Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 55(8): 1466-1482.e9, 2022 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-35863346

RESUMEN

Although many studies have addressed the regulatory circuits affecting neuronal activities, local non-synaptic mechanisms that determine neuronal excitability remain unclear. Here, we found that microglia prevented overactivation of pre-sympathetic neurons in the hypothalamic paraventricular nucleus (PVN) at steady state. Microglia constitutively released platelet-derived growth factor (PDGF) B, which signaled via PDGFRα on neuronal cells and promoted their expression of Kv4.3, a key subunit that conducts potassium currents. Ablation of microglia, conditional deletion of microglial PDGFB, or suppression of neuronal PDGFRα expression in the PVN elevated the excitability of pre-sympathetic neurons and sympathetic outflow, resulting in a profound autonomic dysfunction. Disruption of the PDGFBMG-Kv4.3Neuron pathway predisposed mice to develop hypertension, whereas central supplementation of exogenous PDGFB suppressed pressor response when mice were under hypertensive insult. Our results point to a non-immune action of resident microglia in maintaining the balance of sympathetic outflow, which is important in preventing cardiovascular diseases.


Asunto(s)
Hipertensión , Microglía , Animales , Hipertensión/metabolismo , Ratones , Neuronas/fisiología , Potasio/metabolismo , Proteínas Proto-Oncogénicas c-sis/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo
2.
Neurotox Res ; 39(4): 1181-1188, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33871814

RESUMEN

Multiple sclerosis is a chronic demyelinating disease with a functional disturbance in the immune system and axonal damages. It was shown that Apamin as a blood-brain barrier shuttle acts as a Ca2+ activated K+ channels (SK channels) blocker. In this study, the effects of Apamin on oligodendrocyte differentiation markers were evaluated on an induced model of MS. Briefly, C57BL/6 male mice (22 ± 5 g) except the control group were fed with 0.2% (w/w) cuprizone pellets for 6 weeks. After cuprizone withdrawal, mice were divided randomly into six groups. Apamin (100 µg/kg/BW) was administered intraperitoneally as a co-treatment during phase I (demyelination) or post-treatment phase II (remyelination) twice a week. Mice were anesthetized, perfused with phosphate-buffered saline, then fixed brains were coronally sectioned and the changes in oligodendrocytes markers such as Olig2, PDGFR-α, and BrdU incorporation were assessed by immunohistochemistry assay. Apamin administration increased Olig2+ cells in phase I as compared to the control group (p < 0.0001). Also, a decreasing trend in PDGFRa+ cells observed after cuprizone withdrawal (p < 0.001). 5-Bromo-2'-deoxyuridine (BrdU) incorporation test was confirmed stimulation of oligodendrocyte progenitor cell proliferation in phase I in the Apamin exposed group (p < 0.0001), especially at the subventricular zone. This study highlights the potential therapeutic effects of Apamin as a bee venom-derived peptide on oligodendrocyte precursor proliferation and elevation in myelin content in an oxidative induced multiple sclerosis model due to cuprizone exposure.


Asunto(s)
Venenos de Abeja/uso terapéutico , Barrera Hematoencefálica/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cuprizona/toxicidad , Esclerosis Múltiple/tratamiento farmacológico , Oligodendroglía/efectos de los fármacos , Animales , Venenos de Abeja/farmacología , Barrera Hematoencefálica/química , Barrera Hematoencefálica/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Proliferación Celular/fisiología , Quelantes/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple/inducido químicamente , Esclerosis Múltiple/metabolismo , Factor de Transcripción 2 de los Oligodendrocitos/análisis , Factor de Transcripción 2 de los Oligodendrocitos/metabolismo , Oligodendroglía/química , Oligodendroglía/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/análisis , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo
3.
AAPS J ; 23(1): 4, 2020 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-33210183

RESUMEN

Tovetumab (MEDI-575) is a fully human IgG2κ monoclonal antibody that specifically binds to human platelet-derived growth factor receptor alpha (PDGFRα) and blocks receptor signal transduction by PDGF ligands. The affinity of tovetumab determined using surface plasmon resonance technology and flow cytometry demonstrated comparable binding affinity for human and monkey PDGFRα. In single and repeat-dose monkey pharmacokinetic-pharmacodynamic (PK-PD) studies, tovetumab administration resulted in dose-dependent elevation of circulating levels of PDGF-AA, a member of the PDGF ligand family, due to displacement of PDGF-AA from PDGFRα by tovetumab and subsequent blockade of PDGFRα-mediated PDGF-AA degradation. As such, PDGF-AA accumulation is an indirect measurement of receptor occupancy and is a novel PD biomarker for tovetumab. The nonlinear PK of tovetumab and dose-dependent increase in circulating PDGF-AA profiles were well described by a novel mechanistic model, in which tovetumab and PDGF-AA compete for the binding to PDGFRα. To facilitate translational simulation, the internalization half-lives of PDGF-AA and tovetumab upon binding to PDGFRα were determined using confocal imaging to be 14 ± 4 min and 30 ± 8 min, respectively. By incorporating PDGFRα internalization kinetics, the model not only predicted the target receptor occupancy by tovetumab, but also the biologically active agonistic ligand-receptor complex. This work described a novel PD biomarker approach applicable for anti-receptor therapeutics and the first mechanistic model to delineate the in vivo tri-molecular system of a drug, its target receptor, and a competing endogenous ligand, which collectively have been used for optimal dose recommendation supporting clinical development of tovetumab.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales/farmacología , Neoplasias/tratamiento farmacológico , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/aislamiento & purificación , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Monoclonales Humanizados/aislamiento & purificación , Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Biomarcadores Farmacológicos/análisis , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos , Semivida , Humanos , Macaca fascicularis , Ratones , Modelos Biológicos , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo
4.
Cell Rep ; 30(2): 555-570.e7, 2020 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-31940496

RESUMEN

PDGFRα+ mesenchymal progenitor cells are associated with pathological fibro-adipogenic processes. Conversely, a beneficial role for these cells during homeostasis or in response to revascularization and regeneration stimuli is suggested, but remains to be defined. We studied the molecular profile and function of PDGFRα+ cells in order to understand the mechanisms underlying their role in fibrosis versus regeneration. We show that PDGFRα+ cells are essential for tissue revascularization and restructuring through injury-stimulated remodeling of stromal and vascular components, context-dependent clonal expansion, and ultimate removal of pro-fibrotic PDGFRα+-derived cells. Tissue ischemia modulates the PDGFRα+ phenotype toward cells capable of remodeling the extracellular matrix and inducing cell-cell and cell-matrix adhesion, likely favoring tissue repair. Conversely, pathological healing occurs if PDGFRα+-derived cells persist as terminally differentiated mesenchymal cells. These studies support a context-dependent "yin-yang" biology of tissue-resident mesenchymal progenitor cells, which possess an innate ability to limit injury expansion while also promoting fibrosis in an unfavorable environment.


Asunto(s)
Fibrosis/metabolismo , Células Madre Mesenquimatosas/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Animales , Diferenciación Celular/fisiología , Células Cultivadas , Femenino , Fibrosis/patología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Células Madre Mesenquimatosas/patología , Ratones , Ratones Desnudos , Ratones Transgénicos , Músculo Esquelético/citología , Músculo Esquelético/metabolismo
5.
J Ethnopharmacol ; 217: 36-48, 2018 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-29428242

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Bu Shen Yi Sui capsule (BSYSC), based on traditional Chinese formula Liu Wei Di Huang pill, is effective for the treatment of multiple sclerosis (MS) in clinical experience and trials. Our previous studies confirmed that BSYSC had the neuroprotective effect in MS and its animal model, experimental autoimmune encephalomyelitis (EAE); however, its mechanism of action was not clear. Thus, the effect of BSYSC on remyelination and the underlying mechanisms were investigated in the EAE mice. MATERIALS AND METHODS: The EAE model was established by injecting subcutaneously myelin oligodendrocyte protein (MOG) 35-55 in mice. Mice were treated with BSYSC (3.02 g/kg) or vehicle daily by oral gavage for 40 days. The body weight and clinical score of mice were evaluated. Brain was observed by magnetic resonance imaging. The inflammation infiltrate of brain and spinal cord was determined by hematoxylin-eosin staining, while the structure of myelin sheath was visualized by transmission electron microscopy on days 23 and 40 post immunization (dpi), respectively. The protein and mRNA levels of platelets-derived growth factor receptor (PDGFR) α and 2', 3'-cyclic nucleotide-3'-phosphodiesterase (CNPase) were measured by immunohistochemistry, western blot and quantitative real-time polymerase chain reaction. The protein expressions of semaphorins (Sema) 3A, Neuropilin (NRP) - 1, leukemia inhibitory factor (LIF), LIF receptor (LIFR) and Nkx6.2 were further investigated by western blot. RESULTS: BSYSC treatment improved the body weight and clinical score of EAE mice, alleviated inflammatory infiltration and nerve fiber injuries. It also protected the ultrastructural integrity of myelin sheath. BSYSC significantly increased expressions of PDGFRα and CNPase in mice with EAE on 40 dpi. Furthermore, BSYSC treatment increased the expressions of LIF, LIFR and Nkx6.2 and reduced Sema3A and NRP-1 in EAE mice on 40 dpi. CONCLUSIONS: The data demonstrated that BSYSC exhibited the neuroprotective effect against EAE by promoting oligodendrocyte progenitor cells (OPCs) proliferation and differentiation, thus facilitating remyelination. Sema3A/NRP-1, LIF/LIFR and Nkx6.2 are likely contributed to the effects of BSYSC on OPCs.


Asunto(s)
Encéfalo/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Proteínas de Homeodominio/metabolismo , Subunidad alfa del Receptor del Factor Inhibidor de Leucemia/metabolismo , Factor Inhibidor de Leucemia/metabolismo , Vaina de Mielina/efectos de los fármacos , Neuropilina-1/metabolismo , Fármacos Neuroprotectores/farmacología , Semaforina-3A/metabolismo , Médula Espinal/efectos de los fármacos , Factores de Transcripción/metabolismo , 2',3'-Nucleótido Cíclico Fosfodiesterasas/metabolismo , Administración Oral , Animales , Encéfalo/metabolismo , Encéfalo/ultraestructura , Cápsulas , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Medicamentos Herbarios Chinos/administración & dosificación , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Femenino , Ratones Endogámicos C57BL , Vaina de Mielina/metabolismo , Vaina de Mielina/ultraestructura , Glicoproteína Mielina-Oligodendrócito , Fármacos Neuroprotectores/administración & dosificación , Células Precursoras de Oligodendrocitos/efectos de los fármacos , Células Precursoras de Oligodendrocitos/metabolismo , Células Precursoras de Oligodendrocitos/patología , Fragmentos de Péptidos , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal/efectos de los fármacos , Médula Espinal/metabolismo , Médula Espinal/ultraestructura , Factores de Tiempo
6.
Mol Med Rep ; 17(2): 2586-2592, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29207159

RESUMEN

Antioxidant of bamboo leaves (AOB) has been proven to have antioxidant activity and an inhibitory effect on free radicals that induce deterioration of macromolecules. The multi­target regulation of microRNAs (miRs) in the complicated process of vasculogenesis and angiogenesis lead to the use of miRNA therapy in vascular development. In the present study, the role of miRNAs on early embryo vascular development upon AOB stimulation was investigated. For this purpose, mouse embryonic stem cells were spontaneously differentiated as embryoid bodies (EBs) and were examined by phase contrast microscopy. miR­146a mimic and scramble control were transfected into EBs and potential targets of miR­146a were predicted. Cell proliferation and migration were detected by cell viability and wound­healing and migration assays, respectively. Angiogenesis was determined by the Spheroid sprouting assay. It was demonstrated that EBs transfected with miR­146a mimic had an increased growth rate compared with the control cells. miR­146a­transfected cells were very susceptible to AOB treatment. Furthermore, among the predicted miR­146a targets, platelet­derived growth factor receptor alpha (PDGFRA) was identified as a bona fide target of miR­146a. In conclusion, PDGFRA was demonstrated to participate in the modulation of cell migration and proliferation of mouse EBs. The present study expanded the current understanding of AOB biology and elucidated the mechanisms underlying early embryo vascular development upon AOB stimulation.


Asunto(s)
Antioxidantes/farmacología , Cuerpos Embrioides/efectos de los fármacos , Cuerpos Embrioides/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , MicroARNs/genética , Papio , Extractos Vegetales/farmacología , Hojas de la Planta/química , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Animales , Línea Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Células Madre Embrionarias , Expresión Génica , Silenciador del Gen , Genes Reporteros , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt , ARN Interferente Pequeño/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo
7.
J Allergy Clin Immunol Pract ; 6(1): 190-195, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28757367

RESUMEN

BACKGROUND: Glucocorticoids (GCs) are considered first-line treatment for platelet-derived growth factor α (PDGFRA)-negative hypereosinophilic syndromes (HESs). Despite this, little is known about clinical predictors of GC responsiveness in HES. OBJECTIVE: Knowledge of clinical and laboratory predictors of GC response before initiation of GC could lead to more rational selection of subjects with HES for whom earlier institution of second-line and alternative therapies would be appropriate. METHODS: Response to GC, as defined by the reduction of the absolute eosinophil count to below 1000/mm3 and control of symptoms, was assessed by a retrospective chart review of subjects with PDGFRA-negative HES evaluated on an institutional review board-approved protocol. Demographic, clinical, and laboratory parameters obtained before institution of GC, as well as final diagnosis, were evaluated to determine predictors of GC response. Proportional odds models were used for univariate and multivariate assessment of predictors with permutation adjusted P values to correct for multiple comparisons. RESULTS: A total of 164 subjects with PDGFRA-negative HES were categorized according to GC response. Of them, 39% of the subjects responded to low dose (≤10 mg) prednisone, 9% did not respond to GC, and the remainder (52%) had variable responses to GC. The HES subtype diagnosis was the best predictor of response to GC with myeloid forms and lymphocytic variants of HES being the least responsive to GC. CONCLUSIONS: In a large cohort of well-characterized subjects with HES, the odds of response to GC was predicted by HES subtype. Using this model, clinicians may more readily proceed to second-line agents in subjects with confirmed lymphocytic or myeloid forms of HES.


Asunto(s)
Eosinófilos/inmunología , Síndrome Hipereosinofílico/diagnóstico , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores Farmacológicos , Recuento de Células , Niño , Preescolar , Estudios de Cohortes , Resistencia a Medicamentos , Femenino , Glucocorticoides/uso terapéutico , Humanos , Síndrome Hipereosinofílico/tratamiento farmacológico , Masculino , Persona de Mediana Edad , Fenotipo , Pronóstico , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Estudios Retrospectivos , Adulto Joven
8.
EBioMedicine ; 18: 288-299, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28408241

RESUMEN

Maternal vitamin A intake varies but its impact on offspring metabolic health is unknown. Here we found that maternal vitamin A or retinoic acid (RA) administration expanded PDGFRα+ adipose progenitor population in progeny, accompanied by increased blood vessel density and enhanced brown-like (beige) phenotype in adipose tissue, protecting offspring from obesity. Blockage of retinoic acid signaling by either BMS493 or negative RA receptor (RARαDN) over-expression abolished the increase in blood vessel density, adipose progenitor population, and beige adipogenesis stimulated by RA. Furthermore, RA-induced beige adipogenesis was blocked following vascular endothelial growth factor receptor (VEGFR) 2 knock out in PDGFRα+ cells, suggesting its mediatory role. Our data reveal an intrinsic link between maternal retinoid level and offspring health via promoting beige adipogenesis. Thus, enhancing maternal retinoids is an amiable therapeutic strategy to prevent obesity in offspring, especially for those born to obese mothers which account for one third of all pregnancies.


Asunto(s)
Adipogénesis/efectos de los fármacos , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Tretinoina/farmacología , Tejido Adiposo Beige/citología , Tejido Adiposo Beige/metabolismo , Tejido Adiposo Beige/patología , Animales , Temperatura Corporal , Células Cultivadas , Cromatografía Líquida de Alta Presión , Dieta Alta en Grasa , Suplementos Dietéticos , Femenino , Prueba de Tolerancia a la Glucosa , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/patología , Obesidad/prevención & control , Consumo de Oxígeno/efectos de los fármacos , Embarazo , Retinaldehído/sangre , Transducción de Señal/efectos de los fármacos , Células del Estroma/citología , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Tretinoina/sangre , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Vitamina A/sangre , Vitamina A/farmacología
9.
Eur J Cancer ; 69: 158-165, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27821319

RESUMEN

BACKGROUND: Pre-clinical and clinical evidence suggests a rationale for the use of anti-angiogenic agents, including sorafenib, in recurrent and/or metastatic salivary gland carcinomas (RMSGCs). This study evaluates the activity of sorafenib in patients with RMSGCs and also investigates whether the activity of sorafenib could be related to its main tailored targets (i.e. BRAF, vascular endothelial growth factor receptor 2 [VEGFR2], platelet-derived growth factor receptor α [PDGFRα] and ß, RET, KIT). PATIENTS AND METHODS: Patients received sorafenib at 400 mg BID. The primary end-point was response rate (RR) including complete response or partial response (PR); secondary end-points included RR according to Choi criteria, disease control rate (DCR), overall survival (OS), and progression-free survival (PFS). RESULTS: Thirty-seven patients (19 adenoid cystic cancers, ACC) were enrolled. Six PRs were recorded. RR was 16% (95% confidence interval [CI]: 6-32; 11% in ACC and 22% in non-ACC). Choi criteria could be applied in 30 out of 37 cases with a RR of 50% (95% CI: 31-69%); DCR was 76% (95% CI: 59-88%). Incidence of ≥G3 adverse events was 29.7%. Median PFS and OS for the entire population were 5.9 months and 23.4 months, respectively. Median PFS and OS were 8.9 and 26.4 months for ACC versus 4.2 and 12.3 months for non-ACC patients. All the cases showed expression of PDGFRß in the stroma and VEGFR2 in endothelial cells; PDGFRα positivity was found in the stroma of four (27%) cases. All except for two cases showed no PDGFRß, VEGFR2 and PDGFRα expression in the tumour cells. KIT expression was restricted to ACC and a weak RET expression was limited to one adenocarcinoma, not otherwise specified (NOS). No BRAF mutation was found. No correlation was observed between the sorafenib activity and the expression of its markers although all six responders (two ACC, one adenocarcinoma, NOS, one salivary duct cancer [SDC], one high-grade mucoepidermoid [HG-MEC] and one poorly-differentiated cancer) are enriched in the stromal component showing a PDGFRß immunodecoration. In ACCs, immunohistochemistry revealed MYB protein expression in 15/16 cases (94%) and the MYB-NFIB fusion oncogene was observed in 9/14 (64%). CONCLUSIONS: Sorafenib is the first anti-angiogenic agent to demonstrate activity in RMSGC patients, particularly in some histotypes such as HG-MEC, SDC and adenocarcinoma, NOS. The PDGFRß-positive rich stromal component characterising these histotypes and the lack of correlation between the activity of sorafenib and its targets suggests anti-angiogenic effect as the prevalent mechanism of action of sorafenib in SGCs.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/uso terapéutico , Carcinoma Adenoide Quístico/tratamiento farmacológico , Carcinoma Mucoepidermoide/tratamiento farmacológico , Mioepitelioma/tratamiento farmacológico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Niacinamida/análogos & derivados , Compuestos de Fenilurea/uso terapéutico , Neoplasias de las Glándulas Salivales/tratamiento farmacológico , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Adenocarcinoma/secundario , Adulto , Anciano , Carcinoma Adenoide Quístico/metabolismo , Carcinoma Adenoide Quístico/patología , Carcinoma Adenoide Quístico/secundario , Carcinoma Mucoepidermoide/metabolismo , Carcinoma Mucoepidermoide/patología , Carcinoma Mucoepidermoide/secundario , Diarrea/inducido químicamente , Supervivencia sin Enfermedad , Erupciones por Medicamentos/etiología , Fatiga/inducido químicamente , Síndrome Mano-Pie/etiología , Humanos , Hipertensión/inducido químicamente , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Mioepitelioma/metabolismo , Mioepitelioma/patología , Mioepitelioma/secundario , Metástasis de la Neoplasia , Recurrencia Local de Neoplasia/metabolismo , Niacinamida/uso terapéutico , Proteínas Proto-Oncogénicas c-kit/metabolismo , Proteínas Proto-Oncogénicas c-ret/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Neoplasias de las Glándulas Salivales/metabolismo , Neoplasias de las Glándulas Salivales/patología , Sorafenib , Tasa de Supervivencia , Resultado del Tratamiento , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Adulto Joven
10.
Brain Res ; 1639: 28-37, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-26944297

RESUMEN

We investigated the effects of auraptene on mouse oligodendroglial cell lineage in an animal model of demyelination induced by cuprizone. Auraptene, a citrus coumarin, was intraperitoneally administered to mice fed the demyelinating agent cuprizone. Immunohistochemical analysis of the corpus callosum and/or Western blotting analysis of brain extracts revealed that cuprizone reduced immunoreactivity for myelin-basic protein, a marker of myelin, whereas it increased immunoreactivity to platelet derived-growth factor receptor-α, a marker of oligodendrocyte precursor cells. Administration of auraptene enhanced the immunoreactivity to oligodendrocyte transcription factor 2, a marker of oligodendrocyte precursor cells and oligodendrocyte lineage precursor cells, but had no effect on immunoreactivity to myelin-basic protein or platelet-derived growth factor receptor-α. These findings suggest that auraptene promotes the production of oligodendrocyte lineage precursor cells in an animal model of demyelination and may be useful for individuals with demyelinating diseases.


Asunto(s)
Cumarinas/farmacología , Enfermedades Desmielinizantes/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Oligodendroglía/efectos de los fármacos , Células Madre/efectos de los fármacos , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Cuprizona , Enfermedades Desmielinizantes/metabolismo , Enfermedades Desmielinizantes/patología , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Expresión Génica/efectos de los fármacos , Inyecciones Intraperitoneales , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/metabolismo , Proteína Básica de Mielina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Factor de Transcripción 2 de los Oligodendrocitos , Oligodendroglía/metabolismo , Oligodendroglía/patología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Células Madre/fisiología
11.
Lab Chip ; 16(4): 734-42, 2016 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-26775648

RESUMEN

3D organoids using stem cells to study development and disease are now widespread. These models are powerful to mimic in vivo situations but are currently associated with high variability and low throughput. For biomedical research, platforms are thus necessary to increase reproducibility and allow high-throughput screens (HTS). Here, we introduce a microwell platform, integrated in standard culture plates, for functional HTS. Using micro-thermoforming, we form round-bottom microwell arrays from optically clear cyclic olefin polymer films, and assemble them with bottom-less 96-well plates. We show that embryonic stem cells aggregate faster and more reproducibly (centricity, circularity) as compared to a state-of-the-art microwell array. We then run a screen of a chemical library to direct differentiation into primitive endoderm (PrE) and, using on-chip high content imaging (HCI), we identify molecules, including regulators of the cAMP pathway, regulating tissue size, morphology and PrE gene activity. We propose that this platform will benefit to the systematic study of organogenesis in vitro.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Cuerpos Embrioides/citología , Cuerpos Embrioides/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento/métodos , Microtecnología/métodos , Temperatura , Animales , Agregación Celular/efectos de los fármacos , Línea Celular , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Activación Enzimática/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Cinética , Ratones , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Reproducibilidad de los Resultados
12.
Sci Rep ; 5: 11008, 2015 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-26046579

RESUMEN

High-mobility group box 1 (HMGB1) mobilizes platelet-derived growth factor receptor alpha-positive (PDGFRα(+)) mesenchymal cells from bone marrow (BM) into circulation. However, whether HMGB1-induced endogenous PDGFRα(+) mesenchymal cells stimulate skin regeneration has been unclear. Here, we investigated the functions of the HMGB1/BM-PDGFRα(+) mesenchymal cell axis in the regeneration of mouse skin grafts. We found that intravenous HMGB1 administration induced an accumulation of endogenous BM-PDGFRα(+) mesenchymal cells followed by significant inflammatory suppression in the grafts. In contrast, mice with reduced BM-PDGFRα(+) mesenchymal cells showed massive inflammation of the grafts compared to mice that had normal levels of these cells even after HMGB1 administration, suggesting that BM-PDGFRα(+) mesenchymal cells contribute to the HMGB1-induced anti-inflammatory effect. We also found that intravenously administered HMGB1 augmented the local migration of BM-PDGFRα(+) mesenchymal cells from circulation to skin graft by inducing the expression of CXCR4, an SDF-1 receptor, on these cells. Finally, we showed the therapeutic activity of the HMGB1/BM-PDGFRα(+) mesenchymal cell axis in an allergic contact dermatitis model. The results illustrated the contribution of the HMGB1/BM-PDGFRα(+) mesenchymal cell axis in suppressing the inflammation of injured/inflamed skin. These findings may provide future perspectives on the use of HMGB1-based medicines for intractable diseases.


Asunto(s)
Antiinflamatorios/administración & dosificación , Proteína HMGB1/administración & dosificación , Células Madre Mesenquimatosas/fisiología , Animales , Células de la Médula Ósea/fisiología , Movimiento Celular , Células Cultivadas , Dermatitis Alérgica por Contacto/tratamiento farmacológico , Dermatitis Alérgica por Contacto/inmunología , Evaluación Preclínica de Medicamentos , Masculino , Ratones Endogámicos C57BL , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Piel/efectos de los fármacos , Piel/inmunología , Piel/patología , Trasplante de Piel , Regulación hacia Arriba , Cicatrización de Heridas
13.
J Neurooncol ; 122(3): 471-80, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25732621

RESUMEN

Solid tumors arising from malignant transformation of glial cells are one of the leading causes of central nervous system tumor-related death in children. Recurrence in spite of rigorous surgical and chemoradiation therapies remains a major hurdle in management of these tumors. Here, we investigate the efficacy of the second-generation receptor tyrosine kinase inhibitor nilotinib as a therapeutic option for the management of pediatric gliomas. We have utilized two independent pediatric high-grade glioma cell lines with either high platelet-derived growth factor receptor alpha (PDGFRα) or high PDGFRß expression in in vitro assays to investigate the specific downstream effects of nilotinib treatment. Using in vitro cell-based assays we show that nilotinib inhibits PDGF-BB-dependent activation of PDGFRα. We further show that nilotinib is able to decrease cell proliferation and anchorage-independent growth via suppression of AKT and ERK1/2 signaling pathways. Our results suggest that nilotinib may be effective for management of a PDGFRα-dependent group of pediatric gliomas.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioma/patología , Pirimidinas/farmacología , Animales , Becaplermina , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ensayo de Unidades Formadoras de Colonias , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/uso terapéutico , Glioma/tratamiento farmacológico , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Ratones SCID , Proteína Oncogénica v-akt/metabolismo , Proteínas Proto-Oncogénicas c-sis/metabolismo , Pirimidinas/uso terapéutico , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factores de Tiempo , Vinculina/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Methods Mol Biol ; 1263: 43-61, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25618335

RESUMEN

Chemical genomics has the unique potential to expose novel mechanisms of complex cellular biology through screening of small molecules in in vitro assays of a biological phenotype of interest, followed by target identification. In the case of disease-specific assays, the cellular proteins identified might constitute novel drug targets, and the small molecules themselves might be developed as drug leads. In cardiovascular biology, a chemical genomics approach to study the formation of cardiomyocyte, vascular endothelial, and smooth muscle lineages might contribute to therapeutic regeneration. Here, we describe methods used to develop high content screening assays implementing multipotent cardiovascular progenitors derived from human pluripotent stem cells and have identified novel compounds that direct cardiac differentiation.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos/métodos , Ensayos Analíticos de Alto Rendimiento , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/efectos de los fármacos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Técnicas de Cultivo de Célula , Línea Celular , Separación Celular/métodos , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Fenotipo , Células Madre Pluripotentes/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo
15.
Lasers Med Sci ; 29(6): 1867-74, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24870411

RESUMEN

Telocytes (TCs) are a brand-new cell type frequently observed in the interstitial space of many organs (see www.telocytes.com ). TCs are defined by very long (tens of micrometers) and slender prolongations named telopodes. At their level, dilations-called podoms (~300 nm), alternate with podomers (80-100 nm). TCs were identified in a myometrial interstitial cell culture based on morphological criteria and by CD34 and PDGF receptor alpha (PDGFRα) immunopositivity. However, the mechanism(s) of telopodes formation and/or elongation and ramification is not known. We report here the low-level laser stimulation (LLLS) using a 1,064-nm neodymium-doped yttrium aluminum garnet (Nd:YAG) laser (with an output power of 60 mW) of the telopodal lateral extension (TLE) growth in cell culture. LLLS of TCs determines a higher growth rate of TLE in pregnant myometrium primary cultures (10.3 ± 1.0 µm/min) compared to nonpregnant ones (6.6 ± 0.9 µm/min). Acute exposure (30 min) of TCs from pregnant myometrium to 1 µM mibefradil, a selective inhibitor of T-type calcium channels, determines a significant reduction in the LLLS TLE growth rate (5.7 ± 0.8 µm/min) compared to LLLS per se in same type of samples. Meanwhile, chronic exposure (24 h) completely abolishes the LLLS TLE growth in both nonpregnant and pregnant myometria. The initial direction of TLE growth was modified by LLLS, the angle of deviation being more accentuated in TCs from human pregnant myometrium than in TCs from nonpregnant myometrium. In conclusion, TCs from pregnant myometrium are more susceptible of reacting to LLLS than those from nonpregnant myometrium. Therefore, some implications are emerging for low-level laser therapy (LLLT) in uterine regenerative medicine.


Asunto(s)
Terapia por Luz de Baja Intensidad/métodos , Miometrio/patología , Telocitos/patología , Adulto , Antígenos CD34/metabolismo , Biopsia , Femenino , Humanos , Histerectomía , Láseres de Estado Sólido , Persona de Mediana Edad , Miometrio/efectos de la radiación , Neodimio/química , Embarazo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Telocitos/efectos de la radiación , Útero/efectos de la radiación
16.
ASN Neuro ; 5(1): e00107, 2013 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-23368675

RESUMEN

NSC (neural stem cells)/NPC (neural progenitor cells) are multipotent and self-renew throughout adulthood in the SVZ (subventricular zone) of the mammalian CNS (central nervous system). These cells are considered interesting targets for CNS neurodegenerative disorder cell therapies, and understanding their behaviour in vitro is crucial if they are to be cultured prior to transplantation. We cultured the SVZ tissue belonging to newborn rats under the form of NS (neurospheres) to evaluate the effects of Tf (transferrin) on cell proliferation. The NS were heterogeneous in terms of the NSC/NPC markers GFAP (glial fibrillary acidic protein), Nestin and Sox2 and the OL (oligodendrocyte) progenitor markers NG2 (nerve/glia antigen 2) and PDGFRα (platelet-derived growth factor receptor α). The results of this study indicate that aTf (apoTransferrin) is able to increase cell proliferation of SVZ-derived cells in vitro, and that these effects were mediated at least in part by the TfRc1 (Tf receptor 1). Since OPCs (oligodendrocyte progenitor cells) represent a significant proportion of the proliferating cells in the SVZ-derived primary cultures, we used the immature OL cell line N20.1 to show that Tf was able to augment the proliferation rate of OPC, either by adding aTf to the culture medium or by overexpressing rat Tf in situ. The culture medium supplemented with ferric iron, together with aTf, increased the DNA content, while ferrous iron did not. The present work provides data that could have a potential application in human cell replacement therapies for neurodegenerative disease and/or CNS injury that require the use of in vitro amplified NPCs.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Ventrículos Laterales/citología , Células-Madre Neurales/efectos de los fármacos , Oligodendroglía/efectos de los fármacos , Transferrina/farmacología , Animales , Animales Recién Nacidos , Antígenos/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Bromodesoxiuridina/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteínas de Filamentos Intermediarios/metabolismo , Hierro/administración & dosificación , Ventrículos Laterales/crecimiento & desarrollo , Masculino , Proteínas del Tejido Nervioso/metabolismo , Nestina , Factor de Transcripción 2 de los Oligodendrocitos , Proteoglicanos/metabolismo , Ratas , Ratas Wistar , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factores de Transcripción SOXB1/metabolismo , Transferrina/metabolismo
17.
Gastroenterology ; 144(1): 134-144.e6, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23041331

RESUMEN

BACKGROUND & AIMS: A fraction of gastrointestinal stromal tumor (GIST) cells overexpress the platelet-derived growth factor receptor (PDGFR)A, although most overexpress KIT. It is not known if this is because these receptor tyrosine kinases have complementary oncogenic potential, or because of heterogeneity in the cellular origin of GIST. Little also is known about why Hedgehog (HH) signaling is activated in some GIST. HH binds to and inactivates the receptor protein patched homolog (PTCH). METHODS: Ptch was conditionally inactivated in mice (to achieve constitutive HH signaling) using a Cre recombinase regulated by the lysozyme M promoter. Cre-expressing cells were traced using R26R-LacZ reporter mice. Tumors were characterized by in situ hybridization, immunohistochemistry, immunoblot, and quantitative reverse-transcriptase polymerase chain reaction analyses. Cell transformation was assessed by soft agar assay. RESULTS: Loss of Ptch from lysozyme M-expressing cells resulted in the development of tumors of GIST-like localization and histology; these were reduced when mice were given imatinib, a drug that targets KIT and PDGFRA. The Hh signaling pathway was activated in the tumor cells, and Pdgfrα, but not Kit, was overexpressed and activated. Lineage tracing revealed that Cre-expressing intestinal cells were Kit-negative. These cells sometimes expressed Pdgfrα and were located near Kit-positive interstitial cells of Cajal. In contrast to KIT, activation of PDGFRA increased anchorage-independent proliferation and was required for tumor formation in mice by cells with activated HH signaling. CONCLUSIONS: Inactivation of Ptch in mice leads to formation of GIST-like tumors that express Pdgfrα, but not Kit. Activation of Pdgfrα signaling appears to facilitate tumorigenesis.


Asunto(s)
Neoplasias Gastrointestinales/genética , Tumores del Estroma Gastrointestinal/metabolismo , Proteínas Hedgehog/genética , Leiomiosarcoma/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptores de Superficie Celular/genética , Animales , Benzamidas , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Neoplasias Gastrointestinales/tratamiento farmacológico , Neoplasias Gastrointestinales/metabolismo , Neoplasias Gastrointestinales/patología , Tumores del Estroma Gastrointestinal/tratamiento farmacológico , Tumores del Estroma Gastrointestinal/genética , Tumores del Estroma Gastrointestinal/patología , Expresión Génica , Genotipo , Proteínas Hedgehog/metabolismo , Humanos , Mesilato de Imatinib , Integrasas/genética , Integrasas/metabolismo , Mucosa Intestinal/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Leiomiosarcoma/metabolismo , Ratones , Muramidasa/genética , Muramidasa/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores Patched , Receptor Patched-1 , Piperazinas/uso terapéutico , Regiones Promotoras Genéticas , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirimidinas/uso terapéutico , Receptores de Superficie Celular/metabolismo , Transducción de Señal/genética , Proteína con Dedos de Zinc GLI1 , Proteína Gli2 con Dedos de Zinc , Proteína Gli3 con Dedos de Zinc
18.
Cancer Res ; 72(17): 4515-25, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22787122

RESUMEN

Akt activation by the IGF-1 receptor (IGF-1R) has been posited to be a mechanism of intrinsic resistance to mTORC1 inhibitors (rapalogues) for sarcomas. Here we show that rapamycin-induced phosphorylation of Akt can occur in an IGF-1R-independent manner. Analysis of synovial sarcoma cell lines showed that either IGF-1R or the PDGF receptor alpha (PDGFRA) can mediate intrinsic resistance to rapamycin. Repressing expression of PDGFRA or inhibiting its kinase activity in synovial sarcoma cells blocked rapamycin-induced phosphorylation of Akt and decreased tumor cell viability. Expression profiling of clinical tumor samples revealed that PDGFRA was the most highly expressed kinase gene among several sarcoma disease subtypes, suggesting that PDGFRA may be uniquely significant for synovial sarcomas. Tumor biopsy analyses from a synovial sarcoma patient treated with the mTORC1 inhibitor everolimus and PDGFRA inhibitor imatinib mesylate confirmed that this drug combination can impact both mTORC1 and Akt signals in vivo. Together, our findings define mechanistic variations in the intrinsic resistance of synovial sarcomas to rapamycin and suggest therapeutic strategies to address them.


Asunto(s)
Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Sarcoma Sinovial/metabolismo , Sirolimus/farmacología , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Antineoplásicos/farmacología , Benzamidas , Línea Celular Tumoral , Activación Enzimática/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Humanos , Mesilato de Imatinib , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Terapia Molecular Dirigida , Complejos Multiproteicos , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Piperazinas/farmacología , Proteínas/antagonistas & inhibidores , Proteínas/metabolismo , Pirimidinas/farmacología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Sarcoma Sinovial/tratamiento farmacológico , Sarcoma Sinovial/genética , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR , Transcripción Genética
20.
Am J Physiol Cell Physiol ; 302(11): C1590-8, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22322974

RESUMEN

Wound healing is a complex dynamic process involving a variety of cell types, including fibroblasts that express and respond to cytokines and growth factors in the local microenvironment. The mechanisms controlling gene expression after injury at a transcriptional level are poorly understood. Here we show that decreased expression of a key receptor, PDGF-receptor (R)-α, after fibroblast injury is due to the release and paracrine activity of TNF-α. TNF-α inhibits PDGF-R-α expression and this involves formation of a c-Fos-Yin Yang 1 (YY1) complex and histone deacetylase (HDAC) activity. c-Fos, induced by TNF-α, negatively regulates PDGF-R-α transcription. Small interfering RNA (siRNA) targeting c-Fos or the zinc finger transcription factor YY1 inhibits TNF-α suppression of PDGF-R-α expression. Coimmunoprecipitation studies show that TNF-α stimulates the formation of a complex between c-Fos with YY1. Furthermore, chromatin immunoprecipitation (ChIP) analysis reveals the enrichment of c-Fos, YY1, and HDAC-1 at the PDGF-R-α promoter in cells exposed to TNF-α. With suberoylanilide hydroxamic acid (SAHA) and HDAC-1 siRNA, we demonstrate that HDAC mediates TNF-α repression of PDGF-R-α. These findings demonstrate that transcriptional repression of PDGF-R-α after fibroblast injury involves paracrine activity of endogenous TNF-α, the formation of a c-Fos-YY1 complex, and negative regulatory activity by HDAC.


Asunto(s)
Histona Desacetilasas/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Transcripción YY1/metabolismo , Animales , Secuencia de Bases , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Histona Desacetilasas/genética , Ácidos Hidroxámicos/metabolismo , Ratones , Datos de Secuencia Molecular , Células 3T3 NIH , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-fos/genética , Interferencia de ARN , ARN Interferente Pequeño , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Secuencias Reguladoras de Ácidos Nucleicos , Análisis de Secuencia de ADN , Vorinostat , Factor de Transcripción YY1/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA