Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros












Intervalo de ano de publicação
1.
J Vet Pharmacol Ther ; 47(4): 288-293, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38407510

RESUMO

In the United States, a generic Type A medicated article product can gain the FDA approval by demonstrating bioequivalence (BE) to the pioneer product by successfully conducting a blood level, pharmacodynamic, or clinical BE study. A biowaiver can be granted based on several criteria, assuming the dissolution of the test and reference products represents the only factor influencing the relative bioavailability of both products. Monensin is practically insoluble in H2O per the USP definition. Previously published data from a comparison study of monensin dissolution profiles from the pioneer product and four generic products using biorelevant media showed that generic monensin products demonstrated different dissolution profiles to the pioneer product in these USP biorelevant rumen media. This follow-up study compared the solubility profiles in simulated intestinal fluid (cFaSSIF, pH 7.5) for the pioneer product and four generic products. The generic monensin products demonstrated different in vitro dissolution profiles to the pioneer product in biorelevant media. The differences demonstrated in solubility and dissolution profiles are of concern regarding the potential efficacy of generic monensin in cattle. There are also additional concerns for the potential development of Eimeria resistance in cattle receiving a sub-therapeutic dose of monensin from a less soluble generic product.


Assuntos
Medicamentos Genéricos , Monensin , Solubilidade , Monensin/farmacocinética , Monensin/química , Monensin/administração & dosagem , Medicamentos Genéricos/farmacocinética , Medicamentos Genéricos/química , Equivalência Terapêutica , Animais
2.
Molecules ; 24(24)2019 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-31817501

RESUMO

Lincomycin, monensin, and roxarsone are commonly used veterinary drugs. This study investigated their behaviours in different soils and their toxic effects on environmental organisms. Sorption and mobility analyses were performed to detect the migration capacity of drugs in soils. Toxic effects were evaluated by inhibition or acute toxicity tests on six organism species: algae, plants, daphnia, fish, earthworms and quails. The log Kd values (Freundlich model) of drugs were: lincomycin in laterite soil was 1.82; monensin in laterite soil was 2.76; and roxarsone in black soil was 1.29. The Rf value of lincomycin, roxarsone, monensin were 0.4995, 0.4493 and 0.8348 in laterite soil, and 0.5258, 0.5835 and 0.8033 in black soil, respectively. The EC50 for Scenedesmus obliquus, Arabidopsis thaliana, Daphnia magna and LC50/LD50 for Eisenia fetida, Danio rerio, and Coturnix coturnix were: 13.15 mg/L,32.18 mg/kg dry soil,292.6 mg/L,452.7 mg/L,5.74 g/kg dry soil and 103.9 mg/kg (roxarsone); 1.085 mg/L, 25 mg/kg dry soil, 21.1 mg/L, 4.76 mg/L, 0.346 g/kg dry soil and 672.8 mg/kg (monensin); 0.813 mg/L, 35.40 mg/kg dry soil, >400 mg/L, >2800 mg/L, >15 g/kg dry soil, >2000 mg/kg (lincomycin). These results showed that the environmental effects of veterinary drug residues should not be neglected, due to their mobility in environmental media and potential toxic effects on environmental organisms.


Assuntos
Arabidopsis/metabolismo , Coturnix/metabolismo , Daphnia/metabolismo , Lincomicina , Monensin , Oligoquetos/metabolismo , Roxarsona , Scenedesmus/metabolismo , Peixe-Zebra/metabolismo , Animais , Lincomicina/efeitos adversos , Lincomicina/farmacocinética , Monensin/administração & dosagem , Monensin/farmacocinética , Roxarsona/efeitos adversos , Roxarsona/farmacocinética
3.
J Vet Pharmacol Ther ; 40(4): 370-382, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27943298

RESUMO

We developed a flow-limited physiologically based pharmacokinetic model for residues of monensin in chickens and evaluated its predictive ability by comparing it with an external data set describing concentration decays after the end of treatment. One advantage of this model is that the values for most parameters (34 of 38) were taken directly from the literature or from field data (for growth and feed intake). Our model included growth (changes in body weight) to describe exposure throughout the life of the chicken. We carried out a local sensitivity analysis to evaluate the relative importance of model parameters on model outputs and revealed the predominant influence of 19 parameters (including three estimated ones): seven pharmacokinetic parameters, five physiological parameters and seven animal performance parameters. Our model estimated the relative bioavailability of monensin as feed additive at 3.9%, which is even lower than the absolute bioavailability in solution (29.91%). Our model can be used for extrapolations of farming conditions, such as monensin supplementation or building lighting programme (which may have a significant impact for short half-life molecules such as monensin). This validated PBPK model may also be useful for interspecies extrapolations or withdrawal period calculations for modified dosage regimens.


Assuntos
Galinhas/metabolismo , Monensin/farmacocinética , Ração Animal , Animais , Disponibilidade Biológica , Peso Corporal , Meia-Vida
4.
J Vet Pharmacol Ther ; 39(4): 363-72, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26763112

RESUMO

Used in both beef cattle and dairy cows, monensin can provide many health benefits but can, when unintended overexposures occur, result in adverse effects. Information on serum and tissue concentrations following overexposure and/or overt toxicosis which may aid in diagnostics and clinical outcome is lacking. The aim of this study was to determine concentrations of monensin in biological specimens following oral exposure for 10 days to an approved dose (1 mg/kg) and a higher dose (5 mg/kg) of monensin given daily on a body weight basis to 10 dairy cows. No deaths were reported; cows receiving 5 mg/kg showed early signs of toxicosis including depression, decreased feed intake, and diarrhea after 4 days of exposure. Histopathological findings were minimal in most cows. Pharmacokinetic modeling of the detected serum concentrations for the 1 and 5 mg/kg dose groups determined the Cmax , Tmax, and t1/2λ to be 0.87 and 1.68 ng/mL, 2.0 and 1.0 h, and 1.76 and 2.32 days, respectively. Mixed regression models showed that the dose level and days since last dose were significantly associated with monensin concentrations in all four tissues, and with cardiac troponin levels. The high dose resulted in a significant elevation of monensin in tissues at approximately 4.7 times compared to the monensin concentrations in the tissues of animals from the low-dose group. The cTnI concentrations in the high-dose group were 2.1 times that of cTnI in the low-dose group. Thus, the ability to diagnose monensin overexposure and/or toxicosis will improve from knowledge of biological monensin concentrations from this study.


Assuntos
Leite/química , Monensin/análise , Administração Oral , Animais , Bovinos , Feminino , Rim/química , Fígado/química , Monensin/efeitos adversos , Monensin/sangue , Monensin/farmacocinética , Músculo Esquelético/química , Miocárdio/química , Troponina C/sangue
5.
Antimicrob Agents Chemother ; 60(3): 1304-18, 2015 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-26666937

RESUMO

The global emergence of drug resistance in malaria is impeding the therapeutic efficacy of existing antimalarial drugs. Therefore, there is a critical need to develop an efficient drug delivery system to circumvent drug resistance. The anticoccidial drug monensin, a carboxylic ionophore, has been shown to have antimalarial properties. Here, we developed a liposome-based drug delivery of monensin and evaluated its antimalarial activity in lipid formulations of soya phosphatidylcholine (SPC) cholesterol (Chol) containing either stearylamine (SA) or phosphatidic acid (PA) and different densities of distearoyl phosphatidylethanolamine-methoxy-polyethylene glycol 2000 (DSPE-mPEG-2000). These formulations were found to be more effective than a comparable dose of free monensin in Plasmodium falciparum (3D7) cultures and established mice models of Plasmodium berghei strains NK65 and ANKA. Parasite killing was determined by a radiolabeled [(3)H]hypoxanthine incorporation assay (in vitro) and microscopic counting of Giemsa-stained infected erythrocytes (in vivo). The enhancement of antimalarial activity was dependent on the liposomal lipid composition and preferential uptake by infected red blood cells (RBCs). The antiplasmodial activity of monensin in SA liposome (50% inhibitory concentration [IC50], 0.74 nM) and SPC:Chol-liposome with 5 mol% DSPE-mPEG 2000 (IC50, 0.39 nM) was superior to that of free monensin (IC50, 3.17 nM), without causing hemolysis of erythrocytes. Liposomes exhibited a spherical shape, with sizes ranging from 90 to 120 nm, as measured by dynamic light scattering and high-resolution electron microscopy. Monensin in long-circulating liposomes of stearylamine with 5 mol% DSPE-mPEG 2000 in combination with free artemisinin resulted in enhanced killing of parasites, prevented parasite recrudescence, and improved survival. This is the first report to demonstrate that monensin in PEGylated stearylamine (SA) liposome has therapeutic potential against malaria infections.


Assuntos
Antimaláricos/farmacologia , Artemisininas/farmacologia , Lipossomos/administração & dosagem , Malária/tratamento farmacológico , Monensin/farmacologia , Aminas/administração & dosagem , Aminas/química , Animais , Antimaláricos/administração & dosagem , Sangue/efeitos dos fármacos , Sangue/parasitologia , Sistemas de Liberação de Medicamentos/métodos , Quimioterapia Combinada , Feminino , Lipossomos/química , Lipossomos/farmacologia , Malária/parasitologia , Camundongos , Monensin/farmacocinética , Plasmodium berghei/patogenicidade , Plasmodium falciparum/efeitos dos fármacos , Distribuição Tecidual
6.
Biomed Res Int ; 2014: 152102, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24987668

RESUMO

Monensin A is a commercially important natural product isolated from Streptomyces cinnamonensins that is primarily employed to treat coccidiosis. Monensin A selectively complexes and transports sodium cations across lipid membranes and displays a variety of biological properties. In this study, we evaluated the Jacobsen catalyst as a cytochrome P450 biomimetic model to investigate the oxidation of monensin A. Mass spectrometry analysis of the products from these model systems revealed the formation of two products: 3-O-demethyl monensin A and 12-hydroxy monensin A, which are the same ones found in in vivo models. Monensin A and products obtained in biomimetic model were tested in a mitochondrial toxicity model assessment and an antimicrobial bioassay against Staphylococcus aureus, S. aureus methicillin-resistant, Staphylococcus epidermidis, Pseudomonas aeruginosa, and Escherichia coli. Our results demonstrated the toxicological effects of monensin A in isolated rat liver mitochondria but not its products, showing that the metabolism of monensin A is a detoxification metabolism. In addition, the antimicrobial bioassay showed that monensin A and its products possessed activity against Gram-positive microorganisms but not for Gram-negative microorganisms. The results revealed the potential of application of this biomimetic chemical model in the synthesis of drug metabolites, providing metabolites for biological tests and other purposes.


Assuntos
Antifúngicos , Sistema Enzimático do Citocromo P-450/metabolismo , Mitocôndrias Hepáticas/metabolismo , Modelos Biológicos , Monensin , Animais , Antifúngicos/farmacocinética , Antifúngicos/farmacologia , Bactérias/crescimento & desenvolvimento , Monensin/farmacocinética , Monensin/farmacologia , Oxirredução/efeitos dos fármacos , Ratos
7.
Environ Sci Technol ; 48(5): 2724-31, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24494860

RESUMO

Ionophore antibiotics (IPAs) are polyether compounds used in broiler feed to promote growth and control coccidiosis. Most of the ingested IPAs are excreted into broiler litter (BL), a mixture of excreta and bedding material. BL is considered a major source of IPAs released into the environment as BL is commonly used to fertilize agricultural fields. This study investigated IPA biodegradation in BL and soil microcosms, as a process affecting the fate of IPAs in the environment. The study focused on the most widely used IPAs, monensin (MON), salinomycin (SAL), and narasin (NAR). MON was stable in BL microcosms at 24-72% water content (water/wet litter, w/w) and 35-60 °C, whereas SAL and NAR degraded under certain conditions. Factor analysis was conducted to delineate the interaction of water and temperature on SAL and NAR degradation in the BL. A major transformation product of SAL and NAR was identified. Abiotic reaction(s) were primarily responsible for the degradation of MON and SAL in nonfertilized soil microcosms, whereas biodegradation contributed significantly in BL-fertilized soil microcosms. SAL biotransformation in soil microcosms yielded the same product as in the BL microcosms. A new primary biotransformation product of MON was identified in soil microcosms. A field study showed that MON and SAL were stable during BL stacking, whereas MON degraded after BL was applied to grassland. The biotransformation product of MON was also detected in the top soil layer where BL was applied.


Assuntos
Antibacterianos/farmacocinética , Poluentes do Solo/farmacocinética , Animais , Biodegradação Ambiental , Galinhas , Fertilizantes , Ionóforos/farmacocinética , Esterco , Monensin/farmacocinética , Piranos/farmacocinética , Microbiologia do Solo , Temperatura , Drogas Veterinárias/farmacocinética , Água
8.
J Vet Pharmacol Ther ; 36(5): 494-501, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23067134

RESUMO

Drug-drug interactions (DDIs) may adversely affect the prevention and cure of diseases. The effects of three polyether ionophore antibiotics, salinomycin (SAL), monensin (MON), and maduramycin (MAD) on the pharmacokinetics of florfenicol (FFC) were investigated in broilers. The chickens were fed rations with or without SAL (60 mg/kg feeds), MON (120 mg/kg feeds), or MAD (5 mg/kg feeds) for 14 consecutive days. FFC was given to the chickens either intravenously (i.v.) or orally (p.o.) at a single dose of 30 mg/kg body weight. Blood samples were taken from each chicken at 0-24 h postadministration of FFC. The plasma concentration of FFC was detected by high-performance liquid chromatography. The plasma concentration of FFC decreased with i.v. or p.o. co-administration of SAL, MON, or MAD in broilers, implying occurrence of DDIs during the co-administration of FFC with these ionophores. Our findings suggest that more attention should be given to the use of FFC to treat bacterial infections in chickens supplemented with polyether ionophore antibiotics.


Assuntos
Antibacterianos/farmacocinética , Coccidiostáticos/farmacocinética , Ionóforos/farmacocinética , Lactonas/farmacocinética , Monensin/farmacocinética , Piranos/farmacocinética , Tianfenicol/análogos & derivados , Administração Oral , Animais , Antibacterianos/administração & dosagem , Antibacterianos/sangue , Galinhas/sangue , Galinhas/metabolismo , Cromatografia Líquida de Alta Pressão/veterinária , Coccidiostáticos/administração & dosagem , Interações Medicamentosas , Quimioterapia Combinada , Injeções Intravenosas/veterinária , Ionóforos/administração & dosagem , Lactonas/administração & dosagem , Masculino , Monensin/administração & dosagem , Piranos/administração & dosagem , Tianfenicol/administração & dosagem , Tianfenicol/sangue , Tianfenicol/farmacocinética
9.
Artigo em Inglês | MEDLINE | ID: mdl-22963507

RESUMO

Recent legislation has addressed the unavoidable carry-over of coccidiostats and histomonostats in feed, which may lead to the presence of residues of these compounds in eggs. In this study, laying hens received cross-contaminated feed at a ratio of 2.5%, 5% and 10% of the therapeutic dose of monensin and lasalocid for broilers. The eggs were collected during the treatment and depletion period and were analysed using liquid chromatography-tandem mass spectrometry. The different egg matrices were separated and analysed during the plateau phase. High lasalocid concentrations, which exceeded the maximum residue level, and low monensin concentrations were found in whole egg. Plateau levels were reached at days 7-9 for lasalocid and at days 3-5 for monensin. For lasalocid, the highest concentrations were measured in egg yolk; residue concentrations in egg white were very low.


Assuntos
Ração Animal/análise , Galinhas/fisiologia , Coccidiostáticos/farmacocinética , Resíduos de Drogas/análise , Ovos/análise , Contaminação de Alimentos , Oviposição , Animais , Animais Endogâmicos , Bélgica , Coccidiostáticos/administração & dosagem , Coccidiostáticos/análise , Relação Dose-Resposta a Droga , Clara de Ovo/química , Gema de Ovo/química , Feminino , Lasalocida/administração & dosagem , Lasalocida/análise , Lasalocida/farmacocinética , Monensin/administração & dosagem , Monensin/análise , Monensin/farmacocinética , Distribuição Aleatória , Distribuição Tecidual
10.
J Biomed Nanotechnol ; 8(1): 172-81, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22515105

RESUMO

PLGA nanoparticles loaded with monensin (carboxylic ionophore) were prepared by emulsion solvent evaporation method using PLGA of molecular weight (Mw.) 19000 and 110000 Da. The nanoparticles were characterized by applying dynamic light scattering (DLS), atomic force microscopy (AFM), differential scanning calorimetry (DSC) and Fourier transformed infrared spectroscopy (FTIR). Negatively charged and spherical smooth surfaced nanoparticles of size range between 147-167 nm were obtained. The nanoparticles of monensin-PLGA showed no chemical interaction between monensin and the polymer molecules. The release kinetics in vitro studies exhibited biphasic release profile characterized by an initial fast release followed by a slower release. The antimalarial efficacy of monensin-PLGA nanoparticles was also examined. Monensin loaded in nanoparticles was 10-fold more effective in inhibiting the growth of P. falciparum in vitro as compared to free monensin. The antimalarial efficacy of monensin-PLGA nanoparticles was significantly dependent on the Mw. of the polymer.


Assuntos
Antimaláricos/administração & dosagem , Ácido Láctico/química , Monensin/administração & dosagem , Nanopartículas/química , Plasmodium falciparum/efeitos dos fármacos , Ácido Poliglicólico/química , Antimaláricos/química , Antimaláricos/farmacocinética , Varredura Diferencial de Calorimetria , Células Cultivadas , Emulsões/administração & dosagem , Emulsões/química , Emulsões/farmacocinética , Eritrócitos/efeitos dos fármacos , Hemólise/efeitos dos fármacos , Humanos , Ácido Láctico/administração & dosagem , Ácido Láctico/farmacocinética , Peso Molecular , Monensin/química , Monensin/farmacocinética , Nanopartículas/administração & dosagem , Tamanho da Partícula , Ácido Poliglicólico/administração & dosagem , Ácido Poliglicólico/farmacocinética , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Espectroscopia de Infravermelho com Transformada de Fourier
11.
Small ; 8(6): 943-8, 2012 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-22315201

RESUMO

The concept of a long-term sensor for ion changes in the lysosome is presented. The sensor is made by layer-by-layer assembly of oppositely charged polyelectrolytes around ion-sensitive fluorophores, in this case for protons. The sensor is spontaneously incorporated by cells and resides over days in the lysosome. Intracellular changes of the concentration of protons upon cellular stimulation with pH-active agents are monitored by read-out of the sensor fluorescence at real time. With help of this sensor concept it is demonstrated that the different agents used (Monensin, Chloroquine, Bafilomycin A1, Amiloride) possessed different kinetics and mechanisms of action in affecting the intracellular pH values.


Assuntos
Concentração de Íons de Hidrogênio , Lisossomos/metabolismo , Amilorida/farmacocinética , Cloroquina/farmacocinética , Fluorescência , Macrolídeos/farmacocinética , Monensin/farmacocinética
12.
J Vet Pharmacol Ther ; 35(1): 73-81, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21615753

RESUMO

The current study describes the pharmacokinetic parameters of two carboxylic polyether ionophores: monensin in turkeys and salinomycin in chickens. These data can be used to understand and predict the occurrence of undesirable residues of coccidiostats in edible tissues of these animal species. Special attention is paid to the distribution of residues between the different edible tissues during and at the end of the treatment period. For the bioavailability studies, monensin was administered to turkeys intravenously, in the left wing vein, at a dose of 0.4 mg /kg and orally at a dose of 20 mg /kg. Salinomycin was administered to chickens intravenously, in the left wing vein, at a dose of 0.25 mg /kg and orally at a dose of 2.5 mg /kg. Residue studies were carried out with supplemented feed at the rate of 100 mg /kg of feed for monensin in turkeys and 70 mg /kg for salinomycin in chickens, respectively. Coccidiostats had a low bioavailability in poultry (around 30% for monensin in chickens, around 1% for monensin in turkeys and around 15% for salinomycin in chickens). Monensin in chickens had a longer terminal half-life (between 3.07 and 5.55 h) than both monensin in turkeys (between 1.36 and 1.55 h) and salinomycin in chickens (between 1.33 and 1.79 h). The tissue /plasma partition coefficients showed a higher affinity of both monensin and salinomycin for fat, followed by liver and muscle tissue. The depletion data showed a fairly rapid elimination of coccidiostats in all the tissues after cessation of treatment. According to the results of depletion studies, a withdrawal period of 1 day seems sufficient to avoid undesirable exposure of consumers.


Assuntos
Galinhas/metabolismo , Coccidiostáticos/farmacocinética , Monensin/farmacocinética , Piranos/farmacocinética , Perus/metabolismo , Tecido Adiposo/metabolismo , Administração Oral , Animais , Área Sob a Curva , Disponibilidade Biológica , Galinhas/sangue , Coccidiostáticos/sangue , Feminino , Meia-Vida , Fígado/metabolismo , Masculino , Monensin/administração & dosagem , Monensin/sangue , Músculo Esquelético/metabolismo , Piranos/administração & dosagem , Piranos/sangue , Distribuição Tecidual , Perus/sangue
13.
J Vet Pharmacol Ther ; 32(5): 451-6, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19754911

RESUMO

The pharmacokinetics of monensin including apparent volume of distribution, total body clearance, systemic bioavailability, partition coefficients and tissue residues were determined in chickens. The drug was given by intravenous injection in the left wing vein at the dose of 0.46 mg/kg and by intracrop administration at the dose of 4 mg/kg according to a destructive sampling. The pharmacokinetic variables were compared after noncompartmental, naïve averaged, naïve pooled and nonlinear mixed-effects modelling analyses. Partition coefficients and tissue residues were determined after a treatment with feed additives (125 mg/kg of feed) of 33 days. The clearance, volume of distribution and bioavailabilty were approximately 2.2 L/h/kg, approximately 9 L/kg and approximately 30% respectively except with nonlinear mixed effects models that presented values of 1.77 L/h/kg, 14.05 L/kg and 11.36% respectively. Tissue/plasma partition coefficients were estimated to 0.83, 3.39 and 0.51 for liver, fat and thigh muscle respectively. Monensin residues after treatment were not detected 6 h after withdrawal except for fat where monensin was still quantifiable 12 h after. Pharmacokinetic variables seem to be inaccurate when assessed with non linear mixed-effects modelling associated to destructive sampling in chickens. Values varied slightly with noncompartmental, naïve averaged and naïve pooled analyses. The absorption, elimination and partition parameters will be incorporated into a physiologically based pharmacokinetic model and the depletion study will be used to test the ability of this model to describe monensin residues in edible tissues under different dosage regimens.


Assuntos
Coccidiostáticos/farmacocinética , Monensin/farmacocinética , Administração Oral , Animais , Disponibilidade Biológica , Galinhas/metabolismo , Cromatografia Líquida de Alta Pressão , Coccidiostáticos/sangue , Feminino , Injeções Intravenosas/veterinária , Masculino , Taxa de Depuração Metabólica , Monensin/sangue , Distribuição Tecidual
15.
Arq. bras. med. vet. zootec ; 59(6): 1516-1522, dez. 2007. ilus, tab
Artigo em Português | LILACS | ID: lil-476126

RESUMO

Dois estudos foram realizados com vacas lactantes utilizadas como unidade experimental e doadoras de líquido ruminal, sendo as populações de bactérias utilizadas para avaliar a ação de níveis crescentes de lasalocida e monensina na resistência à perda de potássio intracelular, e para produção de gases in vitro. A perda de potássio (Kmax) da lasalocida foi menor para a população de bactérias obtidas do líquido de rúmen de vacas submetidas a dietas com monensina, óleo de soja e monensina mais óleo de soja (19,4 a 25,4 por cento) quando comparada com a perda de potássio em vacas submetidas a dietas sem ionóforo e óleo de soja (30,1 por cento). O mesmo ocorreu para a perda de potássio da monensina, em que o menor valor foi de 6,5 por cento para monensina mais óleo e o maior, de 29,5 por cento, para o controle. Necessita-se de alta concentração de monensina (Kd= 2,3µM), porém baixa de lasalocida (Kd= 0,2µM) para causar a metade da perda máxima de potássio intracelular da população de bactérias do rúmen de vacas submetidas a dietas com monensina. As populações de bactérias de vacas submetidas às dietas com monensina foram sensíveis à lasalocida. As amostras incubadas com própolis produziram menor volume de gases (12,9ml/100g de MS)


Two studies were carried out with lactating cows as experimental units and ruminal fluid donors. The ruminal bacteria population was used to evaluate the action of increasing levels of lasalocid and monensin on resistance of intracellular potassium depletion and in vitro gas production intracellular depletion potassium (Kmax) of lasalocid was lower to ruminal bacteria population obtained from rumen of cows fed diets with monensin, soybean oil and monensin plus soybean oil (19.4 to 25.4 percent) when compared to cows fed with control diet (30.1 percent). The same occurred for intracellular depletion potassium (Kmax) of monensin, in which the lowest value was 6.5 percent to monensin plus soybean oil and the greatest was 29.5 percent to control. High monensin concentration (Kd= 2.3µM) and low lasalocid concentration (Kd= 0.2µM) were necessary to cause half of maximum potassium depletion in ruminal bacteria population from cows fed diet with monensin. The ruminal bacteria population from cows feed diet with monensin were sensible to lasalocid. In vitro gas production showed the lowest volume when diets were incubated with propolis (12,9ml/100g of DM)


Assuntos
Animais , Feminino , Bovinos , Canais de Potássio/farmacocinética , Flatulência/induzido quimicamente , Flatulência/veterinária , Ionóforos/administração & dosagem , Lasalocida/administração & dosagem , Lasalocida/farmacocinética , Monensin/administração & dosagem , Monensin/farmacocinética
16.
J Chromatogr B Analyt Technol Biomed Life Sci ; 850(1-2): 15-23, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17141581

RESUMO

A liquid chromatography-tandem mass spectrometric (LC-MS/MS) method was developed and validated for use in pharmacokinetic studies in order to determine the concentrations of monensin in plasma and edible tissues of chicken. Two sample preparations were performed, one for determining monensin concentrations in plasma using acetonitrile for protein precipitation and another one for determining monensin concentrations in muscle, liver, and fat using methanol-water followed by a clean up on a solid-phase extraction cartridge. Sample extracts were injected into the LC-MS/MS system, and a gradient elution was performed on a C18 column. Narasin was used as internal standard. The LC-MS/MS method was validated using an approach based on accuracy profiles, and applicability of the method was demonstrated for the determination of monensin in chicken plasma, muscle, liver, and fat in a pharmacokinetic study.


Assuntos
Antiprotozoários/sangue , Cromatografia Líquida/métodos , Monensin/sangue , Espectrometria de Massas por Ionização por Electrospray/métodos , Animais , Antiprotozoários/farmacocinética , Calibragem , Galinhas , Monensin/farmacocinética , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Espectrometria de Massas em Tandem/métodos , Distribuição Tecidual
17.
Can J Vet Res ; 69(3): 180-5, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16187547

RESUMO

Milk residues and performance were evaluated in lactating cows that were fed up to 10 times the recommended dose of monensin. Following an acclimatization period of 14 d, during which cows were fed a standard lactating cow total mixed ration containing 24 ppm monensin, 18 lactating Holstein dairy cows were grouped according to the level of feed intake and then randomly assigned within each group to 1 of 3 challenge rations delivering 72, 144, and 240 ppm monensin. Outcome measurements included individual cow daily feed intakes, daily milk production, body weights, and monensin residues in composite milk samples from each cow. There were no detectable monensin residues (< 0.005 microg/mL) in any of the milk samples collected. Lactating cows receiving a dose of 72 ppm monensin exhibited up to a 20% reduction in dry matter intake, and a 5% to 15% drop in milk production from the pre-challenge period. Cows receiving doses of 144 and 240 ppm monensin exhibited rapid decreases in feed intake of up to 50% by the 2nd d and milk production losses of up to 20% and 30%, respectively, within 4 d. Lactating cows receiving up to 4865 mg monensin per day had no detectable monensin residues (< 0.005 microg/mL) in any of the milk samples collected. Results of this study confirm that food products derived from lactating dairy cattle receiving monensin at recommended levels are safe for human consumption.


Assuntos
Bovinos/fisiologia , Resíduos de Drogas/análise , Ionóforos/farmacocinética , Lactação/efeitos dos fármacos , Leite/química , Monensin/farmacocinética , Animais , Bovinos/metabolismo , Qualidade de Produtos para o Consumidor , Relação Dose-Resposta a Droga , Ingestão de Energia/efeitos dos fármacos , Feminino , Ionóforos/administração & dosagem , Lactação/fisiologia , Leite/metabolismo , Monensin/administração & dosagem , Distribuição Aleatória
18.
Vet Hum Toxicol ; 46(5): 242-5, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15487643

RESUMO

Enrofloxacin, a fluoroquinolone, and its interaction with monensin, an ionophore drug, was studied to explore the influence of enrofloxacin on drug metabolizing enzymes that can lead to physiological and toxicological consequences upon coadministration with monensin in broiler chickens. Group I, treated with 100 mg monesin/kg feed from 1 d old to 41st d of age, did not show any influence on aniline hydroxylase and cytochrome b5 levels. Group II, treated with 10 mg enrofloxacin/kg body weight per os for three consecutive days on 33rd, 34th, 35th d of age, had a highly significant decrease in aniline hydroxylase on 38th d (ie on 3rd d post-treatment with enrofloxacin); a reversal effect was noticed on the 41st day (ie on 6th d post-treatment with enrofloxacin). There was no alteration in cytochrome b5 level. Group III with monensin and enrofloxacin coadministration 100 mg monensin/kg feed from 1 d old to the 41st day + 10 mg enrofloxacin/kg body weight, per os for 3 consecutive days on the 33rd, 34th, 35th d of age) had a significant decrease in aniline hydroxylase level on the 3rd d post-treatment with enrofloxacin, but an elevation tending to reach normal on the 6th d post-treatment with enrofloxacin. Monensin + enrofloxacin coadministration did not produce any alteration in cytochrome b5 level. Creatine kinase (CK) and alanine amino transferase (ALT) levels significantly increased on the 3rd d post-treatment with enrofloxacin, but on the 6th d post-treatment with enrofloxacin the increase declined. Aspartate amino transferase (AST) significantly increased on the 6th d post enrofloxacin treatment. This study demonstrated the reversible competitive type of inhibition of enrofloxacin on CYP450 enzymes, and with coadministration with monensin produced increased CK, AST and ALT serum enzymes suggesting heart and liver injury. Simultaneous administration of enrofloxacin and monensin even at recommended levels could result in adverse interactions.


Assuntos
Antineoplásicos/toxicidade , Antiprotozoários/toxicidade , Sistema Enzimático do Citocromo P-450/farmacologia , Fluoroquinolonas/toxicidade , Monensin/toxicidade , Quinolonas/toxicidade , Animais , Antineoplásicos/farmacocinética , Antiprotozoários/farmacocinética , Galinhas , Sistema Enzimático do Citocromo P-450/efeitos dos fármacos , Interações Medicamentosas , Enrofloxacina , Fluoroquinolonas/farmacocinética , Fígado/patologia , Masculino , Monensin/farmacocinética , Miocárdio/patologia , Quinolonas/farmacocinética
19.
Avian Dis ; 48(2): 317-23, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15283418

RESUMO

Medicinal feed additives bacitracin, chlortetracycline (CTC), laidlomycin, lasalocid, and salinomycin inhibited the transfer of multiresistance-conferring plasmid pBR325 (Tet(r) Amp(r) Cp(r), 6.0 kb) into selected gram-negative strains with the use of an in vitro model. High concentrations of ampicillin-sensitive competence-pretreated Escherichia coli HB 101 cells were exposed to 10% (v/v) of 1:10 dimethyl sulfoxide/agent : water containing test mixtures for 0.5 hr prior to plasmid addition and transforming conditions. Transformation was inhibited for all antimicrobials and showed a positive association wich higher concentration. Additional testing of ionophore compounds separately and in combination with bacitracin, chlortetracycline, lincomycin, roxarsone, tylosin, and virginiamycin at representative feed concentrations demonstrated 80.6% to >99.9% inhibition (P < 0.001) of resistance transfer. Bacitracin alone inhibited transformation within the range of 50-500 ppm. No increase in resistance transfer was observed when poultry-derived and reference gram-negative isolates having low or no transformation efficiency were additionally tested. The results suggest that these compounds, at relevant concentrations used in animal feed, may interfere with cell envelope-associated DNA uptake channels or other transformation competence mechanisms. Through these mechanisms, ionophores and cell membrane-interactive feed agents such as CTC and bacitracin may act to inhibit resistance transfer mechanisms within poultry and livestock.


Assuntos
Bacitracina/farmacologia , Clortetraciclina/farmacologia , Farmacorresistência Bacteriana , Quimioterapia Combinada/farmacologia , Escherichia coli/efeitos dos fármacos , Ionóforos/farmacologia , Monensin/análogos & derivados , Transformação Bacteriana/efeitos dos fármacos , Lasalocida/farmacocinética , Lasalocida/farmacologia , Monensin/farmacocinética , Plasmídeos , Piranos/farmacologia , Roxarsona/farmacocinética , Transformação Bacteriana/genética , Tilosina/farmacologia , Virginiamicina/farmacologia
20.
J Pharm Pharmacol ; 53(5): 617-27, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11370701

RESUMO

The carboxylic ionophore monensin was formulated into long-circulating nanoparticles with the help of polyethylene glycol/poly (DL-lactide-co-glycolide) diblock copolymers, in an attempt to enhance the cytotoxicity of a ricin-based immunotoxin, anti-My9, and anticancer drugs like adriamycin and tamoxifen. This study looked into various aspects involving the preparation (using a homogenizer and an EmulsiFlex homogenizer-extrusion device) and lyophilization of long-circulating monensin nanoparticles (LMNP) of particle size < 200 nm in diameter. The particle size of LMNP was reduced from 194 nm to 160 nm by passing the nanoparticles through an EmulsiFlex, before freeze-drying. There was a 4.8-83.7% increase in the particle size of LMNP after freeze-drying, which was dependent upon the manufacturing conditions such as use of the EmulsiFlex for size reduction before freeze-drying, the freezing method (rapid/slow) and the concentration of lyoprotectant (mannitol or trehalose) employed for freeze-drying. LMNP freeze-dried with 2.4% of trehalose showed minimal size change (< 9%) after freeze-drying. Further, the freezing method was found to have negligible effect on the particle size of LMNP freeze-dried with trehalose in comparison with mannitol. The entrapment efficiency of monensin in LMNP was found to be 14.2 +/- 0.3%. The LMNP were found to be spherical in shape and smooth in surface texture as observed by atomic force microscopy. In-vitro release of monensin from LMNP in phosphate buffered saline (PBS) pH 7.4 or PBS supplemented with 10% human serum indicated that there was an initial rapid release of about 40% in the first 8 h followed by a fairly slow release (about 20%) in the next 88 h. In-vivo studies conducted with Sprague-Dawley rats showed that 20% of monensin remained in circulation 4-8 h after the intravenous administration of LMNP. An in-vitro dye-based cytotoxicity assay (MTS/PMS method) showed that there was 500 times and 5 times potentiation of the cytotoxicity of anti-My9 immunotoxin by LMNP (5 x 10(-8) M of monensin) in HL-60 sensitive and resistant human tumour cell lines, respectively. Further, LMNP (5 x 10(-8) M of monensin) potentiated the cytotoxicity of adriamycin in MCF 7 and SW 620 cell lines by 100 fold and 10 fold, respectively, and that of tamoxifen by 44 fold in MCF 7 cell line as assessed by crystal violet dye uptake assay. Our results suggest that it is possible to prepare LMNP possessing appropriate particle size (< 200 nm), monensin content and in-vitro and in-vivo release characteristics with the help of a homogenizer and an EmulsiFlex homogenizer-extrusion device. LMNP can be freeze-dried with minimal increase in particle size by using a suitable concentration of a lyoprotectant like trehalose. Furthermore, LMNP could potentiate the cytotoxicity of immunotoxin, adriamycin and tamoxifen by 5-500 fold in-vitro, which will be further investigated in-vivo in a suitable animal model.


Assuntos
Antifúngicos/farmacocinética , Antineoplásicos Fitogênicos/farmacologia , Antineoplásicos/farmacologia , Doxorrubicina/farmacologia , Imunotoxinas/farmacologia , Monensin/farmacocinética , Ricina/farmacologia , Tamoxifeno/farmacologia , Animais , Antifúngicos/imunologia , Antifúngicos/farmacologia , Interações Medicamentosas , Excipientes/química , Liofilização , Meia-Vida , Concentração de Íons de Hidrogênio , Monensin/imunologia , Monensin/farmacologia , Tamanho da Partícula , Polietilenoglicóis/química , Polímeros , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...