Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Cereb Blood Flow Metab ; 40(1_suppl): S34-S48, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33208001

RESUMEN

Microglia are key regulators of inflammatory response after stroke and brain injury. To better understand activation of microglia as well as their phenotypic diversity after ischemic stroke, we profiled the transcriptome of microglia after 75 min transient focal cerebral ischemia in 3-month- and 12-month-old male spontaneously hypertensive rats. Microglia were isolated from the brains by FACS sorting on days 3 and 14 after cerebral ischemia. GeneChip Rat 1.0ST microarray was used to profile the whole transcriptome of sorted microglia. We identified an evolving and complex pattern of activation from 3 to 14 days after stroke onset. M2-like patterns were extensively and persistently upregulated over time. M1-like patterns were only mildly upregulated, mostly at day 14. Younger 3-month-old brains showed a larger microglial response in both pro- and anti-inflammatory pathways, compared to older 12-month-old brains. Importantly, our data revealed that after stroke, most microglia are activated towards a wide spectrum of novel polarization states beyond the standard M1/M2 dichotomy, especially in pathways related to TLR2 and dietary fatty acid signaling. Finally, classes of transcription factors that might potentially regulate microglial activation were identified. These findings should provide a comprehensive database for dissecting microglial mechanisms and pursuing neuroinflammation targets for acute ischemic stroke.


Asunto(s)
Isquemia Encefálica/fisiopatología , Microglía/metabolismo , Accidente Cerebrovascular/fisiopatología , Transcriptoma/genética , Animales , Modelos Animales de Enfermedad , Masculino , Fenotipo , Ratas
2.
Stem Cell Res ; 20: 105-114, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28334554

RESUMEN

A fast track "Hot Start" process was implemented to launch the European Bank for Induced Pluripotent Stem Cells (EBiSC) to provide early release of a range of established control and disease linked human induced pluripotent stem cell (hiPSC) lines. Established practice amongst consortium members was surveyed to arrive at harmonised and publically accessible Standard Operations Procedures (SOPs) for tissue procurement, bio-sample tracking, iPSC expansion, cryopreservation, qualification and distribution to the research community. These were implemented to create a quality managed foundational collection of lines and associated data made available for distribution. Here we report on the successful outcome of this experience and work flow for banking and facilitating access to an otherwise disparate European resource, with lessons to benefit the international research community. ETOC: The report focuses on the EBiSC experience of rapidly establishing an operational capacity to procure, bank and distribute a foundational collection of established hiPSC lines. It validates the feasibility and defines the challenges of harnessing and integrating the capability and productivity of centres across Europe using commonly available resources currently in the field.


Asunto(s)
Bancos de Muestras Biológicas , Células Madre Pluripotentes Inducidas/citología , Línea Celular , Criopreservación , Europa (Continente) , Humanos
3.
Arch Toxicol ; 91(3): 1385-1400, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27344343

RESUMEN

Assessing the potential of a new drug to cause drug-induced liver injury (DILI) is a challenge for the pharmaceutical industry. We therefore determined whether cell models currently used in safety assessment (HepG2, HepaRG, Upcyte and primary human hepatocytes in conjunction with basic but commonly used endpoints) are actually able to distinguish between novel chemical entities (NCEs) with respect to their potential to cause DILI. A panel of thirteen compounds (nine DILI implicated and four non-DILI implicated in man) were selected for our study, which was conducted, for the first time, across multiple laboratories. None of the cell models could distinguish faithfully between DILI and non-DILI compounds. Only when nominal in vitro concentrations were adjusted for in vivo exposure levels were primary human hepatocytes (PHH) found to be the most accurate cell model, closely followed by HepG2. From a practical perspective, this study revealed significant inter-laboratory variation in the response of PHH, HepG2 and Upcyte cells, but not HepaRG cells. This variation was also observed to be compound dependent. Interestingly, differences between donors (hepatocytes), clones (HepG2) and the effect of cryopreservation (HepaRG and hepatocytes) were less important than differences between the cell models per se. In summary, these results demonstrate that basic cell health endpoints will not predict hepatotoxic risk in simple hepatic cells in the absence of pharmacokinetic data and that a multicenter assessment of more sophisticated signals of molecular initiating events is required to determine whether these cells can be incorporated in early safety assessment.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Pruebas de Toxicidad Aguda/métodos , Células Cultivadas , Criopreservación , Células Hep G2/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Humanos , Reproducibilidad de los Resultados , Pruebas de Toxicidad Aguda/normas
4.
NPJ Regen Med ; 2: 28, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29302362

RESUMEN

Regenerative medicine therapies hold enormous potential for a variety of currently incurable conditions with high unmet clinical need. Most progress in this field to date has been achieved with cell-based regenerative medicine therapies, with over a thousand clinical trials performed up to 2015. However, lack of adequate safety and efficacy data is currently limiting wider uptake of these therapies. To facilitate clinical translation, non-invasive in vivo imaging technologies that enable careful evaluation and characterisation of the administered cells and their effects on host tissues are critically required to evaluate their safety and efficacy in relevant preclinical models. This article reviews the most common imaging technologies available and how they can be applied to regenerative medicine research. We cover details of how each technology works, which cell labels are most appropriate for different applications, and the value of multi-modal imaging approaches to gain a comprehensive understanding of the responses to cell therapy in vivo.

5.
Stroke ; 47(4): 1094-100, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26965847

RESUMEN

BACKGROUND AND PURPOSE: Pentraxin 3 (PTX3) is released on inflammatory responses in many organs. However, roles of PTX3 in brain are still mostly unknown. Here we asked whether and how PTX3 contributes to blood-brain barrier dysfunction during the acute phase of ischemic stroke. METHODS: In vivo, spontaneously hypertensive rats were subjected to focal cerebral ischemia by transient middle cerebral artery occlusion. At day 3, brains were analyzed to evaluate the cellular origin of PTX3 expression. Correlations with blood-brain barrier breakdown were assessed by IgG staining. In vitro, rat primary astrocytes and rat brain endothelial RBE.4 cells were cultured to study the role of astrocyte-derived PTX3 on vascular endothelial growth factor-mediated endothelial permeability. RESULTS: During the acute phase of stroke, reactive astrocytes in the peri-infarct area expressed PTX3. There was negative correlation between gradients of IgG leakage and PTX3-positive astrocytes. Cell culture experiments showed that astrocyte-conditioned media increased levels of tight junction proteins and reduced endothelial permeability under normal conditions. Removing PTX3 from astrocyte-conditioned media by immunoprecipitation increased endothelial permeability. PTX3 strongly bound vascular endothelial growth factor in vitro and was able to decrease vascular endothelial growth factor-induced endothelial permeability. CONCLUSIONS: Astrocytes in peri-infarct areas upregulate PTX3, which may support blood-brain barrier integrity by regulating vascular endothelial growth factor-related mechanisms. This response in astrocytes may comprise a compensatory mechanism for maintaining blood-brain barrier function after ischemic stroke.


Asunto(s)
Astrocitos/metabolismo , Barrera Hematoencefálica/metabolismo , Proteína C-Reactiva/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Componente Amiloide P Sérico/metabolismo , Accidente Cerebrovascular/metabolismo , Animales , Barrera Hematoencefálica/patología , Encéfalo/metabolismo , Encéfalo/patología , Muerte Celular , Medios de Cultivo Condicionados , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Infarto de la Arteria Cerebral Media/patología , Masculino , Ratas , Ratas Endogámicas SHR , Accidente Cerebrovascular/patología , Factor A de Crecimiento Endotelial Vascular/metabolismo
6.
Cell Transplant ; 25(4): 705-14, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26811151

RESUMEN

Aging and vascular comorbidities such as hypertension comprise critical cofactors that influence how the brain responds to stroke. Ischemic stress induces neurogenesis and oligodendrogenesis in younger brains. However, it remains unclear whether these compensatory mechanisms can be maintained even under pathologically hypertensive and aged states. To clarify the age-related remodeling capacity after stroke under hypertensive conditions, we assessed infarct volume, behavioral outcomes, and surrogate markers of neurogenesis and oligodendrogenesis in acute and subacute phases after transient focal cerebral ischemia in 3- and 12-month-old spontaneously hypertensive rats (SHRs). Hematoxylin and eosin staining showed that 3- and 12-month-old SHRs exhibited similar infarction volumes at both 3 and 14 days after focal cerebral ischemia. However, recovery of behavioral deficits (neurological score assessment and adhesive removal test) was significantly less in 12-month-old SHRs compared to 3-month-old SHRs. Concomitantly, numbers of nestin(+) neural stem/progenitor cells (NSPCs) near the infarct border area or subventricular zone in 12-month-old SHRs were lower than 3-month-old SHRs at day 3. Similarly, numbers of PDGFR-α(+) oligodendrocyte precursor cells (OPCs) in the corpus callosum were lower in 12-month-old SHRs at day 3. Lower levels of NSPC and OPC numbers were accompanied by lower expression levels of phosphorylated CREB. By day 14 postischemia, NSPC and OPC numbers in 12-month-old SHRs recovered to similar levels as in 3-month-old SHRs, but the numbers of proliferating NSPCs (Ki-67(+)nestin(+) cells) and proliferating OPCs (Ki-67(+)PDGFR-α(+) cells) remained lower in the older brains even at day 14. Taken together, these findings suggest that aging may also decrease poststroke compensatory responses for neurogenesis and oligodendrogenesis even under hypertensive conditions.


Asunto(s)
Envejecimiento/metabolismo , Infarto Encefálico/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis , Oligodendroglía/metabolismo , Envejecimiento/patología , Animales , Infarto Encefálico/patología , Masculino , Células-Madre Neurales/patología , Oligodendroglía/patología , Ratas , Ratas Endogámicas SHR
7.
PLoS One ; 10(9): e0138724, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26407349

RESUMEN

Adult mammalian brain can be plastic after injury and disease. Therefore, boosting endogenous repair mechanisms would be a useful therapeutic approach for neurological disorders. Isoxazole-9 (Isx-9) has been reported to enhance neurogenesis from neural stem/progenitor cells (NSPCs). However, the effects of Isx-9 on other types of progenitor/precursor cells remain mostly unknown. In this study, we investigated the effects of Isx-9 on the three major populations of progenitor/precursor cells in brain: NSPCs, oligodendrocyte precursor cells (OPCs), and endothelial progenitor cells (EPCs). Cultured primary NSPCs, OPCs, or EPCs were treated with various concentrations of Isx-9 (6.25, 12.5, 25, 50 µM), and their cell numbers were counted in a blinded manner. Isx-9 slightly increased the number of NSPCs and effectively induced neuronal differentiation of NSPCs. However, Isx-9 significantly decreased OPC number in a concentration-dependent manner, suggesting cytotoxicity. Isx-9 did not affect EPC cell number. But in a matrigel assay of angiogenesis, Isx-9 significantly inhibited tube formation in outgrowth endothelial cells derived from EPCs. This potential anti-tube-formation effect of Isx-9 was confirmed in a brain endothelial cell line. Taken together, our data suggest that mechanisms and targets for promoting stem/progenitor cells in the central nervous system may significantly differ between cell types.


Asunto(s)
Diferenciación Celular , Células Progenitoras Endoteliales/efectos de los fármacos , Isoxazoles/farmacología , Células-Madre Neurales/efectos de los fármacos , Animales , Biomarcadores , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Células Progenitoras Endoteliales/citología , Células Progenitoras Endoteliales/metabolismo , Ratones , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis , Neuronas/citología , Neuronas/metabolismo , Oligodendroglía/citología , Oligodendroglía/metabolismo , Ratas
8.
Bioorg Med Chem Lett ; 25(17): 3748-53, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26142946

RESUMEN

A novel neurogenic compound (1), discovered from a mouse neural progenitor cell (NPC) screen, showed profound neurogenic effect on human NPCs. Synthesis and SAR of this novel 2,3,11,11a-tetrahydro-1H-pyrazino[1,2-b]isoquinoline-1,4(6H)-dione series are described. Compound 20 is brain penetrable in rodents, and promotes neurogenesis in wild type mice, therefore it is a good tool molecule to study neurogenesis induction as a potential treatment for conditions associated with neurogenesis impairment diseases.


Asunto(s)
Isoquinolinas/química , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Pirazinas/farmacocinética , Relación Estructura-Actividad , Administración Oral , Animales , Encéfalo/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Química Sintética , Relación Dosis-Respuesta a Droga , Descubrimiento de Drogas , Evaluación Preclínica de Medicamentos/métodos , Semivida , Humanos , Isoquinolinas/administración & dosificación , Isoquinolinas/farmacocinética , Isoquinolinas/farmacología , Ratones Endogámicos C57BL , Células-Madre Neurales/citología , Pirazinas/administración & dosificación , Pirazinas/química
9.
Toxicol Pathol ; 42(5): 844-54, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23960163

RESUMEN

Whether biliary proliferative lesions in nonclinical species are predictive of potential hepatotoxicity in humans depends, at least in part, on the nature and severity of such changes in the nonclinical species. We reviewed published literature (clinical and nonclinical) and experimental data from rat toxicology studies conducted by GlaxoSmithKline and the National Institute of Environmental Health Sciences' National Toxicology Program in an effort to better characterize the relative risk of hepatobiliary effects in humans. Available evidence supports the interpretation that minimal "typical" appearing bile duct hyperplasia limited to the portal triads may be considered non-adverse in the rat and is of little to no concern to humans. The toxicological relevance of mild to moderate "typical" hyperplasia is less certain, and may be considered adverse in the rat and potentially pose a risk for humans, particularly if accompanied by evidence of hepatobiliary injury or functional compromise. In addition, any proliferative lesion that includes atypical or dysplastic epithelial changes, oval cell proliferation, and/or significant extension beyond the portal tracts is considered more ominous and may be considered adverse in the rat.


Asunto(s)
Neoplasias de los Conductos Biliares/patología , Conductos Biliares/citología , Conductos Biliares/patología , Proliferación Celular , Animales , Conductos Biliares/metabolismo , Biomarcadores/metabolismo , Femenino , Hiperplasia/patología , Masculino , Ratas , Ratas Sprague-Dawley
10.
Toxicol Sci ; 126(2): 306-16, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22262563

RESUMEN

Certain human diseases affecting the biliary tree can be modeled in rats by ingestion of the hepatobiliary toxin alpha-naphthylisothiocyanate (ANIT). Phosphorus magnetic resonance spectroscopy (MRS) allows the noninvasive monitoring of cell dynamics through detection of phosphodiesters (PDE) and phosphomonoesters (PME). Hepatic (31)P MRS techniques were therefore used to study the toxic effects of low-dose chronic ANIT ingestion, with a view toward providing biomarkers sensitive to hepatobiliary dysfunction and cholestatic liver injury. Rats were fed an ANIT supplemented diet at three doses (ANIT_0.05%, ANIT_0.04%, and ANIT_0.025%) for 2 weeks. Data from in vivo MRS were compared with results from pair-fed controls (PFCs). Blood and tissue samples were collected at 2 weeks for clinical chemistry, histology, and (1)H magic angle spinning MRS. Increases in PDE, relative to total phosphorus (tPh), were detected in both the ANIT_0.05% and ANIT_0.04% groups (0.07 ± 0.01 and 0.08 ± 0.01, respectively) relative to PFC groups (0.03 ± 0.01 and 0.05 ± 0.01, respectively). An increase in PME/tPh was observed in the ANIT_0.05% group only (0.17 ± 0.02) relative to PFC_0.05% (0.12 ± 0.01). Ex vivo (1)H MRS findings supported this, wherein measured phosphocholines (PCs) were increased in ANIT_0.05% and ANIT_0.04% groups. Increases in relative total choline (tCho) distinguished the ANIT_0.05% group from the ANIT_0.04% group. Markers of hepatotoxicity such as raised total bilirubin and alkaline phosphatase were found at all ANIT doses. Histological findings included a dose-related increase in both severity of biliary hyperplasia and focal hepatocellular necrosis. Here, we found that ANIT-induced moderate hepatobiliary dysfunction was associated with a relative increase in phosphodiesters in vivo and PCs ex vivo. Raised PME/tPh in vivo and tCho ex vivo were also present at high doses corresponding to a higher incidence of marked biliary hyperplasia and moderate hepatocellular necrosis.


Asunto(s)
1-Naftilisotiocianato/toxicidad , Hígado/efectos de los fármacos , Espectroscopía de Resonancia Magnética/métodos , 1-Naftilisotiocianato/administración & dosificación , Análisis de Varianza , Animales , Peso Corporal , Conducta Alimentaria , Técnicas In Vitro , Hígado/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley
11.
PLoS One ; 7(12): e51404, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23284691

RESUMEN

Transforming growth factor (TGF) ß has diverse and sometimes paradoxical effects on cell proliferation and differentiation, presumably reflecting a fundamental but incompletely-understood role in regulating tissue homeostasis. It is generally considered that downstream activity is modulated at the ligand:receptor axis, but microarray analysis of proliferative versus differentiating normal human bladder epithelial cell cultures identified unexpected transcriptional changes in key components of the canonical TGFß R/activin signalling pathway associated with cytodifferentiation. Changes included upregulation of the transcriptional modulator SMAD3 and downregulation of inhibitory modulators SMURF2 and SMAD7. Functional analysis of the signalling pathway revealed that non-differentiated normal human urothelial cells responded in paracrine mode to TGFß by growth inhibition, and that exogenous TGFß inhibited rather than promoted differentiation. By contrast, in differentiated cell cultures, SMAD3 was activated upon scratch-wounding and was involved in promoting tissue repair. Exogenous TGFß enhanced the repair and resulted in hyperplastic scarring, indicating a feedback loop implicit in an autocrine pathway. Thus, the machinery for autocrine activation of the SMAD3-mediated TGFßR pathway is established during urothelial differentiation, but signalling occurs only in response to a trigger, such as wounding. Our study demonstrates that the circuitry of the TGFßR pathway is defined transcriptionally within a tissue-specific differentiation programme. The findings provide evidence for re-evaluating the role of TGFßR signalling in epithelial homeostasis as an autocrine-regulated pathway that suppresses differentiation and promotes tissue repair. This provides a new paradigm to help unravel the apparently diverse and paradoxical effect of TGFß signalling on cell proliferation and differentiation.


Asunto(s)
Comunicación Autocrina , Diferenciación Celular , Comunicación Paracrina , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal , Urotelio/citología , Animales , Comunicación Autocrina/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células HeLa , Homeostasis/efectos de los fármacos , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Comunicación Paracrina/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Transcriptoma/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología , Cicatrización de Heridas/efectos de los fármacos
12.
Drug Metab Dispos ; 38(1): 143-9, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19797606

RESUMEN

The intestinal-derived secondary bile acid (BA) lithocholic acid (LCA) is hepatotoxic and is implicated in the pathogenesis of cholestatic diseases. LCA is an endogenous ligand of the xenobiotic nuclear receptor pregnane X receptor (PXR), but there is currently no consensus on the respective roles of hepatic and intestinal PXR in mediating protection against LCA in vivo. Under the conditions reported here, we show that mice lacking Pxr are resistant to LCA-mediated hepatotoxicity. This unexpected phenotype is found in association with enhanced urinary BA excretion and elevated basal expression of drug metabolism enzymes and the hepatic sulfate donor synthesis enzyme Papss2 in Pxr(-/-) mice. By subsequently comparing molecular responses to dietary and intraperitoneal administration of LCA, we made two other significant observations: 1) LCA feeding induces intestinal, but not hepatic, drug-metabolizing enzymes in a largely Pxr-independent manner; and 2) in contrast to LCA feeding, bypassing first-pass gut transit by intraperitoneal administration of LCA did induce hepatic detoxification machinery and in a Pxr-dependent manner. These data reconcile important discrepancies in the reported molecular responses to this BA and suggest that Pxr plays only a limited role in mediating responses to gut-derived LCA. Furthermore, the route of administration must be considered in the future planning and interpretation of experiments designed to assess hepatic responses to BAs, orally administered pharmaceuticals, and dietary toxins.


Asunto(s)
Inactivación Metabólica/fisiología , Mucosa Intestinal/metabolismo , Ácido Litocólico/farmacocinética , Hígado/metabolismo , Receptores de Esteroides/metabolismo , Administración Oral , Alanina Transaminasa/sangre , Animales , Hidrocarburo de Aril Hidroxilasas/genética , Ácidos y Sales Biliares/sangre , Ácidos y Sales Biliares/metabolismo , Ácidos y Sales Biliares/orina , Peso Corporal/efectos de los fármacos , Enfermedad Hepática Inducida por Sustancias y Drogas/sangre , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Citocromo P-450 CYP3A/genética , Sistema Enzimático del Citocromo P-450/genética , Sistema Enzimático del Citocromo P-450/metabolismo , Familia 2 del Citocromo P450 , Inducción Enzimática/efectos de los fármacos , Femenino , Expresión Génica/efectos de los fármacos , Inyecciones Intraperitoneales , Intestinos/efectos de los fármacos , Ácido Litocólico/administración & dosificación , Ácido Litocólico/farmacología , Hígado/efectos de los fármacos , Hígado/patología , Masculino , Proteínas de la Membrana/genética , Proteínas de Transporte de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Complejos Multienzimáticos/genética , Receptor X de Pregnano , Esteroide Hidroxilasas/genética , Sulfato Adenililtransferasa/genética , Sulfotransferasas/genética
13.
Mol Cancer Res ; 6(1): 53-63, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18234962

RESUMEN

Bladder cancer evolves via the accumulation of numerous genetic alterations, with loss of p53 and p16 function representing key events in the development of malignant disease. In addition, components of the epidermal growth factor receptor (EGFR) signaling pathway are frequently overexpressed, providing potential chemotherapeutic targets. We have previously described the generation of "paramalignant" human urothelial cells with disabled p53 or p16 functions. In this study, we investigated the relative responses of normal, paramalignant, and malignant human urothelial cells to EGFR tyrosine kinase inhibitors (PD153035 and GW572016), a mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) kinase (MEK) inhibitor (U0126), and a phosphatidylinositol 3-kinase inhibitor (LY294002). The proliferation of normal human urothelial cells was dependent on signaling via the EGFR and MEK pathways and was abolished reversibly by inhibitors of EGFR or downstream MEK signaling pathways. Inhibitors of phosphatidylinositol 3-kinase resulted in only transient cytostasis, which was most likely mediated via cross-talk with the MEK pathway. These responses were maintained in cells with disabled p16 function, whereas cells with loss of p53 function displayed reduced sensitivity to PD153035 and malignant cell lines were the most refractory to PD153035 and U0126. These results indicate that urothelial cells acquire insensitivity to inhibitors of EGFR signaling pathways as a result of malignant transformation. This has important implications for the use of EGFR inhibitors for bladder cancer therapy, as combination treatments with conventional chemotherapy or radiotherapy may protect normal cells and enable better selective targeting of malignant cells.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Receptores ErbB/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Neoplasias Urológicas/patología , Urotelio/efectos de los fármacos , Urotelio/patología , Apoptosis/efectos de los fármacos , Biomarcadores/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Volumetría , Neoplasias Urológicas/enzimología , Urotelio/enzimología
14.
Toxicology ; 213(1-2): 117-28, 2005 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-16084005

RESUMEN

A pilot toxicology database system has been created which is accessible on-line via the world-wide web or in-house via an intranet. It is intended to be suitable as a source of toxicological information and to support structure-activity relationship studies, and it can be searched on chemical structural and substructural as well as toxicological and physico-chemical data. Successful completion of the pilot has led to an ongoing project to develop and expand the system.


Asunto(s)
Bases de Datos como Asunto , Toxicología , Estudios de Factibilidad , Cooperación Internacional , Proyectos Piloto , Programas Informáticos , Relación Estructura-Actividad
15.
Arch Toxicol ; 79(4): 208-23, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15838709

RESUMEN

The present study was designed to provide further information about the relevance of raised urinary levels of N-methylnicotinamide (NMN), and/or its metabolites N-methyl-4-pyridone-3-carboxamide (4PY) and N-methyl-2-pyridone-3-carboxamide (2PY), to peroxisome proliferation by dosing rats with known peroxisome proliferator-activated receptor alpha (PPARalpha) ligands [fenofibrate, diethylhexylphthalate (DEHP) and long-chain fatty acids (LCFA)] and other compounds believed to modulate lipid metabolism via PPARalpha-independent mechanisms (simvastatin, hydrazine and chlorpromazine). Urinary NMN was correlated with standard markers of peroxisome proliferation and serum lipid parameters with the aim of establishing whether urinary NMN could be used as a biomarker for peroxisome proliferation in the rat. Data from this study were also used to validate a previously constructed multivariate statistical model of peroxisome proliferation (PP) in the rat. The predictive model, based on 1H nuclear magnetic resonance (NMR) spectroscopy of urine, uses spectral patterns of NMN, 4PY and other endogenous metabolites to predict hepatocellular peroxisome count. Each treatment induced pharmacological (serum lipid) effects characteristic of their class, but only fenofibrate, DEHP and simvastatin increased peroxisome number and raised urinary NMN, 2PY and 4PY, with simvastatin having only a transient effect on the latter. These compounds also reduced mRNA expression for aminocarboxymuconate-semialdehyde decarboxylase (ACMSDase, EC 4.1.1.45), the enzyme believed to be involved in modulating the flux of tryptophan through this pathway, with decreasing order of potency, fenofibrate (-10.39-fold) >DEHP (-3.09-fold) >simvastatin (-1.84-fold). Of the other treatments, only LCFA influenced mRNA expression of ACMSDase (-3.62-fold reduction) and quinolinate phosphoribosyltransferase (QAPRTase, EC 2.4.2.19) (-2.42-fold) without any change in urinary NMN excretion. Although there were no correlations between urinary NMN concentration and serum lipid parameters, NMN did correlate with peroxisome count (r2=0.63) and acyl-CoA oxidase activity (r2=0.61). These correlations were biased by the large response to fenofibrate compared to the other treatments; nevertheless the data do indicate a relationship between the tryptophan-NAD+ pathway and PPARalpha-dependent pathways, making this metabolite a potentially useful biomarker to detect PP. In order to strengthen the observed link between the metabolites associated with the tryptophan-NAD+ pathway and more accurately predict PP, other urinary metabolites were included in a predictive statistical model. This statistical model was found to predict the observed PP in 26/27 instances using a pre-determined threshold of 2-fold mean control peroxisome count. The model also predicted a time-dependent increase in peroxisome count for the fenofibrate group, which is important when considering the use of such modelling to predict the onset and progression of PP prior to its observation in samples taken at autopsy.


Asunto(s)
NAD/metabolismo , Proliferadores de Peroxisomas/metabolismo , Triptófano/metabolismo , Animales , Biomarcadores , Carboxiliasas/metabolismo , Cromatografía Líquida de Alta Presión , Expresión Génica , Hepatocitos/ultraestructura , Hipolipemiantes/farmacología , Inmunohistoquímica , Lípidos/sangre , Lipoproteínas/sangre , Pruebas de Función Hepática , Espectroscopía de Resonancia Magnética , Masculino , Espectrometría de Masas , Microscopía Electrónica de Transmisión , Modelos Estadísticos , Tamaño de los Órganos/efectos de los fármacos , Pentosiltransferasa/metabolismo , Peroxisomas/ultraestructura , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley
16.
J Pharmacol Exp Ther ; 313(2): 780-9, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15665138

RESUMEN

The mild analgesic drug acetaminophen (AAP) induces severe hepatic injury when taken at excessive doses. Recent evidence shows that the initial form of damage is through apoptosis, but this fails to go to completion and degenerates into necrosis. The aim of this study was to elucidate the mechanism through which AAP induces apoptosis using human HuH7 hepatoma cells as an in vitro model system to investigate the initial phase of AAP-induced hepatic injury. AAP-induced apoptosis in HuH7 cells as evidenced by chromatin condensation was preceded by the translocation of Bax to mitochondria and the cytoplasmic release of the proapoptotic factors cytochrome c and Smac/DIABLO. A concomitant loss of mitochondrial membrane potential occurred. Activation of the mitochondrial pathway of apoptosis led to the activation of execution caspases-3 and -7. AAP-induced apoptosis and cell death was blocked by inhibitors of caspases but not by inhibitors of calpains, cathepsins, and serine proteases. Apoptosis was unaffected by inhibitors of the mitochondrial permeability transition pore and by inhibitors of Jun NH(2)-terminal kinases, p38 mitogen-activated protein kinase, or mitogen-activated protein kinase kinase 1/2. However, pharmacological inhibition of glycogen synthase kinase-3 (GSK-3) delayed and decreased the extent of AAP-induced apoptosis. In comparison, endoplasmic reticulum stress-induced but not prooxidant-induced apoptosis of HuH7 cells was sensitive to GSK-3 inhibition. It is concluded that AAP-induced apoptosis involves the mitochondrial pathway of apoptosis that is mediated by GSK-3 and most likely initiated through an endoplasmic reticulum stress response.


Asunto(s)
Acetaminofén/toxicidad , Apoptosis/fisiología , Carcinoma Hepatocelular/enzimología , Carcinoma Hepatocelular/patología , Glucógeno Sintasa Quinasa 3/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Humanos , Inhibidores de Proteínas Quinasas/farmacología
17.
Metabolism ; 53(10): 1322-30, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15375789

RESUMEN

Glycogen synthase kinase-3 (GSK-3) protein levels and activity are elevated in skeletal muscle in type 2 diabetes, and inversely correlated with both glycogen synthase activity and insulin-stimulated glucose disposal. To explore this relationship, we have produced transgenic mice that overexpress human GSK-3beta in skeletal muscle. GSK-3beta transgenic mice were heavier, by up to 20% (P < .001), than their age-matched controls due to an increase in fat mass. The male GSK-3beta transgenic mice had significantly raised plasma insulin levels and by 24 weeks of age became glucose-intolerant as determined by a 50% increase in the area under their oral glucose tolerance curve (P < .001). They were also hyperlipidemic with significantly raised serum cholesterol (+90%), nonesterified fatty acids (NEFAs) (+55%), and triglycerides (+170%). At 29 weeks of age, GSK-3beta protein levels were 5-fold higher, and glycogen synthase activation (-27%), glycogen levels (-58%) and insulin receptor substrate-1 (IRS-1) protein levels (-67%) were significantly reduced in skeletal muscle. Hepatic glycogen levels were significantly increased 4-fold. Female GSK-3beta transgenic mice did not develop glucose intolerance despite 7-fold overexpression of GSK-3beta protein and a 20% reduction in glycogen synthase activation in skeletal muscle. However, plasma NEFAs and muscle IRS-1 protein levels were unchanged in females. We conclude that overexpression of human GSK-3beta in skeletal muscle of male mice resulted in impaired glucose tolerance despite raised insulin levels, consistent with the possibility that elevated levels of GSK-3 in type 2 diabetes are partly responsible for insulin resistance.


Asunto(s)
Intolerancia a la Glucosa/genética , Glucógeno Sintasa Quinasa 3/biosíntesis , Glucógeno Sintasa Quinasa 3/genética , Músculo Esquelético/fisiología , Regiones Promotoras Genéticas/fisiología , Animales , Western Blotting , Composición Corporal/fisiología , Peso Corporal/fisiología , Cartilla de ADN , ADN Complementario/biosíntesis , ADN Complementario/genética , Femenino , Prueba de Tolerancia a la Glucosa , Glucógeno/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Proteínas Sustrato del Receptor de Insulina , Resistencia a la Insulina/genética , Resistencia a la Insulina/fisiología , Lípidos/sangre , Hígado/metabolismo , Masculino , Ratones , Ratones Transgénicos , Músculo Esquelético/metabolismo , Fenotipo , Fosfoproteínas/biosíntesis , Fosfoproteínas/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
J Invest Dermatol ; 122(4): 971-83, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15102088

RESUMEN

Peroxisome proliferator-activated receptor (PPAR) are nuclear hormone receptors that are activated by endogenous lipid metabolites. Previous studies have demonstrated that PPAR-alpha activation stimulates keratinocyte differentiation in vitro and in vivo, is anti-inflammatory, and improves barrier homeostasis. Recent studies have shown that PPAR-beta/delta activation induces keratinocyte differentiation in vitro. This study demonstrated that topical treatment of mice with a selective PPAR-beta/delta agonist (GW1514) in vivo had pro-differentiating effects, was anti-inflammatory, improved barrier homeostasis, and stimulated differentiation in a disease model of epidermal hyperproliferation [corrected]. In contrast to PPAR-alpha activation, PPAR-beta/deltain vivo did not display anti-proliferative or pro-apoptotic effects. The pro-differentiating effects persisted in mice lacking PPAR-alpha, but were decreased in mice deficient in retinoid X receptor-alpha, the major heterodimerization partner of PPAR. Furthermore, in vitro PPAR-beta/delta activation, aside from stimulating differentiation-related genes, additionally induced adipose differentiation-related protein (ADRP) and fasting induced adipose factor (FIAF) mRNA in cultures keratinocytes, which was paralleled by increased oil red O staining indicative of lipid accumulation, the bulk of which were triglycerides (TG). Comparison of differentially expressed genes between PPAR-beta/delta and PPAR-alpha activation revealed distinct profiles. Together, these studies indicate that PPAR-beta/delta activation stimulates keratinocyte differentiation, is anti-inflammatory, improves barrier homeostasis, and stimulates TG accumulation in keratinocytes.


Asunto(s)
Queratinocitos/citología , Queratinocitos/metabolismo , Metabolismo de los Lípidos , Receptores Citoplasmáticos y Nucleares/fisiología , Factores de Transcripción/fisiología , Administración Tópica , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Citocinas/farmacología , Dermatitis/prevención & control , Células Epidérmicas , Epidermis/metabolismo , Regulación de la Expresión Génica , Humanos , Queratinocitos/efectos de los fármacos , Queratinocitos/efectos de la radiación , Masculino , Ratones , Ratones Pelados , Permeabilidad , Receptores Citoplasmáticos y Nucleares/agonistas , Tiazoles/administración & dosificación , Tiazoles/farmacología , Factores de Transcripción/agonistas , Rayos Ultravioleta
19.
J Cell Sci ; 117(Pt 10): 2029-36, 2004 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15054105

RESUMEN

Recently, considerable interest has focused on the ability of activated peroxisome proliferator-activated receptor gamma (PPARgamma) to promote cytodifferentiation in adipocytes and some carcinoma cells; however, the role of PPARgamma in normal epithelial cytodifferentiation is unknown. Using uroplakin (UP) gene expression as a specific correlate of terminal urothelial cytodifferentiation, we investigated the differentiation-inducing effects of PPARgamma activation in normal human urothelial (NHU) cells grown as finite cell lines in monoculture. Two high-affinity activators of PPARgamma, troglitazone (TZ) and rosiglitazone (RZ) induced the expression of mRNA for UPII and UPIb and, to a lesser extent, UPIa. The specificity of the effect was shown by pretreating cells with a PPARgamma antagonist, GW9662, which attenuated the TZ-induced response in a dose-specific manner. The PPARgamma-mediated effect on UP gene expression was maximal when there was concurrent inhibition of autocrine-activated epidermal growth factor receptor (EGFR) signalling through either the phosphatidylinositol 3-kinase or extracellular signal-regulated kinase (ERK) pathways. The use of a specific EGFR tyrosine kinase inhibitor, PD153035, correlated with PPARgamma dephosphorylation and translocation to the nucleus, indicating a mechanism for regulating the balance between proliferation and differentiation. This is the first identification of specific factors involved in regulating differentiation-associated gene changes in urothelium and the first unambiguous evidence of a role for PPARgamma signalling in the terminal differentiation programme of a normal epithelium.


Asunto(s)
Receptores ErbB/metabolismo , PPAR gamma/fisiología , Urotelio/metabolismo , Transporte Activo de Núcleo Celular , Adipocitos/metabolismo , Anilidas/farmacología , Animales , Western Blotting , Bovinos , Diferenciación Celular , Línea Celular , Proliferación Celular , Cromanos/metabolismo , Inhibidores Enzimáticos/farmacología , Factor de Crecimiento Epidérmico/metabolismo , Epitelio/metabolismo , Regulación de la Expresión Génica , Humanos , Inmunoprecipitación , Ligandos , Glicoproteínas de Membrana/metabolismo , Proteínas de la Membrana/metabolismo , Microscopía Fluorescente , PPAR gamma/metabolismo , Fosforilación , Regiones Promotoras Genéticas , Quinazolinas/farmacología , ARN Mensajero/metabolismo , Ribonucleasas/metabolismo , Transducción de Señal , Tiazolidinedionas/metabolismo , Factores de Tiempo , Troglitazona , Uroplaquina II , Uroplaquina III , Uroplaquina Ia , Uroplaquina Ib , Urotelio/citología
20.
Diabetes ; 52(1): 43-50, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12502492

RESUMEN

Since leptin levels are independently correlated with risk of coronary heart disease, we have identified signaling pathways important in mediating leptin production and lipogenesis in human preadipocytes. We used inhibitors of p70(S6) kinase, p42/44 mitogen-activated protein kinase (MAPK), p38 MAPK, and phosphatidylinositol 3-kinase (PI3K). Human preadipocytes were induced to differentiate in insulin, dexamethasone, triiodothyronine, and 3-isobutyl-1-methylxanthine in the presence or absence of inhibitors and the peroxisome proliferator-activated receptor (PPAR)-gamma activator rosiglitazone. Differentiation was assessed by measuring leptin secretion, lipid content, and lipogenic activity. Rosiglitazone increased cell protein by 15%, the lipid content of the cell layer was doubled, and the lipogenic activity increased sevenfold but did not stimulate leptin secretion. None of the inhibitors significantly inhibited protein content over 20 days, but lipid content and lipogenic activity were inhibited by p70(S6) kinase and p38 MAPK inhibition but not by p42/44 MAPK or PI3K inhibition. All of the inhibitors significantly decreased leptin secretion, and these inhibitory effects were increased by coincubation with rosiglitazone. We conclude that PI3K and p42/44 MAPK pathways are not critical to the differentiation program leading to lipid accumulation, but stimulation of leptin secretion is dependent on these as well as the p70(S6) kinase and p38 MAPK signaling pathways.


Asunto(s)
Adipocitos/citología , Adipocitos/metabolismo , Leptina/biosíntesis , Lípidos/biosíntesis , Transducción de Señal/fisiología , Tiazoles/farmacología , Tiazolidinedionas , Androstadienos/farmacología , Diferenciación Celular/fisiología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Humanos , Imidazoles/farmacología , Leptina/metabolismo , Metabolismo de los Lípidos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Piridinas/farmacología , Rosiglitazona , Sirolimus/farmacología , Células Madre/citología , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Wortmanina , Proteínas Quinasas p38 Activadas por Mitógenos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...