Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
Angew Chem Int Ed Engl ; 60(6): 3131-3137, 2021 02 08.
Article in English | MEDLINE | ID: mdl-33125829

ABSTRACT

1,4-Dihydropyridines (DHP), the most commonly used antihypertensives, function by inhibiting the L-type voltage-gated Ca2+ (Cav ) channels. DHP compounds exhibit chirality-specific antagonistic or agonistic effects. The structure of rabbit Cav 1.1 bound to an achiral drug nifedipine reveals the general binding mode for DHP drugs, but the molecular basis for chiral specificity remained elusive. Herein, we report five cryo-EM structures of nanodisc-embedded Cav 1.1 in the presence of the bestselling drug amlodipine, a DHP antagonist (R)-(+)-Bay K8644, and a titration of its agonistic enantiomer (S)-(-)-Bay K8644 at resolutions of 2.9-3.4 Å. The amlodipine-bound structure reveals the molecular basis for the high efficacy of the drug. All structures with the addition of the Bay K8644 enantiomers exhibit similar inactivated conformations, suggesting that (S)-(-)-Bay K8644, when acting as an agonist, is insufficient to lock the activated state of the channel for a prolonged duration.


Subject(s)
Calcium Channel Blockers/chemistry , Calcium Channels, L-Type/chemistry , Dihydropyridines/chemistry , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/chemistry , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/metabolism , Amlodipine/chemistry , Amlodipine/metabolism , Binding Sites , Calcium Channel Agonists/chemistry , Calcium Channel Agonists/metabolism , Calcium Channel Blockers/metabolism , Calcium Channels, L-Type/metabolism , Cryoelectron Microscopy , Dihydropyridines/metabolism , Molecular Dynamics Simulation , Nanostructures/chemistry , Protein Structure, Tertiary , Stereoisomerism
2.
Cardiovasc Drugs Ther ; 35(4): 719-732, 2021 08.
Article in English | MEDLINE | ID: mdl-33245463

ABSTRACT

PURPOSE: In the present study, the therapeutic efficacy of a selective BKCa channel opener (compound X) in the treatment of monocrotaline (MCT)-induced pulmonary arterial hypertension (PAH) was investigated. METHODS: PAH was induced in male Wistar rats by a single injection of MCT. After two weeks, the MCT-treated group was divided into two groups that were either treated with compound X or vehicle. Compound X was administered daily at 28 mg/kg. Electrocardiographic, echocardiographic, and haemodynamic analyses were performed; ex vivo evaluations of pulmonary artery reactivity, right ventricle (RV) and lung histology as well as expression levels of α and ß myosin heavy chain, brain natriuretic peptide, and cytokines (TNFα and IL10) in heart tissue were performed. RESULTS: Pulmonary artery rings of the PAH group showed a lower vasodilatation response to acetylcholine, suggesting endothelial dysfunction. Compound X promoted strong vasodilation in pulmonary artery rings of both control and MCT-induced PAH rats. The untreated hypertensive rats presented remodelling of pulmonary arterioles associated with increased resistance to pulmonary flow; increased systolic pressure, hypertrophy and fibrosis of the RV; prolongation of the QT and Tpeak-Tend intervals (evaluated during electrocardiogram); increased lung and liver weights; and autonomic imbalance with predominance of sympathetic activity. On the other hand, treatment with compound X reduced pulmonary vascular remodelling, pulmonary flow resistance and RV hypertrophy and afterload. CONCLUSION: The use of a selective and potent opener to activate the BKCa channels promoted improvement of haemodynamic parameters and consequent prevention of RV maladaptive remodelling in rats with MCT-induced PAH.


Subject(s)
Calcium Channel Agonists , Large-Conductance Calcium-Activated Potassium Channels , Pulmonary Arterial Hypertension , Quinolines/pharmacology , Vascular Resistance/drug effects , Vasoconstriction/drug effects , Vasodilation/drug effects , Animals , Calcium Channel Agonists/metabolism , Calcium Channel Agonists/pharmacokinetics , Disease Models, Animal , Large-Conductance Calcium-Activated Potassium Channels/agonists , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Pulmonary Arterial Hypertension/drug therapy , Pulmonary Arterial Hypertension/metabolism , Pulmonary Arterial Hypertension/physiopathology , Rats , Rats, Wistar , Treatment Outcome , Vascular Remodeling/drug effects , Ventricular Function, Right/drug effects
3.
Bioorg Med Chem ; 28(24): 115829, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33191084

ABSTRACT

To develop the novel ryanodine receptors (RyRs) insecticides, encouraged by our previous research work, a series of novel N-phenylpyrazole derivatives containing a polysubstituted phenyl ring scaffold were designed and synthesized. The bioassays results indicated that some title compounds exhibited excellent insecticidal activity. For oriental armyworm (Mythimna separata), compounds 7f, 7g, 7i and 7o at 0.5 mg L-1 displayed 100% larvicidal activity, and even at 0.1 mg L-1, 7o was 30% larvicidal activity, comparable to chlorantraniliprole (30%) and better than cyantraniliprole (10%). Compounds 7f and 7o had the median lethal concentrations (LC50) of 8.83 × 10-2 and 7.12 × 10-2 mg L-1, respectively, close to chlorantraniliprole (6.79 × 10-2 mg L-1). Additionally, for diamondback moth (Plutella xylostella), the larvicidal activity of compounds 7f and 7i were 90% and 70% at 0.01 mg L-1, respectively, better than chlorantraniliprole (50%) and cyantraniliprole (40%). More impressively, the LC50 value of 7f was 4.2 × 10-3 mg L-1, slightly lower than that of chlorantraniliprole (5.0 × 10-3 mg L-1). The molecular docking between compound 7f and RyRs of diamondback moth validated our molecular designation. Furthermore, the calcium imaging experiment explored the influence of compound 7o on the calcium homeostasis in the central neurons of the third larvae of oriental armyworm. The results of this study indicated that 7o is a potent novel lead targeting at RyRs.


Subject(s)
Calcium Channel Agonists/chemistry , Pyrazoles/chemistry , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Binding Sites , Calcium Channel Agonists/metabolism , Calcium Channel Agonists/pharmacology , Calcium Signaling/drug effects , Drug Design , Drug Evaluation, Preclinical , Insecticides/chemistry , Insecticides/metabolism , Insecticides/pharmacology , Larva/drug effects , Molecular Docking Simulation , Moths/drug effects , Moths/growth & development , Pyrazoles/metabolism , Pyrazoles/pharmacology , Ryanodine Receptor Calcium Release Channel/chemistry , Structure-Activity Relationship
4.
Eur J Pharmacol ; 870: 172873, 2020 Mar 05.
Article in English | MEDLINE | ID: mdl-31866408

ABSTRACT

Hydroxy-safflor yellow A (HSYA) can exert a variety of effects upon the vascular system. However, the underlying mechanisms are not clear. The present study is to investigate its vasodilating effect and the mechanisms. Wistar-Kyoto (WKY) rats and spontaneously hypertensive rats (SHR) were enrolled for studying effects of HSYA on blood pressure, vasodilation, intracellular Ca2+ transient and membrane ion channels. Vasodilation and intracellular Ca2+ transient were measured by using vasomotor assay and fluorescence imaging system, respectively. The effect of HSYA on the large conductance Ca2+ activated and voltage-gated potassium channel (BKCa channel) currents in rat mesentery artery and on L-type calcium channel (Ca-L) currents in HEK293cells expressed with Ca-L were investigated using patch clamp techniques. Blood pressure of SHR and WKY rats were concentration dependently reduced by HSYA with a larger effect of HSYA in SHR than that in WKY rats. The tension of mesenteric arteries induced by 3 µM phenylephrine was attenuated by HSYA (IC50 = 90.8 µΜ). Patch clamp study showed that HSYA could activate BKCa channels and suppress Ca-L channels in a concentration dependent manner. The results of calcium signaling assays indicated that HSYA could reduce the intracellular free Ca2+ level. These findings demonstrate that HSYA could activate BKCa channels and inhibit Ca-L channels and reduce intracellular free Ca2+ level, which are probably important for its vasodilatory effect.


Subject(s)
Calcium Channel Agonists/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/metabolism , Chalcone/analogs & derivatives , Potassium Channels, Voltage-Gated/agonists , Quinones/pharmacology , Animals , Blood Pressure/drug effects , Calcium Channel Agonists/metabolism , Calcium Channel Blockers/metabolism , Calcium Signaling , Chalcone/metabolism , Chalcone/pharmacology , HEK293 Cells , Humans , Male , Membrane Potentials/drug effects , Mesenteric Arteries/drug effects , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Patch-Clamp Techniques , Phenylephrine/metabolism , Quinones/metabolism , Rats, Inbred SHR , Rats, Inbred WKY , Vasodilation/drug effects
5.
Neurosci Lett ; 709: 134343, 2019 09 14.
Article in English | MEDLINE | ID: mdl-31279915

ABSTRACT

Calcium influx through N-methyl-D-aspartate receptors (NMDAR) and voltage-gated calcium channels (VGCC) play major roles in postsynaptic signaling mechanisms. NMDAR subunit GluN2B is phosphorylated at Ser1303. Phosphorylation at this site is a prominent event in cell culture systems as well as in vivo. However, the functional significance of phosphorylation at this site is not completely understood. In this study, we compared the effect of calcium signaling through NMDAR and VGCC on the phosphorylation status of GluN2B-Ser1303 in the rat in vivo. VGCC was activated by intraperitoneal (IP) injection of the activator, BayK8644 and NMDAR was activated by intracerebroventricular (ICV) injection of NMDA in separate experimental groups. We found that the level of phospho-GluN2B-Ser1303 in the cortex and in the hippocampus increased in response to activation of either channel. The effects could be prevented by prior ICV administration of the specific blockers of these channels such as MK-801 for NMDAR and nifedipine for VGCC. The effect was also blocked by pretreatment with ICV administration of KN-93 indicating that it is mediated through CaM kinase. Both during NMDAR activation and VGCC activation, cell survival associated signals such as phospho-AKT and phospho-CREB showed decrease, consistent with activation of cell death pathways during these treatments. We conclude that under in vivo conditions, calcium influx through either NMDAR or VGCC activates CaM kinase, which in turn phosphorylates GluN2B-Ser1303.


Subject(s)
Calcium Channel Agonists/metabolism , Calcium Channels, L-Type/metabolism , N-Methylaspartate/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/metabolism , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Animals , Calcium Channel Agonists/pharmacology , Male , N-Methylaspartate/pharmacology , Phosphorylation/drug effects , Phosphorylation/physiology , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/agonists
6.
Mol Pharmacol ; 94(3): 973-983, 2018 09.
Article in English | MEDLINE | ID: mdl-29980657

ABSTRACT

Nifedipine and FPL 64176 (FPL), which block and potentiate L-type voltage-gated Ca2+ channels, respectively, modulate Cav1.2 more potently than Cav1.3. To identify potential strategies for developing subtype-selective inhibitors, we investigated the role of divergent amino acid residues in transmembrane domains IIIS5 and the extracellular IIIS5-3P loop region in modulation of these channels by nifedipine and FPL. Insertion of the extracellular IIIS5-3P loop from Cav1.2 into Cav1.3 (Cav1.3+) reduced the IC50 of nifedipine from 289 to 101 nM, and substitution of S1100 with an A residue, as in Cav1.2, accounted for this difference. Substituting M1030 in IIIS5 to V in Cav1.3+ (Cav1.3+V) further reduced the IC50 of nifedipine to 42 nM. FPL increased current amplitude with an EC50 of 854 nM in Cav1.3, 103 nM in Cav1.2, and 99 nM in Cav1.3+V. In contrast to nifedipine block, substitution of M1030 to V in Cav1.3 had no effect on potency of FPL potentiation of current amplitude, but slowed deactivation in the presence and absence of 10 µM FPL. FPL had no effect on deactivation of Cav1.3/dihydropyridine-insensitive (DHPi), a channel with very low sensitivity to nifedipine block (IC50 ∼93 µM), but did shift the voltage-dependence of activation by ∼-10 mV. We conclude that the M/V variation in IIIS5 and the S/A variation in the IIIS5-3P loop of Cav1.2 and Cav1.3 largely determine the difference in nifedipine potency between these two channels, but the difference in FPL potency is determined by divergent amino acids in the IIIS5-3P loop.


Subject(s)
Calcium Channel Agonists/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/physiology , Nifedipine/pharmacology , Pyrroles/pharmacology , Amino Acid Sequence , Calcium Channel Agonists/metabolism , Calcium Channel Blockers/metabolism , Calcium Channels, L-Type/chemistry , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Nifedipine/metabolism , Protein Structure, Secondary , Pyrroles/metabolism
7.
Bull Exp Biol Med ; 165(2): 272-275, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29923011

ABSTRACT

Store-operated channels activated in response to intracellular calcium store depletion represent the main pathway of calcium entry from the extracellular space in nonelectroexcitable cells. Adapter proteins organize the components of this system into integral complex. We studied the influence of adapter proteins of the Homer family on endogenous store-operated calcium Imin channels in A431 cells. Monomeric Homer 1a proteins increase activity of Imin channels, but did not modulate their electrophysiological properties. Recombinant Homer 1c protein did not block the induced calcium currents.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Homer Scaffolding Proteins/physiology , Action Potentials/drug effects , Calcium Channel Agonists/metabolism , Calcium Channel Agonists/pharmacology , Calcium Channels/drug effects , Calcium Channels/physiology , Calcium Signaling/drug effects , Cytoplasm/metabolism , Electrophysiological Phenomena/drug effects , Homer Scaffolding Proteins/pharmacology , Humans , Ion Channel Gating/drug effects , Patch-Clamp Techniques , Protein Multimerization/physiology , Recombinant Proteins/pharmacology , Tumor Cells, Cultured
8.
J Biol Chem ; 292(31): 12947-12958, 2017 08 04.
Article in English | MEDLINE | ID: mdl-28584051

ABSTRACT

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.


Subject(s)
Calcium Signaling , Lipid Bilayers/chemistry , Models, Molecular , Ryanodine Receptor Calcium Release Channel/chemistry , Amino Acid Substitution , Animals , Caffeine/chemistry , Caffeine/metabolism , Caffeine/pharmacology , Calcium Channel Agonists/chemistry , Calcium Channel Agonists/metabolism , Calcium Channel Agonists/pharmacology , Calcium Signaling/drug effects , Glutamine/chemistry , HEK293 Cells , Humans , Isoleucine/chemistry , Ligands , Molecular Dynamics Simulation , Mutation , Peptide Fragments/agonists , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , Rabbits , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Ryanodine/chemistry , Ryanodine/metabolism , Ryanodine/pharmacology , Ryanodine Receptor Calcium Release Channel/genetics , Ryanodine Receptor Calcium Release Channel/metabolism
9.
Biophys J ; 112(10): 2127-2137, 2017 May 23.
Article in English | MEDLINE | ID: mdl-28538150

ABSTRACT

Ca2+ regulates ryanodine receptor's (RyR) activity through an activating and an inhibiting Ca2+-binding site located on the cytoplasmic side of the RyR channel. Their altered sensitivity plays an important role in the pathology of malignant hyperthermia and heart failure. We used lanthanide ions (Ln3+) as probes to investigate the Ca2+ sensors of RyR, because they specifically bind to Ca2+-binding proteins and they are impermeable to the channel. Eu3+'s and Sm3+'s action was tested on single RyR1 channels reconstituted into planar lipid bilayers. When the activating binding site was saturated by 50 µM Ca2+, Ln3+ potently inhibited RyR's open probability (Kd Eu3+ = 167 ± 5 nM and Kd Sm3+ = 63 ± 3 nM), but in nominally 0 [Ca2+], low [Eu3+] activated the channel. These results suggest that Ln3+ acts as an agonist of both Ca2+-binding sites. More importantly, the voltage-dependent characteristics of Ln3+'s action led to the conclusion that the activating Ca2+ binding site is located within the electrical field of the channel (in the vestibule). This idea was tested by applying the pore blocker toxin maurocalcine on the cytoplasmic side of RyR. These experiments showed that RyR lost reactivity to changing cytosolic [Ca2+] from 50 µM to 100 nM when the toxin occupied the vestibule. These results suggest that maurocalcine mechanically prevented Ca2+ from dissociating from its binding site and support our vestibular Ca2+ sensor-model further.


Subject(s)
Calcium/metabolism , Lanthanoid Series Elements/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Binding Sites , Calcium/chemistry , Calcium Channel Agonists/chemistry , Calcium Channel Agonists/metabolism , Calcium Channel Agonists/pharmacology , Calcium Channel Blockers/chemistry , Calcium Channel Blockers/pharmacology , Cations/chemistry , Cations/metabolism , Cytosol/chemistry , Cytosol/metabolism , Dose-Response Relationship, Drug , Lanthanoid Series Elements/chemistry , Lipid Bilayers/chemistry , Membrane Potentials/drug effects , Membrane Potentials/physiology , Microscopy, Electron , Microsomes/chemistry , Microsomes/metabolism , Models, Molecular , Rabbits , Ryanodine Receptor Calcium Release Channel/chemistry , Sarcoplasmic Reticulum/chemistry , Sarcoplasmic Reticulum/metabolism , Scorpion Venoms/pharmacology
10.
J Agric Food Chem ; 64(18): 3697-704, 2016 May 11.
Article in English | MEDLINE | ID: mdl-27109555

ABSTRACT

To discover potent insecticides targeting ryanodine receptors (RyRs), a series of novel anthranilic diamides analogues (12a-12u) containing N-substituted phenylpyrazole were designed and synthesized. These compounds were characterized by (1)H NMR, (13)C NMR, and HRMS, and the structure of compound 12u was confirmed by X-ray diffraction. Their insecticidal activities indicated that these compounds displayed moderate to excellent activities. In particular, 12i showed 100 and 37% larvicidal activities against oriental armyworm (Mythimna separata) at 0.25 and 0.05 mg L(-1), equivalent to that of chlorantraniliprole (100%, 0.25 mg L(-1); and 33%, 0.05 mg L(-1)). The activity of 12i against diamondback moth (Plutella xylostella) was 95% at 0.05 mg L(-1), whereas the control was 100% at 0.05 mg L(-1). The calcium-imaging technique experiment results showed that the effects of 12i on the intracellular calcium ion concentration ([Ca(2+)]i) in neurons were concentration-dependent. After the central neurons of Helicoverpa armigera were dyed by loading with fluo-5N and treated with 12i, the free calcium released in endoplasmic reticulum indicated the target of compound 12i is RyRs or IP3Rs. The activation of RyRs by natural ryanodine completely blocked the calcium release induced by 12i, which indicated that RyRs in the central neurons of H. armigera third-instar larvae is the possible target of compound 12i.


Subject(s)
Calcium Channel Agonists/chemical synthesis , Insecticides/chemistry , Isoxazoles/chemistry , Ryanodine Receptor Calcium Release Channel/chemistry , Animals , Calcium Channel Agonists/chemistry , Calcium Channel Agonists/metabolism , Diamide , Drug Design , Insect Proteins/antagonists & inhibitors , Insect Proteins/chemistry , Insect Proteins/metabolism , Insecticides/chemical synthesis , Insecticides/pharmacology , Larva/drug effects , Larva/growth & development , Molecular Structure , Moths/drug effects , Moths/growth & development , Ryanodine Receptor Calcium Release Channel/metabolism , Structure-Activity Relationship , X-Ray Diffraction
11.
Nat Commun ; 5: 3897, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24941892

ABSTRACT

Cav1.2 and Cav1.3 are the main L-type Ca(2+) channel subtypes in the brain. Cav1.3 channels have recently been implicated in the pathogenesis of Parkinson's disease. Therefore, Cav1.3-selective blockers are developed as promising neuroprotective drugs. We studied the pharmacological properties of a pyrimidine-2,4,6-trione derivative (1-(3-chlorophenethyl)-3-cyclopentylpyrimidine-2,4,6-(1H,3H,5H)-trione, Cp8) recently reported as the first highly selective Cav1.3 blocker. Here we show, in contrast to this previous study, that Cp8 reproducibly increases inward Ca(2+) currents of Cav1.3 and Cav1.2 channels expressed in tsA-201 cells by slowing activation, inactivation and enhancement of tail currents. Similar effects are also observed for native Cav1.3 and Cav1.2 channels in mouse chromaffin cells, while non-L-type currents are unaffected. Evidence for a weak and non-selective inhibition of Cav1.3 and Cav1.2 currents is only observed in a minority of cells using Ba(2+) as charge carrier. Therefore, our data identify pyrimidine-2,4,6-triones as Ca(2+) channel activators.


Subject(s)
Barbiturates/metabolism , Calcium Channel Agonists/metabolism , Calcium Channels, L-Type/metabolism , Animals , Barbiturates/chemistry , Calcium Channel Agonists/chemistry , Calcium Channels, L-Type/chemistry , Calcium Channels, L-Type/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
12.
Toxicon ; 85: 5-16, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24751366

ABSTRACT

Crotoxin (Crtx), the main toxin in the venom of Crotalus durissus terrificus snake, is a heterodimer with a basic subunit, CB, and an acidic subunit, CA. CB is a phospholipase A2 that depends on CA to specifically bind to the cell membrane. This toxin acts in the central nervous system (CNS) causing chronic seizure effects and other cytotoxic effects. Here, we report its action on glutamate release in rat cerebral cortex synaptosomes. Aiming at a better understanding of the mechanism of action of Crtx, calcium channel blockers were used and internalization studies were performed in cerebellar granule neurons. Our results show that Crtx induces calcium-dependent glutamate release via N and P/Q calcium channels. In addition, the CB subunit of Crtx is shown to be internalized. This internalization does not depend on the presence of CA subunit neither on the PLA2 activity of CB. A correlation between CB internalization and glutamate release remains to be established.


Subject(s)
Calcium Channel Agonists/pharmacology , Calcium Channels, N-Type/metabolism , Cerebral Cortex/drug effects , Crotalid Venoms/chemistry , Crotalus , Crotoxin/pharmacology , Synaptosomes/drug effects , Animals , Calcium Channel Agonists/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels, N-Type/chemistry , Calcium Signaling/drug effects , Cells, Cultured , Cerebral Cortex/metabolism , Crotalid Venoms/enzymology , Crotoxin/antagonists & inhibitors , Crotoxin/metabolism , Glutamic Acid/metabolism , Group II Phospholipases A2/antagonists & inhibitors , Group II Phospholipases A2/metabolism , Group II Phospholipases A2/pharmacology , Male , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Neurotoxins/antagonists & inhibitors , Neurotoxins/metabolism , Neurotoxins/pharmacology , Protein Subunits/antagonists & inhibitors , Protein Subunits/metabolism , Protein Subunits/pharmacology , Protein Transport/drug effects , Rats, Wistar , Reptilian Proteins/antagonists & inhibitors , Reptilian Proteins/metabolism , Reptilian Proteins/pharmacology , Synaptic Transmission/drug effects , Synaptosomes/metabolism
13.
J Biotechnol ; 167(3): 248-54, 2013 Sep 10.
Article in English | MEDLINE | ID: mdl-23830902

ABSTRACT

Inositol 1,4,5-trisphosphate (IP3) receptors consist of three subtypes: IP3R1, IP3R2, and IP3R3. Although numerous IP3 receptor ligands have been synthesized, none of the subtype-selective ligands are known. We have developed a simple fluorescence method to examine the subtype selectivity of IP3 receptor ligands using FRET-based IP3 biosensors LIBRAvI, LIBRAvII, and LIBRAvIII. The addition of IP3 or adenophostin A (ADA) to permeabilized biosensor-expressing cells increased the fluorescence ratios of these biosensors in a concentration-dependent manner, and the potency of ADA relative to that of IP3 in terms of the changes in the fluorescence ratios of LIBRAvI, LIBRAvII, and LIBRAvIII was 43-, 22-, and 28-fold, respectively. This fluorescence-based method further showed that several ADA analogs had significant differences with respect to subtype selectivity and potency. These results highlight the important role played by the O-glycosidic structure of ADA in the selectivity of the ligands for IP3R1, as evidenced by the modified selectivity following replacement of the 5'-hydroxyl with a phenyl or phenethyl group. We also found that one ADA analog 5'-deoxy-5'-phenyladenophostin A possessed a partial agonistic effect on IP3R1. Together, the novel fluorescent methods described herein are useful for the evaluation of properties of IP3R ligands, including potency, efficacy, and subtype selectivity.


Subject(s)
Biosensing Techniques/methods , Calcium Channel Agonists/metabolism , Fluorescence Resonance Energy Transfer/methods , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Adenosine/analogs & derivatives , Calcium Channel Agonists/analysis , Cell Line , Humans , Inositol 1,4,5-Trisphosphate Receptors/agonists , Ligands
14.
Pflugers Arch ; 464(5): 425-58, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23001121

ABSTRACT

The Transient Receptor Potential Ankyrin 1 channel (TRPA1), is a member of the large TRP family of ion channels, and functions as a Ca(2+) permeable non-selective cation channel in many different cell processes, ranging from sensory to homeostatic tasks. TRPA1 is highly conserved across the animal kingdom. The only mammalian TRPA subfamily member, TRPA1, is widely expressed in neuronal (e.g. sensory dorsal root and trigeminal ganglia neurons)- and in non-neuronal cells (e.g. epithelial cells, hair cells). It exhibits 14-19 amino-(N-)terminal ankyrin repeats, an unusual structural feature. The TRPA1 channel is activated by noxious cold (<17 °C) as well as by a plethora of chemical compounds that includes not only electrophilic compounds and oxidants that can modify, in an alkylative or oxidative fashion, nucleophilic cysteine residues in the channel's N-terminus, but also compounds that do not covalently bind to the channel proteins (e.g. menthol, nifedipin). Based on localization and functional properties, TRPA1 is considered a key player in acute and chronic (neuropathic) pain and inflammation. Moreover, its role in the (patho)physiology of nearly all organ systems is anticipated, and will be discussed along with the potential of TRPA1 as a drug target for the management of various pathological conditions.


Subject(s)
Transient Receptor Potential Channels/genetics , Transient Receptor Potential Channels/physiology , Animals , Calcium Channel Agonists/metabolism , Channelopathies/genetics , Humans , Inflammation/metabolism , Ion Channel Gating , Nociception , Pain/metabolism , Phylogeny , Protein Structure, Tertiary , TRPA1 Cation Channel , Transient Receptor Potential Channels/chemistry , Transient Receptor Potential Channels/metabolism
15.
J Environ Pathol Toxicol Oncol ; 31(3): 285-93, 2012.
Article in English | MEDLINE | ID: mdl-23339702

ABSTRACT

Polycystic kidney disease (PKD) is associated with alterations in developmental processes that severely affect kidney integrity, often leading to fatal consequences. It has been suggested that dysfunctional calcium (Ca2+) regulation associated with the PKD phenotype is consequent to mutations affecting the pkd1 gene. Previously, it has been observed that blocking calcium along with cAMP allowed tubular epithelial cells to enter the proliferative phase that culminated in a cyst-like phenotype. In this regard, mouse metanephroi, (embryonic day 13.5, E13.5) were used to study morphological and ultrastructural effects of calcium replenishment on 8-bromocyclic 3'5'cyclic adenosine monophosphate (8-Br-cAMP)-induced cyst-like tubular dilations. Phase contrast microscopy of 8-Br-cAMP-treated metanephroi exhibited numerous dilated tubules that continued to increase in size for 4 days in culture. The effects of 8-Br-cAMP on renal tubular epithelia were assessed by histopathological and electron microscopic analyses. Transmission electron microscopy revealed changes such as increased vacuolation, swollen mitochondria, chromatin condensation, and disrupted cell membrane in tubular epithelia of 8-Br-cAMP-treated metanephroi. Concurrent treatments with calcium-channel agonists (calcium ionophore A23187 and phorbol-12-myristate-13-acetate) and 8-Br-cAMP abolished cAMP-induced morphometric and ultrastructural alterations. Calcium replenishment rescued tubular epithelial cells from mitogenic effects of cAMP and restored normal morphology at cellular and sub-cellular levels as verified by histopathological and ultrastructural examinations.


Subject(s)
8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Calcimycin/metabolism , Calcium Channel Agonists/metabolism , Calcium/metabolism , Kidney/drug effects , Polycystic Kidney Diseases/embryology , Tetradecanoylphorbol Acetate/metabolism , Animals , Kidney/embryology , Mice , Mice, Inbred BALB C , Microscopy, Electron, Transmission , Microscopy, Phase-Contrast , Organ Culture Techniques , Polycystic Kidney Diseases/physiopathology
16.
Nephrol Ther ; 7(2): 118-38, 2011 Apr.
Article in French | MEDLINE | ID: mdl-21273150

ABSTRACT

Calcium and phosphate play a key role in bone mineralization but have also many other physiological functions. The control of serum phosphate concentration is mandatory to avoid the occurrence of severe metabolic disorders, but is less tightly regulated than serum ionized calcium concentration, which is maintained in a very limited range thanks to parathyroid hormone (PTH) and the active vitamin D metabolite calcitriol. Any change in serum ionized calcium concentration is detected by the calcium sensing receptor (CaSR), a membranous protein located principally in the parathyroid glands and the kidney. A decrease in ionized calcium level inactivates the CaSR, thus stimulating PTH secretion. PTH in turn stimulates the release of calcium and phosphate from bone, renal calcium reabsorption and calcium and phosphate intestinal absorption by inducing renal calcitriol production. Moreover, PTH inhibits phosphate reabsorption in proximal tubular cells, thus contributing towards phosphate homeostasis. Fibroblast growth factor 23 (FGF23) is a circulating factor that decreases serum levels of inorganic phosphate by inhibiting renal phosphate reabsorption and calcitriol production and may have a great physiological role in phosphate homeostasis. Recently, vitamin D actions independent of calcium and phosphate homeostasis were discovered. Basal exploration of phosphocalcic metabolism abnormalities consists in measurement of serum calcium (ionized calcium if possible), phosphate, 25-hydroxy vitamine D and PTH and of 24 hours urinary calcium excretion as well as renal function. Hence, the understanding of physiopathological mechanisms has been improved by newly identified genetic disorders responsible for phophocalcic homeostasis disturbances.


Subject(s)
Calcitriol/metabolism , Calcium Channel Agonists/metabolism , Calcium/metabolism , Metabolic Diseases/prevention & control , Parathyroid Hormone/metabolism , Phosphates/metabolism , Bone and Bones/metabolism , Calcitriol/blood , Calcium/blood , Calcium Channel Agonists/blood , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/metabolism , Homeostasis , Humans , Kidney/metabolism , Kidney Failure, Chronic/metabolism , Kidney Failure, Chronic/therapy , Metabolic Diseases/metabolism , Parathyroid Glands/metabolism , Parathyroid Hormone/blood , Phosphates/blood , Receptors, Calcium-Sensing/metabolism
17.
Am J Physiol Cell Physiol ; 300(4): C937-49, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21228322

ABSTRACT

L-type voltage-gated calcium channels (LTCCs) have long been considered as crucial regulators of neuronal excitability. This role is thought to rely largely on coupling of LTCC-mediated Ca(2+) influx to Ca(2+)-dependent conductances, namely Ca(2+)-dependent K(+) (K(Ca)) channels and nonspecific cation (CAN) channels, which mediate afterhyperpolarizations (AHPs) and afterdepolarizations (ADPs), respectively. However, in which manner LTCCs, K(Ca) channels, and CAN channels co-operate remained scarcely known. In this study, we examined how activation of LTCCs affects neuronal depolarizations and analyzed the contribution of Ca(2+)-dependent potassium- and cation-conductances. With the use of hippocampal neurons in primary culture, pulsed current-injections were applied in the presence of tetrodotoxin (TTX) for stepwise depolarization and the availability of LTCCs was modulated by BAY K 8644 and isradipine. By varying pulse length and current strength, we found that weak depolarizing stimuli tend to be enhanced by LTCC activation, whereas in the course of stronger depolarizations LTCCs counteract excitation. Both effect modes appear to involve the same channels that mediate ADP and AHP, respectively. Indeed, ADPs were activated at lower stimulation levels than AHPs. In the absence of TTX, activation of LTCCs prolonged or shortened burst firing, depending on the initial burst duration, and invariably augmented brief unprovoked (such as excitatory postsynaptic potentials) and provoked electrical events. Hence, regulation of membrane excitability by LTCCs involves synchronous activity of both excitatory and inhibitory Ca(2+)-activated ion channels. The overall enhancing or dampening effect of LTCC stimulation on excitability does not only depend on the relative abundance of the respective coupling partner but also on the stimulus intensity.


Subject(s)
Calcium Channels, L-Type/metabolism , Membrane Potentials/physiology , Neurons/metabolism , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/metabolism , Action Potentials/physiology , Animals , Apamin/metabolism , Calcium/metabolism , Calcium Channel Agonists/metabolism , Calcium Channel Blockers/metabolism , Cells, Cultured , Hippocampus/cytology , Isradipine/metabolism , Neurons/cytology , Neurons/drug effects , Patch-Clamp Techniques , Potassium/metabolism , Rats , Rats, Sprague-Dawley , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology
18.
Br J Pharmacol ; 161(5): 1070-85, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20977457

ABSTRACT

BACKGROUND AND PURPOSE: Adenophostin A (AdA) is a potent agonist of inositol 1,4,5-trisphosphate receptors (IP(3) R). AdA shares with IP(3) the essential features of all IP(3) R agonists, namely structures equivalent to the 4,5-bisphosphate and 6-hydroxyl of IP(3) , but the basis of its increased affinity is unclear. Hitherto, the 2'-phosphate of AdA has been thought to provide a supra-optimal mimic of the 1-phosphate of IP(3) . EXPERIMENTAL APPROACH: We examined the structural determinants of AdA binding to type 1 IP(3) R (IP(3) R1). Chemical synthesis and mutational analysis of IP(3) R1 were combined with (3) H-IP(3) binding to full-length IP(3) R1 and its N-terminal fragments, and Ca(2+) release assays from recombinant IP(3) R1 expressed in DT40 cells. KEY RESULTS: Adenophostin A is at least 12-fold more potent than IP(3) in functional assays, and the IP(3) -binding core (IBC, residues 224-604 of IP(3) R1) is sufficient for this high-affinity binding of AdA. Removal of the 2'-phosphate from AdA (to give 2'-dephospho-AdA) had significantly lesser effects on its affinity for the IBC than did removal of the 1-phosphate from IP(3) (to give inositol 4,5-bisphosphate). Mutation of the only residue (R568) that interacts directly with the 1-phosphate of IP(3) decreased similarly (by ~30-fold) the affinity for IP(3) and AdA, but mutating R504, which has been proposed to form a cation-π interaction with the adenine of AdA, more profoundly reduced the affinity of IP(3) R for AdA (353-fold) than for IP(3) (13-fold). CONCLUSIONS AND IMPLICATIONS: The 2'-phosphate of AdA is not a major determinant of its high affinity. R504 in the receptor, most likely via a cation-π interaction, contributes specifically to AdA binding.


Subject(s)
Adenosine/analogs & derivatives , Calcium Channel Agonists/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Inositol 1,4,5-Trisphosphate/metabolism , Adenosine/chemistry , Adenosine/metabolism , Animals , Calcium/metabolism , Calcium Channel Agonists/chemistry , Inositol 1,4,5-Trisphosphate Receptors/agonists , Inositol 1,4,5-Trisphosphate Receptors/genetics , Male , Mutation , Protein Binding , Rats , Rats, Wistar
19.
J Appl Physiol (1985) ; 109(3): 830-9, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20576841

ABSTRACT

Skeletal muscle weakness is a reported ailment in individuals working in commercial hog confinement facilities. To date, specific mechanisms responsible for this symptom remain undefined. The purpose of this study was to assess whether hog barn dust (HBD) contains components that are capable of binding to and modulating the activity of type 1 ryanodine receptor Ca2+-release channel (RyR1), a key regulator of skeletal muscle function. HBD collected from confinement facilities in Nebraska were extracted with chloroform, filtered, and rotary evaporated to dryness. Residues were resuspended in hexane-chloroform (20:1) and precipitates, referred to as HBDorg, were air-dried and studied further. In competition assays, HBDorg dose-dependently displaced [3H]ryanodine from binding sites on RyR1 with an IC50 of 1.5±0.1 microg/ml (Ki=0.4±0.0 microg/ml). In single-channel assays using RyR1 reconstituted into a lipid bilayer, HBDorg exhibited three distinct dose-dependent effects: first it increased the open probability of RyR1 by increasing its gating frequency and dwell time in the open state, then it induced a state of reduced conductance (55% of maximum) that was more likely to occur and persist at positive holding potentials, and finally it irreversibly closed RyR1. In differentiated C2C12 myotubes, addition of HBD triggered a rise in intracellular Ca2+ that was blocked by pretreatment with ryanodine. Since persistent activation and/or closure of RyR1 results in skeletal muscle weakness, these new data suggest that HBD is responsible, at least in part, for the muscle ailment reported by hog confinement workers.


Subject(s)
Calcium Channel Agonists/pharmacology , Calcium Channel Blockers/pharmacology , Chloroform/chemistry , Dust/analysis , Housing, Animal , Ion Channel Gating/drug effects , Muscle, Skeletal/drug effects , Ryanodine Receptor Calcium Release Channel/drug effects , Solvents/chemistry , Agricultural Workers' Diseases/chemically induced , Animals , Binding Sites , Binding, Competitive , Calcium/metabolism , Calcium Channel Agonists/adverse effects , Calcium Channel Agonists/isolation & purification , Calcium Channel Agonists/metabolism , Calcium Channel Blockers/adverse effects , Calcium Channel Blockers/isolation & purification , Calcium Channel Blockers/metabolism , Cell Line , Chromatography, Thin Layer , Dose-Response Relationship, Drug , Humans , Membrane Potentials , Mice , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Weakness/chemically induced , Muscle, Skeletal/metabolism , Occupational Health , Rabbits , Risk Assessment , Ryanodine/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Sus scrofa
20.
Mol Cell Endocrinol ; 315(1-2): 308-13, 2010 Feb 05.
Article in English | MEDLINE | ID: mdl-19815053

ABSTRACT

Activation of GPR40 is reported to enhance insulin secretion in the presence of glucose. We determined whether sulfonylureas could replace glucose for GPR40-mediated enhancement of insulin secretion and investigated underlying mechanisms using INS-1E cells. GW9508, a specific agonist of GPR40, significantly enhanced insulin secretion in the presence of high concentrations of glucose. In contrast, sulfonylureas increased insulin secretion in the absence of glucose. In the presence of sulfonylureas, activation of GPR40 significantly enhanced insulin secretion. The L-type calcium channel (LTCC) activator S-(-)-Bay K8644 also concentration-dependently increased insulin secretion in the absence of glucose. In the presence of 10 micromol/L S-(-)-Bay K8644, GW9508 significantly increased insulin secretion. On the other hand, the LTCC blocker nifedipine significantly inhibited insulin secretion mediated by either glucose, glipizide or glucose plus GW9508. Thus, sulfonylureas could replace glucose to support GPR40-mediated enhancement of insulin secretion, whereas blockage of LTCC reduced both glucose and sulfonylurea-mediated insulin secretion.


Subject(s)
Glucose/metabolism , Receptors, G-Protein-Coupled/metabolism , Sulfonylurea Compounds/metabolism , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/metabolism , Animals , Calcium Channel Agonists/metabolism , Calcium Channel Blockers/metabolism , Calcium Channels, L-Type/metabolism , Cell Line/drug effects , Cyclic AMP/metabolism , Glipizide/pharmacology , Hypoglycemic Agents/pharmacology , Insulin/metabolism , Insulin Secretion , Nifedipine/metabolism , Rats , Receptors, G-Protein-Coupled/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...