Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Cell Calcium ; 96: 102406, 2021 06.
Article in English | MEDLINE | ID: mdl-33848733

ABSTRACT

The effect of brain extracellular matrix (ECM) on synaptic plasticity remains controversial. Here, we show that targeted enzymatic attenuation with chondroitinase ABC (ChABC) of ECM triggers the appearance of new glutamatergic synapses on hippocampal pyramidal neurons, thereby increasing the amplitude of field EPSPs while decreasing both the mean miniature EPSC amplitude and AMPA/NMDA ratio. Although the increased proportion of 'unpotentiated' synapses caused by ECM attenuation should promote long-term potentiation (LTP), surprisingly, LTP was suppressed. The upregulation of small conductance Ca2+-activated K+ (SK) channels decreased the excitability of pyramidal neurons, thereby suppressing LTP. A blockade of SK channels restored cell excitability and enhanced LTP; this enhancement was abolished by a blockade of Rho-associated protein kinase (ROCK), which is involved in the maturation of dendritic spines. Thus, targeting ECM elicits the appearance of new synapses, which can have potential applications in regenerative medicine. However, this process is compensated for by a reduction in postsynaptic neuron excitability, preventing network overexcitation at the expense of synaptic plasticity.


Subject(s)
Extracellular Matrix/metabolism , Neuronal Plasticity/physiology , Small-Conductance Calcium-Activated Potassium Channels/biosynthesis , Synapses/metabolism , Up-Regulation/physiology , Animals , Apamin/pharmacology , Chondroitinases and Chondroitin Lyases/pharmacology , Extracellular Matrix/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Neuronal Plasticity/drug effects , Organ Culture Techniques , Small-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Synapses/drug effects , Up-Regulation/drug effects
2.
Biochem Pharmacol ; 183: 114337, 2021 01.
Article in English | MEDLINE | ID: mdl-33186592

ABSTRACT

The opening of endothelial small-conductance calcium-activated potassium channels (KCa2.3) is essential for endothelium-dependent hyperpolarization (EDH), which predominantly occurs in small resistance arteries. Adenosine monophosphate-activated protein kinase (AMPK), an important metabolic regulator, has been implicated in regulating endothelial nitric oxide synthase activity. However, it was unclear whether AMPK regulated endothelial KCa2.3-mediated EDH-type vasodilation. Using bioinformatics analysis and myograph system, we investigated the regulation by AMPK of KCa2.3 in human umbilical vein endothelial cells (HUVECs) or mouse second-order mesenteric resistance arteries. In HUVECs, AMPK activation either by activators (AICAR, A769662 and MK-8722) or expression of the constitutively active form of AMPK significantly upregulated KCa2.3 expression. Such effects were abolished by AMPK inhibitor (compound C) or AMPK α1-/α2-siRNA, extracellular-signal-regulated-kinase 5 (ERK5) inhibitor (ERK5-IN-1), and specific siRNA to myocyte-enhancer factor 2 (MEF2) or krüppel-like factor 2/4 (KLF2/4). KCa2.3 expression was significantly reduced in mesenteric resistance arteries in AMPKα2 knockout mice when compared with littermate control mice. Furthermore, in high-fat diet fed mice, 2-week treatment with AICAR restored endothelial KCa2.3 expression in mesenteric resistance arteries with improved endothelial dysfunction. Our results demonstrate that activation of AMPK upregulates KCa2.3 channel expression through the ERK5-MEF2-KLF2/4 signaling pathway in vascular endothelium, which contributes to benefits through KCa2.3-mediated EDH-type vasodilation in mesenteric resistance arteries.


Subject(s)
AMP-Activated Protein Kinases/biosynthesis , Diet, High-Fat/adverse effects , Endothelium, Vascular/metabolism , Obesity/metabolism , Small-Conductance Calcium-Activated Potassium Channels/biosynthesis , Up-Regulation/physiology , AMP-Activated Protein Kinases/antagonists & inhibitors , Animals , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Indoles/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/etiology , Oximes/pharmacology , RNA, Small Interfering/pharmacology , Small-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Up-Regulation/drug effects
3.
Int Urol Nephrol ; 52(10): 1851-1861, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32399771

ABSTRACT

PURPOSE: Overactive bladder (OAB) is related to detrusor overactivity (DO), which is caused by the increased detrusor smooth muscle (DSM) cells excitability. Small-conductance Ca2+-activated K+ (SK) channels is a fundamental regulator of excitability and contractility in DSM cells. Obesity-related OAB is associated with the decreased expression and regulatory function of SK channels in DSM layer. However, the regulation role of SK channels in obesity-related OAB DSM cell excitability is still unknown. Here, we tested the hypothesis that obesity-related OAB is associated with reduced expression and activity of SK channels in DSM cells. METHODS: Female Sprague-Dawley rats were fed a normal diet (ND) or a high-fat diet (HFD) and weighed after 12 weeks. We performed urodynamic study, single-cell quantitative reverse transcription-polymerase chain reaction (qRT-PCR), and patch-clamp electrophysiology. RESULTS: Increased average body weights and urodynamically demonstrated OAB were observed in HFD rats. Single-cell qRT-PCR experiments discovered the decreased mRNA expression level of SK channel in DSM cell from HFD rats. Patch-clamp studies revealed that NS309, a SK channel activator, had an attenuated effect on membrane potential hyperpolarization in HFD DSM cells. In addition, the reduced whole cell SK channel currents were recorded in HFD DSM cells. CONCLUSIONS: Attenuated SK channels expression and function, which results in the increased DSM cells excitability and contributes to DO, is discovered in obesity-related OAB DSM cells, suggesting that SK channels might be potential therapeutic targets to control OAB.


Subject(s)
Myocytes, Smooth Muscle/physiology , Obesity/physiopathology , Small-Conductance Calcium-Activated Potassium Channels/physiology , Urinary Bladder, Overactive/physiopathology , Urinary Bladder/physiology , Animals , Disease Models, Animal , Female , Myocytes, Smooth Muscle/metabolism , Obesity/complications , Rats , Rats, Sprague-Dawley , Small-Conductance Calcium-Activated Potassium Channels/biosynthesis , Urinary Bladder, Overactive/etiology
4.
Neuropharmacology ; 141: 66-75, 2018 10.
Article in English | MEDLINE | ID: mdl-30142400

ABSTRACT

Anesthetics exposure to neonates leads to impairment of hippocampal synaptic plasticity and cognitive functions later in life. This phenomenon complies with the concept of metaplasticity: a priming stimulation can affect induction of synaptic plasticity mins or days later. We aimed to understand whether small conductance Ca2+-activated potassium channel type2 (SK2) and subunit composition of AMPA receptors are altered and contribute to sevoflurane-induced metaplasticity. To fulfill this goal, we exposed neonatal rats (postnatal day 7) to 2% sevoflurane for 2 h (sevoflurane rats) and examined synaptic plasticity in the hippocampus and cognitive function in juvenile rats (postnatal day 30-35). We observed that the juvenile sevoflurane rats showed elevation in the threshold for LTP induction, facilitation of LTD induction, and cognitive dysfunctions. Meanwhile, these rats also exhibited increased surface expression of SK2 and enhanced synaptic recruitment of GluA2-lacking AMPA receptors, which possess stronger inward rectification. Blocking SK2 eliminated inward rectification of AMPA receptors in juvenile sevoflurane rats. Interestingly, blocking either SK2 channels or GluA2-lacking AMPA receptors normalized LTP, LTD, and spatial memory in juvenile sevoflurane rats. Our data indicate that neonatal sevoflurane anesthesia have negative impact on cognitive function extended to juvenile rats probably through increasing surface expression of SK2 and synaptic recruitment of GluA2-lacking AMPA receptors. This study provides a new sight for sevoflurane induced metaplasticity.


Subject(s)
Cognitive Dysfunction/physiopathology , Hippocampus/drug effects , Receptors, AMPA/metabolism , Sevoflurane/administration & dosage , Sevoflurane/adverse effects , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Animals , Animals, Newborn , Apamin/pharmacology , Cognitive Dysfunction/chemically induced , Excitatory Postsynaptic Potentials/physiology , Hippocampus/metabolism , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Long-Term Synaptic Depression/drug effects , Long-Term Synaptic Depression/physiology , Male , Neuronal Plasticity/physiology , Rats , Receptors, AMPA/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/biosynthesis , Sevoflurane/antagonists & inhibitors , Small-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Small-Conductance Calcium-Activated Potassium Channels/biosynthesis , Spatial Memory/drug effects
5.
Mol Med Rep ; 18(2): 1377-1386, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29901154

ABSTRACT

Drug resistance is a principal contributor to the poor prognosis of ovarian cancer (OC). Therefore, identifying factors that affect drug resistance in OC is critical. In the present study, 51 OC specimens from lab collections were immunohistochemically tested, public data for 489 samples from The Cancer Genome Atlas cohort and 1,656 samples from the Kaplan­Meier Plotter were downloaded, and data were retrieved from Oncomine. It was identified that the mRNA and protein expression of the potassium calcium­activated channel subfamily N member 3 (KCNN3) was markedly lower in OC tissues compared with normal tissues, and in drug­resistant OC tissues compared with sensitive OC tissues. Low KCNN3 expression consistently predicted shorter disease­free and overall survival (OS). Specifically, low KCNN3 expression predicted shorter OS in 395 patients with low expression levels of mucin­16. There was additional evidence that KCNN3 expression is mediated by microRNA­892b. Furthermore, text mining and analyses of protein and gene interactions indicated that KCNN3 affects drug resistance. To the best of the authors' knowledge, this is the first report to associate KCNN3 with poor prognosis and drug resistance in OC. The present findings indicated that KCNN3 is a potential prognostic marker and therapeutic target for OC.


Subject(s)
Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/biosynthesis , Ovarian Neoplasms/metabolism , Small-Conductance Calcium-Activated Potassium Channels/biosynthesis , Adolescent , Adult , Aged , Aged, 80 and over , Disease-Free Survival , Female , Humans , Middle Aged , Neoplasm Proteins/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/mortality , Small-Conductance Calcium-Activated Potassium Channels/genetics , Survival Rate
6.
Mol Neurobiol ; 54(2): 1078-1091, 2017 03.
Article in English | MEDLINE | ID: mdl-26803493

ABSTRACT

The dysfunction of the small-conductance calcium-activated K+ channel SK3 has been described as one of the factors responsible for the progress of psychoneurological diseases, but the molecular basis of this is largely unknown. This report reveals through use of immunohistochemistry and computational tomography that long-term increased expression of the SK3 small-conductance calcium-activated potassium channel (SK3-T/T) in mice induces a notable bilateral reduction of the hippocampal area (more than 50 %). Histological analysis showed that SK3-T/T mice have cellular disarrangements and neuron discontinuities in the hippocampal formation CA1 and CA3 neuronal layer. SK3 overexpression resulted in cognitive loss as determined by the object recognition test. Electrophysiological examination of hippocampal slices revealed that SK3 channel overexpression induced deficiency of long-term potentiation in hippocampal microcircuits. In association with these results, there were changes at the mRNA levels of some genes involved in Alzheimer's disease and/or linked to schizophrenia, epilepsy, and autism. Taken together, these features suggest that augmenting the function of SK3 ion channel in mice may present a unique opportunity to investigate the neural basis of central nervous system dysfunctions associated with schizophrenia, Alzheimer's disease, or other neuropsychiatric/neurodegenerative disorders in this model system. As a more detailed understanding of the role of the SK3 channel in brain disorders is limited by the lack of specific SK3 antagonists and agonists, the results observed in this study are of significant interest; they suggest a new approach for the development of neuroprotective strategies in neuropsychiatric/neurodegenerative diseases with SK3 representing a potential drug target.


Subject(s)
Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Hippocampus/metabolism , Hippocampus/pathology , Small-Conductance Calcium-Activated Potassium Channels/biosynthesis , Animals , Atrophy , Cognitive Dysfunction/genetics , Gene Expression , Mice , Mice, Knockout , Mice, Transgenic , Organ Culture Techniques , Small-Conductance Calcium-Activated Potassium Channels/genetics
7.
Hypertension ; 69(1): 143-153, 2017 01.
Article in English | MEDLINE | ID: mdl-27872234

ABSTRACT

Endothelium-dependent hyperpolarization (EDH)-mediated responses are impaired in hypertension, but the underlying mechanisms have not yet been determined. The activation of small- and intermediate-conductance of Ca2+-activated K+ channels (SKCa and IKCa) underpins EDH-mediated responses. It was recently reported that Ca2+ influx through endothelial transient receptor potential vanilloid type 4 channel (TRPV4) is a prerequisite for the activation of SKCa/IKCa in endothelial cells in specific beds. Here, we attempted to determine whether the impairment of EDH in hypertension is attributable to the dysfunction of TRPV4 and S/IKCa, using isolated superior mesenteric arteries of 20-week-old stroke-prone spontaneously hypertensive rats (SHRSP) and age-matched Wistar-Kyoto (WKY) rats. In the WKY arteries, EDH-mediated responses were reduced by a combination of SKCa/IKCa blockers (apamin plus TRAM-34; 1-[(2-chlorophenyl)diphenylmethl]-1H-pyrazole) and by the blockade of TRPV4 with the selective antagonist RN-1734 or HC-067047. In the SHRSP arteries, EDH-mediated hyperpolarization and relaxation were significantly impaired when compared with WKY. GSK1016790A, a selective TRPV4 activator, evoked robust hyperpolarization and relaxation in WKY arteries. In contrast, in SHRSP arteries, the GSK1016790A-evoked hyperpolarization was small and relaxation was absent. Hyperpolarization and relaxation to cyclohexyl-[2-(3,5-dimethyl-pyrazol-1-yl)-6-methyl-pyrimidin-4-yl]-amine, a selective SKCa activator, were marginally decreased in SHRSP arteries compared with WKY arteries. The expression of endothelial TRPV4 and SKCa protein was significantly decreased in the SHRSP mesenteric arteries compared with those of WKY, whereas function and expression of IKCa were preserved in SHRSP arteries. These findings suggest that EDH-mediated responses are impaired in superior mesenteric arteries of SHRSP because of a reduction in both TRPV4 and SKCa input to EDH.


Subject(s)
Down-Regulation , Endothelium, Vascular/metabolism , Hypertension/genetics , Small-Conductance Calcium-Activated Potassium Channels/genetics , TRPV Cation Channels/genetics , Vasodilation , Animals , Blotting, Western , DNA/genetics , Disease Models, Animal , Endothelium, Vascular/physiopathology , Hypertension/metabolism , Hypertension/physiopathology , Male , Mesenteric Arteries/metabolism , Mesenteric Arteries/pathology , Mesenteric Arteries/physiopathology , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Small-Conductance Calcium-Activated Potassium Channels/biosynthesis , TRPV Cation Channels/biosynthesis
8.
Neuropharmacology ; 113(Pt A): 519-532, 2017 02.
Article in English | MEDLINE | ID: mdl-27825825

