Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Biochem Pharmacol ; 223: 116140, 2024 May.
Article in English | MEDLINE | ID: mdl-38513740

ABSTRACT

Cancer cells consume more glucose and usually overexpress glucose transporters which have become potential targets for the development of anticancer drugs. It has been demonstrated that selective SGLT2 inhibitors, such as canagliflozin and dapagliflozin, display anticancer activity. Here we demonstrated that canagliflozin and dapagliflozin synergistically enhanced the growth inhibitory effect of paclitaxel in cancer cells including ovarian cancer and oral squamous cell carcinoma cells. Canagliflozin also inhibited glucose uptake via GLUTs. The combination of paclitaxel and WZB117, a GLUT inhibitor, exhibited a strong synergy, supporting the notion that inhibition of GLUTs by canagliflozin may also account for the synergy between canagliflozin and paclitaxel. Mechanistic studies in ES-2 ovarian cancer cells revealed that canagliflozin potentiated paclitaxel-induced apoptosis and DNA damaging effect. Paclitaxel in the nanomolar range elevated abnormal mitotic cells as well as aneuploid cells, and canagliflozin further enhanced this effect. Furthermore, canagliflozin downregulated cyclin B1 and phospho-BUBR1 upon spindle assembly checkpoint (SAC) activation by paclitaxel, and may consequently impair SAC. Thus, paclitaxel disturbed microtubule dynamics and canagliflozin compromised SAC activity, together they may induce premature mitotic exit, accumulation of aneuploid cells with DNA damage, and ultimately apoptosis.


Subject(s)
Benzhydryl Compounds , Carcinoma, Squamous Cell , Glucosides , Mouth Neoplasms , Ovarian Neoplasms , Female , Humans , Paclitaxel/pharmacology , Canagliflozin/pharmacology , Mitosis , Apoptosis , Ovarian Neoplasms/genetics , Glucose/pharmacology , Aneuploidy
2.
CPT Pharmacometrics Syst Pharmacol ; 12(12): 1960-1971, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37735924

ABSTRACT

The anticonvulsant valproic acid (VPA) despite complex pharmacokinetics has been in clinical use for nearly 6 decades. Previous reports indicated neonates, infants, and toddlers/preschoolers had higher risk of valproate hepatotoxicity than adults. However, dosing recommendations for those less than 10 years of age are lacking. To decipher clinical puzzles, physiologically-based pharmacokinetic (PBPK) models of VPA and its hepatotoxic metabolite 4-ene-VPA were constructed and simulated with particularly integrated information of drug-metabolizing enzyme ontogeny. Adult and pediatric PK data of VPA (n = 143 subjects) and 4-ene-VPA (n = 8 subjects) collected from previous reports were used for model development and validation. Sensitivity analyses were performed to characterize ontogeny impacts of CYP2C9 and UGT2B7 on dispositions of VPA and 4-ene-VPA across age groups. Optimal VPA dosing for each pediatric age group was also predicted and objectively judged by ensuring VPA efficacy and avoiding 4-ene-VPA hepatotoxicity. The study revealed UGT2B7 ontogeny was quite influential on VPA clearance even in neonates and small children. Intrinsic clearance of CYP2C9 was the most prominent determinant for areas under the concentration-time curve of VPA and 4-ene-VPA in infants, and toddlers/preschoolers, reflecting higher hepatotoxicity risk due to noxious 4-ene-VPA accumulation in these groups. The ontogeny-based PBPK approach complements conventional allometric methods in dosing estimation for the young by providing more mechanistic insight of the processes changing with age. The established ontogeny-based PBPK approach for VPA therapy deserves further corroboration by real-world therapeutic data to affirm its clinical applicability.


Subject(s)
Chemical and Drug Induced Liver Injury , Drug-Related Side Effects and Adverse Reactions , Adult , Infant , Infant, Newborn , Humans , Child , Valproic Acid/adverse effects , Valproic Acid/pharmacokinetics , Cytochrome P-450 CYP2C9 , Anticonvulsants/therapeutic use , Chemical and Drug Induced Liver Injury/etiology
3.
Molecules ; 28(5)2023 Mar 03.
Article in English | MEDLINE | ID: mdl-36903603

ABSTRACT

Hepatocellular carcinoma is the third most common cause of cancer-related death according to the International Agency for Research on Cancer. Dihydroartemisinin (DHA), an antimalarial drug, has been reported to exhibit anticancer activity but with a short half-life. We synthesized a series of bile acid-dihydroartemisinin hybrids to improve its stability and anticancer activity and demonstrated that an ursodeoxycholic-DHA (UDC-DHA) hybrid was 10-fold more potent than DHA against HepG2 hepatocellular carcinoma cells. The objectives of this study were to evaluate the anticancer activity and investigate the molecular mechanisms of UDCMe-Z-DHA, a hybrid of ursodeoxycholic acid methyl ester and DHA via a triazole linkage. We found that UDCMe-Z-DHA was even more potent than UDC-DHA in HepG2 cells with IC50 of 1 µM. Time course experiments and stability in medium determined by cell viability assay as well as HPLC-MS/MS analysis revealed that UDCMe-Z-DHA was more stable than DHA, which in part accounted for the increased anticancer activity. Mechanistic studies revealed that UDCMe-Z-DHA caused G0/G1 arrest and induced reactive oxygen species (ROS), mitochondrial membrane potential loss and autophagy, which may in turn lead to apoptosis. Compared to DHA, UDCMe-Z-DHA displayed much lower cytotoxicity toward normal cells. Thus, UDCMe-Z-DHA may be a potential drug candidate for hepatocellular carcinoma.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/metabolism , Ursodeoxycholic Acid , Liver Neoplasms/pathology , Tandem Mass Spectrometry , Apoptosis , Artemether , Cell Line, Tumor
4.
Molecules ; 27(22)2022 Nov 21.
Article in English | MEDLINE | ID: mdl-36432207

ABSTRACT

Tumor cells rely on aerobic glycolysis to support growth and survival, thus require more glucose supply. Glucose transporters GLUTs, primarily GLUT1, are overexpressed in various cancers. Targeting GLUTs has been regarded as a promising anticancer strategy. In this study, we first evaluated 75 potential GLUT1 inhibitors obtained from virtual screening of the NCI chemical library by a high-throughput cell-based method using a fluorescent glucose analogue 2-(N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)amino)-2-deoxy-d-glucose (2-NBDG) in COS-7 and SKOV3 cells that express high levels of GLUT1. Four compounds, #12, #16, #43 and #69, that significantly inhibited glucose uptake were further evaluated using flow cytometry directly measuring 2-NBDG uptake at the single-cell level and a Glucose Uptake-GloTM assay indirectly measuring 2-deoxy-d-glucose uptake in SKOV3, COS-7 or MCF-7 cells. The inhibitory effect on cancer cell growth was also determined in SKOV3 and MCF-7 cells, and #12 exhibited the best growth inhibitory effect equivalent to a known GLUT1 inhibitor WZB117. Although the anticancer effect of the identified potential GLUT1 inhibitors was moderate, they may enhance the activity of other anticancer drugs. Indeed, we found that #12 synergistically enhanced the anticancer activity of metformin in SKOV3 ovarian cancer cells.


Subject(s)
Antineoplastic Agents , Glucose , Glucose Transporter Type 1 , Biological Transport , Antineoplastic Agents/pharmacology , Flow Cytometry
5.
Eur J Pharmacol ; 933: 175256, 2022 Oct 15.
Article in English | MEDLINE | ID: mdl-36088983

ABSTRACT

Many studies have indicated that the risk of cognitive impairment is higher in patients with rheumatoid arthritis (RA). Additionally, patients with RA may have a lower incidence of cognitive impairment with long-term use of ibuprofen. This study was aimed at investigating the impacts of RA on memory function and the mechanisms that ibuprofen may exhibit to improve memory function in rats with collagen-induced arthritis (CIA). Ibuprofen (30 mg/kg) was given twice daily to CIA rats for two weeks starting from Day 18 following the first immunization. Memory function was measured by the Morris water maze (MWM) test and long-term potentiation (LTP). The proinflammatory cytokine levels and downstream signaling pathways, including mitogen-activated protein kinase (MAPK) and nuclear factor kappa B (NF-κB), were examined. Furthermore, the glutamatergic system, including glutamate transporters/receptors and brain extracellular levels of glutamate, was investigated. The results showed that the impaired learning memory in CIA rats, examined by the MWM test and LTP, can be ameliorated by ibuprofen treatment. Along with the improvement in memory deficits, ibuprofen attenuated both neuroinflammation and the associated elevated levels of phosphorylated p38, JNK, and p65 in the hippocampus of CIA rats. In addition, the decreased excitatory amino acid transporter 2 level, the increased extracellular glutamate, and the upregulated hippocampal NMDA receptor 2B of CIA rats were all normalized by ibuprofen treatment. These findings suggest that the effect of ibuprofen on the memory improvement in CIA rats is associated with the normalization of the activated MAPK and NF-κB pathways and the aberrant glutamatergic system.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/complications , Arthritis, Experimental/drug therapy , Cytokines/metabolism , Excitatory Amino Acid Transporter 2 , Glutamates , Ibuprofen/pharmacology , Ibuprofen/therapeutic use , Memory Disorders/drug therapy , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Rats
6.
Front Pharmacol ; 13: 879748, 2022.
Article in English | MEDLINE | ID: mdl-35662690

ABSTRACT

Breast cancer is the most prevalent cancer and the second leading cause of cancer death in women. Cisplatin is a commonly used chemotherapeutic drug for breast cancer treatment. Owing to serious side effects, the combination of cisplatin with other drugs is an effective strategy to simultaneously reduce side effects and increase the anticancer efficacy. GLUT1 is an emerging target for cancer treatment since cancer cells usually consume more glucose, a phenomenon called the Warburg effect. In this study, we found that the combination of cisplatin and a novel GLUT1 inhibitor #43 identified from our previous high-throughput screening exerted a synergistic anticancer effect in MCF-7 and MDA-MB-231 breast cancer cells. Mechanism studies in MCF-7 cells revealed that combination of cisplatin and #43 significantly induced apoptosis, intracellular reactive oxygen species, and loss of mitochondrial membrane potential. Furthermore, #43 enhanced the DNA damaging effect of cisplatin. Akt/mTOR downstream signaling and the ERK signaling pathway usually involved in cell growth and survival were inhibited by the combination treatment. On the other hand, phosphorylation of p38 and JNK, which may be associated with apoptosis, was induced by the combination treatment. Altogether, our data indicate that oxidative stress, DNA damage, the Akt/mTOR and MAPK signaling pathways, and apoptosis may be involved in the synergism of cisplatin and #43 in breast cancer cells.

7.
AJR Am J Roentgenol ; 205(4): 703-8, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26397318

ABSTRACT

OBJECTIVE: For unknown reasons, there is discordance among previous reports with regard to the association of contrast medium (CM) with nephropathy and the incidence of nephropathy after contrast-enhanced CT. This study aimed to determine the frequency of and possible factors related to CM-induced nephropathy in hospitalized patients, with an emphasis on detailing coprescriptions with nephrotoxic potential. MATERIALS AND METHODS: Of 1378 inpatients who underwent CT, 208 (15.1%) met the inclusion criteria: receipt of IV iodinated CM and baseline serum creatinine level obtained within 45 days before and within 2 weeks after CT. Patient demographics, clinical characteristics, comorbidity, nephrotoxic comedications (nine classes of drugs), and type of CM administered were retrospectively reviewed. Relationships between CM-induced nephropathy (serum creatinine level increase ≥ 25% or ≥ 0.5 mg/dL after CT) and risk factors were assessed by stepwise multivariate logistic regression. RESULTS: The cohort of 208 subjects had a high number of comorbidities (mean [± SD], 5.8 ± 3.5 diagnoses) and a high rate of receiving nephrotoxic comedications (45.2%). CM-induced nephropathy was detected in 27 (13.0%) patients. Concurrent use of four nephrotoxic agents (odds ratio [OR], 26.250; 95% CI, 3.673-233.993) was the most influential factor associated with CM-induced nephropathy; other predictors included preexisting renal disease (OR, 8.218; 95% CI, 1.622-42.357), baseline serum creatinine level less than 0.7 or greater than or equal to 1.3 mg/dL (OR, 3.463; 95% CI, 1.341-9.025), and hemoglobin level less than 9.3 g/dL (OR, 3.141; 95% CI, 1.087-8.946). CONCLUSION: Among the known risk factors, such as preexisting renal disease, high serum creatinine level, and low hemoglobin level, a statistically significant association was identified between CM-induced nephropathy and concurrent receipt of four nephrotoxic medications. Relevant preventive measures are warranted for individuals at risk, especially hospitalized patients receiving multiple nephrotoxic medications who require contrast-enhanced CT.


Subject(s)
Contrast Media/adverse effects , Kidney Diseases/chemically induced , Polypharmacy , Tomography, X-Ray Computed , Adolescent , Adult , Aged , Aged, 80 and over , Creatinine , Female , Follow-Up Studies , Hospitalization , Humans , Incidence , Kidney Diseases/diagnosis , Kidney Diseases/epidemiology , Logistic Models , Male , Middle Aged , Retrospective Studies , Risk Factors , Taiwan , Time Factors , Young Adult
8.
Oncotarget ; 6(27): 24032-46, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26125228

ABSTRACT

Epi-reevesioside F, a new cardiac glycoside isolated from the root of Reevesia formosana, displayed potent activity against glioblastoma cells. Epi-reevesioside F was more potent than ouabain with IC50 values of 27.3±1.7 vs. 48.7±1.8 nM (P < 0.001) and 45.0±3.4 vs. 81.3±4.3 nM (P < 0.001) in glioblastoma T98 and U87 cells, respectively. However, both Epi-reevesioside F and ouabain were ineffective in A172 cells, a glioblastoma cell line with low Na+/K+-ATPase α3 subunit expression. Epi-reevesioside F induced cell cycle arrest at S and G2 phases and apoptosis. It also induced an increase of intracellular concentration of Na+ but not Ca2+, cleavage and exposure of N-terminus of Bak, loss of mitochondrial membrane potential, inhibition of Akt activity and induction of caspase cascades. Potassium supplements significantly inhibited Epi-reevesioside F-induced effects. Notably, Epi-reevesioside F caused cytosolic acidification that was highly correlated with the anti-proliferative activity. In summary, the data suggest that Epi-reevesioside F inhibits Na+/K+-ATPase, leading to overload of intracellular Na+ and cytosolic acidification, Bak activation and loss of mitochondrial membrane potential. The PI3-kinase/Akt pathway is inhibited and caspase-dependent apoptosis is ultimately triggered in Epi-reevesioside F-treated glioblastoma cells.


Subject(s)
Apoptosis/drug effects , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Ouabain/chemistry , Saponins/chemistry , Sodium-Potassium-Exchanging ATPase/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , Antineoplastic Agents/chemistry , Brain Neoplasms/drug therapy , Calcium/chemistry , Cell Line, Tumor , Cell Proliferation , Cytosol/metabolism , Flow Cytometry , Glioblastoma/drug therapy , Humans , Hydrogen-Ion Concentration , Inhibitory Concentration 50 , Membrane Potential, Mitochondrial , Potassium/chemistry , Protein Structure, Tertiary , Rhodamines/chemistry , Sodium/chemistry
9.
Neuropharmacology ; 92: 146-57, 2015 May.
Article in English | MEDLINE | ID: mdl-25596491

ABSTRACT

Alzheimer's disease is a neurodegenerative disorder characterized by deposition of ß-amyloid (Aß) fibrils accompanied with progressive neurite loss. None of the clinically approved anti-Alzheimer's agents target both pathological processes. We hypothesized that conjugation of a metal chelator to destabilize Aß fibrils (fAßs) and a long-chain fatty alcohol to induce neurite outgrowth may generate a novel molecular scaffold that targets both pathologies. The hydroxyalkylquinoline J2326 was designed and synthesized by joining an 11-carbon alcohol to 5-chloro-8-methoxyquinoline at the 2-position and its anti-neurodegenerative potentials in vitro and in vivo were characterized. It attenuated fAß formation and disaggregated the existing fAß zinc-dependently as well as zinc-independently. It also triggered extracellular signal-regulated kinase-dependent neurite outgrowth and increased synaptic activity in neuronal cells. In fAß-driven neurodegeneration in vitro, J2326 reversed neurite collapse and neurotoxicity. These roles of J2326 were also demonstrated in vivo and were pivotal to the observed improvement in memory of mice with hippocampal fAß lesions. These results show that the effectiveness of J2326 on fAß-driven neurodegeneration is ascribed to its novel scaffold. This might give clues to evolving attractive therapy for future clinical trials.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Amyloid/metabolism , Antipsychotic Agents/chemistry , Antipsychotic Agents/therapeutic use , Drug Design , Models, Molecular , Neurites/drug effects , Animals , Chlorides/pharmacology , Disease Models, Animal , Fatty Alcohols/pharmacology , Mice , Quinolines/pharmacology , Rats , Signal Transduction/drug effects , Zinc/metabolism , Zinc Compounds/pharmacology
10.
Eur J Pharmacol ; 730: 90-101, 2014 May 05.
Article in English | MEDLINE | ID: mdl-24607408

ABSTRACT

Glioblastoma, a highly malignant glioma, is resistant to both radiation and chemotherapy and is an intractable problem in clinical treatment. New therapeutic approaches are in urgent need. Calanquinone A, an herbal constituent, displayed anti-proliferative activity against glioblastoma cells, including A172, T98 and U87. Flow cytometric analysis showed an S phase arrest and a subsequent apoptosis to calanquinone A action. Further identification demonstrated a rapid increase of γH2A.X formation at S phase. The data together with comet tail formation and Chk1 activation indicated DNA damage response. N-acetyl cysteine (an antioxidant and a glutathione precursor) and exogenously applied glutathione, but not trolox (an antioxidant), completely abolished calanquinone A-induced effects. Immunofluorescence assay revealed that calanquinone A decreased the intracellular glutathione levels in both A172 and T98 cells. However, calanquinone A, by itself, did not conjugate glutathione. The data suggested that the decrease of cellular glutathione predominantly contributed to the anticancer mechanism. Furthermore, calanquinone A induced the activation of AMP-activated protein kinase (AMPK) and the inhibition of p70S6K activity. Rhodamine efflux assay showed that calanquinone A did not block efflux activity, indicating that calanquinone A was not a P-glycoprotein substrate. In summary, the data suggest that calanquinone A displays anti-glioblastoma activity through a decrease of cellular glutathione levels that subsequently induces DNA damage stress and AMPK activation, leading to cell cycle arrest at S-phase and apoptotic cell death. Furthermore, calanquinone A does not serve as a P-glycoprotein substrate, suggesting a potential for further development in anti-glioblastoma therapy.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Antineoplastic Agents/pharmacology , DNA Damage , Glioblastoma/pathology , Glutathione/metabolism , Quinones/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , S Phase Cell Cycle Checkpoints/drug effects
11.
J Biosci ; 39(1): 85-95, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24499793

ABSTRACT

One of the pathological hallmarks of Alzheimer's disease is the presence of insoluble extracellular amyloid plaques. These plaques are mainly constituted of amyloid beta peptide (A beta), a proteolytic product of amyloid precursor protein (APP). APP processing also generates the APP intracellular domain (AICD). We have previously demonstrated that AICD interacts with FKBP12, a peptidyl-prolyl cis-trans isomerase (PPIase) ubiquitous in nerve systems. This interaction was interfered by FK506, a clinically used immunosuppressant that has recently been reported to be neuroprotective. To elucidate the roles of FKBP12 in the pathogenesis of Alzheimer's disease, the effect of FKBP12 overexpression on APP processing was evaluated. Our results revealed that APP processing was shifted towards the amyloidogenic pathway, accompanied by a change in the subcellular localization of APP, upon FKBP12 overexpression. This FKBP12-overexpression-induced effect was reverted by FK506. These findings support our hypothesis that FKBP12 may participate in the regulation of APP processing. FKBP12 overexpression may lead to the stabilization of a certain isomer (presumably the cis form) of the Thr668-Pro669 peptide bond in AICD, therefore change its affinity to flotillin-1 or other raft-associated proteins, and eventually change the localization pattern and cause a shift in the proteolytic processing of APP.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Protein Processing, Post-Translational/physiology , Tacrolimus Binding Protein 1A/metabolism , Tacrolimus/metabolism , Alzheimer Disease/genetics , Humans , Membrane Proteins/metabolism , Protein Processing, Post-Translational/genetics , Tacrolimus Binding Protein 1A/genetics
12.
ChemMedChem ; 7(5): 850-60, 2012 May.
Article in English | MEDLINE | ID: mdl-22383290

ABSTRACT

The metal ion chelating ß-N-hydroxy-γ-ketocarboxamide pharmacophore was integrated into a quinazolinone scaffold, leading to N-arylalkyl-3-hydroxy-4-oxo-3,4-dihydroquinazolin-2-carboxamide derivatives as hepatitis C virus (HCV) NS5B polymerase inhibitors. Lead optimization led to the identification of N-phenylpropyl carboxamide 9 k (IC(50) =8.8 µM). Compound 9 k possesses selectivity toward HCV1b replicon Ava.5 cells (EC(50) =17.5 µM) over parent Huh-7 cells (CC(50) =187.5 µM). Compound 9 k effects a mixed mode of NS5B inhibition, with NTP-competitive displacement properties. The interaction between 9 k and NS5B is stabilized by the presence of magnesium ions. Docking studies showed that the binding orientation of 9 k occupies the central portions of both magnesium-mediated and NTP-ribose-response binding sites within the active site region of NS5B. As a result, 3-hydroxy-4-oxo-3,4-dihydroquinazolin-2-carboxamide derivatives are disclosed herein as novel, mainly active site inhibitors of HCV NS5B polymerase.


Subject(s)
Antiviral Agents/chemistry , Drug Discovery , Hepacivirus , Quinazolinones/chemistry , Viral Nonstructural Proteins/antagonists & inhibitors , Antiviral Agents/pharmacology , Catalytic Domain , Humans , Models, Molecular , Molecular Structure , Polymerase Chain Reaction , Quinazolinones/pharmacology , Structure-Activity Relationship
13.
J Urol ; 186(6): 2434-41, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22019170

ABSTRACT

PURPOSE: Emerging evidence shows that the translocation of apoptosis related factors on cellular organelles, such as mitochondria, endoplasmic reticulum, Golgi apparatus and nucleus, has a crucial role in the apoptotic process. We characterized the effect of paclitaxel (Sigma®) on Golgi involved apoptosis in human hormone refractory prostate cancer. MATERIALS AND METHODS: FACScan™ flow cytometric analysis was used to determine cell cycle distribution and the subG1 (apoptosis) population. Protein expression and localization were detected by Western blot, confocal microscopic examination and the sucrose gradient separation technique. RESULTS: Paclitaxel induced Golgi apparatus disassembly and interaction between Golgi complexes and mitochondria. Discontinuous sucrose gradient fractionation was used to determine and collect Golgi containing fractions. Data revealed that paclitaxel induced an increase of Cdk1 activity and DR5 expression on the Golgi complex that was associated with increased cleavage of caspase-8, a DR5 downstream factor, and caspase-3 into catalytically active fragments. Data were validated by confocal immunofluorescence microscopy. Golgi associated effects were inhibited by the Cdk1 inhibitor roscovitine (Sigma), suggesting a critical role for Golgi-Cdk1. Also, paclitaxel caused an increase of nuclear but not of Golgi associated PKC-δ activity. The selective PKC-δ inhibitor rottlerin (Sigma) completely inhibited the increase of Golgi-Cdk1 activity, suggesting that nuclear PKC-δ served as an upstream regulator of Golgi-Cdk1. CONCLUSIONS: Data suggest that paclitaxel induces nuclear translocation and activation of PKC-δ, which in turn causes Golgi-Cdk1 activation, leading to Golgi associated DR5 up-regulation, and caspase-8 and 3 activation. Golgi mediated signaling cascades facilitate mitochondria involved apoptotic pathways and at least partly explain the anticancer activity of paclitaxel action.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , CDC2 Protein Kinase/physiology , Paclitaxel/pharmacology , Prostatic Neoplasms/enzymology , Protein Kinase C-delta/drug effects , Protein Kinase C-delta/physiology , Golgi Apparatus/enzymology , Humans , Male , Prostatic Neoplasms/drug therapy , Tumor Cells, Cultured
14.
J Urol ; 185(5): 1967-74, 2011 May.
Article in English | MEDLINE | ID: mdl-21421237

ABSTRACT

PURPOSE: Costunolide is a natural sesquiterpene lactone. We elucidated what to our knowledge is a novel mechanism to highlight its potential in chemotherapy for prostate cancer, particularly androgen refractory prostate cancer. MATERIALS AND METHODS: Several pharmacological and biochemical assays were used to characterize the apoptotic signaling pathways of costunolide (ChromaDex™) in prostate cancer cells. RESULTS: Costunolide showed effective antiproliferative activity against hormone dependent (LNCaP) and independent (PC-3 and DU-145) prostate cancer cells (ATCC®) by sulforhodamine B assay, clonogenic test and flow cytometric analysis of carboxyfluorescein succinimidyl ester labeling. In PC-3 cells data showed that costunolide induced a rapid overload of nuclear Ca(2+), DNA damage response and ATR phosphorylation. Costunolide induced G1-phase cell cycle arrest, which was supported by p21 up-regulation and its association with the cyclin dependent kinase 2/cyclin E complex. The association resulted in inhibition of the complex activity and inhibition of Rb phosphorylation. Costunolide mediated effects were substantially inhibited by glutathione, the reactive oxygen species scavenger and glutathione precursor N-acetylcysteine, and the Ca(2+) chelator BAPTA-AM other than the reactive oxygen species scavenger Trolox®. This indicated the crucial role of intracellular Ca(2+) mobilization and thiol depletion but not of reactive oxygen species production in apoptotic signaling. CONCLUSIONS: Data suggest that costunolide induces the depletion of intracellular thiols and overload of nuclear Ca(2+) that cause DNA damage and p21 up-regulation. The association of p21 with the cyclin dependent kinase 2/cyclin E complex blocks cyclin dependent kinase 2 activity and inhibits Rb phosphorylation, leading to G1 arrest of the cell cycle and subsequent apoptotic cell death in human prostate cancer cells.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Calcium/metabolism , Prostatic Neoplasms/drug therapy , Sesquiterpenes/pharmacology , Analysis of Variance , Blotting, Western , Cell Cycle/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Comet Assay , DNA Damage , Flow Cytometry , Humans , Immunoprecipitation , Male , Prostatic Neoplasms/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Tumor Cells, Cultured
15.
J Urol ; 183(6): 2409-18, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20403609

ABSTRACT

PURPOSE: Androgen refractory prostate cancer is a major clinical challenge. Treatment approaches to prostate cancer are based on various mechanisms that cause malignant cell apoptosis. Of these strategies the anticancer effect of triggering death receptors is well substantiated. MATERIALS AND METHODS: Several pharmacological and biochemical assays were used to characterize the apoptotic signaling pathways of the natural dihydrochalcone cryptocaryone in prostate cancer cells. RESULTS: Cryptocaryone induced antiproliferative and apoptotic effects in human androgen independent prostate cancer cells. It induced caspase-8 and 3 activation but did not change total protein levels of death receptors and their ligands. DR5 surface expression was moderately increased by cryptocaryone. Confocal immunofluorescence examination showed that cryptocaryone induced Fas clustering and the association of downstream signaling molecules, including FADD and procaspase-8. DR4 and DR5 aggregation was also induced by cryptocaryone. Data were confirmed by protein profile analysis of detergent resistant membranes showing that Fas, DR4, DR5, FADD and procaspase-8 levels were increased 1.3, 3.5, 4.1, 13.1 and 4.1-fold, respectively, in the lipid raft compartment. Cryptocaryone mediated clustering of death receptors and associated molecules was also detected in nonraft compartments. The distribution between lipid raft and nonraft compartments was validated by the cholesterol depleting agent methyl-beta-cyclodextrin. Cryptocaryone significantly potentiated FasL induced apoptosis in PC-3 cells. CONCLUSIONS: We suggest that cryptocaryone has anticancer activity via the stimulation of death receptor and associated molecule clustering, leading to caspase-8 and 3 activation, and apoptosis in prostate cancer cells.


Subject(s)
Apoptosis/drug effects , Membrane Microdomains/drug effects , Membrane Microdomains/physiology , Prostatic Neoplasms/pathology , Pyrones/pharmacology , Receptors, Death Domain/drug effects , Receptors, Death Domain/physiology , Androgens , Drug Screening Assays, Antitumor , Humans , Male , Tumor Cells, Cultured
16.
Anal Chem ; 81(16): 6944-51, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19621898

ABSTRACT

Zinc, which is abundant in senile plaques consisting mainly of fibrillar beta-amyloid (Abeta), plays a critical role in the pathogenesis of Alzheimer's disease. Treatment with zinc chelators such as clioquinol has been used to prevent Abeta aggregation in Alzheimer's patients; however, clioquinol produces severe side effects. A simple, easy, inexpensive, and versatile screen to identify zinc chelators for inhibition of Abeta aggregation is currently unavailable. We thus developed a high-throughput screen that identifies zinc chelators with anti-Abeta aggregation activity. The recombinant Abeta peptides, aggregated on solid-phase microplates, formed Abeta-immunopositive beta-sheet-containing structures in the presence of zinc. Formation of these Abeta fibrils was specifically blocked by metal ion chelators. This screening model improves identification of zinc-enhanced Abeta fibrils and anti-Abeta aggregation mediated by zinc chelating. The convenient system could qualitatively and quantitatively assay a large sample pool for Abeta aggregation inhibition and dissolution of Abeta aggregates. This screen is practical, reliable, and versatile for comprehensive detection of amyloid fibrillation and identification of inhibitors of Abeta aggregation.


Subject(s)
Amyloid beta-Peptides/metabolism , Chelating Agents/pharmacology , Models, Theoretical , Peptide Fragments/metabolism , Zinc/pharmacology , Amyloid beta-Peptides/antagonists & inhibitors , Humans , Peptide Fragments/antagonists & inhibitors , Recombinant Proteins/metabolism
17.
J Formos Med Assoc ; 107(8): 600-8, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18678543

ABSTRACT

BACKGROUND/PURPOSE: Inward rectifying potassium channel 6.2 (Kir6.2DelataC26 channel) is closely related to ATP-sensitive potassium channels. Whether sodium azide, barium ion, d-amphetamine or procaine acts directly on the Kir6.2DeltaC26 channel remains unclear. We studied the effects of these compounds on Kir6.2DeltaC26 channel expressed in Xenopus oocytes. METHODS: The coding sequence of a truncated form of mouse Kir6.2 (GenBank accession number NP_034732.1), Kir6.2(1-364) (i.e. Kir6.2DeltaC26), was subcloned into the pET20b(+) vector. Plasmid containing the correct T7 promoter-Kir6.2(1-364) cDNA fragment [Kir6.2/pET20b(+)] was then subject to NotI digestion to generate the templates for in vitro run-off transcriptions. The channel was expressed in Xenopus laevis oocytes. Two-electrode voltage clamping was used to measure the effects of sodium azide, barium ion, d-amphetamine and procaine on Kir6.2DeltaC26 channel current. RESULTS: Sodium azide activated and barium ion and d-amphetamine inhibited the Kir6.2DeltaC26 channel. Procaine did not have any significant effect on the Kir6.2DeltaC26 channel. CONCLUSION: Kir6.2DeltaC26 channel expressed in Xenopus oocytes can be used as a pharmacological tool for the study of inward rectifying potassium channels.


Subject(s)
Barium/pharmacology , Dextroamphetamine/pharmacology , Potassium Channels, Inwardly Rectifying/drug effects , Procaine/pharmacology , Sodium Azide/pharmacology , Amino Acid Sequence , Animals , Base Sequence , KATP Channels/drug effects , Molecular Sequence Data , Recombinant Proteins/drug effects , Xenopus laevis
18.
Eur J Pharmacol ; 556(1-3): 45-54, 2007 Feb 05.
Article in English | MEDLINE | ID: mdl-17140562

ABSTRACT

The marine organisms produce many metabolic substances with numerous pharmacological activities. It has been suggested that ilimaquinone, a metabolite of sea sponge, can induce vesiculation of the Golgi apparatus and display several biological activities, such as anti-human immunodeficiency virus, anti-inflammation as well as anti-microbial activities. In this study, the sulforhodamine B assays showed that ilimaquinone induced a concentration-dependent anti-proliferative effect in several types of cancer cell lines, including prostate cancer PC-3 and LNCaP, non-small cell lung cancer A549 and hepatocellular carcinoma Hep3B cells. The anticancer mechanism of ilimaquinone in the representative PC-3 cells was identified. Ilimaquinone induced a time-dependent increase of G(1) phase arrest and a subsequent increase of hypodiploid sub-G(1) phase (apoptosis) of the cell cycle. The arrest of the cell cycle was associated with a sustained high level of nuclear cyclin E but the absence of DNA synthesis by flow cytometric analysis, indicating an incomplete S phase. Although ilimaquinone-induced Golgi vesiculation, the data showed that the inhibition of cancer cell growth was not through the Golgi fragmentation. Several biological kinases and transcription factors were examined in this study. The data demonstrated that ilimaquinone did not activate extracellular signal-regulated kinase and phosphatidylinositol 3-kinase but induce the up-regulation and nuclear translocation of growth arrest and DNA damage inducible gene 153 (CHOP/GADD153). Furthermore, ilimaquinone-mediated anti-proliferative effect is significantly reduced in the antisense CHOP/GADD153-overexpressing cells. Ilimaquinone also inhibited DNA binding of NF-kappaB; however, this inhibitory effect could not explain ilimaquinone-induced anticancer effect. In summary, it is suggested that ilimaquinone induces the anti-proliferative effect through the G(1) arrest of the cell cycle and the up-regulation and nuclear translocation of CHOP/GADD153.


Subject(s)
Antineoplastic Agents/pharmacology , Porifera/chemistry , Quinones/pharmacology , Sesquiterpenes/pharmacology , Transcription Factor CHOP/metabolism , Active Transport, Cell Nucleus , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cyclin E/metabolism , Elafin/metabolism , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Golgi Apparatus/drug effects , Golgi Apparatus/ultrastructure , Humans , NF-kappa B/metabolism , Signal Transduction , Up-Regulation
19.
Naunyn Schmiedebergs Arch Pharmacol ; 374(3): 223-33, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17102938

ABSTRACT

Microtubules are crucial targets for cancer chemotherapeutic drugs, and new microtubule-directed agents are of continued interest in drug development. A novel microtubule-directed agent, ethyl-2-[N-rho-chlorobenzyl-(2'-methoxy)]-anilino-4-oxo -4, 5-dihydro-furan-3-carboxylate, was identified. The compound, designated K2154, inhibited cell proliferation, with IC(50) values of 10.3, 15.3, 9.6, 11.2, 12.8 and 12.1 muM in prostate cancer PC-3, hepatocellular carcinoma Hep3B, non-small cell lung cancer A549, colorectal cancer HT29 and HCT116, and P-glycoprotein-rich breast cancer NCI/ADR-RES cells, respectively. Because NCI/ADR-RES cells were susceptible to inhibition by K2154, it indicated that this compound is a poor substrate for P-glycoprotein. In this study, PC-3 cells were used to identify the anticancer mechanisms of K2154. K2154 induced an arrest of the cell cycle at G2/M phase and a subsequent increase of hypodiploid phase in PC-3 cells, whereas it only induced a moderate level of G2/M arrest with little increase of hypodiploid phase in normal prostate cells. K2154 inhibited microtubule assembly in both in vitro turbidity assay and in vivo microtubule spin-down experiment. Immunochemical examination showed that K2154 caused formation of abnormal mitotic characteristics with bipolar spindles, particularly, in beta(II)- and beta(III)-tubulin staining. It also induced several pathways, including cyclin B1 up-regulation, dephosphorylation on Tyr(15) and phosphorylation on Thr(161) of Cdk1 and Cdc25C phosphorylation, and roscovitine (a Cdk1 inhibitor) significantly inhibited K2154-induced apoptosis, suggesting a pro-apoptotic role of Cdk1. Phosphorylation of Bcl-2 and Bcl-xL and cleavage of Mcl-1, together with activation of caspase-9 and -3, indicated that mitochondrial pathway played a central role in K2154-mediated apoptotic cell death. Additionally, AIF contributed to a late phase of K2154-induced apoptotic pathway. In conclusion, it is suggested that K2154 displays an anticancer activity through a target on microtubules and a subsequent signaling cascade on cell cycle regulation and apoptotic machinery.


Subject(s)
Aniline Compounds/pharmacology , Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Cell Proliferation/drug effects , Furans/pharmacology , Tubulin/drug effects , ATP Binding Cassette Transporter, Subfamily B, Member 1 , Apoptosis/drug effects , Apoptosis Inducing Factor/metabolism , Caspases/drug effects , Caspases/metabolism , Cell Line, Tumor , Cyclin B/drug effects , Cyclin B/metabolism , Cyclin B1 , Cytochromes c/metabolism , Drug Delivery Systems , Humans , Inhibitory Concentration 50 , Microtubules/drug effects , Mitochondria/metabolism , Mitosis/drug effects , Phosphorylation , Tubulin/metabolism , Up-Regulation
20.
Biochem Biophys Res Commun ; 350(2): 472-7, 2006 Nov 17.
Article in English | MEDLINE | ID: mdl-17011518

ABSTRACT

To elucidate the roles of the APP intracellular domain (AICD) in the development of Alzheimer's disease, a yeast two-hybrid system was used to screen for AICD-interacting proteins. Our result revealed that FKBP12, an immunophilin with a peptidyl-prolyl cis-trans isomerase (PPIase) activity, may interact with AICD. This interaction was confirmed by coimmunoprecipitation studies. FKBP12 has been shown to be expressed at a higher level in areas of pathology of patients with neurodegenerative diseases. In addition, Pin1, a member of another PPIase family, has been suggested to be involved in the amyloidogenic APP processing and Abeta production. The interaction between FKBP12 and AICD might hint at a possible role FKBP12 plays, probably in a fashion similar to Pin1, in the amyloidogenesis of APP. We also found that the interaction was interfered, in a dose-dependent manner, by FK506, whose neuroprotective effect has been suggested to be correlated with its PPIase inhibitory activity.


Subject(s)
Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/metabolism , Tacrolimus Binding Protein 1A/metabolism , Binding Sites , Humans , Immunoprecipitation , Protein Structure, Tertiary , Recombinant Fusion Proteins/analysis , Tacrolimus Binding Protein 1A/genetics , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...