2013年1月26日星期六

Exposure to Extremely Low-Frequency Electromagnetic Fields Modulates Na(+) Currents in Rat Cerebellar Granule Cells through Increase of AA/PGE(2) and EP Receptor-Mediated cAMP/PKA Pathway.

Exposure to Extremely Low-Frequency Electromagnetic Fields Modulates Na(+) Currents in Rat Cerebellar Granule Cells through Increase of AA/PGE(2) and EP Receptor-Mediated cAMP/PKA Pathway.

PLoS One. 2013;8(1):e54376

Authors: He YL, Liu DD, Fang YJ, Zhan XQ, Yao JJ, Mei YA

Abstract
Although the modulation of Ca(2+) channel activity by extremely low-frequency electromagnetic fields (ELF-EMF) has been studied previously, few reports have addressed the effects of such fields on the activity of voltage-activated Na(+) channels (Na(v)). Here, we investigated the effects of ELF-EMF on Na(v) activity in rat cerebellar granule cells (GCs). Our results reveal that exposing cerebellar GCs to ELF-EMF for 10-60 min significantly increased Na(v) currents (I(Na)) by 30-125% in a time- and intensity-dependent manner. The Na(v) channel steady-state activation curve, but not the steady-state inactivation curve, was significantly shifted (by 5.2 mV) towards hyperpolarization by ELF-EMF stimulation. This phenomenon is similar to the effect of intracellular application of arachidonic acid (AA) and prostaglandin E(2) (PGE(2)) on I(Na) in cerebellar GCs. Increases in intracellular AA, PGE(2) and phosphorylated PKA levels in cerebellar GCs were observed following ELF-EMF exposure. Western blottings indicated that the Na(V) 1.2 protein on the cerebellar GCs membrane was increased, the total expression levels of Na(V) 1.2 protein were not affected after exposure to ELF-EMF. Cyclooxygenase inhibitors and PGE(2) receptor (EP) antagonists were able to eliminate this ELF-EMF-induced increase in phosphorylated PKA and I(Na). In addition, ELF-EMF exposure significantly enhanced the activity of PLA(2) in cerebellar GCs but did not affect COX-1 or COX-2 activity. Together, these data demonstrate for the first time that neuronal I(Na) is significantly increased by ELF-EMF exposure via a cPLA2 AA PGE(2) EP receptors PKA signaling pathway.

PMID: 23349866 [PubMed - in process]

coxinhibitors c-met inhibitors zm-447439

The Blocking of c-Met Signaling Induces Apoptosis through the Increase of p53 Protein in Lung Cancer.

The Blocking of c-Met Signaling Induces Apoptosis through the Increase of p53 Protein in Lung Cancer.

Cancer Res Treat. 2012 Dec;44(4):251-61

Authors: Jung HY, Joo HJ, Park JK, Kim YH

Abstract
PURPOSE: c-Met is an attractive potential target for novel therapeutic inhibition of human cancer, and c-Met tyrosine kinase inhibitors (TKIs) are effective growth inhibitors of various malignancies. However, their mechanisms in anticancer effects are not clear. In the present study, we investigated the possibility that blocking c-Met signaling induces p53-mediated growth inhibition in lung cancer.
MATERIALS AND METHODS: The growth inhibitory effects of c-Met TKI (SU11274) on lung cancer cells and a xenograft model were assessed using the MTT assay, flow cytometry, and terminal deoxyribonucleotide transferase-mediated nick-end labeling staining. The role of p53 protein in the sensitivity of c-Met TKI (SU11274) was examined by Western blot analysis and immunohistochemistry.
RESULTS: SU11274 significantly induced apoptosis in A549 cells with wild-type p53, compared with that in Calu-1 cells with null-type p53. SU11274 increased p53 protein by enhancing the stability of p53 protein. Increased p53 protein by SU11274 induced up-regulation of Bax and PUMA expression and down-regulation of Bcl-2 expression, subsequently activating caspase 3. In p53 knock-out and knock-in systems, we confirmed that SU11274 caused apoptosis through the p53-mediated apoptotic pathway. Likewise, in the A549 xenograft model, SU11274 effectively shrank tumor volume and induced apoptosis via increased p53 protein expression. Blocking c-Met signaling increased the level of p53 protein.
CONCLUSION: Our finding suggested that p53 plays an important role in SU11274-induced apoptosis, and p53 status seems to be related to the sensitivity to SU11274 in lung cancer.

PMID: 23341789 [PubMed - in process]

dna-pk coxinhibitors c-met inhibitors

Role of mammalian target of rapamycin inhibitor in the treatment of metastatic epithelioid angiomyolipoma: A case report.

Role of mammalian target of rapamycin inhibitor in the treatment of metastatic epithelioid angiomyolipoma: A case report.

Int J Urol. 2013 Jan 24;

Authors: Kohno J, Matsui Y, Yamasaki T, Shibasaki N, Kamba T, Yoshimura K, Sumiyoshi S, Mikami Y, Ogawa O

Abstract
Epithelioid angiomyolipoma has malignant potential; however, no effective therapy has been established for advanced cases. A 50-year-old woman with a history of right nephrectomy for epithelioid angiomyolipoma was referred to our institution. Computed tomography and magnetic resonance imaging showed multiple tumors in her lung, liver and pelvic cavity. The liver and pelvic tumor specimens obtained by needle biopsy confirmed the diagnosis of epithelioid angiomyolipoma recurrence. The patient was treated with everolimus (10?mg/day). Three months later, pulmonary lesions disappeared; liver and pelvic tumors significantly shrank in size, but the pelvic tumor gradually enlarged again. We carried out surgical resection of the residual liver and pelvic cavity tumors. Although the mammalian target of rapamycin inhibitor seems to be effective for treating epithelioid angiomyolipoma, its long-term effects remain unknown. Thus, aggressive administration of a multidisciplinary treatment including molecular target therapy and surgical resection is required to improve the prognosis of epithelioid angiomyolipoma.

PMID: 23347205 [PubMed - as supplied by publisher]

c-met inhibitors zm-447439 rad001

Enhanced NF?B Activity Impairs Vascular Function through PARP-1, SP-1 and COX2-Dependent Mechanisms in Type 2 Diabetes.

Enhanced NF?B Activity Impairs Vascular Function through PARP-1, SP-1 and COX2-Dependent Mechanisms in Type 2 Diabetes.

Diabetes. 2013 Jan 24;

Authors: Kassan M, Choi SK, Galan M, Bishop A, Umezawa K, Trebak M, Belmadani S, Matrougui K

Abstract
Type 2 diabetes (T2D) is associated with vascular dysfunction. We hypothesized that increased nuclear factor kappa B (NF?B) signaling contributes to vascular dysfunction in T2D. We have treated type 2 diabetic (db(-)/db(-)) and control (db(-)/db(+)) mice with two NF?B inhibitors (DHMEQ, 6mg/kg, twice a week and IKK-NBD peptide, 500 ?g/kg/day) for four weeks. Pressure-induced myogenic tone (MT) was significantly potentiated, while endothelium dependent relaxation (EDR) was impaired in small coronary arterioles (CA) and mesenteric resistance artery (MRA) from diabetic mice compared to control. Interestingly, diabetic mice treated with NF?B inhibitors significantly reduced MT potentiation and improved EDR. Importantly, vascular function was also rescued in db(-)/db(-p50NF?B-/-) and db(-)/db(-PARP-1-/-) double knockout mice compared to db(-)/db(-) mice. Additionally, the acute in vitro down regulation of NF?B-p65 using p65NF?B shRNA lentivirus in arteries from db(-)/db(-) mice also improved vascular function. The NF?B inhibition did not affect blood glucose level and body weight. The RNA levels for Sp-1 and eNOS phosphorylation were decreased, while p65NF?B phosphorylation, cleaved PARP-1 and COX-2 expression were increased in arteries from diabetic mice, which were restored after NF?B inhibition and in db(-)/db(-p50NF?B-/-) and db(-)/db(-PARP-1-/-) mice.In the present study, we provided evidence that enhanced NF?B activity impairs vascular function by PARP-1, Sp-1 and COX-2-dependent mechanisms in male type 2 diabetic mice. Therefore, NF?B could be a potential target to overcome diabetes-induced vascular dysfunction.

PMID: 23349490 [PubMed - as supplied by publisher]

ecdysone chir-258 dovitinib

2013年1月25日星期五

Disposition and metabolism of 14C-dovitinib (TKI258), an inhibitor of FGFR and VEGFR, after oral administration in patients with advanced solid tumors.

Related Articles

Disposition and metabolism of 14C-dovitinib (TKI258), an inhibitor of FGFR and VEGFR, after oral administration in patients with advanced solid tumors.

Cancer Chemother Pharmacol. 2012 Nov;70(5):653-63

Authors: Dubbelman AC, Upthagrove A, Beijnen JH, Marchetti S, Tan E, Krone K, Anand S, Schellens JH

Abstract
PURPOSE: This study investigated the metabolism and excretion of dovitinib (TKI258), a tyrosine kinase inhibitor that inhibits fibroblast, vascular endothelial, and platelet-derived growth factor receptors, in patients with advanced solid tumors.
METHODS: Four patients (cohort 1) received a single 500 mg oral dose of (14)C-dovitinib, followed by the collection of blood, urine, and feces for ?10�days. Radioactivity concentrations were measured by liquid scintillation counting and plasma concentrations of dovitinib by liquid chromatography-tandem mass spectrometry. Both techniques were applied for metabolite profiling and identification. A continuous-dosing extension phase (nonlabeled dovitinib 400�mg daily) was conducted with the 3 patients from cohort 1 and 9 additional patients from cohort 2.
RESULTS: The majority of radioactivity was recovered in feces (mean 61�%; range 52-69�%), as compared with urine (mean 16�%; range 13-21�%). Only 6-19�% of the radioactivity was recovered in feces as unchanged dovitinib, suggesting high oral absorption. (14)C-dovitinib was eliminated predominantly via oxidative metabolism, with prominent primary biotransformations including hydroxylation on the fluorobenzyl ring and N-oxidation and carbon oxidation on the methylpiperazine moiety. Dovitinib was the most prominent radioactive component in plasma. The high apparent volume of distribution (2,160 L) may indicate that dovitinib distributes extensively to tissues. Adverse events were predominantly mild to moderate, and most common events included nausea, vomiting, constipation, diarrhea, and fatigue.
CONCLUSIONS: Dovitinib was well absorbed, extensively distributed, and eliminated mainly by oxidative metabolism, followed by excretion, predominantly in feces. The adverse events were as expected for this class of drug.

PMID: 23010851 [PubMed - indexed for MEDLINE]

zm-447439 rad001 ecdysone

Warburg effect and translocation-induced genomic instability: two yeast models for cancer cells.

Warburg effect and translocation-induced genomic instability: two yeast models for cancer cells.

Front Oncol. 2012;2:212

Authors: Tosato V, Gr�ning NM, Breitenbach M, Arnak R, Ralser M, Bruschi CV

Abstract
Yeast has been established as an efficient model system to study biological principles underpinning human health. In this review we focus on yeast models covering two aspects of cancer formation and progression (i) the activity of pyruvate kinase (PK), which recapitulates metabolic features of cancer cells, including the Warburg effect, and (ii) chromosome bridge-induced translocation (BIT) mimiking genome instability in cancer. Saccharomyces cerevisiae is an excellent model to study cancer cell metabolism, as exponentially growing yeast cells exhibit many metabolic similarities with rapidly proliferating cancer cells. The metabolic reconfiguration includes an increase in glucose uptake and fermentation, at the expense of respiration and oxidative phosphorylation (the Warburg effect), and involves a broad reconfiguration of nucleotide and amino acid metabolism. Both in yeast and humans, the regulation of this process seems to have a central player, PK, which is up-regulated in cancer, and to occur mostly on a post-transcriptional and post-translational basis. Furthermore, BIT allows to generate selectable translocation-derived recombinants ("translocants"), between any two desired chromosomal locations, in wild-type yeast strains transformed with a linear DNA cassette carrying a selectable marker flanked by two DNA sequences homologous to different chromosomes. Using the BIT system, targeted non-reciprocal translocations in mitosis are easily inducible. An extensive collection of different yeast translocants exhibiting genome instability and aberrant phenotypes similar to cancer cells has been produced and subjected to analysis. In this review, we hence provide an overview upon two yeast cancer models, and extrapolate general principles for mimicking human disease mechanisms in yeast.

PMID: 23346549 [PubMed - in process]

dna-pk coxinhibitors c-met inhibitors

Optimizing megakaryocyte polyploidization by targeting multiple pathways of cytokinesis.

Related Articles

Optimizing megakaryocyte polyploidization by targeting multiple pathways of cytokinesis.

Transfusion. 2012 Nov;52(11):2406-13

Authors: Avanzi MP, Chen A, He W, Mitchell WB

Abstract
BACKGROUND: Large-scale in vitro production of platelets (PLTs) from cord blood stem cells is one goal of stem cell research. One step toward this goal will be to produce polyploid megakaryocytes capable of releasing high numbers of PLTs. Megakaryocyte polyploidization requires distinct cytoskeletal and cellular mechanisms, including actin polymerization, myosin activation, microtubule formation, and increased DNA production. In this study we variably combined inhibition of these principal mechanisms of cytokinesis with the goal of driving polyploidization in megakaryocytes.
STUDY DESIGN AND METHODS: Megakaryocytes were derived from umbilical cord blood and cultured with reagents that inhibit distinct mechanisms of cytokinesis: Rho-Rock inhibitor (RRI), Src inhibitor (SI), nicotinamide (NIC), aurora B inhibitor (ABI), and myosin light chain kinase inhibitor (MLCKI). Combinations of reagents were used to determine their interactions and to maximize megakaryocyte ploidy.
RESULTS: Treatment with RRI, NIC, SI, and ABI, but not with MLCKI, increased the final ploidy and RRI was the most effective single reagent. RRI and MLCKI, both inhibitors of MLC activation, resulted in opposite ploidy outcomes. Combinations of reagents also increased ploidy and the use of NIC, SI, and ABI was as effective as RRI alone. Addition of MLCKI to NIC, SI, and ABI reached the highest level of polyploidization.
CONCLUSION: Megakaryocyte polyploidization results from modulation of a combination of distinct cytokinesis pathways. Reagents targeting distinct cytoskeletal pathways produced additive effects in final megakaryocyte ploidy. The RRI, however, showed no additive effect but produced a high final ploidy due to overlapping inhibition of multiple cytokinesis pathways.

PMID: 22612069 [PubMed - indexed for MEDLINE]

dna-pk coxinhibitors c-met inhibitors

Combinatorial targeting of FGF and ErbB receptors blocks growth and metastatic spread of breast cancer models.

Combinatorial targeting of FGF and ErbB receptors blocks growth and metastatic spread of breast cancer models.

Breast Cancer Res. 2013 Jan 23;15(1):R8

Authors: Issa A, Gill JW, Heideman MR, Sahin O, Wiemann S, Dey JH, Hynes NE

Abstract
ABSTRACT: INTRODUCTION: Targeting receptor tyrosine kinases (RTKs) with kinase inhibitors is a clinically validated anti-cancer approach, however, the effectiveness of individual inhibitors is often short-lived and resistance emerges. Experimental approaches have revealed numerous feed-back loops in tumor cells and that blocking one signaling pathway, be it the receptor or downstream targets, is often not sufficient to cause tumor regression. Alterations in fibroblast growth factor receptor (FGFR) activity have been implicated in breast cancer. Using breast cancer models with autocrine activation of fibroblast growth factor receptors (FGFR), we have examined the impact of targeting FGFRs in vivo with a selective kinase inhibitor in combination with an inhibitor of PI3K/mTOR or with a pan-ErbB inhibitor. METHODS: The 4T1 or 67NR mammary cancer cells are models for basal-like breast cancer and display constitutive FGFR activity. Upon their injection into fat pads of female Balb/c mice both tumor cell lines form tumors; 4T1 tumors, but not 67NR tumors metastasize to lungs. Tumor growth was measured in mice treated with an FGFR inhibitor (dovitinib/TKI258), a PI3K/ mTOR inhibitor (NVP-BEZ235) or with a pan-ErbB inhibitor (AEE788). Inhibitors were administered individually or in combination and lung metastases were quantified in the 4T1 model. To uncover mechanisms underlying inhibitor activity, tumor lysates were examined by western analyses for signaling activity of FGFR/FRS2, ErbB2, the Erk and the PI3K/Akt/mTOR pathways. Tumor sections were examined for proliferation, apoptosis and vessel density using antibodies for P-Histone H3, cleaved Caspase-3 and CD31, respectively. A transcriptome analysis was carried out on tumors treated with dovitinib for different times to identify pathways upregulated by FGFR inhibition. Anti-phosphotyrosine receptor antibody arrays (P-Tyr RTK) were used to screen in an unbiased manner for active receptors in 4T1 tumors. RESULTS: Treatment of 4T1 and 67NR tumor-bearing mice with the combination of dovitinib + NVP-BEZ235 causes tumor stasis. Western analysis of tumor lysates shows strong down-regulation of the FRS2/Erk and PI3K/Akt/mTOR signaling pathways. Examination of tumor sections revealed that the combination treatment results in a significant decrease in proliferation and high numbers of apoptotic cells, in comparison to tumors treated with individual inhibitors. Using P-Tyr RTK arrays, we identified high levels of P-EGFR and P-ErbB2 in 4T1 tumors. Testing AEE788 in the 4T1 and 67NR models revealed that only the combination of dovitinib + AEE788 resulted in blockade of the PI3K/Akt/mTOR pathway, prolonged tumor stasis and in the 4T1 model, a highly significant decrease in lung metastasis. Analyses of the tumor sections revealed that the combination of dovitinib + AEE788 caused a significant decrease in proliferation and high levels of apoptosis. The results show that in vivo these breast cancer models become dependent upon co-activation of FGFR and ErbB receptors for PI3K pathway activity. CONCLUSIONS: The work presented here shows that in the 4T1 and 67NR breast cancer models the combination of dovitinib + NVP-BEZ235 or dovitinib + AEE788 leads to a strong inhibition of tumor growth and, for the 4T1 model, a block in metastatic spread. Only these combinations strongly down-regulate the FGFR/FRS2/Erk and the PI3K/Akt/mTOR signaling pathways and cause high levels of apoptosis. Interestingly, the decrease in mitosis and increase in apoptosis was consistently stronger in the dovitinib + AEE788 treatment-group suggesting that targeting ErbB receptors has broader downstream effects compared to targeting only PI3K/mTOR. In experiments aimed at testing the durability of treatment-response, this combination was also more effective. Considering that sub-classes of human breast tumors co-express ErbB receptors and FGFRs, these results might have implications for targeted therapy.

PMID: 23343422 [PubMed - as supplied by publisher]

rad001 ecdysone chir-258

Novel molecular targets for the treatment of gastroenteropancreatic endocrine tumors: answers and unsolved problems.

Novel molecular targets for the treatment of gastroenteropancreatic endocrine tumors: answers and unsolved problems.

Int J Mol Sci. 2012;14(1):30-45

Authors: Capurso G, Fendrich V, Rinzivillo M, Panzuto F, Bartsch DK, Delle Fave G

Abstract
As more knowledge on molecular alterations favoring carcinogenesis and spreading of gastroenteropancreatic endocrine tumors has become available, a number of targeted agents interfering with key growth and angiogenic pathways have been explored in preclinical and clinical studies. The mTOR inhibitor Everolimus, and the multi-target antiangiogenetic agent Sunitinib, have been shown to be effective and thus have been approved by the FDA for treatment of pancreatic endocrine tumors. However, there is little data on the primary resistance to targeted agents on these tumors. The goals of the present review are to elucidate the possible advantage of combined treatments in overcoming induced resistances, and to identify biomarkers able to predict clinical efficacy. Moreover, the role of interesting targets for which a strong biological rationale exists, and specific inhibitors are available, such as the Src Family Kinases and the Hedgehog Pathway, are discussed. There is now need for more preclinical studies on cell lines and animal models to provide a stronger preclinical background in this field, as well as clinical trials specifically comparing one targeted therapy with another or combining different targeted agents.

PMID: 23344019 [PubMed - in process]

c-met inhibitors zm-447439 rad001

2013年1月24日星期四

Phosphorylation of 4E-BP1 predicts sensitivity to everolimus in gastric cancer cells.

Phosphorylation of 4E-BP1 predicts sensitivity to everolimus in gastric cancer cells.

Cancer Lett. 2013 Jan 19;

Authors: Nishi T, Iwasaki K, Ohashi N, Tanaka C, Kobayashi D, Nakayama G, Koike M, Fujiwara M, Kobayashi T, Kodera Y

Abstract
We studied the effect of everolimus, an inhibitor of the mammalian target of rapamycin (mTOR) on human gastric cancer cell lines. Cell proliferation in 3 of 8 cell lines was effectively inhibited by everolimus. Basal phosphorylation level of 4E-BP1 (T37/46, T70) was significantly higher in everolimus-sensitive cells than in everolimus-resistant cells. In subcutaneous xenograft model, immunohistochemistry analysis revealed that everolimus-sensitive cells expressed high levels of phospho-4E-BP1 (T37/46). In conclusion, phosphorylation of 4E-BP1 may be a predictive biomarker of everolimus sensitivity in gastric cancer.

PMID: 23340172 [PubMed - as supplied by publisher]

coxinhibitors c-met inhibitors zm-447439

The Blocking of c-Met Signaling Induces Apoptosis through the Increase of p53 Protein in Lung Cancer.

The Blocking of c-Met Signaling Induces Apoptosis through the Increase of p53 Protein in Lung Cancer.

Cancer Res Treat. 2012 Dec;44(4):251-61

Authors: Jung HY, Joo HJ, Park JK, Kim YH

Abstract
PURPOSE: c-Met is an attractive potential target for novel therapeutic inhibition of human cancer, and c-Met tyrosine kinase inhibitors (TKIs) are effective growth inhibitors of various malignancies. However, their mechanisms in anticancer effects are not clear. In the present study, we investigated the possibility that blocking c-Met signaling induces p53-mediated growth inhibition in lung cancer.
MATERIALS AND METHODS: The growth inhibitory effects of c-Met TKI (SU11274) on lung cancer cells and a xenograft model were assessed using the MTT assay, flow cytometry, and terminal deoxyribonucleotide transferase-mediated nick-end labeling staining. The role of p53 protein in the sensitivity of c-Met TKI (SU11274) was examined by Western blot analysis and immunohistochemistry.
RESULTS: SU11274 significantly induced apoptosis in A549 cells with wild-type p53, compared with that in Calu-1 cells with null-type p53. SU11274 increased p53 protein by enhancing the stability of p53 protein. Increased p53 protein by SU11274 induced up-regulation of Bax and PUMA expression and down-regulation of Bcl-2 expression, subsequently activating caspase 3. In p53 knock-out and knock-in systems, we confirmed that SU11274 caused apoptosis through the p53-mediated apoptotic pathway. Likewise, in the A549 xenograft model, SU11274 effectively shrank tumor volume and induced apoptosis via increased p53 protein expression. Blocking c-Met signaling increased the level of p53 protein.
CONCLUSION: Our finding suggested that p53 plays an important role in SU11274-induced apoptosis, and p53 status seems to be related to the sensitivity to SU11274 in lung cancer.

PMID: 23341789 [PubMed - in process]

coxinhibitors c-met inhibitors zm-447439

The Blocking of c-Met Signaling Induces Apoptosis through the Increase of p53 Protein in Lung Cancer.

The Blocking of c-Met Signaling Induces Apoptosis through the Increase of p53 Protein in Lung Cancer.

Cancer Res Treat. 2012 Dec;44(4):251-61

Authors: Jung HY, Joo HJ, Park JK, Kim YH

Abstract
PURPOSE: c-Met is an attractive potential target for novel therapeutic inhibition of human cancer, and c-Met tyrosine kinase inhibitors (TKIs) are effective growth inhibitors of various malignancies. However, their mechanisms in anticancer effects are not clear. In the present study, we investigated the possibility that blocking c-Met signaling induces p53-mediated growth inhibition in lung cancer.
MATERIALS AND METHODS: The growth inhibitory effects of c-Met TKI (SU11274) on lung cancer cells and a xenograft model were assessed using the MTT assay, flow cytometry, and terminal deoxyribonucleotide transferase-mediated nick-end labeling staining. The role of p53 protein in the sensitivity of c-Met TKI (SU11274) was examined by Western blot analysis and immunohistochemistry.
RESULTS: SU11274 significantly induced apoptosis in A549 cells with wild-type p53, compared with that in Calu-1 cells with null-type p53. SU11274 increased p53 protein by enhancing the stability of p53 protein. Increased p53 protein by SU11274 induced up-regulation of Bax and PUMA expression and down-regulation of Bcl-2 expression, subsequently activating caspase 3. In p53 knock-out and knock-in systems, we confirmed that SU11274 caused apoptosis through the p53-mediated apoptotic pathway. Likewise, in the A549 xenograft model, SU11274 effectively shrank tumor volume and induced apoptosis via increased p53 protein expression. Blocking c-Met signaling increased the level of p53 protein.
CONCLUSION: Our finding suggested that p53 plays an important role in SU11274-induced apoptosis, and p53 status seems to be related to the sensitivity to SU11274 in lung cancer.

PMID: 23341789 [PubMed - in process]

dovitinib dna-pk coxinhibitors

Cloning and functional analysis of the ecdysteroid receptor complex in the opossum shrimp Neomysis integer (Leach, 1814).

Cloning and functional analysis of the ecdysteroid receptor complex in the opossum shrimp Neomysis integer (Leach, 1814).

Aquat Toxicol. 2013 Jan 3;130-131C:31-40

Authors: De Wilde R, Swevers L, Soin T, Christiaens O, Roug� P, Cooreman K, Janssen CR, Smagghe G

Abstract
In this paper, the non-target effects of tebufenozide were evaluated on the estuarine crustacean, the opposum shrimp Neomysis integer (Leach, 1814). Tebufenozide is a synthetic non-steroidal ecdysone agonist insecticide and regarded as potential endocrine-disrupting chemical (EDC). N. integer is the most used crustacean in ecotoxicological research in parallel to Daphnia sp. and has been proposed for the regulatory testing of potential EDCs in the US, Europe and Japan. Major results were: (i) cDNAs encoding the ecdysteroid receptor (EcR) and the retinoid-X-receptor (RXR), were cloned and sequenced, and subsequent molecular phylogenetic analysis (maximum likelihood and neighbor-joining) revealed that the amino acid sequence of the ligand binding domain (LBD) of N. integer EcR (NiEcR) clusters as an outgroup of the Crustacea, while NiRXR-LBD clusters in the Malacostracan clade (bootstrap percentage=75%). (ii) 3D-modeling of ligand binding to NiEcR-LBD demonstrated an incompatibility of the insecticide tebufenozide to fit into the NiEcR-ligand binding pocket. This was in great contrast to ponasterone A (PonA) that is the natural molting hormone in Crustacea and for which efficient docking was demonstrated. In addition, the heterodimerization of NiEcR-LBD with the common shrimp Crangon crangon (Linnaeus, 1758) RXR-LBD (CrcRXR-LBD) was also modeled in silico. (iii) With use of insect Hi5 cells, chimeric constructs of NiEcR-LBD and CrcRXR-LBD fused to either the yeast Gal4-DNA binding domain (DBD) or Gal4-activation domain (AD) were cloned into expression plasmids and co-transfected with a Gal4 reporter to quantify the protein-protein interactions of NiEcR-LBD with CrcRXR-LBD. Investigation of the ligand effect of PonA and tebufenozide revealed that only the presence of PonA could induce dimerization of this heterologous receptor complex. (iv) Finally, in an in vivo toxicity assay, N. integer juveniles were exposed to tebufenozide at a concentration of 100?g/L, and no effects against the molting process and nymphal development were scored. In conclusion, the in vitro cell reporter assay, based on NiEcR-LBD/CrcRXR-LBD heterodimerization in Hi5 cells and validated with the natural ecdysteroid hormone PonA, represents a useful tool for the screening of putative EDCs. As a test example for non-steroidal ecdysone agonist insecticides, tebufenozide had no negative effects on NiEcR/RXR receptor dimerization in vitro, nor on the molting process and nymphal development of N. integer at the tested concentration (100?g/L) in vivo.

PMID: 23337090 [PubMed - as supplied by publisher]

coxinhibitors c-met inhibitors zm-447439

Proteomic (antibody microarray) exploration of the molecular mechanism of action of the specific COX-2 inhibitor DuP 697.

Proteomic (antibody microarray) exploration of the molecular mechanism of action of the specific COX-2 inhibitor DuP 697.

Int J Oncol. 2013 Jan 22;

Authors: Agarwal V, Hodgkinson VC, Eagle GL, Scaife L, Lind MJ, Cawkwell L

Abstract
We have previously shown that specific COX-2 inhibitors, including DuP�697, have anti-proliferative effects on mesothelioma cells and potentiate the cytotoxicity of pemetrexed. Here, we used a novel proteomic approach to explore the mechanism of action of this agent. COX-2-positive cell lines MSTO-211H (mesothelioma) and A549 (lung cancer) were exposed to DuP�697 for 72 h. Drug carrier only was added to control cells. Extracted proteins from treated and control cells were analysed using a comparative proteomic platform. Differentially expressed proteins, identified by the Panorama Xpress Profiler725 antibody microarray were submitted to Ingenuity Pathway Analysis. A total of 32 unique differentially expressed proteins were identified with a significant (>1.8-fold) difference in expression between treated and untreated cells in at least one cell line. Five molecules, BCL2L1 (Bcl-xL), BID, CHUK (IKK), FASLG and RAF1, were mapped to the Apoptosis Signaling pathway following Ingenuity Pathway Analysis. BCL2L1 (Bcl-xL) and BID were analysed using immuno-blotting and differential expression was confirmed. Proteomic (antibody microarray) analysis suggests that the mechanism of action of DuP�697 may be exerted via the induction of apoptosis. The antibody microarray platform can be utilised to explore the molecular mechanism of action of novel anticancer agents.

PMID: 23338544 [PubMed - as supplied by publisher]

dna-pk coxinhibitors c-met inhibitors

2013年1月23日星期三

Cytoprotective and anti-inflammatory effects of melatonin in hydrogen peroxide-stimulated CHON-001 human chondrocyte cell line and rabbit model of osteoarthritis via the SIRT1 pathway.

Related Articles

Cytoprotective and anti-inflammatory effects of melatonin in hydrogen peroxide-stimulated CHON-001 human chondrocyte cell line and rabbit model of osteoarthritis via the SIRT1 pathway.

J Pineal Res. 2012 Oct;53(3):225-37

Authors: Lim HD, Kim YS, Ko SH, Yoon IJ, Cho SG, Chun YH, Choi BJ, Kim EC

Abstract
Melatonin has potent antioxidant, analgesic, and antinociceptive properties. However, the effects of melatonin against oxidative stress-induced cytotoxicity and inflammatory mediators in human chondrocytes remain poorly understood. This study examined the effects and underlying mechanism of melatonin in hydrogen peroxide (H(2) O(2) )-stimulated human chondrocytes and rabbit osteoarthritis (OA) model. Melatonin markedly inhibited hydrogen peroxide (H(2) O(2) )-stimulated cytotoxicity, iNOS, and COX-2 protein and mRNA expression, as well as the downstream products, NO and PGE(2) . Incubation of cells with melatonin decreased H(2) O(2) -induced Sirtuin 1 (SIRT1) mRNA and protein expression. SIRT1 inhibition by sirtinol or Sirt1 siRNA reversed the effects of melatonin on H(2) O(2) -mediated induction of pro-inflammatory cytokines (NO, PGE(2) , TNF-?, IL-1?, and IL-8) and the expression of iNOS, COX-2, and cartilage destruction molecules. Melatonin blocked H(2) O(2) -induced phosphorylation of PI3K/Akt, p38, ERK, JNK, and MAPK, as well as activation of NF-?B, which was reversed by sirtinol and SIRT1 siRNA. In rabbit with OA, intra-articular injection of melatonin significantly reduced cartilage degradation, which was reversed by sirtinol. Taken together, this study shows that melatonin exerts cytoprotective and anti-inflammatory effects in an oxidative stress-stimulated chondrocyte model and rabbit OA model, and that the SIRT1 pathway is strongly involved in this effect.

PMID: 22507555 [PubMed - indexed for MEDLINE]

zm-447439 rad001 ecdysone

Proteomic (antibody microarray) exploration of the molecular mechanism of action of the specific COX-2 inhibitor DuP 697.

Proteomic (antibody microarray) exploration of the molecular mechanism of action of the specific COX-2 inhibitor DuP 697.

Int J Oncol. 2013 Jan 22;

Authors: Agarwal V, Hodgkinson VC, Eagle GL, Scaife L, Lind MJ, Cawkwell L

Abstract
We have previously shown that specific COX-2 inhibitors, including DuP�697, have anti-proliferative effects on mesothelioma cells and potentiate the cytotoxicity of pemetrexed. Here, we used a novel proteomic approach to explore the mechanism of action of this agent. COX-2-positive cell lines MSTO-211H (mesothelioma) and A549 (lung cancer) were exposed to DuP�697 for 72 h. Drug carrier only was added to control cells. Extracted proteins from treated and control cells were analysed using a comparative proteomic platform. Differentially expressed proteins, identified by the Panorama Xpress Profiler725 antibody microarray were submitted to Ingenuity Pathway Analysis. A total of 32 unique differentially expressed proteins were identified with a significant (>1.8-fold) difference in expression between treated and untreated cells in at least one cell line. Five molecules, BCL2L1 (Bcl-xL), BID, CHUK (IKK), FASLG and RAF1, were mapped to the Apoptosis Signaling pathway following Ingenuity Pathway Analysis. BCL2L1 (Bcl-xL) and BID were analysed using immuno-blotting and differential expression was confirmed. Proteomic (antibody microarray) analysis suggests that the mechanism of action of DuP�697 may be exerted via the induction of apoptosis. The antibody microarray platform can be utilised to explore the molecular mechanism of action of novel anticancer agents.

PMID: 23338544 [PubMed - as supplied by publisher]

chir-258 dovitinib dna-pk

Mechanism of P-glycoprotein expression in the SGC7901 human gastric adenocarcinoma cell line induced by cyclooxygenase-2.

Related Articles

Mechanism of P-glycoprotein expression in the SGC7901 human gastric adenocarcinoma cell line induced by cyclooxygenase-2.

Asian Pac J Cancer Prev. 2012;13(5):2379-83

Authors: Gu KS, Chen Y

Abstract
OBJECTIVE: To investigate possible signal pathway involvement in multi-drug resistant P-glycoprotein (P-gp) expression induced by cyclooxygenase-2 (COX-2) in a human gastric adenocarcinoma cell line stimulated with pacliaxel (TAX).
METHODS: The effects of TAX on SGC7901 cell growth with different doses was assessed by MTT assay, along with the effects of the COX-2 selective inhibitor NS-398 and the nuclear factor-KB (NF-KB) pathway inhibitor pyrrolidine dithiocarbamate (PDTC). Influence on COX-2, NF-KB p65 and P-gp expression was determined by Western blotting.
RESULTS: TAX, NS-398 and PDTC all reduced SGC7901 growth, with dose- dependence. With increasing dose of TAX, the expression of COX-2, p65 and P-gp showed rising trends, this being reversed by NS-398. PDTC also caused decrease in expression of p65 and P-gp over time.
CONCLUSION: COX-2 may induce the expression of P-gp in SGC7901 cell line via the NF-kappa B pathway with pacliaxel stimulation.

PMID: 22901225 [PubMed - indexed for MEDLINE]

chir-258 dovitinib dna-pk

ALK inhibitors: a new targeted therapy in the treatment of advanced NSCLC.

Related Articles

ALK inhibitors: a new targeted therapy in the treatment of advanced NSCLC.

Target Oncol. 2013 Jan 17;

Authors: Casaluce F, Sgambato A, Maione P, Rossi A, Ferrara C, Napolitano A, Palazzolo G, Ciardiello F, Gridelli C

Abstract
The anaplastic lymphoma kinase (ALK) fusion gene is a key oncogenic driver in a subset of patients with advanced non-small cell lung cancer (NSCLC). Oncogenic fusion genes, including echinoderm microtubule-associated protein-like 4 (EML4) and ALK, have been detected in approximately 2-7�% of NSCLC patients. Fluorescence in situ hybridization (FISH) is the recommended method for detecting ALK gene rearrangement. EML4-ALK fusion genes define a molecular subset of NSCLC with distinct clinical characteristic (lung adenocarcinoma, never or former smoker, usually mutually exclusive with EGFR mutations). Crizotinib (PF-02341066) is an orally bioavailable, ATP-competitive, small molecule inhibitor of both the receptor tyrosine kinases ALK and c-MET (hepatocyte growth factor receptor). Crizotinib has been shown to yield important clinical benefit such as objective response rate, progression-free survival (PFS), and anticipated improvements in quality of life when used in pretreated patients with advanced NSCLC harboring EML4-ALK gene rearrangement. Preliminary phase II data suggested that crizotinib is safe and well tolerated with rapid and robust antitumor activity. A phase III randomized trial in a second-line setting showed response rate and PFS (primary study endpoint) advantage for crizotinib as compared to second-line chemotherapy. Treatment-related adverse events, predominantly restricted to the gastrointestinal and visual systems, are generally self-limiting or easily managed. Crizotinib is a new standard of care for patients with advanced, ALK-positive, NSCLC. In this review, we will discuss the discovery of ALK rearrangements, the clinical epidemiology of lung cancer driven by ALK, the clinical data for ALK-targeted therapy in NSCLC, and ongoing ALK inhibitor-based clinical trials.

PMID: 23325296 [PubMed - as supplied by publisher]

c-met inhibitors zm-447439 rad001

ALK inhibitors: a new targeted therapy in the treatment of advanced NSCLC.

Related Articles

ALK inhibitors: a new targeted therapy in the treatment of advanced NSCLC.

Target Oncol. 2013 Jan 17;

Authors: Casaluce F, Sgambato A, Maione P, Rossi A, Ferrara C, Napolitano A, Palazzolo G, Ciardiello F, Gridelli C

Abstract
The anaplastic lymphoma kinase (ALK) fusion gene is a key oncogenic driver in a subset of patients with advanced non-small cell lung cancer (NSCLC). Oncogenic fusion genes, including echinoderm microtubule-associated protein-like 4 (EML4) and ALK, have been detected in approximately 2-7�% of NSCLC patients. Fluorescence in situ hybridization (FISH) is the recommended method for detecting ALK gene rearrangement. EML4-ALK fusion genes define a molecular subset of NSCLC with distinct clinical characteristic (lung adenocarcinoma, never or former smoker, usually mutually exclusive with EGFR mutations). Crizotinib (PF-02341066) is an orally bioavailable, ATP-competitive, small molecule inhibitor of both the receptor tyrosine kinases ALK and c-MET (hepatocyte growth factor receptor). Crizotinib has been shown to yield important clinical benefit such as objective response rate, progression-free survival (PFS), and anticipated improvements in quality of life when used in pretreated patients with advanced NSCLC harboring EML4-ALK gene rearrangement. Preliminary phase II data suggested that crizotinib is safe and well tolerated with rapid and robust antitumor activity. A phase III randomized trial in a second-line setting showed response rate and PFS (primary study endpoint) advantage for crizotinib as compared to second-line chemotherapy. Treatment-related adverse events, predominantly restricted to the gastrointestinal and visual systems, are generally self-limiting or easily managed. Crizotinib is a new standard of care for patients with advanced, ALK-positive, NSCLC. In this review, we will discuss the discovery of ALK rearrangements, the clinical epidemiology of lung cancer driven by ALK, the clinical data for ALK-targeted therapy in NSCLC, and ongoing ALK inhibitor-based clinical trials.

PMID: 23325296 [PubMed - as supplied by publisher]

dna-pk coxinhibitors c-met inhibitors

2013年1月22日星期二

MET increases the sensitivity of gefitinib-resistant cells to SN-38, an active metabolite of irinotecan, by up-regulating the topoisomerase I activity.

Related Articles

MET increases the sensitivity of gefitinib-resistant cells to SN-38, an active metabolite of irinotecan, by up-regulating the topoisomerase I activity.

J Thorac Oncol. 2012 Sep;7(9):1337-44

Authors: Sakai A, Kasahara K, Ohmori T, Kimura H, Sone T, Fujimura M, Nakao S

Abstract
INTRODUCTION: Most non-small-cell lung cancer tumors with epidermal growth factor receptor mutations are responsive to EGFR tyrosine kinase inhibitors, such as gefitinib and erlotinib, but almost all such tumors ultimately acquire resistance. We previously found that a gefitinib-resistant cell line, PC-9/Met in which MET (MNNG-HOS transforming gene) is amplified, was more sensitive than its parent cell line (PC-9) to 7-ethyl-10-hydroxy-camptothecin (SN-38), an active metabolite of irinotecan. The purpose of this study was to investigate the mechanisms responsible for the increased sensitivity of the gefitinib-resistant cell line to SN-38.
METHODS: The sensitivity of PC-9 and PC-9/Met to SN-38 was assessed by performing water soluble tetrazolium salt (WST-1) assays. Topoisomerase I (topo I) activities were determined for the cell lines cultured in the presence of hepatocyte growth factor and for those of which MET expression was knocked down by introducing a MET-specific small interfering RNA.
RESULTS: PC-9/Met exhibited higher topo I activities, and higher topo I gene and protein expression levels than PC-9 did. Suppression of MET expression by a MET-specific small interfering RNA led to a decrease in the topo I protein expression in the PC-9/Met cells. The stimulation of PC-9 with hepatocyte growth factor caused an increase in the topo I protein level via the activation of MET.
CONCLUSIONS: The increased sensitivity of PC-9/Met cells to SN-38 compared with that of PC-9 cells was partially because of topo I activities resulting from increased topo I mRNA and protein expression caused by MET signaling.

PMID: 22722827 [PubMed - indexed for MEDLINE]

rad001 ecdysone chir-258

ALK inhibitors: a new targeted therapy in the treatment of advanced NSCLC.

Related Articles

ALK inhibitors: a new targeted therapy in the treatment of advanced NSCLC.

Target Oncol. 2013 Jan 17;

Authors: Casaluce F, Sgambato A, Maione P, Rossi A, Ferrara C, Napolitano A, Palazzolo G, Ciardiello F, Gridelli C

Abstract
The anaplastic lymphoma kinase (ALK) fusion gene is a key oncogenic driver in a subset of patients with advanced non-small cell lung cancer (NSCLC). Oncogenic fusion genes, including echinoderm microtubule-associated protein-like 4 (EML4) and ALK, have been detected in approximately 2-7�% of NSCLC patients. Fluorescence in situ hybridization (FISH) is the recommended method for detecting ALK gene rearrangement. EML4-ALK fusion genes define a molecular subset of NSCLC with distinct clinical characteristic (lung adenocarcinoma, never or former smoker, usually mutually exclusive with EGFR mutations). Crizotinib (PF-02341066) is an orally bioavailable, ATP-competitive, small molecule inhibitor of both the receptor tyrosine kinases ALK and c-MET (hepatocyte growth factor receptor). Crizotinib has been shown to yield important clinical benefit such as objective response rate, progression-free survival (PFS), and anticipated improvements in quality of life when used in pretreated patients with advanced NSCLC harboring EML4-ALK gene rearrangement. Preliminary phase II data suggested that crizotinib is safe and well tolerated with rapid and robust antitumor activity. A phase III randomized trial in a second-line setting showed response rate and PFS (primary study endpoint) advantage for crizotinib as compared to second-line chemotherapy. Treatment-related adverse events, predominantly restricted to the gastrointestinal and visual systems, are generally self-limiting or easily managed. Crizotinib is a new standard of care for patients with advanced, ALK-positive, NSCLC. In this review, we will discuss the discovery of ALK rearrangements, the clinical epidemiology of lung cancer driven by ALK, the clinical data for ALK-targeted therapy in NSCLC, and ongoing ALK inhibitor-based clinical trials.

PMID: 23325296 [PubMed - as supplied by publisher]

dna-pk coxinhibitors c-met inhibitors

Disposition and metabolism of 14C-dovitinib (TKI258), an inhibitor of FGFR and VEGFR, after oral administration in patients with advanced solid tumors.

Related Articles

Disposition and metabolism of 14C-dovitinib (TKI258), an inhibitor of FGFR and VEGFR, after oral administration in patients with advanced solid tumors.

Cancer Chemother Pharmacol. 2012 Nov;70(5):653-63

Authors: Dubbelman AC, Upthagrove A, Beijnen JH, Marchetti S, Tan E, Krone K, Anand S, Schellens JH

Abstract
PURPOSE: This study investigated the metabolism and excretion of dovitinib (TKI258), a tyrosine kinase inhibitor that inhibits fibroblast, vascular endothelial, and platelet-derived growth factor receptors, in patients with advanced solid tumors.
METHODS: Four patients (cohort 1) received a single 500 mg oral dose of (14)C-dovitinib, followed by the collection of blood, urine, and feces for ?10�days. Radioactivity concentrations were measured by liquid scintillation counting and plasma concentrations of dovitinib by liquid chromatography-tandem mass spectrometry. Both techniques were applied for metabolite profiling and identification. A continuous-dosing extension phase (nonlabeled dovitinib 400�mg daily) was conducted with the 3 patients from cohort 1 and 9 additional patients from cohort 2.
RESULTS: The majority of radioactivity was recovered in feces (mean 61�%; range 52-69�%), as compared with urine (mean 16�%; range 13-21�%). Only 6-19�% of the radioactivity was recovered in feces as unchanged dovitinib, suggesting high oral absorption. (14)C-dovitinib was eliminated predominantly via oxidative metabolism, with prominent primary biotransformations including hydroxylation on the fluorobenzyl ring and N-oxidation and carbon oxidation on the methylpiperazine moiety. Dovitinib was the most prominent radioactive component in plasma. The high apparent volume of distribution (2,160 L) may indicate that dovitinib distributes extensively to tissues. Adverse events were predominantly mild to moderate, and most common events included nausea, vomiting, constipation, diarrhea, and fatigue.
CONCLUSIONS: Dovitinib was well absorbed, extensively distributed, and eliminated mainly by oxidative metabolism, followed by excretion, predominantly in feces. The adverse events were as expected for this class of drug.

PMID: 23010851 [PubMed - indexed for MEDLINE]

dna-pk coxinhibitors c-met inhibitors

ALK inhibitors: a new targeted therapy in the treatment of advanced NSCLC.

Related Articles

ALK inhibitors: a new targeted therapy in the treatment of advanced NSCLC.

Target Oncol. 2013 Jan 17;

Authors: Casaluce F, Sgambato A, Maione P, Rossi A, Ferrara C, Napolitano A, Palazzolo G, Ciardiello F, Gridelli C

Abstract
The anaplastic lymphoma kinase (ALK) fusion gene is a key oncogenic driver in a subset of patients with advanced non-small cell lung cancer (NSCLC). Oncogenic fusion genes, including echinoderm microtubule-associated protein-like 4 (EML4) and ALK, have been detected in approximately 2-7�% of NSCLC patients. Fluorescence in situ hybridization (FISH) is the recommended method for detecting ALK gene rearrangement. EML4-ALK fusion genes define a molecular subset of NSCLC with distinct clinical characteristic (lung adenocarcinoma, never or former smoker, usually mutually exclusive with EGFR mutations). Crizotinib (PF-02341066) is an orally bioavailable, ATP-competitive, small molecule inhibitor of both the receptor tyrosine kinases ALK and c-MET (hepatocyte growth factor receptor). Crizotinib has been shown to yield important clinical benefit such as objective response rate, progression-free survival (PFS), and anticipated improvements in quality of life when used in pretreated patients with advanced NSCLC harboring EML4-ALK gene rearrangement. Preliminary phase II data suggested that crizotinib is safe and well tolerated with rapid and robust antitumor activity. A phase III randomized trial in a second-line setting showed response rate and PFS (primary study endpoint) advantage for crizotinib as compared to second-line chemotherapy. Treatment-related adverse events, predominantly restricted to the gastrointestinal and visual systems, are generally self-limiting or easily managed. Crizotinib is a new standard of care for patients with advanced, ALK-positive, NSCLC. In this review, we will discuss the discovery of ALK rearrangements, the clinical epidemiology of lung cancer driven by ALK, the clinical data for ALK-targeted therapy in NSCLC, and ongoing ALK inhibitor-based clinical trials.

PMID: 23325296 [PubMed - as supplied by publisher]

ecdysone chir-258 dovitinib

A role of DNA-dependent protein kinase for the activation of AMP-activated protein kinase in response to glucose deprivation.

Related Articles

A role of DNA-dependent protein kinase for the activation of AMP-activated protein kinase in response to glucose deprivation.

Biochim Biophys Acta. 2012 Dec;1823(12):2099-108

Authors: Amatya PN, Kim HB, Park SJ, Youn CK, Hyun JW, Chang IY, Lee JH, You HJ

Abstract
The catalytic subunit of DNA-dependent protein kinase (DNA-PKcs) plays an essential role in double-strand break repair by initially recognizing and binding to DNA breaks. Here, we show that DNA-PKcs interacts with the regulatory ?1 subunit of AMP-activated protein kinase (AMPK), a heterotrimeric enzyme that has been proposed to function as a "fuel gauge" to monitor changes in the energy status of cells and is controlled by the upstream kinases LKB1 and Ca�?/calmodulin-dependent kinase kinase (CaMKK). In co-immunoprecipitation analyses, DNA-PKcs and AMPK?1 interacted physically in DNA-PKcs-proficient M059K cells but not in DNA-PKcs-deficient M059J cells. Glucose deprivation-stimulated phosphorylation of AMPK? on Thr172 and of acetyl-CoA carboxylase (ACC), a downstream target of AMPK, is substantially reduced in M059J cells compared with M059K cells. The inhibition or down-regulation of DNA-PKcs by the DNA-PKcs inhibitors, wortmannin and Nu7441, or by DNA-PKcs siRNA caused a marked reduction in AMPK phosphorylation, AMPK activity, and ACC phosphorylation in response to glucose depletion in M059K, WI38, and IMR90 cells. In addition, DNA-DNA-PKcs(-/-) mouse embryonic fibroblasts (MEFs) exhibited decreased AMPK activation in response to glucose-free conditions. Furthermore, the knockdown of DNA-PKcs led to the suppression of AMPK (Thr172) phosphorylation in LKB1-deficient HeLa cells under glucose deprivation. Taken together, these findings support the positive regulation of AMPK activation by DNA-PKcs under glucose-deprived conditions in mammalian cells.

PMID: 22982065 [PubMed - indexed for MEDLINE]

dovitinib dna-pk coxinhibitors

2013年1月21日星期一

The microRNA miR-7 regulates Tramtrack69 in a developmental switch in Drosophila follicle cells.

Related Articles

The microRNA miR-7 regulates Tramtrack69 in a developmental switch in Drosophila follicle cells.

Development. 2013 Jan 16;

Authors: Huang YC, Smith L, Poulton J, Deng WM

Abstract
Development in multicellular organisms includes both small incremental changes and major switches of cell differentiation and proliferation status. During Drosophila oogenesis, the follicular epithelial cells undergo two major developmental switches that cause global changes in the cell-cycle program. One, the switch from the endoreplication cycle to a gene-amplification phase, during which special genomic regions undergo repeated site-specific replication, is attributed to Notch downregulation, ecdysone signaling activation and upregulation of the zinc-finger protein Tramtrack69 (Ttk69). Here, we report that the microRNA miR-7 exerts an additional layer of regulation in this developmental switch by regulating Ttk69 transcripts. miR-7 recognizes the 3_ UTR of ttk69 transcripts and regulates Ttk69 expression in a dose-dependent manner. Overexpression of miR-7 effectively blocks the switch from the endocycle to gene amplification through its regulation of ttk69. miR-7 and Ttk69 also coordinate other cell differentiation events, such as vitelline membrane protein expression, that lead to the formation of the mature egg. Our studies reveal the important role miR-7 plays in developmental decision-making in association with signal-transduction pathways.

PMID: 23325762 [PubMed - as supplied by publisher]

chir-258 dovitinib dna-pk

Rhynchophylline attenuates LPS-induced pro-inflammatory responses through down-regulation of MAPK/NF-?B signaling pathways in primary microglia.

Related Articles

Rhynchophylline attenuates LPS-induced pro-inflammatory responses through down-regulation of MAPK/NF-?B signaling pathways in primary microglia.

Phytother Res. 2012 Oct;26(10):1528-33

Authors: Song Y, Qu R, Zhu S, Zhang R, Ma S

Abstract
Excessive activation of microglial cells has been implicated in various types of neuroinflammation. Suppression of microglial activation would have therapeutic benefits, leading to the alleviation of the progression of neurodegeneration. In this study, the inhibitory effects of rhynchophylline (RIN), a tetracyclic oxindole alkaloid component isolated from Uncaria rhynchophylla (Miq.) Jacks., on the production of pro-inflammatory mediators were investigated in lipopolysaccharide (LPS)-stimulated microglia. The results showed that RIN markedly reduced the production of nitric oxide (NO), prostaglandins E(2) (PGE(2) ), monocyte chemoattractant protein (MCP-1), tumor necrosis factor-? (TNF-?) and interleukin-1? (IL-1?) in LPS-activated microglia. The mRNA expression levels of iNOS and COX-2 were also depressed by RIN in a concentration-dependent manner. Further studies revealed that RIN blocked I?B? phosphorylation and degradation, inhibited the phosphorylation of mitogen-activated protein kinases (MAPKs). In summary, these data suggest that RIN suppresses inflammatory responses of microglia and may act as a potential therapeutic agent for various neurodegenerative diseases involving neuroinflammation.

PMID: 22322985 [PubMed - indexed for MEDLINE]

coxinhibitors c-met inhibitors zm-447439

Effects of Cardiovascular Drugs on Mortality in Severe COPD: A Time-Dependent Analysis.

Effects of Cardiovascular Drugs on Mortality in Severe COPD: A Time-Dependent Analysis.

Am J Respir Crit Care Med. 2013 Jan 17;

Authors: Ekstr�m MP, Bornefalk Hermansson A, Str�m KE

Abstract
RATIONALE: Cardiovascular drugs may improve survival in Chronic Obstructive Pulmonary Disease (COPD). However, previous studies did not account for major sources of bias and drug effects have not been evaluated in severe COPD. OBJECTIVE: To estimate the time-dependent effects of cardiovascular drugs on survival in oxygen-dependent COPD, accounting for immortal and immeasurable time bias. DESIGN, SETTING AND PATIENTS: Prospective national study of patients starting long-term oxygen therapy (LTOT) for COPD in Sweden between 1 October 2005 and 30 June 2009. Effects on mortality were estimated using extended Cox regression adjusted for age, sex, PaO2, PaCO2, WHO performance status, body mass index, comorbidity and concomitant medications. Immortal and immeasurable time bias was addressed by analyzing all medications as time-dependent variables and accounting for hospitalized time, respectively. MEASUREMENTS: Time-dependent effects of angiotensin-converting enzyme inhibitors or angiotensin receptor blockers (ACEI/ARB), antiplatelet drugs, beta blockers, and statins on all-cause mortality. RESULTS: Of the 2,249 included patients, 1,129 (50%) died under observation. No patient was lost to follow-up. The adjusted time-dependent model was compatible with reduced mortality (hazard ratio (HR); 95% confidence interval) for antiplatelet drugs (HR 0.86; 0.75 to 0.99; P=0.030) and trends for ACEI/ARB (HR 0.90; 0.79 to 1.04; P=0.166) and statins (HR 0.86; 0.72 to 1.03; P=0.105), whereas beta blockers increased mortality (HR 1.19; 1.04 to 1.37; P=0.010). CONCLUSIONS: This study supports that antiplatelet drugs improve survival, whereas beta blockers decrease survival in oxygen-dependent COPD.

PMID: 23328521 [PubMed - as supplied by publisher]

coxinhibitors c-met inhibitors zm-447439

Effects of Cardiovascular Drugs on Mortality in Severe COPD: A Time-Dependent Analysis.

Effects of Cardiovascular Drugs on Mortality in Severe COPD: A Time-Dependent Analysis.

Am J Respir Crit Care Med. 2013 Jan 17;

Authors: Ekstr�m MP, Bornefalk Hermansson A, Str�m KE

Abstract
RATIONALE: Cardiovascular drugs may improve survival in Chronic Obstructive Pulmonary Disease (COPD). However, previous studies did not account for major sources of bias and drug effects have not been evaluated in severe COPD. OBJECTIVE: To estimate the time-dependent effects of cardiovascular drugs on survival in oxygen-dependent COPD, accounting for immortal and immeasurable time bias. DESIGN, SETTING AND PATIENTS: Prospective national study of patients starting long-term oxygen therapy (LTOT) for COPD in Sweden between 1 October 2005 and 30 June 2009. Effects on mortality were estimated using extended Cox regression adjusted for age, sex, PaO2, PaCO2, WHO performance status, body mass index, comorbidity and concomitant medications. Immortal and immeasurable time bias was addressed by analyzing all medications as time-dependent variables and accounting for hospitalized time, respectively. MEASUREMENTS: Time-dependent effects of angiotensin-converting enzyme inhibitors or angiotensin receptor blockers (ACEI/ARB), antiplatelet drugs, beta blockers, and statins on all-cause mortality. RESULTS: Of the 2,249 included patients, 1,129 (50%) died under observation. No patient was lost to follow-up. The adjusted time-dependent model was compatible with reduced mortality (hazard ratio (HR); 95% confidence interval) for antiplatelet drugs (HR 0.86; 0.75 to 0.99; P=0.030) and trends for ACEI/ARB (HR 0.90; 0.79 to 1.04; P=0.166) and statins (HR 0.86; 0.72 to 1.03; P=0.105), whereas beta blockers increased mortality (HR 1.19; 1.04 to 1.37; P=0.010). CONCLUSIONS: This study supports that antiplatelet drugs improve survival, whereas beta blockers decrease survival in oxygen-dependent COPD.

PMID: 23328521 [PubMed - as supplied by publisher]

rad001 ecdysone chir-258

2013年1月20日星期日

ALK inhibitors: a new targeted therapy in the treatment of advanced NSCLC.

Related Articles

ALK inhibitors: a new targeted therapy in the treatment of advanced NSCLC.

Target Oncol. 2013 Jan 17;

Authors: Casaluce F, Sgambato A, Maione P, Rossi A, Ferrara C, Napolitano A, Palazzolo G, Ciardiello F, Gridelli C

Abstract
The anaplastic lymphoma kinase (ALK) fusion gene is a key oncogenic driver in a subset of patients with advanced non-small cell lung cancer (NSCLC). Oncogenic fusion genes, including echinoderm microtubule-associated protein-like 4 (EML4) and ALK, have been detected in approximately 2-7�% of NSCLC patients. Fluorescence in situ hybridization (FISH) is the recommended method for detecting ALK gene rearrangement. EML4-ALK fusion genes define a molecular subset of NSCLC with distinct clinical characteristic (lung adenocarcinoma, never or former smoker, usually mutually exclusive with EGFR mutations). Crizotinib (PF-02341066) is an orally bioavailable, ATP-competitive, small molecule inhibitor of both the receptor tyrosine kinases ALK and c-MET (hepatocyte growth factor receptor). Crizotinib has been shown to yield important clinical benefit such as objective response rate, progression-free survival (PFS), and anticipated improvements in quality of life when used in pretreated patients with advanced NSCLC harboring EML4-ALK gene rearrangement. Preliminary phase II data suggested that crizotinib is safe and well tolerated with rapid and robust antitumor activity. A phase III randomized trial in a second-line setting showed response rate and PFS (primary study endpoint) advantage for crizotinib as compared to second-line chemotherapy. Treatment-related adverse events, predominantly restricted to the gastrointestinal and visual systems, are generally self-limiting or easily managed. Crizotinib is a new standard of care for patients with advanced, ALK-positive, NSCLC. In this review, we will discuss the discovery of ALK rearrangements, the clinical epidemiology of lung cancer driven by ALK, the clinical data for ALK-targeted therapy in NSCLC, and ongoing ALK inhibitor-based clinical trials.

PMID: 23325296 [PubMed - as supplied by publisher]

rad001 ecdysone chir-258

High performance liquid chromatography used for quality control of Achyranthis Radix.

Related Articles

High performance liquid chromatography used for quality control of Achyranthis Radix.

Arch Pharm Res. 2012 Aug;35(8):1449-55

Authors: Zhao BT, Jeong SY, Moon DC, Son KH, Son JK, Woo MH

Abstract
To establish a standard of quality control and to identify reliable Achyranthis Radix, three phytoecdysones including ecdysterone (1), 25R-inokosterone (2) and 25S-inokosterone (3) were determined by quantitative HPLC/UV analysis. Three phytoecdysones were separated with an YMC J'sphere ODS C(18) column (250 mm � 4.6 mm, 4 ?m) by isocratic elution using 0.1% formic acid in water and acetonitrile (85:15, v/v%) as the mobile phase. The flow rate was 1.0 mL/min and the UV detector wavelength was set at 245 nm. The standards were quantified by HPLC/UV from Achyranthes bidentata Blume and Achyranthes japonica Nakai, as well as Cyathula capitata Moq. and Cyathula officinalis Kuan, which are of a different genus but are comparative herbs. The method was successfully used in the analysis of Achyranthis Radix of different geographical origin or genera with relatively simple conditions and procedures, and the assay results were satisfactory for linearity, recovery, precision, accuracy, stability and robustness. The HPLC analytical method for pattern recognition analysis was validated by repeated analysis of eighteen A. bidentata Blume samples and ten A. japonica Nakai samples. The results indicate that the established HPLC/UV method is suitable for quantitation and pattern recognition analyses for quality evaluation of Achyranthis Radix.

PMID: 22941488 [PubMed - indexed for MEDLINE]

dna-pk coxinhibitors c-met inhibitors

MET increases the sensitivity of gefitinib-resistant cells to SN-38, an active metabolite of irinotecan, by up-regulating the topoisomerase I activity.

Related Articles

MET increases the sensitivity of gefitinib-resistant cells to SN-38, an active metabolite of irinotecan, by up-regulating the topoisomerase I activity.

J Thorac Oncol. 2012 Sep;7(9):1337-44

Authors: Sakai A, Kasahara K, Ohmori T, Kimura H, Sone T, Fujimura M, Nakao S

Abstract
INTRODUCTION: Most non-small-cell lung cancer tumors with epidermal growth factor receptor mutations are responsive to EGFR tyrosine kinase inhibitors, such as gefitinib and erlotinib, but almost all such tumors ultimately acquire resistance. We previously found that a gefitinib-resistant cell line, PC-9/Met in which MET (MNNG-HOS transforming gene) is amplified, was more sensitive than its parent cell line (PC-9) to 7-ethyl-10-hydroxy-camptothecin (SN-38), an active metabolite of irinotecan. The purpose of this study was to investigate the mechanisms responsible for the increased sensitivity of the gefitinib-resistant cell line to SN-38.
METHODS: The sensitivity of PC-9 and PC-9/Met to SN-38 was assessed by performing water soluble tetrazolium salt (WST-1) assays. Topoisomerase I (topo I) activities were determined for the cell lines cultured in the presence of hepatocyte growth factor and for those of which MET expression was knocked down by introducing a MET-specific small interfering RNA.
RESULTS: PC-9/Met exhibited higher topo I activities, and higher topo I gene and protein expression levels than PC-9 did. Suppression of MET expression by a MET-specific small interfering RNA led to a decrease in the topo I protein expression in the PC-9/Met cells. The stimulation of PC-9 with hepatocyte growth factor caused an increase in the topo I protein level via the activation of MET.
CONCLUSIONS: The increased sensitivity of PC-9/Met cells to SN-38 compared with that of PC-9 cells was partially because of topo I activities resulting from increased topo I mRNA and protein expression caused by MET signaling.

PMID: 22722827 [PubMed - indexed for MEDLINE]

chir-258 dovitinib dna-pk

A role of DNA-dependent protein kinase for the activation of AMP-activated protein kinase in response to glucose deprivation.

Related Articles

A role of DNA-dependent protein kinase for the activation of AMP-activated protein kinase in response to glucose deprivation.

Biochim Biophys Acta. 2012 Dec;1823(12):2099-108

Authors: Amatya PN, Kim HB, Park SJ, Youn CK, Hyun JW, Chang IY, Lee JH, You HJ

Abstract
The catalytic subunit of DNA-dependent protein kinase (DNA-PKcs) plays an essential role in double-strand break repair by initially recognizing and binding to DNA breaks. Here, we show that DNA-PKcs interacts with the regulatory ?1 subunit of AMP-activated protein kinase (AMPK), a heterotrimeric enzyme that has been proposed to function as a "fuel gauge" to monitor changes in the energy status of cells and is controlled by the upstream kinases LKB1 and Ca�?/calmodulin-dependent kinase kinase (CaMKK). In co-immunoprecipitation analyses, DNA-PKcs and AMPK?1 interacted physically in DNA-PKcs-proficient M059K cells but not in DNA-PKcs-deficient M059J cells. Glucose deprivation-stimulated phosphorylation of AMPK? on Thr172 and of acetyl-CoA carboxylase (ACC), a downstream target of AMPK, is substantially reduced in M059J cells compared with M059K cells. The inhibition or down-regulation of DNA-PKcs by the DNA-PKcs inhibitors, wortmannin and Nu7441, or by DNA-PKcs siRNA caused a marked reduction in AMPK phosphorylation, AMPK activity, and ACC phosphorylation in response to glucose depletion in M059K, WI38, and IMR90 cells. In addition, DNA-DNA-PKcs(-/-) mouse embryonic fibroblasts (MEFs) exhibited decreased AMPK activation in response to glucose-free conditions. Furthermore, the knockdown of DNA-PKcs led to the suppression of AMPK (Thr172) phosphorylation in LKB1-deficient HeLa cells under glucose deprivation. Taken together, these findings support the positive regulation of AMPK activation by DNA-PKcs under glucose-deprived conditions in mammalian cells.

PMID: 22982065 [PubMed - indexed for MEDLINE]

coxinhibitors c-met inhibitors zm-447439

High performance liquid chromatography used for quality control of Achyranthis Radix.

Related Articles

High performance liquid chromatography used for quality control of Achyranthis Radix.

Arch Pharm Res. 2012 Aug;35(8):1449-55

Authors: Zhao BT, Jeong SY, Moon DC, Son KH, Son JK, Woo MH

Abstract
To establish a standard of quality control and to identify reliable Achyranthis Radix, three phytoecdysones including ecdysterone (1), 25R-inokosterone (2) and 25S-inokosterone (3) were determined by quantitative HPLC/UV analysis. Three phytoecdysones were separated with an YMC J'sphere ODS C(18) column (250 mm � 4.6 mm, 4 ?m) by isocratic elution using 0.1% formic acid in water and acetonitrile (85:15, v/v%) as the mobile phase. The flow rate was 1.0 mL/min and the UV detector wavelength was set at 245 nm. The standards were quantified by HPLC/UV from Achyranthes bidentata Blume and Achyranthes japonica Nakai, as well as Cyathula capitata Moq. and Cyathula officinalis Kuan, which are of a different genus but are comparative herbs. The method was successfully used in the analysis of Achyranthis Radix of different geographical origin or genera with relatively simple conditions and procedures, and the assay results were satisfactory for linearity, recovery, precision, accuracy, stability and robustness. The HPLC analytical method for pattern recognition analysis was validated by repeated analysis of eighteen A. bidentata Blume samples and ten A. japonica Nakai samples. The results indicate that the established HPLC/UV method is suitable for quantitation and pattern recognition analyses for quality evaluation of Achyranthis Radix.

PMID: 22941488 [PubMed - indexed for MEDLINE]

c-met inhibitors zm-447439 rad001