Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.175
Filtrar
1.
Biochemistry ; 63(9): 1170-1177, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38587906

RESUMEN

The MbnBC enzyme complex converts cysteine residues in a peptide substrate, MbnA, to oxazolone/thioamide groups during the biosynthesis of copper chelator methanobactin (Mbn). MbnBC belongs to the mixed-valent diiron oxygenase (MVDO) family, of which members use an Fe(II)Fe(III) cofactor to react with dioxygen for substrate modification. Several crystal structures of the inactive Fe(III)Fe(III) form of MbnBC alone and in complex with MbnA have been reported, but a mechanistic understanding requires determination of the oxidation states of the crystallographically observed Fe ions in the catalytically active Fe(II)Fe(III) state, along with the site of MbnA binding. Here, we have used electron nuclear double resonance (ENDOR) spectroscopy to determine such structural and electronic properties of the active site, in particular, the mode of substrate binding to the MV state, information not accessible by X-ray crystallography alone. The oxidation states of the two Fe ions were determined by 15N ENDOR analysis. The presence and locations of both bridging and terminal exogenous solvent ligands were determined using 1H and 2H ENDOR. In addition, 2H ENDOR using an isotopically labeled MbnA substrate indicates that MbnA binds to the Fe(III) ion of the cluster via the sulfur atom of its N-terminal modifiable cysteine residue, with displacement of a coordinated solvent ligand as shown by complementary 1H ENDOR. These results, which underscore the utility of ENDOR in studying MVDOs, provide a molecular picture of the initial steps in Mbn biosynthesis.


Asunto(s)
Imidazoles , Oligopéptidos , Imidazoles/metabolismo , Imidazoles/química , Oligopéptidos/metabolismo , Oligopéptidos/química , Oligopéptidos/biosíntesis , Oxidación-Reducción , Cristalografía por Rayos X , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/química , Espectroscopía de Resonancia por Spin del Electrón , Oxigenasas/metabolismo , Oxigenasas/química , Dominio Catalítico , Especificidad por Sustrato , Modelos Moleculares , Hierro/metabolismo , Hierro/química
2.
Pharm Res ; 41(5): 849-861, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38485855

RESUMEN

PURPOSE: Olmesartan medoxomil (olmesartan-MX), an ester-type prodrug of the angiotensin II receptor blocker (ARB) olmesartan, is predominantly anionic at intestinal pH. Human organic anion transporting polypeptide 2B1 (OATP2B1) is expressed in the small intestine and is involved in the absorption of various acidic drugs. This study was designed to test the hypothesis that OATP2B1-mediated uptake contributes to the enhanced intestinal absorption of olmesartan-MX, even though olmesartan itself is not a substrate of OATP2B1. METHODS: Tetracycline-inducible human OATP2B1- and rat Oatp2b1-overexpressing HEK 293 cell lines (hOATP2B1/T-REx-293 and rOatp2b1/T-REx-293, respectively) were established to characterize OATP2B1-mediated uptake. Rat jejunal permeability was measured using Ussing chambers. ARBs were quantified by liquid chromatography-tandem mass spectrometry. RESULTS: Significant olmesartan-MX uptake was observed in hOATP2B1/T-REx-293 and rOatp2b1/T-REx-293 cells, whereas olmesartan uptake was undetectable or much lower than olmesartan-MX uptake, respectively. Furthermore, olmesartan-MX exhibited several-fold higher uptake in Caco-2 cells and greater permeability in rat jejunum compared to olmesartan. Olmesartan-MX uptake in hOATP2B1/T-REx-293 cells and in Caco-2 cells was significantly decreased by OATP2B1 substrates/inhibitors such as 1 mM estrone-3-sulfate, 100 µM rifamycin SV, and 100 µM fluvastatin. Rat Oatp2b1-mediated uptake and rat jejunal permeability of olmesartan-MX were significantly decreased by 50 µM naringin, an OATP2B1 inhibitor. Oral administration of olmesartan-MX with 50 µM naringin to rats significantly reduced the area under the plasma concentration-time curve of olmesartan to 76.9%. CONCLUSION: Olmesartan-MX is a substrate for OATP2B1, and the naringin-sensitive transport system contributes to the improved intestinal absorption of olmesartan-MX compared with its parent drug, olmesartan.


Asunto(s)
Imidazoles , Absorción Intestinal , Olmesartán Medoxomilo , Transportadores de Anión Orgánico , Profármacos , Tetrazoles , Animales , Humanos , Absorción Intestinal/efectos de los fármacos , Olmesartán Medoxomilo/metabolismo , Profármacos/farmacocinética , Profármacos/metabolismo , Células HEK293 , Tetrazoles/farmacocinética , Tetrazoles/metabolismo , Transportadores de Anión Orgánico/metabolismo , Transportadores de Anión Orgánico/antagonistas & inhibidores , Masculino , Imidazoles/farmacocinética , Imidazoles/metabolismo , Ratas , Ratas Sprague-Dawley , Yeyuno/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacocinética , Bloqueadores del Receptor Tipo 1 de Angiotensina II/metabolismo , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Permeabilidad/efectos de los fármacos , Células CACO-2
3.
J Pharmacol Sci ; 154(3): 166-174, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38395517

RESUMEN

Imidazole derivatives are commonly used as antifungal agents. Here, we aimed to investigate the functions of imidazole derivatives on macrophage lineage cells. We assessed the expression levels of inflammatory cytokines in mouse monocyte/macrophage lineage (RAW264.7) cells. All six imidazole derivatives examined, namely ketoconazole, sulconazole, isoconazole, luliconazole, clotrimazole, and bifonazole, reduced the expression levels of inflammatory cytokines, such as interleukin (IL)-6 and tumor necrosis factor-α, after induction by lipopolysaccharide (LPS) in RAW264.7 cells. These imidazole derivatives also induced cell death in RAW264.7 cells, regardless of the presence or absence of LPS. Since the cell death was characteristic in morphology, we investigated the mode of the cell death. An imidazole derivative, sulconazole, induced gasdermin D degradation together with caspase-11 activation, namely, pyroptosis in RAW264.7 cells and peritoneal macrophages. Furthermore, priming with interferon-γ promoted sulconazole-induced pyroptosis in RAW264.7 cells and macrophages and reduced the secretion of the inflammatory cytokine, IL-1ß, from sulconazole-treated macrophages. Our results suggest that imidazole derivatives suppress inflammation by inducing macrophage pyroptosis, highlighting their modulatory potential for inflammatory diseases.


Asunto(s)
Interferón gamma , Piroptosis , Ratones , Animales , Interferón gamma/metabolismo , Monocitos/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/metabolismo , Imidazoles/farmacología , Imidazoles/metabolismo , Citocinas/metabolismo
4.
Mediators Inflamm ; 2024: 9528976, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38405621

RESUMEN

Traditionally, the treatment of inflammatory conditions has focused on the inhibition of inflammatory mediator production; however, many conditions are refractory to this classical approach. Recently, an alternative has been presented by researchers to solve this problem: The immunomodulation of cells closely related to inflammation. Hence, macrophages, a critical key in both innate and acquired immunity, have been presented as an alternative target for the development of new medicines. In this work, we tested the fluorophenyl-imidazole for its anti-inflammatory activity and possible immunomodulatory effect on RAW 264.7 macrophages. We also evaluated the anti-inflammatory effect of the compound, and the macrophage repolarization to M2 was confirmed by the ability of the compound to reduce the M1 markers TNF-α, IL-6, MCP-1, IL-12p70, IFN-γ, and TLR4, the high levels of p65 phosphorylated, iNOS and COX-2 mRNA expression, and the fact that the compound was not able to induce the production of M1 markers when used in macrophages without lipopolysaccharide (LPS) stimulation. Moreover, fluorophenyl-imidazole had the ability to increase the M2 markers IL-4, IL-13, CD206, apoptosis and phagocytosis levels, arginase-1, and FIZZ-1 mRNA expression before LPS stimulation. Similarly, it was also able to induce the production of these same M2 markers in macrophages without being induced with LPS. These results reinforce the affirmation that the fluorophenyl-imidazole has an important anti-inflammatory effect and demonstrates that this effect is due to immunomodulatory activity, having the ability to trigger a repolarization of macrophages from M1 to M2a. These facts suggest that this molecule could be used as an alternative scaffold for the development of a new medicine to treat inflammatory conditions, where the anti-inflammatory and proregenerative properties of M2a macrophages are desired.


Asunto(s)
Lipopolisacáridos , Macrófagos , Lipopolisacáridos/metabolismo , Macrófagos/metabolismo , Interleucina-12/metabolismo , Imidazoles/farmacología , Imidazoles/metabolismo , ARN Mensajero/metabolismo
5.
Molecules ; 28(14)2023 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-37513289

RESUMEN

Mammalian 15-lipoxygenases (ALOX15) are lipid peroxidizing enzymes that exhibit variable functionality in different cancer and inflammation models. The pathophysiological role of linoleic acid- and arachidonic acid-derived ALOX15 metabolites rendered this enzyme a target for pharmacological research. Several indole and imidazole derivatives inhibit the catalytic activity of rabbit ALOX15 in a substrate-specific manner, but the molecular basis for this allosteric inhibition remains unclear. Here, we attempt to define a common pharmacophore, which is critical for this allosteric inhibition. We found that substituted imidazoles induce weaker inhibitory effects when compared with the indole derivatives. In silico docking studies and molecular dynamics simulations using a dimeric allosteric enzyme model, in which the inhibitor occupies the substrate-binding pocket of one monomer, whereas the substrate fatty acid is bound at the catalytic center of another monomer within the ALOX15 dimer, indicated that chemical modification of the core pharmacophore alters the enzyme-inhibitor interactions, inducing a reduced inhibitory potency. In our dimeric ALOX15 model, the structural differences induced by inhibitor binding are translated to the hydrophobic dimerization cluster and affect the structures of enzyme-substrate complexes. These data are of particular importance since substrate-specific inhibition may contribute to elucidation of the putative roles of ALOX15 metabolites derived from different polyunsaturated fatty acids in mammalian pathophysiology.


Asunto(s)
Ácido Linoleico , Farmacóforo , Animales , Conejos , Ácido Linoleico/metabolismo , Mamíferos/metabolismo , Ácidos Linoleicos/metabolismo , Araquidonato 15-Lipooxigenasa/química , Imidazoles/farmacología , Imidazoles/metabolismo
6.
ACS Chem Biol ; 18(10): 2240-2248, 2023 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-37463352

RESUMEN

DNA hydroxymethylation is involved in many biological processes, including nuclear reprogramming, embryonic development, and tumor suppression. In this study, we report that an anticancer agent, nutlin-3, selectively stimulates global DNA hydroxymethylation in TP53 wild-type cancer cells as manifested by the elevation of 5-hydroxymethylcytosine (5hmC) in genomic DNA. In contrast, nutlin 3 fails to enhance DNA hydroxymethylation in TP53-mutated cancer cells. Consistently, nutlin-3 as a MDM2 antagonist only activates wild-type but not mutated TP53. Furthermore, nutlin-3 does not alter the expression of TET1 but slightly reduces the expression of TET2 and TET3 proteins. These TET family proteins are responsible for converting 5-methylcytosine (5mC) to 5hmC. Interestingly, TET1 knockdown could significantly block the nutlin-3-induced DNA hydroxymethylation as well as TP53 and P21 activation. Immunoprecipitation analysis supports that p53 strongly interacts with TET1 proteins. These results suggest that nutlin-3 activates TP53 and promotes p53-TET1 interaction. As positive feedback, the p53-TET1 interaction further enhances p53 activation and promotes apoptosis. Collectively, we demonstrate that nutlin-3 stimulates DNA hydroxymethylation and apoptosis via a positive feedback mechanism.


Asunto(s)
Proteínas Proto-Oncogénicas , Proteína p53 Supresora de Tumor , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Imidazoles/farmacología , Imidazoles/metabolismo , ADN , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Apoptosis , Línea Celular Tumoral
7.
Chembiochem ; 24(6): e202300006, 2023 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-36602436

RESUMEN

Nutlin-3a is a reversible inhibitor of the p53/MDM2 interaction. We have synthesized the derivative Nutlin-3a-aa bearing an additional exocyclic methylene group in the piperazinone moiety. Nutlin-3a-aa is more active than Nutlin-3a against purified wild-type MDM2, and is more effective at increasing p53 levels and releasing transcription of p53 target genes from MDM2-induced repression. X-ray analysis of wild-type MDM2-bound Nutlin-3a-aa indicated that the orientation of its modified piperazinone ring was altered in comparison to the piperazinone ring of MDM2-bound Nutlin-3a, with the exocyclic methylene group of Nutlin-3a-aa pointing away from the protein surface. Our data point to the introduction of exocyclic methylene groups as a useful approach by which to tailor the conformation of bioactive molecules for improved biological activity.


Asunto(s)
Antineoplásicos , Proteína p53 Supresora de Tumor , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Proto-Oncogénicas c-mdm2 , Antineoplásicos/farmacología , Imidazoles/farmacología , Imidazoles/metabolismo , Línea Celular Tumoral , Apoptosis
8.
Drug Metab Pharmacokinet ; 47: 100475, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36370616

RESUMEN

Dapaconazole is a new antifungal imidazole that has been shown a high efficacy against several pathogenic fungi. This study aimed to investigate the interspecies variation in the in vitro metabolic profiles and in vivo hepatic clearance (CLH,in vivo) prediction of dapaconazole using liver microsomes from male Sprague Dawley rat, male Beagle dog and mixed gender human using a liquid chromatography coupled to tandem mass spectrometry (UHPLC-MS/MS) method. In addition, the produced metabolites were identified by ultra-high-performance liquid chromatography with quadrupole time-of-flight mass spectrometer (UHPLC-QTOF-MS/MS). The microsomal protein concentration of 0.1 mg/mL and the incubation time of 10 min were employed for the kinetics determination, resulting in a sigmoidal kinetic profile for all species evaluated. The predicted CLH,in vivo was 6.5, 11.6 and 7.5 mL/min/kg for human, rat and dog, respectively. Furthermore, five metabolized products were identified. These findings provide preliminary information for understanding dapaconazole metabolism and the interspecies differences in catalytic behaviours, supporting the choice of a suitable laboratory animal for future pharmacokinetics and metabolism studies.


Asunto(s)
Microsomas Hepáticos , Espectrometría de Masas en Tándem , Masculino , Animales , Ratas , Humanos , Perros , Microsomas Hepáticos/metabolismo , Espectrometría de Masas en Tándem/métodos , Antifúngicos , Ratas Sprague-Dawley , Cromatografía Líquida de Alta Presión/métodos , Imidazoles/metabolismo
9.
Cell Biochem Biophys ; 80(4): 633-645, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36184717

RESUMEN

The MDM2-p53 protein-protein interaction is a promising model for researchers to design, study, and discover new anticancer drugs. The design of therapeutically active compounds that can maintain or restore the binding of MDM2 to p53 has been found to limit the oncogenic activities of both. This led to the current development of a group of xanthone-core and cis-imidazoline analogs compounds, among which γ-Mangostin (GM), α-Mangostin (AM), and Nutlin exhibited their MDM2-p53 interaction inhibitory effects. Therefore, in this study, we seek to determine the mechanisms by which these compounds elicit MDM2-p53 interaction targeting. Unique to the binding of GM, AM, and Nutlin, from our findings, they share the same three active site residues Val76, Tyr50, and Gly41, which represent the top active side residues that contribute to high electrostatic energy. Consequently, the free binding energy contributed enormously to the binding of these compounds, which culminated in the high binding affinities of GM, AM, and Nutlin with high values. Furthermore, GM, AM, and Nutlin commonly interrupted the stable and compact conformation of MDM2 coupled with its active site, where Cα deviations were relatively high. We believe that our findings would assist in the design of more potent active anticancer drugs.


Asunto(s)
Antineoplásicos , Garcinia mangostana , Imidazolinas , Xantonas , Dominio Catalítico , Garcinia mangostana/metabolismo , Imidazoles/química , Imidazoles/metabolismo , Imidazoles/farmacología , Simulación de Dinámica Molecular , Piperazinas/farmacología , Unión Proteica , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Xantonas/farmacología
10.
Proc Natl Acad Sci U S A ; 119(36): e2205608119, 2022 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-36037385

RESUMEN

Cop9 signalosome (CSN) regulates the function of cullin-RING E3 ubiquitin ligases (CRLs) by deconjugating the ubiquitin-like protein NEDD8 from the cullin subunit. To understand the physiological impact of CSN function on the CRL network and cell proliferation, we combined quantitative mass spectrometry and genome-wide CRISPR interference (CRISPRi) and CRISPR activation (CRISPRa) screens to identify factors that modulate cell viability upon inhibition of CSN by the small molecule CSN5i-3. CRL components and regulators strongly modulated the antiproliferative effects of CSN5i-3, and in addition we found two pathways involved in genome integrity, SCFFBXO5-APC/C-GMNN and CUL4DTL-SETD8, that contribute substantially to the toxicity of CSN inhibition. Our data highlight the importance of CSN-mediated NEDD8 deconjugation and adaptive exchange of CRL substrate receptors in sustaining CRL function and suggest approaches for leveraging CSN inhibition for the treatment of cancer.


Asunto(s)
Replicación del ADN , Ubiquitina-Proteína Ligasas , Azepinas/metabolismo , Complejo del Señalosoma COP9/antagonistas & inhibidores , Complejo del Señalosoma COP9/genética , Complejo del Señalosoma COP9/metabolismo , Supervivencia Celular , Proteínas Cullin/genética , Proteínas Cullin/metabolismo , Imidazoles/metabolismo , Proteína NEDD8/metabolismo , Pirazoles/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
11.
Cells ; 11(14)2022 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-35883606

RESUMEN

Immunotherapy is an attractive therapeutic strategy for the treatment of osteosarcoma (OS). The unique features of γδ T cells have made them popular for cancer immunotherapy. Here, we expanded γδ T cells using human peripheral blood mononuclear cells (PBMCs) and investigated their therapeutic potential against OS cells. PBMCs from healthy donors were cultured for 10 days with CON medium (unstimulated control); EX media, CON with recombinant human interleukin-2 (rhIL-2) and zoledronate; and EX28 media, CON with rhIL-2, zoledronate, and CD3/CD28 activator. The expanded γδ T cells were isolated by magnetic cell separation or fluorescence-activated cell sorting, cultured with two OS cell lines (KHOS/NP and MG-63) at various cell ratios with or without doxorubicin or ifosfamide, and analyzed for cytotoxicity and cytokine secretion. The number of CD3+γδTCR+Vγ9+ triple-positive γδ T cells and concentrations of IFN-γ and TNF-α were highest in the rhIL-2 (100 IU) and zoledronate (1 µM) supplemented culture conditions. The CD3/CD28 agonist did not show any additional effects on γδ T cell expansion. The expanded γδ T cells exhibited potent in vitro cytotoxicity against OS in a ratio- and time-dependent manner. The γδ T cells may enhance the effect of chemotherapeutic agents against OS and may be a new treatment strategy, including chemo-immunotherapy, for OS.


Asunto(s)
Neoplasias Óseas , Osteosarcoma , Receptores de Antígenos de Linfocitos T gamma-delta , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/terapia , Antígenos CD28/metabolismo , Difosfonatos/metabolismo , Difosfonatos/farmacología , Humanos , Imidazoles/metabolismo , Imidazoles/farmacología , Leucocitos Mononucleares/metabolismo , Osteosarcoma/metabolismo , Osteosarcoma/terapia , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/uso terapéutico , Linfocitos T/metabolismo , Linfocitos T/trasplante , Ácido Zoledrónico/farmacología
12.
Int J Mol Sci ; 23(10)2022 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-35628239

RESUMEN

Triazole and imidazole fungicides represent an emerging class of pollutants with endocrine-disrupting properties. Concerning mammalian reproduction, a possible causative role of antifungal compounds in inducing toxicity has been reported, although currently, there is little evidence about potential cooperative toxic effects. Toxicant-induced oxidative stress (OS) may be an important mechanism potentially involved in male reproductive dysfunction. Thus, to clarify the molecular mechanism underlying the effects of azoles on male reproduction, the individual and combined potential of fluconazole (FCZ), prochloraz (PCZ), miconazole (MCZ), and ketoconazole (KCZ) in triggering in vitro toxicity, redox status alterations, and OS in mouse TM4 Sertoli cells (SCs) was investigated. In the present study, we demonstrate that KCZ and MCZ, alone or in synergistic combination with PCZ, strongly impair SC functions, and this event is, at least in part, ascribed to OS. In particular, azoles-induced cytotoxicity is associated with growth inhibitory effects, G0/G1 cell cycle arrest, mitochondrial dysfunction, reactive oxygen species (ROS) generation, imbalance of the superoxide dismutase (SOD) specific activity, glutathione (GSH) depletion, and apoptosis. N-acetylcysteine (NAC) inhibits ROS accumulation and rescues SCs from azole-induced apoptosis. PCZ alone exhibits only cytostatic and pro-oxidant properties, while FCZ, either individually or in combination, shows no cytotoxic effects up to 320 µM.


Asunto(s)
Cetoconazol , Miconazol , Animales , Apoptosis , Glutatión/metabolismo , Imidazoles/metabolismo , Imidazoles/farmacología , Cetoconazol/farmacología , Masculino , Mamíferos/metabolismo , Ratones , Miconazol/farmacología , Mitocondrias/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo
13.
Mar Drugs ; 20(5)2022 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-35621964

RESUMEN

Balenine is one of the endogenous imidazole dipeptides derived from marine products. It is composed of beta-alanine and 3-methyl-L-histidine, which exist mainly in the muscles of marine organisms. The physiological functions of dietary balenine are not well-known. In this study, we investigated whether the supplementation of dietary balenine was associated with muscle function in a cardiotoxin-indued muscle degeneration/regeneration model. Through morphological observation, we found that the supplementation of balenine-enriched extract promoted the regeneration stage. In addition, the expression of regeneration-related myogenic marker genes, such as paired box protein 7, MyoD1, myogenin, and Myh3, in a group of mice fed a balenine-enriched extract diet was higher than that in a group fed a normal diet. Moreover, the supplementation of balenine-enriched extract promoted the expression of anti-inflammatory cytokines as well as pro-inflammatory cytokines at the degeneration stage. Interestingly, phagocytic activity in the balenine group was significantly higher than that in the control group in vitro. These results suggest that balenine may promote the progress of muscle regeneration by increasing the phagocytic activity of macrophages.


Asunto(s)
Dipéptidos , Macrófagos , Músculo Esquelético , Fagocitosis , Animales , Citocinas/metabolismo , Dipéptidos/metabolismo , Dipéptidos/farmacología , Imidazoles/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/fisiología , Ratones , Músculo Esquelético/inmunología , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiología , Fagocitosis/efectos de los fármacos
14.
Food Chem Toxicol ; 164: 113051, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35460824

RESUMEN

Individuals of all ages, including children and teenagers, consume 4-methylimidazole (4-MI) in their food. 4-MI is a caramel-colored waste product that has previously been linked to human carcinogenesis and has shown possible signs of reproductive toxicity. This study aimed to determine whether 4-MI is harmful to oocytes during meiosis and fertilization. Female mice were intragastrically administered 0, 50, or 100 mg/kg body weight of 4-MI daily for 10 days. We found that 4-MI affects the quality of oocytes by affecting their meiotic ability and fertility potential. Specifically, 4-MI rendered the meiotic spindles and chromosomes less stable, which halted oocyte maturation and resulted in aneuploidy. 4-MI also slowed the decrease in the levels of cortical granules and their component ovastacin; consequently, sperms could not be bound and fertilization could not occur. We also found that mitochondrial dysfunction was associated with oocytes deterioration. This led to reactive oxygen species accumulation and cell death. Altogether, our findings reveal that the poor condition of oocytes subjected to 4-MI is primarily attributable to mitochondrial malfunction and redox alterations.


Asunto(s)
Meiosis , Oocitos , Animales , Femenino , Fertilización , Imidazoles/metabolismo , Ratones
15.
Biochem Biophys Res Commun ; 598: 15-19, 2022 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-35151199

RESUMEN

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.


Asunto(s)
Imidazoles/química , Imidazoles/farmacología , Inhibidores de Proteínas Quinasas/química , Piridazinas/química , Piridazinas/farmacología , Familia-src Quinasas/química , Sitios de Unión , Cristalografía por Rayos X , Humanos , Imidazoles/metabolismo , Modelos Moleculares , Mutación , Conformación Proteica , Dominios Proteicos , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-hck/química , Proteínas Proto-Oncogénicas c-hck/metabolismo , Piridazinas/metabolismo , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
16.
ACS Appl Mater Interfaces ; 14(3): 3675-3684, 2022 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-35020346

RESUMEN

Exploiting zeolitic imidazolate framework (ZIF)-based nanoparticles to synergistically enhance starvation-combined chemotherapy strategies remains an urgent demand. Herein, glucose oxidase (GOX) and doxorubicin (DOX) were facilely incorporated into ZIFs for starvation-combined chemotherapy. The as-prepared DOX/GOX-loaded ZIF (DGZ) exhibited uniform size with good dispersity, effective protection of the GOX activity, and stable delivery of the drugs into tumor. Correspondingly, it could achieve the glucose- and pH-responsive degradation and thus the controllable drug release. As a result, the acidification of glucose accompanied with reactive oxygen species (ROS) production was observed for the starvation-enhanced chemotherapy and the improved degradation. Most importantly, adjustable Zn2+ release was achieved with the biodegradation of DGZ, which thus contributed to an augmented therapeutic outcome via the Zn2+-induced mitochondrial dysfunction and antioxidation dyshomeostasis. These findings, synergized with the enhancement of starvation-combined chemotherapy by inhibiting the mitochondrial energy metabolism and boosting the ROS accumulation using pristine ZIF-based nanoparticles, provide a new insight into the metal-organic framework-based nanomedicine for further cancer treatments.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Antioxidantes/farmacología , Materiales Biocompatibles/farmacología , Doxorrubicina/farmacología , Estructuras Metalorgánicas/farmacología , Neoplasias/tratamiento farmacológico , Antibióticos Antineoplásicos/química , Antibióticos Antineoplásicos/metabolismo , Antioxidantes/química , Antioxidantes/metabolismo , Materiales Biocompatibles/química , Materiales Biocompatibles/metabolismo , Doxorrubicina/química , Doxorrubicina/metabolismo , Glucosa Oxidasa/metabolismo , Homeostasis/efectos de los fármacos , Humanos , Imidazoles/química , Imidazoles/metabolismo , Imidazoles/farmacología , Ensayo de Materiales , Estructuras Metalorgánicas/química , Estructuras Metalorgánicas/metabolismo , Dinámicas Mitocondriales/efectos de los fármacos , Neoplasias/metabolismo , Zeolitas/química , Zeolitas/metabolismo , Zeolitas/farmacología
17.
J Med Chem ; 65(3): 2342-2360, 2022 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-35007061

RESUMEN

Chemical probes for epigenetic proteins are essential tools for dissecting the molecular mechanisms for gene regulation and therapeutic development. The bromodomain and extra-terminal (BET) proteins are master transcriptional regulators. Despite promising therapeutic targets, selective small molecule inhibitors for a single bromodomain remain an unmet goal due to their high sequence similarity. Here, we address this challenge via a structure-activity relationship study using 1,4,5-trisubstituted imidazoles against the BRD4 N-terminal bromodomain (D1). Leading compounds 26 and 30 have 15 and 18 nM affinity against BRD4 D1 and over 500-fold selectivity against BRD2 D1 and BRD4 D2 via ITC. Broader BET selectivity was confirmed by fluorescence anisotropy, thermal shift, and CETSA. Despite BRD4 engagement, BRD4 D1 inhibition was unable to reduce c-Myc expression at low concentration in multiple myeloma cells. Conversely, for inflammation, IL-8 and chemokine downregulation were observed. These results provide new design rules for selective inhibitors of an individual BET bromodomain.


Asunto(s)
Proteínas de Ciclo Celular/antagonistas & inhibidores , Imidazoles/farmacología , Factores de Transcripción/antagonistas & inhibidores , Sitios de Unión , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Diseño de Fármacos , Humanos , Imidazoles/química , Imidazoles/metabolismo , Estructura Molecular , Unión Proteica , Dominios Proteicos , Proteínas Proto-Oncogénicas c-myc/metabolismo , Relación Estructura-Actividad , Factores de Transcripción/química , Factores de Transcripción/metabolismo
18.
Carbohydr Polym ; 275: 118717, 2022 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-34742441

RESUMEN

In this paper, a reinforced Biomass Gel Artificial Muscle (BGAM) was fabricated by natural polysaccharide of Sodium Alginate (SA) doped with an Ionic Liquid (IL) of 1-ethyl-3-methylimidazolium tetrafluoroborate ([EMIm][BF4]). Micro-nano regulation effect and reinforcement mechanism of IL doping content on electromechanical response performance of BGAM were researched during a single cycle and repeated cycles. Then, a green fabrication process and a set of valid test methods for BGAM were proposed in detail. The experimental results showed that when IL doping content was 4 mL, the BGAM achieved optimal modification, with a porosity of 70.47%, where it internally adopted the porous polymer structure of ion channels. Additionally, specific capacitance of BGAM attained a maximum value of 126.98 mF/g, and the inner resistance and elastic modulus reached minimum values of 2.018 Ω and 1.871 MPa, separately. Thus, the optimal working life and output-force density values, namely, 1720 s and 13.072 mN/g, respectively, were also determined for the BGAM.


Asunto(s)
Alginatos/metabolismo , Imidazoles/metabolismo , Líquidos Iónicos/metabolismo , Polisacáridos/metabolismo , Alginatos/química , Biomasa , Capacidad Eléctrica , Geles/química , Geles/metabolismo , Imidazoles/química , Líquidos Iónicos/química , Tamaño de la Partícula , Polisacáridos/química
19.
ISME J ; 16(1): 211-220, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34290379

RESUMEN

Aerobic methanotrophy is strongly controlled by copper, and methanotrophs are known to use different mechanisms for copper uptake. Some methanotrophs secrete a modified polypeptide-methanobactin-while others utilize a surface-bound protein (MopE) and a secreted form of it (MopE*) for copper collection. As different methanotrophs have different means of sequestering copper, competition for copper significantly impacts methanotrophic activity. Herein, we show that Methylomicrobium album BG8, Methylocystis sp. strain Rockwell, and Methylococcus capsulatus Bath, all lacking genes for methanobactin biosynthesis, are not limited for copper by multiple forms of methanobactin. Interestingly, Mm. album BG8 and Methylocystis sp. strain Rockwell were found to have genes similar to mbnT that encodes for a TonB-dependent transporter required for methanobactin uptake. Data indicate that these methanotrophs "steal" methanobactin and such "theft" enhances the ability of these strains to degrade methylmercury, a potent neurotoxin. Further, when mbnT was deleted in Mm. album BG8, methylmercury degradation in the presence of methanobactin was indistinguishable from when MB was not added. Mc. capsulatus Bath lacks anything similar to mbnT and was unable to degrade methylmercury either in the presence or absence of methanobactin. Rather, Mc. capsulatus Bath appears to rely on MopE/MopE* for copper collection. Finally, not only does Mm. album BG8 steal methanobactin, it synthesizes a novel chalkophore, suggesting that some methanotrophs utilize both competition and cheating strategies for copper collection. Through a better understanding of these strategies, methanotrophic communities may be more effectively manipulated to reduce methane emissions and also enhance mercury detoxification in situ.


Asunto(s)
Compuestos de Metilmercurio , Methylosinus trichosporium , Cobre/metabolismo , Imidazoles/metabolismo , Compuestos de Metilmercurio/metabolismo , Methylosinus trichosporium/genética , Methylosinus trichosporium/metabolismo , Oligopéptidos/metabolismo
20.
Rapid Commun Mass Spectrom ; 36(5): e9240, 2022 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-34904306

RESUMEN

RATIONALE: Tirabrutinib is an orally administered Bruton's tyrosine kinase (BTK) inhibitor developed for the treatment of autoimmune disorders and haematological malignancies. The goals of this study were to identify the metabolites of tirabrutinib and to propose the metabolic pathways. METHODS: Tirabrutinib was individually incubated with rat, dog and human liver microsomes at 37°C for 1 h. To trap the potential reactive metabolites, glutathione (GSH) was incorporated into the incubation samples. The incubation samples were analysed using ultra-high-performance liquid chromatography combined with high-resolution mass spectrometry (UHPLC-HRMS). The metabolites were identified and characterized by exact masses, product ions and retention times. RESULTS: A total of 18 metabolites, including four GSH conjugates, were identified and characterized in terms of elemental compositions and product ions. The metabolic pathways of tirabrutinib included amide hydrolysis, O-dealkylation, mono-oxygenation, di-oxygenation and GSH conjugation. Among these metabolites, M10 was the most abundant metabolite. Compared with dog, rat has the closer metabolic profiles to humans, and thus it would be more suitable for toxicity study. CONCLUSIONS: This study provides valuable data regarding the in vitro metabolism of tirabrutinib, which may be helpful for further safety assessment of this drug.


Asunto(s)
Imidazoles/química , Imidazoles/metabolismo , Microsomas Hepáticos/metabolismo , Pirimidinas/química , Pirimidinas/metabolismo , Animales , Cromatografía Líquida de Alta Presión , Perros , Humanos , Espectrometría de Masas , Metaboloma , Microsomas Hepáticos/química , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA