Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 138
Filtrar
1.
Stem Cells Dev ; 30(7): 374-385, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33599158

RESUMEN

The course of differentiation of pluripotent stem cells into cardiomyocytes and the intermediate cell types are characterized using molecular markers for different stages of development. These markers have been selected primarily from studies in the mouse and from a limited number of human studies. However, it is not clear how well mouse cardiogenesis compares with human cardiogenesis at the molecular level. We tackle this issue by analyzing and comparing the expression of common cardiomyogenesis markers [platelet-derived growth factor receptor, alpha polypeptide (PDGFR-α), fetal liver kinase 1 (FLK1), ISL1, NK2 homeobox 5 (NKX2.5), cardiac troponin T (CTNT), connexin43 (CX43), and myosin heavy chain 7 (MYHC-B)] in the developing pig heart at embryonic day (E)15, E16, E18, E20, E22, and E24 and in differentiating cardiomyocytes from human induced pluripotent stem cells (hiPSCs). We found that porcine expression of the mesoderm marker FLK1 and the cardiac progenitor marker ISL1 was in line with our differentiating hiPSC and reported murine expression. The cardiac lineage marker NKX2.5 was expressed at almost all stages in the pig and hiPSC, with an earlier onset in the hiPSC compared with reported murine expression. Markers of immature cardiomyocytes, CTNT, and MYHC-B were consistently expressed throughout E16-E70 in the pig, which is comparable with mouse development, whereas the markers increased over time in the hiPSC. However, the commonly used mature cardiomyocyte marker, CX43, should be used with caution, as it was also expressed in the pig mesoderm, as well as hiPSC immature cardiomyocytes, while this has not been reported in mice. Based on our observations in the various species, we suggest to use FLK1/PDGFR-α for identifying cardiac mesoderm and ISL1/NKX2.5 for cardiac progenitors. Furthermore, a combination of two or more of the following, CTNT+/MYHC-B+/ISL1+ could mark immature cardiomyocytes and CTNT+/ISL1- mature cardiomyocytes. CX43 should be used together with sarcomeric proteins. This knowledge may help improving differentiation of hiPSC into more in vivo-like cardiac tissue in the future.


Asunto(s)
Biomarcadores/metabolismo , Diferenciación Celular , Corazón/embriología , Células Madre Pluripotentes Inducidas/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Línea Celular , Femenino , Proteína Homeótica Nkx-2.5/biosíntesis , Humanos , Inmunohistoquímica/métodos , Células Madre Pluripotentes Inducidas/citología , Ratones , Miocardio/citología , Miocitos Cardíacos/citología , Factor 3 de Transcripción de Unión a Octámeros/biosíntesis , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Factores de Transcripción SOXB1/biosíntesis , Porcinos
2.
Respir Res ; 22(1): 57, 2021 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-33596914

RESUMEN

BACKGROUND: MicroRNA (miR) are small conserved RNA that regulate gene expression post-transcription. Previous genome-wide analysis studies in preterm infants indicate that pathways of miR 219-5p are important in infants with Bronchopulmonary Dysplasia (BPD). METHODS: Here we report a prospective cohort study of extremely preterm neonates wherein infants diagnosed with severe BPD expressed increased airway miR-219-5p and decreased platelet derived growth factor receptor alpha (PDGFR-α), a target of mir-219-5p and a key regulator of alveolarization, compared to post-conception age-matched term infants. RESULTS: miR-219-5p was highly expressed in the pulmonary epithelial lining in lungs of infants with BPD by in situ hybridization of human infant lungs. In both in vitro and in vivo (mouse) models of BPD, miR-219-5p was increased on exposure to hyperoxia compared with the normoxia control, with a complementary decrease of PDGFR-α. To further confirm the target relationship between miR-219 and PDGFR-α, pulmonary epithelial cells (MLE12) and lung primary fibroblasts were treated with a mimic of miR-219-5p and a locked nucleic acid (LNA) based inhibitor of miR-219-5p. In comparison with the control group, the level of miR-219 increased significantly after miR-219 mimic treatment, while the level of PDGFR-α declined markedly. LNA exposure increased PDGFR-α. Moreover, in BPD mouse model, over-expression of miR-219-5p inhibited alveolar development, indicated by larger alveolar spaces accompanied by reduced septation. CONCLUSIONS: Taken together, our results demonstrate that increased miR-219-5p contributes to the pathogenesis of BPD by targeting and reducing PDGFR-α. The use of specific miRNA antagonists may be a therapeutic strategy for preventing the development of BPD.


Asunto(s)
Displasia Broncopulmonar/metabolismo , MicroARNs/biosíntesis , Alveolos Pulmonares/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Animales , Animales Recién Nacidos , Displasia Broncopulmonar/patología , Displasia Broncopulmonar/terapia , Estudios de Cohortes , Presión de las Vías Aéreas Positiva Contínua/métodos , Humanos , Recién Nacido , Recien Nacido Prematuro/metabolismo , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Estudios Prospectivos , Alveolos Pulmonares/patología
3.
J Cell Biochem ; 120(3): 4355-4365, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30246400

RESUMEN

MicroRNAs (miRNAs) have been identified as key players in cardiogenesis and heart pathophysiological processes. However, many miRNAs are still not recognized for their roles in cardiomyocytes differentiation. In this study, we evaluated the effects of microRNA-218 (miR-218) in cardiomyocyte differentiation of the mouse embryonic stem cells (ESCs) in vitro. The percentage of the beating embryoid bodies (EBs) in miR-218 mimic-treated cells was reduced to 32% compared with miR-218 mimic negative control (56%) on day 5 + 3. The amplitude of the intracellular Ca2+ transients in the cardiomyocytes derived from ESCs was reduced upon miR-218 overexpression, followed by the decreased calcium-related proteins and cell junction proteins expressions. Besides, miR-218 expression in ESCs was related to the directional spreading ability of EBs during differentiation. The increased expression of miR-218 could promote the migration of ESCs in vitro, while the decreased expression of miR-218 could inhibit the migration by the transwell experiment. Meanwhile, miR-218 could regulate cell migration-related proteins Cdc42 and Rac1. Platelet-derived growth factor receptor α (PDGFRα) was further confirmed to be a direct target of miR-218 both physically and functionally by dual-luciferase reporter assay. Our data further described that overexpression of PDGFRα rescued the miR-218-mediated inhibition of cardiomyocyte differentiation and restored the miR-218-mediated promotion of cell migration. In conclusion, miR-218 was demonstrated to exert an inhibitory function and promoted cell migration via targeting PDGFRα during cardiomyocyte differentiation from ESCs. The current study revealed the role of miR-218 and may provide an important hint for cardiomyocyte differentiation of ESCs and induced pluripotent stem cells.


Asunto(s)
Diferenciación Celular , Movimiento Celular , MicroARNs/biosíntesis , Células Madre Embrionarias de Ratones/metabolismo , Miocitos Cardíacos/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Animales , Señalización del Calcio/genética , Línea Celular , Ratones , MicroARNs/genética , Células Madre Embrionarias de Ratones/citología , Miocitos Cardíacos/citología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética
4.
Aesthetic Plast Surg ; 43(2): 514-520, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30552469

RESUMEN

BACKGROUND: PGGFRα+ preadipocytes are the major subpopulations that can regenerate into adipocytes. Two different types of macrophages exist in the fat tissue: the classically activated macrophage (M1) and the alternatively activated macrophage (M2). In this study, we investigated whether M1/M2 macrophages play distinct roles in adipogenic differentiation of PDGFRα+ preadipocytes. METHODS: Mouse preadipocytes and macrophages were isolated from C57BL/6 male mice of 6-8 weeks. The culture supernate of M1 and M2 macrophages was collected and co-cultured with the PDGFRα+ preadipocytes. After 3 days, Oil Red O staining was used to evaluate to adipogenic differentiation of PDGFRα+ preadipocytes and the expression of adipogenic-related transcription factors (C/EBP-α, PPARγ) were also tested. RESULTS: The results showed that when cultured in vitro, M1 macrophages could significantly suppress the adipogenesis of PDGFRα+ preadipocytes as well as the C/EBP-α and PPARγ expression, but M2 macrophages did not have significant influence on the adipogenesis of PDGFRα+ preadipocytes nor on C/EBP-α and PPARγ expression compared with the control group. CONCLUSIONS: M1 macrophages significantly suppress PDGFRα+ preadipocyte adipogenesis which provides a possible way to improve adipogenesis and fat retention after fat-free grafting by mitigating acute inflammation and manipulating M1 macrophage levels. NO LEVEL ASSIGNED: This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .


Asunto(s)
Adipocitos , Adipogénesis/fisiología , Diferenciación Celular , Macrófagos/fisiología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas , Adipocitos/metabolismo , Animales , Células Cultivadas , Masculino , Ratones , Ratones Endogámicos C57BL , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/biosíntesis
5.
J Neurosci ; 38(44): 9505-9513, 2018 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-30242047

RESUMEN

PDGFRα, specifically expressed by immature oligodendrocyte progenitor cells (OPCs) in the CNS, plays a critical role in OPC proliferation and migration. However, it has been uncertain whether all cells of oligodendrocyte lineage are derived from the PDGFRα-expressing OPCs. In the present study, we uncovered a PDGFRα-independent oligodendrocyte lineage in the developing cortex. This OPC subpopulation originates from the local ventricular/subventricular zone after birth and contributes to the earliest mature oligodendrocytes in the cortex. PDGFRα signaling does not regulate the generation and differentiation of cortical OPCs. Fate-mapping studies in the PDGFRαCreER; Sox10-GFP/tdTom double-transgenic mice of either sex have further corroborated the PDGFRα-independent oligodendrocyte lineage. This study provides additional missing genetic evidence for PDGFRα-independent oligodendrocyte lineage in the developing hindbrain.SIGNIFICANCE STATEMENT This is the first report of a subpopulation of oligodendrocyte lineage in the developing mouse cortex independent of PDGFRα signaling. These oligodendrocyte progenitor cells are generated from the local ventral ventricular zone/subventricular zone after birth, and contribute to the earliest mature oligodendrocytes in the cortex.


Asunto(s)
Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Corteza Cerebral/metabolismo , Células Precursoras de Oligodendrocitos/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Rombencéfalo/metabolismo , Animales , Corteza Cerebral/embriología , Corteza Cerebral/crecimiento & desarrollo , Femenino , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Embarazo , Rombencéfalo/embriología , Rombencéfalo/crecimiento & desarrollo
6.
Histol Histopathol ; 33(11): 1151-1165, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29693711

RESUMEN

Telocytes (TCs) represent a new distinct type of cells found in the stromal compartment of many organs, including the skeletal, cardiac and smooth muscles. TCs are morphologically defined as interstitial cells with a small cellular body from which arise very long (up to hundreds of micrometers) and thin moniliform processes (named telopodes) featuring the alternation of slender segments (called podomers) and small dilated portions (called podoms) accommodating some organelles. Although these stromal cells are mainly characterized by their ultrastructural traits, in the last few years TCs have been increasingly studied for their immunophenotypes, microRNA profiles, and gene expression and proteomic signatures. By their long-distance spreading telopodes, TCs build a three-dimensional network throughout the whole stromal space and communicate with each other and neighboring cells through homocellular and heterocellular junctions, respectively. Moreover, increasing evidence suggests that TCs may exert paracrine functions being able to transfer genetic information and signaling molecules to other cells via the release of different types of extracellular vesicles. A close relationship between TCs and stem/progenitor cell niches has also been described in several organs. However, the specific functions of TCs located in the muscle interstitium remain to be unraveled. Here, we review the morphological and possible functional aspects of TCs in skeletal, cardiac and smooth muscle tissues. The potential involvement of TCs in muscle tissue pathological changes and future possibilities for targeting TCs as a novel promising therapeutic strategy to foster muscle tissue regeneration and repair are also discussed.


Asunto(s)
Músculo Esquelético/citología , Músculo Liso/citología , Miocardio/citología , Telocitos , Animales , Antígenos CD34/biosíntesis , Humanos , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Células del Estroma , Telocitos/citología , Telocitos/fisiología
7.
Anticancer Res ; 38(2): 795-802, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29374704

RESUMEN

AIM: The purpose of this study was to explore the parallel expression of platelet-derived growth factor receptor α (PDGFRα) and human epidermal growth factor receptor 2 (HER2) or p53 in relation to clinicopathological parameters of oral squamous cell carcinoma (OSCC) to define their role in progressive growth of tumor. MATERIALS AND METHODS: Expression of PDGFRα, HER2 and p53 was evaluated in 71 OSCC samples by immunohistochemistry. HER2 status was verified by fluorescence in situ hybridization. RESULTS: PDGFRα and p53 expression were associated with tumor grade (p=0.043 and p=0.040, respectively). HER2 expression was more frequent in advanced (III/IV) cancer (p=0.006). A positive correlation of PDGFRα with HER2 (r=0.267; p=0.024) and with p53 (r=0.266; p=0.025) was noted. PDGFRα/HER2 and PDGFRα/p53 co-expression was found more often in G3 than in G1 and G2 tumors (p=0.008 and p=0.015, respectively). CONCLUSION: Our study revealed that PDGFRα/HER2 and PDGFRα/p53 co-expression exists in poorly differentiated OSCCs, suggesting that cooperation between these proteins might enhance aggressive behavior of tumor.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de la Boca/metabolismo , Receptor ErbB-2/biosíntesis , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Proteína p53 Supresora de Tumor/biosíntesis , Biomarcadores de Tumor/biosíntesis , Carcinoma de Células Escamosas/patología , Femenino , Neoplasias de Cabeza y Cuello/patología , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Neoplasias de la Boca/patología , Carcinoma de Células Escamosas de Cabeza y Cuello
8.
Stem Cells Dev ; 27(3): 184-198, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29205098

RESUMEN

Cardiac progenitor cells (CPCs) are being developed as a promising treatment for heart failure. Although clinical trials have predominantly used donor cardiac biopsies to derive CPCs, a better solution could be to use previously cryopreserved human heart tissue. This would enable timely and convenient access to healthy and young heart samples for CPC production. However, few studies have attempted to isolate CPCs from previously cryopreserved heart tissue. In this study, we isolated CPCs from eight nondiseased human heart samples previously cryopreserved as part of the Sydney Heart Bank. Resulting cells were strongly positive for known fibroblast (DDR2, Vimentin), mesenchymal/CPC (PDGFRα, CD90) markers, and for pluripotency genes (SOX2, NANOG, MYC, KLF4), whereas being negative for the pan-hematopoietic marker (CD45). Outgrowth cells from aged hearts had decreased proliferative and self-renewing capacity that correlated with shorter telomere lengths compared with cells from young hearts. No telomerase activity was detected in any cells isolated. Colony-forming assays and fluorescence-activated cell sorting were used to enrich PDGFRα+/CD90+/CD31- CPCs. Multipotent potential was confirmed using in vitro differentiation assays with smooth muscle (MYH11+), endothelial cell (vWF+), and cardiomyocyte-like (cTnT+, α-actinin+) cell formation. Single cell assays demonstrated clonogenicity of PDGFRα+ CPCs with maintenance of prolonged self-renewing capacity (>2 months), and pluripotency gene expression at both early and late culture passages. Our results demonstrate that multipotent PDGFRα+ CPCs can be harvested and expanded from previously banked cryopreserved human heart samples. These data support cardiac tissue banking as a strategy for improved access to CPCs for future clinical therapies.


Asunto(s)
Antígenos de Diferenciación/biosíntesis , Separación Celular , Criopreservación , Regulación de la Expresión Génica , Miocardio , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Células Cultivadas , Humanos , Factor 4 Similar a Kruppel , Mioblastos Cardíacos/citología , Mioblastos Cardíacos/metabolismo , Miocardio/citología , Miocardio/metabolismo
9.
Cancer Res ; 77(16): 4279-4292, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28634201

RESUMEN

Despite intensive multimodal treatment of sarcomas, a heterogeneous group of malignant tumors arising from connective tissue, survival remains poor. Candidate-based targeted treatments have demonstrated limited clinical success, urging an unbiased and comprehensive analysis of oncogenic signaling networks to reveal therapeutic targets and personalized treatment strategies. Here we applied mass spectrometry-based phosphoproteomic profiling to the largest and most heterogeneous set of sarcoma cell lines characterized to date and identified novel tyrosine phosphorylation patterns, enhanced tyrosine kinases in specific subtypes, and potential driver kinases. ALK was identified as a novel driver in the Aska-SS synovial sarcoma (SS) cell line via expression of an ALK variant with a large extracellular domain deletion (ALKΔ2-17). Functional ALK dependency was confirmed in vitro and in vivo with selective inhibitors. Importantly, ALK immunopositivity was detected in 6 of 43 (14%) of SS patient specimens, one of which exhibited an ALK rearrangement. High PDGFRα phosphorylation also characterized SS cell lines, which was accompanied by enhanced MET activation in Yamato-SS cells. Although Yamato-SS cells were sensitive to crizotinib (ALK/MET-inhibitor) but not pazopanib (VEGFR/PDGFR-inhibitor) monotherapy in vitro, synergistic effects were observed upon drug combination. In vivo, both drugs were individually effective, with pazopanib efficacy likely attributable to reduced angiogenesis. MET or PDGFRα expression was detected in 58% and 84% of SS patients, respectively, with coexpression in 56%. Consequently, our integrated approach has led to the identification of ALK and MET as promising therapeutic targets in SS. Cancer Res; 77(16); 4279-92. ©2017 AACR.


Asunto(s)
Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-met/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Sarcoma Sinovial/tratamiento farmacológico , Sarcoma Sinovial/enzimología , Quinasa de Linfoma Anaplásico , Animales , Línea Celular Tumoral , Estudios de Cohortes , Crizotinib , Femenino , Humanos , Indazoles , Ratones , Ratones Endogámicos BALB C , Terapia Molecular Dirigida , Fosforilación , Proteómica , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-met/genética , Pirazoles/farmacología , Piridinas/farmacología , Pirimidinas/farmacología , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Sarcoma Sinovial/genética , Transducción de Señal , Sulfonamidas/farmacología , Sulfonas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Intern Med ; 56(7): 823-826, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28381750

RESUMEN

Pulmonary artery sarcoma is highly malignant and easily metastasizes to the systemic organs. Both the introduction of novel diagnostic procedures and the development of new treatment modalities are required to achieve long-term survival. Several studies have shown that platelet-derived growth factor receptor α (PDGFRα) gene amplification is frequently observed in pulmonary artery sarcoma. PDGFRα is known to be involved in cell proliferation in certain malignancies. PDGFRα may become a potential biological marker in pulmonary artery sarcoma. We report a case in which a diagnosis of pulmonary artery sarcoma overexpressing PDGFRα was made using endovascular catheter biopsy following positron emission tomography with integrated computed tomography (PET/CT) scans.


Asunto(s)
Arteria Pulmonar/patología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Sarcoma/metabolismo , Sarcoma/patología , Neoplasias Vasculares/metabolismo , Neoplasias Vasculares/patología , Biomarcadores , Proliferación Celular , Femenino , Humanos , Pulmón/patología , Persona de Mediana Edad , Tomografía Computarizada por Tomografía de Emisión de Positrones , Sarcoma/diagnóstico , Neoplasias Vasculares/diagnóstico
11.
Tissue Cell ; 48(3): 242-51, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26993973

RESUMEN

Demyelination was induced by two weeks cuprizone treatment. Rats of +ve control and triiodothyronine (T3) then received three subcutaneous injections of either saline or T3 day after day and sacrificed at the end of the third and fifth weeks. Animals in -ve control group received only standard rodent chow. After one week of cuprizone withdrawal the corpus callosum in +ve control and T3 treated rats was still demyelinated as revealed by MBP immunohistochemistry. The assay of PLP gene showed significant increase of T3 treated group compared to both the -ve control and +ve control groups. After three weeks, significant improvement in myelination was detected in T3-treated group compared to +ve control as detected by both MBP immunohistochemistry and electron microscopy. After one week of cuprizone withdrawal, PDGFRα positive cells and gene expression showed significant increase in +ve control and T3-treated groups as compared to -ve control with insignificant difference in between the former two groups. After three weeks of cuprizone withdrawal, PDGFRα positive cells in T3-treated and +ve control groups decreased to the control levels. These results suggest that T3 was effective in improving remyelination when administered during acute phase and might direct progenitor lineage toward oligodendrocytes.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Oligodendroglía/efectos de los fármacos , Células Madre/efectos de los fármacos , Triyodotironina/administración & dosificación , Animales , Cuerpo Calloso/efectos de los fármacos , Cuerpo Calloso/ultraestructura , Cuprizona/administración & dosificación , Enfermedades Desmielinizantes/metabolismo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Inyecciones Subcutáneas , Masculino , Ratones , Microscopía Electrónica , Oligodendroglía/metabolismo , Ratas , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Células Madre/metabolismo
12.
Pathol Res Pract ; 212(3): 204-9, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26778387

RESUMEN

BACKGROUND: To investigate the clinical and prognostic significance of Ku80 and PDGFR-α in nasal type NK/T cell lymphoma (NKTCL). METHODS: Immunohistochemistry for Ku80 and PDGFR-α was performed on tissue sections from 35 patients diagnosed with NKTCL. We analyzed the relationship between Ku80 and PDGFR-α expression and the clinicopathological features of NKTCL. We further performed multivariate analyses to identify prognostic factors for progression free survival (PFS) of patients. RESULTS: Ku80 expression rate in NKTCL was 94.3% compared with that in reactive lymphoid hyperplasia of nasopharynx (P=0.003). The positive expression rate of PDGFR-α in the group of NKTCL was 91.4% compared with that in control group (P=0.004). The positive correlation between Ku80 and PDGFR-α was also found (r=0.496, P=0.002). Ku80 and PDGFR-α expressions were not correlated with patient's gender, age, B symptoms, LDH, Ann Arbor stage, IPI score and treatment (P>0.05). High expression of Ku80 and PDGFR-α was shown to be correlated with worse PFS (P=0.003 and 0.034, respectively). Multivariate analysis with a Cox proportional hazards model further suggested that Ku80 high expression rate (HR, 11.495; P=0.009), PDGFR-α high expression rate (HR, 4.836; P=0.031) and International Prognostic Index (IPI) score of 3-4 (HR, 7.308; P=0.001) were statistically independent prognostic factors for patients' PFS. Our results suggest that Ku80 and PDGFR-α may be valuable indicators for predicting the survival of NKTCL patients. CONCLUSION: Ku80 and PDGFR-α might be effective predictive indicators for the prognosis of NKTCL. A large prospective study is required to confirm the prognostic significance of Ku80 and PDGFR-α in NKTCL.


Asunto(s)
Biomarcadores de Tumor/análisis , Autoantígeno Ku/biosíntesis , Linfoma Extranodal de Células NK-T/patología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Adolescente , Adulto , Anciano , Supervivencia sin Enfermedad , Femenino , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Autoantígeno Ku/análisis , Linfoma Extranodal de Células NK-T/metabolismo , Linfoma Extranodal de Células NK-T/mortalidad , Masculino , Persona de Mediana Edad , Pronóstico , Modelos de Riesgos Proporcionales , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/análisis , Estudios Retrospectivos , Adulto Joven
13.
Cells Tissues Organs ; 201(1): 26-37, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26613250

RESUMEN

The Apert syndrome is a rare congenital disorder most often arising from S252W or P253R mutations in fibroblast growth factor receptor (FGFR2). Numerous studies have focused on the regulatory role of Apert FGFR2 signaling in bone formation, whereas its functional role in tooth development is largely unknown. To investigate the role of FGFR signaling in cell proliferation and odontogenic differentiation of human dental cells in vitro, we isolated dental pulp and enamel organ epithelia (EOE) tissues from an Apert patient carrying the S252W FGFR2 mutation. Apert primary pulp and EOE cells were established and shown to exhibit normal morphology and express alkaline phosphatase under differentiation conditions. Similar to control cells, Apert dental pulp and EOE cells expressed all FGFRs, with highest levels of FGFR1 followed by FGFR2 and low levels of FGFR3 and FGFR4. However, Apert cells had increased cell growth compared with control cells. Distinct from previous findings in osteoblast cells, gain-of-function S252W FGFR2 mutation did not upregulate the expression of epidermal growth factor receptor (EGFR) and platelet-derived growth factor receptor (PDGFRα), but elevated extracellular signal-regulated kinase (ERK) signaling in cells after EGF stimulation. Unexpectedly, there was little effect of the S252W mutation on odontogenic gene expression in dental pulp and EOE cells. However, after inhibition of total FGFR signaling or ERK signaling, the expression of odontogenic genes was upregulated in both dental cell types, indicating the negative effect of whole FGFR signaling on odontogenic differentiation. This study provides novel insights on FGFR signaling and a common Apert FGFR2 mutation in the regulation of odontogenic differentiation of dental mesenchymal and epithelial cells.


Asunto(s)
Acrocefalosindactilia/genética , Pulpa Dental/citología , Órgano del Esmalte/citología , Odontogénesis/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Diente/embriología , Fosfatasa Alcalina/biosíntesis , Diferenciación Celular/genética , Proliferación Celular/genética , Células Cultivadas , Receptores ErbB/biosíntesis , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación de la Expresión Génica/genética , Humanos , Masculino , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Transducción de Señal
14.
Cancer Lett ; 365(2): 174-81, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26049020

RESUMEN

INTRODUCTION: Glioma initiating cells (GICs) play important roles in tumor initiation and progression. However, interactions between tumor cells and host cells of local tumor microenvironment are kept largely unknown. Besides GICs and their progeny cells, whether adjacent normal glial cells contribute to tumorigenesis during glioma tissue remodeling deserves further investigation. METHODS: Red fluorescence protein (RFP) gene was stably transfected into human GIC cells lines SU3 and U87, then were transplanted intracerebrally into athymic nude mice with whole-body green fluorescence protein (GFP) expression. The interactions between GICs and host cells in vivo were observed during tissue remodeling processes initiated by hGICs. The biological characteristics of host glial cells with high proliferation capability cloned from the xenograft were further assayed. RESULTS: In a SU3 initiated dual-fluorescence xenograft glioma model, part of host cells cloned from the intracerebral tumors were found acquiring the capability of unlimited proliferation. PCR and FISH results indicated that malignant transformed cells were derived from host cells; cell surface marker analysis showed these cells expressed murine oligodendrocyte specific marker CNP, and oligodendrocyte progenitor cells (OPCs) specific markers PDGFR-α and NG2. Chromosomal analysis showed these cells were super tetraploid. In vivo studies showed they behaved with high invasiveness activity and nearly 100% tumorigenic ratio. Compared with SU3 cells with higher PDGF-B expression, GICs derived from U87 cells with low level of PDGF-B expression failed to induce host cell transformation. CONCLUSIONS: Primary high invasive GICs SU3 contribute to transformation of adjacent normal host glial cells in local tumor microenvironment possibly via PDGF/PDGFR signaling activation, which deserved further investigation.


Asunto(s)
Neoplasias Encefálicas/patología , Glioma/patología , Células Madre Neoplásicas/patología , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica , Modelos Animales de Enfermedad , Proteínas Fluorescentes Verdes/genética , Humanos , Proteínas Luminiscentes/genética , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Trasplante de Neoplasias , Neuroglía/citología , Proteínas Proto-Oncogénicas c-sis/biosíntesis , Proteínas Proto-Oncogénicas c-sis/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Transducción de Señal , Trasplante Heterólogo , Microambiente Tumoral , Proteína Fluorescente Roja
15.
Eur J Haematol ; 95(5): 480-3, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25941032

RESUMEN

Rare patients suffering from myeloid neoplasms share clinical and cytological features indistinguishable from chronic myeloid leukemia (CML) but lack the BCR-ABL1 fusion gene. Several studies provide evidence that alterations in genes encoding tyrosine kinase receptors such as the platelet-derived growth factor receptor (PDGFR) may be involved in the pathogenesis of these disorders. Here we describe a patient with a rare CML-like disease in whom we identified a novel in-frame BCR-PDGFRA rearrangement joining BCR exon 17 to PDGFRA exon 13, resulting in overexpression of PDGFRA. The design of a specific quantitative PCR assay to monitor the molecular response during treatment with imatinib, a multitargeted tyrosine kinase inhibitor (TKI) with activity against ABL, c-Kit, and PDGFRA revealed an outstanding disease control with durably undetectable BCR-PDGFRA transcripts. Multiple TKIs are currently available yet with distinct target profiles; thus, accurate molecular diagnosis and monitoring tools are essential to establish tailored treatments and assess response to therapy in this type of rare hematological malignancy.


Asunto(s)
Exones , Mesilato de Imatinib/administración & dosificación , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Proteínas de Fusión Oncogénica/genética , Proteínas Proto-Oncogénicas c-bcr/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Anciano , Femenino , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/enzimología , Neoplasia Residual , Proteínas de Fusión Oncogénica/metabolismo , Proteínas Proto-Oncogénicas c-bcr/biosíntesis , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/biosíntesis
16.
BMC Cancer ; 14: 281, 2014 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-24758355

RESUMEN

BACKGROUND: Zalypsis(®) is a marine compound in phase II clinical trials for multiple myeloma, cervical and endometrial cancer, and Ewing's sarcoma. However, the determinants of the response to Zalypsis are not well known. The identification of biomarkers for Zalypsis activity would also contribute to broaden the spectrum of tumors by selecting those patients more likely to respond to this therapy. METHODS: Using in vitro drug sensitivity data coupled with a set of molecular data from a panel of sarcoma cell lines, we developed molecular signatures that predict sensitivity to Zalypsis. We verified these results in culture and in vivo xenograft studies. RESULTS: Zalypsis resistance was dependent on the expression levels of PDGFRα or constitutive phosphorylation of c-Kit, indicating that the activation of tyrosine kinase receptors (TKRs) may determine resistance to Zalypsis. To validate our observation, we measured the levels of total and active (phosphorylated) forms of the RTKs PDGFRα/ß, c-Kit, and EGFR in a new panel of diverse solid tumor cell lines and found that the IC50 to the drug correlated with RTK activation in this new panel. We further tested our predictions about Zalypsis determinants for response in vivo in xenograft models. All cells lines expressing low levels of RTK signaling were sensitive to Zalypsis in vivo, whereas all cell lines except two with high levels of RTK signaling were resistant to the drug. CONCLUSIONS: RTK activation might provide important signals to overcome the cytotoxicity of Zalypsis and should be taken into consideration in current and future clinical trials.


Asunto(s)
Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Sarcoma/tratamiento farmacológico , Sarcoma/genética , Biomarcadores Farmacológicos , Línea Celular Tumoral , Resistencia a Antineoplásicos , Receptores ErbB/biosíntesis , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas Proto-Oncogénicas c-kit/biosíntesis , ARN Mensajero/biosíntesis , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Sarcoma/patología , Tetrahidroisoquinolinas/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
17.
APMIS ; 122(9): 867-72, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24750504

RESUMEN

Anaplastic lymphoma kinase (ALK) gene aberrations are found in several tumor types including anaplastic large cell lymphoma (ALCL) and non-small cell lung cancer (NSCLC). Crizotinib, an inhibitor of ALK-fusion proteins, has shown clinical activity, but resistance mechanisms limit long-lasting disease control. It has been shown that ALK-NPM fusion upregulates platelet-derived growth factor receptor beta (PDGFRB) via JUN and transcription factor Jun B (JUNB) in ALCL. PDGFRB inhibition has been identified as therapy option for ALK-positive ALCL. Here, we investigated the ALK/JUN/JUNB/transcription factor Jun C (JUNC)/PDGFR axis in metastatic NSCLC with regard to ALK aberrations. We performed immunohistochemical analysis of platelet-derived growth factor receptor alpha (PDGFRA), PDGFRB, JUNB and JUNC expression in formalin-fixed, paraffin-embedded specimens of 15 NSCLC brain metastases (5 ALK translocations, 3 of them involving EML4, 5 ALK amplifications, 5 without ALK aberrations). We did not find a statistically significant difference in expression of PDGFRA, PDGFRB, JUNB or JUNC in tumor samples with normal ALK status, ALK amplification or ALK translocations (Kruskal-Wallis test p > 0.05). Interestingly, PDGFRB was not expressed in tumor cells (but in vascular and stromal cells) in any of our cases. Our data argue against PDGFRB activation in association with ALK gene aberrations in metastatic NSCLC and thus seem to imply differential pathobiological roles of ALK alterations in lung cancer and lymphoma, possibly depending on different fusion partner genes. These results may be relevant for targeted therapies, as they indicate that inhibition of PDGFRB in ALK translocated NSCLC seems to be no therapeutic opportunity.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares/genética , Proteínas Tirosina Quinasas Receptoras/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Anciano , Quinasa de Linfoma Anaplásico , Proteínas de Ciclo Celular/biosíntesis , Crizotinib , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Linfoma/genética , MAP Quinasa Quinasa 4/biosíntesis , Masculino , Proteínas Asociadas a Microtúbulos/biosíntesis , Persona de Mediana Edad , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Tirosina Quinasas , Pirazoles/uso terapéutico , Piridinas/uso terapéutico , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Serina Endopeptidasas/biosíntesis , Factores de Transcripción/biosíntesis
19.
Stem Cells ; 32(8): 2072-83, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24677751

RESUMEN

Embryonic stem cells (ESCs) represent an ideal model to study how lineage decisions are established during embryonic development. Using a doxycycline-inducible mouse ESC line, we have previously shown that expression of the transcriptional activator Pax3 in early mesodermal cells leads to the robust generation of paraxial mesoderm progenitors that ultimately differentiate into skeletal muscle precursors. Here, we show that the ability of this transcription factor to induce the skeletal myogenic cell fate occurs at the expenses of the cardiac lineage. Our results show that the PDGFRα+FLK1--subfraction represents the main population affected by Pax3, through downregulation of several transcripts encoding for proteins involved in cardiac development. We demonstrate that although Nkx2-5, Tbx5, and Gata4 negatively affect Pax3 skeletal myogenic activity, the cardiac potential of embryoid body-derived cultures is restored solely by forced expression of Tbx5. Taking advantage of this model, we used an unbiased genome-wide approach to identify genes whose expression is rescued by Tbx5, and which could represent important regulators of cardiac development. These findings elucidate mechanisms regulating the commitment of mesodermal cells in the early embryo and identify the Tbx5 cardiac transcriptome.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Embrionarias/metabolismo , Músculo Esquelético/citología , Miocardio/citología , Factores de Transcripción Paired Box/metabolismo , Proteínas de Dominio T Box/metabolismo , Animales , Western Blotting , Linaje de la Célula , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Regulación del Desarrollo de la Expresión Génica , Corazón/embriología , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Músculo Esquelético/embriología , Factor de Transcripción PAX3 , Técnicas de Placa-Clamp , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/biosíntesis
20.
Anticancer Res ; 34(2): 623-30, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24510991

RESUMEN

AIM: To investigate the expression of platelet-derived growth factor (PDGF) receptor-A (PDGFRα), epidermal growth factor receptor (EGFR) and c-Met in spinal chordoma. To the authors' knowledge, little is known regarding the prognostic significance of receptor tyrosine kinase in spinal chordoma. MATERIALS AND METHODS: Using immunohistochemical techniques, the authors investigated PDGFR-α, EGFR and c-MET expression in 52 primary and 104 recurrent lesions, and compared these data with clinicopathological parameters. RESULTS: PDGFR-α, EGFR and c-MET were found to be expressed in 75.0%, 83% and 77% of primary, and in 97.0% of recurrent lesions in all investigated receptor tyrosine kinases. Higher PDGFR-α and c-MET expression was found to be correlated with younger patient age. Lesions with a higher expression of PDGFR-α demonstrated significantly higher EGFR scores in both primary and recurrent lesions compared to those with lower PDGFR-α expression. In recurrent lesions, higher c-MET expression was found to be associated with significantly better prognosis than those with lower c-MET expression (p=0.033). Lesions with a higher level of PDGFR-α expression were found to have significantly poorer prognosis than those with lower PDGFR-α expression (p=0.024). Those patients with lower EGFR expression were found to have significantly better prognosis than those with higher EGFR expression (p=0.005). CONCLUSION: In the current study, c-MET expression in patients with spinal chordoma was found to be correlated with a younger patient age and a favorable prognosis. Patients with a higher level of PDGFR-α and EGFR expression were found to have a significantly poorer prognosis than those with lower PDGFR-α and EGFR expression.


Asunto(s)
Cordoma/enzimología , Receptores ErbB/biosíntesis , Proteínas Proto-Oncogénicas c-met/biosíntesis , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Neoplasias de la Médula Espinal/enzimología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Cordoma/patología , Estudios de Cohortes , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Neoplasias de la Médula Espinal/patología , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...