Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 168
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(11)2024 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-38891932

RESUMEN

4-O-Methyl-ascochlorin (MAC), a derivative of the prenyl-phenol antibiotic ascochlorin extracted from the fungus Ascochyta viciae, shows anticarcinogenic effects on various cancer cells. 5-Fluorouracil (5-FU) is used to treat colorectal cancer (CRC); however, its efficacy must be enhanced. In this study, we investigated the molecular mechanisms by which MAC acts synergistically with 5-FU to inhibit cell proliferation and induce apoptosis in CRC cells. MAC enhanced the cytotoxic effects of 5-FU by suppressing the Akt/mTOR/p70S6K and Wnt/ß-catenin signaling pathways. It also reduced the viability of 5-FU-resistant (5-FU-R) cells. Furthermore, expression of anti-apoptosis-related proteins and cancer stem-like cell (CSC) markers by 5-FU-R cells decreased in response to MAC. Similar to MAC, the knockdown of CTNNB1 induced apoptosis and reduced expression of mRNA encoding CRC markers in 5-FU-R cells. In summary, these results suggest that MAC and other ß-catenin modulators may be useful in overcoming the 5-FU resistance of CRC cells.


Asunto(s)
Apoptosis , Proliferación Celular , Neoplasias Colorrectales , Sinergismo Farmacológico , Fluorouracilo , Vía de Señalización Wnt , beta Catenina , Humanos , Fluorouracilo/farmacología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Vía de Señalización Wnt/efectos de los fármacos , Apoptosis/efectos de los fármacos , beta Catenina/metabolismo , beta Catenina/genética , Proliferación Celular/efectos de los fármacos , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo
2.
Int J Mol Sci ; 24(16)2023 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-37628997

RESUMEN

Ubiquitin-specific protease 2 (USP2) is a deubiquitinase belonging to the USPs subfamily. USP2 has been known to display various biological effects including tumorigenesis and inflammation. Therefore, we aimed to examine the sensitization effect of USP2 in TRAIL-mediated apoptosis. The pharmacological inhibitor (ML364) and siRNA targeting USP2 enhanced TNF-related apoptosis-inducing ligand (TRAIL)-induced cancer cell death, but not normal cells. Mechanistically, USP2 interacted with survivin, and ML364 degraded survivin protein expression by increasing the ubiquitination of survivin. Overexpression of survivin or USP2 significantly prevented apoptosis through cotreatment with ML364 and TRAIL, whereas a knockdown of USP2 increased sensitivity to TRAIL. Taken together, our data suggested that ML364 ubiquitylates and degrades survivin, thereby increasing the reactivity to TRAIL-mediated apoptosis in cancer cells.


Asunto(s)
Neoplasias , Ligando Inductor de Apoptosis Relacionado con TNF , Humanos , Regulación hacia Abajo , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Survivin/genética , Muerte Celular , Neoplasias/genética , Ubiquitina Tiolesterasa/genética
3.
Gen Physiol Biophys ; 41(4): 263-274, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35938960

RESUMEN

Platycodin D (PD) is a triterpenoid saponin, a major bioactive constituent of the roots of Platycodon grandiflorum, which is well known for possessing various pharmacological properties. However, the anti-cancer mechanism of PD in bladder cancer cells remains poorly understood. In the current study, we investigated the effect of PD on the growth of human bladder urothelial carcinoma cells. PD treatment significantly reduced the cell survival of bladder cancer cells associated with induction of apoptosis and DNA damage. PD inhibited the expression of inhibitor of apoptosis family members, activated caspases, and induced cleavage of poly (ADP-ribose) polymerase. PD also increased the release of cytochrome c into the cytoplasm by disrupting the mitochondrial membrane potential while upregulating the expression ratio of Bax to Bcl-2. The PD-mediated anti-proliferative effect was significantly inhibited by pre-treatment with a pancaspase inhibitor, but not by an inhibitor of necroptosis. Moreover, PD suppressed the phosphoinositide 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) signaling pathway, and the apoptosis-inducing effect of PD was further enhanced by a PI3K inhibitor. In addition, PD increased the accumulation of reactive oxygen species (ROS), whereas N-acetyl cysteine (NAC), an ROS inhibitor, significantly attenuated the growth inhibition and inactivation of the PI3K/Akt/mTOR signaling caused by PD. Furthermore, NAC significantly suppressed apoptosis, DNA damage, and decreased cell viability induced by PD treatment. Collectively, our findings indicated that PD blocked the growth of bladder urothelial carcinoma cells by inducing ROS-mediated inactivation of the PI3K/Akt/mTOR signaling.


Asunto(s)
Carcinoma de Células Transicionales , Saponinas , Triterpenos , Neoplasias de la Vejiga Urinaria , Apoptosis , Humanos , Fosfatidilinositol 3-Quinasa/metabolismo , Fosfatidilinositol 3-Quinasa/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Saponinas/farmacología , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Triterpenos/farmacología , Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/patología
4.
Int J Mol Sci ; 23(23)2022 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-36499465

RESUMEN

4-O-methylascochlorin (MAC) is a 4-fourth carbon-substituted derivative of ascochlorin, a compound extracted from a phytopathogenic fungus Ascochyta viciae. MAC induces apoptosis and autophagy in various cancer cells, but the effects of MAC on apoptosis and autophagy in cervical cancer cells, as well as how the interaction between apoptosis and autophagy mediates the cellular anticancer effects are not known. Here, we investigated that MAC induced apoptotic cell death of cervical cancer cells without regulating the cell cycle and promoted autophagy by inhibiting the phosphorylation of serine-threonine kinase B (Akt), mammalian target of rapamycin (mTOR), and 70-kDa ribosomal protein S6 kinase (p70S6K). Additional investigations suggested that Bcl-2/adenovirus E1B 19 kDa protein-interacting protein 3 (BNIP-3), but not Hypoxia-inducible factor 1 alpha (HIF-1α), is a key regulator of MAC-induced apoptosis and autophagy. BNIP-3 siRNA suppressed MAC-induced increases in cleaved- poly (ADP-ribose) polymerase (PARP) and LC3II expression. The pan-caspase inhibitor Z-VAD-FMK suppressed MAC-induced cell death and enhanced MAC-induced autophagy. The autophagy inhibitor chloroquine (CQ) enhanced MAC-mediated cell death by increasing BNIP-3 expression. These results indicate that MAC induces apoptosis to promote cell death and stimulates autophagy to promote cell survival by increasing BNIP-3 expression. This study also showed that co-treatment of cells with MAC and CQ further enhanced the death of cervical cancer cells.


Asunto(s)
Neoplasias del Cuello Uterino , Femenino , Humanos , Neoplasias del Cuello Uterino/genética , Línea Celular Tumoral , Autofagia , Apoptosis , Cloroquina/farmacología
5.
J Biochem Mol Toxicol ; 34(10): e22552, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32562591

RESUMEN

A prior study identified that 4-O-methylascochlorin (MAC), a methylated derivative of ascochlorin (ASC) from the fungus Ascochyta viciae, activates autophagy in leukemia cells by suppressing c-Myc phosphorylation. However, the effects of MAC on autophagy in other cancer cells remain unknown. In the present study, we demonstrated that MAC activated autophagy in human glioblastoma. MAC increased expression of autophagy-related proteins, such as LC3-II and Beclin-1. Moreover, MAC stimulated AMP-activated protein kinase (AMPK) phosphorylation and suppressed phosphorylation of the mTOR, p70S6K, and 4EBP1. The well-known AMPK activator metformin increased LC3-II levels, which were augmented by MAC cotreatment. AMPK knockdown decreased LC3-II levels and inhibited MAC activation of autophagy. Furthermore, MAC suppression of c-Myc expression activated autophagy. Treatment with the c-MYC inhibitor, 10058-FA, induced autophagy, as did c-Myc small interfering RNA knockdown. These effects were augmented by MAC cotreatment. Taken together, these findings indicated that MAC induces autophagy in human glioblastoma by activating AMPK signaling and inhibiting c-Myc protein expression in human glioblastoma.


Asunto(s)
Adenilato Quinasa/metabolismo , Autofagia/efectos de los fármacos , Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Proteínas Proto-Oncogénicas c-myc/antagonistas & inhibidores , Terpenos/farmacología , Animales , Beclina-1/metabolismo , Neoplasias Encefálicas/enzimología , Línea Celular Tumoral , Regulación hacia Abajo , Activación Enzimática , Glioblastoma/enzimología , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
6.
J Cell Biochem ; 120(2): 1742-1753, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30324762

RESUMEN

In our previous studies, structurally similar compounds of ascochlorin and ascofuranone exhibited anti-inflammatory activity. Neural inflammation plays a significant role in the commence and advancement of neurodegenerative diseases. It is not known whether 4-O-carboxymethylascochlorin (AS-6) regulates the initial stage of inflammatory responses at the cellular level in BV2 microglia cells. We here investigated the anti-inflammatory effects of AS-6 treatment in microglia cells with the microglial protection in neurons. We found that the lipopolysaccharide (LPS)-stimulated production of nitric oxide, a main regulator of inflammation, is suppressed by AS-6 in BV2 microglial cells. In addition, AS-6 dose-dependently suppressed the increase in COX-2 protein and messenger RNA levels in LPS-stimulated BV2 cells. Moreover, AS-6 inhibited the expression and secretion of proinflammatory cytokines in BV2 microglial cells. At the intracellular level, AS-6 inhibited LPS-activated nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) in BV2 microglial cells. AS-6 negatively affected mitogen-activated protein kinases (MAPK) and Akt phosphorylation: Phosphorylated forms of ERK, JNK, p38, and Akt decreased. To check whether AS-6 protects against inflammatory inducer-mediated neurotoxicity, neuronal SH-SY5Y cells were coincubated with BV2 cells in conditioned medium. AS-6 exerted a neuroprotective effect by suppressing microglial activation by LPS or amyloid-ß peptide. AS-6 is a promising suppressor of inflammatory responses in LPS-induced BV2 cells by attenuating NF-κB and MAPKs signaling. AS-6 protected against microglial-mediated neurotoxicity in SH-SY5Y and BV2 cocultured cells from LPS-induced neuroinflammation and death via inhibiting MAPK, NF-κB, and Akt pathways.

7.
Biosci Biotechnol Biochem ; 83(12): 2244-2248, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31392931

RESUMEN

Hypoxia-inducible factor-1 (HIF-1) is a transcription factor that plays essential roles in human diseases including cancer. The synthetic ascochlorin derivative 4-O-methylascochlorin stabilizes HIF-1α protein, and activates its transcriptional activity, resulting to induce gene expression of its downstream targets such as VEGF and GLUT-1. Here, we quantified protein level of HIF-1α in human osteosarcoma U2OS cells treated with ascochlorin-related compounds and typical HIF-1α stabilizers to characterize properties of HIF-1α stabilization by 4-O-methylascochlorin. Structure-activity relationship studies suggested that the aromatic moiety and hydrophobic substitution of the 4'-hydroxyl group are important for HIF-1α stabilization by ascochlorin-related compounds. 4-O-Methylascochlorin-induced HIF-1α stabilization was suppressed by ascorbic acid and compound C, but not by Fe(II), whereas ascorbic acid only suppressed HIF-1α stabilization by dimethyloxaloylglycine, an analog of the HIF-1 hydroxylase substrate. Fe(II) completely suppressed iron chelator-induced stabilization. These results suggest that ascochlorin-related compounds stabilize HIF-1α in a manner distinct from iron chelating or substrate competition.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/efectos de los fármacos , Quelantes del Hierro/farmacología , Oxigenasas de Función Mixta/metabolismo , Terpenos/farmacología , Unión Competitiva , Línea Celular Tumoral , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Relación Estructura-Actividad , Especificidad por Sustrato , Terpenos/química
8.
Phytother Res ; 33(5): 1384-1393, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30887612

RESUMEN

Evidence suggests that auranofin (AF) exhibits anticancer activity by inhibiting thioredoxin reductase (TrxR). Here, in this study, we have investigated the synergistic effects of AF and morin and their mechanism for the anticancer effects focusing on apoptosis in Hep3B human hepatocellular carcinoma cells. We assessed the anticancer activities by annexin V/PI double staining, caspase, and TrxR activity assay. Morin enhances the inhibitory effects on TrxR activity of AF as well as reducing cell viability. Annexin V/PI double staining revealed that morin/AF cotreatment induced apoptotic cell death. Morin enhances AF-induced mitochondrial membrane potential (ΔΨm) loss and cytochrome c release. Further, morin/AF cotreatment upregulated death receptor DR4/DR5, modulated Bcl-2 family members (upregulation of Bax and downregulation of Bcl-2), and activated caspase-3, -8, and -9. Morin also enhances AF-induced reactive oxygen species (ROS) generation. The anticancer effects results from caspase-dependent apoptosis, which was triggered via extrinsic pathway by upregulating TRAIL receptors (DR4/DR5) and enhanced via intrinsic pathway by modulating Bcl-2 and inhibitor of apoptosis protein family members. These are related to ROS generation. In conclusion, this study provides evidence that morin can enhance the anticancer activity of AF in Hep3B human hepatocellular carcinoma cells, indicating that its combination could be an alternative treatment strategy for the hepatocellular carcinoma.


Asunto(s)
Auranofina/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Flavonoides/farmacología , Neoplasias Hepáticas/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/patología , Caspasas/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Citocromos c/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Humanos , Proteínas Inhibidoras de la Apoptosis/metabolismo , Neoplasias Hepáticas/patología , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
9.
Int J Mol Sci ; 20(15)2019 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-31387245

RESUMEN

Licochalcone A (LCA) is a chalcone that is predominantly found in the root of Glycyrrhiza species, which is widely used as an herbal medicine. Although previous studies have reported that LCA has a wide range of pharmacological effects, evidence for the underlying molecular mechanism of its anti-cancer efficacy is still lacking. In this study, we investigated the anti-proliferative effect of LCA on human bladder cancer cells, and found that LCA induced cell cycle arrest at G2/M phase and apoptotic cell death. Our data showed that LCA inhibited the expression of cyclin A, cyclin B1, and Wee1, but increased the expression of cyclin-dependent kinase (Cdk) inhibitor p21WAF1/CIP1, and increased p21 was bound to Cdc2 and Cdk2. LCA activated caspase-8 and -9, which are involved in the initiation of extrinsic and intrinsic apoptosis pathways, respectively, and also increased caspase-3 activity, a typical effect caspase, subsequently leading to poly (ADP-ribose) polymerase cleavage. Additionally, LCA increased the Bax/Bcl-2 ratio, and reduced the integrity of mitochondria, which contributed to the discharge of cytochrome c from the mitochondria to the cytoplasm. Moreover, LCA enhanced the intracellular levels of reactive oxygen species (ROS); however, the interruption of ROS generation using ROS scavenger led to escape from LCA-mediated G2/M arrest and apoptosis. Collectively, the present data indicate that LCA can inhibit the proliferation of human bladder cancer cells by inducing ROS-dependent G2/M phase arrest and apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Chalconas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Neoplasias de la Vejiga Urinaria/metabolismo , Biomarcadores , Caspasas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo
10.
J Cell Mol Med ; 22(12): 6345-6356, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30338933

RESUMEN

4-O-methylascochlorin (MAC) is a derivative of ascochlorin, a prenyl-phenol compound antibiotic isolated from the fungus Ascochyta viciae. MAC induces caspase/poly (ADP-ribose) polymerase-mediated apoptosis in leukemia cells. However, the effects of MAC on autophagy in cancer cells and the underlying molecular mechanisms remain unknown. Here, we show that MAC induces autophagy in lung cancer cells. MAC significantly induced the expression of autophagy marker proteins including LC3-II, Beclin1, and ATG7. MAC promoted AMP-activated protein kinase (AMPK) phosphorylation and inhibited the phosphorylation of mammalian target of rapamycin (mTOR) and its downstream signalling proteins P70S6K and 4EBP1. The AMPK activator AICAR upregulated LC3-II expression through the AMPK/mTOR pathway similar to the effects of MAC. MAC-induced LC3-II protein expression was slightly reduced in AMPK siRNA transfected cells. MAC upregulated hypoxia-inducible factor-1α (HIF-1α) and BNIP3, which are HIF-1α-dependent autophagic proteins. Treatment with CoCl2 , which mimics hypoxia, induced autophagy similar to the effect of MAC. The HIF-1α inhibitor YC-1 and HIF-1α siRNA inhibited the MAC-induced upregulation of LC3-II and BNIP3. These results suggest that MAC induces autophagy via the AMPK/mTOR signalling pathway and by upregulating HIF-1α and BNIP3 protein expression in lung cancer cells.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas de la Membrana/genética , Proteínas Proto-Oncogénicas/genética , Terpenos/farmacología , Antibacterianos/química , Antibacterianos/farmacología , Apoptosis/efectos de los fármacos , Ascomicetos/química , Autofagia/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Proteínas Asociadas a Microtúbulos/genética , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/genética , Terpenos/química , Activación Transcripcional/efectos de los fármacos
11.
J Cell Biochem ; 119(1): 300-313, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28569433

RESUMEN

Human glioblastomas express higher levels of matrix metalloprotease-2 (MMP-2) than low-grade brain tumors and normal brain tissues. Ascochlorin (ASC) has anti-metastatic, anti-angiogenic, and synergistic effect in various types of cancer cells. However, it remains unknown whether ASC can affect cell migration and invasion in malignant human glioma cells. In this study, we found that ASC indeed inhibits cell migration and invasion in U373MG and A172. ASC significantly suppresses the MMP-2 gelatinolytic activity and expression in U373MG and A172. To determine the molecular mechanism by which ASC suppressed cell migration and invasion, we investigated whether ASC could modulate metastasis via focal adhesion kinase (FAK) and janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling, a potential drug target. ASC strongly inhibits the phosphorylation of FAK, and treatment with a FAK inhibitor significantly suppresses cancer cell migration in the presence of ASC. In addition, ASC significantly decreased phosphorylation of JAK2/STAT3, cancer cell migration and nuclear translocation of STAT3. Taken together, these results suggest that ASC inhibits cell migration and invasion by blocking FAK and JAK/STAT signaling, resulting in reduced MMP-2 activity. J. Cell. Biochem. 119: 300-313, 2018. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Alquenos/farmacología , Movimiento Celular/efectos de los fármacos , Quinasa 1 de Adhesión Focal/metabolismo , Glioblastoma/tratamiento farmacológico , Janus Quinasa 2/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Proteínas de Neoplasias/metabolismo , Fenoles/farmacología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Línea Celular Tumoral , Glioblastoma/enzimología , Glioblastoma/patología , Humanos , Invasividad Neoplásica
12.
J Cell Biochem ; 119(2): 2036-2047, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28833404

RESUMEN

Numerous anti-cancer agents inhibit cell cycle progression via a p53-dependent mechanism; however, other genes such as the proto-oncogene c-Myc are promising targets for anticancer therapy. In the present study, we provide evidence that ascochlorin, an isoprenoid antibiotic, is a non-toxic anti-cancer agent that induces G1 cell cycle arrest and p21WAF1/CIP1 expression by downregulating of c-Myc protein expression. Ascochlorin promoted the G1 arrest, upregulated p53 and p21WAF1/CIP1 , and downregulated c-Myc in HCT116 cells. In p53-deficient cells, ascochlorin enhanced the expression of G1 arrest-related genes except p53. Small interfering RNA (siRNA) mediated c-Myc silencing indicated that the transcriptional repression of c-Myc was related to ascochlorin-mediated modulation of p21WAF1/CIP1 expression. Ascochlorin suppressed the stabilization of the c-Myc protein by inhibiting ERK and P70S6K/4EBP1 phosphorylation, whereas it had no effect on c-Myc degradation mediated by PI3K/Akt/GSK3ß. The ERK inhibitor PD98059 and siRNA-mediated ERK silencing induced G1 arrest and p21WAF1/CIP1 expression by downregulating c-Myc in p53-deficient cells. These results indicated that ascochlorin-induced G1 arrest is associated with the repression of ERK phosphorylation and c-Myc expression. Thus, we reveal a role for ascochlorin in inhibiting tumor growth via G1 arrest, and identify a novel regulatory mechanism for ERK/c-Myc.


Asunto(s)
Alquenos/farmacología , Antibióticos Antineoplásicos/farmacología , Neoplasias Colorrectales/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Fenoles/farmacología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Puntos de Control del Ciclo Celular , Neoplasias Colorrectales/tratamiento farmacológico , Regulación hacia Abajo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HCT116 , Humanos , Fosforilación/efectos de los fármacos , Proto-Oncogenes Mas
13.
J Cell Biochem ; 119(1): 1173-1182, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28708322

RESUMEN

Gangliosides are known to specifically inhibit vascular leukocyte recruitment and consequent interaction with the injured endothelium, the basic inflammatory process. In this study, we have found that the production of nitric oxide (NO), a main regulator of inflammation, is suppressed by GM3 on murine macrophage RAW 264.7 cells, when induced by LPS. In addition, GM3 attenuated the increase in cyclooxyenase-2 (COX-2) protein and mRNA levels in lipopolysaccharide (LPS)-activated RAW 264.7 cells in a dose-dependent manner. Moreover, GM3 inhibited the expression and release of pro-inflammatory cytokines of tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and interleukin-1ß (IL-1ß) in RAW 264.7 macrophages. At the intracellular level, GM3 inhibited LPS-induced nuclear translocation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and activator protein (AP)-1 in RAW 264.7 macrophages. We, therefore, investigated whether GM3 affects mitogen-activated protein kinase (MAPK) phosphorylation, a process known as the upstream signaling regulator. GM3 dramatically reduced the expression levels of the phosphorylated forms of ERK, JNK, and p38 in LPS-activated RAW 264.7 cells. These results indicate that GM3 is a promising suppressor of the vascular inflammatory responses and ganglioside GM3 suppresses the LPS-induced inflammatory response in RAW 264.7 macrophages by suppression of NF-κB, AP-1, and MAPKs signaling. Accordingly, GM3 is suggested as a beneficial agent for the treatment of diseases that are associated with inflammation.


Asunto(s)
Antiinflamatorios/farmacología , Gangliósido G(M3)/farmacología , Lipopolisacáridos/efectos adversos , Macrófagos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Citocinas , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica , Macrófagos/química , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Óxido Nítrico/metabolismo , Fosforilación/efectos de los fármacos , Células RAW 264.7 , Factor de Transcripción AP-1/metabolismo
14.
J Cell Biochem ; 118(12): 4672-4685, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28498494

RESUMEN

Histone deacetylase inhibitors (HDACIs) have emerged as potential anticancer agents for the treatment of solid and hematopoietic cancers. Several HDACIs delay cell growth, induce differentiation, or activate apoptosis in multiple types of tumors, including glioblastomas. In the present study, we showed that the mercaptoacetamide-based HDACI W2 inhibits cell migration and invasion in monomorphic malignant human glioma cells. W2 treatment significantly decreased the activity and expression levels of matrix metalloprotease-2 in malignant A172 cells but not in U373MG cells. Key signaling pathways involved in cell migration and invasion, including PI3K-AKT, ERK-JNK-P38, and FAK/STAT3, were examined to identify the mechanism of action of W2. W2 increased the phosphorylation of AKT and altered cell migration and invasion in an AKT-independent manner. W2 inhibited the phosphorylation of FAK/STAT3, and treatment with a FAK/STAT3 inhibitor significantly suppressed cancer cell migration and MMP-2 activity in the presence of W2. In addition, W2 significantly inhibited the nuclear translocation of phospho-STAT3. Taken together, our results suggest that W2 suppresses cancer cell migration and invasion by inhibiting FAK/STAT3 signaling and STAT3 translocation to the nucleus in monomorphic malignant human glioma cells. J. Cell. Biochem. 118: 4672-4685, 2017. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Quinasa 1 de Adhesión Focal/metabolismo , Glioma/tratamiento farmacológico , Inhibidores de Histona Desacetilasas/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas de Neoplasias/metabolismo , Factor de Transcripción STAT3/metabolismo , Tioacetamida/análogos & derivados , Línea Celular Tumoral , Quinasa 1 de Adhesión Focal/genética , Glioma/genética , Glioma/metabolismo , Glioma/patología , Humanos , Invasividad Neoplásica , Proteínas de Neoplasias/genética , Factor de Transcripción STAT3/genética , Tioacetamida/farmacología
15.
J Cell Biochem ; 117(4): 978-87, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26399466

RESUMEN

A natural compound C23 H32 O4 Cl, ascochlorin (ASC) isolated from an incomplete fungus, Ascochyta viciae has been known to have several biological activities as an antibiotic, antifungal, anti-cancer, anti-hypolipidemic, and anti-hypertension agent. In this study, anti-inflammatory activity has been investigated in lipopolysaccharide (LPS)-induced murine macrophage RAW 264.7 cells, since ASC has not been observed on the inflammatory events. The present study has clearly shown that ASC (1-50 µM) significantly suppressed the production of nitric oxide (NO) and prostaglandin E2 (PGE2 ) and decreased the gene expression of inducible NO synthase (iNOS) and cyclooxygenase-2 (COX-2) in a dose-dependent manner. Moreover, ASC inhibited the mRNA expression and the protein secretion of interleukin (IL)-1ß and IL-6 but not tumor necrosis factor (TNF)-α in LPS-stimulated RAW 264.7 macrophage cells. In addition, ASC suppressed nuclear translocation and DNA binding affinity of nuclear factor-κB (NF-κB). Furthermore, ASC down-regulated phospho-extracellular signal-regulated kinase 1/2 (p-ERK1/2) and p-p38. These results demonstrate that ASC exhibits anti-inflammatory effects in RAW 264.7 macrophage cells.


Asunto(s)
Alquenos/farmacología , Antiinflamatorios no Esteroideos/farmacología , Ciclooxigenasa 2/genética , Lipopolisacáridos/antagonistas & inhibidores , Macrófagos/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Fenoles/farmacología , Transducción de Señal/efectos de los fármacos , Alquenos/aislamiento & purificación , Animales , Antiinflamatorios no Esteroideos/aislamiento & purificación , Línea Celular , Ciclooxigenasa 2/metabolismo , Dinoprostona/antagonistas & inhibidores , Dinoprostona/biosíntesis , Regulación de la Expresión Génica , Lipopolisacáridos/farmacología , Activación de Macrófagos/efectos de los fármacos , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , FN-kappa B/metabolismo , Óxido Nítrico/antagonistas & inhibidores , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Fenoles/aislamiento & purificación , Transporte de Proteínas , Saccharomycetales/química , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
16.
Apoptosis ; 21(5): 657-68, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26922069

RESUMEN

4-O-Methyl-ascochlorin (MAC) is a methylated derivative of the prenyl-phenol antibiotic ascochlorin, which was isolated from an incomplete fungus, Ascochyta viciae. Although the effects of MAC on apoptosis have been reported, the underlying mechanisms remain unknown. Here, we show that MAC promoted apoptotic cell death and downregulated c-Myc expression in K562 human leukemia cells. The effect of MAC on apoptosis was similar to that of 10058-F4 (a c-Myc inhibitor) or c-Myc siRNA, suggesting that the downregulation of c-Myc expression plays a role in the apoptotic effect of MAC. Further investigation showed that MAC downregulated c-Myc by inhibiting protein synthesis. MAC promoted the phosphorylation of AMP-activated protein kinase (AMPK) and inhibited the phosphorylation of mammalian target of rapamycin (mTOR) and its target proteins, including p70S6 K and 4E-BP-1. Treatment of cells with AICAR (an AMPK activator), rapamycin (an mTOR inhibitor), or mTOR siRNA downregulated c-Myc expression and induced apoptosis to a similar extent to that of MAC. These results suggest that the effect of MAC on apoptosis induction in human leukemia cells is mediated by the suppression of c-Myc protein synthesis via an AMPK/mTOR-dependent mechanism.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Antineoplásicos/farmacología , Apoptosis , Proteínas Proto-Oncogénicas c-myc/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Terpenos/farmacología , Línea Celular , Línea Celular Tumoral , Puntos de Control de la Fase G1 del Ciclo Celular , Humanos , Células K562 , Biosíntesis de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Proteínas Proto-Oncogénicas c-myc/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
17.
Glycoconj J ; 33(5): 779-88, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27188588

RESUMEN

In the present study, we isolated pCMAH house-keeping promoter regions (Ph), which are responsible for transcriptional regulation and which are located upstream of the alternative transcript pcmah-2. Luciferase reporter assays using serial construction of each deleted promoter demonstrated that the Ph promoter was highly active in pig-derived kidney PK15. Ph promoter of pcmah lacked a TATA box, but contained three putative Sp1 binding sites. Mutations of these Sp1 binding sites always resulted in the reduction of luciferase activities in Ph-334. In addition, treatment with mithramycin A (25-100 nM) decreased the luciferase activities of the Ph promoters and NeuGc expression in a dose-dependent manner. Electrophoretic mobility shift assay analysis revealed that the probes containing each Sp1 binding site bound to Sp1. Taken together, the results indicate that Sp1 bind to their putative binding sites on the Ph promoter regions of pcmah and positively regulate the promoter activity in pig kidney cells. Interspecies comparison of 5'UTRs and 5'flanking regions shows high homology between pig and cattle, and Sp1 binding sites existing in genomic regions corresponding Ph region are evolutionally conserved.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Genes Esenciales/fisiología , Oxigenasas de Función Mixta/biosíntesis , Ácidos Neuramínicos/metabolismo , Elementos de Respuesta/fisiología , Animales , Línea Celular , Plicamicina/farmacología , Porcinos
18.
Int J Mol Sci ; 17(5)2016 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-27144558

RESUMEN

Gangliosides have been known to play a role in the regulation of apoptosis in cancer cells. This study has employed disialyl-ganglioside GD1b to apoptosis in human breast cancer MCF-7 cells using exogenous treatment of the cells with GD1b and endogenous expression of GD1b in MCF-7 cells. First, apoptosis in MCF-7 cells was observed after treatment of GD1b. Treatment of MCF-7 cells with GD1b reduced cell growth rates in a dose and time dependent manner during GD1b treatment, as determined by XTT assay. Among the various gangliosides, GD1b specifically induced apoptosis of the MCF-7 cells. Flow cytometry and immunofluorescence assays showed that GD1b specifically induces apoptosis in the MCF-7 cells with Annexin V binding for apoptotic actions in early stage and propidium iodide (PI) staining the nucleus of the MCF-7 cells. Treatment of MCF-7 cells with GD1b activated apoptotic molecules such as processed forms of caspase-8, -7 and PARP (Poly(ADP-ribose) polymerase), without any change in the expression of mitochondria-mediated apoptosis molecules such as Bax and Bcl-2. Second, to investigate the effect of endogenously produced GD1b on the regulation of cell function, UDP-gal: ß1,3-galactosyltransferase-2 (GD1b synthase, Gal-T2) gene has been transfected into the MCF-7 cells. Using the GD1b synthase-transfectants, apoptosis-related signal proteins linked to phenotype changes were examined. Similar to the exogenous GD1b treatment, the cell growth of the GD1b synthase gene-transfectants was significantly suppressed compared with the vector-transfectant cell lines and transfection activated the apoptotic molecules such as processed forms of caspase-8, -7 and PARP, but not the levels of expression of Bax and Bcl-2. GD1b-induced apoptosis was blocked by caspase inhibitor, Z-VAD. Therefore, taken together, it was concluded that GD1b could play an important role in the regulation of breast cancer apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Gangliósidos/toxicidad , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Caspasa 7/metabolismo , Caspasa 8/metabolismo , Inhibidores de Caspasas/farmacología , Femenino , Galactosiltransferasas/genética , Galactosiltransferasas/metabolismo , Gangliósidos/biosíntesis , Humanos , Células MCF-7 , Microscopía Fluorescente , Oligopéptidos/farmacología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína X Asociada a bcl-2/metabolismo
19.
Arch Biochem Biophys ; 583: 79-86, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26271443

RESUMEN

Obesity increases the risk of developing many chronic diseases, including type 2 diabetes and certain cancers, and is thereby associated with premature death. The present study was conducted to identify the inhibitory effect of the ascochlorin derivative 4-O-methylascochlorin (MAC) on the differentiation of 3T3-L1 preadipocytes. MAC suppressed the differentiation of 3T3-L1 preadipocytes and inhibited the expression of adipocyte differentiation marker genes, FABP4, PPARγ and C/EBPα. In addition, we found that the inhibitory effects of MAC on differentiation of 3T3-L1 preadipocytes were caused by suppression of mTORC1 via inhibition of mTOR/p70S6K/4E-BP1 phosphorylation and activation of Raptor phosphorylation. MAC also regulated the PPARγ expression and the mTORC1 activation by increasing AMPK phosphorylation and inhibiting PI3K/Akt, which suggest that MAC suppresses the differentiation of 3T3-L1 adipocytes by regulating the AMPK- and PI3K-mTOR-PPARγ signaling pathways. Furthermore, animal model results showed that the phosphorylation of AMPK was enhanced in the liver of C57BL/6 mice intraperitoneally injected with MAC. These results indicate that MAC could be a therapeutic agent for obesity involving PPARγ and AMPK.


Asunto(s)
Adenilato Quinasa/metabolismo , Adipocitos/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , PPAR gamma/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Terpenos/farmacología , Células 3T3-L1 , Adipocitos/citología , Adipocitos/metabolismo , Animales , Ratones , Fosforilación
20.
Mar Drugs ; 13(6): 3936-49, 2015 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-26090845

RESUMEN

For innate immune defense, lower animals such as fish and amphibian are covered with skin mucus, which acts as both a mechanical and biochemical barrier. Although several mucus sources have been isolated and studied for their biochemical and immunological functions, the precise mechanism(s) of action remains unknown. In the present study, we additionally found the eel skin mucus (ESM) to be a promising candidate for use in anti-tumor therapy. Our results showed that the viability of K562 cells was decreased in a dose-dependent manner by treatment with the isolated ESM. The cleaved forms of caspase-9, caspase-3 and poly adenosine diphosphate-ribose polymerase were increased by ESM. The levels of Bax expression and released cytochrome C were also increased after treatment with ESM. Furthermore, during the ESM mediated-apoptosis, phosphorylation levels of ERK1/2 and p38 but not JNK were increased and cell viabilities of the co-treated cells with ESM and inhibitors of ERK 1/2 or p38 were also increased. In addition, treatment with lactose rescued the ESM-mediated decrease in cell viability, indicating lactose-containing glycans in the leukemia cells acted as a counterpart of the ESM for interaction. Taken together, these results suggest that ESM could induce mitochondria-mediated apoptosis through membrane interaction of the K562 human leukemia cells. To the best of our knowledge, this is the first observation that ESM has anti-tumor activity in human cells.


Asunto(s)
Anguilla/metabolismo , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Moco/metabolismo , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/aislamiento & purificación , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Células K562 , Lactosa/metabolismo , Leucemia/tratamiento farmacológico , Leucemia/patología , Mitocondrias/metabolismo , Polisacáridos/metabolismo , Piel/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA