Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 179
Filter
1.
Insect Mol Biol ; 26(1): 25-34, 2017 02.
Article in English | MEDLINE | ID: mdl-27743460

ABSTRACT

Loxosceles intermedia venom comprises a complex mixture of proteins, glycoproteins and low molecular mass peptides that act synergistically to immobilize envenomed prey. Analysis of a venom-gland transcriptome from L. intermedia revealed that knottins, also known as inhibitor cystine knot peptides, are the most abundant class of toxins expressed in this species. Knottin peptides contain a particular arrangement of intramolecular disulphide bonds, and these peptides typically act upon ion channels or receptors in the insect nervous system, triggering paralysis or other lethal effects. Herein, we focused on a knottin peptide with 53 amino acid residues from L. intermedia venom. The recombinant peptide, named U2 -sicaritoxin-Li1b (Li1b), was obtained by expression in the periplasm of Escherichia coli. The recombinant peptide induced irreversible flaccid paralysis in sheep blowflies. We screened for knottin-encoding sequences in total RNA extracts from two other Loxosceles species, Loxosceles gaucho and Loxosceles laeta, which revealed that knottin peptides constitute a conserved family of toxins in the Loxosceles genus. The insecticidal activity of U2 -SCTX-Li1b, together with the large number of knottin peptides encoded in Loxosceles venom glands, suggests that studies of these venoms might facilitate future biotechnological applications of these toxins.


Subject(s)
Brown Recluse Spider/genetics , Cystine-Knot Miniproteins/chemistry , Insecticides/analysis , Phosphoric Diester Hydrolases/chemistry , Spider Venoms/chemistry , Amino Acid Sequence , Animals , Base Sequence , Brown Recluse Spider/metabolism , Conserved Sequence , Cystine-Knot Miniproteins/biosynthesis , Cystine-Knot Miniproteins/genetics , Cystine-Knot Miniproteins/isolation & purification , Diptera , Electrophoresis, Polyacrylamide Gel , Escherichia coli , Molecular Sequence Data , Proteome , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Toxicity Tests , Transcriptome
2.
J Clin Periodontol ; 32(5): 535-8, 2005 May.
Article in English | MEDLINE | ID: mdl-15842271

ABSTRACT

BACKGROUND: Sonic scalers have an elliptical and piezoceramic ultrasonic scalers a linear oscillation pattern. Thus, a sonic scaler "hammers" the tooth surface, irrespective of its alignment to the tooth, whereas a piezoceramic ultrasonic scaler may oscillate parallel to the tooth surface and gently remove calculus if the alignment is correct. The aim of this study was to measure pain on a visual analogue scale (VAS) during removal of supragingival calculus on mandibular incisors with a sonic or an ultrasonic scaler. MATERIAL AND METHODS: Seventy-four periodontally healthy subjects with supragingival calculus on the mandibular incisors were treated with both a sonic and a piezoceramic ultrasonic scaler in a split-mouth design. The sequence of instrument application and allocation of instruments to jaw side were randomized. Patient comfort was assessed with a VAS after treatment. RESULTS: The VAS results did not show any difference between the two instrumentation modalities. CONCLUSION: For calculus removal during prophylaxis the type of power-driven instrument does not seem to have an impact on perceived pain. This means that the oscillation pattern does not influence the pain experience.


Subject(s)
Dental High-Speed Equipment/adverse effects , Dental Scaling/adverse effects , Dentin Sensitivity/etiology , Sonication/adverse effects , Ultrasonic Therapy/adverse effects , Adult , Dental Calculus/therapy , Dental Scaling/instrumentation , Equipment Design , Facial Pain/etiology , Female , Humans , Incisor , Male , Mandible , Pain Measurement , Single-Blind Method , Sonication/instrumentation , Statistics, Nonparametric , Ultrasonic Therapy/instrumentation
3.
Exp Neurol ; 187(2): 410-7, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15144867

ABSTRACT

For the treatment of neurodegenerative disorders such as Parkinson's disease cell or gene therapeutical options are increasingly verified. For such approaches, neural stem cells or astrocytes are discussed as possible cell candidates. As also fetal retinal pigment epithelial cells have been successfully tested for such therapeutical options, we investigated the potential of iris pigment epithelial cells as an autologous source for future cell replacement therapies. Using the ELISA technique, we looked for the secretion of neurotrophic factors under basal and stimulated conditions by iris pigment epithelial cells (IPE) cells and compared them with the secretion of retinal pigment epithelial cells (RPE) cells. As iron plays a causative role in cell death during Parkinson's disease, the iron-binding capacity by IPE cells was investigated. Furthermore, we checked the integrative capacity of IPE cells after transplantation into the striatum of adult rats. Our data reveal that IPE cells produce and secrete a variety of neurotrophic factors which can be stimulated after treatment with cytokines. Following transplantation, the cells can be easily detected by their pigmentation, survive for at least 8 weeks and as shown by electron microscopy integrate within the host tissue. Moreover, cells can be transduced with high efficiency using a third generation adenoviral vector, making them promising vehicles to locally deliver therapeutic proteins for the treatment of neurodegenerative diseases in a combined cell and gene therapeutical approach.


Subject(s)
Epithelial Cells/transplantation , Epithelial Cells/ultrastructure , Iris/cytology , Pigment Epithelium of Eye/transplantation , Pigment Epithelium of Eye/ultrastructure , Animals , Cell Differentiation , Cell Movement , Cell Separation , Epithelial Cells/metabolism , Glial Cell Line-Derived Neurotrophic Factor , Graft Survival/physiology , Humans , Iron/metabolism , Neostriatum/cytology , Neostriatum/surgery , Nerve Growth Factors/biosynthesis , Nerve Growth Factors/metabolism , Neurodegenerative Diseases/therapy , Pigment Epithelium of Eye/metabolism , Rats , Rats, Long-Evans , Stereotaxic Techniques
5.
Proc Natl Acad Sci U S A ; 98(20): 11158-62, 2001 Sep 25.
Article in English | MEDLINE | ID: mdl-11562475

ABSTRACT

We have shown previously that at physiologically relevant oxygen tension (pO(2) approximately 10 mmHg), NO S-nitrosylates 1 of approximately 50 free cysteines per ryanodine receptor 1 (RyR1) subunit and transduces a calcium-sensitizing effect on the channel by means of calmodulin (CaM). It has been suggested that cysteine-3635 is part of a CaM-binding domain, and its reactivity is attenuated by CaM [Porter Moore, C., Zhang, J. Z., Hamilton, S. L. (1999) J. Biol. Chem. 274, 36831-36834]. Therefore, we tested the hypothesis that the effect of NO was mediated by C3635. The full-length RyR1 single-site C3635A mutant was generated and expressed in HEK293 cells. The mutation resulted in the loss of CaM-dependent NO modulation of channel activity and reduced S-nitrosylation by NO to background levels but did not affect NO-independent channel modulation by CaM or the redox sensitivity of the channel to O(2) and glutathione. Our results reveal that different cysteines within the channel have been adapted to serve in nitrosative and oxidative responses, and that S-nitrosylation of the cysteine-containing CaM-binding domain underlies the mechanism of CaM-dependent regulation of RyR1 by NO.


Subject(s)
Calmodulin/metabolism , Cysteine , Muscle, Skeletal/metabolism , Nitric Oxide/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Ryanodine/metabolism , Amino Acid Substitution , Animals , Binding Sites , Cell Line , Glutathione/pharmacology , Humans , Intracellular Membranes/metabolism , Microsomes/metabolism , Mutagenesis, Site-Directed , Nitric Oxide/pharmacology , Oxidation-Reduction , Oxygen/pharmacology , Rabbits , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Ryanodine Receptor Calcium Release Channel/chemistry , Ryanodine Receptor Calcium Release Channel/drug effects , Sarcoplasmic Reticulum/metabolism , Transfection
6.
Neuromuscul Disord ; 11(6-7): 530-7, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11525881

ABSTRACT

Malignant hyperthermia is a pharmacogenetic disorder associated with mutations in Ca(2+) regulatory proteins. It manifests as a hypermetabolic crisis triggered by commonly used anesthetics. Malignant hyperthermia susceptibility is a dominantly inherited predisposition to malignant hyperthermia that can be diagnosed by using caffeine/halothane contracture tests. In a multigenerational North American family with a severe form of malignant hyperthermia that has caused four deaths, a novel RYR1 A2350T missense mutation was identified in all individuals testing positive for malignant hyperthermia susceptibility. The same A2350T mutation was identified in an Argentinean family with two known fatal MH reactions. Functional analysis in HEK-293 cells revealed an altered Ca(2+) dependence and increased caffeine sensitivity of the expressed mutant protein thus confirming the pathogenic potential of the RYR1 A2350T mutation.


Subject(s)
Malignant Hyperthermia/genetics , Mutation, Missense , Ryanodine Receptor Calcium Release Channel/genetics , Amino Acid Substitution , Argentina , Family Health , Female , Genetic Linkage , Genotype , Humans , Male , Pedigree , Phenotype , Radioligand Assay , Ryanodine/metabolism , Ryanodine/pharmacology , Ryanodine Receptor Calcium Release Channel/metabolism , Tritium , United States
7.
Biophys J ; 81(3): 1419-29, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11509356

ABSTRACT

The II-III cytoplasmic loop of the skeletal muscle dihydropyridine receptor (DHPR) alpha(1)-subunit is essential for skeletal-type excitation-contraction coupling. Single channel and [(3)H]ryanodine binding studies with a full-length recombinant peptide (p(666-791)) confirmed that this region specifically activates skeletal muscle Ca2+ release channels (CRCs). However, attempts to identify shorter domains of the II-III loop specific for skeletal CRC activation have yielded contradictory results. We assessed the specificity of the interaction of five truncated II-III loop peptides by comparing their effects on skeletal and cardiac CRCs in lipid bilayer experiments; p(671-680) and p(720-765) specifically activated the submaximally Ca2+-activated skeletal CRC in experiments using both mono and divalent ions as current carriers. A third peptide, p(671-690), showed a bimodal activation/inactivation behavior indicating a high-affinity activating and low-affinity inactivating binding site. Two other peptides (p(681-690) and p(681-685)) that contained an RKRRK-motif and have previously been suggested in in vitro studies to be important for skeletal-type E-C coupling, failed to specifically stimulate skeletal CRCs. Noteworthy, p(671-690), p(681-690), and p(681-685) induced similar subconductances and long-lasting channel closings in skeletal and cardiac CRCs, indicating that these peptides interact in an isoform-independent manner with the CRCs.


Subject(s)
Calcium Channels, L-Type/chemistry , Calcium Channels, L-Type/metabolism , Muscle, Skeletal/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Binding Sites , Calcium/metabolism , Calcium Channels, L-Type/pharmacology , Dose-Response Relationship, Drug , Electric Conductivity , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Molecular Sequence Data , Muscle, Skeletal/drug effects , Myocardium/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Protein Isoforms/metabolism , Protein Structure, Tertiary , Rabbits , Ryanodine/metabolism , Substrate Specificity
8.
J Biol Chem ; 276(25): 22579-85, 2001 Jun 22.
Article in English | MEDLINE | ID: mdl-11306590

ABSTRACT

Fusion proteins and full-length mutants were generated to identify the Ca(2+)-free (apoCaM) and Ca(2+)-bound (CaCaM) calmodulin binding sites of the skeletal muscle Ca(2+) release channel/ryanodine receptor (RyR1). [(35)S]Calmodulin (CaM) overlays of fusion proteins revealed one potential Ca(2+)-dependent (aa 3553-3662) and one Ca(2+)-independent (aa 4302-4430) CaM binding domain. W3620A or L3624D substitutions almost abolished completely, whereas V3619A or L3624A substitutions reduced [(35)S]CaM binding to fusion protein (aa 3553-3662). Three full-length RyR1 single-site mutants (V3619A,W3620A,L3624D) and one deletion mutant (Delta4274-4535) were generated and expressed in human embryonic kidney 293 cells. L3624D exhibited greatly reduced [(35)S]CaM binding affinity as indicated by a lack of noticeable binding of apoCaM and CaCaM (nanomolar) and the requirement of CaCaM (micromolar) for the inhibition of RyR1 activity. W3620A bound CaM (nanomolar) only in the absence of Ca(2+) and did not show inhibition of RyR1 activity by 3 microm CaCaM. V3619A and the deletion mutant bound apoCaM and CaCaM at levels compared with wild type. V3619A activity was inhibited by CaM with IC(50) approximately 200 nm, as compared with IC(50) approximately 50 nm for wild type and the deletion mutant. [(35)S]CaM binding experiments with sarcoplasmic reticulum vesicles suggested that apoCaM and CaCaM bind to the same region of the native RyR1 channel complex. These results indicate that the intact RyR1 has a single CaM binding domain that is shared by apoCaM and CaCaM.


Subject(s)
Calcium/metabolism , Calmodulin/metabolism , Muscle, Skeletal/metabolism , Protein Isoforms/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Calmodulin/chemistry , Cell Line , Humans , Kinetics , Protein Binding , Ryanodine Receptor Calcium Release Channel/chemistry , Sulfur Radioisotopes
9.
J Biol Chem ; 276(23): 20144-53, 2001 Jun 08.
Article in English | MEDLINE | ID: mdl-11274202

ABSTRACT

Metabolically (35)S-labeled calmodulin (CaM) was used to determine the CaM binding properties of the cardiac ryanodine receptor (RyR2) and to identify potential channel domains for CaM binding. In addition, regulation of RyR2 by CaM was assessed in [(3)H]ryanodine binding and single-channel measurements. Cardiac sarcoplasmic reticulum vesicles bound approximately four CaM molecules per RyR2 tetramer in the absence of Ca(2+); in the presence of 100 microm Ca(2+), the vesicles bound 7.5 CaM molecules per tetramer. Purified RyR2 bound approximately four [(35)S]CaM molecules per RyR tetramer, both in the presence and absence of Ca(2+). At least four CaM binding domains were identified in [(35)S]CaM overlays of fusion proteins spanning the full-length RyR2. The affinity (but not the stoichiometry) of CaM binding was altered by redox state as controlled by the presence of either GSH or GSSG. Inhibition of RyR2 activity by CaM was influenced by Ca(2+) concentration, redox state, and other channel modulators. Parallel experiments with the skeletal muscle isoform showed major differences in the CaM binding properties and regulation by CaM of the skeletal and cardiac ryanodine receptors.


Subject(s)
Calmodulin/metabolism , Myocardium/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Allosteric Regulation , Animals , Binding Sites , Calmodulin/antagonists & inhibitors , Dogs , Membrane Potentials , Protein Binding , Rabbits , Ryanodine Receptor Calcium Release Channel/physiology
10.
J Biol Chem ; 276(19): 15625-30, 2001 May 11.
Article in English | MEDLINE | ID: mdl-11278999

ABSTRACT

The skeletal muscle Ca(2+) release channel/ryanodine receptor (RyR1) is a prototypic redox-responsive ion channel. Nearly half of the 101 cysteines per RyR1 subunit are kept in a reduced (free thiol) state under conditions comparable with resting muscle. Here we assessed the effects of physiological determinants of cellular redox state (oxygen tension, reduced (GSH) or oxidized (GSSG) glutathione, and NO/O(2) (released by 3-morpholinosydnonimine)) on RyR1 redox state and activity. Oxidation of approximately 10 RyR1 thiols (from approximately 48 to approximately 38 thiols/RyR1 subunit) had little effect on channel activity. Channel activity increased reversibly as the number of thiols was further reduced to approximately 23/subunit, whereas more extensive oxidation (to approximately 13 thiols/subunit) inactivated the channel irreversibly. Neither S-nitrosylation nor tyrosine nitration contributed to these effects. The results identify at least three functional classes of RyR1 thiols and suggest that 1) the channel may be protected from oxidation by a large reservoir of functionally inert thiols, 2) the channel may be designed to respond to moderate oxidative stress by a change in activation setpoint, and 3) the channel is susceptible to oxidative injury under more extensive conditions.


Subject(s)
Glutathione/metabolism , Molsidomine/pharmacology , Muscle, Skeletal/physiology , Nitric Oxide Donors/pharmacology , Ryanodine Receptor Calcium Release Channel/physiology , Sarcoplasmic Reticulum/physiology , Sulfhydryl Compounds/metabolism , Animals , Glutathione Disulfide/metabolism , Kinetics , Molsidomine/analogs & derivatives , Nitric Oxide/metabolism , Oxidation-Reduction , Protein Subunits , Rabbits , Ryanodine/pharmacokinetics , Ryanodine Receptor Calcium Release Channel/drug effects , Superoxides/metabolism
11.
Physiol Rev ; 81(1): 209-237, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11152758

ABSTRACT

In the past five years, skeletal muscle has emerged as a paradigm of "nitric oxide" (NO) function and redox-related signaling in biology. All major nitric oxide synthase (NOS) isoforms, including a muscle-specific splice variant of neuronal-type (n) NOS, are expressed in skeletal muscles of all mammals. Expression and localization of NOS isoforms are dependent on age and developmental stage, innervation and activity, history of exposure to cytokines and growth factors, and muscle fiber type and species. nNOS in particular may show a fast-twitch muscle predominance. Muscle NOS localization and activity are regulated by a number of protein-protein interactions and co- and/or posttranslational modifications. Subcellular compartmentalization of the NOSs enables distinct functions that are mediated by increases in cGMP and by S-nitrosylation of proteins such as the ryanodine receptor-calcium release channel. Skeletal muscle functions regulated by NO or related molecules include force production (excitation-contraction coupling), autoregulation of blood flow, myocyte differentiation, respiration, and glucose homeostasis. These studies provide new insights into fundamental aspects of muscle physiology, cell biology, ion channel physiology, calcium homeostasis, signal transduction, and the biochemistry of redox-related systems.


Subject(s)
Drosophila Proteins , Muscle, Skeletal/metabolism , Nitric Oxide/metabolism , Animals , Carrier Proteins/metabolism , Caveolin 3 , Caveolins/metabolism , Cell Differentiation/drug effects , Cell Respiration/physiology , Dyneins , Glucose/metabolism , Humans , Isoenzymes/classification , Isoenzymes/metabolism , Mammals , Membrane Potentials/physiology , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle, Skeletal/blood supply , Muscle, Skeletal/drug effects , Muscular Dystrophy, Duchenne/metabolism , Nitric Oxide/pharmacology , Nitric Oxide Synthase/classification , Nitric Oxide Synthase/metabolism , Physical Exertion/physiology , Regional Blood Flow/drug effects , Regional Blood Flow/physiology , Ryanodine Receptor Calcium Release Channel/metabolism
12.
Am J Physiol Regul Integr Comp Physiol ; 279(5): R1889-98, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11049875

ABSTRACT

Two distinct skeletal muscle ryanodine receptors (RyR1s) are expressed in a fiber type-specific manner in fish skeletal muscle (11). In this study, we compare [(3)H]ryanodine binding and single channel activity of RyR1-slow from fish slow-twitch skeletal muscle with RyR1-fast and RyR3 isolated from fast-twitch skeletal muscle. Scatchard plots indicate that RyR1-slow has a lower affinity for [(3)H]ryanodine when compared with RyR1-fast. In single channel recordings, RyR1-slow and RyR1-fast had similar slope conductances. However, the maximum open probability (P(o)) of RyR1-slow was threefold less than the maximum P(o) of RyR1-fast. Single channel studies also revealed the presence of two populations of RyRs in tuna fast-twitch muscle (RyR1-fast and RyR3). RyR3 had the highest P(o) of all the RyR channels and displayed less inhibition at millimolar Ca(2+). The addition of 5 mM Mg-ATP or 2.5 mM beta, gamma-methyleneadenosine 5'-triphosphate (AMP-PCP) to the channels increased the P(o) and [(3)H]ryanodine binding of both RyR1s but also caused a shift in the Ca(2+) dependency curve of RyR1-slow such that Ca(2+)-dependent inactivation was attenuated. [(3)H]ryanodine binding data also showed that Mg(2+)-dependent inhibition of RyR1-slow was reduced in the presence of AMP-PCP. These results indicate differences in the physiological properties of RyRs in fish slow- and fast-twitch skeletal muscle, which may contribute to differences in the way intracellular Ca(2+) is regulated in these muscle types.


Subject(s)
Adenosine Triphosphate/analogs & derivatives , Fishes , Muscle Fibers, Fast-Twitch/physiology , Muscle, Skeletal/physiology , Ryanodine Receptor Calcium Release Channel/physiology , Tuna , Adenine Nucleotides/pharmacology , Adenosine Triphosphate/pharmacology , Air Sacs/metabolism , Animals , Calcium/pharmacology , Electric Conductivity , Muscle Fibers, Slow-Twitch/metabolism , Ryanodine/metabolism , Tritium
13.
Biophys J ; 79(5): 2494-508, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11053125

ABSTRACT

Calcium release units (CRUs) are junctions between the sarcoplasmic reticulum (SR) and exterior membranes that mediates excitation contraction (e-c) coupling in muscle cells. In skeletal muscle CRUs contain two isoforms of the sarcoplasmic reticulum Ca(2+)release channel: ryanodine receptors type 1 and type 3 (RyR1 and RyR3). 1B5s are a mouse skeletal muscle cell line that carries a null mutation for RyR1 and does not express either RyR1 or RyR3. These cells develop dyspedic SR/exterior membrane junctions (i.e., dyspedic calcium release units, dCRUs) that contain dihydropyridine receptors (DHPRs) and triadin, two essential components of CRUs, but no RyRs (or feet). Lack of RyRs in turn affects the disposition of DHPRs, which is normally dictated by a linkage to RyR subunits. In the dCRUs of 1B5 cells, DHPRs are neither grouped into tetrads nor aligned in two orthogonal directions. We have explored the structural role of RyR3 in the assembly of CRUs in 1B5 cells independently expressing either RyR1 or RyR3. Either isoform colocalizes with DHPRs and triadin at the cell periphery. Electron microscopy shows that expression of either isoform results in CRUs containing arrays of feet, indicating the ability of both isoforms to be targeted to dCRUs and to assemble in ordered arrays in the absence of the other. However, a significant difference between RyR1- and RyR3-rescued junctions is revealed by freeze fracture. While cells transfected with RyR1 show restoration of DHPR tetrads and DHPR orthogonal alignment indicative of a link to RyRs, those transfected with RyR3 do not. This indicates that RyR3 fails to link to DHPRs in a specific manner. This morphological evidence supports the hypothesis that activation of RyR3 in skeletal muscle cells must be indirect and provides the basis for failure of e-c coupling in muscle cells containing RyR3 but lacking RyR1 (see the accompanying report, ).


Subject(s)
Muscle, Skeletal/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Amino Acid Sequence , Animals , Biophysical Phenomena , Biophysics , Calcium Channels, L-Type/metabolism , Cell Differentiation , Cell Line , Mice , Microscopy, Confocal , Microscopy, Electron , Muscle, Skeletal/cytology , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ryanodine Receptor Calcium Release Channel/chemistry , Ryanodine Receptor Calcium Release Channel/genetics , Sarcoplasmic Reticulum/metabolism , Transduction, Genetic
14.
Cell ; 102(4): 499-509, 2000 Aug 18.
Article in English | MEDLINE | ID: mdl-10966111

ABSTRACT

Ion channels have been studied extensively in ambient O2 tension (pO2), whereas tissue PO2 is much lower. The skeletal muscle calcium release channel/ryanodine receptor (RyR1) is one prominent example. Here we report that PO2 dynamically controls the redox state of 6-8 out of 50 thiols in each RyR1 subunit and thereby tunes the response to NO. At physiological pO2, nanomolar NO activates the channel by S-nitrosylating a single cysteine residue. Among sarcoplasmic reticulum proteins, S-nitrosylation is specific to RyR1 and its effect on the channel is calmodulin dependent. Neither activation nor S-nitrosylation of the channel occurs at ambient PO2. The demonstration that channel cysteine residues subserve coupled O2 sensor and NO regulatory functions and that these operate through the prototypic allosteric effector calmodulin may have general implications for the regulation of redox-related systems.


Subject(s)
Muscle, Skeletal/physiology , Nitric Oxide/metabolism , Oxygen/metabolism , Ryanodine Receptor Calcium Release Channel/physiology , Signal Transduction , Animals , Calcium/metabolism , Calcium-Transporting ATPases/metabolism , Calmodulin/metabolism , Oxidation-Reduction , Rabbits , Ryanodine/metabolism , Sarcoplasmic Reticulum/metabolism
15.
Biophys J ; 79(2): 828-40, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10920015

ABSTRACT

We tested the hypothesis that part of the lumenal amino acid segment between the two most C-terminal membrane segments of the skeletal muscle ryanodine receptor (RyR1) is important for channel activity and conductance. Eleven mutants were generated and expressed in HEK293 cells focusing on amino acid residue I4897 homologous to the selectivity filter of K(+) channels and six other residues in the M3-M4 lumenal loop. Mutations of amino acids not absolutely conserved in RyRs and IP(3)Rs (D4903A and D4907A) showed cellular Ca(2+) release in response to caffeine, Ca(2+)-dependent [(3)H]ryanodine binding, and single-channel K(+) and Ca(2+) conductances not significantly different from wild-type RyR1. Mutants with an I4897 to A, L, or V or D4917 to A substitution showed a decreased single-channel conductance, loss of high-affinity [(3)H]ryanodine binding and regulation by Ca(2+), and an altered caffeine-induced Ca(2+) release in intact cells. Mutant channels with amino acid residue substitutions that are identical in the RyR and IP(3)R families (D4899A, D4899R, and R4913E) exhibited a decreased K(+) conductance and showed a loss of high-affinity [(3)H]ryanodine binding and loss of single-channel pharmacology but maintained their response to caffeine in a cellular assay. Two mutations (G4894A and D4899N) were able to maintain pharmacological regulation both in intact cells and in vitro but had lower single-channel K(+) and Ca(2+) conductances than the wild-type channel. The results support the hypothesis that amino acid residues in the lumenal loop region between the two most C-terminal membrane segments constitute a part of the ion-conducting pore of RyR1.


Subject(s)
Calcium/metabolism , Membrane Potentials/physiology , Muscle, Skeletal/physiology , Ryanodine Receptor Calcium Release Channel/chemistry , Ryanodine Receptor Calcium Release Channel/physiology , Ryanodine/pharmacokinetics , Amino Acid Sequence , Amino Acid Substitution , Animals , Cell Line , Humans , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Potassium Channels/chemistry , Protein Structure, Secondary , Rabbits , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Transfection
16.
J Biol Chem ; 274(46): 32680-91, 1999 Nov 12.
Article in English | MEDLINE | ID: mdl-10551824

ABSTRACT

The effects of ruthenium red (RR) on the skeletal and cardiac muscle ryanodine receptors (RyRs) were studied in vesicle-Ca(2+) flux, [(3)H]ryanodine binding, and single channel measurements. In vesicle-Ca(2+) flux measurements, RR was more effective in inhibiting RyRs at 0.2 microM than 20 microM free Ca(2+). [(3)H]Ryanodine binding measurements suggested noncompetitive interactions between RR inhibition and Ca(2+) regulatory sites of RyRs. In symmetric 0.25 M KCl with 10-20 microM cytosolic Ca(2+), cytosolic RR decreased single channel activities at positive and negative holding potentials. In close to fully activated skeletal (20 microM Ca(2+) + 2 mM ATP) and cardiac (200 microM Ca(2+)) RyRs, cytosolic RR induced a predominant subconductance at a positive but not negative holding potential. Lumenal RR induced a major subconductance in cardiac RyR at negative but not positive holding potentials and several subconductances in skeletal RyR. The RR-related subconductances of cardiac RyR showed a nonlinear voltage dependence, and more than one RR molecule appeared to be involved in their formation. Cytosolic and lumenal RR also induced subconductances in Ca(2+)-conducting skeletal and cardiac RyRs recorded at 0 mV holding potential. These results suggest that RR inhibits RyRs and induces subconductances by binding to cytosolic and lumenal sites of skeletal and cardiac RyRs.


Subject(s)
Muscle, Skeletal/metabolism , Myocardium/metabolism , Ruthenium Red/pharmacology , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Calcium/metabolism , Dogs , Electric Conductivity , Electrophysiology , Kinetics , Protein Binding/drug effects , Proteolipids/metabolism , Rabbits , Ryanodine/metabolism , Sarcoplasmic Reticulum/metabolism
17.
FEBS Lett ; 459(2): 154-8, 1999 Oct 08.
Article in English | MEDLINE | ID: mdl-10518010

ABSTRACT

Six chimeras of the skeletal muscle (RyR1) and cardiac muscle (RyR2) Ca(2+) release channels (ryanodine receptors) previously used to identify RyR1 dihydropyridine receptor interactions [Nakai et al. (1998) J. Biol. Chem. 273, 13403] were expressed in HEK293 cells to assess their Ca(2+) dependence in [(3)H]ryanodine binding and single channel measurements. The results indicate that the C-terminal one-fourth has a major role in Ca(2+) activation and inactivation of RyR1. Further, our results show that replacement of RyR1 regions with corresponding RyR2 regions can result in loss and/or reduction of [(3)H]ryanodine binding affinity while maintaining channel activity.


Subject(s)
Calcium/metabolism , Muscle, Skeletal/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Cells, Cultured , Humans , Muscle, Skeletal/cytology , Recombinant Fusion Proteins/metabolism , Ryanodine/metabolism , Ryanodine Receptor Calcium Release Channel/physiology , Tritium
18.
J Membr Biol ; 169(3): 189-98, 1999 Jun 01.
Article in English | MEDLINE | ID: mdl-10354465

ABSTRACT

Single channel and [3H]ryanodine binding measurements were performed to test for a direct functional interaction between 2,3-butanedione 2-monoxime (BDM) and the skeletal and cardiac muscle sarcoplasmic reticulum Ca2+ release channels (ryanodine receptors). Single channel measurements were carried out in symmetric 0.25 m KCl media using the planar lipid bilayer method. BDM (1-10 mm) activated suboptimally Ca2+-activated (0.5-1 microM free Ca2+) single, purified and native cardiac and skeletal release channels in a concentration-dependent manner by increasing the number of channel events without a change of single channel conductances. BDM activated the two channel isoforms when added to either side of the bilayer. At a maximally activating cytosolic Ca2+ concentration of 20 microM, BDM was without effect on the cardiac channel, whereas it inhibited skeletal channel activities with IC50 approximately 2.5 mm. In agreement with single channel measurements, high-affinity [3H]ryanodine binding to the two channel isoforms was increased in a concentration-dependent manner at

Subject(s)
Cholinesterase Reactivators/pharmacology , Diacetyl/analogs & derivatives , Heart/physiology , Muscle, Skeletal/physiology , Ryanodine Receptor Calcium Release Channel/physiology , Animals , Calcium/physiology , Diacetyl/pharmacology , Dogs , Dose-Response Relationship, Drug , Ion Channel Gating/drug effects , Muscle Contraction/drug effects , Myocardial Contraction/drug effects , Patch-Clamp Techniques , Rabbits , Ryanodine Receptor Calcium Release Channel/drug effects
20.
Biophys J ; 76(3): 1346-66, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10049318

ABSTRACT

Current was measured from single open channels of the calcium release channel (CRC) of cardiac sarcoplasmic reticulum (over the range +/-180 mV) in pure and mixed solutions (e.g., biionic conditions) of the alkali metal ions Li+, K+, Na+, Rb+, Cs+, ranging in concentration from 25 mM to 2 M. The current-voltage (I-V) relations were analyzed by an extension of the Poisson-Nernst-Planck (PNP) formulation of electrodiffusion, which includes local chemical interaction described by an offset in chemical potential, which likely reflects the difference in dehydration/solvation/rehydration energies in the entry/exit steps of permeation. The theory fits all of the data with few adjustable parameters: the diffusion coefficient of each ion species, the average effective charge distribution on the wall of the pore, and an offset in chemical potential for lithium and sodium ions. In particular, the theory explains the discrepancy between "selectivities" defined by conductance sequence and "selectivities" determined by the permeability ratios (i.e., reversal potentials) in biionic conditions. The extended PNP formulation seems to offer a successful combined treatment of selectivity and permeation. Conductance selectivity in this channel arises mostly from friction: different species of ions have different diffusion coefficients in the channel. Permeability selectivity of an ion is determined by its electrochemical potential gradient and local chemical interaction with the channel. Neither selectivity (in CRC) seems to involve different electrostatic interaction of different ions with the channel protein, even though the ions have widely varying diameters.


Subject(s)
Calcium Channels/metabolism , Myocardium/metabolism , Animals , Biophysical Phenomena , Biophysics , Cations, Monovalent/metabolism , Diffusion , Electric Conductivity , In Vitro Techniques , Membrane Potentials , Models, Biological , Permeability , Static Electricity
SELECTION OF CITATIONS
SEARCH DETAIL
...