Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Oncol Rep ; 45(5)2021 05.
Article in English | MEDLINE | ID: mdl-34105734

ABSTRACT

The aim of the present study was to explore and verify the potential mechanism of seminoma progression. Data on 132 RNA­seq and 156 methylation sites from stage II/III and I seminoma specimens were downloaded from The Cancer Genome Atlas database. An initial filter of |fold­change| >2 and false discovery rate <0.05 were used to identify differentially expressed genes (DEGs) which were associated with differential methylation site genes; these genes were considered potential candidates for further investigation by survival analysis. Potassium voltage­gated channel subfamily C member 1 (KCNC1) expression was verified in seminoma human tissues and three seminoma cell lines. The invasive, proliferative and apoptotic abilities of the human testicular tumor Ntera­2 and normal human testis Hs1.Tes cell lines were assessed following aberrant KCNC1 expression. KCNC1 was identified as a DEG, in which hypermethylation inhibited its expression and it was associated with poor overall survival in patients with seminoma. The present results demonstrated that KCNC1 is negatively correlated with methylation. Due to the abnormal expression of KCNC1 in seminoma cells, it was suggested that KCNC1 could be used as a diagnostic indicator and therapeutic target for the progression of seminoma.


Subject(s)
DNA Methylation , Seminoma/genetics , Shaw Potassium Channels/genetics , Testicular Neoplasms/genetics , Adult , Apoptosis/genetics , Cell Proliferation/genetics , Gene Knockout Techniques , Humans , Immunohistochemistry , Male , Neoplasm Invasiveness , Neoplasm Metastasis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Seminoma/metabolism , Seminoma/mortality , Seminoma/pathology , Shaw Potassium Channels/biosynthesis , Survival Rate , Testicular Neoplasms/metabolism , Testicular Neoplasms/mortality , Testicular Neoplasms/pathology , Transfection
2.
FASEB J ; 35(1): e21241, 2021 01.
Article in English | MEDLINE | ID: mdl-33368632

ABSTRACT

The voltage-gated potassium channel Kv3.4 is a crucial regulator of nociceptive signaling in the dorsal root ganglion (DRG) and the dorsal horn of the spinal cord. Moreover, Kv3.4 dysfunction has been linked to neuropathic pain. Although kinases and phosphatases can directly modulate Kv3.4 gating, the signaling mechanisms regulating the expression and stability of the Kv3.4 protein are generally unknown. We explored a potential role of PKCε and found an unexpected interaction that has a positive effect on Kv3.4 expression. Co-immunoprecipitation studies revealed a physical association between PKCε and Kv3.4 in both heterologous cells and rat DRG neurons. Furthermore, in contrast to the wild-type and constitutively active forms of PKCε, expression of a catalytically inactive form of the enzyme inhibits Kv3.4 expression and membrane localization through a dominant negative effect. Co-expression of Kv3.4 with the wild-type, constitutively active, or catalytically inactive forms of PKCε had no significant effects on Kv3.4 gating. These results suggest that a novel physical interaction of the Kv3.4 channel with functional PKCε primarily determines its stability and localization in DRG neurons. This interaction is akin to those of previously identified accessory ion channel proteins, which could be significant in neural tissues where Kv3.4 regulates electrical signaling.


Subject(s)
Ganglia, Spinal/metabolism , Gene Expression Regulation , Neurons/metabolism , Protein Kinase C-epsilon/metabolism , Shaw Potassium Channels/biosynthesis , Animals , CHO Cells , Cricetulus , HEK293 Cells , Humans , Protein Kinase C-epsilon/genetics , Rats , Shaw Potassium Channels/genetics
3.
BMC Pulm Med ; 20(1): 260, 2020 Oct 08.
Article in English | MEDLINE | ID: mdl-33032555

ABSTRACT

BACKGROUND: Hypoxic pulmonary vasoconstriction (HPV) is a reaction of the pulmonary vasculature upon hypoxia, diverting blood flow into ventilated areas to preserve oxygenation. It is impaired in endotoxemia or ARDS. Voltage gated potassium channels have been shown to play a key role in the regulation of HPV. The aim of the study was to identify a voltage gated potassium channel involved in dysregulated HPV during endotoxemia. METHODS: Lungs of male C57BL/6 mice with and without endotoxemia (n = 6 ea. group) were analyzed for Kv3.4 gene and protein expression. HPV was examined in isolated perfused lungs of mice with and without endotoxemia and with and without selective Kv3.4 blocker BDS-I (n = 7 ea. group). Pulmonary artery pressure (PAP) and pressure-flow curves were measured during normoxic (FiO2 0.21) and hypoxic (FiO2 0.01) ventilation. HPV was quantified as the increase in perfusion pressure in response to hypoxia in percent of baseline perfusion pressure (ΔPAP) in the presence and absence of BDS-I. RESULTS: Kv3.4 gene (3.2 ± 0.5-fold, p < 0.05) and protein (1.5 ± 0.1-fold p < 0.05) expression levels were increased in endotoxemic mouse lungs. Endotoxemia reduced HPV (∆PAP control: 121.2 ± 8.7% vs. LPS 19.5 ± 8.0%, means ± SEM) while inhibition of Kv3.4 with 50 nM BDS-I augmented HPV in endotoxemic but not in control lungs (∆PAP control BDS-I: 116.6 ± 16.0% vs. LPS BDS-I 84.4 ± 18.2%, means ± SEM). CONCLUSIONS: Kv3.4 gene and protein expressions are increased in endotoxemic mouse lungs. Selective inhibition of Kv3.4 augments HPV in lungs of endotoxemic mice, but not in lungs of control mice.


Subject(s)
Endotoxemia/metabolism , Hypoxia/metabolism , Lung/metabolism , Shaw Potassium Channels/biosynthesis , Vasoconstriction/physiology , Animals , Endotoxemia/pathology , Hypoxia/pathology , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Lung/blood supply , Lung/drug effects , Male , Mice , Mice, Inbred C57BL , Organ Culture Techniques , Perfusion , Pulmonary Artery/drug effects , Pulmonary Artery/physiopathology , Pulmonary Circulation/drug effects , Shaw Potassium Channels/antagonists & inhibitors , Vasoconstriction/drug effects
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.
Sci Rep ; 6: 33043, 2016 09 14.
Article in English | MEDLINE | ID: mdl-27623749

ABSTRACT

Obesity is a major risk factor for type 2 diabetes. To unravel the genetic determinants of obesity-associated diabetes, we performed a genome-wide study using the 1,000 Genomes-based imputation in a Korean childhood cohort (KoCAS-1, n = 484) and carried out de novo replication in an independent population (KoCAS-2, n = 1,548). A novel variant (rs10879834) with multiple diverse associations for obesity-related traits was also found to be replicated in an adult cohort (KARE, n = 8,842). Functional annotations using integrative epigenetic analyses identified biological significance and regulatory effects with an inverse methylation-expression correlation (cg27154343 in the 5'-UTR of the KCNC2 gene), tissue-specific enhancer mark (H3K4me1), and pathway enrichment (insulin signaling). Further functional studies in cellular and mouse models demonstrated that KCNC2 is associated with anti-obesogenic effects in the regulation of obesity-induced insulin resistance. KCNC2 shRNA transfection induced endoplasmic reticulum (ER) stress and hepatic gluconeogenesis. Overproduction of KCNC2 decreased ER stress, and treatment with metformin enhanced KCNC2 expression. Taken together, these data suggest that reduction of KCNC2 is associated with modified hepatic gluconeogenesis and increased ER stress on obesity-mediated diabetic risk. An integrative multi-omics analysis might reveal new functional and clinical implications related to the control of energy and metabolic homeostasis in humans.


Subject(s)
5' Untranslated Regions , DNA Methylation , Diabetes Mellitus , Genetic Predisposition to Disease , Obesity , Quantitative Trait, Heritable , Shaw Potassium Channels , Adolescent , Animals , Child , Diabetes Mellitus/epidemiology , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Female , Humans , Male , Mice , Obesity/epidemiology , Obesity/genetics , Obesity/metabolism , Republic of Korea , Risk Factors , Shaw Potassium Channels/biosynthesis , Shaw Potassium Channels/genetics
6.
PLoS One ; 11(2): e0148633, 2016.
Article in English | MEDLINE | ID: mdl-26849432

ABSTRACT

Voltage-gated K+ (Kv) channels are well known to be involved in cell proliferation. However, even though cell proliferation is closely related to cell differentiation, the relationship between Kv channels and cell differentiation remains poorly investigated. This study demonstrates that Kv3.3 is involved in K562 cell erythroid differentiation. Down-regulation of Kv3.3 using siRNA-Kv3.3 increased hemin-induced K562 erythroid differentiation through decreased activation of signal molecules such as p38, cAMP response element-binding protein, and c-fos. Down-regulation of Kv3.3 also enhanced cell adhesion by increasing integrin ß3 and this effect was amplified when the cells were cultured with fibronectin. The Kv channels, or at least Kv3.3, appear to be associated with cell differentiation; therefore, understanding the mechanisms of Kv channel regulation of cell differentiation would provide important information regarding vital cellular processes.


Subject(s)
Cell Differentiation/drug effects , Down-Regulation/drug effects , Hemin/pharmacology , Response Elements/physiology , Shaw Potassium Channels/biosynthesis , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Differentiation/genetics , Down-Regulation/genetics , Fibronectins/pharmacology , Humans , Integrin beta3/biosynthesis , K562 Cells , Shaw Potassium Channels/genetics
7.
J Comp Neurol ; 520(16): 3650-72, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22473424

ABSTRACT

Precise axon pathfinding is crucial for establishment of the initial neuronal network during development. Pioneer axons navigate without the help of preexisting axons and pave the way for follower axons that project later. Voltage-gated ion channels make up the intrinsic electrical activity of pioneer axons and regulate axon pathfinding. To elucidate which channel molecules are present in pioneer axons, immunohistochemical analysis was performed to examine 14 voltage-gated ion channels (Kv1.1-Kv1.3, Kv3.1-Kv3.4, Kv4.3, Cav1.2, Cav1.3, Cav2.2, Nav1.2, Nav1.6, and Nav1.9) in nine axonal tracts in the developing rat forebrain, including the optic nerve, corpus callosum, corticofugal fibers, thalamocortical axons, lateral olfactory tract, hippocamposeptal projection, anterior commissure, hippocampal commissure, and medial longitudinal fasciculus. We found A-type K⁺ channel Kv3.4 in both pioneer axons and early follower axons and L-type Ca²âº channel Cav1.2 in pioneer axons and early and late follower axons. Spatially, Kv3.4 and Cav1.2 were colocalized with markers of pioneer neurons and pioneer axons, such as deleted in colorectal cancer (DCC), in most fiber tracts examined. Temporally, Kv3.4 and Cav1.2 were expressed abundantly in most fiber tracts during axon pathfinding but were downregulated beginning in synaptogenesis. By contrast, delayed rectifier Kv channels (e.g., Kv1.1) and Nav channels (e.g., Nav1.2) were absent from these fiber tracts (except for the corpus callosum) during pathfinding of pioneer axons. These data suggest that Kv3.4 and Cav1.2, two high-voltage-activated ion channels, may act together to control Ca²âº -dependent electrical activity of pioneer axons and play important roles during axon pathfinding.


Subject(s)
Calcium Channels, L-Type/biosynthesis , Prosencephalon/embryology , Shaw Potassium Channels/biosynthesis , Animals , Cell Movement , Female , Fluorescent Antibody Technique , Immunohistochemistry , Male , Microscopy, Confocal , Neurogenesis/physiology , Prosencephalon/metabolism , Rats , Rats, Sprague-Dawley
8.
J Mol Neurosci ; 46(3): 606-15, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21912965

ABSTRACT

In neurons, voltage-dependent Kv3 potassium channels are essential for the generation of action potentials at high frequency. A dysregulation of the Kv3.1 and Kv3.4 channel subunits has been suggested to contribute to neuronal and glial alterations in Alzheimer's disease, but a quantitative evaluation of these subunits in a mouse model of the pathology is still lacking. We analysed the profile of expression of the four Kv3 subunits by quantitative reverse transcription PCR and Western blot in the whole mouse brain and in dissected brain regions (olfactory bulb, septum, neocortex, hippocampus, brainstem and cerebellum) from 14 days after conception to 18 months after birth. In addition, we measured the levels of Kv3.1 and Kv3.4 messenger RNAs (mRNAs) and proteins in neocortex and hippocampus of APPPS1 mice, a transgenic model of Alzheimer's disease. Although all Kv3 transcripts were significantly expressed in embryonic age in whole brain extracts, only Kv3.1, Kv3.2 and Kv3.4 subunit proteins were present, suggesting a novel role for Kv3 channels at this developmental stage. With the exception of Kv3.4, during postnatal development, Kv3 transcripts and proteins showed a progressive increase in expression and reached an asymptote in adulthood, suggesting that the increase in Kv3 expression during development might contribute to the maturation of the electrical activity of neurons. During aging, Kv3 expression was rather stable. In contrast, in the neocortex of aged APPPS1 mice, Kv3.1 mRNA and protein levels were significantly lower compared to wild type, suggesting that a decrease in Kv3 currents could play a role in the cognitive symptoms of Alzheimer's disease.


Subject(s)
Aging/genetics , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Gene Expression Regulation, Developmental/physiology , Shaw Potassium Channels/genetics , Alzheimer Disease/pathology , Animals , Disease Models, Animal , Female , Mice , Mice, Inbred Strains , Mice, Transgenic , Shaw Potassium Channels/biosynthesis
9.
J Biol Chem ; 287(3): 1755-69, 2012 Jan 13.
Article in English | MEDLINE | ID: mdl-22105078

ABSTRACT

Synaptic inputs received at dendrites are converted into digital outputs encoded by action potentials generated at the axon initial segment in most neurons. Here, we report that alternative splicing regulates polarized targeting of Kv3.1 voltage-gated potassium (Kv) channels to adjust the input-output relationship. The spiking frequency of cultured hippocampal neurons correlated with the level of endogenous Kv3 channels. Expression of axonal Kv3.1b, the longer form of Kv3.1 splice variants, effectively converted slow-spiking young neurons to fast-spiking ones; this was not the case for Kv1.2 or Kv4.2 channel constructs. Despite having identical biophysical properties as Kv3.1b, dendritic Kv3.1a was significantly less effective at increasing the maximal firing frequency. This suggests a possible role of channel targeting in regulating spiking frequency. Mutagenesis studies suggest the electrostatic repulsion between the Kv3.1b N/C termini, created by its C-terminal splice domain, unmasks the Kv3.1b axonal targeting motif. Kv3.1b axonal targeting increased the maximal spiking frequency in response to prolonged depolarization. This finding was further supported by the results of local application of channel blockers and computer simulations. Taken together, our studies have demonstrated that alternative splicing controls neuronal firing rates by regulating the polarized targeting of Kv3.1 channels.


Subject(s)
Alternative Splicing/physiology , Axons/metabolism , Dendrites/metabolism , Hippocampus/metabolism , Nerve Tissue Proteins/biosynthesis , Shaw Potassium Channels/biosynthesis , Animals , HEK293 Cells , Humans , Mutagenesis , Nerve Tissue Proteins/genetics , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Protein Structure, Tertiary , Rats , Shaw Potassium Channels/genetics
10.
J Chem Neuroanat ; 42(1): 30-8, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21440618

ABSTRACT

GABAergic interneurones, including those within spinal dorsal horn, contain one of the two isoforms of the synthesizing enzyme glutamate decarboxylase (GAD), either GAD65 or GAD67. The physiological significance of these two GABAergic phenotypes is unknown but a more detailed anatomical and functional characterization may help resolve this issue. In this study, two transgenic Green Fluorescent Protein (GFP) knock-in murine lines, namely GAD65-GFP and GAD67-GFP (Δneo) mice, were used to profile expression of Shaw-related Kv3.1b and Kv3.3 K(+)-channel subunits in dorsal horn interneurones. Neuronal expression of these subunits confers specific biophysical characteristic referred to as 'fast-spiking'. Immuno-labelling for Kv3.1b or Kv3.3 revealed the presence of both of these subunits across the dorsal horn, most abundantly in laminae I-III. Co-localization studies in transgenic mice indicated that Kv3.1b but not Kv3.3 was associated with GAD65-GFP and GAD67-GFP immunopositive neurones. For comparison the distributions of Kv4.2 and Kv4.3 K(+)-channel subunits which are linked to an excitatory neuronal phenotype were characterized. No co-localization was found between GAD-GFP +ve neurones and Kv4.2 or Kv4.3. In functional studies to evaluate whether either GABAergic population is activated by noxious stimulation, hindpaw intradermal injection of capsaicin followed by c-fos quantification in dorsal horn revealed co-expression c-fos and GAD65-GFP (quantified as 20-30% of GFP +ve population). Co-expression was also detected for GAD67-GFP +ve neurones and capsaicin-induced c-fos but at a much reduced level of 4-5%. These data suggest that whilst both GAD65-GFP and GAD67-GFP +ve neurones express Kv3.1b and therefore may share certain biophysical traits, their responses to peripheral noxious stimulation are distinct.


Subject(s)
Interneurons/metabolism , Posterior Horn Cells/metabolism , Shaw Potassium Channels/biosynthesis , Animals , Gene Knock-In Techniques , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pain/metabolism , Pain/physiopathology , gamma-Aminobutyric Acid/metabolism
11.
Neurol Sci ; 32(4): 571-7, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21234782

ABSTRACT

The expression of voltage-gated potassium channels belonging to the Kv3 family has been studied in the sensori-motor cortex of rats exposed to alcohol inhalation during the first postnatal week (P2-P6). The study was carried out using comparative RT-PCR. At P9, a significant reduction of the expression of Kv3.2 and Kv3.4 subunits occurred in alcohol-treated animals, as compared with controls. The expression of the Kv3.4a splicing variant, which is thought to be critically involved in the high-frequency firing of some cortical interneurons, was also correspondingly reduced. The downregulation of Kv3.2 and Kv3.4a subunits represented a long-lasting effect of alcohol exposure, since it was also observed in P24 animals. The expression of both Kv3.1 and Kv3.3 channels appeared to be not significantly affected by alcohol exposure. An increased susceptibility to apoptotic neuronal death after early postnatal exposure to ethanol was confirmed by the lower bcl-2/bax ratio observed in alcohol-treated animals. Although Kv3.4 subunits are thought to trigger apoptosis, the lack of upregulation in our model argues against their involvement in the mechanism leading to alcohol-induced apoptosis. The possible consequences of the selective downregulation of Kv3 subunits on the cortical function, as well as their relevance for the genesis of fetal alcohol effects, are discussed.


Subject(s)
Central Nervous System Depressants/toxicity , Cerebral Cortex/metabolism , Ethanol/toxicity , Shaw Potassium Channels/biosynthesis , Animals , Animals, Newborn , Apoptosis/genetics , Cerebral Cortex/drug effects , DNA Primers , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Electrophysiological Phenomena , Female , Fetal Alcohol Spectrum Disorders/metabolism , Male , Neurons/physiology , Pregnancy , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
12.
J Pathol ; 221(4): 402-10, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20593490

ABSTRACT

The concept of ion channels as membrane therapeutic targets and diagnostic/prognostic biomarkers has attracted growing attention. We therefore investigated the expression pattern and clinical significance of the Kv3.4 potassium channel subunit during the development and progression of head and neck squamous cell carcinomas (HNSCCs). KCNC4 mRNA levels were determined by real-time RT-PCR in both HNSCC tissue specimens and derived cell lines. Kv3.4 protein expression was evaluated by immunohistochemistry in paraffin-embedded tissue specimens from 84 patients with laryngeal/pharyngeal squamous cell carcinomas and 67 patients with laryngeal dysplasias. Molecular alterations were correlated with clinicopathological parameters and patient outcome. Increased KCNC4 mRNA levels were found in 15 (54%) of 28 tumours, compared to the corresponding normal epithelia and varied mRNA levels were detected in 12 HNSCC-derived cell lines analysed. Increased Kv3.4 protein expression was observed in 34 (40%) of 84 carcinomas and also at early stages of HNSCC tumourigenesis. Thus, 35 (52%) of 67 laryngeal lesions displayed Kv3.4-positive staining in the dysplastic areas, whereas both stromal cells and normal adjacent epithelia exhibited negligible expression. No significant correlations were found between Kv3.4-positive expression in HNSCC and clinical data; however, Kv3.4 expression tended to diminish in advanced-stage tumours. Interestingly, patients carrying Kv3.4-positive dysplasias experienced a significantly higher laryngeal cancer incidence than did those with negative lesions (p = 0.0209). In addition, functional studies using HNSCC cells revealed that inhibition of Kv3.4 expression by siRNA leads to the inhibition of cell proliferation via selective cell cycle arrest at the G2/M phase without affecting apoptosis. Collectively, these data demonstrate for the first time that Kv3.4 expression is frequently increased during HNSCC tumourigenesis and correlated significantly with a higher cancer risk. Our findings support a role for Kv3.4 in malignant transformation and provide original evidence for the potential clinical utility of Kv3.4 expression as a biomarker for cancer risk assessment.


Subject(s)
Biomarkers, Tumor/biosynthesis , Carcinoma, Squamous Cell/metabolism , Head and Neck Neoplasms/metabolism , Shaw Potassium Channels/biosynthesis , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/pathology , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Disease Progression , Epidemiologic Methods , Female , Gene Expression , Head and Neck Neoplasms/pathology , Humans , Male , Middle Aged , Neoplasm Invasiveness , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Staging , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Prognosis , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Shaw Potassium Channels/genetics , Tumor Cells, Cultured
13.
J Neurosci ; 29(50): 15735-44, 2009 Dec 16.
Article in English | MEDLINE | ID: mdl-20016089

ABSTRACT

The role of cerebellar Kv3.1 and Kv3.3 channels in motor coordination was examined with an emphasis on the deep cerebellar nuclei (DCN). Kv3 channel subunits encoded by Kcnc genes are distinguished by rapid activation and deactivation kinetics that support high-frequency, narrow action potential firing. Previously we reported that increased lateral deviation while ambulating and slips while traversing a narrow beam of ataxic Kcnc3-null mice were corrected by restoration of Kv3.3 channels specifically to Purkinje cells, whereas Kcnc3-mutant mice additionally lacking one Kcnc1 allele were partially rescued. Here, we report mice lacking all Kcnc1 and Kcnc3 alleles exhibit no such rescue. For Purkinje cell output to reach the rest of the brain it must be conveyed by neurons of the DCN or vestibular nuclei. As Kcnc1, but not Kcnc3, alleles are lost, mutant mice exhibit increasing gait ataxia accompanied by spike broadening and deceleration in DCN neurons, suggesting the facet of coordination rescued by Purkinje-cell-restricted Kv3.3 restoration in mice lacking just Kcnc3 is hypermetria, while gait ataxia emerges when additionally Kcnc1 alleles are lost. Thus, fast repolarization in Purkinje cells appears important for normal movement velocity, whereas DCN neurons are a prime candidate locus where fast repolarization is necessary for normal gait patterning.


Subject(s)
Gene Targeting , Psychomotor Performance/physiology , Purkinje Cells/physiology , Shaw Potassium Channels/deficiency , Shaw Potassium Channels/genetics , Shaw Potassium Channels/physiology , Action Potentials/genetics , Alleles , Animals , Cerebellar Nuclei/metabolism , Cerebellar Nuclei/pathology , Female , Gait Ataxia/genetics , Gait Ataxia/pathology , Male , Mice , Mice, Knockout , Purkinje Cells/cytology , Purkinje Cells/pathology , Shaw Potassium Channels/biosynthesis
14.
J Gen Physiol ; 133(4): 361-74, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19332619

ABSTRACT

Crystal structures of potassium (K(+)) channels reveal that the selectivity filter, the narrow portion of the pore, is only approximately 3-A wide and buttressed from behind, so that its ability to expand is highly constrained, and the permeation of molecules larger than Rb(+) (2.96 A in diameter) is prevented. N-methyl-d-glucamine (NMDG(+)), an organic monovalent cation, is thought to be a blocker of Kv channels, as it is much larger (approximately 7.3 A in mean diameter) than K(+) (2.66 A in diameter). However, in the absence of K(+), significant NMDG(+) currents could be recorded from human embryonic kidney cells expressing Kv3.1 or Kv3.2b channels and Kv1.5 R487Y/V, but not wild-type channels. Inward currents were much larger than outward currents due to the presence of intracellular Mg(2+) (1 mM), which blocked the outward NMDG(+) current, resulting in a strong inward rectification. The NMDG(+) current was inhibited by extracellular 4-aminopyridine (5 mM) or tetraethylammonium (10 mM), and largely eliminated in Kv3.2b by an S6 mutation that prevents the channel from opening (P468W) and by a pore helix mutation in Kv1.5 R487Y (W472F) that inactivates the channel at rest. These data indicate that NMDG(+) passes through the open ion-conducting pore and suggest a very flexible nature of the selectivity filter itself. 0.3 or 1 mM K(+) added to the external NMDG(+) solution positively shifted the reversal potential by approximately 16 or 31 mV, respectively, giving a permeability ratio for K(+) over NMDG(+) (P(K)(+)/P(NMDG)(+)) of approximately 240. Reversal potential shifts in mixtures of K(+) and NMDG(+) are in accordance with P(K)(+)/P(NMDG)(+), indicating that the ions compete for permeation and suggesting that NMDG(+) passes through the open state. Comparison of the outer pore regions of Kv3 and Kv1.5 channels identified an Arg residue in Kv1.5 that is replaced by a Tyr in Kv3 channels. Substituting R with Y or V allowed Kv1.5 channels to conduct NMDG(+), suggesting a regulation by this outer pore residue of Kv channel flexibility and, as a result, permeability.


Subject(s)
Amino Acid Substitution , Ion Channel Gating/physiology , Meglumine/metabolism , Potassium Channels, Voltage-Gated/metabolism , Amino Acid Substitution/genetics , Animals , Arginine/genetics , Cell Line , Humans , Ion Channel Gating/genetics , Mutation , Permeability , Potassium Channel Blockers/metabolism , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Potassium Channels, Voltage-Gated/genetics , Rats , Shaw Potassium Channels/antagonists & inhibitors , Shaw Potassium Channels/biosynthesis , Shaw Potassium Channels/genetics , Shaw Potassium Channels/metabolism , Tyrosine/genetics , Valine/genetics
15.
Eur J Pharmacol ; 587(1-3): 187-95, 2008 Jun 10.
Article in English | MEDLINE | ID: mdl-18430420

ABSTRACT

We have previously reported that subacute hypoxia activates lung 15-lipoxygenase (15-LOX), which catalyzes arachidonic acid to produce 15-HETE, leading to constriction of neonatal rabbit pulmonary arteries. Subacute hypoxia suppresses Kv3.4 channel expression and results in an inhibition of whole-cell K(+) currents (I(K)). Although the Kv channel inhibition is likely to be mediated through 15-HETE, direct evidence is still lacking. To reveal the role of the 15-LOX/15-HETE pathway in the hypoxia-induced down-regulation of Kv3.4 channel expression and inhibition of I(K), we performed studies using 15-LOX blockers, whole-cell patch-clamp, semi-quantitative PCR, ELISA and Western blot analysis. We found that Kv3.4 channel expression at the mRNA and protein levels was greatly up-regulated in pulmonary arterial smooth muscle cells after blockade of 15-LOX by CDC or NDGA. The 15-LOX blockade also partially restored I(K). In comparison, 15-HETE had a stronger effect than 12-HETE on the expression of Kv3.4 channels. 5-HETE had no noticeable effect on Kv3.4 channel expression. These data indicate that the 15-LOX pathway via its metabolite, 15-HETE, seems to play a role in the down-regulation of Kv3.4 expression and I(K) inhibition after subacute hypoxia.


Subject(s)
Hydroxyeicosatetraenoic Acids/physiology , Hypoxia/metabolism , Myocytes, Smooth Muscle/physiology , Pulmonary Artery/physiology , Shaw Potassium Channels/biosynthesis , Animals , Arachidonate 15-Lipoxygenase/metabolism , Blotting, Western , Calcium/metabolism , Cell Separation , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Hydroxyeicosatetraenoic Acids/metabolism , Indicators and Reagents , Myocytes, Smooth Muscle/metabolism , Patch-Clamp Techniques , Pulmonary Artery/cytology , Pulmonary Artery/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Shaw Potassium Channels/genetics
16.
J Neurosci ; 27(37): 9855-65, 2007 Sep 12.
Article in English | MEDLINE | ID: mdl-17855600

ABSTRACT

A-type K+ channels (A-channels) are crucial in controlling neuronal excitability, and their downregulation in pain-sensing neurons may increase pain sensation. To test this hypothesis, we first characterized the expression of two A-channels, Kv3.4 and Kv4.3, in rat dorsal root ganglion (DRG) neurons. Kv3.4 was expressed mainly in the nociceptive DRG neurons, in their somata, axons, and nerve terminals innervating the dorsal horn of spinal cord. In contrast, Kv4.3 appeared selectively in the somata of a subset of nonpeptidergic nociceptive DRG neurons. Most Kv4.3(+) DRG neurons also expressed Kv3.4. In a neuropathic pain model induced by spinal nerve ligation in rats, the protein levels of Kv3.4 and Kv4.3 in the DRG neurons were greatly reduced. After Kv3.4 or Kv4.3 expression in lumbar DRG neurons was suppressed by intrathecal injections of antisense oligodeoxynucleotides, mechanical but not thermal hypersensitivity developed. Together, our data suggest that reduced expression of A-channels in pain-sensing neurons may induce mechanical hypersensitivity, a major symptom of neuropathic pain.


Subject(s)
Neurons, Afferent/metabolism , Pain/metabolism , Shal Potassium Channels/antagonists & inhibitors , Shal Potassium Channels/biosynthesis , Shaw Potassium Channels/antagonists & inhibitors , Shaw Potassium Channels/biosynthesis , Animals , Gene Expression Regulation/physiology , Male , Pain/genetics , Pain Measurement/methods , Physical Stimulation/methods , Rats , Rats, Sprague-Dawley , Shal Potassium Channels/genetics , Shaw Potassium Channels/genetics , Spinal Cord/metabolism
17.
J Neurosci ; 25(50): 11531-41, 2005 Dec 14.
Article in English | MEDLINE | ID: mdl-16354911

ABSTRACT

Pyramidal neurons of the electrosensory lateral line lobe (ELL) of Apteronotus leptorhynchus express Kv3-type voltage-gated potassium channels that give rise to high-threshold currents at the somatic and dendritic levels. Two members of the Kv3 channel family, AptKv3.1 and AptKv3.3, are coexpressed in these neurons. AptKv3.3 channels are expressed at uniformly high levels in each of four ELL segments, whereas AptKv3.1 channels appear to be expressed in a graded manner with higher levels of expression in segments that process high-frequency electrosensory signals. Immunohistochemical and recombinant channel expression studies show a differential distribution of these two channels in the dendrites of ELL pyramidal neurons. AptKv3.1 is concentrated in somas and proximal dendrites, whereas AptKv3.3 is distributed throughout the full extent of the large dendritic tree. Recombinant channel expression of AptKv3 channels through in vivo viral injections allowed directed retargeting of AptKv3 subtypes over the somadendritic axis, revealing that the sequence responsible for targeting channels to distal dendrites lies within the C-terminal domain of the AptKv3.3 protein. The targeting domain includes a consensus sequence predicted to bind to a PDZ (postsynaptic density-95/Discs large/zona occludens-1)-type protein-protein interaction motif. These findings reveal that different functional roles for Kv3 potassium channels at the somatic and dendritic level of a sensory neuron are attained through specific targeting that selectively distributes Kv3.3 channels to the dendritic compartment.


Subject(s)
Dendrites/physiology , Fish Proteins/physiology , Neurons, Afferent/physiology , Peptide Fragments/physiology , Shaw Potassium Channels/physiology , Amino Acid Sequence , Animals , Cell Line , Cricetinae , Dendrites/genetics , Drosophila , Fish Proteins/biosynthesis , Fish Proteins/genetics , Gymnotiformes , Mice , Molecular Sequence Data , Neurons, Afferent/metabolism , Peptide Fragments/biosynthesis , Peptide Fragments/genetics , Protein Structure, Tertiary/genetics , Rats , Shaw Potassium Channels/biosynthesis , Shaw Potassium Channels/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...