Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
3.
Brain ; 139(Pt 6): 1762-82, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27084575

RESUMEN

The p75 neurotrophin receptor is important in multiple physiological actions including neuronal survival and neurite outgrowth during development, and after central nervous system injury. We have discovered a novel piperazine-derived compound, EVT901, which interferes with p75 neurotrophin receptor oligomerization through direct interaction with the first cysteine-rich domain of the extracellular region. Using ligand binding assays with cysteine-rich domains-fused p75 neurotrophin receptor, we confirmed that EVT901 interferes with oligomerization of full-length p75 neurotrophin receptor in a dose-dependent manner. Here we report that EVT901 reduces binding of pro-nerve growth factor to p75 neurotrophin receptor, blocks pro-nerve growth factor induced apoptosis in cells expressing p75 neurotrophin receptor, and enhances neurite outgrowth in vitro Furthermore, we demonstrate that EVT901 abrogates p75 neurotrophin receptor signalling by other ligands, such as prion peptide and amyloid-ß. To test the efficacy of EVT901 in vivo, we evaluated the outcome in two models of traumatic brain injury. We generated controlled cortical impacts in adult rats. Using unbiased stereological analysis, we found that EVT901 delivered intravenously daily for 1 week after injury, reduced lesion size, protected cortical neurons and oligodendrocytes, and had a positive effect on neurological function. After lateral fluid percussion injury in adult rats, oral treatment with EVT901 reduced neuronal death in the hippocampus and thalamus, reduced long-term cognitive deficits, and reduced the occurrence of post-traumatic seizure activity. Together, these studies provide a new reagent for altering p75 neurotrophin receptor actions after injury and suggest that EVT901 may be useful in treatment of central nervous system trauma and other neurological disorders where p75 neurotrophin receptor signalling is affected.


Asunto(s)
Oligodendroglía/efectos de los fármacos , Piperazinas/farmacología , Receptor de Factor de Crecimiento Nervioso/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/patología , Recuento de Células , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Enfermedades Desmielinizantes/patología , Relación Dosis-Respuesta a Droga , Humanos , Masculino , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Oligodendroglía/metabolismo , Fosforilación/efectos de los fármacos , Cultivo Primario de Células , Ensayo de Unión Radioligante , Ratas , Receptor de Factor de Crecimiento Nervioso/biosíntesis , Receptor trkA/metabolismo , Recuperación de la Función
4.
Biosens Bioelectron ; 78: 58-66, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26594887

RESUMEN

Detection of thrombin in plasma raises timely challenges to enable therapeutic management of thrombosis in patients under vital threat. Thrombin binding aptamers represent promising candidates as sensing elements for the development of real-time thrombin biosensors; however implementation of such biosensor requires the clear understanding of thrombin-aptamer interaction properties in real-like environment. In this study, we used Surface Plasmon Resonance technique to answer the questions of specificity and sensitivity of thrombin detection by the thrombin-binding aptamers HD1, NU172 and HD22. We systematically characterized their properties in the presence of thrombin, as well as interfering molecular species such as the thrombin precursor prothrombin, thrombin in complex with some of its natural inhibitors, nonspecific serum proteins, and diluted plasma. Kinetic experiments show the multiple binding modes of HD1 and NU172, which both interact with multiple sites of thrombin with low nanomolar affinities and show little specificity of interaction for prothrombin vs. thrombin. HD22, on the other hand, binds specifically to thrombin exosite II and has no affinity to prothrombin at all. While thrombin in complex with some of its inhibitors could not be recognized by any aptamer, the binding of HD1 and NU172 properties is compromised by thrombin inhibitors alone, as well as with serum albumin. Finally, the complex nature of plasma was overwhelming for HD1, but we define conditions for the thrombin detection at 10nM range in 100-fold diluted plasma by HD22. Consequently HD22 showed key advantage over HD1 and NU172, and appears as the only alternative to design an aptasensor.


Asunto(s)
Aptámeros de Nucleótidos/química , Técnicas Biosensibles/métodos , Trombina/aislamiento & purificación , Trombosis/diagnóstico , Animales , Sitios de Unión , Humanos , Ratones , Plasma/química , Unión Proteica , Albúmina Sérica/química , Resonancia por Plasmón de Superficie , Trombina/química , Trombosis/patología
5.
J Biol Chem ; 290(6): 3405-17, 2015 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-25384978

RESUMEN

The formation of new vessels in the tumor, termed angiogenesis, is essential for primary tumor growth and facilitates tumor invasion and metastasis. Hypoxia has been described as one trigger of angiogenesis. Indeed, hypoxia, which is characterized by areas of low oxygen levels, is a hallmark of solid tumors arising from an imbalance between oxygen delivery and consumption. Hypoxic conditions have profound effects on the different components of the tumoral environment. For example, hypoxia is able to activate endothelial cells, leading to angiogenesis but also thereby initiating a cascade of reactions involving neutrophils, smooth muscle cells, and fibroblasts. In addition, hypoxia directly regulates the expression of many genes for which the role and the importance in the tumoral environment remain to be completely elucidated. In this study, we used a method to selectively label sialoglycoproteins to identify new membrane and secreted proteins involved in the adaptative process of endothelial cells by mass spectrometry-based proteomics. We used an in vitro assay under hypoxic condition to observe an increase of protein expression or modifications of glycosylation. Then the function of the identified proteins was assessed in a vasculogenesis assay in vivo by using a morpholino strategy in zebrafish. First, our approach was validated by the identification of sialoglycoproteins such as CD105, neuropilin-1, and CLEC14A, which have already been described as playing key roles in angiogenesis. Second, we identified several new proteins regulated by hypoxia and demonstrated for the first time the pivotal role of GLUT-1, TMEM16F, and SDF4 in angiogenesis.


Asunto(s)
Neovascularización Fisiológica , Procesamiento Proteico-Postraduccional , Sialoglicoproteínas/metabolismo , Adaptación Fisiológica , Animales , Anoctaminas , Antígenos CD/genética , Antígenos CD/metabolismo , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Hipoxia de la Célula , Endoglina , Transportador de Glucosa de Tipo 1/genética , Transportador de Glucosa de Tipo 1/metabolismo , Glicoproteínas/genética , Glicoproteínas/metabolismo , Glicosilación , Células Endoteliales de la Vena Umbilical Humana , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Neuropilina-1/genética , Neuropilina-1/metabolismo , Proteínas de Transferencia de Fosfolípidos/genética , Proteínas de Transferencia de Fosfolípidos/metabolismo , Proteoma/química , Proteoma/metabolismo , Proteómica/métodos , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Sialoglicoproteínas/genética , Pez Cebra
6.
J Cell Physiol ; 230(1): 43-51, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24760775

RESUMEN

Tumor angiogenesis is accompanied by vasculogenesis, which is involved in the differentiation and mobilization of human bone marrow cells. In order to further characterize the role of vasculogenesis in the tumor growth process, the effects of FGF2 on the differentiation of human bone marrow AC133(+) cells (BM-AC133(+)) into vascular precursors were studied in vitro. FGF2, like VEGFA, induced progenitor cell differentiation into cell types with endothelial cell characteristics. SSR128129E, a newly discovered specific FGFR antagonist acting by allosteric interaction with FGFR, abrogated FGF2-induced endothelial cell differentiation, showing that FGFR signaling is essential during this process. To assess the involvement of the FGF/FRGR signaling in vivo, the pre-clinical model of Lewis lung carcinoma (LL2) in mice was used. Subcutaneous injection of LL2 cells into mice induced an increase of circulating EPCs from peripheral blood associated with tumor growth and an increase of intra-tumoral vascular index. Treatment with the FGFR antagonist SSR128129E strongly decreased LL2 tumor growth as well as the intra-tumoral vascular index (41% and 50% decrease vs. vehicle-treated mice respectively, P < 0.01). Interestingly, SSR128129E treatment significantly decreased the number of circulating EPCs from the peripheral blood (53% inhibition vs. vehicle-treated mice, P < 0.01). These results demonstrate for the first time that the blockade of the FGF/FGFR pathway by SSR128129E reduces EPC recruitment during angiogenesis-dependent tumor growth. In this context, circulating EPCs could be a reliable surrogate marker for tumor growth and angiogenic activity.


Asunto(s)
Carcinoma Pulmonar de Lewis/irrigación sanguínea , Células Madre Hematopoyéticas/citología , Indolizinas/farmacología , Neovascularización Patológica/patología , Receptores de Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , ortoaminobenzoatos/farmacología , Antígeno AC133 , Animales , Antígenos CD/biosíntesis , Células de la Médula Ósea/metabolismo , Adhesión Celular , Diferenciación Celular/efectos de los fármacos , Línea Celular , Movimiento Celular , Células Endoteliales/citología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Glicoproteínas/biosíntesis , Humanos , Ratones , Ratones Endogámicos C57BL , Péptidos , ARN Mensajero/biosíntesis , Receptores de Factores de Crecimiento de Fibroblastos/genética , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal
7.
PLoS One ; 9(11): e113215, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25405900

RESUMEN

RGS18 is a myeloerythroid lineage-specific regulator of G-protein signaling, highly expressed in megakaryocytes (MKs) and platelets. In the present study, we describe the first generation of a RGS18 knockout mouse model (RGS18-/-). Interesting phenotypic differences between RGS18-/- and wild-type (WT) mice were identified, and show that RGS18 plays a significant role in both platelet generation and function. RGS18 deficiency produced a gain of function phenotype in platelets. In resting platelets, the level of CD62P expression was increased in RGS18-/- mice. This increase correlated with a higher level of plasmatic serotonin concentration. RGS18-/- platelets displayed a higher sensitivity to activation in vitro. RGS18 deficiency markedly increased thrombus formation in vivo. In addition, RGS18-/- mice presented a mild thrombocytopenia, accompanied with a marked deficit in MK number in the bone marrow. Analysis of MK maturation in vitro and in vivo revealed a defective megakaryopoiesis in RGS18-/- mice, with a lower bone marrow content of only the most committed MK precursors. Finally, RGS18 deficiency was correlated to a defect of platelet recovery in vivo under acute conditions of thrombocytopenia. Thus, we highlight a role for RGS18 in platelet generation and function, and provide additional insights into the physiology of RGS18.


Asunto(s)
Megacariocitos/metabolismo , Activación Plaquetaria/fisiología , Proteínas RGS/genética , Proteínas RGS/metabolismo , Transducción de Señal/fisiología , Análisis de Varianza , Animales , Recuento de Células Sanguíneas , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Filogenia , Activación Plaquetaria/genética , Regiones Promotoras Genéticas/genética , Serotonina/sangre , Transducción de Señal/genética , Trombosis/metabolismo
8.
Chem Biol ; 21(10): 1310-1317, 2014 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-25200605

RESUMEN

Angiogenesis contributes to the development of numerous disorders. Even though fibroblast growth factors (FGFs) were discovered as mediators of angiogenesis more than 30 years ago, their role in developmental angiogenesis still remains elusive. We use a recently described chemical probe, SSR128129E (SSR), that selectively inhibits the action of multiple FGF receptors (FGFRs), in combination with the zebrafish model to examine the role of FGF signaling in vascular development. We observe that while FGFR signaling is less important for vessel guidance, it affects vascular outgrowth and is especially required for the maintenance of blood vessel integrity by ensuring proper cell-cell junctions between endothelial cells. In conclusion, our work illustrates the power of a small molecule probe to reveal insights into blood vessel formation and stabilization and thus of broad interest to the vascular biology community.


Asunto(s)
Vasos Sanguíneos/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Proteínas de Pez Cebra/metabolismo , Aletas de Animales/fisiología , Animales , Vasos Sanguíneos/efectos de los fármacos , Cadherinas/metabolismo , Embrión no Mamífero/metabolismo , Indolizinas/química , Indolizinas/metabolismo , Indolizinas/farmacología , Uniones Intercelulares/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Receptores de Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Regeneración , Transducción de Señal/efectos de los fármacos , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo , Proteínas de Pez Cebra/antagonistas & inhibidores , ortoaminobenzoatos/química , ortoaminobenzoatos/metabolismo , ortoaminobenzoatos/farmacología
9.
J Med Chem ; 57(17): 7293-316, 2014 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-25075638

RESUMEN

In the search of a potential backup for clopidogrel, we have initiated a HTS campaign designed to identify novel reversible P2Y12 antagonists. Starting from a hit with low micromolar binding activity, we report here the main steps of the optimization process leading to the identification of the preclinical candidate SAR216471. It is a potent, highly selective, and reversible P2Y12 receptor antagonist and by far the most potent inhibitor of ADP-induced platelet aggregation among the P2Y12 antagonists described in the literature. SAR216471 displays potent in vivo antiplatelet and antithrombotic activities and has the potential to differentiate from other antiplatelet agents.


Asunto(s)
Indoles/farmacología , Inhibidores de Agregación Plaquetaria/farmacología , Agregación Plaquetaria/efectos de los fármacos , Antagonistas del Receptor Purinérgico P2/farmacología , Piridazinas/farmacología , Receptores Purinérgicos P2Y12/metabolismo , Síndrome Coronario Agudo/prevención & control , Adenosina Difosfato/farmacología , Administración Oral , Animales , Unión Competitiva , Células CHO , Cricetinae , Cricetulus , Humanos , Indoles/síntesis química , Indoles/metabolismo , Inyecciones Intravenosas , Masculino , Modelos Químicos , Estructura Molecular , Inhibidores de Agregación Plaquetaria/síntesis química , Inhibidores de Agregación Plaquetaria/metabolismo , Antagonistas del Receptor Purinérgico P2/síntesis química , Antagonistas del Receptor Purinérgico P2/metabolismo , Piridazinas/síntesis química , Piridazinas/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Purinérgicos P2Y12/genética , Trombosis/prevención & control
10.
J Med Chem ; 56(23): 9441-56, 2013 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-24175584

RESUMEN

Compound 15 (SAR107375), a novel potent dual thrombin and factor Xa inhibitor resulted from a rational optimization process. Starting from compound 14, with low factor Xa and modest anti-thrombin inhibitory activities (IC50's of 3.5 and 0.39 µM, respectively), both activities were considerably improved, notably through the incorporation of a neutral chlorothiophene P1 fragment and tuning of P2 and P3-P4 fragments. Final optimization of metabolic stability with microsomes led to the identification of 15, which displays strong activity in vitro vs factor Xa and thrombin (with Ki's of 1 and 8 nM, respectively). In addition 15 presents good selectivity versus related serine proteases (roughly 300-fold), including trypsin (1000-fold), and is very active (0.39 µM) in the thrombin generation time (TGT) coagulation assay in human platelet rich plasma (PRP). Potent in vivo activity in a rat model of venous thrombosis following iv and, more importantly, po administration was also observed (ED50 of 0.07 and 2.8 mg/kg, respectively). Bleeding liability was reduced in the rat wire coil model, more relevant to arterial thrombosis, with 15 (blood loss increase of 2-fold relative to the ED80 value) compared to rivaroxaban 2 and dabigatran etexilate 1a.


Asunto(s)
Anticoagulantes/síntesis química , Inhibidores del Factor Xa , Fibrinolíticos/síntesis química , Piperazinas/síntesis química , Sulfonamidas/síntesis química , Trombina/antagonistas & inhibidores , Animales , Anticoagulantes/farmacología , Cristalografía por Rayos X , Diseño de Fármacos , Fibrinolíticos/farmacología , Humanos , Piperazinas/farmacología , Piperazinas/uso terapéutico , Ratas , Sulfonamidas/farmacología , Sulfonamidas/uso terapéutico , Trombosis de la Vena/tratamiento farmacológico
11.
PLoS One ; 8(7): e70479, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23936211

RESUMEN

Radiation therapy in the treatment of cancer is dose limited by radiation injury in normal tissues such as the intestine and the heart. To identify the mechanistic involvement of transforming growth factor-beta 1 (TGF-ß1) in intestinal and cardiac radiation injury, we studied the influence of pharmacological induction of TGF-ß1 with xaliproden (SR 57746A) in rat models of radiation enteropathy and radiation-induced heart disease (RIHD). Because it was uncertain to what extent TGF-ß induction may enhance radiation injury in heart and intestine, animals were exposed to irradiation schedules that cause mild to moderate (acute) radiation injury. In the radiation enteropathy model, male Sprague-Dawley rats received local irradiation of a 4-cm loop of rat ileum with 7 once-daily fractions of 5.6 Gy, and intestinal injury was assessed at 2 weeks and 12 weeks after irradiation. In the RIHD model, male Sprague-Dawley rats received local heart irradiation with a single dose of 18 Gy and were followed for 6 months after irradiation. Rats were treated orally with xaliproden starting 3 days before irradiation until the end of the experiments. Treatment with xaliproden increased circulating TGF-ß1 levels by 300% and significantly induced expression of TGF-ß1 and TGF-ß1 target genes in the irradiated intestine and heart. Various radiation-induced structural changes in the intestine at 2 and 12 weeks were significantly enhanced with TGF-ß1 induction. Similarly, in the RIHD model induction of TGF-ß1 augmented radiation-induced changes in cardiac function and myocardial fibrosis. These results lend further support for the direct involvement of TGF-ß1 in biological mechanisms of radiation-induced adverse remodeling in the intestine and the heart.


Asunto(s)
Corazón/efectos de los fármacos , Intestinos/efectos de los fármacos , Naftalenos/efectos adversos , Piridinas/efectos adversos , Traumatismos Experimentales por Radiación/inducido químicamente , Factor de Crecimiento Transformador beta1/agonistas , Animales , Biomarcadores/metabolismo , Rayos gamma , Expresión Génica/efectos de los fármacos , Expresión Génica/efectos de la radiación , Corazón/fisiopatología , Corazón/efectos de la radiación , Inflamación/inducido químicamente , Inflamación/metabolismo , Inflamación/patología , Mucosa Intestinal/metabolismo , Intestinos/patología , Intestinos/efectos de la radiación , Masculino , Dosis de Radiación , Traumatismos Experimentales por Radiación/metabolismo , Traumatismos Experimentales por Radiación/patología , Ratas , Ratas Sprague-Dawley , Factor de Crecimiento Transformador beta1/sangre
12.
Cancer Cell ; 23(4): 489-501, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23597563

RESUMEN

The fibroblast growth factor (FGF)/fibroblast growth factor receptor (FGFR) signaling network plays an important role in cell growth, survival, differentiation, and angiogenesis. Deregulation of FGFR signaling can lead to cancer development. Here, we report an FGFR inhibitor, SSR128129E (SSR), that binds to the extracellular part of the receptor. SSR does not compete with FGF for binding to FGFR but inhibits FGF-induced signaling linked to FGFR internalization in an allosteric manner, as shown by crystallography studies, nuclear magnetic resonance, Fourier transform infrared spectroscopy, molecular dynamics simulations, free energy calculations, structure-activity relationship analysis, and FGFR mutagenesis. Overall, SSR is a small molecule allosteric inhibitor of FGF/FGFR signaling, acting via binding to the extracellular part of the FGFR.


Asunto(s)
Inhibidores de Proteínas Quinasas/farmacología , Receptores de Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Regulación Alostérica/efectos de los fármacos , Unión Competitiva , Procesos de Crecimiento Celular/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Conformación Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad
13.
Nat Methods ; 10(1): 77-83, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23202434

RESUMEN

Lineage conversion of one somatic cell type to another is an attractive approach for generating specific human cell types. Lineage conversion can be direct, in the absence of proliferation and multipotent progenitor generation, or indirect, by the generation of expandable multipotent progenitor states. We report the development of a reprogramming methodology in which cells transition through a plastic intermediate state, induced by brief exposure to reprogramming factors, followed by differentiation. We use this approach to convert human fibroblasts to mesodermal progenitor cells, including by non-integrative approaches. These progenitor cells demonstrated bipotent differentiation potential and could generate endothelial and smooth muscle lineages. Differentiated endothelial cells exhibited neo-angiogenesis and anastomosis in vivo. This methodology for indirect lineage conversion to angioblast-like cells adds to the armamentarium of reprogramming approaches aimed at the study and treatment of ischemic pathologies.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Reprogramación Celular , Endotelio Vascular/citología , Fibroblastos/citología , Miocitos del Músculo Liso/citología , Células Madre/citología , Animales , Biomarcadores/metabolismo , Western Blotting , Movimiento Celular , Proliferación Celular , Células Cultivadas , Endotelio Vascular/metabolismo , Fibroblastos/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Ratones , Miocitos del Músculo Liso/metabolismo , Neovascularización Fisiológica , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/metabolismo
14.
Protein Cell ; 3(12): 934-42, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23161332

RESUMEN

Articular cartilage, which is mainly composed of collagen II, enables smooth skeletal movement. Degeneration of collagen II can be caused by various events, such as injury, but degeneration especially increases over the course of normal aging. Unfortunately, the body does not fully repair itself from this type of degeneration, resulting in impaired movement. Microfracture, an articular cartilage repair surgical technique, has been commonly used in the clinic to induce the repair of tissue at damage sites. Mesenchymal stem cells (MSC) have also been used as cell therapy to repair degenerated cartilage. However, the therapeutic outcomes of all these techniques vary in different patients depending on their age, health, lesion size and the extent of damage to the cartilage. The repairing tissues either form fibrocartilage or go into a hypertrophic stage, both of which do not reproduce the equivalent functionality of endogenous hyaline cartilage. One of the reasons for this is inefficient chondrogenesis by endogenous and exogenous MSC. Drugs that promote chondrogenesis could be used to induce self-repair of damaged cartilage as a non-invasive approach alone, or combined with other techniques to greatly assist the therapeutic outcomes. The recent development of human induced pluripotent stem cell (iPSCs), which are able to self-renew and differentiate into multiple cell types, provides a potentially valuable cell resource for drug screening in a "more relevant" cell type. Here we report a screening platform using human iPSCs in a multi-well plate format to identify compounds that could promote chondrogenesis.


Asunto(s)
Condrogénesis/efectos de los fármacos , Evaluación Preclínica de Medicamentos/métodos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Diferenciación Celular/efectos de los fármacos , Condrocitos/citología , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Genes Reporteros/genética , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Queratinocitos/citología , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Luciferasas/genética , Péptidos/síntesis química , Péptidos/metabolismo , Reproducibilidad de los Resultados
15.
Blood ; 115(16): 3390-7, 2010 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-20139094

RESUMEN

Growth arrest-specific gene 6 (Gas6) is expressed in antigen-presenting cells and endothelial cells (ECs) but not in T cells. When wild-type (WT) or Gas6(-/-) mice received allogeneic non-T cell-depleted bone marrow cells, hepatic graft-versus-host disease (GVHD) was alleviated in Gas6(-/-) recipients regardless of donor genotype, but not in WT recipients. T-cell infiltration was more prominent and diffuse in WT than in Gas6(-/-) recipients' liver. When mice received 0.5 x 10(6) allogeneic T cells with T cell-depleted allogeneic bone marrow, clinical signs indicated that GVHD was less severe in Gas6(-/-) than in WT recipients, as shown by a significant improvement of the survival and reduced liver GVHD. These data demonstrate that donor cells were not involved in the protection mechanism. In addition, lack of Gas6 in antigen-presenting cells did not affect WT or Gas6(-/-) T-cell proliferation. We therefore assessed the response of WT or Gas6(-/-) ECs to tumor necrosis factor-alpha. Lymphocyte transmigration was less extensive through Gas6(-/-) than WT ECs and was not accompanied by increases in adhesion molecule levels. Thus, the lack of Gas6 in ECs impaired donor T-cell transmigration into the liver, providing a rationale for considering Gas6 pathway as a potential nonimmunosuppressive target to minimize GVHD in patients receiving allogeneic hematopoietic stem cell transplantation.


Asunto(s)
Enfermedad Injerto contra Huésped/prevención & control , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Hígado/inmunología , Animales , Separación Celular , Quimiotaxis de Leucocito/genética , Quimiotaxis de Leucocito/inmunología , Células Endoteliales/metabolismo , Citometría de Flujo , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/inmunología , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intercelular/genética , Hígado/patología , Activación de Linfocitos/inmunología , Prueba de Cultivo Mixto de Linfocitos , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Trasplante Homólogo
16.
Hypertension ; 54(3): 676-83, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19597037

RESUMEN

Recent advances in basic and clinical research have identified Rho kinase as an important target potentially implicated in a variety of cardiovascular diseases. Rho kinase is a downstream mediator of RhoA that leads to stress fiber formation, membrane ruffling, smooth muscle contraction, and cell motility. Increased Rho-kinase activity is associated with vasoconstriction and elevated blood pressure. We identified a novel inhibitor of Rho kinase (SAR407899) and characterized its effects in biochemical, cellular, tissue-based, and in vivo assays. SAR407899 is an ATP-competitive Rho-kinase inhibitor, equipotent against human and rat-derived Rho-kinase 2 with inhibition constant values of 36 nM and 41 nM, respectively. It is highly selective in panel of 117 receptor and enzyme targets. SAR407899 is approximately 8-fold more active than fasudil. In vitro, SAR407899 demonstrated concentration-dependent inhibition of Rho-kinase-mediated phosphorylation of myosin phosphatase, thrombin-induced stress fiber formation, platelet-derived growth factor-induced proliferation, and monocyte chemotactic protein-1-stimulated chemotaxis. SAR407899 potently (mean IC(50) values: 122 to 280 nM) and species-independently relaxed precontracted isolated arteries of different species and different vascular beds. In vivo, over the dose range 3 to 30 mg/kg PO, SAR407899 lowered blood pressure in a variety of rodent models of arterial hypertension. The antihypertensive effect of SAR407899 was superior to that of fasudil and Y-27632. In conclusion, SAR407899 is a novel and potent selective Rho-kinase inhibitor with promising antihypertensive activity.


Asunto(s)
Fosfatasa de Miosina de Cadena Ligera/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Quinasas Asociadas a rho/antagonistas & inhibidores , Animales , Arterias/efectos de los fármacos , Arterias/fisiología , Presión Sanguínea/efectos de los fármacos , Catálisis/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Quimiotaxis/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Humanos , Immunoblotting , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Fosforilación/efectos de los fármacos , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Ratas Sprague-Dawley , Fibras de Estrés/metabolismo , Vasoconstricción/efectos de los fármacos , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo
17.
Proc Natl Acad Sci U S A ; 106(15): 6152-7, 2009 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-19369214

RESUMEN

VEGF-B, a homolog of VEGF discovered a long time ago, has not been considered an important target in antiangiogenic therapy. Instead, it has received little attention from the field. In this study, using different animal models and multiple types of vascular cells, we revealed that although VEGF-B is dispensable for blood vessel growth, it is critical for their survival. Importantly, the survival effect of VEGF-B is not only on vascular endothelial cells, but also on pericytes, smooth muscle cells, and vascular stem/progenitor cells. In vivo, VEGF-B targeting inhibited both choroidal and retinal neovascularization. Mechanistically, we found that the vascular survival effect of VEGF-B is achieved by regulating the expression of many vascular prosurvival genes via both NP-1 and VEGFR-1. Our work thus indicates that the function of VEGF-B in the vascular system is to act as a "survival," rather than an "angiogenic" factor and that VEGF-B inhibition may offer new therapeutic opportunities to treat neovascular diseases.


Asunto(s)
Neovascularización Patológica , Factor B de Crecimiento Endotelial Vascular/metabolismo , Animales , Supervivencia Celular , Células Cultivadas , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Genoma , Miembro Posterior/irrigación sanguínea , Isquemia/genética , Isquemia/metabolismo , Ratones , Ratones Noqueados , Ratas , Retina/metabolismo , Células Madre/citología , Células Madre/metabolismo , Regulación hacia Arriba , Factor B de Crecimiento Endotelial Vascular/deficiencia , Factor B de Crecimiento Endotelial Vascular/genética
18.
Blood ; 111(8): 4096-105, 2008 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-18156494

RESUMEN

The role of Gas6 in endothelial cell (EC) function remains incompletely characterized. Here we report that Gas6 amplifies EC activation in response to inflammatory stimuli in vitro. In vivo, Gas6 promotes and accelerates the sequestration of circulating platelets and leukocytes on activated endothelium as well as the formation and endothelial sequestration of circulating platelet-leukocyte conjugates. In addition, Gas6 promotes leukocyte extravasation, inflammation, and thrombosis in mouse models of inflammation (endotoxinemia, vasculitis, heart transplantation). Thus, Gas6 amplifies EC activation, thereby playing a key role in enhancing the interactions between ECs, platelets, and leukocytes during inflammation.


Asunto(s)
Plaquetas/patología , Comunicación Celular , Células Endoteliales/patología , Inflamación/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Leucocitos/patología , Animales , Plaquetas/metabolismo , Línea Celular , Células Endoteliales/enzimología , Endotelio/metabolismo , Trasplante de Corazón , Humanos , Leucocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patología , Proteínas Oncogénicas/metabolismo , Selectina-P/metabolismo , Proteínas Proto-Oncogénicas , Proteínas Tirosina Quinasas Receptoras/metabolismo , Vasculitis/metabolismo , Vasculitis/patología , Tirosina Quinasa del Receptor Axl
19.
Exp Cell Res ; 313(14): 2980-92, 2007 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17619016

RESUMEN

The biological complexity of NGF action is achieved by binding two distinct neurotrophin receptors, TrkA and p75(NTR). While several reports have provided lines of evidence on the interaction between TrkA and p75(NTR) at the plasma membrane, much fewer data are available on the consequence of such an interaction in terms of intracellular signaling. In this study, we have focused on how p75(NTR) may affect TrkA downstream signaling with respect to neuronal differentiation. Here, we have shown that cooperation between p75(NTR) and TrkA results in an increased NGF-mediated TrkA autophosphorylation, leads to a sustained activation of ERK1/2 and accelerates neurite outgrowth. Interestingly, neurite outgrowth is concomitant with a selective enhancement of the AP-1 activity and the transcriptional activation of genes such as GAP-43 and p21(CIP/WAF), known to be involved in the differentiation process. Collectively, our results unveil a functional link between the specific expression profile of neurotrophin receptors in neuronal cells and the NGF-mediated regulation of the differentiation process possibly through a persistent ERKs activation and the selective control of the AP-1 activity.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Proteína GAP-43 , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Neuronas/fisiología , Receptor de Factor de Crecimiento Nervioso/metabolismo , Receptor trkA/metabolismo , Factor de Transcripción AP-1/metabolismo , Animales , Diferenciación Celular/fisiología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Activación Enzimática , Proteína GAP-43/genética , Proteína GAP-43/metabolismo , Humanos , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/genética , Factor de Crecimiento Nervioso/metabolismo , Neuronas/citología , Fosforilación , Receptor de Factor de Crecimiento Nervioso/genética , Receptor trkA/genética , Transducción de Señal/fisiología , Factor de Transcripción AP-1/genética , Transcripción Genética , Células Tumorales Cultivadas
20.
Vasc Med ; 12(2): 113-22, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17615799

RESUMEN

Recent advances in our understanding of cardiovascular disease have revealed that atherothrombotic events, such as myocardial infarction and ischemic stroke, are the end result of a complex inflammatory response to multifaceted vascular pathology. As well as initiating thrombus formation at the site of a ruptured atherosclerotic plaque, platelets play a key role in vascular inflammation, through release of their own pro-inflammatory mediators and interactions with other relevant cell types (endothelial cells, leukocytes, and smooth muscle cells). An increasing body of literature shows that inflammatory biomarkers can be used to predict atherothrombotic risk and that antiplatelet therapy may reduce the levels of these markers. Acetylsalicylic acid (ASA) has been attributed with reducing levels of the transcription factor nuclear factor kappaB (NF-kappaB), C-reactive protein, and soluble CD40 ligand, although the evidence relating to the latter two markers is conflicting. There is also substantial evidence that therapy with clopidogrel, a specific antagonist of the platelet P2Y12 ADP-receptor, also leads to reductions in serum levels of CD40 ligand, C-reactive protein, P-selectin, and platelet-leukocyte aggregate formation. Beneficial effects of clopidogrel on inflammatory markers have been demonstrated across the spectrum of atherothrombotic disease (acute coronary syndrome patients, patients undergoing percutaneous coronary intervention (PCI), acute ischemic stroke patients, and those with peripheral arterial disease). Oral glycoprotein (GP) IIb/IIIa receptor antagonists, at doses that achieve moderate levels of receptor blockade, may paradoxically be associated with platelet-mediated pro-inflammatory effects. A similar phenomenon has been observed with intravenous GP IIb/IIIa antagonists in vitro, but most often at low doses, and data from clinical studies suggest that these agents may actually attenuate release of inflammatory mediators when administered at doses producing more complete receptor blockade.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Inhibidores de Agregación Plaquetaria/uso terapéutico , Trombosis/tratamiento farmacológico , Angina Inestable/prevención & control , Aspirina/uso terapéutico , Biomarcadores/análisis , Plaquetas/efectos de los fármacos , Plaquetas/fisiología , Clopidogrel , Humanos , Inflamación/tratamiento farmacológico , Infarto del Miocardio/prevención & control , Enfermedades Vasculares Periféricas/prevención & control , Accidente Cerebrovascular/prevención & control , Ticlopidina/análogos & derivados , Ticlopidina/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...