Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
Add more filters










Publication year range
1.
Regul Toxicol Pharmacol ; 120: 104842, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33346014

ABSTRACT

Non-dioxin like polychlorinated biphenyls (NDL PCB) are recognized neurotoxicants with implications on altered neurodevelopment and neurodegeneration in exposed organisms. NDL PCB neurotoxic relative potency schemes have been developed for a single mechanism, namely activity toward the ryanodine receptor (RyR), or combined mechanisms including, but not limited to, alterations of RyR and dopaminergic pathways. We compared the applicability of the two neurotoxic equivalency (NEQ) schemes and applied each scheme to PCB mixtures found in environmental and human serum samples. A multiple mechanistic NEQ predicts higher neurotoxic exposure concentrations as compared to a scheme based on the RyR alone. Predictions based on PCB ortho categorization, versus homologue categorization, lead to a higher prediction of neurotoxic exposure concentrations, especially for the mMOA. The application of the NEQ schemes to PCB concentration data suggests that PCBs found in fish from US lakes represent a considerable NEQ exposure to fish consuming individuals, that indoor air of schools contained high NEQ concentrations representing an exposure concern when inhaled by children, and that levels already detected in the serum of adults and children may contribute to neurotoxicity. With further validation and in vivo exposure data the NEQ scheme would help provide a more inclusive measure of risk presented by PCB mixtures.


Subject(s)
Dioxins , Environmental Monitoring/methods , Polychlorinated Biphenyls/toxicity , United States Environmental Protection Agency , Water Pollutants, Chemical/toxicity , Animals , Fishes , Humans , Neurotoxicity Syndromes/diagnosis , Neurotoxicity Syndromes/epidemiology , Polychlorinated Biphenyls/analysis , United States/epidemiology , Water Pollutants, Chemical/analysis
2.
Transl Psychiatry ; 3: e277, 2013 Jul 09.
Article in English | MEDLINE | ID: mdl-23838888

ABSTRACT

Autism spectrum disorders (ASDs) are neurodevelopmental in origin, affecting an estimated 1 in 88 children in the United States. We previously described ASD-specific maternal autoantibodies that recognize fetal brain antigens. Herein, we demonstrate that lactate dehydrogenase A and B (LDH), cypin, stress-induced phosphoprotein 1 (STIP1), collapsin response mediator proteins 1 and 2 (CRMP1, CRMP2) and Y-box-binding protein to comprise the seven primary antigens of maternal autoantibody-related (MAR) autism. Exclusive reactivity to specific antigen combinations was noted in 23% of mothers of ASD children and only 1% of controls. ASD children from mothers with specific reactivity to LDH, STIP1 and CRMP1 and/or cypin (7% vs 0% in controls; P<0.0002; odds ratios of 24.2 (95% confidence interval: 1.45-405)) had elevated stereotypical behaviors compared with ASD children from mothers lacking these antibodies. We describe the first panel of clinically significant biomarkers with over 99% specificity for autism risk thereby advancing our understanding of the etiologic mechanisms and therapeutic possibilities for MAR autism.


Subject(s)
Autistic Disorder/immunology , Autoantibodies/immunology , Brain/growth & development , Nerve Tissue Proteins/immunology , Antibody Specificity/immunology , Blotting, Western , Brain/immunology , Child , Electrophoresis, Gel, Two-Dimensional , Female , Guanine Deaminase/immunology , Humans , Intercellular Signaling Peptides and Proteins/immunology , Isoenzymes/immunology , L-Lactate Dehydrogenase/immunology , Lactate Dehydrogenase 5 , Maternal-Fetal Exchange/immunology , Pregnancy , Stereotyped Behavior , Y-Box-Binding Protein 1/immunology
3.
Proc Natl Acad Sci U S A ; 104(18): 7646-51, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17460041

ABSTRACT

Noncoplanar polychlorinated biphenyls (PCBs) are widely dispersed in human environment and tissues. Here, an exemplar noncoplanar PCB was fed to rat dams during gestation and throughout three subsequent nursing weeks. Although the hearing sensitivity and brainstem auditory responses of pups were normal, exposure resulted in the abnormal development of the primary auditory cortex (A1). A1 was irregularly shaped and marked by internal nonresponsive zones, its topographic organization was grossly abnormal or reversed in about half of the exposed pups, the balance of neuronal inhibition to excitation for A1 neurons was disturbed, and the critical period plasticity that underlies normal postnatal auditory system development was significantly altered. These findings demonstrate that developmental exposure to this class of environmental contaminant alters cortical development. It is proposed that exposure to noncoplanar PCBs may contribute to common developmental disorders, especially in populations with heritable imbalances in neurotransmitter systems that regulate the ratio of inhibition and excitation in the brain. We conclude that the health implications associated with exposure to noncoplanar PCBs in human populations merit a more careful examination.


Subject(s)
Auditory Cortex/drug effects , Maternal Exposure , Neuronal Plasticity/drug effects , Polychlorinated Biphenyls/pharmacology , Animals , Electrophysiology , Female , Hearing/drug effects , Rats , Rats, Sprague-Dawley , Sensitivity and Specificity
4.
Pest Manag Sci ; 57(10): 941-5, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11695187

ABSTRACT

Ryanoids have not attained importance as insecticides, but the increasing number of xenobiotic effectors known to influence Ca2+ signalling by interaction with ryanodine receptors (RyRs) may serve to identify new targets for insect control. A historical review of redox control of microsomal Ca2+ transport is given here, followed by recent evidence indicating that hyperactive Cys residues are an essential component of a transmembrane redox sensor. Focus is on the role of sulfhydryl chemistry in RyR regulation; metabolic quinonoid intermediates from pesticides and other environmental contaminants are of interest in this context.


Subject(s)
Calcium/metabolism , Microsomes/physiology , Ryanodine Receptor Calcium Release Channel/metabolism , Biological Transport , Calcium Signaling , Cell Membrane/physiology , Membrane Potentials , Oxidation-Reduction , Oxidative Stress
5.
Biophys J ; 81(6): 3216-30, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11720987

ABSTRACT

In this investigation we use a "dyspedic" myogenic cell line, which does not express any ryanodine receptor (RyR) isoform, to examine the local Ca(2+) release behavior of RyR3 and RyR1 in a homologous cellular system. Expression of RyR3 restored caffeine-sensitive, global Ca(2+) release and causes the appearance of relatively frequent, spontaneous, spatially localized elevations of [Ca(2+)], as well as occasional spontaneous, propagating Ca(2+) release, in both intact and saponin-permeabilized myotubes. Intact myotubes expressing RyR3 did not, however, respond to K(+) depolarization. Expression of RyR1 restored depolarization-induced global Ca(2+) release in intact myotubes and caffeine-induced global release in both intact and permeabilized myotubes. Both intact and permeabilized RyR1-expressing myotubes exhibited relatively infrequent spontaneous Ca(2+) release events. In intact myotubes, the frequency of occurrence and properties of these RyR1-induced events were not altered by partial K(+) depolarization or by application of nifedipine, suggesting that these RyR1 events are independent of the voltage sensor. The events seen in RyR1-expressing myotubes were spatially more extensive than those seen in RyR3-expressing myotubes; however, when analysis was limited to spatially restricted "Ca(2+) spark"-like events, events in RyR3-expressing myotubes were larger in amplitude and duration compared with those in RyR1. Thus, in this skeletal muscle context, differences exist in the spatiotemporal properties and frequency of occurrence of spontaneous release events generated by RyR1 and RyR3. These differences underscore functional differences between the Ca(2+) release behavior of RyR1 and RyR3 in this homologous expression system.


Subject(s)
Calcium/metabolism , Myocardium/cytology , Potassium/metabolism , Ryanodine Receptor Calcium Release Channel/chemistry , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Blotting, Western , Caffeine/pharmacology , Cell Line , Cell Membrane Permeability , Cells, Cultured , Immunohistochemistry , Mice , Microscopy, Confocal , Muscle, Skeletal/chemistry , Muscle, Skeletal/metabolism , Protein Binding , Protein Isoforms , Recombinant Proteins/metabolism , Ryanodine/pharmacology , Spectrometry, Fluorescence , Time Factors
6.
J Neurosci ; 21(11): 3860-70, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11356874

ABSTRACT

Investigating how calcium release from the endoplasmic reticulum (ER) is triggered and coordinated is crucial to our understanding of how oligodendrocyte progenitor cells (OPs) develop into myelinating cells. Sparks and puffs represent highly localized Ca(2+) release from the ER through ryanodine receptors (RyRs) and inositol trisphosphate receptors (IP(3)Rs), respectively. To study whether sparks or puffs trigger Ca(2+) waves in OPs, we performed rapid high-resolution line scan recordings in fluo-4-loaded OP processes. We found spontaneous and evoked sparks and puffs, and we have identified functional cross talk between IP(3)Rs and RyRs. Local events evoked using the IP(3)-linked agonist methacholine (MeCh) showed significantly different morphology compared with events evoked using the caffeine analog 3,7-dimethyl-1-propargylxanthine (DMPX). Pretreatment with MeCh potentiated DMPX-evoked events, whereas inhibition of RyRs potentiated events evoked by low concentrations of MeCh. Furthermore, activation of IP(3)Rs but not RyRs was critical for Ca(2+) wave initiation. Using immunocytochemistry, we show OPs express the specific Ca(2+) release channel subtypes RyR3 and IP(3)R2 in patches along OP processes. RyRs are coexpressed with IP(3)Rs in some patches, but IP(3)Rs are also found alone. This differential distribution pattern may underlie the differences in local and global Ca(2+) signals mediated by these two receptors. Thus, in OPs, interactions between IP(3)Rs and RyRs determine the spatial and temporal characteristics of calcium signaling, from microdomains to intracellular waves.


Subject(s)
Calcium Channels/metabolism , Calcium Signaling/physiology , Receptor Cross-Talk/physiology , Receptors, Cytoplasmic and Nuclear/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Stem Cells/metabolism , Aniline Compounds , Animals , Calcium/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Fluorescent Dyes , Immunohistochemistry , Inositol 1,4,5-Trisphosphate Receptors , Macrocyclic Compounds , Methacholine Chloride/pharmacology , Muscarinic Agonists/pharmacology , Oligodendroglia/cytology , Oxazoles/pharmacology , Purinergic P1 Receptor Antagonists , Rats , Receptor Aggregation/physiology , Receptors, Muscarinic/metabolism , Ryanodine Receptor Calcium Release Channel/drug effects , Stem Cells/cytology , Theobromine/analogs & derivatives , Theobromine/pharmacology , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/metabolism , Xanthenes
7.
Mol Pharmacol ; 59(3): 506-13, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11179446

ABSTRACT

Polycyclic aromatic hydrocarbons are environmental pollutants known to be carcinogenic and immunotoxic. In intact cell assays, benzo[a]pyrene (B[a]P) disrupts Ca(2+) homeostasis in both immune and nonimmune cells, but the molecular mechanism is undefined. In this study, B[a]P and five metabolites are examined for their ability to alter Ca(2+) transport across microsomal membranes. Using a well-defined model system, junctional SR vesicles from skeletal muscle, we show that a single o-quinone metabolite of B[a]P, B[a]P-7,8-dione, can account for altered Ca(2+) transport across microsomal membranes. B[a]P-7,8-dione induces net Ca(2+) release from actively loaded vesicles in a dose-, time-, and Ca(2+)-dependent manner. In the presence of 5 microM extravesicular Ca(2+), B[a]P-7,8-dione exhibited threshold and EC(50) values of 0.4 and 2 microM, respectively, and a maximal release rate of 2 micromol of Ca(2+) min(-1) mg(-1). The mechanism by which B[a]P-7,8-dione enhanced Ca(2+) efflux was further investigated by measuring macroscopic fluxes and single RyR1 channels reconstituted in bilayer lipid membranes and direct measurements of SERCA catalytic activity. B[a]P-7,8-dione (< or = 20 microM) had no measurable effect on initial rates of Ca(2+) accumulation in the presence of ruthenium red to block ryanodine receptor (RyR1), nor did it alter Ca(2+)-dependent (thapsigargin-sensitive) ATPase activity. B[a]P-7,8-dione selectively altered the function of RyR1 in a time-dependent diphasic manner, first activating then inhibiting channel activity. Considering that RyR1 and its two alternate isoforms are broadly expressed in mammalian cells and their important role in Ca(2+)-signaling, the present results reveal a mechanism by which metabolic bioactivation of B[a]P may mediate RyR dysfunction of pathophysiological significance.


Subject(s)
Benzo(a)pyrene/pharmacology , Benzopyrenes/pharmacology , Calcium/metabolism , Microsomes/drug effects , Ryanodine Receptor Calcium Release Channel/physiology , Animals , Biological Transport/drug effects , Calcium-Transporting ATPases/metabolism , In Vitro Techniques , Microsomes/enzymology , Microsomes/metabolism , Oxidation-Reduction , Quinones/metabolism , Rabbits , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/enzymology , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases
8.
Proc Natl Acad Sci U S A ; 98(5): 2865-70, 2001 Feb 27.
Article in English | MEDLINE | ID: mdl-11226332

ABSTRACT

The ryanodine receptor (RyR) family of proteins constitutes a unique type of calcium channel that mediates Ca(2+) release from endoplasmic reticulum/sarcoplasmic reticulum stores. Ryanodine has been widely used to identify contributions made by the RyR to signaling in both muscle and nonmuscle cells. Ryanodine, through binding to high- and low-affinity sites, has been suggested to block the channel pore based on its ability to induce partial conductance states and irreversible inhibition. We examined the effect of ryanodine on an RyR type 1 (RyR1) point mutant (E4032A) that exhibits a severely compromised phenotype. When expressed in 1B5 (RyR null/dyspedic) myotubes, E4032A is relatively unresponsive to stimulation by cell membrane depolarization or RyR agonists, although the full-length protein is correctly targeted to junctions and interacts with dihydropyridine receptors (DHPRs) inducing their arrangement into tetrads. However, treatment of E4032A-expressing cells with 200-500 microM ryanodine, concentrations that rapidly activate and then inhibit wild-type (wt) RyR1, restores the responsiveness of E4032A-expressing myotubes to depolarization and RyR agonists. Moreover, the restored E4032A channels remain resistant to subsequent exposure to ryanodine. In single-channel studies, E4032A exhibits infrequent (channel-open probability, P(o) < 0.005) and brief (<250 micros) gating events and insensitivity to Ca(2+). Addition of ryanodine restores Ca(2+)-dependent channel activity exhibiting full, 3/4, 1/2, and 1/4 substates. This evidence suggests that, whereas ryanodine does not occlude the RyR pore, it does bind to sites that allosterically induce substantial conformational changes in the RyR. In the case of E4032A, these changes overcome unfavorable energy barriers introduced by the E4032A mutation to restore channel function.


Subject(s)
Point Mutation , Ryanodine Receptor Calcium Release Channel/metabolism , Ryanodine/metabolism , Allosteric Regulation , Base Sequence , Cells, Cultured , DNA Primers , Microscopy, Electron , Protein Binding , Ryanodine Receptor Calcium Release Channel/genetics
9.
J Neurochem ; 76(2): 450-63, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11208908

ABSTRACT

ortho-Substituted PCBs mobilize Ca2+ from isolated brain microsomes by interaction with FKBP12/RyR complexes. Investigation into the cellular importance of this mechanism was undertaken using PC12 cells by fluoroimaging the actions of specific PCB congeners on [Ca2+]i and pH. RyR and IP3R share a common intracellular Ca2+ store in PC12 cells. Perfusion of nM to low microM PCB95 caused a transient rise of [Ca2+]i that was not completely dependent on extracellular Ca2+. Pre-incubation of the cells with ryanodine or FK506 completely eliminated PCB95 responses, suggesting a primary action on the FKPP12/RyR-sensitive store. PCB95, but not PCB126, induced a gradual decrease in cytosolic pH that could be completely eliminated by FK506 pre-incubation of the cells. Direct respiration measurement using isolated brain mitochondria demonstrated that neither of the PCBs directly altered any stage of mitochondrial respiration. These results revealed that PCB95 disrupts intracellular Ca2+ signaling in PC12 cells by interaction with the FKBP12/RyR complex that in turn accelerated cellular metabolism, possibly affecting signaling between ER and mitochondria. Since ortho-substituted PCBs have been shown to be neurotoxic and may affect neurodevelopment, studies on the molecular mechanism by which they alter cellular signaling may provide valuable information on the physiological roles of FKPB12 and RyR on neuronal functions.


Subject(s)
Acidosis/chemically induced , Calcium Signaling/drug effects , Immunophilins/metabolism , Pheochromocytoma/metabolism , Polychlorinated Biphenyls/pharmacology , Animals , Bradykinin/pharmacology , Brain Chemistry , Calcium/metabolism , Cell Respiration/drug effects , Enzyme Inhibitors/pharmacology , Estrogen Antagonists/pharmacology , Hydrogen-Ion Concentration/drug effects , Inositol Phosphates/metabolism , Intracellular Fluid/drug effects , Intracellular Fluid/metabolism , Male , Mitochondria/chemistry , Mitochondria/drug effects , PC12 Cells , Pheochromocytoma/drug therapy , Rats , Rats, Sprague-Dawley , Ryanodine/pharmacology , Structure-Activity Relationship , Tacrolimus/pharmacology
10.
Biophys J ; 79(5): 2509-25, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11053126

ABSTRACT

Of the three known ryanodine receptor (RyR) isoforms expressed in muscle, RyR1 and RyR2 have well-defined roles in contraction. However, studies on mammalian RyR3 have been difficult because of low expression levels relative to RyR1 or RyR2. Using the herpes simplex virus 1 (HSV-1) helper-free amplicon system, we expressed either RyR1 or RyR3 in 1B5 RyR-deficient myotubes. Western blot analysis revealed that RyR1- or RyR3-transduced cells expressed the appropriate RyR isoform of the correct molecular mass. Although RyR1 channels exhibited the expected unitary conductance for Cs(+) in bilayer lipid membranes, 74 of 88 RyR3 channels exhibited pronounced subconductance behavior. Western blot analysis with an FKBP12/12.6-selective antibody reveals that differences in gating behavior exhibited by RyR1 and RyR3 may be, in part, the result of lower affinity of RyR3 for FKBP12. In calcium imaging studies, RyR1 restored skeletal-type excitation-contraction coupling, whereas RyR3 did not. Although RyR3-expressing myotubes were more sensitive to caffeine than those expressing RyR1, they were much less sensitive to 4-chloro-m-cresol (CMC). In RyR1-expressing cells, regenerative calcium oscillations were observed in response to caffeine and CMC but were never seen in RyR3-expressing 1B5 cells. In [(3)H]ryanodine binding studies, only RyR1 exhibited sensitivity to CMC, but both RyR isoforms responded to caffeine. These functional differences between RyR1 and RyR3 expressed in a mammalian muscle context may reflect differences in association with accessory proteins, especially FKBP12, as well as structural differences in modulator binding sites.


Subject(s)
Muscle, Skeletal/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Biophysical Phenomena , Biophysics , Caffeine/pharmacology , Calcium Signaling , Cell Line , Cresols/pharmacology , Electrophysiology , Gene Expression , Herpesvirus 1, Human/genetics , Mice , Muscle Contraction/physiology , 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 , Tacrolimus Binding Protein 1A/metabolism , Transduction, Genetic
11.
J Cell Physiol ; 185(2): 184-99, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11025440

ABSTRACT

Nerve-evoked contractile activity in skeletal muscle regulates transcript and protein levels of many metabolic genes in a coordinate fashion, including the muscle isozyme of glycogen phosphorylase (MGP). Cellular signaling mechanisms mediating the activity-dependent modulation of MGP transcript levels were investigated in a spontaneously contractile rat skeletal muscle cell line (Rmo). Mechanisms regulating MGP mRNA levels in Rmo myotubes were compared with those previously shown to modulate the gene encoding the alpha subunit of the acetylcholine receptor (alphaAChR). Reducing the resting membrane potential from -78 to -30 mV, either electrochemically (KCl) or by increasing Na(+) permeability (veratridine): (1) prevented activation of transverse tubules, (2) impeded calcium release by the sarcoplasmic reticulum (SR), and (3) blocked Rmo contractility. MGP mRNA levels decreased to 30% of control levels and alphaAChR levels increased to 350% following 24 h of depolarization. Differing mechanisms appear to mediate this voltage-dependent regulation of MGP and alphaAChR. Inhibition of SR calcium efflux selectively decreased MGP mRNA levels by 30-50% when using dantrolene, thapsigargin, or a dose of ryanodine shown to inactivate Ca(2+)-induced SR Ca(2+) release (CICR). By contrast, blockade of voltage sensors in transverse tubules with nifedipine, a dihydroaminopyridine (DHAP) antagonist, selectively increased alphaAChR mRNA levels by twofold. These data indicate that the voltage-dependent regulation of AChR gene expression differs from that modulating the MGP gene. KCl-induced depolarization and dantrolene both inhibit pulsatile SR Ca(2+) efflux in Rmo myotubes, but by differing mechanisms. Depolarization and dantrolene comparably reduced MGP mRNA levels and decreased MGP transcript stability from a t(1/2) of 24 h to 14.5 and 16 h, respectively. Reduced transcript stability can account for the observed reduction in mRNA levels of MGP in noncontractile Rmo myotubes and could be a significant regulatory mechanism in skeletal muscle that coordinates the activity-dependent expression of MGP with other glycogenolytic genes.


Subject(s)
Gene Expression Regulation/physiology , Muscle Contraction , Muscle, Skeletal/physiology , Phosphorylases/genetics , Sarcoplasmic Reticulum/physiology , Aminopyridines/antagonists & inhibitors , Animals , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Cell Line , Electrophysiology , Glycolysis , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Nifedipine/pharmacology , Protein Isoforms/genetics , RNA Stability/physiology , RNA, Messenger/metabolism , Rats , Receptors, Cholinergic/genetics , Sarcolemma/physiology
12.
Mol Cell ; 6(2): 421-31, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10983988

ABSTRACT

We report here that RyRs interact with and gate the store-operated hTrp3 and Icrac channels. This gating contributes to activation of hTrp3 and Icrac by agonists. Coupling of hTrp3 to IP3Rs or RyRs in the same cells was found to be mutually exclusive. Biochemical and functional evidence suggest that mutually exclusive coupling reflects clustering and segregation of hTrp3-IP3R and hTrp3-RyR complexes in plasma membrane microdomains. Gating of CCE by RyRs indicates that gating by conformational coupling is not unique to skeletal muscle but is a general mechanism for communication between events in the plasma and endoplasmic reticulum membranes.


Subject(s)
Calcium Channels/physiology , Calcium Signaling , Ion Channel Gating/physiology , Ryanodine Receptor Calcium Release Channel/physiology , Barium/pharmacokinetics , Caffeine/pharmacology , Calcium/metabolism , Calcium Channels/drug effects , Carbachol/pharmacology , Cell Line , Cell Membrane/physiology , Heart/physiology , Humans , Ion Channel Gating/drug effects , Muscle, Skeletal/physiology , Patch-Clamp Techniques , Protein Isoforms/physiology , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Ryanodine Receptor Calcium Release Channel/drug effects , TRPC Cation Channels , Transfection
13.
J Biol Chem ; 275(46): 35902-7, 2000 Nov 17.
Article in English | MEDLINE | ID: mdl-10998414

ABSTRACT

Inositol 1,4,5-trisphosphate receptors (IP(3)R) and ryanodine receptors (RyR) mediate the release of endoplasmic and sarcoplasmic reticulum (ER/SR) Ca(2+) stores and regulate Ca(2+) entry through voltage-dependent or ligand-gated channels of the plasma membrane. A prominent property of ER/SR Ca(2+) channels is exquisite sensitivity to sulfhydryl-modifying reagents. A plausible role for sulfhydryl chemistry in physiologic regulation of Ca(2+) release channels and the fidelity of Ca(2+) release from ER/SR is lacking. This study reveals the existence of a transmembrane redox sensor within the RyR1 channel complex that confers tight regulation of channel activity in response to changes in transmembrane redox potential produced by cytoplasmic and luminal glutathione. A transporter selective for glutathione is co-localized with RyR1 within the SR membrane to maintain local redox potential gradients consistent with redox regulation of ER/SR Ca(2+) release. Hyperreactive sulfhydryls previously shown to reside within the RyR1 complex (Liu, G., and Pessah, I. N. (1994) J. Biol. Chem. 269, 33028-33034) are an essential biochemical component of a transmembrane redox sensor. Transmembrane redox sensing may represent a fundamental mechanism by which ER/SR Ca(2+) channels respond to localized changes in transmembrane glutathione redox potential produced by physiologic and pathophysiologic modulators of Ca(2+) release from stores.


Subject(s)
Ion Channel Gating , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Calcium/metabolism , Carrier Proteins/metabolism , Coumarins/metabolism , Flufenamic Acid , Fluorescent Dyes , Fluorometry , Glutathione/metabolism , Glutathione Disulfide/metabolism , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Membrane Potentials , Oxidation-Reduction , Rabbits , Ryanodine Receptor Calcium Release Channel/chemistry , Sarcoplasmic Reticulum/metabolism , Sulfhydryl Compounds/metabolism
14.
Arch Biochem Biophys ; 379(1): 109-18, 2000 Jul 01.
Article in English | MEDLINE | ID: mdl-10864448

ABSTRACT

The ryanodine-sensitive calcium channels, also called ryanodine receptors, are intracellular Ca(2+)-release channels that have been shown to bind the neutral plant alkaloid ryanodine with nanomolar affinity. The activity of the skeletal muscle (RyR1), cardiac muscle (RyR2), and brain (RyR3) ryanodine receptor isoforms have been shown to be highly regulated by physiological factors including pH, temperature, and ionic strength; endogenous compounds including Ca(2+), Mg(2+), and adenosine triphosphate (ATP); and pharmacological agents including caffeine, ruthenium red, and neomycin. RyR3 is reportedly expressed in diverse tissues including lung; however, specific [(3)H]ryanodine binding sites in mammalian lung tissue have not been characterized. In this study, hamster lung ryanodine binding proteins were shown to specifically bind [(3)H]ryanodine with an affinity similar to that of RyR isoforms found in other tissues and this binding was shown to be sensitive to Ca(2+) concentration, stimulation by caffeine and spermine, and inhibition by Mg(2+), ruthenium red, and neomycin. The solubilized, intact ryanodine binding protein from hamster lung demonstrated approximately the same 30S sedimentation coefficient as RyR1 and RyR2, but a putative ryanodine receptor subunit from hamster lung was not found to cross-react with antibodies specific for the three known isoforms. We conclude that the hamster lung ryanodine binding protein demonstrates sedimentation and binding characteristics that are similar to those of the known RyR isoforms, but may exhibit antigenic dissimilarity from the typical RyR isoforms found in muscle and brain.


Subject(s)
Lung/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Ryanodine/metabolism , Animals , Antibodies/immunology , Binding Sites , Caffeine/pharmacology , Calcium/pharmacology , Centrifugation, Density Gradient , Cricetinae , Hydrogen-Ion Concentration , Kinetics , Magnesium/pharmacology , Neomycin/pharmacology , Osmolar Concentration , Protein Binding/drug effects , Protein Isoforms/immunology , Ruthenium Red/pharmacology , Ryanodine Receptor Calcium Release Channel/immunology , Ryanodine Receptor Calcium Release Channel/isolation & purification , Spermine/pharmacology , Tritium
15.
Proc Natl Acad Sci U S A ; 97(8): 4380-5, 2000 Apr 11.
Article in English | MEDLINE | ID: mdl-10759554

ABSTRACT

In many types of muscle, intracellular Ca(2+) release for contraction consists of brief Ca(2+) sparks. Whether these result from the opening of one or many channels in the sarcoplasmic reticulum is not known. Examining massive numbers of sparks from frog skeletal muscle and evaluating their Ca(2+) release current, we provide evidence that they are generated by multiple channels. A mode is demonstrated in the distribution of spark rise times in the presence of the channel activator caffeine. This finding contradicts expectations for single channels evolving reversibly, but not for channels in a group, which collectively could give rise to a stereotyped spark. The release channel agonists imperatoxin A, ryanodine, and bastadin 10 elicit fluorescence events that start with a spark, then decay to steady levels roughly proportional to the unitary conductances of 35%, 50%, and 100% that the agonists, respectively, promote in bilayer experiments. This correspondence indicates that the steady phase is produced by one open channel. Calculated Ca(2+) release current decays 10- to 20-fold from spark to steady phase, which requires that six or more channels be open during the spark.


Subject(s)
Calcium Channels/physiology , Calcium/physiology , Muscle, Skeletal/physiology , Animals , Ion Channel Gating , Membrane Potentials , Rana pipiens
16.
Am J Physiol Cell Physiol ; 278(3): C619-26, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10712251

ABSTRACT

Analysis of RyR1 structure function in muscle cells is made difficult by the low (<5%) transfection efficiencies of myoblasts or myotubes using calcium phosphate or cationic lipid techniques. We inserted the full-length 15.3-kb RyR1 cDNA into a herpes simplex virus type 1 (HSV-1) amplicon vector, pHSVPrPUC between the ori/IE 4/5 promoter sequence and the HSV-1 DNA cleavage/packaging signal (pac). pHSVGN and pHSVGRyR1, two amplicons that expressed green fluorescent protein, were used for fluorescence-activated cell sorter analysis of transduction efficiency. All amplicons were packaged into HSV-1 virus particles using a helper virus-free packaging system and yielded 10(6) transducing vector units/ml. HSVRyR1, HSVGRyR1, and HSVGN virions efficiently transduced mouse myoblasts and myotubes, expressing the desired product in 70-90% of the cells at multiplicity of infection 5. The transduced cells appeared healthy and RyR1 produced by this method was targeted properly and restored skeletal excitation-contraction coupling in dyspedic myotubes. The myotubes produced sufficient protein to allow single-channel analyses from as few as 10 100-mm dishes. In most cases this method could preclude the need for permanent transfectants for the study of RyR1 structure function.


Subject(s)
Gene Transfer Techniques , Herpesvirus 1, Human/genetics , Muscle, Skeletal/physiology , Ryanodine Receptor Calcium Release Channel/physiology , Transfection/methods , Animals , Animals, Newborn , Calcium/pharmacology , Cell Differentiation , Cells, Cultured , Genetic Vectors , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/cytology , Promoter Regions, Genetic , Recombinant Proteins/biosynthesis , Ryanodine/pharmacology , Ryanodine Receptor Calcium Release Channel/genetics
17.
FEBS Lett ; 469(2-3): 179-85, 2000 Mar 10.
Article in English | MEDLINE | ID: mdl-10713267

ABSTRACT

Maurocalcine is a novel toxin isolated from the venom of the chactid scorpion Scorpio maurus palmatus. It is a 33-mer basic peptide cross-linked by three disulfide bridges, which shares 82% sequence identity with imperatoxin A, a scorpion toxin from the venom of Pandinus imperator. Maurocalcine is peculiar in terms of structural properties since it does not possess any consensus motif reported so far in other scorpion toxins. Due to its low concentration in venom (0.5% of the proteins), maurocalcine was chemically synthesized by means of an optimized solid-phase method, and purified after folding/oxidation by using both C18 reversed-phase and ion exchange high-pressure liquid chromatographies. The synthetic product (sMCa) was characterized. The half-cystine pairing pattern of sMCa was identified by enzyme-based cleavage and Edman sequencing. The pairings were Cys3-Cys17, Cys10-Cys21, and Cys16-Cys32. In vivo, the sMCa was lethal to mice following intracerebroventricular inoculation (LD(50), 20 microg/mouse). In vitro, electrophysiological experiments based on recordings of single channels incorporated into planar lipid bilayers showed that sMCa potently and reversibly modifies channel gating behavior of the type 1 ryanodine receptor by inducing prominent subconductance behavior.


Subject(s)
Ryanodine Receptor Calcium Release Channel/chemistry , Scorpion Venoms/chemical synthesis , Amino Acid Sequence , Animals , Chromatography, High Pressure Liquid , Cystine/chemistry , Electrophysiology , Injections, Intraventricular , Ion Channel Gating , Lethal Dose 50 , Lipid Bilayers , Mice , Molecular Sequence Data , Scorpion Venoms/chemistry , Scorpion Venoms/toxicity , Sequence Analysis, Protein , Sequence Homology, Amino Acid
18.
Antioxid Redox Signal ; 2(1): 17-25, 2000.
Article in English | MEDLINE | ID: mdl-11232595

ABSTRACT

Several laboratories using chemically heterogeneous sulfhydryl modifying agents have shown that sarcoplasmic reticulum (SR) Ca2+ channels known as ryanodine receptors (RyRs) are especially sensitive to modification of functionally important cysteine residues. The functional consequence of sulfhydryl modification of RyRs can include phases of activation and inhibition that are very much dependent on the concentration of the reagent used, the length of exposure, and the nature of the chemical reaction the reagent undertakes with sulfhydryl groups. Most challenging is understanding the relationship for how specific sulfhydryl moieties ascribe specific aspects of RyR function. Considering the structural complexity of the RyR complex with its associated proteins, this task is likely to be a formidable one. A small number of hyperreactive thiols have been shown to exist within the RyR complex. Their functional role does not appear to impact directly on channel gating. Rather hyperreactive cysteine (Cys) moieties may represent biochemical components of a redox sensor that conveys information about localized changes in redox potential produced by physiologic (e.g., glutathione, nitric oxide) and pathophysiologic (quinones, reactive oxygen species) channel modulators to the Ca2+ release process. The molecular and functional details of such a redox sensor remains to be elucidated.


Subject(s)
Calcium Signaling/physiology , Carrier Proteins , Cystine/metabolism , Muscle Proteins/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/metabolism , Sulfhydryl Compounds/physiology , Animals , Calcium Signaling/drug effects , Calcium-Transporting ATPases/metabolism , Feedback , Glutathione/physiology , Humans , Intracellular Signaling Peptides and Proteins , Ion Transport/drug effects , Macromolecular Substances , Metals, Heavy/pharmacology , Muscle Proteins/chemistry , Muscle Proteins/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/ultrastructure , Myocardium/metabolism , Myocardium/ultrastructure , Nitric Oxide/physiology , Oxidation-Reduction , Oxidative Stress , Protein Conformation/drug effects , Quinones/pharmacology , Quinones/toxicity , Rabbits , Rats , Reactive Oxygen Species , Ryanodine Receptor Calcium Release Channel/chemistry , Ryanodine Receptor Calcium Release Channel/drug effects , Sarcoplasmic Reticulum/drug effects , Sulfhydryl Compounds/pharmacology
19.
J Biol Chem ; 274(46): 32603-12, 1999 Nov 12.
Article in English | MEDLINE | ID: mdl-10551814

ABSTRACT

The marine sponge Ianthella basta synthesizes at least 25 tetrameric bromotyrosine structures that possess a stringent structural requirement for modifying the gating behavior of ryanodine-sensitive Ca(2+) channels (ryanodine receptors) (RyR)). Bastadin 5 (B5) was shown to stabilize open and closed channel states with little influence on the sensitivity of the channel to activation by Ca(2+) (Mack, M. M., Molinski, T. F., Buck, E. D., and Pessah, I. N. (1994) J. Biol. Chem. 269, 23236-23249). In the present paper, we utilize single channel analysis and measurements of Ca(2+) flux across the sarcoplasmic reticulum to identify bastadin 10 (B10) as the structural congener responsible for dramatically stabilizing the open conformation of the RyR channel, possibly by reducing the free energy associated with closed to open channel transitions (DeltaG*c --> o). The stability of the channel open state induced by B10 sensitized the channel to activation by Ca(2+) to such an extent that it essentially obviated regulation by physiological concentrations of Ca(2+) and relieved inhibition by physiological Mg(2+). These actions of B10 were produced only on the cytoplasmic face of the channel, were selectively eliminated by pretreatment of channels with FK506 or rapamycin, and were reconstituted by human recombinant FKBP12. The actions of B10 were found to be reversible. A structure-activity model is proposed by which substitutions on the Eastern and Western hemispheres of the bastarane macrocycle may confer specificity toward the RyR1-FKBP12 complex to stabilize either the closed or open channel conformation. These results indicate that RyR1-FKBP12 complexes possesses a novel binding domain for phenoxycatechols and raise the possibility of molecular recognition of an endogenous ligand.


Subject(s)
Immunophilins/metabolism , Phenyl Ethers/pharmacology , Ryanodine Receptor Calcium Release Channel/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/pharmacology , Animals , Caffeine/pharmacology , Calcium/metabolism , Electric Conductivity , Halogenated Diphenyl Ethers , Humans , Ion Channel Gating/drug effects , Kinetics , Magnesium/metabolism , Muscle, Skeletal/drug effects , Peptides, Cyclic , Porifera , Protein Conformation/drug effects , Rabbits , Recombinant Proteins , Ryanodine Receptor Calcium Release Channel/drug effects , Sarcoplasmic Reticulum/metabolism , Tacrolimus/pharmacology , Tacrolimus Binding Proteins
20.
Mol Pharmacol ; 55(5): 821-31, 1999 May.
Article in English | MEDLINE | ID: mdl-10220560

ABSTRACT

Quinones undergo redox cycling and/or arylation reactions with key biomolecules involved with cellular Ca2+ regulation. The present study utilizes nanomolar quantities of the fluorogenic maleimide 7-diethylamino-3-(4'-maleimidylphenyl)-4-methylcoumarin (CPM) to measure the reactivity of hyperreactive sulfhydryl moieties on sarcoplasmic reticulum (SR) membranes in the presence and absence of quinones by analyzing the kinetics of forming CPM-thioether adducts and localization of fluorescence by SDS-polyacrylamide gel electrophoresis. Doxorubicin, 1,4-naphthoquinone (NQ), and 1, 4-benzoquinone (BQ) are found to selectively and dose-dependently interact with a class of hyperreactive sulfhydryl groups localized on ryanodine-sensitive Ca2+ channels [ryanodine receptor (RyR)], and its associated protein, triadin, of skeletal type channels. NQ and BQ are the most potent compounds tested for reducing the rate of CPM labeling of hyperreactive SR thiols (IC50 = 0.3 and 1.8 microM, respectively) localized on RyR and associated protein. The reduced forms of quinone, tert-butylhydroquinone, and 5-imino-daunorubicin do not alter significantly the pattern or kinetics of CPM labeling up to 100 microM, demonstrating that the quinone group is essential for modulating the state of hyperreactive SR thiols. Nanomolar NQ is shown to enhance the association of [3H]ryanodine for its high-affinity binding site and directly enhance channel-open probability in bilayer lipid membrane in a reversible manner. By contrast, micromolar NQ produces a time-dependent biphasic action on channel function, leading to irreversible channel inactivation. These results provide evidence that nanomolar quinone selectively and reversibly alters the redox state of hyperreactive sulfhydryls localized in the RyR/Ca2+ channel complex, resulting in enhanced channel activation. The Ca2+-dependent cytotoxicities observed with reactive quinones formed at the microsomal surface by oxidative metabolism may be related to their ability to selectively modify hyperreactive thiols regulating normal functioning of microsomal Ca2+ release channels.


Subject(s)
Carrier Proteins , Cysteine/metabolism , Quinones/pharmacology , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Biological Transport/drug effects , Calcium/metabolism , Coumarins/metabolism , Dose-Response Relationship, Drug , Kinetics , Muscle Proteins/metabolism , Rabbits , Ryanodine Receptor Calcium Release Channel/drug effects , Ryanodine Receptor Calcium Release Channel/physiology , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...