Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Appl Biochem Biotechnol ; 195(8): 4796-4817, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37184724

ABSTRACT

Islet transplantation offers improved glycemic control in individuals with type 1 diabetes mellitus. However, in vitro islet culture is associated with islet apoptosis and eventually will lose their functionality prior to transplantation. In this study, we examined the effects of mesenchymal stem cells (MSCs) secretome preconditioned with diazoxide (DZ) and trimetazidine (TMZ) on rat islet cells during pre-transplant culture. With and without preconditioned hAD-MSCs' concentrated conditioned media (CCM) were added to the culture medium containing rat islets every 12 h for 24 and 48 h, after testing for selected cytokine concentrations (interleukin (IL)-4, IL-6, IL-13). Insulin content, glucose-stimulated insulin secretion, islet cell apoptosis, and mRNA expression of pro-apoptotic (BAX, BAK-1, and PUMA) and anti-apoptotic factors (BCL-2, BCL-xL, and XIAP) in rat islets were assessed after 24 and 48 h of culture. The protein level of IL-6 and IL-4 was significantly higher in TMZ-MSC-CM compared to MSC-non-CM. In rat isolated islets, normalized secreted insulin in the presence of 16.7 mM glucose was significantly higher in treated islet groups compared to control islets at both 24 and 48 h cultivation. Also, the percentage of apoptotic islet cells TMZ-MSC-CCM-treated islets was significantly lower compared to MSC-CM and MSC-CCM-treated islets in both 24 and 48 h cultivation. Consistent with the number of apoptotic cells, after 24 h culture, the expression of BCL-2 and BCL-xL genes in the control islets was lower than all treatment islet groups and in 48 h was lower than only TMZ-MSC-CM-treated islets. Also, the expression of the XIAP gene in control islets was significantly lower compared to the TMZ-MSC-CCM-treated islets at both at 24 and 48 h. In addition, mRNA level of the BAX gene in TMZ-MSC-CCM-treated islets was significantly lower compared to other groups at 48 h. Our findings revealed that TMZ proved to be more effective than DZ and could enhance the potential of hAD-MSCs-CM to improve the function and viability of islets prior to transplantation.


Subject(s)
Islets of Langerhans , Mesenchymal Stem Cells , Trimetazidine , Rats , Animals , Trimetazidine/pharmacology , Trimetazidine/metabolism , Interleukin-6/metabolism , Secretome , bcl-2-Associated X Protein/metabolism , Islets of Langerhans/metabolism , Insulin/metabolism , Diazoxide/metabolism , Diazoxide/pharmacology , Glucose/metabolism
2.
Ecotoxicol Environ Saf ; 256: 114829, 2023 May.
Article in English | MEDLINE | ID: mdl-36989557

ABSTRACT

Heavy metals are ubiquitous environmental pollutants that are extremely dangerous for public health, but the molecular mechanisms of their cytotoxic action are still not fully understood. In the present work, the possible contribution of the mitochondrial ATP-sensitive potassium channel (mK(ATP)), which is usually considered protective for the cell, to hepatotoxicity caused by heavy metals was investigated using polarography and swelling techniques as well as flow cytometry. Using isolated liver mitochondria from adult male Wistar rats and various potassium media containing or not containing penetrating anions (KNO3, KSCN, KAcet, KCl), we studied the effect of mK(ATP) modulators, namely its blockers (5-hydroxydecanoate, glibenclamide, ATP, ADP) and activators (diazoxide, malonate), on respiration and/or membrane permeability in the presence of hepatotoxins such as Cd2+, Hg2+, and Cu2+. It has been shown for the first time that, contrary to Hg2+ and depending on media used, the mK(ATP) modulators affect Cd2+- and/or Cu2+-induced alterations in mitochondrial swelling and respiration rates, although differently, nevertheless, in the ways compatible with mK(ATP) participation in both these cases. On rat AS-30D ascites hepatoma cells, it was found that, unlike Cd2+, an increase in the production of reactive oxygen species was observed with the simultaneous use of Cu2+ and diazoxide; in addition, there was no protective effect of diazoxide against cell death, which also occurred in the presence of Cu2+. In conclusion, the relationships (functional, structural and/or regulatory) between mK(ATP), components of the mitochondrial electron transport chain (CI, CII-CIII and/or ATP synthase, CV) and mitochondrial permeability transition pores were discussed, as well as the role of these molecular structures in the mechanisms of the cytotoxic action of heavy metals.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Mercury , Metals, Heavy , Rats , Male , Animals , Mitochondria, Liver , KATP Channels/metabolism , KATP Channels/pharmacology , Diazoxide/metabolism , Diazoxide/pharmacology , Cadmium/toxicity , Ascites/metabolism , Carcinoma, Hepatocellular/metabolism , Rats, Wistar , Metals, Heavy/metabolism , Mercury/metabolism , Liver Neoplasms/metabolism , Adenosine Triphosphate/metabolism
3.
J Bioenerg Biomembr ; 55(1): 71-78, 2023 02.
Article in English | MEDLINE | ID: mdl-36723797

ABSTRACT

AIM/INTRODUCTION: Diabetes Mellitus is a chronic degenerative disease, and its main biochemical characteristic is hyperglycemia due to impaired insulin secretion, resistance to peripheral actions of insulin, or both. Hyperglycemia causes dyslipidemia and stimulates oxidative damage, leading to the main symptoms, such as fatigue and culminates in diabetic complications. Previous studies have shown that ATP-sensitive potassium channels counteract muscle fatigue and metabolic stress in healthy mouse models. To determine the effect of diazoxide on muscle strength development during diabetes, we tested the effect of diazoxide in streptozotocin-diabetic rats in muscle function, lipid profile and oxidative stress biomarkers. MATERIALS AND METHODS: Wistar rats were divided into 4 groups of six animals each: (1) Control group, (2) diabetes group, (3) Control group + diazoxide, and (4) Diabetic + diazoxide (DB + DZX). 4 weeks after rats were sacrificed, soleus and extensor digitorum longus muscles (EDL) were extracted to prepare homogenates and serum was obtained for biochemical measurements. Oxidative damage was evaluated by the thiobarbituric acid method and the fluorescent for reactive oxygen species (ROS) probe 2,4-H2DCFDA, respectively. RESULTS: Diabetic rats with diazoxide administration showed an increase in the development of muscle strength in both muscles; in turn, the onset of fatigue was longer compared to the group of diabetic rats without treatment. Regarding the lipid profile, diazoxide decreased total cholesterol levels in the group of diabetic rats treated with diazoxide (x̅46.2 mg/dL) compared to the untreated diabetic group (x̅=104.4 mg/dL); secondly, diazoxide decreased triglyceride concentrations (x̅=105.3 mg/dL) compared to the untreated diabetic rats (x̅=412.2 mg/dL) as well as the levels of very low-density lipoproteins (x̅=20.4 mg/dL vs. x̅=82.44 mg/dL). Regarding the various markers of oxidative stress, the diabetic group treated with diazoxide was able to reduce the concentrations of TBARS and total reactive oxygen species as well as preserve the concentrations of reduced glutathione. CONCLUSION: Diazoxide administration in diabetic rats increases muscle strength development in EDL and soleus muscle, decreases fatigue, reduces cholesterol and triglyceride concentrations and improves oxidative stress parameters such as TBARS, ROS, and glutathione status.


Subject(s)
Diabetes Mellitus, Experimental , Hyperglycemia , Mice , Rats , Animals , Diazoxide/adverse effects , Diazoxide/metabolism , Streptozocin/adverse effects , Streptozocin/metabolism , Rats, Wistar , Reactive Oxygen Species/metabolism , Diabetes Mellitus, Experimental/complications , Thiobarbituric Acid Reactive Substances/adverse effects , Thiobarbituric Acid Reactive Substances/metabolism , Oxidative Stress , Hyperglycemia/complications , Muscle, Skeletal/metabolism , Lipids , Triglycerides/adverse effects , Triglycerides/metabolism , Cholesterol/metabolism
4.
Anatol J Cardiol ; 27(2): 88-99, 2023 02.
Article in English | MEDLINE | ID: mdl-36747448

ABSTRACT

BACKGROUND: Myocardial infarction is associated with the autophagy and apoptosis of cardiomyocytes, and the protein kinase B/mammalian target of rapamycin (AKT/mTOR) pathway plays a crucial role in this mechanism. METHODS: Acute myocardial infarction rat models were assessed 0.5, 2, 4, and 6 hours after the induction of the myocardial infarction using hematoxylin and eosin staining, triphenyl tetrazolium chloride staining, myocardial enzyme measurements, and levels of autophagic activity. Additionally, diazoxide, 5-hydroxydecanoate, and LY294002 were intraperitoneally administered to rat models at peak myocardial injury to assess their effects on cardiac injury. The expression levels of autophagy-related and apoptosis-related proteins, as well as p-AKT and p-mTOR, were measured. Electron microscopy was used to assess the ultrastructure and the number of autophagosomes in the cardiac tissue. RESULTS: We demonstrated that the degree of myocardial injury and the level of autophagy were significantly elevated in the experimental cohort compared with the control cohort. In addition, the myocardial infarct size was significantly smaller in diazoxide-treated acute myocardial infarction rats compared with untreated rats. Diazoxide also decreased the levels of myocardial injury markers, autophagy, and apoptosis, while it also induced the levels of AKT and mTOR phosphorylation, decreased the number of autophagosomes, and improved the myocardial ultrastructure of the acute myocardial infarction rats. 5-Hydroxydecanoate treatment resulted in an opposite effect to those observed upon diazoxide treatment. LY294002 was also able to reverse diazoxide treatment effects. CONCLUSION: Peak levels of myocardial tissue injury and autophagy were observed 2 hours post-acute myocardial infarction induction in rats. Diazoxide treatment inhibited myocardial autophagy and apoptosis while protecting cardiac tissue from ischemic injury, which is likely to have proceeded through activation of the AKT/mTOR pathway.


Subject(s)
Myocardial Infarction , Proto-Oncogene Proteins c-akt , Rats , Animals , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Diazoxide/pharmacology , Diazoxide/therapeutic use , Diazoxide/metabolism , Rats, Sprague-Dawley , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/pharmacology , Myocardial Infarction/drug therapy , Myocytes, Cardiac , Mammals/metabolism
5.
Pflugers Arch ; 475(4): 477-488, 2023 04.
Article in English | MEDLINE | ID: mdl-36707457

ABSTRACT

Connexin43 (Cx43) exits as hemichannels in the inner mitochondrial membrane. We examined how mitochondrial Cx43 and mitochondrial KATP channels affect the occurrence of triggered arrhythmias. To generate cardiac-specific Cx43-deficient (cCx43-/-) mice, Cx43flox/flox mice were crossed with α-MHC (Myh6)-cre+/- mice. The resulting offspring, Cx43flox/flox/Myh6-cre+/- mice (cCx43-/- mice) and their littermates (cCx43+/+ mice), were used. Trabeculae were dissected from the right ventricles of mouse hearts. Cardiomyocytes were enzymatically isolated from the ventricles of mouse hearts. Force was measured with a strain gauge in trabeculae (22°C). To assess arrhythmia susceptibility, the minimal extracellular Ca2+ concentration ([Ca2+]o,min), at which arrhythmias were induced by electrical stimulation, was determined in trabeculae. ROS production was estimated with 2',7'-dichlorofluorescein (DCF), mitochondrial membrane potential with tetramethylrhodamine methyl ester (TMRM), and Ca2+ spark frequency with fluo-4 and confocal microscopy in cardiomyocytes. ROS production within the mitochondria was estimated with MitoSoxRed and mitochondrial Ca2+ with rhod-2 in trabeculae. Diazoxide was used to activate mitochondrial KATP. Most of cCx43-/- mice died suddenly within 8 weeks. Cx43 was present in the inner mitochondrial membrane in cCx43+/+ mice but not in cCx43-/- mice. In cCx43-/- mice, the [Ca2+]o,min was lower, and Ca2+ spark frequency, the slope of DCF fluorescence intensity, MitoSoxRed fluorescence, and rhod-2 fluorescence were higher. TMRM fluorescence was more decreased in cCx43-/- mice. Most of these changes were suppressed by diazoxide. In addition, in cCx43-/- mice, antioxidant peptide SS-31 and N-acetyl-L-cysteine increased the [Ca2+]o,min. These results suggest that Cx43 deficiency activates Ca2+ leak from the SR, probably due to depolarization of mitochondrial membrane potential, an increase in mitochondrial Ca2+, and an increase in ROS production, thereby causing triggered arrhythmias, and that Cx43 hemichannel deficiency may be compensated by activation of mitochondrial KATP channels in mouse hearts.


Subject(s)
Connexin 43 , Heart Ventricles , Mice , Animals , Heart Ventricles/metabolism , Connexin 43/metabolism , Diazoxide/adverse effects , Diazoxide/metabolism , Reactive Oxygen Species/metabolism , Myocytes, Cardiac/metabolism , Arrhythmias, Cardiac/metabolism , Mitochondria , Adenosine Triphosphate/metabolism
6.
Biochemistry ; 51(45): 9211-22, 2012 Nov 13.
Article in English | MEDLINE | ID: mdl-23078514

ABSTRACT

ATP sensitive potassium (K(ATP)) channels are composed of four copies of a pore-forming inward rectifying potassium channel (Kir6.1 or Kir6.2) and four copies of a sulfonylurea receptor (SUR1, SUR2A, or SUR2B) that surround the pore. SUR proteins are members of the ATP-binding cassette (ABC) superfamily of proteins. Binding of MgATP at the SUR nucleotide binding domains (NBDs) results in NBD dimerization, and hydrolysis of MgATP at the NBDs leads to channel opening. The SUR proteins also mediate interactions with K(ATP) channel openers (KCOs) that activate the channel, with KCO binding and/or activation involving residues in the transmembrane helices and cytoplasmic loops of the SUR proteins. Because the cytoplasmic loops make extensive interactions with the NBDs, we hypothesized that the NBDs may also be involved in KCO binding. Here, we report nuclear magnetic resonance (NMR) spectroscopy studies that demonstrate a specific interaction of the KCO pinacidil with the first nucleotide binding domain (NBD1) from SUR2A, the regulatory SUR protein in cardiac K(ATP) channels. Intrinsic tryptophan fluorescence titrations also demonstrate binding of pinacidil to SUR2A NBD1, and fluorescent nucleotide binding studies show that pinacidil binding increases the affinity of SUR2A NBD1 for ATP. In contrast, the KCO diazoxide does not interact with SUR2A NBD1 under the same conditions. NMR relaxation experiments and size exclusion chromatography indicate that SUR2A NBD1 is monomeric under the conditions used in drug binding studies. These studies identify additional binding sites for commonly used KCOs and provide a foundation for testing binding of drugs to the SUR NBDs.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Receptors, Drug/metabolism , ATP-Binding Cassette Transporters/physiology , Adenosine Triphosphate/metabolism , Animals , Binding Sites/genetics , Diazoxide/metabolism , Nuclear Magnetic Resonance, Biomolecular , Nucleotides/metabolism , Pinacidil/metabolism , Potassium Channels, Inwardly Rectifying/physiology , Protein Structure, Tertiary , Rats , Receptors, Drug/physiology , Sulfonylurea Receptors
7.
Br J Pharmacol ; 159(3): 669-77, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20128805

ABSTRACT

BACKGROUND AND PURPOSE: ATP-sensitive potassium channels (K(ATP) channels) in beta cells are a major target for insulinotropic drugs. Here, we studied the effects of selected stimulatory and inhibitory pharmacological agents in islets lacking K(ATP) channels. EXPERIMENTAL APPROACH: We compared insulin secretion (IS) and cytosolic calcium ([Ca(2+)](c)) changes in islets isolated from control mice and mice lacking sulphonylurea receptor1 (SUR1), and thus K(ATP) channels in their beta cells (Sur1KO). KEY RESULTS: While similarly increasing [Ca(2+)](c) and IS in controls, agents binding to site A (tolbutamide) or site B (meglitinide) of SUR1 were ineffective in Sur1KO islets. Of two non-selective blockers of potassium channels, quinine was inactive, whereas tetraethylammonium was more active in Sur1KO compared with control islets. Phentolamine, efaroxan and alinidine, three imidazolines binding to K(IR)6.2 (pore of K(ATP) channels), stimulated control islets, but only phentolamine retained weaker stimulatory effects on [Ca(2+)](c) and IS in Sur1KO islets. Neither K(ATP) channel opener (diazoxide, pinacidil) inhibited Sur1KO islets. Calcium channel blockers (nimodipine, verapamil) or diphenylhydantoin decreased [Ca(2+)](c) and IS in both types of islets, verapamil and diphenylhydantoin being more efficient in Sur1KO islets. Activation of alpha(2)-adrenoceptors or dopamine receptors strongly inhibited IS while partially (clonidine > dopamine) lowering [Ca(2+)](c) (control > Sur1KO islets). CONCLUSIONS AND IMPLICATIONS: Those drugs retaining effects on IS in islets lacking K(ATP) channels, also affected [Ca(2+)](c), indicating actions on other ionic channels. The greater effects of some inhibitors in Sur1KO than in control islets might be relevant to medical treatment of congenital hyperinsulinism caused by inactivating mutations of K(ATP) channels.


Subject(s)
Insulin-Secreting Cells/metabolism , Insulin/metabolism , KATP Channels/deficiency , Potassium Channels/metabolism , Tolbutamide/pharmacology , Animals , Benzofurans , Calcium/metabolism , Calcium/pharmacology , Cytosol/metabolism , Diazoxide/metabolism , Diazoxide/pharmacology , Female , Imidazoles , Imidazolines/metabolism , Imidazolines/pharmacology , Insulin/pharmacology , Insulin Secretion , Mice , Mice, Knockout , Phentolamine/metabolism , Phentolamine/pharmacology , Pinacidil/metabolism , Pinacidil/pharmacology , Potassium Channels/pharmacology , Tolbutamide/metabolism
8.
Drug Metab Dispos ; 38(2): 232-40, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19875500

ABSTRACT

SUR1-selective ATP-sensitive potassium channel openers (PCOs) have been shown to be of clinical value for the treatment of several metabolic disorders, including type I and type II diabetes, obesity, and hyperinsulinemia. Taking into account these promising therapeutic benefits, different series of 3-alkylamino-4H-1,2,4-benzothiadiazine 1,1-dioxides structurally related to diazoxide were developed. In view of the lead optimization process of the series, knowledge of absorption, distribution, metabolism, excretion, and toxicity parameters, and more particularly the metabolic fate of these compounds, is a fundamental requirement. For such a purpose, two selected promising compounds [7-chloro-3-isopropylamino-4H-1,2,4-benzothiadiazine 1,1-dioxide (BPDZ 73) and 7-chloro-3-(3-pentylamino)-4H-1,2,4-benzothiadiazine 1,1-dioxide (BPDZ 157)] were incubated in the presence of phenobarbital-induced rat liver microsomes to produce expected mammal in vivo phase I metabolites. The resulting major metabolites were then analyzed by both mass spectrometry (MS) and NMR to completely elucidate their chemical structures. The two compounds were also further incubated in the presence of nontreated rats and human microsomes to compare the metabolic profiles. In the present study, the combined use of an exact mass liquid chromatography (LC)/tandem MS platform and an LC/solid-phase extraction/NMR system allowed the clarification of some unresolved structural assessments in the accurate chemical structure elucidation process of the selected PCO drugs. These results greatly help the optimization of the lead compounds.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Benzothiadiazines/metabolism , Cyclic S-Oxides/metabolism , Diazoxide/analogs & derivatives , Ion Channel Gating/drug effects , KATP Channels/metabolism , Membrane Transport Modulators/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Receptors, Drug/metabolism , Animals , Chromatography, High Pressure Liquid/methods , Diazoxide/metabolism , Humans , Isomerism , Magnetic Resonance Spectroscopy/methods , Male , Metabolic Detoxication, Phase I , Microsomes, Liver/metabolism , Phenobarbital/pharmacology , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Rats , Rats, Sprague-Dawley , Solid Phase Extraction/methods , Sulfonylurea Receptors , Tandem Mass Spectrometry/methods
9.
J Cardiovasc Pharmacol ; 51(5): 483-91, 2008 May.
Article in English | MEDLINE | ID: mdl-18437094

ABSTRACT

Mitochondrial (m) KATP channel opening has been implicated in triggering cardiac preconditioning. Its consequence on mitochondrial respiration, however, remains unclear. We investigated the effects of two different KATP channel openers and antagonists on mitochondrial respiration under two different energetic conditions. Oxygen consumption was measured for complex I (pyruvate/malate) or complex II (succinate with rotenone) substrates in mitochondria from fresh guinea pig hearts. One of two mKATP channel openers, pinacidil or diazoxide, was given before adenosine diphosphate in the absence or presence of an mKATP channel antagonist, glibenclamide or 5-hydroxydecanoate. Without ATP synthase inhibition, both mKATP channel openers differentially attenuated mitochondrial respiration. Neither mKATP channel antagonist abolished these effects. When ATP synthase was inhibited by oligomycin to decrease [ATP], both mKATP channel openers accelerated respiration for both substrate groups. This was abolished by mKATP channel blockade. Thus, under energetically more physiological conditions, the main effect of mKATP channel openers on mitochondrial respiration is differential inhibition independent of mKATP channel opening. In contrast, under energetically less physiological conditions, mKATP channel opening can be evidenced by accelerated respiration and blockade by antagonists. Therefore, the effects of mKATP channel openers on mitochondrial function likely depend on the experimental conditions and the cell's underlying energetic state.


Subject(s)
Diazoxide/pharmacology , Mitochondria, Heart/metabolism , Oxygen Consumption/drug effects , Pinacidil/pharmacology , Potassium Channels/agonists , Animals , Decanoic Acids/pharmacology , Diazoxide/metabolism , Electron Transport Complex I/metabolism , Electron Transport Complex II/metabolism , Glyburide/pharmacology , Guinea Pigs , Hydroxy Acids/pharmacology , In Vitro Techniques , Mitochondria, Heart/drug effects , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Potassium Channel Blockers/pharmacology
10.
J Biol Chem ; 282(24): 17433-41, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17430885

ABSTRACT

We describe the existence of a potassium ion transport mechanism in the mitochondrial inner membrane of a lower eukaryotic organism, Acanthamoeba castellanii. We found that substances known to modulate potassium channel activity influenced the bioenergetics of A. castellanii mitochondria. In isolated mitochondria, the rate of resting respiration is increased by about 10% in response to potassium channel openers, i.e. diazoxide and BMS-191095, during succinate-, malate-, or NADH-sustained respiration. This effect is strictly dependent on the presence of potassium ions in an incubation medium and is reversed by glibenclamide (a potassium channel blocker). Diazoxide and BMS-191095 also caused a slight but statistically significant depolarization of mitochondrial membrane potential (measured with a TPP(+)-specific electrode), regardless of the respiratory substrate used. The resulting steady state value of membrane potential was restored after treatment with glibenclamide or 1 mM ATP. Additionally, the electrophysiological properties of potassium channels present in the A. castellanii inner mitochondrial membrane are described in the reconstituted system, using black lipid membranes. Conductance from 90 +/- 7 to 166 +/- 10 picosiemens, inhibition by 1 mM ATP/Mg(2+) or glibenclamide, and activation by diazoxide were observed. These results suggest that an ATP-sensitive potassium channel similar to that of mammalian mitochondria is present in A. castellanii mitochondria.


Subject(s)
Acanthamoeba castellanii/metabolism , Adenosine Triphosphate/metabolism , Potassium Channels/metabolism , Animals , Anti-Arrhythmia Agents/metabolism , Cell Respiration/physiology , Diazoxide/metabolism , Electrophysiology , Glyburide/metabolism , Membrane Potential, Mitochondrial/physiology , Mitochondria/metabolism , Mitochondria/ultrastructure , Potassium/metabolism , Potassium Channels/genetics
11.
J Endocrinol ; 189(1): 127-36, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16614387

ABSTRACT

Ocimum sanctum leaves have previously been reported to reduce blood glucose when administered to rats and humans with diabetes. In the present study, the effects of ethanol extract and five partition fractions of O. sanctum leaves were studied on insulin secretion together with an evaluation of their mechanisms of action. The ethanol extract and each of the aqueous, butanol and ethylacetate fractions stimulated insulin secretion from perfused rat pancreas, isolated rat islets and a clonal rat beta-cell line in a concentration-dependent manner. The stimulatory effects of ethanol extract and each of these partition fractions were potentiated by glucose, isobutylmethylxanthine, tolbutamide and a depolarizing concentration of KCl. Inhibition of the secretory effect was observed with diazoxide, verapamil and Ca2+ removal. In contrast, the stimulatory effects of the chloroform and hexane partition fractions were associated with decreased cell viability and were unaltered by diazoxide and verapamil. The ethanol extract and the five fractions increased intracellular Ca2+ in clonal BRIN-BD11 cells, being partly attenuated by the addition of verapamil. These findings indicated that constituents of O. sanctum leaf extracts have stimulatory effects on physiological pathways of insulin secretion which may underlie its reported antidiabetic action.


Subject(s)
Insulin/metabolism , Ocimum/metabolism , Pancreas/metabolism , Plant Extracts/metabolism , Plant Leaves/metabolism , 1-Methyl-3-isobutylxanthine/metabolism , Acetates/metabolism , Animals , Butanols/metabolism , Cell Line , Diazoxide/metabolism , Ethanol/metabolism , Glucose/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Perfusion , Potassium Chloride/metabolism , Rats , Rats, Long-Evans , Tissue Culture Techniques/methods , Tolbutamide/metabolism , Verapamil/metabolism
12.
Dev Biol ; 289(2): 395-405, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16343479

ABSTRACT

Mammalian sperm must undergo a series of physiological changes after leaving the testis to become competent for fertilization. These changes, collectively known as capacitation, occur in the female reproductive tract where the sperm plasma membrane is modified in terms of its components and ionic permeability. Among other events, mouse sperm capacitation leads to an increase in the intracellular Ca(2+) and pH as well as to a hyperpolarization of the membrane potential. It is well known that ion channels play a crucial role in these events, though the molecular identity of the particular channels involved in capacitation is poorly defined. In the present work, we report the identification and potential functional role of K(ATP) channels in mouse spermatogenic cells and sperm. By using whole-cell patch clamp recordings in mouse spermatogenic cells, we found K(+) inwardly rectifying (K(ir)) currents that are sensitive to Ba(2+), glucose and the sulfonylureas (tolbutamide and glibenclamide) that block K(ATP) channels. The presence of these channels was confirmed using inhibitors of the ATP synthesis and K(ATP) channel activators. Furthermore, RT-PCR assays allowed us to detect transcripts for the K(ATP) subunits SUR1, SUR2, K(ir)6.1 and K(ir)6.2 in total RNA from elongated spermatids. In addition, immunoconfocal microscopy revealed the presence of these K(ATP) subunits in mouse spermatogenic cells and sperm. Notably, incubation of sperm with tolbutamide during capacitation abolished hyperpolarization and significantly decreased the percentage of AR in a dose-dependent fashion. Together, our results provide evidence for the presence of K(ATP) channels in mouse spermatogenic cells and sperm and disclose the contribution of these channels to the capacitation-associated hyperpolarization.


Subject(s)
Potassium Channels, Inwardly Rectifying/metabolism , Sperm Capacitation/physiology , Spermatogenesis/physiology , Spermatozoa/metabolism , ATP-Binding Cassette Transporters/metabolism , Adenosine Triphosphate/metabolism , Animals , Barium/metabolism , Barium/pharmacology , Diazoxide/metabolism , Diazoxide/pharmacology , Dose-Response Relationship, Drug , Glyburide/metabolism , Glyburide/pharmacology , KATP Channels , Male , Membrane Potentials , Mice , Microscopy, Confocal , Multidrug Resistance-Associated Proteins/metabolism , Pinacidil/metabolism , Pinacidil/pharmacology , RNA, Messenger/metabolism , Receptors, Drug , Spermatozoa/cytology , Sulfonylurea Receptors , Time Factors , Tolbutamide/metabolism , Tolbutamide/pharmacology
13.
J Cell Sci ; 118(Pt 19): 4463-71, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-16159958

ABSTRACT

Phospholipase C (PLC) regulates various cellular processes by catalyzing the formation of inositol-1,4,5-trisphosphate (IP3) and diacylglycerol from phosphatidylinositol-4,5-bisphosphate (PIP2). Here, we have investigated the influence of Ca2+ on receptor-triggered PLC activity in individual insulin-secreting beta-cells. Evanescent wave microscopy was used to record PLC activity using green fluorescent protein (GFP)-tagged PIP2/IP3-binding pleckstrin homology domain from PLCdelta1, and the cytoplasmic Ca2+ concentration ([Ca2+]i) was simultaneously measured using the indicator Fura Red. Stimulation of MIN6 beta-cells with the muscarinic-receptor agonist carbachol induced rapid and sustained PLC activation. By contrast, only transient activation was observed after stimulation in the absence of extracellular Ca2+ or in the presence of the non-selective Ca2+ channel inhibitor La3+. The Ca2+-dependent sustained phase of PLC activity did not require voltage-gated Ca2+ influx, as hyperpolarization with diazoxide or direct Ca2+ channel blockade with nifedipine had no effect. Instead, the sustained PLC activity was markedly suppressed by the store-operated channel inhibitors 2-APB and SKF96365. Depletion of intracellular Ca2+ stores with the sarco(endo)plasmic reticulum Ca2+-ATPase inhibitors thapsigargin or cyclopiazonic acid abolished Ca2+ mobilization in response to carbachol, and strongly suppressed the PLC activation in Ca2+-deficient medium. Analogous suppressions were observed after loading cells with the Ca2+ chelator BAPTA. Stimulation of primary mouse pancreatic beta-cells with glucagon elicited pronounced [Ca2+]i spikes, reflecting protein kinase A-mediated activation of Ca2+-induced Ca2+ release via IP3 receptors. These [Ca2+]i spikes were found to evoke rapid and transient activation of PLC. Our data indicate that receptor-triggered PLC activity is enhanced by positive feedback from Ca2+ entering the cytoplasm from intracellular stores and via store-operated channels in the plasma membrane. Such amplification of receptor signalling should be important in the regulation of insulin secretion by hormones and neurotransmitters.


Subject(s)
Calcium/metabolism , Feedback, Physiological , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Isoenzymes/metabolism , Signal Transduction/physiology , Type C Phospholipases/metabolism , Animals , Boron Compounds/metabolism , Calcium Channel Blockers/metabolism , Calcium Channels/metabolism , Calcium-Transporting ATPases/antagonists & inhibitors , Calcium-Transporting ATPases/metabolism , Cells, Cultured , Diazoxide/metabolism , Enzyme Activation , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Inositol 1,4,5-Trisphosphate/metabolism , Insulin-Secreting Cells/cytology , Isoenzymes/genetics , Lanthanum/metabolism , Mice , Microscopy, Fluorescence/methods , Phospholipase C delta , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Type C Phospholipases/genetics
14.
J Exp Biol ; 208(Pt 14): 2765-72, 2005 Jul.
Article in English | MEDLINE | ID: mdl-16000545

ABSTRACT

The activation of ATP-sensitive potassium (K ATP) ion channels in the heart is thought to exert a cardioprotective effect under low oxygen conditions, possibly enhancing tolerance of environmental hypoxia in aquatic vertebrates. The purpose of this study was to examine the possibility that hypoxia-induced activation of cardiac K ATP channels, whether in the sarcolemma (sarcK ATP) or mitochondria (mitoK ATP), enhances viability in cardiac muscle cells from a species highly tolerant of low oxygen environments, the goldfish Carassius auratus. During moderate hypoxia (6-7 kPa), the activation of sarcK ATP channels was indicated by a reduction in transmembrane action potential duration (APD). This response to hypoxia was mimicked by the NO-donor SNAP (100 micromol l(-1)) and the stable cGMP analog 8-Br-cGMP, but abolished by glibenclamide or l-NAME, an inhibitor of NO synthesis. The mitoK ATP channel opener diazoxide did not affect APD. Isolated ventricular muscle cells were then incubated under normoxic and hypoxic conditions. Cell viability was decreased in hypoxia; however, the negative effects of low oxygen were reduced during simultaneous exposure to SNAP, 8-Br-cGMP, and diazoxide. The cardioprotective effect of diazoxide, but not 8-Br-cGMP, was reduced by the mitoK ATP channel blocker 5-HD. These data suggest that hypoxia-induced activation of sarcK ATP or mitoK ATP channels could enhance tolerance of low-oxygen environments in this species, and that sarcK ATP activity is increased through a NO and cGMP-dependent pathway.


Subject(s)
Acclimatization/physiology , Goldfish/metabolism , Hypoxia/metabolism , Models, Biological , Myocardium/metabolism , Potassium Channels/metabolism , Action Potentials/physiology , Analysis of Variance , Animals , Cyclic GMP/analogs & derivatives , Cyclic GMP/metabolism , Diazoxide/metabolism , Glyburide/metabolism , Mitochondria/metabolism , NG-Nitroarginine Methyl Ester/metabolism , Penicillamine/analogs & derivatives , Penicillamine/metabolism , Sarcolemma/metabolism
15.
Am J Physiol Heart Circ Physiol ; 286(3): H955-63, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14592940

ABSTRACT

The cardiac Na(+)/Ca(2+) exchanger (NCX) contributes to cellular injury during hypoxia, as its altered function is largely responsible for a rise in cytosolic Ca(2+) concentration ([Ca(2+)](i)). In addition, the NCX in guinea pig ventricular myocytes undergoes profound inhibition during hypoxia and rapid reactivation during reoxygenation. The mechanisms underlying these changes in NCX activity are likely complex due to the participation of multiple inhibitory factors including altered cytosolic Na(+) concentration, pH, and ATP. Our main hypothesis is that oxidative stress is an essential trigger for rapid NCX reactivation in guinea pig ventricular myocytes and is thus a critical factor in determining the timing and magnitude of Ca(2+) overload. This hypothesis was evaluated in cardiac myocytes using fluorescent indicators to measure [Ca(2+)](i) and oxidative stress. An NCX antisense oligonucleotide was used to decrease NCX protein expression in some experiments. Our results indicate that NCX activity is profoundly inhibited in hypoxic guinea pig ventricular myocytes but is reactivated within 1-2 min of reoxygenation at a time of rising oxidative stress. We also found that several interventions to decrease oxidative stress including antioxidants and diazoxide prevented NCX reactivation and Ca(2+) overload during reoxygenation. Furthermore, application of exogenous H(2)O(2) was sufficient by itself to reactivate the NCX during sustained hypoxia and could reverse the suppression of reoxygenation-mediated NCX reactivation by diazoxide. These data suggest that elevated oxidative stress in reoxygenated guinea pig ventricular myocytes is required for rapid NCX reactivation, and thus reactivation should be viewed as an active process rather than being due to the simple decline of NCX inhibition.


Subject(s)
Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/metabolism , Reactive Oxygen Species/metabolism , Sodium-Calcium Exchanger/metabolism , Animals , Antioxidants/pharmacology , Cells, Cultured , Chromans/pharmacology , Diazoxide/metabolism , Free Radical Scavengers/pharmacology , Guinea Pigs , Heart Ventricles/cytology , Hypoxia/metabolism , Metalloporphyrins/pharmacology , Myocytes, Cardiac/cytology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Resveratrol , Stilbenes/pharmacology
16.
Ann N Y Acad Sci ; 1009: 167-74, 2003 Dec.
Article in English | MEDLINE | ID: mdl-15028583

ABSTRACT

The pancreatic beta-cell expresses an imidazoline-binding site that is involved in the regulation of insulin secretion. This site is pharmacologically atypical in comparison with the I(1) and I(2) sites described in other tissues, and it has been classified as I(3). The structural requirements for binding of ligands to the I(3) site have not been fully defined, although a range of synthetic I(3) ligands have been characterized in functional terms. Evidence has been presented that an endogenous I(3) ligand may exist, because extracts of brain contain an active principle that stimulates insulin secretion in a manner consistent with the involvement of I(3) sites. The active component has not been identified but has been equated with the long-sought clonidine displacing substance (CDS) that is proposed as the endogenous ligand for imidazoline-binding sites. Recent evidence has indicated that one active component of CDS may be a beta-carboline, but it is not known whether beta-carbolines can stimulate insulin secretion. Thus, we have studied the effects of beta-carbolines on insulin secretion and cytosolic Ca(2+) levels in rodent and human islet cells. The results reveal that harmane, pinoline, and norharmane cause a dose- and glucose-dependent increase in insulin secretion but show that this response differs in a number of ways from that elicited by the well-characterized I(3)-agonist, efaroxan. Thus, beta-carbolines represent a new class of insulin secretagogues, although it remains unclear whether their action is mediated solely by I(3) sites in the beta cell.


Subject(s)
Adrenergic alpha-Antagonists/pharmacology , Benzofurans/pharmacology , Carbolines/pharmacology , Imidazoles/pharmacology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Animals , Binding Sites , Calcium/metabolism , Cells, Cultured , Diazoxide/metabolism , Glucose/metabolism , Humans , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/cytology , Ligands , Male , Rats , Rats, Wistar
17.
Diabetes ; 51(6): 1896-906, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12031979

ABSTRACT

ATP-sensitive K(+) (K(ATP)) channels are activated by a diverse group of compounds known as potassium channel openers (PCOs). Here, we report functional studies of the Kir6.2/SUR1 Selective PCO 3-isopropylamino-7-methoxy-4H-1,2,4-benzothiadiazine 1,1-dioxide (NNC 55-9216). We recorded cloned K(ATP) channel currents from inside-out patches excised from Xenopus laevis oocytes heterologously expressing Kir6.2/SUR1, Kir6.2/SUR2A, or Kir6.2/SUR2B, corresponding to the beta-cell, cardiac, and smooth muscle types of the K(ATP) channel. NNC 55-9216 reversibly activated Kir6.2/SUR1 currents (EC(50) = 16 micromol/l). This activation was dependent on intracellular MgATP and was abolished by mutation of a single residue in the Walker A motifs of either nucleotide-binding domain of SUR1. The drug had no effect on Kir6.2/SUR2A or Kir6.2/SUR2B currents. We therefore used chimeras of SUR1 and SUR2A to identify regions of SUR1 involved in the response to NNC 55-9216. Activation was completely abolished and significantly reduced by swapping transmembrane domains 8-11. The reverse chimera consisting of SUR2A with transmembrane domains 8-11 and NBD2 consisting SUR1 was activated by NNC 55-9216, indicating that these SUR1 regions are important for drug activation. [(3)H]glibenclamide binding to membranes from HEK293 cells transfected with SUR1 was displaced by NNC 55-9216 (IC(50) = 105 micromol/l), and this effect was impaired when NBD2 of SUR1 was replaced by that of SUR2A. These results suggest NNC 55-9216 is a SUR1-selective PCO that requires structural determinants, which differ from those needed for activation of the K(ATP) channel by pinacidil and cromakalim. The high selectivity of NNC 55-9216 may prove to be useful for studies of the molecular mechanism of PCO action.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Benzothiadiazines , Diazoxide/pharmacology , Ion Channel Gating/drug effects , Potassium Channels, Inwardly Rectifying/drug effects , Adenosine Triphosphate/pharmacology , Animals , Binding Sites , Cell Line , Cell Membrane/metabolism , Diazoxide/analogs & derivatives , Diazoxide/metabolism , Electric Conductivity , Gene Expression , Glyburide/metabolism , Hypoglycemic Agents/metabolism , Mice , Mutagenesis , Oocytes/metabolism , Potassium Channels, Inwardly Rectifying/genetics , Rats , Recombinant Fusion Proteins , Transfection , Tritium , Xenopus laevis
18.
Trends Pharmacol Sci ; 21(11): 439-45, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11121575

ABSTRACT

K(ATP) channel openers are a diverse group of drugs with a wide range of potential therapeutic uses. Their molecular targets, the K(ATP) channels, exhibit tissue-specific responses because they possess different types of regulatory sulfonylurea receptor subunits. It is well recognized that complex interactions occur between K(ATP) channel openers and nucleotides, but the cloning of the K(ATP) channel has introduced a new dimension to the study of these events and has furthered our understanding of the molecular basis of the action of K(ATP) channel openers.


Subject(s)
Minoxidil/analogs & derivatives , Potassium Channels/drug effects , Adenosine Triphosphate/physiology , Animals , Binding Sites , Cromakalim/metabolism , Cromakalim/pharmacology , Diazoxide/metabolism , Diazoxide/pharmacology , Humans , Minoxidil/metabolism , Minoxidil/pharmacology , Nicorandil/metabolism , Nicorandil/pharmacology , Pinacidil/metabolism , Pinacidil/pharmacology , Potassium Channels/metabolism , Potassium Channels/physiology
19.
EMBO J ; 17(19): 5529-35, 1998 Oct 01.
Article in English | MEDLINE | ID: mdl-9755153

ABSTRACT

KATP channels are composed of a small inwardly rectifying K+ channel subunit, either KIR6.1 or KIR6.2, plus a sulfonylurea receptor, SUR1 or SUR2 (A or B), which belong to the ATP-binding cassette superfamily. SUR1/KIR6.2 reconstitute the neuronal/pancreatic beta-cell channel, whereas SUR2A/KIR6.2 and SUR2B/KIR6.1 (or KIR6.2) are proposed to reconstitute the cardiac and the vascular-smooth-muscle-type KATP channels, respectively. We report that potassium channel openers (KCOs) bind to and act through SURs and that binding to SUR1, SUR2A and SUR2B requires ATP. Non-hydrolysable ATP-analogues do not support binding, and Mg2+ or Mn2+ are required. Point mutations in the Walker A motifs or linker regions of both nucleotide-binding folds (NBFs) abolish or weaken [3H]P1075 binding to SUR2B, rendering reconstituted SUR2B/KIR6.2 channels insensitive towards KCOs. The C-terminus of SUR affects KCO affinity with SUR2B approximately SUR1 > SUR2A. KCOs belonging to different structural classes inhibited specific [3H]P1075 binding to SUR2B in a monophasic manner, with the exception of minoxidil sulfate, which induced a biphasic displacement. The affinities of KCO binding to SUR2B were 3.5-8-fold higher than their potencies for activation of SUR2B/KIR6.2 channels. The results establish that SURs are the KCO receptors of KATP channels and suggest that KCO binding requires a conformational change induced by ATP hydrolysis in both NBFs.


Subject(s)
ATP-Binding Cassette Transporters , Adenosine Triphosphate/metabolism , Potassium Channels, Inwardly Rectifying , Potassium Channels/metabolism , Receptors, Drug/metabolism , Animals , Binding Sites/genetics , Binding, Competitive , Cricetinae , Diazoxide/metabolism , Glyburide/metabolism , Guanidines/metabolism , Humans , Mutation , Peptide Fragments/genetics , Peptide Fragments/metabolism , Pinacidil/metabolism , Potassium Channels/agonists , Potassium Channels/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Pyridines/metabolism , Receptors, Drug/genetics , Recombinant Proteins/metabolism , Sulfonylurea Receptors
SELECTION OF CITATIONS
SEARCH DETAIL
...