Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 163
Filtrar
1.
J Histochem Cytochem ; 67(11): 825-844, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31411936

RESUMEN

There is a growing recognition that myogenic stem cells are influenced by their microenvironment during regeneration. Several interstitial cell types have been described as supportive for myoblasts. In this role, both the pericyte as a possible progenitor for mesenchymal stem cells, and interstitial cells in the endomysium have been discussed. We have applied immunohistochemistry on normal and pathological human skeletal muscle using markers for pericytes, or progenitor cells and found a cell type co-expressing CD10, CD34, CD271, and platelet-derived growth factor receptor α omnipresent in the endomysium. The marker profile of these cells changed dynamically in response to muscle damage and atrophy, and they proliferated in response to damage. The cytology and expression profile of the CD10+ cells indicated a capacity to participate in myogenesis. Both morphology and indicated function of these cells matched properties of several previously described interstitial cell types. Our study suggests a limited number of cell types that could embrace many of these described cell types. Our study indicate that the CD10+, CD34+, CD271+, and platelet-derived growth factor receptor α+ cells could have a supportive role in human muscle regeneration, and thus the mechanisms by which they exert their influence could be implemented in stem cell therapy.


Asunto(s)
Células Madre Mesenquimatosas/citología , Músculo Esquelético/citología , Pericitos/citología , Telocitos/citología , Adapaleno/análisis , Antígenos CD34/análisis , Humanos , Inmunohistoquímica , Músculo Esquelético/ultraestructura , Neprilisina/análisis , Receptores del Factor de Crecimiento Derivado de Plaquetas/análisis
2.
Br J Cancer ; 121(2): 139-149, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31235865

RESUMEN

BACKGROUND: Crenolanib is a tyrosine kinase inhibitor targeting PDGFR-α, PDGFR-ß and Fms related tyrosine kinase-3 (FLT3) that is currently evaluated in several clinical trials. Although platelet-derived growth factor receptor (PDGFR) signalling pathway is believed to play an important role in angiogenesis and maintenance of functional vasculature, we here demonstrate a direct angiostatic activity of crenolanib independently of PDGFR signalling. METHODS: The activity of crenolanib on cell viability, migration, sprouting, apoptosis and mitosis was assessed in endothelial cells, tumour cells and fibroblasts. Alterations in cell morphology were determined by immunofluorescence experiments. Flow-cytometry analysis and mRNA expression profiles were used to investigate cell differentiation. In vivo efficacy was investigated in human ovarian carcinoma implanted on the chicken chorioallantoic membrane (CAM). RESULTS: Crenolanib was found to inhibit endothelial cell viability, migration and sprout length, and induced apoptosis independently of PDGFR expression. Treated cells  showed altered actin arrangement and nuclear aberrations. Mitosis was affected at several levels including mitosis entry and centrosome clustering. Crenolanib suppressed human ovarian carcinoma tumour growth and angiogenesis in the CAM model. CONCLUSIONS: The PDGFR/FLT3 inhibitor crenolanib targets angiogenesis and inhibits tumour growth in vivo unrelated to PDGFR expression. Based on our findings, we suggest a broad mechanism of action of crenolanib.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antineoplásicos/farmacología , Bencimidazoles/farmacología , Moduladores de la Mitosis/farmacología , Piperidinas/farmacología , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Tirosina Quinasa 3 Similar a fms/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Pollos , Femenino , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Neoplasias Ováricas/irrigación sanguínea , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Receptores del Factor de Crecimiento Derivado de Plaquetas/análisis , Receptores del Factor de Crecimiento Derivado de Plaquetas/fisiología
3.
Anal Chem ; 91(12): 7603-7612, 2019 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-31088078

RESUMEN

Clinical and biomedical research seeks single-cell quantification to better understand their roles in a complex, multicell environment. Recently, quantification of vascular endothelial growth factor receptors (VEGFRs) provided important insights into endothelial cell characteristics and response in tumor microenvironments. However, existing technologies for quantifying plasma membrane receptor tyrosine kinases (RTKs) lack multiplexing capabilities, limiting detailed characterization. Here, we use the unique spectral properties of quantum dots (Qdots) to optimize and dually quantify VEGFR1 and VEGFR2 on human umbilical vein endothelial cells (HUVECs). To enable this quantification, we reduce nonspecific binding between Qdot-conjugated antibodies and cells via buffer optimization. Second, we identify optimal labeling conditions by examining Qdot-conjugated antibody binding to five receptors: VEGFRs (VEGFR1 and VEGFR2), their coreceptor neuropilin1 (NRP1), and platelet-derived growth factor receptor (PDGFRα and PDGFRß). We establish that 800-20 000 is the dynamic range where accurate Qdot-enabled quantification can be achieved. Through these optimizations, we demonstrate measurement of 1 100 VEGFR1 and 6 900 VEGFR2 per HUVEC. We induce ∼90% upregulation of VEGFR1 and ∼30% downregulation of VEGFR2 concentration via 24 h VEGF-A165 treatment. We observe no change in VEGFR1 or VEGFR2 concentration with 24 h VEGF-B167 treatment. We further apply Qdots to analyze HUVEC heterogeneity and observe that 24 h VEGF-A165 treatment induces a ∼15% decrease in VEGFR2 heterogeneity, but little to no change in VEGFR1 heterogeneity. We observe that VEGF-B167 induces little to no change in either VEGFR1 or VEGFR2 heterogeneity. Overall, we demonstrate experimental and analytical strategies for quantifying two or more RTKs at single-level using Qdots, which will help provide new insights into biological systems.


Asunto(s)
Puntos Cuánticos/química , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Anticuerpos Inmovilizados/química , Anticuerpos Inmovilizados/inmunología , Regulación hacia Abajo/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Receptores del Factor de Crecimiento Derivado de Plaquetas/análisis , Receptores del Factor de Crecimiento Derivado de Plaquetas/inmunología , Factor A de Crecimiento Endotelial Vascular/farmacología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/inmunología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/inmunología
4.
Biochem Biophys Res Commun ; 508(3): 965-972, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30545632

RESUMEN

Renal fibrosis is the final common pathway of various renal injuries and it leads to chronic kidney disease. Recent studies reported that FOXD1-lineage pericyte plays a critical role in tubulointerstitial fibrosis (TIF). However the regulatory mechanisms remain unclear. Autophagy is a cellular process of degradation of damaged cytoplasmic components that regulates cell death and proliferation. To investigate the role of autophagy in FOXD1-lineage pericytes on renal TIF, we generated the FOXD1-lineage stromal cell-specific Atg7 deletion (Atg7△FOXD1) mice. FOXD1-lineage stromal cell-specific Atg7 deletion enhanced renal TIF through Smad-dependent transforming growth factor (TGF)-ß signaling after unilateral ureteral obstruction (UUO). FOXD1-lineage stromal cell-specific Atg7 deletion increased the accumulation of interstitial myofibroblasts and enhanced the differentiation of pericytes into myofibroblasts after UUO. Peritubular capillary rarefaction was accelerated in Atg7△FOXD1 mice after UUO. Atg7△FOXD1 mice increased the accumulation of SQSTM1/p62-positive aggregates in the obstructed kidney and resulted in increased expression of NLRP3 inflammasome, interleukin (IL) 1-ß and caspase-1 signaling pathway, which enhanced apoptosis of interstitial cells after UUO. In summary, our data showed that autophagy in FOXD1-lineage stromal cells plays a protective role in renal TIF through regulating the Smad4 dependent TGF-ß an NLRP3 inflammasome signaling pathway.


Asunto(s)
Autofagia , Factores de Transcripción Forkhead/análisis , Inflamasomas/metabolismo , Riñón/patología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Apoptosis , Proteína 7 Relacionada con la Autofagia/genética , Diferenciación Celular , Linaje de la Célula , Fibrosis , Factores de Transcripción Forkhead/genética , Riñón/citología , Riñón/metabolismo , Masculino , Ratones , Ratones Noqueados , Miofibroblastos/citología , Pericitos/citología , Receptores del Factor de Crecimiento Derivado de Plaquetas/análisis , Transducción de Señal , Proteínas Smad/metabolismo , Células del Estroma/química , Obstrucción Ureteral/complicaciones
5.
Hum Pathol ; 65: 157-165, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28506734

RESUMEN

Kaposi sarcoma (KS) is a locally progressive, intermediate-grade vascular neoplasm with no known cure, high recurrence rates, and potential for wide dissemination. Low efficacy and high toxicity limit current therapeutic options for advanced disease. Activation of mammalian target of rapamycin (mTOR), platelet-derived growth factor (PDGF), vascular endothelial growth factor (VEGF), and c-kit signaling pathways has been implicated in KS pathogenesis and may suggest a role for targeted inhibitors. KS cases were retrospectively retrieved (N=274), most (90%) associated with human immunodeficiency virus. Tissue microarray slides were stained with human herpes virus-8, Friend leukemia integration 1 transcription factor, CD117 (c-kit), phospho-S6 (pS6), PDGF receptor-ß, VEGF, and phospho-mTOR. Both intensity and extent of staining were scored. Multiplying these scores for each core yielded total staining H-scores. Human herpes virus-8 was positive in 87% and Friend leukemia integration 1 transcription factor in 95.7% of cases. Most were also VEGF+ (97.6%), pS6+ (95.7%), CD117+ (92.5%), and PDGFRB+ (87.4%). Approximately half (55.6%) were phospho-mTOR+. There was no significant difference in staining among patients with low (<500 cells/mm3) or preserved CD4 T-cell counts. Immunohistochemistry confirms upregulation of the mTOR, PDGF, VEGF, and c-kit pathways in a large cohort of KS samples. Of proteins tested, pS6, downstream of mTOR, demonstrated the highest proportion of strong positivity (67.1%). These results support the possibility of using targeted inhibitors in KS. Overexpression was independent of CD4 count, suggesting that even patients with low counts may be targeted therapy candidates.


Asunto(s)
Biomarcadores de Tumor/análisis , Proteínas Proto-Oncogénicas c-kit/análisis , Receptores del Factor de Crecimiento Derivado de Plaquetas/análisis , Sarcoma de Kaposi/química , Transducción de Señal , Serina-Treonina Quinasas TOR/análisis , Factor A de Crecimiento Endotelial Vascular/análisis , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/uso terapéutico , Recuento de Linfocito CD4 , Niño , Preescolar , Femenino , Infecciones por VIH/inmunología , Infecciones por VIH/virología , Herpesvirus Humano 8/aislamiento & purificación , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Terapia Molecular Dirigida , Fosforilación , Valor Predictivo de las Pruebas , Proteínas Proto-Oncogénicas c-kit/antagonistas & inhibidores , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Estudios Retrospectivos , Proteína S6 Ribosómica/análisis , Sarcoma de Kaposi/tratamiento farmacológico , Sarcoma de Kaposi/patología , Sarcoma de Kaposi/virología , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Análisis de Matrices Tisulares , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Adulto Joven
6.
World J Gastroenterol ; 22(19): 4662-72, 2016 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-27217698

RESUMEN

AIM: To determine if expression of colonic tryptophan hydroxylase-2 (TPH2), a surrogate marker of neuronal 5-hydroxytryptamine, is altered in Hirschsprung's-associated enterocolitis. METHODS: Entire resected colonic specimens were collected at the time of pull-through operation in children with Hirschsprung's disease (HSCR, n = 12). Five of these patients had a history of pre-operative Hirschsprung's-associated enterocolitis (HAEC). Controls were collected at colostomy closure in children with anorectal malformation (n = 10). The distribution of expression of TPH2 was evaluated using immunofluorescence and confocal microscopy. Protein expression of TPH2 was quantified using western blot analysis in the deep smooth muscle layers. RESULTS: TPH2 was co-expressed in nitrergic and cholinergic ganglia in the myenteric and submucosal plexuses in ganglionic colon in HSCR and healthy controls. Co-expression was also seen in submucosal interstitial cells of Cajal and PDGFRα(+) cells. The density of TPH2 immuno-positive fibers decreased incrementally from ganglionic bowel to transition zone bowel to aganglionic bowel in the myenteric plexus. Expression of TPH2 was reduced in ganglionic bowel in those affected by pre-operative HAEC compared to those without HAEC and healthy controls. However, expression of TPH2 was similar or high compared to controls in the colons of children who had undergone diverting colostomy for medically refractory HAEC. CONCLUSION: Altered TPH2 expression in colonic serotonergic nerves of patients with HSCR complicated by HAEC may contribute to intestinal secretory and motor disturbances, including recurrent HAEC.


Asunto(s)
Colon/inervación , Sistema Nervioso Entérico/enzimología , Enterocolitis/enzimología , Enfermedad de Hirschsprung/enzimología , Neuronas Serotoninérgicas/enzimología , Triptófano Hidroxilasa/análisis , Anoctamina-1 , Biomarcadores/análisis , Western Blotting , Estudios de Casos y Controles , Canales de Cloruro/análisis , Colon/patología , Colon/cirugía , Sistema Nervioso Entérico/patología , Sistema Nervioso Entérico/fisiopatología , Enterocolitis/patología , Enterocolitis/fisiopatología , Enterocolitis/cirugía , Femenino , Técnica del Anticuerpo Fluorescente , Enfermedad de Hirschsprung/patología , Enfermedad de Hirschsprung/fisiopatología , Enfermedad de Hirschsprung/cirugía , Humanos , Lactante , Masculino , Microscopía Confocal , Proteínas de Neoplasias/análisis , Receptores del Factor de Crecimiento Derivado de Plaquetas/análisis
7.
Tumour Biol ; 37(8): 10053-66, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27193823

RESUMEN

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


Asunto(s)
Biomarcadores de Tumor/análisis , Proteínas de Neoplasias/análisis , Neoplasias/química , Factor de Crecimiento Derivado de Plaquetas/análisis , Receptores del Factor de Crecimiento Derivado de Plaquetas/análisis , Transducción de Señal , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Aptámeros de Péptidos/uso terapéutico , Ensayos Clínicos como Asunto , Monitoreo de Drogas , Humanos , Terapia Molecular Dirigida , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/química , Proteínas de Neoplasias/fisiología , Neoplasias/mortalidad , Neoplasias/terapia , Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Proteínas de Fusión Oncogénica/genética , Factor de Crecimiento Derivado de Plaquetas/química , Factor de Crecimiento Derivado de Plaquetas/fisiología , Pronóstico , Isoformas de Proteínas/análisis , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Interferencia de ARN , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Receptores del Factor de Crecimiento Derivado de Plaquetas/química , Receptores del Factor de Crecimiento Derivado de Plaquetas/fisiología , Resultado del Tratamiento
8.
Domest Anim Endocrinol ; 51: 46-55, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25498237

RESUMEN

This study evaluated the messenger RNA (mRNA) expression and immunolocalization of all members of the platelet-derived growth factor (PDGF) family in caprine ovaries by quantitative PCR and immunohistochemistry, respectively. Detectable levels of PDGF-A mRNA were not observed in primordial follicles. Higher levels of PDGF-B mRNA were observed in primary follicles than in primordial follicles (P < 0.05). PDGF-D mRNA levels were higher in secondary follicles than in the other preantral follicle categories (P < 0.05). PDGF-B mRNA expression was higher than PDGF-C mRNA expression in primary follicles (P < 0.05). In antral follicles, PDGF-A mRNA expression was higher in cumulus-oocyte complexes (COCs) from small antral follicles than in those from large antral follicles and their respective granulosa/theca (GT) cells (P < 0.05). Furthermore, in COCs from small and large antral follicles, PDGF-A mRNA expression was higher than that of the other PDGF isoforms (P < 0.05). The mRNA levels of PDGF-B and PDGF-D and PDGFR-α and PDGFR-ß were higher in GT cells from large antral follicles than in GT cells from small antral follicles and in their respective COCs (P < 0.05). In COCs and GT cells from small antral follicles, the mRNA levels of PDGFR-α were higher than those of PDGFR-ß (P < 0.05). All proteins were observed in the cytoplasm of oocytes from all follicular categories. In granulosa cells, all PDGFs and PDGFR-ß were detected from starting at the secondary stage, and in theca cells, all proteins, except PDGF-C, were detected starting at the antral stage. In conclusion, PDGF and its receptors are differentially expressed in the oocytes and ovarian cells according to the stage of follicular development, suggesting their role in the regulation of folliculogenesis in goats.


Asunto(s)
Expresión Génica , Cabras/metabolismo , Ovario/metabolismo , Factor de Crecimiento Derivado de Plaquetas/genética , Receptores del Factor de Crecimiento Derivado de Plaquetas/genética , Animales , Células del Cúmulo/química , Femenino , Células de la Granulosa/química , Inmunohistoquímica/veterinaria , Oocitos/química , Folículo Ovárico/química , Ovario/química , Factor de Crecimiento Derivado de Plaquetas/análisis , Reacción en Cadena de la Polimerasa/veterinaria , ARN Mensajero/análisis , Receptores del Factor de Crecimiento Derivado de Plaquetas/análisis , Células Tecales/química
9.
J Comp Pathol ; 151(4): 322-8, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25172054

RESUMEN

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


Asunto(s)
Linfoma/veterinaria , Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Receptores del Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Animales , Enfermedades de los Perros/metabolismo , Enfermedades de los Perros/patología , Perros , Inmunohistoquímica , Linfoma/metabolismo , Linfoma/patología , Factor de Crecimiento Derivado de Plaquetas/análisis , Receptores del Factor de Crecimiento Derivado de Plaquetas/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
Ann Surg Oncol ; 21 Suppl 4: S584-90, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24743909

RESUMEN

BACKGROUND: Various kinds of molecular targeted drugs to inhibit receptor tyrosine kinases (RTKs) have been recently developed. The relationship between the expression status of major RTKs and prognosis in gastric cancer remains unclear. We conducted a multicenter study to evaluate the prognostic impact of the expression of epidermal growth factor receptor (EGFR), c-Met, platelet-derived growth factor receptor (PDGFR), and c-Kit in gastric cancer. METHODS: This study included 153 gastric cancer patients who underwent gastrectomy at 9 institutions between 2000 and 2006. Expression status of EGFR, c-Met, PDGFR, and c-Kit were evaluated with immunohistochemistry (IHC) centrally. Overall survival based on RTK expression status was statistically compared. Cox multivariate analysis was conducted to adjust for potentially confounding factors. RESULTS: The positive rates for EGFR, c-Met, PDGFR, and c-Kit were 14.4, 24.8, 41.2, and 11.1 %, respectively. Significant interactions with expression status were observed for pathological N stage with EGFR; HER2-status with c-Met; tumor location, histology, and pathological N stage with PDGFR; and no examined variables with c-Kit. Concomitant HER2 positivity was observed for 0.7 % of tumors positive for EGFR, 3.9 % for c-Met, 4.6 % for PDGFR, and 1.3 % for c-Kit. There were some differences in overall survival between patients with or without RTK expression, but only c-Kit expression showed a significant survival difference in Cox multivariate analysis (P = 0.046). CONCLUSIONS: Our multicenter study indicated that IHC expression of 4 RTKs had some prognostic impact and that c-Kit-positive status may be a significant indicator of good prognosis in gastric cancer patients.


Asunto(s)
Adenocarcinoma/química , Adenocarcinoma/patología , Proteínas Tirosina Quinasas Receptoras/análisis , Neoplasias Gástricas/química , Neoplasias Gástricas/patología , Adenocarcinoma/cirugía , Adulto , Anciano , Anciano de 80 o más Años , Receptores ErbB/análisis , Femenino , Gastrectomía , Humanos , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Pronóstico , Proteínas Proto-Oncogénicas c-kit/análisis , Receptor ErbB-2/análisis , Receptores del Factor de Crecimiento Derivado de Plaquetas/análisis , Estudios Retrospectivos , Neoplasias Gástricas/cirugía , Tasa de Supervivencia
11.
Auris Nasus Larynx ; 38(1): 123-6, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20580173

RESUMEN

OBJECTIVE: We reported an extremely rare case of atypical laryngeal carcinoid, and examined the expression of several proteins for application of molecular targeted therapy. METHOD: Case report and review of the literature concerning atypical carcinoid arising from the larynx. The expressions of proteins were determined by immunohistochemical analysis. RESULTS: We present here a case of atypical laryngeal carcinoid in a 79-year-old Japanese man, which was completely resected, and with no evidence of recurrence. On immunohistochemical analysis, neoplastic elements revealed, strong positivity for platelet-derived growth factor receptor α (PDGFRα), vascular endothelial growth factor receptor 2 (VEGFR2), and epidermal growth factor receptor (EGFR), and were mild positivity for KIT. CONCLUSION: Our findings suggest that atypical laryngeal carcinoid could be completely removed if it is located in the limited lesion. PDGFRα, VEGFR2, and EGFR expressions in this case provide the evidence that atypical laryngeal carcinoid is the candidate for molecular targeted therapy, although further investigations are necessary.


Asunto(s)
Tumor Carcinoide/química , Receptores ErbB/análisis , Neoplasias Laríngeas/química , Terapia Molecular Dirigida , Receptores del Factor de Crecimiento Derivado de Plaquetas/análisis , Receptor 2 de Factores de Crecimiento Endotelial Vascular/análisis , Anciano , Biomarcadores/análisis , Tumor Carcinoide/tratamiento farmacológico , Humanos , Inmunohistoquímica , Neoplasias Laríngeas/tratamiento farmacológico , Masculino
12.
J Orthop Sci ; 15(5): 654-60, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20953927

RESUMEN

BACKGROUND: Imatinib myselate is a molecularly targeted drug that inhibits Abl tyrosine kinase, as well as type III tyrosine kinase receptors such as platelet-derived growth factor receptor (PDGFR), KIT, colony-stimulating factor 1 receptor (CSF-1R), and discoidin domain receptor (DDR). Ph1 chromosome-positive chronic myeloid leukemias (CMLs), KIT-positive gastrointestinal stromal tumors (GISTs), and PDGFR-positive dermatofibrosarcoma protuberans (DFSP) have been reported to be responsive to imatinib treatment. We conducted a multicenter Phase II trial of imatinib in patients with relapsed or refractory KIT-positive (excluding GISTs) or PDGFR-positive sarcomas. METHODS: Patient ages ranged from 12 and 75 years. Eligibility criteria included (1) metastatic sarcomas with a definitive diagnosis based on histopathology or that were completely unresectable and locally advanced; (2) relapsed or refractory cases that had completed standard treatment; and (3) a tumor confirmed by immunohistochemical staining to be KIT- or PDGFR-positive. A 600-mg dose of imatinib was administered to patients once a day, with each patient receiving six courses of the drug and each course lasting 4 weeks. In cases categorized as stable or progressive, the imatinib dose was increased to 800 mg/day administered twice daily. RESULTS: A total of 25 patients who met the eligibility criteria were enrolled in the trial; 22 were evaluated for response. The response rate with a 600 mg/day dose of imatinib was 4.5% (0 complete response, 1 partial response). There were no other objective responses after increasing imatinib to 800 mg/day (0/10). We estimated 50% progression-free survival to be 61.0 days for an imatinib dose of 600 mg/day based on the Kaplan-Meier method. Side effects of imatinib were generally similar to those observed in previous clinical trials. CONCLUSIONS: Our results did not indicate effectiveness of imatinib monotherapy at a dose of 600 or 800 mg/day in patients with relapsed or refractory KIT-positive (excluding GISTs) or PDGFR-positive sarcomas. Our findings suggest the need to evaluate the synergistic effect of combination therapy with other anticancer drugs.


Asunto(s)
Antineoplásicos/uso terapéutico , Piperazinas/uso terapéutico , Proteínas Proto-Oncogénicas c-kit/análisis , Pirimidinas/uso terapéutico , Receptores del Factor de Crecimiento Derivado de Plaquetas/análisis , Sarcoma/tratamiento farmacológico , Adolescente , Adulto , Anciano , Antineoplásicos/efectos adversos , Benzamidas , Niño , Supervivencia sin Enfermedad , Femenino , Humanos , Mesilato de Imatinib , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Piperazinas/efectos adversos , Pirimidinas/efectos adversos , Sarcoma/química , Sarcoma/patología , Sarcoma/secundario , Adulto Joven
13.
Thyroid ; 20(9): 975-80, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20718683

RESUMEN

BACKGROUND: Currently, there is no standard treatment for metastatic anaplastic thyroid cancer (ATC). DNA microarray analysis has shown platelet-dervived growth factor receptor (PDGFR) overexpression in ATC relative to well-differentiated thyroid cancer. In p53-mutated/deficient ATC cell lines, cABL is overexpressed, and selective inhibition of cABL results in a cytostatic effect. Imatinib inhibits tyrosine kinase activity of Bcr-ABL and PDGF. We hypothesize that patients with ATC that over-expresses PDGF receptors or cABL will respond to imatinib. METHODS: Patients with histologically confirmed ATC who had measurable disease and whose disease expressed PDGF receptors by immunohistochemistry were eligible for study. Imatinib was administered at 400 mg orally twice daily without drug holiday. Response to treatment was assessed every 8 weeks. Patients with complete response, partial responses, or stable disease were treated until disease progression. The study was terminated early due to poor accrual. RESULTS: From February 2004 to May 2007, 11 patients were enrolled and were started on imatinib. At baseline, 4/11 had locoregional disease, 5/11 had distant metastases, and 2/11 had both. Nine of 11 had prior chemoradiation, and 7/11 had thyroidectomy. Eight of 11 were evaluable for response; 4 were excluded for lack of follow-up with radiologic evaluation. The overall response rates at 8 weeks were complete response 0/8, partial response 2/8, and stable disease 4/8. The median time to follow-up was 26 months (ranges 23-30 months). The rate of 6-month progression-free survival was 36% (95% confidence interval, 9%-65%). The rate of 6-month overall survival was 45% (95% confidence interval, 16%-70%). The most common grade 3 toxicity was edema in 25%; other grade 3 toxicities included fatigue and hyponatremia (12.5% each). There were no grade 4 toxicities or treatment related deaths. CONCLUSIONS: Imatinib appears to have activity in advanced ATC and is well tolerated. Due to difficulty of accruing patients with a rare malignancy at a single institution, further investigation of imatinib in ATC may be warranted in a multi-institutional setting.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma/tratamiento farmacológico , Piperazinas/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirimidinas/uso terapéutico , Neoplasias de la Tiroides/tratamiento farmacológico , Anciano , Antineoplásicos/efectos adversos , Benzamidas , Carcinoma/radioterapia , Supervivencia sin Enfermedad , Femenino , Humanos , Mesilato de Imatinib , Masculino , Persona de Mediana Edad , Piperazinas/efectos adversos , Inhibidores de Proteínas Quinasas/efectos adversos , Pirimidinas/efectos adversos , Receptores del Factor de Crecimiento Derivado de Plaquetas/análisis , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores
14.
Lasers Med Sci ; 25(4): 531-7, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20162318

RESUMEN

Irradiation by light emitting diode (LED) promotes fibroblast proliferation and wound healing. However, its mechanism is still unknown. The purpose of this study was to clarify the mechanism of fibroblast proliferation by LED irradiation. Cultured NIH3T3 fibroblasts from normal mice were irradiated by LED with a center wavelength of 627 nm. LED irradiation was performed with an energy density of 4 J/cm(2), at subculture and 24 h later. The expression of several growth factors and their receptors was analyzed by reverse transcriptase-polymerase chain reaction (RT-PCR): platelet-derived growth factor (PDGF)-A, PDGF-B, and PDGF-C, transforming growth factor-beta (TGF-beta), basic fibroblast growth factor (bFGF), PDGF-alpha receptor, and TGF-beta receptor. Then, the activation of the extracellular signal-regulated kinase (ERK) pathway was examined by Western blotting with and without the PDGF receptor inhibitor. LED irradiation induced cell growth of NIH3T3 fibroblasts. The expression of PDGF-C had significantly increased in the irradiated group (P < 0.01). Although strong activation of the ERK pathway was observed in the irradiated group, its activation was completely suppressed by the PDGF receptor inhibitor. We concluded that LED irradiation promotes fibroblast proliferation by increasing autocrine production of PDGF-C and activating the ERK pathway through phosphorylation of the PDGF receptor.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Luz , Animales , División Celular/efectos de la radiación , Células Cultivadas , Activación Enzimática/efectos de la radiación , Factor 2 de Crecimiento de Fibroblastos/análisis , Fibroblastos/efectos de la radiación , Linfocinas/análisis , Ratones , Factor de Crecimiento Derivado de Plaquetas/análisis , Proteínas Proto-Oncogénicas c-sis/análisis , Receptores del Factor de Crecimiento Derivado de Plaquetas/análisis , Receptores de Factores de Crecimiento Transformadores beta/análisis , Transducción de Señal/efectos de la radiación , Factor de Crecimiento Transformador beta/análisis
15.
Pediatr Nephrol ; 25(5): 867-75, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20020158

RESUMEN

Congenital nephrotic syndrome of the Finnish type (NPHS1, CNF) is an autosomal recessive disease caused by mutations in a major podocyte protein, nephrin. NPHS1 is associated with heavy proteinuria and the development of glomerular scarring. We studied the cellular and molecular changes affecting the glomerular mesangium in NPHS1 kidneys. Marked hyperplasia of mesangial cells (MC) was mainly responsible for the early mesangial expansion in NPHS1 glomeruli. The levels of the proliferation marker, mindbomb homolog 1 and the major MC mitogen, platelet-derived growth factor, and its receptors, however, were quite normal. Only a small number of cells were positive for CD68 (marker for phagocytic cells) and CD34 (marker for mesenchymal precursor cells) in the NPHS1 mesangium. MCs strongly expressed alpha-smooth muscle actin, indicating myofibloblast transformation. The expression levels of the profibrotic mediators osteopontin and transforming growth factor beta were up-regulated in NPHS1 glomeruli by 3.2 and 1.6-fold, respectively, compared to the controls. The synthesis by MCs of the typical fibroblast products collagen I, fibronectin, and tenascin, however, was low, and the extracellular matrix increase was caused by the accumulation of a normal MC product, collagen IV. The results indicate that severe glomerular sclerosis can develop without major qualitative cellular or molecular changes in the mesangium.


Asunto(s)
Proliferación Celular , Mesangio Glomerular/patología , Células Mesangiales/patología , Síndrome Nefrótico/patología , Actinas/análisis , Adolescente , Antígenos CD/análisis , Antígenos CD34/análisis , Antígenos de Diferenciación Mielomonocítica/análisis , Biopsia , Estudios de Casos y Controles , Niño , Preescolar , Progresión de la Enfermedad , Proteínas de la Matriz Extracelular/análisis , Genotipo , Mesangio Glomerular/química , Mesangio Glomerular/cirugía , Humanos , Hiperplasia , Inmunohistoquímica , Lactante , Proteínas de la Membrana/genética , Células Mesangiales/química , Persona de Mediana Edad , Mutación , Nefrectomía , Síndrome Nefrótico/clasificación , Síndrome Nefrótico/congénito , Síndrome Nefrótico/metabolismo , Síndrome Nefrótico/cirugía , Osteopontina/análisis , Fenotipo , Factor de Crecimiento Derivado de Plaquetas/análisis , Receptores del Factor de Crecimiento Derivado de Plaquetas/análisis , Esclerosis , Ubiquitina-Proteína Ligasas/análisis
16.
Anal Chem ; 81(18): 7795-800, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19691297

RESUMEN

Tunable aptamer capillary electrophoresis (CE) techniques were developed to enable the separation and detection of platelet derived growth factor (PDGF) isomers and their receptors. Using an aptamer that formed a stable complex with the B chain but not with the A chain of PDGF, we were able to tweak the electrophoretic mobilities of the PDGF isomers for their separation. PDGF-AB bound to a single aptamer molecule was well resolved from PDGF-BB bound to two aptamer molecules. Simultaneous determination of 50 pM of two isomers was accomplished in a single analysis. Furthermore, PDGF-AB was used as a connector to bring receptor alpha and fluorescent aptamer into a single complex molecule. As a result, the formation of a (receptor alpha)-(PDGF-AB)-(fluorescent aptamer) ternary complex enabled the detection of the receptor alpha by tunable aptamer CE. A competitive assay was developed to determine receptor beta, making use of the competition between the receptor beta and fluorescent aptamer in binding to PDGF-BB. Detection limits were 0.5 nM for PDGF receptor alpha and 3 nM for receptor beta. Determination of PDGF isomers and their receptors in diluted serum samples showed no interference from the sample matrix.


Asunto(s)
Aptámeros de Nucleótidos/metabolismo , Electroforesis Capilar/métodos , Factor de Crecimiento Derivado de Plaquetas/análisis , Receptores del Factor de Crecimiento Derivado de Plaquetas/análisis , Aptámeros de Nucleótidos/química , Colorantes Fluorescentes/química , Factor de Crecimiento Derivado de Plaquetas/química , Isoformas de Proteínas/análisis , Isoformas de Proteínas/química , Receptores del Factor de Crecimiento Derivado de Plaquetas/sangre , Receptores del Factor de Crecimiento Derivado de Plaquetas/química , Proteínas Recombinantes/análisis , Proteínas Recombinantes/química
17.
Orthod Craniofac Res ; 12(3): 168-77, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19627518

RESUMEN

Our goal was to discover genes differentially expressed in the perichondrium (PC) of the mandibular condylar cartilage (MCC) that might enhance regenerative medicine or orthopaedic therapies directed at the tissues of the temporomandibular joint. We used targeted gene arrays (osteogenesis, stem cell) to identify genes preferentially expressed in the PC and the cartilaginous (C) portions of the MCC in 2-day-old mice. Genes with higher expression in the PC sample related to growth factor ligand-receptor interactions [FGF-13 (6.4x), FGF-18 (4x), NCAM (2x); PGDF receptors, transforming growth factor (TGF)-beta and IGF-1], the Notch isoforms (especially Notch 3 and 4) and their ligands or structural proteins/proteoglycans [collagen XIV (21x), collagen XVIII (4x), decorin (2.5x)]. Genes with higher expression in the C sample consisted mostly of known cartilage-specific genes [aggrecan (11x), procollagens X (33x), XI (14x), IX (4.5x), Sox 9 (4.4x) and Indian hedgehog (6.7x)]. However, the functional or structural roles of several genes that were expressed at higher levels in the PC sample are unclear [myogenic factor (Myf) 9 (9x), tooth-related genes such as tuftelin (2.5x) and dentin sialophosphoprotein (1.6x), VEGF-B (2x) and its receptors (3-4x) and sclerostin (1.7x)]. FGF, Notch and TGF-beta signalling may be important regulators of MCC proliferation and differentiation; the relatively high expression of genes such as Myf6 and VEGF-B and its receptors suggests a degree of unsuspected plasticity in PC cells.


Asunto(s)
Cartílago Articular/metabolismo , Expresión Génica/genética , Cóndilo Mandibular/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Agrecanos/análisis , Animales , Animales Recién Nacidos , Proteínas Morfogenéticas Óseas/análisis , Colágeno/análisis , Colágeno Tipo IX/análisis , Colágeno Tipo X/análisis , Colágeno Tipo XI/análisis , Decorina , Proteínas del Esmalte Dental/análisis , Proteínas de la Matriz Extracelular/análisis , Factores de Crecimiento de Fibroblastos/análisis , Marcadores Genéticos , Glicoproteínas , Proteínas Hedgehog/análisis , Factor I del Crecimiento Similar a la Insulina/análisis , Péptidos y Proteínas de Señalización Intercelular , Ratones , Factores Reguladores Miogénicos/análisis , Moléculas de Adhesión de Célula Nerviosa/análisis , Fosfoproteínas/análisis , Procolágeno/análisis , Precursores de Proteínas/análisis , Proteoglicanos/análisis , Proteínas Proto-Oncogénicas/análisis , Receptor Notch3 , Receptor Notch4 , Receptores Notch/análisis , Receptores del Factor de Crecimiento Derivado de Plaquetas/análisis , Receptores de Factores de Crecimiento Endotelial Vascular/análisis , Factor de Transcripción SOX9/análisis , Sialoglicoproteínas , Factor de Crecimiento Transformador beta/análisis , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor B de Crecimiento Endotelial Vascular/análisis
18.
Clin Breast Cancer ; 8(6): 511-5, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19073506

RESUMEN

BACKGROUND: Imatinib mesylate is a potent inhibitor of the Bcr-Abl, c-Kit, and platelet-derived growth factor receptor (PDGFR) tyrosine kinases. On the basis of variable expression of c-Kit and PDGFR in breast cancer and of in vitro data supporting synergy between imatinib and capecitabine, the Southwest Oncology Group conducted a phase II trial of the combination in metastatic breast cancer. PATIENTS AND METHODS: Eligible patients had progressive, measurable metastatic breast cancer and received

Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/patología , Desoxicitidina/análogos & derivados , Fluorouracilo/análogos & derivados , Piperazinas/administración & dosificación , Pirimidinas/administración & dosificación , Receptores del Factor de Crecimiento Derivado de Plaquetas/análisis , Administración Oral , Adulto , Anciano , Benzamidas , Neoplasias de la Mama/química , Capecitabina , Desoxicitidina/administración & dosificación , Sinergismo Farmacológico , Femenino , Fluorouracilo/administración & dosificación , Humanos , Mesilato de Imatinib , Persona de Mediana Edad , Proteínas Proto-Oncogénicas c-kit/análisis , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/análisis
19.
Eur J Haematol ; 81(1): 58-64, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18331602

RESUMEN

Transient myeloproliferative disorder (TMD) is experienced by approximately 10% of neonates with Down syndrome (DS). Most TMD is asymptomatic and the patients undergo spontaneous remission within a few months. However, some cases are fatal because of systemic organ dysfunctions including hepatic fibrosis. Some cytokines such as platelet-derived growth factor (PDGF) may be involved in the development of hepatic fibrosis in TMD. The report describes a fatal case of TMD accompanying DS. The patient presented with pulmonary hypertension and hepatic failure. An autopsy disclosed severe fibrosis in the lung, liver, kidney and pancreas. Immunohistochemical analysis revealed high expression of PDGF receptor beta in the severe fibrotic areas of the fibrotic tissues. A real-time polymerase chain reaction (PCR) analysis demonstrated the expression of PDGFalpha and PDGFbeta in the peripheral blood samples of the patient. The finding indicates that the PDGF pathway may play an important role in the fibrosis of several organs in patients with TMD.


Asunto(s)
Síndrome de Down/complicaciones , Fibrosis/etiología , Trastornos Mieloproliferativos/patología , Factor de Crecimiento Derivado de Plaquetas/fisiología , Receptores del Factor de Crecimiento Derivado de Plaquetas/análisis , Humanos , Recién Nacido , Masculino , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas , Receptor beta de Factor de Crecimiento Derivado de Plaquetas
20.
Anal Chem ; 80(5): 1497-504, 2008 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-18237154

RESUMEN

This study describes the first instance of the use of two differently sized Au nanoparticles (Au NPs), acting separately as donor and acceptor, in homogeneous photoluminescence quenching assays developed for the analysis of proteins. Introduction of a breast cancer marker protein, platelet-derived growth factor AA (PDGF AA), to a solution of 11-mercaptoundecanoic acid-protected, 2.0-nm photoluminescent Au nanodots (L(AuND)) led to the preparation of PDGF AA-L(AuND) as the donor. Thiol-derivative PDGF binding aptamers (Apt) and 13-nm spherical Au NPs were used to synthesize the Apt-Q(AuNP) acceptor. The photoluminescence of PDGF AA-L(AuND) at 520 nm decreased when photoluminescence quenching occurred between Apt-Q(AuNP) and PDGF AA-L(AuND). We used the PDGF AA-L(AuND)/Apt-Q(AuNP)-based molecular light switching system to analyze PDGFs and PDGF alpha-receptor in separate homogeneous solutions. In the presence of PDGFs, the interaction between Apt-Q(AuNP) and PDGF AA-L(AuND) decreased as a result of competitive reactions between the PDGFs and Apt-Q(AuNP). Similarly, the interaction between Apt-Q(AuNP) and PDGF AA-L(AuND) reduced as a result of competitive reactions between PDGF alpha-receptor and PDGF AA-L(AuND). The limits of detection (LODs) for PDGF AA and PDGF alpha-receptor were 80 pM and 0.25 nM, respectively, resulting from a low background photoluminescence signal. When using the Apt-Q(AuNP) as selectors for (a) the enrichment of PDGF AA and (b) the removal of matrixes possessing intense background fluorescence from cell media and urine samples, the LOD for PDGF AA decreased to 10 pM. Unlike quantum dots, the L(AuND) provide the advantages of biocompatibility, ease of bioconjugation, and minimal toxicity.


Asunto(s)
Oro/química , Luminiscencia , Nanoestructuras/química , Factor de Crecimiento Derivado de Plaquetas/análisis , Receptores del Factor de Crecimiento Derivado de Plaquetas/análisis , Trombina/análisis , Línea Celular Tumoral , Humanos , Óptica y Fotónica , Factor de Crecimiento Derivado de Plaquetas/química , Receptores del Factor de Crecimiento Derivado de Plaquetas/química , Trombina/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA