Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Korean J Radiol ; 25(3): 257-266, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38413110

RESUMEN

OBJECTIVE: To investigate molecular and functional consequences of additional exposures to iodine- or gadolinium-based contrast agents within 24 hours from the initial intravenous administration of iodine-based contrast agents through an animal study. MATERIALS AND METHODS: Fifty-six Sprague-Dawley male rats were equally divided into eight groups: negative control, positive control (PC) with single-dose administration of CT contrast agent, and additional administration of either CT or MR contrast agents 2, 4, or 24 hours from initial CT contrast agent injection. A 12 µL/g of iodinated contrast agent or a 0.47 µL/g of gadolinium-based contrast agent were injected into the tail vein. Serum levels of blood urea nitrogen, creatinine, cystatin C (Cys C), and malondialdehyde (MDA) were measured. mRNA and protein levels of kidney injury molecule-1 (KIM-1) and neutrophil gelatinase-associated lipocalin (NGAL) were evaluated. RESULTS: Levels of serum creatinine (SCr) were significantly higher in repeated CT contrast agent injection groups than in PC (0.21 ± 0.02 mg/dL for PC; 0.40 ± 0.02, 0.34 ± 0.03, and 0.41 ± 0.10 mg/dL for 2-, 4-, and 24-hour interval groups, respectively; P < 0.001). There was no significant difference in the average Cys C and MDA levels between PC and repeated CT contrast agent injection groups (Cys C, P = 0.256-0.362; MDA, P > 0.99). Additional doses of MR contrast agent did not make significant changes compared to PC in SCr (P > 0.99), Cys C (P = 0.262), and MDA (P = 0.139-0.771) levels. mRNA and protein levels of KIM-1 and NGAL were not significantly different among additional CT or MR contrast agent groups (P > 0.05). CONCLUSION: A sufficient time interval, probably more than 24 hours, between repeated contrast-enhanced CT examinations may be necessary to avoid deterioration in renal function. However, conducting contrast-enhanced MRI on the same day as contrast-enhanced CT may not induce clinically significant kidney injury.


Asunto(s)
Lesión Renal Aguda , Yodo , Ratas , Animales , Masculino , Medios de Contraste/efectos adversos , Lipocalina 2 , Ratas Sprague-Dawley , Gadolinio , Riñón , Imagen por Resonancia Magnética , Tomografía Computarizada por Rayos X , Administración Intravenosa , ARN Mensajero , Creatinina , Biomarcadores
2.
Biochem Pharmacol ; 215: 115761, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37604294

RESUMEN

Although the use of iodinated contrast agents is sometimes unavoidable for accurate diagnosis, contrast-induced acute kidney injury (CI-AKI) is a possible complication of its administration. The pathogenesis of CI-AKI has not been fully elucidated, but oxidative stress is thought to be a major factor. Sestrin2 plays an important role in cellular and mitochondrial homeostasis by regulating oxidative stress. In this study, we aimed to investigate whether recombinant adenovirus containing sestrin2 (RS) can attenuate CI-AKI by reducing oxidative stress in a CI-AKI mice model. Our results showed that RS decreases oxidative stress, pro-inflammatory cytokines (TNF-α, IL-1α, IL-1ß and IL-6) and apoptosis (Bax/Bcl2 and cleaved caspase-3) in the CI-AKI model. Additionally, RS alleviated mitochondrial damage, as evidenced by morphological changes, are restored ATP synthesis. Furthermore, RS administration resulted in a decrease in mitochondrial fission marker (Drp1) that was increased in the CI-AKI model, while the mitochondrial fusion marker (Mfn2) increased, indicating a restoration of mitochondrial dynamics. Decreased relative blood volume, as evaluated on computed tomography (CT), significantly increased compared to the CI-AKI group after RS administration. Finally, renal injury markers such as Kim-1, Ngal, IL-18 also decreased and kidney function was preserved with RS. These results suggested that RS can mitigate the deterioration of renal function in CI-AKI model.


Asunto(s)
Lesión Renal Aguda , Estrés Oxidativo , Animales , Ratones , Adenoviridae , Apoptosis , Citocinas , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/prevención & control
3.
Eur J Med Chem ; 239: 114501, 2022 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-35716517

RESUMEN

Inhibition of translation initiation has emerging implications for the development of mechanism-based anticancer therapeutics. Phosphorylation of eIF2α is recognized as a key target that regulates the translation initiation cascade. Based on the bioisosteric replacement of urea-derived eIF2α phosphorylation activator 1, a novel series of N-aryl-N'-[4-(aryloxy)cyclohexyl]squaramide derivatives was designed and synthesized; their effects on the activation of eIF2α phosphorylation was assessed systematically. A brief structure-activity relationship analysis was established by stepwise structural optimization of the squaramide series. Subsequently, the antiproliferative activities of the selected analogues were determined in human leukemia K562 cells. We then identified 10 potent eIF2α phosphorylation activators with considerable anticancer activity. The most promising analogues 19 and 40 possessed higher cancer cell selectivity (SI = 6.16 and 4.83, respectively) than parent 1 (SI = 2.20). Finally, protein expression analysis revealed that compounds 19 and 40 induced eIF2α phosphorylation and its downstream effectors ATF4 and CHOP.


Asunto(s)
Factor 2 Eucariótico de Iniciación , Quinina , Humanos , Fosforilación , Quinina/análogos & derivados , Relación Estructura-Actividad
4.
Anticancer Drugs ; 33(1): e370-e380, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34419957

RESUMEN

The dysregulation of glycolysis regardless of oxygen availability is one of the major characteristics of cancer cells. While the drug resistance of ovarian cancer cells has been extensively studied, the molecular mechanism of anticancer drug resistance under low-glucose conditions remains unknown. In this study, we investigated the pathway mediating drug resistance under low-glucose conditions by examining the relationship between embryonic lethal abnormal vision Drosophila homolog-like (ELAVL) protein and glycolysis-related enzymes. Ovarian cancer cells resistant to 2.5 nM paclitaxel were exposed to low-glucose media for 2 weeks, and the expression levels of ELAVL2, ELAVL4, glycolytic enzymes, and drug resistance-related proteins were elevated to levels comparable to those in cells resistant to 100 nM paclitaxel. Gene silencing of ELAVL2/4 using small interfering RNA prevented the upregulation of glycolysis-related enzymes, reduced lactate production, and sensitized 2.5 nM paclitaxel-resistant ovarian cancer cells to anticancer agents under hypoglycemic conditions. Furthermore, pharmacological inhibition of glycolytic enzymes with 2-deoxyglucose, a specific inhibitor of glycolysis, triggered caspase-dependent apoptosis, reduced lactate generation, and blocked the expression of drug resistance-related proteins under low-glucose conditions. These results suggest that the level of ELAVL2/4 is responsible for the development of chemoresistance through activation of the glycolysis pathway under glucose deprivation conditions.


Asunto(s)
Resistencia a Antineoplásicos/genética , Proteína 2 Similar a ELAV/genética , Proteína 4 Similar a ELAV/genética , Glucólisis/genética , Neoplasias Ováricas/genética , Antineoplásicos/farmacología , Caspasas/metabolismo , Línea Celular Tumoral , Femenino , Glucosa/metabolismo , Humanos , Ácido Láctico/metabolismo , Neoplasias Ováricas/patología , Paclitaxel/farmacología
5.
Mol Med Rep ; 23(6)2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33846772

RESUMEN

The abnormal expression of tropomyosin receptor kinase (Trk) serves an important role in the promotion of cancer progression. Homeobox C6 (HOXC6) and A disintegrin and metalloproteinase domain­containing 8 (ADAM8) are associated with the invasiveness of cancer cells. However, the exact relationship between these molecules and their downstream signaling pathways in chemoresistant colon cancer cells are largely unknown. Therefore, the current study investigated the association between TrkB/C with HOXC6 and ADAM8 in the induction of drug­resistant colon cancer cell metastasis. The results demonstrated that chemoresistant colon cancer cells exhibited upregulated TrkB/C, HOXC6 and ADAM8 expression. Additionally, but also chemoresistant colon cancer cells demonstrated higher migratory activities compared with parent colon cancer cells. The pharmacological inhibition of TrkB/C activity reduced the phosphorylation of mitogen­activated protein kinase kinase/ERK and subsequently suppressed HOXC6 and ADAM8 expression. In addition, gene silencing of HOXC6 inhibited ADAM8 and MMP activity, and inhibited the migration and invasion of drug­resistant cancer cells. However, the targeted downregulation of ADAM8 using small interfering RNA failed to suppress TrkB/C­associated ERK­mediated HOXC6 signaling activity. Furthermore, pre­treatment with ADAM10­ and ADAM17­specific inhibitors had no effect on attenuating the invasiveness of chemoresistant colon cancer cells. The results indicated that TrkB/C­mediated ERK activation serves an important role in the metastasis of drug­resistant colon cancer cells through the regulation of HOXC6/ADAM8 activity.


Asunto(s)
Proteínas ADAM/metabolismo , Neoplasias del Colon/metabolismo , Genes Homeobox/efectos de los fármacos , Proteínas de Homeodominio/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de la Membrana/metabolismo , Receptor trkB/metabolismo , Receptor trkC/metabolismo , Proteínas ADAM/genética , Proteína ADAM10 , Proteína ADAM17 , Antineoplásicos/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Neoplasias del Colon/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas de Homeodominio/genética , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/farmacología , Proteínas de la Membrana/genética , Receptor trkB/genética , Receptor trkB/farmacología , Receptor trkC/genética , Receptor trkC/farmacología , Transducción de Señal , Regulación hacia Arriba
6.
Carcinogenesis ; 41(10): 1329-1340, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32649737

RESUMEN

In cancer, resistance to chemotherapy is one of the main reasons for therapeutic failure. Cells that survive after treatment with anticancer drugs undergo various changes, including in cell metabolism. In this study, we investigated the effects of AKT-mediated miR-125b-5p alteration on metabolic changes and examined how these molecules enhance migration and induce drug resistance in colon cancer cells. AKT1 and AKT3 activation in drug-resistant colon cancer cells caused aberrant downregulation of miR-125b-5p, leading to GLUT5 expression. Targeted inhibition of AKT1 and AKT3 restored miR-125b-5p expression and prevented glycolysis- and lipogenesis-related enzyme activation. In addition, restoring the level of miR-125b-5p by transfection with the mimic sequence not only significantly blocked the production of lactate and intracellular fatty acids but also suppressed the migration and invasion of chemoresistant colon cancer cells. GLUT5 silencing with small interfering RNA attenuated mesenchymal marker expression and migratory activity in drug-resistant colon cancer cells. Additionally, treatment with 2,5-anhydro-d-mannitol resensitized chemoresistant cancer cells to oxaliplatin and 5-fluorouracil. In conclusion, our findings suggest that changes in miR-125b-5p and GLUT5 expression after chemotherapy can serve as a new marker to indicate metabolic change-induced migration and drug resistance development.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Resistencia a Antineoplásicos , Transportador de Glucosa de Tipo 5/metabolismo , MicroARNs/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Antineoplásicos/farmacología , Movimiento Celular , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Regulación hacia Abajo , Activación Enzimática , Glucólisis/genética , Células HT29 , Humanos , Lipogénesis/genética , MicroARNs/genética , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/uso terapéutico , Receptores Toll-Like/metabolismo
7.
Mol Carcinog ; 59(2): 154-167, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31746054

RESUMEN

CD248, also called endosialin or tumor endothelial marker-1, is markedly upregulated in almost all cancers, including colon cancers. Changes in microRNA profiles are one of the direct causes of cancer development and progression. In this study, we investigated whether a change in CD248 expression in colon cancer cells could induce drug resistance after chemotherapy, and we explored the relationship between miR-125b-5p levels and CD248 expression in Toll-like receptor (TLR)-modified chemoresistant colon cancer cells. TLR2/6 and TLR5 upregulation in drug-resistant colon cancer cells contributed to miR-125b-5p downregulation and specificity protein 1 (Sp1)-mediated CD248 upregulation via nuclear factor-kappa B (NF-κB) activation. Exposure to specific TLR2/6 or TLR5 ligands enhanced the expression of mesenchymal markers as well as the migratory activity of oxaliplatin- or 5-fluorouracil-resistant colon cancer cells. The transfection of a synthetic miR-125b-5p mimic into chemoresistant cells prevented Sp1 and CD248 activation and significantly impaired invasive activity. Furthermore, Sp1 or CD248 gene silencing as well as miR-125b-5p overexpression markedly reversed drug resistance and inhibited epithelial-mesenchymal transition in colon cancer cells. Taken together, these results suggest that changes in miR-125b-5p levels play an important role in Sp1-mediated CD248 expression and the development of drug resistance in TLR-mutated colon cancer cells.


Asunto(s)
Antígenos CD/genética , Antígenos de Neoplasias/genética , Neoplasias Colorrectales/genética , Fluorouracilo/farmacología , Regulación Neoplásica de la Expresión Génica/genética , MicroARNs/genética , Receptores Toll-Like/genética , Antígenos CD/metabolismo , Antígenos de Neoplasias/metabolismo , Antimetabolitos Antineoplásicos/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Regulación hacia Abajo/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HCT116 , Células HT29 , Humanos , FN-kappa B/metabolismo , Interferencia de ARN , Receptores Toll-Like/metabolismo
8.
Mar Drugs ; 17(9)2019 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-31505769

RESUMEN

Intracellular reactive oxygen species (ROS) play an important role in the proliferation and differentiation of hematopoietic stem and progenitor cells (HSPCs). HSPCs are difficult to be expanded ex vivo while maintaining their stemness when they are exposed to oxidative damage after being released from the bone marrow. There have been efforts to overcome this limitation by using various cytokine cocktails and antioxidants. In this study, we investigated the effects of echinochrome A (Ech A)-a well-established and non-toxic antioxidant-on the ex vivo expansion of HSPCs by analyzing a CD34+ cell population and their biological functions. We observed that Ech A-induced suppression of ROS generation and p38-MAPK/JNK phosphorylation causes increased expansion of CD34+ cells. Moreover, p38-MAPK/JNK inhibitors SB203580 and SP600125 promoted ex vivo expansion of CD34+ cells. We also demonstrated that the activation of Lyn kinase and p110δ is a novel mechanism for Ech A to enhance ex vivo expansion of CD34+ cells. Ech A upregulated phospho-Src, phospho-Lyn, and p110δ expression. Furthermore, the Ech A-induced ex vivo expansion of CD34+ cells was inhibited by pretreatment with the Src family inhibitor PP1 and p110δ inhibitor CAL-101; PP1 blocked p110δ upregulation and PI3K/Akt activation, whereas CAL-101 and PI3K/Akt pathway inhibitor LY294002 did not block Src/Lyn activation. These results suggest that Ech A initially induces Src/Lyn activation, upregulates p110δ expression, and finally activates the PI3K/Akt pathway. CD34+ cells expanded in the presence of Ech A produced equal or more hematopoietic colony-forming cells than unexpanded CD34+ cells. In conclusion, Ech A promotes the ex vivo expansion of CD34+ cells through Src/Lyn-mediated p110δ expression, suppression of ROS generation, and p38-MAPK/JNK activation. Hence, Ech A is a potential candidate modality for the ex vivo, and possibly in vivo, expansion of CD34+ cells.


Asunto(s)
Antígenos CD34/metabolismo , Células Sanguíneas/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Naftoquinonas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Familia-src Quinasas/metabolismo , Antracenos/farmacología , Antioxidantes/metabolismo , Células Sanguíneas/metabolismo , Células Cultivadas , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Humanos , Imidazoles/farmacología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Piridinas/farmacología , Células Madre/efectos de los fármacos , Células Madre/metabolismo
9.
Mar Drugs ; 17(7)2019 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-31336860

RESUMEN

Death-associated protein kinase 1 (DAPK1) expression induced by diverse death stimuli mediates apoptotic activity in various cancers, including ovarian cancer. In addition, mutual interaction between the tumor suppressor p53 and DAPK1 influences survival and death in several cancer cell lines. However, the exact role and connection of DAPK1 and p53 family proteins (p53, p63, and p73) in drug-resistant ovarian cancer cells have not been studied previously. In this study, we investigated whether DAPK1 induction by gliotoxin derived from marine fungus regulates the level of transcriptionally active p63 (TAp63) to promote apoptosis in an autophagy-dependent manner. Pre-exposure of paclitaxel-resistant ovarian cancer cells to gliotoxin inhibited the expression of multidrug resistant-associated proteins (MDR1 and MRP1-3), disrupted the mitochondrial membrane potential, and induced caspase-dependent apoptosis through autophagy induction after subsequent treatment with paclitaxel. Gene silencing of DAPK1 prevented TAp63-mediated downregulation of MDR1 and MRP1-3 and autophagic cell death after sequential treatment with gliotoxin and then paclitaxel. However, pretreatment with 3-methyladenine (3-MA), an autophagy inhibitor, had no effect on the levels of DAPK1 and TAp63 or on the inhibition of MDR1 and MRP1-3. These results suggest that DAPK1-mediated TAp63 upregulation is one of the critical pathways that induce apoptosis in chemoresistant cancer cells.


Asunto(s)
Muerte Celular Autofágica/efectos de los fármacos , Gliotoxina/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Línea Celular Tumoral , Proteínas Quinasas Asociadas a Muerte Celular/genética , Proteínas Quinasas Asociadas a Muerte Celular/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Silenciador del Gen , Gliotoxina/uso terapéutico , Humanos , Neoplasias Ováricas/patología , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , ARN Interferente Pequeño/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Regulación hacia Arriba/efectos de los fármacos
10.
J Diabetes Res ; 2019: 2376512, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30729133

RESUMEN

Metformin and pioglitazone are two commonly prescribed oral hypoglycemic agents for diabetes. Recent evidence suggests that these drugs may contribute to bladder cancer. This study investigated molecular mechanism underlying effects of metformin and pioglitazone in bladder epithelial carcinogenesis in type 2 diabetes. The cells derived from human bladder epithelial cells (HBlEpCs) were treated with metformin or pioglitazone with high glucose and insulin. Cell viability and proliferation were evaluated using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and a bromodeoxyuridine incorporation assay, respectively, while cell cycle regulatory factors and oncogene expression were analyzed using western blotting. Metformin or pioglitazone suppressed cell viability concentration and time dependently, which was reversed by exposure to high glucose with or without insulin. Prolonged exposure to high glucose and insulin enhanced cyclin D, cyclin-dependent kinase 4 (Cdk4), and Cdk2 expression and suppressed cyclin-dependent kinase inhibitors p21 and p15/16 in HBlEpC cotreated with pioglitazone and metformin. Levels of tumor suppressor proteins p53 and cav-1 were downregulated while those of the oncogenic protein as c-Myc were upregulated under high glucose and insulin supplementation in HBlEpC cotreated with pioglitazone and metformin. Prolonged exposure to high glucose with or without insulin downregulated B cell lymphoma 2-associated X (Bax) and failed to enhance the expression of extracellular signal-regulated kinase (ERK) and p38 mitogen-activated protein kinase (p38MAPK) in drug-treated cells. These results suggest that hyperglycemic and insulinemic conditions promote cell cycle progression and oncogenic signaling in drug-treated bladder epithelial cells and uncontrolled hyperglycemia and hyperinsulinemia are probably greater cancer risk factors than diabetes drugs.


Asunto(s)
Ciclo Celular/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Glucosa/farmacología , Insulina/farmacología , Metformina/farmacología , Pioglitazona/farmacología , Vejiga Urinaria/efectos de los fármacos , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Epiteliales/citología , Humanos , Hipoglucemiantes/farmacología , Transducción de Señal/efectos de los fármacos , Vejiga Urinaria/citología
11.
Neoplasia ; 21(2): 206-215, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30622051

RESUMEN

CD97 shows a strong relationship with metastasis and poor clinical outcome in various tumors, including ovarian cancer. The expression of CD97 in metastatic ovarian cancer cells was higher than that in primary ovarian cancer cells. Mature miRNAs are frequently de-regulated in cancer and incorporated into a specific mRNA, leading to post-transcriptional silencing. In this study, we investigated whether the miR-503-5p targeting of the CD97 3'-untranslated region (3'-UTR) contributes to ovarian cancer metastasis as well as the underlying mechanism regulating cancer progression. In LPS-stimulated or paclitaxel-resistant ovarian cancer cells, stimulation with recombinant human CD55 (rhCD55) of CD97 in ovarian cancer cells activated NF-κB-dependent miR-503-5p down-regulation and the JAK2/STAT3 pathway, consequently promoting the migratory and invasive capacity. Furthermore, restoration of miR-503-5p by transfection with mimics or NF-κB inhibitor efficiently blocked CD97 expression and the downstream JAK2/STAT3 signaling pathway. Target inhibition of JAK with siRNA also impaired colony formation and metastasis of LPS-stimulated and paclitaxel-resistant ovarian cancer cells. Taken together, these results suggest that high CD97 expression, which is controlled through the NF-κB/miR-503-5p signaling pathway, plays an important role in the invasive activity of metastatic and drug-resistant ovarian cancer cells by activating the JAK2/STAT3 pathway.


Asunto(s)
Antígenos CD/genética , Regulación Neoplásica de la Expresión Génica , Janus Quinasa 2/metabolismo , MicroARNs/genética , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Regiones no Traducidas 3' , Antígenos CD/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Resistencia a Antineoplásicos/genética , Femenino , Humanos , FN-kappa B , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Paclitaxel/farmacología , Unión Proteica , Interferencia de ARN , Receptores Acoplados a Proteínas G
12.
Oncol Lett ; 16(4): 4526-4536, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30214587

RESUMEN

Treatment with celecoxib and bortezomib as single chemotherapeutic agents reduces the viability and proliferation of colorectal cancer cells. The use of these agents in combination with other chemotherapeutic agents is usually associated with adverse effects. In the present study, a combination of celecoxib and bortezomib was investigated for potential synergistic effects in colon cancer cells. The sequential exposure to celecoxib with bortezomib synergistically induced apoptotic death in human colon cancer cells compared with groups treated with a single drug or other drug combinations. c-Jun N-terminal kinase/p38-mitogen-activated protein kinase-induced endoplasmic reticulum (ER) stress through serial exposure to celecoxib and bortezomib may have induced the intracellular Ca2+ release, leading to the generation of autophagosomes in p53-expressing HCT-116 cells. Targeted inhibition of p53 activity or ER stress or treatment with the Ca2+-chelating agent BAPTA-AM suppressed the ER stress-mediated Ca2+ release and apoptosis. Although p53-/- HCT-116 cells were less sensitive to sequential treatment with celecoxib and bortezomib, co-localization of autophagosomes was detected in the absence of CCAAT-enhancer-binding protein homologous protein expression. Treatment of p53-/- HCT-116 cells with BAPTA-AM did not inhibit apoptosis following serial treatment with celecoxib and bortezomib. These results suggest that the order of drug administration is important in treating cancer and that the sequential treatment with celecoxib and bortezomib enhances the ER stress-mediated autophagy-associated cell death of colon cancer cells, regardless of p53 expression.

13.
Mol Cell Biochem ; 439(1-2): 199-211, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28819788

RESUMEN

The activation of phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) is critical for the induction of epithelial-mesenchymal transition (EMT) by growth factors, including insulin-like growth factor 1 (IGF-1). The activation of intracellular lipogenesis provides proliferative and survival signals for cancer cells. In this study, we investigated the connection between lipogenesis-related EMT processes and IGF-1-mediated PI3K p110 isoform activation in primary (SW480 cells) and metastatic (SW620) colon carcinoma cells. We also examined the underlying signaling pathway that promotes fatty acid synthesis in IGF-1-activated colon cancer cells. IGF-1 stimulation upregulated the expression of lipogenic enzymes as well as the activation of Nardilysin (N-arginine dibasic convertase, NRD1) and its downstream targets, a disintegrin and metalloproteases 10 (ADAM10) and ADAM17. The upregulation of the Lyn/Syk-mediated PI3K p110δ isoform in SW480 cells and the Lyn-dependent PI3K p110α isoform in SW620 cells triggered fatty acid production and cell motility in IGF-1-activated colon cancer cells. Pharmacological inhibition with A66 (PI3K p110α specific inhibitor) and CAL-101 (PI3K p110δ specific inhibitor) efficiently inhibited EMT in colon cancer cells by blocking the NRD1/ADAM family protein signaling pathway. Gene silencing of NRD1 and ADAM family proteins attenuated the generation of intracellular fatty acid and the migratory activity of colon cancer cells. Our results suggest that the different isoforms of the PI3K p110 subunit could be therapeutic targets for primary and metastatic colon cancer and that regulation of the NRD1/ADAM signaling pathway controls lipogenesis-mediated EMT in IGF-1-stimulated colon cancer cells.


Asunto(s)
Proteína ADAM10/metabolismo , Proteína ADAM17/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Neoplasias del Colon/metabolismo , Transición Epitelial-Mesenquimal , Factor I del Crecimiento Similar a la Insulina/metabolismo , Lipogénesis , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Proteína ADAM10/genética , Proteína ADAM17/genética , Secretasas de la Proteína Precursora del Amiloide/genética , Fosfatidilinositol 3-Quinasa Clase I/genética , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Humanos , Factor I del Crecimiento Similar a la Insulina/genética , Proteínas de la Membrana/genética , Proteínas de Neoplasias/genética
14.
Mol Med Rep ; 17(3): 3563-3574, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29286114

RESUMEN

Epithelial-mesenchymal transition (EMT) of retinal pigment epithelial (RPE) cells is inevitable change of age­related macular degeneration (AMD). Smoking is a major risk factor for the development of EMT in several diseases, including lung cancer. Cigarette smoke­induced stress promotes the production of epidermal growth factor (EGF) in RPE cells. However, the underlying signaling pathways induced by aberrant EGF receptor (EGFR) expression in cigarette smoke-exposed RPE cells remain largely unknown. In the present study, the morphological transformation and production of EMT-associated cytokines were investigated to analyze the effect of smoking on the retina. Furthermore, EGF­treated or cigarette smoke­exposed RPE cells, as well as the downstream targets of EGFR, were investigated to identify the key molecules involved in EMT of cigarette smoke­stimulated RPE cells via immunoblotting. Exposure of RPE cells to cigarette smoke extract (CSE) induced secretion of VEGF and TGF­ß1, and increased the expression of EMT markers. CSE­mediated focal adhesion kinase (FAK) activation resulted in the phosphorylation and activation of spleen associated tyrosine kinase (Syk)/Src proto­oncogene, non­receptor tyrosine kinase (Src), leading to migration and invasion of RPE cells. Knockdown of FAK or pharmacological inhibition of Syk/Src abrogated CSE­mediated VEGF and TGF­ß1 production and blocked the phosphorylation of Smad2/3 in CSE­stimulated RPE cells. Erlotinib (an EGFR inhibitor) suppressed EGF and CSE­mediated switch from an epithelial to mesenchymal phenotype. Baicalein, an inhi-bitor of 12/15­lipooxygenase, also efficiently suppressed CSE­induced EMT processes by inhibiting EGFR­associated downstream signaling transduction. The results identified a novel signaling pathway mediated by EGFR in CSE­activated RPE cells, and suggest baicalein as a potential new therapeutic drug for CSE­associated retinopathy.


Asunto(s)
Transición Epitelial-Mesenquimal , Receptores ErbB/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Humo/efectos adversos , Quinasa Syk/metabolismo , Familia-src Quinasas/metabolismo , Araquidonato 12-Lipooxigenasa/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Quinasa 1 de Adhesión Focal/genética , Humanos , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Retina/citología , Transducción de Señal/efectos de los fármacos , Proteínas Smad/metabolismo , Nicotiana/química , Nicotiana/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
15.
Oncol Lett ; 14(6): 7947-7956, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29250183

RESUMEN

Ampelopsin (Amp) is bioactive natural product and exerts anti-cancer effects against several cancer types. The present study investigated the anti-colon cancer activity of Amp and explored its mechanism of action. The treatment of colon cancer cells with Amp resulted in the dose- and time-dependent induction of apoptosis via the activation of endoplasmic reticulum (ER) stress, 5' adenosine monophosphate-activated protein kinase (AMPK), and c-Jun N-terminal protein kinase (JNK)/p38 mitogen-activated protein kinases (MAPKs). Salubrinal, an ER stress inhibitor, prevented the upregulation of ER stress-associated proteins, including phosphorylated protein kinase RNA-like ER kinase, phosphorylated eukaryotic translation initiation factor 2α, glucose-regulated protein 78, and CCAAT/enhancer-binding protein homologous protein, as well as suppressing AMPK activation and the MAPK signaling pathway. Knockdown of AMPK by RNA interference failed to block ER stress. Additionally, SP600125 (a JNK inhibitor) and SB203580 (a p38-MAPK inhibitor) effectively inhibited apoptosis and attenuated the expression of X-linked IAP-associated factor 1 (XAF1) and apoptotic Bcl-2 family proteins (BCL2 antagonist/killer 1 and BCL2-associated X protein) in Amp-treated colon cancer cells. Furthermore, reactive oxygen species (ROS)-mediated ER stress/AMPK apoptotic signaling pathway in Amp-treated colon cancer cells were markedly inhibited by treatment with N-acetyl-L-cysteine, a ROS scavenger. These results demonstrate that treatment with Amp induces the apoptotic death of colon cancer cells through ER stress-initiated AMPK/MAPK/XAF1 signaling. These results also provide experimental information for developing Amp as therapeutic drug against colon cancer.

16.
Oncol Rep ; 38(5): 3167-3176, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28901470

RESUMEN

Toll-like receptor (TLR)-mediated signaling induces cell migration or invasion in several tumors and various stages of cancer. Interactions of mesothelin, a 40-kDa cell surface glycoprotein, with cancer antigen 125 (CA125) is associated with drug resistance, metastasis, and poor clinical outcome of ovarian cancer patients. In this study, we examined the role of TLR5 and TLR7 in the metastasis of ovarian cancer through the induction of mesothelin/CA125 expression and investigated its underlying mechanism. TLR5 agonist (flagellin) and TLR7 agonist (imiquimod) upregulated mesenchymal phenotypes and produced epithelial-mesenchymal transition (EMT)-related cytokines in the SKOV3 cells; however, TLR7 expressing CaOV3 cells had no response to the specific ligand, imiquimod, for enhancing its EMT processes. Stimulation of the SKOV3 cells with flagellin or imiquimod activated Wiskott-Aldrich syndrome protein verprolin-homologous 3 (WAVE3) and mesothelin/CA125, whereas it suppressed the expression of TAp63. Moreover, knockdown of TLR5 or TLR7 in SKOV3 cells profoundly impaired the TLR5- or TLR7-intiated downstream signaling pathway. Loss of WAVE3 in SKOV3 cells led to the inhibition of invasion, suppression of mesenchymal characteristics, prevention of OCT4/SOX2 secretion, and attenuation of mesothelin/CA125 expression after stimulation with flagellin or imiquimod. Although the disruption of mesothelin decreased the migratory activity of the TLR5/7-activated SKOV3 cells, knockdown of mesothelin failed to reduce the expression of mesenchymal markers, OCT4, and SOX2. In addition, targeting OCT4 or SOX2 with siRNA had no effect on the expression of mesothelin and the suppression of transcriptionally active p63 (TAp63) in the TLR5/7-stimulated SKOV3 cells. Our results suggest that TLR5/7-mediated WAVE3 activation not only controls the mesothelin-related EMT processes but also modulates OCT4/SOX2-mediated mesenchymal marker expression. Taken together, both TLR5 and TLR7 expression are critical for the TLR5/7-induced metastasis of ovarian cancer and the inhibition of WAVE3 might be a new therapeutic target to control ovarian cancer metastasis.


Asunto(s)
Proteínas Ligadas a GPI/metabolismo , Neoplasias Ováricas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Receptor Toll-Like 5/metabolismo , Receptor Toll-Like 7/metabolismo , Familia de Proteínas del Síndrome de Wiskott-Aldrich/metabolismo , Antígeno Ca-125/metabolismo , Línea Celular Tumoral , Transición Epitelial-Mesenquimal , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas de la Membrana/metabolismo , Mesotelina , Invasividad Neoplásica , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Factores de Transcripción SOXB1/metabolismo , Activación Transcripcional
17.
Mol Med Rep ; 16(2): 1826-1836, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28627617

RESUMEN

Cluster of differentiation (CD) 44 and epidermal growth factor (EGF) are closely involved in cellular migration and have been used as stem cell markers. Although the hyaluronan (HA)­binding CD44 is responsible for enhanced cellular motility, the mechanism underlying its actions in various cell types and clinical conditions have yet to be elucidated. In the present study, the multikinase inhibitor sorafenib was used to investigate the diverse effects of EGF stimulation on epithelial­mesenchymal transition (EMT) in ovarian cancer cells using immunoblotting and reverse transcription­polymerase chain reaction. In addition, the association between EGF and CD44/HA signaling pathways in the control of mesenchymal phenotype was determined by gene silencing with small interfering RNA transfection. EGF stimulation of ovarian cancer cells increased cellular migration, mesenchymal transition, CD44 expression and the activation of matrix metalloproteinase (MMP)­2 and MMP­9. Sorafenib effectively suppressed the loss of epithelial characteristics in EGF­treated SK­OV­3 ovarian cancer cells, via targeting the mitogen­activated protein kinase (MAPK)/extracellular signal­regulated kinase (ERK) pathway. Although treatment of Caov­3 ovarian cancer cells with sorafenib blocked the expression of mesenchymal phenotypes following EGF stimulation, EGF­activated Caov­3 cells exhibited reduced MAPK/ERK signaling. Furthermore, EGF­activated Caov­3 cells increased the expression of hyaluronan synthase 2 and HA­CD44 ligation in EGF­exposed Caov­3 cells, which resulted in the activation of the Ras/Raf/MEK signaling pathway, amplification of migratory activity and the expression of mesenchymal markers, including N­cadherin and vimentin. Furthermore, silencing EGFR in SK­OV­3 cells and CD44 in Caov­3 cells suppressed their migratory activity, through inhibition of the MAPK/ERK pathway. The present results suggested that EGF­mediated signaling may regulate metastasis and invasion of ovarian cancer cells, in a cancer cell type­dependent manner.


Asunto(s)
Factor de Crecimiento Epidérmico/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Receptores de Hialuranos/metabolismo , Ácido Hialurónico/metabolismo , Niacinamida/análogos & derivados , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Compuestos de Fenilurea/farmacología , Transducción de Señal/efectos de los fármacos , Basigina/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Femenino , Humanos , Hialuronano Sintasas/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Invasividad Neoplásica , Niacinamida/farmacología , Fenotipo , ARN Interferente Pequeño/metabolismo , Sorafenib
18.
Exp Ther Med ; 13(4): 1415-1425, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28413487

RESUMEN

The extracellular signals induced by vascular endothelial growth factor (VEGF) are implicated in choroidal neovascularization (CNV) and thus, are associated with vision-limiting complications in the human retina. Vandetanib is an oral anticancer drug that selectively inhibits the activities of VEGF receptor and epidermal growth factor receptor tyrosine kinase; however, the effects of vandetanib on VEGF in retinal pigment epithelial (RPE) cells have not yet been studied. In the present study, a combined treatment of vandetanib and a disintegrin and metalloproteinase (ADAM) protein inhibitors were used to assess the regulation of Epstein-Barr virus (EBV)-infected ARPE19 cells (ARPE19/EBV) migration as a model of CNV. Vandetanib suppressed the expression of the mesenchymal markers ADAM10 and ADAM17 in ARPE19/EBV cells, and also upregulated epithelial cell markers of the RPE cells, E-cadherin and N-cadherin. The migratory activity of ARPE19/EBV induced by VEGF was efficiently blocked by vandetanib. Furthermore, co-treatment with vandetanib and an ADAM10 inhibitor (GI254023X) or ADAM17 inhibitor (Marimastat) synergistically prevented migration and the expression of vimentin, Snail and α-smooth muscle actin by regulating extracellular signal-regulated kinase and p38 mitogen-activated protein kinase. These results suggest that a combination treatment of vandetanib and ADAM inhibitors may be developed as a novel therapeutic regimen to control retina neovascular disease.

19.
Anticancer Res ; 37(4): 1805-1818, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28373445

RESUMEN

AIM: To investigate the molecular mechanisms by which long-term exposure to cigarette smoke extract (CSE) contributes to ovarian cancer metastasis. MATERIALS AND METHODS: Western blot analysis for diverse p110 isoforms of phosphoinositide 3-kinase (PI3K)-related signaling pathway and epithelial-mesenchymal transition (EMT) markers was performed to analyze the underlying mechanisms. Migratory activity of CSE-exposed ovarian cancer cells was determined by transendothelial migration and invasion assay. RESULTS: After exposure to CSE for four weeks, CaOV3 (primary) and SKOV3 (metastatic) ovarian cancer cells showed enhanced mesenchymal characteristics and produced EMT-related cytokines [intwerleukin-8 (IL-8), vascular endothelial growth factor (VEGF) and transforming growth factor-beta 1 (TGF-ß1)]. CSE exposure activated the Src-p110δ-p21 protein-activated kinase 1 (PAK1) in CaOV3 cells and the Lyn-p110ß-Rho-associated kinases 1/2 (ROCK1/2) in SKOV3 cells, which led to the stimulation of LIM kinase 1 (LIMK1) phosphorylation and TGF-ß1 release. LIMK1 knockdown efficiently blocked the migratory activity and suppressed the mesenchymal phenotypes of CSE-treated ovarian cancer cells. Reactive oxygen species (ROS) initiated the CSE-mediated EMT processes in ovarian cancer cells. CONCLUSION: Characterization of the p110 isotypes of PI3K is critical for regulating cancer metastasis; LIMK1 could be a common therapeutic target of ovarian cancer metastasis.


Asunto(s)
Quinasas Lim/metabolismo , Neoplasias Ováricas/secundario , Fosfatidilinositol 3-Quinasas/metabolismo , Fumar/efectos adversos , Quinasas p21 Activadas/metabolismo , Quinasas Asociadas a rho/metabolismo , Western Blotting , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Citocinas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Humanos , Quinasas Lim/antagonistas & inhibidores , Quinasas Lim/genética , Metástasis de la Neoplasia , Neoplasias Ováricas/etiología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/genética , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta1/metabolismo , Células Tumorales Cultivadas
20.
Int J Oncol ; 50(5): 1711-1720, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28339079

RESUMEN

Epstein-Barr virus (EBV) infection is closely related to carcinogenesis of various cancers, and is also associated with the development of drug resistance in cancer stem cells. However, in EBV-positive cancer cells, the mechanistic details of the downstream signaling and the connection of PI3K with the NF-κB pathway for development of drug resistance remain controversial. Diffuse large B-cell lymphoma (DLBCL) and multiple myeloma (MM) cells infected by EBV display drug resistance-related proteins (MDR1, MRP1 and MRP2) and stem cell markers (OCT4 and SOX2). EBV-infected HT (HT/EBV) and H929 (H929/EBV) cells activated p110δ expression, but downregulated the expression of p110α and p110ß. A combination of CAL-101, a p110δ-specific inhibitor, with bortezomib treatment of HT/EBV cells synergistically suppressed proliferation, reduced levels of drug resistance-related proteins, activated caspase cleavage and recovered expression of p110α/p110ß. Additionally, co-treatment with CAL-101 and bortezomib attenuated the expression of OCT4 and SOX2 via inhibition of activated NF-κB. Co-treatment with CAL-101 and bortezomib also attenuated drug resistance and NF-κB activity of EBV-infected H929 cells. Our results provide supportive evidence for the clinical application of CAL-101 and bortezomib to treat EBV-infected hematologic cancer.


Asunto(s)
Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Mieloma Múltiple/tratamiento farmacológico , FN-kappa B/genética , Fosfatidilinositol 3-Quinasas/genética , Bortezomib/administración & dosificación , Carcinogénesis/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica , Herpesvirus Humano 4/efectos de los fármacos , Humanos , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/patología , Linfoma de Células B Grandes Difuso/virología , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Mieloma Múltiple/virología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Factor 3 de Transcripción de Unión a Octámeros/biosíntesis , Fosfatidilinositol 3-Quinasas/biosíntesis , Purinas/administración & dosificación , Quinazolinonas/administración & dosificación , Factores de Transcripción SOXB1/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA