Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Kaohsiung J Med Sci ; 33(11): 543-549, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29050671

ABSTRACT

Gambogic acid (GA) has been shown to inhibit cancer cell proliferation, induce apoptosis, and enhance reactive oxygen species accumulation. However, whether GA could improve multidrug resistance through modulating autophagy has never been explored. We demonstrated that the combination of GA and cisplatin (CDDP) resulted in a stronger growth inhibition effect on A549 and NCI-H460 cells using the MTT assay. Furthermore, treatment with GA significantly increased autophagy in these cells. More importantly, GA-induced cell death could be largely abolished by 3-methyladenine (3-MA) or chloroquine (CQ) treatment, suggesting that GA-induced cell death was dependent on autophagy. Western blot analysis showed that GA treatment suppressed the activation of Akt, mTOR, and S6. In addition, using a GA and rapamycin combination induced more cell death compared to either GA or rapamycin alone. In summary, GA may have utility as an adjunct therapy for non-small cell lung cancer (NSCLC) patients through autophagy-dependent cell death, even when cancer cells have developed resistance to apoptosis.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Autophagy/drug effects , Garcinia/chemistry , Gene Expression Regulation, Neoplastic , Xanthones/pharmacology , A549 Cells , Adenine/analogs & derivatives , Adenine/pharmacology , Antineoplastic Agents, Phytogenic/isolation & purification , Autophagy/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Chloroquine/pharmacology , Cisplatin/pharmacology , Drug Combinations , Drug Synergism , Humans , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Plant Extracts/chemistry , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6/antagonists & inhibitors , Ribosomal Protein S6/genetics , Ribosomal Protein S6/metabolism , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Xanthones/isolation & purification
2.
Toxicol Lett ; 233(2): 102-13, 2015 Mar 04.
Article in English | MEDLINE | ID: mdl-25597859

ABSTRACT

Postnatal isoflurane exposure leads to neurodegeneration and deficits of spatial learning and memory in the adulthood. However, the underlying mechanisms remain unclear. Ribosomal protein S6 is demonstrated to play a pivotal role in control of cell survival, protein synthesis and synaptogenesis for brain development. In this study, the possible role of S6 and its upstream signaling pathways in the developmental neurotoxicity of isoflurane was evaluated using models of primary cultured hippocampal neurons and postnatal day 7 rats. We found that isoflurane decreased IGF-1 level and suppressed activation of IGF-1 receptor, sequentially inhibiting S6 activity via IGF-1/MEK/ERK and IGF-1/PI3K/Akt signaling pathways. S6 inhibition enhanced isoflurane-induced decreased Bcl-xL and increased cleaved caspase-3 and Bad, also reduced PSD95 expression and aggravated deficits of spatial learning and memory. S6 activation could reverse the damages above. These results indicate that S6 inhibition, led by suppression of upstream IGF-1/MEK/ERK and IGF-1/PI3K/Akt signaling pathways, is involved in the neuroapoptosis, synaptogenesis impairment and spatial learning and memory decline caused by postnatal isoflurane exposure. S6 activation may exhibit protective potential against developmental neurotoxicity of isoflurane.


Subject(s)
Anesthetics, Inhalation/toxicity , Brain/growth & development , Isoflurane/toxicity , Neurotoxicity Syndromes/pathology , Ribosomal Protein S6/antagonists & inhibitors , Animals , Apoptosis/drug effects , Hippocampus/cytology , Hippocampus/drug effects , Learning Disabilities/chemically induced , Learning Disabilities/psychology , Maze Learning/drug effects , Memory Disorders/chemically induced , Memory Disorders/psychology , Motor Activity/drug effects , Primary Cell Culture , Rats , Rats, Sprague-Dawley
3.
Eur J Pharm Sci ; 63: 87-95, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25008115

ABSTRACT

The PI3K/Akt/mTOR/S6 ribosomal protein signalling pathway is a key potential target in breast cancer therapy, playing a central role in proliferation and cell survival. In this study, we found that the seleno-compound 2,4-dihydroselenoquinazoline (3a) generally inhibited this signalling axis in MCF-7 breast cancer cells and caused downregulation of S6 ribosomal protein phosphorylation in a dose- and time-dependent manner. Furthermore, 3a caused a dose- and time-dependent decrease in MCF-7 cell viability as well as cell cycle arrest in G2/M. Interestingly 3a also induced apoptosis, as evidenced by cleavage of PARP and caspase-7, and inhibited autophagy, as demonstrated by accumulation of LC3-II and p62/SQSTM1. Given that induction of autophagy has been previously described as a mechanism by which some breast cancer cells counteract proapoptotic signalling and develop resistance to anti-hormone therapy, this suggests that this derivative, which both triggers apoptosis and inhibits autophagy, may be beneficial in preventing and overcoming resistance in breast cancer cells. The data also show the complexity of this signalling axis which is far from being understood.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Organoselenium Compounds/pharmacology , Quinazolines/pharmacology , Ribosomal Protein S6/antagonists & inhibitors , Signal Transduction/drug effects , Cell Cycle/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , MCF-7 Cells , Molecular Structure , Organoselenium Compounds/chemistry , Quinazolines/chemistry , Ribosomal Protein S6/metabolism , Structure-Activity Relationship , Tumor Cells, Cultured
4.
Exp Cell Res ; 326(2): 201-9, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24768699

ABSTRACT

Everolimus is an orally administered mTOR inhibitor. The effect, and mechanism of action, of everolimus on lung cancers with an epidermal growth factor receptor (EGFR) mutation remain unclear. Four gefitinib-sensitive and -resistant cell lines were used in the present work. Growth inhibition was determined using the MTT assay. Transgenic mice carrying the EGFR L858R mutation were treated with everolimus (10 mg/kg/day), or vehicle alone, from 5 to 20 weeks of age, and were then sacrificed. To evaluate the efficacy of everolimus in prolonging survival, everolimus (10 mg/kg/day) or vehicle was administered from 5 weeks of age. The four cell lines were similarly sensitive to everolimus. Expression of phosphorylated (p) mTOR and pS6 were suppressed upon treatment with everolimus in vitro, whereas the pAKT level increased. The numbers of lung tumors with a long axis exceeding 1mm in the everolimus-treated and control groups were 1.9 ± 0.9 and 9.4 ± 3.2 (t-test, p<0.001), respectively. pS6 was suppressed during eve r olimus treatment. Although apoptosis and autophagy were not induced in everolimus-treated EGFR transgenic mice, angiogenesis was suppressed. The median survival time in the everolimus-treated group (58.0 weeks) was significantly longer than that in the control group (31.2 weeks) (logrank test, p<0.001). These findings suggest that everolimus had an indirect effect on tumor formation by inhibiting angiogenesis and might be effective to treat lung tumors induced by an activating EGFR gene mutation.


Subject(s)
ErbB Receptors/genetics , ErbB Receptors/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mutation , Sirolimus/analogs & derivatives , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Everolimus , Gefitinib , Humans , Lung Neoplasms/pathology , Mice , Mice, Transgenic , Mutant Proteins/genetics , Mutant Proteins/metabolism , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Phosphorylation , Quinazolines/pharmacology , Ribosomal Protein S6/antagonists & inhibitors , Ribosomal Protein S6/metabolism , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism
5.
Am J Respir Cell Mol Biol ; 45(5): 1028-35, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21622293

ABSTRACT

Inhibiting hypoxia-inducible factor (HIF)-1α activity has been proposed as a novel therapeutic target in LPS-induced sepsis syndrome. We have reported that tanshinone IIA (TIIA) can reduce LPS-induced lethality and lung injury in mice, but the precise mechanisms have not been fully described. Therefore, the present study investigated whether the protective effect of TIIA was related to the inhibition of LPS-induced HIF-1α expression and what mechanisms accounted for it. This study showed that TIIA pretreatment improved LPS-induced biochemical and cellular changes and reduced the production of inflammatory cytokines. Pretreatment with TIIA decreased LPS-induced HIF-1α expression in vivo and in vitro. TIIA did not affect the LPS-induced HIF-1α mRNA level but inhibited HIF-1α protein translation by the inhibition of the PI3K/AKT and MAPK pathways and related protein translational regulators, such as p70S6K1, S6 ribosomal protein, 4E-BP1, and eIF4E, and promoted HIF-1α protein degradation via the proteasomal pathway in LPS-stimulated macrophages. These observations partially explain the antiinflammatory effects of TIIA, which provides scientific basis for its application for the treatment of acute lung injury/acute respiratory distress syndrome or sepsis.


Subject(s)
Abietanes/pharmacology , Acute Lung Injury/drug therapy , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Endotoxemia/drug therapy , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/antagonists & inhibitors , Cell Cycle Proteins , DNA-Binding Proteins/antagonists & inhibitors , Endotoxemia/chemically induced , Eukaryotic Initiation Factors , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Lipopolysaccharides/toxicity , MAP Kinase Signaling System/drug effects , Macrophages/drug effects , Male , Mice , Mice, Inbred BALB C , Phosphoproteins/antagonists & inhibitors , Proteasome Endopeptidase Complex/drug effects , Protein Biosynthesis/drug effects , Ribosomal Protein S6/antagonists & inhibitors , Ribosomal Protein S6 Kinases, 70-kDa/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors
6.
Amino Acids ; 39(5): 1487-92, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20464435

ABSTRACT

The ribosomal protein S6 is essential for the formation of the subunits of higher eukaryotic ribosomes, and S6 heterozygosity leads to early embryonal lethality in mice. S6 is phosphorylated at clustered residues S235/236 and S240/244 upon numerous physiological and pathological stimuli. So far, the S6Kinases, S6K1 and S6K2 are the only proven S6 S240/244 phosphorylating enzymes in mammalian cells. The activity of these S6Kinases is strictly regulated via the mammalian target of rapamycin (mTOR) enzyme complex with raptor, named mTORC1. In time course experiments with the mTORC1 inhibitor rapamycin we here demonstrate rapamycin-resistant phosphorylation of the ribosomal protein S6 at S240/244. Serum-restimulation experiments further demonstrated that this rapamycin-resistant S6 240/244 phosphorylation is induced via serum factors in a cell cycle-dependent manner. Our data allow new insights into the regulation of S6 phosphorylation and provide evidence for the existence of rapamycin-resistant S6 phosphorylating kinase activities.


Subject(s)
Ribosomal Protein S6/antagonists & inhibitors , Sirolimus/pharmacology , Cell Cycle , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes , Phosphorylation , Proteins/antagonists & inhibitors , Proteins/metabolism , Ribosomal Protein S6/metabolism , Structure-Activity Relationship , TOR Serine-Threonine Kinases
8.
Blood ; 113(26): 6669-80, 2009 Jun 25.
Article in English | MEDLINE | ID: mdl-19401561

ABSTRACT

Detailed genomic studies have shown that cytogenetic abnormalities contribute to multiple myeloma (MM) pathogenesis and disease progression. Nevertheless, little is known about the characteristics of MM at the epigenetic level and specifically how microRNAs regulate MM progression in the context of the bone marrow milieu. Therefore, we performed microRNA expression profiling of bone marrow derived CD138(+) MM cells versus their normal cellular counterparts and validated data by qRT-PCR. We identified a MM-specific microRNA signature characterized by down-expression of microRNA-15a/-16 and overexpression of microRNA-222/-221/-382/-181a/-181b (P < .01). We investigated the functional role of microRNA-15a and -16 and showed that they regulate proliferation and growth of MM cells in vitro and in vivo by inhibiting AKT serine/threonine-protein-kinase (AKT3), ribosomal-protein-S6, MAP-kinases, and NF-kappaB-activator MAP3KIP3. Moreover, miRNA-15a and -16 exerted their anti-MM activity even in the context of the bone marrow milieu in vitro and in vivo. These data indicate that microRNAs play a pivotal role in the biology of MM and represent important targets for novel therapies in MM.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/physiology , Multiple Myeloma/pathology , RNA, Neoplasm/physiology , Angiogenesis Inhibitors/physiology , Animals , Cell Adhesion , Cell Division/physiology , Clinical Trials, Phase II as Topic/statistics & numerical data , Coculture Techniques , Endothelial Cells/cytology , Gene Expression Profiling , Humans , Mice , Mice, SCID , MicroRNAs/biosynthesis , MicroRNAs/genetics , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/metabolism , Prognosis , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Ribosomal Protein S6/antagonists & inhibitors , Ribosomal Protein S6/metabolism , Stromal Cells/cytology
9.
J Biol Chem ; 282(19): 14056-64, 2007 May 11.
Article in English | MEDLINE | ID: mdl-17360704

ABSTRACT

Converging signals from the mammalian target of rapamycin (mTOR) and phosphoinositide 3-kinase (PI3K) pathways are well established to modulate translation initiation. Less is known regarding the molecular basis of protein synthesis regulated by other inputs, such as agonists of the Ras/extracellular signal-regulated kinase (ERK) signaling cascade. Ribosomal protein (rp) S6 is a component of the 40S ribosomal subunit that becomes phosphorylated at several serine residues upon mitogen stimulation, but the exact molecular mechanisms regulating its phosphorylation and the function of phosphorylated rpS6 is poorly understood. Here, we provide evidence that activation of the p90 ribosomal S6 kinases (RSKs) by serum, growth factors, tumor promoting phorbol esters, and oncogenic Ras is required for rpS6 phosphorylation downstream of the Ras/ERK signaling cascade. We demonstrate that while ribosomal S6 kinase 1 (S6K1) phosphorylates rpS6 at all sites, RSK exclusively phosphorylates rpS6 at Ser(235/236) in vitro and in vivo using an mTOR-independent mechanism. Mutation of rpS6 at Ser(235/236) reveals that phosphorylation of these sites promotes its recruitment to the 7-methylguanosine cap complex, suggesting that Ras/ERK signaling regulates assembly of the translation preinitiation complex. These data demonstrate that RSK provides an mTOR-independent pathway linking the Ras/ERK signaling cascade to the translational machinery.


Subject(s)
Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Ribosomal Protein S6/metabolism , Small-Conductance Calcium-Activated Potassium Channels/metabolism , ras Proteins/metabolism , Cells, Cultured , HeLa Cells , Humans , Immunoblotting , Immunoprecipitation , Kidney/metabolism , Luciferases/metabolism , MAP Kinase Kinase Kinases , Mutation , Phosphorylation , Polyribosomes/metabolism , Protein Biosynthesis , RNA, Small Interfering/pharmacology , Ribosomal Protein S6/antagonists & inhibitors , Ribosomal Protein S6/genetics , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Ribosomes/metabolism , Signal Transduction , Small-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Small-Conductance Calcium-Activated Potassium Channels/genetics
10.
Cancer Res ; 65(16): 7462-9, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-16103100

ABSTRACT

Activation of protein kinase Cbeta (PKCbeta) has been repeatedly implicated in tumor-induced angiogenesis. The PKCbeta-selective inhibitor, Enzastaurin (LY317615.HCl), suppresses angiogenesis and was advanced for clinical development based upon this antiangiogenic activity. Activation of PKCbeta has now also been implicated in tumor cell proliferation, apoptosis, and tumor invasiveness. Herein, we show that Enzastaurin has a direct effect on human tumor cells, inducing apoptosis and suppressing the proliferation of cultured tumor cells. Enzastaurin treatment also suppresses the phosphorylation of GSK3betaser9, ribosomal protein S6(S240/244), and AKT(Thr308). Oral dosing with Enzastaurin to yield plasma concentrations similar to those achieved in clinical trials significantly suppresses the growth of human glioblastoma and colon carcinoma xenografts. As in cultured tumor cells, Enzastaurin treatment suppresses the phosphorylation of GSK3beta in these xenograft tumor tissues. Enzastaurin treatment also suppresses GSK3beta phosphorylation to a similar extent in peripheral blood mononuclear cells (PBMCs) from these treated mice. These data show that Enzastaurin has a direct antitumor effect and that Enzastaurin treatment suppresses GSK3beta phosphorylation in both tumor tissue and in PBMCs, suggesting that GSK3beta phosphorylation may serve as a reliable pharmacodynamic marker for Enzastaurin activity. With previously published reports, these data support the notion that Enzastaurin suppresses tumor growth through multiple mechanisms: direct suppression of tumor cell proliferation and the induction of tumor cell death coupled to the indirect effect of suppressing tumor-induced angiogenesis.


Subject(s)
Apoptosis/drug effects , Colonic Neoplasms/drug therapy , Glioblastoma/drug therapy , Indoles/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Animals , Cell Growth Processes/drug effects , Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Female , Glioblastoma/enzymology , Glioblastoma/pathology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , HCT116 Cells , Humans , Male , Mice , Mice, Nude , Phosphorylation/drug effects , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Protein Kinase C beta , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Ribosomal Protein S6/antagonists & inhibitors , Ribosomal Protein S6/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
11.
Biosci Biotechnol Biochem ; 66(11): 2437-43, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12506984

ABSTRACT

An expression library of Arabidopsis thaliana cDNAs was randomly introduced into A. thaliana. The transformant pool was used to obtain a line, c105, with reduced apical dominance and irregular positioning of leaves and flowers. The inserted DNA was a 3'-fragment of the ribosomal protein S6 gene with antisense orientation. The transcriptional level of the ribosomal protein S6 was lower in c105 than in the wild-type plant. Introduction of the same fragment into the wild-type plant gave phenotypes similar to those of c105, so the phenotypes of c105 were due to the S6 antisense expression. The phenotypes suggest selectively reduced function of specific proteins rather than an overall decrease in protein function caused by defective ribosomal biogenesis.


Subject(s)
Arabidopsis Proteins/genetics , Arabidopsis/growth & development , Ribosomal Protein S6/antagonists & inhibitors , Arabidopsis/genetics , Blotting, Northern , DNA Probes/genetics , DNA, Complementary/metabolism , DNA, Plant/genetics , DNA, Plant/metabolism , Flowers/genetics , Flowers/growth & development , Gene Expression Regulation, Plant , Mutation , Phenotype , Plant Leaves/genetics , Plant Leaves/growth & development , Plants, Genetically Modified/metabolism , RNA, Antisense/biosynthesis , RNA, Antisense/genetics , RNA, Ribosomal/biosynthesis , RNA, Ribosomal/genetics , Ribosomal Protein S6/genetics , Ribosomal Protein S6/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...