Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 214
Filtrar
1.
JCI Insight ; 2(24)2017 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-29263300

RESUMEN

The scaffold protein synectin plays a critical role in the trafficking and regulation of membrane receptor pathways. As platelet-derived growth factor receptor (PDGFR) is essential for hepatic stellate cell (HSC) activation and liver fibrosis, we sought to determine the role of synectin on the PDGFR pathway and development of liver fibrosis. Mice with deletion of synectin from HSC were found to be protected from liver fibrosis. mRNA sequencing revealed that knockdown of synectin in HSC demonstrated reductions in the fibrosis pathway of genes, including PDGFR-ß. Chromatin IP assay of the PDGFR-ß promoter upon synectin knockdown revealed a pattern of histone marks associated with decreased transcription, dependent on p300 histone acetyltransferase. Synectin knockdown was found to downregulate PDGFR-α protein levels, as well, but through an alternative mechanism: protection from autophagic degradation. Site-directed mutagenesis revealed that ubiquitination of specific PDGFR-α lysine residues was responsible for its autophagic degradation. Furthermore, functional studies showed decreased PDGF-dependent migration and proliferation of HSC after synectin knockdown. Finally, human cirrhotic livers demonstrated increased synectin protein levels. This work provides insight into differential transcriptional and posttranslational mechanisms of synectin regulation of PDGFRs, which are critical to fibrogenesis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Cirrosis Hepática/metabolismo , Receptores del Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Autofagia/fisiología , Movimiento Celular/fisiología , Regulación hacia Abajo/fisiología , Técnicas de Silenciamiento del Gen , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/patología , Células Estrelladas Hepáticas/fisiología , Humanos , Cirrosis Hepática/genética , Cirrosis Hepática/patología , Cirrosis Hepática/prevención & control , Ratones Noqueados , Fibrosis Pulmonar/metabolismo , Receptores del Factor de Crecimiento Derivado de Plaquetas/genética , Receptores del Factor de Crecimiento Derivado de Plaquetas/fisiología , Ubiquitina/metabolismo , Regulación hacia Arriba/fisiología
2.
J Invest Dermatol ; 137(8): 1671-1681, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28433542

RESUMEN

Systemic sclerosis (SSc) is a multi-organ fibrotic disease with few treatment options. Activated fibroblasts are the key effector cells in SSc responsible for the excessive production of collagen and the development of fibrosis. Platelet-derived growth factor (PDGF), a potent mitogen for cells of mesenchymal origin, has been implicated in the activation of SSc fibroblasts. Our aim was to examine the therapeutic potential of crenolanib, an inhibitor of PDGF receptor signaling, in cultured fibroblasts and in angiotensin II-induced skin and heart fibrosis. Crenolanib effectively inhibited proliferation and migration of SSc and healthy control fibroblasts and attenuated basal and transforming growth factor-ß-induced expression of CCN2/CTGF and periostin. In contrast to healthy control fibroblasts, SSc fibroblasts proliferated in response to PDGFAA, whereas a combination of PDGFAA and CCN2 was required to elicit a similar response in healthy control fibroblasts. PDGF receptor α mRNA correlated with CCN2 and other fibrotic markers in the skin of SSc patients. In mice challenged with angiotensin II, PDGF receptor α-positive cells were increased in the skin and heart. These PDGF receptor α-positive cells co-localized with PDGF receptor ß, procollagen, and periostin. Treatment with crenolanib attenuated the skin and heart fibrosis. Our data indicate that inhibition of PDGF signaling presents an attractive therapeutic approach for SSc.


Asunto(s)
Bencimidazoles/farmacología , Piperidinas/farmacología , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Esclerodermia Sistémica/tratamiento farmacológico , Animales , Moléculas de Adhesión Celular/biosíntesis , Moléculas de Adhesión Celular/genética , Células Cultivadas , Factor de Crecimiento del Tejido Conjuntivo/biosíntesis , Factor de Crecimiento del Tejido Conjuntivo/genética , Modelos Animales de Enfermedad , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Regulación de la Expresión Génica , Humanos , Immunoblotting , Ratones , Ratones Endogámicos C57BL , ARN/genética , ARN Mensajero , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores del Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Receptores del Factor de Crecimiento Derivado de Plaquetas/genética , Esclerodermia Sistémica/metabolismo , Esclerodermia Sistémica/patología , Transducción de Señal
3.
Pediatr Blood Cancer ; 63(2): 348-51, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26375879

RESUMEN

Clinical and radiological diagnosis of infantile fibrosarcoma (IFS) is challenging because of its similarity to vascular origin tumors. Treatment involves complete resection. Although chemotherapy may allow more conservative resection, treatment guidelines are not strictly defined. One IFS patient with an unresectable tumor had disease progression during chemotherapy. A primary tumor sample showed high VEGFR-1/2/3 and PDGFR-α/ß expression. After pazopanib therapy, most tumor showed necrosis within 29 days and could be removed completely, with no relapse in 8 months post-resection. When IFS features hypervascularity, VEGFR and PDGFR expression may be high, thus allowing consideration of VEGFR inhibitors such as pazopanib.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Fibrosarcoma/tratamiento farmacológico , Terapia Neoadyuvante/métodos , Pirimidinas/uso terapéutico , Sulfonamidas/uso terapéutico , Axila/patología , Resistencia a Antineoplásicos , Fibrosarcoma/patología , Humanos , Indazoles , Lactante , Masculino , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Receptores del Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptores de Factores de Crecimiento Endotelial Vascular/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
4.
J Comp Pathol ; 151(4): 322-8, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25172054

RESUMEN

Platelet-derived growth factors (PDGFs) belong to a family of polypeptide growth factors that signal through cell surface tyrosine kinase receptors to stimulate growth, proliferation and differentiation. Platelet-derived growth factor receptors (PDGFRs) are also considered important targets for specific kinase inhibitors in the treatment of several human tumours. The aim of this study was to investigate the role of PDGF-A, PDGF-B, PDGFR-α and PDGFR-ß in canine lymphoma by determining gene and protein expression in lymph nodes of dogs with diffuse large B-cell lymphoma (DLBCL), peripheral T-cell lymphoma (PTCL), T-lymphoblastic lymphoma (T-LBL) and in healthy control dogs. One lymph node was also studied at the end of therapy in a subset of dogs in remission for DLBCL. In controls, PDGF-A, PDGFR-α and PDGFR-ß mRNA levels were significantly higher than in DLBCLs, PTCLs and T-LBLs. However, PDGFR-α and PDGFR-ß were minimally expressed by lymphocytes and plasma cells in normal lymph nodes as determined by immunohistochemistry, while neoplastic B and T cells showed the highest score (P <0.05). This discordant result may be compatible with the constitutive expression of these molecules by endothelial cells and fibroblasts in normal lymph nodes, thereby influencing gene expression results. Furthermore, these cells were not included in the immunohistochemical analysis. Similarly, dogs with DLBCL that were in remission at the end of therapy showed significantly higher gene expression of PDGFs and receptors than at the time of diagnosis and with an opposite trend to the protein assay. PDGF-B protein and mRNA were overexpressed in PTCLs and T-LBLs when compared with DLBCLs and controls (P <0.05). Additionally, there was a correlation between protein expression of PDGF-B and both PDGFRs in PTCLs and T-LBLs, suggesting an autocrine or paracrine loop in the aetiology of aggressive canine T-cell lymphomas. These data provide a rationale for the use of PDGFR antagonists in the therapy of aggressive T-cell lymphomas, but not in DLBCLs.


Asunto(s)
Linfoma/veterinaria , Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Receptores del Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Animales , Enfermedades de los Perros/metabolismo , Enfermedades de los Perros/patología , Perros , Inmunohistoquímica , Linfoma/metabolismo , Linfoma/patología , Factor de Crecimiento Derivado de Plaquetas/análisis , Receptores del Factor de Crecimiento Derivado de Plaquetas/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
5.
J Immunol ; 193(4): 1636-44, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-25024385

RESUMEN

Lymph node (LN) stromal cells (LNSCs) form the functional structure of LNs and play an important role in lymphocyte survival and the maintenance of immune tolerance. Despite their broad spectrum of function, little is known about LNSC responses during microbial infection. In this study, we demonstrate that LNSC subsets display distinct kinetics following vaccinia virus infection. In particular, compared with the expansion of other LNSC subsets and the total LN cell population, the expansion of fibroblastic reticular cells (FRCs) was delayed and sustained by noncirculating progenitor cells. Notably, newly generated FRCs were preferentially located in perivascular areas. Viral clearance in reactive LNs preceded the onset of FRC expansion, raising the possibility that viral infection in LNs may have a negative impact on the differentiation of FRCs. We also found that MHC class II expression was upregulated in all LNSC subsets until day 10 postinfection. Genetic ablation of radioresistant stromal cell-mediated Ag presentation resulted in slower contraction of Ag-specific CD4(+) T cells. We propose that activated LNSCs acquire enhanced Ag-presentation capacity, serving as an extrinsic brake system for CD4(+) T cell responses. Disrupted function and homeostasis of LNSCs may contribute to immune deregulation in the context of chronic viral infection, autoimmunity, and graft-versus-host disease.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Ganglios Linfáticos/citología , Células del Estroma/inmunología , Virus Vaccinia/inmunología , Traslado Adoptivo , Animales , Presentación de Antígeno/inmunología , Antígeno B7-1/biosíntesis , Linfocitos T CD4-Positivos/trasplante , Diferenciación Celular/inmunología , Antígenos de Histocompatibilidad Clase I/biosíntesis , Antígenos de Histocompatibilidad Clase II/biosíntesis , Receptores de Hialuranos/biosíntesis , Tolerancia Inmunológica , Integrina beta1/biosíntesis , Ganglios Linfáticos/virología , Receptor beta de Linfotoxina/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Parabiosis , Receptores del Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Transducción de Señal/inmunología , Vaccinia/inmunología , Molécula 1 de Adhesión Celular Vascular/biosíntesis
6.
Mol Neurobiol ; 49(2): 658-72, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24018979

RESUMEN

Among matrix metalloproteinases (MMPs), MMP-9 has been observed in patients with brain inflammatory diseases and may contribute to the pathology of brain diseases. Thrombin has been known as a regulator of MMP-9 expression and cells migration. However, the mechanisms underlying thrombin-induced MMP-9 expression in rat brain astrocytes (RBA-1 cells) were not completely understood. Here, we demonstrated that thrombin induced the expression of pro-form MMP-9 in RBA-1 cells and cells migration which were attenuated by pretreatment with the inhibitor of receptor tyrosine kinase (Genistein), c-Src (PP1), Jak2 (AG490), PDGFR (AG1296), PI3K (LY294002), Akt (SH-5), PKCs (Ro318220), PKCδ (Rottlerin), or NF-κB (Bay11-7082) and transfection with siRNA of c-Src, PDGFR, Akt, PKCδ, ATF2, p65, IKKα, or IKKß. In addition, thrombin-stimulated c-Src, Jak2, or PDGFR phosphorylation was inhibited by a thrombin inhibitor (PPACK), PP1, AG490, or AG1296. Thrombin further stimulated c-Src and PDGFR complex formation in RBA-1 cells. Thrombin also stimulated Akt and PKCδ phosphorylation and PKCδ translocation which were reduced by PPACK, PP1, AG490, AG1296, or LY294002. We further observed that thrombin markedly stimulated ATF2 or IκBα phosphorylation and NF-κB p65 translocation which were inhibited by Rottlerin or LY294002. Finally, thrombin stimulated in vivo binding of p65 to the MMP-9 promoter, which was reduced by pretreatment with Rottlerin or LY294002. These results concluded that in RBA-1 cells, thrombin activated a c-Src/Jak2/PDGFR/PI3K/Akt/PKCδ pathway, which in turn triggered ATF2 and NF-κB activation and ultimately induced MMP-9 expression associated with cell migration.


Asunto(s)
Astrocitos/metabolismo , Genes src/fisiología , Janus Quinasa 2/biosíntesis , Metaloproteinasa 9 de la Matriz/biosíntesis , Proteína Quinasa C-delta/biosíntesis , Receptores del Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Animales , Astrocitos/efectos de los fármacos , Encéfalo/citología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Células Cultivadas , Inducción Enzimática/efectos de los fármacos , Inducción Enzimática/fisiología , Genes src/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Trombina/farmacología
7.
Sci Signal ; 6(287): ra66, 2013 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-23921085

RESUMEN

The relationship between drug resistance, changes in signaling, and emergence of an invasive phenotype is well appreciated, but the underlying mechanisms are not well understood. Using machine learning analysis applied to the Cancer Cell Line Encyclopedia database, we identified expression of AXL, the gene that encodes the epithelial-to-mesenchymal transition (EMT)-associated receptor tyrosine kinase (RTK) AXL, as exceptionally predictive of lack of response to ErbB family receptor-targeted inhibitors. Activation of EGFR (epidermal growth factor receptor) transactivated AXL, and this ligand-independent AXL activity diversified EGFR-induced signaling into additional downstream pathways beyond those triggered by EGFR alone. AXL-mediated signaling diversification was required for EGF (epidermal growth factor)-elicited motility responses in AXL-positive TNBC (triple-negative breast cancer) cells. Using cross-linking coimmunoprecipitation assays, we determined that AXL associated with EGFR, other ErbB receptor family members, MET (hepatocyte growth factor receptor), and PDGFR (platelet-derived growth factor receptor) but not IGF1R (insulin-like growth factor 1 receptor) or INSR (insulin receptor). From these AXL interaction data, we predicted AXL-mediated signaling synergy for additional RTKs and validated these predictions in cells. This alternative mechanism of receptor activation limits the use of ligand-blocking therapies and indicates against therapy withdrawal after acquired resistance. Further, subadditive interaction between EGFR- and AXL-targeted inhibitors across all AXL-positive TNBC cell lines may indicate that increased abundance of EGFR is principally a means to transactivation-mediated signaling.


Asunto(s)
Neoplasias de la Mama/metabolismo , Receptores ErbB/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal , Antígenos CD/biosíntesis , Antígenos CD/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular/genética , Receptores ErbB/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-met/biosíntesis , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Tirosina Quinasas Receptoras/genética , Receptor IGF Tipo 1/biosíntesis , Receptor IGF Tipo 1/genética , Receptor de Insulina/biosíntesis , Receptor de Insulina/genética , Receptores del Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Receptores del Factor de Crecimiento Derivado de Plaquetas/genética , Activación Transcripcional/genética , Tirosina Quinasa del Receptor Axl
8.
Am J Physiol Regul Integr Comp Physiol ; 304(9): R702-11, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23447134

RESUMEN

The present study investigated transient receptor potential vanilloid type 4 (TRPV4) ion channels in pancreatic stellate cells (PSCs) isolated from rats with high-fat and alcohol diet (HFA)-induced chronic pancreatitis. TRPV4 is a calcium-permeable nonselective ion channel responsive to osmotic changes, alcohol metabolites arachidonic acid, anandamide, their derivatives, and injury-related lipid mediators. Male Lewis rats were fed HFA for 6-8 wk before isolation and primary culture of PSCs. Control PSCs were harvested from rats fed standard chow. Immunoreactivity for cytoskeletal protein activation product α-smooth muscle actin (α-SMA) and platelet-derived growth factor receptor-ß subunit (PDGFR-ß) characterized the cells as PSCs. TRPV4 expression increased in PSCs of HFA-fed rats and control cultures after alcohol treatment (50 mM). Cell responses to activation of inducible TRPV4 were assessed with live cell calcium imaging. Threefold increased and sustained intracellular calcium mobilization responses occurred in 70% of pancreatic stellate cells from HFA-fed rats in response to TRPV4 activators arachidonic acid, lipid second messenger, phorbol ester 4 α-phorbol 12,13-didecanoate (4αPDD), and 50% hypoosmotic media compared with relatively unresponsive PSCs from control rats. Activation responses were attenuated by nonselective TRPV channel blocker ruthenium red. Tumor necrosis factor-α (TNF-α, 1 ng/ml, 16 h) increased responses to 4αPDD in control PSCs. These findings implicate TRPV4-mediated calcium responses inducible after HFA exposure and inflammation in reactive responses of activated PSCs that impair pancreatic function, such as responsiveness to cytokines and the deposition of collagen fibrosis that precipitates ductal blockage and pain.


Asunto(s)
Depresores del Sistema Nervioso Central/toxicidad , Grasas de la Dieta/toxicidad , Etanol/toxicidad , Células Estrelladas Pancreáticas/fisiología , Canales Catiónicos TRPV/biosíntesis , Actinas/metabolismo , Animales , Ácido Araquidónico/farmacología , Western Blotting , Calcio/metabolismo , Separación Celular , Células Cultivadas , Citofotometría , Fibrosis , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Masculino , Células Estrelladas Pancreáticas/efectos de los fármacos , Pancreatitis/genética , Pancreatitis/patología , Ratas , Ratas Endogámicas Lew , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores del Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Canales Catiónicos TRPV/antagonistas & inhibidores , Canales Catiónicos TRPV/genética , Vitamina A/farmacología , Vitaminas/farmacología
9.
Endocrine ; 43(1): 147-53, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22815044

RESUMEN

Autonomously functioning thyroid nodules (AFTN) are known to receive an increased blood influx necessary to sustain their high rate of growth and hormone production. Here, we investigated the expression of hematic and lymphatic vases in a series of 20 AFTN compared with the contralateral non-tumor tissues of the same patients, and the transcript levels of proteins involved in the control of vascular proliferation, including the vascular endothelial growth factor (VEGF) and platelet-derived growth factors (PDGF) and their receptors and the endothelial nitric oxide synthase (eNOS). In parallel, the expression of the differentiation markers sodium/iodide symporter (NIS), thyroperoxidase (TPO), thyroglobulin (Tg), and TSH receptor (TSHR) was also investigated. The data were further analyzed comparing subgroups of tumors with or without mutations in the TSHR gene. Analysis by means of CD31 and D2-40 immunostaining showed in AFTN an increased number of hematic, but not lymphatic, vessels in parallel with an enhanced proliferation rate shown by increased Ki67 staining. Quantitative RT-PCR analysis revealed an increase of VEGF, VEGFR1 and 2, PDGF-A, PDGF-B, and eNOS expression in tumor versus normal tissues. Also, higher transcript levels of NIS, TPO, and Tg were detected. Comparison of the two subgroups of samples revealed only few differences in the expression of the genes examined. In conclusion, these data demonstrate an increased expression of angiogenesis-related factors associated with an enhanced proliferation of hematic, but not lymphatic, vessels in AFTNs. In this context, the presence of TSHR mutations may only slightly influence the expression of pro-angiogenic growth factors.


Asunto(s)
Proteínas Angiogénicas/biosíntesis , Bocio Nodular/metabolismo , Mutación , Neovascularización Patológica/metabolismo , Receptores de Tirotropina/metabolismo , Glándula Tiroides/metabolismo , Tirotoxicosis/metabolismo , Regulación hacia Arriba , Proteínas Angiogénicas/genética , Proteínas Angiogénicas/metabolismo , Biomarcadores/metabolismo , Proliferación Celular , Bocio Nodular/inmunología , Bocio Nodular/patología , Bocio Nodular/fisiopatología , Humanos , Sistema Linfático/inmunología , Sistema Linfático/metabolismo , Sistema Linfático/patología , Microvasos/metabolismo , Microvasos/patología , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neovascularización Patológica/patología , Óxido Nítrico Sintasa de Tipo III/biosíntesis , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Factor de Crecimiento Derivado de Plaquetas/genética , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , ARN Mensajero/metabolismo , Receptores del Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Receptores del Factor de Crecimiento Derivado de Plaquetas/genética , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptores de Tirotropina/deficiencia , Receptores de Tirotropina/genética , Receptores de Tirotropina/inmunología , Receptores de Factores de Crecimiento Endotelial Vascular/biosíntesis , Receptores de Factores de Crecimiento Endotelial Vascular/genética , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Glándula Tiroides/irrigación sanguínea , Glándula Tiroides/inmunología , Glándula Tiroides/patología , Tirotoxicosis/inmunología , Tirotoxicosis/patología , Tirotoxicosis/fisiopatología , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
10.
Stem Cells ; 31(1): 146-55, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23034897

RESUMEN

Tumor tropism of human bone marrow-derived mesenchymal stem cells (MSC) has been exploited for the delivery of therapeutic genes for anticancer therapy. However, the exact contribution of these cells in the tumor microenvironment remains unknown. In this study, we examined the biological effect of MSC on tumor cells. The results showed that MSC inhibited the growth of human glioma cell lines and patient-derived primary glioma cells in vitro. Coadministration of MSC and glioma cells resulted in significant reduction in tumor volume and vascular density, which was not observed when glioma was injected with immortalized normal human astrocytes. Using endothelial progenitor cells (EPC) from healthy donors and HUVEC endothelial cells, the extent of EPC recruitment and capacity to form endothelial tubes was significantly impaired in conditioned media derived from MSC/glioma coculture, suggesting that MSC suppressed tumor angiogenesis through the release of antiangiogenic factors. Further studies using antibody array showed reduced expression of platelet-derived growth factor (PDGF)-BB and interleukin (IL)-1ß in MSC/glioma coculture when compared with controls. In MSC/glioma coculture, PDGF-BB mRNA and the corresponding proteins (soluble and membrane bound forms) as well as the receptors were found to be significantly downregulated when compared with that of glioma cocultured with normal human astrocytes or glioma monoculture. Furthermore, IL-1ß, phosphorylated Akt, and cathepsin B proteins were also reduced in MSC/glioma. Taken together, these data indicated that the antitumor effect of MSC may be mediated through downregulation of PDGF/PDGFR axis, which is known to play a key role in glioma angiogenesis. STEM Cells2013;31:146-155.


Asunto(s)
Glioma/patología , Células Madre Mesenquimatosas/fisiología , Neovascularización Patológica , Proteínas Proto-Oncogénicas c-sis/genética , Receptores del Factor de Crecimiento Derivado de Plaquetas/genética , Animales , Astrocitos , Becaplermina , Células de la Médula Ósea/fisiología , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/terapia , Catepsina B/biosíntesis , Línea Celular Tumoral , Técnicas de Cocultivo , Regulación hacia Abajo , Glioma/terapia , Células Endoteliales de la Vena Umbilical Humana , Humanos , Interleucina-1beta/biosíntesis , Trasplante de Células Madre Mesenquimatosas , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas Proto-Oncogénicas c-akt/biosíntesis , Proteínas Proto-Oncogénicas c-sis/biosíntesis , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores del Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Microambiente Tumoral
11.
Inhal Toxicol ; 24(8): 468-75, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22746397

RESUMEN

Pulmonary artery hypertension (PAH) is a severe disease characterized with progressive increase of pulmonary vascular resistance that finally causes right ventricular failure and premature death. Cigarette smoke (CS) is a major factor of Chronic Obstructive Pulmonary Disease (COPD) that can lead to PAH. However, the mechanism of CS-induced PAH is poorly understood. Mounting evidence supports that pulmonary vascular remodeling play an important role in the development of PAH. PDGF signaling has been demonstrated to be a major mediator of vascular remodeling implicated in PAH. However, the association of PDGF signaling with CS-induced PAH has not been documented. In this study, we investigated CS-induced PAH in rats and the expression of platelet derived growth factor (PDGF) and PDGF receptor (PDGFR) in pulmonary artery. Forty male rats were randomly divided into control group and three experimental groups that were exposed to CS for 1, 2, and 3 months, respectively. CS significantly increased right ventricular systolic pressure (RVSP) and right ventricular hypertrophy index (RVHI). Histology staining demonstrated that CS significantly increased the thickness of pulmonary artery wall and collagen deposition. The expression of PDGF isoform B (PDGF-B) and PDGF receptor beta (PDGFRß) were significantly increased at both protein and mRNA levels in pulmonary artery of rats with CS exposure. Furthermore, Cigarette smoke extract (CSE) significantly increased rat pulmonary artery smooth muscle cell (PASMC) proliferation, which was inhibited by PDGFR inhibitor Imatinib. Thus, our data suggest PDGF signaling is implicated in CS-induced PAH.


Asunto(s)
Hipertensión Pulmonar/etiología , Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Transducción de Señal/efectos de los fármacos , Fumar/efectos adversos , Contaminación por Humo de Tabaco/efectos adversos , Animales , Western Blotting , Proliferación Celular/efectos de los fármacos , Hipertensión Pulmonar Primaria Familiar , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/patología , Hipertrofia Ventricular Derecha/etiología , Hipertrofia Ventricular Derecha/metabolismo , Hipertrofia Ventricular Derecha/patología , Exposición por Inhalación , Pulmón/irrigación sanguínea , Pulmón/efectos de los fármacos , Masculino , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/metabolismo , Arteria Pulmonar/patología , Ratas , Ratas Sprague-Dawley , Receptores del Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Presión Ventricular/efectos de los fármacos
12.
Am J Physiol Lung Cell Mol Physiol ; 302(11): L1159-66, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22447953

RESUMEN

The pathophysiology of congenital diaphragmatic hernia (CDH) is constituted by pulmonary hypoplasia and pulmonary hypertension (PH). We previously reported successful treatment with imatinib of a patient with CDH. This study examines the effect of antenatal imatinib administration on the pulmonary vasculature in a rat model of CDH. Pregnant rats were given nitrofen to induce CDH. Controls were given olive oil. Half of the CDH fetuses and half of the controls were treated with imatinib antenatally E17-E21, rendering four groups: Control, Control+Imatinib, CDH, and CDH+Imatinib. Lung sections were obtained for morphometry and immunohistochemistry, and protein was purified for Western blot. Effects of nitrofen and imatinib on Ki-67, caspase-3, PDGF-B, and PDGF receptors were analyzed. Imatinib significantly reduced medial wall thickness in pulmonary arteries of rats with CDH. It also normalized lumen area and reduced the proportion of fully muscularized arteries. Imatinib also caused medial thinning in the control group. Cell proliferation was increased in CDH, and this proliferation was significantly reduced by imatinib. PDGF-B and PDGFR-ß were upregulated in CDH, and imatinib treatment resulted in a downregulation. PDGFR-α remained unchanged in CDH but was significantly downregulated by imatinib. Antenatal imatinib treatment reduces development of medial wall thickness and restores lumen area in pulmonary arteries in nitrofen-induced CDH. The mechanism is reduced cell proliferation. Imatinib is an interesting candidate for antenatal therapy for PH in CDH, but potential side effects need to be investigated and more specific targeting of PDGF signaling is needed.


Asunto(s)
Hernias Diafragmáticas Congénitas , Pulmón/irrigación sanguínea , Pulmón/patología , Piperazinas/farmacología , Pirimidinas/farmacología , Remodelación de las Vías Aéreas (Respiratorias)/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Benzamidas , Caspasa 3/biosíntesis , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Hernia Diafragmática/inducido químicamente , Hernia Diafragmática/tratamiento farmacológico , Hernia Diafragmática/patología , Hernia Diafragmática/fisiopatología , Mesilato de Imatinib , Antígeno Ki-67/biosíntesis , Pulmón/efectos de los fármacos , Éteres Fenílicos/farmacología , Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Embarazo , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores del Factor de Crecimiento Derivado de Plaquetas/biosíntesis
13.
J Neurooncol ; 106(2): 235-42, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21789698

RESUMEN

Abnormal signaling through the platelet-derived growth factor receptor (PDGFR) has been proposed as a possible mechanism of spinal cord glioma initiation and progression. However, the extent of PDGFR expression in human spinal cord gliomas remains unknown. In this study we perform immunohistochemical analysis of PDGFRα expression in a series of 33 primary intramedullary spinal cord gliomas of different types and grades. PDGFRα was seen to be expressed in a significant subset of these tumors across all major glioma types including ependymoma, oligodendroglioma, pilocytic astrocytoma, astrocytoma, and glioblastoma. These results support the hypothesis that growth factor signaling through the PDGFR may be important for the development of at least a subset of human spinal cord gliomas. Further studies investigating the prognostic significance of PDGFR expression as well as the role of PDGF signaling on the development of intramedullary spinal cord gliomas are warranted.


Asunto(s)
Glioma/metabolismo , Glioma/patología , Receptores del Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Neoplasias de la Médula Espinal/metabolismo , Neoplasias de la Médula Espinal/patología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Niño , Preescolar , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Transducción de Señal/fisiología , Adulto Joven
15.
Oncotarget ; 2(7): 562-8, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21725138

RESUMEN

Better understanding the mechanisms underlying the metastatic process is essential to developing novel targeted therapeutics. Recently, invadopodia have been increasingly recognized as important drivers of local invasion in metastasis. Invadopodia are basally-localized, actin-rich structures that concentrate protease activity to areas of the cell in contact with the extracellular matrix. We recently found that the transcription factor Twist1, a central regulator of the epithelial-mesenchymal transition (EMT), promotes invadopodia formation via upregulation of platelet-derived growth factor receptor (PDGFR) expression and activity. This finding, combined with other investigations into the mechanisms of invadopodia formation, reveal several novel targets for clinical inhibition of invadopodia. Here, we provide an overview of clinically-relevant targets for intervention in invadopodia, including Src signaling, PDGFR signaling, and metalloprotease activity.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Extensiones de la Superficie Celular/efectos de los fármacos , Transición Epitelial-Mesenquimal/fisiología , Terapia Molecular Dirigida , Proteínas Nucleares/metabolismo , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteína 1 Relacionada con Twist/metabolismo , Actinas/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Extensiones de la Superficie Celular/ultraestructura , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Matriz Extracelular/metabolismo , Femenino , Humanos , Metaloproteasas/metabolismo , Ratones , Invasividad Neoplásica , Metástasis de la Neoplasia/prevención & control , Péptido Hidrolasas/metabolismo , Receptores del Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Transducción de Señal/efectos de los fármacos , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/metabolismo
16.
J Pharmacol Exp Ther ; 338(3): 758-66, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21622732

RESUMEN

We recently showed that suramin treatment prevents the onset of renal fibrosis in a model of obstructive nephropathy induced by unilateral ureteral obstruction (UUO). In this study, we further assessed the effect of delayed administration of suramin on the progression of tubulointerstitial fibrosis. Mice were given a single dose of suramin at 20 mg/kg starting at day 3 of obstruction, and kidneys were harvested after an additional 7 or 14 days of obstruction. Suramin completely blocked further increase in expression of type I collagen and fibronectin and largely suppressed expression of α-smooth muscle actin (α-SMA) in both treatment groups. UUO injury induced phosphorylation of Smad-3, a key mediator of transforming growth factor-ß (TGF-ß) signaling, epidermal growth factor receptor, and platelet-derived growth factor receptor after 3 days and further increased at 10 days after UUO injury. When suramin was administered at 3 days after obstruction, phosphorylation of these molecules was not further increased in the obstructed kidney. Suramin treatment also inhibited activation of signal transducer and activator of transcription 3 and extracellular signal-regulated kinase 1 and 2, two signaling pathways associated with renal fibrogenesis. Furthermore, delayed application of suramin suppressed TGF-ß1-induced expression of α-SMA and fibronectin in cultured renal interstitial fibroblasts. These results indicate that administration of suramin is effective in attenuating the progression of renal fibrosis after injury and suggest the potential clinical application of suramin as an antifibrotic treatment in patients with chronic kidney disease.


Asunto(s)
Nefritis Intersticial/tratamiento farmacológico , Suramina/uso terapéutico , Obstrucción Ureteral/tratamiento farmacológico , Actinas/metabolismo , Animales , Western Blotting , Células Cultivadas , Colágeno Tipo I/biosíntesis , Progresión de la Enfermedad , Receptores ErbB/biosíntesis , Receptores ErbB/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibronectinas/biosíntesis , Fibrosis , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Nefritis Intersticial/etiología , Nefritis Intersticial/patología , Fosforilación , Receptores del Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Receptores del Factor de Crecimiento Derivado de Plaquetas/genética , Proteína smad3/metabolismo , Obstrucción Ureteral/complicaciones , Obstrucción Ureteral/patología
17.
Int J Cancer ; 128(9): 2050-62, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21387285

RESUMEN

Recent studies have revealed that platelet-derived growth factor (PDGF) plays a role in promoting progressive tumor growth in several organs; however, whether PDGF plays such a role in gastric carcinoma is undetermined. We examined whether inhibition of PDGF receptor (PDGF-R) tyrosine kinase signaling by imatinib affects tumor growth and metastasis in an orthotopic nude mouse model of human gastric carcinoma. TMK-1 human gastric carcinoma cells were injected into the gastric wall of nude mice. Groups of mice (n = 10 each) received sterile water (control), low-dose imatinib (50 mg/kg/day), high-dose imatinib (200 mg/kg/day), cancer chemotherapeutic agent irinotecan (5 mg/kg/week), or imatinib (50 mg/kg/day or 200 mg/kg/day) and irinotecan (5 mg/kg/week) in combination for 28 days. Tumor growth and metastasis were assessed. Resected tumors were analyzed immunohistochemically. Carcinoma-associated fibroblasts, pericytes and lymphatic endothelial cells in stroma expressed high levels of PDGF-R; carcinoma cells did not. Treatment with imatinib alone did not inhibit tumor growth and metastasis; however, treatment with irinotecan alone or combined with imatinib significantly inhibited tumor growth. Only treatment with high-dose imatinib and irinotecan in combination inhibited lymph node and peritoneal metastases. Immunohistochemically, only imatinib alone or in combination with irinotecan was shown to significantly decrease the stromal reaction, microvessel area and pericyte coverage of tumor microvessels. These effects were marked with high-dose imatinib. In conclusion, administration of PDGF-R tyrosine kinase inhibitor in combination with irinotecan appears to impair the progressive growth of gastric carcinoma by blockade of PDGF-R signaling pathways in stromal cells.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Piperazinas/uso terapéutico , Pirimidinas/uso terapéutico , Neoplasias Gástricas/tratamiento farmacológico , Adenocarcinoma/patología , Animales , Benzamidas , Western Blotting , Camptotecina/análogos & derivados , Camptotecina/uso terapéutico , Proliferación Celular/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Humanos , Mesilato de Imatinib , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Irinotecán , Masculino , Ratones , Ratones Desnudos , Microscopía Confocal , Metástasis de la Neoplasia/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Piperazinas/administración & dosificación , Pirimidinas/administración & dosificación , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Receptores del Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias Gástricas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
18.
J Biol Chem ; 286(12): 10593-604, 2011 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-21209081

RESUMEN

The developing and mature central nervous system contains neural precursor cells expressing the proteoglycan NG2. Some of these cells continuously differentiate to myelin-forming oligodendrocytes; knowledge of the destiny of NG2(+) precursors would benefit from the characterization of new key functional players. In this respect, the G protein-coupled membrane receptor GPR17 has recently emerged as a new timer of oligodendrogliogenesis. Here, we used purified oligodendrocyte precursor cells (OPCs) to fully define the immunophenotype of the GPR17-expressing cells during OPC differentiation, unveil its native signaling pathway, and assess the functional consequences of GPR17 activation by its putative endogenous ligands, uracil nucleotides and cysteinyl leukotrienes (cysLTs). GPR17 presence was restricted to very early differentiation stages and completely segregated from that of mature myelin. Specifically, GPR17 decorated two subsets of slowly proliferating NG2(+) OPCs: (i) morphologically immature cells expressing other early proteins like Olig2 and PDGF receptor-α, and (ii) ramified preoligodendrocytes already expressing more mature factors, like O4 and O1. Thus, GPR17 is a new marker of these transition stages. In OPCs, GPR17 activation by either uracil nucleotides or cysLTs resulted in potent inhibition of intracellular cAMP formation. This effect was counteracted by GPR17 antagonists and receptor silencing with siRNAs. Finally, uracil nucleotides promoted and GPR17 inhibition, by either antagonists or siRNAs, impaired the normal program of OPC differentiation. These data have implications for the in vivo behavior of NG2(+) OPCs and point to uracil nucleotides and cysLTs as main extrinsic local regulators of these cells under physiological conditions and during myelin repair.


Asunto(s)
Diferenciación Celular/fisiología , Proliferación Celular , Regulación de la Expresión Génica/fisiología , Oligodendroglía/metabolismo , Receptores Acoplados a Proteínas G/biosíntesis , Células Madre/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Silenciador del Gen , Vaina de Mielina/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Factor de Transcripción 2 de los Oligodendrocitos , Oligodendroglía/citología , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores del Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Células Madre/citología , Nucleótidos de Uracilo/metabolismo , Nucleótidos de Uracilo/farmacología
19.
J Clin Neurosci ; 17(12): 1557-62, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20822908

RESUMEN

The aim of this study was to investigate the expression of platelet-derived growth factor (PDGF) ligands A and B and receptors α and ß in cerebral arteriovenous and cavernous malformations. Fifteen arteriovenous malformation (AVM) and 15 cerebral cavernous malformation (CCM) tissue samples were immunostained for PDGF ligands A and B, PDGF receptors (PDGFR) α and ß, and vascular endothelial growth factor. Tissues were compared in terms of expression levels within various vascular layers, and the results were confirmed using western blotting. AVM had higher levels of PDGF-A expression than CCM (p = 0.004, 0.009, 0.001, and 0.027, for endothelium, media, adventitia, and perilesional tissue, respectively) and western blotting showed that there was higher expression of PDGFR-α in AVM tissues. In contrast, CCM endothelium, media, and adventitia had higher PDGF-B expression compared with AVM (p = 0.007, 0.001, and 0.039, respectively). PDGFR-ß expression was also significantly higher in the endothelium of CCM tissue (p = 0.007). Overexpression of PDGF ligands and receptors in AVM and CCM may mean that therapeutic strategies targeting the PDGF pathway could be useful in the treatment of these two malformations.


Asunto(s)
Neoplasias del Sistema Nervioso Central/metabolismo , Hemangioma Cavernoso del Sistema Nervioso Central/metabolismo , Malformaciones Arteriovenosas Intracraneales/metabolismo , Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Proteínas Proto-Oncogénicas c-sis/biosíntesis , Receptores del Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Western Blotting , Humanos , Inmunohistoquímica , Ligandos
20.
J Orthop Res ; 28(8): 1107-12, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20186834

RESUMEN

Tissue engineering approaches that harness the stimulatory power of platelet-rich plasma have produced encouraging results in anterior cruciate ligament (ACL) repair. However, a number of recent studies have demonstrated age-dependent differences in cellular responses to such an approach. Identifying the reasons for these differences would allow counteracting them and consequently improve outcomes. In this study we hypothesized that these age-related effects are caused by differences in the expression of the receptors for growth factors released from platelet-rich plasma (PRP). Porcine ACL fibroblasts from a predetermined number of animals of different ages were obtained, and mRNA levels of the receptors of platelet-derived growth factor (PDGF), transforming growth factor beta (TGF-beta), fibroblast growth factor (FGF), and vascular endothelial growth factor (VEGF) were determined. Expression levels were compared across age groups (young and adolescent) and regressed on age in days. While no significant difference was seen across groups, the regression analysis showed decreases in receptor expression with increasing age. These differences were statistically significant for TGF-beta receptor 1, FGF receptor, and VEGF receptor 2; and borderline significant for TGF-beta receptor 3 and PDGF receptor. The only receptor that was not associated with age was VEGF receptor 1, a regulator of VEGF receptor 2. These findings suggest that the decrease in growth factor receptor expression as a likely reason for reduced PRP action with increasing age.


Asunto(s)
Ligamento Cruzado Anterior/citología , Receptores de Factores de Crecimiento de Fibroblastos/biosíntesis , Receptores del Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Receptores de Factores de Crecimiento Transformadores beta/biosíntesis , Receptores de Factores de Crecimiento Endotelial Vascular/biosíntesis , Envejecimiento , Animales , Ligamento Cruzado Anterior/metabolismo , Femenino , Plasma Rico en Plaquetas/metabolismo , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...