Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Cells ; 10(4)2021 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-33920284

RESUMEN

Telomerase is known to contribute to telomere maintenance and to provide cancer cell immortality. However, numerous reports are showing that the function of the enzyme goes far beyond chromosome ends. The study aimed to explore how telomerase downregulation in MCF7 and MDA-MB-231 breast cancer cells affects their ability to survive. Consequently, sensitivity to drug resistance, proliferation, and adhesion were assessed. The lentiviral-mediated human telomerase reverse transcriptase (hTERT) downregulation efficiency was performed at gene expression and protein level using qPCR and Western blot, respectively. Telomerase activity was evaluated using the Telomeric Repeat Amplification Protocol (TRAP) assay. The study revealed that hTERT downregulation led to an increased sensitivity of breast cancer cells to doxorubicin which was demonstrated in MTT and clonogenic assays. During a long-term doubling time assessment, a decreased population doubling level was observed. Interestingly, it did not dramatically affect cell cycle distribution. hTERT downregulation was accompanied by an alteration in ß1-integrin- and by focal adhesion kinase (FAK)-driven pathways together with the reduction of target proteins phosphorylation, i.e., paxillin and c-Src. Additionally, autophagy activation was observed in MDA-MB-231 cells manifested by alternations in Atg5, Beclin 1, LC3II/I ratio, and p62. These results provide new evidence supporting the possible therapeutic potential of telomerase downregulation leading to induction of autophagy and cancer cells elimination.


Asunto(s)
Autofagia , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Regulación hacia Abajo , Doxorrubicina/farmacología , Resistencia a Antineoplásicos , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Telomerasa/metabolismo , Autofagia/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Silenciador del Gen/efectos de los fármacos , Humanos , Proteínas de Neoplasias/metabolismo , Ensayo de Tumor de Célula Madre
2.
Pharmacol Rep ; 72(1): 166-178, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32016855

RESUMEN

BACKGROUND: Drosera spatulata is a source of many compounds such as naphthoquinones, phenolic acids, flavonoids, anthocyanins, and naphthalene derivatives. Unfortunately, the information regarding the biological activity and chemical profile of those compounds is still incomplete. Herein, we investigated the biological activity of 3-O-acetylaleuritolic acid (3-O-AAA) in cancer cell lines. METHODS: The cell viability of HeLa, HT-29, MCF7, and MCF12A cells was assessed using MTT assay. Proliferation potential was assessed using the clonogenic assay and flow cytometry. Migration modulation was tested using a scratch assay. Protein expression was analyzed by immunoblotting. RESULTS: 3-O-AAA significantly inhibited the growth of all tested tumor cells. The results of the colony formation assay suggested cytostatic properties of the studied compound. The scratch assay showed that 3-O-AAA was an efficient migration inhibitor in a dose-dependent manner. Moreover, it caused modulation of mTOR, beclin1, and Atg5 proteins suggesting a possible role of the compound in autophagy induction. CONCLUSION: Collectively, these results demonstrated that 3-O-AAA inhibited the proliferation and migration of cancer cell lines as well as contributed to autophagy induction showing some anticancer properties.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Proliferación Celular/efectos de los fármacos , Drosera/química , Ácidos Palmíticos/farmacología , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/aislamiento & purificación , Autofagia/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Células HT29 , Células HeLa , Humanos , Células MCF-7 , Ácidos Palmíticos/administración & dosificación , Ácidos Palmíticos/aislamiento & purificación
3.
Int J Mol Sci ; 20(11)2019 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-31151281

RESUMEN

Human telomeres were one of the first discovered and characterized sequences forming quadruplex structures. Association of these structures with oncogenic and tumor suppressor proteins suggests their important role in cancer development and therapy efficacy. Since cationic porphyrin TMPyP4 is known as G-quadruplex stabilizer and telomerase inhibitor, the aim of the study was to analyze the anticancer properties of this compound in two different human breast-cancer MCF7 and MDA-MB-231 cell lines. The cytotoxicity of TMPyP4 alone or in combination with doxorubicin was measured by MTT (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromid) and clonogenic assays, and the cell-cycle alterations were analyzed by flow cytometry. Telomerase expression and activity were evaluated using qPCR and telomeric repeat amplification protocol (TRAP) assays, respectively. The contribution of G-quadruplex inhibitor to protein pathways engaged in cell survival, DNA repair, adhesion, and migration was performed using immunodetection. Scratch assay and functional assessment of migration and cell adhesion were also performed. Consequently, it was revealed that in the short term, TMPyP4 neither revealed cytotoxic effect nor sensitized MCF7 and MDA-MB-231 to doxorubicin, but altered breast-cancer cell adhesion and migration. It suggests that TMPyP4 might substantially contribute to a significant decrease in cancer cell dissemination and, consequently, cancer cell survival reduction. Importantly, this effect might not be associated with telomeres or telomerase.


Asunto(s)
Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Porfirinas/farmacología , Telomerasa/antagonistas & inhibidores , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Daño del ADN , Reparación del ADN , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Células MCF-7 , Transducción de Señal , Telomerasa/genética , Telomerasa/metabolismo
4.
Sci Rep ; 8(1): 12940, 2018 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-30154415

RESUMEN

Chronic obstructive pulmonary disease (COPD) is a risk factor for the development of lung cancer (LC). The mechanism of interplay between both diseases remains poorly recognized. This report examines whether COPD may cause a senescence response in human bronchial epithelial cells (HBECs), leading to the progression of LC in a senescence-dependent manner. The results show that HBECs exposed to serum from COPD patients manifest increased expression of markers of cellular senescence, including senescence-associated ß-galactosidase (SA-ß-Gal), histone γ-H2A.X, and p21, as compared to the serum of healthy donors. This effect coincides with an increased generation of reactive oxygen species by these cells. The clinical analysis demonstrated that COPD may cause the senescence, independently on smoking status and disease severity. The concentrations of CXCL5, CXCL8/IL-8 and VEGF were higher in conditioned medium (CM) harvested from HBECs after exposure to COPD serum as compared to controls. In addition, CM treated with serum from COPD patients stimulated adhesion of A549 cancer cells to HBECs, as well as accelerating cancer cell proliferation and migration in vitro. Collectively, these findings indicate that COPD may induce senescence-like changes in HBECs and thus enhance some processes associated with the progression of lung cancer.


Asunto(s)
Bronquios/metabolismo , Senescencia Celular , Células Epiteliales/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/sangre , Suero , Anciano , Anciano de 80 o más Años , Antígenos de Diferenciación/metabolismo , Bronquios/patología , Células Cultivadas , Citocinas/metabolismo , Células Epiteliales/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Enfermedad Pulmonar Obstructiva Crónica/patología
5.
BMC Cancer ; 18(1): 185, 2018 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-29439667

RESUMEN

BACKGROUND: Several efforts have been focused on identification of pathways involved in malignancy, progression, and response to treatment in Glioblastoma (GB). Overexpression of PKCε was detected in histological samples from GB, anaplastic astrocytoma, and gliosarcoma and is considered an important marker of negative disease outcome. In multiple studies on GB, autophagy has been shown as a survival mechanism during cellular stress, contributing to resistance against anti-cancer agents. The main object of this research was to determine the influence of PKCε downregulation on the expression of genes involved in autophagy pathways in glioblastoma cell lines U-138 MG and U-118 MG with high PKCε level. METHODS: We conducted siRNA-mediated knockdown of PKCε in glioblastoma cell lines and studied the effects of autophagy pathway. The expression of autophagy-related genes was analyzed using qPCR and Western blot analysis was carried out to assess protein levels. Immunostaining was used to detect functional autophagic maturation process. RESULTS: We found that these cell lines exhibited a high basal expression of autophagy-related genes. Our results suggest that the loss of PKCε contributes to the downregulation of genes involved in autophagy pathways. Moreover, most of the changes we observed in Western blot analysis and endogenous immunofluorescence experiments confirmed dysfunction of autophagy programs. We found that knockdown of PKCε induced a decrease in the expression of Beclin1, Atg5, PI3K, whereas the expression of other autophagy-related proteins mTOR and Bcl2 was increased. Treatment of control siRNA glioma cells with rapamycin-induced autophagosome formation and increase in LC3-II level and caused a decrease in the expression of p62. Additionally, PKCε siRNA caused a diminution in the Akt phosphorylation at Ser473 and in the protein level in both cell lines. Moreover, we observed reduction in the adhesion of glioblastoma cells, accompanied by the decrease in total FAK protein level and phosphorylation. CONCLUSIONS: Effects of down-regulation of PKCε in glioma cells raised the possibility that the expression of PKCε is essential for the autophagic signal transduction pathways in these cells. Thus, our results identify an important role of PKCε in autophagy and may, more importantly, identifyit as a novel therapeutic target.


Asunto(s)
Autofagia/genética , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Proteína Quinasa C-epsilon/genética , Proteína 5 Relacionada con la Autofagia/genética , Proteína 5 Relacionada con la Autofagia/metabolismo , Línea Celular Tumoral , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteína Quinasa C-epsilon/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Interferencia de ARN , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA