Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Front Bioeng Biotechnol ; 10: 873531, 2022.
Article in English | MEDLINE | ID: mdl-35620470

ABSTRACT

Catecholaminergic polymorphic ventricular tachycardia (CPVT) is characterized by an arrhythmogenic mechanism involving disruption of calcium handling. This genetic disease can lead to sudden death in children and young adults during physical or emotional stress. Prior CPVT studies have focused on calcium handling, but mechanical functionality has rarely been investigated in vitro. In this research we combine stem cell-derived cardiomyocytes from a CPVT patient (RyR2-H2464D mutation) and a healthy familial control with an engineered culture platform to evaluate mechanical function of cardiomyocytes. Substrates with Young's modulus ranging from 10 to 50 kPa were used in conjunction with microcontact printing of ECM proteins into defined patterns for subsequent attachment. Digital Image Correlation (DIC) was used to evaluate collections of contracting cells. The amplitude of contractile strain was utilized as a quantitative indicator of functionality and disease severity. We found statistically significant differences: the maximum contractile strain was consistently higher in patient samples compared to control samples on all substrate stiffnesses. Additionally, the patient cell line had a statistically significantly slower intrinsic contraction rate than the control, which agrees with prior literature. Differences in mechanical strain have not been previously reported, and hypercontractility is not a known characteristic of CPVT. However, functional changes can occur as the disease progresses, thus this observation may not represent behavior observed in adolescent and adult patients. These results add to the limited studies of mechanical function of CPVT CMs reported in literature and identify functional differences that should be further explored.

2.
Toxicon ; 76: 328-42, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-23891887

ABSTRACT

The number and types of venom components that affect ion-channel function are reviewed. These are the most important venom components responsible for human intoxication, deserving medical attention, often requiring the use of specific anti-venoms. Special emphasis is given to peptides that recognize Na(+)-, K(+)- and Ca(++)-channels of excitable cells. Knowledge generated by direct isolation of peptides from venom and components deduced from cloned genes, whose amino acid sequences are deposited into databanks are nowadays in the order of 1.5 thousands, out of an estimate biodiversity closed to 300,000. Here the diversity of components is briefly reviewed with mention to specific references. Structural characteristic are discussed with examples taken from published work. The principal mechanisms of action of the three different types of peptides are also reviewed. Na(+)-channel specific venom components usually are modifier of the open and closing kinetic mechanisms of the ion-channels, whereas peptides affecting K(+)-channels are normally pore blocking agents. The Ryanodine Ca(++)-channel specific peptides are known for causing sub-conducting stages of the channels conductance and some were shown to be able to internalize penetrating inside the muscle cells.


Subject(s)
Ion Channels/chemistry , Scorpion Venoms/chemistry , Amino Acid Sequence , Models, Molecular , Scorpion Venoms/classification , Structure-Activity Relationship
3.
FEBS Lett ; 508(1): 5-10, 2001 Nov 09.
Article in English | MEDLINE | ID: mdl-11707258

ABSTRACT

The effect of imperatoxin A (IpTx(a)) on the ryanodine receptor type 3 (RyR3) was studied. IpTx(a) stimulates [(3)H]ryanodine binding to RyR3-containing microsomes, but this effect requires toxin concentrations higher than those required to stimulate RyR1 channels. The effect of IpTx(a) on RyR3 channels was observed at calcium concentrations in the range 0.1 microM to 10 mM. By contrast, RyR2 channels were not significantly affected by IpTx(a) in the same calcium ranges. Single channel current measurements indicated that IpTx(a) induced subconductance state in RyR3 channels that was similar to those observed with RyR1 and RyR2 channels. These results indicate that IpTx(a) is capable of inducing similar subconductance states in all three RyR isoforms, while stimulation of [(3)H]ryanodine binding by this toxin results in isoform-specific responses, with RyR1 being the most sensitive channel, RyR3 displaying an intermediate response and RyR2 the least responsive ones.


Subject(s)
Calcium/metabolism , Microsomes/drug effects , Ryanodine Receptor Calcium Release Channel/metabolism , Ryanodine/metabolism , Scorpion Venoms/pharmacology , Animals , Cattle , Cells, Cultured , Electrophysiology , Immunohistochemistry , Microsomes/chemistry , Muscle, Skeletal/metabolism , Protein Isoforms/metabolism , Ryanodine/chemistry , Ryanodine Receptor Calcium Release Channel/genetics , Scorpions/chemistry , Tritium/chemistry
5.
Am J Physiol Heart Circ Physiol ; 280(1): H208-15, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11123235

ABSTRACT

The present study was designed to test the hypothesis that cADP-ribose (cADPR) increases Ca(2+) release through activation of ryanodine receptors (RYR) on the sarcoplasmic reticulum (SR) in coronary arterial smooth muscle cells (CASMCs). We reconstituted RYR from the SR of CASMCs into planar lipid bilayers and examined the effect of cADPR on the activity of these Ca(2+) release channels. In a symmetrical cesium methanesulfonate configuration, a 245 pS Cs(+) current was recorded. This current was characterized by the formation of a subconductance and increase in the open probability (NP(o)) of the channels in the presence of ryanodine (0.01-1 microM) and imperatoxin A (100 nM). A high concentration of ryanodine (50 microM) and ruthenium red (40-80 microM) substantially inhibited the activity of RYR/Ca(2+) release channels. Caffeine (0.5-5 mM) markedly increased the NP(o) of these Ca(2+) release channels of the SR, but D-myo-inositol 1,4,5-trisphospate and heparin were without effect. Cyclic ADPR significantly increased the NP(o) of these Ca(2+) release channels of SR in a concentration-dependent manner. Addition of cADPR (0.01 microM) into the cis bath solution produced a 2.9-fold increase in the NP(o) of these RYR/Ca(2+) release channels. An eightfold increase in the NP(o) of the RYR/Ca(2+) release channels (0.0056 +/- 0.001 vs. 0.048 +/- 0.017) was observed at a concentration of cADPR of 1 microM. The effect of cADPR was completely abolished by ryanodine (50 microM). In the presence of cADPR, Ca(2+)-induced activation of these channels was markedly enhanced. These results provide evidence that cADPR activates RYR/Ca(2+) release channels on the SR of CASMCs. It is concluded that cADPR stimulates Ca(2+) release through the activation of RYRs on the SR of these smooth mucle cells.


Subject(s)
Adenosine Diphosphate Ribose/analogs & derivatives , Coronary Vessels/metabolism , Muscle, Smooth, Vascular/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Adenosine Diphosphate Ribose/pharmacology , Adenosine Diphosphate Ribose/physiology , Animals , Calcium/metabolism , Calcium Channels/drug effects , Calcium Channels/physiology , Cattle , Coronary Vessels/drug effects , Coronary Vessels/ultrastructure , Cyclic ADP-Ribose , In Vitro Techniques , Lipid Bilayers , Membranes , Microsomes/drug effects , Microsomes/metabolism , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/ultrastructure , Ruthenium Red/pharmacology , Ryanodine Receptor Calcium Release Channel/drug effects , Sarcoplasmic Reticulum/diagnostic imaging , Sarcoplasmic Reticulum/metabolism , Ultrasonography
6.
Biochem J ; 351(Pt 1): 57-65, 2000 Oct 01.
Article in English | MEDLINE | ID: mdl-10998347

ABSTRACT

A negatively charged region of the N-terminal portion of the skeletal ryanodine receptor (RyR), located between residues 1872-1923, is involved in Ca (2+)-dependent regulation of the Ca(2+)-release channel. This region is divergent between the skeletal (RyR1) and cardiac (RyR2) isoforms of the channel, and is known as D3. Ca(2+) exerts important regulatory functions on the RyR, being involved in both activation and inactivation functions of the channel, i.e. the effects occurring at micromolar and millimolar Ca(2+) concentrations respectively. To characterize the role of D3 in the Ca(2+)-dependent regulation of the Ca(2+)-release channel, we studied the functional consequences of deleting the D3 region from RyR1 (DeltaD3-RyR1) using a heterologous expression system, [(3)H]ryanodine binding assays and single-channel recordings in lipid bilayers. Deletion of the D3 region selectively affected Ca(2+)-dependent regulation of RyR1, but did not alter [(3)H]ryanodine binding or the effect of other modulators on the RyR. Compared with full-length RyR1 (wt-RyR1), the Ca(2+)-dependence curve of DeltaD3-RyR1 is broader, reflecting increased sensitivity to Ca(2+) activation and decreased sensitivity to Ca(2+) inactivation. In addition, DeltaD3-RyR1 was more resistant to inhibition by Mg(2+). Comparison of the effect of caffeine on wt-RyR1 and DeltaD3-RyR1 suggested that D3 is an important region of RyR that participates in Ca(2+)-dependent activation and inactivation of the Ca(2+)-release channel.


Subject(s)
Calcium/pharmacology , Muscle, Skeletal/metabolism , Ryanodine Receptor Calcium Release Channel/chemistry , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , CHO Cells , Caffeine/pharmacology , Calcium/metabolism , Calcium Channels/metabolism , Cricetinae , Ion Channel Gating/drug effects , Lipid Bilayers/metabolism , Magnesium/pharmacology , Membrane Potentials/drug effects , Protein Binding/drug effects , Protein Structure, Tertiary , Ryanodine/metabolism , Ryanodine Receptor Calcium Release Channel/genetics , Sequence Deletion/genetics
7.
Biophys J ; 79(2): 814-27, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10920014

ABSTRACT

We have investigated the effects of imperatoxin A (IpTx(a)) on local calcium release events in permeabilized frog skeletal muscle fibers, using laser scanning confocal microscopy in linescan mode. IpTx(a) induced the appearance of Ca(2+) release events from the sarcoplasmic reticulum that are approximately 2 s and have a smaller amplitude (31 +/- 2%) than the "Ca(2+) sparks" normally seen in the absence of toxin. The frequency of occurrence of long-duration imperatoxin-induced Ca(2+) release events increased in proportion to IpTx(a) concentrations ranging from 10 nM to 50 nM. The mean duration of imperatoxin-induced events in muscle fibers was independent of toxin concentration and agreed closely with the channel open time in experiments on isolated frog ryanodine receptors (RyRs) reconstituted in planar lipid bilayer, where IpTx(a) induced opening of single Ca(2+) release channels to prolonged subconductance states. These results suggest involvement of a single molecule of IpTx(a) in the activation of a single Ca(2+) release channel to produce a long-duration event. Assuming the ratio of full conductance to subconductance to be the same in the fibers as in bilayer, the amplitude of a spark relative to the long event indicates involvement of at most four RyR Ca(2+) release channels in the production of short-duration Ca(2+) sparks.


Subject(s)
Calcium/metabolism , Muscle, Skeletal/physiology , Sarcoplasmic Reticulum/physiology , Scorpion Venoms/pharmacology , Algorithms , Animals , Calcium Channels/drug effects , Calcium Channels/physiology , In Vitro Techniques , Kinetics , Lipid Bilayers , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/physiology , Rana pipiens , Sarcoplasmic Reticulum/drug effects , Software
8.
J Cell Biol ; 146(2): 493-9, 1999 Jul 26.
Article in English | MEDLINE | ID: mdl-10427100

ABSTRACT

Cryo-electron microscopy and three-dimensional, single-particle image analysis have been used to reveal the specific binding site of imperatoxin A (IpTx(a)) on the architecture of the calcium release channel/ryanodine receptor from skeletal muscle (RyR1). IpTx(a) is a peptide toxin that binds with high affinity to RyR1 and affects its functioning. The toxin was derivatized with biotin to enhance its detection with streptavidin. IpTx(a) binds to the cytoplasmic moiety of RyR1 between the clamp and handle domains, 11 nm away from the transmembrane pore. The proposed mimicry by IpTx(a) of the dihydropyridine receptor (DHPR) II-III loop, thought to be a main physiological excitation-contraction trigger, suggests that the IpTx(a) binding location is a potential excitation-contraction signal transduction site.


Subject(s)
Ryanodine Receptor Calcium Release Channel/metabolism , Ryanodine Receptor Calcium Release Channel/ultrastructure , Scorpion Venoms/metabolism , Allosteric Regulation , Animals , Binding Sites , Biotin , Calcium Channels/metabolism , Calcium Channels, L-Type , Cryoelectron Microscopy , Cytoplasm , Dose-Response Relationship, Drug , Ion Channel Gating , Models, Molecular , Molecular Mimicry , Muscle Contraction/physiology , Rabbits , Ryanodine/metabolism , Ryanodine Receptor Calcium Release Channel/chemistry , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism , Scorpion Venoms/pharmacology , Streptavidin
9.
FEBS Lett ; 450(3): 221-6, 1999 May 07.
Article in English | MEDLINE | ID: mdl-10359078

ABSTRACT

A 25 amino acid segment (Glu666-Pro691) of the II-III loop of the alpha1 subunit of the skeletal dihydropyridine receptor, but not the corresponding cardiac segment (Asp788-Pro814), activates skeletal ryanodine receptors. To identify the structural domains responsible for activation of skeletal ryanodine receptors, we systematically replaced amino acids of the cardiac II-III loop with their skeletal counterparts. A cluster of five basic residues of the skeletal II-III loop (681RKRRK685) was indispensable for activation of skeletal ryanodine receptors. In the cardiac segment, a negatively charged residue (Glu804) appears to diminish the electrostatic potential created by this basic cluster. In addition, Glu800 in the group of negatively charged residues 798EEEEE802 of the cardiac II-III loop may serve to prevent the binding of the activation domain.


Subject(s)
Calcium Channels/metabolism , Peptides/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Amino Acid Sequence , Animals , Calcium Channels/chemistry , Calcium Channels, L-Type , Models, Molecular , Molecular Sequence Data , Muscle, Skeletal/metabolism , Myocardium/metabolism , Peptides/chemistry , Protein Conformation , Rabbits , Ryanodine/metabolism , Swine , Tritium
10.
J Biol Chem ; 274(12): 7879-86, 1999 Mar 19.
Article in English | MEDLINE | ID: mdl-10075681

ABSTRACT

Excitation-contraction coupling in skeletal muscle is believed to be triggered by direct protein-protein interactions between the sarcolemmal dihydropyridine-sensitive Ca2+ channel and the Ca2+ release channel/ryanodine receptor (RyR) of sarcoplasmic reticulum. A 138-amino acid cytoplasmic loop between repeats II and III of the alpha1 subunit of the skeletal dihydropyridine receptor (the II-III loop) interacts with a region of the RyR to elicit Ca2+ release. In addition, small segments (10-20 amino acid residues) of the II-III loop retain the capacity to activate Ca2+ release. Imperatoxin A, a 33-amino acid peptide from the scorpion Pandinus imperator, binds directly to the RyR and displays structural and functional homology with an activating segment of the II-III loop (Glu666-Leu690). Mutations in a structural motif composed of a cluster of basic amino acids followed by Ser or Thr dramatically reduce or completely abolish the capacity of the peptides to activate RyRs. Thus, the Imperatoxin A-RyR interaction mimics critical molecular characteristics of the II-III loop-RyR interaction and may be a useful tool to elucidate the molecular mechanism that couples membrane depolarization to sarcoplasmic reticulum Ca2+ release in vivo.


Subject(s)
Calcium Channels/chemistry , Peptide Fragments/pharmacology , Ryanodine Receptor Calcium Release Channel/metabolism , Scorpion Venoms/pharmacology , Amino Acid Sequence , Animals , CHO Cells , Calcium/metabolism , Calcium Channels/pharmacology , Calcium Channels, L-Type , Chromatography, High Pressure Liquid , Cricetinae , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Binding , Scorpion Venoms/chemistry , Scorpion Venoms/genetics , Scorpions , Sequence Homology, Amino Acid , Structure-Activity Relationship
11.
J Mol Cell Cardiol ; 30(5): 913-21, 1998 May.
Article in English | MEDLINE | ID: mdl-9618232

ABSTRACT

It has been suggested that adenosine cardioprotection occurs via adenosine A1 receptor-mediated activation of protein kinase C (PKC). However, adenosine has well-known vasodilatory effects in the myocardium, whereas PKC is a vasoconstrictor. This study examined whether adenosine A1 receptor activation alters the effects of the PKC activator. 1,2-dioctanoyl-s,n-glycerol (DOG) in isolated perfused rat hearts (left-ventricular developed pressure) and rat ventricular myocytes ([Ca2+]i and cell shortening). Exposure to DOG decreased left-ventricular developed pressure by 30%, an effect that was completely reversible. Pretreatment of isolated hearts with either the PKC inhibitor chelerythrine or the adenosine A1 agonist 2-chloro-N6-cyclo-cyclo-isolated pentlyadenosine (CCPA) attenuated the negative inotropic effects of DOG. In the isolated myocytes, DOG decreased [Ca2+]i and cell shortening by 25 and 28%, respectively, effects that were attenuated by both chelerythrine and CCPA. The CCPA attenuation of the DOG-induced decrease in [Ca2+]i and cell shortening was blocked by pretreating the myocytes with the adenosine A1 antagonist, 8-cyclopentyl-1,3-dipropylxanthine (DPCPX). These results indicate that in rat ventricular myocardium, adenosine A1 receptor activation attenuates the apparent PKC-dependent negative inotropic effects of DOG via preservation of [Ca2+]i levels.


Subject(s)
Diglycerides/pharmacology , Myocardial Contraction/drug effects , Purinergic P1 Receptor Agonists , Adenosine/analogs & derivatives , Adenosine/pharmacology , Analysis of Variance , Animals , Depression, Chemical , Diglycerides/antagonists & inhibitors , Enzyme Activation , Heart Ventricles/cytology , Heart Ventricles/drug effects , In Vitro Techniques , Male , Rats , Rats, Sprague-Dawley , Stimulation, Chemical , Xanthines/pharmacology
12.
J Gen Physiol ; 111(5): 679-90, 1998 May.
Article in English | MEDLINE | ID: mdl-9565405

ABSTRACT

Single-channel and [3H]ryanodine binding experiments were carried out to examine the effects of imperatoxin activator (IpTxa), a 33 amino acid peptide isolated from the venom of the African scorpion Pandinus imperator, on rabbit skeletal and canine cardiac muscle Ca2+ release channels (CRCs). Single channel currents from purified CRCs incorporated into planar lipid bilayers were recorded in 250 mM KCl media. Addition of IpTxa in nanomolar concentration to the cytosolic (cis) side, but not to the lumenal (trans) side, induced substates in both ryanodine receptor isoforms. The substates displayed a slightly rectifying current-voltage relationship. The chord conductance at -40 mV was approximately 43% of the full conductance, whereas it was approximately 28% at a holding potential of +40 mV. The substate formation by IpTxa was voltage and concentration dependent. Analysis of voltage and concentration dependence and kinetics of substate formation suggested that IpTxa reversibly binds to the CRC at a single site in the voltage drop across the channel. The rate constant for IpTxa binding to the skeletal muscle CRC increased e-fold per +53 mV and the rate constant of dissociation decreased e-fold per +25 mV applied holding potential. The effective valence of the reaction leading to the substate was approximately 1.5. The IpTxa binding site was calculated to be located at approximately 23% of the voltage drop from the cytosolic side. IpTxa induced substates in the ryanodine-modified skeletal CRC and increased or reduced [3H]ryanodine binding to sarcoplasmic reticulum vesicles depending on the level of channel activation. These results suggest that IpTxa induces subconductance states in skeletal and cardiac muscle Ca2+ release channels by binding to a single, cytosolically accessible site different from the ryanodine binding site.


Subject(s)
Muscle, Skeletal/metabolism , Myocardium/metabolism , Ryanodine Receptor Calcium Release Channel/drug effects , Ryanodine Receptor Calcium Release Channel/metabolism , Scorpion Venoms/pharmacology , Animals , Dogs , Electric Conductivity , Electrophysiology , Kinetics , Models, Biological , Osmolar Concentration , Rabbits , Ryanodine/metabolism , Sarcoplasmic Reticulum/metabolism
13.
J Physiol ; 510 ( Pt 1): 155-64, 1998 Jul 01.
Article in English | MEDLINE | ID: mdl-9625874

ABSTRACT

1. Immunoblot analysis, [3H]ryanodine binding, and planar lipid bilayer techniques were used to identify and characterize the functional properties of ryanodine receptors (RyRs) from Lytechinus pictus and Strongylocentrotus purpuratus sea urchin eggs. 2. An antibody against mammalian skeletal RyRs identified an approximately 400 kDa band in the cortical microsomes of sea urchin eggs while a cardiac-specific RyR antibody failed to recognize this protein. [3H]Ryanodine binding to cortical microsomes revealed the presence of a high-affinity (Kd = 13 nM), saturable (maximal density of receptor sites, Bmax = 1.56 pmol (mg protein)-1) binding site that exhibited a biphasic response to Ca2+. 3. Upon reconstitution of cortical microsomes into lipid bilayers, only sparse and unstable openings of a high-conductance cation channel were detected. Addition of crude sea urchin egg homogenate to the cytosolic (cis side) of the channel increased the frequency of openings and stabilized channel activity. The homogenate-activated channels were Ca2+ sensitive, selective for Ca2+ over Cs+, and driven by ryanodine into a long-lived subconductance state that represented approximately 40 % of the full conductance level. Homogenate dialysed in membranes with a molecular weight cut-off <= 2000 lacked the capacity to increase the frequency of RyR openings and to stabilize channel activity. 4. Direct application of cyclic adenosine diphosphoribose (cADPR) or photolysis of NPE-cADPR ('caged' cADPR) by ultraviolet laser pulses produced transient activation of sea urchin egg RyRs. Calmodulin (CaM) failed to activate reconstituted RyRs; however, channel activity was inhibited by the CaM blocker trifluoroperazine, suggesting that CaM was necessary but not sufficient to sustain RyR activity. 5. These findings suggest that a functional Ca2+ release unit in sea urchin eggs is a complex of several molecules, one of which corresponds to a protein functionally similar to mammalian RyRs.


Subject(s)
Ovum/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Sea Urchins/metabolism , ADP-ribosyl Cyclase , Animals , Calmodulin/antagonists & inhibitors , Electric Conductivity , Immunoblotting , Microsomes/metabolism , Phosphorus-Oxygen Lyases/pharmacology , Ryanodine/metabolism , Ryanodine Receptor Calcium Release Channel/drug effects , Ryanodine Receptor Calcium Release Channel/physiology , Trifluoperazine/pharmacology
14.
Am J Physiol ; 274(1): H8-17, 1998 01.
Article in English | MEDLINE | ID: mdl-9458846

ABSTRACT

Pyruvate has been shown to be a metabolic inotrope in the myocardium. In millimolar concentrations, it has been shown to increase both myocardial phosphorylation potential and the cytosolic [NAD+]-to-[NADH] ratio. To determine if changes in these parameters can alter intracellular Ca2+ concentration ([Ca2+]i) and hence contractile function, Ca2+ transients and cell shortening (CS) were measured in isolated rat ventricular myocytes superfused with a physiological N-2-hydroxyethylpiperazine-N'-2-ethanesulfonic acid buffer (11 mmol/l glucose) with and without additional pyruvate, L-lactate, acetate, or isoproterenol. The addition of 5 mmol/l pyruvate resulted in a 33% increase in CS and a 39% increase in systolic [Ca2+]i. These pyruvate effects were 70% of those observed with 100 nmol/l isoproterenol. The mitochondrial monocarboxylate transport inhibitor alpha-cyano-4-hydroxycinnamate (250 mumol/l) strongly inhibited pyruvate inotropy, suggesting a substantial obligatory coupling between pyruvate inotropism and its oxidation by the mitochondria. A possible role of the cytosolic [NAD+]-to-[NADH] ratio was assessed by comparing the effects of 20 mmol/l L-lactate to those of equimolar pyruvate. In contrast to 20 mmol/l pyruvate, excess L-lactate failed to appreciably increase CS or systolic [Ca2+]i. The findings imply that, at levels substantially above 5 mmol/l, a portion of pyruvate inotropism might be due to extreme cytosolic [NAD+]-to-[NADH] ratios. This study is the first evidence that augmented [Ca2+]i transients are most likely the mechanism of cardiac pyruvate inotropism.


Subject(s)
Heart/physiology , Myocardial Contraction/drug effects , Myocardium/metabolism , Pyruvic Acid/pharmacology , Animals , Calcium/metabolism , Cells, Cultured , Cytosol/metabolism , Glucose/metabolism , Glucose/pharmacology , Heart Ventricles , Lactic Acid/pharmacology , Male , Mitochondria, Heart/metabolism , Models, Cardiovascular , Myocardium/cytology , NAD/metabolism , NADP/metabolism , Phosphorylation , Rats , Rats, Wistar
16.
Trends Cardiovasc Med ; 8(3): 111-8, 1998 Apr.
Article in English | MEDLINE | ID: mdl-21235920

ABSTRACT

Toxins from scorpion venom are emerging as useful ligands for structure/function studies of ryanodine receptors (RyR), the sarcoplasmic reticulum Ca(2+) release channels that elevate intracellular Ca(2+) to elicit contraction of cardiac and skeletal muscle. Imperatoxin A (IpTx(a)), a 3.7 kDa peptide from the African scorpion P. imperator, is an agonist of RyRs which, similar to the alkaloid ryanodine, binds with high affinity to the RyR protein and induces the appearance of a long-lived subconductance state. Imperatoxin I (IpTx(i)), a 15 kDa heterodimeric protein from the same venom that displays phospholipase A(2) activity, inhibits RyRs without a physical interaction with the channel protein, by releasing free fatty acids into the incubation medium. IpTx(a) and IpTx(i) are the first of a group of peptide probes of RyRs with diverse mechanism of action which overcome some of the undesirable characteristics of ryanodine.

18.
J Biol Chem ; 272(40): 25333-8, 1997 Oct 03.
Article in English | MEDLINE | ID: mdl-9312152

ABSTRACT

Sorcin is a widely expressed, 22-kDa Ca2+-binding protein initially identified in multidrug-resistant cells. In the heart, sorcin localizes to the dyadic junctions of transverse tubules and sarcoplasmic reticulum and coimmunoprecipitates with the Ca2+ release channel/ryanodine receptor (RyR) (Meyers, M. B., Pickel, V. M., Sheu, S.-S., Sharma, V. K., Scotto, K. W., and Fishman, G. I. (1995) J. Biol. Chem. 270, 26411-26418). We have investigated a possible functional interaction between sorcin and cardiac RyR using purified recombinant sorcin in [3H]ryanodine binding experiments and single channel recordings of RyR. The open probability of single RyR was decreased significantly by the addition of sorcin to the cytoplasmic side of the channel (IC50 approximately 480 nM). In addition, sorcin completely inhibited [3H]ryanodine binding with an IC50 approximately 700 nM. Inhibition occurred over a wide range of [Ca2+], and sorcin-modulated RyR remained Ca2+-dependent. Furthermore, caffeine-activated RyRs were also inhibited by sorcin at low [Ca2+] (pCa 7), suggesting that Ca2+ is not an obligatory factor for sorcin inhibition of RyR. Comparisons of these inhibitory effects with those of calmodulin and calpain, proteins structurally related to sorcin, suggested that the interaction of sorcin with cardiac RyR was distinct from and independent of either of these modulatory proteins. Phosphorylation of sorcin with the catalytic subunit of protein kinase A significantly decreased the ability of sorcin to modulate RyR. These results suggest that sorcin may modulate RyR function in a normal cell environment and that the level of modulation is in turn influenced by signaling pathways that increase protein kinase A activity.


Subject(s)
Calcium Channels/physiology , Calcium-Binding Proteins/pharmacology , Heart/physiology , Microsomes/physiology , Muscle Proteins/physiology , Muscle, Skeletal/physiology , Animals , Caffeine/pharmacology , Calcium/pharmacology , Calcium Channels/drug effects , Calcium-Binding Proteins/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Ion Channel Gating/drug effects , Kinetics , Membrane Potentials/drug effects , Microsomes/drug effects , Muscle Proteins/drug effects , Phosphoproteins/metabolism , Phosphorylation , Probability , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Ryanodine/metabolism , Ryanodine Receptor Calcium Release Channel , Sarcoplasmic Reticulum/physiology , Swine
19.
Am J Physiol ; 273(2 Pt 2): H796-804, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9277497

ABSTRACT

We investigated the effects of myocardial stunning on the function of the two main Ca2+ transport proteins of the sarcoplasmic reticulum (SR), the Ca(2+)-adenosinetriphosphatase and the Ca(2+)-release channel or ryanodine receptor. Regional myocardial stunning was induced in open-chest pigs (n = 6) by a 10-min occlusion of the left anterior descending coronary artery (LAD) and 2 h reperfusion. SR vesicles isolated from the LAD-perfused region (stunned) and the normal left circumflex coronary artery (LC)-perfused region were used to assess the oxalate-supported 45Ca2+ uptake, [3H]ryanodine binding, and single-channel recordings of ryanodine-sensitive Ca(2+)-release channels in planar lipid bilayers. Myocardial stunning decreased LAD systolic wall thickening to 20% of preischemic values. The rate of SR 45Ca2+ uptake in the stunned LAD bed was reduced by 37% compared with that of the normal LC bed (P < 0.05). Stunning was also associated with a 38% reduction in the maximal density of high-affinity [3H]ryanodine binding sites (P < 0.05 vs. normal LC) but had no effect on the dissociation constant. The open probability of ryanodine-sensitive Ca(2+)-release channels determined by single channel recordings in planar lipid bilayers was 26 +/- 2% for control SR (n = 33 channels from 3 animals) and 14 +/- 2% for stunned SR (n = 21 channels; P < 0.05). This depressed activity of SR function observed in postischemic myocardium could be one of the mechanisms underlying myocardial stunning.


Subject(s)
Calcium Channels/physiology , Muscle Proteins/physiology , Myocardial Stunning/metabolism , Myocardium/metabolism , Animals , Calcium/metabolism , Calcium Channels/metabolism , Female , Hemodynamics , Male , Ryanodine Receptor Calcium Release Channel , Sarcoplasmic Reticulum/metabolism , Swine , Ventricular Function
20.
Science ; 276(5313): 800-6, 1997 May 02.
Article in English | MEDLINE | ID: mdl-9115206

ABSTRACT

Cardiac hypertrophy and heart failure caused by high blood pressure were studied in single myocytes taken from hypertensive rats (Dahl SS/Jr) and SH-HF rats in heart failure. Confocal microscopy and patch-clamp methods were used to examine excitation-contraction (EC) coupling, and the relation between the plasma membrane calcium current (ICa) and evoked calcium release from the sarcoplasmic reticulum (SR), which was visualized as "calcium sparks." The ability of ICa to trigger calcium release from the SR in both hypertrophied and failing hearts was reduced. Because ICa density and SR calcium-release channels were normal, the defect appears to reside in a change in the relation between SR calcium-release channels and sarcolemmal calcium channels. beta-Adrenergic stimulation largely overcame the defect in hypertrophic but not failing heart cells. Thus, the same defect in EC coupling that develops during hypertrophy may contribute to heart failure when compensatory mechanisms fail.


Subject(s)
Calcium/metabolism , Cardiomegaly/physiopathology , Heart Failure/physiopathology , Myocardial Contraction/physiology , Myocardium/metabolism , Adrenergic beta-Agonists/pharmacology , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels/metabolism , Calcium Channels, L-Type , Cardiomegaly/etiology , Cell Membrane/metabolism , Heart Failure/etiology , Hypertension/complications , Isoproterenol/pharmacology , Microscopy, Confocal , Muscle Proteins/metabolism , Myocardial Contraction/drug effects , Nifedipine/pharmacology , Patch-Clamp Techniques , Rats , Rats, Inbred Strains , Ryanodine Receptor Calcium Release Channel , Sarcoplasmic Reticulum/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...