Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 471
Filtrar
1.
J Mol Endocrinol ; 66(3): 233-243, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33640869

RESUMEN

Hemolysis, elevated liver enzymes, and low platelet count (HELLP) syndrome is a life-threatening pregnancy complication. Though there are several medications widely used to treat HELLP syndrome, delivery is the only efficient treatment. The goal of the present study was to investigate the effects of platelet-derived growth factor-D (PDGF-D), a newly identified PDGF, in a rat model of HELLP syndrome which was accomplished by sFlt-1 and sEng injection. The expression levels of PDGF-D in pregnant women diagnosed with HELLP syndrome was determined. A HELLP rat model was established and the PDGF-D expression level in the plasma and the placenta tissue was evaluated. To evaluate the effects of PDGF-D in HELLP syndrome model, siPDGF-D was injected into the rats and the HELLP syndrome-related parameters were measured. The levels of inflammatory cytokines and PDGF-D were determined by ELISA. The oxidative stress activities in the plasma were also determined. Furthermore, the expression of PDGF-D/PDGFR-ß/nuclear factor κB (NF-κB) p65 in placenta tissues was evaluated by Western blotting. Compared to the normal pregnant (NP) group, the levels of PDGF-D were augmented regardless of species. Knockdown of PDGF-D can result in the alleviation of HELLP syndrome development and progression in the HELLP rat model. Importantly, as a result of PDGF-D knockdown, the serum levels of inflammatory cytokines and oxidative stress activities were modulated, and the phosphorylation of PDGFR-ß and NF-κB p65 in placenta tissue was inhibited. Taking together, our findings indicate that targeting PDGF-D could be used as a novel strategy to treat patients with HELLP syndrome.


Asunto(s)
Síndrome HELLP/metabolismo , Linfocinas/antagonistas & inhibidores , Linfocinas/metabolismo , FN-kappa B/metabolismo , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Adulto , Animales , Modelos Animales de Enfermedad , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Inflamación/patología , Estrés Oxidativo , Fosforilación , Embarazo , Ratas Sprague-Dawley , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Adulto Joven
2.
Exp Eye Res ; 204: 108446, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33476605

RESUMEN

Neovascular age-related macular degeneration (neoAMD) is the leading cause of blindness in AMD and manifests as choroidal neovascularization (CNV). Anti-vascular endothelial growth factor (VEGF) therapies are the mainstay treatments but with limited efficacy and cause detrimental effects on the retina after long-term application. These disadvantages warrant alternative strategy. Herein, we examined the effect on CNV by intravitreal injection of bortezomib, a reversible proteasome inhibitor, and further dissected the mechanism. Krypton red Laser was used to create CNV model in mice. The angiogenesis volume was assessed in choroidal flat-mount with isolectin GS-IB4 labeling and the leakage was examined with fluorescein fundus angiography. Injection of Borsub inhibited angiogenesis in the CNV model which was dose-dependent; the injection significantly inhibited leakage as well. Furthermore, Borsub injection reduced the contents of VEGF-A, macrophage chemotactic factor 1 (MCP-1), and platelet-derived growth factor (PDGF)-D but not PDGF-B, examined by enzyme-linked immunosorbent assay, in choroid/retinal pigment epithelium (RPE) tissue. These injections also reduced phospho-VEGFR-2 and phospho-PDGFRß in choroid/RPE tissue examined by immunoblotting. Moreover, Borsub inhibited the recruitment of mural cells or macrophages to laser-injured spots. Injection of Borsub indicated negative effect on scotopic and photopic responses recorded by electroretinogram. Altogether, intravitreal injection of Borsub significantly reduced CNV by antagonizing VEGF-A/Flk-1 and PDGF-D/PDGFRß pathways without impacting electroretinography parameters. Thus, Borsub may offer an invaluable therapy for the prevention and treatment of neoAMD.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Antineoplásicos/uso terapéutico , Bortezomib/uso terapéutico , Neovascularización Coroidal/tratamiento farmacológico , Modelos Animales de Enfermedad , Linfocinas/antagonistas & inhibidores , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Western Blotting , Quimiocina CCL2/antagonistas & inhibidores , Quimiocina CCL2/metabolismo , Neovascularización Coroidal/metabolismo , Neovascularización Coroidal/fisiopatología , Reposicionamiento de Medicamentos , Electrorretinografía/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Angiografía con Fluoresceína , Etiquetado Corte-Fin in Situ , Inyecciones Intravítreas , Linfocinas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo
3.
Eur J Pharmacol ; 895: 173868, 2021 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-33460613

RESUMEN

Complex of platelet-derived growth factor (PDGF) isoforms and PDGF receptors have important functions in the regulation of growth and survival of various cell types. Herein, it was found that aberrant PDGFC expression is closely associated with survival rates in triple-negative breast cancer (TNBC) patients. In addition, PDGFC expression was identified to be significantly increased in TNBC cells unlike other subtypes such as PDGFA, PDGFB, and PDGFD. Apparently, the effects of specific PDGF receptor (PDGFR) inhibitors such as sunitinib and ponatinib on HCC1806 and Hs578T TNBC cells were investigated. Both inhibitors decreased cell viability in a dose-dependent manner. In addition, the inhibitors completely inhibited cell growth in both the cell lines and decreased the expression of matrix metalloproteinase-1 (MMP-1), one of the metastasis-related genes. Cell migration was also decreased by the inhibitors. Finally, the combined effects of the inhibitors with doxorubicin (DOX) were investigated. The results showed that the combination of two PDGFR inhibitors with DOX inhibited the growth of cells and enhanced the apoptotic cell death more uniformly than DOX. Consequently, it is demonstrated that PDGFR inhibitors, sunitinib and ponatinib hold the potential for effective treatment of TNBC.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Doxorrubicina/farmacología , Imidazoles/farmacología , Linfocinas/antagonistas & inhibidores , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Piridazinas/farmacología , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Sunitinib/farmacología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Linfocinas/genética , Linfocinas/metabolismo , Metaloproteinasa 1 de la Matriz/metabolismo , Factor de Crecimiento Derivado de Plaquetas/genética , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptores del Factor de Crecimiento Derivado de Plaquetas/genética , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología
4.
Sci Rep ; 10(1): 22383, 2020 12 24.
Artículo en Inglés | MEDLINE | ID: mdl-33361796

RESUMEN

Disruption of blood-brain barrier (BBB) integrity is a feature of various neurological disorders. Here we found that the BBB is differently affected during the preclinical, progression and remission phase of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). We have identified an upregulation of pro-inflammatory and pro-angiogenic factors in the BBB transcriptome and down-regulation of endothelial tight junction members coinciding with elevated BBB leakage specifically during the progression phase. These changes were antagonized by blocking PDGFRα signaling with the small tyrosine kinase inhibitor imatinib. Moreover, targeting the PDGFRα ligand PDGF-CC using a neutralizing antibody, facilitated recovery of BBB integrity and improvement of EAE symptoms. Intracerebroventricular injection of PDGF-CC induced upregulation, whereas blocking PDGF-CC during EAE led to downregulation of Tnfa and Il1a at the BBB. Our findings suggest that blocking PDGF-CC counteracts fundamental aspects of endothelial cell activation and disruption of the BBB by decreasing Tnfa and Il1a expression. We also demonstrate that both PDGF-CC and its receptor PDGFRα were upregulated in MS lesions indicating that blocking PDGF-CC may be considered a novel treatment for MS.


Asunto(s)
Anticuerpos Neutralizantes/farmacología , Barrera Hematoencefálica/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Linfocinas/antagonistas & inhibidores , Esclerosis Múltiple/inmunología , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Animales , Barrera Hematoencefálica/patología , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/inmunología , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/patología , Inflamación/tratamiento farmacológico , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Interleucina-1alfa/genética , Interleucina-1alfa/inmunología , Linfocinas/genética , Linfocinas/inmunología , Ratones , Ratones Transgénicos , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/genética , Esclerosis Múltiple/patología , Factor de Crecimiento Derivado de Plaquetas/genética , Factor de Crecimiento Derivado de Plaquetas/inmunología , Transducción de Señal/genética , Transducción de Señal/inmunología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
5.
Kidney Int ; 95(5): 1103-1119, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30827511

RESUMEN

Platelet-derived growth factors (PDGF) have been implicated in kidney disease progression. We previously found that PDGF-C is upregulated at sites of renal fibrosis and that antagonism of PDGF-C reduces fibrosis in the unilateral ureteral obstruction model. We studied the role of PDGF-C in collagen 4A3-/- ("Alport") mice, a model of progressive renal fibrosis with greater relevance to human kidney disease. Alport mice were crossbred with PDGF-C-/- mice or administered a neutralizing PDGF-C antibody. Both PDGF-C deficiency and neutralization reduced serum creatinine and blood urea nitrogen levels and mitigated glomerular injury, renal fibrosis, and renal inflammation. Unexpectedly, systolic blood pressure was also reduced in both Alport and wild-type mice treated with a neutralizing PDGF-C antibody. Neutralization of PDGF-C reduced arterial wall thickness in the renal cortex of Alport mice. Aortic rings isolated from anti-PDGF-C-treated wildtype mice exhibited reduced tension and faster relaxation than those of untreated mice. In vitro, PDGF-C upregulated angiotensinogen in aortic tissue and in primary hepatocytes and induced nuclear factor κB (NFκB)/p65-binding to the angiotensinogen promoter in hepatocytes. Neutralization of PDGF-C suppressed transcript expression of angiotensinogen in Alport mice and angiotensin II receptor type 1 in Alport and wildtype mice. Finally, administration of neutralizing PDGF-C antibodies ameliorated angiotensin II-induced hypertension in healthy mice. Thus, in addition to its key role in mediating renal fibrosis, we identified PDGF-C as a mediator of hypertension via effects on renal vasculature and on the renin-angiotensin system. The contribution to both renal fibrosis and hypertension render PDGF-C an attractive target in progressive kidney disease.


Asunto(s)
Hipertensión/patología , Riñón/patología , Linfocinas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Angiotensinógeno/metabolismo , Animales , Presión Sanguínea/genética , Células Cultivadas , Colágeno Tipo IV/genética , Modelos Animales de Enfermedad , Fibrosis , Hepatocitos , Humanos , Hipertensión/etiología , Hipertensión/genética , Linfocinas/antagonistas & inhibidores , Masculino , Ratones , Ratones Noqueados , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Cultivo Primario de Células , Regulación hacia Arriba , Uréter/cirugía
6.
PLoS One ; 13(7): e0201089, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30052660

RESUMEN

PDGF-CC is a member of the platelet-derived growth factor (PDGF) family that stimulates PDGFRα phosphorylation and thereby activates intracellular signalling events essential for development but also in cancer, fibrosis and neuropathologies involving blood-brain barrier (BBB) disruption. In order to elucidate the biological and pathological role(s) of PDGF-CC signalling, we have generated high affinity neutralizing monoclonal antibodies (mAbs) recognizing human PDGF-CC. We determined the complementarity determining regions (CDRs) of the selected clones, and mapped the binding epitope for clone 6B3. Using the monoclonal 6B3, we determined the expression pattern for PDGF-CC in different human primary tumours and control tissues, and explored its ability to neutralize PDGF-CC-induced phosphorylation of PDGFRα. In addition, we showed that PDGF-CC induced disruption of the blood-retinal barrier (BRB) was significantly reduced upon intraperitoneal administration of a chimeric anti-PDGF-CC antibody. In summary, we report on high affinity monoclonal antibodies against PDGF-CC that have therapeutic efficacy in vivo.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Linfocinas/antagonistas & inhibidores , Linfocinas/inmunología , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Factor de Crecimiento Derivado de Plaquetas/inmunología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Células A549 , Animales , Barrera Hematorretinal/efectos de los fármacos , Barrera Hematorretinal/metabolismo , Barrera Hematorretinal/patología , Permeabilidad Capilar , Expresión Génica/efectos de los fármacos , Humanos , Inmunohistoquímica , Ratones Endogámicos C57BL , Ratones Transgénicos , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Recombinantes/inmunología , Transducción de Señal/efectos de los fármacos
7.
PLoS One ; 13(7): e0200649, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30021009

RESUMEN

Platelet-derived growth factor CC (PDGF-CC) is important during foetal development but also in pathogenesis of neurologic diseases, cancer and fibrosis. We have previously demonstrated that blocking the PDGF-CC/PDGF receptor alpha (PDGFRα) axis resulted in reduction of stroke volume and cerebrovascular permeability after experimentally induced stroke. Recently, we could translate these findings into the clinic showing that imatinib, a small tyrosine kinase inhibitor targeting PDGF receptors, can significantly improve neurological outcome after ischemic stroke in human. Herein we report preclinical toxicological analyses of our newly generated monoclonal anti-human PDGF-CC antibody 6B3 (mAb 6B3) in PDGF-CC humanized mice. Beside histological organ assessment, we also analysed serum, urine, haematological parameters and the general health status of the treated mice. We could not find any indications that mAb 6B3 is toxic or has other significant side effects neither in short, nor in long treatment regimens. Our results indicate that mAb 6B3 can be further developed for clinical use. This opens up the possibility to assess the therapeutic potential of blocking PDGF-CC in diverse pathological conditions such as neurologic diseases, cancer and fibrosis.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/farmacología , Anticuerpos Neutralizantes/farmacología , Linfocinas/antagonistas & inhibidores , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales de Origen Murino/efectos adversos , Anticuerpos Monoclonales de Origen Murino/inmunología , Anticuerpos Neutralizantes/administración & dosificación , Anticuerpos Neutralizantes/inmunología , Evaluación Preclínica de Medicamentos , Humanos , Linfocinas/inmunología , Ratones , Ratones Transgénicos , Factor de Crecimiento Derivado de Plaquetas/inmunología
8.
Diabetes ; 67(8): 1549-1560, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29794241

RESUMEN

Obesity increases the risk of vascular diseases, including aortic aneurysm (AA). Perivascular adipose tissue (PVAT) surrounding arteries are altered during obesity. However, the underlying mechanism of adipose tissue, especially PVAT, in the pathogenesis of AA is still unclear. Here we showed that angiotensin II (AngII) infusion increases the incidence of AA in leptin-deficient obese mice (ob/ob) and high-fat diet-induced obese mice with adventitial inflammation. Furthermore, transcriptome analysis revealed that platelet-derived growth factor-D (PDGF-D) was highly expressed in the PVAT of ob/ob mice. Therefore, we hypothesized that PDGF-D mediates adventitial inflammation, which provides a direct link between PVAT dysfunction and AA formation in AngII-infused obese mice. We found that PDGF-D promotes the proliferation, migration, and inflammatory factors expression in cultured adventitial fibroblasts. In addition, the inhibition of PDGF-D function significantly reduced the incidence of AA in AngII-infused obese mice. More importantly, adipocyte-specific PDGF-D transgenic mice are more susceptible to AA formation after AngII infusion accompanied by exaggerated adventitial inflammatory and fibrotic responses. Collectively, our findings reveal a notable role of PDGF-D in the AA formation during obesity, and modulation of this cytokine might be an exploitable treatment strategy for the condition.


Asunto(s)
Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/etiología , Grasa Intraabdominal/metabolismo , Linfocinas/metabolismo , Obesidad/fisiopatología , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Adventicia/efectos de los fármacos , Adventicia/inmunología , Adventicia/metabolismo , Adventicia/patología , Angiotensina II/administración & dosificación , Angiotensina II/efectos adversos , Animales , Aorta Abdominal/diagnóstico por imagen , Aorta Abdominal/efectos de los fármacos , Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/diagnóstico por imagen , Aneurisma de la Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/patología , Bencimidazoles/farmacología , Células Cultivadas , Dieta Alta en Grasa/efectos adversos , Implantes de Medicamentos , Regulación de la Expresión Génica/efectos de los fármacos , Mediadores de Inflamación/metabolismo , Grasa Intraabdominal/efectos de los fármacos , Grasa Intraabdominal/inmunología , Grasa Intraabdominal/patología , Linfocinas/agonistas , Linfocinas/antagonistas & inhibidores , Linfocinas/genética , Masculino , Ratones , Ratones Mutantes , Ratones Transgénicos , Obesidad/etiología , Obesidad/metabolismo , Obesidad/patología , Especificidad de Órganos , Factor de Crecimiento Derivado de Plaquetas/agonistas , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Factor de Crecimiento Derivado de Plaquetas/genética , Quinolinas/farmacología , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Grasa Subcutánea Abdominal/efectos de los fármacos , Grasa Subcutánea Abdominal/inmunología , Grasa Subcutánea Abdominal/metabolismo , Grasa Subcutánea Abdominal/patología , Análisis de Supervivencia
9.
Biochem Biophys Res Commun ; 478(2): 845-51, 2016 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-27507215

RESUMEN

Platelet-derived growth factor D (PDGF-D) signaling plays significant roles during the development and progression of human malignancies via interacting with the receptor of PDGF-D (PDGFR). Meanwhile, the majority of human tumor metastasis is closely associated with epithelial-mesenchymal transition (EMT). However, the underlying mechanism between PDGF-D/PDGFR signaling and EMT which involved in tumor metastasis remain dismal. This study aimed to investigate the role of PDGF-D signaling during EMT process of tongue squamous cell carcinoma (TSCC). In our study, the expression of PDGF-D and PDGFR were examined in primary TSCC samples and the expression of PDGF-D was also determined in TSCC cell lines. In addition, the correlation between PDGF-D expression and TSCC aggressive histopathological features was analyzed. Our results implied that upregulation of PDGFRß in UM1 cells induced with exogenous PDGF-D can remarkably promote tumor cells invasiveness; conversely, when using small interfering RNA (siRNA), the invasiveness can be severely prohibited. Furthermore, PDGF-D downstream signal molecules p38, AKT, ERK and EMT biomarkers (E-cadherin, N-cadherin, Vimentin and snail) were measured using Western blot. Our results showed that PDGF-D can induce p38, AKT and ERK phosphorylation; downregulate epithelial markers and upregulate mesenchymal markers. On the contrary, PDGFRß siRNA significantly prohibited p38, AKT and ERK phosphorylation; inhibited EMT process. Function analysis revealed that PDGFRß siRNA obviously interfered with UM1 cell migration and invasion, according to transwell and wound healing assay. In conclusion, this study suggested that EMT process can be triggered by the PDGF-D/PDGFRß axis in TSCC, and then involved in the tumor cell invasion via activation of p38/AKT/ERK/EMT pathway.


Asunto(s)
Carcinoma de Células Escamosas/genética , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Proteínas Proto-Oncogénicas c-akt/genética , Neoplasias de la Lengua/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Antígenos CD/genética , Antígenos CD/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Movimiento Celular , Progresión de la Enfermedad , Humanos , Linfocinas/antagonistas & inhibidores , Linfocinas/genética , Linfocinas/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Invasividad Neoplásica , Fosforilación , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Factor de Crecimiento Derivado de Plaquetas/genética , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal , Factores de Transcripción de la Familia Snail/genética , Factores de Transcripción de la Familia Snail/metabolismo , Neoplasias de la Lengua/metabolismo , Neoplasias de la Lengua/patología , Vimentina/genética , Vimentina/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
10.
Biochem Biophys Res Commun ; 457(3): 307-13, 2015 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-25576870

RESUMEN

Platelet-derived growth factor-D (PDGF-D) is a more recent recognized growth factor involved in the regulation of several cellular processes, including cell proliferation, transformation, invasion, and angiogenesis by binding to and activating its cognate receptor PDGFR-ß. After bile duct ligation or in the carbon tetrachloride-induced hepatic fibrosis model, PDGF-D showed upregulation comparable to PDGF-B. Moreover, adenoviral PDGF-D gene transfer induced hepatic stellate cell proliferation and liver fibrosis. We here investigated the molecular mechanism of PDGF-D involvement in liver fibrogenesis. Therefore, the GRX mouse cell line was stimulated with PDGF-D and evaluated for fibrotic markers and PDGF-D signaling pathways in comparison to the other PDGF isoforms. We found that PDGF-D failed to enhance Col I and α-smooth muscle actin (α-SMA) production but has capacity to upregulate expression of the tissue inhibitor of metalloprotease 1 (TIMP-1) resulting in attenuation of MMP-2 and MMP-9 gelatinase activity as indicated by gelatinase zymography. This phenomenon was restored through application of a PDGF-D neutralizing antibody. Unexpectedly, PDGF-D incubation decreased both PDGFR-α and -ß in mRNA and protein levels, and PDGF-D phosphorylated typrosines specific for PDGFR-α and -ß. We conclude that PDGF-D intensifies fibrogenesis by interfering with the fibrolytic activity of the TIMP-1/MMP system and that PDGF-D signaling is mediated through both PDGF-α and -ß receptors.


Asunto(s)
Linfocinas/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Inhibidor Tisular de Metaloproteinasa-1/biosíntesis , Animales , Anticuerpos Neutralizantes , Becaplermina , Línea Celular , Colágeno Tipo I/genética , Regulación hacia Abajo , Matriz Extracelular/metabolismo , Fibrinólisis , Homeostasis , Cirrosis Hepática/etiología , Cirrosis Hepática/genética , Cirrosis Hepática/metabolismo , Linfocinas/antagonistas & inhibidores , Linfocinas/inmunología , Sistema de Señalización de MAP Quinasas , Ratones , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Factor de Crecimiento Derivado de Plaquetas/inmunología , Proteínas Proto-Oncogénicas c-sis/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal
11.
J Vasc Surg ; 59(5): 1402-9.e1-4, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-23856609

RESUMEN

BACKGROUND: Platelet-derived growth factor C (PDGF-C) has been reported to promote angiogenesis independently of vascular endothelial growth factor (VEGF), although its significance in postnatal angiogenesis in vivo remains poorly understood. VEGF has been employed as a major molecular tool to induce therapeutic angiogenesis. However, VEGF therapy is not very effective in models of cardiovascular diseases associated with diabetes, and the mechanisms of this phenomenon still remain to be elucidated. METHODS: We used a murine model of hind limb ischemia and of streptozotocin-induced diabetes. RESULTS: Expression of PDGF-C and its receptor PDGFR-α were markedly upregulated in ischemic limbs. Treatment with a neutralizing antibody against PDGF-C significantly impaired blood flow recovery and neovascularization after ischemia almost to the same extent as a VEGF-neutralizing antibody. Mice deficient in PDGF-C exhibited reduced blood flow recovery after ischemia compared with wild-type mice, confirming a strong proangiogenic activity of PDGF-C. Next, we injected an expression vector encoding PDGF-C into ischemic limbs. Blood flow recovery and neovascularization after ischemia were significantly improved in the groups treated with PDGF-C compared with controls. Attenuation of angiogenic responses to ischemia has been reported in patients with diabetes even after VEGF treatment, although a precise mechanism remains unknown. We hypothesized that PDGF-C might relate to the impaired angiogenesis of diabetes. We tested this hypothesis by inducing diabetes by intraperitoneal injection of streptozotocin. Expression levels of PDGF-C at baseline and after ischemia were significantly lower in limb tissues of diabetic mice than in those of control mice, whereas expression levels of other members of the PDGF family and VEGF were not changed or were even higher in diabetic mice. Introduction of VEGF complementary DNA expression plasmid vector into ischemic limbs did not improve blood flow recovery. However, these changes were effectively reversed by additional introduction of the PDGF-C complementary DNA plasmid vector. CONCLUSIONS: These results indicate that downregulation of PDGF-C expression in limb tissues of diabetic mice contributes to impaired angiogenesis and suggest that introduction of PDGF-C might be a novel strategy for therapeutic angiogenesis, especially in the diabetic state. CLINICAL RELEVANCE: Angiogenesis and arteriogenesis after ischemia are attenuated in most diabetic patients, although the precise mechanisms remain unclear. Platelet-derived growth factors (PDGFs) have a variety of functions on many cell types, and PDGF-C stimulates angiogenesis and revascularizes ischemic tissues. This study indicates the role for PDGF-C as a critical regulator of impaired angiogenesis of diabetes and suggests that PDGF-C might be a novel target for the treatment of ischemic cardiovascular diseases in diabetes.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Angiopatías Diabéticas/metabolismo , Isquemia/metabolismo , Linfocinas/metabolismo , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Animales , Anticuerpos Neutralizantes/administración & dosificación , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/fisiopatología , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/fisiopatología , Angiopatías Diabéticas/genética , Angiopatías Diabéticas/fisiopatología , Angiopatías Diabéticas/terapia , Técnicas de Transferencia de Gen , Miembro Posterior , Isquemia/genética , Isquemia/fisiopatología , Isquemia/terapia , Linfocinas/antagonistas & inhibidores , Linfocinas/deficiencia , Linfocinas/genética , Linfocinas/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuropilina-1/metabolismo , Neuropilina-2/metabolismo , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Factor de Crecimiento Derivado de Plaquetas/deficiencia , Factor de Crecimiento Derivado de Plaquetas/genética , Factor de Crecimiento Derivado de Plaquetas/inmunología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Recuperación de la Función , Flujo Sanguíneo Regional , Transducción de Señal , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/inmunología , Factor A de Crecimiento Endotelial Vascular/metabolismo
12.
Oncol Rep ; 30(6): 2887-96, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24126726

RESUMEN

The molecular mechanisms responsible for the elevated metastatic potential of malignant melanoma are still not fully understood. In order to shed light on the molecules involved in the acquisition by melanoma of a highly aggressive phenotype, we compared the gene expression profiles of two cell clones derived from the human cutaneous metastatic melanoma cell line M14: a highly invasive clone (M14C2/MK18) and a clone (M14C2/C4) with low ability to invade the extracellular matrix (ECM). The highly invasive phenotype of M14C2/MK18 cells was correlated with overexpression of neuropilin-1, activation of a vascular endothelial growth factor (VEGF)-A/VEGFR-2 autocrine loop and secretion of matrix metalloprotease-2. Moreover, in an in vivo murine model, M14C2/MK18 cells displayed a higher growth rate as compared with M14C2/C4 cells, even though in vitro both clones possessed comparable proliferative potential. Microarray analysis in M14C2/MK18 cells showed a strong upregulation of platelet-derived growth factor (PDGF)-C, a cytokine that contributes to angiogenesis, and downregulation of calpain-3, a calcium-dependent thiol-protease that regulates specific signalling cascade components. Inhibition of PDGF-C with a specific antibody resulted in a significant decrease in ECM invasion by M14C2/MK18 cells, confirming the involvement of PDGF-C in melanoma cell invasiveness. Moreover, the PDGF-C transcript was found to be upregulated in a high percentage of human melanoma cell lines (17/20), whereas only low PDGF-C levels were detected in a few melanocytic cultures (2/6). By contrast, inhibition of calpain-3 activity in M14C2/C4 control cells, using a specific chemical inhibitor, markedly increased ECM invasion, strongly suggesting that downregulation of calpain-3 plays a role in the acquisition of a highly invasive phenotype. The results indicate that PDGF-C upregulation and calpain-3 downregulation are involved in the aggressiveness of malignant melanoma and suggest that modulators of these proteins or their downstream effectors may synergise with VEGF­A therapies in combating tumour-associated angiogenesis and melanoma spread.


Asunto(s)
Calpaína/genética , Linfocinas/genética , Melanoma/genética , Proteínas Musculares/genética , Invasividad Neoplásica/genética , Factor de Crecimiento Derivado de Plaquetas/genética , Animales , Calpaína/biosíntesis , Línea Celular Tumoral , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Linfocinas/antagonistas & inhibidores , Linfocinas/biosíntesis , Metaloproteinasa 2 de la Matriz/genética , Melanoma/patología , Ratones , Proteínas Musculares/biosíntesis , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Transducción de Señal/genética , Factor A de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Am J Pathol ; 182(1): 107-17, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23141925

RESUMEN

Platelet-derived growth factors (PDGF) are key mediators of organ fibrosis. We investigated whether PDGF-C(-/-) mice or mice treated with neutralizing PDGF-C antibodies are protected from bile duct ligation-induced liver fibrosis, and we compared the effects with those of PDGF-C deficiency or neutralization on kidney fibrosis induced by unilateral ureteral obstruction. Unexpectedly, and in contrast to kidney fibrosis, PDGF-C deficiency or antagonism did not protect from liver fibrosis or functional liver impairment. Furthermore, the hepatic infiltration of monocytes/macrophages/dendritic cells and chemokine mRNA expression (CC chemokine ligand [CCL]5, CCL2, and CC chemokine receptor 2 [CCR2]) remained unchanged. Transcript expression of PDGF ligands increased in both liver and kidney fibrosis and was not affected by neutralization of PDGF-C. In kidney fibrosis, PDGF-C deficiency or antagonism led to reduced expression and signaling of PDGF-receptor (R)-α- and PDGFR-ß-chains. In contrast, in liver fibrosis there was either no difference (PDGF-C(-/-) mice) or even an upregulation of PDGFR-ß and signaling (anti-PDGF-C group). Finally, in vitro studies in portal myofibroblasts pointed to a predominant role of PDGF-B and PDGF-D signaling in liver fibrosis. In conclusion, our study revealed significant differences between kidney and liver fibrosis in that PDGF-C mediates kidney fibrosis, whereas antagonism of PDGF-C in liver fibrosis appears to be counteracted by significant upregulation and increased PDGFR-ß signaling. PDGF-C antagonism, therefore, may not be effective to treat liver fibrosis.


Asunto(s)
Riñón/patología , Cirrosis Hepática/metabolismo , Linfocinas/fisiología , Factor de Crecimiento Derivado de Plaquetas/fisiología , Receptores del Factor de Crecimiento Derivado de Plaquetas/fisiología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Fibrosis , Cirrosis Hepática/etiología , Cirrosis Hepática/prevención & control , Linfocinas/antagonistas & inhibidores , Linfocinas/deficiencia , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miofibroblastos/metabolismo , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Factor de Crecimiento Derivado de Plaquetas/deficiencia , Ratas , Ratas Sprague-Dawley , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal/fisiología , Regulación hacia Arriba/fisiología , Obstrucción Ureteral/complicaciones
14.
Clin Exp Metastasis ; 29(5): 409-17, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22362252

RESUMEN

In a nude mouse model of colorectal liver metastases, we have identified a paracrine tumor cell/host cell signalling pathway that is apparently required for successful tumor growth. Whereas recombinant platelet derived growth factor-C (PDGF-C) and supernatants from PDGF-C secreting wild type LS174T colon carcinoma cells could rescue tumor promoting hepatic stellate cells (HSC) from growth inhibition by serum starvation, supernatants from LS174T colon carcinoma cells with reduced secretion of PDGF-C had much less effect on serum starved HSC. Autocrine growth inhibition of LS174T cells by PDGF-C knock-down was only marginal. In vivo, a prominent inhibition of liver metastasis was observed if PDGF-C was knocked-down in LS174T cells. By whole genome array analysis of host cells of the invasion front and subsequent immunohistochemical staining we identified p21 activated kinase-2 (PAK-2) as being strongly and specifically expressed by HSC. The above described effect of PDGF-C on HSC was found to be dependent on PAK-2 because in contrast to wild type HSC, silencing of PAK-2 in HSC only allowed for a partial PDGF-C-mediated rescue from serum starvation leading to only a slight increase of proliferation. These data indicate that PDGF-C promotes tumor growth via a growth promoting effect on HSC that is at least in part dependent on the presence of functional PAK-2.


Asunto(s)
Adenocarcinoma/secundario , Neoplasias del Colon/patología , Células Estrelladas Hepáticas/patología , Neoplasias Hepáticas/secundario , Linfocinas/metabolismo , Comunicación Paracrina , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Quinasas p21 Activadas/metabolismo , Adenocarcinoma/metabolismo , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Western Blotting , Proliferación Celular , Neoplasias del Colon/metabolismo , Femenino , Perfilación de la Expresión Génica , Células Estrelladas Hepáticas/metabolismo , Humanos , Técnicas para Inmunoenzimas , Neoplasias Hepáticas/metabolismo , Linfocinas/antagonistas & inhibidores , Linfocinas/genética , Ratones , Ratones Desnudos , Análisis de Secuencia por Matrices de Oligonucleótidos , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Factor de Crecimiento Derivado de Plaquetas/genética , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Células Tumorales Cultivadas , Quinasas p21 Activadas/antagonistas & inhibidores , Quinasas p21 Activadas/genética
15.
Prostate ; 72(12): 1328-38, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22213159

RESUMEN

BACKGROUND: The major cause of death in prostate cancer (PCa) cases is due to distant metastatic lesions, with the bone being the most prevalent site for secondary colonization. Utilization of small molecule inhibitors to treat bone metastatic PCa have had limited success either as monotherapies or in combination with other chemotherapeutics due to intolerable toxicities. In the current study, we developed a clinically relevant in vivo intraosseous tumor model overexpressing the platelet-derived growth factor D (PDGF D) to test the efficacy of a newly characterized vascular endothelial growth factor receptor (VEGFR)/PDGFR inhibitor, cediranib (also called AZD2171). METHODS: An intratibial-injection model was established utilizing DU145 cells with or without increased PDGF D expression. Tumor-bearing mice were treated by daily gavage administration of cediranib and/or weekly i.p. injection of docetaxel for 7 weeks. Tibiae were monitored by in vivo/ex vivo X-rays and histomorphometry analysis was performed to estimate tumor volume and tumor-associated trabecular bone growth. RESULTS: Cediranib reduced intraosseous growth of prostate tumors as well as tumor-associated bone responses. When compared to the standard chemotherapeutic agent docetaxel, cediranib exhibited a stronger inhibition of tumor-associated bone response. The efficacy of cediranib was further enhanced when the drug was co-administered with docetaxel. Importantly, the therapeutic benefits of cediranib and docetaxel are more prominent in intraosseous prostate tumors overexpressing PDGF D. CONCLUSION: These novel findings support the utilization of cediranib, either alone or in combination with docetaxel, to treat bone metastatic PCa exhibiting PDGF D expression.


Asunto(s)
Neoplasias Óseas/tratamiento farmacológico , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/patología , Inhibidores de Crecimiento/uso terapéutico , Linfocinas/biosíntesis , Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Quinazolinas/uso terapéutico , Animales , Antineoplásicos/uso terapéutico , Neoplasias Óseas/metabolismo , Neoplasias Óseas/secundario , Línea Celular Tumoral , Transformación Celular Neoplásica/metabolismo , Humanos , Linfocinas/antagonistas & inhibidores , Masculino , Ratones , Ratones SCID , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Neoplasias de la Próstata/metabolismo , Distribución Aleatoria , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
16.
Biochem J ; 441(3): 909-18, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22035541

RESUMEN

The PDGF (platelet-derived growth factor) family members are potent mitogens for cells of mesenchymal origin and serve as important regulators of cell migration, survival, apoptosis and transformation. Tumour-derived PDGF ligands are thought to function in both autocrine and paracrine manners, activating receptors on tumour and surrounding stromal cells. PDGF-C and -D are secreted as latent dimers, unlike PDGF-A and -B. Cleavage of the CUB domain from the PDGF-C and -D dimers is required for their biological activity. At present, little is known about the proteolytic processing of PDGF-C, the rate-limiting step in the regulation of PDGF-C activity. In the present study we show that the breast carcinoma cell line MCF7, engineered to overexpress PDGF-C, produces proteases capable of cleaving PDGF-C to its active form. Increased PDGF-C expression enhances cell proliferation, anchorage-independent cell growth and tumour cell motility by autocrine signalling. In addition, MCF7-produced PDGF-C induces fibroblast cell migration in a paracrine manner. Interestingly, PDGF-C enhances tumour cell invasion in the presence of fibroblasts, suggesting a role for tumour-derived PDGF-C in tumour-stromal interactions. In the present study, we identify tPA (tissue plasminogen activator) and matriptase as major proteases for processing of PDGF-C in MCF7 cells. In in vitro studies, we also show that uPA (urokinase-type plasminogen activator) is able to process PDGF-C. Furthermore, by site-directed mutagenesis, we identify the cleavage site for these proteases in PDGF-C. Lastly, we provide evidence suggesting a two-step proteolytic processing of PDGF-C involving creation of a hemidimer, followed by GFD-D (growth factor domain dimer) generation.


Asunto(s)
Neoplasias de la Mama/patología , Carcinoma/patología , Linfocinas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Serina Proteasas/fisiología , Animales , Comunicación Autocrina/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Carcinoma/genética , Carcinoma/metabolismo , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Humanos , Linfocinas/antagonistas & inhibidores , Linfocinas/genética , Linfocinas/fisiología , Ratones , Células 3T3 NIH , Comunicación Paracrina/genética , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Factor de Crecimiento Derivado de Plaquetas/genética , Factor de Crecimiento Derivado de Plaquetas/fisiología , Multimerización de Proteína/genética , Multimerización de Proteína/fisiología , Procesamiento Proteico-Postraduccional , Serina Proteasas/genética , Serina Proteasas/metabolismo , Transfección
17.
Int J Cancer ; 131(5): E603-13, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22095574

RESUMEN

Recent studies have revealed that the maturation state of vessels in tumors, in addition to vascularity, is a critical determinant of tumor growth. The role of oxygen-dependent signaling pathways in hypoxia-stimulated angiogenesis is well established, however, little is known about their impact on vessel maturation in tumors. Here, we have studied the function of the cellular oxygen sensor, factor inhibiting HIF-1 (FIH), which controls the activity of hypoxia-inducible factor-1. FIH silencing in mouse LM8 osteosarcoma stimulated angiogenesis but did not influence tumor growth. In contrast, FIH overexpression led to increased pericyte coverage of the tumor vasculature, reduced vessel leakiness and enhanced tumor growth. Vessel maturation was paralleled by up-regulation of platelet-derived growth factor (PDGF)-C in tumors and expression of PDGF receptor-α on pericytes. Ablation of PDGF-C in FIH-overexpressing tumor cells reduced pericyte coverage and tumor growth. Our data suggest that FIH-mediated PDGF-C induction in LM8 osteosarcoma stimulates the recruitment of PDGFR-α positive pericytes to the tumor vasculature, leading to vessel maturation and enhanced tumor growth.


Asunto(s)
Neoplasias Óseas/irrigación sanguínea , Neoplasias Óseas/patología , Proliferación Celular , Linfocinas/metabolismo , Oxigenasas de Función Mixta/metabolismo , Osteosarcoma/irrigación sanguínea , Osteosarcoma/patología , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Inductores de la Angiogénesis , Animales , Apoptosis , Western Blotting , Neoplasias Óseas/metabolismo , Femenino , Factor 1 Inducible por Hipoxia/genética , Factor 1 Inducible por Hipoxia/metabolismo , Técnicas para Inmunoenzimas , Linfocinas/antagonistas & inhibidores , Linfocinas/genética , Ratones , Ratones Endogámicos C3H , Oxigenasas de Función Mixta/genética , Neovascularización Patológica , Osteosarcoma/metabolismo , Pericitos/metabolismo , Pericitos/patología , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Factor de Crecimiento Derivado de Plaquetas/genética , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Células Tumorales Cultivadas
18.
Breast Cancer Res Treat ; 126(1): 15-25, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20379844

RESUMEN

Platelet-derived growth factor-D (PDGF-D) has been linked with several human malignancies; however, its role in breast cancer progression is not known. We found that PDGF-D expressing breast cancer cell lines MDA-MB-231 and SUM-149 are more invasive compared to cell lines with little or no expression of PDGF-D such as MDA-MB-468 and MCF-7 cells. Over-expression of PDGF-D in PDGF-D low expressing MDA-MB-468 and MCF-7 cells by cDNA transfection showed increased cell proliferation while silencing the expression of PDGF-D by siRNA in PDGF-D high expressing MDA-MB-231 and SUM-149 cells showed decreased cell proliferation and increased apoptosis. Moreover, PDGF-D over-expression was positively correlated with the expression of Notch-1 and Jagged-1, and the expression of mesenchymal markers (Vimentin and ZEB-2) with concomitant decreased expression of epithelial marker E-cadherin. Since NF-κB activation plays a crucial role in Notch signaling as well as in epithelial-mesenchymal transition and tumor aggressiveness, we determined the DNA binding activity of NF-κB and our findings are consistent showing that PDGF-D over-expression led to increased DNA binding activity of NF-κB while it was found to be decreased by inactivation of PDGF-D. These results were also consistent with the expression and activity of MMP-9 and VEGF, as well as invasive characteristics. Further, forced expression of Notch-1/Jagged-1 by cDNA transfection de-repressed the effects of PDGF-D silencing on NF-κB activity and invasion. From these results, we conclude that PDGF-D plays an important role in breast tumor aggressiveness and this process is mechanistically linked with the activation of Notch and NF-κB signaling.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Linfocinas/metabolismo , FN-kappa B/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor Notch1/metabolismo , Transducción de Señal , Apoptosis , Western Blotting , Neoplasias de la Mama/genética , Proteínas de Unión al Calcio , Adhesión Celular , Movimiento Celular , Proliferación Celular , Ensayo de Unidades Formadoras de Colonias , Ensayo de Cambio de Movilidad Electroforética , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular , Proteína Jagged-1 , Luciferasas/metabolismo , Linfocinas/antagonistas & inhibidores , Linfocinas/genética , Proteínas de la Membrana , FN-kappa B/genética , Invasividad Neoplásica , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Factor de Crecimiento Derivado de Plaquetas/genética , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Receptor Notch1/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Serrate-Jagged , Células Tumorales Cultivadas
19.
J Exp Med ; 207(4): 867-80, 2010 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-20231377

RESUMEN

Platelet-derived growth factor CC (PDGF-CC) is the third member of the PDGF family discovered after more than two decades of studies on the original members of the family, PDGF-AA and PDGF-BB. The biological function of PDGF-CC remains largely to be explored. We report a novel finding that PDGF-CC is a potent neuroprotective factor that acts by modulating glycogen synthase kinase 3beta (GSK3beta) activity. In several different animal models of neuronal injury, such as axotomy-induced neuronal death, neurotoxin-induced neuronal injury, 6-hydroxydopamine-induced Parkinson's dopaminergic neuronal death, and ischemia-induced stroke, PDGF-CC protein or gene delivery protected different types of neurons from apoptosis in both the retina and brain. On the other hand, loss-of-function assays using PDGF-C null mice, neutralizing antibody, or short hairpin RNA showed that PDGF-CC deficiency/inhibition exacerbated neuronal death in different neuronal tissues in vivo. Mechanistically, we revealed that the neuroprotective effect of PDGF-CC was achieved by regulating GSK3beta phosphorylation and expression. Our data demonstrate that PDGF-CC is critically required for neuronal survival and may potentially be used to treat neurodegenerative diseases. Inhibition of the PDGF-CC-PDGF receptor pathway for different clinical purposes should be conducted with caution to preserve normal neuronal functions.


Asunto(s)
Apoptosis/fisiología , Encéfalo/citología , Supervivencia Celular/fisiología , Glucógeno Sintasa Quinasa 3/metabolismo , Linfocinas/fisiología , Neuronas/citología , Factor de Crecimiento Derivado de Plaquetas/fisiología , Retina/citología , Animales , Anticuerpos/inmunología , Anticuerpos/farmacología , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/genética , Encéfalo/irrigación sanguínea , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Isquemia Encefálica/patología , Isquemia Encefálica/fisiopatología , Isquemia Encefálica/prevención & control , Permeabilidad Capilar/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Corteza Cerebral/irrigación sanguínea , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/patología , Femenino , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , Perfilación de la Expresión Génica , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Peróxido de Hidrógeno/farmacología , Linfocinas/antagonistas & inhibidores , Linfocinas/farmacología , Linfocinas/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , N-Metilaspartato/farmacología , Neovascularización Patológica/inducido químicamente , Factores de Crecimiento Nervioso/genética , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Traumatismos del Nervio Óptico/tratamiento farmacológico , Traumatismos del Nervio Óptico/patología , Enfermedad de Parkinson Secundaria/inducido químicamente , Enfermedad de Parkinson Secundaria/tratamiento farmacológico , Enfermedad de Parkinson Secundaria/metabolismo , Enfermedad de Parkinson Secundaria/patología , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Factor de Crecimiento Derivado de Plaquetas/farmacología , Factor de Crecimiento Derivado de Plaquetas/uso terapéutico , Ratas , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/inmunología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética
20.
Curr Drug Targets ; 10(1): 38-41, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19149534

RESUMEN

Platelet-derived growth factor-D (PDGF-D) is a newly recognized growth factor that can regulate many cellular processes, including cell proliferation, transformation, invasion, and angiogenesis by specifically binding to and activating its cognate receptor PDGFR-beta. The functions of PDGF-D in human cancer progression are largely unknown. We discuss here the role of PDGF-D signaling pathway in cancer and how its deregulation is involved in tumor development and progression to metastatic disease.


Asunto(s)
Antineoplásicos/uso terapéutico , Linfocinas/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Humanos , Linfocinas/biosíntesis , Linfocinas/fisiología , Metástasis de la Neoplasia , Neoplasias/metabolismo , Neoplasias/patología , Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Factor de Crecimiento Derivado de Plaquetas/fisiología , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Receptores del Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Receptores del Factor de Crecimiento Derivado de Plaquetas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...