Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Respir Res ; 16: 122, 2015 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-26438053

RESUMO

BACKGROUND: Poor interferon gamma (IFNγ) production during respiratory syncytial virus (RSV) is associated with prolonged viral clearance and increased disease severity in neonatal mice and humans. We previously showed that intra-nasal delivery of IFNγ significantly enhances RSV clearance from neonatal lungs prior to observed T-lymphocyte recruitment or activation, suggesting an innate immune mechanism of viral clearance. We further showed that alveolar macrophages dominate the RSV-infected neonatal airways relative to adults, consistent with human neonatal autopsy data. Therefore, the goal of this work was to determine the role of neonatal alveolar macrophages in IFNγ-mediated RSV clearance. METHODS: Clodronate liposomes, flow cytometry, viral plaque assays, and histology were used to examine the role of alveolar macrophages (AMs) and the effects of intra-nasal IFNγ in RSV infected neonatal Balb/c mice. The functional outcomes of AM depletion were determined quantitatively by viral titers using plaque assay. Illness was assessed by measuring reduced weight gain. RESULTS: AM activation during RSV infection was age-dependent and correlated tightly with IFNγ exposure. Higher doses of IFNγ more efficiently stimulated AM activation and expedited RSV clearance without significantly affecting weight gain. The presence of AMs were independently associated with improved RSV clearance, whereas AM depletion but not IFNγ exposure, significantly impaired weight gain in RSV-infected neonates. CONCLUSION: We show here for the first time, that IFNγ is critical for neonatal RSV clearance and that it depends, in part, on alveolar macrophages (AMs) for efficient viral clearing effects. Early reductions in viral burden are likely to have profound short- and long-term immune effects in the vulnerable post-natally developing lung environment. Studies are ongoing to elucidate the pathologic effects associated with early versus delayed RSV clearance in developing neonatal airways.


Assuntos
Antivirais/administração & dosagem , Interferon gama/administração & dosagem , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos Alveolares/efeitos dos fármacos , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Vírus Sinciciais Respiratórios/efeitos dos fármacos , Administração Intranasal , Fatores Etários , Animais , Animais Recém-Nascidos , Antivirais/farmacocinética , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Interferon gama/farmacocinética , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/virologia , Masculino , Camundongos Endogâmicos BALB C , Camundongos Knockout , Receptores de Interferon/deficiência , Receptores de Interferon/genética , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/metabolismo , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sinciciais Respiratórios/imunologia , Vírus Sinciciais Respiratórios/patogenicidade , Fatores de Tempo , Carga Viral , Aumento de Peso , Receptor de Interferon gama
2.
Synapse ; 68(12): 613-623, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25088028

RESUMO

The serotonin subtype-4 (5-HT4 ) receptor, which is known to be involved physiologically in learning and memory, and pathologically in Alzheimer's disease, anxiety, and other neuropsychiatric disorders-has few radioligands readily available for imaging in vivo. We have previously reported two novel 5-HT4 receptor radioligands, namely [methoxy-11 C](1-butylpiperidin-4-yl)methyl 4-amino-3-methoxybenzoate; [11 C]RX-1), and the [18 F]3-fluoromethoxy analog ([18 F]RX-2), and in this study we evaluated them by PET in rhesus monkey. Brain scans were performed at baseline, receptor preblock or displacement conditions using SB 207710, a 5-HT4 receptor antagonist, on the same day for [11 C]RX-1 and on different days for [18 F]RX-2. Specific-to-nondisplaceable ratio (BPND ) was measured with the simplified reference tissue model from all baseline scans. To determine specific binding, total distribution volume (VT ) was also measured in some monkeys by radiometabolite-corrected arterial input function after ex vivo inhibition of esterases from baseline and blocked scans. Both radioligands showed moderate to high peak brain uptake of radioactivity (2-6 SUV). Regional BPND values were in the rank order of known 5-HT4 receptor distribution with a trend for higher BPND values from [18 F]RX-2. One-tissue compartmental model provided good fits with well identified VT values for both radioligands. In the highest 5-HT4 receptor density region, striatum, 50-60% of total binding was specific. The VT in receptor-poor cerebellum reached stable values by about 60 min for both radioligands indicating little influence of radiometabolites on brain signal. In conclusion, both [11 C]RX-1 and [18 F]RX-2 showed positive attributes for PET imaging of brain 5-HT4 receptors, validating the radioligand design strategy. Synapse 68:613-623, 2014. © 2014 Wiley Periodicals, Inc.

3.
Crit Care Med ; 41(10): 2379-87, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23896831

RESUMO

OBJECTIVE: Preclinical and clinical studies have suggested that therapeutic hypothermia, while decreasing neurologic injury, may also lead to drug toxicity that may limit its benefit. Cooling decreases cytochrome P450 (CYP)-mediated drug metabolism, and limited clinical data suggest that drug levels are elevated. Fosphenytoin is metabolized by cytochrome P450 2C, has a narrow therapeutic range, and is a commonly used antiepileptic medication. The objective of this study was to evaluate the impact of therapeutic hypothermia on phenytoin levels and pharmacokinetics in children with severe traumatic brain injury. DESIGN: Pharmacokinetic analysis of subjects participating in a multicenter randomized phase III study of therapeutic hypothermia for severe traumatic brain injury. SETTING: ICU at the Children's Hospital of Pittsburgh. PATIENTS: Nineteen children with severe traumatic brain injury. INTERVENTIONS: None. MEASUREMENTS AND MAIN RESULTS: A sum of 121 total and 114 free phenytoin levels were evaluated retrospectively in 10 hypothermia-treated and nine normothermia-treated children who were randomized to 48 hours of cooling to 32-33°C followed by slow rewarming or controlled normothermia. Drug dosing, body temperatures, and demographics were collected during cooling, rewarming, and posttreatment periods (8 d). A trend toward elevated free phenytoin levels in the hypothermia group (p=0.051) to a median of 2.2 mg/L during rewarming was observed and was not explained by dosing differences. Nonlinear mixed-effects modeling incorporating both free and total levels demonstrated that therapeutic hypothermia specifically decreased the time-variant component of the maximum velocity of phenytoin metabolism (Vmax) 4.6-fold (11.6-2.53 mg/hr) and reduced the overall Vmax by ~50%. Simulations showed that the increased risk for drug toxicity extends many days beyond the end of the cooling period. CONCLUSIONS: Therapeutic hypothermia significantly reduces phenytoin elimination in children with severe traumatic brain injury leading to increased drug levels for an extended period of time after cooling. Pharmacokinetic interactions between hypothermia and medications should be considered when caring for children receiving this therapy.


Assuntos
Anticonvulsivantes/farmacocinética , Lesões Encefálicas/metabolismo , Hipotermia Induzida/métodos , Fenitoína/farmacocinética , Adolescente , Anticonvulsivantes/sangue , Lesões Encefálicas/terapia , Criança , Pré-Escolar , Feminino , Humanos , Hipotermia Induzida/efeitos adversos , Unidades de Terapia Intensiva Pediátrica , Masculino , Pennsylvania , Fenitoína/sangue , Estudos Retrospectivos , Índices de Gravidade do Trauma
4.
Artigo em Inglês | MEDLINE | ID: mdl-28111064

RESUMO

A rapid and sensitive method using ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) was developed to simultaneously quantify hydroxyeicosatetraenoic (HETE), dihydroxyeicosatrienoic (DiHETrE), epoxyeicosatrienoic acid (EET), and prostaglandin metabolites of arachidonic acid in human plasma. Sample preparation consisted of solid phase extraction with Oasis HLB (30mg) cartridges for all metabolites. Separation of HETEs, EETs, and DiHETrEs was achieved on an Acquity UPLC BEH C18, 1.7µm (100×2.1mm) reversed-phase column (Waters Corp, Millford, MA) with negative electrospray ionization mass spectrometric detection. A second injection of the same extracted sample allowed for separation and assessment of prostaglandin metabolites under optimized UPLC-MS/MS conditions. Additionally, the endogenous levels of these metabolites in five different matrices were determined in order to select the optimal matrix for assay development. Human serum albumin was shown to have the least amount of endogenous metabolites, a recovery efficiency of 79-100% and a matrix effect of 71 - 100%. Linear calibration curves ranging from 0.416 to 66.67ng/ml were validated. Inter-assay and intra-assay variance was less than 15% at most concentrations. This method was successfully applied to quantify metabolite levels in plasma samples of healthy control subjects receiving niacin administration to evaluate the association between niacin administration and eicosanoid plasma level response.


Assuntos
Cromatografia Líquida/métodos , Eicosanoides/sangue , Niacina/administração & dosagem , Espectrometria de Massas em Tandem/métodos , Voluntários Saudáveis , Humanos , Albumina Sérica Humana/metabolismo , Extração em Fase Sólida
5.
Ther Hypothermia Temp Manag ; 6(4): 169-179, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27622966

RESUMO

Targeted temperature management (TTM) has been shown to reduce mortality and improve neurological outcomes in out-of-hospital cardiac arrest (CA) patients and in neonates with hypoxic-ischemic encephalopathy (HIE). TTM has also been associated with adverse drug events in the critically ill patient due to its effect on drug pharmacokinetics (PKs) and pharmacodynamics (PDs). We aim to evaluate the current literature on the effect of TTM on drug PKs and PDs following CA. MEDLINE/PubMed databases were searched for publications, which include the MeSH terms hypothermia, drug metabolism, drug transport, P450, critical care, cardiac arrest, hypoxic-ischemic encephalopathy, pharmacokinetics, and pharmacodynamics between July 2006 and October 2015. Twenty-three studies were included in this review. The studies demonstrate that hypothermia impacts PK parameters and increases concentrations of cytochrome-P450-metabolized drugs in the cooling and rewarming phase. Furthermore, the current data demonstrate a combined effect of CA and hypothermia on drug PK. Importantly, these effects can last greater than 4-5 days post-treatment. Limited evidence suggests hypothermia-mediated changes in the Phase II metabolism and the Phase III transport of drugs. Hypothermia also has been shown to potentially decrease the effect of specific drugs at the receptor level. Therapeutic hypothermia, as commonly deployed/applied during TTM, alters PK, and elevates concentrations of several commonly used medications. Hypothermia-mediated effects are an important factor when dosing and monitoring patients undergoing TTM treatment.


Assuntos
Anti-Infecciosos/farmacocinética , Anticonvulsivantes/farmacocinética , Regulação da Temperatura Corporal , Parada Cardíaca/terapia , Hipnóticos e Sedativos/farmacocinética , Hipotermia Induzida/métodos , Inibidores da Agregação Plaquetária/farmacocinética , Animais , Anti-Infecciosos/administração & dosagem , Anti-Infecciosos/efeitos adversos , Anticonvulsivantes/administração & dosagem , Anticonvulsivantes/efeitos adversos , Disponibilidade Biológica , Cálculos da Dosagem de Medicamento , Interações Medicamentosas , Parada Cardíaca/metabolismo , Parada Cardíaca/fisiopatologia , Humanos , Hipnóticos e Sedativos/administração & dosagem , Hipnóticos e Sedativos/efeitos adversos , Hipotermia Induzida/efeitos adversos , Inibidores da Agregação Plaquetária/administração & dosagem , Inibidores da Agregação Plaquetária/efeitos adversos , Reaquecimento , Fatores de Risco , Resultado do Tratamento
6.
Am J Pharm Educ ; 80(1): 3, 2016 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-26941429

RESUMO

OBJECTIVE: To develop, implement, and evaluate "Test2Learn" a program to enhance pharmacogenomics education through the use of personal genomic testing (PGT) and real genetic data. DESIGN: One hundred twenty-two second-year doctor of pharmacy (PharmD) students in a required course were offered PGT as part of a larger program approach to teach pharmacogenomics within a robust ethical framework. The program added novel learning objectives, lecture materials, analysis tools, and exercises using individual-level and population-level genetic data. Outcomes were assessed with objective measures and pre/post survey instruments. ASSESSMENT: One hundred students (82%) underwent PGT. Knowledge significantly improved on multiple assessments. Genotyped students reported a greater increase in confidence in understanding test results by the end of the course. Similarly, undergoing PGT improved student's self-perceived ability to empathize with patients compared to those not genotyped. Most students (71%) reported feeling PGT was an important part of the course, and 60% reported they had a better understanding of pharmacogenomics specifically because of the opportunity. CONCLUSION: Implementation of PGT in the core pharmacy curriculum was feasible, well-received, and enhanced student learning of pharmacogenomics.


Assuntos
Educação de Pós-Graduação em Farmácia/métodos , Testes Genéticos , Genômica/educação , Farmacogenética/educação , Estudantes de Farmácia , Currículo , Humanos , Aprendizagem
7.
PLoS One ; 10(8): e0129709, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26237219

RESUMO

BACKGROUND AND PURPOSE: Reducing body temperature can prolong tolerance to ischemic injury such as stroke or myocardial infarction, but is difficult and uncomfortable in awake patients because of shivering. We tested the efficacy and safety of the alpha-2-adrenergic agonist dexmedetomidine for suppressing shivering induced by a rapid infusion of cold intravenous fluids. METHODS: Ten subjects received a rapid intravenous infusion of two liters of cold (4°C) isotonic saline on two separate test days, and we measured their core body temperature, shivering, hemodynamics and sedation for two hours. On one test day, fluid infusion was preceded by placebo infusion. On the other test day, fluid infusion was preceded by 1.0 µg/kg bolus of dexmedetomidine over 10 minutes. RESULTS: All ten subjects experienced shivering on placebo days, with shivering beginning at a mean (SD) temperature of 36.6 (0.3)°C. The mean lowest temperature after placebo was 36.0 (0.3)°C (range 35.7-36.5°C). Only 3/10 subjects shivered on dexmedetomidine days, and the mean lowest temperature was 35.7 (0.4)°C (range 35.0-36.3°C). Temperature remained below 36°C for the full two hours in 6/10 subjects. After dexmedetomidine, subjects had moderate sedation and a mean 26 (13) mmHg reduction in blood pressure that resolved within 90 minutes. Heart rate declined a mean 23 (11) bpm after both placebo and dexmedetomidine. Dexmedetomidine produced no respiratory depression. CONCLUSION: Dexmedetomidine decreases shivering in normal volunteers. This effect is associated with decreased systolic blood pressure and sedation, but no respiratory depression.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/uso terapêutico , Dexmedetomidina/uso terapêutico , Hipnóticos e Sedativos/uso terapêutico , Hipotermia Induzida , Estremecimento/efeitos dos fármacos , Agonistas de Receptores Adrenérgicos alfa 2/efeitos adversos , Adulto , Pressão Sanguínea/efeitos dos fármacos , Temperatura Corporal/efeitos dos fármacos , Dexmedetomidina/efeitos adversos , Feminino , Frequência Cardíaca/efeitos dos fármacos , Humanos , Hipnóticos e Sedativos/efeitos adversos , Hipotermia Induzida/métodos , Masculino , Adulto Jovem
8.
J Pharm Sci ; 101(3): 1028-39, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22170327

RESUMO

Drug interactions with plasma proteins influence their pharmacokinetics and pharmacodynamics. We aimed to test whether a strong quantitative relationship exists between plasma free fraction (f(P) ) and lipophilicity for low molecular weight nonacidic drug-like compounds. We measured the n-octanol-buffer distribution coefficients at pH 7.4 ((m) logD) of 18 diverse radiotracers (<470 Da) used for brain imaging with positron emission tomography in vivo. Lipophilicities were also computed as (c) logD with two software packages. The f(P) values for monkeys and humans were determined by ultrafiltration and transformed into m logD(pr/pl) values representing the log(10) of the within phase partition of the radiotracers between plasma proteins and remaining plasma. (m) logD(pr/pl) correlated strongly with (m) logD for human ((m) logD(pr/pl) = 0.733(m) logD-0.780, r(2) = 0.74) and monkey ((m) logD(pr/pl) = 0.780(m) logD-1.15, r(2) = 0.83), but less strongly with (c) logD. These relationships were significantly different between species (P = 0.006). Removal of eight fluorinated compounds from the datasets raised r(2) to 0.81 and 0.91 for humans and monkeys, respectively. For the tested compounds, we conclude that n-octanol-buffer (pH 7.4) distribution strongly models that between plasma proteins and remaining plasma and moreover that (m) logD accounts for over 74% of compound (m) logD(pr/pl) and is a strong determinant of f(P).


Assuntos
Proteínas Sanguíneas/metabolismo , Preparações Farmacêuticas/química , Preparações Farmacêuticas/metabolismo , Compostos Radiofarmacêuticos/química , Compostos Radiofarmacêuticos/metabolismo , 1-Octanol/química , Adulto , Animais , Haplorrinos , Humanos , Lipídeos/química , Masculino , Plasma/metabolismo , Tomografia por Emissão de Pósitrons , Ligação Proteica
9.
Nucl Med Biol ; 39(8): 1128-36, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22898316

RESUMO

INTRODUCTION: Rhodamine-123 is a known substrate for the efflux transporter, P-glycoprotein (P-gp). We wished to assess whether rhodamine-123 might serve as a useful substrate for developing probes for imaging efflux transporters in vivo with positron emission tomography (PET). For this purpose, we aimed to label rhodamine-123 with carbon-11 (t(1/2)=20.4min) and to study its biodistribution in rodents. METHODS: [¹¹C]Rhodamine-123 was prepared by treating rhodamine-110 (desmethyl-rhodamine-123) with [¹¹C]methyl iodide. The biodistribution of this radiotracer was studied with PET in wild-type mice and rats, in efflux transporter knockout mice, in wild-type rats pretreated with DCPQ (an inhibitor of P-gp) or with cimetidine (an inhibitor of organic cation transporters; OCT), and in P-gp knockout mice pretreated with cimetidine. Unchanged radiotracer in forebrain, plasma and peripheral tissues was also measured ex vivo at 30min after radiotracer administration to wild-type and efflux transporter knockout rodents. RESULTS: [(¹¹C]Rhodamine-123 was obtained in 4.4% decay-corrected radiochemical yield from cyclotron-produced [¹¹C]carbon dioxide. After intravenous administration of [¹¹C]rhodamine-123 to wild-type rodents, PET and ex vivo measurements showed radioactivity uptake was very low in brain, but relatively high in some other organs such as heart, and especially liver and kidney. Inhibition of P-gp increased uptake in brain, heart, kidney and liver, but only by up to twofold. Secretion of radioactivity from kidney was markedly reduced by OCT knockout or pretreatment with cimetidine. CONCLUSIONS: [¹¹C]Rhodamine-123 was unpromising as a PET probe for P-gp function and appears to be a strong substrate of OCT in kidney. Cimetidine appears effective for blocking OCT in kidney in vivo.


Assuntos
Rodamina 123/síntese química , Rodamina 123/farmacocinética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/antagonistas & inibidores , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/deficiência , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Radioisótopos de Carbono , Técnicas de Química Sintética , Cimetidina/farmacologia , Dibenzocicloeptenos/farmacologia , Técnicas de Inativação de Genes , Rim/efeitos dos fármacos , Rim/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Camundongos , Proteínas de Transporte de Cátions Orgânicos/antagonistas & inibidores , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Tomografia por Emissão de Pósitrons , Quinolinas/farmacologia , Traçadores Radioativos , Ratos , Rodamina 123/metabolismo , Distribuição Tecidual
10.
Schizophr Res ; 141(2-3): 185-8, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22910406

RESUMO

This study sought to determine whether cannabinoid-1 (CB(1)) receptor binding was altered in the postmortem dorsolateral prefrontal cortex (DLPFC) of individuals with schizophrenia (schizophrenia; n=47) compared to controls (n=43). The CB(1) receptor inverse agonist radioligand [(3)H]MePPEP was used to measure specific binding to CB(1) receptors. The specific binding of [(3)H]MePPEP to CB(1) receptors was 20% higher in patients with schizophrenia than in controls. Power analyses suggested that 53 subjects per group would be needed to detect a similar difference in vivo with positron emission tomography (PET) and the structurally related inverse agonist radioligand [(18)F]FMPEP-d(2) (80% statistical power, p<0.05).


Assuntos
Córtex Pré-Frontal/diagnóstico por imagem , Receptor CB1 de Canabinoide/metabolismo , Esquizofrenia/diagnóstico por imagem , Esquizofrenia/patologia , Adulto , Idoso , Estudos de Coortes , Feminino , Radioisótopos de Flúor/farmacocinética , Humanos , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único/genética , Tomografia por Emissão de Pósitrons , Córtex Pré-Frontal/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Pirrolidinonas/farmacocinética , Receptor CB1 de Canabinoide/genética , Análise de Regressão , Esquizofrenia/genética , Distribuição Tecidual/efeitos dos fármacos , Trítio/farmacocinética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA