117 results on '"Pessah, I. N."'
Search Results
2. A Transgenic Myogenic Cell Line Lacking Ryanodine Receptor Protein for Homologous Expression Studies: Reconstitution of Ry 1 R Protein and Function
- Author
-
Moore, R. A., Nguyen, H., Galceran, J., Pessah, I. N., and Allen, P. D.
- Published
- 1998
3. Rare coding variation provides insight into the genetic architecture and phenotypic context of autism
- Author
-
J. M., Fu, Satterstrom, F. K., Peng, M., Brand, H., Collins, R. L., Dong, S., Wamsley, B., Klei, L., Wang, L., Hao, S. P., Stevens, C. R., Cusick, C., Babadi, M., Banks, E., Collins, B., Dodge, S., Gabriel, S. B., Gauthier, L., Lee, S. K., Liang, L., Ljungdahl, A., Mahjani, B., Sloofman, L., Smirnov, A. N., Barbosa, M., Betancur, C., Brusco, A., Chung, B. H. Y., Cook, E. H., Cuccaro, M. L., Domenici, E., Ferrero, G. B., Gargus, J. J., Herman, G. E., Hertz-Picciotto, I., Maciel, P., Manoach, D. S., Passos-Bueno, M. R., Persico, A., Renieri, A., Sutcliffe, J. S., Tassone, F., Trabetti, E., Campos, G., Cardaropoli, S., Carli, D., Chan, M. C. Y., Fallerini, C., Giorgio, E., Girardi, A. C., Hansen-Kiss, E., Lee, S. L., Lintas, C., Ludena, Y., Nguyen, R., Pavinato, L., Pericak-Vance, M., Pessah, I. N., Schmidt, R. J., Smith, M., Costa, C. I. S., Trajkova, S., Wang, J. Y. T., M. H. C., Yu, Aleksic, B., Artomov, M., Benetti, E., Biscaldi-Schafer, M., Borglum, A. D., Carracedo, A., Chiocchetti, A. G., Coon, H., Doan, R. N., Fernandez-Prieto, M., Freitag, C. M., Gerges, S., Guter, S., Hougaard, D. M., Hultman, C. M., Jacob, S., Kaartinen, M., Kolevzon, A., Kushima, I., Lehtimaki, T., Rizzo, C. L., Maltman, N., Manara, M., Meiri, G., Menashe, I., Miller, J., Minshew, N., Mosconi, M., Ozaki, N., Palotie, A., Parellada, M., Puura, K., Reichenberg, A., Sandin, S., Scherer, S. W., Schlitt, S., Schmitt, L., Schneider-Momm, K., Siper, P. M., Suren, P., Sweeney, J. A., Teufel, K., del Pilar Trelles, M., Weiss, L. A., Yuen, R., Cutler, D. J., De Rubeis, S., Buxbaum, J. D., Daly, M. J., Devlin, B., Roeder, K., Sanders, S. J., Talkowski, M. E., Massachusetts General Hospital [Boston], Broad Institute of MIT and Harvard (BROAD INSTITUTE), Harvard Medical School [Boston] (HMS)-Massachusetts Institute of Technology (MIT)-Massachusetts General Hospital [Boston], Carnegie Mellon University [Pittsburgh] (CMU), Harvard Medical School [Boston] (HMS), University of California [San Francisco] (UC San Francisco), University of California (UC), University of California [Los Angeles] (UCLA), University of Pittsburgh School of Medicine, Pennsylvania Commonwealth System of Higher Education (PCSHE), Icahn School of Medicine at Mount Sinai [New York] (MSSM), Neuroscience Paris Seine (NPS), Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS)-Institut de Biologie Paris Seine (IBPS), Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), Università degli studi di Torino = University of Turin (UNITO), Azienda Ospedalerio - Universitaria Città della Salute e della Scienza di Torino = University Hospital Città della Salute e della Scienza di Torino, The University of Hong Kong (HKU), University of Illinois [Chicago] (UIC), University of Illinois System, University of Miami Leonard M. Miller School of Medicine (UMMSM), University of Trento [Trento], University of California [Irvine] (UC Irvine), Nationwide Children's Hospital, University of California [Davis] (UC Davis), Universidade do Minho = University of Minho [Braga], Massachusetts General Hospital [Boston, MA, USA], Escola Politecnica da Universidade de Sao Paulo [Sao Paulo], Università degli Studi di Messina = University of Messina (UniMe), Università degli Studi di Siena = University of Siena (UNISI), Azienda Ospedaliera Universitaria Senese, Vanderbilt University [Nashville], Vanderbilt University School of Medicine [Nashville], Università degli studi di Verona = University of Verona (UNIVR), University of Texas Health Science Center, The University of Texas Health Science Center at Houston (UTHealth), Università Campus Bio-Medico di Roma / University Campus Bio-Medico of Rome ( UCBM), Emory University School of Medicine, Emory University [Atlanta, GA], Helsingin yliopisto = Helsingfors universitet = University of Helsinki, Autism Sequencing Consortium (ASC), Broad Institute Center for Common Disease Genomics (Broad-CCDG), iPSYCH-BROAD Consortium : Branko Aleksic, Mykyta Artomov, Elisa Benetti, Monica Biscaldi-Schafer, Anders D Børglum, Angel Carracedo, Andreas G Chiocchetti, Hilary Coon, Ryan N Doan, Montserrat Fernández-Prieto, Christine M Freitag, Sherif Gerges, Stephen Guter, David M Hougaard, Christina M Hultman, Suma Jacob, Miia Kaartinen, Alexander Kolevzon, Itaru Kushima, Terho Lehtimäki, Caterina Lo Rizzo, Nell Maltman, Marianna Manara, Gal Meiri, Idan Menashe, Judith Miller, Nancy Minshew, Matthew Mosconi, Norio Ozaki, Aarno Palotie, Mara Parellada, Kaija Puura, Abraham Reichenberg, Sven Sandin, Stephen W Scherer, Sabine Schlitt, Lauren Schmitt, Katja Schneider-Momm, Paige M Siper, Pål Suren, John A Sweeney, Karoline Teufel, Maria Del Pilar Trelles, Lauren A Weiss, Ryan Yuen., and Betancur, Catalina
- Subjects
Broad Institute Center for Common Disease Genomics ,Autism Sequencing Consortium ,DNA Copy Number Variations ,Autism Spectrum Disorder ,[SDV.NEU.NB]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Neurobiology ,Autism ,Intellectual and Developmental Disabilities (IDD) ,iPSYCH-BROAD Consortium ,autism spectrum disorders ,disease gene ,copy number variants ,neuropsychiatric disorders ,[SDV.GEN.GH] Life Sciences [q-bio]/Genetics/Human genetics ,GENOMAS ,Medical and Health Sciences ,Article ,Clinical Research ,Genetics ,Humans ,2.1 Biological and endogenous factors ,Genetic Predisposition to Disease ,Autistic Disorder ,Aetiology ,Genetic association study ,Pediatric ,Human Genome ,Neurodevelopmental disorders ,[SDV.NEU.NB] Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Neurobiology ,Biological Sciences ,Autism spectrum disorders ,Brain Disorders ,Mental Health ,[SDV.GEN.GH]Life Sciences [q-bio]/Genetics/Human genetics ,Mutation ,Gene expression ,Biotechnology ,Developmental Biology - Abstract
International audience; Some individuals with autism spectrum disorder (ASD) carry functional mutations rarely observed in the general population. We explored the genes disrupted by these variants from joint analysis of protein-truncating variants (PTVs), missense variants and copy number variants (CNVs) in a cohort of 63,237 individuals. We discovered 72 genes associated with ASD at false discovery rate (FDR) ≤ 0.001 (185 at FDR ≤ 0.05). De novo PTVs, damaging missense variants and CNVs represented 57.5%, 21.1% and 8.44% of association evidence, while CNVs conferred greatest relative risk. Meta-analysis with cohorts ascertained for developmental delay (DD) (n = 91,605) yielded 373 genes associated with ASD/DD at FDR ≤ 0.001 (664 at FDR ≤ 0.05), some of which differed in relative frequency of mutation between ASD and DD cohorts. The DD-associated genes were enriched in transcriptomes of progenitor and immature neuronal cells, whereas genes showing stronger evidence in ASD were more enriched in maturing neurons and overlapped with schizophrenia-associated genes, emphasizing that these neuropsychiatric disorders may share common pathways to risk.
- Published
- 2022
4. Perinatal Exposure to a Noncoplanar Polychlorinated Biphenyl Alters Tonotopy, Receptive Fields, and Plasticity in Rat Primary Auditory Cortex
- Author
-
Kenet, T., Froemke, R. C., Schreiner, C. E., Pessah, I. N., and Merzenich, M. M.
- Published
- 2007
- Full Text
- View/download PDF
5. NON-COPLANAR ENVIRONMENTAL CHEMICALS: IMPLICATIONS FOR AUTISM RISK: C15-03
- Author
-
Pessah, I. N.
- Published
- 2009
6. A Transgenic Myogenic Cell Line Lacking Ryanodine Receptor Protein for Homologous Expression Studies: Reconstitution of Ry1R Protein and Function
- Author
-
Moore, R. A., Nguyen, H., Galceran, J., Pessah, I. N., and Allen, P. D.
- Published
- 1998
7. HSV-1 amplicon vectors are a highly efficient gene delivery system for skeletal muscle myoblasts and myotubes
- Author
-
WANG, Y., FRAEFEL, C., PROTASI, F., MOORE, R. A., FESSENDEN, J. D., PESSAH, I. N., DIFRANCESCO, A., BREAKEFIELD, X., and ALLEN, P. D.
- Subjects
Herpes simplex virus -- Research ,Cellular signal transduction -- Research ,Excitation (Physiology) -- Research ,Virus research -- Research ,Biological sciences - Abstract
Wang, Y., C. Fraefel, F. Protasi, R. A. Moore, J. D. Fessenden, I. N. Pessah, A. DiFrancesco, X. Breakefield, and P. D. Allen. HSV-1 amplicon vectors are a highly efficient gene delivery system for skeletal muscle myoblasts and myotubes. Am. J. Physiol. Cell Physiol. 278: C619-C626, 2000.--Analysis of RyR1 structure function in muscle cells is made difficult by the low ([is less than] 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.sup.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. helper virus-free herpes simplex virus type 1 packaging; RyR1; excitation-contraction coupling; large complementary DNA
- Published
- 2000
8. Polybrominated dipheny ehters in relation to autism and developmental delay : A case-control study
- Author
-
Hertz-Picciotto, I., Bergman, Åke, Fängström, Britta, Rose, M. B., Krakowiak, P., Pessah, I. N., Hansen, R. L., Bennett, D. H., Hertz-Picciotto, I., Bergman, Åke, Fängström, Britta, Rose, M. B., Krakowiak, P., Pessah, I. N., Hansen, R. L., and Bennett, D. H.
- Abstract
Background: Polybrominated diphenyl ethers (PBDEs) are flame retardants used widely and in increasing amounts in the U.S. over the last few decades. PBDEs and their metabolites cross the placenta and studies in rodents demonstrate neurodevelopmental toxicity from prenatal exposures. PBDE exposures occur both via breastfeeding and hand-to-mouth activities in small children. Methods: Participants were 100 children from the CHARGE (CHildhood Autism Risk from Genetics and the Environment) Study, a case-control epidemiologic investigation of children with autism/autism spectrum disorder, with developmental delay and from the general population. Diagnoses of autism were confirmed by the Autism Diagnostic Observation Schedule and Autism Diagnostic Inventory-Revised, and of developmental delay using the Mullen's Scales of Early Learning and the Vineland Adaptive Behavior Scales. Typically developing controls were those with no evidence of delay, autism, or autism spectrum disorder. Eleven PBDE congeners were measured by gas chromatography/mass spectrometry from serum specimens collected after children were assessed. Logistic regression was used to evaluate the association between plasma PBDEs and autism. Results: Children with autism/autism spectrum disorder and developmental delay were similar to typically developing controls for all PBDE congeners, but levels were high for all three groups. Conclusions: Plasma samples collected post-diagnosis in this study may not represent early life exposures due to changes in diet and introduction of new household products containing PBDEs. Studies with direct measurements of prenatal or infant exposures are needed to assess the possible causal role for these compounds in autism spectrum disorders.
- Published
- 2011
- Full Text
- View/download PDF
9. Autism-specific maternal autoantibodies recognize critical proteins in developing brain
- Author
-
Braunschweig, D, primary, Krakowiak, P, additional, Duncanson, P, additional, Boyce, R, additional, Hansen, R L, additional, Ashwood, P, additional, Hertz-Picciotto, I, additional, Pessah, I N, additional, and Van de Water, J, additional
- Published
- 2013
- Full Text
- View/download PDF
10. Murine hippocampal neurons expressing Fmr1 gene premutations show early developmental deficits and late degeneration
- Author
-
Chen, Y., primary, Tassone, F., additional, Berman, R. F., additional, Hagerman, P. J., additional, Hagerman, R. J., additional, Willemsen, R., additional, and Pessah, I. N., additional
- Published
- 2009
- Full Text
- View/download PDF
11. Ryanodine receptor point mutant E4032A reveals an allosteric interaction with ryanodine
- Author
-
Fessenden, J. D., primary, Chen, L., additional, Wang, Y., additional, Paolini, C., additional, Franzini-Armstrong, C., additional, Allen, P. D., additional, and Pessah, I. N., additional
- Published
- 2001
- Full Text
- View/download PDF
12. Bastadins relate ryanodine-sensitive and -insensitive Ca2+ efflux pathways in skeletal SR and BC3H1 cells
- Author
-
Pessah, I. N., primary, Molinski, T. F., additional, Meloy, T. D., additional, Wong, P., additional, Buck, E. D., additional, Allen, P. D., additional, Mohr, F. C., additional, and Mack, M. M., additional
- Published
- 1997
- Full Text
- View/download PDF
13. Molecular interaction between ryanodine receptor and glycoprotein triadin involves redox cycling of functionally important hyperreactive sulfhydryls
- Author
-
Liu, G, primary and Pessah, I N, additional
- Published
- 1994
- Full Text
- View/download PDF
14. Ryanodine and dihydropyridine binding patterns and ryanodine receptor mRNA levels in myopathic hamster heart
- Author
-
Lachnit, W. G., primary, Phillips, M., additional, Gayman, K. J., additional, and Pessah, I. N., additional
- Published
- 1994
- Full Text
- View/download PDF
15. (+)-7S-Hydroxyxestospongin A from the Marine Sponge Xestospongia sp. and Absolute Configuration of (+)-Xestospongin D
- Author
-
Moon, S.-S., MacMillan, J. B., Olmstead, M. M., Ta, T. A., Pessah, I. N., and Molinski, T. F.
- Abstract
The structure of the title compound, (+)-7S-hydroxyxestospongin A was solved by single-crystal X-ray diffraction analysis and the absolute stereochemistry obtained by analysis of the derived R and S Mosher's esters. The absolute configuration of xestospongin D was determined for the first time by analysis of anomalous scattering from the X-ray crystal diffraction data set. Xestospongins A, C, and D, araguspongine C, and demethylxestospongin B exhibited modest antifungal activity (MIC 30−100 g/mL) against various fluconazole-resistant Candida spp., but 7S-hydroxyxestospongin A was inactive.
- Published
- 2002
- Full Text
- View/download PDF
16. Chemical synthesis and characterization of maurocalcine, a scorpion toxin that activates Ca2+ release channel/ryanodine receptors
- Author
-
Fajloun, Z., Kharrat, R., Chen, L., Lecomte, C., Luccio, E. Di, Bichet, D., Ayeb, M. El, Rochat, H., Allen, P. D., and Pessah, I. N.
- Published
- 2000
- Full Text
- View/download PDF
17. Bastadin 10 stabilizes the open conformation of the ryanodine-sensitive Ca(2+) channel in an FKBP12-dependent manner.
- Author
-
Chen, L, Molinski, T F, and Pessah, I N
- 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.
- Published
- 1999
18. Antithetical actions of mitoxantrone and doxorubicin on ryanodine-sensitive Ca++ release channels of rat cardiac sarcoplasmic reticulum: evidence for a competitive mechanism.
- Author
-
Kim, E, Giri, S N, and Pessah, I N
- Abstract
The anthracenedione mitoxantrone (MTX) is examined for activity toward ryanodine-sensitive Ca++ release channels (i.e., ryanodine receptors; RyR) from rat cardiac SR singly or in combination with doxorubicin (DXR). MTX and DXR exhibit antithetical activities toward RyR. Under conditions promoting channel closure, DXR enhances the binding of [3H]ryanodine (EC50 = 44 microM), whereas MTX has higher affinity (EC50 = 25 microM) but > 10-fold lower activity towards activating RyR. Unlike DXR, MTX assayed under conditions promoting channel activation inhibits the binding of [3H]ryanodine (IC50 = 3.3 microM) and does not alter the potency with which Ca++ activates RyR in the presence or absence of Mg++. In the presence of Mg++, MTX does not alter kobs and slows k-1 for [3H]ryanodine binding, whereas DXR accelerates kobs with little change in k-1. The antithetical behavior of MTX and DXR at the RyR raises the possibility that MTX antagonizes the ability of DXR to activate the RyR when the drugs are present in combination. In consonance with this hypothesis, MTX inhibits DXR (60 microM)-sensitized [3H]ryanodine-binding sites in a dose-dependent manner (IC50 = 8.4 microM) and shifts the EC50 for DXR-activated [3H]ryanodine-binding without reducing the maximum occupancy attained at high DXR concentration, revealing the competitive nature of the interaction of DXR and MTX. MTX singly does not alter active Ca++ accumulation by SR but is shown to fully inhibit DXR-induced Ca++ release. The results suggest that MTX antagonizes DXR-activated RyR by binding to mutually exclusive sites and may provide a rational basis for combination therapy aimed at extending the current dose limits of DXR.
- Published
- 1994
19. Reconstitution of N1-methylnicotinamide and p-aminohippuric acid transport in phospholipid vesicles with a protein fraction isolated from dog kidney membranes.
- Author
-
Holohan, P D, Pessah, N I, Pessah, I N, and Ross, C R
- Published
- 1979
20. The delta isomer of hexachlorocyclohexane induces rapid release of the myo-inositol-1,4,5-trisphosphate-sensitive Ca2+ store and blocks capacitative Ca2+ entry in rat basophilic leukemia cells.
- Author
-
Mohr, F C, Alojipan, S V, Dunston, S K, and Pessah, I N
- Abstract
Antigenic stimulation of rat basophilic leukemia cells releases Ca2+ from internal stores and increases membrane permeability to Ca2+. The delta isomer of hexachlorocyclohexane (delta-HCH) is structurally similar to myo-inositol-1,4,5-trisphosphate (IP3) and is a potent releaser of stored Ca2+ from permeabilized cells. This release of Ca2+ is not mediated by a competitive interaction with the IP3 receptor on the Ca2+ release channel on the endoplasmic reticulum. In intact cells, delta-HCH and, to a lesser extent, lindane (gamma-hexachlorocyclohexane) transiently increase the intracellular Ca2+ concentration. The return to basal concentrations is mediated by the plasma membrane Ca2+ pumps and not by resequestration of Ca2+ into intracellular stores. Treatment of cells with delta-HCH (25-100 microM), but not lindane, leads to a progressive inhibition of the antigen- and thapsigargin-stimulated Ca2+ signal. Caffeine, a modulator of the ryanodine receptor Ca2+ channel, attenuates the rise in intracellular Ca2+ induced by delta-HCH, suggesting that ryanodine receptor-like Ca2+ channels may be present in RBL cells. At 25 microM delta-HCH, a concentration that does not inhibit the antigen-stimulated Ca2+ signal, the release of [3H]serotonin from antigen-stimulated cells is enhanced as is secretion of [3H]serotonin from cells pretreated with 25-100 microM lindane. The depletion of Ca2+ from intracellular stores by delta-HCH should evoke Ca2+ entry into the cells by a capacitative mechanism; however; divalent cation permeability across the plasma membrane (Mn2+ influx) is not increased but rather is decreased by delta-HCH. An understanding of the mechanism of action of delta-HCH in releasing stored Ca2+ and blocking Ca2+ influx across the plasma membrane may provide insights into the regulation of capacitative Ca2+ entry in nonexcitable cells.
- Published
- 1995
21. Anthraquinone-sensitized Ca2+ release channel from rat cardiac sarcoplasmic reticulum: possible receptor-mediated mechanism of doxorubicin cardiomyopathy.
- Author
-
Pessah, I N, Durie, E L, Schiedt, M J, and Zimanyi, I
- Abstract
Rat cardiac membrane vesicles enriched in biochemical markers of the junctional region of sarcoplasmic reticulum (SR) and exhibiting ruthenium red-sensitive rapid Ca2+ release have been prepared. Doxorubicin and seven congeners are shown to enhance the binding of [3H]ryanodine to the ryanodine receptor with a strong structural requirement. Doxorubicin enhances the binding of [3H]ryanodine to SR membranes and soluble receptor preparations and induces Ca2+ release from SR vesicles in a highly Ca2(+)-dependent manner, suggesting that anthraquinones promote the open state of the junctional Ca2+ release channel by increasing the affinity of the Ca2+ activator site for Ca2+. Doxorubicin reduces the Kd of [3H]ryanodine binding solely by enhancing the rat of association. Caffeine competes for the same site with anthraquinones, because the caffeine-activated binding of [3H]ryanodine is inhibited by doxorubicin and vice versa. The acute effect of doxorubicin on the cardiac Ca2+ release channel is fully reversible; however, long term treatment (up to 24 hr) with doxorubicin increases the sensitivity of the preparation to subsequent acute challenge with doxorubicin. The thiol-reductive agent dithiothreitol enhances, whereas the reactive disulfide 4,4'-dithiodipyridine reduces, the doxorubicin-enhanced binding of [3H]ryanodine. These results demonstrate that the acute and chronic cardiotoxicity of anthraquinones may be accounted for by a receptor-mediated mechanism. Our findings suggest that the chronic effects observed with the clinical use of anthraquinones may be the result of a receptor-mediated shift in the redox equilibrium of allosteric thiols at the ryanodine receptor complex, which in turn leads to long term sensitization of the Ca2+ release channel.
- Published
- 1990
22. Ryanodine receptor type III (Ry3R) identification in mouse parotid acini. Properties and modulation of [3H]ryanodine-binding sites.
- Author
-
DiJulio, D H, Watson, E L, Pessah, I N, Jacobson, K L, Ott, S M, Buck, E D, and Singh, J C
- Abstract
Immunoblot analysis and [3H]ryanodine binding were used to characterize and identify ryanodine receptors (RyRs) in nonexcitable mouse parotid acini. Western analysis revealed ryanodine receptor type III (Ry3R) to be the only detectable isoform in parotid microsomal membranes. Binding of [3H]ryanodine to microsomal fractions was dependent on Ca2+, salt, pH, and temperature. At 23 degrees C, and in the presence of 0.5 M KCl and 100 microM Ca2+, [3H]ryanodine bound specifically to membranes with high affinity (Kd = 6 nM); maximum binding capacity (Bmax) was 275 fmol/mg protein. Mg2+ and ruthenium red inhibited [3H]ryanodine binding (IC50 = 1.4 mM and 0.5 microM, respectively). 4-Chloro-3-ethylphenol enhanced the binding of [3H]ryanodine 2.5-fold; whereas ATP and caffeine were much less efficacious toward activating Ry3R (56% and 18% maximal enhancement, respectively). Bastadin, a novel modulator of the 12-kDa FK506 binding protein.RyR complex, increased [3H]ryanodine binding 3-4-fold by enhancing Kd. The immunosuppressant FK506 enhanced [3H]ryanodine receptor occupancy at >100 microM and antagonized the action of bastadin, suggesting that an immunophilin modulates Ry3R in parotid acini. These results suggest that Ry3R may play an important role in Ca2+ homeostasis in mouse parotid acini.
- Published
- 1997
23. Mechanism of anthraquinone-induced calcium release from skeletal muscle sarcoplasmic reticulum.
- Author
-
Abramson, J J, Buck, E, Salama, G, Casida, J E, and Pessah, I N
- Abstract
The anthraquinones, doxorubicin, mitoxantrone, daunorubicin and rubidazone are shown to be potent stimulators of Ca2+ release from skeletal muscle sarcoplasmic reticulum (SR) vesicles and to trigger transient contractions in chemically skinned psoas muscle fibers. These effects of anthraquinones are the direct consequence of their specific interaction with the [3H] ryanodine receptor complex, which constitutes the Ca2+ release channel from the triadic junction. In the presence of adenine nucleotides and physiological Mg2+ concentrations (approximately 1.0 mM), channel activation by doxorubicin and daunorubicin exhibits a sharp dependence on submicromolar Ca2+ which is accompanied by a selective, dose-dependent increase in the apparent affinity of the ryanodine binding sites for Ca2+, in a manner similar to that previously reported with caffeine. Unlike caffeine, however, anthraquinones increase [3H]ryanodine receptor occupancy to the level observed in the presence of adenine nucleotides. A strong interaction between the anthraquinone and the caffeine binding sites on the Ca2+ release channel is also observed when monitoring Ca2+ fluxes across the SR. Millimolar caffeine both inhibits anthraquinone-stimulated Ca2+ release, and reduces anthraquinone-stimulated [3H]ryanodine receptor occupancy, without changing the effective binding constant of the anthraquinone for its binding site. The degree of cooperativity for daunorubicin activation of Ca2+ release from SR also increases in the presence of caffeine. These results demonstrate that the mechanism by which anthraquinones stimulate Ca2+ release is caused by a direct interaction with the [3H]ryanodine receptor complex, and by sensitization of the Ca2+ activator site for Ca2+.
- Published
- 1988
- Full Text
- View/download PDF
24. Ortho-substituted polychlorinated biphenyls alter microsomal calcium transport by direct interaction with ryanodine receptors of mammalian brain.
- Author
-
Wong, P W, Brackney, W R, and Pessah, I N
- Abstract
A stringent structure-activity relationship among polychlorinated biphenyls (PCBs) possessing two or more ortho-chlorine substituents is observed for activation of ryanodine receptors in mammalian brain, revealing an arylhydrocarbon receptor-independent mechanism through which non-coplanar PCBs disrupt neuronal Ca2+ signaling. Of the congeners assayed, non-coplanar PCB 95 exhibits the highest potency (EC50 = 12-24 microM) toward activating high affinity [3H]ryanodine-binding in rat hippocampus, cerebellum, and cerebral cortex. Coplanar PCB 66 and PCB 126 have no ryanodine receptor activity in all brain regions examined. PCB 95 enhances [3H]ryanodine-binding affinity and capacity by significantly altering modulation by Ca2+ and Mg2+, thereby stabilizing a high affinity conformation of the ryanodine receptor. Ca2+ transport measurements using cortical microsomes reveal that PCB 95 discriminates between inositol 1,4,5-trisphosphate- and ryanodine-sensitive stores. PCB 95 selectively mobilizes Ca2+ from ryanodine-sensitive stores in a dose-dependent manner (EC50 = 3.5 microM) and is completely inhibited by ryanodine receptor blockers, whereas coplanar PCBs are inactive. These data demonstrate that ortho-substituted PCBs disrupt Ca2+ transport in central neurons by direct interaction with ryanodine receptors, showing high selectivity and specificity. Alteration of Ca2+ signaling mediated by ryanodine receptors in specific regions of the central nervous system may account, at least in part, for the significant impact of these agents toward neurodevelopment and neuroplasticity in mammals.
- Published
- 1997
25. Discrimination of multiple binding sites for antagonists of the calcium release channel complex of skeletal and cardiac sarcoplasmic reticulum.
- Author
-
Mack, W M, Zimányi, I, and Pessah, I N
- Abstract
The mechanisms by which ruthenium red (RR), neomycin and FLA 365 ([2,6-dichloro-4-aminophenyl]isopropylamine) inhibit calcium channels of skeletal and cardiac sarcoplasmic reticulum (SR) are characterized. Neomycin and FLA 365 inhibit ryanodine-enhanced calcium release from skeletal SR vesicles in a dose-dependent manner. The apparent affinity of [3H]ryanodine is reduced in a dose-dependent manner by each inhibitor indicative of competitive mechanisms. Displacement studies with skeletal and cardiac SR demonstrate that the order of inhibitory potency is RR greater than neomycin greater than FLA 365 and RR greater than FLA 365 greater than neomycin, respectively. Neomycin is 100-fold less potent in cardiac SR and inhibition of [3H]ryanodine binding is biphasic in both tissues. Neomycin induces a greater proportion of [3H]ryanodine binding states recalcitrant to inhibition in cardiac SR. The ability of neomycin to increase the apparent affinity of [3H]ryanodine for its binding sites is potentiated by RR and attenuated by FLA 365. Kinetic binding studies reveal that increasing neomycin concentrations decreases the association of [3H]ryanodine as predicted for competitive inhibition. However, high (much greater than Kn) neomycin increases [3H]ryanodine binding affinity by slowing dissociation of the radioligand demonstrating that, like micromolar ryanodine, neomycin induces allosterism. Studies with combinations of antagonists demonstrate the existence of two non-overlapping inhibitor recognition sites within the ryanoid site, one polycationic inhibitor site and one FLA 365 inhibitor site. These results suggest that aminoglycoside-induced muscle paralysis may be mediated by direct block of pre- and postsynaptic calcium release channels of endoplasmic reticulum.
- Published
- 1992
26. Stereoselective modulation of ryanodine-sensitive calcium channels by the delta isomer of hexachlorocyclohexane (delta-HCH).
- Author
-
Pessah, I N, Mohr, F C, Schiedt, M, and Joy, R M
- Abstract
delta-Hexachlorocyclohexane (delta-HCH) is shown to be 30-fold more potent as a positive inotropic agent with rat atrial strips compared with lindane (gamma-HCH). Threshold and ED50 values for enhanced contractile force at a pacing frequency of 0.5 Hz are less than 1 microM and 2.2 microM for delta-HCH and 40 microM and 63 microM for gamma-HCH, respectively. Contracture developed in atria exposed to greater than 4 microM delta-HCH (ED50 = 11 microM) but not in atria exposed to gamma-HCH. Uptake and release of Ca++ measured from actively loaded cardiac sarcoplasmic reticulum (SR) vesicles is measured with antipyrylazo III. Although delta-HCH (30 microM) decreases Ca(++)-dependent ATPase by 20%, it does not significantly alter Ca++ loading in the presence of ruthenium red. Addition of delta-HCH (5-50 microM) after loading is complete causes rapid, dose-dependent release of Ca++ from SR. Ca++ release induced by delta-HCH is markedly stereoselective. Compared with gamma-HCH (50 microM), delta-HCH (50 microM) induces a nearly 20-fold higher initial rate of Ca++ release (4.3 nmol of Ca++/mg/sec). Studies with [3H]ryanodine demonstrate that delta-HCH sharply inhibits Ca(++)- or daunorubicin-activated radioligand binding (IC50 = 37 and 25 microM, respectively, logit slope = 2). Inhibition of [3H]ryanodine-binding by delta-HCH is stereoselective inasmuch as IC50 values for alpha, beta and gamma isomers are greater than 100 microM. The delta-HCH modified Ca++ channel appears to proceed by a noncompetitive mechanism (reducing Bmax in equilibrium experiments) with respect to the conformationally sensitive binding site for [3H]ryanodine.(ABSTRACT TRUNCATED AT 250 WORDS)
- Published
- 1992
27. Comparison of [3H]ryanodine receptors and Ca++ release from rat cardiac and rabbit skeletal muscle sarcoplasmic reticulum.
- Author
-
Zimányi, I and Pessah, I N
- Abstract
Sarcoplasmic reticulum (SR) vesicles prepared from rat ventricle muscle are isolated, and their [3H]ryanodine-binding and calcium transport properties are studied in detail under active loading conditions in the presence of pyrophosphate. Experiments are performed in tandem with rabbit skeletal SR under identical conditions to allow direct comparisons of the mechanisms by which activators and inhibitors influence the calcium release channel. Ca(++)-induced Ca++ release is demonstrated with both preparations and the cardiac channel is about 1.5-fold more sensitive to activation by Ca++, which is in excellent quantitative agreement with the ability of Ca++ to activate [3H]ryanodine-binding sites. The cardiac and skeletal receptors show major quantitative differences with respect to sensitivity to pharmacologic modulators, cations and pH. The inhibitors ruthenium red, Mg++ and neomycin are significantly more potent in inhibiting the skeletal receptor, whereas the activators daunorubicin and caffeine are significantly more potent towards the cardiac receptor. The ATP analog, beta,gamma-methyleneadenosine 5'-triphosphate, enhances the binding of [3H]ryanodine to the high-affinity site in skeletal SR by a factor of 4 but has a negligible effect on the cardiac receptor, although at suboptimal Ca++ for the binding of ryanodine, beta,gamma-methyleneadenosine 5'-triphosphate activates the cardiac receptor to a greater extent. High levels of salt (1 M NaCl) enhance the rate of [3H]ryanodine association with its binding sites in both preparations, although they selectively reduce the binding-site capacity in skeletal SR due to a failure to maintain a stable equilibrium. Although high- and low-affinity binding of [3H]ryanodine have a similar response to changing pH, the skeletal receptors are significantly more sensitive to pH.(ABSTRACT TRUNCATED AT 250 WORDS)
- Published
- 1991
28. Ortho-substituted polychlorinated biphenyls alter calcium regulation by a ryanodine receptor-mediated mechanism: structural specificity toward skeletal- and cardiac-type microsomal calcium release channels.
- Author
-
Wong, P W and Pessah, I N
- Abstract
We investigated a novel molecular mechanism by which polychlorinated biphenyls (PCBs) alter microsomal Ca2+ transport with sarcoplasmic reticulum (SR) membranes isolated from skeletal and cardiac muscles. Aroclors with an intermediate weight percent of chlorine enhance by >6-fold the binding of 1 nM[3H]ryanodine to its conformationally sensitive site on the SR Ca2+ -release channel [i.e., ryanodine receptor (RyR)] with high potency (EC50=1.4 microM), whereas Aroclors with either high or low chlorine composition show little activity. Structure-activity studies with selected pentachlorobiphenyl congeners reveal a stringent structural requirement for chlorine substitution at the ortho-positions, with 2,2',3,5',6-pentachlorobiphenyl having the highest potency toward skeletal and cardiac isoforms of RyR (EC50=330 nM and 2 microM, respectively). In contrast, 3,3',4,4',5-pentachlorobiphenyl does not enhance ryanodine binding, suggesting that noncoplanarity of the biphenyl rings is required for channel activation. However, 2,2',4,6,6'-pentachlorobiphenyl is significantly less active toward RyR, suggesting that some degree of rotation about the biphenyl bond is required. 2,2',3,5',6-Pentachlorobiphenyl induces a dose-dependent release of Ca2+ from actively loaded SR vesicles with a maximum rate of 1.2 micromol mg-1 min-1 (EC50=1 microM), whereas 3,3',4,4',5-pentachlorobiphenyl (< / = microM) does not alter Ca2+ transport. The mechanism of PCB-induced channel activation involves a significant decrease in the inhibitory potency of Ca2+ and Mg2+ (20-fold and 100-fold, respectively). Neither 2,2',3,5',6- nor 3,3',4,4',5-pentachlorobiphenyl (< / = 10 microM) alters the activity of the skeletal isoform of sarcoplasmic/endoplasmic reticulum Ca2+ -ATPase or the cardiac isoform of sarcoplasmic/endoplasmic reticulum Ca2+ -ATPase, and PCB-induced Ca2+ release can be fully blocked by either microM ryanodine or ruthenium red. These results are the first to demonstrate a selective ryanodine receptor-mediated mechanism by which ortho-substituted PCBs alter microsomal Ca2+ transport and may have toxicological relevance.
- Published
- 1996
29. Calcium-ryanodine receptor complex. Solubilization and partial characterization from skeletal muscle junctional sarcoplasmic reticulum vesicles.
- Author
-
Pessah, I N, Francini, A O, Scales, D J, Waterhouse, A L, and Casida, J E
- Abstract
The Ca2+-ryanodine receptor complex is solubilized in functional form on treating sarcoplasmic reticulum (SR) vesicles from rabbit fast skeletal muscle with 3-[(3-cholamidopropyl)dimethylammonio]-1-propane-sulfonate (CHAPS) (1 mg/mg protein) and 1 M NaCl at pH 7.1 by shaking for 30 min at 5 degrees C. The heavy membrane preparations obtained from pyrophosphate homogenates frequently exhibit junctional feet and appear to be derived primarily from the terminal cisternae of the SR. The characteristics of [3H]ryanodine binding are similar for the soluble receptor and the heavy SR vesicles with respect to dependence on Ca2+, pharmacological specificity for inhibition by six ryanoids and ruthenium red, and lack of sensitivity to voltage-dependent Ca2+-channel blockers, inositol 1,4,5-trisphosphate, or doxorubicin. In contrast, the cation sensitivity is decreased on receptor solubilization. The soluble receptor is modulated by cyclic nucleotides and rapidly denatured at 50 degrees C. Saturation experiments reveal a single class of receptors (Kd = 9.6 nM), whereas kinetic measurements yield a calculated association constant of 5.5 X 10(6) min-1 M-1 and a dissociation constant of 5.7 X 10(-4) min-1, suggesting that the [3H]ryanodine receptor complex ages with time to a state which is recalcitrant to dissociation. Sepharose chromatography shows that the receptor complex consists primarily of two protein fractions, one of apparent Mr 150,000-300,000 and a second, the [3H]ryanodine binding component, of approximately Mr 1.2 X 10(6). Preliminary analysis of the soluble receptor preparation by sodium dodecyl sulfate-polyacrylamide gel electrophoresis reveals subunits of Mr greater than 200,000 and major bands of calsequestrin and Ca2+-transport ATPase. These findings indicate that [3H]ryanodine binds to the Ca2+-induced open state of the channel involved in the release of contractile Ca2+.
- Published
- 1986
- Full Text
- View/download PDF
30. Ryanodine induces persistent inactivation of the Ca2+ release channel from skeletal muscle sarcoplasmic reticulum.
- Author
-
Zimányi, I, Buck, E, Abramson, J J, Mack, M M, and Pessah, I N
- Abstract
Junctional sarcoplasmic reticulum (SR) membranes isolated from rabbit skeletal muscle were pretreated with 0.1-500 microM ryanodine under equilibrium conditions optimal for receptor binding, followed by the removal of bound alkaloid by several washes in Ca(2+)- and ryanodine-free buffer. Pretreatment with > 100 nM ryanodine results in a concentration-dependent decrease in the Bmax of the high affinity sites and a complete loss of measurable low affinity binding sites that persist for > 48 hr. Quantitative analysis of residual ryanodine using three different methods demonstrates that the inhibition is not the result of residual ryanodine bound to its receptor. Ca2+ transport measurements made with antipyrylazo III show that actively loaded ryanodine-pretreated SR exhibits a persistent insensitivity to ryanodine- and daunomycin-induced Ca2+ release that is not seen with washed control vesicles. Lipid bilayer membranes fused with SR vesicles exhibit rapidly fluctuating single-channel events with a conductance of 468 pS in asymmetric CsCl solutions. Ryanodine (10 microM) produces a unidirectional transition to a slowly fluctuating half-conductance state that is not reversed by perfusion of the bilayer with Ca(2+)-free buffer and subsequent addition of dithiothreitol. However, dithiothreitol added in the ryanodine pretreatment medium offers marked protection against ryanodine-induced loss of binding sites and allows complete restoration of native gating behavior of single channels in bilayer lipid membrane. Using three different experimental approaches, the data demonstrate that the alkaloid at micromolar concentration persistently alters SR Ca2+ release channel function, perhaps by uncoupling four negatively cooperative binding sites. The oxidation of critical receptor thiols is implicated in the process.
- Published
- 1992
31. Characterization of multiple [3H]ryanodine binding sites on the Ca2+ release channel of sarcoplasmic reticulum from skeletal and cardiac muscle: evidence for a sequential mechanism in ryanodine action.
- Author
-
Pessah, I N and Zimanyi, I
- Abstract
Kinetic and equilibrium measurements of [3H]ryanodine binding to the Ca2+ release channel of rabbit skeletal and rat cardiac sarcoplasmic reticulum (SR) are examined to ascertain the nature of cooperative interactions among high and low affinity binding sites and to quantitate their distribution. Equilibrium studies reveal affinities of 1-4 nM for the highest affinity binding site and of 30-50 nM, 500-800 nM, and 2-4 microM for the lower affinity sites in both preparations, with Hill coefficients of significantly less than 1, and initial rates of association and dissociation increase with increasing concentrations of ryanodine. SR vesicles are actively loaded in the presence of pyrophosphate, and fluctuations in extravesicular Ca2+ are measured by the absorbance change of antipyrylazo III. The data demonstrate a biphasic, time- and concentration-dependent action of ryanodine on the release of Ca2+, with an initial activation and a subsequent inactivation phase. Kinetic analysis of the activation of Ca2+ release by ryanodine, in consonance with the binding data, demonstrates the existence of multiple binding sites for the alkaloid on the channel complex, with nanomolar to micromolar affinities. Based on the present findings obtained by receptor binding analysis and Ca2+ transport measurements, we suggest a model that describes four, most plausibly negatively cooperative, binding sites on the Ca2+ release channel. Occupation of ryanodine binding sites produces sequential activation followed by inactivation of the SR channel, revealing the strong possibility of an irreversible uncoupling of the native function of the receptor/channel complex by high concentrations of ryanodine. A model relating ryanodine receptor occupancy with SR Ca2+ release stresses two important new findings regarding the interaction of ryanodine with its receptor. First, ryanodine binds to four sites on the oligomeric channel complex with decreasing affinities, which can be best described by allosteric negative cooperativity. Second, binding of ryanodine to its receptor activates the Ca2+ release channel in a concentration-dependent and saturable manner in the range of 20 nM to 1 mM and produces a kinetically limited and sequential inactivation of the Ca2+ channel, with the concomitant attainment of full negative cooperativity. The results presented suggest that driving of the complex toward full negative cooperativity with high concentrations of ryanodine promotes a long-lived conformational state in which ryanodine is physically occluded and hindered from free diffusion from its binding site.
- Published
- 1991
32. Ca2+-activated ryanodine binding: mechanisms of sensitivity and intensity modulation by Mg2+, caffeine, and adenine nucleotides.
- Author
-
Pessah, I N, Stambuk, R A, and Casida, J E
- Abstract
The Ca2+-ryanodine receptor complex is a functional unit at the terminal cisternae (TC) of the sarcoplasmic reticulum (SR) whose proteins comprise the Ca2+ release channels which may be involved in excitation-contraction coupling. Ca2+, Mg2+, caffeine, and adenine nucleotides, but not inositol 1,4,5-trisphosphate, may exert their inotropic effects on skeletal muscle SR by direct allosteric modulation of the [3H]ryanodine-binding site. Micromolar Ca2+ is primarily responsible for activating [3H]ryanodine binding by regulating receptor site density, affinity, and cooperativity. Mg2+ reduces the sensitivity to Ca2+ activation by directly competing with Ca2+ for the activator site. However, inhibition by Mg2+ is overcome in the presence of beta,gamma-methyleneadenosine 5'-triphosphate (AMP-PCP; 1 mM) or caffeine (20 mM). Caffeine dramatically increases the affinity of the Ca2+ activator site for Ca2+, whereas AMP-PCP or cAMP enhances the gating efficiency or the lifetime of the open state of the TC SR channel. A kinetic model is proposed for four functional domains of the Ca2+-ryanodine receptor complex: the Ca2+-regulatory domain which binds Ca2+ with microM affinity is primarily responsible for gating the Ca2+ channel of the TC SR in a cooperative manner, and is inhibited by mM Mg2+ by direct competition for the activator site which appears to contain critical sulfhydryl groups; a Ca2+-activate alkaloid binding domain in close proximity to the channel which binds ryanodine with nM affinity and rapidly occludes upon complex formation; a domain which binds caffeine with low (greater than mM) affinity and directly influences the sensitivity of the Ca2+-regulatory site; and a domain which binds adenine nucleotides with intermediate affinity (less than mM), does not require phosphorylation, and intensifies the Ca2+ signal which triggers opening of the Ca2+-release channel.
- Published
- 1987
33. Partial characterization of specific cantharidin binding sites in mouse tissues.
- Author
-
Graziano, M J, Pessah, I N, Matsuzawa, M, and Casida, J E
- Abstract
The mode of action of cantharidin, the natural vesicant of blister beetles, is examined by radioligand binding studies with mouse tissues. [3H]Cantharidin undergoes specific and saturable binding with the liver cytosol, which is characterized as follows: Kd and Bmax values of 30 nM and 1.8 pmol/mg of protein, respectively; linearity with respect to protein concentration; pH optimum of 6.5 to 7.5; association and dissociation half-times of 20 min and 12 hr, respectively; and 50% inhibition by Mg2+ at 70 microM, Ca2+ at 224 microM, pyrophosphate at 27 microM, and nucleotide triphosphates at 52-81 microM. The binding site undergoes a loss of activity at 45 degrees or higher. The toxicological relevance of this specific [3H]cantharidin binding site of mouse liver cytosol is established in three ways. First, the potency of 15 active cantharidin analogs for inhibiting [3H]cantharidin binding is correlated with their acute toxicity to mice (r = 0.829). Second, 26 related compounds that are inactive in inhibiting [3H]cantharidin binding are also of little or no toxicity to mice. Finally, the binding of [3H] cantharidin to liver cytosol from mice poisoned with increasing amounts of unlabeled cantharidin is inhibited in a dose-dependent manner. [3H]Cantharidin also specifically binds to cytosol fractions of blood, brain, heart, kidney, lung, pancreas, skin, spleen, and stomach. The characteristics of the specific binding site in brain are very similar to those determined in liver with respect to Kd, Bmax, association/dissociation kinetics, and sensitivity to inhibitors. It therefore appears that the toxicity of cantharidin and related oxabicycloheptanes, including the herbicide endothal, is attributable to binding at a specific site in liver and possibly other tissues.
- Published
- 1988
34. Dyspedic mouse skeletal muscle expresses major elements of the triadic junction but lacks detectable ryanodine receptor protein and function.
- Author
-
Buck, E D, Nguyen, H T, Pessah, I N, and Allen, P D
- Abstract
The ry1(53) dyspedic mouse contains two disrupted alleles for ryanodine receptor type 1 (skeletal isoform of ryanodine receptor; Ry1R) resulting in perinatal death. In the present study, whole skeletal muscle homogenates and sucrose gradient-purified junctional sarcoplasmic reticulum from neonatal wild-type and dyspedic mice were assayed for biochemical and functional markers. Equilibrium binding experiments performed with 1-120 nM [3H]ryanodine reveal saturable high and low affinity binding to membrane preparations from wild-type mice, but not to preparations from dyspedic mice. Binding experiments performed with [3H]PN200 show a 2-fold reduction in [3H]PN200 binding capacity in dyspedic muscle, compared to age-matched wild-type muscle, with no change in receptor affinity. The presence or absence of proteins known to be critical for normal ryanodine receptor/Ca2+ channel complex function was assessed by Western blot analysis. Results indicate that FKBP-12, DHPRalpha1, triadin, calsequestrin, SERCA1 (sarco(endo)plasmic reticulum Ca2+ ATPase), and skeletal muscle myosin heavy chain are present in both dyspedic and wild-type muscle. Only wild-type membranes showed immunoreactivity toward Ry1R antibody. Neither dyspedic nor wild-type mouse muscle showed detectable immunoreactivity toward Ry2R or Ry3R antibodies, even after sucrose gradient purification of sarcoplasmic reticulum. These results indicate that proteins critical for ryanodine receptor function are expressed in dyspedic skeletal muscle in the absence of Ry1R. Ca2+ transport measurements show that membranes from wild-type controls, but not dyspedic mice, release Ca2+ upon exposure to ryanodine. Dyspedic mice and cells derived from them serve as excellent homologous expression systems in which to study how Ry1R structure relates to function.
- Published
- 1997
35. Direct evidence for the existence and functional role of hyperreactive sulfhydryls on the ryanodine receptor-triadin complex selectively labeled by the coumarin maleimide 7-diethylamino-3-(4'-maleimidylphenyl)-4-methylcoumarin.
- Author
-
Liu, G, Abramson, J J, Zable, A C, and Pessah, I N
- Abstract
The fluorogenic sulfhydryl probe 7-diethylamino-3-(4'-maleimidylphenyl)-4-methylcoumarin (CPM) (1-50 nM) is used to characterize the functional role and location of highly reactive thiol groups on the ryanodine-sensitive Ca2+ release channel complex [i.e., ryanodine receptors (RyRs)] of skeletal and cardiac junctional sarcoplasmic reticulum (SR). The kinetics of forming fluorescent CPM adducts with junctional but not longitudinal SR membrane proteins (0.02-1 pmol of CPM/microgram of SR protein) are found to be markedly dependent on the presence of physiological and pharmacological modulators of the RyR Ca2+ channel. RyR agonists, micromolar Ca2+, and nanomolar ryanodine promote a slow SR thiol-CPM reaction, with an apparent rate constant k of 0.0021 +/- 0.0002 sec-1, and > 89% of the fluorescence is associated with the 110-kDa Ca2+ pump, which constitutes 68% of the protein in the SR preparations. However, in the presence of Ca2+ channel antagonists (millimolar Mg2+, millimolar Ca2+, or micromolar ryanodine), CPM rapidly forms adducts with a single class of highly reactive (hyperreactive) SR thiols (k = 0.025 +/- 0.002 sec-1). Nonreducing sodium dodecyl sulfate-polyacrylamide gel electrophoresis of CPM-labeled SR protein and Western blot analyses with antiryanodine or antitriadin antibodies reveal that the hyperreactive thiols labeled by CPM under conditions favoring channel closure are localized principally to the RyR protomer and triadin, which constitute < 6% of the protein in the SR preparation. Immunoprecipitation experiments with antiryanodine and antitriadin monoclonal antibodies confirm the location of CPM-labeled thiol groups on RyR and triadin, respectively. The results indicate that the RyR and triadin contain a small number of highly reactive cysteine residues that selectively conjugate with CPM only when channel closure is favored. It is shown that either 1) the redox state (sulfhydryl/disulfide status) or 2) the accessibility of the hyperreactive thiols on the RyR and triadin is determined by the conformational state of the channel. Covalent modification of hyperreactive thiols with nanomolar CPM inhibits both Ca(2+)-induced Ca2+ release and the gating activity of single channels reconstituted in bilayers, revealing the essential functional importance of hyperreactive thiols on channel-associated proteins. 1,4-Naphthoquinone (0.4-40 pmol/micrograms of protein) selectively oxidizes hyperreactive thiols on RyR and triadin and releases Ca2+ from SR vesicles, without inhibiting Ca(2+)-ATPase activity. The results provide direct evidence of the existence and functional role of hyperreactive cysteine residues on the RyR and triadin in regulating the gating of ryanodine-sensitive intracellular Ca2+ channels and strongly suggest that these important Ca2+ regulatory channels may be an important target for oxidative cell damage mediated by quinones.
- Published
- 1994
36. Bastadin 20 and Bastadin O-Sulfate Esters from Ianthella basta: Novel Modulators of the Ry<INF>1</INF>R FKBP12 Receptor Complex
- Author
-
Franklin, M. A., Penn, S. G., Lebrilla, C. B., Lam, T. H., Pessah, I. N., and Molinski, T. F.
- Abstract
New compounds bastadin 20 (
9 ), 15,34-O-disulfatobastadin 7 (10 ), and 10-O-sulfatobastadin 3 (11 ) were isolated from Ianthella basta collected in Exmouth Gulf, Western Australia. Compounds10 and11 exhibited moderate differential activity as SR Ca2+ channel agonists (EC50 13.6 and 100 μM, respectively) of the Ry1 R FKBP12 complex, while the potency of9 was almost half that of10 (EC50 20.6 μM). The problem of dereplication of bastadins was addressed using 1H-NMR fingerprinting of MeO signals in the corresponding permethyl bastadin derivatives.- Published
- 1996
37. Sparks and puffs in oligodendrocyte progenitors: Cross talk between ryanodine receptors and inositol trisphosphate receptors
- Author
-
Haak, L. L., Long-Sheng Song, Molinski, T. F., Pessah, I. N., Cheng, H., and Russell, J. T.
38. Ryanodine Induces Maturation of Embryonic Acetylcholinesterase Forms in Cultured Quail Myotubes
- Author
-
Pessah, I. N., Nieberg, P. S., and Wilson, B. W.
- Published
- 1993
- Full Text
- View/download PDF
39. Non-dioxin-like polychlorinated biphenyl neurotoxic equivalents found in environmental and human samples.
- Author
-
Holland EB and Pessah IN
- Subjects
- Animals, Fishes, Humans, Neurotoxicity Syndromes diagnosis, Neurotoxicity Syndromes epidemiology, Polychlorinated Biphenyls analysis, United States epidemiology, Water Pollutants, Chemical analysis, Dioxins, Environmental Monitoring methods, Polychlorinated Biphenyls toxicity, United States Environmental Protection Agency, Water Pollutants, Chemical toxicity
- 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., (Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
40. Type 1 and type 3 ryanodine receptors generate different Ca(2+) release event activity in both intact and permeabilized myotubes.
- Author
-
Ward CW, Protasi F, Castillo D, Wang Y, Chen SR, Pessah IN, Allen PD, and Schneider MF
- Subjects
- 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, Calcium metabolism, Myocardium cytology, Potassium metabolism, Ryanodine Receptor Calcium Release Channel chemistry, Ryanodine Receptor Calcium Release Channel metabolism
- 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.
- Published
- 2001
- Full Text
- View/download PDF
41. Ryanodine receptor acts as a sensor for redox stress.
- Author
-
Pessah IN
- Subjects
- Biological Transport, Calcium Signaling, Cell Membrane physiology, Membrane Potentials, Oxidation-Reduction, Oxidative Stress, Calcium metabolism, Microsomes physiology, Ryanodine Receptor Calcium Release Channel metabolism
- 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.
- Published
- 2001
- Full Text
- View/download PDF
42. Sparks and puffs in oligodendrocyte progenitors: cross talk between ryanodine receptors and inositol trisphosphate receptors.
- Author
-
Haak LL, Song LS, Molinski TF, Pessah IN, Cheng H, and Russell JT
- Subjects
- 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, Calcium Channels metabolism, Calcium Signaling physiology, Receptor Cross-Talk physiology, Receptors, Cytoplasmic and Nuclear metabolism, Ryanodine Receptor Calcium Release Channel metabolism, Stem Cells metabolism
- 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.
- Published
- 2001
43. A bioactive metabolite of benzo[a]pyrene, benzo[a]pyrene-7,8-dione, selectively alters microsomal Ca2+ transport and ryanodine receptor function.
- Author
-
Pessah IN, Beltzner C, Burchiel SW, Sridhar G, Penning T, and Feng W
- Subjects
- 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, Benzo(a)pyrene pharmacology, Benzopyrenes pharmacology, Calcium metabolism, Microsomes drug effects, Ryanodine Receptor Calcium Release Channel physiology
- 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.
- Published
- 2001
- Full Text
- View/download PDF
44. ortho-substituted PCB95 alters intracellular calcium signaling and causes cellular acidification in PC12 cells by an immunophilin-dependent mechanism.
- Author
-
Wong PW, Garcia EF, and Pessah IN
- Subjects
- 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, Acidosis chemically induced, Calcium Signaling drug effects, Immunophilins metabolism, Pheochromocytoma metabolism, Polychlorinated Biphenyls pharmacology
- 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.
- Published
- 2001
- Full Text
- View/download PDF
45. Transmembrane redox sensor of ryanodine receptor complex.
- Author
-
Feng W, Liu G, Allen PD, and Pessah IN
- Subjects
- 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, Ion Channel Gating, Ryanodine Receptor Calcium Release Channel metabolism
- 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.
- Published
- 2000
- Full Text
- View/download PDF
46. Divergent functional properties of ryanodine receptor types 1 and 3 expressed in a myogenic cell line.
- Author
-
Fessenden JD, Wang Y, Moore RA, Chen SR, Allen PD, and Pessah IN
- Subjects
- 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, Muscle, Skeletal metabolism, Ryanodine Receptor Calcium Release Channel metabolism
- 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.
- Published
- 2000
- Full Text
- View/download PDF
47. Gating of store-operated channels by conformational coupling to ryanodine receptors.
- Author
-
Kiselyov KI, Shin DM, Wang Y, Pessah IN, Allen PD, and Muallem S
- Subjects
- 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, Calcium Channels physiology, Calcium Signaling, Ion Channel Gating physiology, Ryanodine Receptor Calcium Release Channel physiology
- 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.
- Published
- 2000
- Full Text
- View/download PDF
48. Characterization of [(3)H]ryanodine binding sites in mammalian lung.
- Author
-
Wild JS, Giri SN, Moore R, and Pessah IN
- Subjects
- 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, Lung metabolism, Ryanodine metabolism, Ryanodine Receptor Calcium Release Channel metabolism
- 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., (Copyright 2000 Academic Press.)
- Published
- 2000
- Full Text
- View/download PDF
49. Functional role of hyperreactive sulfhydryl moieties within the ryanodine receptor complex.
- Author
-
Pessah IN and Feng W
- Subjects
- 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, Calcium Signaling physiology, Carrier Proteins, Cystine metabolism, Muscle Proteins metabolism, Ryanodine Receptor Calcium Release Channel metabolism, Sarcoplasmic Reticulum metabolism, Sulfhydryl Compounds physiology
- 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.
- Published
- 2000
- Full Text
- View/download PDF
50. Site-selective modification of hyperreactive cysteines of ryanodine receptor complex by quinones.
- Author
-
Feng W, Liu G, Xia R, Abramson JJ, and Pessah IN
- Subjects
- 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, Carrier Proteins, Cysteine metabolism, Quinones pharmacology, Ryanodine Receptor Calcium Release Channel metabolism
- 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.
- Published
- 1999
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.