Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.606
Filter
1.
Nutrients ; 16(17)2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39275318

ABSTRACT

Studies have demonstrated the therapeutic effects of Lindera plants. This study was undertaken to reveal the antihypertensive properties of Lindera erythrocarpa leaf ethanolic extract (LEL). Aorta segments of Sprague-Dawley rats were used to study the vasodilatory effect of LEL, and the mechanisms involved were evaluated by treating specific inhibitors or activators that affect the contractility of blood vessels. Our results revealed that LEL promotes a vasorelaxant effect through the nitric oxide/cyclic guanosine 3',5'-monophosphate pathway, blocking the Ca2+ channels, opening the K+ channels, and inhibiting the vasoconstrictive action of angiotensin II. In addition, the effects of LEL on blood pressure were investigated in spontaneously hypertensive rats by the tail-cuff method. LEL (300 or 1000 mg/kg) was orally administered to the rats, and 1000 mg/kg of LEL significantly lowered the blood pressure. Systolic blood pressure decreased by -20.06 ± 4.87%, and diastolic blood pressure also lowered by -30.58 ± 5.92% at 4 h in the 1000 mg/kg LEL group. Overall, our results suggest that LEL may be useful to treat hypertensive diseases, considering its vasorelaxing and hypotensive effects.


Subject(s)
Antihypertensive Agents , Blood Pressure , Cyclic GMP , Hypertension , Lindera , Nitric Oxide , Plant Extracts , Rats, Inbred SHR , Rats, Sprague-Dawley , Animals , Antihypertensive Agents/pharmacology , Plant Extracts/pharmacology , Nitric Oxide/metabolism , Blood Pressure/drug effects , Cyclic GMP/metabolism , Male , Hypertension/drug therapy , Rats , Lindera/chemistry , Potassium Channels/metabolism , Potassium Channels/drug effects , Calcium Channels/metabolism , Calcium Channels/drug effects , Plant Leaves/chemistry , Vasodilation/drug effects , Signal Transduction/drug effects , Vasodilator Agents/pharmacology
2.
Nat Commun ; 15(1): 7189, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39168976

ABSTRACT

Biological ion channels usually conduct the high-flux transport of 107 ~ 108 ions·s-1; however, the underlying mechanism is still lacking. Here, by applying the KcsA potassium channel as a typical example, and performing multitimescale molecular dynamics simulations, we demonstrate that there is coherence of the K+ ions confined in biological channels, which determines transport. The coherent oscillation state of confined K+ ions with a nanosecond-level lifetime in the channel dominates each transport event, serving as the physical basis for the high flux of ~108 ions∙s-1. The coherent transfer of confined K+ ions only takes several picoseconds and has no perturbation effect on the ion coherence, acting as the directional key of transport. Such ion coherence is allowed by quantum mechanics. An increase in the coherence can significantly enhance the ion conductance. These findings provide a potential explanation from the perspective of coherence for the high-flux ion transport with ultralow energy consumption of biological channels.


Subject(s)
Ion Transport , Molecular Dynamics Simulation , Potassium Channels , Potassium , Quantum Theory , Potassium Channels/metabolism , Potassium Channels/chemistry , Potassium/metabolism , Potassium/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Ions/metabolism
3.
J Transl Med ; 22(1): 785, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39175013

ABSTRACT

BACKGROUND: Activation of the NLRP3 inflammasome is critical in the inflammatory response to gout. Potassium ion (K+) efflux mediated by the TWIK2 channel is an important upstream mechanism for NLRP3 inflammasome activation. Therefore, the TWIK2 channel may be a promising therapeutic target for MSU crystal-induced inflammation. In the present study, we investigated the effect of ML335, a known K2P channel modulator, on MSU crystal-induced inflammatory responses and its underlying molecular mechanisms. METHODS: By molecular docking, we calculated the binding energies and inhibition constants of five K2P channel modulators (Hydroxychloroquine, Fluoxetine, DCPIB, ML365 and ML335) with TWIK2. Intracellular potassium ion concentration and mitochondrial function were assessed by flow cytometry. The interaction between MARCH5 and SIRT3 was demonstrated by immunoprecipitation and Western blotting assay. MSU suspensions were injected into mouse paw and peritoneal cavity to induce acute gout model. RESULTS: ML335 has the highest binding energy and the lowest inhibition constant with TWIK2 in the five calculated K2P channel modulators. In comparison, among these five compounds, ML335 efficiently inhibited the release of IL-1ß from MSU crystal-treated BMDMs. ML335 decreased MSU crystal-induced K+ efflux mainly dependent on TWIK2 channel. More importantly, ML335 can effectively inhibit the expression of the mitochondrial E3 ubiquitin ligase MARCH5 induced by MSU crystals, and MARCH5 can interact with the SIRT3 protein. ML335 blocked MSU crystal-induced ubiquitination of SIRT3 protein by MARCH5. In addition, ML335 improved mitochondrial dynamics homeostasis and mitochondrial function by inhibiting MARCH5 protein expression. ML335 attenuated the inflammatory response induced by MSU crystals in vivo and in vitro. CONCLUSION: Inhibition of TWIK2-mediated K+ efflux by ML335 alleviated mitochondrial injury via suppressing March5 expression, suggesting that ML335 may be an effective candidate for the future treatment of gout.


Subject(s)
Inflammation , Mitochondria , Potassium , Animals , Mitochondria/metabolism , Mitochondria/drug effects , Inflammation/pathology , Potassium/metabolism , Mice, Inbred C57BL , Molecular Docking Simulation , Male , Gout/metabolism , Gout/pathology , Gout/drug therapy , Mice , Ubiquitin-Protein Ligases/metabolism , Potassium Channels/metabolism , Sirtuin 3/metabolism , Interleukin-1beta/metabolism , Inflammasomes/metabolism , Humans
4.
Int J Biol Macromol ; 277(Pt 2): 134306, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39094860

ABSTRACT

Fungal infections pose severe and potentially lethal threats to plant, animal, and human health. Ergosterol has served as the primary target for developing antifungal medications. However, many antifungal drugs remain highly toxic to humans due to similarity in cell membrane composition between fungal and animal cells. Iturin A, lipopeptide produced by Bacillus subtilis, efficiently inhibit various fungi, but demonstrated safety in oral administration, indicating the existence of targets different from ergosterol. To pinpoint the exact antifungal target of iturin A, we used homologous recombination to knock out and overexpress erg3, a key gene in ergosterol synthesis. Saccharomyces cerevisiae and Aspergillus carbonarius were transformed using the LiAc/SS-DNNPEG and Agrobacterium-mediated transformation (AMT), respectively. Surprisingly, increasing ergosterol content did not augment antifungal activity. Furthermore, iturin A's antifungal activity against S. cerevisiae was reduced while it pre-incubation with voltage-gated potassium (Kv) channel inhibitor, indicating that Kv activation was responsible for cell death. Iturin A was found to activate the Kv protein, stimulating K+ efflux from cell. In vitro tests confirmed interaction between iturin A and Kv protein. This study highlights Kv as one of the precise targets of iturin A in its antifungal activity, offering a novel target for the development of antifungal medications.


Subject(s)
Antifungal Agents , Bacillus subtilis , Peptides, Cyclic , Saccharomyces cerevisiae , Antifungal Agents/pharmacology , Antifungal Agents/chemistry , Peptides, Cyclic/pharmacology , Peptides, Cyclic/chemistry , Bacillus subtilis/drug effects , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Lipopeptides/pharmacology , Potassium Channels/metabolism , Potassium Channels/genetics , Ergosterol , Aspergillus/drug effects , Aspergillus/metabolism , Potassium/metabolism , Microbial Sensitivity Tests
5.
J Am Heart Assoc ; 13(16): e035415, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39158577

ABSTRACT

BACKGROUND: Cardiovascular disease remains one of the leading causes of death globally. Myocardial ischemia and infarction, in particular, frequently cause disturbances in cardiac electrical activity that can trigger ventricular arrhythmias. We aimed to investigate whether catestatin, an endogenous catecholamine-inhibiting peptide, ameliorates myocardial ischemia-induced ventricular arrhythmias in rats and the underlying ionic mechanisms. METHODS AND RESULTS: Adult male Sprague-Dawley rats were randomly divided into control and catestatin groups. Ventricular arrhythmias were induced by ligation of the left anterior descending coronary artery and electrical stimulation. Action potential, transient outward potassium current, delayed rectifier potassium current, inward rectifying potassium current, and L-type calcium current (ICa-L) of rat ventricular myocytes were recorded using a patch-clamp technique. Catestatin notably reduced ventricular arrhythmia caused by myocardial ischemia/reperfusion and electrical stimulation of rats. In ventricular myocytes, catestatin markedly shortened the action potential duration of ventricular myocytes, which was counteracted by potassium channel antagonists TEACl and 4-AP, and ICa-L current channel agonist Bay K8644. In addition, catestatin significantly increased transient outward potassium current, delayed rectifier potassium current, and inward rectifying potassium current density in a concentration-dependent manner. Catestatin accelerated the activation and decelerated the inactivation of the transient outward potassium current channel. Furthermore, catestatin decreased ICa-L current density in a concentration-dependent manner. Catestatin also accelerated the inactivation of the ICa-L channel and slowed down the recovery of ICa-L from inactivation. CONCLUSIONS: Catestatin enhances the activity of transient outward potassium current, delayed rectifier potassium current, and inward rectifying potassium current, while suppressing the ICa-L in ventricular myocytes, leading to shortened action potential duration and ultimately reducing the ventricular arrhythmia in rats.


Subject(s)
Action Potentials , Chromogranin A , Myocytes, Cardiac , Peptide Fragments , Rats, Sprague-Dawley , Animals , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Chromogranin A/pharmacology , Chromogranin A/metabolism , Action Potentials/drug effects , Peptide Fragments/pharmacology , Calcium Channels, L-Type/metabolism , Calcium Channels, L-Type/drug effects , Arrhythmias, Cardiac/physiopathology , Arrhythmias, Cardiac/prevention & control , Arrhythmias, Cardiac/metabolism , Anti-Arrhythmia Agents/pharmacology , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Heart Ventricles/physiopathology , Potassium Channels, Inwardly Rectifying/metabolism , Potassium Channels, Inwardly Rectifying/drug effects , Disease Models, Animal , Potassium Channel Blockers/pharmacology , Rats , Patch-Clamp Techniques , Delayed Rectifier Potassium Channels/metabolism , Delayed Rectifier Potassium Channels/drug effects , Potassium Channels/metabolism , Potassium Channels/drug effects
6.
Mar Drugs ; 22(8)2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39195449

ABSTRACT

The relative lack of marine venom could be attributed to the difficulty in dealing with venomous marine animals. Moreover, the venom of marine animals consists of various bioactive molecules, many of which are proteins with unique properties. In this study, we investigated the potential toxic proteins of jellyfish collected for ligand screening to understand the mechanism of the toxic effects of jellyfish. Since taxonomic identification is problematic due to the lack of proper keys, we conducted morphological and molecular mitochondrial DNA sequencing from COI and ITS regions. The venom extract from nematocysts found along the bell margins was used for protein characterization using SDS-gel electrophoresis and nano-liquid chromatography-tandem mass spectrometry. Ligand screening for the most potent toxin and antibacterial and cytotoxicity assays were carried out. The phylogenetic tree showed distinct clustering from other Catostylus sp. The proteomic analysis revealed venom with many bioactive proteins. Only 13 venom proteins were identified with molecular weights ranging from 4318 to 184,923 Da, exhibiting the venom's complexity. The overall toxin protein composition of Catostylus sp. venom was dominated by potassium channel toxin alpha-KTx. Molecular docking of toxin alpha-KTx 1.13 revealed high specificity towards the human voltage-gated potassium channel Kv3 with a high fitness score and a minimum energy barrier of -17.9 kcal/mol. Disc diffusion and cytotoxicity assays revealed potent antibacterial activity against Klebsiella pneumoniae with no cytotoxicity. Further studies on detailed characterization and therapeutic potentials are warranted.


Subject(s)
Anti-Bacterial Agents , Cnidarian Venoms , Molecular Docking Simulation , Peptides , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Humans , Cnidarian Venoms/pharmacology , Cnidarian Venoms/chemistry , Peptides/pharmacology , Peptides/chemistry , Scyphozoa , Ligands , Phylogeny , Potassium Channels/drug effects , Potassium Channels/metabolism , Proteomics/methods
7.
J Plant Physiol ; 302: 154322, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39137481

ABSTRACT

Plants often face high salinity as a significant environmental challenge with roots being the first responders to this stress. Maintaining K+/Na+ ratio within plant cells is crucial for survival, as the intracellular K+ level decreases and the intracellular Na+ level increases under saline conditions. However, knowledge about the molecular regulatory mechanisms of K+ loss in response to salt stress through outward-rectifying K+ channels in plants is largely unknown. In this study, we found that the Arabidopsis double mutant gorkskor, in which the GORK and SKOR genes are disrupted, showed an improved primary root growth under salt stress compared to wild-type (WT) and the gork and skor single-mutant plants. No significant differences in the sensitivity to mannitol stress between the WT and gorkskor mutant were observed. Accumulation of ROS induced by salt stress was reduced in the gorkskor roots. The gorkskor mutant seedlings had significantly higher K+ content, lower Na+ content, and a greater resultant K+/Na+ ratio than the WT under salt stress. Moreover, salt-stress-induced elevation of cytosolic free Ca2+ concentration was reduced in the gorkskor roots. Taken together, these results suggest that Arabidopsis Shaker-type outward-rectifying K+ channels GORK and SKOR may redundantly function in regulation of primary root growth under salt stress and are involved in not only the late-stage response (e.g. K+ leakage) but also the early response including ROS production and [Ca2+]cyt elevation.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Plant Roots , Potassium Channels , Salt Stress , Arabidopsis/growth & development , Arabidopsis/genetics , Arabidopsis/physiology , Arabidopsis/metabolism , Plant Roots/growth & development , Plant Roots/metabolism , Plant Roots/drug effects , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Potassium Channels/metabolism , Potassium Channels/genetics , Potassium/metabolism , Sodium/metabolism , Reactive Oxygen Species/metabolism , Mutation , Shaker Superfamily of Potassium Channels
8.
Int J Mol Sci ; 25(16)2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39201487

ABSTRACT

Previous studies have suggested a role for selective serotonin reuptake inhibitors (SSRIs) such as fluoxetine (Prozac®) in the treatment of dizziness and inner ear vestibular dysfunction. The potential mechanism of action within the vestibular system remains unclear; however, fluoxetine has been reported to block certain types of K+ channel in other systems. Here, we investigated the direct actions of fluoxetine on membrane currents in presynaptic hair cells and postsynaptic calyx afferents of the gerbil peripheral vestibular system using whole cell patch clamp recordings in crista slices. We explored differences in K+ currents in peripheral zone (PZ) and central zone (CZ) calyces of the crista and their response to fluoxetine application. Outward K+ currents in PZ calyces showed greater inactivation at depolarized membrane potentials compared to CZ calyces. The application of 100 µM fluoxetine notably reduced K+ currents in calyx terminals within both zones of the crista, and the remaining currents exhibited distinct traits. In PZ cells, fluoxetine inhibited a non-inactivating K+ current and revealed a rapidly activating and inactivating K+ current, which was sensitive to blocking by 4-aminopyridine. This was in contrast to CZ calyces, where low-voltage-activated and non-inactivating K+ currents persisted following application of 100 µM fluoxetine. Additionally, marked inhibition of transient inward Na+ currents by fluoxetine was observed in calyces from both crista zones. Different concentrations of fluoxetine were tested, and the EC50 values were found to be 40 µM and 32 µM for K+ and Na+ currents, respectively. In contrast, 100 µM fluoxetine had no impact on voltage-dependent K+ currents in mechanosensory type I and type II vestibular hair cells. In summary, micromolar concentrations of fluoxetine are expected to strongly reduce both Na+ and K+ conductance in afferent neurons of the peripheral vestibular system in vivo. This would lead to inhibition of action potential firing in vestibular sensory neurons and has therapeutic implications for disorders of balance.


Subject(s)
Fluoxetine , Gerbillinae , Fluoxetine/pharmacology , Animals , Membrane Potentials/drug effects , Vestibule, Labyrinth/drug effects , Vestibule, Labyrinth/metabolism , Patch-Clamp Techniques , Selective Serotonin Reuptake Inhibitors/pharmacology , Potassium Channels/metabolism , Male , Hair Cells, Vestibular/drug effects , Hair Cells, Vestibular/metabolism
9.
Int J Mol Sci ; 25(16)2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39201565

ABSTRACT

The TRESK (K2P18.1, KCNK18) background potassium channel is expressed in primary sensory neurons and has been reported to contribute to the regulation of pain sensations. In the present study, we examined the interaction of TRESK with NDFIP1 (Nedd4 family-interacting protein 1) in the Xenopus oocyte expression system by two-electrode voltage clamp and biochemical methods. We showed that the coexpression of NDFIP1 abolished the TRESK current under the condition where the other K+ channels were not affected. Mutations in the three PPxY motifs of NDFIP1, which are responsible for the interaction with the Nedd4 ubiquitin ligase, prevented a reduction in the TRESK current. Furthermore, the overexpression of a dominant-negative Nedd4 construct in the oocytes coexpressing TRESK with NDFIP1 partially reversed the down-modulating effect of the adaptor protein on the K+ current. The biochemical data were also consistent with the functional results. An interaction between epitope-tagged versions of TRESK and NDFIP1 was verified by co-immunoprecipitation experiments. The coexpression of NDFIP1 with TRESK induced the ubiquitination of the channel protein. Altogether, the results suggest that TRESK is directly controlled by and highly sensitive to the activation of the NDFIP1-Nedd4 system. The NDFIP1-mediated reduction in the TRESK component may induce depolarization, increase excitability, and attenuate the calcium dependence of the membrane potential by reducing the calcineurin-activated fraction in the ensemble background K+ current.


Subject(s)
Carrier Proteins , Oocytes , Potassium Channels , Ubiquitination , Animals , Potassium Channels/metabolism , Potassium Channels/genetics , Oocytes/metabolism , Carrier Proteins/metabolism , Carrier Proteins/genetics , Humans , Membrane Proteins/metabolism , Membrane Proteins/genetics , Xenopus laevis , Nedd4 Ubiquitin Protein Ligases/metabolism , Nedd4 Ubiquitin Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Protein Binding , Potassium/metabolism , Xenopus Proteins
10.
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
11.
J Mol Neurosci ; 74(3): 69, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39017898

ABSTRACT

The objective of this investigation was to examine the impact of multiple exposures to general anesthesia (GA) with sevoflurane on the offspring of pregnant mice, as well as to elucidate the underlying mechanism. Neurodevelopmental assessments, including various reflexes and behavioral tests, were conducted on the offspring in the GA group to evaluate neuronal cell development. Furthermore, neonatal mouse neuronal cells were isolated and transfected with a high-expression CREB vector (pcDNA3.1-CREB), followed by treatment with sevoflurane (0.72 mol/L), ZD7288 (50 µmol/L), and KN-62 (10 µmol/L), or a combination of these compounds. The expression of relevant genes was then analyzed using qRT-PCR and western blot techniques. In comparison to the sham group, neonatal mice in the GA group exhibited significantly prolonged latencies in surface righting reflex, geotaxis test, and air righting reflex. Furthermore, there was a notable deceleration in the development of body weight and tail in the GA group. These mice also displayed impairments in social ability, reduced reciprocal social interaction behaviors, diminished learning capacity, and heightened levels of anxious behaviors. Additionally, synaptic trigger malfunction was observed, along with decreased production of c-Fos and neurotrophic factors. Sevoflurane was found to notably decrease cellular c-Fos and neurotrophic factor production, as well as the expression of HCN2 and CaMKII/CREB-related proteins. The inhibitory effects of sevoflurane on HCN2 or CaMKII channels were similar to those observed with ZD7288 or KN-62 inhibition. However, overexpression of CREB mitigated the impact of sevoflurane on neuronal cells. Repetitive exposure to sevoflurane general anesthesia while pregnant suppresses the CaMKII/CREB pathway, leading to the development of autism-like characteristics in offspring mice through the reduction of HCN2 expression.


Subject(s)
Anesthetics, Inhalation , Autistic Disorder , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Down-Regulation , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Prenatal Exposure Delayed Effects , Sevoflurane , Animals , Sevoflurane/pharmacology , Sevoflurane/toxicity , Mice , Pregnancy , Female , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Anesthetics, Inhalation/pharmacology , Anesthetics, Inhalation/toxicity , Anesthetics, Inhalation/adverse effects , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Autistic Disorder/genetics , Autistic Disorder/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP Response Element-Binding Protein/genetics , Potassium Channels/metabolism , Potassium Channels/genetics , Cells, Cultured , Neurons/metabolism , Neurons/drug effects , Male , Mice, Inbred C57BL
12.
Eur J Pharmacol ; 979: 176822, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39047965

ABSTRACT

BACKGROUND & AIMS: The treatment of cardiovascular diseases (CVD) could greatly benefit from using nitric oxide (NO) donors. This study aimed to investigate the mechanisms of action of NONO2P that contribute to the observed responses in the mesenteric artery. The hypothesis was that NONO2P would have similar pharmacological actions to sodium nitroprusside (SNP) and NO. METHODS: Male Wistar rats were euthanized to isolate the superior mesenteric artery for isometric tension recordings. NO levels were measured using the DAF-FM/DA dye, and cyclic guanosine monophosphate (cGMP) levels were determined using a cGMP-ELISA Kit. RESULTS: NONO2P presented a similar maximum efficacy to SNP. The free radical of NO (NO•) scavengers (PTIO; 100 µM and hydroxocobalamin; 30 µM) and nitroxyl anion (NO-) scavenger (L-cysteine; 3 mM) decreased relaxations promoted by NONO2P. The presence of the specific soluble guanylyl cyclase (sGC) inhibitor (ODQ; 10 µM) nearly abolished the vasorelaxation. The cGMP-dependent protein kinase (PKG) inhibition (KT5823; 1 µM) attenuated the NONO2P relaxant effect. The vasorelaxant response was significantly attenuated by blocking inward rectifying K+ channels (Kir), voltage-operated K+ channels (KV), and large conductance Ca2+-activated K+ channels (BKCa). NONO2P-induced relaxation was attenuated by cyclopiazonic acid (10 µM), indicating that sarcoplasmic reticulum Ca2+-ATPase (SERCA) activation is involved in this relaxation. Moreover, NONO2P increased NO levels in endothelial cells and cGMP production. CONCLUSIONS: NONO2P induces vasorelaxation with the same magnitude as SNP, releasing NO• and NO-. Its vasorelaxant effect involves sGC, PKG, K+ channels opening, and SERCA activation, suggesting its potential as a therapeutic option for CVD.


Subject(s)
Cyclic GMP-Dependent Protein Kinases , Cyclic GMP , Nitric Oxide Donors , Nitric Oxide , Potassium Channels , Rats, Wistar , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Signal Transduction , Soluble Guanylyl Cyclase , Vasodilation , Animals , Male , Vasodilation/drug effects , Nitric Oxide Donors/pharmacology , Nitric Oxide/metabolism , Soluble Guanylyl Cyclase/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Rats , Potassium Channels/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Cyclic GMP/metabolism , Signal Transduction/drug effects , Mesenteric Arteries/drug effects , Mesenteric Arteries/physiology , Guanylate Cyclase/metabolism , Enzyme Activation/drug effects
13.
Proc Natl Acad Sci U S A ; 121(31): e2310120121, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39058579

ABSTRACT

The axon initial segment (AIS) is a critical compartment in neurons. It converts postsynaptic input into action potentials that subsequently trigger information transfer to target neurons. This process relies on the presence of several voltage-gated sodium (NaV) and potassium (KV) channels that accumulate in high densities at the AIS. TRAAK is a mechanosensitive leak potassium channel that was recently localized to the nodes of Ranvier. Here, we uncover that TRAAK is also present in AISs of hippocampal and cortical neurons in the adult rat brain as well as in AISs of cultured rat hippocampal neurons. We show that the AIS localization is driven by a C-terminal ankyrin G-binding sequence that organizes TRAAK in a 190 nm spaced periodic pattern that codistributes with periodically organized ankyrin G. We furthermore uncover that while the identified ankyrin G-binding motif is analogous to known ankyrin G-binding motifs in NaV1 and KV7.2/KV7.3 channels, it was acquired by convergent evolution. Our findings identify TRAAK as an AIS ion channel that convergently acquired an ankyrin G-binding motif and expand the role of ankyrin G to include the nanoscale organization of ion channels at the AIS.


Subject(s)
Ankyrins , Axon Initial Segment , Hippocampus , Pyramidal Cells , Animals , Ankyrins/metabolism , Rats , Pyramidal Cells/metabolism , Axon Initial Segment/metabolism , Hippocampus/metabolism , Hippocampus/cytology , Axons/metabolism , Amino Acid Motifs , Potassium Channels/metabolism , Protein Binding
14.
PLoS Biol ; 22(7): e3002716, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39008526

ABSTRACT

Heterologous sensitization of adenylyl cyclase (AC) results in elevated cAMP signaling transduction that contributes to drug dependence. Inhibiting cullin3-RING ligases by blocking the neddylation of cullin3 abolishes heterologous sensitization, however, the modulating mechanism remains uncharted. Here, we report an essential role of the potassium channel tetramerization domain (KCTD) protein 2, 5, and 17, especially the dominant isoform KCTD5 in regulating heterologous sensitization of AC1 and morphine dependence via working with cullin3 and the cullin-associated and neddylation-dissociated 1 (CAND1) protein. In cellular models, we observed enhanced association of KCTD5 with Gß and cullin3, along with elevated dissociation of Gß from AC1 as well as of CAND1 from cullin3 in heterologous sensitization of AC1. Given binding of CAND1 inhibits the neddylation of cullin3, we further elucidated that the enhanced interaction of KCTD5 with both Gß and cullin3 promoted the dissociation of CAND1 from cullin3, attenuated the inhibitory effect of CAND1 on cullin3 neddylation, ultimately resulted in heterologous sensitization of AC1. The paraventricular thalamic nucleus (PVT) plays an important role in mediating morphine dependence. Through pharmacological and biochemical approaches, we then demonstrated that KCTD5/cullin3 regulates morphine dependence via modulating heterologous sensitization of AC, likely AC1 in PVT in mice. In summary, the present study revealed the underlying mechanism of heterologous sensitization of AC1 mediated by cullin3 and discovered the role of KCTD proteins in regulating morphine dependence in mice.


Subject(s)
Adenylyl Cyclases , Cullin Proteins , Morphine Dependence , Animals , Adenylyl Cyclases/metabolism , Adenylyl Cyclases/genetics , Cullin Proteins/metabolism , Mice , Morphine Dependence/metabolism , HEK293 Cells , Humans , Potassium Channels/metabolism , Potassium Channels/genetics , Mice, Inbred C57BL , Male , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein beta Subunits/genetics , Morphine/pharmacology , Mice, Knockout , Signal Transduction , Cyclic AMP/metabolism
15.
Mol Med Rep ; 30(3)2024 Sep.
Article in English | MEDLINE | ID: mdl-39027992

ABSTRACT

Triple­negative breast cancer (TNBC) is a highly aggressive and heterogeneous subtype of breast cancer that lacks expression of estrogen receptor, progesterone receptor, and HER2, making it more challenging to treat with targeted therapies. The present study aimed to identify CD8+ T cell­associated genes, which could provide insight into the mechanisms underlying TNBC to facilitate developing novel immunotherapies. TNBC datasets were downloaded from public databases including The Cancer Genome Atlas, Molecular Taxonomy of Breast Cancer International Consortium, and Gene Expression Omnibus. Candidate genes were identified integrating weighted gene co­expression network analysis (WGCNA), differential gene expression, protein­protein­interaction network construction and univariate Cox regression analysis. Kaplan­Meier survival, multivariate Cox regression and receiver operating characteristic analysis were performed to evaluate the prognostic value of hub genes. Knockdown experiments, alongside wound healing, Cell Counting Kit­8 and Transwell migration and invasion assays were performed. In total, seven gene modules were associated with CD8+ T cells using WGCNA, among which potassium channel tetramerization domain 5 (KCTD5) was significantly upregulated in TNBC samples and was associated with poor prognosis. KCTD5 expression inversely associated with infiltration ratios of 'Macrophages M1', 'Plasma cells', and 'γδ T cells', but positively with 'activated Mast cells', 'Macrophages M0', and 'Macrophages M2'. As an independent prognostic indicator for TNBC, KCTD5 was also associated with drug sensitivity and the expression of programmed cell death protein 1, Cytotoxic T­Lymphocyte­Associated Protein 4 (CTLA4), CD274), Cluster of Differentiation 86 (CD86), Lymphocyte­Activation Gene 3 (LAG3), T Cell Immunoreceptor with Ig and ITIM Domains (TIGIT). Knockdown of KCTD5 significantly inhibited viability, migration and invasion of TNBC cells in vitro. KCTD5 was suggested to impact the tumor immune microenvironment by influencing the infiltration of immune cells and may serve as a potential therapeutic target for TNBC.


Subject(s)
CD8-Positive T-Lymphocytes , Gene Expression Regulation, Neoplastic , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/mortality , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Female , Prognosis , Cell Line, Tumor , Potassium Channels/genetics , Potassium Channels/metabolism , Middle Aged , Biomarkers, Tumor/genetics , Disease Progression , Kaplan-Meier Estimate , Protein Interaction Maps , Cell Movement/genetics , Gene Regulatory Networks , Cell Proliferation
16.
J Am Chem Soc ; 146(29): 19896-19908, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-38982560

ABSTRACT

The disturbance of potassium current in cardiac myocytes caused by potassium channel dysfunction can lead to cardiac electrophysiological disorders, resulting in associated cardiovascular diseases. The emergence of artificial potassium ion channels opens up a way to replace dysfunctional natural ion channels and cure related diseases. However, bionic potassium ion channels have not been introduced into living cells to regulate cell function. One of the biggest challenges is that when the bionic channel fuses with the cell, it is difficult to control the inserting angle of the bionic potassium channel to ensure its penetration of the entire cell membrane. In nature, the extracellular vesicles can fuse with living cells with a completely preserved structure of vesicle protein. Inspired by this, we developed a vesicle fusion-based bionic porin (VFBP), which integrates bionic potassium ion channels into cardiomyocytes to replace damaged potassium ion channels. Theoretical and experimental results show that the inserted bionic ion channels have a potassium ion transport rate comparable to that of natural ion channels, which can restore the potassium ion outflow in cardiomyocytes and repair the abnormal action potential and excitation-contraction coupling of cardiomyocytes. Therefore, the bionic potassium ion channel system based on membrane fusion is expected to become the research object in many fields such as ultrafast ion transport, transmembrane delivery, and channelopathies treatment.


Subject(s)
Myocytes, Cardiac , Potassium Channels , Myocytes, Cardiac/metabolism , Potassium Channels/metabolism , Potassium Channels/chemistry , Humans , Potassium/metabolism , Potassium/chemistry , Animals , Porins/metabolism , Porins/chemistry
17.
Int J Mol Sci ; 25(13)2024 Jul 06.
Article in English | MEDLINE | ID: mdl-39000550

ABSTRACT

The effect of the modulators of the mitochondrial ATP-dependent potassium channel (mitoKATP) on the structural and biochemical alterations in the substantia nigra and brain tissues was studied in a rat model of Parkinson's disease induced by rotenone. It was found that, in experimental parkinsonism accompanied by characteristic motor deficits, both neurons and the myelin sheath of nerve fibers in the substantia nigra were affected. Changes in energy and ion exchange in brain mitochondria were also revealed. The nucleoside uridine, which is a source for the synthesis of the mitoKATP channel opener uridine diphosphate, was able to dose-dependently decrease behavioral disorders and prevent the death of animals, which occurred for about 50% of animals in the model. Uridine prevented disturbances in redox, energy, and ion exchanges in brain mitochondria, and eliminated alterations in their structure and the myelin sheath in the substantia nigra. Cytochemical examination showed that uridine restored the indicators of oxidative phosphorylation and glycolysis in peripheral blood lymphocytes. The specific blocker of the mitoKATP channel, 5-hydroxydecanoate, eliminated the positive effects of uridine, suggesting that this channel is involved in neuroprotection. Taken together, these findings indicate the promise of using the natural metabolite uridine as a new drug to prevent and, possibly, stop the progression of Parkinson's disease.


Subject(s)
Mitochondria , Potassium Channels , Rotenone , Uridine , Animals , Uridine/pharmacology , Uridine/metabolism , Rats , Potassium Channels/metabolism , Mitochondria/metabolism , Mitochondria/drug effects , Male , Disease Models, Animal , Parkinson Disease/metabolism , Parkinson Disease/drug therapy , Parkinson Disease/etiology , Parkinson Disease/pathology , Substantia Nigra/metabolism , Substantia Nigra/drug effects , Substantia Nigra/pathology , Neuroprotective Agents/pharmacology , Oxidative Phosphorylation/drug effects , Rats, Wistar , Decanoic Acids/pharmacology , Hydroxy Acids/pharmacology
18.
BMC Biol ; 22(1): 164, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39075558

ABSTRACT

BACKGROUND: The identification of novel toxins from overlooked and taxonomically exceptional species bears potential for various pharmacological applications. The remipede Xibalbanus tulumensis, an underwater cave-dwelling crustacean, is the only crustacean for which a venom system has been described. Its venom contains several xibalbin peptides that have an inhibitor cysteine knot (ICK) scaffold. RESULTS: Our screenings revealed that all tested xibalbin variants particularly inhibit potassium channels. Xib1 and xib13 with their eight-cysteine domain similar to spider knottins also inhibit voltage-gated sodium channels. No activity was noted on calcium channels. Expanding the functional testing, we demonstrate that xib1 and xib13 increase PKA-II and Erk1/2 sensitization signaling in nociceptive neurons, which may initiate pain sensitization. Our phylogenetic analysis suggests that xib13 either originates from the common ancestor of pancrustaceans or earlier while xib1 is more restricted to remipedes. The ten-cysteine scaffolded xib2 emerged from xib1, a result that is supported by our phylogenetic and machine learning-based analyses. CONCLUSIONS: Our functional characterization of synthesized variants of xib1, xib2, and xib13 elucidates their potential as inhibitors of potassium channels in mammalian systems. The specific interaction of xib2 with Kv1.6 channels, which are relevant to treating variants of epilepsy, shows potential for further studies. At higher concentrations, xib1 and xib13 activate the kinases PKA-II and ERK1/2 in mammalian sensory neurons, suggesting pain sensitization and potential applications related to pain research and therapy. While tested insect channels suggest that all probably act as neurotoxins, the biological function of xib1, xib2, and xib13 requires further elucidation. A novel finding on their evolutionary origin is the apparent emergence of X. tulumensis-specific xib2 from xib1. Our study is an important cornerstone for future studies to untangle the origin and function of these enigmatic proteins as important components of remipede but also other pancrustacean and arthropod venoms.


Subject(s)
Potassium Channels , Animals , Potassium Channels/metabolism , Potassium Channels/genetics , MAP Kinase Signaling System/drug effects , Phylogeny , Mice , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Evolution, Molecular , Humans , Arthropod Venoms/chemistry
19.
FEBS Lett ; 598(16): 1955-1966, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38880762

ABSTRACT

This study investigated how membrane thickness and tension modify the gating of KcsA potassium channels when simultaneously varied. The KcsA channel undergoes global conformational changes upon gating: expansion of the cross-sectional area and longitudinal shortening upon opening. Thus, membranes impose differential effects on the open and closed conformations, such as hydrophobic mismatches. Here, the single-channel open probability was recorded in the contact bubble bilayer, by which variable thickness membranes under a defined tension were applied. A fully open channel in thin membranes turned to sporadic openings in thick membranes, where the channel responded moderately to tension increase. Quantitative gating analysis prompted the hypothesis that tension augmented the membrane deformation energy when hydrophobic mismatch was enhanced in thick membranes.


Subject(s)
Bacterial Proteins , Ion Channel Gating , Potassium Channels , Potassium Channels/metabolism , Potassium Channels/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Lipid Bilayers/metabolism , Lipid Bilayers/chemistry , Cell Membrane/metabolism , Cell Membrane/chemistry , Hydrophobic and Hydrophilic Interactions , Protein Conformation
20.
Neurochem Int ; 178: 105797, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38942118

ABSTRACT

K+ channels exist in all living systems. They allow a selective transition to the K+ ion, which enables the activity of various vital tissues such as muscle cells, neurons, and even bacteria and plants. Despite the mechanism variation in the gating process of K+ channels in different tissues, the selectivity for the K+ ion is preserved and the electrochemical cascade is maintained in these tissues. The electrochemical gradient of the K+ ion is very close to the diffusion rate of K+ ions in bulk water. On the molecular level, how does a K+ ion move across the ion conduction pathway? There are many molecular models that describe and answer this question, however, this is rarely described on the macro level. Here, a physical model can serve as a very good basis for enabling a deeper understanding of the K+ ion for ion transport. Classical physical energy and linear and angular momentum laws can provide a good explanation as to how and what happens to K+ ions when they pass through an ion conduction pathway. This model describes the passage of the ion even before it enters the ion conduction path until the last ion at the end exits. The simulation described here is fascinating and depicts the state of the ion at the farthest end released at almost the same speed as the first ion initially, while all the other ions remain almost at rest. How does this occur? What happens if we change the size or mass of the ion? In this work, I describe this principle and the related problems that could be studied.


Subject(s)
Potassium Channels , Potassium , Potassium/metabolism , Potassium Channels/metabolism , Potassium Channels/chemistry , Ion Channel Gating/physiology , Humans , Animals , Ion Transport/physiology
SELECTION OF CITATIONS
SEARCH DETAIL