Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Differentiation ; 115: 53-61, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32891959

RESUMEN

Hematopoietic stem cell-containing intra-aortic hematopoietic cell clusters (IAHCs) emerge in the dorsal aorta of the aorta-gonad-mesonephros (AGM) region during midgestation mouse embryos. We previously showed that transduction of Sox17 in CD45lowc-Kithigh cells, which are one component of IAHCs, maintained the cluster formation and the undifferentiated state, but the mechanism of the cluster formation by Sox17 has not been clarified. By microarray gene expression analysis, we found that genes for vascular endothelial-cadherin (VE-cad) and endothelial cell-selective adhesion molecule (ESAM) were expressed at high levels in Sox17-transduced c-Kit+ cells. Here we show the functional role of these adhesion molecules in the formation of IAHCs and the maintenance of the undifferentiated state by in vitro experiments. We detected VE-cad and ESAM expression in endothelial cells of dorsal aorta and IAHCs in E10.5 embryos by whole mount immunohistochemistry. Cells with the middle expression level of VE-cad and the low expression level of ESAM had the highest colony-forming ability. Tamoxifen-dependent nuclear translocation of Sox17-ERT fusion protein induced the formation of cell clusters and the expression of Cdh5 (VE-cad) and ESAM genes. We showed the induction of the Cdh5 (VE-cad) and ESAM expression and the direct interaction of Sox17 with their promoter by luciferase assay and chromatin immunoprecipitation assay, respectively. Moreover, shRNA-mediated knockdown of either Cdh5 (VE-cad) or ESAM gene in Sox17-transduced cells decreased the multilineage-colony forming potential. These findings suggest that VE-cad and ESAM play an important role in the high hematopoietic activity of IAHCs and cluster formation.


Asunto(s)
Antígenos CD/genética , Cadherinas/genética , Moléculas de Adhesión Celular/genética , Diferenciación Celular/genética , Proteínas HMGB/genética , Hematopoyesis/genética , Factores de Transcripción SOXF/genética , Animales , Aorta/crecimiento & desarrollo , Aorta/metabolismo , Cadherinas/antagonistas & inhibidores , Moléculas de Adhesión Celular/antagonistas & inhibidores , Embrión de Mamíferos , Células Endoteliales/citología , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas HMGB/antagonistas & inhibidores , Células Madre Hematopoyéticas/citología , Humanos , Ratones , Embarazo , ARN Interferente Pequeño/farmacología , Factores de Transcripción SOXF/antagonistas & inhibidores
2.
Front Immunol ; 10: 709, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31024546

RESUMEN

The release of the prototypic DAMP High Mobility Group Box 1 (HMGB1) into extracellular environment and its binding to the Receptor for Advanced Glycation End Products (RAGE) has been described to trigger sterile inflammation and regulate healing outcome. However, their role on host response to Ti-based biomaterials and in the subsequent osseointegration remains unexplored. In this study, HMGB1 and RAGE inhibition in the Ti-mediated osseointegration were investigated in C57Bl/6 mice. C57Bl/6 mice received a Ti-device implantation (Ti-screw in the edentulous alveolar crest and a Ti-disc in the subcutaneous tissue) and were evaluated by microscopic (microCT [bone] and histology [bone and subcutaneous]) and molecular methods (ELISA, PCR array) during 3, 7, 14, and 21 days. Mice were divided into 4 groups: Control (no treatment); GZA (IP injection of Glycyrrhizic Acid for HMGB1 inhibition, 4 mg/Kg/day); RAP (IP injection of RAGE Antagonistic Peptide, 4 mg/Kg/day), and vehicle controls (1.5% DMSO solution for GZA and 0.9% saline solution for RAP); treatments were given at all experimental time points, starting 1 day before surgeries. HMGB1 was detected in the Ti-implantation sites, adsorbed to the screws/discs. In Control and vehicle groups, osseointegration was characterized by a slight inflammatory response at early time points, followed by a gradual bone apposition and matrix maturation at late time points. The inhibition of HMGB1 or RAGE impaired the osseointegration, affecting the dynamics of mineralized and organic bone matrix, and resulting in a foreign body reaction, with persistence of macrophages, necrotic bone, and foreign body giant cells until later time points. While Control samples were characterized by a balance between M1 and M2-type response in bone and subcutaneous sites of implantation, and also MSC markers, the inhibition of HMGB1 or RAGE caused a higher expression M1 markers and pro-inflammatory cytokines, as well chemokines and receptors for macrophage migration until later time points. In conclusion, HMGB1 and RAGE have a marked role in the osseointegration, evidenced by their influence on host inflammatory immune response, which includes macrophages migration and M1/M2 response, MSC markers expression, which collectively modulate bone matrix deposition and osseointegration outcome.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Artroplastia/métodos , Materiales Biocompatibles/metabolismo , Proteínas HMGB/metabolismo , Inflamación/inmunología , Macrófagos/inmunología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Titanio/metabolismo , Animales , Materiales Biocompatibles/química , Biomarcadores/metabolismo , Matriz Ósea/efectos de los fármacos , Movimiento Celular , Ácido Glicirrínico/administración & dosificación , Proteínas HMGB/antagonistas & inhibidores , Humanos , Inmunomodulación , Ratones , Ratones Endogámicos C57BL , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Oseointegración , Péptidos/administración & dosificación , Titanio/química
3.
Mol Biol Rep ; 39(1): 227-31, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21556772

RESUMEN

High mobility group box 1 protein (HMGB1) plays an important role in myocardial ischemia and reperfusion (I/R) injury. Ethyl pyruvate (EP), a potent reactive oxygen species scavenger, has been reported to inhibit myocardial apoptosis and reduce myocardial I/R injury. The aim of this study was to investigate the mechanism by which EP reduces myocardial I/R injury in rats. Anesthetized male rats were once treated with EP (50 mg/kg, i.p.) before ischemia, and then subjected to ischemia for 30 min followed by reperfusion for 4 h. Lactate dehydrogenase (LDH), creatine kinase (CK), malondialdehyde (MDA), superoxide dismutase (SOD) activity and infarct size were measured. HMGB1 expression was assessed by immunoblotting. The results showed that pretreatment of EP (50 mg/kg) could significantly reduce the infarct size and the levels of LDH and CK after 4 h reperfusion (all P<0.05). EP could also significantly inhibit the increase of the MDA level, the decrease of the SOD level (both P<0.05). Meanwhile, EP could significantly inhibit the expression of HMGB1 induced by I/R. The present study suggested that ethyl pyruvate could attenuate myocardial I/R injury by inhibiting HMGB1 expression.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Proteínas HMGB/antagonistas & inhibidores , Daño por Reperfusión Miocárdica/prevención & control , Piruvatos/farmacología , Análisis de Varianza , Animales , Creatina Quinasa/metabolismo , Immunoblotting , L-Lactato Deshidrogenasa/metabolismo , Masculino , Malondialdehído/metabolismo , Daño por Reperfusión Miocárdica/patología , Ratas , Superóxido Dismutasa/metabolismo
4.
J Neurosci ; 31(39): 13921-35, 2011 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-21957254

RESUMEN

The SRY-box (Sox) transcription factors regulate oligodendrocyte differentiation, but their signaling targets are largely unknown. We have identified a major signal transduction pathway regulated by Sox containing gene 17 (Sox17) in the oligodendrocyte lineage. Microarray analysis in oligodendrocyte progenitor cells (OPCs) after Sox17 attenuation revealed upregulated genes associated with cell cycle control and activation of the Wingless and integration site (Wnt)/ß-catenin pathway. Sox17 knockdown also increases the levels of cyclin D1, Axin2, and activated ß-catenin. In OPCs, the expression pattern of Sox17, cyclin D1, and secreted Frizzled-related protein-1 in the presence of platelet-derived growth factor (PDGF) was coordinately accelerated by addition of thyroid hormone, indicating differentiation-induced regulation of Sox17 targets. In developing white matter, decreased total ß-catenin, activated ß-catenin, and cyclin D1 levels coincided with the peak of Sox17 expression, and immunoprecipitates showed a developmentally regulated interaction among Sox17, T-cell transcription factor 4, and ß-catenin proteins. In OPCs, PDGF stimulated phosphorylation of glycogen synthase 3ß and the Wnt coreceptor LRP6, and enhanced ß-catenin-dependent gene expression. Sox17 overexpression inhibited PDGF-induced TOPFLASH and cyclin D1 promoter activity, and decreased endogenous cyclin D1, activated ß-catenin, as well as total ß-catenin levels. Recombinant Sox17 prevented Wnt3a from repressing myelin protein expression, and inhibition of Sox17-mediated proteasomal degradation of ß-catenin blocked myelin protein induction. These results indicate that Sox17 suppresses cyclin D1 expression and cell proliferation by directly antagonizing ß-catenin, whose activity in OPCs is stimulated not only by Wnt3a, but also by PDGF. Our identification of downstream targets of Sox17 thus defines signaling pathways and molecular mechanisms in OPCs that are regulated by Sox17 during cell cycle exit and the onset of differentiation in oligodendrocyte development.


Asunto(s)
Proteínas HMGB/fisiología , Oligodendroglía/fisiología , Factores de Transcripción SOXF/fisiología , Transducción de Señal/fisiología , Células Madre/fisiología , Proteínas Wnt/fisiología , beta Catenina/fisiología , Animales , Células Cultivadas , Técnicas de Sustitución del Gen , Proteínas HMGB/antagonistas & inhibidores , Proteínas HMGB/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de la Mielina/antagonistas & inhibidores , Proteínas de la Mielina/biosíntesis , Células 3T3 NIH , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Factores de Transcripción SOXF/antagonistas & inhibidores , Factores de Transcripción SOXF/genética , beta Catenina/antagonistas & inhibidores
5.
Expert Rev Anti Infect Ther ; 9(7): 807-22, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21810053

RESUMEN

Influenza A virus is a significant cause of morbidity and mortality worldwide. Severe influenza is recognized as a clinical syndrome, characterized by hyperinduction of proinflammatory cytokine production, otherwise known as hypercytokinemia or a 'cytokine storm'. Research focused on therapeutics to modulate influenza virus-induced inflammation is currently underway. In this review, we discuss the limitations of current antiviral drug treatment strategies, describe the influenza viral and host pathogenicity determinants, and present the evidence supporting the use of immunomodulatory therapy to target the host inflammatory response as a means to improve clinical outcome in severe influenza. We then review the experimental data on investigational immunomodulatory agents targeting the host inflammatory response in severe influenza, including anti-TNF therapy, statins, glucocorticoids, cyclooxygenase-2 inhibitors, macrolides, peroxisome proliferator-activated receptor agonists, AMP-activated protein kinase agonists and high mobility group box 1 antagonists. We then conclude with a rationale for the use of mesenchymal stromal (stem) cells and angiopoietin-1 therapy against deleterious influenza-induced host responses that mediate end-organ injury and dysfunction.


Asunto(s)
Factores Inmunológicos/uso terapéutico , Inmunomodulación , Virus de la Influenza A/efectos de los fármacos , Gripe Humana , Proteínas Quinasas Activadas por AMP/metabolismo , Angiopoyetina 1/administración & dosificación , Angiopoyetina 1/uso terapéutico , Antivirales/administración & dosificación , Antivirales/uso terapéutico , Ciclooxigenasa 2/metabolismo , Inhibidores de la Ciclooxigenasa 2/uso terapéutico , Glucocorticoides/administración & dosificación , Glucocorticoides/uso terapéutico , Proteínas HMGB/antagonistas & inhibidores , Proteínas HMGB/metabolismo , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Virus de la Influenza A/crecimiento & desarrollo , Virus de la Influenza A/inmunología , Vacunas contra la Influenza/uso terapéutico , Gripe Humana/inmunología , Gripe Humana/patología , Gripe Humana/prevención & control , Gripe Humana/terapia , Gripe Humana/virología , Macrólidos/uso terapéutico , Células Madre Mesenquimatosas/citología , Receptores Activados del Proliferador del Peroxisoma/agonistas , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Trasplante de Células Madre , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/biosíntesis
6.
Br J Cancer ; 98(4): 824-31, 2008 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-18268498

RESUMEN

SOX transcription factors are essential for embryonic development and play critical roles in cell fate determination, differentiation and proliferation. We previously reported that the SOX2 protein is expressed in normal gastric mucosae but downregulated in some human gastric carcinomas. To clarify the roles of SOX2 in gastric carcinogenesis, we carried out functional characterisation of SOX2 in gastric epithelial cell lines. Exogenous expression of SOX2 suppressed cell proliferation in gastric epithelial cell lines. Flow cytometry analysis revealed that SOX2-overexpressing cells exhibited cell-cycle arrest and apoptosis. We found that SOX2-mediated cell-cycle arrest was associated with decreased levels of cyclin D1 and phosphorylated Rb, and an increased p27(Kip1) level. These cells exhibited further characteristics of apoptosis, such as DNA laddering and caspase-3 activation. SOX2 hypermethylation signals were observed in some cultured and primary gastric cancers with no or weak SOX2 expression. Among the 52 patients with advanced gastric cancers, those with cancers showing SOX2 methylation had a significantly shorter survival time than those without this methylation (P=0.0062). Hence, SOX2 plays important roles in growth inhibition through cell-cycle arrest and apoptosis in gastric epithelial cells, and the loss of SOX2 expression may be related to gastric carcinogenesis and poor prognosis.


Asunto(s)
Apoptosis , Proliferación Celular , Proteínas de Unión al ADN/metabolismo , Silenciador del Gen , Proteínas HMGB/metabolismo , Neoplasias Gástricas/metabolismo , Factores de Transcripción/metabolismo , Western Blotting , Caspasas/metabolismo , Ciclo Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Metilación de ADN , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Regulación hacia Abajo , Citometría de Flujo , Proteínas HMGB/antagonistas & inhibidores , Proteínas HMGB/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Fosforilación , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/farmacología , Proteína de Retinoblastoma , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción SOXB1 , Neoplasias Gástricas/patología , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Células Tumorales Cultivadas
7.
DNA Cell Biol ; 21(3): 199-212, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12015897

RESUMEN

Overcoming local DNA rigidity is required to perform three-dimensional DNA-protein configuration at promoter regions. The abundant architectural nonhistone chromosomal HMG box proteins are nonsequence-specific; however, they have been established to specifically recognize distorted DNA. Using transient transfection to overexpress two different members of the HMGB-1/2 family of DNA architectural factors, we demonstrate that these proteins provide a general enhancement in reporter gene expression irrespective of the promoter being considered. Evidences are also provided indicating that stimulation may not be achieved by recruitment of the proteins by regulatory factors or as a consequence of major chromatin unfolding as previously suggested. Interestingly, the influence of the HMG box proteins under study was overridden when the promoters were either induced or stimulated by Trichostatin A (TSA) but recovered upon extended induction period. These results also support the concept that the architectural role of these proteins can contribute to the preinitiation complex assembly required for basal transcription, but to a much lesser extent to the poised promoter scaffolding characteristic of activated transcription.


Asunto(s)
Proteínas HMGB/fisiología , Activación Transcripcional , Línea Celular , Cromosomas/química , Genes Reporteros , Proteínas HMGB/antagonistas & inhibidores , Proteína HMGB1/fisiología , Proteína HMGB2/fisiología , Humanos , Ácidos Hidroxámicos/farmacología , Cinética , Regiones Promotoras Genéticas , Transactivadores/fisiología , Activación Transcripcional/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA