Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 9(2): e88516, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24520393

RESUMO

Our laboratory has previously demonstrated that peripheral inflammatory pain (PIP), induced by subcutaneous plantar injection of λ-carrageenan, results in increased expression and activity of the ATP-dependent efflux transporter P-glycoprotein (P-gp) that is endogenously expressed at the blood-brain barrier (BBB). The result of increased P-gp functional expression was a significant reduction in CNS uptake of morphine and, subsequently, reduced morphine analgesic efficacy. A major concern in the treatment of acute pain/inflammation is the potential for drug-drug interactions resulting from P-gp induction by therapeutic agents co-administered with opioids. Such effects on P-gp activity can profoundly modulate CNS distribution of opioid analgesics and alter analgesic efficacy. In this study, we examined the ability of diclofenac, a non-steroidal anti-inflammatory drug (NSAID) that is commonly administered in conjunction with the opioids during pain therapy, to alter BBB transport of morphine via P-gp and whether such changes in P-gp morphine transport could alter morphine analgesic efficacy. Administration of diclofenac reduced paw edema and thermal hyperalgesia in rats subjected to PIP, which is consistent with the known mechanism of action of this NSAID. Western blot analysis demonstrated an increase in P-gp expression in rat brain microvessels not only following PIP induction but also after diclofenac treatment alone. Additionally, in situ brain perfusion studies showed that both PIP and diclofenac treatment alone increased P-gp efflux activity resulting in decreased morphine brain uptake. Critically, morphine analgesia was significantly reduced in animals pretreated with diclofenac (3 h), as compared to animals administered diclofenac and morphine concurrently. These novel findings suggest that administration of diclofenac and P-gp substrate opioids during pain pharmacotherapy may result in a clinically significant drug-drug interaction.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Anti-Inflamatórios não Esteroides/farmacologia , Sistema Nervoso Central/metabolismo , Diclofenaco/farmacologia , Morfina/metabolismo , Animais , Anti-Inflamatórios não Esteroides/administração & dosagem , Anti-Inflamatórios não Esteroides/uso terapêutico , Transporte Biológico/efeitos dos fármacos , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Carragenina , Sistema Nervoso Central/efeitos dos fármacos , Sistema Nervoso Central/patologia , Diclofenaco/administração & dosagem , Diclofenaco/uso terapêutico , Edema/sangue , Edema/complicações , Edema/tratamento farmacológico , Edema/patologia , Feminino , Imunofluorescência , Hiperalgesia/sangue , Hiperalgesia/complicações , Hiperalgesia/tratamento farmacológico , Hiperalgesia/patologia , Microvasos/efeitos dos fármacos , Microvasos/metabolismo , Microvasos/patologia , Nociceptividade/efeitos dos fármacos , Dor/sangue , Dor/complicações , Dor/tratamento farmacológico , Dor/patologia , Ratos , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/sangue
2.
J Cereb Blood Flow Metab ; 34(4): 699-707, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24473481

RESUMO

Cerebral hypoxia and subsequent reoxygenation stress (H/R) is a component of several diseases. One approach that may enable neural tissue rescue after H/R is central nervous system (CNS) delivery of drugs with brain protective effects such as 3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors (i.e., statins). Our present in vivo data show that atorvastatin, a commonly prescribed statin, attenuates poly (ADP-ribose) polymerase (PARP) cleavage in the brain after H/R, suggesting neuroprotective efficacy. However, atorvastatin use as a CNS therapeutic is limited by poor blood-brain barrier (BBB) penetration. Therefore, we examined regulation and functional expression of the known statin transporter organic anion transporting polypeptide 1a4 (Oatp1a4) at the BBB under H/R conditions. In rat brain microvessels, H/R (6% O2, 60 minutes followed by 21% O2, 10 minutes) increased Oatp1a4 expression. Brain uptake of taurocholate (i.e., Oap1a4 probe substrate) and atorvastatin were reduced by Oatp inhibitors (i.e., estrone-3-sulfate and fexofenadine), suggesting involvement of Oatp1a4 in brain drug delivery. Pharmacological inhibition of transforming growth factor-ß (TGF-ß)/activin receptor-like kinase 5 (ALK5) signaling with the selective inhibitor SB431542 increased Oatp1a4 functional expression, suggesting a role for TGF-ß/ALK5 signaling in Oatp1a4 regulation. Taken together, our novel data show that targeting an endogenous BBB drug uptake transporter (i.e., Oatp1a4) may be a viable approach for optimizing CNS drug delivery for treatment of diseases with an H/R component.


Assuntos
Barreira Hematoencefálica/metabolismo , Sistemas de Liberação de Medicamentos , Hipóxia Encefálica/metabolismo , Transportadores de Ânions Orgânicos/metabolismo , Traumatismo por Reperfusão/metabolismo , Animais , Atorvastatina , Transporte Biológico , Gasometria , Barreira Hematoencefálica/efeitos dos fármacos , Western Blotting , Dióxido de Carbono/sangue , Eletrólitos/sangue , Feminino , Ácidos Heptanoicos/administração & dosagem , Ácidos Heptanoicos/farmacocinética , Ácidos Heptanoicos/uso terapêutico , Hipóxia Encefálica/complicações , Hipóxia Encefálica/tratamento farmacológico , Microvasos/metabolismo , Fármacos Neuroprotetores/administração & dosagem , Fármacos Neuroprotetores/farmacocinética , Fármacos Neuroprotetores/uso terapêutico , Oxigênio/sangue , Pirróis/administração & dosagem , Pirróis/farmacocinética , Pirróis/uso terapêutico , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/etiologia
3.
Curr Pharm Des ; 20(10): 1422-49, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-23789948

RESUMO

The blood-brain barrier (BBB) and blood-cerebrospinal fluid (BCSF) barriers are critical determinants of CNS homeostasis. Additionally, the BBB and BCSF barriers are formidable obstacles to effective CNS drug delivery. These brain barrier sites express putative influx and efflux transporters that precisely control permeation of circulating solutes including drugs. The study of transporters has enabled a shift away from "brute force" approaches to delivering drugs by physically circumventing brain barriers towards chemical approaches that can target specific compounds of the BBB and/or BCSF barrier. However, our understanding of transporters at the BBB and BCSF barriers has primarily focused on understanding efflux transporters that efficiently prevent drugs from attaining therapeutic concentrations in the CNS. Recently, through the characterization of multiple endogenously expressed uptake transporters, this paradigm has shifted to the study of brain transporter targets that can facilitate drug delivery (i.e., influx transporters). Additionally, signaling pathways and trafficking mechanisms have been identified for several endogenous BBB/BCSF transporters, thereby offering even more opportunities to understand how transporters can be exploited for optimization of CNS drug delivery. This review presents an overview of the BBB and BCSF barrier as well as the many families of transporters functionally expressed at these barrier sites. Furthermore, we present an overview of various strategies that have been designed and utilized to deliver therapeutic agents to the brain with a particular emphasis on those approaches that directly target endogenous BBB/BCSF barrier transporters.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Sistema Nervoso Central/efeitos dos fármacos , Sistemas de Liberação de Medicamentos , Proteínas de Membrana Transportadoras/química , Preparações Farmacêuticas/administração & dosagem , Animais , Barreira Hematoencefálica/metabolismo , Sistema Nervoso Central/metabolismo , Humanos , Proteínas de Membrana Transportadoras/metabolismo
4.
Mol Pharmacol ; 84(5): 774-86, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24019224

RESUMO

Effective pharmacologic treatment of pain with opioids requires that these drugs attain efficacious concentrations in the central nervous system (CNS). A primary determinant of CNS drug permeation is P-glycoprotein (P-gp), an endogenous blood-brain barrier (BBB) efflux transporter that is involved in brain-to-blood transport of opioid analgesics (i.e., morphine). Recently, the nuclear receptor constitutive androstane receptor (CAR) has been identified as a regulator of P-gp functional expression at the BBB. This is critical to pharmacotherapy of pain/inflammation, as patients are often administered acetaminophen (APAP), a CAR-activating ligand, in conjunction with an opioid. Our objective was to investigate, in vivo, the role of CAR in regulation of P-gp at the BBB. Following APAP treatment, P-gp protein expression was increased up to 1.4-1.6-fold in a concentration-dependent manner. Additionally, APAP increased P-gp transport of BODIPY-verapamil in freshly isolated rat brain capillaries. This APAP-induced increase in P-gp expression and activity was attenuated in the presence of CAR pathway inhibitor okadaic acid or transcriptional inhibitor actinomycin D, suggesting P-gp regulation is CAR-dependent. Furthermore, morphine brain accumulation was enhanced by P-gp inhibitors in APAP-treated animals, suggesting P-gp-mediated transport. A warm-water (50°C) tail-flick assay revealed a significant decrease in morphine analgesia in animals treated with morphine 3 or 6 hours after APAP treatment, as compared with animals treated concurrently. Taken together, our data imply that inclusion of APAP in a pain treatment regimen activates CAR at the BBB and increases P-gp functional expression, a clinically significant drug-drug interaction that modulates opioid analgesic efficacy.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Acetaminofen/farmacologia , Analgésicos não Narcóticos/farmacologia , Barreira Hematoencefálica/metabolismo , Receptores Citoplasmáticos e Nucleares/fisiologia , Animais , Transporte Biológico/efeitos dos fármacos , Encéfalo/irrigação sanguínea , Receptor Constitutivo de Androstano , Feminino , Morfina/farmacocinética , Permeabilidade , Ratos , Ratos Sprague-Dawley
5.
J Neurochem ; 122(5): 962-75, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22716933

RESUMO

P-glycoprotein (ABCB1/MDR1, EC 3.6.3.44), the major efflux transporter at the blood-brain barrier (BBB), is a formidable obstacle to CNS pharmacotherapy. Understanding the mechanism(s) for increased P-glycoprotein activity at the BBB during peripheral inflammatory pain is critical in the development of novel strategies to overcome the significant decreases in CNS analgesic drug delivery. In this study, we employed the λ-carrageenan pain model (using female Sprague-Dawley rats), combined with confocal microscopy and subcellular fractionation of cerebral microvessels, to determine if increased P-glycoprotein function, following the onset of peripheral inflammatory pain, is associated with a change in P-glycoprotein trafficking which leads to pain-induced effects on analgesic drug delivery. Injection of λ-carrageenan into the rat hind paw induced a localized, inflammatory pain (hyperalgesia) and simultaneously, at the BBB, a rapid change in colocalization of P-glycoprotein with caveolin-1, a key scaffolding/trafficking protein. Subcellular fractionation of isolated cerebral microvessels revealed that the bulk of P-glycoprotein constitutively traffics to membrane domains containing high molecular weight, disulfide-bonded P-glycoprotein-containing structures that cofractionate with membrane domains enriched with monomeric and high molecular weight, disulfide-bonded, caveolin-1-containing structures. Peripheral inflammatory pain promoted a dynamic redistribution between membrane domains of P-glycoprotein and caveolin-1. Disassembly of high molecular weight P-glycoprotein-containing structures within microvascular endothelial luminal membrane domains was accompanied by an increase in ATPase activity, suggesting a potential for functionally active P-glycoprotein. These results are the first observation that peripheral inflammatory pain leads to specific structural changes in P-glycoprotein responsible for controlling analgesic drug delivery to the CNS.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Barreira Hematoencefálica/fisiopatologia , Hiperalgesia/etiologia , Hiperalgesia/patologia , Inflamação Neurogênica/complicações , Adenosina Trifosfatases/metabolismo , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Carragenina/toxicidade , Caveolina 1/metabolismo , Modelos Animais de Doenças , Feminino , Hiperalgesia/tratamento farmacológico , Microvasos/efeitos dos fármacos , Microvasos/metabolismo , Microvasos/patologia , Peso Molecular , Inflamação Neurogênica/induzido quimicamente , Transporte Proteico/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
6.
Am J Physiol Heart Circ Physiol ; 302(3): H582-93, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22081706

RESUMO

Our laboratory has shown that λ-carrageenan-induced peripheral inflammatory pain (CIP) can alter tight junction (TJ) protein expression and/or assembly leading to changes in blood-brain barrier xenobiotic permeability. However, the role of reactive oxygen species (ROS) and subsequent oxidative stress during CIP is unknown. ROS (i.e., superoxide) are known to cause cellular damage in response to pain/inflammation. Therefore, we examined oxidative stress-associated effects at the blood-brain barrier (BBB) in CIP rats. During CIP, increased staining of nitrosylated proteins was detected in hind paw tissue and enhanced presence of protein adducts containing 3-nitrotyrosine occurred at two molecular weights (i.e., 85 and 44 kDa) in brain microvessels. Tempol, a pharmacological ROS scavenger, attenuated formation of 3-nitrotyrosine-containing proteins in both the hind paw and in brain microvessels when administered 10 min before footpad injection of λ-carrageenan. Similarly, CIP increased 4-hydroxynoneal staining in brain microvessels and this effect was reduced by tempol. Brain permeability to [(14)C]sucrose and [(3)H]codeine was increased, and oligomeric assemblies of occludin, a critical TJ protein, were altered after 3 h CIP. Tempol attenuated both [(14)C]sucrose and [(3)H]codeine brain uptake as well as protected occludin oligomers from disruption in CIP animals, suggesting that ROS production/oxidative stress is involved in modulating BBB functional integrity during pain/inflammation. Interestingly, tempol administration reduced codeine analgesia in CIP animals, indicating that oxidative stress during pain/inflammation may affect opioid delivery to the brain and subsequent efficacy. Taken together, our data show for the first time that ROS pharmacological scavenging is a viable approach for maintaining BBB integrity and controlling central nervous system drug delivery during acute inflammatory pain.


Assuntos
Barreira Hematoencefálica , Permeabilidade Capilar/efeitos dos fármacos , Óxidos N-Cíclicos/farmacologia , Proteínas de Membrana/metabolismo , Neuralgia , Xenobióticos/farmacocinética , Doença Aguda , Aldeídos/farmacocinética , Analgésicos Opioides/farmacocinética , Animais , Antioxidantes/farmacologia , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/metabolismo , Permeabilidade Capilar/imunologia , Radioisótopos de Carbono , Codeína/farmacocinética , Inibidores de Cisteína Proteinase/farmacocinética , Hiperalgesia/tratamento farmacológico , Hiperalgesia/imunologia , Hiperalgesia/metabolismo , Masculino , Proteínas de Membrana/imunologia , Neuralgia/tratamento farmacológico , Neuralgia/imunologia , Neuralgia/metabolismo , Neurite (Inflamação)/tratamento farmacológico , Neurite (Inflamação)/imunologia , Neurite (Inflamação)/metabolismo , Ocludina , Estresse Oxidativo/imunologia , Ratos , Ratos Sprague-Dawley , Marcadores de Spin , Sacarose/farmacocinética , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/imunologia , Junções Íntimas/metabolismo , Tirosina/análogos & derivados , Tirosina/metabolismo
7.
J Cereb Blood Flow Metab ; 29(6): 1084-98, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19319146

RESUMO

Our laboratory has shown that peripheral inflammatory pain induced by lambda-carrageenan (CIP) can increase blood-brain barrier (BBB) permeability and alter tight junction (TJ) protein expression leading to changes in BBB functional integrity. However, the intracellular signaling mechanisms involved in this pathophysiologic response have not been elucidated. Transforming growth factor (TGF)-beta signaling pathways are known to regulate vascular integrity and permeability. Therefore, we examined the function of TGF-beta signaling at the BBB in rats subjected to CIP. During CIP, serum TGF-beta1 and protein expression of the TGF-beta receptor activin receptor-like kinase-5 (ALK5) were reduced. Brain permeability to (14)C-sucrose was increased and expression of TJ proteins (i.e., claudin-5, occludin, zonula occluden (ZO-1)) were also altered after 3 h CIP. Pharmacological inhibition of ALK5 with the selective inhibitor SB431542 further enhanced brain uptake of (14)C-sucrose, increased TJ protein expression (i.e., claudin-3, claudin-5, occludin, ZO-1), and decreased nuclear expression of TGF-beta/ALK5 signaling molecules (i.e., Smad2, Smad3), which suggests a role for TGF-beta/ALK5 signaling in the regulation of BBB integrity. Interestingly, administration of exogenous TGF-beta1 before CIP activated the TGF-beta/ALK5 pathway and reduced BBB permeability to (14)C-sucrose. Taken together, our data show that TGF-beta/ALK5 signaling is, in part, involved in the regulation of BBB functional integrity.


Assuntos
Barreira Hematoencefálica/metabolismo , Inflamação/complicações , Dor/etiologia , Dor/metabolismo , Transdução de Sinais , Junções Íntimas/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Receptores de Ativinas/metabolismo , Animais , Encéfalo/irrigação sanguínea , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Carragenina/farmacologia , Edema/induzido quimicamente , Edema/metabolismo , Feminino , Temperatura Alta , Humanos , Inflamação/induzido quimicamente , Inflamação/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases , Ratos , Ratos Sprague-Dawley , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta , Proteínas Smad/metabolismo , Especificidade por Substrato , Sacarose/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA