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













Base de datos
Intervalo de año de publicación
1.
Ecotoxicol Environ Saf ; 273: 116090, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38364346

RESUMEN

Airway epithelium, the first defense barrier of the respiratory system, facilitates mucociliary clearance against inflammatory stimuli, such as pathogens and particulates inhaled into the airway and lung. Inhaled particulate matter 2.5 (PM2.5) can penetrate the alveolar region of the lung, and it can develop and exacerbate respiratory diseases. Although the pathophysiological effects of PM2.5 in the respiratory system are well known, its impact on mucociliary clearance of airway epithelium has yet to be clearly defined. In this study, we used two different 3D in vitro airway models, namely the EpiAirway-full-thickness (FT) model and a normal human bronchial epithelial cell (NHBE)-based air-liquid interface (ALI) system, to investigate the effect of diesel exhaust particles (DEPs) belonging to PM2.5 on mucociliary clearance. RNA-sequencing (RNA-Seq) analyses of EpiAirway-FT exposed to DEPs indicated that DEP-induced differentially expressed genes (DEGs) are related to ciliary and microtubule function and inflammatory-related pathways. The exposure to DEPs significantly decreased the number of ciliated cells and shortened ciliary length. It reduced the expression of cilium-related genes such as acetylated α-tubulin, ARL13B, DNAH5, and DNAL1 in the NHBEs cultured in the ALI system. Furthermore, DEPs significantly increased the expression of MUC5AC, whereas they decreased the expression of epithelial junction proteins, namely, ZO1, Occludin, and E-cadherin. Impairment of mucociliary clearance by DEPs significantly improved the release of epithelial-derived inflammatory and fibrotic mediators such as IL-1ß, IL-6, IL-8, GM-CSF, MMP-1, VEGF, and S100A9. Taken together, it can be speculated that DEPs can cause ciliary dysfunction, hyperplasia of goblet cells, and the disruption of the epithelial barrier, resulting in the hyperproduction of lung injury mediators. Our data strongly suggest that PM2.5 exposure is directly associated with ciliary and epithelial barrier dysfunction and may exacerbate lung injury.


Asunto(s)
Lesión Pulmonar , Emisiones de Vehículos , Humanos , Emisiones de Vehículos/toxicidad , Lesión Pulmonar/metabolismo , Mucosa Respiratoria , Material Particulado/metabolismo , Células Epiteliales , Epitelio
2.
Antioxidants (Basel) ; 12(6)2023 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-37371919

RESUMEN

Particulate matter 2.5 (PM2.5) induces lung injury by increasing the generation of reactive oxygen species (ROS) and inflammation. ROS aggravates NLRP3 inflammasome activation, which activates caspase-1, IL-1ß, and IL-18 and induces pyroptosis; these factors propagate inflammation. In contrast, treatment with exogenous 8-hydroxydeoxyguanosine (8-OHdG) decreases RAC1 activity and eventually decreases dinucleotide phosphate oxidase (NOX) and ROS generation. To establish modalities that would mitigate PM2.5-induced lung injury, we evaluated whether 8-OHdG decreased PM2.5-induced ROS generation and NLRP3 inflammasome activation in BEAS-2B cells. CCK-8 and lactate dehydrogenase assays were used to determine the treatment concentration. Fluorescence intensity, Western blotting, enzyme-linked immunosorbent assay, and immunoblotting assays were also performed. Treatment with 80 µg/mL PM2.5 increased ROS generation, RAC1 activity, NOX1 expression, NLRP3 inflammasome (NLRP3, ASC, and caspase-1) activity, and IL-1ß and IL-18 levels in cells; treatment with 10 µg/mL 8-OHdG significantly attenuated these effects. Furthermore, similar results, such as reduced expression of NOX1, NLRP3, ASC, and caspase-1, were observed in PM2.5-treated BEAS-2B cells when treated with an RAC1 inhibitor. These results show that 8-OHdG mitigates ROS generation and NLRP3 inflammation by inhibiting RAC1 activity and NOX1 expression in respiratory cells exposed to PM2.5.

3.
Stem Cell Res ; 24: 148-150, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-29034882

RESUMEN

We generated human induced pluripotent stem cells (KSCBi002-B and KSCBi002-B-1) from the dermal fibroblasts of a donor using a modified RNA-based gene delivery method. According to GTG-banding analysis, the generated KSCBi002-B line has a cytogenetic abnormality (46,XY, t(1;4)(q21;q25)) that is distinct from that of the donor, whereas KSCBi002-B-1 has a normal karyotype (46,XY). These cell lines can be useful as a model for characterizing the hiPSCs generated by a non-viral and non-integrative system, or as a chromosomal balanced translocation model. These two cell lines are registered and available from the National Stem Cell Bank, Korea National Institute of Health.


Asunto(s)
Fibroblastos/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , ARN/metabolismo , Línea Celular , Humanos , Masculino
4.
Stem Cell Res ; 21: 13-15, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28677527

RESUMEN

We generated human induced pluripotent stem cells (hiPSCs) from dermal fibroblasts using a Sendai virus (SeV)-based gene delivery method. The generated hiPSC line, KSCBi002-A, has a normal karyotype (46,XY). The pluripotency and differentiation capacity were characterized by comparison with those of a human embryonic stem cell line. This cell line is registered and available from the National Stem Cell Bank, Korea National Institute of Health.


Asunto(s)
Dermis/metabolismo , Fibroblastos/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Virus Sendai , Transducción Genética , Línea Celular , Dermis/citología , Fibroblastos/citología , Humanos , Células Madre Pluripotentes Inducidas/citología , Masculino
5.
Stem Cell Res ; 21: 44-46, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28677537

RESUMEN

Urinary cells can be an ideal source for generating hiPSCs and progenitors, as they are easily accessible, non-invasive, and universally available. We generated human induced pluripotent stem cells (hiPSCs) from the urinary cells of a healthy donor using a Sendai virus-based gene delivery method. The generated hiPSC line, KSCBi001-A, has a normal karyotype (46,XY). The pluripotency and capacity of multilineage differentiation were characterized by comparison with those of a human embryonic stem cell line. This cell line is registered and available from National Stem Cell Bank, Korea National Institute of Health.


Asunto(s)
Células Madre Pluripotentes Inducidas/metabolismo , Virus Sendai , Transducción Genética , Orina/citología , Humanos , Células Madre Pluripotentes Inducidas/citología , Masculino
6.
J Alzheimers Dis ; 43(1): 243-57, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25079792

RESUMEN

The cytoplasmic C-terminal domain of amyloid-ß protein precursor (AßPP) binds to several proteins that regulate the trafficking and processing of AßPP and affects amyloid-ß (Aß) production. We previously reported that levels of AT-motif binding factor 1 (ATBF1) are increased in the brains of 17-month-old Tg2576 mice compared with wild-type controls, and that Aß42 increases ATBF1 expression, inducing death in primary rat cortical neurons. Here, we show that ATBF1 levels are increased in the cytoplasm of hippocampal neurons in Alzheimer's disease (AD) brains compared with non-AD brains. Furthermore, cotransfection of human embryonic kidney (HEK293T) and human neuroblastoma (SH-SY5Y) cells with ATBF1 and AßPP695 increased steady-state levels of AßPP via the binding of ATBF1 to the AßPP cytoplasmic domain (amino acids 666-690), resulting in increased Aß production and cellular and soluble AßPP (sAßPP) levels without affecting the activity or levels of AßPP processing enzymes (α-, ß-, or γ-secretase). Conversely, knockdown of endogenous ATBF1 reduced levels of cellular AßPP, sAßPP, and Aß in HEK293 cells overexpressing human AßPP695. Our findings provide insight into the dynamics of AßPP processing and Aß production, and suggest that ATBF1 is a novel AßPP binding protein that may be a suitable therapeutic target for AD.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Proteínas de Homeodominio/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Línea Celular Tumoral , Citoplasma/metabolismo , Femenino , Expresión Génica/fisiología , Técnicas de Silenciamiento del Gen , Células HEK293 , Hipocampo/metabolismo , Proteínas de Homeodominio/genética , Humanos , Masculino , Neuronas/metabolismo , Fragmentos de Péptidos/metabolismo
7.
J Alzheimers Dis ; 43(4): 1215-28, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25147119

RESUMEN

Amyloid-ß (Aß) peptide plays a major role in the pathogenesis of Alzheimer's disease (AD), and is generated by ß- and γ-secretase-mediated proteolytic processing of amyloid-ß protein precursor (AßPP). In the present study, we investigated the effect of 118 natural compounds on Aß production in the medium of HEK293 cells stably expressing human AßPP695 (HEK293-AßPP) using Aß42 sandwich ELISA to find natural compounds that can modulate Aß production. We found that a coumarin derivative of citrus fruits, auraptene, increased Aß production. Treatment of HEK293-AßPP cells and rat primary cortical neurons with auraptene significantly increased the secretion of Aß40, Aß42, and the Aß42/40 ratio. However, auraptene did not change the protein levels of the AßPP processing enzymes, a disintegrin and metalloproteinases 10 (ADAM10, α-secretase), ß-site AßPP cleaving enzyme-1 (BACE-1, ß-secretase), and presenilin 1 (PS1, γ-secretase component). Auraptene increased the activity of γ-secretase but not that of α- and ß-secretase. Furthermore, auraptene enhanced γ-secretase-mediated production of Aß from AßPP or AßPP-C99, but not through α- and ß-secretase. Auraptene also phosphorylated c-Jun N-terminal kinase (JNK), and pretreatment with the JNK inhibitor, SP600125, reduced auraptene-induced γ-secretase activity. Overall, our results suggest that auraptene-mediated activation of JNK may contribute to the production of Aß by promoting γ-secretase activity.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Fármacos del Sistema Nervioso Central/farmacología , Cumarinas/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Fragmentos de Péptidos/metabolismo , Proteínas ADAM/metabolismo , Proteína ADAM10 , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Antracenos/farmacología , Ácido Aspártico Endopeptidasas/metabolismo , Supervivencia Celular/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Ensayo de Inmunoadsorción Enzimática , Células HEK293 , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas de la Membrana/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fosforilación/efectos de los fármacos , Presenilina-1/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Ratas Sprague-Dawley
8.
Mol Neurodegener ; 6: 47, 2011 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-21729327

RESUMEN

BACKGROUND: Recently, several lines of evidence have shown the aberrant expression of cell-cycle-related proteins and tumor suppressor proteins in vulnerable neurons of the Alzheimer's disease (AD) brain and transgenic mouse models of AD; these proteins are associated with various paradigms of neuronal death. It has been reported that ATBF1 induces cell cycle arrest associated with neuronal differentiation in the developing rat brain, and that gene is one of the candidate tumor suppressor genes for prostate and breast cancers in whose cells overexpressed ATBF1 induces cell cycle arrest. However, the involvement of ATBF1 in AD pathogenesis is as yet unknown. RESULTS: We found that ATBF1 was up-regulated in the brains of 17-month-old Tg2576 mice compared with those of age-matched wild-type mice. Moreover, our in vitro studies showed that Aß1-42 and DNA-damaging drugs, namely, etoposide and homocysteine, increased the expression ATBF1 level in primary rat cortical neurons, whereas the knockdown of ATBF1 in these neurons protected against neuronal death induced by Aß1-42, etoposide, and homocysteine, indicating that ATBF1 mediates neuronal death in response to these substances. In addition, we found that ATBF1-mediated neuronal death is dependent on ataxia-telangiectasia mutated (ATM) because the blockage of ATM activity by treatment with ATM inhibitors, caffeine and KU55933, abolished ATBF1 function in neuronal death. Furthermore, Aß1-42 phosphorylates ATM, and ATBF1 interacts with phosphorylated ATM. CONCLUSIONS: To the best of our knowledge, this is the first report that Aß1-42 and DNA-damaging drugs increased the ATBF1 expression level in primary rat cortical neurons; this increase, in turn, may activate ATM signaling responsible for neuronal death through the binding of ATBF1 to phosphorylated ATM. ATBF1 may therefore be a suitable target for therapeutic intervention of AD.

9.
J Cell Biochem ; 112(5): 1259-67, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21465524

RESUMEN

Insulin resistance is the primary cause responsible for type 2 diabetes. Phosphatase and tensin homolog (PTEN) plays a negative role in insulin signaling and its inhibition improves insulin sensitivity. Metformin is a widely used insulin-sensitizing drug; however, the mechanism by which metformin acts is poorly understood. To gain insight into the role of PTEN, we examined the effect of metformin on PTEN expression. Metformin suppressed the expression of PTEN in an AMP-activated protein kinase (AMPK)-dependent manner in preadipocyte 3T3-L1 cells. Knock-down of PTEN potentiated the increase in insulin-mediated phosphorylation of Akt/ERK. Metformin also increased the phosphorylation of c-Jun N-terminal kinase (JNK)-c-Jun and mammalian target of rapamycin (mTOR)-p70S6 kinase pathways. Both pharmacologic inhibition and knock-down of AMPK blocked metformin-induced phosphorylation of JNK and mTOR. Knock-down of AMPK recovered the metformin-induced PTEN down-regulation, suggesting the involvement of AMPK in PTEN regulation. PTEN promoter activity was suppressed by metformin and inhibition of mTOR and JNK by pharmacologic inhibitors blocked metformin-induced PTEN promoter activity suppression. These findings provide evidence for a novel role of AMPK on PTEN expression and thus suggest a possible mechanism by which metformin may contribute to its beneficial effects on insulin signaling.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Hipoglucemiantes/farmacología , Resistencia a la Insulina , Insulina/metabolismo , Metformina/farmacología , Fosfohidrolasa PTEN/metabolismo , Células 3T3-L1 , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/genética , Adipocitos/metabolismo , Animales , Diabetes Mellitus Tipo 2/genética , Regulación hacia Abajo/efectos de los fármacos , Glucosa/metabolismo , Ratones , Fosforilación/efectos de los fármacos , Transducción de Señal/genética , Serina-Treonina Quinasas TOR
10.
J Biol Chem ; 286(9): 7567-76, 2011 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-21193401

RESUMEN

Homocysteine sulfinic acid (HCSA) is a homologue of the amino acid cysteine and a selective metabotropic glutamate receptor (mGluR) agonist. However, the metabolic role of HCSA is poorly understood. In this study, we showed that HCSA and glutamate stimulated glucose uptake in C2C12 mouse myoblast cells and increased AMP-activated protein kinase (AMPK) phosphorylation. RT-PCR and Western blot analysis revealed that C2C12 expresses mGluR5. HCSA transiently increased the intracellular calcium concentration. Although α-methyl-4-carboxyphenylglycine, a metabotropic glutamate receptor antagonist, blocked the action of HCSA in intracellular calcium response and AMPK phosphorylation, 6-cyano-7-nitroquinoxaline-2,3-dione, an AMPA antagonist, did not exhibit such effects. Knockdown of mGluR5 with siRNA blocked HCSA-induced AMPK phosphorylation. Pretreatment of cells with STO-609, a calmodulin-dependent protein kinase kinase (CaMKK) inhibitor, blocked HCSA-induced AMPK phosphorylation, and knockdown of CaMKK blocked HCSA-induced AMPK phosphorylation. In addition, HCSA activated p38 mitogen-activated protein kinase (MAPK). Expression of dominant-negative AMPK suppressed HCSA-mediated phosphorylation of p38 MAPK, and inhibition of AMPK and p38 MAPK blocked HCSA-induced glucose uptake. Phosphorylation of protein kinase C ζ (PKCζ) was also increased by HCSA. Pharmacologic inhibition or knockdown of p38 MAPK blocked HCSA-induced PKCζ phosphorylation, and knockdown of PKCζ suppressed the HCSA-induced increase of cell surface GLUT4. The stimulatory effect of HCSA on cell surface GLUT4 was impaired in FITC-conjugated PKCζ siRNA-transfected cells. Together, the above results suggest that HCSA may have a beneficial role in glucose metabolism in skeletal muscle cells via stimulation of AMPK.


Asunto(s)
Glucosa/farmacocinética , Homocisteína/análogos & derivados , Sistema de Señalización de MAP Quinasas/fisiología , Fibras Musculares Esqueléticas/enzimología , Proteína Quinasa C/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Calcio/metabolismo , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/genética , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Células Cultivadas , Transportador de Glucosa de Tipo 4/metabolismo , Homocisteína/metabolismo , Homocisteína/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/efectos de los fármacos , Músculo Esquelético/citología , Músculo Esquelético/enzimología , Fosforilación/fisiología , Proteína Quinasa C/genética , Receptor del Glutamato Metabotropico 5 , Receptores de Glutamato Metabotrópico/genética , Receptores de Glutamato Metabotrópico/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/genética
11.
J Cell Physiol ; 226(4): 974-81, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20857458

RESUMEN

Metformin is a major oral anti-diabetic drug and is known as an insulin sensitizer. However, the mechanism by which metformin acts is unclear. In this study, we found that AICAR, an AMPK activator, and metformin increased the expression of Rab4 mRNA and protein levels in skeletal muscle C2C12 cells. The promoter activity of Rab4 was increased by metformin in an AMPK-dependent manner. Metformin stimulated the phosphorylation of AS160, Akt substrate, and Rab GTPase activating protein (GAP), and also increased the phosphorylation of PKC-zeta, which is a critical molecule for glucose uptake. Knockdown of AMPK blocked the metformin-induced phosphorylation of AS160/PKC-zeta. In addition, a colorimetric absorbance assay showed that insulin-induced translocation of GLUT4 was suppressed in Rab4 knockdown cells. Moreover, Rab4 interacted with PKC-zeta but not with GLUT4. The C-terminal-deleted Rab4 mutant, Rab4ΔCT, showed diffuse sub-cellular localization, while wild-type Rab4 localized exclusively to the perinuclear membrane. Unlike Rab4ΔCT, wild-type Rab4 co-localized with PKC-zeta. Together, these results demonstrate that metformin induces Rab4 expression via AMPK-AS160-PKC-zeta and modulates insulin-mediated GLUT4 translocation.


Asunto(s)
Adenilato Quinasa/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Metformina/farmacología , Células Musculares/efectos de los fármacos , Células Musculares/enzimología , Músculo Esquelético/citología , Proteínas de Unión al GTP rab4/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Línea Celular , Regulación de la Expresión Génica/efectos de los fármacos , Células HeLa , Humanos , Insulina/farmacología , Ratones , Proteínas Mutantes/metabolismo , Fosforilación/efectos de los fármacos , Fosfoserina/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , Proteína Quinasa C/metabolismo , Estructura Terciaria de Proteína , Transporte de Proteínas/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ribonucleótidos/farmacología , Transducción de Señal/efectos de los fármacos , Proteínas de Unión al GTP rab4/química , Proteínas de Unión al GTP rab4/genética
12.
Biol Pharm Bull ; 33(7): 1105-11, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20606297

RESUMEN

ABCA1, a member of the ATP-binding cassette transporter family, regulates high-density lipoprotein (HDL) metabolism and reverses cholesterol transport. Its expression is upregulated mainly by the activation of the liver X receptor (LXR), retinoid X receptor (RXR), and peroxisome proliferator-activated receptors (PPARs). To identify natural compounds that can upregulate ABCA1 expression, we developed a reporter assay using U251-MG (human glioma cell line) cells that stably express a human ABCA1 promoter-luciferase and performed a cell-based high-throughput screening of 118 natural compounds. Using this system, we identified honokiol, a compound extracted from Magnolia officinalis, as an activator of the ABCA1 promoter. We found that honokiol also increased ABCA1 mRNA and protein expression levels in a dose-dependent manner in U251-MG cells without significant cell death and also increased ABCA1, ABCG1 and apolipoprotein E (apoE) expression levels in THP-1 macrophages. PPAR antagonists did not diminish the induction of ABCA1 expression by honokiol in U251-MG cells. Cotreatment of the cells with honokiol and T0901317 (synthetic LXR ligand) further increased the ABCA1 expression level, whereas cotreatment with 9-cis retinoic acid had no additive effect compared with treatment with honokiol alone. We also found that honokiol has binding affinity to RXRbeta. In this study, we identified for the first time honokiol as an upregulator of ABCA1 expression, which is mediated by the binding of honokiol to RXRbeta as a ligand.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Compuestos de Bifenilo/farmacología , Lignanos/farmacología , Receptor beta X Retinoide/agonistas , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/genética , Secuencia de Bases , Western Blotting , Línea Celular Tumoral , Cartilla de ADN , Humanos , Etiquetado Corte-Fin in Situ , Ligandos , Regiones Promotoras Genéticas
13.
J Cancer Res Clin Oncol ; 136(2): 187-94, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19655168

RESUMEN

PURPOSE: Although the altered expression of integrin alpha4 is known to be associated with transformation or metastasis in several human cancers, the information on cholangiocarcinoma (CC) is still poor. In this study, we investigated the promoter methylation status of integrin alpha4 gene in CC. METHODS: A total of 29 CC, 19 adjacent non-tumor-containing tissue and 15 normal liver specimens were used for identification of gene methylation status by methylation-specific polymerase chain reaction. RESULTS: The frequency of DNA methylation was 55.17% (16 of 29) in the CC specimens (P < 0.001). Also, transcripts of the integrin alpha4 gene were decreased in all CC tissues in which there was DNA methylation of the integrin alpha4 gene. In addition, the downregulated expression of integrin alpha4 in CC cells with hypermethylation of the integrin alpha4 gene was restored by treatment with 5-aza-2'-deoxycytidine, a DNA methyltransferase inhibitor. Moreover, we found that DNA methylation of integrin alpha4 was detected in all CC tissues obtained from patients with LN metastasis (7/7). Furthermore, phosphorylation of paxillin, cell migration-related molecule, was regulated by silencing of integrin alpha4. CONCLUSIONS: Taken together, these results suggest that loss of the integrin alpha4 gene is caused by aberrant DNA methylation of the 5'-CpG island site of the gene, and methylation of the integrin alpha4 gene can be a useful marker of metastasis of CC.


Asunto(s)
Neoplasias de los Conductos Biliares/patología , Conductos Biliares Intrahepáticos , Biomarcadores de Tumor/genética , Colangiocarcinoma/secundario , Metilación de ADN , Integrina alfa4/genética , Adulto , Anciano , Antimetabolitos Antineoplásicos/farmacología , Azacitidina/farmacología , Secuencia de Bases , Neoplasias de los Conductos Biliares/genética , Línea Celular Tumoral , Colangiocarcinoma/genética , Islas de CpG/genética , Metilación de ADN/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Silenciador del Gen , Humanos , Integrina alfa4/efectos de los fármacos , Integrina beta1/genética , Metástasis Linfática , Masculino , Metiltransferasas/antagonistas & inhibidores , Persona de Mediana Edad , Datos de Secuencia Molecular , Invasividad Neoplásica , Paxillin/metabolismo , Fosforilación , ARN Interferente Pequeño , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN , Transfección
14.
J Korean Med Sci ; 24(3): 493-7, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19543515

RESUMEN

To gain insight into the differential mechanism of gene promoter hypermethylation in acute and chronic leukemia, we identified the methylation status on one part of 5'CpG rich region of 8 genes, DAB2IP, DLC-1, H-cadherin, ID4, Integrin alpha4, RUNX3, SFRP1, and SHP1 in bone marrows from acute myeloid leukemia (AML) and chronic myeloid leukemia (CML) patients. Also, we compared the methylation status of genes in AML and CML using methylation-specific PCR (MSP). The frequencies of DNA methylation of ID4, SFRP1, and SHP1 were higher in AML patients compared to those in CML patients. In contrast, no statistical difference between AML and CML was detected for other genes such as DLC-1, DAB2IP, H-cadherin, Integrin alpha4, and RUNX3. Taken together, these results suggest that these methylation-controlled genes may have different roles in AML and CML, and thus, may act as a biological marker of AML.


Asunto(s)
Metilación de ADN , Proteínas Inhibidoras de la Diferenciación/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Leucemia Mieloide Aguda/genética , Proteínas de la Membrana/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Adolescente , Adulto , Anciano , Islas de CpG , Femenino , Humanos , Proteínas Inhibidoras de la Diferenciación/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Leucemia Mieloide Aguda/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Persona de Mediana Edad , Regiones Promotoras Genéticas , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo
15.
Oncol Res ; 17(4): 151-7, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18773859

RESUMEN

Aberrant DNA methylation of 5'-CpG islands located within gene promoters has been identified as a mechanism for transcriptional inactivation of tumor suppressor genes. To ascertain the mechanism of gene promoter hypermethylation in cholangiocarcinoma (CC), we investigated promoter methylation status of the candidate genes ID4, DLC-1, and SFRP1 in 41 CCs, 19 adjacent nontumor tissues, and 15 normal liver tissues using methylation-specific PCR (MSP). The frequencies of DNA methylation were: 57.5% (23 of 40) for ID4, 14.3% (5 of 35) for DLC-1, and 63.4% (26 of 41) for SFRP1, respectively. In contrast, a low frequency of methylation was detected in nontumor tissues. In addition, hypermethylated status of these genes was detected in three kinds of CC cell lines. Moreover, the downregulated expression of these genes in these cells was restored by treatment with 5-aza-2'-deoxycytidine, a DNA methyltransferase inhibitor. Taken together, these results suggest that aberrant DNA methylation may contribute to the tumorigenesis of cholangiocarcinoma.


Asunto(s)
Neoplasias de los Conductos Biliares/genética , Conductos Biliares Intrahepáticos , Colangiocarcinoma/genética , Islas de CpG , Metilación de ADN , Genes Supresores de Tumor , Regiones Promotoras Genéticas , Adulto , Anciano , Neoplasias de los Conductos Biliares/patología , Línea Celular Tumoral , Colangiocarcinoma/patología , Epigénesis Genética , Humanos , Persona de Mediana Edad
16.
Regul Pept ; 151(1-3): 71-4, 2008 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-18555543

RESUMEN

Oxytocin is a mammalian hormone that is released mainly after distension of the uterine cervix. In this study, we report that oxytocin stimulates intracellular release of calcium, and also activates AMPK (AMP-activated protein kinase) in C2C12 myoblast cells in a time/dose-dependent manner. Oxytocin receptor mRNA was detected in C2C12 cells. In addition, oxytocin stimulated glucose uptake and, moreover, inhibition of either CaMKK (Ca(2+)/calmodulin-dependent protein kinase kinase) or AMPK blocked oxytocin-mediated AMPK activation and glucose uptake. Taken together, our findings suggest that oxytocin may serve a peripheral metabolic function in skeletal muscle cells through the calcium-CaMKK-AMPK pathway.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Calcio/metabolismo , Glucosa/metabolismo , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Oxitocina/farmacología , Animales , Secuencia de Bases , Transporte Biológico Activo/efectos de los fármacos , Línea Celular , Cartilla de ADN/genética , Activación Enzimática/efectos de los fármacos , Femenino , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Oxitocina/genética
17.
Biochem Biophys Res Commun ; 370(4): 641-5, 2008 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-18406347

RESUMEN

AM251, a cannabinoid antagonist, has various biological activities. In this study, we found that AM251 suppressed the viability of hepatoma HepG2 cells and also increased phosphorylation of JNK (c-jun N-terminal kinase) and ATF3 (activating transcription factor 3). In addition, AM251 phosphorylated AMPK (AMP-activated protein kinase) in a time and dose-dependent manner. Inhibition of AMPK blocked AM251-induced JNK/ATF3 phosphorylation. Expression of AMPK or treatment with AICAR (5-aminoimidazole-4-carboxy-amide-1-d-ribofuranoside), an AMPK activator, activated the JNK/ATF3 pathways. Together, these results suggest that AM251 may have anti-tumor effects in hepatoma through activation of the AMPK-JNK-ATF3 signal pathway.


Asunto(s)
Factor de Transcripción Activador 3/metabolismo , Antineoplásicos/farmacología , Supervivencia Celular/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Complejos Multienzimáticos/metabolismo , Piperidinas/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Pirazoles/farmacología , Proteínas Quinasas Activadas por AMP , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Cannabinoides/antagonistas & inhibidores , Línea Celular Tumoral , Humanos , Fosforilación/efectos de los fármacos , Ribonucleótidos/farmacología , Transducción de Señal/efectos de los fármacos
18.
Biochem Biophys Res Commun ; 361(4): 854-8, 2007 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-17689496

RESUMEN

Caffeic acid phenethyl ester (CAPE), a flavonoid-like compound, is one of the major components of honeybee propolis. In the present study, we investigated the metabolic effects of CAPE in skeletal muscle cells and found that CAPE stimulated glucose uptake in differentiated L6 rat myoblast cells and also activated AMPK (AMP-activated protein kinase). In addition, the inhibition of AMPK blocked CAPE-induced glucose uptake, and CAPE activated the Akt pathway in a PI3K (phosphoinositide 3-kinase)-dependent manner. Furthermore, CAPE enhanced both insulin-mediated Akt activation and glucose uptake. In summary, our results suggest that CAPE may have beneficial roles in glucose metabolism via stimulation of the AMPK pathway.


Asunto(s)
Ácidos Cafeicos/farmacología , Glucosa/metabolismo , Complejos Multienzimáticos/metabolismo , Fibras Musculares Esqueléticas/enzimología , Alcohol Feniletílico/análogos & derivados , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Quinasas Activadas por AMP , Animales , Transporte Biológico/efectos de los fármacos , Diferenciación Celular , Línea Celular , Activación Enzimática , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Mioblastos/citología , Alcohol Feniletílico/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas
19.
Cancer Genet Cytogenet ; 157(1): 37-41, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15676145

RESUMEN

Intrahepatic cholangiocarcinoma (ICC) arises from epithelial cells in the intrahepatic bile duct. Until now, only few reports have been available concerning the genetic changes during the progression of ICC. In this study, we analyzed chromosomal aberrations in 19 frozen ICC samples using comparative genomic hybridization. The common chromosomal gains were observed in 8q22 approximately qter (11 cases, 58%), 5p14 approximately pter (32%), 2q33 approximately qter (26%), 7p (26%), 17q21 approximately q22 (26%), 18q12 approximately q21 (26%), and 19q13.1 (26%). DNA amplification was identified in nine tumors (47%). Chromosomal loss was found in Y (60%), 1p34 approximately pter (37%), 4q(32%), 18q21 approximately qter (32%) 19p (32%), X (32%), 5q11 approximately q14 (26%), 8p(26%), 9p (26%), and 17p (26%). Chromosomal aberrations identified in this study provide candidate regions involved in the tumorigenesis and progression of ICC.


Asunto(s)
Neoplasias de los Conductos Biliares/genética , Conductos Biliares Intrahepáticos , Colangiocarcinoma/genética , Aberraciones Cromosómicas , Hibridación de Ácido Nucleico/métodos , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad
20.
J Korean Med Sci ; 19(5): 682-7, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15483344

RESUMEN

Intrahepatic cholangiocarcinoma (ICC), a malignant neoplasm of the biliary epithelium,is usually fatal because of difficulty in early diagnosis and lack of availability of effective therapy. The genetic mechanisms involved in the development of ICC are not well understood and only a few cytogenetic studies of ICC have been published. Recently, technique of degenerate oligonucleotide primed (DOP)-PCR comparative genomic hybridization (CGH) permits genetic imbalances screening of the entire genome using only small amounts of tumor DNA. In this study chromosomal aberrations in 33 Korean ICC were investigated by DOP-PCR CGH. The common sites of copy number increases were 20q (67%), 17 (61%), 11q11-q13 (42%), 8p12- qter (39%), 18p (39%), 15q22-qter (36%), 16p (36%), 6p21 (30%), 3q25-qter (27%), 1q41-qter (24%), and 5p14-q11.2 (24%). DNA amplification was identified in 16 carcinomas (48%). The frequent sites of amplification were 20q, 17p, 17q23-qter, and 7p. The most frequent sites of copy number decreases were 1p32-pter (21%) and 4q (21%). The recurrent chromosomal aberrations identified in this study provide candidate regions involved in the tumorigenesis and progression of ICC.


Asunto(s)
Neoplasias de los Conductos Biliares/genética , Conductos Biliares Intrahepáticos , Colangiocarcinoma/genética , Aberraciones Cromosómicas , Reacción en Cadena de la Polimerasa/métodos , Adulto , Anciano , Cartilla de ADN , Femenino , Dosificación de Gen , Humanos , Masculino , Persona de Mediana Edad , Hibridación de Ácido Nucleico/métodos , Oligonucleótidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA