Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
ACS Chem Biol ; 18(10): 2290-2299, 2023 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-37769131

RESUMEN

Hyperactivity of cardiac sarcoplasmic reticulum (SR) ryanodine receptor (RyR2) Ca2+-release channels contributes to heart failure and arrhythmias. Reducing the RyR2 activity, particularly during cardiac relaxation (diastole), is a desirable therapeutic goal. We previously reported that the unnatural enantiomer (ent) of an insect-RyR activator, verticilide, inhibits porcine and mouse RyR2 at diastolic (nanomolar) Ca2+ and has in vivo efficacy against atrial and ventricular arrhythmia. To determine the ent-verticilide structural mode of action on RyR2 and guide its further development via medicinal chemistry structure-activity relationship studies, here, we used fluorescence lifetime (FLT)-measurements of Förster resonance energy transfer (FRET) in HEK293 cells expressing human RyR2. For these studies, we used an RyR-specific FRET molecular-toolkit and computational methods for trilateration (i.e., using distances to locate a point of interest). Multiexponential analysis of FLT-FRET measurements between four donor-labeled FKBP12.6 variants and acceptor-labeled ent-verticilide yielded distance relationships placing the acceptor probe at two candidate loci within the RyR2 cryo-EM map. One locus is within the Ry12 domain (at the corner periphery of the RyR2 tetrameric complex). The other locus is sandwiched at the interface between helical domain 1 and the SPRY3 domain. These findings document RyR2-target engagement by ent-verticilide, reveal new insight into the mechanism of action of this new class of RyR2-targeting drug candidate, and can serve as input in future computational determinations of the ent-verticilide binding site on RyR2 that will inform structure-activity studies for lead optimization.


Asunto(s)
Depsipéptidos , Canal Liberador de Calcio Receptor de Rianodina , Ratones , Porcinos , Humanos , Animales , Rianodina/química , Rianodina/metabolismo , Rianodina/uso terapéutico , Canal Liberador de Calcio Receptor de Rianodina/química , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Transferencia Resonante de Energía de Fluorescencia/métodos , Células HEK293 , Arritmias Cardíacas/tratamiento farmacológico , Arritmias Cardíacas/metabolismo , Depsipéptidos/metabolismo , Calcio/metabolismo , Miocitos Cardíacos/metabolismo
2.
J Biol Chem ; 295(46): 15622-15635, 2020 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-32878990

RESUMEN

Structural analyses identified the central domain of ryanodine receptor (RyR) as a transducer converting conformational changes in the cytoplasmic platform to the RyR gate. The central domain is also a regulatory hub encompassing the Ca2+-, ATP-, and caffeine-binding sites. However, the role of the central domain in RyR activation and regulation has yet to be defined. Here, we mutated five residues that form the Ca2+ activation site and 10 residues with negatively charged or oxygen-containing side chains near the Ca2+ activation site. We also generated eight disease-associated mutations within the central domain of RyR2. We determined the effect of these mutations on Ca2+, ATP, and caffeine activation and Mg2+ inhibition of RyR2. Mutating the Ca2+ activation site markedly reduced the sensitivity of RyR2 to Ca2+ and caffeine activation. Unexpectedly, Ca2+ activation site mutation E3848A substantially enhanced the Ca2+-independent basal activity of RyR2, suggesting that E3848A may also affect the stability of the closed state of RyR2. Mutations in the Ca2+ activation site also abolished the effect of ATP/caffeine on the Ca2+-independent basal activity, suggesting that the Ca2+ activation site is also a critical determinant of ATP/caffeine action. Mutating residues with negatively charged or oxygen-containing side chains near the Ca2+ activation site significantly altered Ca2+ and caffeine activation and reduced Mg2+ inhibition. Furthermore, disease-associated RyR2 mutations within the central domain significantly enhanced Ca2+ and caffeine activation and reduced Mg2+ inhibition. Our data demonstrate that the central domain plays an important role in channel activation, channel regulation, and closed state stability.


Asunto(s)
Miocardio/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Adenosina Trifosfato/farmacología , Sitios de Unión , Cafeína/farmacología , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Células HEK293 , Humanos , Magnesio/química , Magnesio/metabolismo , Simulación de Dinámica Molecular , Mutagénesis Sitio-Dirigida , Unión Proteica/efectos de los fármacos , Estabilidad Proteica , Estructura Terciaria de Proteína , Rianodina/química , Rianodina/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/química , Canal Liberador de Calcio Receptor de Rianodina/genética
3.
J Am Chem Soc ; 142(30): 12937-12941, 2020 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-32609506

RESUMEN

A stereoselective entry to ryanoids is described that culminates in the synthesis of anhydroryanodol and thus the formal total synthesis of ryanodol. The pathway described features an annulation reaction conceived to address the uniquely complex and highly oxygenated polycyclic skeleton common to members of this natural product class. It is demonstrated that metallacycle-mediated intramolecular coupling of an alkyne and a 1,3-diketone can proceed with a highly functionalized enyne and with outstanding levels of stereoselection. Furthermore, the first application of this technology in natural product synthesis is demonstrated here. More broadly, the advances described demonstrate the value that programs in natural product total synthesis have in advancing organic chemistry, here through the design and realization of an annulation reaction that accomplishes what previously established reactions do not.


Asunto(s)
Productos Biológicos/síntesis química , Rianodina/análogos & derivados , Productos Biológicos/química , Ciclización , Estructura Molecular , Rianodina/síntesis química , Rianodina/química , Estereoisomerismo
4.
Org Lett ; 21(15): 6126-6129, 2019 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-31298546

RESUMEN

An annulation reaction is described to access a range of polycyclic and highly oxygenated carbocycles. First developed in an approach to the synthesis of ryanodol, metallacycle-mediated annulative diketone-alkyne coupling defines a framework for realization of new retrosynthetic relationships for complex molecule synthesis. In addition to demonstrating this reaction in the context of forging distinct carbocyclic systems, including those featuring a seven-membered ring, the choice of quenching reagent leads to unique reaction outcomes.


Asunto(s)
Carbono/química , Rianodina/análogos & derivados , Alquinos/química , Ciclización , Cetonas/química , Oxígeno/química , Rianodina/química
5.
J Biol Chem ; 292(31): 12947-12958, 2017 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-28584051

RESUMEN

The type 1 ryanodine receptor (RyR1) mediates Ca2+ release from the sarcoplasmic reticulum to initiate skeletal muscle contraction and is associated with muscle diseases, malignant hyperthermia, and central core disease. To better understand RyR1 channel function, we investigated the molecular mechanisms of channel gating and ion permeation. An adequate model of channel gating requires accurate, high-resolution models of both open and closed states of the channel. To this end, we generated an open-channel RyR1 model using molecular simulations to pull Ca2+ through the pore constriction site of a closed-channel RyR1 structure determined at 3.8-Šresolution. Importantly, we find that our open-channel model is consistent with the RyR1 and cardiac RyR (RyR2) open-channel structures reported while this paper was in preparation. Both our model and the published structures show similar rotation of the upper portion of the pore-lining S6 helix away from the 4-fold channel axis and twisting of Ile-4937 at the channel constriction site out of the channel pore. These motions result in a minimum open-channel pore radius of ∼3 Šformed by Gln-4933, rather than Ile-4937 in the closed-channel structure. We also present functional support for our model by mutations around the closed- and open-channel constriction sites (Gln-4933 and Ile-4937). Our results indicate that use of ion-pulling simulations produces a RyR1 open-channel model, which can provide insights into the mechanisms of channel opening complementing those from the structural data.


Asunto(s)
Señalización del Calcio , Membrana Dobles de Lípidos/química , Modelos Moleculares , Canal Liberador de Calcio Receptor de Rianodina/química , Sustitución de Aminoácidos , Animales , Cafeína/química , Cafeína/metabolismo , Cafeína/farmacología , Agonistas de los Canales de Calcio/química , Agonistas de los Canales de Calcio/metabolismo , Agonistas de los Canales de Calcio/farmacología , Señalización del Calcio/efectos de los fármacos , Glutamina/química , Células HEK293 , Humanos , Isoleucina/química , Ligandos , Simulación de Dinámica Molecular , Mutación , Fragmentos de Péptidos/agonistas , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Conformación Proteica en Hélice alfa , Dominios y Motivos de Interacción de Proteínas , Conejos , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Rianodina/química , Rianodina/metabolismo , Rianodina/farmacología , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo
6.
Biophys J ; 112(8): 1645-1653, 2017 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-28445755

RESUMEN

Ryanodine (Ryd) irreversibly targets ryanodine receptors (RyRs), a family of intracellular calcium release channels essential for many cellular processes ranging from muscle contraction to learning and memory. Little is known of the atomistic details about how Ryd binds to RyRs. In this study, we used all-atom molecular dynamics simulations with both enhanced and bidirectional sampling to gain direct insights into how Ryd interacts with major residues in RyRs that were experimentally determined to be critical for its binding. We found that the pyrrolic ring of Ryd displays preference for the R4892AGGG-F4921 residues in the cavity of RyR1, which explain the effects of the corresponding mutations in RyR2 in experiments. Particularly, the mutant Q4933A (or Q4863A in RyR2) critical for both the gating and Ryd binding not only has significantly less interaction with Ryd than the wild-type, but also yields more space for Ryd and water molecules in the cavity. These results describe clear binding modes of Ryd in the RyR cavity and offer structural mechanisms explaining functional data collected on RyR blockade.


Asunto(s)
Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Rianodina/metabolismo , Animales , Sitios de Unión , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Mutación , Estructura Secundaria de Proteína , Rianodina/química , Canal Liberador de Calcio Receptor de Rianodina/química , Canal Liberador de Calcio Receptor de Rianodina/genética , Termodinámica , Agua/química
7.
Biophys J ; 111(11): 2387-2394, 2016 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-27926840

RESUMEN

We have used chemical synthesis, electron paramagnetic resonance (EPR), and circular dichroism to detect and analyze the structural dynamics of a ryanodine receptor (RyR) peptide bound to calmodulin (CaM). The skeletal muscle calcium release channel RyR1 is activated by Ca2+-free CaM and inhibited by Ca2+-bound CaM. To probe the structural mechanism for this regulation, wild-type RyRp and four spin-labeled derivatives were synthesized, each containing the nitroxide probe 2,2,6,6-tetramethyl-piperidine-1-oxyl-4-amino-4-carboxylic acid substituted for a single amino acid. In 2,2,6,6-tetramethyl-piperidine-1-oxyl-4-amino-4-carboxylic acid, the probe is rigidly and stereospecifically coupled to the α-carbon, enabling direct detection by EPR of peptide backbone structural dynamics. In the absence of CaM, circular dichroism indicates a complete lack of secondary structure, while 40% trifluoroethanol (TFE) induces >90% helicity and is unperturbed by the spin label. The EPR spectrum of each spin-labeled peptide indicates nanosecond dynamic disorder that is substantially reduced by TFE, but a significant gradient in dynamics is observed, decreasing from N- to C-terminus, both in the presence and absence of TFE. When bound to CaM, the probe nearest RyRp's N-terminus shows rapid rotational motion consistent with peptide backbone dynamics of a locally unfolded peptide, while the other three sites show substantial restriction of dynamics, consistent with helical folding. The two N-terminal sites, which bind to the C-lobe of CaM, do not show a significant Ca2+-dependence in mobility, while both C-terminal sites, which bind to the N-lobe of CaM, are significantly less mobile in the presence of bound Ca2+. These results support a model in which the interaction of RyR with CaM is nonuniform along the peptide, and the primary effect of Ca2+ is to increase the interaction of the C-terminal portion of the peptide with the N-terminal lobe of CaM. These results provide, to our knowledge, new insight into the Ca2+-dependent regulation of RyR by CaM.


Asunto(s)
Calcio/metabolismo , Calmodulina/metabolismo , Rianodina/química , Rianodina/metabolismo , Marcadores de Spin , Secuencia de Aminoácidos , Óxidos N-Cíclicos/química , Modelos Moleculares , Unión Proteica , Conformación Proteica en Hélice alfa
8.
Chem Pharm Bull (Tokyo) ; 64(7): 874-9, 2016 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-27010543

RESUMEN

Ryanodine (1) is a plant-derived natural product with powerful pharmacological and insecticidal action, and is a potent modulator of intracellular calcium release channels. Compound 1 possesses a 1H-pyrrole-2-carboxylate ester at the C3-position of heptahydroxylated terpenoid ryanodol (2). Whereas 2 was readily obtained from 1 by basic hydrolysis, 1 has never been synthesized from 2, due to the extreme difficulty in selectively introducing the bulky pyrrole moiety at the severely hindered C3-hydroxyl group of heptaol 2. Here we report chemical conversion of 2 to 1 for the first time. The derivatization was realized through the use of a new protective group strategy and the application of on-site construction of the pyrrole-2-carboxylate ester from the glycine ester and 1,3-bis(dimethylamino)allylium tetrafluoroborate.


Asunto(s)
Rianodina/análogos & derivados , Rianodina/síntesis química , Estructura Molecular , Rianodina/química
9.
Chemistry ; 22(1): 222-9, 2016 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-26616151

RESUMEN

Ryanodine (1) is a potent modulator of intracellular calcium release channels, designated as ryanodine receptors. The exceptionally complex molecular architecture of 1 comprises a highly oxygenated pentacyclic system with eleven contiguous stereogenic centers, which makes it a formidable target for organic synthesis. We identified the embedded C2 -symmetric tricyclic substructure within 1. This specific recognition permitted us to design a concise synthetic route to enantiopure tricycle 9 by utilizing a series of pairwise functionalizations. The four tetrasubstituted carbon centers of 9 were effectively constructed by three key reactions, a dearomatizing Diels-Alder reaction, the kinetic resolution of the obtained racemic 14 through asymmetric methanolysis, and the transannular aldol reaction of the eight-membered diketone 10. A new combination of cobalt-catalyzed hydroperoxidation and NfF-promoted elimination enabled conversion of the hindered olefin of 9 into the corresponding ketone, thus realizing the desymmetrization. Finally, the tetrasubstituted carbon was stereospecifically installed by utilizing the α-alkoxy bridgehead radical to deliver the core tetracycle 7 with the six contiguous tetrasubstituted carbon centers. Consequently, the present work not only accomplishes efficient assembly of four out of the five fused rings of 1, but also develops two new powerful methodologies: two-step ketone formation and bridgehead radical reaction.


Asunto(s)
Alquenos/química , Hidrocarburos Aromáticos con Puentes/química , Hidrocarburos Aromáticos con Puentes/síntesis química , Cetonas/química , Rianodina/química , Rianodina/síntesis química , Catálisis , Reacción de Cicloadición , Estructura Molecular , Oxidación-Reducción , Estereoisomerismo
10.
Chemistry ; 22(1): 230-6, 2016 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-26616265

RESUMEN

(+)-Ryanodine (1) is the ester derivative of 1H-pyrrole-2-carboxylic acid and the complex terpenoid (+)-ryanodol (2), which possesses eleven contiguous stereogenic centers on the ABCDE-ring system. Compound 1 is known to be a potent modulator of intracellular calcium release channels, whereas the activity of 2 is significantly weaker. To chemically construct 1, the multiple oxygen functional groups must be installed on the fused pentacycle in stereoselective fashions and the extremely hindered C3-hydroxy group must be acylated in a site-selective manner. First, the total synthesis of 2 was accomplished by introducing the five stereocenters from the previously prepared enantiopure ABDE-ring 7. Stereoselective construction of the C3-secondary, C2- and C6-tertiary alcohols was achieved by three nucleophilic reactions. The C9- and C10-trisubstituted carbon centers were regio- and stereoselectively introduced by hydroboration/oxidation of the six-membered C-ring, which was formed by the ring-closing metathesis reaction. Direct esterification of the C3-alcohol with pyrrole-2-carboxylic acid proved unsuccessful; therefore, we developed a new, two-step protocol for attachment of the pyrrole moiety. The C3-hydroxy group was first converted into the less sterically cumbersome glycine ester, which was then transformed into the pyrrole ring through condensation with 1,3-bis(dimethylamino)allylium tetrafluoroborate. This procedure resulted in the first total synthesis of 1.


Asunto(s)
Alcoholes/química , Pirroles/química , Rianodina/síntesis química , Estructura Molecular , Oxidación-Reducción , Rianodina/química , Estereoisomerismo
11.
Cell Calcium ; 58(2): 160-70, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25931303

RESUMEN

The earliest critical event of pancreatitis is a long lasting high amplitude rise of intracellular Ca(2+) concentration of the acinar cell, which can be triggered by high concentration of bile acids. Although, Ca(2+)-release through ryanodine receptors (RyR) is involved in the process, the significance and the exact mechanism of bile acid's action on RyR has not been fully elucidated yet. Therefore, we aimed to test with various techniques and aspects whether bile acids exert a direct effect on RyR and SERCA pump. Our data show that taurocholic acid (TCA)-induced Ca(2+) release in pancreatic acinar cells was significantly reduced by the RyR antagonist dantrolene. Further, we show that TCA enhanced RyR's (3)H-ryanodine binding and triggered robust Ca(2+)-release from RyR-enriched vesicles in the pathologically relevant concentration range. RyR single channel current analysis demonstrated that 200µM TCA induced a 5-fold increase in the channel's open probability and caused a significant lengthening of the mean open time. TCA also suppressed Ca(2+)-uptake rate and ATP-ase activity of SERCA-enriched vesicles, but interestingly, failed to decrease Ca(2+) elimination rate in intact cells. Overall, our results strongly suggest that TCA opens RyR by an allosteric mechanism, which contribute significantly to bile acid-induced pathologic Ca(2+)-leak from the endoplasmic reticulum in pancreatic acinar cells.


Asunto(s)
Células Acinares/metabolismo , Ácidos y Sales Biliares/farmacología , Páncreas Exocrino/citología , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Células Acinares/citología , Células Acinares/efectos de los fármacos , Animales , Calcio/metabolismo , Colagogos y Coleréticos/farmacología , Dantroleno/farmacología , Ratones , Microsomas/metabolismo , Ratas , Rianodina/química , Rianodina/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Ácido Taurocólico/farmacología
12.
PLoS One ; 9(10): e111804, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25350563

RESUMEN

In rat tail artery (RTA), spinal cord injury (SCI) increases nerve-evoked contractions and the contribution of L-type Ca2+ channels to these responses. In RTAs from unoperated rats, these channels play a minor role in contractions and Bay K8644 (L-type channel agonist) mimics the effects of SCI. Here we investigated the mechanisms underlying the facilitatory actions of SCI and Bay K8644 on nerve-evoked contractions of RTAs and the hypothesis that Ca2+ entering via L-type Ca2+ channels is rapidly sequestered by the sarcoplasmic reticulum (SR) limiting its role in contraction. In situ electrochemical detection of noradrenaline was used to assess if Bay K8644 increased noradrenaline release. Perforated patch recordings were used to assess if SCI changed the Ca2+ current recorded in RTA myocytes. Wire myography was used to assess if SCI modified the effects of Bay K8644 and of interrupting SR Ca2+ uptake on nerve-evoked contractions. Bay K8644 did not change noradrenaline-induced oxidation currents. Neither the size nor gating of Ca2+ currents differed between myocytes from sham-operated (control) and SCI rats. Bay K8644 increased nerve-evoked contractions in RTAs from both control and SCI rats, but the magnitude of this effect was reduced by SCI. By contrast, depleting SR Ca2+ stores with ryanodine or cyclopiazonic acid selectively increased nerve-evoked contractions in control RTAs. Cyclopiazonic acid also selectively increased the blockade of these responses by nifedipine (L-type channel blocker) in control RTAs, whereas ryanodine increased the blockade produced by nifedipine in both groups of RTAs. These findings suggest that Ca2+ entering via L-type channels is normally rapidly sequestered limiting its access to the contractile mechanism. Furthermore, the findings suggest SCI reduces the role of this mechanism.


Asunto(s)
Calcio/metabolismo , Citoplasma/metabolismo , Traumatismos de la Médula Espinal/patología , Ácido 3-piridinacarboxílico, 1,4-dihidro-2,6-dimetil-5-nitro-4-(2-(trifluorometil)fenil)-, Éster Metílico/química , Animales , Canales de Calcio/efectos de los fármacos , Canales de Calcio Tipo L/metabolismo , Electroquímica , Femenino , Indoles/química , Células Musculares/efectos de los fármacos , Contracción Muscular/efectos de los fármacos , Nifedipino/química , Norepinefrina/química , Ratas , Ratas Sprague-Dawley , Rianodina/química , Retículo Sarcoplasmático/metabolismo , Cola (estructura animal)/irrigación sanguínea
13.
Mol Pharmacol ; 86(3): 318-29, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25002270

RESUMEN

Ryanodine receptors (RyRs) are intracellular membrane channels playing key roles in many Ca(2+) signaling pathways and, as such, are emerging novel therapeutic and insecticidal targets. RyRs are so named because they bind the plant alkaloid ryanodine with high affinity and although it is established that ryanodine produces profound changes in all aspects of function, our understanding of the mechanisms underlying altered gating is minimal. We address this issue using detailed single-channel gating analysis, mathematical modeling, and energetic evaluation of state transitions establishing that, with ryanodine bound, the RyR pore adopts an extremely stable open conformation. We demonstrate that stability of this state is influenced by interaction of divalent cations with both activating and inhibitory cytosolic sites and, in the absence of activating Ca(2+), trans-membrane voltage. Comparison of the conformational stability of ryanodine- and Imperatoxin A-modified channels identifies significant differences in the mechanisms of action of these qualitatively similar ligands.


Asunto(s)
Canal Liberador de Calcio Receptor de Rianodina/fisiología , Calcio/metabolismo , Células HEK293 , Humanos , Activación del Canal Iónico , Cinética , Modelos Biológicos , Conformación Proteica , Rianodina/química , Canal Liberador de Calcio Receptor de Rianodina/química , Venenos de Escorpión/química , Termodinámica
14.
J Am Chem Soc ; 136(16): 5916-9, 2014 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-24708178

RESUMEN

Ryanodol (1) exists in nature in the form of the 1H-pyrrole-2-carboxylate ester derivative known as ryanodine, which is a potent modulator of the calcium release channel. The pentacyclic ABCDE-ring system of 1 is fabricated with eight oxy groups, three methyl groups, and one isopropyl group. All the eight tetrasubstituted stereocenters are concentrated within the 10-carbon ABDE framework. The total synthesis of this exceptionally complex molecule was achieved in 22 steps from the simple C2-symmetric tricycle 8. The synthetic route is based on installation of the seven stereogenic centers and formation of the four C-C bonds within the highly congested multicyclic format. The novel and flexible strategy developed here will enable the generation of chemical derivatives with different functional properties toward calcium release channels.


Asunto(s)
Rianodina/química , Rianodina/síntesis química , Canales de Calcio/metabolismo , Técnicas de Química Sintética , Oxidación-Reducción , Rianodina/farmacología
15.
Chem Res Toxicol ; 25(8): 1571-3, 2012 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-22856329

RESUMEN

Radiolabeled anthranilic diamide insecticide [N-C(3)H(3)]chlorantraniliprole was synthesized at high specific activity. It was compared with phthalic diamide insecticide flubendiamide and [(3)H]ryanodine in radioligand binding studies with house fly muscle membranes to provide the first direct evidence with a native insect ryanodine receptor that the major anthranilic and phthalic diamide insecticides bind at different allosterically coupled sites, i.e., there are three distinct Ca(2+)-release channel targets for insecticide action.


Asunto(s)
Benzamidas/metabolismo , Proteínas de Insectos/metabolismo , Insecticidas/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Rianodina/metabolismo , Sulfonas/metabolismo , ortoaminobenzoatos/metabolismo , Animales , Benzamidas/química , Sitios de Unión , Calcio/metabolismo , Carbono/química , Proteínas de Insectos/química , Insectos , Insecticidas/química , Nitrógeno/química , Unión Proteica , Rianodina/química , Canal Liberador de Calcio Receptor de Rianodina/química , Sulfonas/química , Tritio/química , ortoaminobenzoatos/química
16.
J Biol Chem ; 285(44): 34168-80, 2010 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-20610396

RESUMEN

Maurocalcine has been the first demonstrated animal toxin acting as a cell-penetrating peptide. Although it possesses competitive advantages, its use as a cell-penetrating peptide (CPP) requires that analogues be developed that lack its characteristic pharmacological activity on ryanodine-sensitive calcium channels without affecting its cell-penetrating and vector efficiencies. Here, we present the synthesis, three-dimensional (1)H NMR structure, and activity of D-maurocalcine. We demonstrate that it possesses all of the desired features for an excellent CPP: preserved structure, lack of pharmacological action, conserved vector properties, and absence of cell toxicity. This is the first report of a folded/oxidized animal toxin in its D-diastereomer conformation for use as a CPP. The protease resistance of this new peptide analogue, combined with its efficient cell penetration at concentrations devoid of cell toxicity, suggests that D-maurocalcine should be an excellent vector for in vivo applications.


Asunto(s)
Péptidos/química , Venenos de Escorpión/química , Animales , Células CHO , Canales de Calcio/química , Membrana Celular/metabolismo , Dicroismo Circular , Cricetinae , Cricetulus , Fluoresceínas/química , Espectroscopía de Resonancia Magnética/métodos , Microscopía Confocal/métodos , Péptido Hidrolasas/química , Rianodina/química , Venenos de Escorpión/farmacología , Sales de Tetrazolio/farmacología , Tiazoles/farmacología
17.
J Biol Chem ; 285(25): 19219-26, 2010 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-20404344

RESUMEN

The 12-kDa FK506-binding proteins (FKBP12 and FKBP12.6) are regulatory subunits of ryanodine receptor (RyR) Ca(2+) release channels. To investigate the structural basis of FKBP interactions with the RyR1 and RyR2 isoforms, we used site-directed fluorescent labeling of FKBP12.6, ligand binding measurements, and fluorescence resonance energy transfer (FRET). Single-cysteine substitutions were introduced at five positions distributed over the surface of FKBP12.6. Fluorescent labeling at position 14, 32, 49, or 85 did not affect high affinity binding to the RyR1. By comparison, fluorescent labeling at position 41 reduced the affinity of FKBP12.6 binding by 10-fold. Each of the five fluorescent FKBPs retained the ability to inhibit [(3)H]ryanodine binding to the RyR1, although the maximal extent of inhibition was reduced by half when the label was attached at position 32. The orientation of FKBP12.6 bound to the RyR1 and RyR2 was examined by measuring FRET from the different labeling positions on FKBP12.6 to an acceptor attached within the RyR calmodulin subunit. FRET was dependent on the position of fluorophore attachment on FKBP12.6; however, for any given position, the distance separating donors and acceptors bound to RyR1 versus RyR2 did not differ significantly. Our results show that FKBP12.6 binds to RyR1 and RyR2 in the same orientation and suggest new insights into the discrete structural domains responsible for channel binding and inhibition. FRET mapping of RyR-bound FKBP12.6 is consistent with the predictions of a previous cryoelectron microscopy study and strongly supports the proposed structural model.


Asunto(s)
Transferencia Resonante de Energía de Fluorescencia/métodos , Canal Liberador de Calcio Receptor de Rianodina/química , Proteínas de Unión a Tacrolimus/química , Animales , Calmodulina/química , Humanos , Inmunofilinas/química , Músculo Esquelético/metabolismo , Mutación , Miocardio/patología , Unión Proteica , Estructura Terciaria de Proteína , Rianodina/química , Retículo Sarcoplasmático/metabolismo , Porcinos
18.
Biophys J ; 97(7): 1907-16, 2009 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-19804721

RESUMEN

We have shown that physiological levels of Ca(2+)-calmodulin (Ca(2+)CaM; 50-100 nM) activate cardiac ryanodine receptors (RyR2) incorporated into bilayers and increase the frequency of Ca(2+) sparks and waves in cardiac cells. In contrast, it is well known that Ca(2+)CaM inhibits [(3)H]ryanodine binding to cardiac sarcoplasmic reticulum. Since the [(3)H]ryanodine binding technique does not reflect the effects of Ca(2+)CaM on RyR2 open probability (Po), we have investigated, using the reversible ryanoid, ryanodol, whether Ca(2+)CaM can directly influence the binding of ryanoids to single RyR2 channels independently of Po. We demonstrate that Ca(2+)CaM reduces the rate of ryanodol association to RyR2 without affecting the rate of dissociation. We also find that ryanodol-bound channels fluctuate between at least two distinct subconductance states, M(1) and M(2), in a voltage-dependent manner. Ca(2+)CaM significantly alters the equilibrium between these two states. The results suggest that Ca(2+)CaM binding to RyR2 causes a conformation change to regions of the channel that include the ryanoid binding site, thereby leading to a decrease in ryanoid association rate and modulation of gating within the ryanoid/RyR2 bound state. Our data provide a possible explanation for why the effects of Ca(2+)CaM at the single-channel level are not mirrored by [(3)H]ryanodine binding studies.


Asunto(s)
Calcio/metabolismo , Calmodulina/metabolismo , Activación del Canal Iónico , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Rianodina/metabolismo , Animales , Cinética , Miocardio/citología , Probabilidad , Unión Proteica , Rianodina/química , Tritio/química
19.
J Cell Mol Med ; 13(9B): 3082-90, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19298522

RESUMEN

3-iodothyronamine (T(1)AM) is a novel endogenous relative of thyroid hormone, able to interact with trace amine-associated receptors, a class of plasma membrane G protein-coupled receptors, and to produce a negative inotropic and chronotropic effect. In the isolated rat heart 20-25 microM T(1)AM decreased cardiac contractility, but oxygen consumption and glucose uptake were either unchanged or disproportionately high when compared to mechanical work. In adult rat cardiomyocytes acute exposure to 20 microM T(1)AM decreased the amplitude and duration of the calcium transient. In patch clamped cardiomyocytes sarcolemmal calcium current density was unchanged while current facilitation by membrane depolarization was abolished consistent with reduced sarcoplasmic reticulum (SR) calcium release. In addition, T(1)AM decreased transient outward current (I(to)) and I(K1) background current. SR studies involving 20 microM T(1)AM revealed a significant decrease in ryanodine binding due to reduced B(max), no significant change in the rate constant of calcium-induced calcium release, a significant increase in calcium leak measured under conditions promoting channel closure, and no effect on oxalate-supported calcium uptake. Based on these observations we conclude T(1)AM affects calcium and potassium homeostasis and suggest its negative inotropic action is due to a diminished pool of SR calcium as a result of increased diastolic leak through the ryanodine receptor, while increased action potential duration is accounted for by inhibition of I(to) and I(K1) currents.


Asunto(s)
Miocardio/metabolismo , Tironinas/farmacología , Animales , Calcio/metabolismo , Canales de Calcio/química , Membrana Celular/metabolismo , Electrofisiología , Homeostasis , Iones , Masculino , Miocitos Cardíacos/metabolismo , Técnicas de Placa-Clamp , Potasio/metabolismo , Ratas , Ratas Wistar , Receptores Acoplados a Proteínas G/metabolismo , Rianodina/química , Rianodina/farmacología , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/metabolismo
20.
Rapid Commun Mass Spectrom ; 23(8): 1139-46, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19280612

RESUMEN

An important aspect of preventive doping research is the rapid implementation of tests for emerging drugs with potential for misuse into routine doping control assays. New therapeutics of different classes such as PPARdelta-agonists (e.g. GW501516), ryanodine-calstabin-complex stabilizers (e.g. S-107 and JTV-519), and selective androgen receptor modulators (SARMs, e.g. S-40503) are currently used for the treatment of particular medical conditions such as metabolic syndrome, cardiac arrhythmia, debilitating diseases and osteoporosis, respectively. Due to their being at an early stage of clinical trials and the limited availability of data on the metabolism and possible renal elimination of the active drugs, the development of protocols for doping control analyses of plasma specimens could be an option for the detection of the circulating agents. The mass spectrometric fragmentation of four emerging drug candidates (GW501516, S-107, JTV-519, and S-40503) was elucidated by positive electrospray ionization and collision-induced dissociation using a high resolution/high accuracy mass spectrometer. A screening and confirmation procedure was established based on liquid chromatography/tandem mass spectrometry requiring a volume of 100 microL of plasma. Proteins were precipitated using acetonitrile, the specimens were centrifuged and the supernatant analyzed using a triple-quadrupole mass spectrometer employing multiple reaction monitoring of diagnostic ion transitions. The method was validated with regard to specificity, limits of detection (0.4-8.3 ng/mL), recoveries (72-98%), intraday and interday precisions (12-21%), and ion suppression/enhancement effects.


Asunto(s)
Doping en los Deportes/métodos , Quinolinas/sangre , Receptores Androgénicos/sangre , Rianodina/sangre , Proteínas de Unión a Tacrolimus/sangre , Tiazepinas/sangre , Tiazoles/sangre , Cromatografía Liquida/métodos , Humanos , Espectrometría de Masas/métodos , Estructura Molecular , Peso Molecular , Quinolinas/química , Receptores Androgénicos/química , Reproducibilidad de los Resultados , Rianodina/química , Sensibilidad y Especificidad , Espectrometría de Masa por Ionización de Electrospray/métodos , Proteínas de Unión a Tacrolimus/química , Espectrometría de Masas en Tándem/métodos , Tiazepinas/química , Tiazoles/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA