Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
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.
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
3.
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
4.
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
5.
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
6.
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
7.
J Biol Chem ; 290(13): 8232-42, 2015 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-25623072

RESUMEN

Platelet-derived growth factor (PDGF), a potent chemoattractant, induces cell migration via the MAPK and PI3K/Akt pathways. However, the downstream mediators are still elusive. In particular, the role of extracellular mediators is largely unknown. In this study, we identified the matricellular protein Cyr61, which is de novo synthesized in response to PDGF stimulation, as the key downstream mediator of the ERK and JNK pathways, independent of the p38 MAPK and AKT pathways, and, thereby, it mediates PDGF-induced smooth muscle cell migration but not proliferation. Our results revealed that, when Cyr61 was newly synthesized by PDGF, it was promptly translocated to the extracellular matrix and physically interacted with the plasma membrane integrins α6ß1 and αvß3. We further demonstrate that Cyr61 and integrins are integral components of the PDGF signaling pathway via an "outside-in" signaling route to activate intracellular focal adhesion kinase (FAK), leading to cell migration. Therefore, this study provides the first evidence that the PDGF-induced endogenous extracellular matrix component Cyr61 is a key mediator in modulating cell migration by connecting intracellular PDGF-ERK and JNK signals with integrin/FAK signaling. Therefore, extracellular Cyr61 convergence with growth factor signaling and integrin/FAK signaling is a new concept of growth factor-induced cell migration. The discovered signaling pathway may represent an important therapeutic target in growth factor-mediated cell migration/invasion-related vascular diseases and tumorigenesis.


Asunto(s)
Movimiento Celular , Proteína 61 Rica en Cisteína/fisiología , Proteínas Proto-Oncogénicas c-sis/fisiología , Animales , Becaplermina , Proliferación Celular , Células Cultivadas , Activación Enzimática , Matriz Extracelular/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Integrinas/metabolismo , Sistema de Señalización de MAP Quinasas , Ratones , Miocitos del Músculo Liso/fisiología , Activación Transcripcional
8.
J Cell Sci ; 127(Pt 12): 2647-58, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24762811

RESUMEN

DOK1 regulates platelet-derived growth factor (PDGF)-BB-stimulated glioma cell motility. Mechanisms regulating tumour cell motility are essential for invasion and metastasis. We report here that PDGF-BB-mediated glioma cell invasion and migration are dependent on the adaptor protein downstream of kinase 1 (DOK1). DOK1 is expressed in several glioma cell lines and in tumour biopsies from high-grade gliomas. DOK1 becomes tyrosine phosphorylated upon PDGF-BB stimulation of human glioma cells. Knockdown of DOK1 or expression of a DOK1 mutant (DOK1FF) containing Phe in place of Tyr at residues 362 and 398, resulted in inhibition of both the PDGF-BB-induced tyrosine phosphorylation of p130Cas (also known as BCAR1) and the activation of Rap1. DOK1 colocalises with tyrosine phosphorylated p130Cas at the cell membrane of PDGF-BB-treated cells. Expression of a non-tyrosine-phosphorylatable substrate domain mutant of p130Cas (p130Cas15F) inhibited PDGF-BB-mediated Rap1 activation. Knockdown of DOK1 and Rap1 inhibited PDGF-BB-induced chemotactic cell migration, and knockdown of DOK1 and Rap1 and expression of DOK1FF inhibited PDGF-mediated three-dimensional (3D) spheroid invasion. These data show a crucial role for DOK1 in the regulation of PDGF-BB-mediated tumour cell motility through a p130Cas-Rap1 signalling pathway. [Corrected]


Asunto(s)
Neoplasias Encefálicas/metabolismo , Proteína Sustrato Asociada a CrK/metabolismo , Proteínas de Unión al ADN/fisiología , Glioblastoma/metabolismo , Fosfoproteínas/fisiología , Proteínas Proto-Oncogénicas c-sis/fisiología , Proteínas de Unión al ARN/fisiología , Proteínas de Unión a Telómeros/metabolismo , Becaplermina , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Quimiotaxis , Glioblastoma/patología , Humanos , Invasividad Neoplásica , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional , Complejo Shelterina , Transducción de Señal , Familia-src Quinasas/metabolismo
9.
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
10.
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
11.
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
12.
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
13.
Cancer Sci ; 105(9): 1116-23, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24981766

RESUMEN

The lymphatic system plays important roles not only in the physiological processes, such as maintenance of tissue fluid homeostasis, but also in pathological processes including the lymph node metastasis of tumor cells. Therefore, understanding of the molecular mechanisms underlying lymphatic vessel formation is crucial. Previous studies have shown that proliferation and migration of lymphatic endothelial cells (LECs) are activated by multiple types of signals mediated by tyrosine kinase receptors such as vascular endothelial growth factor receptor (VEGFR) 3. Although signals mediated by platelet-derived growth factor receptor ß (PDGFRß) have been implicated in lymphangiogenesis, the mechanisms explaining how PDGFRß expression is maintained in LECs remain to be fully elucidated. In the present study, we show that PDGFRß expression in LECs is maintained by Prox1 transcription factor. Knockdown of Prox1 expression in human dermal LECs decreased the expression of PDGFRß, leading to the lowered migration of human dermal LECs towards PDGF-BB. Furthermore, we found that PDGF signals play important roles in inflammatory lymphangiogenesis in a chronic aseptic peritonitis model. Intraperitoneal administration of imatinib, a potent inhibitor of PDGFRß, and transduction of PDGFRß/Fc chimeric protein by an adenoviral system both reduced inflammatory lymphangiogenesis induced by thioglycollate in mice. We also found that the expression of PDGFRß/Fc reduced tumor lymphangiogenesis in a BxPC3 human pancreatic cancer xenograft model. These findings suggest that PDGFRß is one of the key mediators of lymphatic vessel formation acting downstream of Prox1.


Asunto(s)
Proteínas de Homeodominio/fisiología , Vasos Linfáticos/fisiopatología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Supresoras de Tumor/fisiología , Animales , Antineoplásicos/farmacología , Becaplermina , Benzamidas/farmacología , Línea Celular Tumoral , Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Mesilato de Imatinib , Linfangiogénesis , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-sis/fisiología , Pirimidinas/farmacología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Transducción de Señal
14.
Am J Physiol Heart Circ Physiol ; 306(12): H1714-9, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24727494

RESUMEN

Omentin is a novel adipocytokine expressed in visceral adipose tissue. Secretion and blood concentration of omentin decrease in the obese subjects. We previously demonstrated that omentin is anti-inflammatory in vascular smooth muscle cells (SMCs). While vascular remodeling via migration of SMCs is also important for hypertension development, it remains to be clarified whether omentin affects this process. Here we examined whether omentin controls SMC migration. Omentin (300 ng/ml, 2 h) significantly inhibited platelet-derived growth factor (PDGF)-BB (10 ng/ml, 6 h)-induced migration of rat mesenteric arterial SMCs, as determined by Boyden chamber assay. Omentin (300 ng/ml, 2 h) significantly inhibited PDGF-BB (10 ng/ml, 30 min)-induced phosphorylation of p38 and heat shock protein (HSP) 27. Omentin (300 ng/ml, 2 h) significantly inhibited PDGF-BB (10 ng/ml, 30 min)-induced NADPH oxidase (NOX) activation as determined by lucigenin assay. Omentin (300 ng/ml, 24 h) significantly inhibited fetal bovine serum (5%, 4 days)-induced SMC outgrowth from rat isolated mesenteric artery. In vivo, omentin significantly inhibited carotid intimal hyperplasia in mouse ligation model. In summary, we for the first time demonstrate that omentin prevents PDGF-BB-induced SMC migration by preventing NOX/O2(-)/p38/HSP27 pathways, which might be at least partly responsible for the preventive effects on neointimal hyperplasia. Our data suggest that omentin may be protective against hypertension development by inhibiting vascular structural remodeling.


Asunto(s)
Adipoquinas/farmacología , Antioxidantes/fisiología , Movimiento Celular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Proteínas Proto-Oncogénicas c-sis/farmacología , Transducción de Señal/fisiología , Adipoquinas/fisiología , Animales , Becaplermina , Arterias Carótidas/citología , Arterias Carótidas/efectos de los fármacos , Movimiento Celular/fisiología , Células Cultivadas , Citocinas/farmacología , Proteínas Ligadas a GPI , Proteínas de Choque Térmico HSP27/fisiología , Técnicas In Vitro , Lectinas/farmacología , Masculino , Arterias Mesentéricas/citología , Arterias Mesentéricas/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Modelos Animales , Miocitos del Músculo Liso/citología , NADPH Oxidasas/fisiología , Oxígeno/fisiología , Proteínas Proto-Oncogénicas c-sis/fisiología , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
15.
Am J Pathol ; 182(6): 2132-46, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23570836

RESUMEN

Fibrosis is a deleterious consequence of chronic inflammation in a number of human pathologies ultimately leading to organ dysfunction and failure. Two growth factors that are important in blood vessel physiology and tissue fibrosis, platelet-derived growth factor (PDGF)-B and transforming growth factor (TGF)-ß1, were investigated. Adenoviral vectors were used to induce transient overexpression of these growth factors in mouse skin. Changes in tissue structure and protein and mRNA expressions were investigated. Both PDGF-B and TGF-ß1 could initiate but neither could sustain angiogenesis. Instead, vascular regression was observed. Overexpression of both TGF-ß1 and PDGF-B led to a marked macrophage influx and an expansion of the connective tissue cell population. Over time, this effect was sustained in mice treated with TGF-ß1, whereas it was partially reversible in mice treated with PDGF-B. On the basis of structure and expression of phenotypical markers, the emerging connective tissue cell population may originate from microvascular pericytes. TGF-ß1 induced expansion of connective tissue cells with a myofibroblast phenotype, whereas PDGF-B induced a fibroblast phenotype negative for α-smooth muscle actin. TGF-ß1 and PDGF-B overexpressions mediated distinct effects on mRNA transcript levels of fibrillar procollagens, their modifying enzymes, small leucin-rich repeat proteoglycans, and matricellular proteins affecting both the composition and the quantity of the extracellular matrix. This study offers new insight into the effects of PDGF-B and TGF-ß1 on the vasculature and connective tissue in vivo.


Asunto(s)
Células del Tejido Conectivo/citología , Pericitos/citología , Proteínas Proto-Oncogénicas c-sis/fisiología , Piel/citología , Factor de Crecimiento Transformador beta1/fisiología , Adenoviridae/genética , Animales , Endotelio Vascular/citología , Matriz Extracelular/fisiología , Proteínas de la Matriz Extracelular/metabolismo , Fibromodulina , Regulación de la Expresión Génica/fisiología , Vectores Genéticos , Macrófagos/fisiología , Ratones , Ratones Desnudos , Microvasos/citología , Neovascularización Fisiológica/fisiología , Pericitos/metabolismo , Fenotipo , Proteoglicanos/metabolismo , ARN Mensajero/genética , Piel/irrigación sanguínea , Piel/metabolismo
16.
Exp Eye Res ; 121: 35-40, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24582892

RESUMEN

Myofibroblasts, the primary cells associated with corneal stromal haze (opacity), can be derived from both cornea-derived and bone marrow-derived precursor cells. In the present study, the role of TGFß or PDGF blockage on bone marrow-derived myofibroblast development was investigated using a green fluorescent protein (GFP) chimeric bone marrow mouse model and plasmid vectors that blocked TGFß or PDGF signaling. At the peak of corneal haze one month after irregular phototherapeutic keratectomy the central stroma had significantly less alpha-smooth muscle actin (α-SMA)-positive cells derived from GFP+ bone marrow-derived cells or GFP- keratocyte/corneal fibroblast-derived cells when corneas were treated with the TGFß blocking vector pGFPC1.TGFRBKDEL or the PDGF blocking vector pCMV.PDGFRB.23KDEL compared with the corresponding empty vector treated or untreated control groups. In individual animals, 30-60% of myofibroblasts were derived from bone marrow-derived precursor cells and 40-70% of myofibroblasts were derived from keratocyte-derived precursor cells. TGFß and PDGF regulate corneal myofibroblast development from bone marrow-derived precursor cells and keratocyte/corneal fibroblast-derived precursor cells.


Asunto(s)
Células de la Médula Ósea/citología , Queratocitos de la Córnea/citología , Miofibroblastos/citología , Proteínas Proto-Oncogénicas c-sis/fisiología , Transducción de Señal/efectos de los fármacos , Células Madre/citología , Factor de Crecimiento Transformador beta/fisiología , Actinas/metabolismo , Animales , Células de la Médula Ósea/metabolismo , Queratocitos de la Córnea/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Vectores Genéticos/farmacología , Proteínas Fluorescentes Verdes/metabolismo , Ratones , Ratones Endogámicos C57BL , Miofibroblastos/metabolismo , Queratectomía Fotorrefractiva , Plásmidos , Proteínas Proto-Oncogénicas c-sis/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/antagonistas & inhibidores
17.
Mol Cell Proteomics ; 11(5): 15-30, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22027198

RESUMEN

Stably transfected PC12 cells expressing a chimeric receptor composed of the extracellular domain of the platelet-derived growth factor receptor BB and the transmembrane and intracellular domains of TrkA, the nerve growth factor receptor, were stimulated for 20 min with platelet-derived growth factor and the resulting phosphoproteome was determined from affinity purified tryptic peptides identified by tandem MS (MS/MS) analyses. The changes in the levels of individual phosphorylation sites in stimulated cells versus control were ascertained by the stable isotope labeling of amino acids in cell culture technique. A total of 2035 peptides (806 proteins) were indentified and quantified in both data sets. Of these, 424 phosphopeptides on 259 proteins were found to be up-regulated and 392 sites on 206 proteins were down-regulated (1.8-fold or more). Protein kinases and phosphatases, as well as sites in many proteins involved in G-protein signaling, were prominently represented in the up-regulated group and more than half of the kinase up-regulated phosphosites could be clustered into three sequence motifs; a similar distribution was also found for the down-regulated sites. A comparison of the up-regulated motif profile observed to that calculated from a previous study of the EGFR-induced phosphoproteome in human HeLa cells at the same time point showed a considerable amount of similarity, supporting the view that RTK signal transduction pathways and downstream modifications are likely to be extensively overlapping.


Asunto(s)
Procesamiento Proteico-Postraduccional , Receptor trkA/metabolismo , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Serina/metabolismo , Treonina/metabolismo , Secuencias de Aminoácidos , Animales , Becaplermina , Clonación Molecular , Humanos , Marcaje Isotópico , Factor de Crecimiento Nervioso/fisiología , Neuritas/metabolismo , Células PC12 , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Fosforilación , Mapas de Interacción de Proteínas , Estructura Terciaria de Proteína , Proteoma/química , Proteoma/metabolismo , Proteínas Proto-Oncogénicas c-sis/fisiología , Ratas , Receptor trkA/química , Receptores del Factor de Crecimiento Derivado de Plaquetas/química , Proteínas Recombinantes de Fusión/química , Transducción de Señal , Regulación hacia Arriba
18.
Am J Pathol ; 180(6): 2590-7, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22507839

RESUMEN

The acute response to vascular cell injury, which underpins vasculo-occlusive pathologies such as atherogenesis and restenosis after percutaneous coronary intervention, involves a complex series of molecular events that alter patterns of gene expression and favor a synthetic phenotype. One transcription factor that has been implicated in this process is the evolutionarily conserved mammalian stress response pathway regulator activating transcription factor 4 (ATF-4). Here, we show for the first time that both mRNA and protein levels of ATF-4 are induced in smooth muscle cells (SMCs) by the potent migratory factor PDGF-BB through PDGFR-ß. PDGF-BB also stimulates the expression of tenascin-C (TN-C), an extracellular matrix glycoprotein that regulates the activity of focal adhesion complexes, facilitating the SMC migration that underlies negative vascular remodeling in response to injury. Overexpression of ATF-4 increased transcript levels of the four TN-C isoforms in rat vascular SMCs, and ATF-4 knockdown inhibited PDGF-BB-inducible TN-C expression in vitro and injury-inducible TN-C protein expression in the balloon-injured rat artery wall. Furthermore, we show that ATF-4 is required for PDGF-BB-inducible SMC migration in response to injury. PDGF-BB-induced migration was also compromised in ATF-4 null mEFs, and this effect was rescued by the addition of TN-C. Our findings thus demonstrate the role of ATF-4 in both injury- and PDGF-BB-inducible TN-C expression and cell migration.


Asunto(s)
Factor de Transcripción Activador 4/biosíntesis , Proteínas Proto-Oncogénicas c-sis/fisiología , Tenascina/biosíntesis , Factor de Transcripción Activador 4/deficiencia , Factor de Transcripción Activador 4/genética , Animales , Becaplermina , Traumatismos de las Arterias Carótidas/metabolismo , Traumatismos de las Arterias Carótidas/fisiopatología , Arteria Carótida Común/metabolismo , Relación Dosis-Respuesta a Droga , Fibroblastos/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Ratones , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Proteínas Proto-Oncogénicas c-sis/farmacología , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Ratas , Ratas Sprague-Dawley , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/fisiología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/fisiología , Tenascina/genética , Cicatrización de Heridas/fisiología
19.
Toxicol Appl Pharmacol ; 272(3): 713-25, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23958495

RESUMEN

TRPM7, a non-selective cation channel of the TRP channel superfamily, is implicated in diverse physiological and pathological processes including cell proliferation. Recently, TRPM7 has been reported in hepatic stellate cells (HSCs). Here, we investigated the contribution role of TRPM7 in activated HSC-T6 cell (a rat hepatic stellate cell line) proliferation. TRPM7 mRNA and protein were measured by RT-PCR and Western blot in rat model of liver fibrosis in vivo and PDGF-BB-activated HSC-T6 cells in vitro. Both mRNA and protein of TRPM7 were dramatically increased in CCl4-treated rat livers. Stimulation of HSC-T6 cells with PDGF-BB resulted in a time-dependent increase of TRPM7 mRNA and protein. However, PDGF-BB-induced HSC-T6 cell proliferation was inhibited by non-specific TRPM7 blocker 2-aminoethoxydiphenyl borate (2-APB) or synthetic siRNA targeting TRPM7, and this was accompanied by downregulation of cell cycle proteins, cyclin D1, PCNA and CDK4. Blockade of TRPM7 channels also attenuated PDGF-BB induced expression of myofibroblast markers as measured by the induction of α-SMA and Col1α1. Furthermore, the phosphorylation of ERK and AKT, associated with cell proliferation, decreased in TRPM7 deficient HSC-T6 cells. These observations suggested that TRPM7 channels contribute to perpetuated fibroblast activation and proliferation of PDGF-BB induced HSC-T6 cells via the activation of ERK and PI3K pathways. Therefore, TRPM7 may constitute a useful target for the treatment of liver fibrosis.


Asunto(s)
Proliferación Celular , Células Estrelladas Hepáticas/enzimología , Sistema de Señalización de MAP Quinasas/fisiología , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Proto-Oncogénicas c-sis/fisiología , Canales Catiónicos TRPM/fisiología , Animales , Becaplermina , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Cadena alfa 1 del Colágeno Tipo I , Fibroblastos/enzimología , Fibroblastos/metabolismo , Fibroblastos/patología , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/patología , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratas , Ratas Sprague-Dawley
20.
Neurochem Res ; 38(2): 346-55, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23179587

RESUMEN

The proliferation of neonatal Schwann cells (SCs) in response to mitogenic agents has been well analyzed in vitro (mono-layer-culture method, 2D environment), but not in vivo (3D environment). To assess the mitogenic effect of platelet-derived growth factors-BB (PDGF-BB), Fibroblast Growth Factors-base (bFGF), and their combinations for SCs in collagen gel (three-dimensional, 3D environment), we have developed an integrated microfluidic device on which can reproducibly measure the proliferation from small number of cells (1-100). The rat SCs were cultured for 4 week at the different concentrations of growth factors generated by concentration gradient generator. In the collagen gel culture, almost all of the cells in colonies presented a round cell morphology and maintained their round morphology by the 4th week. The results showed that PDGF-BB and bFGF are all capable of moderately stimulating SCs growth and every group reached the peak in the growth curve at 3 weeks. Moreover, the proliferation test using the conventional method was performed simultaneously and revealed similar results. The biggest difference between 2D and 3D was that cells decrease more remarkable in 3D than that in 2D at 4 weeks. And at 2 and 3 weeks, the growth rate in the collagen gel with 7.14/2.86 and 8.57/1.43 ng/mL groups was higher than that in the mono-layer culture. Our results showed that PDGF-BB and bFGF are capable of moderately stimulating neonatal SCs growth, respectively and synergistically, and the microfluidic technique is highly controllable, contamination free, fully automatic, and inexpensive.


Asunto(s)
Proliferación Celular , Factor 2 de Crecimiento de Fibroblastos/fisiología , Proteínas Proto-Oncogénicas c-sis/fisiología , Células de Schwann/fisiología , Animales , Animales Recién Nacidos , Becaplermina , Técnicas de Cultivo de Célula/instrumentación , Técnicas de Cultivo de Célula/métodos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA