Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 172(3): 534-548.e19, 2018 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-29275861

RESUMEN

Many tumors produce platelet-derived growth factor (PDGF)-DD, which promotes cellular proliferation, epithelial-mesenchymal transition, stromal reaction, and angiogenesis through autocrine and paracrine PDGFRß signaling. By screening a secretome library, we found that the human immunoreceptor NKp44, encoded by NCR2 and expressed on natural killer (NK) cells and innate lymphoid cells, recognizes PDGF-DD. PDGF-DD engagement of NKp44 triggered NK cell secretion of interferon gamma (IFN)-γ and tumor necrosis factor alpha (TNF-α) that induced tumor cell growth arrest. A distinctive transcriptional signature of PDGF-DD-induced cytokines and the downregulation of tumor cell-cycle genes correlated with NCR2 expression and greater survival in glioblastoma. NKp44 expression in mouse NK cells controlled the dissemination of tumors expressing PDGF-DD more effectively than control mice, an effect enhanced by blockade of the inhibitory receptor CD96 or CpG-oligonucleotide treatment. Thus, while cancer cell production of PDGF-DD supports tumor growth and stromal reaction, it concomitantly activates innate immune responses to tumor expansion.


Asunto(s)
Neoplasias Encefálicas/inmunología , Puntos de Control del Ciclo Celular , Glioblastoma/inmunología , Células Asesinas Naturales/inmunología , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Animales , Neoplasias Encefálicas/patología , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Femenino , Glioblastoma/patología , Humanos , Inmunidad Innata , Interferón gamma/metabolismo , Células MCF-7 , Masculino , Ratones , Ratones Endogámicos C57BL , Receptor 2 Gatillante de la Citotoxidad Natural/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
2.
Cell Mol Life Sci ; 81(1): 225, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38769116

RESUMEN

Ischemic stroke induces neovascularization of the injured tissue as an attempt to promote structural repair and neurological recovery. Angiogenesis is regulated by pericytes that potently react to ischemic stroke stressors, ranging from death to dysfunction. Platelet-derived growth factor (PDGF) receptor (PDGFR)ß controls pericyte survival, migration, and interaction with brain endothelial cells. PDGF-D a specific ligand of PDGFRß is expressed in the brain, yet its regulation and role in ischemic stroke pathobiology remains unexplored. Using experimental ischemic stroke mouse model, we found that PDGF-D is transiently induced in brain endothelial cells at the injury site in the subacute phase. To investigate the biological significance of PDGF-D post-ischemic stroke regulation, its subacute expression was either downregulated using siRNA or upregulated using an active recombinant form. Attenuation of PDGF-D subacute induction exacerbates neuronal loss, impairs microvascular density, alters vascular permeability, and increases microvascular stalling. Increasing PDGF-D subacute bioavailability rescues neuronal survival and improves neurological recovery. PDGF-D subacute enhanced bioavailability promotes stable neovascularization of the injured tissue and improves brain perfusion. Notably, PDGF-D enhanced bioavailability improves pericyte association with brain endothelial cells. Cell-based assays using human brain pericyte and brain endothelial cells exposed to ischemia-like conditions were applied to investigate the underlying mechanisms. PDGF-D stimulation attenuates pericyte loss and fibrotic transition, while increasing the secretion of pro-angiogenic and vascular protective factors. Moreover, PDGF-D stimulates pericyte migration required for optimal endothelial coverage and promotes angiogenesis. Our study unravels new insights into PDGF-D contribution to neurovascular protection after ischemic stroke by rescuing the functions of pericytes.


Asunto(s)
Células Endoteliales , Accidente Cerebrovascular Isquémico , Linfocinas , Pericitos , Factor de Crecimiento Derivado de Plaquetas , Pericitos/metabolismo , Pericitos/patología , Animales , Accidente Cerebrovascular Isquémico/metabolismo , Accidente Cerebrovascular Isquémico/patología , Ratones , Linfocinas/metabolismo , Linfocinas/genética , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Humanos , Células Endoteliales/metabolismo , Masculino , Ratones Endogámicos C57BL , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Neovascularización Fisiológica , Movimiento Celular
3.
Proc Natl Acad Sci U S A ; 119(3)2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-35027451

RESUMEN

The axis of platelet-derived growth factor (PDGF) and PDGF receptor-beta (PDGFRß) plays prominent roles in cell growth and motility. In addition, PDGF-D enhances human natural killer (NK) cell effector functions when binding to the NKp44 receptor. Here, we report an additional but previously unknown role of PDGF-D, whereby it mediates interleukin-15 (IL-15)-induced human NK cell survival but not effector functions via its binding to PDGFRß but independent of its binding to NKp44. Resting NK cells express no PDGFRß and only a low level of PDGF-D, but both are significantly up-regulated by IL-15, via the nuclear factor κB signaling pathway, to promote cell survival in an autocrine manner. Both ectopic and IL-15-induced expression of PDGFRß improves NK cell survival in response to treatment with PDGF-D. Our results suggest that the PDGF-D-PDGFRß signaling pathway is a mechanism by which IL-15 selectively regulates the survival of human NK cells without modulating their effector functions.


Asunto(s)
Interleucina-15/metabolismo , Células Asesinas Naturales/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal/fisiología , Animales , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Humanos , Linfocinas , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Receptor 2 Gatillante de la Citotoxidad Natural , Factor de Crecimiento Derivado de Plaquetas/farmacología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética
4.
J Biol Chem ; 298(6): 101981, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35472332

RESUMEN

Mesenchymal stem cells (MSCs) are adult stem cell populations and exhibit great potential in regenerative medicine and oncology. Platelet-derived growth factors (PDGFs) are well known to regulate MSC biology through their chemotactic and mitogenic properties. However, their direct roles in the regulation of MSC lineage commitment are unclear. Here, we show that PDGF D promotes the differentiation of human bone marrow mesenchymal stem cells (hBMSCs) into osteoblasts and inhibits hBMSC differentiation into adipocytes. We demonstrate that PDGF D-induced ß-actin expression and polymerization are essential for mediating this differential regulation of osteoblastogenesis and adipogenesis. Interestingly, we found that PDGF D induces massive upward molecular weight shifts of its cognate receptor, PDGF receptor beta (ß-PDGFR) in hBMSCs, which was not observed in fibroblasts. Proteomic analysis indicated that the E3 ubiquitin ligase HECT, UBA, and WWE domain-containing protein 1 (HUWE1) associates with the PDGF D-activated ß-PDGFR signaling complex in hBMSCs, resulting in ß-PDGFR polyubiquitination. In contrast to the well-known role of ubiquitin in protein degradation, we provide evidence that HUWE1-mediated ß-PDGFR polyubiquitination delays ß-PDGFR internalization and degradation, thereby prolonging AKT signaling. Finally, we demonstrate that HUWE1-regulated ß-PDGFR signaling is essential for osteoblastic differentiation of hBMSCs, while being dispensable for PDGF D-induced hBMSC migration and proliferation as well as PDGF D-mediated inhibition of hBMSC differentiation into adipocytes. Taken together, our findings provide novel insights into the molecular mechanism by which PDGF D regulates the commitment of hBMSCs into the osteoblastic lineage.


Asunto(s)
Linfocinas/metabolismo , Células Madre Mesenquimatosas , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Ubiquitina-Proteína Ligasas , Diferenciación Celular , Proliferación Celular , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Proteómica , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
5.
J Gene Med ; 25(2): e3465, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36413571

RESUMEN

BACKGROUND: Platelet derived growth factors (PDGF)-D and the expression of its receptor increase in neoplastic progression of cancer. Co-silencing of growth factor and receptor can be suggested as an important strategy for effective cancer therapy. In the present study, we hypothesized that suppression of PDGF-D signaling pathway with small interfering RNAs (siRNAs) targeting both PDGF-D and PDGF receptor (PDGFR)-ß is a promising strategy for anticancer therapy. METHODS: Chitosan nanoplexes containing dual and single siRNA were prepared at different weight ratios and controlled by gel retardation assay. Characterization, cellular uptake, gene silencing and invasion studies were performed. The effect of nanoplexes on breast tumor growth, PDGF expression and apoptosis was investigated. RESULTS: We have shown that downregulation of PDGF-D and PDGFR-ß with chitosan/siRNA nanoplex formulations reduced proliferation and invasion in breast cancer cells. In the in vivo breast tumor model, it was determined that the intratumoral administration of chitosan/siPDGF-D/siPDGFR-ß nanoplexes markedly decreased the tumor volume and PDGF-D and PDGFR-ß mRNA and protein expression levels and increased apoptosis. CONCLUSIONS: According to the results obtained, we evaluated the effect of PDGF-D and PDGFR-ß on breast tumor development and showed that RNAi-mediated inhibition of this pathway formulated with chitosan nanoplexes can be considered as a new breast cancer therapy strategy.


Asunto(s)
Neoplasias de la Mama , Quitosano , ARN Interferente Pequeño , Femenino , Humanos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Quitosano/uso terapéutico , Nanoestructuras/uso terapéutico , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/uso terapéutico
6.
J Cell Biochem ; 120(5): 6920-6925, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30652340

RESUMEN

Platelet-derived growth factor-D (PDGF-D) can enhance invasion and metastasis in several human malignancies. Although several studies have been performed to investigate the association between clinicopathological characteristics and prognosis in epithelial ovarian cancer (EOC), the mediation effect of PDGF-D on above-mentioned association have been seldom assessed. In this study, we detected the PDGF-D expression from the tissues of patients with EOC and further collected clinicopathological characteristics and prognostic information to identify whether PDGF-D mediated the effect of differentiated degree on prognosis in patients with EOC. A total of 190 paraffin-embedded tissue samples from patients with EOC between July 2005 and December 2010 were collected. We performed a Kaplan-Meier analysis for the association between differentiated degree and prognosis followed by a causal mediation analysis. The analysis results indicated that differentiated degree was associated with prognosis and PDGF-D mediated the effect of differentiated degree on prognosis in patients with EOC, which might be a potential target for ovarian cancer treatment.

7.
J Cell Sci ; 130(8): 1365-1378, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28254885

RESUMEN

Platelet-derived growth factor (PDGF)-D is a PDGF receptor ß (PDGFRß)-specific ligand implicated in a number of pathological conditions, such as cardiovascular disease and cancer, but its biological function remains incompletely understood. In this study, we demonstrate that PDGF-D binds directly to neuropilin 1 (NRP1), in a manner that requires the PDGF-D C-terminal Arg residue. Stimulation with PDGF-D, but not PDGF-B, induced PDGFRß-NRP1 complex formation in fibroblasts. Additionally, PDGF-D induced translocation of NRP1 to cell-cell junctions in endothelial cells, independently of PDGFRß, altering the availability of NRP1 for VEGF-A-VEGFR2 signaling. PDGF-D showed differential effects on pericyte behavior in ex vivo sprouting assays compared to PDGF-B. Furthermore, PDGF-D-induced PDGFRß-NRP1 interaction can occur in trans between molecules located in different cells (endothelial cells and pericytes). In summary, we show that NRP1 can act as a co-receptor for PDGF-D-PDGFRß signaling and is possibly implicated in intercellular communication in the vascular wall.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Endotelio Vascular/metabolismo , Fibroblastos/metabolismo , Uniones Intercelulares/metabolismo , Neoplasias/metabolismo , Neuropilina-1/metabolismo , Pericitos/metabolismo , Animales , Línea Celular Transformada , Humanos , Linfocinas/metabolismo , Neovascularización Fisiológica , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Unión Proteica , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Porcinos
8.
Cancer Invest ; 37(2): 99-112, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30836770

RESUMEN

Platelet-derived growth factor D (PDGF-D) has been shown to mediate cellular processes of importance in cancer progression. This study aimed to investigate the expression and putative involvement of PDGF-D signaling in colorectal carcinogenesis. PDGF-D was expressed in vascular endothelial cells in tumor and normal tissues. PDGF-D stimulation of cells altered genes of importance in carcinogenic processes. In addition, PDGF-D increased the proliferation rate while imatinib inhibited these effects. PDGF-D and its PDGF receptor beta (PDGFR-ß) are expressed in colorectal cancer and blockage of PDGF-D/PDGFR-ß signaling using tyrosine kinase inhibitors, such as imatinib, might be important in inhibiting tumor-promoting actions.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Linfocinas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal/fisiología , Células CACO-2 , Carcinogénesis/efectos de los fármacos , Carcinogénesis/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Neoplasias Colorrectales/tratamiento farmacológico , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células HT29 , Humanos , Mesilato de Imatinib/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos
9.
Am J Physiol Cell Physiol ; 310(4): C293-304, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26157007

RESUMEN

Activation of ß-platelet-derived growth factor receptor (ß-PDGFR) is associated with prostate cancer (PCa) progression and recurrence after prostatectomy. Analysis of the ß-PDGFR ligands in PCa revealed association between PDGF-D expression and Gleason score as well as tumor stage. During the course of studying the functional consequences of PDGF ligand-specific ß-PDGFR signaling in PCa, we discovered a novel function of PDGF-D for activation/shedding of the serine protease matriptase leading to cell invasion, migration, and tumorigenesis. The present study showed that PDGF-D, not PDGF-B, induces extracellular acidification, which correlates with increased matriptase activation. A cDNA microarray analysis revealed that PDGF-D/ß-PDGFR signaling upregulates expression of the acidosis regulator carbonic anhydrase IX (CAIX), a classic target of the transcriptional factor hypoxia-inducible factor-1α (HIF-1α). Cellular fractionation displayed a strong HIF-1α nuclear localization in PDGF-D-expressing cells. Treatment of vector control or PDGF-B-expressing cells with the HIF-1α activator CoCl2 led to increased CAIX expression accompanied by extracellular acidosis and matriptase activation. Furthermore, the analysis of the CAFTD cell lines, variants of the BPH-1 transformation model, showed that increased PDGF-D expression is associated with enhanced HIF-1α activity, CAIX induction, cellular acidosis, and matriptase shedding. Importantly, shRNA-mediated knockdown of CAIX expression effectively reversed extracellular acidosis and matriptase activation in PDGF-D-transfected BPH-1 cells and in CAFTD variants that express endogenous PDGF-D at a high level. Taken together, these novel findings reveal a new paradigm in matriptase activation involving PDGF-D-specific signal transduction leading to extracellular acidosis.


Asunto(s)
Linfocinas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Neoplasias de la Próstata/enzimología , Serina Endopeptidasas/metabolismo , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Comunicación Autocrina , Anhidrasa Carbónica IX , Anhidrasas Carbónicas/genética , Anhidrasas Carbónicas/metabolismo , Línea Celular Tumoral , Activación Enzimática , Regulación de la Expresión Génica , Humanos , Concentración de Iones de Hidrógeno , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Linfocinas/genética , Masculino , Factor de Crecimiento Derivado de Plaquetas/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Interferencia de ARN , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Serina Endopeptidasas/genética , Transducción de Señal , Factores de Tiempo , Transfección
10.
J Cell Biochem ; 117(3): 599-611, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26252425

RESUMEN

The effect of targeted expression of an anabolic isoform of basic fibroblast growth factor (FGF2) in osteoblastic lineage on tibial fracture healing was assessed in mice. Closed fracture of the tibiae was performed in Col3.6-18 kDaFgf2-IRES-GFPsaph mice in which a 3.6 kb fragment of type I collagen promoter (Col3.6) drives the expression of only the 18 kD isoform of FGF2 (18 kDaFgf2/LMW) with green fluorescent protein-sapphire (GFPsaph) as well as Vector mice (Col3.6-IRES-GFPsaph, Vector) that did not harbor the FGF2 transgene. Radiographic, micro-CT, DEXA, and histologic analysis of fracture healing of tibiae harvested at 3, 10 and 20 days showed a smaller fracture callus but accelerated fracture healing in LMWTg compared with Vector mice. At post fracture day 3, FGF receptor 3 and Sox 9 mRNA were significantly increased in LMWTg compared with Vector. Accelerated fracture healing was associated with higher FGF receptor 1, platelet derived growth factors B, C, and D, type X collagen, vascular endothelial cell growth factor, matrix metalloproteinase 9, tartrate resistant acid phosphatase, cathepsin K, runt-related transcription factor-2, Osterix and Osteocalcin and lower Sox9, and type II collagen expression at 10 days post fracture. We postulate that overexpression of LMW FGF2 accelerated the fracture healing process due to its effects on factors that are important in chondrocyte and osteoblast differentiation and vascular invasion.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/biosíntesis , Curación de Fractura , Tibia/fisiopatología , Animales , Catepsina K/genética , Catepsina K/metabolismo , Colágeno Tipo II/metabolismo , Femenino , Factor 2 de Crecimiento de Fibroblastos/genética , Expresión Génica , Masculino , Ratones Transgénicos , Factor de Crecimiento Derivado de Plaquetas/genética , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Factor de Transcripción SOX9/metabolismo , Fosfatasa Ácida Tartratorresistente/metabolismo , Tibia/diagnóstico por imagen , Tibia/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
11.
Prostate ; 76(6): 534-42, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26732854

RESUMEN

PURPOSE: To determine the functional relationship between androgen receptor (AR) and PDGF D as it relates to the radiation response of PTEN-null prostate cancer (PCa) cells and the effect of enzalutamide on these interactions. METHODS AND MATERIALS: Using murine PTEN-null prostate epithelial cell line and human prostate carcinoma LNCaP (PTEN-mutant) models, nuclear and cytosolic AR levels were determined by immunoblot analysis and the transcriptional activity of nuclear AR was assessed by RT-PCR analysis of its target genes with or without irradiation. Cell survival was evaluated by clonogenic assay or sulforhodamine B (SRB) assay upon irradiation in the absence or presence of the AR antagonist enzalutamide. RESULTS: PTEN loss resulted in upregulation of AR expression in a PDGF-D dependent manner and irradiation selectively increased the nuclear AR protein level and its activity in a murine cell model. When the functional significance of AR in cell survival was tested, treatment with enzalutamide resulted in radiosensitization of human LNCaP cells. Similarly to the murine model, PDGF-D overexpression increased the nuclear AR level and its transcriptional activity in LNCaP cells. PDGF-D over-expression was associated with radioresistance and enzalutamide treatment effectively reversed PDGF-D-mediated radioresistance in LNCaP cells. CONCLUSIONS: We have demonstrated that AR, a target of the PTEN and PDGF D-downstream signaling program, contributes to radiation resistance in human PCa cells. In addition, this study suggests that anti-androgens such as enzalutamide may serve as radiation sensitizers for the treatment of PCa patients, particularly so in patients with loss of PTEN or overexpression of PDGF-D.


Asunto(s)
Linfocinas/metabolismo , Feniltiohidantoína/análogos & derivados , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Neoplasias de la Próstata , Receptores Androgénicos/metabolismo , Transducción de Señal , Animales , Antineoplásicos/farmacología , Benzamidas , Línea Celular Tumoral , Humanos , Masculino , Ratones , Nitrilos , Fosfohidrolasa PTEN/metabolismo , Feniltiohidantoína/farmacología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación , Proteínas Supresoras de Tumor/metabolismo
12.
BMC Med Genet ; 17(1): 62, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27585990

RESUMEN

BACKGROUND: Platelet-derived growth factor (PDGF) D has been reported to be active in fibroblasts, and in areas of myocardial infarction. In this longitudinal study we evaluated the association between PDGF-D polymorphism and cardiovascular mortality, and attempted to discover whether specific genotype differences regarding risk could be observed, and if gender differences could be seen. METHODS: Four hundred seventy-six elderly community participants were included in this study. All participants underwent a clinical examination, echocardiography, and blood sampling including PDGF-D single nucleotide polymorphism (SNP) analyses of the rs974819 A/A, G/A and G/G SNP. The follow-up time was 6.7 years. RESULTS: No specific genotype of rs974819 demonstrated increased cardiovascular mortality in the total population, however, the male group with genotypes A/A and G/A demonstrated an increased risk that persisted in a multivariate evaluation where adjustments were made for well-known cardiovascular risk factors (2.7 fold compared with the G/G genotype). No corresponding finding was observed in the female group. CONCLUSION: We report here for the first time that the genotypes G/A or A/A of the SNP rs974819 near PDGF-D exhibited a 2.7 fold increased cardiovascular mortality risk in males. Corresponding increased risk could not be observed in either the total population and thus not in the female group. However, the sample size is was small and the results should be regarded as hypothesis-generating, and thus more research in the field is recommended.


Asunto(s)
Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/mortalidad , Linfocinas/genética , Factor de Crecimiento Derivado de Plaquetas/genética , Polimorfismo de Nucleótido Simple , Anciano , Anciano de 80 o más Años , Alelos , Índice de Masa Corporal , Enfermedades Cardiovasculares/patología , Ecocardiografía , Femenino , Genotipo , Corazón/diagnóstico por imagen , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Modelos de Riesgos Proporcionales , Factores de Riesgo , Factores Sexuales , Volumen Sistólico
13.
Tumour Biol ; 37(1): 591-9, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26234766

RESUMEN

Increasing attention has been focused on the malignant tumor microenvironment, which plays important roles in tumor occurrence, progression and metastasis. Fibroblasts are recruited by platelet-derived growth factor (PDGFs) and invade the tumor microenvironment. In the PDGF family, PDGF-B has been reported to play an important role in the recruitment and invasion programs. However, whether PDGF-D plays a role in these programs remains unclear. We generated a recombinant plasmid expressing human PDGF-D and transfected the plasmid to dermal fibroblasts to examine the effects on cell invasive activities in 3D type I collagen gels. PDGF-D plasmid transfection enhanced fibroblast invasive activities both in invasive cell numbers and invasion depth in 3D collagen gels. These effects were blocked by Snail-specific siRNA transfection. PDGF-D transfection significantly induced Snail expression at both mRNA and protein levels. PDGF-D further upregulated MT1-MMP mRNA and protein expressions and this was inhibited when Snail was knocked down by siRNA. Both Snail and MT1-MMP expressions in fibroblasts and cellular invasive activities in 3D collagen induced by PDGF-D were inhibited by LY294002, SP600125, and U1026, the inhibitors of PI3K, JNK, and ERK1/2 signaling pathways, respectively. However, no effects were observed in response to the P38MAPK signaling pathway inhibitor SB203580. These effects of PDGF-D were confirmed by using the culture supernatants of the transfectants. Taken together, these data demonstrate that PDGF-D plays important roles in the recruitment and invasion programs of fibroblasts via the activation of PI3K, JNK and ERK1/2 signaling pathways, and upregulation of Snail and downstream effecter MT1-MMP. These findings indicate that PDGF-D is an important player in the tumor microenvironment for fibroblast recruitment.


Asunto(s)
Dermis/citología , Fibroblastos/metabolismo , Linfocinas/genética , Metaloproteinasa 14 de la Matriz/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Factor de Crecimiento Derivado de Plaquetas/genética , Factores de Transcripción de la Familia Snail/metabolismo , Animales , Movimiento Celular , Matriz Extracelular/metabolismo , Expresión Génica , Regulación de la Expresión Génica , Humanos , Linfocinas/metabolismo , Sistema de Señalización de MAP Quinasas , Metaloproteinasa 14 de la Matriz/genética , Metaloproteinasas de la Matriz/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , ARN Interferente Pequeño/genética , Ratas , Transducción de Señal , Factores de Transcripción de la Familia Snail/genética , Transfección
14.
Stem Cells ; 33(2): 542-56, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25332166

RESUMEN

Platelet-derived growth factor-D (PDGF-D) was recently identified, and acts as potent mitogen for mesenchymal cells. PDGF-D also induces cellular transformation and promotes tumor growth. However, the functional role of PDGF-D in adipose-derived stem cells (ASCs) has not been identified. Therefore, we primarily investigated the autocrine and paracrine roles of PDGF-D in this study. Furthermore, we identified the signaling pathways and the molecular mechanisms involved in PDGF-D-induced stimulation of ASCs. It is of interest that PDGF-B is not expressed, but PDGF-D and PDGF receptor-ß are expressed in ASCs. PDGF-D showed the strongest mitogenic effect on ASCs, and PDGF-D regulates the proliferation and migration of ASCs through the PI3K/Akt pathways. PDGF-D also increases the proliferation and migration of ASCs through generation of mitochondrial reactive oxygen species (mtROS) and mitochondrial fission. mtROS generation and fission were mediated by p66Shc phosphorylation, and BCL2-related protein A1 and Serpine peptidase inhibitor, clade E, member 1 mediated the proliferation and migration of ASCs. In addition, PDGF-D upregulated the mRNA expression of diverse growth factors such as vascular endothelial growth factor A, fibroblast growth factor 1 (FGF1), FGF5, leukemia inhibitory factor, inhibin, beta A, interleukin 11, and heparin-binding EGF-like growth factor. Therefore, the preconditioning of PDGF-D enhanced the hair-regenerative potential of ASCs. PDGF-D-induced growth factor expression was attenuated by a pharmacological inhibitor of mitogen-activated protein kinase pathway. In summary, PDGF-D is highly expressed by ASCs, where it acts as a potent mitogenic factor. PDGF-D also upregulates growth factor expression in ASCs. Therefore, PDGF-D can be considered a novel ASC stimulator, and used as a preconditioning agent before ASC transplantation.


Asunto(s)
Tejido Adiposo/metabolismo , Regulación de la Expresión Génica/fisiología , Linfocinas/biosíntesis , Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Células Madre/metabolismo , Tejido Adiposo/citología , Células Cultivadas , Citocinas/biosíntesis , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Linfocinas/farmacología , Mitocondrias/metabolismo , Dinámicas Mitocondriales/efectos de los fármacos , Dinámicas Mitocondriales/fisiología , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Mensajero/biosíntesis , Especies Reactivas de Oxígeno/metabolismo , Células Madre/citología
15.
Curr Drug Deliv ; 20(8): 1176-1187, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-35507787

RESUMEN

BACKGROUND: Gene therapeutics are being developed to treat metastatic breast tumors, which are mostly resistant to conventional therapies. Targeting platelet-derived growth factor-D (PDGF-D) is a viable approach because it is known to play roles in angiogenesis and tumor growth. The success of gene therapy is largely dependent on delivery vectors, but both viral and nonviral delivery vectors have their disadvantages. Evolving hybrid vectors are being used to overcome those disadvantages. OBJECTIVES: In this study, we aimed to prepare a recombinant adenovirus type-5 (Ad5)/chitosan hybrid vector to deliver shPDGF-D in a breast cancer cell line by the noncovalent coating of the Ad5 surface with chitosan, a natural polymer. METHODS: The Ad5/chitosan hybrid vector was prepared by the noncovalent coating of the Ad5 surface with different molecular weights (low and high) and different amounts of chitosan (12.5, 25, and 50 µg), and the effect of silencing PDGF-D was investigated in the MDA-MB-231 cell line. RESULTS: In vitro characterization studies showed that the noncovalent chitosan coating increased the size of the Ad5 particle and changed the surface charge from -16.53 mV to slightly neutral. In vitro cell culture studies also showed that the addition of chitosan with both low (73.61%) and high (65.86%) molecular weight increased the PDGF-D silencing efficiency of the Ad5 vector (42.44%) at 48 hours. While low-molecular-weight chitosan had faster effects, high-molecular-weight chitosan provided a more sustained effect in PDGF-D silencing. CONCLUSION: The results indicate that noncovalent chitosan modification may improve the therapeutic effects of the Ad5 vector, offering the potential for further in vitro and in vivo experiments.


Asunto(s)
Neoplasias de la Mama , Quitosano , Humanos , Femenino , Neoplasias de la Mama/genética , Neoplasias de la Mama/terapia , Terapia Genética/métodos , Vectores Genéticos , Línea Celular , Línea Celular Tumoral
16.
Comput Struct Biotechnol J ; 21: 2405-2418, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37066124

RESUMEN

Platelet-derived growth factor-D (PDGF-D) is abundantly expressed in ocular diseases. Yet, it remains unknown whether and how PDGF-D affects ocular cells or cell-cell interactions in the eye. In this study, using single-cell RNA sequencing (scRNA-seq) and a mouse model of PDGF-D overexpression in retinal pigment epithelial (RPE) cells, we found that PDGF-D overexpression markedly upregulated the key immunoproteasome genes, leading to increased antigen processing/presentation capacity of RPE cells. Also, more than 6.5-fold ligand-receptor pairs were found in the PDGF-D overexpressing RPE-choroid tissues, suggesting markedly increased cell-cell interactions. Moreover, in the PDGF-D-overexpressing tissues, a unique cell population with a transcriptomic profile of both stromal cells and antigen-presenting RPE cells was detected, suggesting PDGF-D-induced epithelial-mesenchymal transition of RPE cells. Importantly, administration of ONX-0914, an immunoproteasome inhibitor, suppressed choroidal neovascularization (CNV) in a mouse CNV model in vivo. Together, we show that overexpression of PDGF-D increased pro-angiogenic immunoproteasome activities, and inhibiting immunoproteasome pathway may have therapeutic value for the treatment of neovascular diseases.

17.
Clin Breast Cancer ; 22(2): e173-e183, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34272173

RESUMEN

BACKGROUND: The platelet derived growth factor-D (PDGF-D) plays an important role in breast tumor aggressiveness. However, limited study has investigated the effect of silencing PDGF-D on the biological function of breast cancer. The purpose of this study is to clarify the potential value of PDGF-D as a target for breast cancer treatment. METHODS: Reverse transcription-polymerase chain reaction and western blot were used to detect PDGF-D expression in 5 different breast cancer cells. The lentiviral vector was usd to silence PDGF-D in MDA-MB-231 cells. Then, Methyl Thiazolyl Tetrazolium was used to detect cell viability, 5-Ethynyl-2'- deoxyuridine and a soft agar assay were used to detect cell proliferation and clonality. Additionally, cell apoptosis after PDGF-D knockdown was measured by Annexin V/ Prodium Iodide staining, and cell migration was detected by trans-well assay. Survival rate and tumor size were measured by nude mice transplantation. RESULTS: The MDA-MB-231 and SK-BR-3 cell lines showed higher PDGF-D expression than the MCF7 cell lines (P<.05). After the PDGF-D gene was silenced, the growth and colony forming abilitys ignificantly decreased (P<.05) together with the induction of apoptosis in MDA-MB-231 cells (P<.05). Moreover, MDA-MB-231 cells with PDGF-D silencing showed significantly diminished aggressive migration and invasion potential compared to other cells (P<.05). In vivo experiments also indicated that PDGF-D silencing inhibited tumor growth and improved the survival rate of tumor-bearing mice. CONCLUSION: Downregulation of PDGF-D had dramatic effects on breast cancer cell proliferation, apoptosis and migration, which indicates that it plays an important role in breast cancer development and progression.


Asunto(s)
Neoplasias de la Mama/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Linfocinas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Femenino , Humanos , ARN Mensajero/metabolismo
19.
Protein Sci ; 31(11): e4468, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36214056

RESUMEN

The vaccinia virus expression system is known for the efficient production of recombinant proteins with "appropriate" posttranslational modification using desired mammalian cell lines. However, being a replication competent virus, vaccinia virus poses a health threat to immunocompromised individuals and requires biosafety level 2 (BSL2) laboratory precautions, thereby restricting its use by the scientific community. Development of the host range restricted modified vaccinia Ankara (MVA) system has allowed researchers to work with a safer virus even at BSL1. Here, we report on the use of an improved second generation MVA viral system incorporating two selective markers and fluorescent proteins for easier recombinant virus identification. Notably, we demonstrate that this novel system is capable of producing secreted recombinant proteins, a finding not previously reported. Through purification and characterization of wild type and mutant platelet-derived growth factor D (PDGF D) dimer species, we demonstrate this system is capable of producing the latent full-length PDGF D dimer, partially processed intermediate dimer (hemidimer), as well as fully processed growth factor domain dimer that show chemical integrity and biological activity. Importantly, this system is amenable to scaling up for the mass production of recombinant PDGF D (rPDGF D) dimer species.


Asunto(s)
Virus Vaccinia , Vaccinia , Humanos , Animales , Virus Vaccinia/genética , Replicación Viral , Factor de Crecimiento Derivado de Plaquetas , Proteínas Recombinantes/genética , Mamíferos
20.
Front Cell Dev Biol ; 9: 686886, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34150781

RESUMEN

Platelet-derived growth factor-D (PDGF-D) is highly expressed in immune cells. However, the potential role of PDGF-D in immune system remains thus far unclear. Here, we reveal a novel function of PDGF-D in activating both classical and alternative complement pathways that markedly increase chemokine and cytokine responses to promote macrophage polarization. Pharmacological targeting of the complement C3a receptor using SB290157 alleviated PDGF-D-induced neuroinflammation by blocking macrophage polarization and inhibited pathological choroidal neovascularization. Our study thus suggests that therapeutic strategies targeting both PDGF-D and the complement system may open up new possibilities for the treatment of neovascular diseases.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA