Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Physiol ; 601(1): 83-98, 2023 01.
Article in English | MEDLINE | ID: mdl-36420836

ABSTRACT

Autosomal dominant polycystic kidney disease is caused by mutations in the membrane receptor PKD1 or the cation channel PKD2. TACAN (also termed TMEM120A), recently reported as an ion channel in neurons for mechanosensing and pain sensing, is also distributed in diverse non-neuronal tissues, such as kidney, heart and intestine, suggesting its involvement in other functions. In this study, we found that TACAN is in a complex with PKD2 in native renal cell lines. Using the two-electrode voltage clamp in Xenopus oocytes, we found that TACAN inhibits the channel activity of PKD2 gain-of-function mutant F604P. TACAN fragments containing the first and last transmembrane domains interacted with the PKD2 C- and N-terminal fragments, respectively. The TACAN N-terminus acted as a blocking peptide, and TACAN inhibited the function of PKD2 by the binding of PKD2 with TACAN. By patch clamping in mammalian cells, we found that TACAN inhibits both the single-channel conductance and the open probability of PKD2 and mutant F604P. PKD2 co-expressed with TACAN, but not PKD2 alone, exhibited pressure sensitivity. Furthermore, we found that TACAN aggravates PKD2-dependent tail curvature and pronephric cysts in larval zebrafish. In summary, this study revealed that TACAN acts as a PKD2 inhibitor and mediates mechanosensitivity of the PKD2-TACAN channel complex. KEY POINTS: TACAN inhibits the function of PKD2 in vitro and in vivo. TACAN N-terminal S1-containing fragment T160X interacts with the PKD2 C-terminal fragment N580-L700, and its C-terminal S6-containing fragment L296-D343 interacts with the PKD2 N-terminal A594X. TACAN inhibits the function of the PKD2 channel by physical interaction. The complex of PKD2 with TACAN, but not PKD2 alone, confers mechanosensitivity.


Subject(s)
Polycystic Kidney, Autosomal Dominant , Zebrafish , Animals , TRPP Cation Channels/genetics , TRPP Cation Channels/metabolism , Ion Channels/genetics , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/metabolism , Kidney/metabolism , Mammals/metabolism
2.
J Mol Cell Cardiol ; 167: 17-31, 2022 06.
Article in English | MEDLINE | ID: mdl-35331696

ABSTRACT

Sodium glucose cotransporter 2 inhibitors (SGLT2i) constitute a promising drug treatment for heart failure patients with either preserved or reduced ejection fraction. Whereas SGLT2i were originally developed to target SGLT2 in the kidney to facilitate glucosuria in diabetic patients, it is becoming increasingly clear that these drugs also have important effects outside of the kidney. In this review we summarize the literature on cardiac effects of SGLT2i, focussing on pro-inflammatory and oxidative stress processes, ion transport mechanisms controlling sodium and calcium homeostasis and metabolic/mitochondrial pathways. These mechanisms are particularly important as disturbances in these pathways result in endothelial dysfunction, diastolic dysfunction, cardiac stiffness, and cardiac arrhythmias that together contribute to heart failure. We review the findings that support the concept that SGLT2i directly and beneficially interfere with inflammation, oxidative stress, ionic homeostasis, and metabolism within the cardiac cell. However, given the very low levels of SGLT2 in cardiac cells, the evidence suggests that SGLT2-independent effects of this class of drugs likely occurs via off-target effects in the myocardium. Thus, while there is still much to be understood about the various factors which determine how SGLT2i affect cardiac cells, much of the research clearly demonstrates that direct cardiac effects of these SGLT2i exist, albeit mediated via SGLT2-independent pathways, and these pathways may play a role in explaining the beneficial effects of SGLT2 inhibitors in heart failure.


Subject(s)
Diabetes Mellitus, Type 2 , Heart Failure , Sodium-Glucose Transporter 2 Inhibitors , Humans , Myocardium/metabolism , Sodium-Glucose Transporter 2/metabolism , Sodium-Glucose Transporter 2/therapeutic use , Sodium-Glucose Transporter 2 Inhibitors/adverse effects
3.
Circulation ; 143(22): 2188-2204, 2021 06.
Article in English | MEDLINE | ID: mdl-33832341

ABSTRACT

BACKGROUND: SGLT2 (sodium/glucose cotransporter 2) inhibitors exert robust cardioprotective effects against heart failure in patients with diabetes, and there is intense interest to identify the underlying molecular mechanisms that afford this protection. Because the induction of the late component of the cardiac sodium channel current (late-INa) is involved in the etiology of heart failure, we investigated whether these drugs inhibit late-INa. METHODS: Electrophysiological, in silico molecular docking, molecular, calcium imaging, and whole heart perfusion techniques were used to address this question. RESULTS: The SGLT2 inhibitor empagliflozin reduced late-INa in cardiomyocytes from mice with heart failure and in cardiac Nav1.5 sodium channels containing the long QT syndrome 3 mutations R1623Q or ΔKPQ. Empagliflozin, dapagliflozin, and canagliflozin are all potent and selective inhibitors of H2O2-induced late-INa (half maximal inhibitory concentration = 0.79, 0.58, and 1.26 µM, respectively) with little effect on peak sodium current. In mouse cardiomyocytes, empagliflozin reduced the incidence of spontaneous calcium transients induced by the late-INa activator veratridine in a similar manner to tetrodotoxin, ranolazine, and lidocaine. The putative binding sites for empagliflozin within Nav1.5 were investigated by simulations of empagliflozin docking to a three-dimensional homology model of human Nav1.5 and point mutagenic approaches. Our results indicate that empagliflozin binds to Nav1.5 in the same region as local anesthetics and ranolazine. In an acute model of myocardial injury, perfusion of isolated mouse hearts with empagliflozin or tetrodotoxin prevented activation of the cardiac NLRP3 (nuclear-binding domain-like receptor 3) inflammasome and improved functional recovery after ischemia. CONCLUSIONS: Our results provide evidence that late-INa may be an important molecular target in the heart for the SGLT2 inhibitors, contributing to their unexpected cardioprotective effects.


Subject(s)
Benzhydryl Compounds/pharmacology , Glucosides/pharmacology , Sodium Channels/drug effects , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Animals , Benzhydryl Compounds/therapeutic use , Glucosides/therapeutic use , Humans , Male , Mice , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use
4.
J Physiol ; 598(19): 4321-4338, 2020 10.
Article in English | MEDLINE | ID: mdl-32721035

ABSTRACT

KEY POINTS: 25-Hydroxyvitamin D (25OHD) is a partial agonist of TRPV1 whereby 25OHD can weakly activate TRPV1 yet antagonize the stimulatory effects of the full TRPV1 agonists capsaicin and oleoyl dopamine. 25OHD binds to TRPV1 within the same vanilloid binding pocket as capsaicin. 25OHD inhibits the potentiating effects of PKC-mediated TRPV1 activity. 25OHD reduces T-cell activation and trigeminal neuron calcium signalling mediated by TRPV1 activity. These results provide evidence that TRPV1 is a novel receptor for the biological actions of vitamin D in addition to the well-documented effects of vitamin D upon the nuclear vitamin D receptor. The results may have important implications for our current understanding of certain diseases where TRPV1 and vitamin D deficiency have been implicated, such as chronic pain and autoimmune diseases, such as type 1 diabetes. ABSTRACT: The capsaicin receptor TRPV1 plays an important role in nociception, inflammation and immunity and its activity is regulated by exogenous and endogenous lipophilic ligands. As vitamin D is lipophilic and involved in similar biological processes as TRPV1, we hypothesized that it directly regulates TRPV1 activity and function. Our calcium imaging and electrophysiological data demonstrate that vitamin D (25-hydroxyvitamin D (25OHD) and 1,25-hydroxyvitamin D (1,25OHD)) can weakly activate TRPV1 at physiologically relevant concentrations (100 nM). Furthermore, both 25OHD and 1,25OHD can inhibit capsaicin-induced TRPV1 activity (IC50  = 34.3 ± 0.2 and 11.5 ± 0.9 nM, respectively), but not pH-induced TRPV1 activity, suggesting that vitamin D interacts with TRPV1 in the same region as the TRPV1 agonist capsaicin. This hypothesis is supported by our in silico TRPV1 structural modelling studies, which place 25OHD in the same binding region as capsaicin. 25OHD also attenuates PKC-dependent TRPV1 potentiation via interactions with a known PKC phospho-acceptor residue in TRPV1. To provide evidence for a physiological role for the interaction of vitamin D with TRPV1, we employed two different cellular models known to express TRPV1: mouse CD4+ T-cells and trigeminal neurons. Our results indicate that 25OHD reduces TRPV1-induced cytokine release from T-cells and capsaicin-induced calcium activity in trigeminal neurons. In summary, we provide evidence that vitamin D is a novel endogenous regulator of TRPV1 channel activity that may play an important physiological role in addition to its known effects through the canonical nuclear vitamin D receptor pathway.


Subject(s)
Transient Receptor Potential Channels , Animals , Capsaicin/pharmacology , Mice , Neurons , Rats, Sprague-Dawley , TRPV Cation Channels , Vitamin D/pharmacology
5.
J Mol Cell Cardiol ; 108: 34-41, 2017 07.
Article in English | MEDLINE | ID: mdl-28483598

ABSTRACT

Cardiac ATP-sensitive K+ (KATP) channel activity plays an important cardio-protective role in regulating excitability in response to metabolic stress. Evidence suggests that these channels are also mechano-sensitive and therefore may couple KATP channel activity to increased cardiac workloads. However, the molecular mechanism that couples membrane stretch to channel activity is not currently known. We hypothesized that membrane stretch may alter the intrinsic MgATPase activity of the cardiac KATP channel resulting in increased channel activation. The inside-out patch-clamp technique was used to record single-channel and macroscopic recombinant KATP channel activity in response to membrane stretch elicited by negative pipette pressure. We found that stretch activation requires the presence of the SUR subunit and that inhibition of MgATPase activity with either the non-hydrolysable ATP analog AMP-PNP or the ATPase inhibitor BeFx significantly reduced the stimulatory effect of stretch. We employed a point mutagenic approach to determine that a single residue (K1337) in the hairpin loop proximal to the major MgATPase catalytic site in the SUR2A subunit is responsible for the difference in mechano-sensitivity between SUR2A and SUR1 containing KATP channels. Moreover, using a double cysteine mutant substitution in the hairpin loop region revealed the importance of a key residue-residue interaction in this region that transduces membrane mechanical forces into KATP channel stimulation via increases in channel MgATPase activity. With respect to KATP channel pharmacology, glibenclamide, but not glicalizide or repaglinide, was able to completely inhibit KATP channel mechano-sensitivity. In summary, our results provide a highly plausible molecular mechanism by which mechanical membrane forces are rapidly converted in changes in KATP channel activity that have implications for our understanding of cardiac KATP channels in physiological or pathophysiological settings that involve increased workload.


Subject(s)
Adenosine Triphosphatases/metabolism , KATP Channels/metabolism , Mechanotransduction, Cellular , Myocardial Contraction , Myocardium/metabolism , Adenosine Triphosphatases/antagonists & inhibitors , Amino Acid Sequence , Amino Acid Substitution , Enzyme Activation , Guanosine Triphosphate/metabolism , HEK293 Cells , Humans , Ion Channel Gating , KATP Channels/chemistry , KATP Channels/genetics , Models, Molecular , Protein Binding , Protein Conformation , Protein Isoforms , Protein Subunits , Structure-Activity Relationship , Sulfonylurea Receptors/chemistry , Sulfonylurea Receptors/genetics , Sulfonylurea Receptors/metabolism
7.
Am J Physiol Heart Circ Physiol ; 313(3): H469-H478, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28667052

ABSTRACT

Cardiac ATP-sensitive K+ (KATP) channels couple changes in cellular metabolism to membrane excitability and are activated during metabolic stress, although under basal aerobic conditions, KATP channels are thought to be predominately closed. Despite intense research into the roles of KATP channels during metabolic stress, their contribution to aerobic basal cardiac metabolism has not been previously investigated. Hearts from Kir6.2+/+ and Kir6.2-/- mice were perfused in working mode, and rates of glycolysis, fatty acid oxidation, and glucose oxidation were measured. Changes in activation/expression of proteins regulating metabolism were probed by Western blot analysis. Despite cardiac mechanical function and metabolic efficiency being similar in both groups, hearts from Kir6.2-/- mice displayed an approximately twofold increase in fatty acid oxidation and a 0.45-fold reduction in glycolytic rates but similar glucose oxidation rates compared with hearts from Kir6.2+/+ mice. Kir6.2-/- hearts also possessed elevated levels of activated AMP-activated protein kinase (AMPK), higher glycogen content, and reduced mitochondrial density. Moreover, activation of AMPK by isoproterenol or diazoxide was significantly blunted in Kir6.2-/- hearts. These data indicate that KATP channel ablation alters aerobic basal cardiac metabolism. The observed increase in fatty acid oxidation and decreased glycolysis before any metabolic insult may contribute to the poor recovery observed in Kir6.2-/- hearts in response to exercise or ischemia-reperfusion injury. Therefore, KATP channels may play an important role in the regulation of cardiac metabolism through AMPK signaling.NEW & NOTEWORTHY In this study, we show that genetic ablation of plasma membrane ATP-sensitive K+ channels results in pronounced changes in cardiac metabolic substrate preference and AMP-activated protein kinase activity. These results suggest that ATP-sensitive K+ channels may play a novel role in regulating metabolism in addition to their well-documented effects on ionic homeostasis during periods of stress.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Cell Membrane/enzymology , Energy Metabolism , Myocytes, Cardiac/enzymology , Potassium Channels, Inwardly Rectifying/deficiency , Animals , Cell Membrane/drug effects , Cell Membrane/ultrastructure , Energy Metabolism/drug effects , Enzyme Activation , Enzyme Activators/pharmacology , Fatty Acids/metabolism , Genotype , Glucose/metabolism , Glycolysis , Isolated Heart Preparation , Kinetics , Male , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Heart/enzymology , Myocardial Contraction , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/ultrastructure , Oxidation-Reduction , Phenotype , Potassium Channels, Inwardly Rectifying/genetics , Time Factors
8.
Biochim Biophys Acta Gen Subj ; 1861(2): 313-322, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27871838

ABSTRACT

BACKGROUND: Although insulin resistance (IR) is a key factor in the pathogenesis of type 2 diabetes (T2D), the precise role of insulin in the development of IR remains unclear. Therefore, we investigated whether chronic basal insulin infusion is causative in the development of glucose intolerance. METHODS: Normoglycemic lean rats surgically instrumented with i.v. catheters were infused with insulin (3mU/kg/min) or physiological saline for 6weeks. At infusion-end, plasma insulin levels along with glucose tolerance were assessed. RESULTS: Six weeks of insulin infusion induced glucose intolerance and impaired insulin response in healthy rats. Interestingly, the effects of chronic insulin infusion were completely normalized following 24h withdrawal of exogenous insulin and plasma insulin response to glucose challenge was enhanced, suggesting improved insulin secretory capacity. As a result of this finding, we assessed whether the effects of insulin therapy followed by a washout could ameliorate established glucose intolerance in obese rats. Obese rats were similarly instrumented and infused with insulin or physiological saline for 7days followed by 24h washout. Seven day-insulin therapy in obese rats significantly improved glucose tolerance, which was attributed to improved insulin secretory capacity and improved insulin signaling in liver and skeletal muscle. CONCLUSION: Moderate infusion of insulin alone is sufficient to cause glucose intolerance and impair endogenous insulin secretory capacity, whereas short-term, intensive insulin therapy followed by insulin removal effectively improves glucose tolerance, insulin response and peripheral insulin sensitivity in obese rats. GENERAL SIGNIFICANCE: New insight into the link between insulin and glucose intolerance may optimize T2D management.


Subject(s)
Blood Glucose/drug effects , Glucose/metabolism , Insulin/administration & dosage , Obesity/blood , Obesity/metabolism , Thinness/blood , Thinness/metabolism , Animals , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Glucose Intolerance/blood , Glucose Tolerance Test/methods , Insulin Resistance/physiology , Liver/drug effects , Liver/metabolism , Male , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Rats , Rats, Sprague-Dawley
10.
J Pharmacol Exp Ther ; 351(1): 164-71, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25118217

ABSTRACT

The trace amine ß-phenylethylamine (PEA) is normally present in the body at low nanomolar concentrations but can reach micromolar levels after ingestion of drugs that inhibit monoamine oxidase and primary amine oxidase. In vivo, PEA elicits a robust pressor response, but there is no consensus regarding the underlying mechanism, with both vasodilation and constriction reported in isolated blood vessels. Using functional and biochemical approaches, we found that at low micromolar concentrations PEA (1-30 µM) enhanced nerve-evoked vasoconstriction in the perfused rat mesenteric bed but at a higher concentration (100 µM) significantly inhibited these responses. The α2-adrenoceptor antagonist rauwolscine (1 µM) also enhanced nerve-mediated vasoconstriction, but in the presence of both rauwolscine (1 µM) and PEA (30 µM) together, nerve-evoked responses were initially potentiated and then showed time-dependent rundown. PEA (10 and 100 µM) significantly increased noradrenaline outflow from the mesenteric bed as determined by high-pressure liquid chromatography coupled with electrochemical detection. In isolated endothelium-denuded arterial segments, PEA (1 µM to 1 mM) caused concentration-dependent reversal of tone elicited by the α1-adrenoceptor agonists noradrenaline (EC50 51.69 ± 10.8 µM; n = 5), methoxamine (EC50 68.21 ± 1.70 µM; n = 5), and phenylephrine (EC50 67.74 ± 16.72 µM; n = 5) but was ineffective against tone induced by prostaglandin F2 α or U46619 (9,11-dideoxy-9α,11α-methanoepoxyprostaglandin F2 α). In rat brain homogenates, PEA displaced binding of both [(3)H]prazosin (Ki ≈ 25 µM) and [(3)H]rauwolscine (Ki ≈ 1.2 µM), ligands for α1- and α2-adrenoceptors, respectively. These data provide the first demonstration that dual indirect sympathomimetic and α1-adrenoceptor blocking actions underlie the vascular effects of PEA in resistance arteries.


Subject(s)
Adrenergic alpha-1 Receptor Antagonists/pharmacology , Mesenteric Arteries/drug effects , Phenethylamines/pharmacology , Receptors, Adrenergic, alpha-1/metabolism , Vasoconstriction/drug effects , Adrenergic alpha-1 Receptor Agonists/pharmacology , Adrenergic alpha-1 Receptor Antagonists/pharmacokinetics , Adrenergic alpha-2 Receptor Antagonists/pharmacology , Animals , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiology , In Vitro Techniques , Male , Mesenteric Arteries/innervation , Mesenteric Arteries/physiology , Phenethylamines/pharmacokinetics , Protein Binding , Rats , Rats, Sprague-Dawley , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/physiology , Yohimbine/pharmacology
11.
BMC Cardiovasc Disord ; 14: 91, 2014 Jul 25.
Article in English | MEDLINE | ID: mdl-25063541

ABSTRACT

BACKGROUND: Nationally, symptomatic heart failure affects 1.5-2% of Canadians, incurs $3 billion in hospital costs annually and the global burden is expected to double in the next 1-2 decades. The current one-year mortality rate after diagnosis of heart failure remains high at >25%. Consequently, new therapeutic strategies need to be developed for this debilitating condition. METHODS/DESIGN: The objective of the Alberta HEART program (http://albertaheartresearch.ca) is to develop novel diagnostic, therapeutic and prognostic approaches to patients with heart failure with preserved ejection fraction. We hypothesize that novel imaging techniques and biomarkers will aid in describing heart failure with preserved ejection fraction. Furthermore, the development of new diagnostic criteria will allow us to: 1) better define risk factors associated with heart failure with preserved ejection fraction; 2) elucidate clinical, cellular and molecular mechanisms involved with the development and progression of heart failure with preserved ejection fraction; 3) design and test new therapeutic strategies for patients with heart failure with preserved ejection fraction. Additionally, Alberta HEART provides training and education for enhancing translational medicine, knowledge translation and clinical practice in heart failure. This is a prospective observational cohort study of patients with, or at risk for, heart failure. Patients will have sequential testing including quality of life and clinical outcomes over 12 months. After that time, study participants will be passively followed via linkage to external administrative databases. Clinical outcomes of interest include death, hospitalization, emergency department visits, physician resource use and/or heart transplant. Patients will be followed for a total of 5 years. DISCUSSION: Alberta HEART has the primary objective to define new diagnostic criteria for patients with heart failure with preserved ejection fraction. New criteria will allow for targeted therapies, diagnostic tests and further understanding of the patients, both at-risk for and with heart failure. TRIAL REGISTRATION: ClinicalTrials.gov NCT02052804.


Subject(s)
Diagnostic Imaging , Heart Failure/diagnosis , Heart Failure/therapy , Research Design , Alberta/epidemiology , Biomarkers/blood , Diagnostic Imaging/methods , Emergency Service, Hospital/statistics & numerical data , Health Resources/statistics & numerical data , Heart Failure/blood , Heart Failure/etiology , Heart Failure/mortality , Heart Transplantation/statistics & numerical data , Hospitalization , Humans , Office Visits/statistics & numerical data , Predictive Value of Tests , Prospective Studies , Risk Assessment , Risk Factors , Stroke Volume , Time Factors , Treatment Outcome , Ventricular Function, Left
12.
Nature ; 452(7190): 1012-6, 2008 Apr 24.
Article in English | MEDLINE | ID: mdl-18401341

ABSTRACT

Energy and glucose homeostasis are regulated by food intake and liver glucose production, respectively. The upper intestine has a critical role in nutrient digestion and absorption. However, studies indicate that upper intestinal lipids inhibit food intake as well in rodents and humans by the activation of an intestine-brain axis. In parallel, a brain-liver axis has recently been proposed to detect blood lipids to inhibit glucose production in rodents. Thus, we tested the hypothesis that upper intestinal lipids activate an intestine-brain-liver neural axis to regulate glucose homeostasis. Here we demonstrate that direct administration of lipids into the upper intestine increased upper intestinal long-chain fatty acyl-coenzyme A (LCFA-CoA) levels and suppressed glucose production. Co-infusion of the acyl-CoA synthase inhibitor triacsin C or the anaesthetic tetracaine with duodenal lipids abolished the inhibition of glucose production, indicating that upper intestinal LCFA-CoAs regulate glucose production in the preabsorptive state. Subdiaphragmatic vagotomy or gut vagal deafferentation interrupts the neural connection between the gut and the brain, and blocks the ability of upper intestinal lipids to inhibit glucose production. Direct administration of the N-methyl-d-aspartate ion channel blocker MK-801 into the fourth ventricle or the nucleus of the solitary tract where gut sensory fibres terminate abolished the upper-intestinal-lipid-induced inhibition of glucose production. Finally, hepatic vagotomy negated the inhibitory effects of upper intestinal lipids on glucose production. These findings indicate that upper intestinal lipids activate an intestine-brain-liver neural axis to inhibit glucose production, and thereby reveal a previously unappreciated pathway that regulates glucose homeostasis.


Subject(s)
Brain/metabolism , Dietary Fats/pharmacology , Glucose/biosynthesis , Intestinal Mucosa/metabolism , Lipid Metabolism , Liver/metabolism , Acyl Coenzyme A/biosynthesis , Acyl Coenzyme A/metabolism , Animals , Brain/drug effects , Dietary Fats/administration & dosage , Dietary Fats/metabolism , Fatty Acids/chemistry , Fatty Acids/metabolism , Glucose/metabolism , Homeostasis/drug effects , Insulin/metabolism , Intestines/drug effects , Intestines/innervation , Liver/drug effects , Liver/innervation , Rats , Satiety Response/drug effects , Tetracaine/pharmacology , Triazenes/pharmacology
13.
Can J Physiol Pharmacol ; 91(4): 316-24, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23627843

ABSTRACT

Triton X-100 (TX-100) is a nonionic detergent frequently used at millimolar concentrations to disrupt cell membranes and solubilize proteins. At low micromolar concentrations, TX-100 has been reported to inhibit the function of potassium channels. Here, we have used electrophysiological and functional techniques to examine the effects of TX-100 on another class of ion channels, L-type voltage-operated calcium channels (VOCCs). TX-100 (30 nmol·L(-1) to 3 µmol·L(-1)) caused reversible concentration-dependent inhibition of recombinant L-type VOCC (CaV 1.2) currents and of native L-type VOCC currents recorded from rat vascular smooth muscle cells and cardiac myocytes, and murine and human pancreatic ß-cells. In functional studies, TX-100 (165 nmol·L(-1) to 3.4 µmol·L(-1)) caused concentration-dependent relaxation of rat isolated mesenteric resistance arteries prestimulated with phenylephrine or KCl. This effect was independent of the endothelium. TX-100 (1.6 µmol·L(-1)) inhibited depolarization-induced exocytosis in both murine and human isolated pancreatic ß-cells. These data indicate that at concentrations within the nanomolar to low micromolar range, TX-100 significantly inhibits L-type VOCC activity in a number of cell types, an effect paralleled by inhibition of cell functions dependent upon activation of these channels. This inhibition occurs at concentrations below those used to solubilize proteins and may compromise the use of solutions containing TX-100 in bioassays.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium/metabolism , Endothelium, Vascular/drug effects , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Octoxynol/pharmacology , Animals , Cell Line , Endothelium, Vascular/metabolism , Exocytosis/drug effects , HEK293 Cells , Humans , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/metabolism , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Smooth Muscle/metabolism , Phenylephrine/pharmacology , Potassium Chloride/pharmacology , Rats , Rats, Sprague-Dawley , Vasodilation/drug effects
14.
Biosens Bioelectron ; 241: 115668, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37774465

ABSTRACT

Continuous glucose monitoring schemes that avoid finger pricking are of utmost importance to enhance the comfort and lifestyle of diabetic patients. To this aim, we propose a microwave planar sensing platform as a potent sensing technology that extends its applications to biomedical analytes. In this paper, a compact planar resonator-based sensor is introduced for noncontact sensing of glucose. Furthermore, in vivo and in-vitro tests using a microfluidic channel system and in clinical trial settings demonstrate its reliable operation. The proposed sensor offers real-time response and a high linear correlation (R2 ∼ 0.913) between the measured sensor response and the blood glucose level (GL). The sensor is also enhanced with machine learning to predict the variation of body glucose levels for non-diabetic and diabetic patients. This addition is instrumental in triggering preemptive measures in cases of unusual glucose level trends. In addition, it allows for the detection of common artifacts of the sensor as anomalies so that they can be removed from the measured data. The proposed system is designed to noninvasively monitor interstitial glucose levels in humans, introducing the opportunity to create a customized wearable apparatus with the ability to learn.


Subject(s)
Biosensing Techniques , Diabetes Mellitus , Humans , Blood Glucose , Blood Glucose Self-Monitoring , Microwaves , Glucose , Diabetes Mellitus/diagnosis , Machine Learning
15.
J Mol Cell Cardiol ; 53(1): 43-52, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22561102

ABSTRACT

AIMS: Epoxyeicosatrienoic acids (EETs) are cytochrome P450 epoxygenase metabolites of arachidonic acid that have known cardioprotective properties. While the mechanism(s) remains unknown, evidence suggests that phosphoinositide 3-kinase (PI3K) and sarcolemmal ATP-sensitive potassium channels (pmK(ATP)) are important. However the role of specific PI3K isoforms and corresponding intracellular mechanisms remains unknown. METHODS AND RESULTS: To study this, mice hearts were perfused in Langendorff mode for 40 min of baseline and subjected to 20 or 30 min of global no-flow ischemia followed by 40 min of reperfusion. C57BL6 mice perfused with 11,12-EET (1 µM) had improved postischemic recovery, whereas co-perfusion with PI3Kα inhibitor, PI-103 (0.1 µM), abolished the EET-mediated effect. In contrast, blocking of PI3Kß or PI3Kγ isoforms failed to inhibit EET-mediated cardioprotection. In addition to the improved post-ischemic recovery, increased levels of p-Akt, decreased calcineurin activity and decreased translocation of proapoptotic protein BAD to mitochondria were noted in EET-treated hearts. Perfusion of 11,12-EET to Kir6.2 deficient mice (pmK(ATP)) failed to improve postischemic recovery, decrease calcineurin activity and translocation of proapoptotic protein BAD, however increased levels of p-Akt were still observed. Patch-clamp experiments demonstrated that 11,12-EET could not activate pmK(ATP) currents in myocytes pre-treated with PI-103. Mechanistic studies in H9c2 cells demonstrate that 11,12-EET limits anoxia-reoxygenation triggered Ca(2+) accumulation and maintains mitochondrial ΔΨm compared to controls. Both PI-103 and glibenclamide (10 µM, pmK(ATP) inhibitor) abolished EET cytoprotection. CONCLUSION: Together our data suggest that EET-mediated cardioprotection involves activation of PI3Kα, upstream of pmK(ATP), which prevents Ca(2+) overload and maintains mitochondrial function.


Subject(s)
8,11,14-Eicosatrienoic Acid/analogs & derivatives , Cardiotonic Agents/pharmacology , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Heart/drug effects , KATP Channels/metabolism , Myocardium/metabolism , Sarcolemma/metabolism , 8,11,14-Eicosatrienoic Acid/pharmacology , Animals , Calcium/metabolism , Cell Line , Hypoxia , Isoenzymes/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/physiopathology , Myocardial Reperfusion Injury/prevention & control , Myocardium/enzymology , Sarcolemma/enzymology
16.
Pharmacogenet Genomics ; 22(3): 206-14, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22209866

ABSTRACT

OBJECTIVES: The common ATP-sensitive potassium (KATP) channel variants E23K and S1369A, found in the KCNJ11 and ABCC8 genes, respectively, form a haplotype that is associated with an increased risk for type 2 diabetes. Our previous studies showed that KATP channel inhibition by the A-site sulfonylurea gliclazide was increased in the K23/A1369 haplotype. Therefore, we studied the pharmacogenomics of seven clinically used sulfonylureas and glinides to determine their structure-activity relationships in KATP channels containing either the E23/S1369 nonrisk or K23/A1369 risk haplotypes. RESEARCH DESIGN AND METHODS: The patch-clamp technique was used to determine sulfonylurea and glinide inhibition of recombinant human KATP channels containing either the E23/S1369 or the K23/A1369 haplotype. RESULTS: KATP channels containing the K23/A1369 risk haplotype were significantly less sensitive to inhibition by tolbutamide, chlorpropamide, and glimepiride (IC50 values for K23/A1369 vs. E23/S1369=1.15 vs. 0.71 µmol/l; 4.19 vs. 3.04 µmol/l; 4.38 vs. 2.41 nmol/l, respectively). In contrast, KATP channels containing the K23/A1369 haplotype were significantly more sensitive to inhibition by mitiglinide (IC50=9.73 vs. 28.19 nmol/l for K23/A1369 vs. E23/S1369) and gliclazide. Nateglinide, glipizide, and glibenclamide showed similar inhibitory profiles in KATP channels containing either haplotype. CONCLUSION: Our results demonstrate that the ring-fused pyrrole moiety in several A-site drugs likely underlies the observed inhibitory potency of these drugs on KATP channels containing the K23/A1369 risk haplotype. It may therefore be possible to tailor existing therapy or design novel drugs that display an increased efficacy in type 2 diabetes patients homozygous for these common KATP channel haplotypes.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Chlorpropamide/pharmacokinetics , Gene Expression Regulation/drug effects , Potassium Channels, Inwardly Rectifying/genetics , Receptors, Drug/genetics , Tolbutamide/pharmacokinetics , ATP-Binding Cassette Transporters/antagonists & inhibitors , Chlorpropamide/administration & dosage , Cyclohexanes/administration & dosage , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Gliclazide/administration & dosage , Glyburide/administration & dosage , Haplotypes , Homozygote , Humans , Isoindoles/administration & dosage , Nateglinide , Patch-Clamp Techniques , Phenylalanine/administration & dosage , Phenylalanine/analogs & derivatives , Polymorphism, Single Nucleotide , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Receptors, Drug/antagonists & inhibitors , Structure-Activity Relationship , Sulfonylurea Compounds/administration & dosage , Sulfonylurea Receptors , Tolbutamide/administration & dosage
17.
J Pharmacol Exp Ther ; 343(2): 325-32, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22879384

ABSTRACT

Excessive reverse-mode (RM) sodium/calcium exchanger 1.1 (NCX1.1) activity, resulting from intracellular sodium accumulation caused by reduced Na+/K+-ATPase activity, increased Na-H exchanger 1 activity. The induction of the voltage-gated sodium channel late current component (late INa), is a major pathway for intracellular calcium (Ca2+i) loading in cardiac ischemia-reperfusion (IR) injury and cardiac glycoside toxicity. Inhibition of late INa with the antianginal agent ranolazine is protective in models of IR injury and cardiac glycoside toxicity. However, whether inhibition of late INa alone is sufficient to provide maximal protection or additional inhibition of RM NCX1.1 provides further benefit remains to be determined conclusively. Therefore, the effects of ranolazine were compared with the INa inhibitor lidocaine in models of IR injury and ouabain toxicity, RM NCX1.1-mediated Ca2+ overload, and patch-clamp assays of RM NCX1.1 currents. Ranolazine and lidocaine (10 µM) similarly reduced Ca2+i overload and improved left ventricle work recovery in whole-heart models of IR injury or exposure to ouabain (80 µM). Ranolazine (10 µM), but not lidocaine (10 µM), reduced RM NCX1.1-mediated Ca2+i overload in ventricular myocytes. Furthermore, ranolazine inhibited RM NCX1.1 currents (IC50 1.7 µM), without affecting forward mode currents, revealing that ranolazine has novel RM NCX1.1 inhibitory actions. However, because lidocaine provides similar protection to ranolazine in whole-heart models but does not inhibit RM NCX1.1, we conclude that induction of late INa is upstream of RM NCX1.1 activity and selective inhibition of late INa alone is sufficient to reduce Ca2+i overload and contractile dysfunction in IR injury and cardiac glycoside toxicity.


Subject(s)
Acetanilides/pharmacology , Calcium/metabolism , Cardiac Glycosides/antagonists & inhibitors , Cardiac Glycosides/pharmacology , Enzyme Inhibitors/pharmacology , Ischemia/metabolism , Myocardial Contraction/drug effects , Piperazines/pharmacology , Sodium Channel Blockers/pharmacology , Sodium-Calcium Exchanger/metabolism , Animals , Animals, Newborn , Calcium Signaling/drug effects , Electrophysiological Phenomena/drug effects , In Vitro Techniques , Lidocaine/pharmacology , Male , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/physiopathology , Patch-Clamp Techniques , Ranolazine , Rats , Rats, Sprague-Dawley , Sodium-Calcium Exchanger/antagonists & inhibitors , Sodium-Calcium Exchanger/genetics , Transfection , Ventricular Dysfunction, Left/drug therapy , Ventricular Dysfunction, Left/physiopathology
18.
IEEE Trans Biomed Eng ; 69(10): 3175-3182, 2022 10.
Article in English | MEDLINE | ID: mdl-35333709

ABSTRACT

Monitoring lactate levels is an established method for determining hyperlactatemia in critically ill patients and assessing aerobic fitness. It is a widely used gold-standard technique in both professional and serious amateur sports. Non-invasive real-time lactate monitoring offers significant advantages over the current technology of finger-prick blood sampling. Possible candidate technology for developing non-invasive real-time lactate monitoring should be highly sensitive, flexible, and capable of real-time monitoring of lactate levels in interstitial fluid or within specific working muscle groups depending on the type of sport. Herein we describe a planar, flexible, passive, chipless tag resonator that is electromagnetically coupled to a reader placed in proximity to the lactate sensor tag. The tag resonator is a thin metallic tracing that can be taped on the skin. The resonance frequency of the tag fluctuates proportionately with changing lactate concentrations in a solution mimicking human interstitial fluid with very high sensitivity. The spectrum of the tag is reflected in the spectrum of the reader, which is a planar microwave resonator designed at a different frequency. The reader could be embedded in a cellphone or an application-specific wearable device for data communication and processing. The tag can accurately and reproducibly measure lactate concentrations in the range of 1 to 10 mM, which is in the physiological range of lactate observed at rest and during intense physical activity. Furthermore, the chrematistics of this technology will allow monitoring of lactate in specific working muscle groups.


Subject(s)
Lactic Acid , Wearable Electronic Devices , Extracellular Fluid , Humans , Microwaves , Monitoring, Physiologic
19.
Cell Metab ; 34(2): 256-268.e5, 2022 02 01.
Article in English | MEDLINE | ID: mdl-35108513

ABSTRACT

In diabetes, glucagon secretion from pancreatic α cells is dysregulated. The underlying mechanisms, and whether dysfunction occurs uniformly among cells, remain unclear. We examined α cells from human donors and mice using electrophysiological, transcriptomic, and computational approaches. Rising glucose suppresses α cell exocytosis by reducing P/Q-type Ca2+ channel activity, and this is disrupted in type 2 diabetes (T2D). Upon high-fat feeding of mice, α cells shift toward a "ß cell-like" electrophysiological profile in concert with indications of impaired identity. In human α cells we identified links between cell membrane properties and cell surface signaling receptors, mitochondrial respiratory chain complex assembly, and cell maturation. Cell-type classification using machine learning of electrophysiology data demonstrated a heterogenous loss of "electrophysiologic identity" in α cells from donors with type 2 diabetes. Indeed, a subset of α cells with impaired exocytosis is defined by an enrichment in progenitor and lineage markers and upregulation of an immature transcriptomic phenotype, suggesting important links between α cell maturation state and dysfunction.


Subject(s)
Diabetes Mellitus, Type 2 , Glucagon-Secreting Cells , Islets of Langerhans , Animals , Diabetes Mellitus, Type 2/metabolism , Exocytosis/physiology , Glucagon/metabolism , Glucagon-Secreting Cells/metabolism , Insulin/metabolism , Islets of Langerhans/metabolism , Mice
20.
J Cell Mol Med ; 15(5): 1136-47, 2011 May.
Article in English | MEDLINE | ID: mdl-20518849

ABSTRACT

Damage to cardiac contractile proteins during ischemia followed by reperfusion is mediated by reactive oxygen species such as peroxynitrite (ONOO(-)), resulting in impairment of cardiac systolic function. However, the pathophysiology of systolic dysfunction during ischemia only, before reperfusion, remains unclear. We suggest that increased ONOO(-) generation during ischemia leads to nitration/nitrosylation of myosin light chain 1 (MLC1) and its increased degradation by matrix metalloproteinase-2 (MMP-2), which leads to impairment of cardiomyocyte contractility. We also postulate that inhibition of ONOO(-) action by use of a ONOO(-) scavenger results in improved recovery from ischemic injury. Isolated rat cardiomyocytes were subjected to 15 and 60 min. of simulated ischemia. Intact MLC1 levels, measured by 2D gel electrophoresis and immunoblot, were shown to decrease with increasing duration of ischemia, which correlated with increasing levels of nitrotyrosine and nitrite/nitrate. In vitro degradation of human recombinant MLC1 by MMP-2 increased after ONOO(-) exposure of MLC1 in a concentration-dependent manner. Mass spectrometry analysis of ischemic rat cardiomyocyte MLC1 showed nitration of tyrosines 78 and 190, as well as of corresponding tyrosines 73 and 185 within recombinant human cardiac MLC1 treated with ONOO(-). Recombinant human cardiac MLC1 was additionally nitrosylated at cysteine 67 and 76 corresponding to cysteine 81 of rat MLC1. Here we show that increased ONOO(-) production during ischemia induces MLC1 nitration/nitrosylation leading to its increased degradation by MMP-2. Inhibition of MLC1 nitration/nitrosylation during ischemia by the ONOO(-) scavenger FeTPPS (5,10,15,20-tetrakis-[4-sulfonatophenyl]-porphyrinato-iron[III]), or inhibition of MMP-2 activity with phenanthroline, provides an effective protection of cardiomyocyte contractility.


Subject(s)
Matrix Metalloproteinase 2/metabolism , Membrane Proteins/metabolism , Myocardial Contraction , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/metabolism , Myosin Light Chains/metabolism , Peroxynitrous Acid/metabolism , Animals , Humans , Matrix Metalloproteinase Inhibitors , Membrane Proteins/antagonists & inhibitors , Myocardial Reperfusion Injury/physiopathology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL