Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Mol Endocrinol ; 27(9): 1502-17, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23831818

ABSTRACT

Steroidogenic acute regulatory protein (StAR) is indispensable for steroid hormone synthesis in the adrenal cortex and the gonadal tissues. This study reveals that StAR is also expressed at high levels in nonsteroidogenic cardiac fibroblasts confined to the left ventricle of mouse heart examined 3 days after permanent ligation of the left anterior descending coronary artery. Unlike StAR, CYP11A1 and 3ß-hydroxysteroid dehydrogenase proteins were not observed in the postinfarction heart, suggesting an apparent lack of de novo cardiac steroidogenesis. Work with primary cultures of rat heart cells revealed that StAR is induced in fibroblasts responding to proapoptotic treatments with hydrogen peroxide or the kinase inhibitor staurosporine (STS). Such induction of StAR in culture was noted before spontaneous differentiation of the fibroblasts to myofibroblasts. STS induction of StAR in the cardiac fibroblasts conferred a marked resistance to apoptotic cell death. Consistent with that finding, down-regulation of StAR by RNA interference proportionally increased the number of STS-treated apoptotic cells. StAR down-regulation also resulted in a marked increase of BAX activation in the mitochondria, an event known to associate with the onset of apoptosis. Last, STS treatment of HeLa cells showed that apoptotic demise characterized by mitochondrial fission, cytochrome c release, and nuclear fragmentation is arrested in individual HeLa cells overexpressing StAR. Collectively, our in vivo and ex vivo evidence suggests that postinfarction expression of nonsteroidogenic StAR in cardiac fibroblasts has novel antiapoptotic activity, allowing myofibroblast precursor cells to survive the traumatized event, probably to differentiate and function in tissue repair at the infarction site.


Subject(s)
Fibroblasts/metabolism , Fibroblasts/pathology , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Phosphoproteins/metabolism , Animals , Animals, Newborn , Apoptosis/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Female , Fibroblasts/drug effects , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , HeLa Cells , Humans , Mice , Mice, Inbred BALB C , Models, Biological , Myocardial Infarction/genetics , Myocardium/pathology , Myocardium/ultrastructure , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Phosphoproteins/genetics , Rats , Recombinant Proteins/pharmacology , Stress, Physiological/drug effects , bcl-2-Associated X Protein/metabolism
2.
Bioelectromagnetics ; 33(8): 634-40, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22532275

ABSTRACT

The purpose of this study was to determine the effect of extremely low frequency and weak magnetic fields (WMF) on cardiac myocyte Ca(2+) transients, and to explore the involvement of potassium channels under the WMF effect. In addition, we aimed to find a physical explanation for the effect of WMF on cardiac myocyte Ca(2+) transients. Indo-1 loaded cells, which were exposed to a WMF at 16 Hz and 40 nT, demonstrated a 75 ± 4% reduction in cytosolic Ca(2+) transients versus control. Treatment with the K(ATP) channel blocker, glibenclamide, followed by WMF at 16 Hz exposure, blocked the reduction in cytosolic calcium transients while treatment with pinacidil, a K(ATP) channel opener, or chromanol 293B, a selective potassium channel blocker of the delayed rectifier K(+) channels, did not inhibit the effect. Based on these finding and the ion cyclotron resonance frequency theory, we further investigated the effect of WMF by changing the direct current (DC) magnetic field (B(0) ). When operating different DC magnetic fields we showed that the WMF value changed correspondingly: for B(0) = 44.5 µT, the effect was observed at 17.05 Hz; for B(0) = 46.5 µT, the effect was observed at 18.15 Hz; and for B(0) = 49 µT the effect was observed at 19.1 Hz. We can conclude that the effect of WMF on Ca(2+) transients depends on the DC magnetic field level.


Subject(s)
Calcium/metabolism , Electric Conductivity , Electromagnetic Fields , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/radiation effects , Animals , Indoles/pharmacology , KATP Channels/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Rats
3.
Lasers Surg Med ; 42(6): 473-80, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20662023

ABSTRACT

BACKGROUND AND OBJECTIVE: Light in the visible and near infrared region stimulates various cellular processes, and thus has been used for therapeutic purposes. One of the proposed mechanisms is based on cellular production of reactive oxygen species (ROS) in response to illumination. In the present study, we followed visible light (VL)-induced hydroxyl radicals in various cell types and cellular sites using the electron paramagnetic resonance (EPR) spin-trapping technique. MATERIALS AND METHODS: Fibroblasts, sperm cells, cardiomyocytes, and skeletal muscle cells were irradiated with broadband (400-800 nm) VL. To detect ROS, the EPR spin-trapping technique coupled with the spin-traps 5,5-dimethyl pyrroline-N-oxide (DMPO) or 5-(diethoxyphosphoryl)-5-methyl-1-pyrroline-N-oxide (DEPMPO) were used. To investigate the cellular sites of ROS formation, the cell-permeable molecule, isopropanol, or the nonpermeable proteins, bovine serum albumin (BSA) and superoxide dismutase (SOD), were introduced to the cells before irradiation. ROS production in mitochondria was measured using the fluorescent probe, MitoTracker Red (MTR). RESULTS AND CONCLUSIONS: The concentration of .OH increased both with illumination time and with cell concentration, and decreased when N(2) was bubbled into the cell culture, suggesting that VL initiates a photochemical reaction via endogenous photosensitizers. VL was found to stimulate ROS generation both in membrane and cytoplasm. In addition, fluorescent measurments confirmed the mitochondria to be target for light-cell interaction. The findings support the hypothesis that ROS are generated in various cellular sites following light illumination.


Subject(s)
Fibroblasts/metabolism , Light , Muscle, Skeletal/cytology , Myocytes, Cardiac/metabolism , Reactive Oxygen Species/metabolism , Spermatozoa/metabolism , Animals , Cattle , Cell Line , Cell Membrane/metabolism , Cyclic N-Oxides , Cytoplasm/metabolism , Electron Spin Resonance Spectroscopy , Fluorescence , Hydroxyl Radical/metabolism , Male , Mitochondria/metabolism , Muscle, Skeletal/metabolism , Pyrroles , Rats
4.
Transplantation ; 87(8): 1155-62, 2009 Apr 27.
Article in English | MEDLINE | ID: mdl-19384161

ABSTRACT

BACKGROUND AND AIM: Mitochondrial calcium overload triggers apoptosis and also regulates ATP production. ATP and uridine-5'-triphosphate (UTP) depletion from hepatic tissue after ischemia causes cell death. ATP and UTP binds to cell membranes of the hepatocytes through P2Y receptors. Our aim was to investigate the role of UTP on the hepatic injury induced by ischemia. METHODS: Isolated mouse livers were randomly divided into five groups: (1) control group; (2) ischemic group (90 min); (3) as group 2, but with the administration of UTP; (4) as group 2, but with the administration of suramin, a P2Y antagonist; and (5) as group 3, but with the simultaneous administration of suramin and UTP. RESULTS: There was a postischemic significant reduction in the release of liver enzymes in the animals pretreated with UTP, the intrahepatic caspase-3 activity was significantly decreased, and the intrahepatic ATP content increased compared with group 2 (ischemic untreated). UTP prevented intracellular Ca overload after hypoxia in hepatocyte cultures. In the UTP-treated groups, significantly fewer apoptotic hepatocyte cells were noted by weaker activation of caspase-3 and by the transferase-mediated dUTP nick end labeling assay. The administration of suramin prevented the beneficial effect of endogenous ATP. UTP treatment attenuated the degradation of IkappaBalpha (nuclear factor-kappaB inhibitor) by 80% during reperfusion with no effect on c-Jun N terminal kinase phosphorylation. CONCLUSION: The administration of UTP before induction of ischemia-reperfusion can attenuate hepatic injury. UTP administration decreased cytosolic Ca overload in hypoxic conditions. UTP-mediated protective effects may be regulated through nuclear factor- kappaB inactivation. These findings have important implications for the potential use of UTP in ischemic hepatic injury.


Subject(s)
Liver/injuries , Reperfusion Injury/prevention & control , Uridine Triphosphate/therapeutic use , Alanine Transaminase/metabolism , Animals , Apoptosis/drug effects , Aspartate Aminotransferases/metabolism , Blood Pressure , Caspase 3/metabolism , Cell Hypoxia , Cells, Cultured , Hepatocytes/drug effects , Hepatocytes/physiology , L-Lactate Dehydrogenase/metabolism , Liver/cytology , Liver/drug effects , Liver/enzymology , Male , Mice , Suramin/therapeutic use , Vena Cava, Inferior/physiology
5.
Cell Calcium ; 43(4): 324-33, 2008 Apr.
Article in English | MEDLINE | ID: mdl-17692373

ABSTRACT

The involvement of nitric oxide (NO) in the late phase of ischemic preconditioning is well established. However, the role of NO as a trigger or mediator of "classic preconditioning" remains to be determined. The present study was designed to investigate the effects of NO on calcium homeostasis in cultured newborn rat cardiomyocytes in normoxia and hypoxia. We found that treatment with the NO donor, sodium nitroprusside (SNP) induced a sustained elevation of intracellular calcium level ([Ca(2+)](i)) followed by a decrease to control levels. Elevation of extracellular calcium, which generally occurs during ischemia, caused an immediate increase in [Ca(2+)](i) and arrhythmia in cultures of newborn cardiomyocytes. Treatment with SNP decreased [Ca(2+)](i) to control levels and re-established synchronized beating of cardiomyocytes. A decrease in extracellular [Na(+)], which inhibits the Na(+)/Ca(2+) exchanger, did not prevent [Ca(2+)](i) reduction by SNP. In contrast, application of thapsigargin, an inhibitor of sarcoplasmic reticulum Ca(2+)-ATPase (SERCA2a), increased [Ca(2+)](i), and in its presence, SNP did not reduce [Ca(2+)](i), indicating that Ca(2+) reduction is achieved via activation of SERCA2a. The results obtained suggest that activation of SERCA2a by SNP increases Ca(2+) uptake into the sarcoplasmic reticulum (SR) and prevents cytosolic Ca(2+) overload, which might explain the protective effect of SNP from hypoxic damage.


Subject(s)
Calcium/metabolism , Ischemic Preconditioning , Myocytes, Cardiac/metabolism , Nitric Oxide/metabolism , Adenosine Triphosphate/metabolism , Animals , Animals, Newborn , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation , Homeostasis , Hypoxia , Myocardial Contraction/physiology , Myocytes, Cardiac/cytology , Nitric Oxide Donors/metabolism , Nitroprusside/metabolism , Rats , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
6.
Life Sci ; 78(10): 1098-102, 2006 Feb 02.
Article in English | MEDLINE | ID: mdl-16202428

ABSTRACT

Leptin, a circulating hormone mainly produced by adipose tissue, regulates fatty acid metabolism and causes multiple systemic biological actions even the regulation of cardiovascular function. It is previously known that leptin is a hypoxia-inducible hormone, that hypoxic conditions increase the expression of this peptide in various tissues such as placenta, pancreas and also in the heart. Since leptin receptors are present in the heart, we hypothesized that whether leptin was a protector response for tissues especially for the heart against the deleterious effects of hypoxia. Cultured cardiomyocytes from newborn rats were initially treated with 3000 ng/ml leptin incubation for 1, 5 and 20 h separately, then subjected to 120 min of hypoxia. Hypoxic damage of myocytes was assayed using the measurements of both lactate dehydrogenase and creatine kinase releases into the medium and performing morphological observations (ultrastructural and immunocytochemical) of plates. The obtained results from leptin treated and non-treated control groups were compared to each other, and these data have demonstrated that 5 h of leptin treatment before hypoxia provides a significant protection for cardiomyocytes against hypoxia. Neither 1- nor 20-h leptin treated groups exhibited sufficient protection against hypoxia. In conclusion, leptin protects the cardiomyocyte cultures from hypoxia, but this effect is selective and evident only in the 5-h treated myocytes.


Subject(s)
Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Leptin/pharmacology , Myocytes, Cardiac/drug effects , Animals , Cells, Cultured , Creatine Kinase/blood , Culture Media , Desmin/pharmacology , Immunohistochemistry , L-Lactate Dehydrogenase/blood , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/ultrastructure , Rats
7.
J Mol Cell Cardiol ; 39(1): 149-58, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15893762

ABSTRACT

Mitochondrial disorder is characteristic of many myocardial injuries such as endotoxemia, shock, acidosis, ischemia/reperfusion, and others. The goal of possible therapy is to increase ATP production. Derivatives of vitamins K may be a potent electron carrier between various mitochondrial electron-donating and electron-accepting enzyme complexes. We aimed to test the possibility that menadione or its water-soluble derivative AK-135, the newly synthesized analogues of vitamin K1--N-derivatives of 2-methyl-3-aminomethyl 1.4-naphthoquinone, would reduce cardiomyocyte damage after hypoxia or mitochondrial respiratory chain inhibition in culture. Menadione, and more effectively, AK-135, restored the electron flow in defective respiratory chain (hypoxia or rotenone) systems. As was shown in this study, 3 microM of AK-135 restored ATP production after blockade of electron flow through mitochondrial complex I with 5 microM rotenone up to 13.18+/-1.56 vs. 3.21+/-1.12 nmol/mg protein in cells treated with rotenone only. In cultures pretreated with 4 microM dicumarol (DT-diaphorase inhibitor), the protective effect of AK-135 and menadione was abolished completely (1.67+/-1.43 and 2.97+/-0.57 nmol/mg protein, respectively). Inhibition of mitochondrial oxidative phosphorylation caused an increase in intracellular Ca(2+) levels. Here we have demonstrated restoration of calcium oscillations and cardiomyocyte contractility by menadione and its derivative after blockade of NADH: ubiquinone oxidoreductase with rotenone, and decrease of Ca(2+) overloading during hypoxia.


Subject(s)
Mitochondrial Diseases/drug therapy , Myocytes, Cardiac/drug effects , Vitamin K 3/pharmacology , Adenosine Triphosphate/metabolism , Animals , Calcium/metabolism , Cell Hypoxia , Cells, Cultured , L-Lactate Dehydrogenase/drug effects , L-Lactate Dehydrogenase/metabolism , Membrane Potentials/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Diseases/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , NAD(P)H Dehydrogenase (Quinone)/drug effects , NAD(P)H Dehydrogenase (Quinone)/metabolism , Naphthoquinones/pharmacology , Rats , Rotenone/pharmacology , Uncoupling Agents/pharmacology , Vitamin K 3/analogs & derivatives
8.
Am J Physiol Heart Circ Physiol ; 288(6): H2792-801, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15681707

ABSTRACT

Activation of either the A(1) or the A(3) adenosine receptor (A(1)R or A(3)R, respectively) elicits delayed cardioprotection against infarction, ischemia, and hypoxia. Mitochondrial contribution to the progression of cardiomyocyte injury is well known; however, the protective effects of adenosine receptor activation in cardiac cells with a respiratory chain deficiency are poorly elucidated. The aim of our study was to further define the role of A(1)R and A(3)R activation on functional tolerance after inhibition of the terminal link of the mitochondrial respiratory chain with sodium azide, in a state of normoxia or hypoxia, compared with the effects of the mitochondrial ATP-sensitive K(+) channel opener diazoxide. Treatment with 10 mM sodium azide for 2 h in normoxia caused a considerable decrease in the total ATP level; however, activation of adenosine receptors significantly attenuated this decrease. Diazoxide (100 muM) was less effective in protection. During treatment of cultured cardiomyocytes with hypoxia in the presence of 1 mM sodium azide, the A(1)R agonist 2-chloro-N(6)-cyclopentyladenosine was ineffective, whereas the A(3)R agonist 2-chloro-N(6)-iodobenzyl-5'-N-methylcarboxamidoadenosine (Cl-IB-MECA) attenuated the decrease in ATP level and prevented cell injury. Cl-IB-MECA delayed the dissipation in the mitochondrial membrane potential during hypoxia in cells impaired in the mitochondrial respiratory chain. In cells with elevated intracellular Ca(2+) concentration after hypoxia and treatment with NaN(3) or after application of high doses of NaN(3), Cl-IB-MECA immediately decreased the elevated intracellular Ca(2+) concentration toward the diastolic control level. The A(1)R agonist was ineffective. This may be especially important for the development of effective pharmacological agents, because mitochondrial dysfunction is a leading factor in the pathophysiological cascade of heart disease.


Subject(s)
Heart/physiology , Mitochondria, Heart/physiology , Muscle Cells/physiology , Receptor, Adenosine A1/physiology , Receptor, Adenosine A3/physiology , Adenosine A1 Receptor Agonists , Adenosine A3 Receptor Agonists , Adenosine Triphosphate/metabolism , Animals , Calcium/physiology , Cell Hypoxia , Cells, Cultured , Diazoxide/pharmacology , Homeostasis , Mitochondria, Heart/pathology , Muscle Cells/cytology , Potassium Channels/physiology , Rats , Sodium Azide/pharmacology
9.
J Mol Histol ; 35(5): 463-70, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15571324

ABSTRACT

This study examined whether triiodo-L-thyronine (T3) affects the expression of the major intercellular channel protein, connexin-43, and contractile protein alpha-sarcomeric actin. Cultured cardiomyocytes from newborn rats were treated on day three in culture with 10 or 100 nM T3 and examined 48 and 72 h thereafter. Treated and untreated cells were examined by immunofluorescence and electron microscopy. Expression levels of Cx43 and sarcomeric alpha-actin were monitored by Western blot analysis. Immunofluorescence labeling showed cell membrane location of Cx43 in punctuate gap junctions, whereby fluorescence signal area was significantly higher in cultured cardiomyocytes exposed to T3. This correlated with electron microscopical findings showing increased numbers and size of gap junction profiles, as well as with a significant dose-dependent increase of Cx43 expression detected by Western blot. Immunofluorescence of sarcomeric a-actin was enhanced and its expression increased dose- and time-dependently in T3-treated cultured heart myocytes. However, exposure to the higher dosage (100 nM) of T3 caused mild disintegration of sarcomeric a-actin in some myocytes, suggesting an over-dosage. The results indicate that T3 up-regulates Cx43 and accelerates gap junction formation in cultured neonatal cardiomyocytes. They suggest that thyroid status cannot only modulate the mechanical function of cardiomyocytes but also cell-to-cell communication essential for myocardial electrical and metabolic synchronizations.


Subject(s)
Actins/biosynthesis , Connexin 43/biosynthesis , Gene Expression Regulation/drug effects , Myocytes, Cardiac/metabolism , Triiodothyronine/pharmacology , Actins/genetics , Animals , Cells, Cultured , Connexin 43/genetics , Dose-Response Relationship, Drug , Myocytes, Cardiac/ultrastructure , Rats
10.
Cell Calcium ; 36(5): 387-96, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15451622

ABSTRACT

Intracellular calcium signaling cascade induced by adenosine A(3) receptor activation was studied in this work. It was found that adenosine A(3) receptor activation (and not A(1) or A(2A) adenosine receptors activation) leads to an increase in cytosolic calcium and its further extrusion. A selective A(3) agonist Cl-IB-MECA (2-chloro-N(6)-(3-iodobenzyl)adenosine-5'-N-methyluronamide) induced an increase in cytoplasmic calcium in a dose-dependent manner, and was independent on extracellular calcium. The Ca(2+) signal in newborn cardiomyocytes, induced by A(3) receptor activation, is dependent on a pertussis toxin-sensitive G-protein. The action of Cl-IB-MECA was not inhibited by an inhibitor of phospholipase C (PLC), and by antagonists to inositol 1,4,5-trisphosphate (IP(3)) receptor. In contrast, inhibition of ryanodine receptor prevented calcium elevation induced by this agonist. It was shown that extrusion of the elevated cytosolic Ca(2+) was achieved via activation of sarcoplasmic reticulum (SR) Ca(2+)-reuptake and of sarcolemmal Na(+)/Ca(2+) exchanger (NCX). The increase in the SR Ca(2+)-uptake and NCX Ca(2+) efflux were sufficient not only for compensation of Ca(2+) release from SR after A(3) receptor activation, but also for an effective prevention of extensive increase in intracellular Ca(2+) and may provide mechanism against cellular Ca(2+) overload. In cells with elevated [Ca(2+)](i) (due to increase of [Ca(2+)](o)), adenosine or Cl-IB-MECA decreased the [Ca(2+)](i) toward diastolic control level, whereas agonist of A(1) receptor was ineffective. The protective effect of A(3) receptor agonist was abolished in the presence of selective A(3) receptor antagonist MRS1523.


Subject(s)
Adenosine/analogs & derivatives , Calcium Signaling/physiology , Myocytes, Cardiac/physiology , Receptor, Adenosine A3/physiology , Adenosine/pharmacology , Adenosine A3 Receptor Agonists , Animals , Animals, Newborn , Calcium Signaling/drug effects , Cells, Cultured , Myocytes, Cardiac/drug effects , Rats , Ryanodine/pharmacology
11.
Biochem Biophys Res Commun ; 300(1): 23-8, 2003 Jan 03.
Article in English | MEDLINE | ID: mdl-12480515

ABSTRACT

The changes measured in intracellular fluorescein fluorescence polarization (IFFP) are used as a new tool for tracing cytoplasmic effects during contractile cycles of cardiac myocytes (1-2-day-old rat hearts), in addition to the established Ca(2+) monitoring and/or videometric methods of tracking cell-shortening. This novel method was found to be non-intrusive to the contraction cycles. The decay of the transient IFFP signal (from 0.220+/-0.01 to 0.170+/-0.013) seems to be closely related to the extended phase of contractile activation. This fact was further supported when Ca(2+) exchanger inhibitor was introduced and significantly decreased (90%) the rate of beats of contraction and IFFP, but not the Ca(2+) beat rate changes. This result suggests that the IFFP indicator is probably associated with the physiological activation, rather than with Ca(2+) alterations. The IFFP measure monitors the average of effective changes in the micro-viscosity of the cytoplasm protein matrix, associated with cellular activation.


Subject(s)
Fluorescence Polarization/methods , Myocardial Contraction/physiology , Animals , Calcium Signaling , Cells, Cultured , Fluorescein , Myocardium/cytology , Myocardium/metabolism , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...