Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
1.
Dev Biol ; 479: 11-22, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34310924

RESUMEN

Platelet derived growth factor beta and its receptor, Pdgfrb, play essential roles in the development of vascular mural cells, including pericytes and vascular smooth muscle cells. To determine if this role was conserved in zebrafish, we analyzed pdgfb and pdgfrb mutant lines. Similar to mouse, pdgfb and pdgfrb mutant zebrafish lack brain pericytes and exhibit anatomically selective loss of vascular smooth muscle coverage. Despite these defects, pdgfrb mutant zebrafish did not otherwise exhibit circulatory defects at larval stages. However, beginning at juvenile stages, we observed severe cranial hemorrhage and vessel dilation associated with loss of pericytes and vascular smooth muscle cells in pdgfrb mutants. Similar to mouse, pdgfrb mutant zebrafish also displayed structural defects in the glomerulus, but normal development of hepatic stellate cells. We also noted defective mural cell investment on coronary vessels with concomitant defects in their development. Together, our studies support a conserved requirement for Pdgfrb signaling in mural cells. In addition, these zebrafish mutants provide an important model for definitive investigation of mural cells during early embryonic stages without confounding secondary effects from circulatory defects.


Asunto(s)
Músculo Liso Vascular/metabolismo , Pericitos/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Animales , Diferenciación Celular , Vasos Coronarios/metabolismo , Desarrollo Embrionario , Músculo Liso Vascular/embriología , Miocitos del Músculo Liso/metabolismo , Proteínas Proto-Oncogénicas c-sis/metabolismo , Proteínas Proto-Oncogénicas c-sis/fisiología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Transducción de Señal/genética , Pez Cebra/embriología , Proteínas de Pez Cebra/metabolismo
2.
JCI Insight ; 6(6)2021 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-33591958

RESUMEN

Excess macrophages and smooth muscle cells (SMCs) characterize many cardiovascular diseases, but crosstalk between these cell types is poorly defined. Pulmonary hypertension (PH) is a lethal disease in which lung arteriole SMCs proliferate and migrate, coating the normally unmuscularized distal arteriole. We hypothesized that increased macrophage platelet-derived growth factor-B (PDGF-B) induces pathological SMC burden in PH. Our results indicate that clodronate attenuates hypoxia-induced macrophage accumulation, distal muscularization, PH, and right ventricle hypertrophy (RVH). With hypoxia exposure, macrophage Pdgfb mRNA was upregulated in mice, and LysM­Cre mice carrying floxed alleles for hypoxia-inducible factor 1a, hypoxia-inducible factor 2a, or Pdgfb had reduced macrophage Pdgfb and were protected against distal muscularization and PH. Conversely, LysM­Cre von-Hippel Lindaufl/fl mice had increased macrophage Hifa and Pdgfb and developed distal muscularization, PH, and RVH in normoxia. Similarly, Pdgfb was upregulated in macrophages from human idiopathic or systemic sclerosis-induced pulmonary arterial hypertension patients, and macrophage-conditioned medium from these patients increased SMC proliferation and migration via PDGF-B. Finally, in mice, orotracheal administration of nanoparticles loaded with Pdgfb siRNA specifically reduced lung macrophage Pdgfb and prevented hypoxia-induced distal muscularization, PH, and RVH. Thus, macrophage-derived PDGF-B is critical for pathological SMC expansion in PH, and nanoparticle-mediated inhibition of lung macrophage PDGF-B has profound implications as an interventional strategy for PH.


Asunto(s)
Hipertensión Pulmonar/patología , Macrófagos/metabolismo , Músculo Liso/fisiopatología , Proteínas Proto-Oncogénicas c-sis/fisiología , Animales , Humanos , Hipertensión Pulmonar/metabolismo , Ratones , Músculo Liso/patología
3.
Cancer Res ; 80(16): 3345-3358, 2020 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-32586981

RESUMEN

Platelet-derived growth factor B (PDGFB) plays a crucial role in recruitment of PDGF receptor ß-positive pericytes to blood vessels. The endothelium is an essential source of PDGFB in this process. Platelets constitute a major reservoir of PDGFB and are continuously activated in the tumor microenvironment, exposing tumors to the plethora of growth factors contained in platelet granules. Here, we show that tumor vascular function, as well as pericyte coverage is significantly impaired in mice with conditional knockout of PDGFB in platelets. A lack of PDGFB in platelets led to enhanced hypoxia and epithelial-to-mesenchymal transition in the primary tumors, elevated levels of circulating tumor cells, and increased spontaneous metastasis to the liver or lungs in two mouse models. These findings establish a previously unknown role for platelet-derived PDGFB, whereby it promotes and maintains vascular integrity in the tumor microenvironment by contributing to the recruitment of pericytes. SIGNIFICANCE: Conditional knockout of PDGFB in platelets demonstrates its previously unknown role in the maintenance of tumor vascular integrity and host protection against metastasis.


Asunto(s)
Movimiento Celular , Endotelio Vascular/metabolismo , Pericitos/fisiología , Proteínas Proto-Oncogénicas c-sis/fisiología , Animales , Vasos Sanguíneos , Neoplasias del Colon/irrigación sanguínea , Transición Epitelial-Mesenquimal , Matriz Extracelular , Técnicas de Inactivación de Genes , Hibridación Genética , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/secundario , Melanoma/irrigación sanguínea , Melanoma/secundario , Ratones , Células Neoplásicas Circulantes , Neoplasias Pancreáticas , Pericitos/metabolismo , Activación Plaquetaria/fisiología , Proteínas Proto-Oncogénicas c-sis/deficiencia , Proteínas Proto-Oncogénicas c-sis/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Trombocitopenia , Hipoxia Tumoral , Microambiente Tumoral
4.
Dev Dyn ; 248(7): 603-612, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31070827

RESUMEN

BACKGROUND: The platelet-derived growth factor (PDGF) family consists of four ligands (PDGF-A, PDGF-B, PDGF-C, PDGF-D) and two tyrosine kinase receptors (PDGFR-α and PDGFR-ß). In vertebrates, PDGF signaling influences cell proliferation, migration, and matrix deposition, and its up-regulation is implicated in cancer progression. Despite this evidence, the role of each family member during embryogenesis is still incomplete and partially controversial. In particular, study of the role of pdgf signaling during craniofacial development has been focused on pdgf-a, while the role of pdgf-b is almost unknown due to the lethal phenotypes of pdgf-b-null mice. RESULTS: By using a pdgf-b splice-blocking morpholino approach, we highlighted impairment of neural crest cell (NCC) migration in Xenopus laevis morphants, leading to alteration of NCC derivatives formation, such as cranial nerves and cartilages. We also uncovered a possible link between pdgf-b and the expression of cadherin superfamily members cdh6 and cdh11, which mediate cell-cell adhesion promoting NCC migration. CONCLUSIONS: Our results suggested that pdgf-b signaling is involved in cranial NCC migration and it is required for proper formation of craniofacial NCC derivatives. Taken together, these data unveiled a new role for pdgf-b during vertebrate development, contributing to complete the picture of pdgf signaling role in craniofacial development.


Asunto(s)
Huesos Faciales/crecimiento & desarrollo , Proteínas Proto-Oncogénicas c-sis/fisiología , Cráneo/crecimiento & desarrollo , Animales , Cadherinas/metabolismo , Adhesión Celular , Movimiento Celular , Embrión no Mamífero , Huesos Faciales/embriología , Ratones , Cresta Neural/citología , Transducción de Señal , Cráneo/embriología , Xenopus laevis/embriología , Xenopus laevis/crecimiento & desarrollo
5.
Brain ; 142(4): 885-902, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30805583

RESUMEN

Brain calcifications are commonly detected in aged individuals and accompany numerous brain diseases, but their functional importance is not understood. In cases of primary familial brain calcification, an autosomally inherited neuropsychiatric disorder, the presence of bilateral brain calcifications in the absence of secondary causes of brain calcification is a diagnostic criterion. To date, mutations in five genes including solute carrier 20 member 2 (SLC20A2), xenotropic and polytropic retrovirus receptor 1 (XPR1), myogenesis regulating glycosidase (MYORG), platelet-derived growth factor B (PDGFB) and platelet-derived growth factor receptor ß (PDGFRB), are considered causal. Previously, we have reported that mutations in PDGFB in humans are associated with primary familial brain calcification, and mice hypomorphic for PDGFB (Pdgfbret/ret) present with brain vessel calcifications in the deep regions of the brain that increase with age, mimicking the pathology observed in human mutation carriers. In this study, we characterize the cellular environment surrounding calcifications in Pdgfbret/ret animals and show that cells around vessel-associated calcifications express markers for osteoblasts, osteoclasts and osteocytes, and that bone matrix proteins are present in vessel-associated calcifications. Additionally, we also demonstrate the osteogenic environment around brain calcifications in genetically confirmed primary familial brain calcification cases. We show that calcifications cause oxidative stress in astrocytes and evoke expression of neurotoxic astrocyte markers. Similar to previously reported human primary familial brain calcification cases, we describe high interindividual variation in calcification load in Pdgfbret/ret animals, as assessed by ex vivo and in vivo quantification of calcifications. We also report that serum of Pdgfbret/ret animals does not differ in calcification propensity from control animals and that vessel calcification occurs only in the brains of Pdgfbret/ret animals. Notably, ossification of vessels and astrocytic neurotoxic response is associated with specific behavioural and cognitive alterations, some of which are associated with primary familial brain calcification in a subset of patients.


Asunto(s)
Astrocitos/metabolismo , Osificación Heterotópica/patología , Proteínas Proto-Oncogénicas c-sis/metabolismo , Anciano , Animales , Encéfalo/patología , Encefalopatías/genética , Calcinosis/patología , Femenino , Humanos , Masculino , Ratones , Mutación , Osteogénesis/fisiología , Estrés Oxidativo , Linaje , Proteínas Proto-Oncogénicas c-sis/genética , Proteínas Proto-Oncogénicas c-sis/fisiología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/genética , Receptor de Retrovirus Xenotrópico y Politrópico
6.
J Cell Biochem ; 119(11): 8897-8908, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30105826

RESUMEN

As a common cause of shoulder pain and disability, rotator cuff injury (RCI) represents a debilitating condition affecting an individual's quality of life. Although surgical repair has been shown to be somewhat effective, many patients may still suffer from reduced shoulder function. The aim of the current study was to identify a more effective mode of RCI treatment by investigating the effect of platelet-derived growth factor subunit B (PDGF-B) on tendon-bone healing after RCI repair by modifying bone marrow-derived mesenchymal stem cells (BMSCs). Surface markers of BMSCs were initially detected by means of flow cytometry, followed by establishment of the rat models and construction of the lentiviral vector. 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide, Thiazolyl Blue Tetrazolium Bromide (MTT) assay, alizarin red staining, and oil red O staining were used to provide verification that PDGF-B was indeed capable of promoting BMSC viability, osteogenic and adipogenic differentiation capability. Furthermore, biomechanical assessment results indicated that PDGF-B could increase the ultimate load and stiffness of the tendon tissue. Real-time reverse-transcription quantitative polymerase chain reaction and Western blot analysis methods provided evidence suggesting that PDGF-B facilitated the expression of tendon-bone healing-related genes and proteins, while contrasting results were obtained after PDGF-B silencing. Taken together, the key findings of the current study provided evidence suggesting that overexpressed PDGF-B could act to enhance tendon-bone healing after RCI repair, thus highlighting the potential of the functional promotion of PDGF-B as a future RCI therapeutic approach.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas/métodos , Proteínas Proto-Oncogénicas c-sis/genética , Proteínas Proto-Oncogénicas c-sis/fisiología , Lesiones del Manguito de los Rotadores/rehabilitación , Tendones/fisiología , Cicatrización de Heridas/fisiología , Adipogénesis/fisiología , Análisis de Varianza , Animales , Supervivencia Celular , Células Cultivadas , Modelos Animales de Enfermedad , Expresión Génica Ectópica/genética , Regulación de la Expresión Génica , Vectores Genéticos , Lentivirus/genética , Masculino , Osteogénesis/fisiología , Ratas , Ratas Sprague-Dawley , Transfección
7.
Cancer Lett ; 395: 20-30, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28286261

RESUMEN

Cancer-associated fibroblast (CAF)-dependent local invasion is the process by which cancer cells invade the extracellular matrix using tracks that have been physically remodeled by CAFs. In the present study, we investigated the process by which the epithelial-mesenchymal transition (EMT) of cancer cells affect CAF-dependent local invasion. Using an in vitro collagen invasion assay, we showed cancer cells undergoing EMT to promote the matrix-remodeling ability of CAFs and thereby enhance CAF-dependent local cancer cell invasion. Platelet-derived growth factor (PDGF)-BB secretion was significantly elevated in cancer cells undergoing EMT, and this induced an increase in the invasion ability of both CAFs and cancer cells. Conversely, knockdown of PDGF-B expression in cancer cells undergoing EMT, or treatment with a PDGF-receptor inhibitor, decreased the invasion ability of both CAFs and cancer cells. By analyzing the gene expression profiles of 442 patients with lung adenocarcinomas, we established that high expression of PDGF-B and presentation of mesenchymal-like tumors were significantly associated with a high rate of disease recurrence and poor patient prognosis. Thus, cancer cells undergoing EMT may accelerate their own ability to invade local tissues via PDGF-BB secretion to promote CAF matrix remodeling. Therefore, targeting PDGF signaling between cancer cells undergoing EMT and CAFs is a promising therapeutic target to inhibit cancer progression and improve patient prognosis.


Asunto(s)
Adenocarcinoma/patología , Fibroblastos Asociados al Cáncer/fisiología , Transición Epitelial-Mesenquimal , Neoplasias Pulmonares/patología , Proteínas Proto-Oncogénicas c-sis/fisiología , Adenocarcinoma/mortalidad , Adenocarcinoma del Pulmón , Becaplermina , Células Cultivadas , Humanos , Neoplasias Pulmonares/mortalidad , Invasividad Neoplásica , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Transducción de Señal
8.
Yonsei Med J ; 58(1): 234-240, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27873518

RESUMEN

PURPOSE: MicroRNAs are small non-coding RNAs that play important roles in vascular smooth muscle cell (VSMC) function. This study investigated the role of miR-379 on proliferation, invasion, and migration of VSMCs and explored underlying mechanisms thereof. MATERIALS AND METHODS: MicroRNA, mRNA, and protein levels were determined by quantitative real-time PCR and western blot. The proliferative, invasive, and migratory abilities of VSMCs were measured by CCK-8, invasion, and wound healing assay, respectively. Luciferase reporter assay was used to confirm the target of miR-379. RESULTS: Platelet-derived growth factor-bb was found to promote cell proliferation and suppress miR-379 expression in VSMCs. Functional assays demonstrated that miR-379 inhibited cell proliferation, cell invasion, and migration. Flow cytometry results further showed that miR-379 induced apoptosis in VSMCs. TargetScan analysis and luciferase report assay confirmed that insulin-like growth factor-1 (IGF-1) 3'UTR is a direct target of miR-379, and mRNA and protein levels of miR-379 and IGF-1 were inversely correlated. Rescue experiments showed that enforced expression of IGF-1 sufficiently overcomes the inhibitory effect of miR-379 on cell proliferation, invasion, and migration in VSMCs. CONCLUSION: Our results suggest that miR-379 plays an important role in regulating VSMCs proliferation, invasion, and migration by targeting IGF-1.


Asunto(s)
Movimiento Celular/fisiología , Proliferación Celular/fisiología , Factor I del Crecimiento Similar a la Insulina/fisiología , MicroARNs/fisiología , Músculo Liso Vascular/citología , Proteínas Proto-Oncogénicas c-sis/fisiología , Apoptosis , Becaplermina , Humanos , Insulina , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Sincalida/fisiología , Cicatrización de Heridas/fisiología
9.
Yonsei Medical Journal ; : 234-240, 2017.
Artículo en Inglés | WPRIM (Pacífico Occidental) | ID: wpr-126252

RESUMEN

PURPOSE: MicroRNAs are small non-coding RNAs that play important roles in vascular smooth muscle cell (VSMC) function. This study investigated the role of miR-379 on proliferation, invasion, and migration of VSMCs and explored underlying mechanisms thereof. MATERIALS AND METHODS: MicroRNA, mRNA, and protein levels were determined by quantitative real-time PCR and western blot. The proliferative, invasive, and migratory abilities of VSMCs were measured by CCK-8, invasion, and wound healing assay, respectively. Luciferase reporter assay was used to confirm the target of miR-379. RESULTS: Platelet-derived growth factor-bb was found to promote cell proliferation and suppress miR-379 expression in VSMCs. Functional assays demonstrated that miR-379 inhibited cell proliferation, cell invasion, and migration. Flow cytometry results further showed that miR-379 induced apoptosis in VSMCs. TargetScan analysis and luciferase report assay confirmed that insulin-like growth factor-1 (IGF-1) 3'UTR is a direct target of miR-379, and mRNA and protein levels of miR-379 and IGF-1 were inversely correlated. Rescue experiments showed that enforced expression of IGF-1 sufficiently overcomes the inhibitory effect of miR-379 on cell proliferation, invasion, and migration in VSMCs. CONCLUSION: Our results suggest that miR-379 plays an important role in regulating VSMCs proliferation, invasion, and migration by targeting IGF-1.


Asunto(s)
Humanos , Apoptosis , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Insulina , Factor I del Crecimiento Similar a la Insulina/fisiología , MicroARNs/fisiología , Músculo Liso Vascular/citología , Proteínas Proto-Oncogénicas c-sis/fisiología , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Sincalida/fisiología , Cicatrización de Heridas/fisiología
10.
PLoS One ; 11(9): e0162758, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27618690

RESUMEN

Systemic sclerosis (SSc) is a chronic autoimmune disorder that can result in extensive tissue damage in the skin and, in advanced cases, internal organs. Vasculopathy, aberrant immune activation, and tissue fibrosis are three hallmarks of the disease that have been identified, with vasculopathy and aberrant immunity being amongst the earliest events. However, a mechanistic link between these processes has not been established. Here, we have identified a novel role of platelet derived growth factor-BB (PDGF-BB)/PDGFRß activation in combination with dermal injury induced by bleomycin as a driver of early, aberrant expression of interferon stimulatory genes (ISGs) and inflammatory monocyte infiltration. Activation of PDGFRß in combination with bleomycin-induced dermal injury resulted in increased dermal thickness, vascular density, monocyte/macrophage infiltration, and exacerbation of tissue injury. Many of these features were dependent on IFNAR-signaling, and an increase in the number of interferon-beta (IFN-ß) producing monocytes cells was found in the skin lesions. Taken together, these results identify a novel link between PDGFRß activation, and Type I IFN-driven vascular maintenance and monocyte/macrophage cell recruitment, and provide a potential explanation linking key features of SSc that were previously thought to be unrelated.


Asunto(s)
Vasos Sanguíneos/patología , Modelos Animales de Enfermedad , Interferón Tipo I/fisiología , Monocitos/patología , Proteínas Proto-Oncogénicas c-sis/fisiología , Esclerodermia Sistémica/patología , Enfermedades de la Piel/patología , Animales , Becaplermina , Bleomicina/farmacología , Fibrosis , Ratones , Ratones Endogámicos C57BL , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo
11.
Anticancer Res ; 36(5): 2291-5, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27127135

RESUMEN

BACKGROUND/AIM: Studies developed in the field of platelet-derived growth factors/platelet-derived growth factor receptors (PDGFs/PDGFRs) inhibition have focused on the therapeutic effects on tumor cells, neglecting their potential effects on tumor blood vessels. We herein propose a differential and critic assessment of platelet-derived growth factor B (PDGF-B) and platelet-derived growth factor receptor ß (PDGFRß) in renal cell carcinoma, correlated with the four main vascular patterns previously reported by our team. MATERIALS AND METHODS: PDGF-B and PDGFRß were evaluated on 50 archival paraffin embedded specimens related to vascular endothelial growth factor (VEGF), its inhibitory isoform VEGF165b and vascular patterns. RESULTS AND CONCLUSION: Our results support the involvement of VEGF165b in the phosphorylation of PDGFRß with an inhibitory effect on endothelial proliferation and migration. The simultaneous action of PDGF-B/PDGFRß and VEGF165b on the same type of receptor may explain the resistance to antiangiogenic therapy, which depends on the degree of modulation of PDGFRß phosphorylation.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antineoplásicos/farmacología , Carcinoma de Células Renales/tratamiento farmacológico , Resistencia a Antineoplásicos/fisiología , Neoplasias Renales/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-sis/fisiología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/fisiología , Factor A de Crecimiento Endotelial Vascular/fisiología , Becaplermina , Carcinoma de Células Renales/irrigación sanguínea , Carcinoma de Células Renales/química , Humanos , Neoplasias Renales/irrigación sanguínea , Neoplasias Renales/química , Neovascularización Patológica/metabolismo , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-sis/análisis , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/análisis , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Estudios Retrospectivos , Factor A de Crecimiento Endotelial Vascular/análisis
12.
Implant Dent ; 25(4): 492-8, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26963746

RESUMEN

PURPOSE: Validation of platelet-rich plasma (PRP) system and assessing its enhancing effect on the healing of iliac crest grafts before implant placement. MATERIALS AND METHODS: Patients randomly allocated to test (n = 13) and control (n = 9) groups. Iliac crest grafts were mixed with PRP in the test group. Tetracycline labeling preceded implant placement. Bone samples were harvested for histomorphometrical analysis. Platelet and growth factor quantifications were performed. ANALYSIS: Data were analyzed using SPSS software package. Independent t test was used and statistical significance was set at 5%. RESULTS: The PRP group showed significantly higher platelet counts, PDGF-BB, and TGF-ß1 concentrations. Tendency to higher volume of woven bone was observed in the PRP group (13 ± 11 vs 4 ± 6, P = 0.1). Histomorphometry showed increased seam separation in the PRP group (8.8 ± 9 µm vs 1.5 ± 3 µm, P = 0.039). Remodeling activity was higher in PRP-woven bone sections and comparable in trabecular sections. CONCLUSION: PRP significantly increased platelet and growth factor concentrations and was of possible enhancing effect on the rate of bone formation at 3 to 4 months of grafting. The clinical significance of this enhancement is yet to be established.


Asunto(s)
Desarrollo Óseo , Trasplante Óseo/métodos , Ilion/cirugía , Péptidos y Proteínas de Señalización Intercelular/análisis , Maxilar/cirugía , Plasma Rico en Plaquetas , Adolescente , Adulto , Anciano , Becaplermina , Desarrollo Óseo/fisiología , Femenino , Humanos , Ilion/patología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Masculino , Maxilar/patología , Persona de Mediana Edad , Recuento de Plaquetas , Plasma Rico en Plaquetas/química , Plasma Rico en Plaquetas/fisiología , Proteínas Proto-Oncogénicas c-sis/análisis , Proteínas Proto-Oncogénicas c-sis/fisiología , Elevación del Piso del Seno Maxilar/métodos , Factor de Crecimiento Transformador beta1/análisis , Factor de Crecimiento Transformador beta1/fisiología , Adulto Joven
13.
PLoS One ; 11(3): e0150500, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26930482

RESUMEN

BACKGROUND: Neointima forming after stent implantation consists of vascular smooth muscle cells (VSMCs) in 90%. Growth factors TGF-ß1, PDGFB, EGF, bFGF and VEGF-A play an important role in VSMC proliferation and migration to the tunica intima after arterial wall injury. The aim of this paper was an analysis of functional polymorphisms in genes encoding TGF-ß1, PDGFB, EGF, bFGF and VEGF-A in relation to in-stent restenosis (ISR). MATERIALS AND METHODS: 265 patients with a stable coronary artery disease (SCAD) hospitalized in our center in the years 2007-2011 were included in the study. All patients underwent stent implantation at admission to the hospital and had another coronary angiography performed due to recurrence of the ailments or a positive result of the test assessing the coronary flow reserve. Angiographically significant ISR was defined as stenosis >50% in the stented coronary artery segment. The patients were divided into two groups-with angiographically significant ISR (n = 53) and without significant ISR (n = 212). Additionally, the assessment of late lumen loss (LLL) in vessel was performed. EGF rs4444903 polymorphism was genotyped using the PCR-RFLP method whilst rs1800470 (TGFB1), rs2285094 (PDGFB) rs308395 (bFGF) and rs699947 (VEGF-A) were determined using the TaqMan method. RESULTS: Angiographically significant ISR was significantly less frequently observed in the group of patients with the A/A genotype of rs1800470 polymorphism (TGFB1) versus patients with A/G and G/G genotypes. In the multivariable analysis, LLL was significantly lower in patients with the A/A genotype of rs1800470 (TGFB1) versus those with the A/G and G/G genotypes and higher in patients with the A/A genotype of the VEGF-A polymorphism versus the A/C and C/C genotypes. The C/C genotype of rs2285094 (PDGFB) was associated with greater LLL compared to C/T heterozygotes and T/T homozygotes. CONCLUSIONS: The polymorphisms rs1800470, rs2285094 and rs6999447 of the TGFB1, PDGFB and VEGF-A genes, respectively, are associated with LLL in patients with SCAD treated by PCI with a metal stent implantation.


Asunto(s)
Enfermedad de la Arteria Coronaria/genética , Reestenosis Coronaria/genética , Factor de Crecimiento Epidérmico/genética , Factor 2 de Crecimiento de Fibroblastos/genética , Polimorfismo de Nucleótido Simple/genética , Proteínas Proto-Oncogénicas c-sis/genética , Stents , Factor de Crecimiento Transformador beta/genética , Factor A de Crecimiento Endotelial Vascular/genética , Anciano , Enfermedad de la Arteria Coronaria/cirugía , Factor de Crecimiento Epidérmico/fisiología , Femenino , Factor 2 de Crecimiento de Fibroblastos/fisiología , Predisposición Genética a la Enfermedad/genética , Humanos , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple/fisiología , Proteínas Proto-Oncogénicas c-sis/fisiología , Stents/efectos adversos , Factor de Crecimiento Transformador beta/fisiología , Factor A de Crecimiento Endotelial Vascular/fisiología
14.
Am J Pathol ; 186(5): 1081-91, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26945107

RESUMEN

Glioma is an aggressive and incurable disease, and is frequently accompanied by augmented platelet-derived growth factor (PDGF) signaling. Overexpression of PDGF-B ligand characterizes a specific subclass of glioblastoma multiforme, but the significance of the ligand remains to be elucidated. For this end, we implanted a glioma-cell line transfected with PDGF-BB-overexpressing vector (GL261-PDGF-BB) or control vector (GL261-vector) into wild-type mouse brain, and examined the effect of glioma-derived PDGF on the tumor microenvironment. The volume of GL261-PDGF-BB rapidly increased compared with GL261-vector. Recruitment of many PDGF receptor (PDGFR)-α and Olig2-positive oligodendrocyte precursor cells and frequent hemorrhages were observed in GL261-PDGF-BB but not in GL261-vector. We then implanted GL261-PDGF-BB into the mouse brain with and without Pdgfra gene inactivation, corresponding to PDGFRα-knockout (KO) and Flox mice, respectively. The recruitment of oligodendrocyte precursor cells was largely suppressed in PDGFRα-KO than in Flox, whereas the volume of GL261-PDGF-BB was comparable between the two genotypes. Frequent hemorrhage and increased IgG-leakage were associated with aberrant vascular structures within the area where many recruited oligodendrocyte precursor cells accumulated in Flox. In contrast, these vascular phenotypes were largely normalized in PDGFRα-KO. Increased matrix metalloproteinase-9 in recruited oligodendrocyte precursor cells and decreased claudin-5 in vasculature may underlie the vascular abnormality. Glioma-derived PDGF-B signal induces cancer stroma characteristically seen in high-grade glioma, and should be therapeutically targeted to improve cancer microenvironment.


Asunto(s)
Neoplasias Encefálicas/patología , Glioblastoma/patología , Oligodendroglía/fisiología , Proteínas Proto-Oncogénicas c-sis/fisiología , Células Madre/fisiología , Animales , Antineoplásicos Hormonales/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Permeabilidad Capilar/fisiología , Línea Celular Tumoral , Hemorragia Cerebral/etiología , Colágeno/fisiología , Femenino , Técnicas de Inactivación de Genes , Vectores Genéticos , Glioblastoma/tratamiento farmacológico , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Trasplante de Neoplasias , Fenotipo , Tamoxifeno/farmacología , Transfección , Carga Tumoral
15.
Exp Cell Res ; 342(1): 20-31, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26945917

RESUMEN

The regulation of vascular smooth muscle cell (VSMC) proliferation is an important issue due to its major implications for the prevention of pathological vascular conditions. The objective of this work was to assess the function of small ubiquitin-like modifier (SUMO)ylated Krϋppel-like transcription factor 4 (KLF4) in the regulation of VSMC proliferation in cultured cells and in animal models with balloon injury. We found that under basal conditions, binding of non-SUMOylated KLF4 to p300 activated p21 (p21(WAF1/CIP1))transcription, leading to VSMC growth arrest. PDGF-BB promoted the interaction between Ubc9 and KLF4 and the SUMOylation of KLF4, which in turn recruited transcriptional corepressors to the p21 promoter. The reduction in p21 enhanced VSMC proliferation. Additionally, the SUMOylated KLF4 did not affect the expression of KLF4, thereby forming a positive feedback loop enhancing cell proliferation. These results demonstrated that SUMOylated KLF4 plays an important role in cell proliferation by reversing the transactivation action of KLF4 on p21 induced with PDGF-BB.


Asunto(s)
Proliferación Celular , Regulación de la Expresión Génica , Factores de Transcripción de Tipo Kruppel/metabolismo , Miocitos del Músculo Liso/fisiología , Sumoilación , Animales , Becaplermina , Células Cultivadas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Arteria Femoral/lesiones , Arteria Femoral/patología , Humanos , Factor 4 Similar a Kruppel , Masculino , Ratones Endogámicos C57BL , Músculo Liso Vascular/patología , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-sis/fisiología , Ratas Sprague-Dawley , Transcripción Genética , Enzimas Ubiquitina-Conjugadoras/genética , Enzimas Ubiquitina-Conjugadoras/metabolismo , Enfermedades Vasculares/metabolismo
16.
Exp Eye Res ; 145: 224-229, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26778750

RESUMEN

Arf encodes an important tumor suppressor, p19(Arf), which also plays a critical role to control hyperplasia in the primary vitreous during mouse eye development. In the absence of Arf, mice are born blind and display a phenotype closely mimicking severe forms of the human eye disease, persistent hyperplastic primary vitreous (PHPV). In this report, we characterize p19(Arf) expression in perivascular cells that normally populate the primary vitreous and express the Arf promoter. Using a new ex vivo model, we show that these cells respond to exogenous Tgfß, despite being isolated at a time when Tgfß has already turned on the Arf promoter. Treatment of the cells with PDGF-B ligand doubles the population of cells in S-phase and ectopic expression of Arf blunts that effect. We show this effect is mediated through Pdgfrß as expression of Arf represses expression of Pdgfrß mRNA and protein to approximately 60%. p53 is not required for Arf-dependent blockade of PDGF-B driven proliferation and repression of Pdgfrß protein as ectopic expression of Arf is still able to inhibit the 2-fold increase in the S-phase fraction of cells upon treatment with PDGF-B. Finally, induction of mature miR-34a, a microRNA previously identified to be regulated by p19(Arf) does not depend on p53 while the expression of the primary transcript does require p53. These data corroborate that, as in vivo, p19(Arf) functions to inhibit PDGF-B driven proliferation ex vivo.


Asunto(s)
Proliferación Celular/fisiología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Proteínas Proto-Oncogénicas c-sis/fisiología , Enfermedades de la Retina/fisiopatología , Cuerpo Vítreo/citología , Animales , Western Blotting , Ciclo Celular/fisiología , Células Cultivadas , Ratones , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteína p53 Supresora de Tumor , Cuerpo Vítreo/efectos de los fármacos
17.
Cardiology ; 133(4): 242-56, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26752520

RESUMEN

OBJECTIVE: To explore the role of platelet-derived growth factor-B (PDGF-B)/platelet-derived growth factor receptor-ß (PDGFR-ß) signaling in chronic atrial fibrillation (AF). METHODS: Thirty-nine AF patients and 33 patients with sinus rhythm (SR) were enrolled. Twenty canines were randomized into 5 groups: control, sham and AF lasting 1, 2 or 4 weeks. The AF canine models were made by rapid atrial pacing. Rat atrial fibroblasts were treated with PDGF-BB or PDGF-BB + PDGFR inhibitor AG1295, respectively. Gene expression in the right atrial appendage of patients, the left atrium of canines and rat atrial fibroblasts was measured by quantitative real-time PCR and Western blot, respectively. The degree of atrial fibrosis was evaluated by Masson trichrome staining. RESULTS: The degree of atrial fibrosis and the expression of PDGF-B, PDGFR-ß and collagen type I (COL1) in AF patients significantly increased compared to patients with SR. The degree of atrial fibrosis and the expression of PDGF-B and COL1 in canines increased progressively with the increased duration of AF. The expression of PDGFR-ß increased progressively 2 weeks after AF. PDGF-BB promoted the proliferation and COL1 secretion of rat atrial fibroblasts. AG1295 attenuated these effects. CONCLUSIONS: Our study suggests that PDGF-B/PDGFR-ß signaling, which promotes the proliferation and COL1 secretion of atrial fibroblasts, is an important contributor to atrial fibrosis in AF and may represent a novel target for the intervention of AF.


Asunto(s)
Fibrilación Atrial/fisiopatología , Proteínas Proto-Oncogénicas c-sis/fisiología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/fisiología , Transducción de Señal , Adulto , Animales , Proliferación Celular , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Modelos Animales de Enfermedad , Perros , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibrosis , Atrios Cardíacos/patología , Humanos , Masculino , Persona de Mediana Edad , Proteínas Proto-Oncogénicas c-sis/genética , ARN Mensajero/metabolismo , Ratas , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Regulación hacia Arriba
18.
Dev Ophthalmol ; 55: 28-37, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26502333

RESUMEN

Systematic study of the mechanisms underlying pathological ocular neovascularization has yielded a wealth of knowledge about pro- and anti-angiogenic factors that modulate diseases such as neovascular age-related macular degeneration. The evidence implicating vascular endothelial growth factor (VEGF) in particular has led to the development of a number of approved anti-VEGF therapies. Additional proangiogenic targets that have emerged as potential mediators of ocular neovascularization include hypoxia-inducible factor-1, angiopoietin-2, platelet-derived growth factor-B and components of the alternative complement pathway. As for VEGF, knowledge of these factors has led to a product pipeline of many more novel agents that are in various stages of clinical development in the setting of ocular neovascularization. These agents are represented by a range of drug classes and, in addition to novel small- and large-molecule VEGF inhibitors, include gene therapies, small interfering RNA agents and tyrosine kinase inhibitors. In addition, combination therapy is beginning to emerge as a strategy to improve the efficacy of individual therapies. Thus, a variety of agents, whether administered alone or as adjunctive therapy with agents targeting VEGF, offer the promise of expanding the range of treatments for ocular neovascular diseases.


Asunto(s)
Angiopoyetina 2/fisiología , Vía Alternativa del Complemento/fisiología , Ojo/irrigación sanguínea , Factor 1 Inducible por Hipoxia/fisiología , Neovascularización Patológica/fisiopatología , Proteínas Proto-Oncogénicas c-sis/fisiología , Factor A de Crecimiento Endotelial Vascular/fisiología , Humanos
19.
Int J Biochem Cell Biol ; 69: 225-32, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26546462

RESUMEN

Vascular smooth muscle cell (VSMC) migration is implicated in atherosclerosis and restenosis. Nuclear receptor subfamily 6, group A, member 1 (NR6A1) is involved in regulating embryonic stem cell differentiation, reproduction, neuronal differentiation. Functional cooperation between cAMP response element modulator tau (CREMtau) and NR6A1 can direct gene expression in cells. cAMP response element binding protein (CREB) plays a key role in VSMC migration. In this study, we sought to determine whether CREB involved in NR6A1-modulated VSMC migration. VSMCs treated with platelet-derived growth factor-BB (PDGF-BB) displayed reduced mRNA and protein levels of NR6A1. Adenovirus-mediated expression of NR6A1 (Ad-NR6A1) could inhibit PDGF-BB- and serum-induced VSMC migration. The mRNA and protein expressions of secreted phosphoprotein 1 (SPP1) were down-regulated by NR6A1 overexpression. SPP1 promoter reporter activity was repressed by NR6A1. NR6A1 was found to physically couple with nuclear actin and the large subunit of RNA polymerase II. Furthermore, we showed that CREB interacted with NR6A1 in VSMCs. NR6A1 overexpression repressed cAMP response element (CRE) activity. ChIP assay revealed that NR6A1 bind to SPP1 promoter. Luciferase reporter assay showed that NR6A1 regulated SPP1 promoter activity via a putative CRE site. Adenovirus mediated local NR6A1 gene transfer attenuated stenosis after balloon-induced arterial injury in Sprague-Dawley rats. Taken together, this study provided experimental evidence that NR6A1 modulated SPP1 expression via its binding with CREB protein in VSMCs. We also revealed a NR6A1-CREB-SPP1 axis that serves as a regulatory mechanism for atherosclerosis and restenosis.


Asunto(s)
Movimiento Celular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/fisiología , Miocitos del Músculo Liso/fisiología , Miembro 1 del Grupo A de la Subfamilia 6 de Receptores Nucleares/fisiología , Animales , Becaplermina , Sitios de Unión , Arterias Carótidas/metabolismo , Arterias Carótidas/patología , Expresión Génica , Silenciador del Gen , Células HEK293 , Humanos , Masculino , Músculo Liso Vascular/patología , Neointima/metabolismo , Osteopontina/genética , Osteopontina/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Proto-Oncogénicas c-sis/fisiología , Ratas Sprague-Dawley
20.
Biol Reprod ; 93(4): 103, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26353894

RESUMEN

We studied the smooth muscle cell differentiation capability of human placental multipotent mesenchymal stromal cells (hPMSCs) and identified how endothelial cells recruit hPMSCs participating in vessel formation. hPMSCs from term placentas were induced to differentiate into smooth muscle cells under induction conditions and different matrix substrates. We assessed endothelial cells from umbilical veins for platelet-derived growth factor (PDGF)-BB expression and to induce hPMSC PDGFR-beta and STAT3 activation. Endothelial cells were co-cultured with hPMSCs for in vitro angiogenesis. Cell differentiation ability was then further assessed by mouse placenta transplantation assay. hPMSCs can differentiate into smooth muscle cells; collagen type I and IV or laminin support this differentiation. Endothelial cells expressed significant levels of PDGF-BB and activated STAT3 transcriptional activity in hPMSCs. Endothelial cell-conditioned medium induced hPMSC migration, which was inhibited by STAT3 small interfering RNA transfection or by pretreatement with PDGFR-beta-blocking antibody but not by PDGFR-alpha-blocking antibody or isotype immunoglobulin G (IgG; P < 0.001). hPMSCs can incorporate into endothelial cells with tube formation and promote endothelial cells, forming capillary-like networks than endothelial cells alone (tube lengths: 12 024.1 ± 960.1 vs. 9404.2 ± 584.7 pixels; P < 0.001). Capillary-like networks were significantly reduced by hPMSCs pretreated with PDGFR-beta-blocking antibody but not by PDGFR-alpha-blocking antibody or isotype IgG (P < 0.001). Transplantation of hPMSCs into mouse placentas revealed incorporation of the hPMSCs into vessel walls, which expressed alpha-smooth muscle actin, calponin, and smooth muscle myosin (heavy chain) in vivo. In conclusion, endothelial cell-hPMSC interactions occur during vessel development of placenta. Placental endothelial cell-derived PDGF-BB recruits hPMSCs involved in vascular development via PDGFR-beta/STAT3 activation.


Asunto(s)
Células Madre Mesenquimatosas/fisiología , Neovascularización Fisiológica/fisiología , Placenta/citología , Células Madre Pluripotentes/fisiología , Proteínas Proto-Oncogénicas c-sis/fisiología , Factor de Transcripción STAT3/fisiología , Animales , Becaplermina , Diferenciación Celular , Células Endoteliales/fisiología , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Músculo Liso Vascular/citología , Placenta/irrigación sanguínea , Placenta/trasplante , Embarazo , Proteínas Proto-Oncogénicas c-sis/genética , Factor de Transcripción STAT3/genética , Transducción de Señal/genética , Transducción de Señal/fisiología , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...