Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Neuroscience ; 532: 103-112, 2023 11 10.
Article in English | MEDLINE | ID: mdl-37778690

ABSTRACT

At the vertebrate neuromuscular junction (NMJ), presynaptic homeostatic potentiation (PHP) refers to an increase in neurotransmitter release that restores the strength of synaptic transmission following a blockade of nicotinic acetylcholine receptors (nAChRs). Mechanisms informing the presynaptic terminal of the loss of postsynaptic receptivity remain poorly understood. Previous research at the mouse NMJ suggests that extracellular protons may function as a retrograde signal that triggers an upregulation of neurotransmitter output (measured by quantal content, QC) through the activation of acid-sensing ion channels (ASICs). We further investigated the pH-dependency of PHP in an ex-vivo mouse muscle preparation. We observed that increasing the buffering capacity of the perfusion saline with HEPES abolishes PHP and that acidifying the saline from pH 7.4 to pH 7.2-7.1 increases QC, demonstrating the necessity and sufficiency of extracellular acidification for PHP. We then sought to uncover how the blockade of nAChRs leads to the pH decrease. Plasma-membrane calcium ATPase (PMCA), a calcium-proton antiporter, is known to alkalize the synaptic cleft following neurotransmission in a calcium-dependent manner. We hypothesize that since nAChR blockade reduces postsynaptic calcium entry, it also reduces the alkalizing activity of the PMCA, thereby causing acidosis, ASIC activation, and QC upregulation. In line with this hypothesis, we found that pharmacological inhibition of the PMCA with carboxyeosin induces QC upregulation and that this effect requires functional ASICs. We also demonstrated that muscles pre-treated with carboxyeosin fail to generate PHP. These findings suggest that reduced PMCA activity causes presynaptic homeostatic potentiation by activating ASICs at the mouse NMJ.


Subject(s)
Calcium , Neuromuscular Junction , Animals , Mice , Calcium/metabolism , Neuromuscular Junction/metabolism , Synaptic Transmission , Presynaptic Terminals/metabolism , Acid Sensing Ion Channels , Neurotransmitter Agents/pharmacology , Hydrogen-Ion Concentration , Calcium-Transporting ATPases/pharmacology
2.
Microvasc Res ; 150: 104572, 2023 11.
Article in English | MEDLINE | ID: mdl-37353069

ABSTRACT

Pathological cardiac hypertrophy is an adaptive reaction in response to pressure or volume overload. Autophagy is critical for damage caused by pathological cardiac hypertrophy. Vacuole membrane protein 1 (VMP1) is an endoplasmic reticulum (ER) transmembrane protein that is effective in activating autophagy. However, the role of VMP1 in pathological cardiac hypertrophy and its underlying mechanisms remain elusive. This study was designed to explore the potential mechanisms of VMP1 on pressure overload-induced pathological cardiac hypertrophy. In this work, abdominal aorta constriction (AAC) surgery was used to induce pathological cardiac hypertrophy in male C57BL/6 mice. H9C2 cardiomyocytes were treated with phenylephrine stimulation (PE) to induce the hypertrophic response. The in vivo results revealed that mice with AAC surgery caused pathological cardiac hypertrophy as evidenced by improved cardiac function according to multiple echocardiographic parameters. Moreover, elevated VMP1 expression was also observed in mice after AAC surgery. VMP1 knockdown aggravated changes in cardiac structure, cardiac dysfunction, and fibrosis. Meanwhile, VMP1 knockdown suppressed autophagy and endoplasmic reticulum calcium ATPase (SERCA) activity in heart tissues. H9C2 cardiomyocytes with VMP1 overexpression were used to investigate the specific mechanism of VMP1 in pathological cardiac hypertrophy, and VMP1 overexpression increased autophagic flux by upregulating SERCA activity. In conclusion, these findings revealed that VMP1 protected against pressure overload-induced pathological cardiac hypertrophy by inducing SERCA-regulated autophagic flux. Our results provide valuable insights regarding the pathophysiology of pathological cardiac hypertrophy and clues to a novel target for the treatment of pathological cardiac hypertrophy.


Subject(s)
Cardiomegaly , Myocytes, Cardiac , Male , Mice , Animals , Mice, Inbred C57BL , Cardiomegaly/metabolism , Myocytes, Cardiac/pathology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Autophagy , Calcium-Transporting ATPases/metabolism , Calcium-Transporting ATPases/pharmacology
3.
Eur J Pharmacol ; 509(1): 77-83, 2005 Feb 10.
Article in English | MEDLINE | ID: mdl-15713432

ABSTRACT

Studies were performed to see if alterations in Ca2+ homeostasis underlie the gastrointestinal motility complications seen in many diabetic patients. Experiments were performed on colonic and ileal tissues taken from streptozotocin-induced diabetic and control rats. Diabetes caused alterations in the responses of the tissues to Ca2+ manipulation but these differed between the colon and ileum. In the colon a small but not significant increase in contractile responses to CaCl2 was observed in diabetic tissues, whereas the responses of the ileum were depressed relative to those of the controls. In contrast, responses of the diabetic ileum to the Ca2+ channel agonist Bay K8644 were greater than those of the controls, whilst the agonist failed to contract the colon. Similarly, the Ca2+-ATPase inhibitors, thapsigargin and cyclopiazonic acid, produced contractions which were greater in diabetic ileal tissues. Thus, alterations in the responses of the diabetic gut to Ca2+ manipulation are complex, and also tissue-specific.


Subject(s)
Calcium Signaling/physiology , Colon, Ascending/physiology , Diabetes Mellitus, Experimental/chemically induced , Ileum/physiology , Streptozocin/adverse effects , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Animals , Calcimycin/pharmacology , Calcium Chloride/pharmacology , Calcium-Transporting ATPases/antagonists & inhibitors , Calcium-Transporting ATPases/pharmacology , Colon, Ascending/drug effects , Colon, Ascending/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Ileum/drug effects , Ileum/pathology , Indoles/pharmacology , Injections, Intraperitoneal , Male , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , Nifedipine/pharmacology , Rats , Rats, Wistar , Streptozocin/administration & dosage , Thapsigargin/pharmacology , Time Factors
4.
J Neurophysiol ; 93(3): 1468-75, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15483058

ABSTRACT

Recent reports on rod photoreceptor neuroprotection by Ca2+ channel blockers have pointed out the need to assess the effect of these blockers on mammalian rods. However, in mammals, rod electrophysiological characterization has been hampered by the small size of these photoreceptors, which were instead extensively studied in nonmammalian vertebrates. To further characterize ionic conductances and to assess the pharmacology of Ca2+ channels in mammalian rods, freshly dissociated pig rod photoreceptors were recorded with the whole cell patch-clamp technique. Rod cells expressed 1) a hyperpolarization-activated inward-rectifying conductance (I(h)) sensitive to external Cs+; 2) a sustained outward K+ current (I(K)) sensitive to tetraethylammonium; 3) a sustained voltage-gated Ca2+ current (I(Ca)) sensitive to benzothiazepine (diltiazem) and phenylalkylamine (verapamil) derivatives; 4) a Ca(2+)-activated Cl- current (I(Cl(Ca))); and 5) a plasma membrane Ca(2+)-ATPase. The Ca2+ current showed a range of activation from positive potentials to -60 mV with a maximum between -30 and -20 mV. In contrast to other L-type Ca2+ channels, rod Ca2+ channels were blocked at similar and relatively high concentrations by the diltiazem isomers and verapamil. The biphasic dose-response for D-diltiazem confirmed the low sensitivity of Ca2+ channels for the molecule. The ATPase, which was localized at the axon terminal, was found to contribute to Ca2+ extrusion. These results suggest that the electrophysiological features of rod photoreceptors had been preserved during evolution from nonmammalian vertebrates to mammals. This work indicates further that mammalian rods express nonclassic L-type Ca2+ channels, showing a low sensitivity to the diltiazem isomers used in neuroprotective studies.


Subject(s)
Calcium Channels/physiology , Calcium/metabolism , Homeostasis/physiology , Ion Channel Gating/physiology , Retinal Rod Photoreceptor Cells/physiology , Animals , Anions/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels/drug effects , Calcium Channels/radiation effects , Calcium-Transporting ATPases/metabolism , Calcium-Transporting ATPases/pharmacology , Cation Transport Proteins , Cations/pharmacology , Cells, Cultured , Cesium/pharmacology , Chlorides/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Electric Stimulation/methods , Immunohistochemistry/methods , Ion Channel Gating/drug effects , Ion Channel Gating/radiation effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Patch-Clamp Techniques/methods , Plasma Membrane Calcium-Transporting ATPases , Potassium Channel Blockers/pharmacology , Retinal Rod Photoreceptor Cells/drug effects , Retinal Rod Photoreceptor Cells/radiation effects , Swine , Tetraethylammonium/pharmacology
5.
Cell Calcium ; 36(1): 1-9, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15126051

ABSTRACT

The effect of sarcoendoplasmic reticulum Ca(2+)-ATPase (SERCA) inhibition on the cytoplasmic Ca(2+) concentration ([Ca(2+)](i)) was studied in primary insulin-releasing pancreatic beta-cells isolated from mice, rats and human subjects as well as in clonal rat insulinoma INS-1 cells. In Ca(2+)-deficient medium the individual primary beta-cells reacted to the SERCA inhibitor cyclopiazonic acid (CPA) with a slow rise of [Ca(2+)](i) followed by an explosive transient elevation. The [Ca(2+)](i) transients were preferentially observed at low intracellular concentrations of the Ca(2+) indicator fura-2 and were unaffected by pre-treatment with 100 microM ryanodine. Whereas 20mM caffeine had no effect on basal [Ca(2+)](i) or the slow rise in response to CPA, it completely prevented the CPA-induced [Ca(2+)](i) transients as well as inositol 1,4,5-trisphosphate-mediated [Ca(2+)](i) transients in response to carbachol. In striking contrast to the primary beta-cells, caffeine readily mobilized intracellular Ca(2+) in INS-1 cells under identical conditions, and such mobilization was prevented by ryanodine pre-treatment. The results indicate that leakage of Ca(2+) from the endoplasmic reticulum after SERCA inhibition is feedback-accelerated by Ca(2+)-induced Ca(2+) release (CICR). In primary pancreatic beta-cells this CICR is due to activation of inositol 1,4,5-trisphosphate receptors. CICR by ryanodine receptor activation may be restricted to clonal beta-cells.


Subject(s)
Calcium Channels/metabolism , Calcium-Transporting ATPases/pharmacology , Calcium/metabolism , Islets of Langerhans/drug effects , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Caffeine/pharmacology , Calcium/pharmacology , Calcium Channels/drug effects , Calcium Signaling/drug effects , Calcium-Transporting ATPases/antagonists & inhibitors , Calcium-Transporting ATPases/metabolism , Cell Line , Chelating Agents/pharmacology , Enzyme Inhibitors/pharmacology , Humans , Inositol 1,4,5-Trisphosphate Receptors , Islets of Langerhans/metabolism , Mice , Mice, Obese , Rats , Rats, Wistar , Receptors, Cytoplasmic and Nuclear/drug effects , Ryanodine/pharmacology , Ryanodine Receptor Calcium Release Channel/drug effects , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/enzymology , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Time Factors
6.
Toxicol Lett ; 148(1-2): 133-9, 2004 Mar 14.
Article in English | MEDLINE | ID: mdl-15019097

ABSTRACT

Metallothionein (MT) is a small sulfhydryl-rich protein whose levels are elevated by various inducers of organelle stresses, such as nuclear stress (cisplatin), mitochondrial stress (antimycin A, 2,4-dinitrophenol) and lysosomal stress (paraquat). Although abnormal folding of protein in the endoplasmic reticulum (ER) causes ER stress, induction of MT synthesis by ER stress has never been investigated. In this study, we examined the induction of MT by an inducer of ER stress, tunicamycin (Tun), which induces ER stress by inhibiting N-linked glycosylation of protein in the ER. Administration of Tun (0.5-1.5 mg/kg, sc) increased hepatic MT levels in C57BL/6J mice (3.1-fold). The maximal increase in hepatic MT was observed 48-96 h after the administration of Tun (1.0 mg/kg). Expressions of MT-I, II and glucose-regulated protein 78 (Bip/GRP78), which is a molecular chaperone induced by ER stress, mRNA were also detected by administration of Tun. Thapsigargin (Thap), a generator of ER stress by inhibiting ER Ca(2+)-ATPase, also increased both hepatic MT levels and expression of MT-I and -II mRNA. The level of expression of Bip/GRP78 mRNA induced by Tun administration in MT-null mice was greater than that in wild-type mice. Taken together, these findings suggest that inhibitors of ER are potent inducers of MT.


Subject(s)
Anti-Bacterial Agents/toxicity , Endoplasmic Reticulum/pathology , Endoplasmic Reticulum/physiology , Metallothionein/biosynthesis , Tunicamycin/toxicity , Animals , Calcium-Transporting ATPases/pharmacology , Endoplasmic Reticulum Chaperone BiP , Genotype , Glycosylation , Liver/chemistry , Male , Metallothionein/pharmacology , Mice , Mice, Inbred C57BL , RNA, Messenger/biosynthesis
7.
Cell Calcium ; 35(2): 131-9, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14706287

ABSTRACT

Using the Drosophila melanogaster S2 cell line, stably expressing a cloned muscarinic acetylcholine receptor (AChR), DM1, we have applied gene silencing by double-stranded RNA interference (RNAi) to knock down gene products involved in DM1-mediated calcium signalling. We have shown that RNAi knock down of either the inositol 1,4,5-trisphosphate receptor (Ins(1,4,5)P(3)R), or the SERCA calcium pump in the S2-DM1 cells blocks the increase in intracellular calcium concentration ([Ca(2+)](i)) resulting from activation of the DM1 receptor by 100 microM carbamylcholine (CCh). When RNAi designed to knock down the ryanodine receptor (RyR) was tested, there was no change in the calcium increase detected in response to CCh, consistent with a failure to detect RyRs in S2-DM1 cells using RT-PCR. A combination of RNAi and calcium imaging has provided a direct demonstration of key roles for the Ins(1,4,5)P(3)R and the SERCA pump in the response to DM1 receptor activation.Thus, we show that silencing of individual genes by RNAi in a well characterised Drosophila S2 cell line offers experimental opportunities for cell-signalling studies. Future investigations with RNAi libraries taking full advantage of the wealth of new information available from sequencing the Drosophila genome, may help identify novel components of cell-signalling pathways and functionally linked gene products.


Subject(s)
Calcium Signaling/genetics , Gene Silencing/physiology , RNA Interference/physiology , RNA, Double-Stranded/genetics , Animals , Calcium Signaling/drug effects , Calcium-Transporting ATPases/genetics , Calcium-Transporting ATPases/pharmacology , Cell Line , Drosophila melanogaster , Gene Silencing/drug effects , RNA, Double-Stranded/pharmacology , Sarcoplasmic Reticulum Calcium-Transporting ATPases
8.
Environ Toxicol Chem ; 22(11): 2761-7, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14587919

ABSTRACT

Increased Cd toxicity at low salinity has been attributed to increased free Cd2+ ion concentration ([Cd2+]sw), but transfer to dilute seawater also stimulates physiological ionic regulation in crabs. In this study, Cd accumulation and Ca homeostasis in the shore crab (Carcinus maenas) were explored at fixed [Cd2+]sw to reveal the physiological events during sublethal Cd exposure. Crabs were exposed to 3.4 or 34 microg/L [Cd2+], in both 100% seawater (SW) and 33% SW for up to 10 d and sampled for hemolymph composition as well as gill and hepatopancreas Ca, Cd, and Ca-ATPase activity. Cadmium exposure ameliorated the expected fall in hemolymph osmotic pressure and NaCl at low salinity and generally protected tissue Ca from decline. Cadmium exposure alone (within salinity) inhibited Ca-ATPase, but this was offset by stimulation of Ca-ATPase at low salinity. The Ca-ATPase activity in the anterior and posterior gills showed different responses to Cd/low salinity stress. Crabs were more sensitive to a 10-fold increase in [Cd2+]sw at low salinity. Overall, we conclude that exposure to a fixed sublethal [Cd2+]sw reveals a compensatory physiological response that is driven primarily by salinity rather than Cd2+ free ion concentration. Physiological responses are therefore important during low-level Cd exposure in dilute seawater.


Subject(s)
Brachyura/physiology , Cadmium/toxicity , Calcium/metabolism , Water Pollutants/toxicity , Animals , Calcium-Transporting ATPases/pharmacology , Gills/physiology , Homeostasis , Sodium Chloride
9.
Cell Calcium ; 34(2): 145-56, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12810056

ABSTRACT

Human bone marrow-derived mesenchymal stem cells (hMSCs) have the potential to differentiate into several types of cells. We have demonstrated spontaneous [Ca(2+)](i) oscillations in hMSCs without agonist stimulation, which result primarily from release of Ca(2+) from intracellular stores via InsP(3) receptors. In this study, we further investigated functions and contributions of Ca(2+) transporters on plasma membrane to generate [Ca(2+)](i) oscillations. In confocal Ca(2+) imaging experiments, spontaneous [Ca(2+)](i) oscillations were observed in 193 of 280 hMSCs. The oscillations did not sustain in the Ca(2+) free solution and were completely blocked by the application of 0.1mM La(3+). When plasma membrane Ca(2+) pumps (PMCAs) were blocked with blockers, carboxyeosin or caloxin, [Ca(2+)](i) oscillations were inhibited. Application of Ni(2+) or KBR7943 to block Na(+)-Ca(2+) exchanger (NCX) also inhibited [Ca(2+)](i) oscillations. Using RT-PCR, mRNAs were detected for PMCA type IV and NCX, but not PMCA type II. In the patch clamp experiments, Ca(2+) activated outward K(+) currents (I(KCa)) with a conductance of 170+/-21.6pS could be recorded. The amplitudes of I(KCa) and membrane potential (V(m)) periodically fluctuated liked to [Ca(2+)](i) oscillations. These results suggest that in undifferentiated hMSCs both Ca(2+) entry through plasma membrane and Ca(2+) extrusion via PMCAs and NCXs play important roles for [Ca(2+)](i) oscillations, which modulate the activities of I(KCa) to produce the fluctuation of V(m).


Subject(s)
Calcium Signaling/physiology , Calcium-Transporting ATPases/pharmacology , Calcium/metabolism , Cell Membrane/metabolism , Mesenchymal Stem Cells/metabolism , Sodium-Calcium Exchanger/pharmacology , Cells, Cultured , Humans , Membrane Potentials , Potassium Channels, Calcium-Activated/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sodium-Calcium Exchanger/antagonists & inhibitors
10.
Jpn J Physiol ; 53(6): 411-6, 2003 Dec.
Article in English | MEDLINE | ID: mdl-15038839

ABSTRACT

Hyperthyroidism is known to affect multiple organ functions, and thyroid hormone has been known to improve myocardial function in a failing heart. The purpose of this study is to elucidate the functional and metabolic effects of thyroid hormone on myocardium in a rat model exposed to long-term excess thyroid hormone, particularly focusing on the SR Ca(2+)-ATPase (SERCA2) function. 3,5,3'-Triiodo-L-thyronine (T3), or the vehicle, was subcutaneously given for 4 weeks (T3 and control [C] group). Bolus I.V. Thapsigargin (TG) was used to test the SERCA2 function (C-TG and T3-TG) in Langendorff perfused heart. Myocardial functions such as LV-developed pressure (LVDP; mmHg), +/- dP/dt (mmHg/s), tau (ms), and oxygen consumption (MVO(2); ml/min/g wt) were measured. SERCA2 and GLUT4 protein level were also evaluated by Western immunoblotting. Left ventricle to body weight (LV/BW) ratio was significantly higher in the T3 group. Both negative dP/dt and tau were significantly decreased by TG. It is interesting that the decrement of negative dP/dt and tau attained by TG was significantly larger in the hyperthyroid group (T3-TG) than in a normal heart (C-TG). SERCA2 and GLUT4 protein levels were not significantly different between control and the T3 group. We conclude that prolonged exposure to thyroid hormone causes hypertrophy of the myocardium and an augmentation of the SR Ca(2+) ATPase activity. Care must be taken in hyperthyroid heart during the ischemia-reperfusion process where the SRECA2 function is inhibited.


Subject(s)
Calcium-Transporting ATPases/pharmacology , Hyperthyroidism/complications , Myocardial Contraction/physiology , Myocardium/metabolism , Reperfusion Injury/physiopathology , Thyroid Hormones/pharmacology , Animals , Disease Models, Animal , Heart Ventricles/pathology , Rats , Rats, Wistar , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Triiodothyronine, Reverse/administration & dosage
11.
Jpn J Physiol ; 52(4): 361-9, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12519471

ABSTRACT

A simulation of some basic features of Ca(2+) release from the sarcoplasmic reticulum (SR) in cardiac muscle was made with a model based on the mechanism of Ca(2+)-induced Ca(2+)-release. The half-sarcomere modeled as a circular cylinder was divided into 20 annular elements in the radial, 50 slices in the axial, and 125 slices in the azimuthal direction. The cylindrical surface of the sarcomere was covered by a layer of the SR. The rate of Ca(2+) release from the terminal sac (TS) is proportional to the product of the open probability of the Ca(2+) release channel and the difference of [Ca(2+)] between the TS and an element facing the TS. Ca(2+) moves from element to element by simple diffusion and is taken up by the tubular SR via Ca(2+)-ATPase. Ca(2+) influx (I(ca)) to trigger the TS Ca(2+) release was introduced to either a single element facing the TS (local I(ca)) or to 20 elements aligned at the level of the Z-line (uniform I(ca)). The simulation showed that with both types of I(ca), TS Ca(2+) release is smoothly graded over a wide range of I(ca) with the TS moderately loaded with Ca(2+). The gain determined by dividing the total amount of TS Ca(2+) release by I(ca) was greater with local than with uniform I(ca). Mechanical alternans was simulated with both the local and uniform I(ca) with an appropriate rate of Ca(2+) replenishment to the TS. A Ca(2+) wave was simulated with a model consisting of 8 longitudinally consecutive sarcomeres with TS heavily loaded with Ca(2+). Thus the present model accounted for graded TS Ca(2+) release, mechanical alternans, and Ca(2+) wave in cardiac muscle at the same time.


Subject(s)
Calcium-Transporting ATPases/pharmacology , Calcium/pharmacokinetics , Heart/physiology , Models, Theoretical , Myocardium , Sarcoplasmic Reticulum/physiology , Cell Culture Techniques , Electrophysiology , Humans
12.
Circulation ; 104(12): 1424-9, 2001 Sep 18.
Article in English | MEDLINE | ID: mdl-11560860

ABSTRACT

BACKGROUND: In heart failure, sarcoplasmic reticulum (SR) Ca(2+)-ATPase (SERCA2a) activity is decreased, resulting in abnormal calcium handling and contractile dysfunction. We have previously shown that increasing SERCA2a expression by gene transfer improves ventricular function in a rat model of heart failure created by ascending aortic constriction. METHODS AND RESULTS: In this study, we tested the effects of gene transfer of SERCA2a on survival, left ventricular (LV) volumes, and metabolism. By 26 to 27 weeks after aortic banding, all animals developed heart failure (as documented by >25% decrease in fractional shortening) and were randomized to receive either an adenovirus carrying the SERCA2a gene (Ad.SERCA2a) or control virus (Ad.betagal-GFP) by use of a catheter-based technique. Sham-operated rats, uninfected or infected with either Ad.betagal-GFP or Ad.SERCA2a, served as controls. Four weeks after gene transfer, survival in rats with heart failure treated with Ad.betagal-GFP was 9%, compared with 63% in rats receiving Ad.SERCA2a. LV volumes were significantly increased in heart failure (0.64+/-0.05 versus 0.35+/-0.03 mL, P<0.02). Overexpression of SERCA2a normalized LV volumes (0.46+/-0.07 mL) in the failing hearts. (31)P NMR analysis showed a reduced ratio of phosphocreatine to ATP content in failing+Ad.betagal-GFP compared with sham+Ad.betagal-GFP (0.82+/-0.13 versus 1.38+/-0.14, P<0.01). Overexpression of SERCA2a in failing hearts improved the phosphocreatine/ATP ratio (1.23+/-0.28). CONCLUSIONS: In this study, we show that unlike inotropic agents that improve contractile function at the expense of increased mortality and worsening metabolism, gene transfer of SERCA2a improves survival and the energy potential in failing hearts.


Subject(s)
Calcium-Transporting ATPases/genetics , Calcium-Transporting ATPases/metabolism , Heart Failure/physiopathology , Heart Failure/therapy , Myocardium/metabolism , Adenoviridae/genetics , Animals , Calcium-Transporting ATPases/pharmacology , Disease Models, Animal , Echocardiography , Gene Expression , Gene Transfer, Horizontal , Genetic Therapy/methods , Genetic Vectors/genetics , Genetic Vectors/metabolism , Genetic Vectors/pharmacology , Heart Failure/pathology , In Vitro Techniques , Isoenzymes/genetics , Isoenzymes/metabolism , Magnetic Resonance Spectroscopy , Myocardial Contraction/drug effects , Myocardium/pathology , Organ Size/drug effects , Rats , Rats, Sprague-Dawley , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Stroke Volume/drug effects , Survival Rate
13.
Gen Pharmacol ; 31(4): 499-501, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9792206

ABSTRACT

1. Ouabain or a related stereoisomer, termed endogenous ouabain, has been identified in adrenal cortex tissue and culture medium from adrenocortical cells. 2. Angiotensin II and adrenocorticotropin, the main activators of aldosterone secretion from adrenal glomerulosa cells appear to increase the production of this compound. 3. The purpose of this review is to briefly discuss recent available experimental evidence suggesting that endogenous ouabain is secreted by the zona glomerulosa of the adrenal gland.


Subject(s)
Adrenal Cortex/metabolism , Ouabain/metabolism , Animals , Calcium-Transporting ATPases/pharmacology , Humans , Ouabain/blood , Sodium-Calcium Exchanger/pharmacology , Zona Glomerulosa/metabolism
14.
J Physiol ; 512 ( Pt 3): 651-67, 1998 Nov 01.
Article in English | MEDLINE | ID: mdl-9769411

ABSTRACT

1. The contribution of the sarcoplasmic reticulum (SR) and Na+-Ca2+ exchanger to intracellular Ca2+ regulation in mouse cardiac myocytes was investigated by measuring contraction after variable rest intervals, rapid cooling contractures (RCCs) and fast application of caffeine. The results obtained showed differences from other species in the roles played by the SR and the Na+-Ca2+ exchanger. They suggest that in mouse ventricular myocytes there is significant Ca2+ entry via the exchanger during rest and during the latter part of the Ca2+ transient. 2. In cardiac myocytes isolated from transgenic mice overexpressing the cardiac Na+-Ca2+ exchanger the time to peak and relaxation of twitches and RCCs were faster than in control littermates. The decline of Ca2+, assessed by indo-1 fluorescence, was faster in transgenic myocytes even in the absence of Na+ and Ca2+ in the superfusing solution. This suggests that SR Ca2+ uptake is faster in these myocytes. However, no difference in the expression of SERCA2a, phospholamban or calsequestrin measured with Western blotting could be found in the two groups. 3. We measured SR Ca2+ content by integrating the caffeine-induced transient inward current. The amount of Ca2+ stored in the SR of transgenic mouse myocytes was 69 % greater than in non-transgenic littermates. The increased SR Ca2+ content may be responsible for the faster rate of SR Ca2+ release and uptake in cells from transgenic mice. 4. We performed experiments to assess whether the reversal potential of the Na+-Ca2+ exchanger (ENa-Ca) was different in transgenic cardiac cells. We measured a Ni2+-sensitive current elicited by voltage ramps in non-dialysed myocytes. The current-voltage relationship showed no difference in the reversal potential of the Na+-Ca2+ exchanger in transgenic and control myocytes. This suggests that the effects on the SR Ca2+ content in transgenic cardiac myocytes can be ascribed to the overexpression of the exchanger and are not secondary to changes in intracellular diastolic Ca2+ and Na+.


Subject(s)
Calcium/metabolism , Myocardium/cytology , Myocardium/metabolism , Sarcoplasmic Reticulum/metabolism , Sodium-Calcium Exchanger/genetics , Sodium-Calcium Exchanger/metabolism , Animals , Caffeine/pharmacology , Calcium-Binding Proteins/pharmacology , Calcium-Transporting ATPases/pharmacology , Calsequestrin/pharmacology , Central Nervous System Stimulants/pharmacology , Electric Stimulation , Electrophysiology , Heart/drug effects , In Vitro Techniques , Membrane Potentials/physiology , Mice , Mice, Transgenic , Muscle Proteins/biosynthesis , Myocardial Contraction/drug effects , Myocardial Contraction/physiology , Myocardium/ultrastructure , Patch-Clamp Techniques , Sarcoplasmic Reticulum/drug effects
15.
Circ Res ; 83(6): 608-13, 1998 Sep 21.
Article in English | MEDLINE | ID: mdl-9742056

ABSTRACT

Alterations in the expression levels of the sarcoplasmic reticulum (SR) Ca2+-ATPase and its regulator, phospholamban, have been implicated in the effects of thyroxine hormone on cardiac function. To determine the role of phospholamban in these effects, hypothyroidism and hyperthyroidism were induced in phospholamban-deficient mice and their isogenic wild types. Hypothyroidism resulted in significant decreases of left ventricular contractility, which could be moderately stimulated by increases in preload or afterload, in both phospholamban-deficient and wild-type mice. However, the basal contractile parameters in hypothyroid phospholamban-deficient hearts were at least as high as those exhibited by hyperthyroid wild-type hearts. In hyperthyroidism, there was no further enhancement of the hyperdynamic contractile parameters in phospholamban-deficient hearts, although the wild-type hearts exhibited significantly increased contractile function compared with their respective euthyroid groups. Furthermore, increases in preload or afterload did not enhance contractility in either phospholamban-deficient or wild-type hyperthyroid hearts. Examination of the relative tissue levels of cardiac SR Ca2+-ATPase revealed increases in hyperthyroidism and decreases in hypothyroidism compared with euthyroidism, and these changes were similar between phospholamban-deficient and wild-type hearts. An opposite trend was observed for phospholamban expression levels in the wild-type group, which were depressed in hyperthyroid hearts but increased in hypothyroid hearts. These findings indicate that (1) thyroid hormones induce similar changes in the cardiac SR Ca2+-ATPase levels in either the presence or absence of phospholamban, (2) the thyroxine-induced increases in SR Ca2+-ATPase levels are not associated with any further stimulation of the hyperdynamic cardiac function in phospholamban-deficient mice, and (3) the decreased contractile parameters in hypothyroid phospholamban-deficient hearts associated with decreases in SR Ca2+-ATPase levels and myosin heavy chain isoform switches are at least as high as those of the stimulated hyperthyroid wild-type hearts. Thus, alterations in the phospholamban level or its activity may be a critical determinant of the contractile responses to altered thyroid states in the mammalian heart.


Subject(s)
Calcium-Binding Proteins/deficiency , Myocardium/chemistry , Thyroid Hormones/pharmacology , Animals , Calcium-Transporting ATPases/pharmacology , Female , Hypothyroidism/metabolism , Male , Mice , Myocardial Contraction/drug effects , Ventricular Function/physiology
16.
J Mol Cell Cardiol ; 30(12): 2683-94, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9990539

ABSTRACT

We examined the hypothesis that the angiotensin converting enzyme inhibitor ramipril at sub-antihypertensive concentrations could improve sarcoplasmic reticulum (SR) CaATPase expression and function in compensated hypertrophied rat hearts. Five weeks after abdominal aortic constriction, rats received a daily dose (50 micrograms/kg/day) of ramipril or vehicle for 4 weeks. Cardiac angiotensin-converting enzyme (ACE) activity increased with cardiac hypertrophy (CH) but returned to normal following ramipril treatment. SR CaATPase protein levels and activity decreased with CH (P < 0.05) and were normalized following ramipril treatment (P < 0.05 for protein and activity). No change in phospholamban (PLB) protein levels could be demonstrated between any of the groups. In contrast, ramipril treatment specifically increased control SR CaATPase and PLB mRNA levels by > 60% (P < 0.01) and > 30%, respectively. In the hypertrophied group, SR CaATPase increased by 35% (P < 0.05 n = 6) after ramipril treatment. Calsequestrin mRNA levels were unaffected by ramipril administration. In conclusion, ramipril normalizes SR CaATPase protein expression and function in pressure-overloaded and compensated CH. The effects of ramipril are however multifaceted, affecting RNA and protein expression differentially.


Subject(s)
Calcium-Transporting ATPases/metabolism , Cardiomegaly/enzymology , Ramipril/pharmacology , Sarcoplasmic Reticulum/enzymology , Animals , Blotting, Northern , Blotting, Western , Calcium/metabolism , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/pharmacology , Calcium-Transporting ATPases/pharmacology , Calsequestrin/pharmacology , Carotid Arteries/drug effects , Dose-Response Relationship, Drug , Guanosine Monophosphate/urine , Immunoblotting , Male , Oxalates/metabolism , Peptidyl-Dipeptidase A/metabolism , Pressure , Rats , Rats, Sprague-Dawley
17.
J Neurosci Res ; 46(6): 678-85, 1996 Dec 15.
Article in English | MEDLINE | ID: mdl-8978502

ABSTRACT

Two enzymes, protein kinase C and microsomal Ca(2+)-ATPase help regulate levels of Ca2+ in many types of cells. Since proteins that regulate Ca2+ often influence sensitivity to Pb2+, we determined the possible roles played by protein kinase C and microsomal Ca(2+)-ATPase for the Pb(2+)-evoked release of norepinephrine (NOR) in PC cells. NOR release was observed at 10 microM Pb2+ when PC 12 cells were stimulated with inhibitors of microsomal Ca(2+)-ATPase such as thapsigargin, cyclopiazonic acid, or 2,5-di-(t-butyl)-hydroquinone. At 5 microM, Pb2+ evoked the release of NOR in PC 12 cells stimulated with activators of protein kinase C such as phorbol 12-myristate 13-acetate (PMA) or (-)-7-octylindolactam. NOR release was observed at 1 microM Pb2+ in the presence of both PMA and thapsigargin. Ni2+ and Cd2+ blocked NOR release stimulated by Pb2+ in the presence of thapsigargin but not by PMA. NOR released by thapsigargin stimulation was not altered in PC 12 cells depleted of protein kinase C. Two proteins found in vesicles, chromogranin B and secretogranin-II were released with NOR. Our results indicate that in PC 12 cells, PB(2+)-evokes the release of neurotransmitters. Furthermore, thapsigargin and PMA increase the cell's sensitivity to Pb2+ by different pathways.


Subject(s)
Lead/pharmacology , Norepinephrine/metabolism , Animals , Cadmium/pharmacology , Calcium Channels/physiology , Calcium-Transporting ATPases/antagonists & inhibitors , Calcium-Transporting ATPases/pharmacology , Carcinogens/pharmacology , Chromogranins/metabolism , Enzyme Inhibitors/pharmacology , Ligands , Microsomes/drug effects , Microsomes/enzymology , Neuropeptides/metabolism , Nickel/pharmacology , Norepinephrine/pharmacokinetics , PC12 Cells/chemistry , PC12 Cells/drug effects , PC12 Cells/enzymology , Protein Kinase C/metabolism , Proteins/metabolism , Rats , Tetradecanoylphorbol Acetate/pharmacology , Thapsigargin/pharmacology , Tritium
18.
Neurochem Res ; 21(4): 471-9, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8734441

ABSTRACT

The ability of mature oligodendrocytes (OLs) to recover from insult is important in repair of damage following demyelination. Since regulation of Ca2+ levels within cells plays a critical role in function and survival, this study investigates the effects of changes in cytoplasmic Ca2+ on the viability of cultured mouse OLs and their ability to maintain membrane sheets. Mature OLs in culture respond rapidly to the calcium ionophore A23187 and promptly return to resting Ca2+ levels when the ionophore is removed. Longer exposure to 0.1-1.0 microM A23187 leads to microtubule disruption, membrane sheet retraction and eventual cell death; nuclear lysis occurs in many of the OLs, as reported by Scolding, et al. (1) for rat OLs. In our cultures, mature OLs were more susceptible to nuclear lysis than were immature OLs or astroglia. Release of intracellular Ca2+ stores with thapsigargin at 5-10 microM also leads to retraction of membrane sheets. Following 6 hours of continuous exposure to thapsigargin, the effects on membrane sheets are reversed over the next 12 hours. After 18 hours of continuous exposure to thapsigargin, only occasional nuclear lysis is observed, but a number of the mature OLs show signs of DNA fragmentation, indicating that apoptotic death is occurring. Our results suggest that mature OLs cannot survive a prolonged influx of extracellular calcium as readily as immature OLs and astroglia, but have mechanism to withstand similar increases in cytoplasmic Ca2+ following sustained release of intracellular stores.


Subject(s)
Calcimycin/pharmacology , Calcium/metabolism , Cell Death/drug effects , Oligodendroglia/metabolism , Animals , Animals, Newborn , Astrocytes/drug effects , Astrocytes/metabolism , Brain/cytology , Calcimycin/analogs & derivatives , Calcium-Transporting ATPases/pharmacology , Cell Membrane/drug effects , Cells, Cultured , Mice , Mice, Inbred BALB C , Oligodendroglia/drug effects , Thapsigargin/pharmacology
19.
Circ Res ; 77(5): 927-35, 1995 Nov.
Article in English | MEDLINE | ID: mdl-7554146

ABSTRACT

Intracellular Ca2+ pools contribute to changes in cytosolic [Ca2+] ([Ca2+]i), which play an important role in endothelial cell signaling. Recently, endothelial ryanodine-sensitive Ca2+ stores were shown to regulate agonist-sensitive intracellular Ca2+ pools. Since caffeine binds the ryanodine Ca2+ release channel on the endoplasmic reticulum in a variety of cell types, we examined the effect of caffeine on [Ca2+]i in human aortic endothelial cell monolayers loaded with the fluorescent probe indo 1. Under baseline conditions, 10 mmol/L caffeine induced a small increase in [Ca2+]i from 86 +/- 10 to 115 +/- 17 nmol/L (mean +/- SEM); this effect was similar to that of 5 mumol/L ryanodine and was unaffected by buffer Ca2+ removal. After depletion of an intracellular Ca2+ store by the irreversible endoplasmic reticulum Ca(2+)-ATPase inhibitor thapsigargin (1 mumol/L), ryanodine did not affect [Ca2+]i. In contrast, caffeine induced a large rapid increase in [Ca2+]i (176 +/- 19 to 338 +/- 35 nmol/L, P < .001) after thapsigargin exposure; this effect of caffeine was only observed when extracellular Ca2+ was present. A similar increase in [Ca2+]i was induced by caffeine after depletion of ryanodine- and histamine-sensitive Ca2+ stores or after pretreatment with the endoplasmic reticulum Ca(2+)-ATPase inhibitor cyclopiazonic acid (10 mumol/L). Thus, under baseline conditions the effect of caffeine on [Ca2+]i is similar to that of ryanodine and appears to be due to the release of an intracellular store. However, after depletion of an endoplasmic reticulum Ca2+ store, caffeine, but not ryanodine, stimulates Ca2+ influx, resulting in a large increase in [Ca2+]i.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Caffeine/pharmacology , Calcium Channels/drug effects , Calcium/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Aorta , Buffers , Calcium Channels/metabolism , Calcium-Transporting ATPases/antagonists & inhibitors , Calcium-Transporting ATPases/pharmacology , Cells, Cultured , Chelating Agents/pharmacology , Endothelium, Vascular/cytology , Enzyme Inhibitors/pharmacology , Fluorescent Dyes , Humans , Indoles/pharmacology , Ryanodine/pharmacology , Terpenes/pharmacology , Thapsigargin
20.
Circ Res ; 77(2): 354-60, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7614721

ABSTRACT

Guinea pig cardiac myocytes were loaded with the fluorescent dye indo 1, and cell contraction was measured by a video edge-detection system. Ca2+ was released from the sarcoplasmic reticulum (SR) by rapidly cooling the myocytes or by rapid application of 10 mmol/L caffeine. Estimates of the amount of Ca2+ released from the SR after different rest intervals (ie, under different loading conditions) were obtained by measuring the current evoked by rapid application of 10 mmol/L caffeine, which we call Na+/Ca2+ exchange current. This current is completely inhibited by removal of extracellular Na+ and Ca2+ or by application of 5 mmol/L Ni2+. SR Ca2+ release after rest intervals of 5 to 120 seconds (assuming cell volume to be 30 x 10(-12) L) was estimated to be 57.8 +/- 5.7 to 25.7 +/- 4.5 mumol/L accessible cell volume, respectively, equivalent to 23 to 10 mumol/kg wet wt, respectively. There was an exponential decline in Ca2+ release from the SR after rest intervals of 2 to 120 seconds (rate constant, 0.029 s-1; t1/2, 24 seconds); thereafter, there remained a portion (56%) of Ca2+ releasable to caffeine application. We found a similar exponential decay (rate constant, 0.020 s-1; t1/2, 35 seconds) of the size of rapid cooling contractures with increasing rest intervals. The time to peak of the Na+/Ca2+ exchange current in the presence of caffeine slowed at long rest intervals, ie, at smaller SR loads. A decrease in SR load of 50% increased the time to peak of the exchange current by 213 +/- 37% (n = 6).(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Calcium/metabolism , Heart Ventricles/cytology , Heart Ventricles/metabolism , Sarcoplasmic Reticulum/metabolism , Animals , Anti-Arrhythmia Agents/pharmacology , Caffeine/pharmacology , Calcium-Transporting ATPases/pharmacology , Cell Separation , Cold Temperature , Fluorescent Dyes , Guinea Pigs , Heart Ventricles/drug effects , In Vitro Techniques , Indoles , Plant Extracts/pharmacology , Plants, Medicinal , Rest , Sarcoplasmic Reticulum/drug effects , Sodium/metabolism , Terpenes/pharmacology , Thapsigargin
SELECTION OF CITATIONS
SEARCH DETAIL
...