Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
J Cell Sci ; 126(Pt 21): 5042-51, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-23943880

ABSTRACT

The ryanodine receptor (RyR) is an ion channel composed of four identical subunits mediating calcium efflux from the endo/sarcoplasmic reticulum of excitable and non-excitable cells. We present several lines of evidence indicating that the RyR2 N-terminus is capable of self-association. A combination of yeast two-hybrid screens, co-immunoprecipitation analysis, chemical crosslinking and gel filtration assays collectively demonstrate that a RyR2 N-terminal fragment possesses the intrinsic ability to oligomerize, enabling apparent tetramer formation. Interestingly, N-terminus tetramerization mediated by endogenous disulfide bond formation occurs in native RyR2, but notably not in RyR1. Disruption of N-terminal inter-subunit interactions within RyR2 results in dysregulation of channel activation at diastolic Ca(2+) concentrations from ryanodine binding and single channel measurements. Our findings suggest that the N-terminus interactions mediating tetramer assembly are involved in RyR channel closure, identifying a crucial role for this structural association in the dynamic regulation of intracellular Ca(2+) release.


Subject(s)
Myocytes, Cardiac/metabolism , Ryanodine Receptor Calcium Release Channel/chemistry , Ryanodine Receptor Calcium Release Channel/metabolism , Amino Acid Motifs , Animals , Calcium/metabolism , Humans , Myocytes, Cardiac/chemistry , Protein Multimerization , Rabbits , Ryanodine Receptor Calcium Release Channel/genetics , Sarcoplasmic Reticulum/metabolism , Swine
2.
Mol Pharmacol ; 85(4): 564-75, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24423447

ABSTRACT

Eudistomin D (EuD) and penaresin (Pen) derivatives are bioactive alkaloids from marine sponges found to induce Ca(2+) release from striated muscle sarcoplasmic reticulum (SR). Although these alkaloids are believed to affect ryanodine receptor (RyR) gating in a "caffeine-like" manner, no single-channel study confirmed this assumption. Here, EuD and MBED (9-methyl-7-bromoeudistomin D) were contrasted against caffeine on their ability to modulate the SR Ca(2+) loading/leak from cardiac and skeletal muscle SR microsomes as well as the function of RyRs in planar bilayers. The effects of these alkaloids on [(3)H]ryanodine binding and SR Ca(2+) ATPase (SERCA) activity were also tested. MBED (1-5 µM) fully mimicked maximal activating effects of caffeine (20 mM) on SR Ca(2+) leak. At the single-channel level, MBED mimicked the agonistic action of caffeine on cardiac RyR gating (i.e., stabilized long openings characteristic of "high-open-probability" mode). EuD was a partial agonist at the maximal doses tested. The tested Pen derivatives displayed mild to no agonism on RyRs, SR Ca(2+) leak, or [(3)H]ryanodine binding studies. Unlike caffeine, EuD and some Pen derivatives significantly inhibited SERCA at concentrations required to modulate RyRs. Instead, MBED's affinity for RyRs (EC50 ∼ 0.5 µM) was much larger than for SERCA (IC50 > 285 µM). In conclusion, MBED is a potent RyR agonist and, potentially, a better choice than caffeine for microsomal and cell studies due to its reported lack of effects on adenosine receptors and phosphodiesterases. As a high-affinity caffeine-like probe, MBED could also help identify the caffeine-binding site in RyRs.


Subject(s)
Calcium-Transporting ATPases/metabolism , Carbolines/pharmacology , Indole Alkaloids/pharmacology , Muscle, Skeletal/drug effects , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/metabolism , Animals , Caffeine/pharmacology , Calcium/metabolism , Calcium Channel Agonists/chemistry , Calcium Channel Agonists/pharmacology , Calcium Channel Blockers/chemistry , Calcium Channel Blockers/pharmacology , Carbolines/chemistry , In Vitro Techniques , Indole Alkaloids/chemistry , Lipid Bilayers/chemistry , Microsomes/drug effects , Microsomes/metabolism , Muscle, Skeletal/metabolism , Myocardium/metabolism , Protein Binding , Rabbits , Ryanodine Receptor Calcium Release Channel/chemistry
3.
Am J Physiol Cell Physiol ; 303(6): C682-97, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22785120

ABSTRACT

Coupled gating (synchronous openings and closures) of groups of skeletal muscle ryanodine receptors (RyR1), which mimics RyR1-mediated Ca(2+) release underlying Ca(2+) sparks, was first described by Marx et al. (Marx SO, Ondrias K, Marks AR. Science 281: 818-821, 1998). The nature of the RyR1-RyR1 interactions for coupled gating still needs to be characterized. Consequently, we defined planar lipid bilayer conditions where ∼25% of multichannel reconstitutions contain mixtures of coupled and independently gating RyR1. In ∼10% of the cases, all RyRs (2-10 channels; most frequently 3-4) gated in coupled fashion, allowing for quantification. Our results indicated that coupling required cytosolic solutions containing ATP/Mg(2+) and high (50 mM) luminal Ca(2+) (Ca(lum)) or Sr(2+) solutions. Bursts of coupled activity (events) started and ended abruptly, with all channels activating/deactivating within ∼300 µs. Coupled RyR1 were heterogeneous, where highly active RyR1 ("drivers") seemed open during the entire coupled event (P(o) = 1), while other RyR1s ("followers") displayed abundant flickering and smaller amplitude. Drivers mean open time increased with cytosolic Ca(2+) (Ca(cyt)) or caffeine, whereas followers flicker frequency was Ca(cyt) independent and more sensitive to inhibition by cytosolic Mg(2+). Coupled events were insensitive to varying lumen-to-cytosol Ca(2+) fluxes from ∼1 to 8 pA, which does not corroborate coupling of neighboring RyR1 by local Ca(2+)-induced Ca(2+) release. However, coupling requires specific Ca(lum) sites, as it was lost when Ca(lum) was replaced by luminal Ba(2+) or Mg(2+). In summary, coupled events reveal complex interactions among heterogeneous RyR1, differentially modulated by cytosolic ATP/Mg(2+), Ca(cyt), and Ca(lum,) which under cell-like ionic conditions may parallel synchronous RyR1 gating during Ca(2+) sparks.


Subject(s)
Adenosine Triphosphate/physiology , Calcium/physiology , Ion Channel Gating/physiology , Magnesium/physiology , Muscle, Skeletal/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Adenosine Triphosphate/chemistry , Animals , Calcium/chemistry , Magnesium/chemistry , Muscle, Skeletal/physiology , Rabbits , Ryanodine Receptor Calcium Release Channel/physiology
4.
Mol Pharmacol ; 79(1): 141-7, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20923851

ABSTRACT

7-Chloro-5-(2-chlorophenyl)-1,5-dihydro-4,1-benzothiazepin-2(3H)-one [CGP-37157 (CGP)], a benzothiazepine derivative of clonazepam, is commonly used as a blocker of the mitochondrial Na+/Ca²+ exchanger. However, evidence suggests that CGP could also affect other targets, such as L-type Ca²+ channels and plasmalemma Na+/Ca²+ exchanger. Here, we tested the possibility of a direct modulation of ryanodine receptor channels (RyRs) and/or sarco/endoplasmic reticulum Ca²+-stimulated ATPase (SERCA) by CGP. In the presence of ruthenium red (inhibitor of RyRs), CGP decreased SERCA-mediated Ca²+ uptake of cardiac and skeletal sarcoplasmic reticulum (SR) microsomes (IC50 values of 6.6 and 9.9 µM, respectively). The CGP effects on SERCA activity correlated with a decreased V(max) of ATPase activity of SERCA-enriched skeletal SR fractions. CGP (≥ 5 µM) also increased RyR-mediated Ca²+ leak from skeletal SR microsomes. Planar bilayer studies confirmed that both cardiac and skeletal RyRs are directly activated by CGP (EC(50) values of 9.4 and 12.0 µM, respectively). In summary, we found that CGP inhibits SERCA and activates RyR channels. Hence, the action of CGP on cellular Ca²+ homeostasis reported in the literature of cardiac, skeletal muscle, and other nonmuscle systems requires further analysis to take into account the contribution of all CGP-sensitive Ca²+ transporters.


Subject(s)
Calcium-Transporting ATPases/antagonists & inhibitors , Calcium-Transporting ATPases/metabolism , Clonazepam/analogs & derivatives , Muscle, Striated/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/metabolism , Thiazepines/pharmacology , Animals , Calcium/metabolism , Clonazepam/pharmacology , Muscle, Striated/drug effects , Rabbits , Sarcoplasmic Reticulum/drug effects
5.
J Biol Chem ; 285(18): 13542-9, 2010 Apr 30.
Article in English | MEDLINE | ID: mdl-20177054

ABSTRACT

In pulmonary arterial smooth muscle, Ca(2+) release from the sarcoplasmic reticulum (SR) via ryanodine receptors (RyRs) may induce constriction and dilation in a manner that is not mutually exclusive. We show here that the targeting of different sarcoplasmic/endoplasmic reticulum Ca(2+)-ATPases (SERCA) and RyR subtypes to discrete SR regions explains this paradox. Western blots identified protein bands for SERCA2a and SERCA2b, whereas immunofluorescence labeling of isolated pulmonary arterial smooth muscle cells revealed striking differences in the spatial distribution of SERCA2a and SERCA2b and RyR1, RyR2, and RyR3, respectively. Almost all SERCA2a and RyR3 labeling was restricted to a region within 1.5 microm of the nucleus. In marked contrast, SERCA2b labeling was primarily found within 1.5 microm of the plasma membrane, where labeling for RyR1 was maximal. The majority of labeling for RyR2 lay in between these two regions of the cell. Application of the vasoconstrictor endothelin-1 induced global Ca(2+) waves in pulmonary arterial smooth muscle cells, which were markedly attenuated upon depletion of SR Ca(2+) stores by preincubation of cells with the SERCA inhibitor thapsigargin but remained unaffected after preincubation of cells with a second SERCA antagonist, cyclopiazonic acid. We conclude that functionally segregated SR Ca(2+) stores exist within pulmonary arterial smooth muscle cells. One sits proximal to the plasma membrane, receives Ca(2+) via SERCA2b, and likely releases Ca(2+) via RyR1 to mediate vasodilation. The other is located centrally, receives Ca(2+) via SERCA2a, and likely releases Ca(2+) via RyR3 and RyR2 to initiate vasoconstriction.


Subject(s)
Calcium/metabolism , Muscle, Smooth, Vascular/metabolism , Pulmonary Artery/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sarcoplasmic Reticulum/metabolism , Animals , Cell Membrane/metabolism , Endothelin-1/pharmacology , Male , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/cytology , Rats , Rats, Wistar , Vasoconstriction/drug effects , Vasoconstriction/physiology , Vasoconstrictor Agents/pharmacology , Vasodilation/drug effects , Vasodilation/physiology
6.
Am J Physiol Cell Physiol ; 299(2): C264-78, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20445169

ABSTRACT

Ryanodine receptors (RyRs) regulate contractility in resistance-size cerebral artery smooth muscle, yet their molecular identity, subcellular location, and phenotype in this tissue remain unknown. Following rat resistance-size cerebral artery myocyte sarcoplasmic reticulum (SR) purification and incorporation into POPE-POPS-POPC (5:3:2; wt/wt) bilayers, unitary conductances of 110 +/- 8, 334 +/- 15, and 441 +/- 27 pS in symmetric 300 mM Cs(+) were usually detected. The most frequent (34/40 bilayers) conductance (334 pS) decreased to

Subject(s)
Cerebral Arteries/cytology , Cerebral Arteries/physiology , Muscle Cells/physiology , Protein Subunits/physiology , Ryanodine Receptor Calcium Release Channel/physiology , Animals , Cells, Cultured , Cerebral Arteries/chemistry , Female , Male , Muscle Cells/chemistry , Protein Subunits/analysis , Rats , Rats, Sprague-Dawley , Ryanodine Receptor Calcium Release Channel/analysis
7.
Cell Calcium ; 44(2): 190-201, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18191199

ABSTRACT

In arterial myocytes the Ca(2+) mobilizing messenger NAADP evokes spatially restricted Ca(2+) bursts from a lysosome-related store that are subsequently amplified into global Ca(2+) waves by Ca(2+)-induced Ca(2+)-release from the sarcoplasmic reticulum (SR) via ryanodine receptors (RyRs). Lysosomes facilitate this process by forming clusters that co-localize with a subpopulation of RyRs on the SR. We determine here whether RyR subtypes 1, 2 or 3 selectively co-localize with lysosomal clusters in pulmonary arterial myocytes using affinity purified specific antibodies. The density of: (1) alphalgP120 labelling, a lysosome-specific protein, in the perinuclear region of the cell (within 1.5mum of the nucleus) was approximately 4-fold greater than in the sub-plasmalemmal (within 1.5mum of the plasma membrane) and approximately 2-fold greater than in the extra-perinuclear (remainder) regions; (2) RyR3 labelling within the perinuclear region was approximately 4- and approximately 14-fold greater than that in the extra-perinuclear and sub-plasmalemmal regions, and approximately 2-fold greater than that for either RyR1 or RyR2; (3) despite there being no difference in the overall densities of fluorescent labelling of lysosomes and RyR subtypes between cells, co-localization with alphalgp120 labelling within the perinuclear region was approximately 2-fold greater for RyR3 than for RyR2 or RyR1; (4) co-localization between alphalgp120 and each RyR subtype declined markedly outside the perinuclear region. Furthermore, selective block of RyR3 and RyR1 with dantrolene (30muM) abolished global Ca(2+) waves but not Ca(2+) bursts in response to intracellular dialysis of NAADP (10nM). We conclude that a subpopulation of lysosomes cluster in the perinuclear region of the cell and form junctions with SR containing a high density of RyR3 to comprise a trigger zone for Ca(2+) signalling by NAADP.


Subject(s)
Calcium Signaling , Calcium/metabolism , Lung/metabolism , Lysosomes/metabolism , Muscle, Smooth, Vascular/metabolism , NADP/analogs & derivatives , Pulmonary Artery/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Cells, Cultured , Fluorescence , Heart/physiology , Lung/cytology , Male , Muscle, Smooth, Vascular/cytology , NADP/metabolism , Protein Isoforms , Pulmonary Artery/cytology , Rats , Rats, Wistar , Ryanodine/pharmacology , Sarcoplasmic Reticulum/metabolism
8.
Biochem Biophys Res Commun ; 369(1): 195-207, 2008 Apr 25.
Article in English | MEDLINE | ID: mdl-18182155

ABSTRACT

The intracellular Ca(2+) release channels are indispensable molecular machinery in practically all eukaryotic cells of multicellular animals. They serve a key role in cell signaling by way of Ca(2+) as a second messenger. In response to a signaling event, the channels release Ca(2+) from intracellular stores. The resulting rise in cytoplasmic Ca(2+) concentration triggers the cell to carry out its specialized role, after which the intracellular Ca(2+) concentration must be reduced so that the signaling event can again be repeated. There are two types of intracellular Ca(2+) release channels, i.e., the ryanodine receptors and the inositol triphosphate receptors. My focus in this minireview is to present a personal account, from the vantage point our laboratory, of the discovery, isolation, and characterization of the ryanodine receptors from mammalian muscle. There are three isoforms: ryanodine receptor 1 (RyR1), first isolated from rabbit fast twitch skeletal muscle; ryanodine receptor 2 (RyR2), first isolated from dog heart; and ryanodine receptor 3 (RyR3), first isolated from bovine diaphragm muscle. The ryanodine receptors are the largest channel structures known. The RyR isoforms are very similar albeit with important differences. Natural mutations in humans in these receptors have already been associated with a number of muscle diseases.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Ion Channel Gating/physiology , Muscle Contraction/physiology , Muscle, Skeletal/physiology , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/physiology , Animals , Humans
9.
Transplantation ; 73(11): 1835-8, 2002 Jun 15.
Article in English | MEDLINE | ID: mdl-12085010

ABSTRACT

BACKGROUND: FK506-binding proteins (FKBP) are immunophilins that interact with the immunosuppressive drugs FK506 and rapamycin. Several FKBP family members such as FKBP12, FKBP12.6, and FKBP51 are expressed in T cells. It has been speculated that these FKBPs are possibly redundant in the immunosuppressant-induced T-cell inactivation. To determine the pharmacological relevance of multiple FKBP members in the immunosuppressant-induced T-cell inactivation, we have investigated the physiological responses of FKBP12-deficient and FKBP12.6-deficient mutant T cells to the immunosuppressive agent FK506. METHODS: FKBP12-deficient and FKBP12.6-deficient T cells were isolated from genetically engineered FKBP12-deficient and FKBP12.6-deficient mice, respectively. T-cell growth inhibitory assay was used to assess their responses to immunosuppressant FK506 treatments. RESULTS: We found that growth inhibition induced by FK506 is abolished in FKBP12-deficient cells but not in FKBP12.6-deficient cells. CONCLUSIONS: FKBP12 is the only FKBP family member that plays a key role in immunosuppressant-mediated immunosuppression.


Subject(s)
Immunosuppressive Agents/pharmacology , T-Lymphocytes/immunology , Tacrolimus Binding Protein 1A/genetics , Tacrolimus Binding Protein 1A/immunology , Tacrolimus/pharmacology , Animals , CD28 Antigens/immunology , CD3 Complex/immunology , Cell Division/drug effects , Cell Division/immunology , Cyclosporine/pharmacology , Dose-Response Relationship, Drug , Mice , Mice, Knockout , T-Lymphocytes/drug effects , Tacrolimus Binding Protein 1A/metabolism
10.
J Cell Sci ; 120(Pt 21): 3784-91, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17925380

ABSTRACT

Ryanodine receptor subtype 1 (RyR1) has been primarily characterized in skeletal muscle but several studies have revealed its expression in smooth muscle. Here, we used Ryr1-null mice to investigate the role of this isoform in Ca(2+) signaling in urinary bladder smooth muscle. We show that RyR1 is required for depolarization-induced Ca(2+) sparks, whereas RyR2 and RyR3 are sufficient for spontaneous or caffeine-induced Ca(2+) sparks. Immunostaining revealed specific subcellular localization of RyR1 in the superficial sarcoplasmic reticulum; by contrast, RyR2 and RyR3 are mainly expressed in the deep sarcoplasmic reticulum. Paradoxically, lack of depolarization-induced Ca(2+) sparks in Ryr1(-/-) myocytes was accompanied by an increased number of cells displaying spontaneous or depolarization-induced Ca(2+) waves. Investigation of protein expression showed that FK506-binding protein (FKBP) 12 and FKBP12.6 (both of which are RyR-associated proteins) are downregulated in Ryr1(-/-) myocytes, whereas expression of RyR2 and RyR3 are unchanged. Moreover, treatment with rapamycin, which uncouples FKBPs from RyR, led to an increase of RyR-dependent Ca(2+) signaling in wild-type urinary bladder myocytes but not in Ryr1(-/-) myocytes. In conclusion, although decreased amounts of FKBP increase Ca(2+) signals in Ryr1(-/-) urinary bladder myocytes the depolarization-induced Ca(2+) sparks are specifically lost, demonstrating that RyR1 is required for depolarization-induced Ca(2+) sparks and suggesting that the intracellular localization of RyR1 fine-tunes Ca(2+) signals in smooth muscle.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Muscle, Smooth/metabolism , Myocytes, Smooth Muscle/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Urinary Bladder/anatomy & histology , Animals , Antibiotics, Antineoplastic/metabolism , Caffeine/metabolism , Cells, Cultured , Central Nervous System Stimulants/metabolism , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myocytes, Smooth Muscle/cytology , Ryanodine Receptor Calcium Release Channel/genetics , Sirolimus/metabolism , Tacrolimus Binding Protein 1A/genetics , Tacrolimus Binding Protein 1A/metabolism , Urinary Bladder/metabolism
11.
Am J Physiol Heart Circ Physiol ; 292(5): H2202-11, 2007 May.
Article in English | MEDLINE | ID: mdl-17209005

ABSTRACT

Patients with mutations in the mitochondrial very-long-chain acyl-CoA dehydrogenase (VLCAD) gene are at risk for cardiomyopathy, myocardial dysfunction, ventricular tachycardia (VT), and sudden cardiac death. The mechanism is not known. Here we report a novel mechanism of VT in mice lacking VLCAD (VLCAD(-/-)). These mice exhibited polymorphic VT and increased incidence of VT after isoproterenol infusion. Polymorphic VT was induced in 10 out of 12 VLCAD(-/-) mice (83%) when isoproterenol was used. One out of 10 VLCAD(-/-) mice with polymorphic VT had VT with the typical bidirectional morphology. At the molecular level, VLCAD(-/-) cardiomyocytes showed increased levels of cardiac ryanodine receptor 2, phospholamban, and calsequestrin with increased [(3)H]ryanodine binding in heart microsomes. At the single cardiomyocyte level, VLCAD(-/-) cardiomyocytes showed significant increase in diastolic indo 1 and fura 2 fluorescence, with increased Ca(2+) transient amplitude. These changes were associated with altered Ca(2+) dynamics, to include: faster sarcomere contraction, larger time derivative of the upstroke, and shorter time-to-minimum sarcomere length compared with VLCAD(+/+) control cells. The L-type Ca(2+) current characteristics were not different under voltage-clamp conditions in the two VLCAD genotypes. Sarcoplasmic reticulum Ca(2+) load measured as normalized integrated Na(+)/Ca(2+) exchange current after rapid caffeine application was increased by 48% in VLCAD(-/-) cells. We conclude that intracellular Ca(2+) handling represents a possible molecular mechanism of arrhythmias in mice and perhaps in VLCAD-deficient humans.


Subject(s)
Acyl-CoA Dehydrogenase, Long-Chain/deficiency , Calcium Signaling , Calcium/metabolism , Heart Ventricles/physiopathology , Tachycardia, Ventricular/physiopathology , Acyl-CoA Dehydrogenase, Long-Chain/genetics , Animals , Female , Male , Mice , Mice, Knockout
12.
J Biol Chem ; 282(48): 34828-38, 2007 Nov 30.
Article in English | MEDLINE | ID: mdl-17921453

ABSTRACT

The 12.6-kDa FK506-binding protein (FKBP12.6) is considered to be a key regulator of the cardiac ryanodine receptor (RyR2), but its precise role in RyR2 function is complex and controversial. In the present study we investigated the impact of FKBP12.6 removal on the properties of the RyR2 channel and the propensity for spontaneous Ca(2+) release and the occurrence of ventricular arrhythmias. Single channel recordings in lipid bilayers showed that FK506 treatment of recombinant RyR2 co-expressed with or without FKBP12.6 or native canine RyR2 did not induce long-lived subconductance states. [(3)H]Ryanodine binding studies revealed that coexpression with or without FKBP12.6 or treatment with or without FK506 did not alter the sensitivity of RyR2 to activation by Ca(2+) or caffeine. Furthermore, single cell Ca(2+) imaging analyses demonstrated that HEK293 cells co-expressing RyR2 and FKBP12.6 or expressing RyR2 alone displayed the same propensity for spontaneous Ca(2+) release or store overload-induced Ca(2+) release (SOICR). FK506 increased the amplitude and decreased the frequency of SOICR in HEK293 cells expressing RyR2 with or without FKBP12.6, indicating that the action of FK506 on SOICR is independent of FKBP12.6. As with recombinant RyR2, the conductance and ligand-gating properties of single RyR2 channels from FKBP12.6-null mice were indistinguishable from those of single wild type channels. Moreover, FKBP12.6-null mice did not exhibit enhanced susceptibility to stress-induced ventricular arrhythmias, in contrast to previous reports. Collectively, our results demonstrate that the loss of FKBP12.6 has no significant effect on the conduction and activation of RyR2 or the propensity for spontaneous Ca(2+) release and stress-induced ventricular arrhythmias.


Subject(s)
Arrhythmias, Cardiac/metabolism , Sarcoplasmic Reticulum/metabolism , Tacrolimus Binding Proteins/metabolism , Animals , Calcium/metabolism , Dogs , Electrocardiography/methods , Humans , Lipid Bilayers/metabolism , Mice , Mice, Nude , Models, Biological , Muscle Cells/metabolism , Rats , Ryanodine/chemistry , Ryanodine Receptor Calcium Release Channel/metabolism
13.
Biophys J ; 90(1): 164-72, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16214874

ABSTRACT

The cardiac isoform of the ryanodine receptor (RyR2) from dog binds predominantly a 12.6-kDa isoform of the FK506-binding protein (FKBP12.6), whereas RyR2 from other species binds both FKBP12.6 and the closely related isoform FKBP12. The role played by FKBP12.6 in modulating calcium release by RyR2 is unclear at present. We have used cryoelectron microscopy and three-dimensional (3D) reconstruction techniques to determine the binding position of FKBP12.6 on the surface of canine RyR2. Buffer conditions that should favor the "open" state of RyR2 were used. Quantitative comparison of 3D reconstructions of RyR2 in the presence and absence of FKBP12.6 reveals that FKBP12.6 binds along the sides of the square-shaped cytoplasmic region of the receptor, adjacent to domain 9, which forms part of the four clamp (corner-forming) structures. The location of the FKBP12.6 binding site on "open" RyR2 appears similar, but slightly displaced (by 1-2 nm) from that found previously for FKBP12 binding to the skeletal muscle ryanodine receptor that was in the buffer that favors the "closed" state. The conformation of RyR2 containing bound FKBP12.6 differs considerably from that depleted of FKBP12.6, particularly in the transmembrane region and in the clamp structures. The x-ray structure of FKBP12.6 was docked into the region of the 3D reconstruction that is attributable to bound FKBP12.6, to show the relative orientations of amino acid residues (Gln-31, Asn-32, Phe-59) that have been implicated as being critical in interactions with RyR2. A thorough understanding of the structural basis of RyR2-FKBP12.6 interaction should aid in understanding the roles that have been proposed for FKBP12.6 in heart failure and in certain forms of sudden cardiac death.


Subject(s)
Biophysics/methods , Ryanodine Receptor Calcium Release Channel/chemistry , Tacrolimus Binding Proteins/chemistry , Tacrolimus Binding Proteins/metabolism , Animals , Binding Sites , Blotting, Western , Calcium/metabolism , Cell Membrane/metabolism , Cryoelectron Microscopy , Dogs , Electrophoresis, Polyacrylamide Gel , Image Processing, Computer-Assisted , Imaging, Three-Dimensional , Microscopy, Electron , Microsomes/metabolism , Models, Molecular , Molecular Conformation , Muscle, Skeletal/metabolism , Myocardium/metabolism , Patch-Clamp Techniques , Protein Binding , Protein Conformation , Protein Isoforms , Protein Structure, Secondary , Protein Structure, Tertiary , Proteins/chemistry , Recombinant Proteins/chemistry , Ryanodine/chemistry , Sarcoplasmic Reticulum/metabolism , Time Factors , X-Rays
14.
J Cell Sci ; 118(Pt 10): 2261-70, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15870112

ABSTRACT

In this study, we characterized the signalling pathway activated by acetylcholine that encodes Ca2+ oscillations in rat duodenum myocytes. These oscillations were observed in intact myocytes after removal of external Ca2+, in permeabilized cells after abolition of the membrane potential and in the presence of heparin (an inhibitor of inositol 1,4,5-trisphosphate receptors) but were inhibited by ryanodine, indicating that they are dependent on Ca2+ release from intracellular stores through ryanodine receptors. Ca2+ oscillations were selectively inhibited by methoctramine (a M2 muscarinic receptor antagonist). The M2 muscarinic receptor-activated Ca2+ oscillations were inhibited by 8-bromo cyclic adenosine diphosphoribose and inhibitors of adenosine diphosphoribosyl cyclase (ZnCl2 and anti-CD38 antibody). Stimulation of ADP-ribosyl cyclase activity by acetylcholine was evaluated in permeabilized cells by measuring the production of cyclic guanosine diphosphoribose (a fluorescent compound), which resulted from the cyclization of nicotinamide guanine dinucleotide. As duodenum myocytes expressed the three subtypes of ryanodine receptors, an antisense strategy revealed that the ryanodine receptor subtype 2 alone was required to initiate the Ca2+ oscillations induced by acetylcholine and also by cyclic adenosine diphosphoribose and rapamycin (a compound that induced uncoupling between 12/12.6 kDa FK506-binding proteins and ryanodine receptors). Inhibition of cyclic adenosine diphosphoribose-induced Ca2+ oscillations, after rapamycin treatment, confirmed that both compounds interacted with the ryanodine receptor subtype 2. Our findings show for the first time that the M2 muscarinic receptor activation triggered Ca2+ oscillations in duodenum myocytes by activation of the cyclic adenosine diphosphoribose/FK506-binding protein/ryanodine receptor subtype 2 signalling pathway.


Subject(s)
Acetylcholine/physiology , Calcium/metabolism , Cyclic ADP-Ribose/physiology , Myocytes, Smooth Muscle/metabolism , Receptor, Muscarinic M2/physiology , Ryanodine Receptor Calcium Release Channel/metabolism , ADP-ribosyl Cyclase/metabolism , Animals , Cells, Cultured , Duodenum/cytology , Myocytes, Smooth Muscle/drug effects , Protein Isoforms/metabolism , Rats , Sarcoplasmic Reticulum/metabolism , Signal Transduction , Sirolimus/pharmacology , Tacrolimus Binding Proteins/metabolism
15.
Am J Physiol Lung Cell Mol Physiol ; 289(2): L338-48, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15863441

ABSTRACT

Ryanodine receptors (RyRs) of pulmonary arterial smooth muscle cells (PASMCs) play important roles in major physiological processes such as hypoxic pulmonary vasoconstriction and perinatal pulmonary vasodilatation. Recent studies show that three subtypes of RyRs are coexpressed and RyR-gated Ca2+ stores are distributed heterogeneously in systemic vascular myocytes. However, the molecular identity and subcellular distribution of RyRs have not been examined in PASMCs. In this study we detected mRNA and proteins of all three subtypes in rat intralobar PASMCs using RT-PCR and Western blot. Quantitative real-time RT-PCR showed that RyR2 mRNA was most abundant, approximately 15-20 times more than the other two subtypes. Confocal fluorescence microscopy revealed that RyRs labeled with BODIPY TR-X ryanodine were localized in the peripheral and perinuclear regions and were colocalized with sarcoplasmic reticulum labeled with Fluo-5N. Immunostaining showed that the subsarcolemmal regions exhibited clear signals of RyR1 and RyR2, whereas the perinuclear compartments contained mainly RyR1 and RyR3. Ca2+ sparks were recorded in both regions, and their activities were enhanced by a subthreshold concentration of caffeine or by endothelin-1, indicating functional RyR-gated Ca2+ stores. Moreover, 18% of the perinuclear sparks were prolonged [full duration/half-maximum (FDHM) = 193.3 +/- 22.6 ms] with noninactivating kinetics, in sharp contrast to the typical fast inactivating Ca2+ sparks (FDHM = 44.6 +/- 3.2 ms) recorded in the same PASMCs. In conclusion, multiple RyR subtypes are expressed differentially in peripheral and perinuclear RyR-gated Ca2+ stores; the molecular complexity and spatial heterogeneity of RyRs may facilitate specific Ca2+ regulation of cellular functions in PASMCs.


Subject(s)
Calcium/metabolism , Myocytes, Smooth Muscle/physiology , Pulmonary Artery/physiology , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/physiology , Animals , Blotting, Western , Boron Compounds/chemistry , Caffeine/pharmacology , Calcium Signaling/drug effects , Central Nervous System Stimulants/pharmacology , Endothelin-1/pharmacology , Fluorescent Dyes/chemistry , Male , Microscopy, Confocal , Microscopy, Fluorescence , Myocytes, Smooth Muscle/drug effects , Pulmonary Artery/drug effects , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Ryanodine/pharmacology , Ryanodine Receptor Calcium Release Channel/drug effects , Ryanodine Receptor Calcium Release Channel/genetics , Sarcoplasmic Reticulum/drug effects
16.
J Biol Chem ; 279(20): 21287-93, 2004 May 14.
Article in English | MEDLINE | ID: mdl-14985349

ABSTRACT

It is generally believed that alterations of calcium homeostasis play a key role in skeletal muscle atrophy and degeneration observed in Duchenne's muscular dystrophy and mdx mice. Mechanical activity is also impaired in gastrointestinal muscles, but the cellular and molecular mechanisms of this pathological state have not yet been investigated. We showed, in mdx duodenal myocytes, that both caffeine- and depolarization-induced calcium responses were inhibited, whereas acetylcholine- and thapsigargin-induced calcium responses were not significantly affected compared with control mice. Calcium-induced calcium release efficiency was impaired in mdx duodenal myocytes depending only on inhibition of ryanodine receptor expression. Duodenal myocytes expressed both type 2 and type 3 ryanodine receptors and were unable to produce calcium sparks. In control and mdx duodenal myocytes, both caffeine- and depolarization-induced calcium responses were dose-dependently and specifically inhibited with the anti-type 2 ryanodine receptor antibody. A strong inhibition of type 2 ryanodine receptor in mdx duodenal myocytes was observed on the mRNA as well as on the protein level. Taken together, our results suggest that inhibition of type 2 ryanodine receptor expression in mdx duodenal myocytes may account for the decreased calcium release from the sarcoplasmic reticulum and reduced mechanical activity.


Subject(s)
Calcium/physiology , Duodenum/physiopathology , Gene Expression Regulation/genetics , Muscle Cells/physiology , Muscular Dystrophy, Animal/physiopathology , Ryanodine Receptor Calcium Release Channel/genetics , Animals , Calcium Signaling , Membrane Potentials , Mice , Mice, Inbred C57BL , Microsomes/metabolism , Muscle, Smooth/physiopathology , Muscular Dystrophy, Animal/genetics , Ryanodine/metabolism
17.
Cell Tissue Res ; 317(2): 137-45, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15205940

ABSTRACT

The ultrastructural localization of ryanodine receptors (RyR) in sinus endothelial cells of the rat spleen was examined by confocal laser scanning and electron microscopy by using isoform-specific antibodies to each of the RyR isoforms. Immunofluorescence microscopy of tissue cryosections revealed RyR3 to be localized, with a strand-like form, in the superficial layer and within the cytoplasm of endothelial cells. Antibodies to RyR1 and RyR2 did not react indicating RyR3 was the predominant isoform. RyR3 was observed over the cortical layer of actin filaments in the apical part and beneath stress fibers in the basal part of the endothelial cells. The distribution of Ca2+-storing tubulovesicular-structures within endothelial cells was established by tissue sections treated with osmium ferricyanide selectively to stain the sarcoplasmic reticulum and transverse tubules in muscle cells; electron microscopy revealed densely stained tubulovesicular structures located throughout the sinus endothelial cells and interconnected at various sites. These structures closely apposed the plasma membrane at the apical, lateral, and basal surfaces of the cells and occasionally ran closely parallel to the plasma membrane and near to the mitochondria. Immunogold electron microscopy revealed RyR in the membranes of the nucleus, tubulovesicular structures, and subplasmalemmal cisternae. In the subplasmalemmal cisternae at the apical, lateral, and basal surfaces, RyR was detected on the membranes near to the plasma membrane. Labeling was also present on the membranes of tubulovesicular structures near to caveolae and on the cristae of the mitochondria. Thus, RyR probably participates in Ca2+ signal transduction and/or mechanosignal transduction in sinus endothelial cells.


Subject(s)
Endothelial Cells/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/metabolism , Spleen/metabolism , Actin Cytoskeleton/metabolism , Animals , Calcium/metabolism , Calcium Signaling/physiology , Caveolae/metabolism , Caveolae/ultrastructure , Cell Communication/physiology , Endothelial Cells/ultrastructure , Mechanotransduction, Cellular/physiology , Microscopy, Electron, Transmission , Mitochondria/metabolism , Mitochondria/ultrastructure , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/ultrastructure , Protein Isoforms/metabolism , Rats , Rats, Wistar , Sarcoplasmic Reticulum/ultrastructure , Spleen/cytology
18.
J Muscle Res Cell Motil ; 23(4): 285-92, 2002.
Article in English | MEDLINE | ID: mdl-12630702

ABSTRACT

The ryanodine receptors (RyRs) are a class of intracellular calcium release channels of which there are three isoforms. In striated muscle, isoform 1 and isoform 2 are mainly expressed in the terminal cisternae of sarcoplasmic reticulum of skeletal muscle and heart, respectively. Isoform 3 is widely distributed in tissues but in minuscule amounts. These channels release calcium ions from intracellular stores in excitation-contraction coupling for cell signaling. Here, we report the presence of skeletal muscle isoform 1 localized in the intercalated discs (IDs) of human and mouse hearts. By using RyR1 and connexin43 specific antibodies and dual immunofluorescent techniques, both were localized in the proximity of the IDs of human and mouse hearts. We confirmed that RyR1 is localized to the IDs by selective immunoprecipitation of RyR isoform 1 from a subcellular fraction containing IDs from human heart tissue. The functional significance of our observation remains to be elucidated as isoform 1 is involved in depolarization induced calcium release, unlike RyR isoforms 2 and 3 which appear to be involved in calcium induced calcium release.


Subject(s)
Calcium Signaling/physiology , Muscle, Skeletal/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Antibodies , Connexin 43/metabolism , Fluorescent Antibody Technique , Humans , Male , Mice , Muscle Contraction/physiology , Myocardium/cytology , Myocytes, Cardiac/cytology , Protein Isoforms/metabolism , Subcellular Fractions
19.
Proc Natl Acad Sci U S A ; 100(24): 14169-74, 2003 Nov 25.
Article in English | MEDLINE | ID: mdl-14605212

ABSTRACT

The molecular basis for the different roles of IL-2 and IL-15 in lymphocyte function has been poorly defined. Searching for differences that underlie the distinct T cell responses to the two cytokines, we observed a marked susceptibility of the IL-15-induced but not of the IL-2-induced proliferation to rapamycin despite a decrease of p70S6 kinase (p70S6K) activation by the drug in response to both cytokines. Activated splenic T lymphocytes deficient in the FK506-binding protein (FKBP) 12, a target of rapamycin activity, had reduced proliferation in response to IL-15 but not to IL-2. This decreased proliferation was accompanied by reduced activation of p70S6K and of the extracellular signal-regulated kinases (ERK) after IL-15 treatment. In contrast to FKBP12-/- cells, splenic FKBP12.6-/- T cells exhibited a decreased proliferative response to IL-2 in the presence of rapamycin without affecting p70S6K or ERK activation. Thus, IL-15 induces T cell proliferation mainly via FKBP12-mediated p70S6K activation. In contrast, IL-2 signaling involves multiple pathways that include at least one additional pathway that depends on FKBP12.6.


Subject(s)
Interleukin-15/pharmacology , Interleukin-2/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tacrolimus Binding Protein 1A/metabolism , Tacrolimus Binding Proteins/metabolism , Animals , Cell Division/drug effects , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Recombinant Proteins/pharmacology , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , T-Lymphocytes/cytology , Tacrolimus Binding Protein 1A/deficiency , Tacrolimus Binding Protein 1A/genetics , Tacrolimus Binding Proteins/deficiency , Tacrolimus Binding Proteins/genetics
20.
Am J Physiol Cell Physiol ; 286(3): C538-46, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14592808

ABSTRACT

Intracellular Ca2+ release through ryanodine receptors (RyRs) plays important roles in smooth muscle excitation-contraction coupling, but the underlying regulatory mechanisms are poorly understood. Here we show that FK506 binding protein of 12.6 kDa (FKBP12.6) associates with and regulates type 2 RyRs (RyR2) in tracheal smooth muscle. FKBP12.6 binds to RyR2 but not other RyR or inositol 1,4,5-trisphosphate receptors, and FKBP12, known to bind to and modulate skeletal RyRs, does not associate with RyR2. When dialyzed into tracheal myocytes, cyclic ADP-ribose (cADPR) alters spontaneous Ca2+ release at lower concentrations and produces macroscopic Ca2+ release at higher concentrations; neurotransmitter-evoked Ca2+ release is also augmented by cADPR. These actions are mediated through FKBP12.6 because they are inhibited by molar excess of recombinant FKBP12.6 and are not observed in myocytes from FKBP12.6-knockout mice. We also report that force development in FKBP12.6-null mice, observed as a decrease in the concentration/tension relationship of isolated trachealis segments, is impaired. Taken together, these findings point to an important role of the FKBP12.6/RyR2 complex in stochastic (spontaneous) and receptor-mediated Ca2+ release in smooth muscle.


Subject(s)
Calcium/metabolism , Cyclic ADP-Ribose/metabolism , Myocytes, Smooth Muscle/physiology , Tacrolimus Binding Proteins/metabolism , Acetylcholine/pharmacology , Animals , Chlorides/metabolism , Horses , Immunosuppressive Agents/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Knockout , RNA, Messenger/analysis , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/metabolism , Tacrolimus/pharmacology , Tacrolimus Binding Proteins/genetics , Trachea/cytology
SELECTION OF CITATIONS
SEARCH DETAIL