Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 541
1.
Nature ; 621(7977): 146-153, 2023 Sep.
Article En | MEDLINE | ID: mdl-37648853

Learning and memory are thought to require hippocampal long-term potentiation (LTP), and one of the few central dogmas of molecular neuroscience that has stood undisputed for more than three decades is that LTP induction requires enzymatic activity of the Ca2+/calmodulin-dependent protein kinase II (CaMKII)1-3. However, as we delineate here, the experimental evidence is surprisingly far from conclusive. All previous interventions inhibiting enzymatic CaMKII activity and LTP4-8 also interfere with structural CaMKII roles, in particular binding to the NMDA-type glutamate receptor subunit GluN2B9-14. Thus, we here characterized and utilized complementary sets of new opto-/pharmaco-genetic tools to distinguish between enzymatic and structural CaMKII functions. Several independent lines of evidence demonstrated LTP induction by a structural function of CaMKII rather than by its enzymatic activity. The sole contribution of kinase activity was autoregulation of this structural role via T286 autophosphorylation, which explains why this distinction has been elusive for decades. Directly initiating the structural function in a manner that circumvented this T286 role was sufficient to elicit robust LTP, even when enzymatic CaMKII activity was blocked.


Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Long-Term Potentiation , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/chemistry , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Glutamic Acid/metabolism , Hippocampus/physiology , Learning/physiology , Long-Term Potentiation/physiology , Optogenetics , Phosphorylation , Protein Binding
2.
Sci Transl Med ; 15(701): eabq7839, 2023 06 21.
Article En | MEDLINE | ID: mdl-37343080

Ca2+/calmodulin-dependent protein kinase II (CaMKII) hyperactivity causes cardiac arrhythmias, a major source of morbidity and mortality worldwide. Despite proven benefits of CaMKII inhibition in numerous preclinical models of heart disease, translation of CaMKII antagonists into humans has been stymied by low potency, toxicity, and an enduring concern for adverse effects on cognition due to an established role of CaMKII in learning and memory. To address these challenges, we asked whether any clinically approved drugs, developed for other purposes, were potent CaMKII inhibitors. For this, we engineered an improved fluorescent reporter, CaMKAR (CaMKII activity reporter), which features superior sensitivity, kinetics, and tractability for high-throughput screening. Using this tool, we carried out a drug repurposing screen (4475 compounds in clinical use) in human cells expressing constitutively active CaMKII. This yielded five previously unrecognized CaMKII inhibitors with clinically relevant potency: ruxolitinib, baricitinib, silmitasertib, crenolanib, and abemaciclib. We found that ruxolitinib, an orally bioavailable and U.S. Food and Drug Administration-approved medication, inhibited CaMKII in cultured cardiomyocytes and in mice. Ruxolitinib abolished arrhythmogenesis in mouse and patient-derived models of CaMKII-driven arrhythmias. A 10-min pretreatment in vivo was sufficient to prevent catecholaminergic polymorphic ventricular tachycardia, a congenital source of pediatric cardiac arrest, and rescue atrial fibrillation, the most common clinical arrhythmia. At cardioprotective doses, ruxolitinib-treated mice did not show any adverse effects in established cognitive assays. Our results support further clinical investigation of ruxolitinib as a potential treatment for cardiac indications.


Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Heart Diseases , Animals , Child , Humans , Mice , Arrhythmias, Cardiac , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Heart Diseases/metabolism , Myocytes, Cardiac/metabolism , Pyrazoles/pharmacology
3.
J Biol Chem ; 299(5): 104693, 2023 05.
Article En | MEDLINE | ID: mdl-37037305

The Ca2+/calmodulin-dependent protein kinase II (CaMKII) is a central regulator of learning and memory, which poses a problem for targeting it therapeutically. Indeed, our study supports prior conclusions that long-term interference with CaMKII signaling can erase pre-formed memories. By contrast, short-term pharmacological CaMKII inhibition with the neuroprotective peptide tatCN19o interfered with learning in mice only mildly and transiently (for less than 1 h) and did not at all reverse pre-formed memories. These results were obtained with ≥500-fold of the dose that protected hippocampal neurons from cell death after a highly clinically relevant pig model of transient global cerebral ischemia: ventricular fibrillation followed by advanced life support and electrical defibrillation to induce the return of spontaneous circulation. Of additional importance for therapy development, our preliminary cardiovascular safety studies in mice and pig did not indicate any concerns with acute tatCN19o injection. Taken together, although prolonged interference with CaMKII signaling can erase memory, acute short-term CaMKII inhibition with tatCN19o did not cause such retrograde amnesia that would pose a contraindication for therapy.


Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Memory , Animals , Mice , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Hippocampus/metabolism , Memory/drug effects , Memory/physiology , Neurons/metabolism , Phosphorylation/physiology , Swine , Peptides/pharmacology
4.
J Med Chem ; 65(22): 15066-15084, 2022 11 24.
Article En | MEDLINE | ID: mdl-36346645

Ca2+/calmodulin-dependent protein kinase II alpha (CaMKIIα) is a brain-relevant kinase and an emerging drug target for ischemic stroke and neurodegenerative disorders. Despite reported CaMKIIα inhibitors, their usefulness is limited by low subtype selectivity and brain permeability. (E)-2-(5-Hydroxy-5,7,8,9-tetrahydro-6H-benzo[7]annulen-6-ylidene)acetic acid (NCS-382) is structurally related to the proposed neuromodulator, γ-hydroxybutyric acid, and is a brain-penetrating high nanomolar-affinity ligand selective for the CaMKIIα hub domain. Herein, we report the first series of NCS-382 analogs displaying improved affinity and preserved brain permeability. Specifically, we present Ph-HTBA (1i) with enhanced mid-nanomolar affinity for the CaMKIIα binding site and a marked hub thermal stabilization effect along with a distinct CaMKIIα Trp403 flip upon binding. Moreover, Ph-HTBA has good cellular permeability and low microsomal clearance and shows brain permeability after systemic administration to mice, signified by a high Kp, uu value (0.85). Altogether, our study highlights Ph-HTBA as a promising candidate for CaMKIIα-associated pharmacological interventions and future clinical development.


Benzocycloheptenes , Brain , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Animals , Mice , Benzocycloheptenes/pharmacology , Binding Sites , Brain/metabolism , Protein Binding , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors
5.
Cell Rep ; 38(1): 110153, 2022 01 04.
Article En | MEDLINE | ID: mdl-34986356

Synaptic plasticity is long-lasting changes in synaptic currents and structure. When neurons are exposed to signals that induce aberrant neuronal excitation, they increase the threshold for the induction of long-term potentiation (LTP), known as metaplasticity. However, the metaplastic regulation of structural LTP (sLTP) remains unclear. We investigate glutamate uncaging/photoactivatable (pa)CaMKII-dependent sLTP induction in hippocampal CA1 neurons after chronic neuronal excitation by GABAA receptor antagonists. We find that the neuronal excitation decreases the glutamate uncaging-evoked Ca2+ influx mediated by GluN2B-containing NMDA receptors and suppresses sLTP induction. In addition, single-spine optogenetic stimulation using paCaMKII indicates the suppression of CaMKII signaling. While the inhibition of Ca2+ influx is protein synthesis independent, the paCaMKII-induced sLTP suppression depends on it. Our findings demonstrate that chronic neuronal excitation suppresses sLTP in two independent ways (i.e., dual inhibition of Ca2+ influx and CaMKII signaling). This dual inhibition mechanism may contribute to robust neuronal protection in excitable environments.


Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Long-Term Potentiation/physiology , Neuronal Plasticity/physiology , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/physiology , Animals , CA1 Region, Hippocampal/metabolism , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Cell Line , Dendritic Spines/metabolism , GABA-A Receptor Antagonists/pharmacology , Glutamic Acid/metabolism , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Receptors, GABA-A/metabolism , Signal Transduction/physiology
6.
J Cell Mol Med ; 26(2): 312-325, 2022 01.
Article En | MEDLINE | ID: mdl-34845819

Persistent cardiac Ca2+ /calmodulin-dependent Kinase II (CaMKII) activation was considered to promote heart failure (HF) development, some studies believed that CaMKII was a target for therapy of HF. However, CaMKII was an important mediator for the ischaemia-induced coronary angiogenesis, and new evidence confirmed that angiogenesis inhibited cardiac remodelling and improved heart function, and some conditions which impaired angiogenesis aggravated ventricular remodelling. This study aimed to investigate the roles and the underlying mechanisms of CaMKII inhibitor in cardiac remodelling. First, we induced cardiac remodelling rat model by ISO, pre-treated by CaMKII inhibitor KN-93, evaluated heart function by echocardiography measurements, and performed HE staining, Masson staining, Tunel staining, Western blot and RT-PCR to test cardiac remodelling and myocardial microvessel density; we also observed ultrastructure of cardiac tissue with transmission electron microscope. Second, we cultured HUVECs, pre-treated by ISO and KN-93, detected cell proliferation, migration, tubule formation and apoptosis, and carried out Western blot to determine the expression of NOX2, NOX4, VEGF, VEGFR2, p-VEGFR2 and STAT3; mtROS level was also measured. In vivo, we found KN-93 severely reduced myocardial microvessel density, caused apoptosis of vascular endothelial cells, enhanced cardiac hypertrophy, myocardial apoptosis, collagen deposition, aggravated the deterioration of myocardial ultrastructure and heart function. In vitro, KN-93 inhibited HUVECs proliferation, migration and tubule formation, and promoted apoptosis of HUVECs. The expression of NOX2, NOX4, p-VEGFR2 and STAT3 were down-regulated by KN-93; mtROS level was severely reduced by KN-93. We concluded that KN-93 impaired angiogenesis and aggravated cardiac remodelling and heart failure via inhibiting NOX2/mtROS/p-VEGFR2 and STAT3 pathways.


Benzylamines , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Heart Failure , Sulfonamides , Ventricular Remodeling , Animals , Benzylamines/adverse effects , Benzylamines/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Endothelial Cells/metabolism , Myocytes, Cardiac/metabolism , NADPH Oxidase 2 , Neovascularization, Physiologic/drug effects , Rats , STAT3 Transcription Factor/metabolism , Sulfonamides/adverse effects , Sulfonamides/pharmacology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Ventricular Remodeling/drug effects
7.
Eur J Pharmacol ; 916: 174603, 2022 Feb 05.
Article En | MEDLINE | ID: mdl-34793771

Acute kidney injury (AKI) is a serious threat to human health. Clinically, ischemia-reperfusion (I/R) injury is considered one of the most common contributors to AKI. Emodin has been reported to alleviate I/R injury in the heart, brain, and small intestine in rats and mice through its anti-inflammatory effects. The present study investigated whether emodin improved AKI induced by I/R and elucidated the molecular mechanisms. We used a mouse model of renal I/R injury and human renal tubular epithelial cell model of hypoxia/reoxygenation (H/R) injury. Ischemia/reperfusion resulted in renal dysfunction. Pretreatment with emodin ameliorated renal injury in mice following I/R injury. Emodin reduced mitochondrial-mediated apoptosis, suppressed the overproduction of mitochondrial reactive oxygen species and accelerated the recovery of adenosine triphosphate both in vivo and in vitro. Emodin prevented mitochondrial fission and restored the balance of mitochondrial dynamics. The phosphorylation of dynamin-related protein 1 (DRP1) at Ser616, a master regulator of mitochondrial fission, was upregulated in both models of I/R and H/R injury, and this upregulation was blocked by emodin. Using computational cognate protein kinase prediction and specific kinase inhibitors, we found that emodin inhibited the phosphorylation of calcium/calmodulin-dependent protein kinase II (https://www.guidetopharmacology.org/GRAC/ObjectDisplayForward?objectId=1554), thereby inhibiting its kinase activity and reducing the phosphorylation of DRP1 at Ser616. The results demonstrated that emodin pretreatment could protect renal function by improving mitochondrial dysfunction induced by I/R.


Acute Kidney Injury/prevention & control , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Dynamins/antagonists & inhibitors , Emodin/pharmacology , Mitochondrial Dynamics/drug effects , Protective Agents/pharmacology , Reperfusion Injury/prevention & control , Acute Kidney Injury/pathology , Animals , Apoptosis/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Line , Disease Models, Animal , Dynamins/metabolism , Emodin/therapeutic use , Humans , Male , Mice, Inbred C57BL , Mitochondria/drug effects , Oxidative Stress/drug effects , Phosphorylation/drug effects , Protective Agents/therapeutic use , Reperfusion Injury/pathology
8.
Oxid Med Cell Longev ; 2021: 4187398, 2021.
Article En | MEDLINE | ID: mdl-34840668

As a calcium-regulated protein, CaMK II is closely related to cell death, and it participates in the development of pathological processes such as reperfusion injury, myocardial infarction, and oligodendrocyte death. The function of CaMK II activation in acute pancreatitis (AP) remains unclear. In our study, we confirmed that the expression of p-CaMK II was increased significantly and consistently in injured pancreatic tissues after caerulein-induced AP. Then, we found that KN93, an inhibitor of CaMK II, could mitigate the histopathological manifestations in pancreatic tissues, reduce serum levels of enzymology, and decrease oxidative stress products. Accordingly, we elucidated the effect of KN93 in vitro and found that KN93 had a protective effect on the pancreatic acinar cell necroptosis pathway by inhibiting the production of ROS and decreasing the expression of RIP3 and p-MLKL. In addition, we identified the protective effect of KN93 on AP through another mouse model induced by pancreatic duct ligation (PDL). Together, these data demonstrated that CaMK II participates in the development of AP and that inhibiting CaMK II activation could protect against AP by reducing acinar cell necroptosis, which may provide a new idea target for the prevention and treatment of AP in the clinic.


Acinar Cells/drug effects , Benzylamines/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Necroptosis , Pancreatitis/prevention & control , Protective Agents/pharmacology , Reactive Oxygen Species/metabolism , Sulfonamides/pharmacology , Acinar Cells/metabolism , Acinar Cells/pathology , Animals , Ceruletide/toxicity , Male , Mice , Mice, Inbred C57BL , Pancreatitis/chemically induced , Pancreatitis/metabolism , Pancreatitis/pathology
9.
Cells ; 10(11)2021 11 05.
Article En | MEDLINE | ID: mdl-34831263

Both, the decreased L-type Ca2+ current (ICa,L) density and increased spontaneous Ca2+ release from the sarcoplasmic reticulum (SR), have been associated with atrial fibrillation (AF). In this study, we tested the hypothesis that remodeling of 3',5'-cyclic adenosine monophosphate (cAMP)-dependent protein kinase A (PKA) signaling is linked to these compartment-specific changes (up- or down-regulation) in Ca2+-handling. Perforated patch-clamp experiments were performed in atrial myocytes from 53 patients with AF and 104 patients in sinus rhythm (Ctl). A significantly higher frequency of transient inward currents (ITI) activated by spontaneous Ca2+ release was confirmed in myocytes from AF patients. Next, inhibition of PKA by H-89 promoted a stronger effect on the ITI frequency in these myocytes compared to myocytes from Ctl patients (7.6-fold vs. 2.5-fold reduction), while the ß-agonist isoproterenol (ISO) caused a greater increase in Ctl patients (5.5-fold vs. 2.1-fold). ICa,L density was larger in myocytes from Ctl patients at baseline (p < 0.05). However, the effect of ISO on ICa,L density was only slightly stronger in AF than in Ctl myocytes (3.6-fold vs. 2.7-fold). Interestingly, a significant reduction of ICa,L and Ca2+ sparks was observed upon Ca2+/Calmodulin-dependent protein kinase II inhibition by KN-93, but this inhibition had no effect on ITI. Fluorescence resonance energy transfer (FRET) experiments showed that although AF promoted cytosolic desensitization to ß-adrenergic stimulation, ISO increased cAMP to similar levels in both groups of patients in the L-type Ca2+ channel and ryanodine receptor compartments. Basal cAMP signaling also showed compartment-specific regulation by phosphodiesterases in atrial myocytes from 44 Ctl and 43 AF patients. Our results suggest that AF is associated with opposite changes in compartmentalized PKA/cAMP-dependent regulation of ICa,L (down-regulation) and ITI (up-regulation).


Atrial Fibrillation/metabolism , Calcium Signaling , Cyclic AMP/metabolism , Adrenergic beta-Antagonists/pharmacology , Aged , Animals , Calcium/metabolism , Calcium Channels, L-Type/metabolism , Calcium Signaling/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Carvedilol/pharmacology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytosol/drug effects , Cytosol/metabolism , Female , Humans , Male , Middle Aged , Receptors, Adrenergic, beta/metabolism , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism
10.
Oxid Med Cell Longev ; 2021: 6673967, 2021.
Article En | MEDLINE | ID: mdl-34527176

Vascular dementia (VaD) is a common cause of cognitive decline and dementia of vascular origin, but the precise pathological mechanisms are unknown, and so effective clinical treatments have not been established. Tilianin, the principal active compound of total flavonoid extract from Dracocephalum moldavica L., is a candidate therapy for cardio-cerebrovascular diseases in China. However, its potential in the treatment of VaD is unclear. The present study is aimed at investigating the protective effects of tilianin on VaD and exploring the underlying mechanism of the action. A model of VaD was established by permanent 2-vessel occlusion (2VO) in rats. Human neurons (hNCs) differentiated from human-induced pluripotent stem cells were used to establish an oxygen-glucose deprivation (OGD) model. The therapeutic effects and potential mechanisms of tilianin were identified using behavioral tests, histochemistry, and multiple molecular biology techniques such as Western blot analysis and gene silencing. The results demonstrated that tilianin modified spatial cognitive impairment, neurodegeneration, oxidation, and apoptosis in rats with VaD and protected hNCs against OGD by increasing cell viability and decreasing apoptosis rates. A study of the mechanism indicated that tilianin restored p-CaMKII/ERK1/2/CREB signaling in the hippocampus, maintaining hippocampus-independent memory. In addition, tilianin inhibited an ox-CaMKII/p38 MAPK/JNK/NF-κB associated inflammatory response caused by cerebral oxidative stress imbalance in rats with VaD. Furthermore, specific CaMKIIα siRNA action revealed that tilianin-exerted neuroprotection involved increase of neuronal viability, inhibition of apoptosis, and suppression of inflammation, which was dependent on CaMKIIα. In conclusion, the results suggested the neuroprotective effect of tilianin in VaD and the potential mechanism associated with dysfunction in the regulation of p-CaMKII-mediated long-term memory and oxidation and inflammation involved with ox-CaMKII, which may lay the foundation for clinical trials of tilianin for the treatment of VaD in the future.


Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Flavonoids/pharmacology , Glycosides/pharmacology , Neuroprotective Agents/pharmacology , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Dementia, Vascular/drug therapy , Dementia, Vascular/metabolism , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Flavonoids/chemistry , Flavonoids/therapeutic use , Glycosides/chemistry , Glycosides/therapeutic use , Hippocampus/metabolism , Humans , Lamiaceae/chemistry , Lamiaceae/metabolism , Male , Maze Learning/drug effects , NF-kappa B/metabolism , Neuroprotective Agents/chemistry , Neuroprotective Agents/therapeutic use , RNA Interference , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley
11.
Cells ; 10(9)2021 09 12.
Article En | MEDLINE | ID: mdl-34572045

Receptor interacting protein kinase 3 (RIPK3)-mediated smooth muscle cell (SMC) necroptosis has been shown to contribute to the pathogenesis of abdominal aortic aneurysms (AAAs). However, the signaling steps downstream from RIPK3 during SMC necroptosis remain unknown. In this study, the roles of mixed lineage kinase domain-like pseudokinase (MLKL) and calcium/calmodulin-dependent protein kinase II (CaMKII) in SMC necroptosis were investigated. We found that both MLKL and CaMKII were phosphorylated in SMCs in a murine CaCl2-driven model of AAA and that Ripk3 deficiency reduced the phosphorylation of MLKL and CaMKII. In vitro, mouse aortic SMCs were treated with tumor necrosis factor α (TNFα) plus Z-VAD-FMK (zVAD) to induce necroptosis. Our data showed that both MLKL and CaMKII were phosphorylated after TNFα plus zVAD treatment in a time-dependent manner. SiRNA silencing of Mlkl-diminished cell death and administration of the CaMKII inhibitor myristoylated autocamtide-2-related inhibitory peptide (Myr-AIP) or siRNAs against Camk2d partially inhibited necroptosis. Moreover, knocking down Mlkl decreased CaMKII phosphorylation, but silencing Camk2d did not affect phosphorylation, oligomerization, or trafficking of MLKL. Together, our results indicate that both MLKL and CaMKII are involved in RIPK3-mediated SMC necroptosis, and that MLKL is likely upstream of CaMKII in this process.


Aortic Aneurysm, Abdominal/pathology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Myocytes, Smooth Muscle/pathology , Necrosis , Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Animals , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/metabolism , Calcium Chloride/toxicity , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Smooth Muscle/metabolism , Phosphorylation , Protein Kinases/chemistry , Protein Kinases/genetics , RNA, Small Interfering/genetics , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
12.
J Pharmacol Sci ; 147(1): 86-94, 2021 Sep.
Article En | MEDLINE | ID: mdl-34294377

We examined the effects of neurotensin (NTS) on the excitability of type II neurons in the rat dorsolateral bed nucleus of the stria terminalis (dlBNST) using whole-cell patch-clamp electrophysiology. Bath-application of NTS depolarized type II dlBNST neurons. Analyses of the steady-state I-V relationships implied that the depolarizing effect of NTS is due to potassium conductance blocking. The depolarizing effect of NTS was abolished in the presence of a PLC inhibitor, but not affected by a protein kinase C inhibitor. In the presence of a CaMKII inhibitor, NTS showed depolarizing effects via the increase in non-selective cation conductance in addition to the decrease in potassium conductance. Unexpectedly, in the presence of a PKA inhibitor, NTS hyperpolarized type II dlBNST neurons. These results reveal that diverse signaling pathways mediate the effects of NTS on the excitability of type II dlBNST neurons. The elevation of intracellular Ca2+ levels via the inositol phosphate-mediated signaling activates both Ca2+-dependent adenylate cyclase (AC) and CaMKII. Activation of the AC-cAMP-PKA pathway exerts depolarizing effects on type II dlBNST neurons by decreasing potassium conductance and increasing non-selective cation conductance, whereas activation of the CaMKII pathway exerts hyperpolarizing effects on dlBNST neurons by decreasing non-selective cation conductance.


Neurons/drug effects , Neurons/physiology , Neurotensin/pharmacology , Septal Nuclei/cytology , Signal Transduction/physiology , Synaptic Transmission/drug effects , Adenylyl Cyclases/metabolism , Animals , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cells, Cultured , Male , Patch-Clamp Techniques/methods , Rats, Sprague-Dawley
13.
Exp Mol Med ; 53(7): 1134-1147, 2021 07.
Article En | MEDLINE | ID: mdl-34234278

Calbindin, a major Ca2+ buffer in dentate granule cells (GCs), plays a critical role in shaping Ca2+ signals, yet how it regulates neuronal function remains largely unknown. Here, we found that calbindin knockout (CBKO) mice exhibited dentate GC hyperexcitability and impaired pattern separation, which co-occurred with reduced K+ current due to downregulated surface expression of Kv4.1. Relatedly, manipulation of calbindin expression in HT22 cells led to changes in CaMKII activation and the level of surface localization of Kv4.1 through phosphorylation at serine 555, confirming the mechanism underlying neuronal hyperexcitability in CBKO mice. We also discovered that Ca2+ buffering capacity was significantly reduced in the GCs of Tg2576 mice to the level of CBKO GCs, and this reduction was restored to normal levels by antioxidants, suggesting that calbindin is a target of oxidative stress. Our data suggest that the regulation of CaMKII signaling by Ca2+ buffering is crucial for neuronal excitability regulation.


Calbindins/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Dentate Gyrus/metabolism , Animals , Antioxidants/pharmacology , Benzylamines/pharmacology , Calbindins/genetics , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Conditioning, Psychological , Dentate Gyrus/cytology , Dentate Gyrus/drug effects , Fear/physiology , HT29 Cells , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Protein Transport , Serine/metabolism , Sulfonamides/pharmacology
14.
Peptides ; 144: 170609, 2021 10.
Article En | MEDLINE | ID: mdl-34242679

Cell pyroptosis, a new type of programmed cell death, has been recently reported to play important roles in the development of cardiac remodeling. How cardiomyocyte pyroptosis is induced remains to be elucidated. Urotensin II (UII) has been known closely related to cardiac remodeling and the development of heart failure. Inhibition of UII receptors has been shown to be effective in the treatment of cardiac hypertrophy and remodeling. However, it is not clear whether UII might induce cardiomyocyte pyroptosis. We here examined the effect of UII treatment on pyroptosis in cultured cardiomyocytes. Treatment of cardiomyocyes of neonatal rats with UII (500 nmol/l) for 48 hours induced a significant pyroptosis as evidenced by not only increased cell death but also upregulated expression levels of NLR family pyrin domain containing 3 (NLRP3), caspase-1, IL-1ß, IL-18 and gasdermin D (GMDSD)-N which are important markers for the identification of cell pyroptosis. All these pyroptosis responses induced by UII were abrogated by an inhibitor of NLRP3. Moreover, the antagonist of UII receptor, Urantide abolished UII- induced cardiomyocyte pyroptosis. Additionally, inhibition of calcineurin by cyclosporin A rather than that of CaMKII by KN93 suppressed the UII-upregulated expression levels of those pyroptosis markers. We therefore demonstrate that UII might induce cardiomyocyte pyroptosis through calcineurin.


Calcineurin/metabolism , Myocytes, Cardiac/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pyroptosis/drug effects , Urotensins/pharmacology , Animals , Benzylamines/pharmacology , Calcineurin Inhibitors/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Cells, Cultured , Cyclosporine/pharmacology , Myocytes, Cardiac/metabolism , Peptide Fragments/pharmacology , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/antagonists & inhibitors , Sulfonamides/pharmacology
15.
Am J Physiol Heart Circ Physiol ; 321(2): H446-H460, 2021 08 01.
Article En | MEDLINE | ID: mdl-34270372

In conditions with abnormally increased activity of the cardiac ryanodine receptor (RyR2), Ca2+/calmodulin-dependent protein kinase II (CaMKII) can contribute to a further destabilization of RyR2 that results in triggered arrhythmias. Therefore, inhibition of CaMKII in such conditions has been suggested as a strategy to suppress RyR2 activity and arrhythmias. However, suppression of RyR2 activity can lead to the development of arrhythmogenic Ca2+ alternans. The aim of this study was to test whether the suppression of RyR2 activity caused by inhibition of CaMKII increases propensity for Ca2+ alternans. We studied spontaneous Ca2+ release events and Ca2+ alternans in isolated left ventricular cardiomyocytes from mice carrying the gain-of-function RyR2 mutation RyR2-R2474S and from wild-type mice. CaMKII inhibition by KN-93 effectively decreased the frequency of spontaneous Ca2+ release events in RyR2-R2474S cardiomyocytes exposed to the ß-adrenoceptor agonist isoprenaline. However, KN-93-treated RyR2-R2474S cardiomyocytes also showed increased propensity for Ca2+ alternans and increased Ca2+ alternans ratio compared with both an inactive analog of KN-93 and with vehicle-treated controls. This increased propensity for Ca2+ alternans was explained by prolongation of Ca2+ release refractoriness. Importantly, the increased propensity for Ca2+ alternans in KN-93-treated RyR2-R2474S cardiomyocytes did not surpass that of wild type. In conclusion, inhibition of CaMKII efficiently reduces spontaneous Ca2+ release but promotes Ca2+ alternans in RyR2-R2474S cardiomyocytes with a gain-of-function RyR2 mutation. The dominant effect in RyR2-R2474S is to reduce spontaneous Ca2+ release, which supports this intervention as a therapeutic strategy in this specific condition. However, future studies on CaMKII inhibition in conditions with increased propensity for Ca2+ alternans should include investigation of both phenomena.NEW & NOTEWORTHY Genetically increased RyR2 activity promotes arrhythmogenic Ca2+ release. Inhibition of CaMKII suppresses RyR2 activity and arrhythmogenic Ca2+ release. Suppression of RyR2 activity prolongs refractoriness of Ca2+ release. Prolonged refractoriness of Ca2+ release leads to arrhythmogenic Ca2+ alternans. CaMKII inhibition promotes Ca2+ alternans by prolonging Ca2+ release refractoriness.


Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium/metabolism , Myocytes, Cardiac/drug effects , Protein Kinase Inhibitors/pharmacology , Ryanodine Receptor Calcium Release Channel/genetics , Sarcoplasmic Reticulum/drug effects , Tachycardia, Ventricular/genetics , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Adrenergic beta-Agonists/pharmacology , Animals , Arrhythmias, Cardiac/metabolism , Benzylamines/pharmacology , Calcium Channel Agonists/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Gain of Function Mutation , Heart Ventricles/cytology , Isoproterenol/pharmacology , Mice , Myocytes, Cardiac/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Sarcoplasmic Reticulum/metabolism , Sulfonamides/pharmacology , Tachycardia, Ventricular/metabolism
16.
Neuroreport ; 32(12): 1058-1064, 2021 08 11.
Article En | MEDLINE | ID: mdl-34232129

The predominant form of edema that occurs during the early stage of ischemic stroke is cytotoxic, resulting in neuronal injury during brain ischemia and reperfusion. Intracellular calcium (Ca2+) is elevated following brain ischemia leading to increased cell membrane permeability. Ca2+/calmodulin-dependent protein kinase II (CaMK II), the downstream molecular signal of N-methyl-d-aspartate receptors (NMDARs), is sensitive to elevations in intracellular Ca2+. Aquaporin-4 (AQP4), which is expressed primarily in the brain, is a water-transport protein. However, it is unclear whether CaMK II regulates AQP4 expression to modulate cellular water permeability. We exposed cultured astrocytes to a hypoxic and glucose-free environment to mimic an ischemic environment in vitro. We investigated the effects of oxygen-glucose deprivation (OGD) on astrocytic viability and swelling, as well as CaMK II and AQP4 expression. We also studied the effects of CaMK II inhibition on cell swelling, viability and AQP4 expression. OGD increased astrocytic swelling and expression of CaMK II and AQP4, and it decreased astrocyte viability. Inhibition of CaMK II resulted in reduced astrocyte water permeability and AQP4 expression. We concluded that the upregulation of CaMK II promoted astrocyte swelling by increasing the expression of AQP4 after OGD.


Aquaporin 4/biosynthesis , Astrocytes/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Hypoxia/physiology , Glucose/deficiency , Water/metabolism , Animals , Animals, Newborn , Aquaporin 4/antagonists & inhibitors , Aquaporin 4/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cell Membrane Permeability/physiology , Cells, Cultured , Gene Expression , Gene Knockdown Techniques/methods , Oxygen/metabolism , Rats , Rats, Sprague-Dawley
17.
Chem Biol Interact ; 346: 109570, 2021 Sep 01.
Article En | MEDLINE | ID: mdl-34217686

Verapamil is reported to prevent scar formation. However, whether verapamil is involved in the ureteral stricture scar and the underlying mechanism need further investigation. Fibroblasts were isolated from ureteral scar tissues. TGF-ß1 stimulation was used to induce fibrosis of fibroblasts. Inhibition of CaMK II was achieved by shRNA transfection. CCK-8 was performed to evaluate cell viability. qRT-PCR was applied to determine the level of mRNA while western blotting was used to determine the level of proteins. Immunofluorescence was used to detect the level of vimentin, collagen I and collagen III. Primary fibroblasts was successfully isolated from ureteral scar tissues. TGF-ß1 stimulation was capable to induce collagen production and fibrosis in primary fibroblasts while inhibition of CaMK II attenuate collagen production. Overexpression of wild type CaMK II lead to further increase of collagen production upon TGF-ß1 stimulation while the mutated CaMK II did not exert this promotion. Treatment of verapamil inhibits TGF-ß1 induced collagen production via inhibiting CaMK II. In present study, we revealed a vital role of Verapamil and CaMK II in the formation of ureteral scar. Verapamil inhibited TGF-ß1 induced collagen fiber formation by regulating CaMK II. Our finding might provide new insight into mechanism of prevention and treatment of ureteral scar.


Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Signal Transduction/drug effects , Smad Proteins/metabolism , Verapamil/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cells, Cultured , Cicatrix/drug therapy , Cicatrix/metabolism , Cicatrix/pathology , Collagen Type I/metabolism , Collagen Type III/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Mutagenesis , RNA Interference , RNA, Small Interfering/metabolism , Transforming Growth Factor beta1/pharmacology , Up-Regulation/drug effects , Verapamil/therapeutic use , Vimentin/metabolism
18.
Bull Exp Biol Med ; 171(3): 297-304, 2021 Jul.
Article En | MEDLINE | ID: mdl-34302204

The study examines the problem whether pyroptosis of U87-MG glioblastoma cells can result from activation of Ca2+/calmodulin-dependent protein kinase II (CaMKII) by a local anesthetic. Glioblastoma cells exposed to various concentrations of typical local anesthetic lidocaine demonstrated augmented cytosolic flux of Ca2+, while suppression of CaMKII expression with the corresponding siRNA significantly inhibited this effect in cells treated with 2 mM lidocaine. Lidocaine up-regulated the expression of mRNA caspase-3 and gasdermin GSDME proteins, whereas silencing of CaMKII gene with siRNA significantly moderated this effect. In addition, lidocaine inhibited proliferation of U87-MG cells, and this effect was prevented by silencing CaMKII gene. Thus, lidocaine activated protein kinase CaMKII, which phosphorylated TRPV1 ion channels and induced calcium overload of U87-MG glioblastoma cells, thereby provoking their pyroptosis.


Anesthetics, Local/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Lidocaine/pharmacology , Neuroglia/drug effects , Pyroptosis/drug effects , Calcium/metabolism , Calcium Signaling , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Caspase 3/genetics , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation , Humans , Ion Transport/drug effects , Neuroglia/cytology , Neuroglia/metabolism , Phosphorylation/drug effects , Pyroptosis/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism
19.
Int J Mol Sci ; 22(14)2021 Jul 09.
Article En | MEDLINE | ID: mdl-34299010

The occurrence and prevalence of heart failure remain high in the United States as well as globally. One person dies every 30 s from heart disease. Recognizing the importance of heart failure, clinicians and scientists have sought better therapeutic strategies and even cures for end-stage heart failure. This exploration has resulted in many failed clinical trials testing novel classes of pharmaceutical drugs and even gene therapy. As a result, along the way, there have been paradigm shifts toward and away from differing therapeutic approaches. The continued prevalence of death from heart failure, however, clearly demonstrates that the heart is not simply a pump and instead forces us to consider the complexity of simplicity in the pathophysiology of heart failure and reinforces the need to discover new therapeutic approaches.


Ca(2+) Mg(2+)-ATPase/metabolism , Calcium/metabolism , Heart Failure/drug therapy , Myocardial Contraction/physiology , Sarcoplasmic Reticulum/metabolism , Adenosine Triphosphatases/metabolism , Adrenergic beta-1 Receptor Agonists/pharmacology , Adrenergic beta-1 Receptor Agonists/therapeutic use , Adrenergic beta-Antagonists/pharmacology , Animals , Antioxidants/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cardiotonic Agents/pharmacology , Dobutamine/pharmacology , Dobutamine/therapeutic use , Heart Failure/physiopathology , Humans
20.
Int J Mol Sci ; 22(11)2021 May 24.
Article En | MEDLINE | ID: mdl-34074030

The ability of organisms to quickly sense and transduce signals of environmental stresses is critical for their survival. Ca2+ is a versatile intracellular messenger involved in sensing a wide variety of stresses and regulating the subsequent cellular responses. So far, our understanding for calcium signaling was mostly obtained from ex vivo tissues and cultured cell lines, and the in vivo spatiotemporal dynamics of stress-triggered calcium signaling in a vertebrate remains to be characterized. Here, we describe the generation and characterization of a transgenic zebrafish line with ubiquitous expression of GCaMP6s, a genetically encoded calcium indicator (GECI). We developed a method to investigate the spatiotemporal patterns of Ca2+ events induced by heat stress. Exposure to heat stress elicited immediate and transient calcium signaling in developing zebrafish. Cells extensively distributed in the integument of the head and body trunk were the first batch of responders and different cell populations demonstrated distinct response patterns upon heat stress. Activity of the heat stress-induced calcium signaling peaked at 30 s and swiftly decreased to near the basal level at 120 s after the beginning of exposure. Inhibition of the heat-induced calcium signaling by LaCl3 and capsazepine and treatment with the inhibitors for CaMKII (Ca²2/calmodulin-dependent protein kinase II) and HSF1 (Heat shock factor 1) all significantly depressed the enhanced heat shock response (HSR). Together, we delineated the spatiotemporal dynamics of heat-induced calcium signaling and confirmed functions of the Ca2+-CaMKII-HSF1 pathway in regulating the HSR in zebrafish.


Calcium Signaling , Calcium/metabolism , Calmodulin/metabolism , Green Fluorescent Proteins/metabolism , Heat-Shock Response/genetics , Zebrafish/genetics , Animals , Animals, Genetically Modified , Calcium Signaling/drug effects , Calcium Signaling/genetics , Calcium Signaling/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calmodulin/genetics , Capsaicin/analogs & derivatives , Capsaicin/pharmacology , Green Fluorescent Proteins/genetics , Heat Shock Transcription Factors/antagonists & inhibitors , Heat Shock Transcription Factors/metabolism , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Heat-Shock Response/physiology , In Situ Hybridization , Lanthanum/pharmacology , Microscopy, Fluorescence , Spatio-Temporal Analysis , Zebrafish/growth & development , Zebrafish/metabolism , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/metabolism
...