Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Int J Biol Sci ; 19(3): 772-788, 2023.
Article in English | MEDLINE | ID: mdl-36778128

ABSTRACT

Xanthine dehydrogenase (XDH) is the rate-limiting enzyme in purine catabolism by converting hypoxanthine to xanthine and xanthine to uric acid. The altered expression and activity of XDH are associated with the development and prognosis of multiple types of cancer, while its role in lung adenocarcinoma (LUAD) remains unknown. Herein, we demonstrated that XDH was highly expressed in LUAD and was significantly correlated with poor prognosis. Though inhibition of XDH displayed moderate effect on the viability of LUAD cells cultured in the complete medium, it significantly attenuated the survival of starved cells. Similar results were obtained in XDH-knockout cells. Nucleosides supplementation rescued the survival of starved LUAD cells upon XDH inhibition, while inhibition of purine nucleoside phosphorylase abrogated the process, indicating that nucleoside degradation is required for the XDH-mediated survival of LUAD cells. Accordingly, metabolic flux revealed that ribose derived from nucleoside fueled key carbon metabolic pathways to sustain the survival of starved LUAD cells. Mechanistically, down-regulation of XDH suppressed unfolded protein response (UPR) and autophagic flux in starved LUAD cells. Inhibition of XDH decreased the level of amino acids produced by autophagic degradation, which was accompanied with down-regulation of mTORC1 signaling. Supplementation of amino acids including glutamine or glutamate rescued the survival of starved LUAD cells upon knockout or inhibition of XDH. Finally, XDH inhibitors potentiated the anti-cancer activity of 2-deoxy-D-glucose that induced UPR and/or autophagy in vitro and in vivo. In summary, XDH plays a crucial role in the survival of starved LUAD cells and targeting XDH may improve the efficacy of drugs that induce UPR and autophagy in the therapy of LUAD.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Humans , Xanthine Dehydrogenase/genetics , Xanthine Dehydrogenase/metabolism , Nucleosides/metabolism , Adenocarcinoma of Lung/genetics , Autophagy/genetics , Unfolded Protein Response , Lung Neoplasms/pathology , Xanthines , Nutrients , Amino Acids/metabolism
2.
World J Clin Cases ; 8(2): 362-369, 2020 Jan 26.
Article in English | MEDLINE | ID: mdl-32047786

ABSTRACT

BACKGROUND: Sacrococcygeal hernia is a very rare condition that is usually secondary to sacrococcygectomy, and its ideal treatment regimen is unclear. Herein, we report a case of sacrococcygeal hernia occurring in a patient who had no history of sacrococcygeal operation, present the operative procedures of mesh repair via a combined laparoscopic and sacrococcygeal approach that has not been described, and discuss our experience in diagnosis and treatment with a review of the literature. CASE SUMMARY: A 54-year-old woman who chiefly complained of a 10-year history of a reversible bulge in her right sacrococcygeal region was admitted to our hospital. The physical examination revealed a bulge in the right sacrococcygeal region upon standing, which disappeared in the prone position but relapsed when performing the Valsalva manoeuvre. Computed tomography displayed an abnormality in the structure of the tissues between the midline of the sacrococcygeal region and the right gluteus muscle. The patient was diagnosed with sacrococcygeal hernia and received hernia repair with mesh through a combined laparoscopic and sacrococcygeal approach. On laparoscopy, the rectum was dissected posterolaterally, and a defect was identified in the right anterior sacrococcygeal region through which part of the rectum protruded. This was followed by the placement of a self-gripping polyester mesh via a sacrococcygeal approach. There were no postoperative complications. The patient was discharged on postoperative day 7 and was followed for more than 6 mo with no recurrence. CONCLUSION: Laparoscopic mesh repair is recommended as a priority of surgical options for sacrococcygeal hernias, while choosing a self-gripping mesh can help avoid the risk of presacral vessel injury by reducing suture fixation.

3.
Acta Pharmacol Sin ; 39(8): 1338-1346, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29345254

ABSTRACT

Tamoxifen, an important endocrine therapeutic agent, is widely used for the treatment of estrogen receptor positive (ER+) breast cancer. However, de novo or acquired resistance prevents patients from benefitting from endocrine approaches and necessitates alternative treatments. In this study, we report that small heat protein beta-8 (HSPB8) may serve as an important molecule in tamoxifen resistance. HSPB8 expression is enhanced in MCF-7 cells resistant to tamoxifen (MCF-7/R) compared to parent cells. Moreover, high expression of HSPB8 associates with poor prognosis in ER+ breast cancer patients but not in patients without classification. Stimulating ER signaling by heterogeneous expression of ERa or 17ß-estradiol promotes HSPB8 expression and reduces the cell population in G1 phase. In contrast, blockage of ER signaling by tamoxifen down-regulates the expression of HSPB8. In addition, knocking down HSPB8 by specific siRNAs induces significant cell cycle arrest at G1 phase. AZD8055 was found to be more potent against the proliferation of MCF-7/R cells than that of parent cells, which was associated with down-regulation of HSPB8. We found that the anti-proliferative activity of AZD8055 was positively correlated with the HSPB8 expression level in ER+ breast cancer cells. Thus, AZD8055 was able to overcome tamoxifen resistance in breast cancer cells, and the expression of HSPB8 may predict the efficacy of AZD8055 in ER+ breast cancer. This hypothesis deserves further investigation.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Heat-Shock Proteins/genetics , Morpholines/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/antagonists & inhibitors , Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , Cell Line, Tumor , Down-Regulation , Estrogen Receptor alpha/metabolism , G1 Phase Cell Cycle Checkpoints/drug effects , Heat-Shock Proteins/metabolism , Humans , Molecular Chaperones , Prognosis , Protein Serine-Threonine Kinases/metabolism , Tamoxifen/pharmacology
4.
Theranostics ; 7(4): 974-986, 2017.
Article in English | MEDLINE | ID: mdl-28382169

ABSTRACT

PI3Kα-selective inhibitor BYL719 is currently in phase II/III clinical trial for the treatment of breast cancer, but highly variable response has been observed among patients. We sought to discover predictive biomarker for the efficacy of BYL719 by dissecting the proliferative signaling pathway mediated by PI3K in breast cancer. BYL719 concurrently inhibited the phosphorylation of AKT and ERK in PIK3CA-mutated human breast cancer cells. PI3K-regulated ERK phosphorylation was independent of canonical PDK1/AKT/mTOR pathway, while it was associated with RAF/MEK. Hyper-activation of EGFR or RAS abrogated inhibition of ERK phosphorylation by BYL719. Furthermore, hyper-activation of receptor tyrosine kinases (RTKs) including EGFR, c-MET, FGFR and HER3 but not IGF-1R restored ERK phosphorylation and cell viability suppressed by BYL719, suggesting the discriminative functions of RTKs in cell signaling and proliferation. By profiling 22 breast cancer cell lines, we found that BYL719 was more potent in cell lines where phosphorylation of both AKT and ERK was attenuated than those where only AKT phosphorylation was inhibited. The potency of BYL719 was further found to be significantly correlated with the expression profile of RTKs in breast cancer cells. Specifically, overexpression of EGFR, c-MET and/or FGFR1 forecasted resistance, while overexpression of IGF-1R and/or HER2 predicted sensitivity to BYL719 in breast cancer cells. Similar correlation between BYL719 efficacy and expression profile of RTKs was found in patient-derived xenograft models of breast cancer. Thus, inhibition of ERK phosphorylation by PI3Kα inhibitor BYL719 contributes to its antitumor efficacy and is determined by the converged signaling from RTKs. The expression profile of RTKs in breast cancer tissue could be potentially developed as a predictive biomarker for the efficacy of PI3Kα inhibitors.


Subject(s)
Breast Neoplasms/drug therapy , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Drug Resistance , Protein-Tyrosine Kinases/biosynthesis , Thiazoles/therapeutic use , Animals , Cell Line, Tumor , Disease Models, Animal , Female , Gene Expression Profiling , Heterografts , Humans , Mice, SCID , Thiazoles/pharmacology , Treatment Failure
5.
Int J Cancer ; 135(10): 2462-74, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-24706460

ABSTRACT

mTOR inhibition led to activation of upstream receptor tyrosine kinases (RTKs) and AKT, which may attenuate the efficacy of mTOR kinase inhibitors. We sought to discover efficient drug combination with mTOR inhibitors by elucidating the survival feedback loops induced by mTOR inhibition in breast cancer. The feedback signaling upon treatment of mTOR inhibitor AZD8055 was determined and the combinatorial activity of AZD8055 and HSP90 inhibitor AUY922 in cell signaling and proliferation were detected. Treatment of breast cancer T47D cells with AZD8055 induced activation of AKT and phosphatidylinositol 3-kinase (PI3K), which was accompanied with increase in expression of multiple upstream proteins including EGFR, HER2, HER3 and IRS-1. Different RTKs were revealed to be responsible for the reactivation of AKT by AZD8055 in different breast cancer cell lines. Down-regulation of these proteins differentially enhanced the antiproliferative activity of AZD8055. AZD8055 and AUY922 displayed synergistic effect against a panel of human breast cancer cells irrespective their genotype, which was associated with enhanced cell cycle arrest and inhibition of DNA synthesis. AUY922 destabilized multiple tested tyrosine kinases and abrogated activation of AKT induced by AZD8055. AZD8055 also inhibited up-regulation of HSP70 and HSP27 upon AUY922 treatment. Cotreatment of these two drugs demonstrated synergistic activity against triple negative MDA-MB-468 xenograft without enhanced toxicity. The combination of AZD8055 and AUY922 demonstrated synergistic activity against various types of breast cancer and established a mechanistic rationale for a combination approach using catalytic mTOR kinase inhibitor and HSP90 inhibitor in the treatment of breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Cell Proliferation , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Isoxazoles/pharmacology , Morpholines/pharmacology , Resorcinols/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Apoptosis , Blotting, Western , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Drug Synergism , Female , Humans , Immunoenzyme Techniques , Mice , Mice, Inbred BALB C , Mice, Nude , Protein Transport , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
6.
J Biol Chem ; 287(15): 12132-41, 2012 Apr 06.
Article in English | MEDLINE | ID: mdl-22337890

ABSTRACT

CXCL12/CXCR4 plays an important role in metastasis of gastric carcinoma. Rapamycin has been reported to inhibit migration of gastric cancer cells. However, the role of mTOR pathway in CXCL12/CXCR4-mediated cell migration and the potential of drugs targeting PI3K/mTOR pathway remains unelucidated. We found that CXCL12 activated PI3K/Akt/mTOR pathway in MKN-45 cells. Stimulating CHO-K1 cells expressing pEGFP-C1-Grp1-PH fusion protein with CXCL12 resulted in generation of phosphatidylinositol (3,4,5)-triphosphate, which provided direct evidence of activating PI3K by CXCL12. Down-regulation of p110ß by siRNA but not p110α blocked phosphorylation of Akt and S6K1 induced by CXCL12. Consistently, p110ß-specific inhibitor blocked the CXCL12-activated PI3K/Akt/mTOR pathway. Moreover, CXCR4 immunoprecipitated by anti-p110ß antibody increased after CXCL12 stimulation and G(i) protein inhibitor pertussis toxin abrogated CXCL12-induced activation of PI3K. Further studies demonstrated that inhibitors targeting the PI3K/mTOR pathway significantly blocked the chemotactic responses of MKN-45 cells triggered by CXCL12, which might be attributed primarily to inhibition of mTORC1 and related to prevention of F-actin reorganization as well as down-regulation of active RhoA, Rac1, and Cdc42. Furthermore, rapamycin inhibited the secretion of CXCL12 and the expression of CXCR4, which might form a positive feedback loop to further abolish upstream signaling leading to cell migration. Finally, we found cells expressing high levels of cxcl12 were sensitive to rapamycin in its activity inhibiting migration as well as proliferation. In summary, we found that the mTOR pathway played an important role in CXCL12/CXCR4-mediated cell migration and proposed that drugs targeting the mTOR pathway may be used for the therapy of metastatic gastric cancer expressing high levels of cxcl12.


Subject(s)
Cell Movement/drug effects , Chemokine CXCL12/physiology , Receptors, CXCR4/metabolism , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Actins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Down-Regulation , Feedback, Physiological , Furans/pharmacology , Gene Expression , Humans , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism , Pyridines/pharmacology , Pyrimidines/pharmacology , Receptors, CXCR4/genetics , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction , Stomach Neoplasms , TOR Serine-Threonine Kinases/metabolism
7.
Biochem Pharmacol ; 83(9): 1183-94, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22305748

ABSTRACT

The mammalian target of rapamycin (mTOR), is deregulated in about 50% of human malignancies and exists in two complexes: mTORC1 and mTORC2. Rapalogs partially inhibit mTORC1 through allosteric binding to mTORC1 and their efficacy is modest as a cancer therapy. A few mTOR kinase inhibitors that inhibit both mTORC1 and mTORC2 have been reported to possess potent anticancer activities. Herein, we designed and synthesized a series of pyrazolopyrimidine derivatives targeting mTOR kinase domain and X-387 was identified as a promising lead. X-387 selectively inhibited mTOR in an ATP-competitive manner while sparing a panel of kinases from the PIKK family. X-387 blocked mTORC1 and mTORC2-mediacted signaling pathway in cell lines with activated mTOR signaling and in rapamycin-resistant cells. Specifically, X-387 inhibited phosphorylation of AKT at T308, which is thought to be a target of PDK1 but not mTOR. Such activity was not due to inhibition of PI3K since X-387 did not inhibit translocation of AKT to the cell membrane. X-387 induced autophagy as observed for other mTOR inhibitors, while induced autophagy is pro-survival since concurrent inhibition of autophagy by 3-MA reinforced the antiproliferative activity of mTOR inhibitors. X-387 also inhibited cell motility, which is associated with decrease in activity of small GTPases such as RhoA, Rac1 and Cdc42. Taken together, X-387 is a promising compound lead targeting mTOR and with a wide spectrum anticancer activity among tumor cell lines. The data also underscores the complexity of the mTOR signaling pathways which are far from being understood.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Phenylurea Compounds/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Autophagy/drug effects , Binding, Competitive , Catalytic Domain , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Size/drug effects , Drug Screening Assays, Antitumor , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Neoplasms/metabolism , Neoplasms/pathology , Oncogene Protein v-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , TOR Serine-Threonine Kinases/metabolism , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein , rhoA GTP-Binding Protein
8.
Mol Cell Biochem ; 360(1-2): 329-37, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21959972

ABSTRACT

Dihydroartemisinin (DHA) is an important artemisinin derivative and presents profound anti-tumor potential. A DHA-resistant cell line named HCT116/R derived from colon carcinoma cell line HCT116 was established in our previous study. Herein, we found that HCT116/R cells were much more resistant to DHA- or artesunate-induced proliferation inhibition and more tolerant to DHA-induced cell cycle arrest and apoptosis compared with those of the parent HCT116 cells. The protein levels of P-glycoprotein and MDR-associated protein 1 and the accumulation of doxorubicin in cells were similar in both cell lines. Moreover, HCT116/R cells were still sensitive to camptothecin- and doxorubicin-induced cell growth inhibition. To further explore the characterization of HCT116/R cell line, a proteomic study employing two-dimensional gel electrophoresis and matrix-assisted laser desorption/ionization time-of-flight mass spectrometry was performed. Eight different expressed proteins between the two cell lines were identified including some heat shock proteins, annexins, etc. This study not only indicates that exposure to DHA may not induce a tumor multi-drug-resistant phenotype but also affords new clues for the further investigation of the anti-cancer mechanisms of DHA and other artemisinin derivatives.


Subject(s)
Antineoplastic Agents/pharmacology , Artemisinins/pharmacology , Drug Resistance, Neoplasm , Annexins/genetics , Annexins/metabolism , Antineoplastic Agents/therapeutic use , Apoptosis , Artemisinins/therapeutic use , Artesunate , Carcinoma/drug therapy , Carcinoma/metabolism , Cell Cycle Checkpoints , Cell Proliferation/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Gene Expression , HCT116 Cells , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Phosphopyruvate Hydratase/genetics , Phosphopyruvate Hydratase/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
9.
Acta Pharmacol Sin ; 32(7): 930-8, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21643004

ABSTRACT

AIM: The insulin-like growth factor-1 receptor (IGF1R) is over-expressed in a wide variety of tumors and contributes to tumor cell proliferation, metastasis and drug resistance. The aim of this study was to establish a sensitive screening platform to identify novel IGF1R inhibitors. METHODS: The catalytic domain of IGF1R was expressed using the Bac-to-Bac baculovirus expression system. The screening platform for IGF1R inhibitors was established based on ELISA. The binding profile of IGF1R with the inhibitors was predicted with molecular docking and then subjected to the surface plasmon resonance (SPR) approach. The growth inhibition of cancer cells by the inhibitors was assessed with MTT assay. Apoptosis was analyzed using flow cytometry and Western blotting. RESULTS: A naturally occurring small molecule compound hematoxylin was identified as the most potent inhibitor (IC50 value=1.8±0.1 µmol/L) within a library of more than 200 compounds tested. Molecular simulation predicted the possible binding mode of hematoxylin with IGF1R. An SPR assay further confirmed that hematoxylin bound directly to IGF1R with high binding affinity (Kd=4.2 × 10⁻6 mol/L). In HL-60 cancer cells, hematoxylin inactivated the phosphorylation of IGF1R and downstream signaling and therefore suppressed cell proliferation. Mechanistic studies revealed that hematoxylin induced apoptosis in HL-60 cells via both extrinsic and intrinsic pathways. CONCLUSION: A simple, sensitive ELISA-based screening platform for identifying IGF1R inhibitors was established. Hematoxylin was identified as a promising IGF1R inhibitor with effective antitumor activity that deserves further investigation.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor/methods , Enzyme-Linked Immunosorbent Assay/methods , Hematoxylin/pharmacology , Protein Kinase Inhibitors/pharmacology , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, IGF Type 1/metabolism , Animals , Cations, Divalent/metabolism , Cell Line , Cell Proliferation/drug effects , Cloning, Molecular , Drug Screening Assays, Antitumor/economics , Enzyme-Linked Immunosorbent Assay/economics , HL-60 Cells , Humans , Models, Molecular , Neoplasms/drug therapy , Phosphorylation/drug effects , Protein Structure, Tertiary , Receptor, IGF Type 1/chemistry , Receptor, IGF Type 1/genetics , Sensitivity and Specificity
10.
Breast Cancer Res Treat ; 130(1): 85-96, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21181437

ABSTRACT

The central role of Src in tumor progression and metastasis has validated it as an attractive therapeutic target for the treatment of human breast cancer. The aim of this study was to identify potential Src kinase inhibitor, explore its activity, and mechanism of action in human breast cancer. A strategy integrating focused combinatorial library design, virtual screening, chemical synthesis, and high-throughput screening was adopted and a novel 6-hydrazinopurine-based inhibitor of c-Src kinase PH006 was obtained. The kinase enzymatic activities were measured by enzyme-linked immunosorbent assay. The binding mode between PH006 and Src was profiled by surface plasmon resonance approach and molecular simulation. The anti-proliferative activity was evaluated by Sulforhodamin B (SRB) and Colony formation. The anti-invasion and anti-migration activities were assessed by trans-well and wound healing assay. Results indicated that PH006 was an ATP-competitive Src inhibitor, which selectively inhibited c-Src with an IC50 of 0.38 µM among a panel of 14 diverse tyrosine kinases. PH006 potently inhibited c-Src phosphorylation and c-Src-dependent signal transduction, resulting in inhibition of cell proliferation, migration, and invasion in human breast cancer MDA-MB-231 cells. Further study demonstrated that the anti-proliferative activity of PH006 was ascribed to its capability to arrest cells in G1 phase, while its anti-motility activity was related to suppression of MMP2/9 and HGF secretion. Moreover, PH006 exhibited potent activity against tumor growth as well as metastasis of human breast cancer MDA-MB-435 xenograft beard in nude mice, which was accompanied with reduced Src/FAK signaling in tumor tissue. Taken together, PH006 is a novel selective inhibitor of c-Src and possesses potent activity against breast cancer growth and metastasis, which could be potentially developed as a lead candidate against breast cancers with elevated Src tyrosine kinase activity.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , src-Family Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Cell Adhesion/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Phosphorylation/drug effects , Protein Binding , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use , Purines/chemistry , Signal Transduction/drug effects , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
11.
Invest New Drugs ; 29(6): 1276-83, 2011 Dec.
Article in English | MEDLINE | ID: mdl-20607588

ABSTRACT

Dihydroartemisinin (DHA), the main active metabolite of artemisinin derivatives, is among the artemisinin derivatives possessing potent anti-malarial and anti-cancer activities. In the present study, we found that DHA displayed significant anti-proliferative activity in human colorectal carcinoma HCT116 cells, which may be attributed to its induction of G1 phase arrest and apoptosis. To further elucidate the mechanism of action of DHA, a proteomic study employed two-dimensional gel electrophoresis (2-DE) and matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) was performed. Glucose-regulated protein 78 (GRP78), which is related with endoplasmic reticulum stress (ER stress), was identified to be significantly up-regulated after DHA treatment. Further study demonstrated that DHA enhanced expression of GRP78 as well as growth arrest and DNA-damage-inducible gene 153 (GADD153, another ER stress-associated molecule) at both mRNA and protein levels. DHA treatment also led to accumulation of GADD153 in cell nucleus. Moreover, pretreatment of HCT116 cells with the iron chelator deferoxamine mesylate salt (DFO) abrogated induction of GRP78 and GADD153 upon DHA treatment, indicating iron is required for DHA-induced ER stress. This result is consistent with the fact that the anti-proliferative activity of DHA is also mediated by iron. We thus suggest the unbalance of redox may result in DHA-induced ER stress, which may contribute, at least in part, to its anti-cancer activity.


Subject(s)
Antineoplastic Agents/pharmacology , Artemisinins/pharmacology , Colorectal Neoplasms/drug therapy , Iron/metabolism , Apoptosis/drug effects , Cell Proliferation/drug effects , Colorectal Neoplasms/pathology , Electrophoresis, Gel, Two-Dimensional , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , G1 Phase/drug effects , HCT116 Cells , Heat-Shock Proteins/genetics , Humans , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Up-Regulation/drug effects
12.
J Pharmacol Exp Ther ; 334(3): 830-8, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20522531

ABSTRACT

The phosphatidylinositol-3-kinase (PI3K)-protein kinase B (Akt)-mammalian target of rapamycin (mTOR) signaling pathway is often constitutively activated in various human cancers, providing validated targets for cancer therapy. Among a series of 5-cyano-6-morpholino-4-substituted-pyrimidine analogs designed and synthesized based on PI3K target, 4-(2-(dimethylamino)vinyl)-2-(3-hydroxyphenyl)-6-morpholinopyrimidine-5-carbonitrile (WJD008) was selected for further pharmacological characterization because of its potent activity against PI3K signaling. WJD008 inhibited kinase activity of PI3Kalpha and mTOR with less activity against PIKK family members. In cellular settings, WJD008 abrogated insulin-like growth factor-I-activated PI3K-Akt-mTOR signaling cascade and blocked the membrane translocation of a pleckstrin homology domain containing enhanced green fluorescent protein-general receptor for phosphoinositides, isoform 1-pleckstrin homology fusion protein, suggesting down-regulation of phosphatidylinositol (3,4,5)-trisphosphate output induced by WJD008 resulted in inactivation of PI3K pathway. Consequently, WJD008 arrested cells in G(1) phase without induction of apoptosis. Furthermore, WJD008 reversed the hyperactivation of the PI3K pathway caused by the oncogenic mutation of p110alpha H1047R and suppressed the proliferation and clonogenesis of transformed RK3E cells harboring this mutant. WJD008 was superior to the pan-PI3K inhibitor wortmannin against proliferation of a panel of cancer cells independently of their status of PI3K pathway or tissue originations. In summary, WJD008 is a potent dual PI3K/mTOR modulator with antiproliferative and anticlonogenic activity in tumor cells and transformed cells with PIK3CA mutant, which provides new clues for the design and development of this chemical scaffold as an anticancer drug.


Subject(s)
Cell Proliferation/drug effects , Cell Transformation, Neoplastic/drug effects , Enzyme Inhibitors/pharmacology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Morpholines/pharmacology , Oncogenes/genetics , Phosphoinositide-3 Kinase Inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyrimidines/pharmacology , Animals , Apoptosis/drug effects , Blotting, Western , CHO Cells , Cell Cycle/drug effects , Cell Line, Tumor , Cells, Cultured , Cricetinae , Cricetulus , Enzyme-Linked Immunosorbent Assay , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/physiology , Mutation/genetics , Oncogene Protein v-akt/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/physiology , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/physiology , Rats , Signal Transduction/drug effects , Structure-Activity Relationship , TOR Serine-Threonine Kinases , Tumor Stem Cell Assay
13.
Expert Opin Investig Drugs ; 19(3): 329-43, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20141350

ABSTRACT

UNLABELLED: IMPORTANCE OF THIS FIELD: Microtubule-inhibiting drugs are among the most commonly prescribed agents in the combat against cancer, though the clinical use of these drugs is limited by acquired resistance, risk of hypersensitivity reactions and intolerable toxicity. With progress in our understanding of cytoskeleton structure and its related signaling pathways, a number of new microtubule-inhibiting agents with diversified structures and modes of action have emerged. WHAT THE READER WILL GAIN: This review mainly describes new microtubule-targeting anticancer agents that have been discovered (especially over the past 2 years), with emphasis on their diversity of structures and distinct modes of action. AREAS COVERED IN THIS REVIEW: Data were obtained exclusively from public sources, including journals and scientific meeting abstracts, up to September 2009. TAKE HOME MESSAGE: A number of new agents have been discovered, and some have entered clinical trials. Even though most of these agents stabilize or destabilize tubulin via binding on the recognized tubulin binding sites, a few compounds bind to tubulin on undefined sites or interrupt microtubule in diverse ways. Moreover, some agents target microtubule indirectly such that they alter the post-translational modification of tubulin. Further investigation into their mechanism of action and evaluation of their anticancer efficacy will help to develop novel regimens that are superior to existing approaches.


Subject(s)
Antineoplastic Agents/pharmacology , Microtubules/drug effects , Neoplasms/drug therapy , Animals , Antineoplastic Agents/adverse effects , Clinical Trials as Topic , Drug Delivery Systems , Drug Design , Drug Resistance, Neoplasm , Humans , Microtubules/metabolism , Neoplasms/physiopathology , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...