ABSTRACT

Parkinson's disease (PD) is a progressive neurodegenerative disease originating from the loss of dopamine (DA) neurons in the substantia nigra pars compacta (SNC). The small-conductance calcium-activated potassium (SK) channels play an essential role in the regulation of midbrain DA neuron activity patterns, as well as excitability of other types of neurons of the basal ganglia. We therefore questioned whether the SK channel expression in the basal ganglia is modified in parkinsonian rats and how this could impact behavioral performance in a reaction time task. We used a rat model of early PD in which the progressive nigrostriatal DA degeneration was produced by bilateral infusions of 6-hydroxydopamine (6-OHDA) into the striatum. In situ hybridization of SK2 and SK3 mRNA and binding of iodinated apamin (SK2/SK3 blocker) were performed at 1, 8 or 21 days postsurgery in sham and 6-OHDA lesion groups. A significant decrease of SK3 channel expression was found in the SNC of lesioned animals at the three time points, with no change of SK2 channel expression. Interestingly, an upregulation of SK2 mRNA and apamin binding was found in the subthalamic nucleus (STN) at 21 days postlesion. These results were confirmed using quantitative real time polymerase chain reaction (qRT-PCR) approach. Functionally, the local infusion of apamin into the STN of parkinsonian rats enhanced the akinetic deficits produced by nigrostriatal DA lesions in a reaction time task while apamin infusion into the SNC had an opposite effect. These effects disappear when the positive modulator of SK channels (CyPPA) is co-administered with apamin. These findings suggest that an upregulation of SK2 channels in the STN may underlie the physiological adjustment to increased subthalamic excitability following partial DA denervation.


Subject(s)
Basal Ganglia/metabolism , Corpus Striatum/metabolism , Dopamine/metabolism , Parkinsonian Disorders/metabolism , Small-Conductance Calcium-Activated Potassium Channels/biosynthesis , Substantia Nigra/metabolism , Animals , Apamin/toxicity , Basal Ganglia/drug effects , Corpus Striatum/drug effects , Gene Expression , Male , Oxidopamine/toxicity , Parkinsonian Disorders/genetics , Rats , Rats, Wistar , Reaction Time/drug effects , Reaction Time/physiology , Small-Conductance Calcium-Activated Potassium Channels/genetics , Substantia Nigra/drug effects
9.
Tumour Biol ; 37(5): 6275-83, 2016 May.
Article in English | MEDLINE | ID: mdl-26619845

ABSTRACT

Metastasis is the survival-determining factor in urothelial carcinoma (UC) of the urinary bladder. The small conductance calcium-activated potassium channel 3 (SK3) enhances tumor cell invasion in breast cancer and malignant melanoma. Since Edelfosine, a glycerophospholipid with antitumoral properties, effectively inhibits SK3 channel activity, our goal was to evaluate SK3 as a potential molecular target to inhibit the gain of an invasive phenotype in UC. SK3 protein expression was analyzed in 208 tissue samples and UC cell lines. Effects of Edelfosine on SK3 expression and intracellular calcium levels as well as on cell morphology, cell survival and proliferation were assessed using immunoblotting, potentiometric fluorescence microscopy, and clonogenic/cell survival assay; furthermore, we analyzed the effect of Edelfosine and SK3 RNAi knockdown on tumor cell migration and invasion in vitro and in vivo. We found that SK3 is strongly expressed in muscle-invasive UC and in the RT112 cellular tumor model. Higher concentrations of Edelfosine have a strong antitumoral effect on UC cells, while 1 µM effectively inhibits migration/invasion of UC cells in vitro and in vivo comparable to the SK3 knockdown phenotype. Taken together, our results show strong expression of SK3 in muscle-invasive UC, consistent with the postulated role of the protein in tumor cell invasion. Edelfosine is able to effectively inhibit migration and invasion of UC cells in vitro and in vivo in an SK3-dependent way, pointing towards a possible role for Edelfosine as an antiinvasive drug to effectively inhibit UC cell invasion and metastasis.


Subject(s)
Carcinoma/drug therapy , Phospholipid Ethers/administration & dosage , Small-Conductance Calcium-Activated Potassium Channels/biosynthesis , Urinary Bladder Neoplasms/drug therapy , Adult , Aged , Calcium/metabolism , Carcinoma/genetics , Carcinoma/pathology , Cell Movement/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Middle Aged , Neoplasm Invasiveness/genetics , Small-Conductance Calcium-Activated Potassium Channels/genetics , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Urothelium/drug effects , Urothelium/pathology
10.
Br J Pharmacol ; 173(1): 177-90, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26454020

ABSTRACT

BACKGROUND AND PURPOSE: Small conductance calcium-activated potassium (KCa 2.x) channels have a widely accepted canonical function in regulating cellular excitability. In this study, we address a potential non-canonical function of KCa 2.x channels in breast cancer cell survival, using in vitro models. EXPERIMENTAL APPROACH: The expression of all KCa 2.x channel isoforms was initially probed using RT-PCR, Western blotting and microarray analysis in five widely studied breast cancer cell lines. In order to assess the effect of pharmacological blockade and siRNA-mediated knockdown of KCa 2.x channels on these cell lines, we utilized MTS proliferation assays and also followed the corresponding expression of apoptotic markers. KEY RESULTS: All of the breast cancer cell lines, regardless of their lineage or endocrine responsiveness, were highly sensitive to KCa 2.x channel blockade. UCL1684 caused cytotoxicity, with LD50 values in the low nanomolar range, in all cell lines. The role of KCa 2.x channels was confirmed using pharmacological inhibition and siRNA-mediated knockdown. This reduced cell viability and also reduced expression of Bcl-2 but increased expression of active caspase-7 and caspase-9. Complementary to these results, a variety of cell lines can be protected from apoptosis induced by staurosporine using the KCa 2.x channel activator CyPPA. CONCLUSIONS AND IMPLICATIONS: In addition to a well-established role for KCa 2.x channels in migration, blockade of these channels was potently cytotoxic in breast cancer cell lines, pointing to modulation of KCa 2.x channels as a potential therapeutic approach to breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Gene Knockdown Techniques , Small-Conductance Calcium-Activated Potassium Channels/deficiency , Alkanes/toxicity , Apoptosis/drug effects , Apoptosis Regulatory Proteins/biosynthesis , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Humans , Lethal Dose 50 , Protein Isoforms/biosynthesis , Protein Isoforms/deficiency , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Quinolinium Compounds/toxicity , RNA, Small Interfering/pharmacology , Small-Conductance Calcium-Activated Potassium Channels/biosynthesis , Small-Conductance Calcium-Activated Potassium Channels/genetics , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Staurosporine/antagonists & inhibitors , Staurosporine/pharmacology
11.
Microcirculation ; 21(8): 747-53, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25047389

ABSTRACT

OBJECTIVE: NO and a non-NO/prostacyclin EDH mechanism are major contributors of vascular tone and cerebral blood flow. However, the effect of metabolic syndrome on EDH-mediated responses in cerebral vessels remains unknown and may offer another avenue for therapeutic targeting. The purpose of this study was to investigate EDH-dependent responses in cerebral arteries during metabolic syndrome. METHODS: EDH-dependent dilations were assessed in MCAs isolated from nondiabetic obese and lean Zucker rats in the presence and absence of NS309, an activator of SKCa and IKCa channels. IKCa channel expression and activity were assessed by western blotting and pressure myography, respectively. RESULTS: EDH-mediated dilations were significantly attenuated in the obese compared to the lean Zucker rat MCA. Luminal delivery of 1 µM NS309 enhanced EDH-mediated responses in lean and obese Zucker cerebral vessels. Both dose-dependent dilations to luminal NS309 and IKCa protein expression in pooled cerebral arteries were comparable between the two groups. CONCLUSIONS: Our results suggest that pharmacological targeting of IKCa channels can rescue EDH-mediated dilations in obese Zucker rat MCAs. Compromised EDH-mediated dilations in obesity are not due to impaired IKCa channel expression or activity.


Subject(s)
Endothelium, Vascular , Indoles/pharmacology , Intermediate-Conductance Calcium-Activated Potassium Channels/biosynthesis , Obesity , Oximes/pharmacology , Small-Conductance Calcium-Activated Potassium Channels/biosynthesis , Vasodilation/drug effects , Animals , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Gene Expression Regulation/drug effects , Male , Obesity/metabolism , Obesity/pathology , Obesity/physiopathology , Rats , Rats, Zucker
12.
PLoS One ; 9(2): e87652, 2014.
Article in English | MEDLINE | ID: mdl-24505302

ABSTRACT

Preterm births accounts for roughly 9% of all births worldwide and can have detrimental or even lethal consequences for the infant. However to develop new treatment that will lower the rate of preterm births, more knowledge is required on the factors contributing to the contraction and relaxation of the myometrium. The small conductance Ca²âº-activated potassium channel subtype 3 (SK3) has been identified in the myometrium of several species including humans, mice and rats, but with great inter species variation of the expression pattern and regulation. The aim of this study was to investigate the expression of SK3 in the uterus of rats stimulated with 17ß-estradiol and progesterone in order to get an in depth understanding of the rat uterine SK3. Using immunohistochemistry SK3 was localized to the glandular and luminal endometrial lamina epitheliali. Furthermore, a weak signal was observed in the myometrium. Using Western blot the protein level of SK3 was found to increase in uteri from animals treated with 17ß-estradiol, an effect that was not reflected at the mRNA level. The levels of mRNA for SK3 were significantly lower in the uterus of 17ß-estradiol-treated animals than in the uterus of ovariectomized animals. We conclude that the SK channels are present in the endometrial epithelium, and possibly also in the myometrium of the rat uterus. Furthermore, the hormonal effect on SK3 caused by 17ß-estradiol includes divergent regulation at mRNA and protein levels.


Subject(s)
Estradiol/pharmacology , Estrogens/pharmacology , Gene Expression Regulation/drug effects , Myometrium/metabolism , Small-Conductance Calcium-Activated Potassium Channels/biosynthesis , Animals , Female , Mice , Myometrium/cytology , Progesterone/pharmacology , Progestins/pharmacology , Rats , Rats, Sprague-Dawley
13.
Pflugers Arch ; 466(2): 307-17, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23873353

ABSTRACT

The present study was designed to investigate the role of advanced glycation end products (AGEs) in intermediate-conductance and small-conductance Ca(2+)-activated potassium channels (KCa3.1 and KCa2.3)-mediated relaxation in rat resistance arteries and the underlying mechanism. The endothelial function of mesenteric arteries was assessed with the use of wire myography. Expression levels of KCa3.1 and KCa2.3 were measured by using Western blot. Reactive oxygen species (ROS) were measured by using dihydroethidium and 2', 7'-dichlorofluorescein diacetate. KCa3.1 and KCa2.3-mediated vasodilatation responses to acetylcholine and NS309 (opener of KCa3.1 and KCa2.3) were impaired by incubation of the third-order mesenteric arteries from normal rats with AGEs (200 µg ml(-1) for 3 h). In cultured human umbilical vein endothelial cells (HUVECs), AGEs increased ROS level and decreased the protein expression of KCa3.1 and KCa2.3. Antioxidant alpha lipoic acid restored the impairment in both vasodilatation function and expression of KCa3.1 and KCa2.3. H2O2 could mimic the effect of AGEs on the protein expression of KCa3.1 and KCa2.3 in cultured HUVECs. These results demonstrate for the first time that AGEs impaired KCa3.1 and KCa2.3-mediated vasodilatation in rat mesenteric arteries via downregulation of both KCa3.1 and KCa2.3, in which the enhanced oxidative stress was involved.


Subject(s)
Glycation End Products, Advanced/pharmacology , Intermediate-Conductance Calcium-Activated Potassium Channels/drug effects , Mesenteric Arteries/physiology , Oxidative Stress/physiology , Small-Conductance Calcium-Activated Potassium Channels/drug effects , Vasodilation/drug effects , Alkanes/pharmacology , Animals , Human Umbilical Vein Endothelial Cells , Humans , In Vitro Techniques , Intermediate-Conductance Calcium-Activated Potassium Channels/biosynthesis , Male , Mesenteric Arteries/drug effects , NG-Nitroarginine Methyl Ester/pharmacology , Pyrazoles/pharmacology , Quinolinium Compounds/pharmacology , Rats , Rats, Sprague-Dawley , Small-Conductance Calcium-Activated Potassium Channels/biosynthesis
14.
Mol Cell Biochem ; 384(1-2): 95-103, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23975505

ABSTRACT

A recent study indicated that apamin-sensitive current (I KAS, mediated by apamin-sensitive small conductance calcium-activated potassium channels subunits) density significantly increased in heart failure and led to recurrent spontaneous ventricular fibrillation. While the underlying molecular correlation with SK channels is still undetermined, we hypothesized that they are remodeled in HF and that bisoprolol could reverse the remodeling. Volume-overload models were created on male Sprague-Dawley rats by producing an abdominal arteriovenous fistula. Confocal microscopy, quantitative real-time PCR, and western blot were performed to investigate the expression of SK channels and observe the influence of ß-blocker bisoprolol on the expression of SK channels I KAS, and the effect of bisoprolol on I KAS and the sensitivity of I KAS to [Ca(2+)]i at single isolated cells were also explored using whole-cell patch clamp techniques. SK channels were remodeled in HF rats, displaying the significant increase of SK1 and SK3 channel expression. After the treatment of HF rats with bisoprolol, the expression of SK1 and SK3 channels was significantly downregulated, and bisoprolol effectively downregulated I KAS density as well as the sensitivity of I KAS to [Ca(2+)]i. Our data indicated that the expression of SK1 and SK3 increased in HF. Bisoprolol effectively attenuated the change and downregulated I KAS density as well as the sensitivity of I KAS to [Ca(2+)]i.


Subject(s)
Adrenergic beta-1 Receptor Antagonists/pharmacology , Bisoprolol/pharmacology , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Animals , Apamin/pharmacology , Arteriovenous Fistula , Cells, Cultured , Down-Regulation , Heart Failure/metabolism , Male , Membrane Potentials/drug effects , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Small-Conductance Calcium-Activated Potassium Channels/biosynthesis , Ventricular Fibrillation/metabolism
15.
Neuroscience ; 247: 164-74, 2013 Sep 05.
Article in English | MEDLINE | ID: mdl-23727510

ABSTRACT

Numerous studies have shown that human endogenous retrovirus W family (HERV-W) envelope gene (env) is related to various diseases but the underlying mechanism has remained poorly understood. Our previous study showed that there was abnormal expression of HERV-W env in sera of patients with schizophrenia. In this paper, we reported that overexpression of the HERV-W env elevated the levels of small conductance Ca(2+)-activated K(+) channel protein 3 (SK3) in human neuroblastoma cells. Using a luciferase reporter system and RNA interference method, we found that functional cAMP response element site was required for the expression of SK3 triggered by HERV-W env. In addition, it was also found that the SK3 channel was activated by HERV-W env. Further study indicated that cAMP response element-binding protein (CREB) was required for the activation of the SK3 channel. Thus, a novel signaling mechanism of how HERV-W env influences neuronal activity and contributes to mental illnesses such as schizophrenia was proposed.


Subject(s)
Cyclic AMP Response Element-Binding Protein/biosynthesis , Endogenous Retroviruses/metabolism , Neuroblastoma/metabolism , Small-Conductance Calcium-Activated Potassium Channels/biosynthesis , Viral Envelope Proteins/biosynthesis , Cell Line, Tumor , Humans
16.
Biochem J ; 452(1): 121-9, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23496660

ABSTRACT

SKCa (small-conductance Ca(2+)-activated K(+)) channels are widely distributed in different tissues, including the brain, pancreatic islets and myocardium and play an important role in controlling electrical activity and cellular functions. However, intracellular signal modulation of SKCa channels is not fully understood. The present study was designed to investigate the potential regulation of hSKCa1 (human SKCa1) channels by PTKs (protein tyrosine kinases) in HEK (human embryonic kidney)-293 cells expressing the hSKCa1 (KCNN1) gene using approaches of whole-cell patch voltage-clamp, immunoprecipitation, Western blotting and mutagenesis. We found that the hSKCa1 current was inhibited by the broad-spectrum PTK inhibitor genistein, the selective EGFR (epidermal growth factor receptor) kinase inhibitors T25 (tyrphostin 25) and AG556 (tyrphostin AG 556), but not by the Src-family kinases inhibitor PP2. The inhibitory effect of these PTK inhibitors was significantly antagonized by the PTP (protein tyrosine phosphatase) inhibitor orthovanadate. The tyrosine phosphorylation level of hSKCa1 channels was reduced by genistein, T25 or AG556. The reduced tyrosine phosphorylation was countered by orthovanadate. Interestingly, the Y109F mutant hSKCa1 channel lost the inhibitory response to T25 or AG556, and showed a dramatic reduction in tyrosine phosphorylation levels and a reduced current density. These results demonstrate the novel information that hSKCa1 channels are inhibited by genistein, T25 and AG556 via EGFR tyrosine kinase inhibition, which is related to the phosphorylation of Tyr(109) in the N-terminus. This effect may affect electrical activity and cellular functions in brain, pancreatic islets and myocardium.


Subject(s)
ErbB Receptors/antagonists & inhibitors , ErbB Receptors/physiology , Gene Expression Regulation, Enzymologic/drug effects , Small-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Genistein/pharmacology , HEK293 Cells , Humans , Protein Kinase Inhibitors/pharmacology , Small-Conductance Calcium-Activated Potassium Channels/biosynthesis , Small-Conductance Calcium-Activated Potassium Channels/genetics , Tyrphostins/pharmacology
17.
Heart Rhythm ; 10(7): 1001-9, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23499625

ABSTRACT

BACKGROUND: MicroRNAs are important regulators of gene expression, including those involving electrical remodeling in atrial fibrillation (AF). Recently, KCNN3, the gene that encodes the small-conductance calcium-activated potassium channel 3 (SK3), was found to be strongly associated with AF. OBJECTIVES: To evaluate the changes in atrial myocardial microRNAs in patients with permanent AF and to determine the role of microRNA on the regulation of cardiac SK3 expression. METHODS: Atrial tissue obtained during cardiac surgery from patients (4 sinus rhythm and 4 permanent AF) was analyzed by using microRNA arrays. Potential targets of microRNAs were predicted by using software programs. The effects of specific microRNAs on target gene expression were evaluated in HL-1 cells from a continuously proliferating mouse hyperplastic atrial cardiomyocyte cell line. Interactions between microRNAs and targets were further evaluated by using luciferase reporter assay and by using Argonaute pull-down assay. RESULTS: Twenty-one microRNAs showed significant (>2-fold) changes in AF. MicroRNA 499 (miR-499) was upregulated by 2.33-fold (P < .01) in AF atria, whereas SK3 protein expression was downregulated by 46% (P < .05). Transfection of miR-499 mimic in HL-1 cells resulted in the downregulation of SK3 protein expression, while that of miR-499 inhibitor upregulated SK3 expression. Binding of miR-499 to the 3' untranslated region of KCNN3 was confirmed by luciferase reporter assay and by the increased presence of SK3 mRNA in Argonaute pulled-down microRNA-induced silencing complexes after transfection with miR-499. CONCLUSION: Atrial miR-499 is significantly upregulated in AF, leading to SK3 downregulation and possibly contributing to the electrical remodeling in AF.


Subject(s)
Atrial Fibrillation/genetics , Gene Expression Regulation , Genetic Predisposition to Disease , MicroRNAs/genetics , Myocytes, Cardiac/metabolism , Small-Conductance Calcium-Activated Potassium Channels/genetics , Aged , Aged, 80 and over , Animals , Atrial Fibrillation/metabolism , Atrial Fibrillation/pathology , Blotting, Western , Cells, Cultured , Female , Humans , Male , Mice , MicroRNAs/biosynthesis , Middle Aged , Myocytes, Cardiac/pathology , Protein Array Analysis , Small-Conductance Calcium-Activated Potassium Channels/biosynthesis
18.
J Neurosci ; 32(40): 13917-28, 2012 Oct 03.
Article in English | MEDLINE | ID: mdl-23035101

ABSTRACT

Sleep spindles are synchronized 11-15 Hz electroencephalographic (EEG) oscillations predominant during nonrapid-eye-movement sleep (NREMS). Rhythmic bursting in the reticular thalamic nucleus (nRt), arising from interplay between Ca(v)3.3-type Ca(2+) channels and Ca(2+)-dependent small-conductance-type 2 (SK2) K(+) channels, underlies spindle generation. Correlative evidence indicates that spindles contribute to memory consolidation and protection against environmental noise in human NREMS. Here, we describe a molecular mechanism through which spindle power is selectively extended and we probed the actions of intensified spindling in the naturally sleeping mouse. Using electrophysiological recordings in acute brain slices from SK2 channel-overexpressing (SK2-OE) mice, we found that nRt bursting was potentiated and thalamic circuit oscillations were prolonged. Moreover, nRt cells showed greater resilience to transit from burst to tonic discharge in response to gradual depolarization, mimicking transitions out of NREMS. Compared with wild-type littermates, chronic EEG recordings of SK2-OE mice contained less fragmented NREMS, while the NREMS EEG power spectrum was conserved. Furthermore, EEG spindle activity was prolonged at NREMS exit. Finally, when exposed to white noise, SK2-OE mice needed stronger stimuli to arouse. Increased nRt bursting thus strengthens spindles and improves sleep quality through mechanisms independent of EEG slow waves (<4 Hz), suggesting SK2 signaling as a new potential therapeutic target for sleep disorders and for neuropsychiatric diseases accompanied by weakened sleep spindles.


Subject(s)
Arousal/physiology , Sleep Stages/physiology , Small-Conductance Calcium-Activated Potassium Channels/physiology , Thalamic Nuclei/physiology , Action Potentials , Animals , Auditory Threshold , Cells, Cultured/physiology , Electroencephalography , Female , Inhibitory Postsynaptic Potentials/physiology , Male , Mice , Mice, Inbred C57BL , Patch-Clamp Techniques , Polysomnography , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/physiology , Small-Conductance Calcium-Activated Potassium Channels/biosynthesis , Small-Conductance Calcium-Activated Potassium Channels/genetics , Specific Pathogen-Free Organisms , Thalamic Nuclei/cytology , Up-Regulation
19.
Am J Physiol Endocrinol Metab ; 303(7): E825-31, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22785240

ABSTRACT

The maternal cardiovascular system undergoes hemodynamic changes during pregnancy via angiogenesis and vasodilation to ensure adequate perfusion of the placenta. Improper vascularization at the maternal-fetal interface can cause pregnancy complications and poor fetal outcomes. Recent evidence indicates that small conductance Ca(2+)-activated K(+) channel subtype 3 (SK3) contributes to vascular remodeling during pregnancy, and we hypothesized that abnormal SK3 channel expression would alter the ability of the maternal cardiovascular system to adapt to pregnancy demands and lead to poor fetal outcomes. We investigated this hypothesis using transgenic Kcnn3(tm1Jpad)/Kcnn3(tm1Jpad) (SK3(T/T)) mice that overexpress the channel. Isolated pressurized uterine arteries from nonpregnant transgenic SK3(T/T) mice had larger basal diameters and decreased agonist-induced constriction than those from their wild-type counterparts; however, non-receptor-mediated depolarization remained intact. In addition to vascular changes, heart rates and ejection fraction were increased, whereas end systolic volume was reduced in SK3(T/T) mice compared with their wild-type littermates. Uterine sonography of the fetuses on pregnancy day 14 showed a significant decrease in fetal size in SK3(T/T) compared with wild-type mice; thus, SK3(T/T) mice displayed an intrauterine growth-restricted phenotype. The SK3(T/T) mice showed decreased placental thicknesses and higher incidence of fetal loss, losing over half of their complement of pups by midgestation. These results establish that the SK3 channel contributes to both maternal and fetal outcomes during pregnancy and point to the importance of SK3 channel regulation in maintaining a healthy pregnancy.


Subject(s)
Fetal Death/metabolism , Fetal Growth Retardation/metabolism , Small-Conductance Calcium-Activated Potassium Channels/biosynthesis , Animals , Female , Fetal Death/genetics , Fetal Growth Retardation/genetics , Heart Rate/genetics , Heart Rate/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Size , Placenta/anatomy & histology , Placenta/diagnostic imaging , Pregnancy , Small-Conductance Calcium-Activated Potassium Channels/genetics , Stroke Volume/genetics , Stroke Volume/physiology , Ultrasonography, Prenatal/methods , Uterine Artery/anatomy & histology , Uterine Artery/diagnostic imaging , Uterus/blood supply , Uterus/diagnostic imaging
20.
J Neurophysiol ; 108(3): 863-70, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22552186

ABSTRACT

Enhanced intrinsic neuronal excitability of hippocampal pyramidal neurons via reductions in the postburst afterhyperpolarization (AHP) has been hypothesized to be a biomarker of successful learning. This is supported by considerable evidence that pharmacologic enhancement of neuronal excitability facilitates learning. However, it has yet to be demonstrated that pharmacologic reduction of neuronal excitability restricted to the hippocampus can retard acquisition of a hippocampus-dependent task. Thus, the present study was designed to address this latter point using a small conductance potassium (SK) channel activator NS309 focally applied to the dorsal hippocampus. SK channels are important contributors to intrinsic excitability, as measured by the medium postburst AHP. NS309 increased the medium AHP and reduced excitatory postsynaptic potential width of CA1 neurons in vitro. In vivo, NS309 reduced the spontaneous firing rate of CA1 pyramidal neurons and impaired trace eyeblink conditioning in rats. Conversely, trace eyeblink conditioning reduced levels of SK2 channel mRNA and protein in the hippocampus. Therefore, the present findings indicate that modulation of SK channels is an important cellular mechanism for associative learning and further support postburst AHP reductions in hippocampal pyramidal neurons as a biomarker of successful learning.


Subject(s)
Association Learning/physiology , Small-Conductance Calcium-Activated Potassium Channels/biosynthesis , Animals , Association Learning/drug effects , Blinking/drug effects , Blinking/physiology , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/physiology , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Indoles/pharmacology , Male , Oximes/pharmacology , Rats , Rats, Inbred F344 , Small-Conductance Calcium-Activated Potassium Channels/agonists
SELECTION OF CITATIONS
SEARCH DETAIL
...