Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 447
Filter
1.
PLoS One ; 19(2): e0297365, 2024.
Article in English | MEDLINE | ID: mdl-38329988

ABSTRACT

The legs of insects play an important role in their daily behaviour, especially reproduction. Entomologists have performed much research on the role of the leg in different behaviours of beetles, an important group in the insect family, but relatively little has been done to study the ultrastructure and transcriptome of their legs. Hence, we systematically studied the ultrastructure and gene expression of the leg of G. cantor, a polygynous beetle, and compared its male and female diversity. In this study, we found the fore-leg, mid-leg and hind-leg of the female were significantly longer than those of the male. From the perspective of intuitive structural differences, we also compared the ultrastructures of the adhesion structure (tarsal) of males and females. The tarsal functional structure of the adult leg mainly includes sensilla and an adhesion structure. The sensilla on the tarsal joint mainly include sensilla chaetica (SCh II, SCh III) and sensilla trichodea (ST II). The adhesion structure includes disc-shaped bristles (di), lanceolate bristles (la), serrated bristles (se), spatula-shaped bristles (spl) and mushroom-shaped bristles (mus). Although there was no significant difference in sensillum distribution or type between males and females, there were significant differences in the distribution and species of adhesion structures between the fore-leg, mid-leg, and hind-leg of the same sex and between males and females. Therefore, different adhesion structures play different roles in various behaviours of beetles. On the other hand, the transcriptome results of male and female legs were screened for a subset of olfaction- and mechanics-related genes. We discovered that the male leg showed upregulation of 1 odorant binding protein (OBP), 2 Olfactory receptors (ORs) and 2 Chemosensory proteins (CSPs). Meanwhile, the female leg showed upregulation of 3 OBPs, 1 OR, 1 Gustatory receptor (GR) and 3 Mechanosensitive proteins (MSPs). An in-depth examination of the ultrastructure and molecular composition of the legs can elucidate its function in the reproductive behavior of G. cantor. Moremore, this investigation will serve as a cornerstone for subsequent research into the underlying behavioral mechanisms.


Subject(s)
Coleoptera , Pyridazines , Animals , Mice , Female , Male , Coleoptera/anatomy & histology , Microscopy, Electron, Scanning , Sensilla/ultrastructure , Pyridazines/metabolism , Arthropod Antennae/anatomy & histology
2.
Biochem Biophys Res Commun ; 598: 15-19, 2022 04 02.
Article in English | MEDLINE | ID: mdl-35151199

ABSTRACT

Ponatinib is a multi-target tyrosine kinase inhibitor that targets ABL, SRC, FGFR, and so on. It was designed to overcome the resistance of BCR-ABL mutation to imatinib, especially the gatekeeper mutation ABLT315I. The molecular mechanism by which ponatinib overcomes mutations of BCR-ABL and some other targets has been explained, but little information is known about the characteristics of ponatinib binding to SRC. Here, we showed that ponatinib inhibited wild type SRC kinase but failed to inhibit SRC gatekeeper mutants in both biochemical and cellular assays. We determined the crystal structure of ponatinib in complex with the SRC kinase domain. In addition, by structural analysis, we provided a possible explanation for why ponatinib showed different effects on SRC and other kinases with gatekeeper mutations. The resistance mechanism of SRC gatekeeper mutations to ponatinib may provide meaningful information for designing inhibitors against SRC family kinases in the future.


Subject(s)
Imidazoles/chemistry , Imidazoles/pharmacology , Protein Kinase Inhibitors/chemistry , Pyridazines/chemistry , Pyridazines/pharmacology , src-Family Kinases/chemistry , Binding Sites , Crystallography, X-Ray , Humans , Imidazoles/metabolism , Models, Molecular , Mutation , Protein Conformation , Protein Domains , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-hck/chemistry , Proto-Oncogene Proteins c-hck/metabolism , Pyridazines/metabolism , src-Family Kinases/genetics , src-Family Kinases/metabolism
3.
Ecotoxicol Environ Saf ; 208: 111645, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33396165

ABSTRACT

The effects of chloridazon (Ch) and its metabolite chloridazon-desphenyl (Ch-D) at the environmentally relevant concentrations of 0.45 µg/L and 2.7 µg/L on signal crayfish Pacifastacus leniusculus were assessed in a 30-day exposure followed by a 15-day depuration period. Locomotion, biochemical haemolymph profile, oxidative and antioxidant parameters, and histopathology were evaluated. Crayfish exposed to Ch at 0.45 µg/L and 2.7 µg/L showed significantly (p < 0.01) higher CAT activity and GSH level in hepatopancreas and gill compared to controls. The concentration of Ch at 2.7 µg/L was associated with significantly (p < 0.01) higher levels of GLU, LACT, ALT, AST in haemolymph compared to controls. Chloridazon-desphenyl exposure at both tested concentrations caused significantly higher (p < 0.01) GLU, LACT, ALT, AST, NH3, and Ca in haemolymph; lipid peroxidation (TBARS) levels in hepatopancreas; and CAT activity and GSH level in hepatopancreas and gill. Alterations of structure including focal dilatation of tubules, increased number of fibrillar cells, and haemocyte infiltration in the interstitium were observed with 2.7 µg/L Ch and with both Ch-D exposures. Locomotion patterns did not vary significantly among groups. A 15-day recovery period was insufficient to restore normal physiological parameters in exposed groups. Chloridazon and its metabolite Ch-D exerts harmful effects on crayfish.


Subject(s)
Astacoidea/drug effects , Herbicides/toxicity , Pyridazines/toxicity , Water Pollutants, Chemical/toxicity , Animals , Antioxidants/metabolism , Astacoidea/physiology , Gills/drug effects , Gills/metabolism , Gills/pathology , Hemolymph/cytology , Hemolymph/drug effects , Hemolymph/metabolism , Hepatopancreas/drug effects , Hepatopancreas/metabolism , Hepatopancreas/pathology , Herbicides/metabolism , Oxidative Stress/drug effects , Pyridazines/metabolism , Water Pollutants, Chemical/metabolism
4.
Bioorg Chem ; 107: 104522, 2021 02.
Article in English | MEDLINE | ID: mdl-33317838

ABSTRACT

A potential microtubule destabilizing series of new thirty-five Pyrrol-2-one, Pyridazin-3(2H)-one and Pyridazin-3(2H)-one/oxime derivatives has been synthesized and tested for their antiproliferative activity against a panel of 60 human cancer cell lines. Compounds IVc, IVg and IVf showed a broad spectrum of growth inhibitory activity against cancer cell lines representing renal, cancer of lung, colon, central nervous system, ovary, and kidney. Among them, compound IVg was found to have broad spectrum anti-tumor activity against the tested nine tumor subpanels with selectivity ratios ranging between 0.21 and 3.77 at the GI50 level. In vitro assaying revealed tubulin polymerization inhibition by all active compounds IVc, IVg and IVf. The results of the docking study revealed nice fitting of compounds IVc, IVf, and IVg into CA-4 binding site in tubulin. The three compounds exhibited high binding affinities (ΔGb = -12.49 to -12.99 kcal/mol) toward tubulin compared to CA-4 (-8.87 kcal/mol). Investigation of the binding modes of the three compounds IVc, IVf, and IVg revealed that they interacted mainly hydrophobically with tubulin and similar binding orientations to that of CA-4. These observations suggest that tubulin is a possible target for these compounds.


Subject(s)
Antineoplastic Agents/pharmacology , Pyridazines/pharmacology , Pyrroles/pharmacology , Tubulin Modulators/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Apoptosis/drug effects , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Design , Drug Screening Assays, Antitumor , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Molecular Docking Simulation , Molecular Structure , Protein Binding , Pyridazines/chemical synthesis , Pyridazines/metabolism , Pyrroles/chemical synthesis , Pyrroles/metabolism , Structure-Activity Relationship , Tubulin/chemistry , Tubulin/metabolism , Tubulin Modulators/chemical synthesis , Tubulin Modulators/metabolism
5.
PLoS One ; 15(12): e0233073, 2020.
Article in English | MEDLINE | ID: mdl-33275615

ABSTRACT

There is unmet need for effective stroke therapies. Numerous neuroprotection attempts for acute cerebral ischemia have failed and as a result there is growing interest in developing therapies to promote functional recovery through increasing synaptic plasticity. For this research study, we hypothesized that in addition to its previously reported role in mediating cell death during the acute phase, the alpha isoform of p38 mitogen-activated protein kinase, p38α, may also contribute to interleukin-1ß-mediated impairment of functional recovery during the subacute phase after acute ischemic stroke. Accordingly, an oral, brain-penetrant, small molecule p38α inhibitor, neflamapimod, was evaluated as a subacute phase stroke treatment to promote functional recovery. Neflamapimod administration to rats after transient middle cerebral artery occlusion at two dose levels was initiated outside of the previously characterized therapeutic window for neuroprotection of less than 24 hours for p38α inhibitors. Six-week administration of neflamapimod, starting at 48 hours after reperfusion, significantly improved behavioral outcomes assessed by the modified neurological severity score at Week 4 and at Week 6 post stroke in a dose-dependent manner. Neflamapimod demonstrated beneficial effects on additional measures of sensory and motor function. It also resulted in a dose-related increase in brain-derived neurotrophic factor (BDNF) protein levels, a previously reported potential marker of synaptic plasticity that was measured in brain homogenates at sacrifice. Taken together with literature evidence on the role of p38α-dependent suppression by interleukin-1ß of BDNF-mediated synaptic plasticity and BDNF production, our findings support a mechanistic model in which inhibition of p38α promotes functional recovery after ischemic stroke by blocking the deleterious effects of interleukin-1ß on synaptic plasticity. The dose-related in vivo efficacy of neflamapimod offers the possibility of having a therapy for stroke that could be initiated outside the short time window for neuroprotection and for improving recovery after a completed stroke.


Subject(s)
Brain Ischemia/drug therapy , Pyridazines/pharmacology , Pyrimidines/pharmacology , Stroke/drug therapy , Animals , Brain/metabolism , Brain Ischemia/complications , Brain Ischemia/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Infarction, Middle Cerebral Artery/complications , Ischemia/complications , Male , Mitogen-Activated Protein Kinase 14/antagonists & inhibitors , Mitogen-Activated Protein Kinase 14/metabolism , Pyridazines/metabolism , Pyrimidines/metabolism , Rats , Rats, Sprague-Dawley , Recovery of Function/drug effects , Recovery of Function/physiology , Stroke/complications , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Drug Des Devel Ther ; 14: 5259-5273, 2020.
Article in English | MEDLINE | ID: mdl-33299299

ABSTRACT

BACKGROUND: Ensartinib (ESB) is a novel anaplastic lymphoma kinase inhibitor (ALK) with additional activity against Abelson murine leukemia (ABL), met proto-oncogene (MET), receptor tyrosine kinase (AXL), and v-ros UR2 sarcoma virus oncogene homolog 1 (ROS1) and is considered a safer alternative for other ALK inhibitors. ESB chemical structure contains a dichloro-fluorophenyl ring and cyclic tertiary amine rings (piperazine) that can be bioactivated generating reactive intermediates. METHODS: In vitro metabolic study of ESB with human liver microsomes (HLMs) was performed and the hypothesis of generating reactive intermediates during metabolism was tested utilizing trapping agents to capture and stabilize reactive intermediates to facilitate their LC-MS/MS detection. Reduced glutathione (GSH) and potassium cyanide (KCN) were utilized as trapping agents for quinone methide and iminium intermediates, respectively. RESULTS: Four in vitro ESB phase I metabolites were characterized. Three reactive intermediates including one epoxide and one iminium intermediates were characterized. ESB bioactivation is proposed to occur through unexpected metabolic pathways. The piperazine ring was bioactivated through iminium ions intermediates generation, while the dichloro-phenyl group was bioactivated through a special mechanism that was revealed by LC-MS/MS. CONCLUSION: These findings lay the foundations for additional work on ESB toxicity. Substituents to the bioactive centers (piperazine ring), either for blocking or isosteric replacement, would likely block or interrupt hydroxylation reaction that will end the bioactivation sequence.


Subject(s)
Antineoplastic Agents/metabolism , Microsomes, Liver/chemistry , Piperazines/metabolism , Pyridazines/metabolism , Antineoplastic Agents/chemistry , Chromatography, Liquid , Drug Stability , Humans , Microsomes, Liver/metabolism , Molecular Conformation , Piperazines/chemistry , Proto-Oncogene Mas , Pyridazines/chemistry , Software , Tandem Mass Spectrometry
7.
Elife ; 92020 11 25.
Article in English | MEDLINE | ID: mdl-33236980

ABSTRACT

Canonical transient receptor potential channels (TRPC) are involved in receptor-operated and/or store-operated Ca2+ signaling. Inhibition of TRPCs by small molecules was shown to be promising in treating renal diseases. In cells, the channels are regulated by calmodulin (CaM). Molecular details of both CaM and drug binding have remained elusive so far. Here, we report structures of TRPC4 in complex with three pyridazinone-based inhibitors and CaM. The structures reveal that all the inhibitors bind to the same cavity of the voltage-sensing-like domain and allow us to describe how structural changes from the ligand-binding site can be transmitted to the central ion-conducting pore of TRPC4. CaM binds to the rib helix of TRPC4, which results in the ordering of a previously disordered region, fixing the channel in its closed conformation. This represents a novel CaM-induced regulatory mechanism of canonical TRP channels.


Subject(s)
Calmodulin/metabolism , Membrane Transport Modulators/pharmacology , Pyridazines/pharmacology , TRPC Cation Channels/drug effects , Zebrafish Proteins/drug effects , Animals , Binding Sites , Calmodulin/chemistry , Calmodulin/genetics , HEK293 Cells , Humans , Ligands , Membrane Potentials , Membrane Transport Modulators/chemistry , Membrane Transport Modulators/metabolism , Models, Molecular , Protein Binding , Protein Conformation , Pyridazines/chemistry , Pyridazines/metabolism , Sf9 Cells , Structure-Activity Relationship , TRPC Cation Channels/chemistry , TRPC Cation Channels/genetics , TRPC Cation Channels/metabolism , Xenopus , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
8.
Molecules ; 25(21)2020 Oct 28.
Article in English | MEDLINE | ID: mdl-33126762

ABSTRACT

Tepotinib (Tepmetko™, Merck) is a potent inhibitor of c-Met (mesenchymal-epithelial transition factor). In March 2020, tepotinib (TEP) was approved for use in Japan for the treatment of patients who suffered from non-small cell lung cancers (NSCLC) harboring an MET exon 14 skipping alteration and have progressed after platinum-based therapy. Practical and in silico experiments were used to screen for the metabolic profile and reactive intermediates of TEP. Knowing the bioactive center and structural alerts in the TEP structure helped in making targeted modifications to improve its safety. First, the prediction of metabolism vulnerable sites and reactivity metabolic pathways was performed using the StarDrop WhichP450™ module and the online Xenosite reactivity predictor tool, respectively. Subsequently, in silico data were used as a guide for the in vitro practical work. Second, in vitro phase I metabolites of TEP were generated from human liver microsome (HLM) incubations. Testing for the generation of unstable reactive intermediates was performed using potassium cyanide as a capturing agent forming stable cyano adduct that can be characterized and identified using liquid chromatography tandem mass spectrometry (LC-MS/MS). Third, in silico toxicity assessment of TEP metabolites was performed, and structural modification was proposed to decrease their side effects and to validate the proposed bioactivation pathway using the DEREK software. Four TEP phase I metabolites and four cyano adducts were characterized. The reactive intermediate generation mechanism of TEP may provide an explanation of its adverse reactions. The piperidine ring is considered a structural alert for toxicity as proposed by the DEREK software and a Xenosite reactivity model, which was confirmed by practical experiments. Steric hindrance or isosteric replacement at α-carbon of the piperidine ring stop the bioactivation sequence that was confirmed using the DEREK software. More drug discovery studies can be performed using this perception permitting the design of new drugs with an increased safety profile. To our knowledge, this is the first study for the identification of in vitro phase I metabolites and reactive intermediates in addition to toxicological properties of the metabolites for TEP that will be helpful for the evaluation of TEP side effects and drug-drug interactions in TEP-treated patients.


Subject(s)
Chromatography, Liquid , Computer Simulation , Piperidines/chemistry , Piperidines/metabolism , Pyridazines/chemistry , Pyridazines/metabolism , Pyrimidines/chemistry , Pyrimidines/metabolism , Tandem Mass Spectrometry
9.
Biomed Pharmacother ; 131: 110732, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32942157

ABSTRACT

Polyphenolic compounds (including flavonoids, chalcones, phenolic acids, and furanocoumarins) represent a common part of our diet, but are also the active ingredients of several dietary supplements and/or medications. These compounds undergo extensive metabolism by human biotransformation enzymes and the microbial flora of the colon. CYP2D6 enzyme metabolizes approximately 25% of the drugs, some of which has narrow therapeutic window. Therefore, its inhibition can lead to the development of pharmacokinetic interactions and the disruption of drug therapy. In this study, the inhibitory effects of 17 plant-derived compounds and 19 colonic flavonoid metabolites on CYP2D6 were examined, employing two assays with different test substrates. The O-demethylation of dextromethorphan was tested employing CypExpress 2D6 kit coupled to HPLC analysis; while the O-demethylation of another CYP2D6 specific substrate (AMMC) was investigated in a plate reader assay with BioVision Fluorometric CYP2D6 kit. Interestingly, some compounds (e.g., bergamottin) inhibited both dextromethorphan and AMMC demethylation; however, certain substances proved to be inhibitors only in one of the assays applied. Our results demonstrate that some polyphenols and colonic metabolites are inhibitors of CYP2D6-catalyzed reactions. Nevertheless, the inhibitory effects showed strong substrate dependence.


Subject(s)
Colon/metabolism , Cytochrome P-450 CYP2D6 Inhibitors/pharmacology , Cytochrome P-450 CYP2D6/metabolism , Polyphenols/pharmacology , Acetamides/metabolism , Dextromethorphan/metabolism , Flavonoids/pharmacology , Humans , Polyphenols/metabolism , Pyridazines/metabolism
10.
Kidney Int ; 98(1): 51-53, 2020 07.
Article in English | MEDLINE | ID: mdl-32571489

ABSTRACT

Mitochondrial dysfunction is thought to be a critical pathway in the development and progression of kidney diseases, but optimal methods to assess kidney mitochondrial dysfunction are not well known. Saeki and colleagues use positron emission tomography imaging with a novel probe, 2-tert-butyl-4-chloro-5-[6-(4-18F-fluorobutoxy)-pyridin-3-ylmethoxy]-2H-pyridazin-3-one (18F-BCPP-BF), to visualize and assess kidney mitochondrial status. The authors demonstrate that reduced uptake of 18F-BCPP-BF, as assessed by positron emission tomography imaging, corresponds to reduced functioning mitochondria in 3 separate animal models of kidney diseases.


Subject(s)
Electron Transport Complex I , Pyridazines , Animals , Brain , Electron Transport Complex I/metabolism , Mitochondria/metabolism , Positron-Emission Tomography , Pyridazines/metabolism , Pyridines , Radiopharmaceuticals/metabolism
11.
J Med Chem ; 63(11): 5816-5840, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32410449

ABSTRACT

Non-BET bromodomain-containing proteins have become attractive targets for the development of novel therapeutics targeting epigenetic pathways. To help facilitate the target validation of this class of proteins, structurally diverse small-molecule ligands and methodologies to produce selective inhibitors in a predictable fashion are in high demand. Herein, we report the development and application of atypical acetyl-lysine (KAc) methyl mimetics to take advantage of the differential stability of conserved water molecules in the bromodomain binding site. Discovery of the n-butyl group as an atypical KAc methyl mimetic allowed generation of 31 (GSK6776) as a soluble, permeable, and selective BRD7/9 inhibitor from a pyridazinone template. The n-butyl group was then used to enhance the bromodomain selectivity of an existing BRD9 inhibitor and to transform pan-bromodomain inhibitors into BRD7/9 selective compounds. Finally, a solvent-exposed vector was defined from the pyridazinone template to enable bifunctional molecule synthesis, and affinity enrichment chemoproteomic experiments were used to confirm several of the endogenous protein partners of BRD7 and BRD9, which form part of the chromatin remodeling PBAF and BAF complexes, respectively.


Subject(s)
Chromosomal Proteins, Non-Histone/antagonists & inhibitors , Lysine/chemistry , Pyridazines/chemistry , Transcription Factors/antagonists & inhibitors , Binding Sites , Chromosomal Proteins, Non-Histone/metabolism , Crystallography, X-Ray , Humans , Ligands , Molecular Dynamics Simulation , Protein Structure, Tertiary , Pyridazines/metabolism , Structure-Activity Relationship , Transcription Factors/metabolism
12.
J Med Chem ; 63(11): 6028-6056, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32298582

ABSTRACT

Neutral sphingomyelinase 2 (nSMase2) catalyzes the cleavage of sphingomyelin to phosphorylcholine and ceramide, an essential step in the formation and release of exosomes from cells that is critical for intracellular communication. Chronic increase of brain nSMase2 activity and related exosome release have been implicated in various pathological processes, including the progression of Alzheimer's disease (AD), making nSMase2 a viable therapeutic target. Recently, we identified phenyl (R)-(1-(3-(3,4-dimethoxyphenyl)-2,6-dimethylimidazo[1,2-b]pyridazin-8-yl)pyrrolidin-3-yl)carbamate 1 (PDDC), the first nSMase2 inhibitor that possesses both favorable pharmacodynamics and pharmacokinetic (PK) parameters, including substantial oral bioavailability, brain penetration, and significant inhibition of exosome release from the brain in vivo. Herein we demonstrate the efficacy of 1 (PDDC) in a mouse model of AD and detail extensive structure-activity relationship (SAR) studies with 70 analogues, unveiling several that exert similar or higher activity against nSMase2 with favorable pharmacokinetic properties.


Subject(s)
Enzyme Inhibitors/chemistry , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Animals , Body Weight/drug effects , Brain/metabolism , Disease Models, Animal , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Exosomes/metabolism , Female , Humans , Male , Mice , Mice, Transgenic , Pyridazines/chemistry , Pyridazines/metabolism , Pyridazines/therapeutic use , Sphingomyelin Phosphodiesterase/metabolism , Sphingomyelin Phosphodiesterase/pharmacology , Structure-Activity Relationship
13.
Chem Biodivers ; 17(6): e2000100, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32239712

ABSTRACT

A facile and convenient synthesis of new pyridazines suitable for use as antimicrobial agents was reported. The hydrazide intermediate was coupled with various benzaldehydes and/or acetophenones and cyclized instantaneously to afford target pyridazine derivatives. The structures of new pyridazines were confirmed by IR, 1 H- and 13 C-NMR, elemental analysis in addition to representative LC/MS. Antimicrobial activity was screened against 10 bacterial and fungal strains. The new pyridazines showed strong to very strong antibacterial activity against Gram-negative (GNB) bacteria, while none of them showed significant antifungal activity at the same concentration range. Chloro derivatives exhibited the highest antibacterial activity with MICs (0.892-3.744 µg/mL) lower than that of chloramphenicol (2.019-8.078 µg/mL) against E. coli, P. aeruginosa, and S. marcescens. Prediction of ADME parameters, pharmacokinetics, and substrate promiscuity revealed that these new pyridazines could be promising drug candidates. Cytotoxic studies on rat hepatocytes showed how much safe these new pyridazines on living organisms (IC50 >64 µg/mL). MOE docking studies showed a good overlay of these new pyridazines with co-crystallized ligand within an E. coli DNA gyrase subunit B active sites (4KFG).


Subject(s)
Anti-Infective Agents/chemical synthesis , Molecular Docking Simulation , Pyridazines/chemistry , Animals , Anti-Infective Agents/metabolism , Anti-Infective Agents/pharmacology , Binding Sites , Catalytic Domain , Cell Survival/drug effects , Cells, Cultured , DNA Gyrase/chemistry , DNA Gyrase/metabolism , Escherichia coli/drug effects , Escherichia coli/enzymology , Fungi/drug effects , Gram-Negative Bacteria/drug effects , Half-Life , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Microbial Sensitivity Tests , Pyridazines/metabolism , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Thermodynamics
14.
Biomed Chromatogr ; 34(6): e4819, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32112427

ABSTRACT

Ponatinib is an oral drug for the treatment of chronic myeloid leukemia and acute lymphoblastic leukemia, which has been reported to increase the risk of hepatotoxicity. The aim of this study was to characterize the metabolites of ponatinib in human liver microsomes as well as its reactive metabolites. Ponatinib was incubated with human liver microsomes in the presence of NADPH and trapping agents (glutathione or potassium cyanide). The metabolites were characterized by liquid chromatography in combination with Q-Exactive-Orbitrap-MS. Under the current conditions, six metabolites were detected and structurally identified on the basis of their accurate masses, fragmentation patterns, and retention times. M3 (N-demethylation) was unambiguously identified by matching its retention time and fragment ions with those of its reference standard. N-demethylation and oxygenation were proved to be the predominant metabolic pathways of ponatinib. In addition, two reactive metabolites (cyano adducts) were detected in human liver microsomes in the presence of potassium cyanide and NADPH, suggesting that ponatinib underwent CYP450-mediated metabolic activation, which could be one of the causative mechanisms for its hepatotoxicity. The current study provides new information regarding the metabolic profiles of ponatinib and would be helpful in understanding the effectiveness and toxicity of ponatinib, especially the mechanism of hepatotoxicity.


Subject(s)
Chromatography, High Pressure Liquid/methods , Imidazoles/analysis , Imidazoles/metabolism , Microsomes, Liver/metabolism , Pyridazines/analysis , Pyridazines/metabolism , Tandem Mass Spectrometry/methods , Cytochrome P-450 Enzyme System/metabolism , Glutathione/metabolism , Humans , Imidazoles/chemistry , NADP/metabolism , Pyridazines/chemistry
15.
Eur J Med Chem ; 190: 112092, 2020 Mar 15.
Article in English | MEDLINE | ID: mdl-32014679

ABSTRACT

Harboring MYD88 L265P mutation triggers tumors growth through the activation of NF-κB by interleukin-1 receptor associated kinase 4 (IRAK4) in diffuse large B-cell lymphoma (DLBCL), highlighting IRAK4 as a therapeutic target for tumors driven by aberrant MYD88 signaling. Herein, we report the design, synthesis, and structure-activity relationships of imidazo[1,2-b]pyridazines as potent IRAK4 inhibitors. The representative compound 5 exhibited excellent IRAK4 potency (IRAK4 IC50 = 1.3 nM) and favorable kinase selectivity profile. It demonstrated cellular selectivity for activated B cell-like (ABC) subtype DLBCL with MYD88 L265P mutation in cytotoxicity assay. The kinase inhibitory efficiency of compound 5 was further validated by Western blot analysis of phosphorylation of IRAK4 and downstream signaling in OCI-LY10 and TMD8 cells. Besides, combination of compound 5 and BTK inhibitor ibrutinib synergistically reduced the viability of TMD8 cells. These results indicated that compound 5 could be a promising IRAK4 inhibitor for the treatment of mutant MYD88 DLBCL.


Subject(s)
Antineoplastic Agents/pharmacology , Imidazoles/pharmacology , Interleukin-1 Receptor-Associated Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyridazines/pharmacology , Adenine/analogs & derivatives , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Cell Line, Tumor , Drug Design , Drug Synergism , Humans , Imidazoles/chemical synthesis , Imidazoles/metabolism , Interleukin-1 Receptor-Associated Kinases/metabolism , Lymphoma, Large B-Cell, Diffuse/drug therapy , Molecular Docking Simulation , Molecular Structure , Mutation , Myeloid Differentiation Factor 88/genetics , Piperidines , Protein Binding , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/metabolism , Pyrazoles/pharmacology , Pyridazines/chemical synthesis , Pyridazines/metabolism , Pyrimidines/pharmacology , Signal Transduction/drug effects , Structure-Activity Relationship
16.
J Med Chem ; 63(5): 2028-2034, 2020 03 12.
Article in English | MEDLINE | ID: mdl-31829628

ABSTRACT

Stimulated Raman scattering (SRS) microscopy represents a powerful method for imaging label-free drug distribution with high resolution. SRS was applied to image label-free ponatinib with high sensitivity and specificity in live human chronic myeloid leukemia (CML) cell lines. This was achieved at biologically relevant, nanomolar concentrations, allowing determination of ponatinib uptake and sequestration into lysosomes during the development of acquired drug resistance and an improved understanding of target engagement.


Subject(s)
Antineoplastic Agents/metabolism , Imidazoles/metabolism , Intracellular Fluid/metabolism , Nonlinear Optical Microscopy/methods , Pyridazines/metabolism , Antineoplastic Agents/analysis , Cell Line, Tumor , Humans , Imidazoles/analysis , Pyridazines/analysis
17.
Bioorg Med Chem Lett ; 30(4): 126811, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31787491

ABSTRACT

This Letter details our efforts to develop new M4 PAM scaffolds with improved pharmacological properties. This endeavor involved replacing the 3,4-dimethylpyridazine core with two novel cores: a 2,3-dimethyl-2H-indazole-5-carboxamide core or a 1-methyl-1H-benzo[d][1,2,3]triazole-6-carboxamide core. Due to shallow SAR, these cores were further evolved into two unique tricyclic cores: an 8,9-dimethyl-8H-pyrazolo[3,4-h]quinazoline core and an 1-methyl-1H-[1,2,3]triazolo[4,5-h]quinazoline core. Both tricyclic cores displayed low nanomolar potency against both human and rat M4.


Subject(s)
Pyridazines/chemistry , Quinazolines/chemistry , Receptor, Muscarinic M4/chemistry , Triazoles/chemistry , Allosteric Regulation , Animals , Drug Design , Half-Life , Humans , Inhibitory Concentration 50 , Pyridazines/metabolism , Pyridazines/pharmacokinetics , Quinazolines/metabolism , Quinazolines/pharmacokinetics , Rats , Receptor, Muscarinic M4/metabolism , Structure-Activity Relationship , Triazoles/metabolism , Triazoles/pharmacokinetics
18.
Environ Monit Assess ; 191(8): 517, 2019 Jul 27.
Article in English | MEDLINE | ID: mdl-31352622

ABSTRACT

The dissipation and residual levels of etoxazole and pyridaben in Goji berry under open field conditions were determined by using GC-NPD (gas chromatography with nitrogen and phosphorus detector) with modified QuEChERS method. At fortification levels of 0.01, 1, and 5 mg/kg in Goji berry, it was shown that recoveries were ranged from 80.40 to 100.9% with relative standard deviation of the method (RSD) for repeatability ranged from 2.20 to 4.25%. The limit of quantification (LOQ) of the method was 0.01 mg/kg. The dissipation rates of etoxazole and pyridaben were described by using first-order kinetics and its half-life, as they are 7.13 days, 5.77 days, and 5.99 days (etoxazole) and 1.02 day, 0.67 day, 1.02 day (pyridaben). The terminal residues of etoxazole and pyridaben were below the European maximum residue limit (MRL, 0.1 mg/kg) in Goji berry when measured 7 days after the final application, which suggested that the use of these insecticides was safe for humans. This study would help in providing the basic information for developing regulation to guard a safe use of etoxazole and pyridaben in Goji berry and prevent health problem from consumers.


Subject(s)
Environmental Monitoring/methods , Lycium/metabolism , Oxazoles/analysis , Pesticide Residues/analysis , Pyridazines/analysis , China , Half-Life , Humans , Kinetics , Lycium/growth & development , Oxazoles/metabolism , Pesticide Residues/metabolism , Pyridazines/metabolism , Tibet
19.
Biochem Pharmacol ; 166: 120-127, 2019 08.
Article in English | MEDLINE | ID: mdl-31078601

ABSTRACT

Overexpression of ABCB1 transporters plays a crucial role in mediating multidrug resistance (MDR). Therefore, it is important to inhibit ABCB1 activity in order to maintain an effective intracellular level of chemotherapeutic drugs. Tepotinib is a MET tyrosine kinase inhibitor with potential anticancer effect and it is currently in clinical trials. In this study, we investigated whether tepotinib could antagonize ABC transporters-mediated MDR. Our results suggest that tepotinib significantly reversed ABCB1-mediated MDR but not ABCG2- or ABCC1-mediated MDR. Mechanistic studies show that tepotinib significantly reversed ABCB1-mediated MDR by attenuating the efflux activity of ABCB1 transporter. The ATPase assay showed that tepotinib inhibited the ATPase activity of ABCB1 in a concentration-dependent manner. Furthermore, treatment with tepotinib did not change protein expression or subcellular localization of ABCB1. Docking analysis indicated that tepotinib interacted with the drug-binding site of the ABCB1 transporter. Our study provides a potential chemotherapeutic strategy of co-administrating tepotinib with other conventional chemotherapeutic agents to overcome MDR and improve therapeutic effect.


Subject(s)
Antineoplastic Agents/metabolism , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Pyridazines/metabolism , Pyrimidines/metabolism , ATP Binding Cassette Transporter, Subfamily B/chemistry , ATP Binding Cassette Transporter, Subfamily B/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Resistance, Multiple/physiology , Drug Resistance, Neoplasm/physiology , HEK293 Cells , Humans , Protein Structure, Secondary , Pyridazines/pharmacology , Pyrimidines/pharmacology
20.
Nat Prod Rep ; 36(12): 1628-1653, 2019 12 11.
Article in English | MEDLINE | ID: mdl-30949650

ABSTRACT

Covering: up to the end of 2018 Piperazic acid is a cyclic hydrazine and a non-proteinogenic amino acid found in diverse non-ribosomal peptide (NRP) and hybrid NRP-polyketide (PK) structures. Piperazic acid was first identified as a residue in the monamycins in 1959. Since then, the piperazic acid residue has been found in >30 families of natural products, representing >140 compounds. Many of these compounds have potent biological activity, ranging from anti-malarial to anti-apoptotic to anti-bacterial activity, although high toxicity often accompanies this potent biological activity. Recently, we identified a piperazate synthase, responsible for N-N bond formation to give piperazic acid. Here, we review piperazic acid-containing natural products discovered from 1959 to 2018, with an emphasis on the biosynthetic routes to these natural products.


Subject(s)
Biological Products/chemistry , Biological Products/metabolism , Pyridazines/chemistry , Biological Products/pharmacology , Molecular Structure , Multigene Family , Peptides/chemistry , Peptides/metabolism , Peptides, Cyclic/biosynthesis , Peptides, Cyclic/chemistry , Pyridazines/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...