Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Carbohydr Polym ; 278: 118944, 2022 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-34973762

RESUMEN

Klebsiella pneumoniae serotype KN2 is a carbapenem-resistant strain and leads to the health care-associated infections, such as bloodstream infections. Its capsular polysaccharide (CPS) was isolated and cleaved by a specific enzyme from a bacteriophage into a hexasaccharide-repeating unit. With GC-MS, NMR, and Mass analyses, the structure of KN2 CPS was determined to be {→3)-ß-D-Glcp-(1→3)-[α-D-GlcpA-(1→4)-ß-D-Glcp-(1→6)]-α-D-Galp-(1→6)-ß-D-Galp-(1→3)-ß-D-Galp-(1→}n. We demonstrated that 1 µg/mL CPS could stimulate J774A.1 murine macrophages to release tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) in vitro. Also, we proved that KN2 CPS induced the immune response through Toll-like receptor 4 (TLR4) in the human embryonic kidney (HEK)-293 cells. Strikingly, the hexasaccharide alone shows the same immune response as the CPS, suggesting that the hexasaccharide can shape the adaptive immunity to be a potential vaccine adjuvant. The glucuronic acid (GlcA) on other polysaccharides can affect the immune response, but the GlcA-reduced KN2 CPS and hexasaccharide still maintain their immunomodulatory activities.


Asunto(s)
Antibacterianos/farmacología , Carbapenémicos/farmacología , Factores Inmunológicos/farmacología , Klebsiella pneumoniae/efectos de los fármacos , Polisacáridos Bacterianos/farmacología , Receptor Toll-Like 4/inmunología , Antibacterianos/química , Carbapenémicos/química , Células HEK293 , Humanos , Factores Inmunológicos/química , Ligandos , Pruebas de Sensibilidad Microbiana , Polisacáridos Bacterianos/química
2.
Chem Commun (Camb) ; 57(50): 6209-6212, 2021 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-34059855

RESUMEN

Toll-like receptor 4 (TLR4) recognizes various protein ligands; however, the protein-TLR4 binding model is unclear. Here we demonstrate a Crenomytilus grayanus lectin (CGL)-TLR4/MD2 model to show that CGL interacts with a TLR4/myeloid differentiation factor 2 (MD2) complex independently of sugar-binding properties. CGL could suppress lipopolysaccharide-induced immune responses significantly, suggesting that TLR4 itself has potential as a therapeutic target.


Asunto(s)
Carbohidratos/química , Lectinas/química , Antígeno 96 de los Linfocitos/química , Receptor Toll-Like 4/química , Animales , Sitios de Unión , Bivalvos , Carbohidratos/inmunología , Humanos , Lectinas/inmunología , Antígeno 96 de los Linfocitos/inmunología , Receptor Toll-Like 4/inmunología
4.
Front Immunol ; 11: 607564, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33424855

RESUMEN

Conjugated polyenes are a class of widely occurring natural products with various biological functions. We previously identified 4-hydroxy auxarconjugatin B (4-HAB) as anti-inflammatory agent with an IC50 of ~20 µM. In this study, we synthesized a new anti-inflammatory 4-HAB analogue, F240B, which has an IC50 of less than 1 µM. F240B dose-dependently induced autophagy by increasing autophagic flux, LC3 speck formation and acidic vesicular organelle formation. F240B inhibited NACHT, LRR and PYD domain-containing protein 3 (NLRP3) inflammasome activation through autophagy induction. In a mechanistic study, F240B inhibited interleukin (IL)-1ß (IL-1ß) precursor expression, promoted degradation of NLRP3 and IL-1ß, and reduced mitochondrial membrane integrity loss in an autophagy-dependent manner. Additionally, F240B inhibited apoptosis-associated speck-like protein containing a CARD (ASC) oligomerization and speck formation without affecting the interaction between NLRP3 and ASC or NIMA-related kinase 7 (NEK7) and double-stranded RNA-dependent kinase (PKR). Furthermore, F240B exerted in vivo anti-inflammatory activity by reducing the intraperitoneal influx of neutrophils and the levels of IL-1ß, active caspase-1, IL-6 and monocyte chemoattractant protein-1 (MCP-1) in lavage fluids in a mouse model of uric acid crystal-induced peritonitis. In conclusion, F240B attenuated the NLRP3 inflammasome through autophagy induction and can be developed as an anti-inflammatory agent in the future.


Asunto(s)
Antiinflamatorios/farmacología , Autofagia/efectos de los fármacos , Inflamasomas/metabolismo , Macrófagos/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Peritonitis/prevención & control , Animales , Antiinflamatorios/síntesis química , Proteínas Relacionadas con la Autofagia/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Mediadores de Inflamación/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/patología , Peritonitis/inducido químicamente , Peritonitis/metabolismo , Peritonitis/patología , Estabilidad Proteica , Células RAW 264.7 , Transducción de Señal , Células THP-1 , Ácido Úrico
5.
J Cell Biochem ; 116(8): 1785-96, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25676701

RESUMEN

We have previously shown that PTP-oc is an enhancer of the functional activity of osteoclasts and that EphA4 is a suppressor. Here, we provide evidence that PTP-oc enhances osteoclast activity in part through inactivation of EphA4 by dephosphorylating key phosphotyrosine (pY) residues of EphA4. We show that EphA4 was pulled down by the PTP-oc trapping mutant but not by the wild-type (WT) PTP-oc and that transgenic overexpression of PTP-oc in osteoclasts drastically decreased pY602 and pY779 residues of EphA4. Consistent with the previous findings that EphA4 deficiency increased pY173-Vav3 level (Rac-GTP exchange factor [GEF]) and enhanced bone resorption activity of osteoclasts, reintroduction of WT-Epha4 in Epha4 null osteoclasts led to ∼50% reduction in the pY173-Vav3 level and ∼2-fold increase in bone resorption activity. Overexpression of Y779F-Epha4 mutant in WT osteoclasts markedly increased in pY173-Vav3 and reduced bone resorption activity, but overexpression of Y602F-Epha4 mutant had no effect, suggesting that pY779 residue plays an important role in the EphA4-mediated suppression of osteoclast activity. Deficient EphA4 in osteoclasts has been shown to up-regulate Rac-GTPase and down-regulate Rho-GTPase. PTP-oc overexpression in osteoclasts also increased the GTP-Rac level to 300% of controls, but decreased the GTP-Rho level to ∼50% of controls. PTP-oc overexpression or deficient Epha4 each also reduced pY87-Ephexin level, which is a Rho GEF. Thus, PTP-oc may differentially regulate Rac signaling versus Rho signaling through dephosphorylation of EphA4, which has shown to have opposing effects on Rac-GTPase versus Rho-GTPase through differential regulation of Vav3 versus Ephexin.


Asunto(s)
Resorción Ósea/metabolismo , Osteoclastos/fisiología , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Receptor EphA4/metabolismo , Tirosina/metabolismo , Animales , Resorción Ósea/genética , Línea Celular , Masculino , Ratones , Ratones Transgénicos , Mutación , Fosforilación , Proteínas Tirosina Fosfatasas no Receptoras/química , Receptor EphA4/genética , Transducción de Señal
6.
PLoS One ; 9(7): e101679, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25007054

RESUMEN

RASSF1C is a major isoform of the RASSF1 gene, and is emerging as an oncogene. This is in contradistinction to the RASSF1A isoform, which is an established tumor suppressor. We have previously shown that RASSF1C promotes lung cancer cell proliferation and have identified RASSF1C target genes with growth promoting functions. Here, we further report that RASSF1C promotes lung cancer cell migration and enhances lung cancer cell tumor sphere formation. We also show that RASSF1C over-expression reduces the inhibitory effects of the anti-cancer agent, betulinic acid (BA), on lung cancer cell proliferation. In previous work, we demonstrated that RASSF1C up-regulates piwil1 gene expression, which is a stem cell self-renewal gene that is over-expressed in several human cancers, including lung cancer. Here, we report on the effects of BA on piwil1 gene expression. Cells treated with BA show decreased piwil1 expression. Also, interaction of IGFBP-5 with RASSF1C appears to prevent RASSF1C from up-regulating PIWIL1 protein levels. These findings suggest that IGFBP-5 may be a negative modulator of RASSF1C/ PIWIL1 growth-promoting activities. In addition, we found that inhibition of the ATM-AMPK pathway up-regulates RASSF1C gene expression.


Asunto(s)
Proteínas Argonautas/metabolismo , Expresión Génica , Proteína 5 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Proteínas Supresoras de Tumor/genética , Antineoplásicos/farmacología , Proteínas Argonautas/genética , Movimiento Celular , Proliferación Celular , Resistencia a Antineoplásicos , Regulación Neoplásica de la Expresión Génica , Humanos , Ácidos Hidroxámicos/farmacología , Proteína 5 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Neoplasias Pulmonares , Triterpenos Pentacíclicos , Pirazoles/farmacología , Pirimidinas/farmacología , Transducción de Señal , Esferoides Celulares , Triterpenos/farmacología , Proteínas Supresoras de Tumor/metabolismo , beta Catenina/genética , beta Catenina/metabolismo , Ácido Betulínico
7.
PLoS One ; 9(5): e98004, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24848992

RESUMEN

This study sought to determine if direct application of the lentiviral (LV)-cyclooxygenase 2 (COX2) vector to the tendon-bone interface would promote osteointegration of the tendon graft in a rat model of biceps tenodesis. The LV-COX2 gene transfer strategy was chosen for investigation because a similar COX2 gene transfer strategy promoted bony bridging of the fracture gap during bone repair, which involves similar histologic transitions that occur in osteointegration. Briefly, a 1.14-mm diameter tunnel was drilled in the mid-groove of the humerus of adult Fischer 344 rats. The LV-COX2 or ßgal control vector was applied directly into the bone tunnel and onto the end of the tendon graft, which was then pulled into the bone tunnel. A poly-L-lactide pin was press-fitted into the tunnel as interference fixation. Animals were sacrificed at 3, 5, or 8 weeks for histology analysis of osteointegration. The LV-COX2 gene transfer strategy enhanced neo-chondrogenesis at the tendon-bone interface but with only marginal effect on de novo bone formation. The tendon-bone interface of the LV-COX2-treated tenodesis showed the well-defined tendon-to-fibrocartilage-to-bone histologic transitions that are indicative of osteointegration of the tendon graft. The LV-COX2 in vivo gene transfer strategy also significantly enhanced angiogenesis at the tendon-bone interface. To determine if the increased osteointegration was translated into an improved pull-out mechanical strength property, the pull-out tensile strength of the LV-COX2-treated tendon grafts was determined with a pull-out mechanical testing assay. The LV-COX2 strategy yielded a significant improvement in the return of the pull-out strength of the tendon graft after 8 weeks. In conclusion, the COX2-based in vivo gene transfer strategy enhanced angiogenesis, osteointegration and improved return of the pull-out strength of the tendon graft. Thus, this strategy has great potential to be developed into an effective therapy to promote tendon-to-bone healing after tenodesis or related surgeries.


Asunto(s)
Huesos/fisiología , Ciclooxigenasa 2/genética , Lentivirus/genética , Tendones/trasplante , Tenodesis/métodos , Resistencia a la Tracción , Extremidad Superior/cirugía , Animales , Huesos/metabolismo , Huesos/cirugía , Condrogénesis , Técnicas de Transferencia de Gen , Oseointegración , Osteogénesis , Ratas , Tendones/cirugía , Cicatrización de Heridas
8.
Mol Biol Int ; 2013: 145096, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24327924

RESUMEN

RASSF1A has been demonstrated to be a tumor suppressor, while RASSF1C is now emerging as a growth promoting protein in breast and lung cancer cells. To further highlight the dual functionality of the RASSF1 gene, we have compared the effects of RASSF1A and RASSF1C on cell proliferation and apoptosis in the presence of TNF- α . Overexpression of RASSF1C in breast and lung cancer cells reduced the effects of TNF- α on cell proliferation, apoptosis, and MST1/2 phosphorylation, while overexpression of RASSF1A had the opposite effect. We also assessed the expression of RASSF1A and RASSF1C in breast and lung tumor and matched normal tissues. We found that RASSF1A mRNA levels are significantly higher than RASSF1C mRNA levels in all normal breast and lung tissues examined. In addition, RASSF1A expression is significantly downregulated in 92% of breast tumors and in 53% of lung tumors. Conversely, RASSF1C was upregulated in 62% of breast tumors and in 47% of lung tumors. Together, these findings suggest that RASSF1C, unlike RASSF1A, is not a tumor suppressor but instead may play a role in stimulating survival in breast and lung cancer cells.

9.
BMC Res Notes ; 5: 239, 2012 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-22591718

RESUMEN

BACKGROUND: RASSF1A and RASSF1C are two major isoforms encoded by the Ras association domain family 1 (RASSF1) gene through alternative promoter selection and mRNA splicing. RASSF1A is a well established tumor suppressor gene. Unlike RASSF1A, RASSF1C appears to have growth promoting actions in lung cancer. In this article, we report on the identification of novel RASSF1C target genes in non small cell lung cancer (NSCLC). METHODS: Over-expression and siRNA techniques were used to alter RASSF1C expression in human lung cancer cells, and Affymetrix-microarray study was conducted using NCI-H1299 cells over-expressing RASSF1C to identify RASSF1C target genes. RESULTS: The microarray study intriguingly shows that RASSF1C modulates the expression of a number of genes that are involved in cancer development, cell growth and proliferation, cell death, and cell cycle. We have validated the expression of some target genes using qRT-PCR. We demonstrate that RASSF1C over-expression increases, and silencing of RASSF1C decreases, the expression of PIWIL1 gene in NSCLC cells using qRT-PCR, immunostaining, and Western blot analysis. We also show that RASSF1C over-expression induces phosphorylation of ERK1/2 in lung cancer cells, and inhibition of the MEK-ERK1/2 pathway suppresses the expression of PIWIL1 gene expression, suggesting that RASSF1C may exert its activities on some target genes such as PIWIL1 through the activation of the MEK-ERK1/2 pathway. Also, PIWIL1 expression is elevated in lung cancer cell lines compared to normal lung epithelial cells. CONCLUSIONS: Taken together, our findings provide significant data to propose a model for investigating the role of RASSF1C/PIWIL1 proteins in initiation and progression of lung cancer.


Asunto(s)
Proteínas Argonautas/genética , Regulación Neoplásica de la Expresión Génica/genética , Células Madre Neoplásicas/metabolismo , Proteínas Supresoras de Tumor/genética , Apoptosis/genética , Proteínas Argonautas/metabolismo , Benzamidas/farmacología , Western Blotting , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Ciclo Celular/genética , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Inmunohistoquímica , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosforilación/efectos de los fármacos , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Supresoras de Tumor/metabolismo
10.
J Bone Miner Res ; 27(7): 1553-65, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22437732

RESUMEN

Claudin 18 (Cldn-18) belongs to a large family of transmembrane proteins that are important components of tight junction strands. Although several claudin members are expressed in bone, the functional role for any claudin member in bone is unknown. Here we demonstrate that disruption of Cldn-18 in mice markedly decreased total body bone mineral density, trabecular bone volume, and cortical thickness in Cldn-18(-/-) mice. Histomorphometric studies revealed that bone resorption parameters were increased significantly in Cldn-18(-/-) mice without changes in bone formation. Serum levels of tartrate-resistant acid phosphatase 5b (TRAP5b) and mRNA expression levels of osteoclast specific markers and signaling molecules were also increased. Loss of Cldn-18 further exacerbated calcium deficiency induced bone loss by influencing bone resorption, thereby resulting in mechanically weaker bone. In vitro studies with bone marrow macrophages revealed Cldn-18 disruption markedly enhanced receptor activator of NF-κB ligand (RANKL)-induced osteoclast differentiation but not macrophage colony-stimulating factor (MCSF)-induced bone marrow macrophage (BMM) proliferation. Consistent with a direct role for Cldn-18 in regulating osteoclast differentiation, overexpression of wild type but not PDZ binding motif deleted Cldn-18 inhibited RANKL-induced osteoclast differentiation. Furthermore, our findings indicate that Cldn-18 interacts with Zonula occludens 2 (ZO-2) to modulate RANKL signaling in osteoclasts. In conclusion, we demonstrate that Cldn-18 is a novel negative regulator of bone resorption and osteoclast differentiation.


Asunto(s)
Resorción Ósea , Claudinas/biosíntesis , Claudinas/fisiología , Osteoclastos/citología , Animales , Células de la Médula Ósea/citología , Diferenciación Celular , Proliferación Celular , Femenino , Genotipo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Ligando RANK/metabolismo , Uniones Estrechas/metabolismo , Proteína de la Zonula Occludens-2
11.
J Gene Med ; 13(10): 511-21, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21898721

RESUMEN

BACKGROUND: The present study aimed to develop a rat model of biceps tenodesis and to assess the feasibility of a lentiviral (LV)-based bone morphogenetic protein (BMP) 4 in vivo gene transfer strategy for healing of biceps tenodesis. METHODS: A rat model of biceps tenodesis was developed with an interference-fit open surgical technique. A LV vector expressing a BMP4 gene or ß-galactosidase (ß-gal) control gene was applied to the bone tunnel and the tendon graft before its insertion into the bone tunnel. Osteointegration was assessed by histology and pull-out tensile strength was measured by a biomechanical test suitable for small rat biceps tendon grafts. RESULTS: Neo-chondrogenesis was seen at the tendon-bone interface of LV-BMP4-treated but not control rats. The LV-BMP4-treated rats showed 32% (p < 0.05) more newly-formed trabecular bone at the tendon-bone junction than the LV-ß-gal-treated controls after 3 weeks. However, the sites of neo-chondrogenesis and new bone formation in the LV-BMP4-treated tenodesis were highly spotty. Although the LV-BMP4 strategy did not promote bony integration of the tendon graft, it yielded a 29.5 ± 11.8% (p = 0.066) increase in improvement the pull-out strength of rat biceps tendons compared to the LV-ß-gal treatment after 5 weeks. CONCLUSIONS: Although the LV-BMP4 in vivo gene transfer strategy did not enhance osteointegration of the tendon graft, it yielded a marked improvement in the return of the pull-out strength of the tendon graft. This presumably was largely a result of the bone formation effect of BMP4 that traps or anchors the tendon graft onto the bony tunnel.


Asunto(s)
Brazo/cirugía , Proteína Morfogenética Ósea 4/genética , Técnicas de Transferencia de Gen , Lentivirus/genética , Oseointegración/fisiología , Tendones/trasplante , Tenodesis , Animales , Brazo/anatomía & histología , Proteína Morfogenética Ósea 4/metabolismo , Condrogénesis/fisiología , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Humanos , Lentivirus/metabolismo , Masculino , Ensayo de Materiales , Modelos Animales , Ratas , Ratas Endogámicas F344 , Procedimientos de Cirugía Plástica/métodos , Estrés Mecánico , Resistencia a la Tracción
12.
Langmuir ; 27(13): 8424-9, 2011 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-21630657

RESUMEN

We describe a simple and effective method to obtain colloidal surface-functionalized Au nanoparticles. The method is primarily based on irradiation of a gold solution with high-flux X-rays from a synchrotron source in the presence of 11-mercaptoundecanoic acid (MUA). Extensive tests of the products demonstrated high colloidal density as well as excellent stability, shelf life, and biocompatibility. Specific tests with X-ray diffraction, UV-visible spectrometry, visible microscopy, Fourier transform infrared spectroscopy, dark-field visible-light scattering microscopy, and transmission electron microscopy demonstrated that MUA, being an effective surfactant, not only allows tunable size control of the nanoparticles, but also facilitates functionalization. The nanoparticle sizes were 6.45 ± 1.58, 1.83 ± 1.21, 1.52 ± 0.37 and 1.18 ± 0.26 nm with no MUA and with MUA-to-Au ratios of 1:2, 1:1, and 3:1. The MUA additionally enabled functionalization with l-glycine. We thus demonstrated flexibility in controlling the nanoparticle size over a large range with narrow size distribution.


Asunto(s)
Oro/química , Nanopartículas del Metal/química , Animales , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Coloides/química , Coloides/farmacología , Relación Dosis-Respuesta a Droga , Ácidos Grasos/química , Oro/farmacología , Ratones , Tamaño de la Partícula , Relación Estructura-Actividad , Compuestos de Sulfhidrilo/química , Propiedades de Superficie
13.
Anal Bioanal Chem ; 401(3): 809-16, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21537916

RESUMEN

Well-dispersed gold nanoparticles (NP) coated with tiopronin were synthesized by X-ray irradiation without reducing agents. High-resolution transmission electron microscopy shows that the average core diameters of the NPs can be systematically controlled by adjusting the tiopronin to Au mole ratio in the reaction. Three methods were used to study the NP uptake by cells: quantitative measurements by inductively coupled plasma mass spectrometry, direct imaging with high lateral resolution transmission electron microscopy and transmission X-ray microscopy. The results confirmed that the NP internalization mostly occurred via endocytosis and concerned the cytoplasm. The particles, in spite of their small sizes, were not found to arrive inside the cell nuclei. The synthesis without reducing agents and solvents increased the biocompatibility as required for potential applications in analysis and biomedicine in general.


Asunto(s)
Endocitosis , Oro/metabolismo , Nanopartículas del Metal/química , Tiopronina/química , Supervivencia Celular , Células/diagnóstico por imagen , Células/metabolismo , Células Cultivadas , Humanos , Espectrometría de Masas , Microscopía Electrónica de Transmisión , Radiografía , Sincrotrones , Tiopronina/síntesis química , Rayos X
14.
Am J Physiol Endocrinol Metab ; 301(1): E40-8, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21467300

RESUMEN

Although thyroid hormone (TH) is known to exert important effects on the skeleton, the nuclear factors constituting the TH receptor coactivator complex and the molecular pathways by which TH mediates its effects on target gene expression in osteoblasts remain poorly understood. A recent study demonstrated that the actions of TH on myoblast differentiation are dependent on diabetes- and obesity-related protein (DOR). However, the role of DOR in osteoblast differentiation is unknown. We found DOR expression increased during in vitro differentiation of bone marrow stromal cells into osteoblasts and also in MC3T3-E1 cells treated with TH. However, DOR expression decreased during cellular proliferation. To determine whether DOR acts as a modulator of TH action during osteoblast differentiation, we examined whether overexpression or knockdown of DOR in MC3T3-E1 cells affects the ability of TH to induce osteoblast differentiation by evaluating alkaline phosphatase (ALP) activity. ALP activity was markedly increased in DOR-overexpressing cells treated with TH. In contrast, loss of DOR dramatically reduced TH stimulation of ALP activity in MC3T3-E1 cells and primary calvaria osteoblasts transduced with lentiviral DOR shRNA. Consistent with reduced ALP activity, mRNA levels of osteocalcin, ALP, and Runx2 were decreased significantly in DOR shRNA cells. In addition, a common single nucleotide polymorphism (SNP), DOR1 found on the promoter of human DOR gene, was associated with circulating osteocalcin levels in nondiabetic subjects. Based on these data, we conclude that DOR plays an important role in TH-mediated osteoblast differentiation, and a DOR SNP associates with plasma osteocalcin in men.


Asunto(s)
Diferenciación Celular/genética , Proteínas Nucleares/fisiología , Osteoblastos/fisiología , Adulto , Anciano , Animales , Animales Recién Nacidos , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteocalcina/sangre , Osteogénesis/efectos de los fármacos , Osteogénesis/genética , Osteogénesis/fisiología , Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas/genética , ARN Interferente Pequeño/farmacología
15.
J Gene Med ; 13(2): 77-88, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21322098

RESUMEN

BACKGROUND: The present study assesses the effect of the stem cell antigen-1 positive (Sca-1(+) ) cell-based human growth hormone (hGH) ex vivo gene transfer strategy on endosteal bone mass in the mouse. METHODS: Sublethally irradiated recipient mice were transplanted with Sca-1(+) cells transduced with lentiviral vectors expressing hGH or ß-galactosidase control genes. Bone parameters were assessed by micro-computed tomography and histomorphometry. RESULTS: This hGH strategy drastically increased hGH mRNA levels in bone marrow cells and serum insulin-like growth factor-I (IGF-I) (by nearly 50%, p < 0.002) in hGH recipient mice. Femoral trabecular bone volume of the hGH mice was significantly reduced by 35% (p < 0.002). The hGH mice also had decreased trabecular number (by 26%; p < 0.0001), increased trabecular separation (by 38%; p < 0.0002) and reduced trabecular connectivity density (by 64%; p < 0.001), as well as significantly more osteoclasts (2.5-fold; p < 0.05) and greater osteoclastic surface per bone surface (2.6-fold; p < 0.01). CONCLUSIONS: Targeted expression of hGH in cells of marrow cavity through the Sca-1(+) cell-based gene transfer strategy increased circulating IGF-I and decreased endosteal bone mass through an increase in resorption in recipient mice. These results indicate that high local levels of hGH or IGF-I in the bone marrow microenvironment enhanced resorption, which is consistent with previous findings in transgenic mice with targeted bone IGF-I expression showing that high local IGF-I expression increased bone remodeling, favoring a net bone loss. Thus, GH and/or IGF-I would not be an appropriate transgene for use in this Sca-1(+) cell-based gene transfer strategy to promote endosteal bone formation. Published 2011 John Wiley & Sons, Ltd.


Asunto(s)
Antígenos Ly/metabolismo , Resorción Ósea , Técnicas de Transferencia de Gen , Hormona de Crecimiento Humana , Proteínas de la Membrana/metabolismo , Animales , Antígenos Ly/genética , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Resorción Ósea/diagnóstico por imagen , Resorción Ósea/genética , Resorción Ósea/metabolismo , Resorción Ósea/patología , Huesos/anatomía & histología , Huesos/citología , Huesos/patología , Fémur/anatomía & histología , Fémur/citología , Dosificación de Gen , Regulación de la Expresión Génica , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Células HEK293 , Hormona de Crecimiento Humana/genética , Hormona de Crecimiento Humana/metabolismo , Humanos , Lentivirus/genética , Lentivirus/metabolismo , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , ARN Mensajero/metabolismo , Radiografía , Transducción Genética , Irradiación Corporal Total
16.
Mol Imaging Biol ; 13(3): 443-451, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20567925

RESUMEN

PURPOSE: The purpose of this study was to evaluate the long-term cellular toxicity, labeling efficiency, chondrogenic differentiation capacity, and intracellular distribution following direct superparamagnetic iron oxide (SPIO) nanoparticle labeling of human mesenchymal stem cells (hMSCs) in the absence of transfection agents. PROCEDURES: hMSCs were incubated with a SPIO, Ferucarbotran, at concentrations of 0, 1, 10, and 100 µg Fe/ml for 24 or 72 h. The cell granularity and size change, reactive oxygen species generation, and mitochondria membrane potential were measured by flow cytometry. The differentiation capacity of the cells into chondrocytes was determined by Alcian blue and Safranin-O staining, immunocytochemical analysis, and reverse transcription polymerase chain reaction. RESULTS: The intracellular distribution of the internalized particles was visualized via confocal microscopy. No significant difference was found in the toxicity of labeled cells relative to controls. Successful chondrogenesis of Ferucarbotran-labeled hMSCs was confirmed. The intracellular SPIO nanoparticles were located within the lysosomes. CONCLUSIONS: In conclusion, we have demonstrated the feasibility of direct labeling with Ferucarbotran without impairment of cellular function, toxicity, or inhibition of differentiation capacity. Furthermore, lysosomal metabolism takes place after intracellular uptake of Ferucarbotran.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Condrogénesis/efectos de los fármacos , Dextranos/toxicidad , Espacio Intracelular/metabolismo , Nanopartículas de Magnetita/toxicidad , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Coloración y Etiquetado , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Citometría de Flujo , Humanos , Inmunohistoquímica , Espacio Intracelular/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Microscopía Confocal , ARN Mensajero/genética , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factores de Tiempo
17.
BMC Cancer ; 10: 562, 2010 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-20955597

RESUMEN

BACKGROUND: The Ras association domain family 1 (RASSF1) gene is a Ras effector encoding two major mRNA forms, RASSF1A and RASSF1C, derived by alternative promoter selection and alternative mRNA splicing. RASSF1A is a tumor suppressor gene. However, very little is known about the function of RASSF1C both in normal and transformed cells. METHODS: Gene silencing and over-expression techniques were used to modulate RASSF1C expression in human breast cancer cells. Affymetrix-microarray analysis was performed using T47D cells over-expressing RASSF1C to identify RASSF1C target genes. RT-PCR and western blot techniques were used to validate target gene expression. Cell invasion and apoptosis assays were also performed. RESULTS: In this article, we report the effects of altering RASSF1C expression in human breast cancer cells. We found that silencing RASSF1C mRNA in breast cancer cell lines (MDA-MB231 and T47D) caused a small but significant decrease in cell proliferation. Conversely, inducible over-expression of RASSF1C in breast cancer cells (MDA-MB231 and T47D) resulted in a small increase in cell proliferation. We also report on the identification of novel RASSF1C target genes. RASSF1C down-regulates several pro-apoptotic and tumor suppressor genes and up-regulates several growth promoting genes in breast cancer cells. We further show that down-regulation of caspase 3 via overexpression of RASSF1C reduces breast cancer cells' sensitivity to the apoptosis inducing agent, etoposide. Furthermore, we found that RASSF1C over-expression enhances T47D cell invasion/migration in vitro. CONCLUSION: Together, our findings suggest that RASSF1C, unlike RASSF1A, is not a tumor suppressor, but instead may play a role in stimulating metastasis and survival in breast cancer cells.


Asunto(s)
Apoptosis , Neoplasias de la Mama/patología , Regulación Neoplásica de la Expresión Génica , Proteínas Supresoras de Tumor/fisiología , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Movimiento Celular , Silenciador del Gen , Humanos , Invasividad Neoplásica , Metástasis de la Neoplasia , Análisis de Secuencia por Matrices de Oligonucleótidos , Plásmidos/metabolismo , Resultado del Tratamiento , Proteínas Supresoras de Tumor/metabolismo
18.
Nanotechnology ; 21(23): 235103, 2010 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-20479509

RESUMEN

Ultrasmall superparamagnetic iron oxide (USPIO) particles are very useful for cellular magnetic resonance imaging (MRI), which plays a key role in developing successful stem cell therapies. However, their low intracellular labeling efficiency, and biosafety concerns associated with their use, have limited their potential usage. In this study we develop a novel system composed of RBC-derived vesicles (RDVs) for efficient delivery of USPIO particles into human bone marrow mesenchymal stem cells (MSCs) for cellular MRI in vitro and in vivo. RDVs are highly biosafe to their autologous MSCs as manifested by cell viability, differentiation, and gene microarray assays. The data demonstrate the potential of RDVs as intracellular delivery vehicles for biomedical applications.


Asunto(s)
Compuestos Férricos/administración & dosificación , Imagen por Resonancia Magnética/métodos , Células Madre/citología , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Eritrocitos/citología , Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Células Madre/metabolismo
19.
Phys Med Biol ; 55(4): 931-45, 2010 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-20090183

RESUMEN

Biocompatible Au nanoparticles with surfaces modified by PEG (polyethylene glycol) were developed in view of possible applications for the enhancement of radiotherapy. Such nanoparticles exhibit preferential deposition at tumor sites due to the enhanced permeation and retention (EPR) effect. Here, we systematically studied their effects on EMT-6 and CT26 cell survival rates during irradiation for a dose up to 10 Gy with a commercial biological irradiator (E(average) = 73 keV), a Cu-Kalpha(1) x-ray source (8.048 keV), a monochromatized synchrotron source (6.5 keV), a radio-oncology linear accelerator (6 MeV) and a proton source (3 MeV). The percentage of surviving cells after irradiation was found to decrease by approximately 2-45% in the presence of PEG-Au nanoparticles ([Au] = 400, 500 or 1000 microM). The cell survival rates decreased as a function of the dose for all sources and nanoparticle concentrations. These results could open the way to more effective cancer irradiation therapies by using nanoparticles with optimized surface treatment. Difficulties in applying MTT assays were also brought to light, showing that this approach is not suitable for radiobiology.


Asunto(s)
Compuestos de Oro , Nanopartículas del Metal , Polietilenglicoles , Radioterapia/métodos , Animales , Materiales Biocompatibles/química , Línea Celular Tumoral , Supervivencia Celular/efectos de la radiación , Coloides/química , Daño del ADN/efectos de la radiación , Compuestos de Oro/química , Nanopartículas del Metal/química , Ratones , Polietilenglicoles/química , Dosificación Radioterapéutica , Factores de Tiempo
20.
Mol Cell Biol ; 30(3): 711-21, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19995908

RESUMEN

Mutations of ephrin B1 in humans result in craniofrontonasal syndrome. Because little is known of the role and mechanism of action of ephrin B1 in bone, we examined the function of osteoblast-produced ephrin B1 in vivo and identified the molecular mechanism by which ephrin B1 reverse signaling regulates bone formation. Targeted deletion of the ephrin B1 gene in type 1alpha2 collagen-producing cells resulted in severe calvarial defects, decreased bone size, bone mineral density, and trabecular bone volume, caused by impairment in osterix expression and osteoblast differentiation. Coimmunoprecipitation of the TAZ complex with TAZ-specific antibody revealed a protein complex containing ephrin B1, PTPN13, NHERF1, and TAZ in bone marrow stromal (BMS) cells. Activation of ephrin B1 reverse signaling with soluble EphB2-Fc led to a time-dependent increase in TAZ dephosphorylation and shuttling from cytoplasm to nucleus. Treatment of BMS cells with exogenous EphB2-Fc resulted in a 4-fold increase in osterix expression as determined by Western blotting. Disruption of TAZ expression using specific lentivirus small hairpin RNA (shRNA) decreased TAZ mRNA by 80% and ephrin B1 reverse signaling-mediated increases in osterix mRNA by 75%. Knockdown of NHERF1 expression reduced basal levels of osterix expression by 90% and abolished ephrin B1-mediated induction of osterix expression. We conclude that locally produced ephrin B1 mediates its effects on osteoblast differentiation by a novel molecular mechanism in which activation of reverse signaling leads to dephosphorylation of TAZ and subsequent release of TAZ from the ephrin B1/NHERF1/TAZ complex to translocate to the nucleus to induce expression of the osterix gene and perhaps other osteoblast differentiation genes. Our findings provide strong evidence that ephrin B1 reverse signaling in osteoblasts is critical for BMS cell differentiation and bone formation.


Asunto(s)
Células de la Médula Ósea/citología , Diferenciación Celular/genética , Efrina-B1/metabolismo , Osteoblastos/citología , Osteogénesis/genética , Fosfoproteínas/metabolismo , Intercambiadores de Sodio-Hidrógeno/metabolismo , Factores de Transcripción/metabolismo , Aciltransferasas , Animales , Células de la Médula Ósea/metabolismo , Efrina-B1/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteoblastos/metabolismo , Fosfoproteínas/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 13/metabolismo , Cráneo/anomalías , Intercambiadores de Sodio-Hidrógeno/genética , Factor de Transcripción Sp7 , Células del Estroma/citología , Células del Estroma/metabolismo , Factores de Transcripción/genética , Activación Transcripcional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA