Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
Int J Biol Sci ; 17(7): 1757-1768, 2021.
Article in English | MEDLINE | ID: mdl-33994860

ABSTRACT

Background: Long noncoding RNA KCNQ1 opposite strand/antisense transcript 1 (lncRNA KCNQ1OT1) is abnormally expressed in various solid tumors. The purpose of this study was to explore the prognostic value and potential functional role of lncRNA KCNQ1OT1 across cancers. Methods: We performed a meta-analysis of published literature to evaluate the prognostic value of lncRNA KCNQ1OT1 across cancers. Verification, functional analysis, and genomic variation analysis were performed using the GEPIA, TIMER, and LnCeVar databases. According to the immune cell infiltration level, we established a prognostic model of lncRNA KCNQ1OT1 expression using public datasets of TIMER. We used quantitative real-time polymerase chain reaction (RT-qPCR) and western blot to detect the expression levels of lncRNA KCNQ1OT1 and the CD155 protein in colorectal cancer (CRC) tissues and cell lines. Then, a lncRNA KCNQ1OT1-knockdown cell line was cocultured to explore the role of lncRNA KCNQ1OT1 and CD155 in the T cell response by flow cytometric analysis. Results: Our results showed that the high expression of lncRNA KCNQ1OT1 was significantly related to poor overall survival across cancers, especially CRC. Interestingly, we found that COAD patients with high lncRNA KCNQ1OT1 expression and high CD8+ T cell infiltration levels had a worse prognosis than those with low lncRNA KCNQ1OT1 expression and high CD8+ T cell infiltration levels. Moreover, lncRNA KCNQ1OT1 and CD155 showed significantly higher expression in CRC tissue than in normal tissue, and lncRNA KCNQ1OT1 expression was positively correlated with CD155 expression in CRC. Finally, knockdown of lncRNA KCNQ1OT1 reduced CD155 expression in HCT116 and SW620 cells and enhanced the immune response in coculture with CD8+ T cells. Conclusions: High lncRNA KCNQ1OT1 expression is significantly correlated with poor prognosis of CRC patients and mediates the CD8+ T cell response in CRC. These findings indicate that lncRNA KCNQ1OT1 is a prognostic biomarker and potential immune therapeutic target for enhancing the CD8+ T cell response in CRC.


Subject(s)
CD8-Positive T-Lymphocytes/pathology , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Apoptosis , CD8-Positive T-Lymphocytes/metabolism , Cell Movement , Cell Proliferation , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Humans , Potassium Channels, Voltage-Gated/biosynthesis , Potassium Channels, Voltage-Gated/genetics , Tumor Cells, Cultured
2.
Exp Eye Res ; 206: 108543, 2021 05.
Article in English | MEDLINE | ID: mdl-33744257

ABSTRACT

Many long non-coding RNAs (lncRNAs) can exert crucial roles in the pathogenesis of cataract, including lncRNA KCNQ1 opposite strand/antisense transcript 1 (KCNQ1OT1). We aimed to further elucidate the biological role and regulatory molecular mechanism of KCNQ1OT1 in cataract. The expression of KCNQ1OT1 and miR-223-3p and BCL2 like 2 (BCL2L2) was examined by qRT-PCR. Cataract cell model was constructed by treatment with hydrogen peroxide (H2O2) in lens epithelial cells (SRA01/04). SRA01/04 cell viability and cell apoptosis were tested using CCK-8 assay and flow cytometry, respectively. Western blot (WB) was performed to measure the levels of apoptosis-related proteins and BCL2L2 protein. The oxidative stress factors were analyzed by corresponding kits. The interaction between miR-223-3p and KCNQ1OT1 or BCL2L2 was validated by dual-luciferase reporter and RNA Immunoprecipitation (RIP) assays. We found that KCNQ1OT1 was upregulated in cataract anterior lens capsule samples and H2O2-induced SRA01/04 cells. Knockdown of KCNQ1OT1 suppressed H2O2-induced SRA01/04 cell apoptosis and oxidative stress. KCNQ1OT1 acted as a sponge of miR-223-3p. Inhibition of miR-223-3p could abate the function of KCNQ1OT1 silence in H2O2-treated SRA01/04 cells. Additionally, BCL2L2 was a direct target of miR-223-3p, and miR-223-3p weakened H2O2-induced SRA01/04 cell apoptosis and oxidative stress by targeting BCL2L2. Collectively, the data suggest a role for the KCNQ1OT1/miR-223-3p/BCL2L2 axis in cataract formation but the data was generated using an epithelial cell line.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Apoptosis/genetics , Cataract/genetics , Gene Expression Regulation , MicroRNAs/genetics , Oxidative Stress/genetics , Aged , Apoptosis Regulatory Proteins/biosynthesis , Cataract/metabolism , Cataract/pathology , Cells, Cultured , DNA/genetics , Female , Humans , Hydrogen Peroxide/toxicity , Male , MicroRNAs/biosynthesis , Middle Aged , Potassium Channels, Voltage-Gated/biosynthesis , Potassium Channels, Voltage-Gated/genetics
3.
Twin Res Hum Genet ; 20(5): 389-394, 2017 10.
Article in English | MEDLINE | ID: mdl-28803575

ABSTRACT

CDKN1C and KCNQ1OT1 are imprinted genes that might be potential regulators of placental development. This study investigated placental expressions of CDKN1C and KCNQ1OT1 in monozygotic twins with and without selective intrauterine growth restriction (sIUGR). Seventeen sIUGR and fifteen normal monozygotic(MZ) twin pairs were examined. Placental mRNA expressions of CDKN1C and KCNQ1OT1 were detected by real-time fluorescent quantitative PCR. CDKN1C protein expression was detected by immunohistochemical assay and Western-blotting. In the sIUGR group, smaller fetuses had a smaller share of the placenta, and CDKN1C protein expression was significantly increased while KCNQ1OT1 mRNA expression was significantly decreased. The CDKN1C/KCNQ1OT1 mRNA ratio was lower in the larger fetus than in the smaller fetus (p < .05). In the control group, CDKN1C protein expression showed no difference between larger and smaller fetuses, while KCNQ1OT1 mRNA expression was significantly lower in the larger fetus, and the CDKN1C/KCNQ1OT1 mRNA ratio was higher in the larger fetus than in the smaller fetus (p < .05). Our findings showed that pathogenesis of sIUGR may be related to the co-effect of the up-regulated protein expression of CDKN1C and down-regulated mRNA expression of KCNQ1OT1 in the placenta.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p57/biosynthesis , Fetal Growth Retardation/metabolism , Gene Expression Regulation, Developmental , Placenta/metabolism , Twins, Monozygotic , Adult , Cyclin-Dependent Kinase Inhibitor p57/genetics , Female , Fetal Growth Retardation/genetics , Humans , Infant, Newborn , Male , Potassium Channels, Voltage-Gated/biosynthesis , Potassium Channels, Voltage-Gated/genetics , Pregnancy
4.
J Neurosci ; 37(34): 8256-8272, 2017 08 23.
Article in English | MEDLINE | ID: mdl-28751455

ABSTRACT

Dysfunction of the fast-inactivating Kv3.4 potassium current in dorsal root ganglion (DRG) neurons contributes to the hyperexcitability associated with persistent pain induced by spinal cord injury (SCI). However, the underlying mechanism is not known. In light of our previous work demonstrating modulation of the Kv3.4 channel by phosphorylation, we investigated the role of the phosphatase calcineurin (CaN) using electrophysiological, molecular, and imaging approaches in adult female Sprague Dawley rats. Pharmacological inhibition of CaN in small-diameter DRG neurons slowed repolarization of the somatic action potential (AP) and attenuated the Kv3.4 current. Attenuated Kv3.4 currents also exhibited slowed inactivation. We observed similar effects on the recombinant Kv3.4 channel heterologously expressed in Chinese hamster ovary cells, supporting our findings in DRG neurons. Elucidating the molecular basis of these effects, mutation of four previously characterized serines within the Kv3.4 N-terminal inactivation domain eliminated the effects of CaN inhibition on the Kv3.4 current. SCI similarly induced concurrent Kv3.4 current attenuation and slowing of inactivation. Although there was little change in CaN expression and localization after injury, SCI induced upregulation of the native regulator of CaN 1 (RCAN1) in the DRG at the transcript and protein levels. Consistent with CaN inhibition resulting from RCAN1 upregulation, overexpression of RCAN1 in naive DRG neurons recapitulated the effects of pharmacological CaN inhibition on the Kv3.4 current and the AP. Overall, these results demonstrate a novel regulatory pathway that links CaN, RCAN1, and Kv3.4 in DRG neurons. Dysregulation of this pathway might underlie a peripheral mechanism of pain sensitization induced by SCI.SIGNIFICANCE STATEMENT Pain sensitization associated with spinal cord injury (SCI) involves poorly understood maladaptive modulation of neuronal excitability. Although central mechanisms have received significant attention, recent studies have identified peripheral nerve hyperexcitability as a driver of persistent pain signaling after SCI. However, the ion channels and signaling molecules responsible for this change in primary sensory neuron excitability are still not well defined. To address this problem, this study used complementary electrophysiological and molecular methods to determine how Kv3.4, a voltage-gated K+ channel robustly expressed in dorsal root ganglion neurons, becomes dysfunctional upon calcineurin (CaN) inhibition. The results strongly suggest that CaN inhibition underlies SCI-induced dysfunction of Kv3.4 and the associated excitability changes through upregulation of the native regulator of CaN 1 (RCAN1).


Subject(s)
Calcineurin Inhibitors/pharmacology , Calcineurin/biosynthesis , Ganglia, Spinal/metabolism , Shaw Potassium Channels/biosynthesis , Spinal Cord Injuries/metabolism , Animals , CHO Cells , Calcineurin Inhibitors/toxicity , Cells, Cultured , Cervical Vertebrae , Cricetinae , Cricetulus , Female , Ganglia, Spinal/drug effects , Neurons/drug effects , Neurons/metabolism , Potassium Channels, Voltage-Gated/biosynthesis , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/physiopathology
5.
Circ Heart Fail ; 10(6)2017 Jun.
Article in English | MEDLINE | ID: mdl-28611128

ABSTRACT

BACKGROUND: KCNE2 is a promiscuous auxiliary subunit of voltage-gated cation channels. A recent work demonstrated that KCNE2 regulates L-type Ca2+ channels. Given the important roles of altered Ca2+ signaling in structural and functional remodeling in diseased hearts, this study investigated whether KCNE2 participates in the development of pathological hypertrophy. METHODS AND RESULTS: We found that cardiac KCNE2 expression was significantly decreased in phenylephrine-induced cardiomyocyte hypertrophy in neonatal rat ventricular myocytes and in transverse aortic constriction-induced cardiac hypertrophy in mice, as well as in dilated cardiomyopathy in human. Knockdown of KCNE2 in neonatal rat ventricular myocytes reproduced hypertrophy by increasing the expression of ANP (atrial natriuretic peptide) and ß-MHC (ß-myosin heavy chain), and cell surface area, whereas overexpression of KCNE2 attenuated phenylephrine-induced cardiomyocyte hypertrophy. Knockdown of KCNE2 increased intracellular Ca2+ transient, calcineurin activity, and nuclear NFAT (nuclear factor of activated T cells) protein levels, and pretreatment with inhibitor of L-type Ca2+ channel (nifedipine) or calcineurin (FK506) attenuated the activation of calcineurin-NFAT pathway and cardiomyocyte hypertrophy. Meanwhile, the phosphorylation levels of p38, extracellular signal-regulated kinase 1/2, and c-Jun N-terminal kinase were increased, and inhibiting the 3 cascades of mitogen-activated protein kinase reduced cardiomyocyte hypertrophy induced by KCNE2 knockdown. Overexpression of KCNE2 in heart by ultrasound-microbubble-mediated gene transfer suppressed the development of hypertrophy and activation of calcineurin-NFAT and mitogen-activated protein kinase pathways in transverse aortic constriction mice. CONCLUSIONS: This study demonstrates that cardiac KCNE2 expression is decreased and contributes to the development of hypertrophy via activation of calcineurin-NFAT and mitogen-activated protein kinase pathways. Targeting KCNE2 is a potential therapeutic strategy for the treatment of hypertrophy.


Subject(s)
Calcineurin/genetics , Cardiomegaly/genetics , Gene Expression Regulation , Mitogen-Activated Protein Kinases/metabolism , NFATC Transcription Factors/genetics , Potassium Channels, Voltage-Gated/genetics , RNA/genetics , Animals , Animals, Newborn , Apoptosis , Calcineurin/metabolism , Cardiomegaly/metabolism , Disease Models, Animal , Humans , Immunohistochemistry , Myocardium/metabolism , Myocardium/pathology , NFATC Transcription Factors/metabolism , Polymerase Chain Reaction , Potassium Channels, Voltage-Gated/biosynthesis , Rats , Rats, Sprague-Dawley
6.
Article in English | MEDLINE | ID: mdl-28611207

ABSTRACT

BACKGROUND: KCNQ1 and KCNE1 assemble to form the slow delayed rectifier (IKs) channel critical for shortening ventricular action potentials during high ß-adrenergic tone. However, too much IKs under basal conditions poses an arrhythmogenic risk. Our objective is to understand how adult ventricular myocytes regulate the IKs amplitudes under basal conditions and in response to stress. METHODS AND RESULTS: We express fluorescently tagged KCNQ1 and KCNE1 in adult ventricular myocytes and follow their biogenesis and trafficking paths. We also study the distribution patterns of native KCNQ1 and KCNE1, and their relationship to IKs amplitudes, in chronically stressed ventricular myocytes, and use COS-7 cell expression to probe the underlying mechanism. We show that KCNQ1 and KCNE1 are both translated in the perinuclear region but traffic by different routes, independent of each other, to their separate subcellular locations. KCNQ1 mainly resides in the jSR (junctional sarcoplasmic reticulum), whereas KCNE1 resides on the cell surface. Under basal conditions, only a small portion of KCNQ1 reaches the cell surface to support the IKs function. However, in response to chronic stress, KCNQ1 traffics from jSR to the cell surface to boost the IKs amplitude in a process depending on Ca binding to CaM (calmodulin). CONCLUSIONS: In adult ventricular myocytes, KCNE1 maintains a stable presence on the cell surface, whereas KCNQ1 is dynamic in its localization. KCNQ1 is largely in an intracellular reservoir under basal conditions but can traffic to the cell surface and boost the IKs amplitude in response to stress.


Subject(s)
Heart Ventricles/metabolism , Hypertension/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , KCNQ1 Potassium Channel/metabolism , Myocytes, Cardiac/metabolism , Potassium Channels, Voltage-Gated/metabolism , Potassium/metabolism , Action Potentials , Animals , COS Cells , Calmodulin/metabolism , Cell Membrane/metabolism , Chlorocebus aethiops , Disease Models, Animal , Dogs , Guinea Pigs , Heart Ventricles/physiopathology , Hypertension/physiopathology , KCNQ1 Potassium Channel/biosynthesis , KCNQ1 Potassium Channel/genetics , Potassium Channels, Voltage-Gated/biosynthesis , Potassium Channels, Voltage-Gated/genetics , Protein Transport , Rats, Inbred SHR , Sarcoplasmic Reticulum/metabolism , Time Factors , Transfection
7.
Article in English | MEDLINE | ID: mdl-28233058

ABSTRACT

In mormyrid weakly electric fish, the electric organ discharge (EOD) is used for species recognition, orientation and prey localization. Produced in the muscle-derived adult electric organ, the EOD exhibits a wide diversity across species in both waveform and duration. While certain defining EOD characteristics can be linked to anatomical features of the electric organ, many factors underlying EOD differentiation are yet unknown. Here, we report the differential expression of 13 Kv1 voltage-gated potassium channel genes, two inwardly rectifying potassium channel genes, two previously studied sodium channel genes and an ATPase pump in two sympatric species of the genus Campylomormyrus in both the adult electric organ and skeletal muscle. Campylomormyrus compressirostris displays a basal EOD, largely unchanged during development, while C. tshokwe has an elongated, putatively derived discharge. We report an upregulation in all Kv1 genes in the electric organ of Campylomormyrus tshokwe when compared to both skeletal muscle and C. compressirostris electric organ. This pattern of upregulation in a species with a derived EOD form suggests that voltage-gated potassium channels are potentially involved in the diversification of the EOD signal among mormyrid weakly electric fish.


Subject(s)
Electric Fish/genetics , Electric Fish/metabolism , Electric Organ/physiology , Potassium Channels, Voltage-Gated/biosynthesis , Potassium Channels, Voltage-Gated/genetics , Animals , Female , Gene Expression
8.
Urology ; 101: 169.e1-169.e5, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27856205

ABSTRACT

OBJECTIVE: To investigate the changes including expression and localization of 2 potassium channels, renal outer medullary K+ channel (ROMK) and voltage-gated K+ channel 7.1 (KCNQ1), after increased urinary potassium leakage in patients with interstitial cystitis/painful bladder syndrome (IC/PBS). MATERIALS AND METHODS: The study group included 24 patients with IC/PBS and a control group consisting of 12 volunteers without any IC/PBS symptoms. Bladder biopsies were taken from both groups. We determined the protein expression and distribution of potassium channels using immunoblotting, immunohistochemistry, and immunofluorescent staining under confocal laser microscopy. RESULTS: The results revealed that ROMK was predominantly expressed in apical cells of the bladder urothelium at significantly higher levels (3.3-fold) in the study group than in the control group. In contrast, KCNQ1 was expressed in the basolateral membrane according to confocal microscopy results and did not significantly differ between groups. CONCLUSION: Our data showed that the abundance of ROMK protein in apical cells was increased in the IC/PBS group, whereas KCNQ1, which was distributed in the basolateral membrane of the bladder urothelium, showed similar abundance between groups. These results suggest that upregulation of the ROMK channel in apical cells might permit avid potassium flux into the bladder lumen to maintain intracellular K+ homeostasis in the dysfunctional urothelium.


Subject(s)
Cystitis, Interstitial/metabolism , Kidney Medulla/metabolism , Potassium Channels, Voltage-Gated/biosynthesis , Urothelium/metabolism , Adult , Biomarkers/metabolism , Cystitis, Interstitial/diagnosis , Female , Humans , Immunoblotting , Immunohistochemistry , Kidney Medulla/pathology , Male , Microscopy, Confocal , Middle Aged , Urothelium/pathology , Young Adult
9.
J Biosci ; 41(4): 659-666, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27966486

ABSTRACT

We examined the effects of the selective serotonin reuptake inhibitor (SSRI) sertraline on voltage-dependent K+ (Kv) channels in freshly isolated rabbit coronary arterial smooth muscle cells using the voltage-clamp technique. Sertraline decreased the Kv channel current in a dose-dependent manner, with an IC50 value of 0.18 mu M and a slope value (Hill coefficient) of 0.61. Although the application of 1 mu M sertraline did not affect the steady-state activation curves, sertraline caused a significant, negative shift in the inactivation curves. Pretreatment with another SSRI, paroxetine, had no significant effect on Kv currents and did not alter the inhibitory effects of sertraline on Kv currents. From these results, we concluded that sertraline dose-dependently inhibited Kv currents independently of serotonin reuptake inhibition by shifting inactivation curves to a more negative potential.


Subject(s)
Potassium Channels, Voltage-Gated/biosynthesis , Selective Serotonin Reuptake Inhibitors/administration & dosage , Serotonin/metabolism , Sertraline/administration & dosage , Animals , Coronary Vessels/drug effects , Coronary Vessels/pathology , Dose-Response Relationship, Drug , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Paroxetine/administration & dosage , Patch-Clamp Techniques , Potassium Channels, Voltage-Gated/metabolism , Rabbits
10.
Clin Exp Hypertens ; 37(5): 381-7, 2015.
Article in English | MEDLINE | ID: mdl-25856227

ABSTRACT

To test the hypothesis that chronic hypoxic pulmonary hypertension (CH-PH) is associated with increased survivin and decreased voltage-gated potassium (KV) channels expression in pulmonary arteries, rats were randomized as: normoxia (N); normoxia + YM155, survivin suppressor (NY); hypoxia (H); hypoxia + YM155 (HY). HY group had significantly reduced pulmonary arterial pressure, right ventricular weight and right ventricular hypertrophy compared with H group. Survivin mRNA and protein were detected in pulmonary arteries of rats with CH-PH, but not rats without CH-PH. YM155 downregulated survivin protein and mRNA. KV channel expression and activity were upregulated after YM155 treatment. Survivin may play a role in the pathogenesis of CH-PH.


Subject(s)
Gene Expression Regulation , Hypertension, Pulmonary/genetics , Hypoxia/genetics , Imidazoles/therapeutic use , Microtubule-Associated Proteins/antagonists & inhibitors , Muscle, Smooth, Vascular/metabolism , Naphthoquinones/therapeutic use , Potassium Channels, Voltage-Gated/genetics , Animals , Chronic Disease , Disease Models, Animal , Hypertension, Pulmonary/complications , Hypertension, Pulmonary/drug therapy , Hypoxia/etiology , Hypoxia/metabolism , Male , Microtubule-Associated Proteins/biosynthesis , Microtubule-Associated Proteins/genetics , Muscle, Smooth, Vascular/physiopathology , Patch-Clamp Techniques , Potassium Channels, Voltage-Gated/biosynthesis , Pulmonary Wedge Pressure , RNA/genetics , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Survivin
11.
J Proteomics ; 87: 89-102, 2013 Jul 11.
Article in English | MEDLINE | ID: mdl-23727489

ABSTRACT

Sea anemones represent one of the emerging groups of interest concerning venomous animals in toxinology and the goal of the present work was the prospection, and the structural and functional characterization of the compounds present in the secretion of the sea anemone Stichodactyla duerdeni from Brazilian coast. We used a combination of offline RPC-MALDI-TOF and online nano-RPC-ESI-LTQ-Orbitrap proteomic techniques as well as functional bioassays. The mucus was milked by electric stimulation and fractionated by gel filtration on Sephadex G-50 yielding 5 main fractions. The low molecular weight fractions were further submitted to RP-HPLC resulting in 35 new subfractions that were subsequently analyzed by offline MALDI-TOF mass spectrometry. MALDI peptide mass fingerprinting yielded up to 134 different molecular masses, ranging from m/z 901 to 10,833. Among these subfractions, a new peptide of 3431Da, named U-SHTX-Sdd1, was purified and completely sequenced by automated Edman's degradation and tandem mass spectrometry. An analysis of U-SHTX-Sdd1 revealed a modified O-HexNAc-Threonine at position 1, which, at the best of our knowledge, constitutes the first sea anemone toxin reported with such post-translational modification. Because of its sequence similarity with other sea anemone toxins, the pharmacological activity of U-SHTX-Sdd1 was assessed by electrophysiological measurements using the two electrode voltage-clamp technique on cloned voltage-gated potassium channel subtypes, expressed in Xenopus laevis oocytes. However, U-SHTX-Sdd1 did not show activity on these channels. A large-scale proteomic approach was also employed to shed lights on the sea anemone compounds, and a total 67 proteins and peptides were identified. BIOLOGICAL SIGNIFICANCE: In this manuscript, we report an extensive characterization of S. duerdeni secretion by means of peptide mass fingerprinting and high-throughput proteome analyses. Also, we report the structure of a new glycopeptide by a combination of biochemical techniques. Despite the previous studies that described proteinaceous compounds present in sea anemone secretions, the number of reported primary sequences is still low. Thus, to access the scenery of protein components from S. duerdeni mucus, including their biological functions, a robust proteomic approach was used together with bioinformatic tools. The demonstrated strategy of analysis is perfectly suitable to other sea anemone secretions and animal venoms. Moreover, new peptide structures can arise contributing to the knowledge of the diversity of these animal peptides.


Subject(s)
Glycopeptides , Ion Channel Gating/drug effects , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Proteomics , Sea Anemones , Animals , Glycopeptides/chemistry , Glycopeptides/genetics , Glycopeptides/metabolism , Glycopeptides/pharmacology , Ion Channel Gating/genetics , Marine Toxins/chemistry , Marine Toxins/genetics , Marine Toxins/metabolism , Marine Toxins/pharmacology , Oocytes , Potassium Channels, Voltage-Gated/biosynthesis , Potassium Channels, Voltage-Gated/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sea Anemones/chemistry , Sea Anemones/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Xenopus laevis
12.
Exp Eye Res ; 116: 424-32, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24416770

ABSTRACT

Human, monkey, and bovine retinal pigment epithelial (RPE) cells exhibit an M-type K+ current, which in many other cell types is mediated by channels composed of KCNQ α-subunits and KCNE auxiliary subunits. Recently, we demonstrated the expression of KCNQ1, KCNQ4, and KCNQ5 in the monkey RPE. Here, we investigated the expression of KCNQ and KCNE subunits in native bovine RPE. RT-PCR analysis revealed the expression of KCNQ1, KCNQ4, and KCNQ5 transcripts in the RPE, but, in Western blot analysis of RPE plasma membranes, only KCNQ5 was detected. Among the five members of the KCNE gene family, transcripts for KCNE1, KCNE2, KCNE3, and KCNE4 were detected in bovine RPE, but only KCNE1 and KCNE2 proteins were detected. Immunohistochemistry of frozen bovine retinal sections revealed KCNE1 expression near the apical and basal membranes of the RPE, in cone outer segments, in the outer nuclear layer, and throughout the inner retina. The localization of KCNE1 in the RPE basal membrane, where KCNQ5 was previously found to be present, suggests that this ß-subunit may contribute to M-type K(+) channels in this membrane.


Subject(s)
Gene Expression Regulation , KCNQ Potassium Channels/genetics , Potassium Channels, Voltage-Gated/genetics , RNA/genetics , Retinal Pigment Epithelium/metabolism , Animals , Blotting, Western , Cattle , Immunohistochemistry , KCNQ Potassium Channels/biosynthesis , Potassium Channels, Voltage-Gated/biosynthesis , Retinal Pigment Epithelium/cytology , Reverse Transcriptase Polymerase Chain Reaction
13.
J Biol Chem ; 287(42): 35565-35575, 2012 Oct 12.
Article in English | MEDLINE | ID: mdl-22927438

ABSTRACT

The Kcnh1 gene encodes a voltage-gated potassium channel highly expressed in neurons and involved in tumor cell proliferation, yet its physiological roles remain unclear. We have used the zebrafish as a model to analyze Kcnh1 function in vitro and in vivo. We found that the kcnh1 gene is duplicated in teleost fish (i.e. kcnh1a and kcnh1b) and that both genes are maternally expressed during early development. In adult zebrafish, kcnh1a and kcnh1b have distinct expression patterns but share expression in brain and testis. Heterologous expression of both genes in Xenopus oocytes revealed a strong conservation of characteristic functional properties between human and fish channels, including a unique sensitivity to intracellular Ca(2+)/calmodulin and modulation of voltage-dependent gating by extracellular Mg(2+). Using a morpholino antisense approach, we demonstrate a strong kcnh1 loss-of-function phenotype in developing zebrafish, characterized by growth retardation, delayed hindbrain formation, and embryonic lethality. This late phenotype was preceded by transcriptional up-regulation of known cell-cycle inhibitors (p21, p27, cdh2) and down-regulation of pro-proliferative factors, including cyclin D1, at 70% epiboly. These results reveal an unanticipated basic activity of kcnh1 that is crucial for early embryonic development and patterning.


Subject(s)
Body Patterning/physiology , Gene Expression Regulation, Developmental/physiology , Potassium Channels, Voltage-Gated/biosynthesis , Transcription, Genetic/physiology , Zebrafish Proteins/biosynthesis , Zebrafish/embryology , Animals , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor Proteins/genetics , Cyclin-Dependent Kinase Inhibitor Proteins/metabolism , Female , Humans , Male , Organ Specificity/physiology , Potassium Channels, Voltage-Gated/genetics , Rhombencephalon/embryology , Xenopus laevis , Zebrafish/genetics , Zebrafish Proteins/genetics
14.
Open Biol ; 2(6): 120072, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22773948

ABSTRACT

Brugada syndrome (BrS) is associated with ventricular tachycardia originating particularly in the right ventricle (RV). We explore electrophysiological features predisposing to such arrhythmic tendency and their possible RV localization in a heterozygotic Scn5a+/- murine model. Na(v)1.5 mRNA and protein expression were lower in Scn5a+/- than wild-type (WT), with a further reduction in the RV compared with the left ventricle (LV). RVs showed higher expression levels of K(v)4.2, K(v)4.3 and KChIP2 in both Scn5a+/- and WT. Action potential upstroke velocity and maximum Na(+) current (I(Na)) density were correspondingly decreased in Scn5a+/-, with a further reduction in the RV. The voltage dependence of inactivation was shifted to more negative values in Scn5a+/-. These findings are predictive of a localized depolarization abnormality leading to slowed conduction. Persistent Na(+) current (I(pNa)) density was decreased in a similar pattern to I(Na). RV transient outward current (I(to)) density was greater than LV in both WT and Scn5a+/-, and had larger time constants of inactivation. These findings were also consistent with the observation that AP durations were smallest in the RV of Scn5a+/-, fulfilling predictions of an increased heterogeneity of repolarization as an additional possible electrophysiological mechanism for arrhythmogenesis in BrS.


Subject(s)
Arrhythmias, Cardiac , Heart Ventricles , Membrane Potentials , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Potassium Channels, Voltage-Gated/biosynthesis , Animals , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/pathology , Arrhythmias, Cardiac/physiopathology , Heart Ventricles/metabolism , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Ion Transport/genetics , Mice , Mice, Mutant Strains , NAV1.5 Voltage-Gated Sodium Channel/genetics , Potassium Channels, Voltage-Gated/genetics
15.
J Vis Exp ; (55)2011 Sep 27.
Article in English | MEDLINE | ID: mdl-21989393

ABSTRACT

In mammalian central nervous system, different types of neurons with diverse molecular and functional characteristics are intermingled with each other, difficult to separate and also not easily identified by their morphology. Thus, it is often difficult to analyze gene expression in a specific neuron type. Here we document a procedure that combines whole-cell patch clamp recording techniques with single-cell reverse transcription polymerase chain reaction (scRT-PCR) to profile mRNA expression in different types of neurons in the substantial nigra. Electrophysiological techniques are first used to record the neurophysiological and functional properties of individual neurons. Then, the cytoplasm of single electrophysiologically characterized nigral neurons is aspirated and subjected to scRT-PCR analysis to obtain mRNA expression profiles for neurotransmitter synthesis enzymes, receptors, and ion channels. The high selectivity and sensitivity make this method particularly useful when immunohistochemistry can not be used due to a lack of suitable antibody or low expression level of the protein. This method is also applicable to neurons in other brain areas.


Subject(s)
Neurons/physiology , Potassium Channels, Voltage-Gated/genetics , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction/methods , Substantia Nigra/physiology , Animals , Female , Male , Neurons/metabolism , Potassium Channels, Voltage-Gated/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Substantia Nigra/cytology , Substantia Nigra/metabolism
16.
Neurosci Res ; 70(4): 361-9, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21624401

ABSTRACT

The medial septum/diagonal band complex (MSDB) controls hippocampal excitability, rhythms and plastic processes. Medial septal neuronal populations display heterogeneous firing patterns. In addition, some of these populations degenerate during age-related disorders (e.g. cholinergic neurons). Thus, it is particularly important to examine the intrinsic properties of theses neurons in order to create new agents that effectively modulate hippocampal excitability and enhance memory processes. Here, we have examined the properties of voltage-gated, K(+) currents in electrophysiologically-identified neurons. These neurons were taken from young rat brain slices containing the MS/DB complex. Whole-cell, patch recordings of outward currents were obtained from slow firing, fast-spiking, regular-firing and burst-firing neurons. Slow firing neurons showed depolarization-activated K(+) current peaks and densities larger than in other neuronal subtypes. Slow firing total current exhibited an inactivating A-type current component that activates at subthreshold depolarization and was reliably blocked by high concentrations of 4-AP. In addition, slow firing neurons expressed a low-threshold delayed rectifier K(+) current component with slow inactivation and intermediate sensitivity to tetraethylammonium. Fast-spiking neurons exhibited the smaller I(K) and I(A) current densities. Burst and regular firing neurons displayed an intermediate firing phenotype with I(K) and I(A) current densities that were larger than the ones observed in fast-spiking neurons but smaller than the ones observed in slow-firing neurons. In addition, the prevalence of each current differed among electrophysiological groups with slow firing and regular firing neurons expressing mostly I(A) and fast spiking and bursting neurons exhibiting mostly delayer rectifier K(+) currents with only minimal contributions of the I(A). The pharmacological or genetic modulations of these currents constitute an important target for the treatment of age-related disorders.


Subject(s)
Action Potentials/physiology , Gene Expression Regulation , Neurons/metabolism , Phenotype , Potassium Channels, Voltage-Gated/biosynthesis , Septum of Brain/cytology , Septum of Brain/metabolism , Animals , Male , Potassium Channels, Voltage-Gated/genetics , Rats , Rats, Sprague-Dawley
17.
Neuroscience ; 178: 68-81, 2011 Mar 31.
Article in English | MEDLINE | ID: mdl-21211550

ABSTRACT

Transient outward rectifying conductances or A-like conductances in sympathetic preganglionic neurons (SPN) are prolonged, lasting for hundreds of milliseconds to seconds and are thought to play a key role in the regulation of SPN firing frequency. Here, a multidisciplinary electrophysiological, pharmacological and molecular single-cell rt-PCR approach was used to investigate the kinetics, pharmacological profile and putative K+ channel subunits underlying the transient outward rectifying conductance expressed in SPN. SPN expressed a 4-aminopyridine (4-AP) sensitive transient outward rectification with significantly longer decay kinetics than reported for many other central neurons. The conductance and corresponding current in voltage-clamp conditions was also sensitive to the Kv4.2 and Kv4.3 blocker phrixotoxin-2 (1-10 µM) and the blocker of rapidly inactivating Kv channels, pandinotoxin-Kα (50 nM). The conductance and corresponding current was only weakly sensitive to the Kv1 channel blocker tityustoxin-Kα and insensitive to dendrotoxin I (200 nM) and the Kv3.4 channel blocker BDS-II (1 µM). Single-cell RT-PCR revealed mRNA expression for the α-subunits Kv4.1 and Kv4.3 in the majority and Kv1.5 in less than half of SPN. mRNA for accessory ß-subunits was detected for Kvß2 in all SPN with differential expression of mRNA for KChIP1, Kvß1 and Kvß3 and the peptidase homologue DPP6. These data together suggest that the transient outwardly rectifying conductance in SPN is mediated by members of the Kv4 subfamily (Kv4.1 and Kv4.3) in association with the ß-subunit Kvß2. Differential expression of the accessory ß subunits, which may act to modulate channel density and kinetics in SPN, may underlie the prolonged and variable time-course of this conductance in these neurons.


Subject(s)
Autonomic Fibers, Preganglionic/physiology , Membrane Potentials/physiology , Potassium Channels, Voltage-Gated/physiology , Sympathetic Nervous System/physiology , 4-Aminopyridine/pharmacology , Animals , Autonomic Fibers, Preganglionic/drug effects , Autonomic Fibers, Preganglionic/metabolism , Female , In Vitro Techniques , Male , Membrane Potentials/drug effects , Patch-Clamp Techniques , Potassium Channels, Voltage-Gated/biosynthesis , Rats , Rats, Inbred WKY , Reverse Transcriptase Polymerase Chain Reaction , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/metabolism , Venoms/pharmacology
18.
Protein Expr Purif ; 76(2): 205-10, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21087668

ABSTRACT

MiRP1 (MinK related protein 1) is a membrane protein in the KCNE family. It can associate with and modulate various voltage gated potassium channels. Mutations in human MiRP1 have been found to cause many congenital and acquired long QT syndromes, which are potentially life-threatening cardiac arrhythmias. Here, human MiRP1 was over-expressed in Escherichia coli, purified and eluted into different detergents. Two dimensional (1)H-(15)N correlated solution nuclear magnetic resonance (NMR) spectra of the human MiRP1 in four different detergent micelles indicated that high resolution solution NMR spectrum can be obtained for human MiRP1 in detergent lyso-myristoylphosphatidylglycerol (LMPG). Circular dichroism (CD) spectroscopy of human MiRP1 indicated a high content of alpha-helical secondary structure in LMPG. Backbone assignments of most MiRP1 residues were achieved through a series of triple resonance NMR experiments. Secondary structure analysis based on backbone chemical shifts showed several stretches of alpha-helices along the primary sequence of MiRP1 in LMPG.


Subject(s)
Potassium Channels, Voltage-Gated/chemistry , Recombinant Proteins/chemistry , Circular Dichroism , Cloning, Molecular , Detergents/chemistry , Electrophoresis, Polyacrylamide Gel , Escherichia coli/genetics , Escherichia coli/metabolism , Humans , Hydrogen-Ion Concentration , Nuclear Magnetic Resonance, Biomolecular/methods , Phosphatidylglycerols/chemistry , Potassium Channels, Voltage-Gated/biosynthesis , Potassium Channels, Voltage-Gated/genetics , Potassium Channels, Voltage-Gated/isolation & purification , Protein Conformation , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification
19.
Dev Neurobiol ; 70(13): 884-96, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20669322

ABSTRACT

A precise organization of gene expression is required for developing neural and muscular systems. Steroid hormones can control the expression of genes that are critical for development. In this study we test the hypothesis that the steroid hormone ecdysone regulates gene expression of the voltage-gated calcium-activated potassium ion channel, Slowpoke or KCNMA1. Late in adult development of the tobacco hawkmoth Manduca sexta, slowpoke (msslo) levels increased contributing to the maturation of the dorsal longitudinal flight muscles (DLMs) and CNS. We show that critical components of ecdysteroid gene regulation were present during upreglation of msslo in late adult DLM and CNS development. Ecdysteroid receptor complex heterodimeric partner proteins, the ecdysteroid receptor (EcR) and ultraspiracle (USP), and the ecdysone-induced early gene, msE75B, were expressed at key developmental time points, suggesting that ecdysteroids direct aspects of gene expression in the DLMs during these late developmental stages. We provide evidence that ecdysteroids suppress msslo transcription in the DLMs; when titers decline msslo transcript levels increase. These results are consistent with msslo being a downstream gene in an ecdysteroid-mediated gene cascade during DLM development. We also show that the ecdysteroids regulate msslo transcript levels in the developing CNS. These results will contribute to our understanding of how the spatiotemporal regulation of slowpoke transcription contributes to tailoring cell excitability to the differing physiological and behavioral demands during development.


Subject(s)
Muscle, Striated/growth & development , Nervous System/growth & development , Potassium Channels, Voltage-Gated/biosynthesis , Potassium Channels, Voltage-Gated/genetics , Steroids/physiology , Animals , Ecdysone/physiology , Gene Expression Regulation, Developmental/physiology , Manduca , Muscle, Striated/cytology , Muscle, Striated/metabolism , Nervous System/cytology , Nervous System/metabolism
20.
Biochem Biophys Res Commun ; 395(3): 330-5, 2010 May 07.
Article in English | MEDLINE | ID: mdl-20381460

ABSTRACT

AIMS: To investigate dynamic changes in the expression of HCN2, HCN4, as well as KCNE1, and KCNE2 mRNA and protein levels in ventricular cells from acute myocardial infarction (AMI) rat hearts. MAIN METHODS: An AMI model was induced by ligating the left anterior descending coronary artery (LAD) of Sprague-Dawley rats. The rats were randomly divided into four experimental groups: 24-hour (24h) post-AMI, 1-week (1w) post-AMI, 2-week (2w) post-AMI, and 4-week (4w) post-AMI; sham-operated control rat groups were established in parallel for each time point. HCN2, HCN4, KCNE1, and KCNE2 mRNA and protein levels were measured by reverse transcription-polymerase chain reaction (RT-PCR) and by immunohistochemistry and Western blot, respectively. KEY FINDINGS: Ventricular arrhythmias occurred in all the post-AMI groups, particularly in the 1w and 2w post-AMI groups. Although HCN2, HCN4, KCNE1, and KCNE2 genes were expressed in the left ventricular myocardium of sham-operated control rats, their expression increased in rat ischemic left ventricular myocardium, with dynamic changes in expression observed 4 weeks after AMI. HCN2, HCN4, and KCNE2 protein levels were highest at 1w and KCNE2 protein levels peaked at 2w post-AMI. SIGNIFICANCE: The expression of the HCN2, HCN4, as well as KCNE1, and KCNE2 genes in ventricular cells from AMI rat hearts underwent dynamic changes, reaching peak levels at 1 or 2weeks post-AMI. The increased expression maybe related to ventricular arrhythmogenesis after AMI.


Subject(s)
Heart Ventricles/metabolism , Ion Channels/biosynthesis , Myocardial Infarction/metabolism , Potassium Channels, Voltage-Gated/biosynthesis , Potassium Channels/biosynthesis , Animals , Disease Models, Animal , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Ion Channels/genetics , Male , Potassium Channels/genetics , Potassium Channels, Voltage-Gated/genetics , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...