Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cardiovasc Drugs Ther ; 35(6): 1291-1304, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33687595

RESUMEN

PURPOSE: Perivascular adipose tissue (PVAT) exerts an anti-contractile effect which is vital in regulating vascular tone. This effect is mediated via sympathetic nervous stimulation of PVAT by a mechanism which involves noradrenaline uptake through organic cation transporter 3 (OCT3) and ß3-adrenoceptor-mediated adiponectin release. In obesity, autonomic dysfunction occurs, which may result in a loss of PVAT function and subsequent vascular disease. Accordingly, we have investigated abnormalities in obese PVAT, and the potential for exercise in restoring function. METHODS: Vascular contractility to electrical field stimulation (EFS) was assessed ex vivo in the presence of pharmacological tools in ±PVAT vessels from obese and exercised obese mice. Immunohistochemistry was used to detect changes in expression of ß3-adrenoceptors, OCT3 and tumour necrosis factor-α (TNFα) in PVAT. RESULTS: High fat feeding induced hypertension, hyperglycaemia, and hyperinsulinaemia, which was reversed using exercise, independent of weight loss. Obesity induced a loss of the PVAT anti-contractile effect, which could not be restored via ß3-adrenoceptor activation. Moreover, adiponectin no longer exerts vasodilation. Additionally, exercise reversed PVAT dysfunction in obesity by reducing inflammation of PVAT and increasing ß3-adrenoceptor and OCT3 expression, which were downregulated in obesity. Furthermore, the vasodilator effects of adiponectin were restored. CONCLUSION: Loss of neutrally mediated PVAT anti-contractile function in obesity will contribute to the development of hypertension and type II diabetes. Exercise training will restore function and treat the vascular complications of obesity.


Asunto(s)
Tejido Adiposo/efectos de los fármacos , Contracción Muscular/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Obesidad/fisiopatología , Obesidad/terapia , Condicionamiento Físico Animal/fisiología , Animales , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Hiperglucemia/inducido químicamente , Hiperinsulinismo/inducido químicamente , Hipertensión/inducido químicamente , Masculino , Ratones , Ratones Endogámicos C57BL , Factor 3 de Transcripción de Unión a Octámeros/efectos de los fármacos , Receptores Adrenérgicos beta 3/efectos de los fármacos , Factor de Necrosis Tumoral alfa/efectos de los fármacos
2.
Can Respir J ; 2020: 2479369, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32849930

RESUMEN

Objective: To investigate the effects of Apatinib on the "stemness" of lung cancer cells in vivo and to explore its related mechanisms. Methods: A xenograft model of lung cancer cells A549 was established in nude mice and randomized into a control group (n = 4) and an Apatinib group (n = 4). Tumor tissues were harvested after 2 weeks, and mRNA was extracted to detect changes in stemness-related genes (CD133, EPCAM, CD13, CD90, ALDH1, CD44, CD45, SOX2, NANOG, and OCT4) and Wnt/ß-catenin, Hedgehog, and Hippo signal pathways. Results: Compared with the control group, the volume and weight of nude mice treated with Apatinib were different and had statistical significance. Apatinib inhibited the expressions of ABCG2, CD24, ICAM-1, OCT4, and SOX2 and upregulated the expressions of CD44, CD13, and FOXD3. Apatinib treatment also inhibited the Wnt/ß-catenin, Hedgehog, and Hippo signaling pathways. Conclusion: Apatinib suppressed the growth of non-small-cell lung cancer cells by repressing the stemness of lung cancer through the inhibition of the Hedgehog, Hippo, and Wnt signaling pathways.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/genética , Proliferación Celular/efectos de los fármacos , Neoplasias Pulmonares/genética , Células Madre Neoplásicas/efectos de los fármacos , Piridinas/farmacología , Células A549 , Animales , Antígenos CD13/efectos de los fármacos , Antígenos CD13/genética , Antígenos CD13/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Proteínas Hedgehog/efectos de los fármacos , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Vía de Señalización Hippo , Humanos , Receptores de Hialuranos/efectos de los fármacos , Receptores de Hialuranos/genética , Receptores de Hialuranos/metabolismo , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Desnudos , Células Madre Neoplásicas/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/efectos de los fármacos , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Factores de Transcripción SOXB1/efectos de los fármacos , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Transducción de Señal , Vía de Señalización Wnt/efectos de los fármacos , Vía de Señalización Wnt/genética , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Cell Reprogram ; 22(2): 82-89, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32125888

RESUMEN

Mesenchymal stem/stromal cells (MSCs) have been identified in almost all adult human tissues and been used in numerous clinical trials for a variety of diseases. Studies have shown that MSCs would undergo cellular senescence when cultured over a long term, which is brought on by increased epigenetic modifications, including DNA methylation. However, the mechanism of MSCs senescence is not well studied. In this study, the effects of RG108, a DNA methyltransferase inhibitor (DNMTi), on senescence, apoptosis, and pluripotency gene expressions in porcine bone marrow (pBM)-MSCs were investigated. First, we determined the optimized dose and time of RG108 treatment in pBM-MSCs to be 10 µM for 48 hours, respectively. Under these conditions, the pluripotency genes (NANOG, POU5F1), the anti-senescence genes (TERT, bFGF), and the anti-apoptosis gene (BCL2) were increased, whereas the apoptotic gene (BAX) was decreased. RG108 protected against apoptosis when pBM-MSC induces apoptosis with H2O2 for 1.5 hours. We also found that RG108 significantly induced the expression of NANOG and POU5F1 by decreasing DNA methylation in gene promoter regions. These results indicate that an optimized dose of RG108 may promote the pluripotency-related character of pBM-MSCs through improving cellular anti-senescence, anti-apoptosis, and pluripotency, which provide a better cell origin for stem cell therapy.


Asunto(s)
Metilasas de Modificación del ADN/antagonistas & inhibidores , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Proteína Homeótica Nanog/efectos de los fármacos , Factor 3 de Transcripción de Unión a Octámeros/efectos de los fármacos , Ftalimidas/farmacología , Triptófano/análogos & derivados , Animales , Apoptosis/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Senescencia Celular/efectos de los fármacos , Metilación de ADN , Epigénesis Genética , Regulación de la Expresión Génica/efectos de los fármacos , Proteína Homeótica Nanog/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Porcinos , Triptófano/farmacología
4.
Neurochem Int ; 123: 46-49, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30055194

RESUMEN

Transporter-mediated uptake determines the peak concentration, duration, and physical spread of released monoamines. Most studies of monoamine clearance focus on the presynaptic uptake1 transporters SERT, NET and DAT. However, recent studies have demonstrated the expression of the uptake2 transporter OCT3 (organic cation transporter 3), throughout the rodent brain. In contrast to NET, DAT and SERT, OCT3 has higher capacity and lower affinity for substrates, is sodium-independent, and is multi-specific, with the capacity to transport norepinephrine, dopamine, serotonin and histamine. OCT3 is insensitive to inhibition by cocaine and antidepressant drugs but is inhibited directly by the glucocorticoid hormone corticosterone. Thus, OCT3 represents a novel, stress hormone-sensitive, monoamine transport mechanism. Incorporating this transporter into current models of monoaminergic neurotransmission requires information on: A) the cellular and subcellular localization of the transporter; B) the effects of OCT3 inhibitors on monoamine clearance; and C) the consequences of decreased OCT3-mediated transport on physiology and/or behavior. This review summarizes studies describing the anatomical distribution of OCT3, its cellular and subcellular localization, its contribution to the regulation of dopaminergic signaling, and its roles in the regulation of behavior. Together, these and other studies suggest that both Uptake1 and Uptake2 transporters play key roles in regulating monoaminergic neurotransmission and the effects of monoamines on behavior.


Asunto(s)
Transporte Biológico/efectos de los fármacos , Encéfalo/efectos de los fármacos , Corticosterona/farmacología , Factor 3 de Transcripción de Unión a Octámeros/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Animales , Encéfalo/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteínas de Transporte de Catión Orgánico/metabolismo
5.
Mol Cell ; 71(6): 1064-1078.e5, 2018 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-30197300

RESUMEN

ß-hydroxybutyrate (ß-HB) elevation during fasting or caloric restriction is believed to induce anti-aging effects and alleviate aging-related neurodegeneration. However, whether ß-HB alters the senescence pathway in vascular cells remains unknown. Here we report that ß-HB promotes vascular cell quiescence, which significantly inhibits both stress-induced premature senescence and replicative senescence through p53-independent mechanisms. Further, we identify heterogeneous nuclear ribonucleoprotein A1 (hnRNP A1) as a direct binding target of ß-HB. ß-HB binding to hnRNP A1 markedly enhances hnRNP A1 binding with Octamer-binding transcriptional factor (Oct) 4 mRNA, which stabilizes Oct4 mRNA and Oct4 expression. Oct4 increases Lamin B1, a key factor against DNA damage-induced senescence. Finally, fasting and intraperitoneal injection of ß-HB upregulate Oct4 and Lamin B1 in both vascular smooth muscle and endothelial cells in mice in vivo. We conclude that ß-HB exerts anti-aging effects in vascular cells by upregulating an hnRNP A1-induced Oct4-mediated Lamin B1 pathway.


Asunto(s)
Ácido 3-Hidroxibutírico/farmacología , Senescencia Celular/efectos de los fármacos , Animales , Células Cultivadas , Regulación de la Expresión Génica , Ribonucleoproteína Nuclear Heterogénea A1/efectos de los fármacos , Ribonucleoproteína Nuclear Heterogénea A1/metabolismo , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Factor 3 de Transcripción de Unión a Octámeros/efectos de los fármacos , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , ARN Mensajero , Activación Transcripcional , Regulación hacia Arriba
6.
Arch Oral Biol ; 74: 69-74, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27886571

RESUMEN

OBJECTIVE: Cancer stem cells contribute to tumor recurrence, and a hypoxic environment is critical for maintaining cancer stem cells. Apigenin is a natural product with anticancer activity. However, the effect of apigenin on cancer stem cells remains unclear. Our aim was to investigate the effect of apigenin on cancer stem cell marker expression in head and neck squamous cell carcinoma cells under hypoxia. DESIGN: We used three head and neck squamous cell carcinoma cell lines; HN-8, HN-30, and HSC-3. The mRNA expression of cancer stem cell markers was determined by semiquantitative RT-PCR and Real-time PCR. The cytotoxic effect of apigenin was determined by MTT colorimetric assay. Flow cytometry was used to reveal the number of cells expressing cancer stem cell surface markers. RESULTS: HN-30 cells, a cancer cell line from the pharynx, showed the greatest response to hypoxia by increasing their expression of CD44, CD105, NANOG, OCT-4, REX-1, and VEGF. Apigenin significantly decreased HN-30 cell viability in dose- and time-dependent manners. In addition, 40µM apigenin significantly down-regulated the mRNA expression of CD44, NANOG, and CD105. Consistent with these results, the hypoxia-induced increase in CD44+ cells, CD105+ cells, and STRO-1+ cells was significantly abolished by apigenin. CONCLUSION: Apigenin suppresses cancer stem cell marker expression and the number of cells expressing cell surface markers under hypoxia.


Asunto(s)
Apigenina/farmacología , Biomarcadores/metabolismo , Carcinoma de Células Escamosas/metabolismo , Hipoxia de la Célula/genética , Neoplasias de Cabeza y Cuello/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Carcinoma de Células Escamosas/genética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Regulación hacia Abajo , Endoglina/efectos de los fármacos , Endoglina/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias de Cabeza y Cuello/genética , Humanos , Receptores de Hialuranos/efectos de los fármacos , Receptores de Hialuranos/metabolismo , Factores de Transcripción de Tipo Kruppel/efectos de los fármacos , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteína Homeótica Nanog/efectos de los fármacos , Proteína Homeótica Nanog/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/efectos de los fármacos , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Faringe , ARN Mensajero/metabolismo , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo
7.
J Pak Med Assoc ; 66(3): 285-91, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26968278

RESUMEN

OBJECTIVE: To investigate the effects of collagen and growth factors on in vitro proliferation of human spermatogonial stem cells obtained from patients with non-obstructive azoospermia. METHODS: The experimental cross-sectional study was conducted from February 2013 to April 2015 after obtaining approval from the ethics committee of Ahvaz Jundishapur University of Medical Sciences, Iran. Testicular sperm extractions of non-obstructive azoospermic patients were obtained from the Clinical Urology and Embryology, In Vitro Fertilization Department of Imam Khomeini Hospital. Spermatogonial stem cells and Sertoli cells, obtained from human testis biopsies by a two-step enzymatic digestion method, were purified using fluorescence- activated cell-sorting and daturastramonium-lectin, and were cultured separately. To investigate a more direct influential factor on colony formation, one control and two experimental groups were formed. Group 1 acted as the control in which spermatogonial stem cells were co-cultured with Sertoli cells alone. In group 2 they were co-cultured with Sertoli cells and growth factors such as leukaemia inhibitory factor, epidermal growth factor and glial cell-derived neurotrophic factor, and in group 3 with Sertoli cells along with growth factors in the presence of collagen-coated dishes. Number and diameter of the colonies were evaluated after 7 weeks. RESULTS: Specimens obtained related to 21 patients. Number and diameter of the colonies in group 3 (18±2.6 and 276.6±45.5) were significantly more than both groups 1 (3.5±1 and D1:81.6±12) and group 2(11±2.2 and 165.2±32.5) (p<0.05 each). Also, the number and diameter of colony in group 2 were significantly better than the control group (p<0.05).Expression profile of the VASA, promyelocytic leukaemia zinc-finger (PLZF), Octamer-binding transcription factor 4 (OCT4) and integrin a6 (INTGa6) were detected in all groups. Based on cytochemical findings, OCT4 was expressed in the colonies of all three groups. CONCLUSIONS: According to positive effects of collagen and growth factors on the colonisation of spermatogonial stem cells, it seems that using the cells may lead to better colonisation of this type of stem cells.


Asunto(s)
Células Madre Germinales Adultas/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Colágeno/farmacología , Células Madre Germinales Adultas/citología , Azoospermia , Técnicas de Cultivo de Célula , Técnicas de Cocultivo , Estudios Transversales , ARN Helicasas DEAD-box/efectos de los fármacos , ARN Helicasas DEAD-box/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Citometría de Flujo , Factor Neurotrófico Derivado de la Línea Celular Glial/farmacología , Humanos , Integrina alfa6/efectos de los fármacos , Integrina alfa6/metabolismo , Factores de Transcripción de Tipo Kruppel/efectos de los fármacos , Factores de Transcripción de Tipo Kruppel/metabolismo , Factor Inhibidor de Leucemia/farmacología , Masculino , Factor 3 de Transcripción de Unión a Octámeros/efectos de los fármacos , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteína de la Leucemia Promielocítica con Dedos de Zinc , Células de Sertoli
8.
J Endod ; 40(10): 1566-72, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25260727

RESUMEN

INTRODUCTION: This study analyzed the detailed biological events underlying pulpal dynamics evoked by 3Mix (the mixture of ciprofloxacin, metronidazole, and minocycline) solution after intentionally delayed tooth replantation because 3Mix improves pulpal healing after tooth injuries. METHODS: The maxillary first molars of 3-week-old mice were extracted and immersed in 3Mix solution for 30 minutes in comparison with phosphate buffered saline (PBS) alone. Cell proliferation, apoptosis, and differentiation were assessed in extracted/replanted teeth during days 0-14 using immunohistochemistry, apoptosis assay, and reverse-transcriptase polymerase chain reaction. RESULTS: 3Mix solution accelerated odontoblast differentiation in the coronal pulp on day 7 and tertiary dentin formation on day 14, whereas the regenerative process was delayed in the PBS group. Cell proliferation and apoptosis occurred in the pulp of the 3Mix group during days 5-7 and subsequently decreased from days 7-14. On day 5, dentin sialophosphoprotein and nestin were first recovered in the 3Mix group, whereas expression levels for alkaline phosphatase, osteopontin, and osteocalcin increased in the PBS group. The expression levels for octamer-binding factor 3/4A and 3/4B reached the maximum level on day 1 and were sharply decreased on day 3 in both groups. High expression levels of Cd11c were first observed in the 3Mix group on day 1 and later at days 5 and 7. CONCLUSIONS: The results suggest that the application of 3Mix may suppress osteoblast differentiation by the migration of dendritic cells to the injury site and via the activation of stem/progenitor cells, resulting in the acceleration of odontoblastlike cell differentiation.


Asunto(s)
Antibacterianos/uso terapéutico , Pulpa Dental/efectos de los fármacos , Soluciones Preservantes de Órganos/uso terapéutico , Reimplante Dental/métodos , Fosfatasa Alcalina/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Tampones (Química) , Antígenos CD11/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ciprofloxacina/uso terapéutico , Pulpa Dental/citología , Dentina Secundaria/efectos de los fármacos , Combinación de Medicamentos , Proteínas de la Matriz Extracelular/efectos de los fármacos , Metronidazol/uso terapéutico , Ratones , Minociclina/uso terapéutico , Nestina/efectos de los fármacos , Factor 3 de Transcripción de Unión a Octámeros/efectos de los fármacos , Odontoblastos/efectos de los fármacos , Osteocalcina/efectos de los fármacos , Osteopontina/efectos de los fármacos , Fosfatos , Fosfoproteínas/efectos de los fármacos , Sialoglicoproteínas/efectos de los fármacos , Cloruro de Sodio , Factores de Tiempo
9.
J Endod ; 40(1): 101-8, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24331999

RESUMEN

INTRODUCTION: Dental pulp surrounded by rigid dentin is vulnerable to inflammatory stress; because of this, the invaded bacteria could cause irreversible pulpitis and necrosis. Octamer-binding transcription factor 4B1 (Oct-4B1), a newly discovered Oct-4 spliced variant belonging to the class V of the POU transcription factor family, serves as a precursor of Oct-4B and an essential functional isoform of Oct-4. However, its specific role in the inflammatory response of dental pulp cells (DPCs) remains unknown. METHODS: To explore the effect of Oct-4B1 on the inflammatory response of DPCs, messenger RNA expression of Oct-4B1 and Oct-4B in DPCs with lipopolysaccharide (LPS) induction was examined by real-time polymerase chain reaction. The expression of Oct-4B1 in DPCs was knocked down by specific small interfering RNA (siRNA); cell proliferation and the apoptosis rate were detected by the Cell Counting Kit-8 (Tokyo, Dojindo, Japan) and Hoechst-propidium iodide staining. The microRNA (miRNA) expression profiles were examined by miRNA microarray and bioinformatic analysis. RESULTS: We showed the messenger RNA expression of Oct-4B1 and Oct-4B was up-regulated in DPCs with LPS stimulation, whereas the knockdown expression of Oct-4B1 led to down-regulation of Oct-4B and an increased number of apoptotic cells in DPCs with LPS stimulation. Moreover, a total of 38 miRNAs were differentially expressed (including 4 up-regulated and 34 down-regulated) in DPCs with Oct-4B1 knockdown. Six of them were confirmed by real-time polymerase chain reaction, among which the target genes of miR-221 were predicted to be enriched in 14 Kyoto Encyclopedia of Genes and Genomes pathways represented by mitogen-activated protein kinase, Wnt, and Toll-like signaling pathways. CONCLUSIONS: Oct-4B1 may play a critical role in the inflammatory response of DPCs through interaction with miRNAs.


Asunto(s)
Pulpa Dental/citología , MicroARNs/inmunología , Factor 3 de Transcripción de Unión a Octámeros/inmunología , Transducción de Señal/inmunología , Adolescente , Adulto , Apoptosis/inmunología , Técnicas de Cultivo de Célula , Supervivencia Celular/inmunología , Células Cultivadas , Niño , Pulpa Dental/efectos de los fármacos , Pulpa Dental/inmunología , Técnicas de Silenciamiento del Gen , Proteínas HSP70 de Choque Térmico/análisis , Proteínas HSP70 de Choque Térmico/efectos de los fármacos , Humanos , Inflamación , Lipopolisacáridos/farmacología , Sistema de Señalización de MAP Quinasas/inmunología , Factor 3 de Transcripción de Unión a Octámeros/efectos de los fármacos , Factor 3 de Transcripción de Unión a Octámeros/genética , ARN Mensajero/análisis , Receptores Toll-Like/inmunología , Vía de Señalización Wnt/inmunología , Adulto Joven
10.
J Oral Pathol Med ; 40(8): 621-8, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21342274

RESUMEN

BACKGROUND: Oral squamous cell carcinoma (OSCC) is the sixth most prevalent cancer worldwide. Cancer stem cells (CSC) model theoretically contribute to tumor growth, metastasis, and chemo-radioresistance. Cisplatin is a widely used chemotherapeutic agent for OSCC treatment. The aim of this study was to compare stemness genes expression in chemo-sensitive and chemo-resistant specimens and further explore the potential markers that may lead to induce chemo-resistance in OSCC. METHODS: The study method is the treatment of OC2 cells with cisplatin select cisplatin-resistant OC2 cells. Self-renewal ability was evaluated by cultivating parental and cisplatin-resistant OC2 cells within sphere-forming assay after serial passages. Differential expression profile of stemness markers between parental and cisplatin-resistant OC2 cells was elucidated. The parental and cisplatin-resistant OC2 cells were assessed for migration/invasion/clonogenicity tumorigenic properties in vitro. Expression of stemness markers in chemo-sensitive and chemo-resistant patients with OSCC was performed by immunohistochemistry staining in vivo. RESULTS: Sphere-forming/self-renewal capability was increased in cisplatin-resistant OC2 cells. Cisplatin-resistant OC2 cells highly expressed the stemness markers (Nanog, Oct4, Bmi1, CD117, CD133, and ABCG2). Furthermore, cisplatin-resistant OC2 cells increased migration/invasion/clonogenicity ability. Notably, up-regulation of Oct4 and Nanog expression was significantly observed in cisplatin-resistant patients with OSCC (**P < 0.01). CONCLUSIONS: These data indicate that cancer stem-like properties were expanded during the acquisition of cisplatin resistance in OSCC. Clinically, oral cancer stemness markers (Oct4 and Nanog) overexpression may promote the OSCC's recurrence to resist cisplatin.


Asunto(s)
Antineoplásicos/uso terapéutico , Cisplatino/uso terapéutico , Proteínas de Homeodominio/metabolismo , Neoplasias de la Boca/metabolismo , Neoplasias de Células Escamosas/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Adulto , Anciano , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Resistencia a Antineoplásicos/genética , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Neoplasias de la Boca/tratamiento farmacológico , Neoplasias de la Boca/genética , Proteína Homeótica Nanog , Neoplasias de Células Escamosas/tratamiento farmacológico , Neoplasias de Células Escamosas/genética , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/efectos de los fármacos , ARN Neoplásico/análisis , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/metabolismo
11.
Eur J Pharmacol ; 591(1-3): 59-65, 2008 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-18616941

RESUMEN

This study was designed to investigate the role of nitric oxide (NO) in bone marrow stem cells and their differentiation into endothelial cells in vitro. Adult mouse bone marrow multipotent progenitor cells (MAPCs) were used as the source of stem cells. Oct-4 expression (both mRNA and protein) was significantly increased by up to 68.0% in MAPCs when incubated with NO donors DETA-NONOate or sodium nitroprusside (SNP) in a concentration-dependant manner (n=3, P<0.05). However, the cell proliferation was dramatically decreased by over 3-folds when treated with DETA-NONOate or SNP for 48 h (n=3, P<0.05). When MAPCs were exposed to DETA-NONOate (100 microM) for the first 48 h during differentiation, the expression (both mRNA and protein) of vWF was significantly increased at day 14 in the differentiating cells. The effects of DETA-NONOate or SNP on cell proliferation, Oct-4 expression and endothelial differentiation of MAPCs were not affected by the guanylyl cyclase inhibitor 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one or cGMP analog 8-Br-cGMP. These data indicate that NO may regulate both the pluripotency and differentiation of MAPCs via a cGMP-independent mechanism.


Asunto(s)
Células Madre Multipotentes/efectos de los fármacos , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Endoteliales/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Células Madre Multipotentes/metabolismo , Donantes de Óxido Nítrico/administración & dosificación , Nitroprusiato/administración & dosificación , Nitroprusiato/farmacología , Compuestos Nitrosos/administración & dosificación , Compuestos Nitrosos/farmacología , Factor 3 de Transcripción de Unión a Octámeros/efectos de los fármacos , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Factores de Tiempo
12.
Mol Reprod Dev ; 73(2): 153-63, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16250007

RESUMEN

RNA interference (RNAi) has become acknowledged as an effective and useful tool to study gene function in diverse groups of cells. We aimed to suppress the expression of the E-cadherin gene during in vitro development of bovine preimplantation embryos using RNAi approach. In this experiment the effect of microinjection of E-cadherin and Oct-4 (as control) double-stranded (ds) RNA on the mRNA and protein expression level of the target E-cadherin gene was investigated. For this, a 496 bp long bovine E-cadherin and 341 bp long Oct-4 dsRNA sample were prepared using in vitro transcription. In vitro produced bovine zygotes were categorized into four treatment groups including those injected with E-cadherin dsRNA, Oct-4 dsRNA, RNase-free water, and uninjected controls. While the injection of E-cadherin dsRNA resulted in the reduction of E-cadherin mRNA and protein levels at the morula and blastocyst stage, the transcript and protein product remained unaffected in the Oct-4 dsRNA, water injected and uninjected control groups. The relative abundance of E-cadherin mRNA in the E-cadherin dsRNA injected morula stage embryos was reduced by 80% compared to the control group (P < 0.05). The Western blot analysis also showed a significant decrease in the E-cadherin protein (119 kDa) in E-cadherin dsRNA injected embryos compared to the other three groups. Microinjection of E-cadherin dsRNA has resulted only 22% blastocyst rate compared to 38%-40% in water injected and uninjected controls. In conclusion, our results indicated the suppression of E-cadherin mRNA and protein has resulted in lower blastocyst rate and the RNAi technology is a promising approach to study the function of genes in early bovine embryogenesis.


Asunto(s)
Blastocisto/efectos de los fármacos , Cadherinas/genética , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Interferencia de ARN/fisiología , ARN Bicatenario/farmacología , Animales , Blastocisto/fisiología , Cadherinas/efectos de los fármacos , Bovinos , Desmocolinas , Fertilización In Vitro/métodos , Técnicas In Vitro , Glicoproteínas de Membrana/efectos de los fármacos , Glicoproteínas de Membrana/genética , Datos de Secuencia Molecular , Factor 3 de Transcripción de Unión a Octámeros/efectos de los fármacos , Factor 3 de Transcripción de Unión a Octámeros/genética , Fenotipo , ARN Mensajero/efectos de los fármacos , ARN Mensajero/genética , Transcripción Genética , beta Catenina/efectos de los fármacos , beta Catenina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...