Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Exp Clin Cancer Res ; 43(1): 1, 2024 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-38163890

RESUMEN

BACKGROUND: Ceramide metabolism is crucial in the progress of brain metastasis (BM). However, it remains unexplored whether targeting ceramide metabolism may arrest BM. METHODS: RNA sequencing was applied to screen different genes in primary and metastatic foci and whole-exome sequencing (WES) to seek crucial abnormal pathway in BM + and BM-patients. Cellular arrays were applied to analyze the permeability of blood-brain barrier (BBB) and the activation or inhibition of pathway. Database and Co-Immunoprecipitation (Co-IP) assay were adopted to verify the protein-protein interaction. Xenograft and zebrafish model were further employed to verify the cellular results. RESULTS: RNA sequencing and WES reported the involvement of RPTOR and ceramide metabolism in BM progress. RPTOR was significantly upregulated in BM foci and increased the permeability of BBB, while RPTOR deficiency attenuated the cell invasiveness and protected extracellular matrix. Exogenous RPTOR boosted the SPHK2/S1P/STAT3 cascades by binding YY1, in which YY1 bound to the regions of SPHK2 promoter (at -353 ~ -365 nt), further promoting the expression of SPHK2. The latter was rescued by YY1 RNAi. Xenograft and zebrafish model showed that RPTOR blockade suppressed BM of non-small cell lung cancer (NSCLC) and impaired the SPHK2/S1P/STAT3 pathway. CONCLUSION: RPTOR is a key driver gene in the brain metastasis of lung cancer, which signifies that RPTOR blockade may serve as a promising therapeutic candidate for clinical application.


Asunto(s)
Neoplasias Encefálicas , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Animales , Humanos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Pez Cebra , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Ceramidas/uso terapéutico , Proteína Reguladora Asociada a mTOR , Factor de Transcripción YY1/genética
2.
Immunol Res ; 70(4): 530-536, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35585420

RESUMEN

Retinoic acid (RA) plays a role in the mounting immune response and controls several functions of the human body, including cholesterol homeostasis. The synthesis, uptake, and efflux of cellular cholesterol are significantly linked to the mammalian target of rapamycin complex-1 (mTORC1). Activation of mTORC1 promotes the synthesis and uptake of the cholesterol and suppresses its efflux, thus causing accumulation of cellular cholesterol. It is intriguing to know the effect of a high dose of RA on cholesterol accumulation in macrophages (mφ) and whether it is via mTOR activation. It is important to note that the long-term treatment of RA in humans is safe. Therefore, we chose a high dose of RA to observe its effect, which may be implicated in diseases like visceral leishmaniasis, where cholesterol deficiency is established. In the present study, we found the increased expression of RAPTOR, a regulatory component of the mTORC1 complex, in mφ upon treatment with RA. We observed the increased expression of SREBP2, LDLR, and PCSK9 in RA-treated mφ under sufficient cholesterol conditions, which further increased cellular cholesterol levels. Notably, their expressions were decreased when the mTOR pathway was inhibited by rapamycin. However, treatment with rapamycin did not result in the loss of cellular cholesterol in RA-treated mφ. Comparison with rapamycin-treated mφ suggests that RA induces cellular cholesterol levels in a mTORC1-independent manner.


Asunto(s)
Proproteína Convertasa 9 , Tretinoina , Colesterol , Humanos , Proproteína Convertasa 9/metabolismo , Proteína Reguladora Asociada a mTOR/metabolismo , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Tretinoina/metabolismo , Tretinoina/farmacología
3.
J Ethnopharmacol ; 278: 114322, 2021 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-34118343

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Duan Teng Yimu decoction is a Chinese herbal medicine compound with proven therapeutic effects on inflammasome-related diseases, such as rheumatoid arthritis. This decoction consists of three Chinese herbal medicines, including Leonurus japonicus (L. japonicus), which promotes the blood circulation and exhibits detumescence activity, traditionally curing gynecologic and inflammasome diseases. AIM OF THE STUDY: To explore the anti-inflammasome activity and the underlying mechanisms of action of the compounds from L. japonicus. MATERIALS AND METHODS: A series of compounds were isolated from L. japonicus. Their anti-inflammasome activities were evaluated in macrophages that were co-stimulated by lipopolysaccharide (LPS) and NLRP3 inflammasome inducers. NLRP3 inflammasome formation and apoptosis speck like containing a CARD (ASC) oligomerization were evaluated by immunofluorescent microscopy and Western blot analysis. The regulation of autophagy after treatment of this compound was also evaluated. Lastly, in vivo activity of Leojaponin was analyzed in a mouse acute gouty arthritis model. RESULTS: Here we show that Leojaponin, a diterpenoid compound from L. japonicus, suppressed lactate dehydrogenase and IL-1ß release in Nigericin-stimulated macrophages in a pyroptosis model. Leojaponin inhibits NLRP3 inflammasome activation in both J774A.1 cells and bone marrow-derived macrophages in a dose dependent manner. Moreover, Leojaponin suppressed NLRP3-mediated ASC specks formation and ASC oligomerization. These activities of Leojaponin depend on restoration of autophagy via promoting RAPTOR phosphorylation. Furthermore, Leojaponin ameliorated monosodium urate (MSU)-induced acute gouty arthritis in vivo. CONCLUSION: Our findings suggest that Leojaponin inhibits NLRP3 inflammasome activation through enhancing autophagy via RAPTOR phosphorylation, thereby highlighting Leojaponin as a potent drug for inflammasome-related diseases.


Asunto(s)
Artritis Gotosa/tratamiento farmacológico , Autofagia/efectos de los fármacos , Diterpenos/uso terapéutico , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteína Reguladora Asociada a mTOR/metabolismo , Animales , Artritis Gotosa/metabolismo , Línea Celular , Diterpenos/química , Regulación de la Expresión Génica/efectos de los fármacos , Inflamasomas/genética , Inflamasomas/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , L-Lactato Deshidrogenasa/antagonistas & inhibidores , Leonurus/química , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Estructura Molecular , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Nigericina/toxicidad , Fosforilación , Proteína Reguladora Asociada a mTOR/genética , Regulación hacia Arriba
4.
Iran J Allergy Asthma Immunol ; 19(2): 183-192, 2020 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-32372631

RESUMEN

Mouse model of multiple sclerosis (MS) is used for the inflammatory demyelinating disease. Rapamycin (RAPA) may contribute to the reduction of inflammatory responses to experimental autoimmune encephalomyelitis (EAE). Due to its adverse side effects, identifying new therapeutic agents is important. We investigated the transcriptional effects of evening primrose/hemp seed oil (EP/HS oil) compared to RAPA on the expression of immunological factors genes in spleen cells of EAE mouse models. We firstly induced EAE mice by injection of myelin oligodendrocyte glycoprotein (MOG). Then, the EAE mice treated and untreated with EP/HS oil were evaluated and compared with naïve mice. The spinal cords were examined histologically. The immunological factors including genes expression of the regulatory-associated protein of mammalian target of rapamycin (RAPTOR), regulatory-associated companion of mammalian target of rapamycin (RICTOR), interferon (IFN)-γ, interleukin (IL)-10, signal transducer and activator of transcription factors (STAT3), forkhead box P3 (FOXP3), and IL-17 of splenocytes were evaluated by real time-polymerase chain reaction (RT-PCR). The data showed that EP/HS oil was able to reduce the severity of EAE and inhibited the development of the disease. EP/HS oil treatment significantly inhibited the expression of RAPTOR, IFN-γ, IL-17, and STAT3 genes and promoted the expression of RICTOR, IL-10, and FOXP3 genes. In conclusion, the EP/HS oil is likely to be involved in transcription of factors in favor of EAE improvement as well as participating in remyelination in the EAE spinal cord and that it suggests to be effective in therapeutic approaches for MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental/terapia , Ácidos Linoleicos/uso terapéutico , Aceite de Linaza/uso terapéutico , Esclerosis Múltiple/terapia , Aceites de Plantas/uso terapéutico , Sirolimus/uso terapéutico , Bazo/metabolismo , Ácido gammalinolénico/uso terapéutico , Animales , Cannabis , Citocinas/metabolismo , Modelos Animales de Enfermedad , Quimioterapia Combinada , Femenino , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Glicoproteína Mielina-Oligodendrócito/inmunología , Oenothera biennis , Proteína Reguladora Asociada a mTOR/genética , Proteína Reguladora Asociada a mTOR/metabolismo , Semillas , Bazo/patología
5.
Basic Res Cardiol ; 115(1): 2, 2019 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-31781960

RESUMEN

The success of therapies targeting myocardial reperfusion injury is limited, while the cardioprotective impact of mitigating ischemia-related damage remains less explored. We have recently shown in a pig model that the intravenous administration of a modified atorvastatin preparation during ischemia attenuates the rise of cardiac ischemia injury biomarkers. In the following study, we sought to investigate the mechanisms behind these ischemia-related cardioprotective effects. Ischemia was induced by 90 min total coronary balloon occlusion in pigs fed a normocholesterolemic regime. Fifteen minutes after the onset of ischemia, animals were randomized to receive intravenous atorvastatin preparation (IV-atorva) or vehicle. After ischemia animals were euthanized to assess the effect of IV-atorva treatment on gene and protein levels/activation of senescence-, apoptosis-, and cardioprotective/metabolic-related markers. Proof-of-concept studies were carried out in mice and rats in which treatments or vehicle were administered 15 min after initiation of ischemia induced by permanent coronary ligation. Western-blot analyses revealed that in the ischemic myocardium of IV-atorva-treated pigs, RhoA was inactivated, phosphorylation of p53 and caspase-3 was reduced and AMPK was activated with the consequent regulation of the mTOR/raptor-signaling pathway. IV-atorva-treated rats showed, as compared to vehicle, a significant reduction (60%) in scar size assessed at 1 month by histological staining, and mice studies demonstrated the causal involvement of AMPK activation in IV-atorva mediated cardioprotective effects. We demonstrate in pigs and rodents that prompt intravenous treatment with atorvastatin during ischemia limits cardiac cell death and reduces infarct size through AMPK signaling.


Asunto(s)
Atorvastatina/farmacología , Corazón/efectos de los fármacos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Isquemia Miocárdica/tratamiento farmacológico , Proteínas Quinasas Activadas por AMP/metabolismo , Administración Intravenosa , Animales , Apoptosis/efectos de los fármacos , Atorvastatina/uso terapéutico , Senescencia Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Masculino , Ratones , Isquemia Miocárdica/metabolismo , Infiltración Neutrófila/efectos de los fármacos , Distribución Aleatoria , Ratas , Proteína Reguladora Asociada a mTOR/metabolismo , Simvastatina/farmacología , Simvastatina/uso terapéutico , Porcinos , Serina-Treonina Quinasas TOR/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
6.
Int J Mol Sci ; 20(10)2019 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-31091659

RESUMEN

Dysregulation of cellular energy metabolism is closely linked to cancer development and progression. Calorie or glucose restriction (CR or GR) inhibits energy-dependent pathways, including IGF-1/PI3K/Akt/mTOR, in cancer cells. However, alterations in proton dynamics and reversal of the pH gradient across the cell membrane, which results in intracellular alkalinization and extracellular acidification in cancer tissues, have emerged as important etiopathogenic factors. We measured glucose, lactate, and ATP production after GR, plant-derived CR-mimetic curcumin treatment, and curcumin plus GR in human hepatoma cells. Intracellular pH regulatory effects, in particular, protein-protein interactions within mTOR complex-1 and its structural change, were investigated. Curcumin treatment or GR mildly inhibited Na+/H+ exchanger-1 (NHE1). vATPase, monocarboxylate transporter (MCT)-1, and MCT4 level. Combination treatment with curcumin and GR further enhanced the inhibitory effects on these transporters and proton-extruding enzymes, with intracellular pH reduction. ATP and lactate production decreased according to pH change. Modeling of mTOR protein revealed structural changes upon treatments, and curcumin plus GR decreased binding of Raptor and GßL to mTOR, as well as of Rag A and Rag B to Raptor. Consequently, 4EBP1 phosphorylation was decreased and cell migration and proliferation were inhibited in a pH-dependent manner. Autophagy was increased by curcumin plus GR. In conclusion, curcumin treatment combined with GR may be a useful supportive approach for preventing intracellular alkalinization and cancer progression.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma Hepatocelular/metabolismo , Curcumina/farmacología , Glucosa/deficiencia , Neoplasias Hepáticas/metabolismo , Álcalis/metabolismo , Línea Celular , Proliferación Celular/efectos de los fármacos , Glucosa/metabolismo , Células Hep G2 , Humanos , Transportadores de Ácidos Monocarboxílicos/metabolismo , Proteína Reguladora Asociada a mTOR/metabolismo , ATPasas de Translocación de Protón Vacuolares/metabolismo
7.
J Cell Physiol ; 232(10): 2741-2749, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28218386

RESUMEN

Osteoarthritis (OA) is currently still an irreversible degenerative disease of the articular cartilage. Recent, dextrose (d-glucose) intraarticular injection prolotherapy for OA patients has been reported to benefit the chondrogenic stimulation of damaged cartilage. However, the detailed mechanism of glucose's effect on cartilage repair remains unclear. Chitosan, a naturally derived polysaccharide, has recently been investigated as a surgical or dental dressing to control breeding. Therefore, in this study, glucose was adsorbed to chitosan membranes (CTS-Glc), and the study aimed to investigate whether CTS-Glc complex membranes could regulate the proliferation of human OA chondrocytes and to explore the underlying mechanism. Human OA and SW1353 chondrocytes were used in this study. The experiments involving the transfection of cells used SW1353 chondrocytes. A specific inhibitor and siRNAs were used to investigate the mechanism underlying the CTS-Glc-regulated proliferation of human chondrocytes. We found that CTS-Glc significantly increased the proliferation of both human OA and SW1353 chondrocytes comparable to glucose- or chitosan-only stimulation. The role of mammalian target of rapamycin complex 1 (mTORC1) signaling, including mTOR, raptor, and S6k proteins, has been demonstrated in the regulation of CTS-Glc-increased human chondrocyte proliferation. mTORC1 signaling increased the expression levels of maturated SREBP-1 and FASN and then induced the expressions of cell cycle regulators, that is, cyclin D, cyclin-dependent kinase-4 and -6 in human chondrocytes. This study elucidates the detailed mechanism behind the effect of CTS-Glc complex membranes in promoting chondrocyte proliferation and proposes a possible clinical application of the CTS-Glc complex in the dextrose intraarticular injection of OA prolotherapy in the future to attenuate the pain and discomfort of OA patients.


Asunto(s)
Antirreumáticos/farmacología , Proliferación Celular/efectos de los fármacos , Quitosano/farmacología , Condrocitos/efectos de los fármacos , Glucosa/farmacología , Membranas Artificiales , Complejos Multiproteicos/metabolismo , Osteoartritis/tratamiento farmacológico , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adsorción , Anciano , Antirreumáticos/química , Técnicas de Cultivo de Célula , Línea Celular , Quitosano/química , Condrocitos/enzimología , Ciclina D1/metabolismo , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/metabolismo , Acido Graso Sintasa Tipo I/metabolismo , Femenino , Glucosa/química , Humanos , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Persona de Mediana Edad , Complejos Multiproteicos/antagonistas & inhibidores , Complejos Multiproteicos/genética , Osteoartritis/enzimología , Inhibidores de Proteínas Quinasas/farmacología , Interferencia de ARN , Proteína Reguladora Asociada a mTOR , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/genética , Factores de Tiempo , Transfección , Homóloga LST8 de la Proteína Asociada al mTOR
8.
Oncogene ; 36(19): 2737-2749, 2017 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-27991928

RESUMEN

Inhibitors of the mechanistic target of rapamycin (mTOR) are currently used to treat advanced metastatic breast cancer. However, whether an aggressive phenotype is sustained through adaptation or resistance to mTOR inhibition remains unknown. Here, complementary studies in human tumors, cancer models and cell lines reveal transcriptional reprogramming that supports metastasis in response to mTOR inhibition. This cancer feature is driven by EVI1 and SOX9. EVI1 functionally cooperates with and positively regulates SOX9, and promotes the transcriptional upregulation of key mTOR pathway components (REHB and RAPTOR) and of lung metastasis mediators (FSCN1 and SPARC). The expression of EVI1 and SOX9 is associated with stem cell-like and metastasis signatures, and their depletion impairs the metastatic potential of breast cancer cells. These results establish the mechanistic link between resistance to mTOR inhibition and cancer metastatic potential, thus enhancing our understanding of mTOR targeting failure.


Asunto(s)
Neoplasias de la Mama/genética , Proteínas de Unión al ADN/genética , Neoplasias Pulmonares/genética , Proto-Oncogenes/genética , Factor de Transcripción SOX9/genética , Serina-Treonina Quinasas TOR/genética , Factores de Transcripción/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Adulto , Anciano , Neoplasias de la Mama/patología , Proteínas Portadoras/genética , Proliferación Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Células MCF-7 , Proteína del Locus del Complejo MDS1 y EV11 , Proteínas de Microfilamentos/genética , Persona de Mediana Edad , Metástasis de la Neoplasia , Osteonectina/genética , Proteína Reguladora Asociada a mTOR , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Oncotarget ; 7(19): 27819-37, 2016 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-27056897

RESUMEN

The aberrant energy homeostasis that characterized by high rate of energy production (glycolysis) and energy consumption (mRNA translation) is associated with the development of cancer. As mammalian target of rapamycin (mTOR) is a critical regulator of aberrant energy homeostasis, it is an attractive target for anti-tumor intervention. The flavonoid compound Icariside II (IS) is a natural mTOR inhibitor derived from Epimedium. Koreanum. Herein, we evaluate the effect of IS on aberrant energy homeostasis. The reduction of glycolysis and mRNA translation in U2OS (osteosarcoma), S180 (fibrosarcoma) and SW1535 (chondrosarcoma) cells observed in our study, indicate that, IS inhibits aberrant energy homeostasis. This inhibition is found to be due to suppression of mammalian target of rapamycin complex 1 (mTORC1)-eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1) axis through blocking the assembly of mTORC1. Furthermore, IS inhibits the cap-dependent translation of c-myc through mTORC1-4E-BP1 axis which links the relationship between mRNA translation and glycolysis. Inhibition of aberrant energy homeostasis by IS, contributes to its in vitro and in vivo anti-proliferation activity. These data indicate that IS disrupts aberrant energy homeostasis of sarcoma cells through suppression of mTORC1-4E-BP1 axis, providing a novel mechanism of IS to inhibit cell proliferation in sarcoma cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Medicamentos Herbarios Chinos/farmacología , Epimedium/química , Flavonoides/farmacología , Homeostasis/efectos de los fármacos , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Sarcoma/tratamiento farmacológico , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteínas de Ciclo Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Medicamentos Herbarios Chinos/uso terapéutico , Flavonoides/uso terapéutico , Glucólisis/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Endogámicos ICR , Fosfoproteínas/genética , Biosíntesis de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-myc/genética , Interferencia de ARN , ARN Interferente Pequeño , Proteína Asociada al mTOR Insensible a la Rapamicina/genética , Proteína Reguladora Asociada a mTOR/genética , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Asian Pac J Cancer Prev ; 16(15): 6549-56, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26434873

RESUMEN

The PI3K-Akt-mTOR, Wnt/ß-catenin and apoptosis signaling pathways have been shown to be involved in genesis of colorectal cancer (CRC). The aim of this study was to elucidate whether combination of Gelam honey and ginger might have chemopreventive properties in HT29 colon cancer cells by modulating the mTOR, Wnt/ß-catenin and apoptosis signaling pathways. Treatment with Gelam honey and ginger reduced the viability of the HT29 cells dose dependently with IC50 values of 88 mg/ml and 2.15 mg/ml respectively, their while the combined treatment of 2 mg/ml of ginger with 31 mg/ml of Gelam honey inhibited growth of most HT29 cells. Gelam honey, ginger and combination induced apoptosis in a dose dependent manner with the combined treatment exhibiting the highest apoptosis rate. The combined treatment downregulated the gene expressions of Akt, mTOR, Raptor, Rictor, ß-catenin, Gsk3ß, Tcf4 and cyclin D1 while cytochrome C and caspase 3 genes were shown to be upregulated. In conclusion, the combination of Gelam honey and ginger may serve as a potential therapy in the treatment of colorectal cancer through inhibiton of mTOR, Wnt/ß catenin signaling pathways and induction of apoptosis pathway.


Asunto(s)
Anticarcinógenos/farmacología , Miel , Extractos Vegetales/farmacología , Serina-Treonina Quinasas TOR/genética , Vía de Señalización Wnt/efectos de los fármacos , Zingiber officinale , Proteínas Adaptadoras Transductoras de Señales/genética , Apoptosis/efectos de los fármacos , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Proteínas Portadoras/genética , Caspasa 3/genética , Supervivencia Celular/efectos de los fármacos , Quimioprevención , Ciclina D1/genética , Citocromos c/genética , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Células HT29 , Humanos , Concentración 50 Inhibidora , Melaleuca , Proteínas Proto-Oncogénicas c-akt/genética , Proteína Asociada al mTOR Insensible a la Rapamicina , Proteína Reguladora Asociada a mTOR , Serina-Treonina Quinasas TOR/metabolismo , Factor de Transcripción 4 , Factores de Transcripción/genética , Regulación hacia Arriba/efectos de los fármacos , beta Catenina/genética
11.
Asian Pac J Cancer Prev ; 15(3): 1099-104, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24606425

RESUMEN

AIMS AND BACKGROUND: Ginsenoside Rh2, which exerts the potent anticancer action both in vitro and in vivo, is one of the most well characterized ginsenosides extracted from ginseng. Although its effects on cancer are significant, the underlying mechanisms remain unknown. In this study, we sought to elucidate possible links between ginsenoside Rh2 and phosphoglucose isomerase/autocrine motility factor (PGI/AMF). METHODS: KG1α, a leukemia cell line highly expressing PGI/AMF was assessed by western blot analysis and reverse transcription- PCR (RT-PCR) assay after transfection of a small interfering (si)-RNA to silence PGI/AMF. The effect of PGI/ AMF on proliferation was measured by typan blue assay and antibody array. A cell counting kit (CCK)-8 and flow cytometry (FCM) were adopted to investigate the effects of Rh2 on PGI/AMF. The relationships between PGI/AMF and Rh2 associated with Akt, mTOR, Raptor, Rag were detected by western blot analysis. RESULTS: KG1α cells expressed PGI/AMF and its down-regulation significantly inhibited proliferation. The antibody array indicated that the probable mechanism was reduced expression of PARP, State1, SAPK/JNK and Erk1/2, while those of PRAS40 and p38 were up-regulated. Silencing of PGI/AMF enhanced the sensibility of KG1α to Rh2 by suppressing the expression of mTOR, Raptor and Akt. CONCLUSION: These results suggested that ginsenoside Rh2 suppressed the proliferation of KG1α, the same as down-regulation of PGI/AMF. Down-regulation of PGI/ AMF enhanced the pharmacological effects of ginsenoside Rh2 on KG1α by reducing Akt/mTOR signaling.


Asunto(s)
Glucosa-6-Fosfato Isomerasa/genética , Leucemia/tratamiento farmacológico , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Panax/metabolismo , Extractos Vegetales/farmacología , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular , Regulación hacia Abajo , Quinasas MAP Reguladas por Señal Extracelular/biosíntesis , Expresión Génica/efectos de los fármacos , Células HL-60 , Proteínas de Homeodominio/biosíntesis , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/biosíntesis , Poli(ADP-Ribosa) Polimerasas/biosíntesis , Proteínas Proto-Oncogénicas c-akt , Interferencia de ARN , ARN Interferente Pequeño , Proteína Reguladora Asociada a mTOR , Factor de Transcripción STAT1/biosíntesis , Serina-Treonina Quinasas TOR/biosíntesis , Proteínas Quinasas p38 Activadas por Mitógenos/biosíntesis
12.
Zhongguo Zhong Yao Za Zhi ; 35(17): 2318-23, 2010 Sep.
Artículo en Chino | MEDLINE | ID: mdl-21137347

RESUMEN

OBJECTIVE: To investigate the antiproliferation effect of cardamonin (CAR) and its possible mechanisms on human umbilical artery smooth muscle cells (HUASMCs) cultured in the mimicking insulin resistance (IR) medium. METHOD: Proliferation of HUASMCs was assayed by MTT method. The mRNA expression of mTOR, Raptor and Rictor was detected by a real-time PCR. The expression content was calculated by Livak method using internal control of beta-actin. RESULT: The proliferation of HUASMCs cultured in the mimicking IR medium was significantly increased. Both in normal and mimic IR culture medium, cells proliferation was inhibited by CAR (1 x 10(-5), 1 x 10(-4) mol x L(-1)). Pretreated with PD98059 and LY294002, cell proliferation induced by phosphatidic acid (PA) was inhibited, and the mRNA expression of mTOR, Raptor and Rictor was significantly decreased by CAR in the mimic IR medium. CONCLUSION: It is implicated that antiproliferation of CAR is involved in mRNA expression decrease of mTOR and its relative protein Raptor and Rictor.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Portadoras/genética , Proliferación Celular/efectos de los fármacos , Chalconas/farmacología , Inhibidores de Crecimiento/farmacología , Miocitos del Músculo Liso/citología , Serina-Treonina Quinasas TOR/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Portadoras/metabolismo , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteína Asociada al mTOR Insensible a la Rapamicina , Proteína Reguladora Asociada a mTOR , Serina-Treonina Quinasas TOR/metabolismo
13.
Cancer Res ; 69(11): 4577-81, 2009 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-19458076

RESUMEN

The Ras/Raf/MEK/extracellular signal-regulated kinase (ERK) and phosphatidylinositol 3-kinase/Akt/mammalian target of rapamycin (mTOR) signaling pathways are aberrantly activated in many tumors, including highly proliferative glioblastomas, but how they are wired with the cell cycle remains imperfectly understood. Inhibitors of MEK/ERK and mTOR pathways are tested as anticancer agents. They are generally considered to induce a G(1) cell cycle arrest through down-regulation of D-type cyclins and up-regulation of p27(kip1). Here, we examined the effect of targeting mTOR by rapamycin and/or MEK by PD184352 in human glioblastoma cell lines. In combination, these drugs cooperatively and potently inhibited the G(1)-S transition and retinoblastoma protein phosphorylation. Their cooperation could not be explained by their partial and differential inhibitory effects on cyclin D1 or D3 but instead by their synergistic inhibition of the activating T172 phosphorylation of cyclin-dependent kinase (CDK) 4. This appeared independent of p27 and unrelated to weak modulations of the CDK-activating kinase activity. The T172 phosphorylation of CDK4 thus appears as a crucial node integrating the activity of both MEK/ERK and mTOR pathways. Combined inhibition of both pathways should be considered as a promising strategy for treatment of tumors harboring a deregulated CDK4 activity.


Asunto(s)
Benzamidas/farmacología , Proliferación Celular/efectos de los fármacos , Quinasa 4 Dependiente de la Ciclina/metabolismo , Glioblastoma/patología , MAP Quinasa Quinasa 1/antagonistas & inhibidores , Sirolimus/farmacología , Factores de Transcripción/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales , Benzamidas/administración & dosificación , Línea Celular Tumoral , Replicación del ADN/efectos de los fármacos , Combinación de Medicamentos , Evaluación Preclínica de Medicamentos , Glioblastoma/metabolismo , Humanos , MAP Quinasa Quinasa 2/antagonistas & inhibidores , Diana Mecanicista del Complejo 1 de la Rapamicina , Complejos Multiproteicos , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/metabolismo , Proteínas/antagonistas & inhibidores , Proteínas/metabolismo , Proteína Reguladora Asociada a mTOR , Proteína de Retinoblastoma/metabolismo , Sirolimus/administración & dosificación , Serina-Treonina Quinasas TOR , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA