Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Stroke ; 30(7): 1472-7, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10390325

RESUMO

BACKGROUND AND PURPOSE: Evidence suggests that glutamate contributes to ischemic brain damage through activation of the alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionate (AMPA) receptor. We tested the novel, selective AMPA receptor antagonist PD152247 (PNQX) in a model of temporary focal ischemia to determine the dose-response relationship and to investigate the contribution of drug-induced hypothermia to the neuroprotective action of AMPA receptor antagonists. METHODS: Temporary focal cerebral ischemia was induced in Sprague-Dawley rats by occluding the middle cerebral artery and both carotid arteries for 3 hours. Body temperature was monitored by telemetry. PNQX was administered intraperitoneally or by intravenous infusion with various doses for 6 hours. Lesion volume was determined after 3 days by stereological methods. RESULTS: PNQX reduced the lesion volume by 51% after intraperitoneal administration. The intravenous dose-response study demonstrated that the lowest effective dose of PNQX was 1.0 mg/kg per hour, which corresponded to a steady state plasma level of 685 ng/mL. Neuroprotection was demonstrated at PNQX plasma concentrations that did not lower body temperature over the entire course of the experiment. CONCLUSIONS: AMPA receptor activation plays an important role in the development of ischemic brain damage. Thus, novel AMPA receptor antagonists may be useful for the treatment of stroke in humans.


Assuntos
Ataque Isquêmico Transitório/tratamento farmacológico , Ataque Isquêmico Transitório/prevenção & controle , Fármacos Neuroprotetores/uso terapêutico , Quinoxalinas/uso terapêutico , Receptores de AMPA/antagonistas & inibidores , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Hipotermia Induzida , Infusões Intravenosas , Masculino , Ratos , Ratos Sprague-Dawley
2.
Epilepsy Res ; 29(3): 233-49, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9551785

RESUMO

Although the cellular mechanisms of pharmacological actions of gabapentin (Neurontin) remain incompletely described, several hypotheses have been proposed. It is possible that different mechanisms account for anticonvulsant, antinociceptive, anxiolytic and neuroprotective activity in animal models. Gabapentin is an amino acid, with a mechanism that differs from those of other anticonvulsant drugs such as phenytoin, carbamazepine or valproate. Radiotracer studies with [14C]gabapentin suggest that gabapentin is rapidly accessible to brain cell cytosol. Several hypotheses of cellular mechanisms have been proposed to explain the pharmacology of gabapentin: 1. Gabapentin crosses several membrane barriers in the body via a specific amino acid transporter (system L) and competes with leucine, isoleucine, valine and phenylalanine for transport. 2. Gabapentin increases the concentration and probably the rate of synthesis of GABA in brain, which may enhance non-vesicular GABA release during seizures. 3. Gabapentin binds with high affinity to a novel binding site in brain tissues that is associated with an auxiliary subunit of voltage-sensitive Ca2+ channels. Recent electrophysiology results suggest that gabapentin may modulate certain types of Ca2+ current. 4. Gabapentin reduces the release of several monoamine neurotransmitters. 5. Electrophysiology suggests that gabapentin inhibits voltage-activated Na+ channels, but other results contradict these findings. 6. Gabapentin increases serotonin concentrations in human whole blood, which may be relevant to neurobehavioral actions. 7. Gabapentin prevents neuronal death in several models including those designed to mimic amyotrophic lateral sclerosis (ALS). This may occur by inhibition of glutamate synthesis by branched-chain amino acid aminotransferase (BCAA-t).


Assuntos
Acetatos/farmacologia , Acetatos/uso terapêutico , Aminas , Ácidos Cicloexanocarboxílicos , Ácido gama-Aminobutírico/metabolismo , Acetatos/farmacocinética , Analgésicos/farmacologia , Analgésicos/uso terapêutico , Animais , Ansiolíticos/administração & dosagem , Ansiolíticos/uso terapêutico , Anticonvulsivantes/farmacologia , Anticonvulsivantes/uso terapêutico , Encéfalo/efeitos dos fármacos , Encéfalo/fisiologia , Canais de Cálcio/química , Canais de Cálcio/efeitos dos fármacos , Canais de Cálcio/fisiologia , Gabapentina , Humanos , Modelos Neurológicos , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Neurotransmissores/fisiologia , Dor , Canais de Sódio/fisiologia , Sinapses/efeitos dos fármacos , Sinapses/fisiologia , Distribuição Tecidual
3.
Neuron ; 16(3): 675-86, 1996 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8785064

RESUMO

Three glutamate transporters have been identified in rat, including astroglial transporters GLAST and GLT-1 and a neuronal transporter EAAC1. Here we demonstrate that inhibition of the synthesis of each glutamate transporter subtype using chronic antisense oligonucleotide administration, in vitro and in vivo, selectively and specifically reduced the protein expression and function of glutamate transporters. The loss of glial glutamate transporters GLAST or GLT-1 produced elevated extracellular glutamate levels, neurodegeneration characteristic of excitotoxicity, and a progressive paralysis. The loss of the neuronal glutamate transporter EAAC1 did not elevate extracellular glutamate in the striatum but did produce mild neurotoxicity and resulted in epilepsy. These studies suggest that glial glutamate transporters provide the majority of functional glutamate transport and are essential for maintaining low extracellular glutamate and for preventing chronic glutamate neurotoxicity.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Astrócitos/metabolismo , Glutamatos/metabolismo , Oligonucleotídeos Antissenso/metabolismo , Medula Espinal/metabolismo , Sistema X-AG de Transporte de Aminoácidos , Animais , Células Cultivadas , Expressão Gênica , Hipocampo/metabolismo , Imuno-Histoquímica , Masculino , Microscopia Eletrônica , Ratos , Ratos Sprague-Dawley
4.
Pharm Res ; 13(3): 398-403, 1996 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8692732

RESUMO

PURPOSE: To determine the apparent bidirectional permeabilities of gabapentin (GBP) across the blood-brain barrier (BBB) using a novel microdialysis-pharmacokinetic approach. METHODS: Rats were administered intravenous infusions of [14C]GBP to achieve clinically relevant steady-state plasma concentrations. Microdialysis was used to monitor GBP concentration in brain extracellular fluid (ECF) in conscious animals. Brain tissue GBP concentration was measured at termination. The BBB influx (CL1) and efflux (CL2) permeabilities of GBP were estimated with a hybrid pharmacokinetic model assuming that transport between intra- and extracellular space was more rapid than transport across the BBB. The time course of GBP concentration in brain tissue was determined independently to validate the model assumption. RESULTS AND CONCLUSIONS: Simulations of the concentration-time course of GBP in brain tissue based on this modeling correlated well with the time-course of brain tissue concentrations determined after intravenous bolus administration and validated this pharmacokinetic-microdialysis approach for estimation of BBB permeabilities. The values for CL1 and CL2 were 0.042 (0.017) and 0.36 (0.16) ml/min.g-brain, respectively, indicating that GBP was more efficiently transported from brain ECF to plasma. The total brain tissue concentration of GBP was significantly higher than the ECF concentration at steady-state due to intracellular accumulation and tissue binding, that if not considered, will lead to underestimated efflux BBB permeability using the tissue homogenate-pharmacokinetic approach.


Assuntos
Acetatos/farmacocinética , Aminas , Anticonvulsivantes/farmacocinética , Barreira Hematoencefálica/fisiologia , Ácidos Cicloexanocarboxílicos , Ácido gama-Aminobutírico , Animais , Encéfalo/metabolismo , Química Encefálica , Diálise , Gabapentina , Masculino , Permeabilidade , Ratos , Ratos Sprague-Dawley , Reprodutibilidade dos Testes
5.
Ann Pharmacother ; 29(11): 1164-7, 1995 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8573965

RESUMO

OBJECTIVE: To provide the biochemical rationale for the use of the new anticonvulsant agent gabapentin as a treatment for amyotrophic lateral sclerosis (ALS). BACKGROUND: ALS is a neuropathologic disorder of the central nervous system characterized by a progressive loss of upper and lower motor neurons. Although the etiopathology of ALS is incompletely known, it is hypothesized that glutamatergic neurotransmission is related to neuropathology. Glutamate is an excitatory amino acid neurotransmitter that is cytotoxic when overexpressed at synaptic terminals, probably through a calcium-related mechanism. The concentration of glutamate in cerebrospinal fluid is increased in patients with ALS. The increased extracellular concentrations of glutamate may be caused by a decreased capacity of glutamate transport in brain tissue and/or abnormal glutamate metabolism. Recent success with the glutamate release inhibitor riluzole in well-controlled clinical trials supports the excitotoxic mechanism of neuropathology in patients with ALS. POTENTIAL TREATMENT FOR ALS: Gabapentin has demonstrated neuroprotective properties in a model of chronic glutamate toxicity in vitro. Although the neuroprotective mechanism of action of gabapentin is currently unknown, it is hypothesized here that gabapentin decreases the rate of formation of glutamate derived from the branched-chain amino acids (BCAAs) leucine, isoleucine, and valine. The proposed decrease in formation of glutamate from BCAAs may decrease the pool of releasable glutamate and therefore compensate for diminished glutamate uptake capacity and/or abnormal glutamate metabolism in patients with ALS. CONCLUSIONS: Based on this rationale, it is proposed that gabapentin may provide a beneficial effect in the treatment of patients with ALS.


Assuntos
Acetatos/uso terapêutico , Aminas , Esclerose Lateral Amiotrófica/tratamento farmacológico , Anticonvulsivantes/uso terapêutico , Ácidos Cicloexanocarboxílicos , Ácido gama-Aminobutírico , Acetatos/farmacologia , Aminoácidos de Cadeia Ramificada/metabolismo , Esclerose Lateral Amiotrófica/etiologia , Esclerose Lateral Amiotrófica/metabolismo , Anticonvulsivantes/farmacologia , Encéfalo/metabolismo , Antagonistas de Aminoácidos Excitatórios/farmacologia , Gabapentina , Glutamatos/metabolismo , Humanos
6.
Epilepsy Res ; 22(1): 1-11, 1995 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8565962

RESUMO

Gabapentin is a novel anticonvulsant drug. The anticonvulsant mechanism of gabapentin is not known. Based on the amino acid structure of gabapentin we explored its possible effects on glutamate and gamma-aminobutyric acid (GABA) metabolism in brain as they may relate to its anticonvulsant mechanisms of action. Gabapentin was tested for its effects on seven enzymes in the metabolic pathways of these two neurotransmitters: alanine aminotransferase (AL-T), aspartate aminotransferase (AS-T), GABA aminotransferase (GABA-T), branched-chain amino acid aminotransferase (BCAA-T), glutamine synthetase (Gln-S), glutaminase (GLNase), and glutamate dehydrogenase (GDH). In the presence of 10 mM gabapentin, only GABA-T, BCAA-T, and GDH activities were affected by this drug. Inhibition of GABA-T by gabapentin was weak (33%). The Ki values for inhibition of cytosolic and mitochondrial forms of GABA-T (17-20 mM) were much higher than the Km values for GABA (1.5-1.9 mM). It is, therefore, unlikely that inhibition of GABA-T by gabapentin is clinically relevant. As with leucine, gabapentin stimulated GDH activity. The GDH activity in rat brain synaptosomes was activated 6-fold and 3.4-fold, respectively, at saturating concentrations (10 mM) of leucine and gabapentin. The half-maximal stimulation by gabapentin was observed at approximately 1.5 mM. Gabapentin is not a substrate of BCAA-T, but it exhibited a potent competitive inhibition of both cytosolic and mitochondrial forms of brain BCAA-T. Inhibition of BCAA-T by this drug was reversible. The Ki values (0.8-1.4 mM) for inhibition of transamination by gabapentin were close to the apparent Km values for the branched-chain amino acids (BCAA) L-leucine, L-isoleucine, and L-valine (0.6-1.2 mM), suggesting that gabapentin may significantly reduce synthesis of glutamate from BCAA in brain by acting on BCAA-T.


Assuntos
Acetatos/farmacologia , Aminas , Anticonvulsivantes/farmacologia , Ácidos Cicloexanocarboxílicos , Ácido Glutâmico/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Citosol/efeitos dos fármacos , Citosol/enzimologia , Gabapentina , Glutamato Desidrogenase/antagonistas & inibidores , Glutamato Desidrogenase/metabolismo , Glutamato-Amônia Ligase/antagonistas & inibidores , Glutamato-Amônia Ligase/metabolismo , Glutaminase/antagonistas & inibidores , Glutaminase/metabolismo , Técnicas In Vitro , Cinética , Masculino , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/enzimologia , Ratos , Ratos Sprague-Dawley , Sinaptossomos/efeitos dos fármacos , Sinaptossomos/enzimologia , Transaminases/antagonistas & inibidores , Transaminases/metabolismo
7.
Fundam Appl Toxicol ; 26(2): 272-81, 1995 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-7589916

RESUMO

PD 138142-15 is a substituted urea hypolipidemic and potential anti-atherosclerotic agent. To determine the toxicity of PD 138142-15, beagle dogs were given oral doses of 1, 10, 30, and 100 mg/kg daily for 13 weeks. Two animals at 100 mg/kg were euthanized during Week 5 due to poor condition. Clinical findings included decreased serum albumin at > or = 30 mg/kg, and increased ALP (up to 30-fold) and 5'-nucleotidase activities (up to 9-fold) at doses > or = 10 mg/kg. ALT and AST activities were elevated only at 100 mg/kg. There was a two- to threefold increase in cytochrome P450 content of hepatic microsomes from all treated animals and increases in liver weights at 10 mg/kg and above. Hepatic changes included hepatocellular hypertrophy and increased cytoplasmic eosinophilia at > or = 10 mg/kg; single cell necrosis of hepatocytes was noted in moribund animals. ACTH-stimulated cortisol levels were decreased at 30 and 100 mg/kg. Adrenal cholesterol esters were decreased at 10 mg/kg and above, while total adrenal cholesterol was decreased at > or = 30 mg/kg. These changes correlated with adrenal cortical zonal atrophy, principally of the zona fasciculata and zona reticularis, present at 30 and 100 mg/kg. Plasma concentrations of PD 131842-15 increased with increasing dose; plasma levels were significantly lower during Week 12 than those on Day 1, possibly due to autoinduction. Overt hepatotoxicity occurred at 100 mg/kg, whereas hepatic changes at 10 and 30 mg/kg were consistent with cytochrome P450 induction. The hepatic lesions were reversible within 4 weeks, while adrenal lesions were still evident after 4 weeks without treatment.


Assuntos
Glândulas Suprarrenais/efeitos dos fármacos , Carbamatos/toxicidade , Hipolipemiantes/toxicidade , Fígado/efeitos dos fármacos , Esterol O-Aciltransferase/antagonistas & inibidores , Administração Oral , Glândulas Suprarrenais/patologia , Animais , Carbamatos/sangue , Sistema Enzimático do Citocromo P-450/metabolismo , Cães , Feminino , Hipolipemiantes/sangue , Fígado/enzimologia , Fígado/patologia , Masculino
8.
Biochem Pharmacol ; 49(6): 799-808, 1995 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-7702638

RESUMO

Induction of hepatic microsomal cytochrome P450 produced by carbamic acid [2,6-bis(1-methylethyl)phenoxy]sulfonyl]-2,6-bis(1-methylethyl) phenyl ester, monosodium salt (PD138142-15), a novel water-soluble inhibitor of acyl-CoA: cholesterol acyltransferase, was examined in male and female rats, dogs, and monkeys, and in male guinea pigs. Relative to control, PD138142-15 increased hepatic microsomal total spectral P450 in all species examined. Hepatic microsomal ethoxyresorufin-O-deethylase, pentoxyresorufin-O-dealkylase, and peroxisomal carnitine acetyltransferase activities and cyanide-insensitive Beta-oxidation were affected only marginally. Erythromycin-N-demethylase activity was increased (2- to 6-fold) in all three species in which it was examined (rat, dog and pig). Marked increases in immunoreactive P450 3A were noted in the rats and dogs, while slight increases were seen in monkeys. Pharmacokinetic studies of PD138142-15 in rats and dogs revealed pronounced decreases (80-90%) in plasma Cmax and AUC within 2 weeks of initiation of daily dosing. In spite of the marked decline in plasma drug levels, efficacy in dogs, as determined by serum cholesterol levels, was maintained for up to 6 weeks with continued dosing. Potential acid (gastric) breakdown products of PD 138142-15 were examined for their hepatic cytochrome P450 induction profiles in rats adn were found to differ both quantitatively and qualitatively from profiles produced by the parent compound. This suggested that induction observed in rats was due to parent PD138142-15 and not to any of the known potential acid breakdown products. The cumulative data establish that PD 138142-15 is an inducer of P450 3A in rats and dogs. The results also suggest that P450 3A is induced in monkeys and pigs as well, although the data are less definitive. Decreases in plasma drug levels imply that the compound may be an autoinducer in dogs and rats. The maintenance of efficacy in spite of decreased drugs levels in dogs suggests that the effects on serum cholesterol are due to a metabolite or that cholesterol lowering effects occur before the compound is metabolized by the liver.


Assuntos
Carbamatos/farmacologia , Sistema Enzimático do Citocromo P-450/biossíntese , Hipolipemiantes/farmacologia , Microssomos Hepáticos/efeitos dos fármacos , Animais , Carbamatos/farmacocinética , Colesterol/sangue , Cães , Indução Enzimática , Feminino , Cobaias , Hipolipemiantes/farmacocinética , Macaca fascicularis , Masculino , Microssomos Hepáticos/enzimologia , Ratos , Especificidade da Espécie , Esterol O-Aciltransferase/antagonistas & inibidores
9.
J Clin Pharmacol ; 34(10): 985-8, 1994 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-7836549

RESUMO

A four-way cross-over study was performed to assess the temporal effect of food on the rate and extent of tacrine (Cognex, THA) absorption after drug administration to healthy, older volunteers. Each volunteer received four single 40-mg THA doses at 1-week intervals. Doses were administered after an 8-hour overnight fast, 1 hour before a standard breakfast, 15 minutes after beginning a standard breakfast, and 2 hours after completion of a standard breakfast. Gastrointestinal side effects were most frequently reported after drug administration to fasted subjects. Mean Cmax and AUC(0-infinity) values after THA administration during breakfast (9.9 ng/mL and 70.2 ng.hr/mL) and 2 hours after breakfast (11.6 ng/mL and 74.2 ng.hour-1.mL-1) were significantly lower than values determined after administration of THA to fasting subjects (15.8 ng/mL, and 91.8 ng.hour-1.mL-1). Little effect was evident when THA was administered 1 hour before breakfast.


Assuntos
Interações Alimento-Droga , Tacrina/farmacocinética , Idoso , Disponibilidade Biológica , Cromatografia Líquida de Alta Pressão , Estudos Cross-Over , Humanos , Absorção Intestinal , Pessoa de Meia-Idade , Tacrina/efeitos adversos , Tacrina/sangue
10.
Epilepsy Res ; 16(3): 175-81, 1993 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8119268

RESUMO

The concentration-time profile of [14C]gabapentin (GBP) in plasma and brain interstitial fluid (ISF) was determined following a single 15 mg/kg intravenous bolus dose to rats. Brain ISF was sampled with a microdialysis probe in striatum. Blood was also collected serially to 4 h postdose. At termination, brain was sectioned into regions and [14C]GBP concentrations were determined. Anticonvulsant effects were determined by maximal electroshock in rats with identical dosing. Plasma [14C]GBP declined linearly after dosing while brain ISF [14C]GBP concentration peaked at approximately 1 h and then declined in parallel with plasma concentration. Throughout, brain ISF [14C]GBP concentration was approximately 3-6% of [14C]GBP concentration in plasma. However, at 4 h postdose, whole brain tissue [14C]GBP concentration was equal to or greater than the concentration of [14C]GBP in plasma. Maximal anticonvulsant effect lagged behind both plasma and brain ISF GBP concentrations. The anticonvulsant effect of GBP is delayed by time-dependent events other than distribution from blood to brain.


Assuntos
Acetatos/farmacocinética , Aminas , Anticonvulsivantes/farmacocinética , Encéfalo/metabolismo , Ácidos Cicloexanocarboxílicos , Ácido gama-Aminobutírico , Acetatos/sangue , Acetatos/farmacologia , Animais , Anticonvulsivantes/sangue , Anticonvulsivantes/farmacologia , Gabapentina , Masculino , Microdiálise , Ratos , Ratos Sprague-Dawley
11.
J Pharm Biomed Anal ; 8(4): 365-72, 1990.
Artigo em Inglês | MEDLINE | ID: mdl-2100211

RESUMO

The "product enantioselectivity" associated with the urinary excretion of the phenobarbital N-glucoside conjugates has not been determined previously. A liquid chromatography method using gradient elution was developed for quantifying both phenobarbital N-glucoside conjugates, phenobarbital, and p-hydroxyphenobarbital. Following a single oral dose of phenobarbital to male Caucasian and Oriental subjects, both phenobarbital N-glucoside conjugates were observed in the urine. In seven subjects, 3.3-10.6% of the phenobarbital dose was detected as a single phenobarbital N-glucoside (S configuration at the C-5 position of the barbiturate ring). The other phenobarbital N-glucoside diastereomer accounted for less than 1.5% of the phenobarbital dose. The urinary excretion of the major phenobarbital N-glucoside diastereomer paralleled the urinary excretion of phenobarbital and was comparable in both Caucasian and Oriental subjects. These results indicate a pronounced selectivity for the formation and/or urinary excretion of the phenobarbital N-glucosides.


Assuntos
Fenobarbital/análogos & derivados , Povo Asiático , Cromatografia Líquida/métodos , Humanos , Fenobarbital/farmacocinética , Fenobarbital/urina , Estereoisomerismo , População Branca
12.
J Clin Endocrinol Metab ; 69(5): 1040-6, 1989 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-2529264

RESUMO

We previously demonstrated a progressive decline in serum dehydroepiandrosterone sulfate (DHEA-S) levels in women during a hyperinsulinemic-euglycemic clamp. To determine whether this fall in serum DHEA-S levels might have been due to insulin-stimulated 1) hydrolysis of DHEA-S to dehydroepiandrosterone (DHEA), 2) conversion of DHEA-S/DHEA to androstenedione, and/or 3) urinary excretion of these steroids, 10 additional men were studied by the hyperinsulinemic-euglycemic clamp technique. Each man received a 0.1 U/kg (0.72 nmol/kg) insulin bolus dose, followed by a 10 mU/kg.min (72 pmol/kg.min) insulin infusion for 4 h. An average insulin level of 12,390 +/- 259 (+/- SE) pmol/L (1,726.8 +/- 36 microU/mL) was achieved; serum glucose was maintained at 5.0 +/- 0.1 mmol/L (90.5 +/- 2.3 mg/dL). During the hyperinsulinemia, serum DHEA-S levels fell progressively and were significantly lower than baseline at 4 and 6 h of study (85.5 +/- 5.9% and 79.1 +/- 3.2% of baseline values, respectively; P less than 0.05). Serum DHEA levels fell concurrently and were significantly lower than baseline at 2, 4, and 6 h of study (66.2 +/- 12.3%, 61.6 +/- 11.2%, and 52.9 +/- 10.2% of baseline values, respectively; P less than 0.05). The percent fall in serum DHEA levels correlated positively with the percent fall in serum DHEA-S levels (r = 0.44; P less than 0.02). Serum androstenedione levels also fell progressively during hyperinsulinemia and were significantly lower than baseline at 2, 4, and 6 h of study (71.5 +/- 4.1%, 71.0 +/- 7.2%, and 48.1 +/- 3.3% of baseline values, respectively; P less than 0.05). No change in serum DHEA-S, DHEA, or androstenedione levels occurred in paired control studies, during which 0.45% saline was infused at rates matched exactly to the rates of the dextrose and insulin infusions during the hyperinsulinemic clamp studies. Despite decreasing serum DHEA-S and DHEA levels during hyperinsulinemia, urinary DHEA-S and DHEA glucuronide excretions were increased by 50% (P less than 0.05) and 86% (P = 0.05), respectively, compared to urinary excretion of these steroids during control studies. In contrast, urinary excretion of unconjugated DHEA was unchanged. Quantitatively, however, increased urinary excretion of conjugated DHEA during hyperinsulinemia accounted for only about 5% of the concomitant fall in serum DHEA-S concentrations.(ABSTRACT TRUNCATED AT 400 WORDS)


Assuntos
Desidroepiandrosterona/análogos & derivados , Insulina/farmacologia , Adulto , Androstenodiona/metabolismo , Creatinina/urina , Desidroepiandrosterona/metabolismo , Desidroepiandrosterona/urina , Sulfato de Desidroepiandrosterona , Glucuronatos/urina , Humanos , Hiperinsulinismo/metabolismo , Insulina/administração & dosagem , Sistemas de Infusão de Insulina , Masculino , Cloreto de Sódio/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...