Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.440
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Dev Dyn ; 250(7): 955-973, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33501723

RESUMEN

BACKGROUND: Neural tube (NT) closure is a complex developmental process that takes place in the early stages of embryogenesis and that is a key step in neurulation. In mammals, the process by which the neural plate generates the NT requires organized cell movements and tissue folding, and it terminates with the fusion of the apposed ends of the neural folds. RESULTS: Here we describe how almost identical cellular and molecular machinery is used to fuse the spinal neural folds as that involved in the repair of epithelial injury in the same area of the embryo. For both natural and wound activated closure of caudal neural tissue, hyaluronic acid and platelet-derived growth factor signaling appear to be crucial for the final fusion step. CONCLUSIONS: There seems to be no general wound healing machinery for all tissues but rather, a tissue-specific epithelial fusion machinery that embryos activate when necessary after abnormal epithelial opening.


Asunto(s)
Células Epiteliales/fisiología , Tubo Neural/embriología , Neurulación/fisiología , Cicatrización de Heridas/fisiología , Animales , Fusión Celular , Células Cultivadas , Embrión de Mamíferos , Desarrollo Embrionario/fisiología , Células Epiteliales/citología , Femenino , Feto/embriología , Ácido Hialurónico/metabolismo , Masculino , Ratones , Cresta Neural/embriología , Cresta Neural/fisiología , Placa Neural/embriología , Placa Neural/fisiología , Defectos del Tubo Neural/embriología , Factor de Crecimiento Derivado de Plaquetas/fisiología , Embarazo
2.
Mol Cell ; 49(4): 751-8, 2013 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-23352452

RESUMEN

Raf kinases are essential for normal Ras-Raf-MEK-ERK pathway signaling, and activating mutations in components of this pathway are associated with a variety of human cancers, as well as the related developmental disorders Noonan, LEOPARD, and cardiofaciocutaneous syndromes. Although the Raf kinases are known to dimerize during normal and disease-associated Raf signaling, the functional significance of Raf dimerization has not been fully elucidated. Here, using mutational analysis and a peptide inhibitor, we show that dimerization is required for normal Ras-dependent Raf activation and for the biological function of disease-associated Raf mutants with moderate, low, or impaired kinase activity. However, dimerization is not needed for the function of B-Raf mutants with high catalytic activity, such as V600E-B-Raf. Importantly, we find that a dimer interface peptide can effectively block Raf dimerization and inhibit Raf signaling when dimerization is required for Raf function, thus identifying the Raf dimer interface as a therapeutic target.


Asunto(s)
Sistema de Señalización de MAP Quinasas , Quinasas raf/metabolismo , Sustitución de Aminoácidos , Animales , Línea Celular , Activación Enzimática , Factor de Crecimiento Epidérmico/fisiología , Humanos , Ratones , Mutagénesis Sitio-Dirigida , Mutación Missense , Neoplasias/enzimología , Fragmentos de Péptidos/farmacología , Factor de Crecimiento Derivado de Plaquetas/fisiología , Dominios y Motivos de Interacción de Proteínas , Inhibidores de Proteínas Quinasas/farmacología , Multimerización de Proteína , Quinasas raf/antagonistas & inhibidores , Quinasas raf/química , Quinasas raf/genética , Proteínas ras/metabolismo
3.
Acta Biochim Biophys Sin (Shanghai) ; 53(12): 1640-1649, 2021 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-34586354

RESUMEN

Osteocytes are the main sensitive cells in bone remodeling due to their potent functional cell processes from the mineralized bone matrix to the bone surface and the bone marrow. Neighboring osteocytes communicate with each other by these cell processes to achieve molecular exchange through gap junction channels. Platelet-derived growth factor-AA (PDGF-AA) has been reported to enhance bone tissue remodeling by promoting cell proliferation, migration, and autocrine secretion in osteoid cell linage. However, the effect of PDGF-AA on intercellular communication between osteocytes is still unclear. In the present study, we elucidated that PDGF-AA could enhance the formation of dendritic processes of osteocytes and the gap junctional intercellular communication by promoting the expression of connexin43 (Cx43). This modulation process was mainly dependent on the activation of phosphorylation of Akt protein by phosphatidylinositol 3-kinase (PI3K)/Akt (also known as protein kinase B, PKB) signaling. Inhibition of PI3K/Akt signaling decreased the Cx43 expression induced by PDGF-AA. These results establish a bridge between PDGF-AA and cell-cell communication in osteocytes, which could help us understand the molecular exchange between bone cells and fracture healing.


Asunto(s)
Comunicación Celular/fisiología , Osteocitos/fisiología , Fosfatidilinositol 3-Quinasa/metabolismo , Factor de Crecimiento Derivado de Plaquetas/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Línea Celular , Cromonas/farmacología , Conexina 43/genética , Conexina 43/metabolismo , Dendritas/metabolismo , Uniones Comunicantes/metabolismo , Ratones , Morfolinas/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Fosforilación/fisiología , Factor de Crecimiento Derivado de Plaquetas/genética , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Transducción de Señal/fisiología , Regulación hacia Arriba
4.
Hepatology ; 70(6): 2107-2122, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31016744

RESUMEN

The growing burden of liver fibrosis and lack of effective antifibrotic therapies highlight the need for identification of pathways and complementary model systems of hepatic fibrosis. A rare, monogenic disorder in which children with mutations in mannose phosphate isomerase (MPI) develop liver fibrosis led us to explore the function of MPI and mannose metabolism in liver development and adult liver diseases. Herein, analyses of transcriptomic data from three human liver cohorts demonstrate that MPI gene expression is down-regulated proportionate to fibrosis in chronic liver diseases, including nonalcoholic fatty liver disease and hepatitis B virus. Depletion of MPI in zebrafish liver in vivo and in human hepatic stellate cell (HSC) lines in culture activates fibrotic responses, indicating that loss of MPI promotes HSC activation. We further demonstrate that mannose supplementation can attenuate HSC activation, leading to reduced fibrogenic activation in zebrafish, culture-activated HSCs, and in ethanol-activated HSCs. Conclusion: These data indicate the prospect that modulation of mannose metabolism pathways could reduce HSC activation and improve hepatic fibrosis.


Asunto(s)
Células Estrelladas Hepáticas/fisiología , Cirrosis Hepática/etiología , Manosa-6-Fosfato Isomerasa/fisiología , Manosa/farmacología , Animales , Células Cultivadas , Glicosilación , Humanos , Masculino , Factor de Crecimiento Derivado de Plaquetas/fisiología , Transducción de Señal/fisiología , Pez Cebra
5.
FASEB J ; 33(6): 7363-7374, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30865840

RESUMEN

Pulmonary arterial hypertension (PAH) is a progressive and fatal disease associated with remodeling of the pulmonary artery. We previously reported that the Ca2+-sensing receptor (CaSR) is up-regulated in pulmonary arterial smooth muscle cells (PASMCs) from patients with idiopathic PAH (IPAH) and contributes to enhanced Ca2+ responses and excessive cell proliferation. However, the mechanisms underlying the up-regulation of CaSR have not yet been elucidated. We herein examined involvement of platelet-derived growth factor (PDGF) on CaSR expression, Ca2+ responses, and proliferation in PASMCs. The expression of PDGF receptors was higher in PASMCs from patients with IPAH than in PASMCs from normal subjects. In addition, PDGF-induced activation of PDGF receptors and their downstream molecules [ERK1/2, p38, protein kinase B, and signal transducer and activator of transcription (STAT) 1/3] were sustained longer in PASMCs from patients with IPAH. The PDGF-induced CaSR up-regulation was attenuated by small interfering RNA knockdown of PDGF receptors and STAT1/3, and by the treatment with imatinib. In monocrotaline-induced pulmonary hypertensive rats, the up-regulation of CaSR was reduced by imatinib. The combination of NPS2143 and imatinib additively inhibited the development of pulmonary hypertension. These results suggest that enhanced PDGF signaling is involved in CaSR up-regulation, leading to excessive PASMC proliferation and vascular remodeling in patients with IPAH. The linkage between CaSR and PDGF signals is a novel pathophysiological mechanism contributing to the development of PAH.-Yamamura, A., Nayeem, M. J., Al Mamun, A., Takahashi, R., Hayashi, H., Sato, M. Platelet-derived growth factor up-regulates Ca2+-sensing receptors in idiopathic pulmonary arterial hypertension.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Hipertensión Pulmonar/fisiopatología , Miocitos del Músculo Liso/metabolismo , Factor de Crecimiento Derivado de Plaquetas/fisiología , Receptores Sensibles al Calcio/biosíntesis , Remodelación Vascular/fisiología , Animales , Calcio/fisiología , División Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Hipertensión Pulmonar/tratamiento farmacológico , Hipertensión Pulmonar/patología , Hipertensión Pulmonar/prevención & control , Mesilato de Imatinib/farmacología , Mesilato de Imatinib/uso terapéutico , Masculino , Monocrotalina/toxicidad , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/efectos de los fármacos , Naftalenos/farmacología , Naftalenos/uso terapéutico , Factor de Crecimiento Derivado de Plaquetas/farmacología , Arteria Pulmonar/patología , Ratas , Ratas Sprague-Dawley , Receptores Sensibles al Calcio/genética , Receptores del Factor de Crecimiento Derivado de Plaquetas/agonistas , Receptores del Factor de Crecimiento Derivado de Plaquetas/fisiología , Transducción de Señal/efectos de los fármacos , Remodelación Vascular/efectos de los fármacos
6.
Dev Biol ; 442(1): 155-161, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30026121

RESUMEN

FGF signaling is known to play a critical role in the specification of primitive endoderm (PrE) and epiblast (Epi) from the inner cell mass (ICM) during mouse preimplantation development, but how FGFs synergize with other growth factor signaling pathways is unknown. Because PDGFRα signaling has also been implicated in the PrE, we investigated the coordinate functions of PDGFRα together with FGFR1 or FGFR2 in PrE development. PrE development was abrogated in Pdgfra; Fgfr1 compound mutants, or significantly reduced in Pdgfra; Fgfr2 or PdgfraPI3K; Fgfr2 compound mutants. We provide evidence that both Fgfr2 and Pdgfra play roles in PrE cell survival while Fgfr1 controls PrE cell specification. Our results suggest a model where FGFR1-engaged ERK1/2 signaling governs PrE specification while PDGFRα- and by analogy possibly FGFR2- engaged PI3K signaling regulates PrE survival and positioning in the embryo. Together, these studies indicate how multiple growth factors and signaling pathways can cooperate in preimplantation development.


Asunto(s)
Factor 4 de Crecimiento de Fibroblastos/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Animales , Blastocisto/metabolismo , Masa Celular Interna del Blastocisto/metabolismo , Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario , Endodermo/metabolismo , Factor 4 de Crecimiento de Fibroblastos/fisiología , Factores de Crecimiento de Fibroblastos/metabolismo , Estratos Germinativos/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Ratones Transgénicos , Fosfatidilinositol 3-Quinasas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/fisiología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal/fisiología
7.
Bull Math Biol ; 81(6): 1645-1664, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30796683

RESUMEN

Paracrine PDGF signaling is involved in many processes in the body, both normal and pathological, including embryonic development, angiogenesis, and wound healing as well as liver fibrosis, atherosclerosis, and cancers. We explored this seemingly dual (normal and pathological) role of PDGF mathematically by modeling the release of PDGF in brain tissue and then varying the dynamics of this release. Resulting simulations show that by varying the dynamics of a PDGF source, our model predicts three possible outcomes for PDGF-driven cellular recruitment and lesion growth: (1) localized, short duration of growth, (2) localized, chronic growth, and (3) widespread chronic growth. Further, our model predicts that the type of response is much more sensitive to the duration of PDGF exposure than the maximum level of that exposure. This suggests that extended duration of paracrine PDGF signal during otherwise normal processes could potentially lead to lesions having a phenotype consistent with pathologic conditions.


Asunto(s)
Encéfalo/patología , Encéfalo/fisiopatología , Modelos Neurológicos , Factor de Crecimiento Derivado de Plaquetas/fisiología , Animales , Encéfalo/crecimiento & desarrollo , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/fisiopatología , Simulación por Computador , Humanos , Conceptos Matemáticos , Células Precursoras de Oligodendrocitos/patología , Células Precursoras de Oligodendrocitos/fisiología , Comunicación Paracrina/fisiología
8.
Med Sci Monit ; 25: 10016-10028, 2019 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-31877561

RESUMEN

BACKGROUND Dental pulp cells (DPCs) play vital roles in the recovery of dental pulp tissue. Concentrated growth factor (CGF) can promote proliferation and mineralization of various cells. However, the functions of CGF on DPCs and dental pulp tissue are unclear. The object of our study was to identify the roles of CGF in DPCs proliferation and mineralization in vitro and to assess the effects of CGF on direct pulp capping in vivo. MATERIAL AND METHODS We performed CCK-8 and Transwell assay to detect proliferation and migration activity of DPCs. Alizarin Red staining was performed to examine mineralized nodules. Alkaline phosphatase activity test was used to measure the mineralization capacity of DPCs. We assessed the odontogenic differentiation gene expression level by Western blot and qPCR. The effect of CGF on direct pulp capping in vivo were evaluated by radiography and histopathology. RESULTS CGF increased the number of proliferative and migratory DPCs. CGF enhanced DPCs mineralized nodules and improved the gene expression levels of DSPP, DMP-1, BSP, and ALP. CGF upregulated the protein levels of ALP, BMP2, SMAD5, Runx2, and p-Smad, and the effect could be partially reversed by Noggin. CGF promoted pulp recovery and kept its vitality in directly pulp capping. CONCLUSIONS CGF promotes DPCs proliferation and mineralization. It regulates the mineralization of DPCs via the BMP2/SMAD5/Runx2 signaling pathway. CGF can be used as the effective graft for direct pulp capping.


Asunto(s)
Pulpa Dental/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/farmacología , Adolescente , Adulto , Calcificación Fisiológica/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Masculino , Tercer Molar , Factor de Crecimiento Derivado de Plaquetas/fisiología , Cultivo Primario de Células/métodos , Diente/metabolismo , Adulto Joven
9.
Am J Physiol Lung Cell Mol Physiol ; 314(4): L593-L605, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29212800

RESUMEN

Pulmonary hypertension (PH) is a lethal condition, and current vasodilator therapy has limited effect. Antiproliferative strategies targeting platelet-derived growth factor (PDGF) receptors, such as imatinib, have generated promising results in animal studies. Imatinib is, however, a nonspecific tyrosine kinase inhibitor and has in clinical studies caused unacceptable adverse events. Further studies are needed on the role of PDGF signaling in PH. Here, mice expressing a variant of PDGF-B with no retention motif ( Pdgfbret/ret), resulting in defective binding to extracellular matrix, were studied. Following 4 wk of hypoxia, right ventricular systolic pressure, right ventricular hypertrophy, and vascular remodeling were examined. Pdgfbret/ret mice did not develop PH, as assessed by hemodynamic parameters. Hypoxia did, however, induce vascular remodeling in Pdgfbret/ret mice; but unlike the situation in controls where the remodeling led to an increased concentric muscularization of arteries, the vascular remodeling in Pdgfbret/ret mice was characterized by a diffuse muscularization, in which cells expressing smooth muscle cell markers were found in the interalveolar septa detached from the normally muscularized intra-acinar vessels. Additionally, fewer NG2-positive perivascular cells were found in Pdgfbret/ret lungs, and mRNA analyses showed significantly increased levels of Il6 following hypoxia, a known promigratory factor for pericytes. No differences in proliferation were detected at 4 wk. This study emphasizes the importance of extracellular matrix-growth factor interactions and adds to previous knowledge of PDGF-B in PH pathobiology. In summary, Pdgfbret/ret mice have unaltered hemodynamic parameters following chronic hypoxia, possibly secondary to a disorganized vascular muscularization.


Asunto(s)
Modelos Animales de Enfermedad , Matriz Extracelular/patología , Hipertensión Pulmonar/patología , Hipoxia/fisiopatología , Linfocinas/fisiología , Músculo Liso Vascular/patología , Factor de Crecimiento Derivado de Plaquetas/fisiología , Remodelación Vascular , Animales , Proliferación Celular , Células Cultivadas , Matriz Extracelular/metabolismo , Femenino , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/metabolismo , Ratones , Ratones Endogámicos C57BL , Músculo Liso Vascular/metabolismo , Pericitos/metabolismo , Pericitos/patología , Transducción de Señal
10.
Cancer Sci ; 109(5): 1513-1523, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29575648

RESUMEN

Glioma is the most common form of malignant brain cancer in adults. The Sleeping Beauty (SB) transposon-based glioma mouse model allows for effective in vivo analysis of candidate genes. In the present study, we developed a transposon vector that encodes the triple combination of platelet-derived growth factor subunit A (PDGFA), and shRNAs against Nf1 and Trp53 (shNf1/shp53). Initiation and progression of glioma in the brain were monitored by expression of a fluorescent protein. Transduction of the vector into neural progenitor and stem cells (NPC) in the subventricular zone (SVZ) of the neonatal brain induced proliferation of oligodendrocyte precursor cells, and promoted formation of highly penetrant malignant gliomas within 2-4 months. Cells isolated from the tumors were capable of forming secondary tumors. Two transposon vectors, encoding either PDGFA or shNf1/shp53 were co-electroporated into NPC. Cells expressing PDGFA or shNf1/shp53 were labeled with unique fluorescent proteins allowing visualization of the spatial distribution of cells with different genetic alterations within the same tumor. Tumor cells located at the center of tumors expressed PDGFA at higher levels than those located at the periphery, indicating that intratumoral heterogeneity in PDGFA expression levels spontaneously developed within the same tumor. Tumor cells comprising the palisading necrosis strongly expressed PDGFA, suggesting that PDGFA signaling is involved in hypoxic responses in glioma. The transposon vectors developed are compatible with any genetically engineered mouse model, providing a useful tool for the functional analysis of candidate genes in glioma.


Asunto(s)
Neoplasias Encefálicas/etiología , Elementos Transponibles de ADN/genética , Modelos Animales de Enfermedad , Glioma/etiología , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Hipoxia de la Célula , Proliferación Celular , Glioma/genética , Glioma/patología , Ratones , Ratones Endogámicos ICR , Células 3T3 NIH , Neurofibromina 1/genética , Factor de Crecimiento Derivado de Plaquetas/genética , Factor de Crecimiento Derivado de Plaquetas/fisiología , Transducción de Señal , Proteína p53 Supresora de Tumor/genética
11.
Osteoarthritis Cartilage ; 26(8): 1045-1054, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29782915

RESUMEN

OBJECTIVE: This study was conducted to identify cytokine profiles associated with radiographic phenotypes of knee osteoarthritis (rKOA) with a focus on early stage of the disease. METHODS: The pilot population study involved 60 middle-aged patients (mean age 50 ± 7.3y.). Standardized weight-bearing anteroposterior and axial radiographs were used to assess rKOA severity in tibiofemoral (TFJ) of patellofemoral joint (PFJ) by grading system (grades 0-3). Luminex (xMAP®) technology was used to simultaneously assess 60 biomarkers (BMs). RESULTS: Several pathways of angiogenic (CXCL10/IP-10, FGF1/2, PDGF-AA/BB, ANG1, RANTES), tissue remodeling/fibrosis (MMP1/3, TIMP2/3/4, TGFß), and fat tissue (leptin) BMs associated with rKOA severity already in very early phase (grade 1). We identified several sets of cytokines as key markers of early knee osteoarthritis (KOA) predicting radiographic features in logistic-regression models (AUC = 0.80-0.97). Marked sex-specificity of rKOA course was detected: upregulation of angiogenesis dominated in females, whereas the activation of tissue remodeling was dominant in males. Several of these shifts, e.g., decrease of CXCL10/IP-10, took place only in grade 1 KOA and disappeared or reversed in later stages. OA of different knee-joint compartments has distinct profiles of cytokines. A broad list of BMs (TIMP2/3/4, MMP1/3, TGFß1/2, vWF-A2, sE-selectin and leptin) associated with OA in the PFJ. CONCLUSION: Our results demonstrate that substantial and time-limited shifts in the angiogenic and TIMP/MMP systems occur in the early stage of KOA. Our study findings highlight the sex-, grade- and compartment-dependent shifts in above processes. The data may contribute to the individualized prevention of KOA in the future.


Asunto(s)
Citocinas/fisiología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Neovascularización Patológica/patología , Osteoartritis de la Rodilla/patología , Biomarcadores/metabolismo , Quimiocina CXCL10/metabolismo , Quimiocina CXCL10/fisiología , Citocinas/metabolismo , Progresión de la Enfermedad , Factor 1 de Crecimiento de Fibroblastos/metabolismo , Factor 1 de Crecimiento de Fibroblastos/fisiología , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Metaloproteinasa 1 de la Matriz/metabolismo , Metaloproteinasa 1 de la Matriz/fisiología , Persona de Mediana Edad , Neovascularización Patológica/metabolismo , Osteoartritis de la Rodilla/metabolismo , Proyectos Piloto , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/fisiología , Factores Sexuales , Inhibidor Tisular de Metaloproteinasa-2/metabolismo , Inhibidor Tisular de Metaloproteinasa-2/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/fisiología
12.
Bull Math Biol ; 80(5): 1292-1309, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-28842831

RESUMEN

Gliomas are the most common of all primary brain tumors. They are characterized by their diffuse infiltration of the brain tissue and are uniformly fatal, with glioblastoma being the most aggressive form of the disease. In recent years, the over-expression of platelet-derived growth factor (PDGF) has been shown to produce tumors in experimental rodent models that closely resemble this human disease, specifically the proneural subtype of glioblastoma. We have previously modeled this system, focusing on the key attribute of these experimental tumors-the "recruitment" of oligodendroglial progenitor cells (OPCs) to participate in tumor formation by PDGF-expressing retrovirally transduced cells-in one dimension, with spherical symmetry. However, it has been observed that these recruitable progenitor cells are not uniformly distributed throughout the brain and that tumor cells migrate at different rates depending on the material properties in different regions of the brain. Here we model the differential diffusion of PDGF-expressing and recruited cell populations via a system of partial differential equations with spatially variable diffusion coefficients and solve the equations in two spatial dimensions on a mouse brain atlas using a flux-differencing numerical approach. Simulations of our in silico model demonstrate qualitative agreement with the observed tumor distribution in the experimental animal system. Additionally, we show that while there are higher concentrations of OPCs in white matter, the level of recruitment of these plays little role in the appearance of "white matter disease," where the tumor shows a preponderance for white matter. Instead, simulations show that this is largely driven by the ratio of the diffusion rate in white matter as compared to gray. However, this ratio has less effect on the speed of tumor growth than does the degree of OPC recruitment in the tumor. It was observed that tumor simulations with greater degrees of recruitment grow faster and develop more nodular tumors than if there is no recruitment at all, similar to our prior results from implementing our model in one dimension. Combined, these results show that recruitment remains an important consideration in understanding and slowing glioma growth.


Asunto(s)
Neoplasias Encefálicas/patología , Glioma/patología , Factor de Crecimiento Derivado de Plaquetas/fisiología , Animales , Simulación por Computador , Humanos , Conceptos Matemáticos , Ratones , Modelos Neurológicos , Invasividad Neoplásica/patología , Células Madre Neoplásicas/patología , Células Precursoras de Oligodendrocitos/patología
13.
Int J Gynecol Cancer ; 28(2): 323-331, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29240605

RESUMEN

OBJECTIVE: This study aimed to investigate whether platelet-derived growth factor D (PDGF-D) is a prognostic biomarker and is associated with platinum resistance in epithelial ovarian cancer, which has not been studied by others previously. METHODS: In this study, we detected expression of PDGF-D in ovarian cancer tissues through immunohistochemistry and Western blotting. Furthermore, we analyzed the association between PDGF-D expression and clinicopathological features including prognosis in epithelial ovarian cancer. Statistical analyses were performed by using χ test, log-rank test, Cox regression test, and Kaplan-Meier method. RESULTS: High PDGF-D expression is positively correlated with International Federation of Gynecology and Obstetrics stage (P < 0.001), histologic grade (P < 0.001), lymph node metastasis (P = 0.022), and poor prognosis (P < 0.001). Platelet-derived growth factor D in platinum-resistant cases is overexpressed compared with that in platinum-sensitive cases (P < 0.001). Obstetrics stage (P = 0.029) and PDGF-D overexpression (P < 0.001) are independently correlated with platinum resistance. CONCLUSIONS: Our study indicates that PDGF-D overexpression is an independent predictor of platinum-based chemotherapy resistance and that it may also be a potential biomarker for targeted therapy and poor prognosis.


Asunto(s)
Biomarcadores de Tumor , Carcinoma Epitelial de Ovario/diagnóstico , Resistencia a Antineoplásicos , Linfocinas/fisiología , Neoplasias Ováricas/diagnóstico , Factor de Crecimiento Derivado de Plaquetas/fisiología , Compuestos de Platino/uso terapéutico , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/metabolismo , Carcinoma Epitelial de Ovario/tratamiento farmacológico , Carcinoma Epitelial de Ovario/metabolismo , Femenino , Humanos , Linfocinas/metabolismo , Persona de Mediana Edad , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Valor Predictivo de las Pruebas , Pronóstico , Adulto Joven
14.
Circ Res ; 116(7): 1269-76, 2015 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-25814687

RESUMEN

Fibrotic diseases are a significant global burden for which there are limited treatment options. The effector cells of fibrosis are activated fibroblasts called myofibroblasts, a highly contractile cell type characterized by the appearance of α-smooth muscle actin stress fibers. The underlying mechanism behind myofibroblast differentiation and persistence has been under much investigation and is known to involve a complex signaling network involving transforming growth factor-ß, endothelin-1, angiotensin II, CCN2 (connective tissue growth factor), and platelet-derived growth factor. This review addresses the contribution of these signaling molecules to cardiac fibrosis.


Asunto(s)
Miocardio/patología , Angiotensina II/fisiología , Bloqueadores del Receptor Tipo 1 de Angiotensina II/uso terapéutico , Animales , Antiinflamatorios/uso terapéutico , Arritmias Cardíacas/etiología , Atrofia , Cicatriz/patología , Factor de Crecimiento del Tejido Conjuntivo/fisiología , Antagonistas de los Receptores de Endotelina/uso terapéutico , Endotelina-1/fisiología , Fibrosis , Humanos , Hipoxia/etiología , Modelos Cardiovasculares , Terapia Molecular Dirigida , Miofibroblastos/fisiología , Factor de Crecimiento Derivado de Plaquetas/fisiología , Piridonas/uso terapéutico , Ratas , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/fisiología
15.
J Cell Sci ; 127(Pt 8): 1684-98, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24481818

RESUMEN

Localization of the non-receptor tyrosine kinase Src to the cell periphery is required for its activation and to mediate focal adhesion turnover, cell spreading and migration. Inactive Src localizes to a perinuclear compartment and the movement of Src to the plasma membrane is mediated by endocytic transport. However, the precise pathways and regulatory proteins that are responsible for SRC transport are incompletely understood. Here, we demonstrate that Src partially colocalizes with the endocytic regulatory protein MICAL-L1 (molecule interacting with CasL-like protein 1) in mammalian cells. Furthermore, MICAL-L1 is required for growth-factor- and integrin-induced Src activation and transport to the cell periphery in HeLa cells and human fibroblasts. Accordingly, MICAL-L1 depletion impairs focal adhesion turnover, cell spreading and cell migration. Interestingly, we find that the MICAL-L1 interaction partner EHD1 (EH domain-containing protein 1) is also required for Src activation and transport. Moreover, the MICAL-L1-mediated recruitment of EHD1 to Src-containing recycling endosomes is required for the release of Src from the perinuclear endocytic recycling compartment in response to growth factor stimulation. Our study sheds new light on the mechanism by which Src is transported to the plasma membrane and activated, and provides a new function for MICAL-L1 and EHD1 in the regulation of intracellular non-receptor tyrosine kinases.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Proteínas del Citoesqueleto/fisiología , Proteínas con Dominio LIM/fisiología , Proteínas de Transporte Vesicular/fisiología , Familia-src Quinasas/metabolismo , Animales , Membrana Celular/enzimología , Movimiento Celular , Forma de la Célula , Endocitosis , Endosomas/enzimología , Activación Enzimática , Factor de Crecimiento Epidérmico/fisiología , Adhesiones Focales/metabolismo , Células HeLa , Humanos , Ratones , Proteínas de Microfilamentos , Oxigenasas de Función Mixta , Factor de Crecimiento Derivado de Plaquetas/fisiología , Transporte de Proteínas , Vesículas Transportadoras/metabolismo
16.
Biochem Biophys Res Commun ; 472(1): 243-9, 2016 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-26920052

RESUMEN

S100A8 is an important member of the S100 protein family, which is involved in intracellular and extracellular regulatory activities. We previously reported that the S100A8 protein was differentially expressed in the asthmatic respiratory tracts. To understand the potential role of S100A8 in asthma, we investigated the effect of recombinant S100A8 protein on the platelet-derived growth factor (PDGF)-induced migration of airway smooth muscle cells (ASMCs) and the underlying molecular mechanism by using multiple methods, such as impedance-based xCELLigence migration assay, transwell migration assays and wound-healing assays. We found that exogenous S100A8 protein significantly inhibited PDGF-induced ASMC migration. Furthermore, the migration inhibition effect of S100A8 was blocked by neutralizing antibody against the receptor for advanced glycation end-products (RAGE), a potential receptor for the S100A8 protein. These findings provide direct evidence that exogenous S100A8 protein inhibits the PDGF-induced migration of ASMCs through the membrane receptor RAGE. Our study highlights a novel role of S100A8 as a potential means of counteracting airway remodeling in chronic airway diseases.


Asunto(s)
Calgranulina A/fisiología , Movimiento Celular/fisiología , Miocitos del Músculo Liso/fisiología , Factor de Crecimiento Derivado de Plaquetas/fisiología , Receptor para Productos Finales de Glicación Avanzada/fisiología , Tráquea/patología , Tráquea/fisiología , Animales , Anticuerpos Neutralizantes , Asma/patología , Asma/fisiopatología , Calgranulina A/administración & dosificación , Calgranulina A/genética , Células Cultivadas , Modelos Animales de Enfermedad , Ratas , Receptor para Productos Finales de Glicación Avanzada/antagonistas & inhibidores , Receptor para Productos Finales de Glicación Avanzada/inmunología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/genética , Cicatrización de Heridas
17.
IUBMB Life ; 68(3): 220-31, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26865509

RESUMEN

As a frequent event following chronic insult, liver fibrosis triggers wound healing reactions, with extracellular matrix components accumulated in the liver. During liver fibrogenesis, activation of hepatic stellate cells (HSCs) is the pivotal event. Fibrosis regression can feasibly be treated through pharmacological induction of HSC apoptosis. Herein we showed that dihydroartemisinin (DHA) improved liver histological architecture, decreased hepatic enzyme levels, and inhibited HSCs activation in the fibrotic rat liver. DHA also induced apoptosis of HSCs in such liver, as demonstrated by reduced distribution of α-SMA-positive cells and the presence of high number of cleaved-caspase-3-positive cells in vivo, as well as by down-regulation of Bcl-2 and up-regulation of Bax. In addition, in vitro experiments showed that DHA significantly inhibited HSC proliferation and led to dramatic morphological alterations in HSCs. we found that DHA disrupted mitochondrial functions and led to activation of caspase cascades in HSCs. Mechanistic investigations revealed that DHA induced HSC apoptosis through disrupting the phosphoinositide 3-kinase (PI3K)/Akt pathway and that PI3K specific inhibitor LY294002 mimicked the pro-apoptotic effect of DHA. DHA is a promising candidate for the prevention and treatment of liver fibrosis.


Asunto(s)
Antiinflamatorios/farmacología , Apoptosis/efectos de los fármacos , Artemisininas/farmacología , Células Estrelladas Hepáticas/fisiología , Cirrosis Hepática/prevención & control , Transducción de Señal/efectos de los fármacos , Animales , Conductos Biliares/patología , Supervivencia Celular , Evaluación Preclínica de Medicamentos , Células Estrelladas Hepáticas/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Masculino , Fosfatidilinositol 3-Quinasas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas Sprague-Dawley
18.
Br J Surg ; 103(8): 950-61, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27169866

RESUMEN

BACKGROUND: Radiation-induced fibrosis (RIF) is a late complication of radiotherapy that results in progressive functional and cosmetic impairment. Autologous fat has emerged as an option for soft tissue reconstruction. There are also sporadic reports suggesting regression of fibrosis following regional lipotransfer. This systematic review aimed to identify cellular mechanisms driving RIF, and the potential role of lipotransfer in attenuating these processes. METHODS: PubMed, OVID and Google Scholar databases were searched to identify all original articles regarding lipotransfer for RIF. All articles describing irradiated fibroblast or myofibroblast behaviour were included. Data elucidating the mechanisms of RIF, role of lipotransfer in RIF and methods to quantify fibrosis were extracted. RESULTS: Ninety-eight studies met the inclusion criteria. A single, definitive model of RIF is yet to be established, but four cellular mechanisms were identified through in vitro studies. Twenty-one studies identified connective tissue growth factor and transforming growth factor ß1 cytokines as drivers of fibrotic cascades. Hypoxia was demonstrated to propagate fibrogenesis in three studies. Oxidative stress from the release of reactive oxygen species and free radicals was also linked to RIF in 11 studies. Purified autologous fat grafts contain cellular and non-cellular properties that potentially interact with these processes. Six methods for quantifying fibrotic changes were evaluated including durometry, ultrasound shear wave elastography, thermography, dark field imaging, and laser Doppler and laser speckle flowmetry. CONCLUSION: Understanding how lipotransfer causes regression of RIF remains unclear; there are a number of new hypotheses for future research.


Asunto(s)
Tejido Adiposo/trasplante , Fibrosis/terapia , Radioterapia/efectos adversos , Piel/patología , Fenómenos Biomecánicos/fisiología , Fibrosis/diagnóstico por imagen , Humanos , Hipoxia/fisiopatología , Estrés Oxidativo/fisiología , Factor de Crecimiento Derivado de Plaquetas/fisiología , Piel/diagnóstico por imagen , Piel/fisiopatología , Factor de Crecimiento Transformador beta1/fisiología , Trasplante Autólogo , Factor de Necrosis Tumoral alfa/fisiología
19.
Tumour Biol ; 37(8): 10053-66, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27193823

RESUMEN

Progress in cancer biology has led to an increasing discovery of oncogenic alterations of the platelet-derived growth factor receptors (PDGFRs) in cancers. In addition, their overexpression in numerous cancers invariably makes PDGFRs and platelet-derived growth factors (PDGFs) prognostic and treatment markers in some cancers. The oncologic alterations of the PDGFR/PDGF system affect the extracellular, transmembrane and tyrosine kinase domains as well as the juxtamembrane segment of the receptor. The receptor is also involved in fusions with intracellular proteins and receptor tyrosine kinase. These discoveries undoubtedly make the system an attractive oncologic therapeutic target. This review covers elementary biology of PDGFR/PDGF system and its role as a prognostic and treatment marker in cancers. In addition, the multifarious therapeutic targets of PDGFR/PDGF system are discussed. Great potential exists in the role of PDGFR/PDGF system as a prognostic and treatment marker and for further exploration of its multifarious therapeutic targets in safe and efficacious management of cancer treatments.


Asunto(s)
Biomarcadores de Tumor/análisis , Proteínas de Neoplasias/análisis , Neoplasias/química , Factor de Crecimiento Derivado de Plaquetas/análisis , Receptores del Factor de Crecimiento Derivado de Plaquetas/análisis , Transducción de Señal , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Aptámeros de Péptidos/uso terapéutico , Ensayos Clínicos como Asunto , Monitoreo de Drogas , Humanos , Terapia Molecular Dirigida , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/química , Proteínas de Neoplasias/fisiología , Neoplasias/mortalidad , Neoplasias/terapia , Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Proteínas de Fusión Oncogénica/genética , Factor de Crecimiento Derivado de Plaquetas/química , Factor de Crecimiento Derivado de Plaquetas/fisiología , Pronóstico , Isoformas de Proteínas/análisis , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Interferencia de ARN , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Receptores del Factor de Crecimiento Derivado de Plaquetas/química , Receptores del Factor de Crecimiento Derivado de Plaquetas/fisiología , Resultado del Tratamiento
20.
Biochem Biophys Res Commun ; 463(4): 575-81, 2015 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-26043684

RESUMEN

Successful development of the conceptus and implantation requires an intimate trophic connection between maternal uterus and conceptus mediated by local regulators including growth factors. Platelet-derived growth factor (PDGF) acts as a chemotactic factor for a variety of cell types. Current studies have determined that PDGF participates in rapid growth and development of cleavage stage embryos, but PDGF-induced effects on the growth and development of peri-implantation conceptus remains unknown. In the present study, PDGF induced phosphorylation of ERK1/2, AKT and RPS6 proteins in porcine trophectoderm (pTr) cells in a dose- and time-dependent manner. Addition of U0126 (an inhibitor of ERK1/2) or LY294002 (a PI3K inhibitor) blocked PDGF-induced effects on phosphorylation of signaling proteins. Combinations of PDGF and U0126 decreased PDGF-induced p-ERK1/2 and p-AKT1, but combinations of PDGF and LY294002 blocked only PDGF-induced AKT phosphorylation. Furthermore, PDGF significantly induced pTr cell migration and these stimulatory effects were blocked by U0126 and LY294002. Immunoreactive p-ERK1/2 and p-RPS6 proteins were abundant in pTr cells treated with PDGF, but U0126 reduced PDGF-induced p-ERK1/2 and p-RPS6 levels to basal amounts. Present study suggests that PDGF secreted into the maternal-conceptus microenvironment stimulates pTr cell migration through signal transduction cascades mediated by the ERK1/2 MAPK and AKT1 pathways.


Asunto(s)
Movimiento Celular/fisiología , Ectodermo/citología , Sistema de Señalización de MAP Quinasas , Factor de Crecimiento Derivado de Plaquetas/fisiología , Trofoblastos/citología , Animales , Línea Celular , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA