Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 166
Filtrar
1.
Fitoterapia ; 168: 105565, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37295753

RESUMEN

Oxymatrine (OMT), was identified as a quinolizidine alkaloid, which was one of the major matrine-type alkaloids extracted from Sophora medicinal plants. Growing studies revealed that OMT has a wide range of beneficial pharmacological values, consisting of anticancer, antidiabetic, antivirus, and antiinflammtion, as well as the protective activities to the brain, liver, heart, lung, vascular, gastrointestinal, bone, kidney, and skin organs. Various in vitro and in vivo models of pharmacological actions were recorded in regard to the usage of alkaloidal OMT. Mechanisms underlying anticancer activity of this compound may have been possibly involved anti-proliferation, invasion, migration, angiogenesis, epithelial-mesenchymal transition of cells, autophagy, especially apoptotic cell deaths. OMT could reduce hyperglycemia and hyperlipemia in a high-fat diet and streptozotocin-stimulated diabetic mice by improving insulin secretion and sensitivity. OMT suppressed gastric ulcer via gastric inflammatory and oxidative inhibitions, and pro-apoptotic actions. It turns out that OMT is relatively safe for cell and animal experiments. In this study, we offer a systematic review of natural occurrence, pharmacological potentials, possible mechanisms of action, pharmacokinetics, and bioavailability. Clinical research with OMT is needed to extensively elucidate its health potential benefits.


Asunto(s)
Alcaloides , Diabetes Mellitus Experimental , Ratones , Animales , Estructura Molecular , Alcaloides/farmacología , Matrinas , Quinolizinas/farmacocinética
2.
J Nat Prod ; 86(5): 1179-1188, 2023 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-37115657

RESUMEN

Apigenin (APG) is a well-known dietary flavonoid with multiple bioactivities, but its poor aqueous solubility may result in low oral bioavailability and thus compromised therapeutic effects. In the present study, APG was complexed with oxymatrine (OMT), a natural quinolizidine alkaloid, for enhanced anti-inflammatory activity, and the related mechanisms in the interaction of APG with OMT were investigated. Fourier transform-infrared spectroscopy, fluorescence spectroscopy, Raman spectroscopy, and proton nuclear magnetic resonance spectroscopy characterizations demonstrated the occurrence of an APG-OMT complex formed at a molar ratio of 1:2. Then, molecular dynamics simulations and quantum chemical calculations were utilized to elucidate that hydrogen bonding, van der Waals forces, and hydrophobic effects were the main forces acting in the formation of the APG-OMT complex. Pharmacokinetic studies in rats demonstrated that the oral bioavailability of APG in the APG-OMT complex was significantly higher than that of APG alone. Finally, bioactivity evaluation in the lipopolysaccharide-induced acute inflammatory injury mouse models showed that the APG-OMT complex exhibited more potent anti-inflammatory effects than APG alone. This study confirmed that APG and OMT exerted enhanced anti-inflammatory effects through self-complexation, which may provide a novel strategy for improving the bioavailability and bioactivity of natural product mixtures.


Asunto(s)
Alcaloides , Apigenina , Ratones , Ratas , Animales , Apigenina/farmacología , Apigenina/química , Alcaloides/farmacocinética , Matrinas , Antiinflamatorios/farmacología , Quinolizinas/farmacocinética
3.
J Nat Prod ; 84(9): 2486-2495, 2021 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-34463097

RESUMEN

This study aims to characterize the pharmacokinetic (PK) profiles and identify important bioavailability barriers and pharmacological pathways of the key active components (KACs) of Antitumor B (ATB), a chemopreventive agent. KACs (matrine, dictamine, fraxinellone, and maackiain) of ATB were confirmed using the antiproliferative assay and COX-2 inhibition activities in oral cancer cells. The observed in vitro activities of KACs were consistent with their cell signaling pathways predicted using the in silico network pharmacology approach. The pharmacokinetics of KACs were determined after i.v., i.p., and p.o. delivery using ATB extract and a mixture of four KACs in mice. Despite good solubilities and permeabilities, poor oral bioavailabilities were estimated for all KACs, mostly because of first-pass metabolism in the liver (for all KACs) and intestines (for matrine and fraxinellone). Multiple-dose PK studies showed 23.2-fold and 8.5-fold accumulation of dictamine and maackiain in the blood, respectively. Moreover, saliva levels of dictamine and matrine were found significantly higher than their blood levels. In conclusion, the systemic bioavailabilities of ATB-KACs were low, but significant levels of dictamine and matrine were found in saliva upon repeated oral administration. Significant salivary concentrations of matrine justified its possible use as a drug-monitoring tool to track patient compliance during chemoprevention trials.


Asunto(s)
Disponibilidad Biológica , Medicamentos Herbarios Chinos/farmacocinética , Neoplasias de la Boca/prevención & control , Alcaloides/farmacocinética , Animales , Benzofuranos/farmacocinética , Quimioprevención , Ratones , Ratones Endogámicos C57BL , Estructura Molecular , Farmacología en Red , Pterocarpanos/farmacocinética , Quinolinas/farmacocinética , Quinolizinas/farmacocinética , Matrinas
4.
Rapid Commun Mass Spectrom ; 35(18): e9157, 2021 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-34182613

RESUMEN

RATIONALE: Characterizing the functional mechanism of quality control marker (Q-marker) was of great importance in revealing the primary pharmacological mechanism of herbs or the other complex system, and drug-related metabolites always contribute to the pharmacological functions. Cortex Phellodendri was used as a core herb in the treatment of diabetes mellitus (DM). As a Q-marker of Cortex Phellodendri, the role of phellodendrine in DM was still unclear. Thus, the characterization of phellodendrine-related metabolites in vivo and the subsequent induced functional mechanism exerted great importance in elucidating the anti-DM mechanism of Cortex Phellodendri. METHODS: An ultra-high-performance liquid chromatography-coupled time-of-flight mass spectrometry (UHPLC/Q-TOF MS) method was developed to profile metabolites of phellodendrine in rats. The potential pharmacological mechanism against DM was predicted by network pharmacology. RESULTS: A total of 19 phellodendrine-related metabolites were screened out in rats for the first time. Among them, M4, M5, M9, and M12 were regarded as the primary metabolites. Meanwhile, phase I metabolic reactions of hydroxylation, demethylation, and isomerization and phase II reactions of glucuronidation and sulfation occurred to phellodendrine; glucuronidation and hydroxylation were the two main metabolic reactions. Moreover, the potential targets of phellodendrine and three main metabolites (M4, M5, and M12) were predicted by a network pharmacological method, and they mainly shared 52 targets, including PDE5A, CHRNA3, SIGMAR1, F3, ESR1, DRD1, DRD2, DRD3, and DRD4. Furthermore, Kyoto Encyclopedia of Genes and Genomes pathway analysis showed that calcium signaling pathway, cGMP-PKG signaling pathway, and cAMP signaling pathway were regarded as the core mechanism of phellodendrine to treat DM. CONCLUSION: The metabolic feature of phellodendrine in vivo was revealed for the first time, and its anti-DM mechanism information for further pharmacological validations was also supplied. It also gave a direction to further elucidation of pharmacological mechanism of Cortex Phellodendri in treating DM.


Asunto(s)
Cromatografía Líquida de Alta Presión/métodos , Diabetes Mellitus/tratamiento farmacológico , Medicamentos Herbarios Chinos/farmacocinética , Espectrometría de Masas/métodos , Quinolizinas/farmacocinética , Animales , Diabetes Mellitus/metabolismo , Medicamentos Herbarios Chinos/administración & dosificación , Medicamentos Herbarios Chinos/metabolismo , Humanos , Masculino , Farmacología en Red , Quinolizinas/administración & dosificación , Quinolizinas/metabolismo , Ratas , Ratas Sprague-Dawley
5.
Pharmacol Res ; 170: 105700, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34087351

RESUMEN

Cytisine, a natural bioactive compound that is mainly isolated from plants of the Leguminosae family (especially the seeds of Laburnum anagyroides), has been marketed in central and eastern Europe as an aid in the clinical management of smoking cessation for more than 50 years. Its main targets are neuronal nicotinic acetylcholine receptors (nAChRs), and pre-clinical studies have shown that its interactions with various nAChR subtypes located in different areas of the central and peripheral nervous systems are neuroprotective, have a wide range of biological effects on nicotine and alcohol addiction, regulate mood, food intake and motor activity, and influence the autonomic and cardiovascular systems. Its relatively rigid conformation makes it an attractive template for research of new derivatives. Recent studies of structurally modified cytisine have led to the development of new compounds and for some of them the biological activities are mediated by still unidentified targets other than nAChRs, whose mechanisms of action are still being investigated. The aim of this review is to describe and discuss: 1) the most recent pre-clinical results obtained with cytisine in the fields of neurological and non-neurological diseases; 2) the effects and possible mechanisms of action of the most recent cytisine derivatives; and 3) the main areas warranting further research.


Asunto(s)
Alcaloides/farmacología , Sistema Nervioso/efectos de los fármacos , Receptores Nicotínicos/efectos de los fármacos , Agentes para el Cese del Hábito de Fumar/farmacología , Cese del Hábito de Fumar , Alcaloides/farmacocinética , Alcaloides/toxicidad , Animales , Azocinas/farmacocinética , Azocinas/farmacología , Azocinas/toxicidad , Humanos , Estructura Molecular , Sistema Nervioso/metabolismo , Quinolizinas/farmacocinética , Quinolizinas/farmacología , Quinolizinas/toxicidad , Receptores Nicotínicos/metabolismo , Agentes para el Cese del Hábito de Fumar/farmacocinética , Agentes para el Cese del Hábito de Fumar/toxicidad , Relación Estructura-Actividad
6.
J Ethnopharmacol ; 267: 113471, 2021 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-33075440

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Vinegar-baked Radix Bupleuri (VBRB) is a processed form of Bupleurum chinense DC. As a well-known meridian-guiding drug, it is traditionally used as a component of traditional Chinese medicine formulations indicated for the treatment of liver diseases. However, the liver targeting component in VBRB remains unclear. Therefore, this study aims to explore the efficacy and mechanism of PSS (polysaccharides in Vinegar-baked Radix Bupleuri) in enhancing liver targeting. MATERIALS AND METHODS: Drug distribution of OM alone or combined with PSS was investigated in vivo. Relative uptake efficiency (RUE) and relative targeting efficiency (RTE) were calculated to evaluate liver targeting efficiency. The mRNA and protein expression of organic cation transporter 1 (OCT1), multi-drug resistance protein 2 (Mrp2), and hepatocyte nuclear factor 4α (HNF4α) in the liver were determined by q-PCR and Western blot. Then, AZT, the inhibitor of OCT1 and BI6015, the inhibitor of HNF4α were used to investigate regulatory mechanisms involved in the uptake of OM in the cell. At last, the role of PSS in the anti-hepatitis B virus (HBV) was explored on HepG2.2.15. RESULTS: PSS increased the AUC of OM in the liver and increase the RUE and RTE in the liver which indicated a liver targeting enhancing effect. The mRNA and protein expression of OCT1 was increased while Mrp2 and HNF4α decreased. PSS could increase the uptake of OM in HepG2 by increasing the protein expression of HNF4α and OCT1, while inhibited Mrp2. Moreover, PSS combined with OM could enhance the anti-HBV effect of OM. CONCLUSION: PSS enhanced the liver targeting efficiency and the underlying mechanism related to up-regulating the expression of OCT1 and HNF4α, while down-regulating of Mrp2. These results suggest that PSS may become a potential excipient and provide a new direction for new targeted research.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Ácido Acético/química , Alcaloides/farmacología , Alcaloides/farmacocinética , Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/metabolismo , Culinaria , Factor Nuclear 4 del Hepatocito/metabolismo , Hígado/efectos de los fármacos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Extractos Vegetales/farmacología , Polisacáridos/farmacología , Quinolizinas/farmacología , Quinolizinas/farmacocinética , Transportadoras de Casetes de Unión a ATP/genética , Animales , Bupleurum/química , Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/genética , Regulación de la Expresión Génica , Células Hep G2 , Factor Nuclear 4 del Hepatocito/genética , Calor , Humanos , Hígado/metabolismo , Masculino , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Extractos Vegetales/química , Polisacáridos/química , Ratas Sprague-Dawley , Distribución Tisular
7.
J Ethnopharmacol ; 269: 113682, 2021 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-33307055

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: "Dogel ebs" was known as Sophora flavescens Ait., which has been widely utilized in the clinical practice of traditional Chinese Mongolian herbal medicine for thousands of years. Shen Nong's Materia Medica (Shen Nong Ben Cao Jing in Chinese pinyin) recorded that it is bitter in taste and cold in nature with the effect of clearing heat and eliminating dampness, insecticide, diuresis. Due to its extensive application in the fields of ethnopharmacological utilization, the pharmaceutical researches of Sophora flavescens Ait.s keeps deepening. Modern pharmacological studies have exhibited that matrine, which is rich in this traditional herbal medicine, mediates its main biological properties. AIMS OF THE REVIEW: This review aimed at summarizing the latest and comprehensive information of matrine on the pharmacology, pharmacokinetics, toxicity, clinical application and preparation researches to explore the therapeutic potential of this natural ingredient. In addition, outlooks and perspective for possible future researches that related are also discussed. MATERIALS AND METHODS: Related information concerning matrine was gathered from the internet database of Google scholar, Pubmed, ResearchGate, Web of Science and Wiley Online Library with the keywords including "matrine", "pharmacology", "toxicology" and "pharmacokinetics", "clinical application", etc. RESULTS: Based on literatures, matrine has a variety of pharmacological effects, including anti-cancer, anti-inflammatory, anti-microbial, detoxification and so on. Nevertheless, there are still some doubts about it due to the toxicity and questionable bioavailability that does exist. CONCLUSIONS: Future researches directions probably include elucidate the mechanism of its toxicity and accurately tracing the in vivo behavior of its drug delivery system. Without doubt, integration of toxicity and efficiency and structure modification based on it are also pivotal methods to enhance pharmacological activity and bioavailability.


Asunto(s)
Alcaloides/farmacocinética , Alcaloides/uso terapéutico , Medicamentos Herbarios Chinos/farmacocinética , Medicamentos Herbarios Chinos/uso terapéutico , Etnofarmacología/métodos , Medicina Tradicional China/métodos , Quinolizinas/farmacocinética , Quinolizinas/uso terapéutico , Alcaloides/toxicidad , Animales , Antihelmínticos/farmacocinética , Antihelmínticos/uso terapéutico , Antihelmínticos/toxicidad , Antiinflamatorios/farmacocinética , Antiinflamatorios/uso terapéutico , Antiinflamatorios/toxicidad , Antineoplásicos Fitogénicos/farmacocinética , Antineoplásicos Fitogénicos/uso terapéutico , Antineoplásicos Fitogénicos/toxicidad , Medicamentos Herbarios Chinos/toxicidad , Humanos , Quinolizinas/toxicidad , Matrinas
8.
J Med Chem ; 63(23): 14700-14723, 2020 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-33297683

RESUMEN

PI3Kδ inhibitors have been approved for B-cell malignancies like CLL, small lymphocytic lymphoma, and so forth. However, currently available PI3Kδ inhibitors are nonoptimal, showing weakness against at least one of the several important properties: potency, isoform selectivity, and/or pharmacokinetic profile. To come up with a PI3Kδ inhibitor that overcomes all these deficiencies, a pharmacophoric expansion strategy was employed. Herein, we describe a systematic transformation of a "three-blade propeller" shaped lead, 2,3-disubstituted quinolizinone 11, through a 1,2-disubstituted quinolizinone 20 to a novel "four-blade propeller" shaped 1,2,3-trisubstituted quinolizinone 34. Compound 34 has excellent potency, isoform selectivity, metabolic stability across species, and exhibited a favorable pharmacokinetic profile. Compound 34 also demonstrated a differentiated efficacy profile in human germinal center B and activated B cell-DLBCL cell lines and xenograft models. Compound 34 qualifies for further evaluation as a candidate for monotherapy or in combination with other targeted agents in DLBCLs and other forms of iNHL.


Asunto(s)
Antineoplásicos/uso terapéutico , Fosfatidilinositol 3-Quinasa Clase I/uso terapéutico , Neoplasias Hematológicas/tratamiento farmacológico , Inhibidores de las Quinasa Fosfoinosítidos-3/uso terapéutico , Quinolizinas/uso terapéutico , Animales , Antineoplásicos/síntesis química , Antineoplásicos/metabolismo , Antineoplásicos/farmacocinética , Línea Celular Tumoral , Fosfatidilinositol 3-Quinasa Clase I/síntesis química , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Fosfatidilinositol 3-Quinasa Clase I/farmacocinética , Perros , Descubrimiento de Drogas , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones SCID , Simulación del Acoplamiento Molecular , Estructura Molecular , Inhibidores de las Quinasa Fosfoinosítidos-3/síntesis química , Inhibidores de las Quinasa Fosfoinosítidos-3/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacocinética , Quinolizinas/síntesis química , Quinolizinas/metabolismo , Quinolizinas/farmacocinética , Células RAW 264.7 , Ratas Sprague-Dawley , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Biol Pharm Bull ; 43(6): 976-984, 2020 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-32188833

RESUMEN

Cytisine N-methylene-(5,7-dihydroxy-4'-methoxy)-isoflavone (CNF2) is a new compound isolated from the Chinese herbal medicine Sophora alopecuroides. Preliminary pharmacodynamic studies demonstrated its activity in inhibiting breast cancer cell metastasis. This study examined the pharmacokinetics, absolute bioavailability, and tissue distribution of CNF2 in rats, and combined computer-aided technology to predict the druggability of CNF2. The binding site of CNF2 and the breast cancer target human epidermal growth factor receptor-2 (HER2) were examined with molecular docking technology. Next, ACD/Percepta software was used to predict the druggability of CNF2 based on the quantitative structure-activity relationship (QSAR). Finally, a simple and effective HPLC method was used to determine plasma pharmacokinetics and tissue distribution of CNF2 in rats. Prediction and experimental results show that compared with the positive control HER2 inhibitor SYR127063, CNF2 has a stronger binding affinity with HER2, suggesting that its efficacy is stronger; and the structure of CNF2 complies with the Lipinski's Rule of Five and has good drug-likeness. The residence time of CNF2 in rats is less than 4 h, and the metabolic rate is relatively fast; But the absolute bioavailability of CNF2 in rats was 6.6%, mainly distributed in the stomach, intestine, and lung tissues, where the CNF2 contents were 401.20, 144.01, and 245.82 µg/g, respectively. This study constructed rapid screening and preliminary evaluation of active compounds, which provided important references for the development and further research of such compounds.


Asunto(s)
Alcaloides/química , Alcaloides/farmacocinética , Antineoplásicos/química , Antineoplásicos/farmacocinética , Isoflavonas/química , Isoflavonas/farmacocinética , Alcaloides/sangre , Animales , Antineoplásicos/sangre , Azocinas/sangre , Azocinas/química , Azocinas/farmacocinética , Femenino , Isoflavonas/sangre , Hígado/metabolismo , Simulación del Acoplamiento Molecular , Quinolizinas/sangre , Quinolizinas/química , Quinolizinas/farmacocinética , Ratas Sprague-Dawley , Distribución Tisular
10.
J Vet Pharmacol Ther ; 43(1): 1-5, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31318080

RESUMEN

Dexmedetomidine is an alpha-2 adrenoceptor agonist, and vatinoxan is an alpha-2 antagonist believed to poorly cross the blood-brain barrier in cats. Dexmedetomidine-vatinoxan combinations are of interest in anesthetized cats because the anesthetic sparing effect of dexmedetomidine may be preserved while vatinoxan attenuates the adverse cardiovascular effects of dexmedetomidine. The aim of this study was to characterize the pharmacokinetics of dexmedetomidine in cats during administration of isoflurane and vatinoxan. Six healthy adult male castrated cats were anesthetized with isoflurane in oxygen. Vatinoxan was administered using a target-controlled infusion system intended to maintain a plasma concentration of 4 µg/ml. Dexmedetomidine, 35 µg/kg was administered intravenously over 5 min. Plasma dexmedetomidine and vatinoxan concentrations were measured at selected time points ranging from prior to 8 hr after dexmedetomidine administration using liquid chromatography/tandem mass spectrometry. Compartment models were fitted to the time-concentration data using nonlinear mixed-effect modeling. A three-compartment model best fitted the data. Typical value (% interindividual variability) for the three-compartment volumes (ml/kg), the metabolic clearance and the two intercompartment distribution clearances (ml min-1 kg-1 ) were 168 (259), 318 (35), 1,425 (18), 12.4 (31), 39.1 (18), and 29.6 (17), respectively. Mean ± standard deviation plasma vatinoxan concentration was 2.6 ± 0.6 µg/ml.


Asunto(s)
Anestesia/veterinaria , Gatos/fisiología , Dexmedetomidina/farmacocinética , Isoflurano/farmacología , Quinolizinas/farmacocinética , Anestésicos por Inhalación/administración & dosificación , Anestésicos por Inhalación/farmacología , Animales , Dexmedetomidina/administración & dosificación , Dexmedetomidina/farmacología , Interacciones Farmacológicas , Hipnóticos y Sedantes/administración & dosificación , Hipnóticos y Sedantes/farmacocinética , Hipnóticos y Sedantes/farmacología , Isoflurano/administración & dosificación , Masculino , Quinolizinas/administración & dosificación , Quinolizinas/farmacología
11.
Vet Anaesth Analg ; 47(1): 70-75, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31806431

RESUMEN

OBJECTIVE: To characterize the pharmacokinetics of vatinoxan in isoflurane-anesthetized cats. STUDY DESIGN: Prospective experimental study. ANIMALS: A group of six adult healthy male neutered cats. METHODS: Cats were anesthetized using isoflurane in oxygen. Venous catheters were placed to administer the drug and sample blood. Vatinoxan, 1 mg kg-1, was administered intravenously over 5 minutes. Blood was sampled before and at various times during and up to 8 hours after vatinoxan administration. Plasma vatinoxan concentration was measured using liquid chromatography/tandem mass spectrometry. Compartment models were fitted to the time-concentration data using population methods and nonlinear mixed effect modeling. RESULTS: A three-compartment model best fitted the data. Typical value (% interindividual variability) for the three volumes (mL kg-1), the metabolic clearance and two distribution clearances (mL minute-1 kg-1) were 34 (55), 151 (35), 306 (18), 2.3 (34), 42.6 (25) and 5.6 (0), respectively. Hypotension increased the second distribution clearance to 10.6. CONCLUSION AND CLINICAL RELEVANCE: The pharmacokinetics of vatinoxan in anesthetized cats were characterized by a small volume of distribution and a low clearance. An intravenous bolus of 100 µg kg-1 of vatinoxan followed by constant rate infusions of 55 µg kg-1 minute-1 for 20 minutes, then 22 µg kg-1 minute-1 for 60 minutes and finally 10 µg kg-1 minute-1 for the remainder of the infusion time is expected to maintain the plasma concentration within 90%-110% of the plasma vatinoxan concentration previously shown to attenuate the cardiovascular effects of dexmedetomidine (25 µg kg-1) in conscious cats.


Asunto(s)
Anestesia/veterinaria , Gatos/metabolismo , Quinolizinas/farmacocinética , Anestésicos por Inhalación/uso terapéutico , Animales , Infusiones Intravenosas , Isoflurano/uso terapéutico , Masculino , Orquiectomía , Quinolizinas/administración & dosificación , Quinolizinas/sangre
12.
Vet Anaesth Analg ; 47(1): 47-52, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31761600

RESUMEN

OBJECTIVE: To quantify the peripheral selectivity of vatinoxan (L-659,066, MK-467) in dogs by comparing the concentrations of vatinoxan, dexmedetomidine and levomedetomidine in plasma and central nervous system (CNS) tissue after intravenous (IV) coadministration of vatinoxan and medetomidine. STUDY DESIGN: Experimental, observational study. ANIMALS: A group of six healthy, purpose-bred Beagle dogs (four females and two males) aged 6.5 ± 0.1 years (mean ± standard deviation). METHODS: All dogs were administered a combination of medetomidine (40 µg kg-1) and vatinoxan (800 µg kg-1) as IV bolus. After 20 minutes, the dogs were euthanized with an IV overdose of pentobarbital (140 mg kg-1) and both venous plasma and CNS tissues (brain, cervical and lumbar spinal cord) were harvested. Concentrations of dexmedetomidine, levomedetomidine and vatinoxan in all samples were quantified by liquid chromatography-tandem mass spectrometry and data were analyzed with nonparametric tests with post hoc corrections where appropriate. RESULTS: All dogs became deeply sedated after the treatment. The CNS-to-plasma ratio of vatinoxan concentration was approximately 1:50, whereas the concentrations of dexmedetomidine and levomedetomidine in the CNS were three- to seven-fold of those in plasma. CONCLUSIONS AND CLINICAL RELEVANCE: With the doses studied, these results confirm the peripheral selectivity of vatinoxan in dogs, when coadministered IV with medetomidine. Thus, it is likely that vatinoxan preferentially antagonizes α2-adrenoceptors outside the CNS.


Asunto(s)
Antagonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Perros/metabolismo , Hipnóticos y Sedantes/farmacocinética , Medetomidina/farmacocinética , Quinolizinas/farmacocinética , Antagonistas de Receptores Adrenérgicos alfa 2/administración & dosificación , Antagonistas de Receptores Adrenérgicos alfa 2/sangre , Animales , Encéfalo/metabolismo , Quimioterapia Combinada/veterinaria , Femenino , Hipnóticos y Sedantes/administración & dosificación , Hipnóticos y Sedantes/sangre , Infusiones Intravenosas/veterinaria , Masculino , Medetomidina/administración & dosificación , Medetomidina/sangre , Tejido Nervioso/metabolismo , Quinolizinas/administración & dosificación , Quinolizinas/sangre
13.
Vet J ; 251: 105345, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31492389

RESUMEN

A constant rate infusion (CRI) of medetomidine is used to balance equine inhalation anesthesia, but its cardiovascular side effects are a concern. This experimental crossover study aimed to evaluate the effects of vatinoxan (a peripheral α2-adrenoceptor antagonist) on cardiorespiratory and gastrointestinal function in anesthetized healthy horses. Six horses received medetomidine hydrochloride 7µg/kg IV alone (MED) or with vatinoxan hydrochloride 140µg/kg IV (MED+V). Anesthesia was induced with midazolam and ketamine and maintained with isoflurane and medetomidine CRI for 60min. Heart rate, carotid and pulmonary arterial pressures, central venous pressure, cardiac output and arterial and mixed venous blood gases were measured. Selected cardiopulmonary parameters were calculated. Plasma drug concentrations were determined. Fecal output was measured over 24h. For statistical comparisons, repeated measures analysis of covariance and paired t-tests were applied. Heart rate decreased slightly from baseline in the MED group. Arterial blood pressures decreased with both treatments, but significantly more dobutamine was needed to maintain normotension with MED+V (P=0.018). Cardiac index (CI) and oxygen delivery index (DO2I) decreased significantly more with MED, with the largest difference observed at 20min: CI was 39±2 and 73±18 (P=0.009) and DO2I 7.4±1.2 and 15.3±4.8 (P=0.014)mL/min/kg with MED and MED+V, respectively. Fecal output or plasma concentrations of dexmedetomidine did not differ between the treatments. In conclusion, premedication with vatinoxan induced hypotension, thus its use in anesthetized horses warrants further studies. Even though heart rate and arterial blood pressures remained clinically acceptable with MED, cardiac performance and oxygen delivery were lower than with MED+V.


Asunto(s)
Motilidad Gastrointestinal/efectos de los fármacos , Isoflurano/farmacología , Medetomidina/farmacología , Quinolizinas/farmacología , Antagonistas de Receptores Adrenérgicos alfa 2 , Anestesia por Inhalación/veterinaria , Anestésicos por Inhalación/administración & dosificación , Anestésicos por Inhalación/farmacología , Animales , Presión Sanguínea/efectos de los fármacos , Fenómenos Fisiológicos Cardiovasculares/efectos de los fármacos , Estudios Cruzados , Femenino , Frecuencia Cardíaca/efectos de los fármacos , Caballos , Hipnóticos y Sedantes/administración & dosificación , Hipnóticos y Sedantes/farmacología , Isoflurano/administración & dosificación , Masculino , Medetomidina/administración & dosificación , Quinolizinas/sangre , Quinolizinas/farmacocinética
14.
Vet Anaesth Analg ; 46(6): 753-764, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31416697

RESUMEN

OBJECTIVE: To characterize the cardiopulmonary effects of dexmedetomidine, with or without vatinoxan, in isoflurane-anesthetized cats. STUDY DESIGN: Randomized, crossover experimental study. ANIMALS: A group of six adult healthy male neutered cats. METHODS: Cats were instrumented during anesthesia with isoflurane in oxygen. Isoflurane end-tidal concentration was set to 1.25 minimum alveolar concentration (MAC). Dexmedetomidine was administered using a target-controlled infusion system to achieve and maintain 10 target plasma concentrations ranging from 0 to 40 ng mL-1. Furthermore, vatinoxan or an equivalent volume of saline was administered using a target-controlled infusion system to achieve and maintain a target plasma concentration of 4 µg mL-1. Isoflurane concentration was adjusted after each change in dexmedetomidine concentration to maintain a concentration equivalent to 1.25 MAC. Heart rate (HR), arterial blood pressure, central venous pressure (CVP), pulmonary artery pressure (PAP), pulmonary artery occlusion pressure (PAOP), body temperature, cardiac output, arterial and mixed-venous blood gas and pH and drug concentrations were measured. Additional variables were calculated from the measurements. RESULTS: Dexmedetomidine alone resulted in decreased HR, cardiac index, stroke index and oxygen delivery, and increased systolic, mean (MAP) and diastolic arterial pressure, CVP, PAP, PAOP, systemic vascular resistance index, rate-pressure product, left ventricular stroke work index and oxygen extraction ratio. Vatinoxan resulted in severe hypotension at target plasma dexmedetomidine concentrations <10 ng mL-1. Vatinoxan attenuated the cardiovascular effects of dexmedetomidine at the 10 and 20 ng mL-1 targets, but MAP could be maintained above 60 mmHg only when isoflurane concentration was <1.25 MAC. Less improvement in cardiovascular function was seen with vatinoxan at the 40 ng mL-1 target plasma dexmedetomidine concentration. CONCLUSIONS AND CLINICAL RELEVANCE: Vatinoxan, at the plasma concentration maintained in this study, attenuated the cardiovascular effects of dexmedetomidine in isoflurane-anesthetized cats. However, its administration resulted in hypotension, which may limit its clinical usefulness.


Asunto(s)
Presión Sanguínea/efectos de los fármacos , Gatos , Dexmedetomidina/farmacocinética , Isoflurano/farmacología , Quinolizinas/farmacocinética , Antagonistas de Receptores Adrenérgicos alfa 2/administración & dosificación , Antagonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Antagonistas de Receptores Adrenérgicos alfa 2/farmacología , Anestésicos por Inhalación/administración & dosificación , Anestésicos por Inhalación/farmacocinética , Anestésicos por Inhalación/farmacología , Animales , Estudios Cruzados , Dexmedetomidina/administración & dosificación , Dexmedetomidina/sangre , Dexmedetomidina/farmacología , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Frecuencia Cardíaca/efectos de los fármacos , Hipnóticos y Sedantes/administración & dosificación , Hipnóticos y Sedantes/farmacocinética , Hipnóticos y Sedantes/farmacología , Hipotensión/inducido químicamente , Hipotensión/veterinaria , Isoflurano/administración & dosificación , Masculino , Quinolizinas/administración & dosificación , Quinolizinas/farmacología , Resistencia Vascular/efectos de los fármacos
15.
Bioorg Chem ; 90: 103032, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31207450

RESUMEN

A series of new cytisine derivatives with a unique endocyclic scaffold were synthesized and evaluated for their inhibitory effect on collagen α1 (I) (COL1A1) promotor in human LX2 cells, taking cytisine as the lead. Structure-activity relationship (SAR) revealed that introducing a 12N-benzyl substitution might significantly enhance the activity. Compound 5f exhibited a promising inhibitory potency against COL1A1 with an IC50 value of 12.8 µM in human LX2 cells, and an inspiring inhibition activity against COL1A1 on both mRNA and protein levels. It also effectively inhibited the expression of α smooth muscle actin (α-SMA), connective tissue growth factor (CTGA), matrix metalloprotein 2 (MMP-2), and transforming growth factor ß1 (TGFß1), indicating an extensive inhibitory effect against fibrogenetic proteins. In addition, compound 5f displayed reasonable PK and safety profiles. The primary mechanism study indicated that it might repress the hepatic fibrogenesis via PI3K/Akt/Smad signaling pathway. The results provided powerful information for further structure optimization, and compound 5f was selected as a novel anti-liver fibrosis agent for further investigation.


Asunto(s)
Alcaloides/uso terapéutico , Cirrosis Hepática/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Alcaloides/síntesis química , Alcaloides/farmacocinética , Animales , Azocinas/síntesis química , Azocinas/farmacocinética , Azocinas/uso terapéutico , Línea Celular , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I , Humanos , Masculino , Ratones , Estructura Molecular , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Quinolizinas/síntesis química , Quinolizinas/farmacocinética , Quinolizinas/uso terapéutico , Ratas Sprague-Dawley , Proteínas Smad/metabolismo , Relación Estructura-Actividad
16.
Vet Anaesth Analg ; 46(4): 443-451, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30982711

RESUMEN

OBJECTIVE: To characterize the effects of dexmedetomidine, with or without vatinoxan, on the minimum alveolar concentration of isoflurane (MACISO) in cats. STUDY DESIGN: Randomized crossover experimental study. ANIMALS: A group of six adult healthy male neutered cats. METHODS: Cats were anesthetized with isoflurane in oxygen and instrumented. Dexmedetomidine was administered using a target-controlled infusion system to achieve 10 target plasma concentrations ranging from 0 to 40 ng mL-1. Additionally, vatinoxan or an equivalent volume of saline was administered using a target-controlled infusion system to achieve a target plasma concentration of 4 µg mL-1. Pulse rate (PR), respiratory rate, systolic arterial pressure (SAP), hemoglobin oxygen saturation, body temperature, end-tidal partial pressure of carbon dioxide and drug concentrations were measured. MACISO was determined at each target plasma dexmedetomidine concentration using the bracketing method and the tail clamp technique. Pharmacodynamic models were fitted to the plasma dexmedetomidine concentration-MACISO. Pharmacodynamic parameters were tested for equivalence, and if rejected, for difference. RESULTS: Dexmedetomidine alone decreased MACISO in a plasma concentration-dependent manner. Maximum reduction was 77 ± 4%; the dexmedetomidine concentration producing 50% of the maximum decrease (IC50) was 0.77 ng mL-1. Vatinoxan increased MACISO in the absence of dexmedetomidine, decreased the potency of dexmedetomidine for its MACISO-reducing effect (IC50 = 12 ng mL-1) and lessened the maximum MACISO reduction (60 ± 14%). PR decreased less and SAP increased less when dexmedetomidine was administered with vatinoxan compared with saline. CONCLUSION AND CLINICAL RELEVANCE: Vatinoxan altered the effect of dexmedetomidine on MACISO. A high plasma dexmedetomidine concentration in the presence of vatinoxan resulted in a large decrease in MACISO, with attenuation of dexmedetomidine-induced cardiovascular effects. The vatinoxan-dexmedetomidine combination may provide clinical benefits in isoflurane-anesthetized cats.


Asunto(s)
Gatos , Dexmedetomidina , Isoflurano , Alveolos Pulmonares , Quinolizinas , Animales , Masculino , Anestesia por Inhalación/veterinaria , Anestésicos por Inhalación/administración & dosificación , Anestésicos por Inhalación/farmacocinética , Anestésicos por Inhalación/farmacología , Estudios Cruzados , Dexmedetomidina/administración & dosificación , Dexmedetomidina/farmacocinética , Dexmedetomidina/farmacología , Interacciones Farmacológicas , Quimioterapia Combinada , Hipnóticos y Sedantes/administración & dosificación , Hipnóticos y Sedantes/farmacocinética , Hipnóticos y Sedantes/farmacología , Isoflurano/farmacología , Quinolizinas/administración & dosificación , Quinolizinas/farmacocinética , Quinolizinas/farmacología
17.
Xenobiotica ; 49(11): 1332-1337, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30526213

RESUMEN

1. Cytisine, a partial agonist for the α4ß2-nAChR, is used as a smoking cessation medication. Cytisine's current dosing is complex and involves taking 1.5 mg several times a day. The aim of this study was to explore the effect of dose on the pharmacokinetics and safety of cytisine after a single dose in healthy adult smokers. 2. Participants were assigned to one of three groups (n = 6 in each group) to receive a single oral dose of 1.5, 3 or 4.5 mg of cytisine. Blood samples were collected up to 24 h post dose. Pulse, blood pressure and respiratory rate were measured. Adverse effects were recorded. 3. Cytisine reached peak plasma concentration 1-2 h post dose in all participants irrespective of dose, with no dose-dependent changes in the elimination phase. Mean (SD) cytisine exposure (AUC0-24h) were 81.9 (15.8), 181.9 (40.8) and 254.5 (48.1) ng.h/mL following 1.5, 3 and 4.5 mg, respectively. 4. Cytisine appears to have predictable pharmacokinetics following a single dose of up to 4.5 mg and may be safe given as a single 4.5 mg dose, which is threefold greater than the recommended dose taken at one time. This study is registered in ClinicalTrials.gov (ID:NCT02585024).


Asunto(s)
Alcaloides/farmacocinética , Fumadores , Administración Oral , Adolescente , Adulto , Alcaloides/administración & dosificación , Alcaloides/efectos adversos , Alcaloides/sangre , Área Bajo la Curva , Azocinas/administración & dosificación , Azocinas/efectos adversos , Azocinas/sangre , Azocinas/farmacocinética , Presión Sanguínea/efectos de los fármacos , Femenino , Semivida , Cefalea/inducido químicamente , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Proyectos Piloto , Quinolizinas/administración & dosificación , Quinolizinas/efectos adversos , Quinolizinas/sangre , Quinolizinas/farmacocinética , Cese del Hábito de Fumar/métodos , Adulto Joven
18.
Med Chem ; 14(8): 764-772, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29473520

RESUMEN

BACKGROUND: Influenza is still a serious threat to human health with significant morbidity and mortality, so it is desirable to develop novel anti-flu drug agents with novel structures. OBJECTIVE: The main purpose of this research was to explore broad-spectrum anti-flu agents and provide antiviral stockpiles in response to potential future influenza pandemics. METHODS: Fifteen novel 12N-substituted tricyclic matrinic derivatives were synthesized and evaluated for their anti-influenza activities against H1N1 subtype taking 12N-p-cyanobenzenesulfonyl matrinane (1) as the lead. All prepared compounds were characterized by 1H NMR, 13C NMR and ESI-HRMS. The pharmacokinetics (PK) profile of the key compound was also examined in this study. RESULT: The structure-activity relationship study indicated that suitable benzyl groups on 12N atom might be beneficial for the activity. Among them, 12N-p-carboxybenzyl matrinic butane (17g) exhibited the most promising activity with an IC50 value of 16.2 µM and a selective index (SI) value of over 33.4. In addition, compound 17g displayed a good in vivo pharmacokinetic profile with an area under the curve (AUC0-∞) value of 9.89 µM·h. CONCLUSION: We consider tricyclic matrinic butane derivatives to be a new class of anti-influenza agents and this study provided useful information on further optimization.


Asunto(s)
Alcaloides/farmacología , Antivirales/farmacología , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Quinolizinas/farmacología , Alcaloides/administración & dosificación , Alcaloides/síntesis química , Alcaloides/farmacocinética , Animales , Antivirales/administración & dosificación , Antivirales/síntesis química , Antivirales/farmacocinética , Perros , Células de Riñón Canino Madin Darby , Masculino , Quinolizinas/administración & dosificación , Quinolizinas/síntesis química , Quinolizinas/farmacocinética , Ratas Sprague-Dawley , Relación Estructura-Actividad
19.
J Vet Pharmacol Ther ; 41(3): 447-456, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29352473

RESUMEN

The effect of MK-467, a peripheral α2 -adrenoceptor antagonist, on plasma drug concentrations, sedation and cardiopulmonary changes induced by intramuscular (IM) medetomidine was investigated in eight sheep. Additionally, the interactions with atipamezole (ATI) used for reversal were also evaluated. Each animal was treated four times in a randomized prospective crossover design with 2-week washout periods. Medetomidine (MED) 30 µg/kg alone or combined in the same syringe with MK-467 300 µg/kg (MMK) was injected intramuscular, followed by ATI 150 µg/kg (MED + ATI and MMK + ATI) or saline intramuscular 30 min later. Plasma was analysed for drug concentrations, and sedation was subjectively assessed with a visual analogue scale. Systemic haemodynamics and blood gases were measured before treatments and at intervals thereafter. With MK-467, medetomidine plasma concentrations were threefold higher prior to ATI, which was associated with more profound sedation and shorter onset. No significant differences were observed in early cardiopulmonary changes between treatments. Atipamezole reversed the medetomidine-related cardiopulmonary changes after both treatments. Sedation scores decreased more rapidly when MK-467 was included. In this study, MK-467 appeared to have a pronounced effect on the plasma concentration and central effects of medetomidine, with minor cardiopulmonary improvement.


Asunto(s)
Antagonistas de Receptores Adrenérgicos alfa 2/farmacología , Imidazoles/farmacocinética , Medetomidina/farmacocinética , Quinolizinas/farmacología , Ovinos/sangre , Antagonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Animales , Presión Sanguínea , Temperatura Corporal , Sedación Consciente/veterinaria , Estudios Cruzados , Interacciones Farmacológicas , Femenino , Hemoglobinas , Hipnóticos y Sedantes/farmacocinética , Hipnóticos y Sedantes/farmacología , Imidazoles/sangre , Imidazoles/farmacología , Inyecciones Intramusculares , Medetomidina/sangre , Medetomidina/farmacología , Oxígeno/sangre , Estudios Prospectivos , Quinolizinas/farmacocinética , Respiración , Escala Visual Analógica
20.
J Feline Med Surg ; 20(8): 721-727, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-28905667

RESUMEN

Objectives The objectives were to evaluate the pharmacokinetics (PK) of subcutaneous (SC) and intravenous (IV) dolasetron and the pharmacodynamics (PD) of SC dolasetron in healthy cats. Methods Five cats with unremarkable complete blood count, serum biochemistry and urinalyses were utilized. In the PK study, cats received 0.8 mg/kg SC and IV dolasetron in a crossover format. Serum samples were obtained via a jugular catheter at 0, 0.25, 0.5, 1, 2, 4, 8, 12, 24, 36 and 48 h after the administration of dolasetron. Dolasetron and the active metabolite hydrodolasetron were measured using liquid chromatography/tandem mass spectrometry. Non-compartmental PK analysis was performed. In the PD study, SC dolasetron (0.8 mg/kg and 1.0 mg/kg) and saline were administered 30 mins prior to administration of 0.44 mg/kg intramuscular xylazine in a randomized three-way crossover. Number of emetic events, lip licks, time to onset of emesis and visual nausea score were scored by a blinded observer. Results In the PK study, dolasetron was quickly metabolized to the active metabolite hydrodolasetron, limiting assessment of dolasetron PK parameters. Median (range) PK parameters for IV hydrodolasetron were as follows: maximum serum concentration (Cmax) 116 ng/ml (69-316 ng/ml), time to maximum concentration (Tmax) 0.5 h (0.3-0.5 h), half-life 3.3 h (2.9-7.2 h) and area under the curve until the last measurable concentration (AUClast) 323 h/ng/ml (138-454 h/ng/ml). Median (range) PK parameters for SC hydrodolasetron were as follows: Cmax 67.9 ng/ml (60.4-117 ng/ml), Tmax 0.5 h (0.5-1.0 h), half-life 3.8 h (2.9-5.3 h) and AUClast 437 h/ng/ml (221.5-621.8 h/ng/ml). There was no significant difference in exposure to hydrodolasetron between the routes of administration. With regard to PD, when dolasetron was administered prior to xylazine, there was no significant difference in the mean number of emetic events, lip licks, time to onset of emesis or visual nausea score when compared with saline. Conclusions and relevance Administration of 0.8 mg/kg dolasetron does not maintain serum concentrations of active metabolite for 24 h. Administration of dolasetron at 0.8 mg/kg and 1 mg/kg did not prevent xylazine-induced vomiting. Additional feline dose studies are needed to determine if a higher dose is efficacious.


Asunto(s)
Gatos/metabolismo , Indoles/administración & dosificación , Indoles/farmacocinética , Quinolizinas/administración & dosificación , Quinolizinas/farmacocinética , Administración Intravenosa , Animales , Cromatografía Liquida , Estudios Cruzados , Método Doble Ciego , Indoles/efectos adversos , Indoles/sangre , Infusiones Subcutáneas , Inyecciones Intramusculares , Quinolizinas/efectos adversos , Quinolizinas/sangre , Distribución Aleatoria , Espectrometría de Masas en Tándem , Xilazina/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...