Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 321
Filter
1.
Int J Biol Macromol ; 264(Pt 2): 130799, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38479663

ABSTRACT

The 3-quinuclidinone reductase plays an irreplaceable role in the biopreparation of (R)-3-quinuclidinol, an intermediate vital for synthesis of various pharmaceuticals. Thermal robustness is a critical factor for enzymatic synthesis in industrial applications. This study characterized a new 3-quinuclidinone reductase, named SaQR, with significant thermal stability. The SaQR was overexpressed in a GST-fused state, and substrate and cofactor screening were conducted. Additionally, three-dimensional structure prediction using AlphaFold and analysis were performed, along with relevant thermostability tests, and the evaluation of factors influencing enzyme activity. The findings highlight the remarkable thermostability of SaQR, retaining over 90% of its activity after 72 h at 50°C, with an optimal operational temperature of 85°C. SaQR showed typical structural traits of the SDR superfamily, with its cofactor-determining residue being aspartic acid, conferring nicotinamide adenine dinucleotide (NAD(H)) preference. Moreover, K+ and Na+, at a concentration of 400 mM, could significantly enhance the activity, while Mg2+ and Mn2+ only display inhibitory effects within the tested concentration range. The findings of molecular dynamics simulations suggest that high temperatures may disrupt the binding of enzyme to substrate by increasing the flexibility of residues 205-215. In conclusion, this study reports a novel 3-quinuclidinone reductase with remarkable thermostability.


Subject(s)
Oxidoreductases , Quinuclidines , Oxidoreductases/metabolism , Quinuclidines/pharmacology , Quinuclidines/metabolism , NAD/metabolism , Molecular Dynamics Simulation , Enzyme Stability
2.
J Med Chem ; 63(3): 1084-1104, 2020 02 13.
Article in English | MEDLINE | ID: mdl-31895562

ABSTRACT

In our pursuit of developing a novel, potent, and selective cell division cycle 7 (Cdc7) inhibitor, we optimized the previously reported thieno[3,2-d]pyrimidinone analogue I showing time-dependent Cdc7 kinase inhibition and slow dissociation kinetics. These medicinal chemistry efforts led to the identification of compound 3d, which exhibited potent cellular activity, excellent kinase selectivity, and antitumor efficacy in a COLO205 xenograft mouse model. However, the issue of formaldehyde adduct formation emerged during a detailed study of 3d, which was deemed an obstacle to further development. A structure-based approach to circumvent the adduct formation culminated in the discovery of compound 11b (TAK-931) possessing a quinuclidine moiety as a preclinical candidate. In this paper, the design, synthesis, and biological evaluation of this series of compounds will be presented.


Subject(s)
Antineoplastic Agents/therapeutic use , Cell Cycle Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyrazolones/therapeutic use , Pyrimidines/therapeutic use , Pyrimidinones/therapeutic use , Quinuclidines/therapeutic use , Thiophenes/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Binding Sites , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Drug Design , Drug Discovery , Formaldehyde/chemistry , Humans , Mice , Molecular Docking Simulation , Molecular Structure , Protein Binding , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/metabolism , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Pyrazolones/pharmacology , Pyrimidines/pharmacology , Pyrimidinones/chemical synthesis , Pyrimidinones/metabolism , Quinuclidines/chemical synthesis , Quinuclidines/metabolism , Structure-Activity Relationship , Thiophenes/chemical synthesis , Thiophenes/metabolism , Xenograft Model Antitumor Assays
3.
J Biol Chem ; 294(10): 3670-3682, 2019 03 08.
Article in English | MEDLINE | ID: mdl-30602570

ABSTRACT

p53 mutants can form amyloid-like structures that accumulate in cells. p53 reactivation with induction of massive apoptosis-1 (PRIMA-1) and its primary active metabolite, 2-methylene-3-quinuclidinone (MQ), can restore unfolded p53 mutants to a native conformation that induces apoptosis and activates several p53 target genes. However, whether PRIMA-1 can clear p53 aggregates is unclear. In this study, we investigated whether PRIMA-1 can restore aggregated mutant p53 to a native form. We observed that the p53 mutant protein is more sensitive to both PRIMA-1 and MQ aggregation inhibition than WT p53. The results of anti-amyloid oligomer antibody assays revealed that PRIMA-1 reverses mutant p53 aggregate accumulation in cancer cells. Size-exclusion chromatography of the lysates from mutant p53-containing breast cancer and ovarian cell lines confirmed that PRIMA-1 substantially decreases p53 aggregates. We also show that MDA-MB-231 cell lysates can "seed" aggregation of the central core domain of recombinant WT p53, corroborating the prion-like behavior of mutant p53. We also noted that this aggregation effect was inhibited by MQ and PRIMA-1. This study provides the first demonstration that PRIMA-1 can rescue amyloid-state p53 mutants, a strategy that could be further explored as a cancer treatment.


Subject(s)
Amyloid/chemistry , Aza Compounds/chemistry , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Mutation , Protein Aggregates , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Cell Line, Tumor , Humans , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Multimerization , Protein Structure, Secondary , Quinuclidines/chemistry , Quinuclidines/metabolism
4.
Sci Rep ; 8(1): 12671, 2018 08 23.
Article in English | MEDLINE | ID: mdl-30140002

ABSTRACT

The tumor suppressor p53 is commonly inactivated in human tumors, allowing evasion of p53-dependent apoptosis and tumor progression. The small molecule APR-246 (PRIMA-1Met) can reactive mutant p53 in tumor cells and trigger cell death by apoptosis. The thioredoxin (Trx) and glutaredoxin (Grx) systems are important as antioxidants for maintaining cellular redox balance and providing electrons for thiol-dependent reactions like those catalyzed by ribonucleotide reductase and peroxiredoxins (Prxs). We show here that the Michael acceptor methylene quinuclidinone (MQ), the active form of APR-246, is a potent direct inhibitor of Trx1 and Grx1 by reacting with sulfhydryl groups in the enzymes. The inhibition of Trx1 and Grx1 by APR-246/MQ is reversible and the inhibitory efficiency is dependent on the presence of glutathione. APR-246/MQ also inhibits Trxs in mutant p53-expressing Saos-2 His-273 cells, showing modification of Trx1 and mitochondrial Trx2. Inhibition of the Trx and Grx systems leads to insufficient reducing power to deoxyribonucleotide production for DNA replication and repair and peroxiredoxin for removal of ROS. We also demonstrate that APR-246 and MQ inhibit ribonucleotide reductase (RNR) in vitro and in living cells. Our results suggest that APR-246 induces tumor cell death through both reactivations of mutant p53 and inhibition of cellular thiol-dependent redox systems, providing a novel strategy for cancer therapy.


Subject(s)
Glutaredoxins/metabolism , Thioredoxins/metabolism , Antioxidants/metabolism , Blotting, Western , Cell Line, Tumor , DNA Repair/genetics , DNA Repair/physiology , Humans , Mass Spectrometry , Mitochondria/metabolism , Oxidation-Reduction , Quinuclidines/metabolism , Reactive Oxygen Species/metabolism , Ribonucleotide Reductases/metabolism , Sulfhydryl Compounds/metabolism
5.
Int J Biol Macromol ; 103: 424-434, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28526345

ABSTRACT

Ni2+-agarose bead-wrapped multi-enzyme/inorganic hybrid sphere composed of the immobilized enzymes as organic component and NaH2PO4 and NaCl as inorganic component was developed by co-immobilizing extracellular His-tagged 3-quinuclidinone reductases and glucose dehydrogenase without pre-purification. The resulting biocatalysts has 3D porous architectures as confirmed by SEM and FESEM, and it enabled the continuous biotransformation of 3-quinuclidone to (R)-3-quinuclidinol with cofactor regeneration in situ. The 3D porous biocatalysts were formed via three steps: First, immobilization of the His-tagged enzymes directly from the cell lysates supernatant. Next, formation of enzyme aggregates, ribbons and gels. Finally, the enzymes, the formed aggregates/ribbons/gels and salt were incorporated to the foam and then covered the Ni2+-agarose bead. The technique made the immobilization of these enzymes effective such that specific enzyme loading of 60.8mg/g support and enzyme loading efficiency of 92.3% were achieved. As a direct consequence, the biocatalyst catalyzed the conversion of 3-quinuclidinone (204g/L) to (R)-3-quinuclidinol in 100% yield and 100% ee at 4.5h, and the recyclability of the biocatalyst was excellent, retaining>95% conversion yield and 100% ee even after the fifteenth runs. Overall, our strategy is demonstrated to be a promising method for developing efficient and robust biocatalyst for asymmetric synthesis.


Subject(s)
Biocatalysis , Coenzymes/metabolism , Enzymes, Immobilized/chemistry , Enzymes, Immobilized/metabolism , Biotransformation , Oxidation-Reduction , Porosity , Quinuclidines/metabolism
6.
Oncotarget ; 7(50): 83017-83030, 2016 Dec 13.
Article in English | MEDLINE | ID: mdl-27806324

ABSTRACT

PRIMA-1Met is the methylated PRIMA-1 (p53 reactivation and induction of massive apoptosis) and could restore tumor suppressor function of mutant p53 and induce p53 dependent apoptosis in cancer cells harboring mutant p53. However, p53 independent activity of PRIMA-1Met remains elusive. Here we reported that PRIMA-1Met attenuated colorectal cancer cell growth irrespective of p53 status. Kinase profiling revealed that mitogen-activated or extracellular signal-related protein kinase (MEK) might be a potential target of PRIMA-1Met. Pull-down binding and ATP competitive assay showed that PRIMA-1Met directly bound MEK in vitro and in cells. Furthermore, the direct binding sites of PRIMA-1Met were explored by using a computational docking model. Treatment of colorectal cancer cells with PRIMA-1Met inhibited p53-independent phosphorylation of MEK, which in turn impaired anchorage-independent cell growth in vitro. Moreover, PRIMA-1Met suppressed colorectal cancer growth in xenograft mouse model by inhibiting MEK1 activity.Taken together, our findings demonstrate a novel p53-independent activity of PRIMA-1Met to inhibit MEK and suppress colorectal cancer growth.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , MAP Kinase Kinase Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Quinuclidines/pharmacology , Tumor Suppressor Protein p53/metabolism , Animals , Antineoplastic Agents/metabolism , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Dose-Response Relationship, Drug , HCT116 Cells , HT29 Cells , Humans , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase Kinases/metabolism , Mice, Nude , Molecular Docking Simulation , Phosphorylation , Protein Binding , Protein Kinase Inhibitors/metabolism , Quinuclidines/metabolism , Signal Transduction/drug effects , Time Factors , Tumor Burden/drug effects , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays
7.
Int J Pharm ; 511(2): 1088-97, 2016 Sep 25.
Article in English | MEDLINE | ID: mdl-27521703

ABSTRACT

Palonosetron (PAL) is recommended for the prevention of chemotherapy-induced nausea and vomiting. The aim of this study was to develop a long-acting PAL transdermal patch to improve patient compliance. We were particularly concerned about the effect of pressure sensitive adhesives (PSAs) on PAL skin permeability. Formulation factors including PSAs, backing films and drug loadings were investigated in the in vitro skin permeation study using rabbit skin. Fourier transform infrared spectrometer study and thermal analysis were conducted to investigate the drug-PSA interaction and thermodynamic activity of PSAs, respectively. The results indicated that high drug skin permeation amount was obtained in PSA DURO-TAK(®)87-2516, which had low interaction potential with PAL and high thermodynamic activity. The optimized patch was composed of PAL of 8 %, DURO-TAK(®)87-2516 as PSA, CoTran™ 9700 as backing film and Scotchpak™ 9744 as release liner. The in vitro skin permeation amount of the optimized patch was 734.0±55.8µg/cm(2) during 3-day administration. The absolute bioavailability of the optimized patch was 43 % in rabbit and a good in vitro-in vivo correlation coefficient was obtained (R(2)=0.989). These results indicated the feasibility of PAL transdermal patch in the prevention of chemotherapy-induced nausea and vomiting.


Subject(s)
Adhesives/administration & dosage , Isoquinolines/administration & dosage , Quinuclidines/administration & dosage , Skin Absorption/drug effects , Transdermal Patch , Adhesives/chemistry , Adhesives/metabolism , Administration, Cutaneous , Animals , Isoquinolines/chemistry , Isoquinolines/metabolism , Male , Organ Culture Techniques , Palonosetron , Pressure , Quinuclidines/chemistry , Quinuclidines/metabolism , Rabbits , Random Allocation , Skin Absorption/physiology
8.
Neuropharmacology ; 110(Pt A): 102-108, 2016 11.
Article in English | MEDLINE | ID: mdl-27422408

ABSTRACT

Neural α7 nicotinic acetylcholine receptors (α7 nAChRs) emerged as a potential pharmacologic target for treating cognitive deficits in schizophrenia and Alzheimer's disease. Experiments modeling these dysfunctions, as well as clinical evidence, demonstrate the relatively consistent procognitive effects of α7 nAChR agonists. One preclinical observation supporting the procognitive role of α7 nAChRs is their ability to modulate neuronal network oscillations closely associated with learning and memory, especially hippocampal oscillations. Due to the high degree of structural similarity between α7 nACh and 5-HT receptors, the majority of α7 nAChR agonists to date also act as 5-HT3 antagonists. To address this confounding property and determine the relevance of α7 nAChR agonist binding to 5-HT3 receptors in modulating hippocampal activity, we tested two well-described α7 nAChR agonists, PNU-282987 and FRM-17874, in mice lacking α7 nAChRs (α7 knock-out, α7KO) using the brainstem simulation-elicited hippocampal theta oscillation assay. Under urethane anesthesia both agonists at equivalent doses demonstrated efficacy in wild-type (WT) mice, significantly enhancing theta power and theta phase-gamma amplitude coupling as compared to saline treated control mice. These effects are comparable to those seen with drugs clinically used to treat Alzheimer's disease. Although α7KO mice showed no alterations in elicited hippocampal oscillations, both α7 nAChR agonists failed to enhance theta power or theta phase - gamma amplitude coupling in these mice. Our findings demonstrate that selective activation of α7 nAChRs can modulate hippocampal oscillation, and these receptors are the primary targets of the tested agonists, PNU-282987 and FRM-17874 and likely underlies their observed procognitive activity.


Subject(s)
Hippocampus/physiology , Nicotinic Agonists/metabolism , Theta Rhythm/physiology , alpha7 Nicotinic Acetylcholine Receptor/agonists , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Animals , Benzamides/metabolism , Benzamides/pharmacology , Bridged Bicyclo Compounds/metabolism , Bridged Bicyclo Compounds/pharmacology , Hippocampus/drug effects , Male , Mice , Mice, Knockout , Nicotinic Agonists/pharmacology , Protein Binding/physiology , Quinuclidines/metabolism , Quinuclidines/pharmacology , Theta Rhythm/drug effects , Thiophenes/metabolism , Thiophenes/pharmacology
9.
Respir Med ; 114: 123-6, 2016 05.
Article in English | MEDLINE | ID: mdl-27109822

ABSTRACT

BACKGROUND: Treatment with long-acting ß2-agonists (LABAs) and long-acting muscarinic antagonists (LAMAs) for chronic obstructive pulmonary disease (COPD) is standard, but response varies. We investigated genetic association with treatment response to umeclidinium (UMEC, a LAMA), vilanterol (VI, a LABA), and combination therapy. METHODS: Data from 17 clinical trials (N = 6075) in patients with COPD receiving once-daily UMEC/VI (125/25mcg or 62.5/25mcg), UMEC (125 or 62.5mcg), VI (25mcg) or placebo were used. Genetic association with change from baseline in trough forced expiratory volume in 1 s (FEV1) ∼24 h post-dosing was assessed for: (i) 3 ß2-adrenoceptor (ADRB2) gene variants; (ii) 298 single-nucleotide polymorphisms (SNPs) with prior evidence of associations; (iii) human leukocyte antigen (HLA) alleles and (iv) genome-wide association study (GWAS) SNPs. Other endpoints were (i) reversibility at screening; and at baseline: (ii) FEV1; (iii) forced vital capacity (FVC), and (iv) FEV1/FVC ratio. Using linear regression, the inverse normal transformed residuals were pooled together, first across treatment group, then across studies for each monotherapy, then combination therapy and finally for every treated patient. RESULTS: Of 6075 patients, 1849 received UMEC/VI, 1390 received UMEC, 1795 received VI, and 1041 received placebo. None of the ADRB2 variants, HLA alleles or GWAS variants tested were associated with treatment response or baseline endpoints. Four SNPs in FAM13A (rs7671167, rs2869967, rs1964516, rs1903003) were significantly associated with baseline FEV1/FVC ratio (p < 3 × 10(-5)) after adjusting for multiple testing. CONCLUSIONS: No genetic association was found with treatment response to UMEC or VI when administered as monotherapies or in combination.


Subject(s)
Benzyl Alcohols/metabolism , Chlorobenzenes/metabolism , Forced Expiratory Volume/genetics , Pulmonary Disease, Chronic Obstructive/drug therapy , Quinuclidines/metabolism , Vital Capacity/genetics , Administration, Inhalation , Adrenergic beta-2 Receptor Agonists/therapeutic use , Aged , Benzyl Alcohols/administration & dosage , Benzyl Alcohols/pharmacology , Bronchodilator Agents/therapeutic use , Chlorobenzenes/administration & dosage , Chlorobenzenes/pharmacology , Clinical Trials as Topic , Drug Combinations , Female , Forced Expiratory Volume/drug effects , Genome-Wide Association Study , HLA Antigens/genetics , Humans , Male , Middle Aged , Muscarinic Antagonists/therapeutic use , Pharmacogenetics , Polymorphism, Single Nucleotide/genetics , Pulmonary Disease, Chronic Obstructive/physiopathology , Quinuclidines/administration & dosage , Quinuclidines/pharmacology , Treatment Outcome , Vital Capacity/drug effects
10.
Cancer Biol Ther ; 16(5): 799-806, 2015.
Article in English | MEDLINE | ID: mdl-25803193

ABSTRACT

PRIMA-1Met has shown promising preclinical activity in various cancer types. However, its effect on Waldenström's Macroglobulinemia (WM) cells as well as its exact mechanism of action is still elusive. In this study, we evaluated the anti- tumor activity of PRIMA-1Met alone and in combination with dexamethasone or bortezomib in WM cell lines and primary samples. Treatment of WM cells with PRIMA-1Met resulted in induction of apoptosis, inhibition of migration and suppression of colony formation. Upon PRIMA-1Met treatment, p73 was upregulated and Bcl-xL was down-regulated while no significant change in expression of p53 was observed. Furthermore, siRNA knockdown of p53 in WM cell line did not influence the PRIMA-1Met-induced apoptotic response whereas silencing of p73 inhibited latter response in WM cells. Importantly, combined treatment of BCWM-1 cells with PRIMA-1Met and dexamethasone or bortezomib induced synergistic reduction in cell survival. Our study provides insights into the mechanisms of anti-WM activity of PRIMA-1Met and supports further clinical evaluation of PRIMA-1Met as a potential novel therapeutic intervention in WM.


Subject(s)
Quinuclidines/metabolism , Waldenstrom Macroglobulinemia/drug therapy , Apoptosis , Cell Line, Tumor , Humans , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
11.
Int J Mol Sci ; 15(9): 15994-6011, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25216334

ABSTRACT

Angiogenesis is the growth of new capillaries from existing blood vessels that supply oxygen and nutrients and provide gateways for immune surveillance. Abnormal vessel growth in term of excessive angiogenesis is a hallmark of cancer, inflammatory and eye diseases. VEGFR-2 (vascular endothelial growth factor receptor 2) dominating the process of angiogenesis has led to approval of therapeutic inhibitors and is becoming a promising target for anti-angiogenic drugs. Notwithstanding these successes, the clinical use of current VEGFR-2 blockers is more challenging than anticipated. Taking axitinib as a reference drug, in our study we found three potent VEGFR-2 inhibitors (ZINC08254217, ZINC08254138, and ZINC03838680) from natural derivatives. Each of the three inhibitors acquired a better grid score than axitinib (-62.11) when docked to VEGFR-2. Molecular dynamics simulations demonstrated that ZINC08254217- and ZINC08254138-VEGFR-2 complexes were more stable than axitinib. Similar to bind free energy for axitinib (-54.68 kcal/mol), such for ZINC03838680, ZINC08254217, and ZINC08254138 was -49.37, -43.32, and -32.73 kcal/mol respectively. These results suggested these three compounds could be candidate drugs against angiogenesis, with comparable VEGFR-2 binding affinity of axitinib. Hence findings in our study are able to provide valuable information on discovery of effective anti-angiogenesis therapy.


Subject(s)
Angiogenesis Inhibitors/chemistry , Quinuclidines/chemistry , Triazoles/chemistry , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Angiogenesis Inhibitors/metabolism , Axitinib , Binding Sites , Imidazoles/chemistry , Imidazoles/metabolism , Indazoles/chemistry , Indazoles/metabolism , Molecular Docking Simulation , Protein Structure, Tertiary , Quinuclidines/metabolism , Thermodynamics , Triazoles/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
12.
Chembiochem ; 15(14): 2050-2, 2014 Sep 22.
Article in English | MEDLINE | ID: mdl-25169816

ABSTRACT

We present the development of highly selective variants of the Parvibaculum lavamentivorans alcohol dehydrogenase. Four amino acids (A158, N162, K202, L224) in the second sphere of the catalytic site were identified to determine the selectivity for 3-quinuclidone reduction significantly. The best variant (A158H/N162G/K202Q/L224W) was able to increase the ee for (R)-3-quinuclidinol production from 84.3 % (wild-type) to ≥99 % and concomitantly to enhance conversion by 43.5 %.


Subject(s)
Alcohol Dehydrogenase/genetics , Alphaproteobacteria/enzymology , Protein Engineering , Quinuclidines/metabolism , Alcohol Dehydrogenase/chemistry , Alcohol Dehydrogenase/metabolism , Alphaproteobacteria/chemistry , Alphaproteobacteria/genetics , Alphaproteobacteria/metabolism , Catalytic Domain , Models, Molecular , Mutation , Oxidation-Reduction
13.
Eur J Pharmacol ; 737: 140-8, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24858620

ABSTRACT

Our laboratory recently reported that a group of novel indole quinuclidine analogs bind with nanomolar affinity to cannabinoid type-1 and type-2 receptors. This study characterized the intrinsic activity of these compounds by determining whether they exhibit agonist, antagonist, or inverse agonist activity at cannabinoid type-1 and/or type-2 receptors. Cannabinoid receptors activate Gi/Go-proteins that then proceed to inhibit activity of the downstream intracellular effector adenylyl cyclase. Therefore, intrinsic activity was quantified by measuring the ability of compounds to modulate levels of intracellular cAMP in intact cells. Concerning cannabinoid type-1 receptors endogenously expressed in Neuro2A cells, a single analog exhibited agonist activity, while eight acted as neutral antagonists and two possessed inverse agonist activity. For cannabinoid type-2 receptors stably expressed in CHO cells, all but two analogs acted as agonists; these two exceptions exhibited inverse agonist activity. Confirming specificity at cannabinoid type-1 receptors, modulation of adenylyl cyclase activity by all proposed agonists and inverse agonists was blocked by co-incubation with the neutral cannabinoid type-1 antagonist O-2050. All proposed cannabinoid type-1 receptor antagonists attenuated adenylyl cyclase modulation by cannabinoid agonist CP-55,940. Specificity at cannabinoid type-2 receptors was confirmed by failure of all compounds to modulate adenylyl cyclase activity in CHO cells devoid of cannabinoid type-2 receptors. Further characterization of select analogs demonstrated concentration-dependent modulation of adenylyl cyclase activity with potencies similar to their respective affinities for cannabinoid receptors. Therefore, indole quinuclidines are a novel structural class of compounds exhibiting high affinity and a range of intrinsic activity at cannabinoid type-1 and type-2 receptors.


Subject(s)
Indoles/chemistry , Quinuclidines/metabolism , Quinuclidines/pharmacology , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism , Adenylyl Cyclase Inhibitors , Adenylyl Cyclases/metabolism , Animals , CHO Cells , Chemical Phenomena , Cricetinae , Cricetulus , Drug Inverse Agonism , Humans , Ligands , Mice , Quinuclidines/chemistry , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/antagonists & inhibitors
14.
PLoS Pathog ; 10(5): e1004114, 2014 May.
Article in English | MEDLINE | ID: mdl-24789335

ABSTRACT

Trypanosomatid parasites are the causative agents of many neglected tropical diseases and there is currently considerable interest in targeting endogenous sterol biosynthesis in these organisms as a route to the development of novel anti-infective drugs. Here, we report the first x-ray crystallographic structures of the enzyme squalene synthase (SQS) from a trypanosomatid parasite, Trypanosoma cruzi, the causative agent of Chagas disease. We obtained five structures of T. cruzi SQS and eight structures of human SQS with four classes of inhibitors: the substrate-analog S-thiolo-farnesyl diphosphate, the quinuclidines E5700 and ER119884, several lipophilic bisphosphonates, and the thiocyanate WC-9, with the structures of the two very potent quinuclidines suggesting strategies for selective inhibitor development. We also show that the lipophilic bisphosphonates have low nM activity against T. cruzi and inhibit endogenous sterol biosynthesis and that E5700 acts synergistically with the azole drug, posaconazole. The determination of the structures of trypanosomatid and human SQS enzymes with a diverse set of inhibitors active in cells provides insights into SQS inhibition, of interest in the context of the development of drugs against Chagas disease.


Subject(s)
Chagas Disease/drug therapy , Enzyme Inhibitors/therapeutic use , Farnesyl-Diphosphate Farnesyltransferase/antagonists & inhibitors , Molecular Targeted Therapy/methods , Trypanocidal Agents/therapeutic use , Animals , Chlorocebus aethiops , Crystallography, X-Ray , Diphosphonates/chemistry , Diphosphonates/metabolism , Diphosphonates/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Farnesyl-Diphosphate Farnesyltransferase/chemistry , Farnesyl-Diphosphate Farnesyltransferase/metabolism , Humans , Models, Molecular , Polyisoprenyl Phosphates/chemistry , Polyisoprenyl Phosphates/metabolism , Protein Binding , Quinuclidines/chemistry , Quinuclidines/metabolism , Quinuclidines/pharmacology , Sesquiterpenes/chemistry , Sesquiterpenes/metabolism , Trypanocidal Agents/chemistry , Trypanocidal Agents/metabolism , Trypanocidal Agents/pharmacology , Trypanosoma cruzi/enzymology , Vero Cells
15.
Biochem Biophys Res Commun ; 446(4): 911-5, 2014 Apr 18.
Article in English | MEDLINE | ID: mdl-24642255

ABSTRACT

(R)-3-Quinuclidinol, a useful compound for the synthesis of various pharmaceuticals, can be enantioselectively produced from 3-quinuclidinone by 3-quinuclidinone reductase. Recently, a novel NADH-dependent 3-quinuclidionone reductase (AtQR) was isolated from Agrobacterium tumefaciens, and showed much higher substrate-binding affinity (>100 fold) than the reported 3-quinuclidionone reductase (RrQR) from Rhodotorula rubra. Here, we report the crystal structure of AtQR at 1.72 Å. Three NADH-bound protomers and one NADH-free protomer form a tetrameric structure in an asymmetric unit of crystals. NADH not only acts as a proton donor, but also contributes to the stability of the α7 helix. This helix is a unique and functionally significant part of AtQR and is related to form a deep catalytic cavity. AtQR has all three catalytic residues of the short-chain dehydrogenases/reductases family and the hydrophobic wall for the enantioselective reduction of 3-quinuclidinone as well as RrQR. An additional residue on the α7 helix, Glu197, exists near the active site of AtQR. This acidic residue is considered to form a direct interaction with the amine part of 3-quinuclidinone, which contributes to substrate orientation and enhancement of substrate-binding affinity. Mutational analyses also support that Glu197 is an indispensable residue for the activity.


Subject(s)
Agrobacterium tumefaciens/enzymology , Oxidoreductases/chemistry , Oxidoreductases/metabolism , Quinuclidines/metabolism , Agrobacterium tumefaciens/chemistry , Agrobacterium tumefaciens/metabolism , Binding Sites , Crystallography, X-Ray , Models, Molecular , NAD/metabolism , Protein Conformation , Stereoisomerism , Substrate Specificity
16.
Biochemistry ; 52(47): 8480-8, 2013 Nov 26.
Article in English | MEDLINE | ID: mdl-24164482

ABSTRACT

The differential action of the novel agonist JN403 at neuronal α7 and muscle nicotinic receptors (AChRs) was explored by using a combination of functional and structural approaches. Single-channel recordings reveal that JN403 is a potent agonist of α7 but a very low-efficacy agonist of muscle AChRs. JN403 elicits detectable openings of α7 and muscle AChRs at concentrations ~1000-fold lower and ~20-fold higher, respectively, than that for ACh. Single-channel activity elicited by JN403 is very similar to that elicited by ACh in α7 but profoundly different in muscle AChRs, where openings are brief and infrequent and do not appear in clusters at any concentration. JN403 elicits single-channel activity of muscle AChRs lacking the ε subunit, with opening events being more frequent and prolonged than those of wild-type AChRs. This finding is in line with the molecular docking studies predicting that JN403 may form a hydrogen bond required for potent activation at the α-δ but not at the α-ε binding site. JN403 does not elicit detectable Ca²âº influx in muscle AChRs but inhibits (±)-epibatidine-elicited influx mainly by a noncompetitive mechanism. Such inhibition is compatible with single-channel recordings revealing that JN403 produces open-channel blockade and early termination of ACh-elicited clusters, and it is therefore also a potent desensitizing enhancer of muscle AChRs. The latter mechanism is supported by the JN403-induced increase in the level of binding of [³H]cytisine and [³H]TCP to resting AChRs. Elucidation of the differences in activity of JN403 between neuronal α7 and muscle AChRs provides further insights into mechanisms underlying selectivity for α7 AChRs.


Subject(s)
Carbamates/pharmacology , Muscle Proteins/agonists , Nerve Tissue Proteins/agonists , Nicotinic Agonists/pharmacology , Quinuclidines/pharmacology , Receptors, Nicotinic/metabolism , alpha7 Nicotinic Acetylcholine Receptor/agonists , Animals , Calcium Signaling/drug effects , Carbamates/metabolism , Cell Line , Fetal Proteins/agonists , Fetal Proteins/chemistry , Fetal Proteins/genetics , Fetal Proteins/metabolism , Humans , Kinetics , Membrane Potentials/drug effects , Mice , Molecular Conformation , Molecular Docking Simulation , Muscle Proteins/chemistry , Muscle Proteins/genetics , Muscle Proteins/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nicotinic Agonists/metabolism , Nicotinic Antagonists/chemistry , Nicotinic Antagonists/metabolism , Nicotinic Antagonists/pharmacology , Protein Binding , Protein Subunits/agonists , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Quinuclidines/metabolism , Receptors, Nicotinic/chemistry , Receptors, Nicotinic/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Torpedo , alpha7 Nicotinic Acetylcholine Receptor/chemistry , alpha7 Nicotinic Acetylcholine Receptor/genetics , alpha7 Nicotinic Acetylcholine Receptor/metabolism
17.
Br J Pharmacol ; 169(6): 1252-62, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23581504

ABSTRACT

BACKGROUND AND PURPOSE: The 5-HT3 receptor antagonist palonosetron is an important treatment for emesis and nausea during cancer therapy. Its clinical efficacy may result from its unique binding and clearance characteristics and receptor down-regulation mechanisms. We investigated the mechanisms by which palonosetron exerts its long-term inhibition of 5-HT3 receptors for a better understanding of its clinical efficacy. EXPERIMENTAL APPROACH: Cell surface receptors (recombinantly expressed 5HT3A or 5HT3AB in COS-7 cells) were monitored using [³H]granisetron binding and ELISA after exposure to palonosetron. Receptor endocytosis was investigated using immunofluorescence microscopy. KEY RESULTS: Chronic exposure to palonosetron reduced the number of available cell surface [³H]granisetron binding sites. This down-regulation was not sensitive to either low temperature or pharmacological inhibitors of endocytosis (dynasore or nystatin) suggesting that internalization did not play a role. This was corroborated by our observation that there was no change in cell surface 5-HT3 receptor levels or increase in endocytic rate. Palonosetron exhibited slow dissociation from the receptor over many hours, with a significant proportion of binding sites being occupied for at least 4 days. Furthermore, our observations suggest that chronic receptor down-regulation involved interactions with an allosteric binding site. CONCLUSIONS AND IMPLICATIONS: Palonosetron acts as a pseudo-irreversible antagonist causing prolonged inhibition of 5-HT3 receptors due to its very slow dissociation. In addition, an irreversible binding mode persists for at least 4 days. Allosteric receptor interactions appear to play a role in this phenomenon.


Subject(s)
Antiemetics/pharmacology , Cell Membrane/drug effects , Down-Regulation/drug effects , Isoquinolines/pharmacology , Quinuclidines/pharmacology , Receptors, Serotonin, 5-HT3/metabolism , Serotonin 5-HT3 Receptor Antagonists/pharmacology , Allosteric Regulation , Animals , Antiemetics/metabolism , COS Cells , Cell Membrane/metabolism , Chlorocebus aethiops , Endocytosis/drug effects , Granisetron/metabolism , Granisetron/pharmacology , Humans , Hydrazones/pharmacology , Isoquinolines/metabolism , Kinetics , Nystatin/pharmacology , Palonosetron , Protein Transport/drug effects , Quinuclidines/metabolism , Radioligand Assay , Receptors, Serotonin, 5-HT3/chemistry , Receptors, Serotonin, 5-HT3/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Serotonin 5-HT3 Receptor Antagonists/metabolism
18.
Xenobiotica ; 43(9): 803-16, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23327533

ABSTRACT

Metabolism of ABT-107 was investigated in in vitro hepatic systems, in rat and monkey receiving [¹4C]ABT-107, and in vivo plasma in rat, dog, monkey and human. In in vitro hepatic systems, ABT-107 was primarily cleared via oxidative metabolism, and proceeded via two parallel pathways. Pathway 1, ABT-107 was oxidized at the nitrogen of quinuclidine moiety to form M1. Pathway 2, oxidation occurred at indole-containing moiety to form M2. Metabolism via N-oxidation was predominant in dog and rat, while in monkey and human, metabolism proceeded primarily via oxidation of indole-containing moiety. ABT-107 was extensively metabolized in vivo in rat and monkey. M1 was primarily found in rat urine and bile; whereas, M2 was the major metabolite in monkey urine and feces. M1 was the predominant circulating metabolite in dog and rat. M2 was the primary circulating metabolite in monkey and human. Enzymatic studies suggested M1 formation was primarily mediated by renal FMO1. CYP3A4, 1A2, 2J2 and 2D6 were primary enzymes catalyzing M2 formation. Biotransformation of ABT-107 in human and monkey is markedly different from that in dog and rat, suggesting that monkey is an appropriate model for predicting human biotransformation and toxicology of ABT-107.


Subject(s)
Indoles/metabolism , Quinuclidines/metabolism , alpha7 Nicotinic Acetylcholine Receptor/agonists , Animals , Biotransformation , Chromatography, High Pressure Liquid , Cytochrome P-450 Enzyme System/metabolism , Dogs , Female , Hepatocytes/metabolism , Humans , Indoles/blood , Indoles/chemistry , Indoles/pharmacokinetics , Macaca fascicularis , Male , Mass Spectrometry , Oxygenases/metabolism , Quinuclidines/blood , Quinuclidines/chemistry , Quinuclidines/pharmacokinetics , Rats , Species Specificity
19.
J Dermatolog Treat ; 24(6): 408-10, 2013 Dec.
Article in English | MEDLINE | ID: mdl-22646507

ABSTRACT

Biomarkers are becoming increasingly important when considering the efficacy, toxicology, mechanism of action, and risk of adverse events in certain drugs. As availability of bio-genomic information increases, more treatments can be tailored to specific individuals, with a net effect of improved health outcomes. Many dermatology drugs have pharmacogenomic information on their labels. Knowing the risks and benefits associated with genomic biomarkers can aid physicians to make more knowledgeable decisions when identifying treatments for their patients.


Subject(s)
Biomarkers/metabolism , Dermatologic Agents/metabolism , Androstenes/adverse effects , Androstenes/metabolism , Aryl Hydrocarbon Hydroxylases/drug effects , Chloroquine/adverse effects , Chloroquine/metabolism , Cytochrome P-450 CYP2C19 , Cytochrome P-450 CYP2D6/deficiency , Cytochrome P-450 CYP2D6/metabolism , Dapsone/adverse effects , Dapsone/metabolism , Dermatologic Agents/adverse effects , Dihydropyrimidine Dehydrogenase Deficiency/metabolism , Ethinyl Estradiol/adverse effects , Ethinyl Estradiol/metabolism , Fluorouracil/adverse effects , Fluorouracil/metabolism , Glucosephosphate Dehydrogenase Deficiency/metabolism , Humans , Naphthalenes/adverse effects , Naphthalenes/metabolism , Primaquine/adverse effects , Primaquine/metabolism , Quinuclidines/adverse effects , Quinuclidines/antagonists & inhibitors , Quinuclidines/metabolism , Terbinafine , Thiophenes/adverse effects , Thiophenes/antagonists & inhibitors , Thiophenes/metabolism
20.
Appl Environ Microbiol ; 79(4): 1378-84, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23263947

ABSTRACT

We used the resting-cell reaction to screen approximately 200 microorganisms for biocatalysts which reduce 3-quinuclidinone to optically pure (R)-(-)-3-quinuclidinol. Microbacterium luteolum JCM 9174 was selected as the most suitable organism. The genes encoding the protein products that reduced 3-quinuclidinone were isolated from M. luteolum JCM 9174. The bacC gene, which consists of 768 nucleotides corresponding to 255 amino acid residues and is a constituent of the bacilysin synthetic gene cluster, was amplified by PCR based on homology to known genes. The qnr gene consisted of 759 nucleotides corresponding to 252 amino acid residues. Both enzymes belong to the short-chain alcohol dehydrogenase/reductase (SDR) family. The genes were expressed in Escherichia coli as proteins which were His tagged at the N terminus, and the recombinant enzymes were purified and characterized. Both enzymes showed narrow substrate specificity and high stereoselectivity for the reduction of 3-quinuclidinone to (R)-(-)-3-quinuclidinol.


Subject(s)
Actinomycetales/enzymology , Coenzymes/metabolism , NAD/metabolism , Oxidoreductases/genetics , Oxidoreductases/metabolism , Quinuclidines/metabolism , Actinomycetales/genetics , Amino Acid Sequence , Cloning, Molecular , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Escherichia coli/genetics , Molecular Sequence Data , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Sequence Alignment , Sequence Analysis, DNA , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...