Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 168
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
J Pharmacol Exp Ther ; 370(3): 350-359, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31201216

RESUMO

Glucose-stimulated insulin secretion from pancreatic ß-cells is controlled by ATP-regulated potassium (KATP) channels composed of Kir6.2 and sulfonylurea receptor 1 (SUR1) subunits. The KATP channel-opener diazoxide is FDA-approved for treating hyperinsulinism and hypoglycemia but suffers from off-target effects on vascular KATP channels and other ion channels. The development of more specific openers would provide critically needed tool compounds for probing the therapeutic potential of Kir6.2/SUR1 activation. Here, we characterize a novel scaffold activator of Kir6.2/SUR1 that our group recently discovered in a high-throughput screen. Optimization efforts with medicinal chemistry identified key structural elements that are essential for VU0071063-dependent opening of Kir6.2/SUR1. VU0071063 has no effects on heterologously expressed Kir6.1/SUR2B channels or ductus arteriole tone, indicating it does not open vascular KATP channels. VU0071063 induces hyperpolarization of ß-cell membrane potential and inhibits insulin secretion more potently than diazoxide. VU0071063 exhibits metabolic and pharmacokinetic properties that are favorable for an in vivo probe and is brain penetrant. Administration of VU0071063 inhibits glucose-stimulated insulin secretion and glucose-lowering in mice. Taken together, these studies indicate that VU0071063 is a more potent and specific opener of Kir6.2/SUR1 than diazoxide and should be useful as an in vitro and in vivo tool compound for investigating the therapeutic potential of Kir6.2/SUR1 expressed in the pancreas and brain.


Assuntos
Ativação do Canal Iônico/efeitos dos fármacos , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Receptores de Sulfonilureias/metabolismo , Xantinas/farmacologia , Xantinas/farmacocinética , Animais , Canal Arterial/efeitos dos fármacos , Canal Arterial/fisiologia , Glucose/farmacologia , Células HEK293 , Humanos , Secreção de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Camundongos , Relação Estrutura-Atividade , Vasodilatação/efeitos dos fármacos , Xantinas/química
2.
Electrophoresis ; 39(19): 2446-2453, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30051931

RESUMO

Discovering hit compounds and optimization processes in medicinal chemistry nowadays could be improved by predictive tools, based on the relationship between structure of molecules and lipophilic properties. Lipophilicity of drug candidate can affect both the pharmacokinetic and pharmacodynamics properties, in particular, the ability of a molecule to cross the cell membrane. Among the new methods for determination of the lipophilicity of compounds, micellar electrokinetic chromatography (MEKC) is considered to be an appropriate one for bioactive molecules, as it closely mimics the physiological conditions. In this paper MEKC was used for the estimation of the lipophilicity of 24 derivatives of 8-alkoxy-7H-purine-2,6-dione, designed and synthesized as potential antidepressant/anxiolytic and antipsychotic agents. The results of experimental method were compared with calculated in silico parameters (AlogPs and milogP by Virtual Computational Laboratory website, log PPallas by Pallas 3.1, Mlog P by Marvin, log PChemS by ChemSketch, log PChemDraw by ChemBioUltra) using Principal Component Analysis (PCA) method. Finally, using estimated log P values for selected compounds ligand - lipophilicity efficiency (LLE), per cent efficiency index (PEI), and binding efficiency index (BEI) parameters were calculated. Applied MEKC procedure could be used for selection of potential lead structure in a group of 7H-purine-2,6-dione derivatives.


Assuntos
Cromatografia Capilar Eletrocinética Micelar/métodos , Psicotrópicos/química , Xantinas/química , Descoberta de Drogas , Interações Hidrofóbicas e Hidrofílicas , Ligantes , Modelos Lineares , Psicotrópicos/análise , Psicotrópicos/farmacocinética , Xantinas/análise , Xantinas/farmacocinética
3.
Acta Pharmacol Sin ; 37(10): 1359-1369, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27264313

RESUMO

AIM: TGR5 agonists stimulate intestinal glucagon-like peptide-1 (GLP-1) release, but systemic exposure causes unwanted side effects, such as gallbladder filling. In the present study, linagliptin, a DPP-4 inhibitor with a large molecular weight and polarity, and MN6, a previously described TGR5 agonist, were linked to produce OL3, a novel low-absorbed TGR5 agonist with reduced side-effects and dual function in lowering blood glucose by activation of TGR5 and inhibition of DPP-4. METHODS: TGR5 activation was assayed in HEK293 cells stably expressing human or mouse TGR5 and a CRE-driven luciferase gene. DPP-4 inhibition was assessed based on the rate of hydrolysis of a surrogate substrate. GLP-1 secretion was measured in human enteroendocrine NCI-H716 cells. OL3 permeability was tested in Caco-2 cells. Acute glucose-lowering effects of OL3 were evaluated in ICR and diabetic ob/ob mice. RESULTS: OL3 activated human and mouse TGR5 with an EC50 of 86.24 and 17.36 nmol/L, respectively, and stimulated GLP-1 secretion in human enteroendocrine NCI-H716 cells (3-30 µmol/L). OL3 inhibited human and mouse DPP-4 with IC50 values of 18.44 and 69.98 µmol/L, respectively. Low permeability of OL3 was observed in Caco-2 cells. In ICR mice treated orally with OL3 (150 mg/kg), the serum OL3 concentration was 101.10 ng/mL at 1 h, and decreased to 13.38 ng/mL at 5.5 h post dose, confirming the low absorption of OL3 in vivo. In ICR mice and ob/ob mice, oral administration of OL3 significantly lowered the blood glucose levels, which was a synergic effect of activating TGR5 that stimulated GLP-1 secretion in the intestine and inhibiting DPP-4 that cleaved GLP-1 in the plasma. In ICR mice, oral administration of OL3 did not cause gallbladder filling. CONCLUSION: OL3 is a low-absorbed TGR5 agonist that lowers blood glucose without inducing gallbladder filling. This study presents a new strategy in the development of potent TGR5 agonists in treating type 2 diabetes, which target to the intestine to avoid systemic side effects.


Assuntos
Glicemia/metabolismo , Inibidores da Dipeptidil Peptidase IV/farmacologia , Hipoglicemiantes/farmacologia , Quinoxalinas/farmacologia , Receptores Acoplados a Proteínas G/agonistas , Xantinas/farmacologia , Animais , Células CACO-2 , Inibidores da Dipeptidil Peptidase IV/efeitos adversos , Inibidores da Dipeptidil Peptidase IV/metabolismo , Humanos , Hipoglicemiantes/efeitos adversos , Hipoglicemiantes/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos ICR , Permeabilidade , Quinoxalinas/metabolismo , Quinoxalinas/farmacocinética , Xantinas/metabolismo , Xantinas/farmacocinética
4.
Neuroimage ; 92: 83-9, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24513151

RESUMO

Adenosine A1 receptors (A1Rs) are implied in the modulation of neuroinflammation. Activation of cerebral A1Rs acts as a brake on the microglial response after traumatic brain injury and has neuroprotective properties in animal models of Parkinson's disease and multiple sclerosis. Neuroinflammatory processes in turn may affect the expression of A1Rs, but the available data is limited and inconsistent. Here, we applied an animal model of encephalitis to assess how neuroinflammation affects the expression of A1Rs. Two groups of animals were studied: Infected rats (n=7) were intranasally inoculated with herpes simplex virus-1 (HSV-1, 1 × 10(7) plaque forming units), sham-infected rats (n=6) received only phosphate-buffered saline. Six or seven days later, microPET scans (60 min with arterial blood sampling) were made using the tracer 8-dicyclopropyl-1-(11)C-methyl-3-propyl-xanthine ((11)C-MPDX). Tracer clearance from plasma and partition coefficient (K1/k2 estimated from a 2-tissue compartment model fit) were not significantly altered after virus infection. PET tracer distribution volume calculated from a Logan plot was significantly increased in the hippocampus (+37%) and medulla (+27%) of virus infected rats. Tracer binding potential (k3/k4 estimated from the model fit) was significantly increased in the cerebellum (+87%) and the medulla (+148%) which may indicate increased A1R expression. This was confirmed by immunohistochemical analysis showing a strong increase of A1R immunoreactivity in the cerebellum of HSV-1-infected rats. Both the quantitative PET data and immunohistochemical analysis indicate that A1Rs are upregulated in brain areas where active virus is present.


Assuntos
Encéfalo/metabolismo , Encefalite Viral/metabolismo , Infecções por HIV/metabolismo , Fatores Imunológicos/metabolismo , Tomografia por Emissão de Pósitrons/veterinária , Receptor A1 de Adenosina/metabolismo , Xantinas/farmacocinética , Animais , Animais não Endogâmicos , Encefalite Viral/diagnóstico por imagem , Infecções por HIV/diagnóstico por imagem , Taxa de Depuração Metabólica , Tomografia por Emissão de Pósitrons/métodos , Compostos Radiofarmacêuticos/farmacocinética , Ratos , Ratos Wistar , Distribuição Tecidual , Regulação para Cima
5.
Neurobiol Dis ; 71: 193-204, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25132555

RESUMO

Adenosine A1 receptor (A1R) stimulation exerts beneficial effects in response to various insults to the brain and, although it was found neuroprotective in a lesional model of Huntington's disease (HD), the features of this receptor in genetic models of HD have never been explored. In the present study we characterized the expression, affinity and functional effects of A1Rs in R6/2 mice (the most widely used transgenic model of HD) and in a cellular model of HD. Binding studies revealed that the density of A1Rs was significantly reduced in the cortex and the striatum of R6/2 mice compared to age-matched wild-type (WT), while receptor affinity was unchanged. The selective A1R agonist cyclopentyladenosine (CPA, 300nM) was significantly more effective in reducing synaptic transmission in corticostriatal slices from symptomatic R6/2 than in age-matched WT mice. Such an effect was due to a stronger inhibition of glutamate release from the pre-synaptic terminal. The different functional activities of A1Rs in HD mice were associated also to a different intracellular signaling pathway involved in the synaptic effect of CPA. In fact, while the PKA pathway was involved in both genotypes, p38 MAPK inhibitor SB203580 partially prevented synaptic effects of CPA in R6/2, but not in WT, mice; moreover, CPA differently modulated the phosphorylation status of p38 in the two genotypes. In vitro studies confirmed a different behavior of A1Rs in HD: CPA (100 nM for 5h) modulated cell viability in STHdh(Q111/Q111) (mhttHD cells), without affecting the viability of STHdh(Q7/Q7) (wthtt cells). This effect was prevented by the application of SB203580. Our results demonstrate that in the presence of the HD mutation A1Rs undergo profound changes in terms of expression, pharmacology and functional activity. These changes have to be taken in due account when considering A1Rs as a potential therapeutic target for this disease.


Assuntos
Adenina/análogos & derivados , Ciclopentanos/farmacologia , Regulação da Expressão Gênica/genética , Doença de Huntington/metabolismo , Receptor A1 de Adenosina/metabolismo , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/genética , Adenina/farmacologia , Antagonistas do Receptor A1 de Adenosina/farmacocinética , Animais , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , AMP Cíclico/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Estimulação Elétrica , Inibidores Enzimáticos/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Ácido Glutâmico/metabolismo , Proteína Huntingtina , Doença de Huntington/genética , Doença de Huntington/patologia , Técnicas In Vitro , Masculino , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Proteínas Nucleares/genética , Cloreto de Potássio/farmacologia , Ligação Proteica/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Estatísticas não Paramétricas , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/genética , Sinaptossomos/efeitos dos fármacos , Sinaptossomos/metabolismo , Transfecção , Expansão das Repetições de Trinucleotídeos/genética , Trítio/farmacocinética , Xantinas/farmacocinética
6.
Eur J Nucl Med Mol Imaging ; 41(6): 1210-20, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24566949

RESUMO

PURPOSE: To study cerebral adenosine receptors (AR) in premanifest and manifest stages of Huntington's disease (HD). METHODS: We quantified the cerebral binding potential (BP ND) of the A1AR in carriers of the HD CAG trinucleotide repeat expansion using the radioligand [(18) F]CPFPX and PET. Four groups were investigated: (i) premanifest individuals far (preHD-A; n = 7) or (ii) near (preHD-B; n = 6) to the predicted symptom onset, (iii) manifest HD patients (n = 8), and (iv) controls (n = 36). RESULTS: Cerebral A1AR values of preHD-A subjects were generally higher than those of controls (by up to 31%, p < .01, in the thalamus on average). Across stages a successive reduction of A1AR BPND was observed to the levels of controls in preHD-B and undercutting controls in manifest HD by down to 25%, p < .01, in the caudatus and amygdala. There was a strong correlation between A1AR BP ND and years to onset. Before onset of HD, the assumed annual rates of change of A1AR density were -1.2% in the caudatus, -1.7% in the thalamus and -3.4% in the amygdala, while the corresponding volume losses amounted to 0.6%, 0.1% and 0.2%, respectively. CONCLUSIONS: Adenosine receptors switch from supra to subnormal levels during phenoconversion of HD. This differential regulation may play a role in the pathophysiology of altered energy metabolism.


Assuntos
Doença de Huntington/diagnóstico por imagem , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos/farmacocinética , Receptor A1 de Adenosina/metabolismo , Xantinas/farmacocinética , Adulto , Idade de Início , Idoso , Encéfalo/metabolismo , Estudos de Casos e Controles , Estudos Transversais , Feminino , Humanos , Doença de Huntington/diagnóstico , Masculino , Pessoa de Meia-Idade , Testes Neuropsicológicos , Ligação Proteica , Distribuição Tecidual
7.
Drug Metab Dispos ; 41(11): 1957-66, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23996078

RESUMO

This study aimed to investigate the impact of methylxanthine abstinence (MA) periods on CYP1A2 activity in individuals with varying levels of caffeine consumption through development of a population pharmacokinetic model of caffeine and its major metabolite paraxanthine. This study developed and evaluated a mixed-effects pharmacokinetic model for caffeine and paraxanthine concentration-time data derived from a sequential single-dose cross-over study in healthy male volunteers (n = 30) who received oral 100 mg caffeine doses. Participants received caffeine with and without a MA period. Participants were classified as low (0-100 mg/d), medium (100-200 mg/d), or high (>200 mg/d) caffeine consumers (LCCs, MCCs, or HCCs, respectively). All caffeine and paraxanthine concentration-time data were simultaneously modeled. Caffeine pharmacokinetics was described by a two-compartment model with first-order absorption and two first-order elimination pathways. Paraxanthine was described by a one-compartment model with first-order absorption and elimination. Among LCCs (n = 16) and MCCs (n = 9), there was no difference in the mean (95% confidence interval) total apparent caffeine clearance (CL) between the MA period [LCCs: 6.88 (5.61-8.16 l/h); MCCs: 10.09 (7.57-12.60 l/h)] versus the no MA period [LCCs: 6.22 (4.97-7.46 l/h); MCCs: 9.68 (7.12-12.24 l/h)]. The mean CL among HCCs (n = 5) was considerably higher in the MA period [10.48 (5.62-15.33 l/h)] compared with the no MA period [6.30 (3.40-9.20 l/h)] (P < 0.05). The decrease in CL in the no MA period among HCC appears to be due to alternative caffeine elimination pathways, rather than CYP1A2.


Assuntos
Cafeína/farmacocinética , Citocromo P-450 CYP1A2/metabolismo , Xantinas/farmacocinética , Adulto , Estudos Cross-Over , Humanos , Masculino , Modelos Biológicos , Teofilina/farmacocinética , Adulto Jovem
8.
J Neurochem ; 123(1): 100-12, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22639925

RESUMO

Studies with multiple sclerosis patients and animal models of experimental autoimmune encephalomyelitis (EAE) implicate adenosine and adenosine receptors in modulation of neuroinflammation and brain injury. Although the involvement of the A(1) receptor has been recently demonstrated, the role of the adenosine A(2A) receptor (A(2A)R) in development of EAE pathology is largely unknown. Using mice with genetic inactivation of the A(2A) receptor, we provide direct evidence that loss of the A(2A)R exacerbates EAE pathology in mice. Compared with wild-type mice, A(2A)R knockout mice injected with myelin oligodendroglia glycoprotein peptide had a higher incidence of EAE and exhibited higher neurological deficit scores and greater decrease in body weight. A(2A)R knockout mice displayed increased inflammatory cell infiltration and enhanced microglial cell activation in cortex, brainstem, and spinal cord. In addition, demyelination and axonal damage in brainstem were exacerbated, levels of Th1 cytokines increased, and Th2 cytokines decreased. Collectively, these findings suggest that extracellular adenosine acting at A(2A)Rs triggers an important neuroprotective mechanism. Thus, the A(2A) receptor is a potential target for therapeutic approaches to multiple sclerosis.


Assuntos
Lesões Encefálicas/etiologia , Lesões Encefálicas/patologia , Encefalomielite Autoimune Experimental/complicações , Regulação da Expressão Gênica/genética , Microglia/patologia , Receptor A2A de Adenosina/genética , Antagonistas do Receptor A1 de Adenosina/farmacocinética , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/patologia , Axônios/patologia , Lesões Encefálicas/complicações , Proliferação de Células , Células Cultivadas , Córtex Cerebral/patologia , Citocinas/metabolismo , Doenças Desmielinizantes/etiologia , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/induzido quimicamente , Encefalomielite Autoimune Experimental/imunologia , Ensaio de Imunoadsorção Enzimática , Feminino , Filtração , Citometria de Fluxo , Adjuvante de Freund/toxicidade , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Glicoproteína Mielina-Oligodendrócito/toxicidade , Fragmentos de Peptídeos/toxicidade , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , RNA Mensageiro/metabolismo , Receptor A2A de Adenosina/deficiência , Medula Espinal/patologia , Baço/citologia , Estatísticas não Paramétricas , Trítio/farmacocinética , Xantinas/farmacocinética
9.
Int J Clin Pharmacol Ther ; 49(9): 563-70, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21888869

RESUMO

OBJECTIVE: The study was conducted to characterize the pharmacokinetics and pharmacodynamics of tonapofylline in subjects with severe renal impairment and in elderly subjects. METHOD: Subjects with severe renal impairment were matched demographically with healthy subjects. Elderly subjects with normal renal function for their ages were also enrolled. All subjects (n = 8 per group) received a single intravenous administration of tonapofylline at 1 mg/kg. RESULTS: The pharmacokinetics of tonapofylline was not significantly different in subjects with severe renal impairment, or in elderly subjects, as compared to healthy subjects. Among all pharmacokinetic parameters, the only statistically significant difference was observed for Cmax between the healthy and the severe renal impairment groups, which was 21% and considered clinically insignificant. Pharmacodynamic assessment demonstrated the natriuretic effects of tonapofylline across groups, with little accompanying kaliuresis. No change in renal function occurred after single dose of tonapofylline, despite substantial increases in excretion of urinary sodium. Single 1 mg/kg intravenous administration of tonapofylline was generally safe. CONCLUSION: The pharmacokinetics of tonapofylline in subjects with severe renal impairment and elderly subjects with normal renal function for age is similar to that in healthy subjects. It has been demonstrated in all groups that tonapofylline has natriuretic effects and is able to maintain renal function, which can be beneficial to patients with congestive heart failure.


Assuntos
Antagonistas do Receptor A1 de Adenosina/farmacocinética , Insuficiência Renal/metabolismo , Xantinas/farmacocinética , Antagonistas do Receptor A1 de Adenosina/efeitos adversos , Antagonistas do Receptor A1 de Adenosina/farmacologia , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Xantinas/efeitos adversos , Xantinas/farmacologia
10.
Biomed Chromatogr ; 25(3): 381-90, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21110389

RESUMO

A sensitive and selective liquid chromatographic-electrospray ionization mass spectrometric method for the simultaneous determination of propentofylline and enantiomers of its active metabolite M1 in rat serum, cortex and hippocampus was developed and validated according to GLP procedures. Sample preparations were carried out by liquid-liquid extraction using diethyl ether after the addition of the internal standard (pentoxifylline). The dried residue was reconstituted in mobile phase and injected onto a Chiralpak AD column (10 µm, 250 × 4.6 mm i.d.). The limit of quantification for propentofylline in serum, cortex and hippocampus was set at 0.25 ng/mL and for enantiomers of its metabolite M1 at 1.25 ng/mL. The established LC/ESI-MS/MS method has been successfully applied to an initial pharmacokinetic study of propentofylline and also to assessment of distribution of parent drug and enantiomers of its pharmacologically active metabolite M1 to cortex and hippocampus after intravenous administration of propentofylline to rats at a dose of 5 mg/kg.


Assuntos
Cromatografia Líquida/métodos , Espectrometria de Massas em Tandem/métodos , Xantinas/análise , Animais , Córtex Cerebral/química , Córtex Cerebral/metabolismo , Estabilidade de Medicamentos , Hipocampo/química , Hipocampo/metabolismo , Masculino , Pentoxifilina/análise , Pentoxifilina/farmacocinética , Ratos , Ratos Wistar , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Estereoisomerismo , Xantinas/sangue , Xantinas/farmacocinética
11.
Am J Ther ; 17(1): 8-16, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20027105

RESUMO

Rolofylline is a potent, selective adenosine A1 receptor antagonist that was under development for the treatment of patients with acute decompensated heart failure and renal function impairment. The 30-mg dose of rolofylline administered by intravenous infusion over 4 hours for 3 days represented the anticipated recommended clinical regimen of rolofylline. This was a randomized, double-blind, double-dummy, placebo-controlled, three-period crossover study performed with a single 2-hour intravenous infusion of 60 mg rolofylline, placebo, or oral moxifloxacin in healthy subjects. Plasma samples were collected for determination of rolofylline, M1-trans, and M1-cis pharmacokinetic parameters. The upper limit of the two-sided 90% confidence interval for the placebo-adjusted least squares mean change from baseline in QTcF interval for rolofylline was less than 5 msec at every time point. Moxifloxacin demonstrated an increase in QTcF of greater than 10 msec at 2, 2.5, and 3 hours postdose, thus establishing the sensitivity of the assay to detect modest increases in QTcF interval. Mean Cmax values of 1947.4, 739.2, and 54.8 nM were attained for rolofylline and its metabolites M1-trans and M1-cis, respectively, which were 2.2- to 3.1-fold higher than historic Cmax values seen at the anticipated clinical dose and regimen. Adenosine A1 receptor antagonism from a single supratherapeutic intravenous dose of 60 mg rolofylline over 2 hours was generally well tolerated and did not prolong the QTcF interval relative to placebo.


Assuntos
Antagonistas do Receptor A1 de Adenosina , Diuréticos/efeitos adversos , Xantinas/efeitos adversos , Administração Oral , Adulto , Compostos Aza/efeitos adversos , Estudos Cross-Over , Diuréticos/administração & dosagem , Diuréticos/farmacocinética , Relação Dose-Resposta a Droga , Método Duplo-Cego , Eletrocardiografia , Feminino , Fluoroquinolonas , Humanos , Infusões Intravenosas , Síndrome do QT Longo/induzido quimicamente , Masculino , Pessoa de Meia-Idade , Moxifloxacina , Quinolinas/efeitos adversos , Xantinas/administração & dosagem , Xantinas/farmacocinética , Adulto Jovem
12.
Pharmacology ; 85(5): 264-71, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20389149

RESUMO

AIMS: Pentoxifylline and lisofylline are methylxanthine derivatives that exhibit anti-inflammatory activity both in vitro and in vivo. This study was designed to develop a pharmacokinetic-pharmacodynamic (PK/PD) model to describe the inhibitory effect of these compounds on TNF-alpha production in mice challenged with bacterial lipopolysaccharide (LPS). METHODS: Male CD-1 mice received increasing intravenous doses of either compound simultaneously with high-dose LPS. A 2-compartment model with Michaelis-Menten-type elimination was used to describe the drug concentration versus time data. Serum TNF-alpha levels were fitted to an indirect response model. RESULTS: Pentoxifylline and lisofylline reduced LPS-induced TNF-alpha serum concentrations in a dose-dependent manner. PK/PD analysis revealed an almost 2-fold higher estimate of K(m) for pentoxifylline in comparison to lisofylline. The production and elimination rates of TNF-alpha were: k(in) = 2,167 pg/ml * h(-1) and k(out) = 1.65 h(-1), respectively. The drug concentration causing 50% of TNF inhibition (IC(50)) was markedly lower for pentoxifylline (0.47 vs. 1.61 microg/ml). CONCLUSIONS: It seems that pentoxifylline is more potent than lisofylline in inhibiting TNF-alpha production in vivo. The proposed PK/PD model allowed a better understanding of the pharmacological properties of both methylxanthine derivatives and may be helpful in appropriate dosage selection for further studies.


Assuntos
Anti-Inflamatórios não Esteroides/farmacocinética , Anti-Inflamatórios não Esteroides/uso terapêutico , Inflamação/tratamento farmacológico , Lipopolissacarídeos/toxicidade , Modelos Biológicos , Xantinas/farmacocinética , Xantinas/uso terapêutico , Animais , Anti-Inflamatórios não Esteroides/sangue , Biomarcadores/sangue , Relação Dose-Resposta a Droga , Meia-Vida , Inflamação/sangue , Inflamação/induzido quimicamente , Lipopolissacarídeos/administração & dosagem , Masculino , Camundongos , Pentoxifilina/análogos & derivados , Pentoxifilina/sangue , Pentoxifilina/farmacocinética , Pentoxifilina/uso terapêutico , Fator de Necrose Tumoral alfa/sangue , Xantinas/sangue
13.
Mol Nutr Food Res ; 63(9): e1801239, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30690879

RESUMO

SCOPE: Dietary polyphenols may protect against breast cancer. However, it is unknown whether polyphenols reach human malignant breast tumors in molecular forms and(or) at concentrations likely to act against cancer. METHODS AND RESULTS: Ninteen breast cancer patients consumed three capsules daily from biopsy-confirmed diagnosis to surgery (6 ± 2 days). The capsules contained pomegranate, orange, lemon, olive, cocoa, and grapeseed extracts plus resveratrol, providing 37 different phenolics (473.7 mg), theobromine and caffeine (19.7 mg). A total of 101 metabolites are identified in urine, 69 in plasma, 39 in normal (NT), and 33 in malignant (MT) tissues by UPLC-ESI-QTOF-MS. Eight control patients did not consume extracts. Phenolic-derived metabolites in MT and NT are mainly glucuronidated and(or) sulfated. Some representative metabolites detected in MT (median and range, pmol g-1 ) are urolithin-A-3-O-glucuronide (26.2; 3.2-66.5), 2,5-dihydroxybenzoic acid (40.2; 27.7-52.2), resveratrol-3-O-sulfate (86.4; 7.8-224.4), dihydroresveratrol-3-O-glucuronide (109.9; 10.3-229.4), and theobromine (715.0; 153.9-3,216). Metabolites, as detected in breast tissues, do not exert antiproliferative or estrogenic/antiestrogenic activities in MCF-7 breast cancer cells. CONCLUSION: This is the first study that describes the metabolic profiling of dietary phenolics and methylxanthines in MT and NT comprehensively. Although phase-II conjugation might hamper a direct anticancer activity, long-term tumor-senescent chemoprevention cannot be discarded.


Assuntos
Antineoplásicos/farmacocinética , Neoplasias da Mama/dietoterapia , Polifenóis/farmacocinética , Xantinas/farmacocinética , Adulto , Idoso , Neoplasias da Mama/metabolismo , Neoplasias da Mama/cirurgia , Neoplasias da Mama Masculina/dietoterapia , Neoplasias da Mama Masculina/metabolismo , Neoplasias da Mama Masculina/cirurgia , Cafeína/farmacocinética , Proliferação de Células/efeitos dos fármacos , Suplementos Nutricionais , Feminino , Humanos , Células MCF-7 , Masculino , Glândulas Mamárias Humanas/efeitos dos fármacos , Glândulas Mamárias Humanas/metabolismo , Pessoa de Meia-Idade , Teobromina/farmacocinética
14.
Pharmacol Rep ; 71(4): 676-681, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31200233

RESUMO

BACKGROUND: Adenosine, an endogenous nucleoside, modulates the release of monoamines, e.g., noradrenaline, serotonin, and dopamine in the brain. Both nonselective and selective stimulation of adenosine receptors produce symptoms of depression in some animal models. Therefore, the main objective of our study was to assess the influence of a selective adenosine A1 receptor antagonist (DPCPX) and a selective adenosine A2A receptor antagonist (DMPX) on the activity of agomelatine and tianeptine. METHODS: The forced swim test (FST) and tail suspension test (TST) were performed to assess the effects of DPCPX and DMPX on the antidepressant-like activity of agomelatine and tianeptine. Drug serum and brain levels were analyzed using HPLC. RESULTS: Co-administration of agomelatine (20 mg/kg) or tianeptine (15 mg/kg) with DMPX (3 mg/kg), but not with DPCPX (1 mg/kg), significantly reduced the immobility time both in the FST and TST in mice. These effects were not associated with an enhancement in animals' spontaneous locomotor activity. The observed changes in the mouse behavior after concomitant injection of DMPX and the tested antidepressant agents were associated with elevated brain concentration of agomelatine and tianeptine. CONCLUSION: Our study shows a synergistic action of the selective A2A receptor antagonist and the studied antidepressant drugs, and a lack of such interaction in the case of the selective A1 receptor antagonist. The interaction between DMPX and agomelatine/tianeptine at least partly occurs in the pharmacokinetic phase. A combination of a selective A2A receptor antagonist and an antidepressant may be a new strategy for treating depression.


Assuntos
Acetamidas/farmacologia , Antagonistas do Receptor A2 de Adenosina/farmacologia , Antidepressivos/farmacologia , Depressão/tratamento farmacológico , Teobromina/análogos & derivados , Tiazepinas/farmacologia , Acetamidas/farmacocinética , Antagonistas do Receptor A1 de Adenosina/farmacocinética , Antagonistas do Receptor A1 de Adenosina/farmacologia , Antagonistas do Receptor A2 de Adenosina/farmacocinética , Animais , Antidepressivos/farmacocinética , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Depressão/sangue , Depressão/metabolismo , Sinergismo Farmacológico , Masculino , Camundongos , Atividade Motora/efeitos dos fármacos , Natação , Teobromina/farmacocinética , Teobromina/farmacologia , Tiazepinas/farmacocinética , Xantinas/farmacocinética , Xantinas/farmacologia
15.
J Neurochem ; 104(2): 435-45, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17953672

RESUMO

Pregnant Wistar rats were orally treated with 1 g/L l-glutamate during the entire gestational period and the status of adenosine A(1) receptor (A(1)R)/adenylyl cyclase transduction pathway from maternal and fetal brain was analyzed. Glutamate consumption, estimated from the loss of water from the drinking bottles, was 110 +/- 4.6 mg/kg/day. In mother brains glutamate intake did not significantly alter the B(max) value, although the K(d) value was significantly decreased. However in fetus brain, a significant decrease in B(max) was observed, without an alteration of K(d) value. Similar results were observed by western blot assays using specific A(1)R antibody, suggesting a down-regulation of A(1)R in fetal brain. Concerning alpha subunits of inhibitory G proteins (Gi), alphaGi(3) protein was slightly but significantly decreased in maternal brain without alterations of either Gi(1) or Gi(2). In contrast, alphaGi(1) and alphaGi(2) isoforms were increased in fetal brain. On the other hand, basal, forskolin, and forskolin plus GTPgammaS-stimulated adenylyl cyclase activity was significantly decreased in both maternal and fetal brain, and this was more prominent in fetal than in maternal brain. Finally, A(1)R functionality was significantly decreased in mother brain whereas no significant differences were detected in fetus brain. These results suggest that glutamate administered to pregnant rats modulates A(1)R signaling pathways in both tissues, showing an A(1)R down-regulation in fetal brain, and desensitization in maternal brain.


Assuntos
Adenilil Ciclases/metabolismo , Encéfalo/metabolismo , Glutamatos/metabolismo , Efeitos Tardios da Exposição Pré-Natal , Receptor A1 de Adenosina/fisiologia , Transdução de Sinais/fisiologia , Antagonistas do Receptor A1 de Adenosina , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/ultraestrutura , Relação Dose-Resposta a Droga , Embrião de Mamíferos , Feminino , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Glutamatos/farmacologia , Plasma/efeitos dos fármacos , Plasma/enzimologia , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Transdução de Sinais/efeitos dos fármacos , Xantinas/farmacocinética
16.
Ann Nucl Med ; 22(10): 841-7, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19142702

RESUMO

OBJECTIVE: Adenosine is an endogenous modulator of synaptic functions in the central nervous system. The effects of adenosine are mediated by at least four adenosine receptor subtypes. Decreased density of adenosine A1 receptors, which is a major subtype adenosine receptor in the hippocampus, has been reported in vitro in Alzheimer's disease. We evaluated adenosine A1 receptor in the brain of elderly normal subjects and patients with Alzheimer's disease (n = 8 and 6, respectively), using positron emission tomography (PET) and 8- dicyclopropylmethyl-1-[(11)C]methyl-3-propylxanthine ([(11)C]MPDX). METHODS: A 60-min PET scan with [(11)C]MPDX was performed. The patients with Alzheimer's disease also underwent PET with [(18)F]fluorodeoxyglucose (FDG). The binding potential of [11C]MPDX was quantitatively calculated in the regions of interest (ROIs) placed on the frontal, medial frontal, temporal, medial temporal, parietal, and occipital cortices, striatum, thalamus, cerebellum, and pons. Statistical parametric mapping (SPM2) was used for analysis of [(11)C]MPDX and FDG-PET. RESULTS: In the ROI-based analysis, the binding potential of [(11)C]MPDX in patients with Alzheimer's disease was significantly lower in the temporal and medial temporal cortices and thalamus than that in elderly normal subjects (P = 0.038, 0.028, and 0.039, respectively). SPM analysis also showed significant decreased binding potential in the temporal and medial temporal cortices and thalamus in patients with Alzheimer's disease. FDG uptake was significantly decreased in the temporoparietal cortex and posterior cingulate gyrus. CONCLUSIONS: Decreased binding of [(11)C]MPDX in patients with Alzheimer's disease was detected in temporal and medial temporal cortices and thalamus. This pattern possibly differed from the hypometabolism pattern of FDG. [(11)C]MPDX PET is valuable for the detection of degeneration in the temporal and medial temporal cortices and corticothalamic transmission, and may provide a different diagnostic tool from FDG-PET in brain disorders such as Alzheimer's disease.


Assuntos
Doença de Alzheimer/diagnóstico por imagem , Doença de Alzheimer/metabolismo , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Receptor A1 de Adenosina/metabolismo , Xantinas/farmacocinética , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Compostos Radiofarmacêuticos/farmacocinética , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Distribuição Tecidual
17.
Neuroscience ; 146(1): 415-26, 2007 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-17350174

RESUMO

Adenosine promotes cytoprotection under conditions of infection, ischemic preconditioning and oxidative stress. Previous studies from our laboratory indicate that the expression of the adenosine A1 receptor (A1AR) is induced by oxidative stress via activation of nuclear factor (NF)-kappaB. The prototypic transcription factor is composed of homo- or heterodimers of p50 and p65 subunits. To determine the role of NF-kappaB in the regulation of the A1AR in vivo, we compared the A1AR RNA and protein levels in the brains of mice lacking the p50 subunit of NF-kappaB (p50-/- mice) and age-matched B6129PF2/J (F2) controls. Radioligand binding assays in the cortex revealed a significantly lower number of A(1)AR (maximal binding capacity, Bmax) in the cortex of p50-/- mice (151+/-62 fmol/mg protein) versus 479+/-181 fmol/mg protein in the F2 (N=5 per strain, P<0.05), but no change in the equilibrium dissociation constant. Similar reductions in A1AR were measured in the hippocampus, brain stem and hypothalamus and in peripheral tissues, such as the adrenal gland, kidney and spleen. Estimation of the A1AR following purification by antibody affinity columns also indicated reduced A1AR in the p50-/- mice cortex, as compared with the F2 mice. A1AR immunocytochemistry indicates distinct neuronal labeling in the F2 cortex, which was substantially reduced in similar sections obtained from p50-/- mice. The p50-/- mice expressed lower levels of A1AR mRNA than F2 mice, as determined by real time PCR. Quantitation of the A1AR transducing G proteins by Western blotting show significantly less Galphai3, no change in Galphai1, but higher levels of Galphao and Gbeta in the cortices of p50-/-, as compared with F2 mice. Administration of bacterial lipopolysaccharide (LPS), an activator of NF-kappaB, increased A1AR expression in the cortices of F2 mice but not p50-/- mice. Cortical neurons cultures prepared from p50-/- mice showed a greater degree of apoptosis, compared with neurons from F2 mice. Activation of the A1AR reduced apoptosis with greater efficacy in cultures from F2 than p50-/- mice. Taken together, these data support a role for NF-kappaB in determining both the basal and LPS-stimulated A1AR expression in vivo which could contribute to neuronal survival.


Assuntos
Encéfalo/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Subunidade p50 de NF-kappa B/deficiência , Receptor A1 de Adenosina/metabolismo , Análise de Variância , Animais , Encéfalo/ultraestrutura , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Imunoprecipitação/métodos , Marcação In Situ das Extremidades Cortadas/métodos , Masculino , Camundongos , Camundongos Knockout , Ligação Proteica/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ensaio Radioligante/métodos , Receptor A1 de Adenosina/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Xantinas/farmacocinética
18.
Naunyn Schmiedebergs Arch Pharmacol ; 375(2): 133-44, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17310264

RESUMO

Antagonists of adenosine A2A receptors (A2A -antagonists) with different chemical structures have been developed by several pharmaceutical companies for the potential treatment of Parkinson's disease. Pharmacological characterization of these antagonists was incomplete, and different assay conditions were used in different labs. Therefore, we characterized the potencies, selectivities, and pharmacokinetic profiles of six prototypical A2A -antagonists. Displacements of [3H]MSX-2 and of [3H]CGS21680 binding to the human cloned and rat A2A receptors were performed. The rank order of potency of antagonists to displace [(3)H]MSX-2 binding to the human A2A was SCH58261 > or = Biogen-34 > or = Ver-6623 > or = MSX-2 > KW-6002 > > DMPX. For the rat striatal A2A, the order of potency was Biogen-34 > or = SCH58261 > or = Ver-6623 > or = MSX-2 > or = KW-6002 > > DMPX. SCH58261 was the most potent antagonist of the human A2A with a K(i) value of 4 nM, whereas Biogen-34 was the most potent antagonist of the rat A2A with a K(i) value of 1.2 nM. Similar results were obtained from cAMP assays. Selectivities of A2A-antagonists were determined using radioligands [3H]DPCPX, [3H]ZM241385, and [125I]-AB-MECA for A1, A2B, and A3 receptors, respectively. KW-6002 and Biogen-34 exhibited the highest selectivity for A2A vs A1 (human and rat), respectively. The pharmacokinetic profiles of antagonists were evaluated in vivo in rats. DMPX and KW-6002 had the greatest oral bioavailability. In contrast, SCH58261, MSX-2, and Ver-6623 had low or poor oral bioavailability. In summary, SCH58261, Biogen-34, MSX-2, and Ver-6623 had high affinities for both human and rat A2A receptors, with reasonable selectivity for A2A over A1 and A2B receptors. They are suitable as A2A -antagonists for in vitro pharmacological studies. Among the six A2A-antagonists, KW-6002 is the best for use in in vivo animal studies, particularly for a CNS target, based on its bioavailability, half life, and brain penetration.


Assuntos
Antagonistas do Receptor A2 de Adenosina , Adenosina/análogos & derivados , Adenosina/química , Adenosina/farmacocinética , Adenosina/farmacologia , Antagonistas do Receptor A3 de Adenosina , Animais , Ligação Competitiva/efeitos dos fármacos , Disponibilidade Biológica , Linhagem Celular , AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Humanos , Masculino , Taxa de Depuração Metabólica , Estrutura Molecular , Células PC12 , Fenetilaminas/química , Fenetilaminas/farmacocinética , Fenetilaminas/farmacologia , Purinas/química , Purinas/farmacocinética , Purinas/farmacologia , Pirimidinas/química , Pirimidinas/farmacocinética , Pirimidinas/farmacologia , Ratos , Ratos Sprague-Dawley , Receptor A2A de Adenosina/genética , Receptor A3 de Adenosina/genética , Receptor A3 de Adenosina/metabolismo , Teobromina/análogos & derivados , Teobromina/química , Teobromina/farmacocinética , Teobromina/farmacologia , Triazinas/química , Triazinas/farmacocinética , Triazinas/farmacologia , Triazóis/química , Triazóis/farmacocinética , Triazóis/farmacologia , Xantinas/química , Xantinas/farmacocinética , Xantinas/farmacologia
19.
Life Sci ; 81(15): 1175-82, 2007 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-17897683

RESUMO

Whether organic anion and cation transporters are involved in the renal excretion of xanthine derivatives, 3-methylxanthie and enprofylline, remains unclear. In this study, we have investigated the effects of typically predominant substrates for organic anion and cation transporters on the tubular secretion of 3-methylxanthine and enprofylline in rats. In the renal clearance experiments using typical substrates for organic anion transporters, probenecid and p-aminohippurate, probenecid (20 mg/kg), but not p-aminohippurate (100 mg/kg), significantly decreased the renal clearance and clearance ratio of 3-methylxanthine and enprofylline. The typical substrates for organic cation transport systems, tetraethylammonium (30.6 mg/kg) and cimetidine (50 or 100 mg/kg), significantly decreased the renal clearance and clearance ratio of 3-methylxanthine and enprofylline. These results suggest that the renal secretory transport of 3-methylxanthine and enprofylline are mediated by probenecid-, cimetidine- and tetraethylammonium-sensitive transport systems. Uric acid, an organic anion, significantly inhibited the renal secretion of 3-methylxanthine, but not enprofylline, suggesting that the renal tubular transport of 3-methylxanthine is also mediated via uric acid-sensitive transport system. These findings suggest the possibility that both organic anion and cation transporters are, at least, involved in the renal tubular transport of 3-methylxanthine and enprofylline in rats.


Assuntos
Rim/metabolismo , Transportadores de Ânions Orgânicos/fisiologia , Proteínas de Transporte de Cátions Orgânicos/fisiologia , Xantinas/farmacocinética , Animais , Cimetidina/farmacologia , Rim/efeitos dos fármacos , Túbulos Renais/efeitos dos fármacos , Túbulos Renais/metabolismo , Masculino , Taxa de Depuração Metabólica , Transportadores de Ânions Orgânicos/biossíntese , Proteínas de Transporte de Cátions Orgânicos/biossíntese , Probenecid/farmacologia , Ratos , Ratos Wistar , Especificidade por Substrato , Tetraetilamônio/farmacologia , Fatores de Tempo , Xantinas/urina , Ácido p-Aminoipúrico/farmacologia
20.
Drug Test Anal ; 9(9): 1372-1384, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27662634

RESUMO

Methylxanthines positives in competition samples have challenged doping control laboratories and racing jurisdictions since methylxanthines are naturally occurring prohibited substances and often constituents of feed. For theobromine, an international threshold (renamed in International Residue Limit, IRL) of 2 µg/mL in urine has been established. On the basis of the data presented herein, a threshold or rather an IRL for theobromine in plasma of 0.3 µg/mL was proposed and was thereupon approved by the International Federation of Horseracing Authorities (IFHA). Official recommendations for reporting caffeine and theophylline are still lacking. The aim of the study was to investigate IRLs for theobromine in blood and for caffeine and theophylline in blood and urine. Therefore, a set of six administrations were carried out including both single i.v. and single oral administrations of caffeine, theobromine and theophylline. Plasma and urine concentrations were determined using a validated liquid chromatography-tandem mass spectrometry (LC-MS/MS). Applying the Toutain model approach an effective plasma concentration (EPC) of caffeine was estimated at 3.05 µg/mL, irrelevant concentrations in blood (IPC) and urine (IUC) approached 6 and 12 ng/mL, respectively. EPC of theobromine was calculated with 3.80 µg/mL, and irrelevant concentrations of theobromine were determined at 8 ng/mL in plasma and at 142 ng/mL in urine. Toutain modelling of the theophylline data produced an EPC, IPC, and IUC of 3.20 µg/mL, 6 ng/mL, and 75 ng/mL, respectively. The obtained irrelevant concentrations were used to postulate IRLs for theobromine in plasma and for caffeine and theophylline in plasma and urine. Copyright © 2016 John Wiley & Sons, Ltd.


Assuntos
Cafeína/química , Cafeína/farmacocinética , Teobromina/química , Teobromina/farmacocinética , Teofilina/química , Teofilina/farmacocinética , Xantinas/farmacocinética , Animais , Cafeína/análise , Cromatografia Líquida/métodos , Cavalos , Teobromina/análise , Teofilina/análise , Xantinas/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA