Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Curr Protoc Pharmacol ; 76: 7.16.1-7.16.16, 2017 Mar 17.
Article in English | MEDLINE | ID: mdl-28306152

ABSTRACT

The blood-brain barrier plays an important role in neuroprotection; however, it can be a major obstacle for drug delivery to the brain. This barrier primarily resides in the brain capillaries and functions as an interface between the brain and peripheral blood circulation. Several anatomical and biochemical elements of the blood-brain barrier are essential to regulate the permeability of nutrients, ions, hormones, toxic metabolites, and xenobiotics into and out of the brain. In particular, high expression of ATP-driven efflux transporters at the blood-brain barrier is a major obstacle in the delivery of CNS pharmacotherapeutics to the brain. The complete understanding of these elements can offer insights on how to modulate barrier functions for neuroprotection against CNS drug toxicity and to enhance drug delivery to the brain. In the literature, preclinical models of the blood-brain barrier are widely utilized to predict drug pharmacokinetics and pharmacodynamics properties in the brain. In addition, these models are essential tools to investigate cellular mechanisms and novel interventions that alter barrier function and permeability. This unit presents procedures to isolate fresh and viable rodent brain capillaries for the assessment of ex vivo transport activity at the blood-brain barrier. © 2017 by John Wiley & Sons, Inc.


Subject(s)
Brain/blood supply , Capillaries/physiology , Central Nervous System Agents/pharmacology , Animals , Biological Transport , Blood-Brain Barrier/metabolism , Brain/metabolism , Capillaries/drug effects , Central Nervous System Agents/chemistry , Central Nervous System Agents/metabolism , Drug Delivery Systems , Humans , Membrane Transport Proteins/metabolism , Permeability , Rodentia , Xenobiotics/chemistry , Xenobiotics/metabolism , Xenobiotics/pharmacology
2.
Neurosci Lett ; 639: 103-113, 2017 02 03.
Article in English | MEDLINE | ID: mdl-28011392

ABSTRACT

P-glycoprotein (P-gp), Breast cancer resistance protein (BCRP) and Multidrug resistance-associated protein 2 (MRP2) residing at the blood-brain barrier (BBB) and the blood-spinal cord barrier (BSCB) are major obstacles for drug delivery to the Central Nervous System (CNS). Disease-induced changes of these xenobiotic transporters at the CNS barriers have been previously documented. Changes in the functional expression of these transporters at the CNS barriers would limit the clinical efficacy of therapeutic agents targeting the CNS. In this study, we characterized the changes in expression and efflux activity of P-gp, BCRP and MRP2 at the BBB and BSCB of an amyotrophic lateral sclerosis (ALS) SOD1-G93A transgenic rat model across the three stages of disease progression: pre-onset, onset and symptomatic. Up-regulation of P-gp and BCRP at the BBB and BSCB during disease progression of ALS would reduce drug entry to the CNS, while any decreases in transport activity would increase drug entry. In SOD rats at the ALS symptomatic stage, we observed increases in both P-gp transport activity and expression compared to age-matched wildtypes. BCRP and MRP2 levels were unchanged in these animals. Immunohistochemical analysis in brain and spinal cord capillaries of SOD rats from all three ALS stages and age-matched wildtypes showed no differences in nuclear localization of a known P-gp regulator, nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB). It suggests that NFκB may have a limited role during P-gp induction observed in our study and additional signaling pathways could be responsible for this response. Our observations imply that novel pharmacological approaches for treating ALS require selecting drugs that are not P-gp substrates in order to improve therapeutic efficacy in the CNS during ALS progression.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Blood-Brain Barrier/metabolism , Central Nervous System/metabolism , Amyotrophic Lateral Sclerosis/drug therapy , Animals , Biological Transport/physiology , Disease Models, Animal , Membrane Transport Proteins/metabolism , Rats , Up-Regulation
3.
Antimicrob Agents Chemother ; 58(3): 1713-22, 2014.
Article in English | MEDLINE | ID: mdl-24379203

ABSTRACT

The blood-testis barrier and blood-brain barrier are responsible for protecting the male genital tract and central nervous system from xenobiotic exposure. In HIV-infected patients, low concentrations of antiretroviral drugs in cerebrospinal fluid and seminal fluid have been reported. One mechanism that may contribute to reduced concentrations is the expression of ATP-binding cassette drug efflux transporters, such as P-glycoprotein (P-gp). The objective of this study was to investigate in vivo the tissue distribution of the HIV protease inhibitor atazanavir in wild-type (WT) mice, P-gp/breast cancer resistance protein (Bcrp)-knockout (Mdr1a-/-, Mdr1b-/-, and Abcg2-/- triple-knockout [TKO]) mice, and Cyp3a-/- (Cyp) mice. WT mice and Cyp mice were pretreated with a P-gp/Bcrp inhibitor, elacridar (5 mg/kg of body weight), and the HIV protease inhibitor and boosting agent ritonavir (2 mg/kg intravenously [i.v.]), respectively. Atazanavir (10 mg/kg) was administered i.v. Atazanavir concentrations in plasma (Cplasma), brain (Cbrain), and testes (Ctestes) were quantified at various times by liquid chromatography-tandem mass spectrometry. In TKO mice, we demonstrated a significant increase in atazanavir Cbrain/Cplasma (5.4-fold) and Ctestes/Cplasma (4.6-fold) ratios compared to those in WT mice (P<0.05). Elacridar-treated WT mice showed a significant increase in atazanavir Cbrain/Cplasma (12.3-fold) and Ctestes/Cplasma (13.5-fold) ratios compared to those in vehicle-treated WT mice. In Cyp mice pretreated with ritonavir, significant (P<0.05) increases in atazanavir Cbrain/Cplasma (1.8-fold) and Ctestes/Cplasma (9.5-fold) ratios compared to those in vehicle-treated WT mice were observed. These data suggest that drug efflux transporters, i.e., P-gp, are involved in limiting the ability of atazanavir to permeate the rodent brain and genital tract. Since these transporters are known to be expressed in humans, they could contribute to the low cerebrospinal and seminal fluid antiretroviral concentrations reported in the clinic.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Brain/metabolism , HIV Protease Inhibitors/pharmacokinetics , Oligopeptides/pharmacokinetics , Pyridines/pharmacokinetics , Seminiferous Tubules/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/physiology , Animals , Atazanavir Sulfate , Brain Chemistry , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/physiology , HIV Protease Inhibitors/analysis , HIV Protease Inhibitors/blood , Male , Mice , Mice, Knockout , Oligopeptides/analysis , Oligopeptides/blood , Pyridines/analysis , Pyridines/blood , Ritonavir/analysis , Ritonavir/blood , Ritonavir/pharmacokinetics , Seminiferous Tubules/chemistry , Testis/chemistry , Testis/metabolism
4.
Curr Pharm Des ; 20(10): 1543-63, 2014.
Article in English | MEDLINE | ID: mdl-23789949

ABSTRACT

Membrane-associated drug transporters are important determinants of antiretroviral drug disposition in the central nervous system during HIV-1 infection. A number of influx and efflux transport proteins expressed at the blood-brain barrier, blood-cerebrospinal fluid barrier and in brain parenchyma cellular compartments (i.e., astrocytes, microglia) have been implicated in the traffic of many antiretroviral drugs into and out of the brain. In particular, members of the ATP-binding cassette membrane associated transporter superfamily and Solute Carrier family are known to be involved in the efflux and/or influx of drugs, respectively. As a result, changes in the functional expression of these transporters can alter the disposition and distribution of drugs in the brain. Moreover, antiretroviral therapy itself and/or pathological events (i.e., inflammation, oxidative stress) associated with viral infection may affect the functional expression of these transporters. This review summarizes recent knowledge on the role of drug transporters in regulating brain antiretroviral drug transport in the context of HIV-1 infection.


Subject(s)
Anti-HIV Agents/therapeutic use , Blood-Brain Barrier/metabolism , Drug Delivery Systems , HIV Infections/virology , HIV-1 , Membrane Transport Proteins/metabolism , Nervous System Diseases/drug therapy , Nervous System Diseases/metabolism , Animals , Blood-Brain Barrier/drug effects , HIV Infections/complications , HIV Infections/drug therapy , Humans , Nervous System Diseases/etiology
5.
Antimicrob Agents Chemother ; 57(9): 4481-8, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23836171

ABSTRACT

The membrane-associated drug transporter P-glycoprotein (P-gp) plays an essential role in drug efflux from the brain. Induction of this protein at the blood-brain barrier (BBB) could further affect the ability of a drug to enter the brain. At present, P-gp induction mediated by antiretroviral drugs at the BBB has not been fully investigated. Since P-gp expression is regulated by ligand-activated nuclear receptors, i.e., human pregnane X receptor (hPXR) and human constitutive androstane receptor (hCAR), these receptors could represent potential pathways involved in P-gp induction by antiretroviral drugs. The aims of this study were (i) to determine whether antiretroviral drugs currently used in HIV pharmacotherapy are ligands for hPXR or hCAR and (ii) to examine P-gp function and expression in human brain microvessel endothelial cells treated with antiretroviral drugs identified as ligands of hPXR and/or hCAR. Luciferase reporter gene assays were performed to examine the activation of hPXR and hCAR by antiretroviral drugs. The hCMEC/D3 cell line, which is known to display several morphological and biochemical properties of the BBB in humans, was used to examine P-gp induction following 72 h of exposure to these agents. Amprenavir, atazanavir, darunavir, efavirenz, ritonavir, and lopinavir were found to activate hPXR, whereas abacavir, efavirenz, and nevirapine were found to activate hCAR. P-gp expression and function were significantly induced in hCMEC/D3 cells treated with these drugs at clinical concentrations in plasma. Together, our data suggest that P-gp induction could occur at the BBB during chronic treatment with antiretroviral drugs identified as ligands of hPXR and/or hCAR.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/agonists , Antiviral Agents/pharmacology , Endothelial Cells/drug effects , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Steroid/agonists , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Brain/blood supply , Brain/cytology , Brain/drug effects , Brain/metabolism , Cell Line , Constitutive Androstane Receptor , Endothelial Cells/cytology , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Gene Expression Regulation , Genes, Reporter , Humans , Luciferases/genetics , Luciferases/metabolism , Microvessels/cytology , Microvessels/drug effects , Microvessels/metabolism , Pregnane X Receptor , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Signal Transduction
6.
Trends Pharmacol Sci ; 34(7): 361-72, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23769624

ABSTRACT

ATP-binding cassette membrane-associated drug efflux transporters and solute carrier influx transporters, expressed at the blood-brain barrier, blood-cerebrospinal fluid barrier, and in brain parenchyma, are important determinants of drug disposition in the central nervous system. Targeting the regulatory pathways that govern the expression of these transporters could provide novel approaches to selectively alter drug permeability into the brain. Nuclear receptors are ligand-activated transcription factors which regulate the gene expression of several metabolic enzymes and drug efflux/influx transporters. Although efforts have primarily been focused on investigating these regulatory pathways in peripheral organs (i.e., liver and intestine), recent findings demonstrate their significance in the brain. This review addresses the role of nuclear receptors in the regulation of drug transporter functional expression in the brain. An in-depth understanding of these pathways could guide the development of novel pharmacotherapy with either enhanced efficacy in the central nervous system or minimal associated neurotoxicity.


Subject(s)
Brain/metabolism , Membrane Transport Proteins/metabolism , Pharmaceutical Preparations/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , ATP-Binding Cassette Transporters/metabolism , Animals , Biological Transport , Brain/drug effects , Humans , Ion Pumps/metabolism , Membrane Transport Proteins/drug effects , Receptors, Cytoplasmic and Nuclear/drug effects
7.
J Neurochem ; 127(3): 342-52, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23777437

ABSTRACT

Intracerebral microdialysis was utilized to investigate the effect of P-glycoprotein (a drug efflux transporter) induction at the mouse blood-brain barrier (BBB) on brain extracellular fluid concentrations of quinidine, an established substrate of P-glycoprotein. Induction was achieved by treating male CD-1 mice for 3 days with 5 mg/kg/day dexamethasone (DEX), a ligand of the nuclear receptor, pregnane X receptor, and a P-glycoprotein inducer. Tandem liquid chromatography mass spectrometric method was used to quantify analytes in dialysate, blood and plasma. P-glycoprotein, pregnane X receptor and Cyp3a11 (metabolizing enzyme for quinidine) protein expression in capillaries and brain homogenates was measured by immunoblot analysis. Following quinidine i.v. administration, the average ratio of unbound quinidine concentrations in brain extracellular fluid (determined from dialysate samples) to plasma at steady state (375-495 min) or Kp, uu, ECF /Plasma in the DEX-treated animals was 2.5-fold lower compared with vehicle-treated animals. In DEX-treated animals, P-glycoprotein expression in brain capillaries was 1.5-fold higher compared with vehicle-treated animals while Cyp3a11 expression in brain capillaries was not significantly different between the two groups. These data demonstrate that P-gp induction mediated by DEX at the BBB can significantly reduce quinidine brain extracellular fluid concentrations by decreasing its brain permeability and further suggest that drug-drug interactions as a result of P-gp induction at the BBB are possible. Applying microdialysis, distribution of quinidine, a P-gp substrate, in mouse brain extracellular fluid (ECF) was investigated following ligand-mediated P-glycoprotein (P-gp) induction at the blood-brain barrier (BBB). We demonstrated that a PXR agonist (dexamethasone) significantly up-regulated P-gp in brain capillaries and reduced quinidine brain ECF concentrations. Our data suggest that drug-drug interactions as a result of P-gp induction at the BBB are possible.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Blood-Brain Barrier/metabolism , Animals , Blotting, Western , Capillaries/metabolism , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP3A/metabolism , Data Interpretation, Statistical , Densitometry , Dexamethasone/pharmacology , Male , Membrane Proteins/metabolism , Mice , Microdialysis , Pregnane X Receptor , Quinidine/blood , Quinidine/metabolism , Receptors, Steroid/metabolism , Tandem Mass Spectrometry
8.
J Neurochem ; 123(6): 944-53, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23035695

ABSTRACT

Induction of the multidrug resistance protein 1 (MDR1)/P-glycoprotein (P-gp) by the vitamin D receptor (VDR) was investigated in isolated rat brain capillaries and rat (RBE4) and human (hCMEC/D3) brain microvessel endothelial cell lines. Incubation of isolated rat brain capillaries with 10 nM of the VDR ligand, 1α,25-dihydroxyvitamin D(3) [1,25(OH)(2)D(3)] for 4 h increased P-gp protein expression fourfold. Incubation with 1,25(OH)(2)D(3) for 4 or 24 h increased P-gp transport activity (specific luminal accumulation of NBD-CSA, the fluorescent P-gp substrate) by 25-30%. In RBE4 cells, Mdr1b mRNA was induced in a concentration-dependent manner by exposure to 1,25(OH)(2)D(3). Concomitantly, P-gp protein expression increased 2.5-fold and was accompanied by a 20-35% reduction in cellular accumulation of the P-gp substrates, rhodamine 6G (R6G), and HiLyte Fluor 488-labeled human amyloid beta 1-42 (hAß(42)). In hCMEC/D3 cells, a 3 day exposure to 100 nM 1,25(OH)(2)D(3) increased MDR1 mRNA expression (40%) and P-gp protein (threefold); cellular accumulation of R6G and hAß(42) was reduced by 30%. Thus, VDR activation up-regulates Mdr1/MDR1 and P-gp protein in isolated rat brain capillaries and rodent and human brain microvascular endothelia, implicating a role for VDR in increasing the brain clearance of P-gp substrates, including hAß(42), a plaque-forming precursor in Alzheimer's disease.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Blood-Brain Barrier/cytology , Brain/blood supply , Calcitriol/metabolism , Calcitriol/physiology , Endothelial Cells/metabolism , Receptors, Calcitriol/physiology , ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/physiology , Brain/cytology , Cell Line , Endothelial Cells/cytology , Endothelial Cells/drug effects , Humans , Ligands , Male , Protein Transport/physiology , Rats , Rats, Sprague-Dawley , Receptors, Calcitriol/metabolism , Up-Regulation/physiology
9.
J Neurochem ; 118(2): 163-75, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21517853

ABSTRACT

In mammalian systems, pregnane X receptor (PXR) and constitutive androstane receptor (CAR) have been recognized as xenobiotic-sensors which can up-regulate the functional expression of drug transporters, such as P-glycoprotein (P-gp). In the brain, an increase in P-gp expression can further limit drug permeability across the blood-brain barrier (BBB) and potentially reduce CNS pharmacotherapy efficacy. At present, the involvement of human PXR (hPXR) and CAR (hCAR) in the regulation of P-gp expression at the human BBB is unknown. In this study, we investigate the role of hPXR and hCAR in the regulation of P-gp expression using a human cerebral microvessel endothelial cell culture system. We demonstrate that activation of hPXR and hCAR by their respective ligands leads to P-gp induction at both mRNA and protein levels, while pharmacological inhibitors of hPXR and hCAR prevent ligand-mediated P-gp induction. Ligand-induced nuclear translocation of hPXR is observed, although such effect could not be demonstrated for hCAR. Furthermore, down-regulation of hPXR and hCAR proteins using small-interfering RNA decreased P-gp expression. Our findings provide first evidence for P-gp regulation by hPXR and hCAR at the human BBB and suggest insights on how to achieve selective P-gp regulation at this site.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Brain/metabolism , Endothelial Cells/physiology , Microvessels/physiology , Orphan Nuclear Receptors/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Receptors, Steroid/physiology , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Animals , Blood-Brain Barrier/cytology , Blood-Brain Barrier/physiology , Brain/blood supply , Brain/cytology , Cell Line, Transformed , Cells, Cultured , Constitutive Androstane Receptor , Endothelial Cells/cytology , Endothelial Cells/metabolism , Hep G2 Cells , Humans , Ligands , Microvessels/cytology , Microvessels/metabolism , Pregnane X Receptor , RNA, Messenger/physiology , Rats , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Steroid/antagonists & inhibitors
10.
Methods Mol Biol ; 686: 313-36, 2011.
Article in English | MEDLINE | ID: mdl-21082379

ABSTRACT

The blood-brain barrier (BBB) physically and metabolically functions as a neurovascular interface between the brain parenchyma and the systemic circulation, and regulates the permeability of several endogenous substrates and xenobiotics in and out of the central nervous system. Several membrane-associated transport proteins, such as P-glycoprotein (P-gp), multidrug resistance-associated proteins, breast cancer resistance protein, and organic anion transporting polypeptides, have been characterized at the BBB and identified to play a major role in regulating the brain bioavailability of several pharmacological agents. This chapter reviews several well-established techniques for the study of the molecular expression, cellular localization, and functional activity of transport proteins in primary and immortalized cell culture systems of the BBB. In particular, we describe the molecular characterization of P-gp/MDR1 at the transcript level using semiquantitative polymerase chain reaction (PCR), at the protein level using immunoblotting, and at the cellular level using immunofluorescence. In addition, the uptake/efflux and transepithelial flux studies, which characterize P-gp transport activity, are described.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Blood-Brain Barrier/metabolism , Cells, Cultured , Humans , Rats
11.
Trends Pharmacol Sci ; 31(1): 22-35, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20004485

ABSTRACT

Treatment of human immunodeficiency virus (HIV) infection involves a combination of several antiviral agents belonging to different pharmacological classes. This combination is referred to as highly active antiretroviral therapy (HAART). This treatment has proved to be very effective in suppressing HIV replication, but antiretroviral drugs have complex pharmacokinetic properties involving extensive drug metabolism and transport by membrane-associated drug carriers. Combination drug therapy often introduces complex drug-drug interactions that can result in toxic or sub-therapeutic drug concentrations, compromising treatment. This review focuses on the role of ATP-binding cassette (ABC) membrane-associated efflux transporters and solute carrier (SLC) uptake transporters in antiretroviral drug disposition, and identifies clinically important antiretroviral drug-drug interactions associated with changes in drug transport.


Subject(s)
Anti-HIV Agents/pharmacology , HIV Infections/drug therapy , Membrane Transport Proteins/drug effects , ATP-Binding Cassette Transporters/drug effects , ATP-Binding Cassette Transporters/metabolism , Animals , Anti-HIV Agents/adverse effects , Anti-HIV Agents/pharmacokinetics , Antiretroviral Therapy, Highly Active/methods , Biological Transport/drug effects , Drug Interactions , Humans , Membrane Transport Proteins/metabolism
12.
J Neurosci Res ; 87(4): 1023-36, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18855943

ABSTRACT

A major concern regarding the chronic administration of antiretroviral drugs is the potential for induction of drug efflux transporter expression (i.e., P-glycoprotein, P-gp) at tissue sites that can significantly affect drug distribution and treatment efficacy. Previous data have shown that the inductive effect of human immunodeficiency virus protease inhibitors (PIs) is mediated through the human orphan nuclear receptor, steroid xenobiotic receptor (SXR or hPXR). The objectives of this study were to investigate transport and inductive properties on efflux drug transporters of two PIs, atazanavir and ritonavir, at the blood-brain barrier by using a human brain microvessel endothelial cell line, hCMEC/D3. Transport properties of PIs by the drug efflux transporters P-gp and multidrug resistance protein 1 (MRP1) were assessed by measuring the cellular uptake of (3)H-atazanavir or (3)H-ritonavir in P-gp and MRP1 overexpressing cells as well as hCMEC/D3. Whereas the P-gp inhibitor, PSC833, increased atazanavir and ritonavir accumulation in hCMEC/D3 cells by 2-fold, the MRP inhibitor MK571 had no effect. P-gp, MRP1, and hPXR expression and localization were examined by Western blot analysis and immunogold cytochemistry at the electron microscope level. Treatment of hCMEC/D3 cells for 72 hr with rifampin or SR12813 (two well-established hPXR ligands) or PIs (atazanavir or ritonavir) resulted in an increase in P-gp expression by 1.8-, 6-, and 2-fold, respectively, with no effect observed for MRP1 expression. In hCMEC/D3 cells, cellular accumulation of these PIs appears to be primarily limited by P-gp efflux activity. Long-term exposure of atazanavir or ritonavir to brain microvessel endothelium may result in further limitations in brain drug permeability as a result of the up-regulation of P-gp expression and function.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Endothelium, Vascular/metabolism , HIV Protease Inhibitors/pharmacology , Up-Regulation , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Atazanavir Sulfate , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/blood supply , Brain/drug effects , Brain/metabolism , Cell Line , Cyclosporins/pharmacology , Diphosphonates/pharmacology , Endothelium, Vascular/drug effects , Humans , Microvessels/drug effects , Microvessels/metabolism , Oligopeptides/pharmacology , Propionates/pharmacology , Pyridines/pharmacology , Quinolines/pharmacology , Rifampin/pharmacology , Ritonavir/pharmacology , Tritium/metabolism
13.
Diabetes ; 56(11): 2722-31, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17682092

ABSTRACT

OBJECTIVE: Prolonged elevation of glucose can adversely affect beta-cell function. In vitro studies have linked glucose-induced beta-cell dysfunction to oxidative stress; however, whether oxidative stress plays a role in vivo is unclear. Therefore, our objective was to investigate the role of oxidative stress in an in vivo model of glucose-induced beta-cell dysfunction. RESEARCH DESIGN AND METHODS: Wistar rats were infused intravenously with glucose for 48 h to achieve 20 mmol/l hyperglycemia with/without co-infusion of one of the following antioxidants: taurine (2-amino ethanesulfonic acid) (TAU), an aldehyde scavenger; N-acetylcysteine (NAC), a precursor of glutathione; or tempol (4-hydroxy-2,2,6,6-tetramethylpiperidine-N-oxyl) (TPO), a superoxide dismutase mimetic. This was followed by islet isolation or hyperglycemic clamp. RESULTS: A 48-h glucose infusion decreased glucose-stimulated insulin secretion (GSIS) and elevated reactive oxygen species (ROS), total superoxide, and mitochondrial superoxide in freshly isolated islets. TPO prevented the increase in total and mitochondrial superoxide and the beta-cell dysfunction induced by high glucose. However, TAU and NAC, despite completely normalizing H(2)DCF-DA (dihydro-dichlorofluorescein diacetate)-measured ROS, did not prevent the increase in superoxide and the decrease in beta-cell function induced by high glucose. TPO but not TAU also prevented beta-cell dysfunction induced by less extreme hyperglycemia (15 mmol/l) for a longer period of time (96 h). To further investigate whether TPO is effective in vivo, a hyperglycemic clamp was performed. Similar to the findings in isolated islets, prolonged glucose elevation (20 mmol/l for 48 h) decreased beta-cell function as assessed by the disposition index (insulin secretion adjusted for insulin sensitivity), and co-infusion of TPO with glucose completely restored beta-cell function. CONCLUSIONS: These findings implicate superoxide generation in beta-cell dysfunction induced by prolonged hyperglycemia.


Subject(s)
Glucose/pharmacology , Insulin-Secreting Cells/metabolism , Superoxides/metabolism , Animals , Glucose/administration & dosage , Hyperglycemia/metabolism , Infusions, Intravenous , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/physiology , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...