Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
1.
J Med Chem ; 67(18): 16209-16221, 2024 Sep 26.
Article in English | MEDLINE | ID: mdl-39238314

ABSTRACT

Therapeutic drugs, whose bioactivity is hindered by a photoremovable cage, offer the advantage of spatiotemporal confinement of their action to the target diseased tissue with improved bioavailability and efficacy. Here, we have applied such an approach to ivabradine (IVA), a bradycardic agent indicated for angina pectoris and heart failure, acting as a specific HCN channel blocker. To overcome the side effects due to its poor discrimination among HCN channel subtypes (HCN1-4), we prepared a caged version of IVA linked to a photocleavable bromoquinolinylmethyl group (BHQ-IVA). We show that upon illumination with blue light (440 nm), BHQ-IVA releases active IVA that blocks HCN channel currents in vitro and exerts a bradycardic effect in vivo. Both BHQ-IVA and the cage are inactive. Caging is stable in aqueous medium and in the dark, and it does not impair aqueous solubility and cell permeation, indispensable for IVA activity. This approach allows for bypassing the poor subtype-specificity of IVA, expanding its prescription to HCN-related diseases besides cardiac.


Subject(s)
Ivabradine , Light , Animals , Humans , Male , Mice , Benzazepines/pharmacology , Benzazepines/chemistry , Benzazepines/pharmacokinetics , Heart Rate/drug effects , HEK293 Cells , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/antagonists & inhibitors , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Ivabradine/pharmacology , Ivabradine/chemistry , Quinolines/chemistry , Quinolines/pharmacology
2.
Nature ; 632(8024): 451-459, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39085604

ABSTRACT

Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels1 are essential for pacemaking activity and neural signalling2,3. Drugs inhibiting HCN1 are promising candidates for management of neuropathic pain4 and epileptic seizures5. The general anaesthetic propofol (2,6-di-iso-propylphenol) is a known HCN1 allosteric inhibitor6 with unknown structural basis. Here, using single-particle cryo-electron microscopy and electrophysiology, we show that propofol inhibits HCN1 by binding to a mechanistic hotspot in a groove between the S5 and S6 transmembrane helices. We found that propofol restored voltage-dependent closing in two HCN1 epilepsy-associated polymorphisms that act by destabilizing the channel closed state: M305L, located in the propofol-binding site in S5, and D401H in S6 (refs. 7,8). To understand the mechanism of propofol inhibition and restoration of voltage-gating, we tracked voltage-sensor movement in spHCN channels and found that propofol inhibition is independent of voltage-sensor conformational changes. Mutations at the homologous methionine in spHCN and an adjacent conserved phenylalanine in S6 similarly destabilize closing without disrupting voltage-sensor movements, indicating that voltage-dependent closure requires this interface intact. We propose a model for voltage-dependent gating in which propofol stabilizes coupling between the voltage sensor and pore at this conserved methionine-phenylalanine interface in HCN channels. These findings unlock potential exploitation of this site to design specific drugs targeting HCN channelopathies.


Subject(s)
Epilepsy , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Ion Channel Gating , Mutation , Potassium Channels , Propofol , Humans , Binding Sites , Cryoelectron Microscopy , Electrophysiology , Epilepsy/drug therapy , Epilepsy/genetics , Epilepsy/metabolism , HEK293 Cells , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/antagonists & inhibitors , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/chemistry , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/ultrastructure , Ion Channel Gating/drug effects , Ion Channel Gating/genetics , Methionine/genetics , Methionine/metabolism , Models, Molecular , Movement/drug effects , Phenylalanine/genetics , Phenylalanine/metabolism , Polymorphism, Genetic , Potassium Channels/chemistry , Potassium Channels/genetics , Potassium Channels/metabolism , Potassium Channels/ultrastructure , Propofol/pharmacology , Propofol/chemistry
3.
Proc Natl Acad Sci U S A ; 121(27): e2402259121, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38917012

ABSTRACT

HCN1-4 channels are the molecular determinants of the If/Ih current that crucially regulates cardiac and neuronal cell excitability. HCN dysfunctions lead to sinoatrial block (HCN4), epilepsy (HCN1), and chronic pain (HCN2), widespread medical conditions awaiting subtype-specific treatments. Here, we address the problem by solving the cryo-EM structure of HCN4 in complex with ivabradine, to date the only HCN-specific drug on the market. Our data show ivabradine bound inside the open pore at 3 Å resolution. The structure unambiguously proves that Y507 and I511 on S6 are the molecular determinants of ivabradine binding to the inner cavity, while F510, pointing outside the pore, indirectly contributes to the block by controlling Y507. Cysteine 479, unique to the HCN selectivity filter (SF), accelerates the kinetics of block. Molecular dynamics simulations further reveal that ivabradine blocks the permeating ion inside the SF by electrostatic repulsion, a mechanism previously proposed for quaternary ammonium ions.


Subject(s)
Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Ivabradine , Molecular Dynamics Simulation , Ivabradine/chemistry , Ivabradine/pharmacology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/chemistry , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/antagonists & inhibitors , Humans , Cryoelectron Microscopy , Animals , Potassium Channels/chemistry , Potassium Channels/metabolism , Muscle Proteins/chemistry , Muscle Proteins/metabolism
4.
Biol Pharm Bull ; 44(11): 1796-1799, 2021.
Article in English | MEDLINE | ID: mdl-34719656

ABSTRACT

Torsadogenic effects of ivabradine, an inhibitor of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, were assessed in an in vivo proarrhythmia model of acute atrioventricular block rabbit. Ivabradine at 0.01, 0.1, and 1 mg/kg was intravenously administered to isoflurane-anesthetized rabbits (n = 5) in the stable idioventricular rhythm. Ivabradine at 0.01 and 0.1 mg/kg hardly affected the atrial and ventricular automaticity, QT interval, or the monophasic action potential duration of the ventricle. Additionally administred ivabradine at 1 mg/kg decreased the atrial and ventricular rate significantly but increased the QT interval and duration of the monophasic action potential. Meanwhile, torsade de pointes arrhythmias were detected in 1 out of 5 animals and in 2 out of 5 animals after the administration of 0.1 and 1 mg/kg, respectively. Importantly, torsade de pointes arrhythmias could be observed only in 2 rabbits showing more potent suppressive effects on ventricular automaticity. These results suggest that the torsadogenic potential of ivabradine may become evident when its expected bradycardic action appears more excessively.


Subject(s)
Atrioventricular Block/drug therapy , Cardiovascular Agents/adverse effects , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/antagonists & inhibitors , Ivabradine/adverse effects , Torsades de Pointes/chemically induced , Animals , Cardiovascular Agents/pharmacology , Cardiovascular Agents/therapeutic use , Disease Models, Animal , Electrocardiography , Heart Rate/drug effects , Hemodynamics , Ivabradine/pharmacology , Ivabradine/therapeutic use , Male , Rabbits
5.
Proc Natl Acad Sci U S A ; 118(35)2021 08 31.
Article in English | MEDLINE | ID: mdl-34429357

ABSTRACT

The development of the cerebral cortex relies on the controlled division of neural stem and progenitor cells. The requirement for precise spatiotemporal control of proliferation and cell fate places a high demand on the cell division machinery, and defective cell division can cause microcephaly and other brain malformations. Cell-extrinsic and -intrinsic factors govern the capacity of cortical progenitors to produce large numbers of neurons and glia within a short developmental time window. In particular, ion channels shape the intrinsic biophysical properties of precursor cells and neurons and control their membrane potential throughout the cell cycle. We found that hyperpolarization-activated cyclic nucleotide-gated cation (HCN) channel subunits are expressed in mouse, rat, and human neural progenitors. Loss of HCN channel function in rat neural stem cells impaired their proliferation by affecting the cell-cycle progression, causing G1 accumulation and dysregulation of genes associated with human microcephaly. Transgene-mediated, dominant-negative loss of HCN channel function in the embryonic mouse telencephalon resulted in pronounced microcephaly. Together, our findings suggest a role for HCN channel subunits as a part of a general mechanism influencing cortical development in mammals.


Subject(s)
Cell Proliferation/physiology , Cerebral Cortex/embryology , Channelopathies/etiology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/physiology , Microcephaly/etiology , Neural Stem Cells/physiology , Neurogenesis/physiology , Animals , Cell Cycle , Cell Death , Cells, Cultured , Cerebral Cortex/cytology , Channelopathies/embryology , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/physiology , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/antagonists & inhibitors , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Mice , Mice, Transgenic , Microcephaly/embryology , Neural Stem Cells/metabolism , Rats
6.
Physiol Rep ; 9(15): e14963, 2021 08.
Article in English | MEDLINE | ID: mdl-34342171

ABSTRACT

Degeneracy, the ability of multiple structural components to elicit the same characteristic functional properties, constitutes an elegant mechanism for achieving biological robustness. In this study, we sought electrophysiological signatures for the expression of ion-channel degeneracy in the emergence of intrinsic properties of rat hippocampal granule cells. We measured the impact of four different ion-channel subtypes-hyperpolarization-activated cyclic-nucleotide-gated (HCN), barium-sensitive inward rectifier potassium (Kir ), tertiapin-Q-sensitive inward rectifier potassium, and persistent sodium (NaP) channels-on 21 functional measurements employing pharmacological agents, and report electrophysiological data on two characteristic signatures for the expression of ion-channel degeneracy in granule cells. First, the blockade of a specific ion-channel subtype altered several, but not all, functional measurements. Furthermore, any given functional measurement was altered by the blockade of many, but not all, ion-channel subtypes. Second, the impact of blocking each ion-channel subtype manifested neuron-to-neuron variability in the quantum of changes in the electrophysiological measurements. Specifically, we found that blocking HCN or Ba-sensitive Kir channels enhanced action potential firing rate, but blockade of NaP channels reduced firing rate of granule cells. Subthreshold measures of granule cell intrinsic excitability (input resistance, temporal summation, and impedance amplitude) were enhanced by blockade of HCN or Ba-sensitive Kir channels, but were not significantly altered by NaP channel blockade. We confirmed that the HCN and Ba-sensitive Kir channels independently altered sub- and suprathreshold properties of granule cells through sequential application of pharmacological agents that blocked these channels. Finally, we found that none of the sub- or suprathreshold measurements of granule cells were significantly altered upon treatment with tertiapin-Q. Together, the heterogeneous many-to-many mapping between ion channels and single-neuron intrinsic properties emphasizes the need to account for ion-channel degeneracy in cellular- and network-scale physiology.


Subject(s)
Bee Venoms/pharmacology , Hippocampus/physiology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/antagonists & inhibitors , Neurons/physiology , Potassium Channels/chemistry , Sodium Channels/chemistry , Animals , Barium/pharmacology , Hippocampus/cytology , Hippocampus/drug effects , Male , Neurons/cytology , Neurons/drug effects , Rats , Rats, Sprague-Dawley
7.
Nat Commun ; 12(1): 2802, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33990563

ABSTRACT

Pacemaker hyperpolarization-activated cyclic nucleotide-gated (HCN) ion channels exhibit a reversed voltage-dependent gating, activating by membrane hyperpolarization instead of depolarization. Sea urchin HCN (spHCN) channels also undergo inactivation with hyperpolarization which occurs only in the absence of cyclic nucleotide. Here we applied transition metal ion FRET, patch-clamp fluorometry and Rosetta modeling to measure differences in the structural rearrangements between activation and inactivation of spHCN channels. We found that removing cAMP produced a largely rigid-body rotation of the C-linker relative to the transmembrane domain, bringing the A' helix of the C-linker in close proximity to the voltage-sensing S4 helix. In addition, rotation of the C-linker was elicited by hyperpolarization in the absence but not the presence of cAMP. These results suggest that - in contrast to electromechanical coupling for channel activation - the A' helix serves to couple the S4-helix movement for channel inactivation, which is likely a conserved mechanism for CNBD-family channels.


Subject(s)
Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/chemistry , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Animals , Cyclic AMP , Cyclic Nucleotide-Gated Cation Channels/antagonists & inhibitors , Cyclic Nucleotide-Gated Cation Channels/chemistry , Cyclic Nucleotide-Gated Cation Channels/metabolism , Female , Fluorescence Resonance Energy Transfer , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/antagonists & inhibitors , Ion Channel Gating , Male , Mechanotransduction, Cellular , Membrane Potentials , Models, Molecular , Oocytes/metabolism , Patch-Clamp Techniques , Protein Conformation, alpha-Helical , Protein Domains , Sea Urchins/metabolism , Spermatozoa/metabolism , Xenopus/metabolism
8.
SLAS Discov ; 26(7): 896-908, 2021 08.
Article in English | MEDLINE | ID: mdl-34041946

ABSTRACT

The hyperpolarization-activated cyclic nucleotide-gated 4 (HCN4) channel underlies the pacemaker currents, called "If," in sinoatrial nodes (SANs), which regulate heart rhythm. Some HCN4 blockers such as ivabradine have been extensively studied for treating various heart diseases. Studies have shown that these blockers have diverse state dependencies and binding sites, suggesting the existence of potential chemical and functional diversity among HCN4 blockers. Here we report approaches for the identification of novel HCN4 blockers through a random screening campaign among 16,000 small-molecule compounds using an automated patch-clamp system. These molecules exhibited various blockade profiles, and their blocking kinetics and associating amino acids were determined by electrophysiological studies and site-directed mutagenesis analysis, respectively. The profiles of these blockers were distinct from those of the previously reported HCN channel blockers ivabradine and ZD7288. Notably, the mutagenesis analysis showed that blockers with potencies that were increased when the channel was open involved a C478 residue, located at the pore cavity region near the cellular surface of the plasma membrane, while those with potencies that were decreased when the channel was open involved residues Y506 and I510, located at the intracellular region of the pore gate. Thus, this study reported for the first time the discovery of novel HCN4 blockers by screening, and their profiling analysis using an automated patch-clamp system provided chemical tools that will be useful to obtain unique molecular insights into the drug-binding modes of HCN4 and may contribute to the expansion of therapeutic options in the future.


Subject(s)
Drug Discovery/methods , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/antagonists & inhibitors , Muscle Proteins/antagonists & inhibitors , Potassium Channel Blockers/chemistry , Data Analysis , Dose-Response Relationship, Drug , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/chemistry , Ion Channel Gating/drug effects , Kinetics , Membrane Potentials/drug effects , Models, Molecular , Molecular Structure , Muscle Proteins/chemistry , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Potassium Channels/chemistry , Structure-Activity Relationship
9.
Neurosci Lett ; 750: 135763, 2021 04 17.
Article in English | MEDLINE | ID: mdl-33617945

ABSTRACT

The cyclic adenosine monophosphate-protein kinase A (cAMP-PKA) signaling acts a pivotal part in hyperpolarization-activated cyclic nucleotide-gated (HCN) channels-mediated neuropathic and inflammatory pain. However, there has been no evidence of cAMP-PKA signaling is involved in regulation of spinal HCN channels function in the occurrence of diabetic neuropathic pain (DNP). The study aimed to elucidate the impact of HCN channels on neuropathic pain in a rat model of diabetes induced by streptozotocin, and whether cAMP-PKA signaling is involved in regulation of HCN channels function. In this report, we evaluated the effect of intrathecal administration of HCN channel blockers ZD7288, cAMP inhibitor SQ22536 and PKA inhibitor H-89 on nociceptive behavior in DNP rats. The mechanical withdrawal threshold (MWT) was measured to evaluate pain behavior in rats. Protein expression levels of HCN2, HCN4 channels and PKA in the spinal dorsal horn of rats were assessed. Furthermore, the levels of cAMP in rat spinal dorsal horn was analyzed. We discovered that DNP rats showed significant mechanical allodynia and are related to the increased HCN2 and HCN4 channels expression, enhanced cAMP production and elevated the expression of PKA protein in the spinal dorsal horn, which were attenuated by intrathecal ZD7288. Furthermore, intrathecal injection of SQ22536 and H-89 significantly reduced the HCN2 and HCN4 channels expression in the spinal dorsal horn of DNP rats. Our findings indicate that HCN channels of the spinal dorsal horn participate in the pathogenesis of allodynia in rats with DNP, which could be regulated by cAMP-PKA signaling. Therefore, HCN channels and cAMP-PKA signaling are potential targets for hyperalgesia treatment in DNP patients.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Diabetic Neuropathies/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Nociception , Spinal Cord/metabolism , Animals , Cyclic AMP/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Diabetic Neuropathies/physiopathology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/antagonists & inhibitors , Isoquinolines/pharmacology , Male , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction , Spinal Cord/physiology , Sulfonamides/pharmacology
10.
Biochem Biophys Res Commun ; 533(4): 952-957, 2020 12 17.
Article in English | MEDLINE | ID: mdl-33008592

ABSTRACT

Quercetin is a natural flavonoid which has been reported to be analgesic in different animal models of pain. However, the mechanism underlying the pain-relieving effects is still unclear. Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels play critical roles in controlling pacemaker activity in cardiac and nervous systems, making the channel a new target for therapeutic exploration. In this study, we explored a series of flavonoids for their modulation on HCN channels. Among all tested flavonoids, quercetin was the most potent inhibitor for HCN channels with an IC50 value of 27.32 ± 1.19 µM for HCN2. Furthermore, quercetin prominently left shifted the voltage-dependent activation curves of HCN channels and decelerated deactivation process. The results presented herein firstly characterize quercetin as a novel and potent inhibitor for HCN channels, which represents a novel structure for future drug design of HCN channel inhibitors.


Subject(s)
Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/antagonists & inhibitors , Quercetin/pharmacology , Animals , COS Cells , Chlorocebus aethiops , Drug Evaluation, Preclinical , Electrophysiological Phenomena , Flavonoids/chemistry , Flavonoids/pharmacology , Flavonols/chemistry , Flavonols/pharmacology , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/genetics , Muscle Proteins/metabolism , Patch-Clamp Techniques , Potassium Channels/genetics , Potassium Channels/metabolism , Quercetin/chemistry , Recombinant Proteins/drug effects , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Structure-Activity Relationship
11.
ASN Neuro ; 12: 1759091420944658, 2020.
Article in English | MEDLINE | ID: mdl-32962418

ABSTRACT

Oxytocin, a hypothalamic neuropeptide essential for breastfeeding, is mainly produced in oxytocin neurons in the supraoptic nucleus (SON) and paraventricular nucleus. However, mechanisms underlying oxytocin secretion, specifically the involvement of hyperpolarization-activated cyclic nucleotide-gated channel 3 (HCN3) in oxytocin neuronal activity, remain unclear. Using a rat model of intermittent and continuous pup deprivation (PD) at the middle stage of lactation, we analyzed the contribution of HCN3 in oxytocin receptor (OTR)-associated signaling cascade to oxytocin neuronal activity in the SON. PD caused maternal depression, anxiety, milk shortage, involution of the mammary glands, and delays in uterine recovery, particularly in continuous PD. PD increased hypothalamic but not plasma oxytocin levels in enzyme-linked immunosorbent assay. In the SON, PD increased c-Fos expression but reduced expressions of cyclooxygenase-2 and HCN3 in Western blots and/or immunohistochemistry. Moreover, PD significantly increased the molecular association of OTR with HCN3 in coimmunoprecipitation. In brain slices, inhibition of HCN3 activity with DK-AH269 blocked prostaglandin E2-evoked increase in the firing activity and burst discharge in oxytocin neurons in patch-clamp recordings. In addition, oxytocin-evoked increase in the molecular association between OTR and HCN3 in brain slices of the SON was blocked by pretreatment with indomethacin, an inhibitor of cyclooxygenase-2. These results indicate that normal activity of oxytocin neurons is under the regulation of an oxytocin receptor-cyclooxygenase-2-HCN3 pathway and that PD disrupts maternal behavior through increasing intranuclear oxytocin secretion in the SON but likely reducing bolus oxytocin release into the blood through inhibition of HCN3 activity.


Subject(s)
Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Lactation/metabolism , Maternal Deprivation , Neurons/metabolism , Oxytocin/metabolism , Potassium Channels/metabolism , Animals , Animals, Newborn , Benzazepines/pharmacology , Female , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/antagonists & inhibitors , Hypothalamus/drug effects , Hypothalamus/metabolism , Lactation/drug effects , Lactation/psychology , Male , Neurons/drug effects , Rats , Rats, Sprague-Dawley
12.
Sci Rep ; 10(1): 11831, 2020 07 16.
Article in English | MEDLINE | ID: mdl-32678143

ABSTRACT

A meta-analysis of genome-wide association studies (GWAS) identified eight loci that are associated with heart rate variability (HRV), but candidate genes in these loci remain uncharacterized. We developed an image- and CRISPR/Cas9-based pipeline to systematically characterize candidate genes for HRV in live zebrafish embryos. Nine zebrafish orthologues of six human candidate genes were targeted simultaneously in eggs from fish that transgenically express GFP on smooth muscle cells (Tg[acta2:GFP]), to visualize the beating heart. An automated analysis of repeated 30 s recordings of beating atria in 381 live, intact zebrafish embryos at 2 and 5 days post-fertilization highlighted genes that influence HRV (hcn4 and si:dkey-65j6.2 [KIAA1755]); heart rate (rgs6 and hcn4); and the risk of sinoatrial pauses and arrests (hcn4). Exposure to 10 or 25 µM ivabradine-an open channel blocker of HCNs-for 24 h resulted in a dose-dependent higher HRV and lower heart rate at 5 days post-fertilization. Hence, our screen confirmed the role of established genes for heart rate and rhythm (RGS6 and HCN4); showed that ivabradine reduces heart rate and increases HRV in zebrafish embryos, as it does in humans; and highlighted a novel gene that plays a role in HRV (KIAA1755).


Subject(s)
Bradycardia/genetics , Heart Rate/physiology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Myocardial Contraction/physiology , RGS Proteins/genetics , Animals , Animals, Genetically Modified , Bradycardia/diagnostic imaging , Bradycardia/metabolism , Bradycardia/physiopathology , CRISPR-Cas Systems , Cardiovascular Agents/pharmacology , Embryo, Nonmammalian , Genes, Reporter , Genome-Wide Association Study , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Heart Rate/drug effects , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/antagonists & inhibitors , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Ivabradine/pharmacology , Meta-Analysis as Topic , Myocardial Contraction/drug effects , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Optical Imaging/methods , Pleckstrin Homology Domains/genetics , RGS Proteins/metabolism , Zebrafish
13.
Neurosci Bull ; 36(8): 875-894, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32519067

ABSTRACT

In the central nervous system, hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are essential to maintain normal neuronal function. Recent studies have shown that HCN channels may be involved in the pathological process of ischemic brain injury, but the mechanisms remain unclear. Autophagy is activated in cerebral ischemia, but its role in cell death/survival remains controversial. In this study, our results showed that the HCN channel blocker ZD7288 remarkably decreased the percentage of apoptotic neurons and corrected the excessive autophagy induced by oxygen-glucose deprivation followed by reperfusion (OGD/R) in hippocampal HT22 neurons. Furthermore, in the OGD/R group, p-mTOR, p-ULK1 (Ser757), and p62 were significantly decreased, while p-ULK1 (Ser317), atg5, and beclin1 were remarkably increased. ZD7288 did not change the expression of p-ULK1 (Ser757), ULK1 (Ser317), p62, Beclin1, and atg5, which are involved in regulating autophagosome formation. Besides, we found that OGD/R induced a significant increase in Cathepsin D expression, but not LAMP-1. Treatment with ZD7288 at 10 µmol/L in the OGD/R group did not change the expression of cathepsin D and LAMP-1. However, chloroquine (CQ), which decreases autophagosome-lysosome fusion, eliminated the correction of excessive autophagy and neuroprotection by ZD7288. Besides, shRNA knockdown of HCN2 channels significantly reduced the accumulation of LC3-II and increased neuron survival in the OGD/R and transient global cerebral ischemia (TGCI) models, and CQ also eliminated the effects of HCN2-shRNA. Furthermore, we found that the percentage of LC3-positive puncta that co-localized with LAMP-1-positive lysosomes decreased in Con-shRNA-transfected HT22 neurons exposed to OGD/R or CQ. In HCN2-shRNA-transfected HT22 neurons, the percentage of LC3-positive puncta that co-localized with LAMP-1-positive lysosomes increased under OGD/R; however, the percentage was significantly decreased by the addition of CQ to HCN2-shRNA-transfected HT22 neurons. The present results demonstrated that blockade of HCN2 channels provides neuroprotection against OGD/R and TGCI by accelerating autophagic degradation attributable to the promotion of autophagosome and lysosome fusion.


Subject(s)
Autophagy , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/antagonists & inhibitors , Neurons , Neuroprotection , Reperfusion Injury , Glucose , Hippocampus/cytology , Humans , Potassium Channels , Pyrimidines
14.
J Neuroinflammation ; 17(1): 100, 2020 Apr 06.
Article in English | MEDLINE | ID: mdl-32248813

ABSTRACT

BACKGROUND: Microglia are essential to maintain cell homeostasis in the healthy brain and are activated after brain injury. Upon activation, microglia polarize towards different phenotypes. The course of microglia activation is complex and depends on signals in the surrounding milieu. Recently, it has been suggested that microglia respond to ion currents, as a way of regulating their activity and function. METHODS AND RESULTS: Under the hypothesis that HCN and KCNQ/Kv7 channels impact on microglia, we studied primary rat microglia in the presence or absence of specific pharmacological blockade or RNA silencing. Primary microglia expressed the subunits HCN1-4, Kv7.2, Kv7.3, and Kv7.5. The expression of HCN2, as well as Kv7.2 and Kv7.3, varied among different microglia phenotypes. The pharmacological blockade of HCN channels by ZD7288 resulted in cell depolarization with slowly rising intracellular calcium levels, leading to enhanced survival and reduced proliferation rates of resting microglia. Furthermore, ZD7288 treatment, as well as knockdown of HCN2 RNA by small interfering RNA, resulted in an attenuation of later microglia activation-both towards the anti- and pro-inflammatory phenotype. However, HCN channel inhibition enhanced the phagocytic capacity of IL4-stimulated microglia. Blockade of Kv7/KCNQ channel by XE-991 exclusively inhibited the migratory capacity of resting microglia. CONCLUSION: These observations suggest that the HCN current contributes to various microglia functions and impacts on the course of microglia activation, while the Kv7/KCNQ channels affect microglia migration. Characterizing the role of HCN channels in microglial functioning may offer new therapeutic approaches for targeted modulation of neuroinflammation as a hallmark of various neurological disorders.


Subject(s)
Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Microglia/metabolism , Potassium Channel Blockers/pharmacology , Potassium Channels, Voltage-Gated/metabolism , Animals , Calcium/metabolism , Cell Proliferation/drug effects , Cell Proliferation/physiology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/antagonists & inhibitors , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Microglia/drug effects , Phagocytosis/drug effects , Phagocytosis/physiology , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Potassium Channels, Voltage-Gated/genetics , Pyrimidines/pharmacology , RNA Interference , Rats , Rats, Wistar
15.
J Membr Biol ; 253(2): 153-166, 2020 04.
Article in English | MEDLINE | ID: mdl-32146488

ABSTRACT

Hyperpolarization-activated cation-nonselective (HCN) channels regulate electrical activity in the brain and heart in a cAMP-dependent manner. The voltage-gating of these channels is mediated by a transmembrane (TM) region but is additionally regulated by direct binding of cAMP to a cyclic nucleotide-binding (CNB) fold in the cytoplasmic C-terminal region. Cyclic AMP potentiation has been explained by an autoinhibition model which views the unliganded CNB fold as an inhibitory module whose influence is disrupted by cAMP binding. However, the HCN2 subtype uses two other CNB fold-mediated mechanisms called open-state trapping and Quick-Activation to respectively slow the deactivation kinetics and speed the activation kinetics, against predictions of an autoinhibition model. To test how these multiple mechanisms are influenced by the TM region, we replaced the TM region of HCN2 with that of HCN4. This HCN4 TM-replacement preserved cAMP potentiation but augmented the magnitude of autoinhibition by the unliganded CNB fold; it moreover disrupted open-state trapping and Quick-Activation so that autoinhibition became the dominant mechanism contributed by the C-terminal region to determine kinetics. Truncation within the CNB fold partially relieved this augmented autoinhibition. This argues against the C-terminal region acting like a portable module with consistent effects on TM regions of different subtypes. Our findings provide evidence that functional interactions between the HCN2 TM region and C-terminal region govern multiple CNB fold-mediated mechanisms, implying that the molecular mechanisms of autoinhibition, open-state trapping, and Quick-Activation include participation of TM region structures.


Subject(s)
Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Protein Interaction Domains and Motifs , Cell Membrane/metabolism , Cyclic AMP/metabolism , Cytoplasm/metabolism , Electrophysiological Phenomena , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/agonists , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/antagonists & inhibitors , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/chemistry , Ion Channel Gating , Kinetics , Ligands , Protein Binding
16.
Cell Rep ; 30(9): 2879-2888.e3, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32130893

ABSTRACT

Peristalsis is indispensable for physiological function of the gut. The enteric nervous system (ENS) plays an important role in regulating peristalsis. While the neural network regulating anterograde peristalsis, which migrates from the oral end to the anal end, is characterized to some extent, retrograde peristalsis remains unresolved with regards to its neural regulation. Using forward genetics in zebrafish, we reveal that a population of neurons expressing a hyperpolarization-activated nucleotide-gated channel HCN4 specifically regulates retrograde peristalsis. When HCN4 channels are blocked by an HCN channel inhibitor or morpholinos blocking the protein expression, retrograde peristalsis is specifically attenuated. Conversely, when HCN4(+) neurons expressing channelrhodopsin are activated by illumination, retrograde peristalsis is enhanced while anterograde peristalsis remains unchanged. We propose that HCN4(+) neurons in the ENS forward activating signals toward the oral end and simultaneously stimulate local circuits regulating the circular muscle.


Subject(s)
Gastrointestinal Tract/innervation , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Neurons/metabolism , Peristalsis , Zebrafish/physiology , Animals , Animals, Genetically Modified , Gastrointestinal Motility/drug effects , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/physiology , Gene Expression Regulation/drug effects , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/antagonists & inhibitors , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Larva/drug effects , Larva/physiology , Morpholinos/pharmacology , Optogenetics , Peristalsis/drug effects , Serotonin/metabolism , Zebrafish/genetics
17.
Int J Mol Sci ; 21(4)2020 Feb 13.
Article in English | MEDLINE | ID: mdl-32070065

ABSTRACT

Croton is an extensive flowering plant genus in the spurge family, Euphorbiaceae. Three croton compounds with the common ent-kaurane skeleton have been purified from Croton tonkinensis. METHODS: We examined any modifications of croton components (i.e., croton-01 [ent-18-acetoxy-7α-hydroxykaur-16-en-15-one], croton-02 [ent-7α,14ß-dihydroxykaur-16-en-15-one] and croton-03 [ent-1ß-acetoxy-7α,14ß-dihydroxykaur-16-en-15-one] on either hyperpolarization-activated cation current (Ih) or erg-mediated K+ current identified in pituitary tumor (GH3) cells and in rat insulin-secreting (INS-1) cells via patch-clamp methods. RESULTS: Addition of croton-01, croton-02, or croton-03 effectively and differentially depressed Ih amplitude. Croton-03 (3 µM) shifted the activation curve of Ih to a more negative potential by approximately 11 mV. The voltage-dependent hysteresis of Ih was also diminished by croton-03 administration. Croton-03-induced depression of Ih could not be attenuated by SQ-22536 (10 µM), an inhibitor of adenylate cyclase, but indeed reversed by oxaliplatin (10 µM). The Ih in INS-1 cells was also depressed effectively by croton-03. CONCLUSION: Our study highlights the evidence that these ent-kaurane diterpenoids might conceivably perturb these ionic currents through which they have high influence on the functional activities of endocrine or neuroendocrine cells.


Subject(s)
Croton/chemistry , Diterpenes, Kaurane/pharmacology , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/antagonists & inhibitors , Pituitary Neoplasms/drug therapy , Adenylyl Cyclases/genetics , Animals , Cell Line, Tumor , Disease Models, Animal , Diterpenes, Kaurane/chemistry , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/chemistry , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Molecular Structure , Pituitary Neoplasms/genetics , Pituitary Neoplasms/pathology , Plant Extracts/chemistry , Plant Extracts/pharmacology , Plant Leaves/chemistry , Rats
18.
Synapse ; 74(3): e22137, 2020 03.
Article in English | MEDLINE | ID: mdl-31584700

ABSTRACT

We examined effects of Group I metabotropic glutamate receptors on the excitability of mouse medial nucleus of the trapezoid body (MNTB) neurons. The selective agonist, S-3,5-dihydroxyphenylglycine (DHPG), evoked a dose-dependent depolarization of the resting potential, increased membrane resistance, increased sag depolarization, and promoted rebound action potential firing. Under voltage-clamp, DHPG evoked an inward current, referred to as IDHPG , which was developmentally stable through postnatal day P56. IDHPG had low temperature dependence in the range 25-34°C, consistent with a channel mechanism. However, the I-V relationship took the form of an inverted U that did not reverse at the calculated Nernst potential for K+ or Cl- . Thus, it is likely that more than one ion type contributes to IDHPG and the mix may be voltage dependent. IDHPG was resistant to the Na+ channel blockers tetrodotoxin and amiloride, and to inhibitors of iGluR (CNQX and MK801). IDHPG was inhibited 21% by Ba2+ (500 µM), 60% by ZD7288 (100 µM) and 73% when the two antagonists were applied together, suggesting that KIR channels and HCN channels contribute to the current. Voltage clamp measurements of IH indicated a small (6%) increase in Gmax by DHPG with no change in the voltage dependence. DHPG reduced action potential rheobase and reduced the number of post-synaptic AP failures during high frequency stimulation of the calyx of Held. Thus, activation of post-synaptic Group I mGlu receptors modifies the excitability of MNTB neurons and contributes to the reliability of high frequency firing in this auditory relay nucleus.


Subject(s)
Action Potentials , Excitatory Amino Acid Agents/pharmacology , Receptors, Metabotropic Glutamate/metabolism , Synaptic Potentials , Trapezoid Body/metabolism , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Amiloride/pharmacology , Animals , Dizocilpine Maleate/pharmacology , Female , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/antagonists & inhibitors , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Male , Methoxyhydroxyphenylglycol/analogs & derivatives , Methoxyhydroxyphenylglycol/pharmacology , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Neurons/physiology , Potassium Channel Blockers/pharmacology , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Potassium Channels, Inwardly Rectifying/metabolism , Pyrimidines/pharmacology , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology , Trapezoid Body/cytology , Trapezoid Body/drug effects , Trapezoid Body/physiology
19.
J Neurophysiol ; 122(2): 691-706, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31268830

ABSTRACT

How neurons filter and integrate their complex patterns of synaptic inputs is central to their role in neural information processing. Synaptic filtering and integration are shaped by the frequency-dependent neuronal membrane impedance. Using single and dual dendritic recordings in vivo, pharmacology, and computational modeling, we characterized the membrane impedance of a collision detection neuron in the grasshopper Schistocerca americana. This neuron, the lobula giant movement detector (LGMD), exhibits consistent impedance properties across frequencies and membrane potentials. Two common active conductances gH and gM, mediated respectively by hyperpolarization-activated cyclic nucleotide-gated (HCN) channels and by muscarine-sensitive M-type K+ channels, promote broadband integration with high temporal precision over the LGMD's natural range of membrane potentials and synaptic input frequencies. Additionally, we found that a model based on the LGMD's branching morphology increased the gain and decreased the delay associated with the mapping of synaptic input currents to membrane potential. More generally, this was true for a wide range of model neuron morphologies, including those of neocortical pyramidal neurons and cerebellar Purkinje cells. These findings show the unexpected role played by two widespread active conductances and by dendritic morphology in shaping synaptic integration.NEW & NOTEWORTHY Neuronal filtering and integration of synaptic input patterns depend on the electrochemical properties of dendrites. We used an identified collision detection neuron in grasshoppers to examine how its morphology and two conductances affect its membrane impedance in relation to the computations it performs. The neuronal properties examined are ubiquitous and therefore promote a general understanding of neuronal computations, including those in the human brain.


Subject(s)
Dendrites/physiology , Excitatory Postsynaptic Potentials/physiology , Motion Perception/physiology , Neurons/physiology , Animals , Electric Impedance , Excitatory Postsynaptic Potentials/drug effects , Female , Grasshoppers , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/antagonists & inhibitors , Models, Biological , Motion Perception/drug effects , Neurons/drug effects , Potassium Channel Blockers/pharmacology
20.
Eur J Cardiothorac Surg ; 55(Suppl 1): i3-i10, 2019 06 01.
Article in English | MEDLINE | ID: mdl-31106335

ABSTRACT

During the last 20 years, the prognosis for heart failure (HF) with reduced ejection fraction has steadily improved due to advances in drug treatment and the consistent implementation of guideline-recommended evidence-based drug therapy. Nevertheless, the morbidity and mortality rates of patients with HF can still be improved. The prevalence of HF is high and continues to increase steadily. Thus, timely and efficient drug treatment plays a central role in improving the quality of life and prognosis for patients with HF. Current therapeutic concepts combine inhibition of the renin-angiotensin-aldosterone system with blockage of the sympathetic system. New therapeutic approaches such as selective heart rate reduction, attenuation of the degradation of natriuretic peptides by neutral endopeptidase inhibition and treatment of comorbidities (e.g. iron deficiency, diabetes mellitus, hyperkalaemia) have led to a further improvement in the survival, time-out-of hospital and quality of life of affected patients. The goal of this article was to give an overview of the current standard drug therapy for HF and the value of new therapeutic approaches implemented in recent years.


Subject(s)
Heart Failure/drug therapy , Adrenergic beta-Antagonists/therapeutic use , Algorithms , Angiotensin Receptor Antagonists/therapeutic use , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Benzhydryl Compounds/therapeutic use , Cardiac Glycosides/therapeutic use , Cardiotonic Agents/therapeutic use , Diuretics/therapeutic use , Enzyme Activators/therapeutic use , Glucosides/therapeutic use , Humans , Hyperkalemia/chemically induced , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/antagonists & inhibitors , Iron/therapeutic use , Ivabradine/therapeutic use , Mineralocorticoid Receptor Antagonists/therapeutic use , Neprilysin/antagonists & inhibitors , Practice Guidelines as Topic , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Renin/antagonists & inhibitors , Renin-Angiotensin System/drug effects , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Stroke Volume , Urea/analogs & derivatives , Urea/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL