441 results on '"Rena, G"'
Search Results
402. Scientific Business Abstracts of the 113th Annual Meeting of the Association of Physicians of Great Britain and Ireland.
- Author
-
Cacciottolo TM, Perikari A, van der Klaauw A, Henning E, Stadler LKJ, Keogh J, Farooqi IS, Tenin G, Keavney B, Ryan E, Budd R, Bewley M, Coelho P, Rumsey W, Sanchez Y, McCafferty J, Dockrell D, Walmsley S, Whyte M, Liu Y, Choy MK, Tenin G, Abraham S, Black G, Keavney B, Ford T, Stanley B, Good R, Rocchiccioli P, McEntegart M, Watkins S, Eteiba H, Shaukat A, Lindsay M, Robertson K, Hood S, McGeoch R, McDade R, Sidik N, McCartney P, Corcoran D, Collison D, Rush C, McConnachie A, Touyz R, Oldroyd K, Berry C, Gazdagh G, Diver L, Marshall J, McGowan R, Ahmed F, Tobias E, Curtis E, Parsons C, Maslin K, D'Angelo S, Moon R, Crozier S, Gossiel F, Bishop N, Kennedy S, Papageorghiou A, Fraser R, Gandhi S, Prentice A, Inskip H, Godfrey K, Schoenmakers I, Javaid MK, Eastell R, Cooper C, Harvey N, Watt ER, Howden A, Mirchandani A, Coelho P, Hukelmann JL, Sadiku P, Plant TM, Cantrell DA, Whyte MKB, Walmsley SR, Mordi I, Forteath C, Wong A, Mohan M, Palmer C, Doney A, Rena G, Lang C, Gray EH, Azarian S, Riva A, Edwards H, McPhail MJW, Williams R, Chokshi S, Patel VC, Edwards LA, Page D, Miossec M, Williams S, Monaghan R, Fotiou E, Santibanez-Koref M, Keavney B, Badat M, Mettananda S, Hua P, Schwessinger R, Hughes J, Higgs D, and Davies J
- Published
- 2019
- Full Text
- View/download PDF
403. Equipotent doses of daunorubicin and idarubicin for AML: a meta-analysis of clinical trials versus in vitro estimation.
- Author
-
Adige S, Lapidus RG, Carter-Cooper BA, Duffy A, Patzke C, Law JY, Baer MR, Ambulos NP, Zou Y, Bentzen SM, and Emadi A
- Subjects
- Daunorubicin administration & dosage, Dose-Response Relationship, Drug, Humans, Idarubicin administration & dosage, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Leukemia, Myeloid, Acute drug therapy
- Abstract
In the treatment of acute myeloid leukemia (AML), the "7 + 3"-based strategy, combining cytarabine 100-200 mg/m
2 for 7 days with an anthracycline for 3 days, remains the standard of care for younger and medically fit patients. Daunorubicin (DNR) and idarubicin (IDA) are the two anthracyclines most commonly used. DNR and IDA are used interchangeably with different conversion factors, as there is no high-level evidence on the equipotency of these two agents for AML treatment. To determine the equipotent doses of DNR and IDA, we first systematically reviewed studies directly comparing the clinical outcomes of AML induction therapy utilizing DNR and IDA. We found 15 articles that met our inclusion criteria and compared time-to-event survival end points as well as complete remission rates post-induction. The DNR:IDA equipotency ratio was estimated at 5.90 with 95% confidence interval (CI) 1.7-20.7. To validate the estimate from our meta-analysis biologically, we conducted in vitro tests comparing anti-AML activity of DNR and IDA against six AML cell lines and two primary AML cells from patients with different cytogenetic and molecular characteristics. Based on these in vitro data, the equipotency dose ratio between DNR and IDA was 4.06 with 95% CI 3.64-4.49. Combining the estimates from the meta-analysis and the in vitro data using inverse-variance weighting, the current best estimate of the DNR:IDA equipotent ratio is 4.1 with 95% CI 3.9-4.3. This estimate, however, is largely driven by the in vitro chemo-sensitivity data. Given clinical studies demonstrating the safety of IDA at higher doses, our work implies that dose intensification of IDA could be investigated in future clinical trials in AML.- Published
- 2019
- Full Text
- View/download PDF
404. Amino alkynylisoquinoline and alkynylnaphthyridine compounds potently inhibit acute myeloid leukemia proliferation in mice.
- Author
-
Naganna N, Opoku-Temeng C, Choi EY, Larocque E, Chang ET, Carter-Cooper BA, Wang M, Torregrosa-Allen SE, Elzey BD, Lapidus RG, and Sintim HO
- Subjects
- Animals, Antineoplastic Agents chemistry, Apoptosis drug effects, Cell Cycle Checkpoints drug effects, Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, Humans, Isoquinolines chemistry, Leukemia, Myeloid, Acute, Mice, Molecular Structure, Mutation, Naphthyridines chemistry, Protein Kinase Inhibitors pharmacology, fms-Like Tyrosine Kinase 3 antagonists & inhibitors, fms-Like Tyrosine Kinase 3 genetics, fms-Like Tyrosine Kinase 3 metabolism, src-Family Kinases antagonists & inhibitors, src-Family Kinases metabolism, Antineoplastic Agents pharmacology, Isoquinolines pharmacology, Naphthyridines pharmacology
- Abstract
Background: Acute myeloid leukemia (AML) remains one of the most lethal, rarely cured cancers, despite decades of active development of AML therapeutics. Currently, the 5-year survival of AML patients is about 30% and for elderly patients, the rate drops to <10%. About 30% of AML patients harbor an activating mutation in the tyrosine kinase domain (TKD) of Fms-Like Tyrosine kinase 3 (FLT3) or a FLT3 internal tandem duplication (FLT3-ITD). Inhibitors of FLT3, such as Rydapt that was recently approved by the FDA, have shown good initial response but patients often relapse due to secondary mutations in the FLT3 TKD, like D835Y and F691 L mutations., Methods: Alkynyl aminoisoquinoline and naphthyridine compounds were synthesized via Sonogashira coupling. The compounds were evaluated for their in vitro and in vivo effects on leukemia growth., Findings: The compounds inhibited FLT3 kinase activity at low nanomolar concentrations. The lead compound, HSN431, also inhibited Src kinase activity. The compounds potently inhibited the viability of MV4-11 and MOLM-14 AML cells with IC50 values <1 nM. Furthermore, the viability of drug-resistant AML cells harboring the D835Y and F691 L mutations were potently inhibited. In vivo efficacy studies in mice demonstrated that the compounds could drastically reduce AML proliferation in mice., Interpretation: Compounds that inhibit FLT3 and downstream targets like Src (for example HSN431) are good leads for development as anti-AML agents. FUND: Purdue University, Purdue Institute for Drug Discovery (PIDD), Purdue University Center for Cancer Research, Elks Foundation and NIH P30 CA023168., (Copyright © 2019 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
405. Exploiting epigenetically mediated changes: Acute myeloid leukemia, leukemia stem cells and the bone marrow microenvironment.
- Author
-
Kogan AA, Lapidus RG, Baer MR, and Rassool FV
- Subjects
- Animals, DNA Methylation, Epigenesis, Genetic, Humans, Leukemia, Myeloid, Acute immunology, Neoplastic Stem Cells immunology, Tumor Microenvironment immunology, Bone Marrow pathology, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, Neoplastic Stem Cells pathology
- Abstract
Acute myeloid leukemia (AML) derives from the clonal expansion of immature myeloid cells in the bone marrow, and results in the disruption of normal hematopoiesis and subsequent bone marrow failure. The bone marrow microenvironment (BME) and its immune and other supporting cells are regarded to facilitate the survival, differentiation and proliferation of leukemia stem cells (LSCs), which enables AML cells to persist and expand despite treatment. Recent studies have identified epigenetic modifications among AML cells and BME constituents in AML, and have shown that epigenetic therapy can potentially reprogram these alterations. In this review, we summarize the interactions between the BME and LSCs, and discuss changes in how the BME and immune cells interact with AML cells. After describing the epigenetic modifications seen across chromatin, DNA, the BME, and the immune microenvironment, we explore how demethylating agents may reprogram these pathological interactions, and potentially re-sensitize AML cells to treatment., (© 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
406. Heart failure and diabetes: metabolic alterations and therapeutic interventions: a state-of-the-art review from the Translational Research Committee of the Heart Failure Association-European Society of Cardiology.
- Author
-
Maack C, Lehrke M, Backs J, Heinzel FR, Hulot JS, Marx N, Paulus WJ, Rossignol P, Taegtmeyer H, Bauersachs J, Bayes-Genis A, Brutsaert D, Bugger H, Clarke K, Cosentino F, De Keulenaer G, Dei Cas A, González A, Huelsmann M, Iaccarino G, Lunde IG, Lyon AR, Pollesello P, Rena G, Riksen NP, Rosano G, Staels B, van Laake LW, Wanner C, Farmakis D, Filippatos G, Ruschitzka F, Seferovic P, de Boer RA, and Heymans S
- Subjects
- Aged, Aged, 80 and over, Comorbidity, Diabetes Mellitus epidemiology, Diabetes Mellitus therapy, Europe, Female, Heart Failure epidemiology, Heart Failure mortality, Heart Failure physiopathology, Humans, Hypoglycemic Agents adverse effects, Hypoglycemic Agents therapeutic use, Insulin Resistance, Male, Obesity complications, Obesity epidemiology, Renal Insufficiency, Chronic complications, Renal Insufficiency, Chronic epidemiology, Socioeconomic Factors, Sulfonylurea Compounds adverse effects, Sulfonylurea Compounds therapeutic use, Diabetes Mellitus drug therapy, Heart Failure therapy, Societies, Medical organization & administration, Translational Research, Biomedical methods
- Published
- 2018
- Full Text
- View/download PDF
407. Regulation of hepatic glucose production and AMPK by AICAR but not by metformin depends on drug uptake through the equilibrative nucleoside transporter 1 (ENT1).
- Author
-
Logie L, Lees Z, Allwood JW, McDougall G, Beall C, and Rena G
- Subjects
- Aminoimidazole Carboxamide analogs & derivatives, Aminoimidazole Carboxamide pharmacokinetics, Animals, Biological Transport drug effects, Female, Liver metabolism, Metformin pharmacokinetics, Mice, Phosphorylation drug effects, Ribonucleotides pharmacokinetics, Signal Transduction drug effects, Thioinosine analogs & derivatives, Thioinosine metabolism, AMP-Activated Protein Kinases metabolism, Equilibrative Nucleoside Transporter 1 drug effects, Glucose metabolism, Hepatocytes drug effects, Hypoglycemic Agents pharmacokinetics
- Abstract
Aim: Recently we have observed differences in the ability of metformin and AICAR to repress glucose production from hepatocytes using 8CPT-cAMP. Previous results indicate that, in addition to activating protein kinase A, 8CPT-modified cAMP analogues suppress the nitrobenzylthioinosine (NBMPR)-sensitive equilibrative nucleoside transporter ENT1. We aimed to exploit 8CPT-cAMP, 8CPT-2-Methyl-O-cAMP and NBMPR, which is highly selective for a high-affinity binding-site on ENT1, to investigate the role of ENT1 in the liver-specific glucose-lowering properties of AICAR and metformin., Methods: Primary mouse hepatocytes were incubated with AICAR and metformin in combination with cAMP analogues, glucagon, forskolin and NBMPR. Hepatocyte glucose production (HGP) and AMPK signalling were measured, and a uridine uptake assay with supporting LC-MS was used to investigate nucleoside depletion from medium by cells., Results: AICAR and metformin increased AMPK pathway phosphorylation and decreased HGP induced by dibutyryl cAMP and glucagon. HGP was also induced by 8CPT-cAMP, 8CPT-2-Methyl-O-cAMP and NBMPR; however, in each case this was resistant to suppression by AICAR but not by metformin. Cross-validation of tracer and mass spectrometry studies indicates that 8CPT-cAMP, 8CPT-2-Methyl-O-cAMP and NBMPR inhibited the effects of AICAR, at least in part, by impeding its uptake into hepatocytes., Conclusions: We report for the first time that suppression of ENT1 induces HGP. ENT1 inhibition also impedes uptake and the effects of AICAR, but not metformin, on HGP. Further investigation of nucleoside transport may illuminate a better understanding of how metformin and AICAR each regulate HGP., (© 2018 The Authors. Diabetes, Obesity and Metabolism published by John Wiley & Sons Ltd.)
- Published
- 2018
- Full Text
- View/download PDF
408. Metformin selectively targets redox control of complex I energy transduction.
- Author
-
Cameron AR, Logie L, Patel K, Erhardt S, Bacon S, Middleton P, Harthill J, Forteath C, Coats JT, Kerr C, Curry H, Stewart D, Sakamoto K, Repiščák P, Paterson MJ, Hassinen I, McDougall G, and Rena G
- Subjects
- AMP-Activated Protein Kinases metabolism, Animals, Cell Line, Tumor, Electron Transport Complex I chemistry, Furans pharmacology, Glucose metabolism, Guanidine analogs & derivatives, Guanidine pharmacology, Mitochondria drug effects, Mitochondria metabolism, Oxidation-Reduction, Oxygen Consumption drug effects, Phosphorylation drug effects, Rats, Ribosomal Protein S6 Kinases metabolism, Signal Transduction drug effects, Electron Transport Complex I metabolism, Energy Metabolism drug effects, Metformin pharmacology
- Abstract
Many guanide-containing drugs are antihyperglycaemic but most exhibit toxicity, to the extent that only the biguanide metformin has enjoyed sustained clinical use. Here, we have isolated unique mitochondrial redox control properties of metformin that are likely to account for this difference. In primary hepatocytes and H4IIE hepatoma cells we found that antihyperglycaemic diguanides DG5-DG10 and the biguanide phenformin were up to 1000-fold more potent than metformin on cell signalling responses, gluconeogenic promoter expression and hepatocyte glucose production. Each drug inhibited cellular oxygen consumption similarly but there were marked differences in other respects. All diguanides and phenformin but not metformin inhibited NADH oxidation in submitochondrial particles, indicative of complex I inhibition, which also corresponded closely with dehydrogenase activity in living cells measured by WST-1. Consistent with these findings, in isolated mitochondria, DG8 but not metformin caused the NADH/NAD
+ couple to become more reduced over time and mitochondrial deterioration ensued, suggesting direct inhibition of complex I and mitochondrial toxicity of DG8. In contrast, metformin exerted a selective oxidation of the mitochondrial NADH/NAD+ couple, without triggering mitochondrial deterioration. Together, our results suggest that metformin suppresses energy transduction by selectively inducing a state in complex I where redox and proton transfer domains are no longer efficiently coupled., (Copyright © 2017 The Authors. Published by Elsevier B.V. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
409. Repurposing Metformin for Cardiovascular Disease.
- Author
-
Rena G and Lang CC
- Subjects
- Anti-Inflammatory Agents adverse effects, Cardiovascular Agents adverse effects, Cardiovascular Diseases epidemiology, Cardiovascular Diseases metabolism, Cardiovascular Diseases physiopathology, Diabetes Mellitus, Type 2 epidemiology, Humans, Hypoglycemic Agents adverse effects, Inflammation Mediators antagonists & inhibitors, Inflammation Mediators metabolism, Metformin adverse effects, Randomized Controlled Trials as Topic, Signal Transduction drug effects, Anti-Inflammatory Agents therapeutic use, Cardiovascular Agents therapeutic use, Cardiovascular Diseases drug therapy, Diabetes Mellitus, Type 2 drug therapy, Drug Repositioning, Hypoglycemic Agents therapeutic use, Metformin therapeutic use
- Published
- 2018
- Full Text
- View/download PDF
410. New Evidence for the Mechanism of Action of a Type-2 Diabetes Drug Using a Magnetic Bead-Based Automated Biosensing Platform.
- Author
-
Uddin R, Nur-E-Habiba, Rena G, Hwu ET, and Boisen A
- Abstract
The mechanism of action (MOA) of the first line type-2 diabetes drug metformin remains unclear despite its widespread usage. However, recent evidence suggests that the mitochondrial copper (Cu)-binding action of metformin may contribute toward the drug's MOA. Here, we present a novel biosensing platform for investigating the MOA of metformin using a magnetic microbead-based agglutination assay which has allowed us to demonstrate for the first time the interaction between Cu and metformin at clinically relevant low micromolar concentrations of the drug, thus suggesting a potential pathway of metformin's blood-glucose lowering action. In this assay, cysteine-functionalized magnetic beadswere agglutinated in the presence of Cu due to cysteine's Cu-chelation property. Addition of clinically relevant doses of metformin resulted in disaggregation of Cu-bridged bead-clusters, whereas the effect of adding a closely related but blood-glucose neutral drug propanediimidamide (PDI) showed completely different responses to the clusters. The entire assay was integrated in an automated microfluidics platform with an advanced optical imaging unit by which we investigated these aggregation-disaggregation phenomena in a reliable, automated, and user-friendly fashion with total assay time of 17 min requiring a sample (metformin/PDI) volume of 30 μL. The marked difference of Cu-binding action between the blood-glucose lowering drug metformin and its inactive analogue PDI thus suggests that metformin's distinctive Cu-binding properties may be required for its effect on glucose homeostasis. The novel automated platform demonstrating this novel investigation thus holds the potential to be utilized for investigating significant and sensitive molecular interactions via magnetic bead-based agglutination assay.
- Published
- 2017
- Full Text
- View/download PDF
411. The mechanisms of action of metformin.
- Author
-
Rena G, Hardie DG, and Pearson ER
- Subjects
- AMP-Activated Protein Kinases metabolism, Animals, Biguanides therapeutic use, Diabetes Mellitus, Type 2 enzymology, Humans, Diabetes Mellitus, Type 2 drug therapy, Hypoglycemic Agents therapeutic use, Metformin therapeutic use
- Abstract
Metformin is a widely-used drug that results in clear benefits in relation to glucose metabolism and diabetes-related complications. The mechanisms underlying these benefits are complex and still not fully understood. Physiologically, metformin has been shown to reduce hepatic glucose production, yet not all of its effects can be explained by this mechanism and there is increasing evidence of a key role for the gut. At the molecular level the findings vary depending on the doses of metformin used and duration of treatment, with clear differences between acute and chronic administration. Metformin has been shown to act via both AMP-activated protein kinase (AMPK)-dependent and AMPK-independent mechanisms; by inhibition of mitochondrial respiration but also perhaps by inhibition of mitochondrial glycerophosphate dehydrogenase, and a mechanism involving the lysosome. In the last 10 years, we have moved from a simple picture, that metformin improves glycaemia by acting on the liver via AMPK activation, to a much more complex picture reflecting its multiple modes of action. More work is required to truly understand how this drug works in its target population: individuals with type 2 diabetes.
- Published
- 2017
- Full Text
- View/download PDF
412. Exploring the Concept of Radiation "Booster Shot" in Combination with an Anti-PD-L1 mAb to Enhance Anti-Tumor Immune Effects in Mouse Pancreas Tumors.
- Author
-
Chuong M, Chang ET, Choi EY, Mahmood J, Lapidus RG, Davila E, and Carrier F
- Abstract
Radiotherapy (RT) has long been known to be immunogenic. Mounting preclinical data demonstrate a synergistic anti-tumor effect when RT is used in combination with immune check point inhibitors (ICI). However, it is unclear how to best integrate RT with an ICI (i.e. dose fractionation, sequence, etc.). Here we explored the concept that RT delivered as an in situ tumor vaccine sequentially to separate tumors over time might stimulate more potent and rapid antitumor immune response than RT delivered to only one tumor. In essence, radiation to a second tumor could be likened to giving a vaccine "booster shot". Mice bearing pancreatic tumors in three different sites were injected with anti-PD-L1 antibody and exposed to three daily consecutive fractions of 4 Gy each at one or two sites with a one week interval. Our data indicate that delivering an RT to one tumor followed by an RT "booster shot" to a second tumor, compared to treating only one tumor with RT, significantly reduced tumor growth at a third non-irradiated site. This abscopal effect to the non-irradiated site was observed earlier (day 9) in mice that received RT to two tumors versusa single tumor (day 17). Decreased growth of the non-irradiated tumor correlated with a transient increase of the CD4/CD8 ratio in the tumor, increase myeloid-derived suppressor cells and tumor associated macrophages in the draining lymph nodes. These data warrant further exploration of sequentially treating multiple lesions with RT and ICI with the intent of generating a robust anti-tumor immune response.
- Published
- 2017
413. Anti-Inflammatory Effects of Metformin Irrespective of Diabetes Status.
- Author
-
Cameron AR, Morrison VL, Levin D, Mohan M, Forteath C, Beall C, McNeilly AD, Balfour DJ, Savinko T, Wong AK, Viollet B, Sakamoto K, Fagerholm SC, Foretz M, Lang CC, and Rena G
- Subjects
- Aged, Animals, Anti-Inflammatory Agents pharmacology, Cells, Cultured, Cohort Studies, Diabetes Mellitus blood, Diabetes Mellitus diagnosis, Double-Blind Method, Female, Hepatocytes drug effects, Hepatocytes metabolism, Hepatocytes pathology, Humans, Hypoglycemic Agents pharmacology, Male, Metformin pharmacology, Mice, Mice, Inbred C57BL, Middle Aged, Piperidines pharmacology, Retrospective Studies, Sulfonamides pharmacology, Anti-Inflammatory Agents therapeutic use, Diabetes Mellitus drug therapy, Hypoglycemic Agents therapeutic use, Metformin therapeutic use
- Abstract
Rationale: The diabetes mellitus drug metformin is under investigation in cardiovascular disease, but the molecular mechanisms underlying possible benefits are poorly understood., Objective: Here, we have studied anti-inflammatory effects of the drug and their relationship to antihyperglycemic properties., Methods and Results: In primary hepatocytes from healthy animals, metformin and the IKKβ (inhibitor of kappa B kinase) inhibitor BI605906 both inhibited tumor necrosis factor-α-dependent IκB degradation and expression of proinflammatory mediators interleukin-6, interleukin-1β, and CXCL1/2 (C-X-C motif ligand 1/2). Metformin suppressed IKKα/β activation, an effect that could be separated from some metabolic actions, in that BI605906 did not mimic effects of metformin on lipogenic gene expression, glucose production, and AMP-activated protein kinase activation. Equally AMP-activated protein kinase was not required either for mitochondrial suppression of IκB degradation. Consistent with discrete anti-inflammatory actions, in macrophages, metformin specifically blunted secretion of proinflammatory cytokines, without inhibiting M1/M2 differentiation or activation. In a large treatment naive diabetes mellitus population cohort, we observed differences in the systemic inflammation marker, neutrophil to lymphocyte ratio, after incident treatment with either metformin or sulfonylurea monotherapy. Compared with sulfonylurea exposure, metformin reduced the mean log-transformed neutrophil to lymphocyte ratio after 8 to 16 months by 0.09 U (95% confidence interval, 0.02-0.17; P=0.013) and increased the likelihood that neutrophil to lymphocyte ratio would be lower than baseline after 8 to 16 months (odds ratio, 1.83; 95% confidence interval, 1.22-2.75; P=0.00364). Following up these findings in a double-blind placebo controlled trial in nondiabetic heart failure (trial registration: NCT00473876), metformin suppressed plasma cytokines including the aging-associated cytokine CCL11 (C-C motif chemokine ligand 11)., Conclusion: We conclude that anti-inflammatory properties of metformin are exerted irrespective of diabetes mellitus status. This may accelerate investigation of drug utility in nondiabetic cardiovascular disease groups., Clinical Trial Registration: Name of the trial registry: TAYSIDE trial (Metformin in Insulin Resistant Left Ventricular [LV] Dysfunction). URL: https://www.clinicaltrials.gov. Unique identifier: NCT00473876., (© 2016 The Authors.)
- Published
- 2016
- Full Text
- View/download PDF
414. Investigation of salicylate hepatic responses in comparison with chemical analogues of the drug.
- Author
-
Cameron AR, Logie L, Patel K, Bacon S, Forteath C, Harthill J, Roberts A, Sutherland C, Stewart D, Viollet B, Sakamoto K, McDougall G, Foretz M, and Rena G
- Subjects
- AMP-Activated Protein Kinases metabolism, Animals, Glucose-6-Phosphate metabolism, Hep G2 Cells, Humans, NF-kappa B metabolism, Rats, TOR Serine-Threonine Kinases metabolism, Hepatocytes metabolism, Liver metabolism, Salicylates chemistry, Salicylates pharmacology, Signal Transduction drug effects
- Abstract
Anti-hyperglycaemic effects of the hydroxybenzoic acid salicylate might stem from effects of the drug on mitochondrial uncoupling, activation of AMP-activated protein kinase, and inhibition of NF-κB signalling. Here, we have gauged the contribution of these effects to control of hepatocyte glucose production, comparing salicylate with inactive hydroxybenzoic acid analogues of the drug. In rat H4IIE hepatoma cells, salicylate was the only drug tested that activated AMPK. Salicylate also reduced mTOR signalling, but this property was observed widely among the analogues. In a sub-panel of analogues, salicylate alone reduced promoter activity of the key gluconeogenic enzyme glucose 6-phosphatase and suppressed basal glucose production in mouse primary hepatocytes. Both salicylate and 2,6 dihydroxybenzoic acid suppressed TNFα-induced IκB degradation, and in genetic knockout experiments, we found that the effect of salicylate on IκB degradation was AMPK-independent. Previous data also identified AMPK-independent regulation of glucose but we found that direct inhibition of neither NF-κB nor mTOR signalling suppressed glucose production, suggesting that other factors besides these cell signalling pathways may need to be considered to account for this response to salicylate. We found, for example, that H4IIE cells were exquisitely sensitive to uncoupling with modest doses of salicylate, which occurred on a similar time course to another anti-hyperglycaemic uncoupling agent 2,4-dinitrophenol, while there was no discernible effect at all of two salicylate analogues which are not anti-hyperglycaemic. This finding supports much earlier literature suggesting that salicylates exert anti-hyperglycaemic effects at least in part through uncoupling., (Copyright © 2016 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
415. A positive feedback loop involving EGFR/Akt/mTORC1 and IKK/NF-kB regulates head and neck squamous cell carcinoma proliferation.
- Author
-
Li Z, Yang Z, Passaniti A, Lapidus RG, Liu X, Cullen KJ, and Dan HC
- Subjects
- Antineoplastic Agents pharmacology, Apoptosis drug effects, Carcinoma, Squamous Cell drug therapy, Carcinoma, Squamous Cell genetics, Carcinoma, Squamous Cell pathology, Cell Line, Tumor, Cisplatin pharmacology, Dose-Response Relationship, Drug, Drug Resistance, Neoplasm, ErbB Receptors genetics, Feedback, Physiological, Head and Neck Neoplasms drug therapy, Head and Neck Neoplasms genetics, Head and Neck Neoplasms pathology, Humans, I-kappa B Kinase antagonists & inhibitors, Mechanistic Target of Rapamycin Complex 1 genetics, Phosphatidylinositol 3-Kinase metabolism, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins c-akt antagonists & inhibitors, RNA Interference, Squamous Cell Carcinoma of Head and Neck, Transfection, Carcinoma, Squamous Cell enzymology, Cell Proliferation drug effects, ErbB Receptors metabolism, Head and Neck Neoplasms enzymology, I-kappa B Kinase metabolism, Mechanistic Target of Rapamycin Complex 1 metabolism, NF-kappa B metabolism, Proto-Oncogene Proteins c-akt metabolism, Signal Transduction drug effects
- Abstract
The overexpression or mutation of epidermal growth factor receptor (EGFR) has been associated with a number of cancers, including head and neck squamous cell carcinoma (HNSCC). Increasing evidence indicates that both the phosphatidylinositol-3-kinase (PI3K)-Akt-mammalian target of Rapamycin (mTOR) and the nuclear factor-kappa B (NF-κB) are constitutively active and contribute to aggressive HNSCC downstream of EGFR. However, whether these two oncogenic signaling pathways exhibit molecular and functional crosstalk in HNSCC is unclear. Our results now reveal that mTORC1, not mTORC2, contributes to NF-κB activation downstream of EGFR/PI3K/Akt signaling. Mechanistically, mTORC1 enhances the inhibitor of nuclear factor kappa-B kinase (IKK) activity to accelerate NF-κB signaling. Concomitantly, activated NF-κB/IKK up-regulates EGFR expression through positive feedback regulation. Blockage of NF-κB/IKK activity by the novel IKKβ specific inhibitor, CmpdA, leads to significant inhibition of cell proliferation and induction of apoptosis. CmpdA also sensitizes intrinsic cisplatin-resistant HNSCC cells to cisplatin treatment. Our findings reveal a new mechanism by which EGFR/PI3K/Akt/mTOR signaling promotes head and neck cancer progression and underscores the need for developing a therapeutic strategy for targeting IKK/NF-κB either as a single agent or in combination with cisplatin in head and neck cancer., Competing Interests: No potential conflicts of interest were disclosed.
- Published
- 2016
- Full Text
- View/download PDF
416. The copper binding properties of metformin--QCM-D, XPS and nanobead agglomeration.
- Author
-
Quan X, Uddin R, Heiskanen A, Parmvi M, Nilson K, Donolato M, Hansen MF, Rena G, and Boisen A
- Abstract
Study of the copper binding properties of metformin is important for revealing its mechanism of action as a first-line type-2 diabetes drug. A quantitative investigation of interactions between metformin and L-cysteine-copper complexes was performed. The results suggest that metformin could interact with biological copper, which plays a key role in mitochondrial function.
- Published
- 2015
- Full Text
- View/download PDF
417. IKK phosphorylation of NF-κB at serine 536 contributes to acquired cisplatin resistance in head and neck squamous cell cancer.
- Author
-
Li Z, Yang Z, Lapidus RG, Liu X, Cullen KJ, and Dan HC
- Abstract
Current treatment methods for advanced head and neck squamous cell carcinoma (HNSCC) include surgery, radiation therapy and chemotherapy. For recurrent and metastatic HNSCC, cisplatin is the most common treatment option, but most of patients will eventually develop cisplatin resistance. Therefore, it is imperative to define the mechanisms involved in cisplatin resistance and find novel therapeutic strategies to overcome this deadly disease. In order to determine the role of nuclear factor-kappa B (NF-κB) in contributing to acquired cisplatin resistance in HNSCC, the expression and activity of NF-κB and its upstream kinases, IKKα and IKKβ, were evaluated and compared in three pairs of cisplatin sensitive and resistant HNSCC cell lines, including a pair of patient derived HNSCC cell line. The experiments revealed that NF-κB p65 activity was elevated in cisplatin resistant HNSCC cells compared to that in their parent cells. Importantly, the phosphorylation of NF-κB p65 at serine 536 and the phosphorylation of IKKα and IKKβ at their activation loops were dramatically elevated in the resistant cell lines. Furthermore, knockdown of NF-κB or overexpression of p65-S536 alanine (p65-S536A) mutant sensitizes resistant cells to cisplatin. Additionally, the novel IKKβ inhibitor CmpdA has been shown to consistently block the phosphorylation of NF-κB at serine 536 while also dramatically improving the efficacy of cisplatin in inhibition of cell proliferation and induction of apoptosis in the cisplatin resistant cancer cells. These results indicated that IKK/NF-κB plays a pivotal role in controlling acquired cisplatin resistance and that targeting the IKK/NF-κB signaling pathway may provide a possible therapeutic method to overcome the acquired resistance to cisplatin in HNSCC.
- Published
- 2015
418. Biomolecular mode of action of metformin in relation to its copper binding properties.
- Author
-
Repiščák P, Erhardt S, Rena G, and Paterson MJ
- Subjects
- Crystallography, X-Ray, Hydrogen-Ion Concentration, Hydrophobic and Hydrophilic Interactions, Mathematical Computing, Models, Molecular, Quantum Theory, Thermodynamics, Coordination Complexes chemistry, Copper chemistry, Hypoglycemic Agents chemistry, Metformin chemistry
- Abstract
Metformin (Metf), the most commonly used type 2 diabetes drug, is known to affect the cellular housekeeping of copper. Recently, we discovered that the structurally closely related propanediimidamide (PDI) shows a cellular behavior different from that of Metf. Here we investigate the binding of these compounds to copper, to compare their binding strength. Furthermore, we take a closer look at the electronic properties of these compounds and their copper complexes such as molecular orbital interactions and electrostatic potential surfaces. Our results clearly show that the copper binding energies cannot alone be the cause of the biochemical differentiation between Metf and PDI. We conclude that other factors such as pKa values and hydrophilicity of the compounds play a crucial role in their cellular activity. Metf in contrast to PDI can occur as an anion in aqueous medium at moderate pH, forming much stronger complexes particularly with Cu(II) ions, suggesting that biguanides but not PDI may induce easy oxidation of Cu(I) ions extracted from proteins. The higher hydrophobicity and the lack of planarity of PDI may further differentiate it from biguanides in terms of their molecular recognition characteristics. These different properties could hold the key to metformin's mitochondrial activity because they suggest that the drug could act at least in part as a pro-oxidant of accessible protein-bound Cu(I) ions.
- Published
- 2014
- Full Text
- View/download PDF
419. Salicylic acid: old and new implications for the treatment of type 2 diabetes?
- Author
-
Rena G and Sakamoto K
- Abstract
Efficacy of salicylic acid as a treatment for diabetes was first established well over a century ago. Antihyperglycaemic effects are thought to include improved peripheral insulin sensitivity and suppression of hepatic glucose production. For most of this period, the molecular mechanisms underlying these effects have been poorly understood and these are still a focus of considerable research, which is reviewed here. Antihyperglycaemic effects are observed only at much higher concentrations than analgesic, antipyretic and antithrombotic properties, suggesting that different targets underlie the antidiabetic aspects of salicylate pharmacology. In the 1950s, antihyperglycaemic responses were linked to mitochondrial uncoupling effects of the drug. Then at the beginning of this century, antihyperglycaemic effects were linked to anti-inflammatory effects of the drug on NF-κB signalling. More recently, new work suggests that direct activation of AMPK may contribute to antihyperglycaemic/antihyperlipidemic actions of salicylates. Better understanding of the mechanism of salicylate's anthyperglycaemic effects may ultimately accelerate the development of new drugs for human use.
- Published
- 2014
- Full Text
- View/download PDF
420. Risk factors for oxygen toxicity seizures in hyperbaric oxygen therapy: case reports from multiple institutions.
- Author
-
Seidel R, Carroll C, Thompson D, Diem RG, Yeboah K, Hayes AJ, Hall B, and Whelan HT
- Subjects
- Aged, Alcoholism complications, Antidepressive Agents adverse effects, Comorbidity, Female, Humans, Hypercapnia complications, Male, Middle Aged, Narcotics adverse effects, Pulmonary Disease, Chronic Obstructive complications, Risk Factors, Substance Withdrawal Syndrome complications, Wisconsin, Hyperbaric Oxygenation adverse effects, Oxygen adverse effects, Seizures etiology
- Abstract
Oxygen toxicity seizures are a rare but recognized complication of hyperbaric oxygen (HBO2) therapy. Many patients undergoing HBO2 therapy have medical conditions or are taking medications that could contribute to seizures. Previous literature has not extensively reported on these factors in patients experiencing oxygen toxicity seizures. We conducted a chart review at several hyperbaric oxygen centers in the Milwaukee, Wisc., area to explore whether the patients who experienced seizures in the hyperbaric chamber had other medical comorbidities or were on medications which lowered their seizure threshold, thereby contributing to oxygen toxicity seizures. There were a total of seven cases of seizures in five patients. Each patient had risk factors for seizures, including hypercapnia secondary to chronic obstructive pulmonary disease, narcotic withdrawal, alcohol dependence, and antidepressant, tramadol or cephalosporin/ceftriaxone use. We hypothesize that patients who experience oxygen toxicity seizures may have other factors which contribute to the development of these seizures.
- Published
- 2013
421. Molecular mechanism of action of metformin: old or new insights?
- Author
-
Rena G, Pearson ER, and Sakamoto K
- Subjects
- AMP-Activated Protein Kinases metabolism, Diabetes Mellitus, Type 2 drug therapy, Diabetes Mellitus, Type 2 metabolism, Energy Metabolism drug effects, Gluconeogenesis drug effects, Humans, Models, Biological, Metformin therapeutic use
- Abstract
Metformin is the first-line drug treatment for type 2 diabetes. Globally, over 100 million patients are prescribed this drug annually. Metformin was discovered before the era of target-based drug discovery and its molecular mechanism of action remains an area of vigorous diabetes research. An improvement in our understanding of metformin's molecular targets is likely to enable target-based identification of second-generation drugs with similar properties, a development that has been impossible up to now. The notion that 5' AMP-activated protein kinase (AMPK) mediates the anti-hyperglycaemic action of metformin has recently been challenged by genetic loss-of-function studies, thrusting the AMPK-independent effects of the drug into the spotlight for the first time in more than a decade. Key AMPK-independent effects of the drug include the mitochondrial actions that have been known for many years and which are still thought to be the primary site of action of metformin. Coupled with recent evidence of AMPK-independent effects on the counter-regulatory hormone glucagon, new paradigms of AMPK-independent drug action are beginning to take shape. In this review we summarise the recent research developments on the molecular action of metformin.
- Published
- 2013
- Full Text
- View/download PDF
422. In vivo screening of S100B inhibitors for melanoma therapy.
- Author
-
Zimmer DB, Lapidus RG, and Weber DJ
- Subjects
- Animals, Antineoplastic Agents adverse effects, Antineoplastic Agents pharmacokinetics, Antineoplastic Agents therapeutic use, Male, Maximum Tolerated Dose, Mice, S100 Calcium Binding Protein beta Subunit, Antineoplastic Agents pharmacology, Drug Screening Assays, Antitumor methods, Melanoma drug therapy, Nerve Growth Factors antagonists & inhibitors, S100 Proteins antagonists & inhibitors
- Abstract
S100 proteins are markers for numerous cancers, and in many cases high S100 protein levels are a prognostic indicator for poor survival. One such case is S100B, which is overproduced in a very large percentage of malignant melanoma cases. Elevated S100B protein was more recently validated to have causative effects towards cancer progression via down-regulating the tumor suppressor protein, p53. Towards eliminating this problem in melanoma, targeting S100B with small molecule inhibitors was initiated. This work relies on numerous chemical biology technologies including structural biology, computer-aided drug design, compound screening, and medicinal chemistry approaches. Another important component of drug development is the ability to test compounds and various molecular scaffolds for their efficacy in vivo. This chapter briefly describes the development of S100B inhibitors, termed SBiXs, for melanoma therapy with a focus on the inclusion of in vivo screening at an early stage in the drug discovery process.
- Published
- 2013
- Full Text
- View/download PDF
423. Cellular responses to the metal-binding properties of metformin.
- Author
-
Logie L, Harthill J, Patel K, Bacon S, Hamilton DL, Macrae K, McDougall G, Wang HH, Xue L, Jiang H, Sakamoto K, Prescott AR, and Rena G
- Subjects
- Adenylate Kinase metabolism, Animals, Cell Line, Cells, Cultured, Chelating Agents pharmacology, Glucose biosynthesis, Hepatocytes metabolism, Oxygen Consumption drug effects, Phosphorylation drug effects, Rats, Ribosomal Protein S6 Kinases metabolism, Signal Transduction drug effects, Trientine pharmacology, Copper metabolism, Hepatocytes drug effects, Hypoglycemic Agents pharmacology, Metformin pharmacology
- Abstract
In recent decades, the antihyperglycemic biguanide metformin has been used extensively in the treatment of type 2 diabetes, despite continuing uncertainty over its direct target. In this article, using two independent approaches, we demonstrate that cellular actions of metformin are disrupted by interference with its metal-binding properties, which have been known for over a century but little studied by biologists. We demonstrate that copper sequestration opposes known actions of metformin not only on AMP-activated protein kinase (AMPK)-dependent signaling, but also on S6 protein phosphorylation. Biguanide/metal interactions are stabilized by extensive π-electron delocalization and by investigating analogs of metformin; we provide evidence that this intrinsic property enables biguanides to regulate AMPK, glucose production, gluconeogenic gene expression, mitochondrial respiration, and mitochondrial copper binding. In contrast, regulation of S6 phosphorylation is prevented only by direct modification of the metal-liganding groups of the biguanide structure, supporting recent data that AMPK and S6 phosphorylation are regulated independently by biguanides. Additional studies with pioglitazone suggest that mitochondrial copper is targeted by both of these clinically important drugs. Together, these results suggest that cellular effects of biguanides depend on their metal-binding properties. This link may illuminate a better understanding of the molecular mechanisms enabling antihyperglycemic drug action.
- Published
- 2012
- Full Text
- View/download PDF
424. The anti-neurodegenerative agent clioquinol regulates the transcription factor FOXO1a.
- Author
-
Cameron AR, Wallace K, Logie L, Prescott AR, Unterman TG, Harthill J, and Rena G
- Subjects
- Active Transport, Cell Nucleus, Forkhead Box Protein O1, Gene Expression Regulation drug effects, Gluconeogenesis genetics, HEK293 Cells, Humans, Hydroxyquinolines pharmacology, Phosphorylation, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins c-akt antagonists & inhibitors, Proto-Oncogene Proteins c-akt metabolism, Signal Transduction drug effects, Structure-Activity Relationship, Zinc pharmacology, Clioquinol pharmacology, Forkhead Transcription Factors metabolism, Neuroprotective Agents pharmacology
- Abstract
Many diseases of aging including AD (Alzheimer's disease) and T2D (Type 2 diabetes) are strongly associated with common risk factors, suggesting that there may be shared aging mechanisms underlying these diseases, with the scope to identify common cellular targets for therapy. In the present study we have examined the insulin-like signalling properties of an experimental AD 8-hydroxyquinoline drug known as CQ (clioquinol). The IIS [insulin/IGF-1 (insulin-like growth factor-1) signalling] kinase Akt/PKB (protein kinase B) inhibits the transcription factor FOXO1a (forkhead box O1a) by phosphorylating it on residues that trigger its exit from the nucleus. In HEK (human embryonic kidney)-293 cells, we found that CQ treatment induces similar responses. A key transcriptional response to IIS is the inhibition of hepatic gluconeogenic gene expression, and, in rat liver cells, CQ represses expression of the key gluconeogenic regulatory enzymes PEPCK (phosphoenolpyruvate carboxykinase) and G6Pase (glucose-6-phosphatase). The effects on FOXO1a and gluconeogenic gene expression require the presence of Zn2+ ions, reminiscent of much earlier studies examining diabetogenic properties of 8-hydroxyquinolines. Comparative investigation of the signalling properties of a panel of these compounds demonstrates that CQ alone exhibits FOXO1a regulation without diabetogenicity. Our results suggest that Zn2+-dependent regulation of FOXOs and gluconeogenesis may contribute to the therapeutic properties of this drug. Further investigation of this signalling response might illuminate novel pharmacological strategies for the treatment of age-related diseases.
- Published
- 2012
- Full Text
- View/download PDF
425. Novel, Broad Spectrum Anti-Cancer Agents Containing the Tricyclic 5:7:5-Fused Diimidazodiazepine Ring System.
- Author
-
Kondaskar A, Kondaskar S, Kumar R, Fishbein JC, Muvarak N, Lapidus RG, Sadowska M, Edelman MJ, Bol GM, Vesuna F, Raman V, and Hosmane RS
- Abstract
Synthesis of a series of novel, broad-spectrum anti-cancer agents containing the tricyclic 5:7:5-fused diimidazo[4,5-d:4',5'-f][1,3]diazepine ring system is reported. Compounds 1, 2, 8, 11, and 12 in the series show promising in vitro antitumor activity with low micromolar IC(50)'s against prostate, lung, breast, and ovarian cancer cell lines. Some notions about structure-activity relationships and a possible mechanism of biological activity are presented. Also presented are preliminary in vivo toxicity studies of 1 using SCID mice.
- Published
- 2010
- Full Text
- View/download PDF
426. Zinc-dependent effects of small molecules on the insulin-sensitive transcription factor FOXO1a and gluconeogenic genes.
- Author
-
Cameron AR, Anil S, Sutherland E, Harthill J, and Rena G
- Subjects
- Blotting, Western, Cell Line, Disulfiram pharmacology, Ditiocarb pharmacology, Forkhead Box Protein O1, Glucose-6-Phosphatase metabolism, Humans, Insulin metabolism, Monoterpenes pharmacology, Phosphoenolpyruvate Carboxykinase (ATP) metabolism, Receptor, IGF Type 1 metabolism, Signal Transduction drug effects, Tropolone analogs & derivatives, Tropolone chemistry, Tropolone pharmacology, Forkhead Transcription Factors metabolism, Gluconeogenesis drug effects, Zinc metabolism
- Abstract
Metal-binding compounds have recently been reported to have anti-hyperglycaemic properties in vivo. In the current study, we have investigated the ability of these compounds and related structures to induce insulin-like signal transduction to downstream effectors such as the transcription factor FOXO1a and the key gluconeogenic regulatory enzymes phosphoenolpyruvate carboxykinase (PEPCK) and glucose 6-phosphatase (G6Pase). Our results indicate that β-thujaplicin, diethyldithiocarbamate (DEDTC) and its clinically-used dimer disulfiram, induce insulin-like dose-dependent effects on signalling to FOXO1a in a manner that is strictly dependent on the presence of zinc ions, as other ions including aluminium, cobalt, copper, lithium and manganese cannot substitute. The most potent compound tested on gluconeogenesis is disulfiram, which in the presence of 10 μM zinc, inhibited both PEPCK and G6Pase with an IC50 of 4 μM. Our results demonstrate that metal-binding compounds with diverse structures can induce zinc-dependent insulin-like effects on signal transduction and gene expression.
- Published
- 2010
- Full Text
- View/download PDF
427. Black tea polyphenols mimic insulin/insulin-like growth factor-1 signalling to the longevity factor FOXO1a.
- Author
-
Cameron AR, Anton S, Melville L, Houston NP, Dayal S, McDougall GJ, Stewart D, and Rena G
- Subjects
- Animals, Biflavonoids analysis, Catechin analogs & derivatives, Catechin analysis, Catechin chemistry, Cell Line, Flavonoids chemistry, Forkhead Box Protein O1, Gene Expression Regulation, Humans, Longevity, Phenols chemistry, Polyphenols, Protein Serine-Threonine Kinases metabolism, Rats, Biflavonoids pharmacology, Catechin pharmacology, Flavonoids pharmacology, Forkhead Transcription Factors metabolism, Insulin metabolism, Insulin-Like Growth Factor I metabolism, Phenols pharmacology, Signal Transduction, Tea chemistry
- Abstract
In vertebrates and invertebrates, relationships between diet and health are controlled by a conserved signalling pathway responsive to insulin-like ligands. In invertebrate models for example, forkhead transcription factor family O (FOXO) transcription factors in this pathway regulate the rate of aging in response to dietary cues, and in vertebrates, obesity and age-induced deficits in the same pathway are thought to contribute to dysregulation of hepatic gluconeogenesis through genes such as phosphoenolpyruvate carboxykinase (PEPCK). Recently, we have begun to screen for dietary constituents capable of regulating this pathway in our cell culture model. Here, we identify three black tea theaflavins, theaflavin 3-O-gallate, theaflavin 3'-O-gallate, theaflavin 3,3'di-O-gallate and thearubigins as novel mimics of insulin/IGF-1 action on mammalian FOXO1a, PEPCK and moreover we provide evidence that the effects on this pathway of the green tea constituent (-)-epigallocatechin gallate depend on its ability to be converted into these larger structures. With the exception of water, tea is the most popular drink globally, but despite this, little is known about the biological availability of black tea polyphenols in vivo or the molecular target(s) mediating the effects presented here. Further investigation in these two areas might provide insight into how age-related metabolic disease may be deferred.
- Published
- 2008
- Full Text
- View/download PDF
428. Epigallocatechin gallate (EGCG) mimics insulin action on the transcription factor FOXO1a and elicits cellular responses in the presence and absence of insulin.
- Author
-
Anton S, Melville L, and Rena G
- Subjects
- Catechin pharmacology, Cell Line, Drug Interactions, Forkhead Box Protein O1, Humans, Molecular Structure, Phosphatidylinositol 3-Kinases metabolism, Phosphoenolpyruvate Carboxykinase (ATP) genetics, Phosphorylation, Promoter Regions, Genetic, Reactive Oxygen Species metabolism, Signal Transduction, Transfection, Catechin analogs & derivatives, Forkhead Transcription Factors metabolism, Insulin pharmacology, Transcription Factors metabolism
- Abstract
The green tea flavonoid epigallocatechin gallate (EGCG) is one of several compounds that have been reported to have insulin-like glucose-lowering properties in mammals. EGCG is understood to act at least in part by repression of gluconeogenic genes such as phosphoenolpyruvate carboxykinase but the transcription factors that are targeted to achieve this are unknown. We show here that EGCG induces phosphorylation of insulin-sensitive residues on the transcription factor FOXO1a. Like insulin, EGCG induced FOXO1a phosphorylation is abolished by the PtdIns 3-kinase inhibitor LY294002 but not by PD98059 (an inhibitor of mitogen-activated protein kinase cascade) or by rapamycin (an inhibitor of signalling to p70 S6 kinase). EGCG differs from insulin and IGF-1 however, in that its induction of FOXO1a phosphorylation is sensitive to scavengers of reactive oxygen species (ROS). These results indicate that EGCG exerts its insulin mimetic effects at least in part by phosphorylation of the FOXOs through a mechanism that is similar but not identical to insulin and IGF-1 induced FOXO phosphorylation. Our results suggest that agents acting in the manner of EGCG may be useful antidiabetic agents.
- Published
- 2007
- Full Text
- View/download PDF
429. Structural elucidation of nitro-substituted five-membered aromatic heterocycles utilizing GIAO DFT calculations.
- Author
-
Katritzky AR, Akhmedov NG, Doskocz J, Hall CD, Akhmedova RG, and Majumder S
- Subjects
- Algorithms, Imidazoles chemistry, Magnetic Resonance Spectroscopy, Molecular Structure, Pyrazoles chemistry, Thiophenes chemistry, Heterocyclic Compounds, 1-Ring chemistry, Hydrocarbons, Aromatic chemistry, Pyrroles chemistry, Quantum Theory
- Abstract
The GIAO (Gauge Including Atomic Orbitals) DFT (Density Functional Theory) method is applied at the B3LYP/6-31+G(d,p)//B3LYP/6-31+G(d), B3LYP/6-311++G(d,p)//B3LYP/6-31+G(d), B3LYP/6-311+G (2d,p)//B3LYP/6-31+G(d) and B3LYP/6-311++G(d,p)//B3LYP/6-311++G(d,p) levels of theory for the calculation of proton and carbon chemicals shifts and coupling constants for 25 nitro-substituted five-membered heterocycles. Difference (1D NOE) spectra in combination with long-range gHMBC experiments were used as tools for the structural elucidation of nitro-substituted five-membered heterocycles. The assigned NMR data (chemical shifts and coupling constants) for all compounds were found to be in good agreement with theoretical calculations using the GIAO DFT method. The magnitudes of one-bond (1JCH) and long-range (nJCH, n>1) coupling constants were utilized for unambiguous differentiation between regioisomers of nitro-substituted five-membered heterocycles., (Copyright (c) 2006 John Wiley & Sons, Ltd.)
- Published
- 2007
- Full Text
- View/download PDF
430. Preparation of nitropyridines by nitration of pyridines with nitric acid.
- Author
-
Katritzky AR, Scriven EF, Majumder S, Akhmedova RG, Vakulenko AV, Akhmedov NG, Murugan R, and Abboud KA
- Abstract
Nitration of pyridines 1a-o with nitric acid in trifluoroacetic anhydride, gave the corresponding 3-nitropyridines 6a-n in yields of 10-83%.
- Published
- 2005
- Full Text
- View/download PDF
431. Anti-tumor effect of combination therapy with intratumoral controlled-release paclitaxel (PACLIMER microspheres) and radiation.
- Author
-
Lapidus RG, Dang W, Rosen DM, Gady AM, Zabelinka Y, O'Meally R, DeWeese TL, and Denmeade SR
- Subjects
- Animals, Cell Line, Tumor, Combined Modality Therapy, Delayed-Action Preparations, Humans, Male, Mice, Mice, Nude, Microspheres, Neoplasm Transplantation, Prostatic Neoplasms pathology, Antineoplastic Agents, Phytogenic administration & dosage, Paclitaxel administration & dosage, Prostatic Neoplasms drug therapy, Prostatic Neoplasms radiotherapy, Radiation-Sensitizing Agents administration & dosage
- Abstract
Background: Paclitaxel is one of the few chemotherapeutics effective in patients with advanced protstate cancer. Paclitaxel has also been reported to have radiosensitizing effects in prostate cancer. Local delivery of a controlled-release paclitaxel product may allow for increase local concentrations of paclitaxel at the tumor site and, in conjunction with radiation, may enhance cell kill by its radiosensitization mechanism., Methods: Orthotopic LNCaP tumors were injected with 40% PACLIMER Microspheres (40% loading; w:w) when tumors were 100-200 mm(3). Twenty-eight days post cell injection, mice were sacrificed, tumors weighed, and serum measured for PSA. TSU-xenografts were injected with PACLIMER Microspheres (10% and 40% loaded; w:w) or placebo microspheres when the tumors were approximately 100 mm(3). Half of xenograft tumors were irradiated with a single dose (10 Gy) of radiation. Tumor volume was followed over time., Results: Forty percent PACLIMER Microspheres significantly reduced tumor growth in the LNCaP orthotopic model. PSA was a good indicator of response. Forty percent PACLIMER Microspheres had a significant effect on slowing TSU growth compared to placebo microspheres. Addition of a single acute dose of radiation significantly enhanced the effect of 10% PACLIMER Microspheres (P < 0.05), had minimal effect on 40% PACLIMER Microspheres, and no enhancing effect on tumors treated with placebo microspheres., Conclusions: A controlled-release formulation of paclitaxel can be very effective in the treatment of prostate cancer. Additionally, PACLIMER Microspheres may be effectively used as a radiosensitizer in genitourinary cancers., (Copyright 2003 Wiley-Liss, Inc.)
- Published
- 2004
- Full Text
- View/download PDF
432. D4476, a cell-permeant inhibitor of CK1, suppresses the site-specific phosphorylation and nuclear exclusion of FOXO1a.
- Author
-
Rena G, Bain J, Elliott M, and Cohen P
- Subjects
- Benzamides metabolism, Cells, Cultured, Culture Media, Serum-Free, Forkhead Box Protein O1, Forkhead Transcription Factors, Humans, Imidazoles metabolism, Immunoprecipitation, Phosphorylation, Serine chemistry, Substrate Specificity, Tumor Cells, Cultured, Benzamides pharmacology, Casein Kinase I metabolism, DNA-Binding Proteins metabolism, Imidazoles pharmacology, Transcription Factors metabolism
- Abstract
The protein kinase CK1 phosphorylates serine residues that are located close to another phosphoserine in the consensus pSer-Xaa-Xaa-Ser. This specificity generates regions in its target proteins containing two or more neighbouring phosphoserine residues, termed here multisite phosphorylation domains (MPDs). In this paper, we demonstrate that D4476 is a potent and rather selective inhibitor of CK1 in vitro and in cells. In H4IIE hepatoma cells, D4476 specifically inhibits the phosphorylation of endogenous forkhead box transcription factor O1a (FOXO1a) on Ser322 and Ser325 within its MPD, without affecting the phosphorylation of other sites. Our results indicate that these residues are targeted by CK1 in vivo and that the CK1-mediated phosphorylation of the MPD is required for accelerated nuclear exclusion of FOXO1a in response to IGF-1 and insulin. D4476 is much more potent and specific than IC261 or CKI-7, and is therefore the most useful CK1 inhibitor currently available for identifying physiological substrates of CK1.
- Published
- 2004
- Full Text
- View/download PDF
433. Insulin regulation of insulin-like growth factor-binding protein-1 gene expression is dependent on the mammalian target of rapamycin, but independent of ribosomal S6 kinase activity.
- Author
-
Patel S, Lochhead PA, Rena G, Fumagalli S, Pende M, Kozma SC, Thomas G, and Sutherland C
- Subjects
- Adenoviridae genetics, Amino Acid Motifs, Animals, Blotting, Western, Cell Line, Culture Media, Serum-Free pharmacology, DNA, Complementary metabolism, Dose-Response Relationship, Drug, Enzyme Activation, Enzyme Inhibitors pharmacology, Humans, Phosphatidylinositol 3-Kinases metabolism, Plasmids metabolism, Precipitin Tests, Promoter Regions, Genetic, Protein Binding, RNA metabolism, Rats, Ribonucleases metabolism, Signal Transduction, Time Factors, Transcription, Genetic, Transfection, Tumor Cells, Cultured, Gene Expression Regulation, Insulin metabolism, Insulin-Like Growth Factor Binding Protein 1 metabolism, Ribosomal Protein S6 Kinases metabolism, Sirolimus pharmacology
- Abstract
Insulin inhibits the expression of the hepatic insulin-like growth factor-binding protein-1 (IGFBP-1) and glucose-6-phosphatase (G6Pase) genes. The signaling pathway that mediates these events requires the activation of phosphatidylinositol 3-kinase, whereas transfection studies have suggested an involvement of Akt (protein kinase B) and FKHR, a transcription factor regulated by Akt. We now demonstrate that insulin repression of endogenous IGFBP-1 gene transcription was blocked by rapamycin or by amino acid starvation. Rapamycin inhibited the mammalian target of rapamycin (mTOR) and the subsequent activation of p70/p85 S6 protein kinase-1 (S6K1) by insulin, whereas amino acid depletion prevented insulin induction of these signaling molecules. Importantly, we demonstrate that insulin regulation of the thymine-rich insulin response element of the IGFBP-1 promoter was also inhibited by rapamycin. However, sustained activation of S6K1 did not repress this promoter. In addition, rapamycin did not affect insulin regulation of G6Pase expression or Akt activation. We propose that these observations indicate that an mTOR-dependent, but S6K-independent mechanism regulates the suppression of IGFBP-1 (but not G6Pase) gene expression by insulin. Therefore, although the insulin-responsive sequence of the G6Pase gene promoter is related to that of the IGFBP-1 promoter, the signaling pathways that mediate suppression of these genes are distinct.
- Published
- 2002
- Full Text
- View/download PDF
434. Antagonistic effects of phorbol esters on insulin regulation of insulin-like growth factor-binding protein-1 (IGFBP-1) but not glucose-6-phosphatase gene expression.
- Author
-
Patel S, Lochhead PA, Rena G, and Sutherland C
- Subjects
- Animals, Benzamides pharmacology, Calcium-Calmodulin-Dependent Protein Kinases metabolism, Culture Media, Serum-Free, Cyclic AMP chemistry, Cyclic AMP pharmacology, DNA-Binding Proteins metabolism, Dexamethasone pharmacology, Enzyme Inhibitors pharmacology, Forkhead Transcription Factors, Gene Expression Regulation physiology, Glucocorticoids pharmacology, Glucose-6-Phosphatase genetics, Glycogen Synthase Kinase 3, Insulin-Like Growth Factor Binding Protein 1 genetics, Mitogen-Activated Protein Kinases metabolism, Promoter Regions, Genetic genetics, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins c-akt, Rats, Ribosomal Protein S6 Kinases metabolism, Tetradecanoylphorbol Acetate metabolism, Transcription Factors metabolism, Tumor Cells, Cultured, Gene Expression Regulation drug effects, Glucose-6-Phosphatase metabolism, Insulin metabolism, Insulin-Like Growth Factor Binding Protein 1 metabolism, Nerve Tissue Proteins, Tetradecanoylphorbol Acetate pharmacology
- Abstract
Glucose-6-phosphatase (G6Pase) and insulin-like growth factor-binding protein-1 (IGFBP-1) genes contain a homologous promoter sequence that is required for gene repression by insulin. Interestingly, this element interacts with members of the forkhead family of transcription factors [e.g. HNF3 (hepatic nuclear factor 3), FKHR (forkhead in rhabdomyosarcoma)] in vitro, while insulin promotes the phosphorylation and inactivation of FKHR in a phosphatidylinositol 3-kinase- and protein kinase B (PKB)-dependent manner. This mechanism has been proposed to underlie insulin action on G6Pase and IGFBP-1 gene transcription. However, we find that treatment of cells with phorbol esters mimics the effect of insulin on G6Pase, but not IGFBP-1, gene expression. Indeed, phorbol ester treatment actually blocks the ability of insulin to repress IGFBP-1 gene expression. In addition, the action of phorbol esters is significantly reduced by inhibition of the p42/p44 mitogen-activated protein (MAP) kinase pathway. However insulin-induced phosphorylation of PKB or FKHR is not affected by the presence of phorbol esters. Therefore we suggest that activation of p42/p44 MAP kinases will reduce the sensitivity of the IGFBP-1 gene promoter, but not the G6Pase gene promoter, to insulin. Importantly, the activation of PKB and phosphorylation of FKHR is not, in itself, sufficient to reduce IGFBP-1 gene expression in the presence of phorbol esters.
- Published
- 2001
- Full Text
- View/download PDF
435. The kinase DYRK1A phosphorylates the transcription factor FKHR at Ser329 in vitro, a novel in vivo phosphorylation site.
- Author
-
Woods YL, Rena G, Morrice N, Barthel A, Becker W, Guo S, Unterman TG, and Cohen P
- Subjects
- Amino Acid Sequence, Animals, Cell Nucleus metabolism, Cells, Cultured, Conserved Sequence, DNA-Binding Proteins genetics, DNA-Binding Proteins immunology, Forkhead Box Protein O1, Forkhead Transcription Factors, Humans, Molecular Sequence Data, Mutagenesis, Phosphorylation, Precipitin Tests, Protein Serine-Threonine Kinases immunology, Protein Serine-Threonine Kinases isolation & purification, Protein-Tyrosine Kinases immunology, Protein-Tyrosine Kinases isolation & purification, Rabbits, Sequence Homology, Amino Acid, Serine genetics, Transcription Factors genetics, Transcription Factors immunology, Transcriptional Activation, Dyrk Kinases, DNA-Binding Proteins metabolism, Protein Serine-Threonine Kinases metabolism, Protein-Tyrosine Kinases metabolism, Serine metabolism, Transcription Factors metabolism
- Abstract
Forkhead in rhabdomyosarcoma (FKHR) is a transcription factor that has been implicated in the control of gene expression by insulin, as well as the regulation of apoptosis by survival factors. These signals trigger the protein kinase B (PKB)-catalysed phosphorylation of FKHR at three residues (Thr(24), Ser(256) and Ser(319)) by a phosphoinositide 3-kinase-dependent pathway that results in the nuclear exit and inactivation of this transcription factor. Here, we have identified a conserved residue (Ser(329)) as a novel in vivo phosphorylation site on FKHR. Ser(329) phosphorylation also decreases the ability of FKHR to stimulate gene transactivation and reduces the proportion of FKHR present in the nucleus. However, unlike the residues targetted by PKB, Ser(329) is phosphorylated in unstimulated HEK-293 cells, and phosphorylation is not increased by stimulation with insulin-like growth factor-1 or by transfection with 3-phosphoinositide-dependent protein kinase-1. We have also purified a protein kinase to near homogeneity from rabbit skeletal muscle that phosphorylates FKHR at Ser(329) specifically and identified it as DYRK1A (dual-specificity tyrosine-phosphorylated and regulated kinase 1A). We find that FKHR and DYRK1A co-localize in discrete regions of the nucleus and can be co-immunoprecipitated from cell extracts. These experiments suggest that DYRK1A may phosphorylate FKHR at Ser(329) in vivo.
- Published
- 2001
- Full Text
- View/download PDF
436. Molecular cloning, genomic positioning, promoter identification, and characterization of the novel cyclic amp-specific phosphodiesterase PDE4A10.
- Author
-
Rena G, Begg F, Ross A, MacKenzie C, McPhee I, Campbell L, Huston E, Sullivan M, and Houslay MD
- Subjects
- 3',5'-Cyclic-AMP Phosphodiesterases metabolism, Amino Acid Sequence, Animals, Base Sequence, COS Cells, Cloning, Molecular, Cyclic Nucleotide Phosphodiesterases, Type 4, DNA, Complementary analysis, Databases, Factual, Enzyme Stability, Exons, Genome, Human, Humans, Introns, Mice, Molecular Sequence Data, Rats, Sequence Homology, Amino Acid, Transfection, 3',5'-Cyclic-AMP Phosphodiesterases genetics, Isoenzymes genetics, Promoter Regions, Genetic genetics
- Abstract
We describe the cloning and expression of HSPDE4A10, a novel long form splice variant of the human cAMP phosphodiesterase PDE4A gene. The 825 amino acid HSPDE4A10 contains a unique N terminus of 46 amino acids encoded by a unique 5' exon. Exon-1(4A10) lies approximately 11 kilobase pairs (kb) downstream of exon-1(4A4) and approximately 13.5 kb upstream of the PDE4A common exon 2. We identify a rat PDE4A10 ortholog and reveal a murine ortholog by nucleotide sequence database searching. PDE4A10 transcripts were detected in various human cell lines and tissues. The 5' sequence flanking exon-1(4A10) exhibited promoter activity with the minimal functional promoter region being highly conserved in the corresponding mouse genomic sequence. Transient expression of the engineered human PDE4A10 open reading frame in COS7 cells allowed detection of a 121-kDa protein in both soluble and particulate fractions. PDE4A10 was localized primarily to the perinuclear region of COS7 cells. Soluble and particulate forms exhibited similar K(m) values for cAMP hydrolysis (3-4 microM) and IC(50) values for inhibition by rolipram (50 nM) but the V(max) value of the soluble form was approximately 3-fold greater than that of the particulate form. At 55 degrees C, soluble HSPDE4A10 was more thermostable (T(0.5) = 11 min) than the particulate enzyme (T(0.5) = 5 min). HSPDE4A10 and HSPDE4A4B are shown here to be similar in size and exhibit similar maximal activities but differ with respect to sensitivity to inhibition by rolipram, thermostability, interaction with the SRC homology 3 domain of LYN, an SRC family tyrosyl kinase, and subcellular localization. We suggest that the unique N-terminal regions of PDE4A isoforms confer distinct properties upon them.
- Published
- 2001
- Full Text
- View/download PDF
437. Roles of the forkhead in rhabdomyosarcoma (FKHR) phosphorylation sites in regulating 14-3-3 binding, transactivation and nuclear targetting.
- Author
-
Rena G, Prescott AR, Guo S, Cohen P, and Unterman TG
- Subjects
- 14-3-3 Proteins, Active Transport, Cell Nucleus, Cell Line, Cell Nucleus metabolism, DNA-Binding Proteins genetics, DNA-Binding Proteins physiology, Forkhead Box Protein O1, Forkhead Box Protein O3, Forkhead Transcription Factors, Glutathione Transferase genetics, Glutathione Transferase metabolism, Green Fluorescent Proteins, Humans, Insulin-Like Growth Factor I pharmacology, Luminescent Proteins genetics, Luminescent Proteins metabolism, Mutagenesis, Site-Directed, Phosphorylation, Phosphoserine metabolism, Phosphothreonine metabolism, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins c-akt, Recombinant Fusion Proteins metabolism, Transcription Factors genetics, Transcription Factors physiology, Transcriptional Activation, DNA-Binding Proteins metabolism, Protein Serine-Threonine Kinases, Transcription Factors metabolism, Tyrosine 3-Monooxygenase metabolism
- Abstract
The transcription factor, forkhead in rhabdomyosarcoma (FKHR), is phosphorylated at three amino acid residues (Thr-24, Ser-256 and Ser-319) by protein kinase B (PKB)alpha. In the present study, mutagenesis has been used to study the roles of these phosphorylation events in regulating FKHR function in transfected HEK-293 cells. We find that the overexpression of FKHR[S256A] (where Ser-256-->Ala) blocks PKB activity in cells, preventing phosphorylation of the endogenous substrates FKHRL1 and glycogen synthase kinase-3. Thus some reported effects of overexpression of this and other mutants may be indirect, and result from suppression of the phosphorylation of other sites on FKHR and/or other PKB substrates. For example, we have shown that Thr-24 phosphorylation alone is critical for interaction with 14-3-3 proteins, and that the substitution of Ser-256 with an alanine residue indirectly blocks 14-3-3 protein binding by preventing the phosphorylation of Thr-24. We also found that insulin-like growth factor (IGF)-1 and serum-induced nuclear exclusion of FKHR[S256A] depends on the degree of overexpression of this mutant. Our results indicated that the interaction of FKHR with 14-3-3 proteins was not required for IGF-1-stimulated exclusion of FKHR from the nucleus. We present evidence in support of another mechanism, which depends on the phosphorylation of Ser-256 and may involve the masking of a nuclear localization signal. Finally, we have demonstrated that the failure of IGF-1 to suppress transactivation by FKHR[S256A] is not explained entirely by its failure to bind 14-3-3 proteins or to undergo nuclear exclusion. This result suggests that Ser-256 phosphorylation may also suppress transactivation by FKHR by yet another mechanism, perhaps by disrupting the interaction of FKHR with target DNA binding sites and/or the function of the transactivation domain.
- Published
- 2001
- Full Text
- View/download PDF
438. Identification and characterization of the human homologue of the short PDE4A cAMP-specific phosphodiesterase RD1 (PDE4A1) by analysis of the human HSPDE4A gene locus located at chromosome 19p13.2.
- Author
-
Sullivan M, Rena G, Begg F, Gordon L, Olsen AS, and Houslay MD
- Subjects
- 3',5'-Cyclic-AMP Phosphodiesterases metabolism, Alternative Splicing, Amino Acid Sequence, Animals, Base Sequence, Cats, Chromosome Mapping, Exons, Humans, Introns, Isoenzymes metabolism, Mice, Molecular Sequence Data, RNA, Messenger metabolism, Rabbits, Rats, Sequence Homology, Amino Acid, Sequence Homology, Nucleic Acid, Swine, 3',5'-Cyclic-AMP Phosphodiesterases genetics, Chromosomes, Human, Pair 19, Isoenzymes genetics
- Abstract
The HSPDE4A gene spans 50 kb, consists of at least 17 exons and is orientated 5'-3', telomere to centromere. It is located at chromosome 19p13.2, being 350 kb proximal to the gene encoding TYK2 and 850 kb distal to the gene encoding the low-density lipoprotein receptor. Its structure is consistent with the production of active 'long' and 'short' isoenzymes as the result of alternative mRNA splicing at two splice junctions. Identified is the single alternatively spliced 5' exon encoding the unique N-terminal region of the long isoenzyme HSPDE4A4B (pde46). The upstream conserved regions, UCR1 and UCR2, which form characteristic domains of PDE4 long forms are each encoded by three exons. The PDE4A-subfamily-specific linker region LR1, which joins UCR1 and UCR2, is encoded by two exons, whereas LR2, which joins UCR2 to the catalytic unit, is encoded by a single exon. Identification of exons encoding an enzymically inactive product of this gene, HSPDE4A8A (2el), indicates that this is an authentic gene product. The 5' exon encoding the unique N-terminal region of the human homologue of the rodent isoform RNPDE4A1A (RD1) was located, and the splice junction used to produce this short PDE4A isoform shown to occur at a different position from that seen in both the rat PDE4B and PDE4D genes. Reverse transcriptase PCR analysis indicates that RD1 homologues are conserved across species, having a conserved membrane-targeting region and a hypervariable LR2 region. Human RD1 was expressed transiently in COS-7 cells and detected as an 83 kDa species primarily associated with the high-speed membrane fraction. Human RD1 exhibited a Km for cAMP of about 3 microM, an IC50 value for inhibition by the PDE4-selective inhibitor rolipram of about 0.3 microM and was considerably more thermostable than rat RD1. Human RD1 was generated as a mature 80 kDa species in an in vitro transcription-translation system and shown to be capable of binding to membranes. Knowledge of the gene structure and the associated sequence information should facilitate analysis of the involvement of PDE4A in hereditary disorders that may result from alterations in enzyme expression, activity, regulation and intracellular targeting and serve as a resource for determining authenticity of cloned PDE4A species.
- Published
- 1998
- Full Text
- View/download PDF
439. Upregulation of cAMP-specific PDE-4 activity following ligation of the TCR complex on thymocytes is blocked by selective inhibitors of protein kinase C and tyrosyl kinases.
- Author
-
Michie AM, Rena G, Harnett MM, and Houslay MD
- Subjects
- 3',5'-Cyclic-AMP Phosphodiesterases antagonists & inhibitors, 3',5'-Cyclic-AMP Phosphodiesterases genetics, Adenylyl Cyclases metabolism, Alkaloids, Animals, Benzophenanthridines, CD3 Complex metabolism, Cells, Cultured, Cyclic AMP analysis, Cyclic Nucleotide Phosphodiesterases, Type 4, Cycloheximide pharmacology, Dactinomycin pharmacology, Enzyme Activation, Enzyme Inhibitors pharmacology, Mice, Mice, Inbred BALB C, Nucleic Acid Synthesis Inhibitors pharmacology, Phenanthridines pharmacology, Protein Kinase C antagonists & inhibitors, Protein Synthesis Inhibitors pharmacology, Protein-Tyrosine Kinases antagonists & inhibitors, Signal Transduction physiology, Staurosporine pharmacology, Tetradecanoylphorbol Acetate pharmacology, Thymus Gland cytology, Up-Regulation physiology, 3',5'-Cyclic-AMP Phosphodiesterases metabolism, Protein Kinase C physiology, Protein-Tyrosine Kinases physiology, Receptors, Antigen, T-Cell metabolism, Thymus Gland enzymology
- Abstract
We have previously shown that the major cAMP phosphodiesterase (PDE) isoforms present in murine thymocytes are the cGMP-stimulated PDE activity (PDE-2) and the cAMP-specific PDE activity (PDE-4), and that these isoforms are differentially regulated following ligation of the TCR (Michie, A.M., Lobban, M. D., Mueller, T., Harnett, M. M., and Houslay, M.D. [1996] Cell. Signalling 8, 97-110). We show here that the anti-CD3-stimulated elevation in PDE-4 activity in murine thymocytes is dependent on protein tyrosine kinase and protein kinase C (PKC)-mediated signals as the TCR-coupled increase in PDE-4 activity can be abrogated by both the tyrosine kinase inhibitor, genistein, and the PKC selective inhibitors chelerythrine and staurosporine. Moreover, the PKC-activating phorbol ester, phorbol-12-myristate, 13-acetate (PMA) caused an increase in PDE-4 activity, similar to that observed in cells challenged with anti-CD3 monoclonal antibodies and which was not additive with cochallenge using anti-CD3 antibodies. Both the PMA- and the anti-CD3 antibody-mediated increases in PDE-4 activity were blocked by treatment with either cycloheximide or actinomycin D. Despite the upregulation of PDE-4 activity consequent to TCR ligation, intracellular cAMP levels increased on challenge of thymocytes with anti-CD3 antibody, indicating that adenylate cyclase activity was also increased by TCR ligation. It is suggested that the anti-CD3-mediated increase in PDE-4 activity was owing to a rapid PKC-dependent induction of PDE-4 activity following crosslinking of the TCR complex. This identifies "crosstalk" occurring between the PKA and PKC signaling pathways initiated by ligation of the antigen receptor in murine thymocytes. That both adenylate cyclase and PDE-4 activities were increased may indicate the presence of compartmentalized cAMP responses present in these cells.
- Published
- 1998
- Full Text
- View/download PDF
440. Receptor-mediated stimulation of lipid signalling pathways in CHO cells elicits the rapid transient induction of the PDE1B isoform of Ca2+/calmodulin-stimulated cAMP phosphodiesterase.
- Author
-
Spence S, Rena G, Sullivan M, Erdogan S, and Houslay MD
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, CHO Cells, Cattle, Cricetinae, Cyclic Nucleotide Phosphodiesterases, Type 1, Dactinomycin pharmacology, Enzyme Induction, Humans, Jurkat Cells, Lipid Metabolism, Molecular Sequence Data, Polymerase Chain Reaction, Sequence Analysis, DNA, Transfection, 3',5'-Cyclic-AMP Phosphodiesterases chemistry, Isoenzymes chemistry, Phosphoric Diester Hydrolases
- Abstract
Chinese hamster ovary cells (CHO cells) do not exhibit any Ca2+/calmodulin-stimulated cAMP phosphodiesterase (PDE1) activity. Challenge of CHO cells with agonists for endogenous P2-purinoceptors, lysophosphatidic acid receptors and thrombin receptors caused a similar rapid transient induction of PDE1 activity in each instance. This was also evident on noradrenaline challenge of a cloned CHO cell line transfected so as to overexpress alpha 1B-adrenoceptors. This novel PDE1 activity appeared within about 15 min of exposure to ligands, rose to a maximum value within 30 min to 1 h and then rapidly decreased. In each case, the expression of novel PDE1 activity was blocked by the transcriptional inhibitor actinomycin D. Challenge with insulin of either native CHO cells or a CHO cell line transfected so as to overexpress the human insulin receptor failed to induce PDE1 activity. Reverse transcriptase-PCR analyses, using degenerate primers able to detect the PDE1C isoform, did not amplify any fragment from RNA preparations of CHO cells expressing PDE1 activity, although they did so from the human thyroid carcinoma FTC133 cell line. Reverse transcriptase-PCR analyses, using degenerate primers able to detect the PDE1A and PDE1B isoforms, successfully amplified a fragment of the predicted size from RNA preparations of both CHO cells expressing PDE1 activity and human Jurkat T-cells. Sequencing of the PCR products, generated using the PDE1A/B primers, yielded a novel sequence which, by analogy with sequences reported for bovine and murine PDE1B forms, suggests that the PDE1 species induced in CHO cells through protein kinase C activation and that expressed in Jurkat T-cells are PDE1B forms.
- Published
- 1997
- Full Text
- View/download PDF
441. Intracellular localization of the PDE4A cAMP-specific phosphodiesterase splice variant RD1 (RNPDE4A1A) in stably transfected human thyroid carcinoma FTC cell lines.
- Author
-
Pooley L, Shakur Y, Rena G, and Houslay MD
- Subjects
- Amino Acid Sequence, Base Sequence, Cyclic Nucleotide Phosphodiesterases, Type 1, Cyclic Nucleotide Phosphodiesterases, Type 4, Fluorescent Antibody Technique, Humans, Molecular Sequence Data, Octoxynol, Phosphodiesterase Inhibitors pharmacology, Transfection, Tumor Cells, Cultured, 3',5'-Cyclic-AMP Phosphodiesterases metabolism, Phosphoric Diester Hydrolases metabolism, RNA Splicing, Thyroid Neoplasms enzymology
- Abstract
Cells of two human follicular thyroid carcinoma cell lines (FTC133, FTC236) were stably transfected with a cDNA encoding the PDE4A cAMP-specific phosphodiesterase (PDE) splice variant RD1 (RNPDE4A1A) so as to generate the cloned cell lines, FTC133A and FTC236A. This allowed the expression of a novel rolipram-inhibited cAMP-specific PDE activity in these cells. Unlike the parent cell lines in which Ca2+/calmodulin caused a profound activation (approx. 3-4-fold) of homogenate PDE activity, no such stimulation was evident in the RD1-expressing cell lines, indicating loss of PDE1 activity. Reverse transcriptase-PCR analysis indicated that this was due to the down-regulation of the PDE1C isoform. The novel PDE4 activity in transfected cells was located exclusively in the membrane fraction, as was immunoreactive RD1. Low concentrations of the detergent Triton X-100, but not high NaCl concentrations, allowed RD1 to be solubilized. Laser scanning confocal immunofluorescence analyses identified RD1 immunoreactivity in a discrete perinuclear region of these RD1-expressing transfected cell lines. A similar pattern of labelling was observed using the antiserum Tex1, which specifically identified the Golgi apparatus. Treatment of FTC133A cells with the Golgi-perturbing agents monensin and brefeldin A led to a similar redistribution of immunoreactive species detected using both the Tex1 and anti-RD1 antisera. It is suggested that the PDE4A splice variant RD1 contains a membrane-association signal which allows the targeted expression of RD1 within the Golgi complex of these human follicular thyroid carcinoma cell lines.
- Published
- 1997
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.