Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Diabet Med ; 37(10): 1715-1722, 2020 10.
Article in English | MEDLINE | ID: mdl-31618475

ABSTRACT

AIM: To examine whether early addition of sitagliptin to metformin is associated with a delay in type 2 diabetes progression. METHODS: Administrative health records from Alberta, Canada, for the period April 2008 to March 2015, were used to conduct a retrospective cohort study in new metformin users. People who started sitagliptin on the same day they initiated metformin therapy were compared with those who added sitagliptin later. Insulin initiation served as a surrogate marker for diabetes progression, and multivariable logistic regression models were used to evaluate the association with sitagliptin addition (costart vs later use). A mixed-effects linear regression model was used to examine the effect of timing of sitagliptin addition on HbA1c change over 1 year. RESULTS: The mean (sd) age of the 8764 people who used sitagliptin was 52.1 (11.1) years, 5665 (64.6%) were men, and 1153 (13.2%) started sitagliptin on the same day as metformin. Insulin was added to the therapy of 173 (15.0%) costarters and 1453 (19.1%) later sitagliptin users. The adjusted odds ratio for adding insulin was 0.76 (95% CI 0.64 to 0.90) in favour of costarting sitagliptin. HbA1c levels decreased in both groups 1 year after starting sitagliptin, with costarters having a significantly greater reduction [absolute between-group difference of 0.5% (95% CI 0.3 to 0.7)] compared with later sitagliptin users. CONCLUSION: Costarting drug therapy with sitagliptin and metformin was associated with a lower likelihood of disease progression in people with type 2 diabetes compared with adding sitagliptin later.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Sitagliptin Phosphate/therapeutic use , Adult , Alberta , Cohort Studies , Diabetes Mellitus, Type 2/metabolism , Disease Progression , Drug Therapy, Combination , Female , Glycated Hemoglobin/metabolism , Humans , Logistic Models , Male , Middle Aged , Retrospective Studies
2.
Diabetes Obes Metab ; 17(6): 523-532, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25711240

ABSTRACT

More than 40 years after publication of the University Group Diabetes Program trial, the cardiovascular safety of sulphonylureas is still contentious. Although several hypotheses linking sulphonylureas to adverse cardiovascular effects exist, none provide conclusive evidence. Adding to the controversy, current clinical trials and observational studies provide inconsistent, and sometimes conflicting, evidence for the cardiovascular effects of sulphonylureas. Overall, observational evidence suggests that an increased risk of adverse cardiovascular outcomes is associated with sulphonylureas; however, these data may be subject to residual confounding and bias. Although evidence from randomized controlled trials has suggested a neutral effect, the majority of these studies were not specifically designed to assess the effect of sulphonylureas on adverse cardiovascular event risk. Current ongoing large clinical trials may provide some clarity on the cardiovascular safety of sulphonylureas, but the results are not expected for several years. With the continued uncertainties concerning the cardiovascular safety of all antidiabetic drugs, a clear answer with regard to sulphonylureas is warranted. The objectives of the present article were to provide an overview of the controversy surrounding sulphonylurea-related cardiovascular effects, to discuss the limitations of the current literature, and to provide recommendations for future studies aiming to elucidate the true relationship between sulphonylureas and adverse cardiovascular effects in people with type 2 diabetes.


Subject(s)
Cardiovascular Diseases/chemically induced , Diabetes Mellitus/drug therapy , Hypoglycemic Agents/adverse effects , Sulfonylurea Compounds/adverse effects , Humans
3.
Cell Death Dis ; 4: e885, 2013 Oct 24.
Article in English | MEDLINE | ID: mdl-24157879

ABSTRACT

Epoxyeicosatrienoic acids (EETs) are cytochrome P450 epoxygenase metabolites of arachidonic acid involved in regulating pathways promoting cellular protection. We have previously shown that EETs trigger a protective response limiting mitochondrial dysfunction and reducing cellular death. Considering it is unknown how EETs regulate cell death processes, the major focus of the current study was to investigate their role in the autophagic response of HL-1 cells and neonatal cardiomyocytes (NCMs) during starvation. We employed a dual-acting synthetic analog UA-8 (13-(3-propylureido)tridec-8-enoic acid), possessing both EET-mimetic and soluble epoxide hydrolase (sEH) inhibitory properties, or 14,15-EET as model EET molecules. We demonstrated that EETs significantly improved viability and recovery of starved cardiac cells, whereas they lowered cellular stress responses such as caspase-3 and proteasome activities. Furthermore, treatment with EETs resulted in preservation of mitochondrial functional activity in starved cells. The protective effects of EETs were abolished by autophagy-related gene 7 (Atg7) short hairpin RNA (shRNA) or pharmacological inhibition of autophagy. Mechanistic evidence demonstrated that sarcolemmal ATP-sensitive potassium channels (pmKATP) and enhanced activation of AMP-activated protein kinase (AMPK) played a crucial role in the EET-mediated effect. Our data suggest that the protective effects of EETs involve regulating the autophagic response, which results in a healthier pool of mitochondria in the starved cardiac cells, thereby representing a novel mechanism of promoting survival of cardiac cells. Thus, we provide new evidence highlighting a central role of the autophagic response in linking EETs with promoting cell survival during deep metabolic stress such as starvation.


Subject(s)
8,11,14-Eicosatrienoic Acid/analogs & derivatives , Amino Acids/deficiency , Autophagy/drug effects , Cardiotonic Agents/pharmacology , Cytoprotection/drug effects , Myocytes, Cardiac/cytology , Oleic Acids/pharmacology , 8,11,14-Eicosatrienoic Acid/pharmacology , AMP-Activated Protein Kinases/metabolism , Adenosine Triphosphate/pharmacology , Animals , Animals, Newborn , Benzamides/pharmacology , Cell Line , Cell Survival/drug effects , Enzyme Activation/drug effects , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Mitochondria, Heart/ultrastructure , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/ultrastructure , Phosphorylation/drug effects , Potassium Channels/metabolism , Rats , Stress, Physiological/drug effects
4.
Diabetes Obes Metab ; 15(11): 1022-8, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23668425

ABSTRACT

AIMS: Adenosine triphosphate sensitive potassium (K(ATP)) channel activity is cardioprotective during ischaemia. One of the purported mechanisms for sulphonylurea adverse effects is through inhibition of these channels. The purpose of this study is to examine whether patients using K(ATP) channel inhibitors at the time of an acute coronary syndrome are at greater risk of death or heart failure (HF) than those not exposed. METHODS: Using linked administrative databases we identified all adults who had an acute coronary syndrome between April 2002 and October 2006 (n = 21 023). RESULTS: Within 30 days of acute coronary syndrome, 5.3% of our cohort died and 15.6% were diagnosed with HF. Individuals with diabetes exhibited significantly higher risk of death (adjusted OR: 1.20, 95% CI: 1.03-1.40) and death or HF (aOR: 1.73, 95% CI: 1.59-1.89) than individuals without diabetes. However, there was no significantly increased risk of death (aOR: 1.00, 95% CI: 0.76-1.33) or death/HF (aOR: 1.06, 95% CI: 0.89-1.26) in patients exposed to K(ATP) channel inhibitors versus patients not exposed to K(ATP) channel inhibitors prior to their acute coronary syndrome. CONCLUSIONS: Diabetes is associated with an increased risk of death or HF within 30 days of an acute coronary syndrome. However, we did not find any excess risk of death or HF associated with use of K(ATP) channel inhibitors at the time of an acute coronary syndrome, raising doubts about the hypothesis that sulphonylureas inhibit the cardioprotective effects of myocardial K(ATP) channels.


Subject(s)
Acute Coronary Syndrome/physiopathology , Diabetes Mellitus, Type 2/drug therapy , Diabetic Cardiomyopathies/physiopathology , Hypoglycemic Agents/adverse effects , Potassium Channel Blockers/adverse effects , Sulfonylurea Compounds/adverse effects , Acute Coronary Syndrome/complications , Acute Coronary Syndrome/diagnosis , Acute Coronary Syndrome/mortality , Aged , Aged, 80 and over , Alberta/epidemiology , Cohort Studies , Diabetes Mellitus, Type 2/complications , Diabetic Cardiomyopathies/diagnosis , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/mortality , Female , Heart Failure/etiology , Humans , Hypoglycemic Agents/therapeutic use , KATP Channels/antagonists & inhibitors , KATP Channels/metabolism , Logistic Models , Male , Medical Record Linkage , Mortality , Potassium Channel Blockers/therapeutic use , Prognosis , Risk Factors , Sulfonylurea Compounds/therapeutic use
5.
Diabetes Obes Metab ; 14(2): 130-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21923736

ABSTRACT

AIM: Insulin secretagogues promote insulin release by binding to sulfonylurea receptors on pancreatic ß-cells (SUR1). However, these drugs also bind to receptor isoforms on cardiac myocytes (SUR2A) and vascular smooth muscle (SUR2B). Binding to SUR2A/SUR2B may inhibit ischaemic preconditioning, an endogenous protective mechanism enabling cardiac tissue to survive periods of ischaemia. This study was designed to identify insulin secretagogues that selectively bind to SUR1 when given at therapeutic doses. METHODS: Using accepted systematic review methods, three electronic databases were searched from inception to 13 June 2011. Original studies measuring the half-maximal inhibitory concentration (IC(50)) for an insulin secretagogue on K(ATP) channels using standard electrophysiological techniques were included. Steady-state concentrations (C(SS)) were estimated from the usual oral dose and clearance values for each drug. RESULTS: Data were extracted from 27 studies meeting all inclusion criteria. IC(50) values for SUR1 were below those for SUR2A/SUR2B for all insulin secretagogues and addition of C(SS) values identified three distinct patterns. The C(SS) for gliclazide, glipizide, mitiglinide and nateglinide lie between IC(50) values for SUR1 and SUR2A/SUR2B, suggesting that these drugs bind selectively to pancreatic receptors. The C(SS) for glimepiride and glyburide (glibenclamide) was above IC(50) values for all three isoforms, suggesting these drugs are non-selective. Tolbutamide and repaglinide may have partial pancreatic receptor selectivity because IC(50) values for SUR1 and SUR2A/SUR2B overlapped somewhat, with the C(SS) in the midst of these values. CONCLUSIONS: Insulin secretagogues display different tissue selectivity characteristics at therapeutic doses. This may translate into different levels of cardiovascular risk.


Subject(s)
ATP-Binding Cassette Transporters/drug effects , Cardiovascular Diseases/metabolism , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/adverse effects , Muscle, Smooth, Vascular/drug effects , Myocytes, Cardiac/drug effects , Potassium Channels, Inwardly Rectifying/drug effects , Receptors, Drug/drug effects , ATP-Binding Cassette Transporters/metabolism , Animals , Carbamates/adverse effects , Cardiovascular Diseases/chemically induced , Cardiovascular Diseases/physiopathology , Cricetinae , Cyclohexanes/adverse effects , Diabetes Mellitus, Type 2/metabolism , Gliclazide/adverse effects , Glipizide/adverse effects , Glyburide/adverse effects , Humans , Hypoglycemic Agents/pharmacology , Ischemic Preconditioning, Myocardial , Isoindoles/adverse effects , Mice , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiopathology , Myocytes, Cardiac/metabolism , Nateglinide , Phenylalanine/adverse effects , Phenylalanine/analogs & derivatives , Piperidines/adverse effects , Potassium Channels, Inwardly Rectifying/metabolism , Rats , Receptors, Drug/metabolism , Risk Factors , Sulfonylurea Compounds/adverse effects , Sulfonylurea Receptors , Tolbutamide/adverse effects
6.
Curr Med Chem ; 18(24): 3640-61, 2011.
Article in English | MEDLINE | ID: mdl-21774762

ABSTRACT

Cardiac atrial and ventricular arrhythmias are major causes of mortality and morbidity. Ischemic heart disease is the most common cause underlying 1) the development of ventricular fibrillation that results in sudden cardiac death and 2) atrial fibrillation that can lead to heart failure and stroke. Current pharmacological agents for the treatment of ventricular and atrial arrhythmias exhibit limited effectiveness and many of these agents can cause serious adverse effects - including the provocation of lethal ventricular arrhythmias. Sarcolemmal ATP-sensitive potassium channels (sarcK(ATP)) couple cellular metabolism to membrane excitability in a wide range of tissues. In the heart, sarcK(ATP) are activated during metabolic stress including myocardial ischemia, and both the opening of sarcK(ATP) and mitochondrial K(ATP) channels protect the ischemic myocardium via distinct mechanisms. Myocardial ischemia leads to a series of events that promote the generation of arrhythmia substrate eventually resulting in the development of life-threatening arrhythmias. In this review, the possible mechanisms of the anti- and proarrhythmic effects of sarcK(ATP) modulation as well as the influence of pharmacological K(ATP) modulators are discussed. It is concluded that in spite of the significant advances made in this field, the possible cardiovascular therapeutic utility of current sarcK(ATP) channel modulators is still hampered by the lack of chamber-specific selectivity. However, recent insights into the chamber-specific differences in the molecular composition of sarcKATP in addition to already existing cardioselective sarcK(ATP) channel modulators with sarcK(ATP) isoform selectivity holds the promise for the future development of pharmacological strategies specific for a variety of atrial and ventricular arrhythmias.


Subject(s)
Arrhythmias, Cardiac/metabolism , KATP Channels/metabolism , Sarcolemma/metabolism , Animals , Anti-Arrhythmia Agents/therapeutic use , Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/pathology , Humans , KATP Channels/antagonists & inhibitors , KATP Channels/genetics , Potassium Channel Blockers/therapeutic use , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism
7.
Br J Pharmacol ; 153(4): 676-83, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18071296

ABSTRACT

BACKGROUND AND PURPOSE: The potent oxidant peroxynitrite (ONOO(-)) induces mechanical dysfunction in the intact heart in part through activation of matrix metalloproteinase-2 (MMP-2). This effect may be independent of the proteolytic actions of MMPs on extracellular matrix proteins. The purpose of this study was to examine the effects of ONOO(-) on contractile function at the level of the single cardiac myocyte and whether this includes the action of MMPs. EXPERIMENTAL APPROACH: Freshly isolated ventricular myocytes from adult rats were superfused with Krebs-Henseleit buffer at 21 degrees C and paced at 0.5 Hz. Contractility was measured using a video edge-detector. ONOO(-) or decomposed ONOO(-) (vehicle control) were co-infused over 40 min to evaluate the contraction cease time (CCT). The effects of ONOO(-) on intracellular [Ca(2+)] were determined in myocytes loaded with calcium green-1 AM. MMP-2 activity was measured by gelatin zymography. KEY RESULTS: ONOO(-) (30-600 microM) caused a concentration-dependent reduction in CCT. Myocytes subjected to 300 microM ONOO(-) had a shorter CCT than decomposed ONOO(-) (14.9+1.5 vs 32.2+3.5 min, n=7-8; P<0.05) and showed increased MMP-2 activity. The MMP inhibitors doxycycline (100 microM) or PD 166793 (2 microM) reduced the decline in CCT induced by 300 microM ONOO(-). ONOO(-) caused shorter calcium transient cease time and significant alterations in intracellular [Ca(2+)] homoeostasis which were partially prevented by doxycycline. CONCLUSIONS AND IMPLICATIONS: This is the first demonstration that inhibition of MMPs protects the cardiac myocyte from ONOO(-)-induced contractile failure via an action unrelated to proteolysis of extracellular matrix proteins.


Subject(s)
Doxycycline/pharmacology , Hydroxamic Acids/pharmacology , Matrix Metalloproteinase Inhibitors , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Oligopeptides/pharmacology , Peroxynitrous Acid/metabolism , Protease Inhibitors/pharmacology , Animals , Calcium/metabolism , Cardiac Pacing, Artificial , Enzyme Activation , Homeostasis , Male , Matrix Metalloproteinase 2/metabolism , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/metabolism , Rats , Rats, Sprague-Dawley , Time Factors
8.
Br J Pharmacol ; 149(6): 657-65, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17016511

ABSTRACT

BACKGROUND AND PURPOSE: The cardiovascular benefits of red wine consumption are often attributed to the antioxidant effects of its polyphenolic constituents, including quercetin, catechin and resveratrol. Inhibition of cardiac voltage-gated sodium channels (VGSCs) is antiarrhythmic and cardioprotective. As polyphenols may also modulate ion channels, and possess structural similarities to several antiarrhythmic VGSC inhibitors, we hypothesised that VGSC inhibition may contribute to cardioprotection by these polyphenols. EXPERIMENTAL APPROACH: The whole-cell voltage-clamp technique was used to record peak and late VGSC currents (INa) from recombinant human heart NaV1.5 channels expressed in tsA201 cells. Right ventricular myocytes from rat heart were isolated and single myocytes were field-stimulated. Either calcium transients or contractility were measured using the calcium-sensitive dye Calcium-Green 1AM or video edge detection, respectively. KEY RESULTS: The red grape polyphenols quercetin, catechin and resveratrol blocked peak INa with IC50s of 19.4 microM, 76.8 microM and 77.3 microM, respectively. In contrast to lidocaine, resveratrol did not exhibit any frequency-dependence of peak INa block. Late INa induced by the VGSC long QT mutant R1623Q was reduced by resveratrol and quercetin. Resveratrol and quercetin also blocked late INa induced by the toxin, ATX II, with IC50s of 26.1 microM and 24.9 microM, respectively. In field-stimulated myocytes, ATXII-induced increases in diastolic calcium were prevented and reversed by resveratrol. ATXII-induced contractile dysfunction was delayed and reduced by resveratrol. CONCLUSIONS AND IMPLICATIONS: Our results indicate that several red grape polyphenols inhibit cardiac VGSCs and that this effect may contribute to the documented cardioprotective efficacy of red grape products.


Subject(s)
Flavonoids/pharmacology , Heart/drug effects , Ion Channel Gating/drug effects , Phenols/pharmacology , Sodium Channels/drug effects , Vitis/chemistry , Animals , Base Sequence , Calcium/metabolism , Cell Line , DNA Primers , Mutagenesis, Site-Directed , Polyphenols , Rats
9.
J Mol Endocrinol ; 32(2): 415-24, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15072548

ABSTRACT

Leptin suppresses insulin secretion by opening ATP-sensitive K(+) (K(ATP)) channels and hyperpolarizing beta-cells. We measured the intracellular concentration of ATP ([ATP](i)) in tumor-derived beta-cells, INS-1, and found that leptin reduced [ATP](i) by approximately 30%, suggesting that the opening of K(ATP) channels by leptin is mediated by decreased [ATP](i). A reduction in glucose availability for metabolism may explain the decreased [ATP](i), since leptin (30 min) reduced glucose transport into INS-1 cells by approximately 35%, compared to vehicle-treated cells. The twofold induction of GLUT2 phosphorylation by GLP-1, an insulin secretagogue, was abolished by leptin. Therefore, the acute effect of leptin could involve covalent modification of GLUT2. These findings suggest that leptin may inhibit insulin secretion by reducing [ATP](i) as a result of reduced glucose availability for the metabolic pathway. In addition, leptin reduced glucose transport by 35% in isolated rat hepatocytes that also express GLUT2, suggesting that glucose transport may also be altered by leptin in other glucose-responsive tissues such as the liver.


Subject(s)
Adenosine Triphosphate/metabolism , Glucose/metabolism , Islets of Langerhans/metabolism , Leptin/pharmacology , Animals , Biological Transport/drug effects , Cells, Cultured , Glucagon/drug effects , Glucagon/metabolism , Glucagon-Like Peptide 1 , Glucose Transporter Type 2 , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Islets of Langerhans/drug effects , Male , Monosaccharide Transport Proteins/drug effects , Monosaccharide Transport Proteins/metabolism , Peptide Fragments/drug effects , Peptide Fragments/metabolism , Phosphorylation , Potassium Channels/drug effects , Potassium Channels/metabolism , Protein Precursors/drug effects , Protein Precursors/metabolism , Rats , Rats, Sprague-Dawley
10.
FASEB J ; 15(14): 2586-94, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11726534

ABSTRACT

The protective roles of sarcolemmal (sarc) and mitochondrial (mito) KATP channels are unclear despite their apparent importance to ischemic preconditioning. We examined these roles by monitoring intracellular calcium ([Ca]int), using fura-2 and fluo-3, in enzymatically isolated rat right ventricular myocytes. Myocyte mortality, estimated using a trypan blue assay, changed approximately in parallel with changes in [Ca]int. Chemically induced hypoxia (CIH), induced by application of cyanide and 2-deoxy-glucose, caused a steady rise in [Ca]int. Calcium increased more rapidly on 'reoxygenation' by return to control solutions. The protein kinase C (PKC) activator PMA abolished both phases of calcium increase. The mitoKATP channel-selective blocker 5-hydroxydecanoate partially prevented the PMA-induced protection during CIH, but not during reoxygenation. In contrast, HMR 1098, a sarcKATP channel-selective blocker, abolished protection only during the reoxygenation. Adenosine (A1) receptor activation prevented or reduced increases in [Ca]int and improved cell viability via a PKC and mito/sarcKATP channel-dependent mechanism. PKC-dependent protection against cytoplasmic calcium increases was also observed in a human cell line (tsA201) transiently expressing sarcKATP channels. Protection was abolished only during the reoxygenation phase by the amino acid substitution (T180A) in the pore-forming Kir6.2 subunit, a mutation previously shown to prevent PKC-dependent modulation. Our data suggest that sarc and mitoKATP channel populations play distinct protective roles, triggered by PKC and/or adenosine, during chemically induced hypoxia/reoxygenation.


Subject(s)
Adenosine/analogs & derivatives , Mitochondria/metabolism , Potassium Channels/physiology , Sarcolemma/metabolism , ATP-Binding Cassette Transporters , Adenosine/pharmacology , Adenosine Triphosphate/pharmacology , Animals , Benzamides/pharmacology , Calcium/metabolism , Cell Hypoxia/physiology , Cell Line , Cell Survival/drug effects , Cells, Cultured , Decanoic Acids/pharmacology , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Heart Ventricles/cytology , Heart Ventricles/drug effects , Humans , Hydroxy Acids/pharmacology , Ischemic Preconditioning, Myocardial , KATP Channels , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Membrane Potentials/drug effects , Myocardium/metabolism , Oxygen/pharmacology , Potassium Channels/drug effects , Potassium Channels/genetics , Potassium Channels, Inwardly Rectifying , Purinergic P1 Receptor Agonists , Purinergic P1 Receptor Antagonists , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/physiology , Time Factors , Ventricular Function , Xanthines/pharmacology
11.
J Biol Chem ; 276(28): 25919-28, 2001 Jul 13.
Article in English | MEDLINE | ID: mdl-11342562

ABSTRACT

We have recently described a novel K(+)-dependent Na(+)/Ca(2+) exchanger, NCKX2, that is abundantly expressed in brain neurons (Tsoi, M., Rhee, K.-H., Bungard, D., Li, X.-F., Lee, S.-L., Auer, R. N., and Lytton, J. (1998) J. Biol. Chem. 273, 4115--4162). The precise role for NCKX2 in neuronal Ca(2+) homeostasis is not yet clearly understood but will depend upon the functional properties of the molecule. Here, we have performed whole-cell patch clamp analysis to characterize cation dependences and ion stoichiometry for rat brain NCKX2, heterologously expressed in HEK293 cells. Outward currents generated by reverse NCKX2 exchange depended on external Ca(2+) with a K(12) of 1.4 or 101 microm without or with 1 mm Mg(2+), and on external K(+) with a K(1/2) of about 12 or 36 mm with choline or Li(+) as counter ion, respectively. Na(+) inhibited outward currents with a K(1/2) of about 60 mm. Inward currents generated by forward NCKX2 exchange depended upon external Na(+) with a K(1/2) of 30 mm and a Hill coefficient of 2.8. K(+) inhibited the inward currents by a maximum of 40%, with a K(1/2) of 2 mm or less, depending upon the conditions. The transport stoichiometry of NCKX2 was determined by observing the change in reversal potential as individual ion gradients were altered. Our data support a stoichiometry for rat brain NCKX2 of 4 Na(+):(1 Ca(2+) + 1 K(+)). These findings provide the first electrophysiological characterization of rat brain NCKX2, and the first evidence that a single recombinantly expressed NCKX polypeptide encodes a K(+)-transporting Na(+)/Ca(2+) exchanger with a transport stoichiometry of 4 Na(+):(1 Ca(2+) + 1 K(+)).


Subject(s)
Carrier Proteins/physiology , Sodium-Calcium Exchanger , Animals , Brain , Ion Transport/physiology , Patch-Clamp Techniques , Rats , Signal Transduction/physiology
12.
Proc Natl Acad Sci U S A ; 97(16): 9058-63, 2000 Aug 01.
Article in English | MEDLINE | ID: mdl-10908656

ABSTRACT

Potassium channels that are inhibited by internal ATP (K(ATP) channels) provide a critical link between metabolism and cellular excitability. Protein kinase C (PKC) acts on K(ATP) channels to regulate diverse cellular processes, including cardioprotection by ischemic preconditioning and pancreatic insulin secretion. PKC action decreases the Hill coefficient of ATP binding to cardiac K(ATP) channels, thereby increasing their open probability at physiological ATP concentrations. We show that PKC similarly regulates recombinant channels from both the pancreas and heart. Surprisingly, PKC acts via phosphorylation of a specific, conserved threonine residue (T180) in the pore-forming subunit (Kir6.2). Additional PKC consensus sites exist on both Kir and the larger sulfonylurea receptor (SUR) subunits. Nonetheless, T180 controls changes in open probability induced by direct PKC action either in the absence of, or in complex with, the accessory SUR1 (pancreatic) or SUR2A (cardiac) subunits. The high degree of conservation of this site among different K(ATP) channel isoforms suggests that this pathway may have wide significance for the physiological regulation of K(ATP) channels in various tissues and organelles.


Subject(s)
Adenosine Triphosphate/metabolism , Potassium Channels, Inwardly Rectifying , Potassium Channels/metabolism , Protein Kinase C/metabolism , Amino Acid Sequence , Animals , Cell Line, Transformed , Humans , Molecular Sequence Data , Phosphorylation , Potassium Channels/chemistry , Rabbits
13.
Circ Res ; 87(2): 112-7, 2000 Jul 21.
Article in English | MEDLINE | ID: mdl-10903994

ABSTRACT

ATP-sensitive K(+) channels (K(ATP)) contribute to the regulation of tone in vascular smooth muscle cells. We determined the effects of protein kinase C (PKC) activation on the nucleoside diphosphate-activated (K(NDP)) subtype of vascular smooth muscle K(ATP) channel. Phorbol 12,13-dibutyrate (PdBu) and angiotensin II inhibited K(NDP) activity of C-A patches of rabbit portal vein (PV) myocytes, but an inactive phorbol ester was without effect, and pretreatment with PKC inhibitor prevented the actions of PdBu. Constitutively active PKC inhibited K(NDP) in I-O patches but was without effect in the presence of a specific peptide inhibitor of PKC. PdBu increased the duration of a long-lived interburst closed state but was without effect on burst duration or intraburst kinetics. PdBu treatment inhibited K(NDP), but not a 70-pS K(ATP) channel of rat PV. The results indicate that the K(NDP) subtype of vascular smooth muscle K(ATP) channel is inhibited by activation of PKC. Control of K(NDP) activity by intracellular signaling cascades involving PKC may, therefore, contribute to control of tone and arterial diameter by vasoconstrictors.


Subject(s)
Muscle, Smooth, Vascular/physiology , Portal Vein/physiology , Potassium Channels/physiology , Protein Kinase C/metabolism , Adenosine Triphosphate/pharmacology , Angiotensin II/pharmacology , Animals , Cells, Cultured , Deoxyglucose/pharmacology , Kinetics , Membrane Potentials/drug effects , Membrane Potentials/physiology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Phorbol 12,13-Dibutyrate/pharmacology , Potassium Channel Blockers , Rabbits , Rats
14.
Can J Cardiol ; 15(10): 1123-30, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10523479

ABSTRACT

Ischemic preconditioning (IPC) is a naturally occurring protective mechanism in the heart and is triggered by brief ischemic insults preceding a sustained ischemic period. The artificial induction of the protective effects of IPC are potentially of great benefit to postinfarct patients and would serve to augment traditional surgical and thrombolytic treatment regimens. As yet, no agent has been developed to trigger IPC specifically. The mechanisms underlying IPC have been the focus of intense study, and many components of the IPC framework have been elucidated. Adenosine, noradrenalin, angiotensin II and endothelin are several of the key inducers of IPC. Signal transduction events downstream of receptor activation converging on protein kinase C stimulation are thought to trigger the intracellular pathways leading to 'preconditioning'. Although the complete sequence of events beneath IPC are not clear, several of the end effectors for the initiation of IPC are. For instance, activation of a class of potassium channels that are sensitive to internal ATP (KATP channels) can induce IPC and may be at least one of the targets for the action of protein kinase C. The precise mechanisms by which stimulation of KATP channels protects the myocardium are still under debate and are discussed in this review. It seems likely that both the plasma membrane and mitochondrial KATP channel populations are involved in IPC. Because KATP channels are rich and varied pharmacologically, it may be possible to target therapeutic agents at the cardiac channel isoform. Such compounds would likely meet the pharmacological criteria for a 'conditioning' agent.


Subject(s)
Adenosine Triphosphate/metabolism , Ischemic Preconditioning, Myocardial , Myocardial Ischemia/physiopathology , Female , Humans , Male , Neurotransmitter Agents/metabolism , Neurotransmitter Agents/pharmacology
15.
Cardiovasc Res ; 44(2): 356-69, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10690312

ABSTRACT

OBJECTIVE: Our goal was to identify the ATP-sensitive potassium (KATP) channels in cardiac Purkinje cells and to document the functional properties that might distinguish them from KATP channels in other parts of the heart. METHODS: Single Purkinje cells and ventricular myocytes were isolated from rabbit heart. Standard patch-clamp techniques were used to record action potential waveforms. and whole-cell and single-channel currents. RESULTS: The KATP channel opener levcromakalim (10 microM) caused marked shortening of the Purkinje cell action potential. Under whole-cell voltage-clamp, levcromakalim induced an outward current, which was blocked by glibenclamide (5 microM), in both Purkinje cells and ventricular myocytes. Metabolic poisoning of Purkinje cells with NaCN and 2-deoxyglucose caused a significant shortening of the action potential (control 376 +/- 51 ms; 6 min NaCN/2-deoxyglucose 153 +/- 21 ms). This effect was reversed with the application of glibenclamide. Inside-out membrane patches from Purkinje cells showed unitary current fluctuations which were inhibited by cytoplasmic ATP with an IC50 of 119 microM and a Hill coefficient of 2.1. This reflects approximately five-fold lower sensitivity to ATP inhibition than for KATP channels from ventricular myocytes under the same conditions. The slope conductance of Purkinje cell KATP channels, with symmetric, 140 mM K+, was 60.1 +/- 2.0 pS (mean +/- SEM). Single-channel fluctuations showed mean open and closed times of 3.6 +/- 1.5 ms and 0.41 +/- 0.2 ms, respectively, at -60 mV and approximately 21 degrees C. At positive potentials. KATP channels exhibited weak inward rectification that was dependent on the concentration of internal Mg2+. Computer simulations, based on the above results, predict significant shortening of the Purkinje cell action potential via activation of KATP channels in the range 1-5 mM cytoplasmic ATP. CONCLUSIONS: Purkinje cell KATP channels may represent a molecular isoform distinct from that present in ventricular myocytes. The presence of KATP channels in the Purkinje network suggests that they may have an important influence on cardiac rhythm and conduction during periods of ischemia.


Subject(s)
Action Potentials/drug effects , Adenosine Triphosphate/metabolism , Computer Simulation , Cromakalim/pharmacology , Potassium Channels/drug effects , Purkinje Fibers/metabolism , Adenosine Triphosphate/pharmacology , Animals , Arrhythmias, Cardiac/metabolism , Cell Separation , Deoxyglucose/pharmacology , Dose-Response Relationship, Drug , Glyburide/pharmacology , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Ion Channel Gating/drug effects , Male , Patch-Clamp Techniques , Purkinje Fibers/drug effects , Rabbits , Sodium Cyanide/pharmacology
16.
Am J Physiol ; 275(4): E568-76, 1998 10.
Article in English | MEDLINE | ID: mdl-9755074

ABSTRACT

The effects of streptozotocin-induced diabetes (5-7 days or 7 wk) on cardiac ATP-sensitive potassium channels (KATP channels) were investigated with the use of single-channel and action potential recordings from dissociated ventricular myocytes isolated from control and diabetic rat hearts. In inside-out patches from diabetic myocytes (5-7 days), the IC50 for ATP inhibition was 82 +/- 7.2 microM (mean +/- SE, n = 8), twice that in controls (43 +/- 3.6 microM, n = 12). For 7-wk diabetic rats, the IC50 was 75 +/- 2.3 microM (n = 6). Increasing internal ADP concentration attenuated ATP-induced inhibition in both controls and diabetics. On reducing the internal pH from 7.4 to 6.8, both control and diabetic myocytes showed a 1.7-fold increase in the IC50 for ATP inhibition. No differences were observed in either intraburst kinetics or unitary conductance of single channels from control and diabetic myocytes. In diabetic myocytes, action potential duration at 90% repolarization (APD90) was longer and more variable than in controls and was significantly shortened by application of the KATP channel opener cromakalim (50 microM). Cromakalim scarcely affected APD90 in controls. Computer simulation of the longer diabetic APD90 required a lower background conductance during the plateau phase in addition to small, measured changes in the delayed rectifier current, transient outward current, and ATP-sensitive K+ current (KATP current, IKATP). The simulations reproduced the enhanced sensitivity of the diabetic APD90 to changes in IKATP. These results have important implications for cardiac function in diabetics and their treatment by sulfonylureas.


Subject(s)
Adenosine Triphosphate/pharmacology , Diabetes Mellitus, Experimental/physiopathology , Heart/physiopathology , Potassium Channels/physiology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Cells, Cultured , Cromakalim/pharmacology , Electric Conductivity , Heart/physiology , Heart Ventricles , Patch-Clamp Techniques , Potassium Channel Blockers , Rats , Rats, Sprague-Dawley , Reference Values
17.
Circ Res ; 79(3): 399-406, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8781473

ABSTRACT

Activation of both ATP-sensitive K+ (KATP) channels and the enzyme protein kinase C (PKC) has been associated with the cardioprotective response of ischemic preconditioning. We recently showed that at low cytoplasmic ATP (< or = 50 mumol/L), PKC inhibits KATP channel activity. This finding is surprising, as both KATP channels and PKC are activated during preconditioning. However, PKC also altered ATP binding to the channel, changing the Hill coefficient from approximately 2 to approximately 1. This apparent change in stoichiometry would lead to a PKC-induced activation of KATP channels at more physiological (millimolar) levels of ATP. The aim of the present study was to determine whether PKC activates cardiac KATP channels at millimolar levels of ATP. The effects of PKC on single KATP channels were studied at millimolar internal ATP levels using excised inside-out membrane patches from rabbit ventricular myocytes. Application of purified constitutively active PKC (20 nmol/L) to the intracellular surface of the patches produced an approximately threefold increase in the channel open probability. The specific PKC inhibitor peptide PKC(19-31) prevented this increase. Heat-inactivated PKC had no effect on KATP channel properties. KATP channel activity spontaneously returned to control levels after washout of PKC. This spontaneous reversal did not occur in the presence of 5 nmol/L okadaic acid, suggesting that the reversal of PKC's action is dependent on activity of a membrane-associated type 2A protein phosphatase (PP2A). In the presence of exogenous PP2A (7.5 nmol/L), PKC had no effect. We conclude that the PKC-induced increase in KATP channel activity at millimolar ATP results from a crossing of the ATP concentration-response curves for inhibition of the phosphorylated and nonphosphorylated forms of the channel. This identifies a mechanism by which PKC activates KATP channels at near physiological levels of ATP and thus could link these two components in a signaling pathway that induces ischemic preconditioning.


Subject(s)
Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Myocardial Ischemia/physiopathology , Myocardial Reperfusion , Myocardium/metabolism , Potassium Channels/metabolism , Protein Kinase C/pharmacology , Animals , Chemical Phenomena , Chemistry , Mathematics , Osmolar Concentration , Patch-Clamp Techniques , Phosphoprotein Phosphatases/pharmacology , Potassium Channels/drug effects , Protein Kinase C/antagonists & inhibitors , Rabbits
18.
J Physiol ; 490 ( Pt 2): 337-50, 1996 Jan 15.
Article in English | MEDLINE | ID: mdl-8821133

ABSTRACT

1. Single myocytes were isolated from rabbit sino-atrial (SA) node by enzymatic dissociation. Spontaneous pacemaker activity, whole-cell and single-channel currents were recorded under conditions known to modulate ATP-sensitive K+ (KATP) channels. 2. The KATP channel openers, cromakalim and pinacidil, slowed or abolished the pacemaker activity, and caused hyperpolarization of the maximum diastolic potential (MDP). Glibenclamide, a KATP channel blocker, reversed these effects. Cromakalim- and pinacidil-activated currents reversed near the potassium equilibrium potential, EK. Glibenclamide had no effect on the L-type calcium current, ICa(L), the hyperpolarization-activated inward current, If, or the delayed rectifier potassium current, IK. 3. Sodium cyanide, which inhibits mitochondrial ATP production, induced a macroscopic current that reversed near EK and was blocked by glibenclamide. 4. In excised, inside-out patches from SA node cells, single KATP channels showed a slope conductance of 52 +/- 8 pS (mean +/- S.D.) when measurements were made at negative voltages in symmetric, 140 mM K+. Channels from ventricular myocytes showed a somewhat larger slope conductance (70 +/- 5 pS). 5. Raising the intracellular ATP concentration caused a concentration-dependent reduction in the open probability of the KATP channels (IC50, 16 microM; Hill coefficient, approximately 1; at both pH 7.4 and 6.8). 6. In excised inside-out patches, cromakalim or pinacidil induced significant increases in KATP channel activity in the presence of 50 microM or 1 mM intracellular ATP. This channel activity was blocked by glibenclamide. 7. Our results suggest that sino-atrial node cells express a distinct isoform of KATP channel which may play an important role in pharmacological and pathophysiological modulation of pacemaker activity.


Subject(s)
Potassium Channels/metabolism , Potassium/metabolism , Sinoatrial Node/metabolism , Action Potentials/drug effects , Adenosine Triphosphate/pharmacology , Animals , Benzopyrans/pharmacology , Cromakalim , Glyburide/pharmacology , Guanidines/pharmacology , Hydrogen-Ion Concentration , Kinetics , Myocardium/metabolism , Patch-Clamp Techniques , Pinacidil , Potassium Channel Blockers , Potassium Channels/drug effects , Pyrroles/pharmacology , Rabbits , Sinoatrial Node/cytology , Sodium Cyanide/pharmacology , Vasodilator Agents/pharmacology
19.
Biochemistry ; 34(21): 7252-7, 1995 May 30.
Article in English | MEDLINE | ID: mdl-7766636

ABSTRACT

Myocytes from rabbit ventricle were enzymatically dissociated and the effects of protein kinase C (PKC) on the properties of single ATP-sensitive (KATP) channels were studied using excised inside-out membrane patches. Application of a purified, constitutively active form of PKC (20 nM) to the intracellular surface of inside-out patches caused a 48% +/- 4% (n = 18) reduction in the open probability of single KATP channels. In the presence of the PKC inhibitors peptide PKC(19-31) or chelerythrine chloride, PKC had no effect on KATP channel properties. Heat-inactivated PKC had no effect on channel properties. KATP channel activity returned spontaneously after removal of PKC. However, application of okadaic acid, at a concentration (5 nM) appropriate for specific inhibition of type 2A protein phosphatase (PP-2A), after removal of PKC, prevented spontaneous recovery of channel activity. Treatment with purified PP-2A during the PKC-mediated inhibition of KATP channel activity caused a partial or full restoration of activity. The Hill coefficient for ATP binding was reduced from 2.2 (control) to 1.2 in the presence of PKC. The apparent inhibition constant (Ki) for ATP was unaffected by PKC [Ki(control) = 21 microM; Ki(PKC) = 20 microM]. PKC is, therefore, capable of inhibiting cardiac KATP channel activity, and the extent to which the channels remain phosphorylated appears to be dependent on membrane-associated PP-2A activity. These enzymes may, therefore, be involved in signal transduction mechanisms which serve to regulate the activity of cardiac KATP channels.


Subject(s)
Adenosine Triphosphate/metabolism , Myocardium/metabolism , Phosphoprotein Phosphatases/metabolism , Potassium Channels/metabolism , Protein Kinase C/metabolism , Animals , Heart Ventricles , Potassium Channel Blockers , Protein Kinase C/antagonists & inhibitors , Rabbits
20.
Eur J Pharmacol ; 259(3): 219-22, 1994 Jul 11.
Article in English | MEDLINE | ID: mdl-7982447

ABSTRACT

Glibenclamide, a blocker of ATP-sensitive K+ (KATP) channels, was tested on three different types of rat skeletal K+ channels incorporated into bilayers. Glibenclamide (10 microM) blocked a class of KATP channels (unitary conductance of 57 pS in symmetric 150 mM KCl), which were inhibited by ATP. High concentrations of glibenclamide (100 microM) had no effect on either voltage-gated K+ channels (37 pS), or Ca(2+)-activated K+ channels (210 pS). Our results show that glibenclamide, even at high concentrations (100 microM) that may be required for quick action in whole muscle experiments, is a selective and specific blocker of skeletal KATP channels.


Subject(s)
Adenosine Triphosphate/pharmacology , Glyburide/pharmacology , Muscle, Skeletal/metabolism , Potassium Channels/metabolism , Animals , Calcium/physiology , In Vitro Techniques , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Lipid Bilayers , Muscle, Skeletal/drug effects , Potassium Channels/drug effects , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...