Fluorescein leakage demonstrating BRB loss was observed in Kimba and Akimba, but not in Akita or wild type mice. In Kimba and Akimba mice fluorescein leakage was associated with focal angiogenesis and correlated significantly with Plvap gene expression. PLVAP is an endothelial cell-specific protein that is absent in intact blood-retinal barrier, but its expression significantly increases in pathological conditions such as DR. Furthermore, in Akimba mice BRB disruption was linked to decreased expression of endothelial junction proteins, pericyte dropout and vessel loss. Despite fluorescein leakage, no alteration in BRB protein levels or pericyte coverage was detected in retinas of Kimba mice. In summary, our data not only demonstrate that hyperglycemia sensitizes retinal vasculature to the effects of VEGF, leading to more severe microvascular changes, but also confirm an important role of PLVAP in the regulation of BRB permeability.


Asunto(s)
Barrera Hematorretinal/patología , Retinopatía Diabética/genética , Modelos Animales de Enfermedad , Neovascularización Retiniana/genética , Vasos Retinianos/patología , Proteínas Angiogénicas/genética , Proteínas Angiogénicas/metabolismo , Animales , Antígenos CD34/genética , Antígenos CD34/metabolismo , Permeabilidad Capilar , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Quimiocinas/genética , Quimiocinas/metabolismo , Retinopatía Diabética/metabolismo , Retinopatía Diabética/patología , Endoglina , Angiografía con Fluoresceína , Expresión Génica , Hiperglucemia/genética , Hiperglucemia/patología , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Edema Macular/genética , Edema Macular/patología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Mutantes , Pericitos/patología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Neovascularización Retiniana/metabolismo , Neovascularización Retiniana/patología , Vasos Retinianos/metabolismo , Factor de von Willebrand/genética , Factor de von Willebrand/metabolismo
14.
J Histochem Cytochem ; 72(5): 329-352, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38733294

RESUMEN

Lactate and ATP formation by aerobic glycolysis, the Warburg effect, is considered a hallmark of cancer. During angiogenesis in non-cancerous tissue, proliferating stalk endothelial cells (ECs) also produce lactate and ATP by aerobic glycolysis. In fact, all proliferating cells, both non-cancer and cancer cells, need lactate for the biosynthesis of building blocks for cell growth and tissue expansion. Moreover, both non-proliferating cancer stem cells in tumors and leader tip ECs during angiogenesis rely on glycolysis for pyruvate production, which is used for ATP synthesis in mitochondria through oxidative phosphorylation (OXPHOS). Therefore, aerobic glycolysis is not a specific hallmark of cancer but rather a hallmark of proliferating cells and limits its utility in cancer therapy. However, local treatment of angiogenic eye conditions with inhibitors of glycolysis may be a safe therapeutic option that warrants experimental investigation. Most types of cells in the eye such as photoreceptors and pericytes use OXPHOS for ATP production, whereas proliferating angiogenic stalk ECs rely on glycolysis for lactate and ATP production. (J Histochem Cytochem XX.XXX-XXX, XXXX).


Asunto(s)
Adenosina Trifosfato , Neoplasias , Neovascularización Patológica , Humanos , Adenosina Trifosfato/metabolismo , Adenosina Trifosfato/biosíntesis , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Animales , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Glucólisis , Oftalmopatías/metabolismo , Oftalmopatías/patología , Fosforilación Oxidativa
15.
Sci Rep ; 13(1): 21436, 2023 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-38052807

RESUMEN

Vascular endothelial growth factor (VEGF)-A induces endothelial hyperpermeability, but the molecular pathways remain incompletely understood. Endothelial nitric oxide synthase (eNOS) regulates acute effects of VEGF-A on permeability of endothelial cells (ECs), but it remains unknown whether and how eNOS regulates late effects of VEGF-A-induced hyperpermeability. Here we show that VEGF-A induces hyperpermeability via eNOS-dependent and eNOS-independent mechanisms at 2 days after VEGF-A stimulation. Silencing of expression of the eNOS gene (NOS3) reduced VEGF-A-induced permeability for dextran (70 kDa) and 766 Da-tracer in human dermal microvascular ECs (HDMVECs), but not in human retinal microvascular ECs (HRECs) and human umbilical vein ECs (HUVECs). However, silencing of NOS3 expression in HRECs increased permeability to dextran, BSA and 766 Da-tracer in the absence of VEGF-A stimulation, suggesting a barrier-protective function of eNOS. We also investigated how silencing of NOS3 expression regulates the expression of permeability-related transcripts, and found that NOS3 silencing downregulates the expression of PLVAP, a molecule associated with trans-endothelial transport via caveolae, in HDMVECs and HUVECs, but not in HRECs. Our findings underscore the complexity of VEGF-A-induced permeability pathways in ECs and the role of eNOS therein, and demonstrate that different pathways are activated depending on the EC phenotype.


Asunto(s)
Óxido Nítrico Sintasa de Tipo III , Factor A de Crecimiento Endotelial Vascular , Humanos , Caveolas/metabolismo , Células Cultivadas , Dextranos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/farmacología
16.
Angiogenesis ; 15(1): 151-63, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22249946

RESUMEN

The functional shift of quiescent endothelial cells into tip cells that migrate and stalk cells that proliferate is a key event during sprouting angiogenesis. We previously showed that the sialomucin CD34 is expressed in a small subset of cultured endothelial cells and that these cells extend filopodia: a hallmark of tip cells in vivo. In the present study, we characterized endothelial cells expressing CD34 in endothelial monolayers in vitro. We found that CD34-positive human umbilical vein endothelial cells show low proliferation activity and increased mRNA expression of all known tip cell markers, as compared to CD34-negative cells. Genome-wide mRNA profiling analysis of CD34-positive endothelial cells demonstrated enrichment for biological functions related to angiogenesis and migration, whereas CD34-negative cells were enriched for functions related to proliferation. In addition, we found an increase or decrease of CD34-positive cells in vitro upon exposure to stimuli that enhance or limit the number of tip cells in vivo, respectively. Our findings suggest cells with virtually all known properties of tip cells are present in vascular endothelial cell cultures and that they can be isolated based on expression of CD34. This novel strategy may open alternative avenues for future studies of molecular processes and functions in tip cells in angiogenesis.


Asunto(s)
Antígenos CD34/metabolismo , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Neovascularización Fisiológica , Biomarcadores/metabolismo , Recuento de Células , Proliferación Celular , Forma de la Célula , Células Cultivadas , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Neovascularización Fisiológica/genética , Fenotipo , Reacción en Cadena de la Polimerasa , Seudópodos/metabolismo , Regulación hacia Arriba
17.
Exp Eye Res ; 96(1): 181-90, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22200486

RESUMEN

Loss of blood-retinal barrier (BRB) properties is an important feature in the pathology of diabetic macular edema (DME), but cellular mechanisms underlying BRB dysfunction are poorly understood. Therefore, we developed and characterized a novel in vitro BRB model, based on primary bovine retinal endothelial cells (BRECs). These cells were shown to maintain specific in vivo BRB properties by expressing high levels of the endothelial junction proteins occludin, claudin-5, VE-cadherin and ZO-1 at cell borders, and the specific pumps glucose transporter-1 (GLUT1) and efflux transporter P-glycoprotein (MDR1). To investigate the influence of pericytes and astrocytes on BRB maintenance in vitro, we compared five different co-culture BRB models, based on BRECs, bovine retinal pericytes (BRPCs) and rat glial cells. Co-cultures of BRECs with BRPCs and glial cells showed the highest trans-endothelial resistance (TEER) as well as decreased permeability of tracers after vascular endothelial growth factor (VEGF) stimulation, suggesting a major role for these cell types in maintaining barrier properties. To mimic the in vivo situation of DME, we stimulated BRECs with VEGF, which downregulated MDR1 and GLUT1 mRNA levels, transiently reduced expression levels of endothelial junctional proteins and altered their organization, increased the number of intercellular gaps in BRECs monolayers and influence the permeability of the model to differently-sized molecular tracers. Moreover, as has been shown in vivo, expression of plasmalemma vesicle-associated protein (PLVAP) was increased in endothelial cells in the presence of VEGF. This in vitro model is the first co-culture model of the BRB that mimicks in vivo VEGF-dependent changes occurring in DME.


Asunto(s)
Astrocitos/citología , Barrera Hematorretinal/fisiología , Endotelio Vascular/citología , Modelos Biológicos , Pericitos/citología , Vasos Retinianos/citología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Actinas/genética , Animales , Astrocitos/metabolismo , Barrera Hematorretinal/efectos de los fármacos , Permeabilidad Capilar , Bovinos , Técnicas de Cocultivo , Impedancia Eléctrica , Endotelio Vascular/metabolismo , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente Indirecta , Transportador de Glucosa de Tipo 1/genética , Microscopía Electrónica , Pericitos/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Uniones Estrechas/fisiología , Factor A de Crecimiento Endotelial Vascular/farmacología
18.
Sci Rep ; 12(1): 21062, 2022 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-36473885

RESUMEN

Extracellular signalling proteins interact in networks rather than in isolation. In this context we investigated vitreous protein levels, including placental growth factor (PlGF), angiopoietin-2 (ANG2) and vascular endothelial growth factor (VEGF), in patients with proliferative diabetic retinopathy (PDR) with variable disease severities, and after anti-VEGF pre-treatment. Vitreous samples of 112 consecutive patients undergoing vitrectomy for PDR and of 52 non-diabetic patients with macular holes as controls were studied. A subset of the PDR patients were treated with either aflibercept (AFB, n = 25) or bevacizumab (BVZ)/ranibizumab (RZB) (n = 13), before surgery. Antibody-based analysis of 35 proteins (growth factors and cytokines) showed a significant increase in expression levels of 27 proteins in PDR patients as compared to controls. In network analysis of co-regulated proteins, a strong correlation in expression levels between VEGF, PlGF, MCP1 and ANG2 was found, mostly clustered around ANG2. In the AFB treatment group, concentrations of several proteins were decreased, including VEGFR1, whereas interleukin 6 and 8 were increased as compared to untreated PDR patients. The observed differences in vitreous protein levels between the different treatments and untreated PDR patients may underlie differences in clinical outcomes in patients with PDR.


Asunto(s)
Diabetes Mellitus , Retinopatía Diabética , Humanos , Femenino , Bevacizumab/farmacología , Bevacizumab/uso terapéutico , Retinopatía Diabética/tratamiento farmacológico , Factor A de Crecimiento Endotelial Vascular , Factor de Crecimiento Placentario
19.
J Histochem Cytochem ; 70(8): 557-569, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35876388

RESUMEN

Endothelial cells (ECs) form a precisely regulated polarized monolayer in capillary walls. Vascular endothelial growth factor-A (VEGF-A) induces endothelial hyperpermeability, and VEGF-A applied to the basolateral side, but not the apical side, has been shown to be a strong barrier disruptor in blood-retinal barrier ECs. We show here that VEGF-A presented to the basolateral side of human umbilical vein ECs (HUVECs) induces higher permeability than apical stimulation, which is similar to results obtained with bovine retinal ECs. We investigated with immunocytochemistry and confocal imaging the distribution of VEGF receptor-2 (VEGFR2) and neuropilin-2 (NRP2) in perinuclear apical and basolateral membrane domains. Orthogonal z-sections of cultured HUVECs were obtained, and the fluorescence intensity at the apical and basolateral membrane compartments was measured. We found that VEGFR2 and NRP2 are evenly distributed throughout perinuclear apical and basolateral membrane compartments in unstimulated HUVECs grown on Transwell inserts, whereas basolateral VEGF-A stimulation induces a shift toward basolateral VEGFR2 and NRP2 localization. When HUVECs were grown on coverslips, the distribution of VEGFR2 and NRP2 across the perinuclear apical and basolateral membrane domains was different. Our findings demonstrate that HUVECs dynamically regulate VEGFR2 and NRP2 localization on membrane microdomains, depending on growth conditions and the polarity of VEGF-A stimulation.


Asunto(s)
Neuropilina-2 , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Bovinos , Membrana Celular/metabolismo , Células Cultivadas , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Neuropilina-2/metabolismo , Retina/metabolismo
20.
Curr Eye Res ; 47(2): 277-286, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34612091

RESUMEN

PURPOSE: The formation of fibrovascular membranes (FVMs) is a serious sight-threatening complication of proliferative diabetic retinopathy (PDR) that may result in retinal detachment and eventual blindness. During the formation of these membranes, neurite/process outgrowth occurs in retinal neurons and glial cells, which may both serve as a scaffold and have guiding or regulatory roles. To further understand this process, we investigated whether previously identified candidate proteins, from vitreous of PDR patients with FVMs, could induce neurite outgrowth in an experimental setting. MATERIALS AND METHODS: Retinal explants of C57BL6/N mouse pups on postnatal day 3 (P3) were cultured in poly-L-lysine- and laminin-coated dishes. Outgrowth stimulation experiments were performed with the addition of potential inducers of neurite outgrowth. Automated analysis of neurite outgrowth was performed by measuring ß-tubulin-immunopositive neurites using Image J. Expression of PDGF receptors was quantified by RT-PCR in FVMs of PDR patients. RESULTS: Platelet-derived growth factor (PDGF) induced neurite outgrowth in a concentration-dependent manner, whilst neuregulin 1 (NRG1) and connective tissue growth factor (CTGF) did not. When comparing three different PDGF dimers, treatment with PDGF-AB resulted in the highest neurite induction, followed by PDGF-AA and -BB. In addition, incubation of retinal explants with vitreous from PDR patients resulted in a significant induction of neurite outgrowth as compared to non-diabetic control vitreous from patients with macular holes, which could be prevented by addition of CP673451, a potent PDGF receptor (PDGFR) inhibitor. Abundant expression of PDGF receptors was detected in FVMs. CONCLUSION: Our findings suggest that PDGF may be involved in the retinal neurite outgrowth, which is associated with the formation of FVMs in PDR.


Asunto(s)
Diabetes Mellitus , Retinopatía Diabética , Animales , Retinopatía Diabética/metabolismo , Humanos , Ratones , Proyección Neuronal , Factor de Crecimiento Derivado de Plaquetas/farmacología , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Cuerpo Vítreo/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA