Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 13 de 13
1.
Epilepsia ; 64(2): 443-455, 2023 02.
Article En | MEDLINE | ID: mdl-36318112

OBJECTIVE: Mutations in the genes encoding neuronal ion channels are a common cause of Mendelian neurological diseases. We sought to identify novel de novo sequence variants in cases with early infantile epileptic phenotypes and neurodevelopmental anomalies. METHODS: Following clinical diagnosis, we performed whole exome sequencing of the index cases and their parents. Identified channel variants were expressed in Xenopus oocytes and their functional properties assessed using two-electrode voltage clamp. RESULTS: We identified novel de novo variants in KCNA6 in four unrelated individuals variably affected with neurodevelopmental disorders and seizures with onset in the first year of life. Three of the four identified mutations affect the pore-lining S6 α-helix of KV 1.6. A prominent finding of functional characterization in Xenopus oocytes was that the channel variants showed only minor effects on channel activation but slowed channel closure and shifted the voltage dependence of deactivation in a hyperpolarizing direction. Channels with a mutation affecting the S6 helix display dominant effects on channel deactivation when co-expressed with wild-type KV 1.6 or KV 1.1 subunits. SIGNIFICANCE: This is the first report of de novo nonsynonymous variants in KCNA6 associated with neurological or any clinical features. Channel variants showed a consistent effect on channel deactivation, slowing the rate of channel closure following normal activation. This specific gain-of-function feature is likely to underlie the neurological phenotype in our patients. Our data highlight KCNA6 as a novel channelopathy gene associated with early infantile epileptic phenotypes and neurodevelopmental anomalies.


Epilepsy , Neurodevelopmental Disorders , Humans , Epilepsy/genetics , Mutation/genetics , Seizures/genetics , Kv1.6 Potassium Channel/genetics
2.
BMC Genomics ; 23(1): 227, 2022 Mar 23.
Article En | MEDLINE | ID: mdl-35321643

BACKGROUND: Genetic variants have been found to influence red blood cell (RBC) susceptibility to hemolytic stress and affect transfusion outcomes and the severity of blood diseases. Males have a higher susceptibility to hemolysis than females, but little is known about the genetic mechanism contributing to the difference. RESULTS: To investigate the sex differences in RBC susceptibility to hemolysis, we conducted a sex-stratified genome-wide association study and a genome-wide gene-by-sex interaction scan in a multi-ethnic dataset with 12,231 blood donors who have in vitro osmotic hemolysis measurements during routine blood storage. The estimated SNP-based heritability for osmotic hemolysis was found to be significantly higher in males than in females (0.46 vs. 0.41). We identified SNPs associated with sex-specific susceptibility to osmotic hemolysis in five loci (SPTA1, KCNA6, SLC4A1, SUMO1P1, and PAX8) that impact RBC function and hemolysis. CONCLUSION: Our study established a best practice to identify sex-specific genetic modifiers for sexually dimorphic traits in datasets with mixed ancestries, providing evidence of different genetic regulations of RBC susceptibility to hemolysis between sexes. These and other variants may help explain observed sex differences in the severity of hemolytic diseases, such as sickle cell and malaria, as well as the viability of red cell storage and recovery.


Blood Preservation , Erythrocytes , Hemolysis , Osmotic Pressure , Female , Genome-Wide Association Study , Humans , Kv1.6 Potassium Channel/genetics , Male , Osmosis , Sex Factors
3.
J Neurosci ; 41(44): 9141-9162, 2021 11 03.
Article En | MEDLINE | ID: mdl-34544832

The potassium channel Kv1.6 has recently been implicated as a major modulatory channel subunit expressed in primary nociceptors. Furthermore, its expression at juxtaparanodes of myelinated primary afferents is induced following traumatic nerve injury as part of an endogenous mechanism to reduce hyperexcitability and pain-related hypersensitivity. In this study, we compared two mouse models of constitutive Kv1.6 knock-out (KO) achieved by different methods: traditional gene trap via homologous recombination and CRISPR-mediated excision. Both Kv1.6 KO mouse lines exhibited an unexpected reduction in sensitivity to noxious heat stimuli, to differing extents: the Kv1.6 mice produced via gene trap had a far more significant hyposensitivity. These mice (Kcna6lacZ ) expressed the bacterial reporter enzyme LacZ in place of Kv1.6 as a result of the gene trap mechanism, and we found that their central primary afferent presynaptic terminals developed a striking neurodegenerative phenotype involving accumulation of lipid species, development of "meganeurites," and impaired transmission to dorsal horn wide dynamic range neurons. The anatomic defects were absent in CRISPR-mediated Kv1.6 KO mice (Kcna6-/-) but were present in a third mouse model expressing exogenous LacZ in nociceptors under the control of a Nav1.8-promoted Cre recombinase. LacZ reporter enzymes are thus intrinsically neurotoxic to sensory neurons and may induce pathologic defects in transgenic mice, which has confounding implications for the interpretation of gene KOs using lacZ Nonetheless, in Kcna6-/- mice not affected by LacZ, we demonstrated a significant role for Kv1.6 regulating acute noxious thermal sensitivity, and both mechanical and thermal pain-related hypersensitivity after nerve injury.SIGNIFICANCE STATEMENT In recent decades, the expansion of technologies to experimentally manipulate the rodent genome has contributed significantly to the field of neuroscience. While introduction of enzymatic or fluorescent reporter proteins to label neuronal populations is now commonplace, often potential toxicity effects are not fully considered. We show a role of Kv1.6 in acute and neuropathic pain states through analysis of two mouse models lacking Kv1.6 potassium channels: one with additional expression of LacZ and one without. We show that LacZ reporter enzymes induce unintended defects in sensory neurons, with an impact on behavioral data outcomes. To summarize we highlight the importance of Kv1.6 in recovery of normal sensory function following nerve injury, and careful interpretation of data from LacZ reporter models.


Gene Knockout Techniques/adverse effects , Genes, Reporter , Kv1.6 Potassium Channel/genetics , Lac Operon , Neuralgia/metabolism , Nociceptors/metabolism , Animals , CRISPR-Cas Systems , Female , Gene Knockout Techniques/methods , Integrases/metabolism , Kv1.6 Potassium Channel/metabolism , Male , Mice , Mice, Inbred C57BL , Nociceptors/pathology , Synapses/metabolism , Synapses/pathology
4.
Circ Genom Precis Med ; 14(1): e003011, 2021 02.
Article En | MEDLINE | ID: mdl-33428857

BACKGROUND: The Val122Ile mutation in Transthyretin (TTR) gene causes a rare, difficult to diagnose hereditary form of cardiac amyloidosis. This mutation is most common in the United States and mainly present in people of African descent. The carriers have an increased risk of congestive heart failure, peripheral edema, and several other noncardiac phenotypes such as carpal tunnel syndrome, and arthroplasty which are top reasons for ambulatory/outpatient surgeries (OSs) in the country. METHODS: We conducted first-ever epigenome-wide association study using the Illumina's EPIC array, in Val122Ile carriers of African descent for heart disease and multiple OSs-an early disease indicator. Differential methylation across genome wide cytosine-phosphate guanine (CpG) sites was tested between carriers with and without heart disease and OS. Significant CpG sites were investigated for cis-mQTLs loci, followed by gene ontology and protein-protein interaction network. We also investigated the significant CpG sites in a secondary cohort of carriers for replication. RESULTS: Five differentially methylated sites (P≤2.1×10-8) in genes-FAM129B, SKI, WDR27, GLS, and an intergenic site near RP11-550A5.2, and one differentially methylated region containing KCNA6 and GALNT3 (P=1.1×10-12) were associated with heart disease. For OS, we observe 4 sites-2 sites in UBE2E3 and SEC14L5, and other 2 in intergenic regions (P≤1.8×10-7) and 3 regions overlapping SH3D21, EVA1B, LTB4R2, and CIDEB (P≤3.9×10-7). Functional protein-interaction module analysis identified ABCA1 (P=0.001) for heart disease. Six cis-mQTLs were associated with one of the significant CpG sites (FAM129B; P=4.1×10-24). We replicated 2 CpG sites (cg18546846 and cg06641417; P<0.05) in an external cohort of biopsy-confirmed cases of TTR (transthyretin) amyloidosis. The genes identified are involved in transport and clearance of amyloid deposits (GLS, ABCA1, FAM129B); cardiac fibrosis (SKI); and muscle tissue regulation (SKI, FAM129B). CONCLUSIONS: These findings highlight the link between a complex amyloid circuit and diverse symptoms of Val122Ile.


Amyloidosis/diagnosis , Black or African American/genetics , Epigenomics , Prealbumin/genetics , ATP Binding Cassette Transporter 1/genetics , Amyloidosis/genetics , DNA Methylation , Gene Regulatory Networks/genetics , Genome-Wide Association Study , Heart Diseases/genetics , Heart Diseases/pathology , Heart Diseases/surgery , Humans , Kv1.6 Potassium Channel/genetics , Phosphoproteins/genetics , Polymorphism, Single Nucleotide , Quantitative Trait Loci , Ubiquitin-Conjugating Enzymes/genetics
5.
Cerebellum ; 20(1): 41-53, 2021 Feb.
Article En | MEDLINE | ID: mdl-32789747

Spinocerebellar ataxia type 3 (SCA3) is the second-most common CAG repeat disease, caused by a glutamine-encoding expansion in the ATXN3 protein. SCA3 is characterized by spinocerebellar degeneration leading to progressive motor incoordination and early death. Previous studies suggest that potassium channel dysfunction underlies early abnormalities in cerebellar cortical Purkinje neuron firing in SCA3. However, cerebellar cortical degeneration is often modest both in the human disease and mouse models of SCA3, raising uncertainty about the role of cerebellar dysfunction in SCA3. Here, we address this question by investigating Purkinje neuron excitability in SCA3. In early-stage SCA3 mice, we confirm a previously identified increase in excitability of cerebellar Purkinje neurons and associate this excitability with reduced transcripts of two voltage-gated potassium (KV) channels, Kcna6 and Kcnc3, as well as motor impairment. Intracerebroventricular delivery of antisense oligonucleotides (ASO) to reduce mutant ATXN3 restores normal excitability to SCA3 Purkinje neurons and rescues transcript levels of Kcna6 and Kcnc3. Interestingly, while an even broader range of KV channel transcripts shows reduced levels in late-stage SCA3 mice, cerebellar Purkinje neuron physiology was not further altered despite continued worsening of motor impairment. These results suggest the progressive motor phenotype observed in SCA3 may not reflect ongoing changes in the cerebellar cortex but instead dysfunction of other neuronal structures within and beyond the cerebellum. Nevertheless, the early rescue of both KV channel expression and neuronal excitability by ASO treatment suggests that cerebellar cortical dysfunction contributes meaningfully to motor dysfunction in SCA3.


Ataxin-3/genetics , Machado-Joseph Disease/drug therapy , Machado-Joseph Disease/genetics , Oligonucleotides, Antisense/therapeutic use , Purkinje Cells/pathology , Repressor Proteins/genetics , Animals , Behavior, Animal , Humans , Injections, Intraventricular , Kv1.6 Potassium Channel/drug effects , Kv1.6 Potassium Channel/genetics , Machado-Joseph Disease/psychology , Mice , Mice, Transgenic , Patch-Clamp Techniques , Phenotype , Potassium Channels, Voltage-Gated/drug effects , Shaw Potassium Channels/drug effects , Shaw Potassium Channels/genetics , Treatment Outcome
6.
Mar Drugs ; 18(8)2020 Aug 13.
Article En | MEDLINE | ID: mdl-32823677

Recently, Conorfamide-Sr3 (CNF-Sr3) was isolated from the venom of Conus spurius and was demonstrated to have an inhibitory concentration-dependent effect on the Shaker K+ channel. The voltage-gated potassium channels play critical functions on cellular signaling, from the regeneration of action potentials in neurons to the regulation of insulin secretion in pancreatic cells, among others. In mammals, there are at least 40 genes encoding voltage-gated K+ channels and the process of expression of some of them may include alternative splicing. Given the enormous variety of these channels and the proven use of conotoxins as tools to distinguish different ligand- and voltage-gated ion channels, in this work, we explored the possible effect of CNF-Sr3 on four human voltage-gated K+ channel subtypes homologous to the Shaker channel. CNF-Sr3 showed a 10 times higher affinity for the Kv1.6 subtype with respect to Kv1.3 (IC50 = 2.7 and 24 µM, respectively) and no significant effect on Kv1.4 and Kv1.5 at 10 µM. Thus, CNF-Sr3 might become a novel molecular probe to study diverse aspects of human Kv1.3 and Kv1.6 channels.


Mollusk Venoms/pharmacology , Potassium Channel Blockers/pharmacology , Shaker Superfamily of Potassium Channels/antagonists & inhibitors , Animals , Conus Snail , Ion Channel Gating , Kv1.3 Potassium Channel/antagonists & inhibitors , Kv1.3 Potassium Channel/genetics , Kv1.3 Potassium Channel/metabolism , Kv1.4 Potassium Channel/antagonists & inhibitors , Kv1.4 Potassium Channel/genetics , Kv1.4 Potassium Channel/metabolism , Kv1.5 Potassium Channel/antagonists & inhibitors , Kv1.5 Potassium Channel/genetics , Kv1.5 Potassium Channel/metabolism , Kv1.6 Potassium Channel/antagonists & inhibitors , Kv1.6 Potassium Channel/genetics , Kv1.6 Potassium Channel/metabolism , Membrane Potentials , Oocytes , Shaker Superfamily of Potassium Channels/genetics , Shaker Superfamily of Potassium Channels/metabolism , Xenopus laevis
7.
J Cell Physiol ; 233(5): 4317-4326, 2018 05.
Article En | MEDLINE | ID: mdl-29139549

Supplementation of 100% oxygen is a very common intervention in intensive care units (ICU) and critical care centers for patients with dysfunctional lung and lung disorders. Although there is advantage in delivering sufficient levels of oxygen, hyperoxia is reported to be directly associated with increasing in-hospital deaths. Our previous studies reported ventricular and electrical remodeling in hyperoxia treated mouse hearts, and in this article, for the first time, we are investigating the effects of hyperoxia on atrial electrophysiology using whole-cell patch-clamp electrophysiology experiments along with assessment of Kv1.5, Kv4.2, and KChIP2 transcripts and protein profiles using real-time quantitative RT-PCR and Western blotting. Our data showed that induction of hyperoxia for 3 days in mice showed larger outward potassium currents with shorter action potential durations (APD). This increase in current densities is due to significant increase in ultrarapid delayed rectifier outward K+ currents (IKur ) and rapidly activating, rapidly inactivating transient outward K+ current (Ito ) densities. We also observed a significant increase in both transcripts and protein levels of Kv1.5 and KChIP2 in hyperoxia treated atrial cardiomyocytes, whereas no significant change was observed in Kv4.2 transcripts or protein. The data presented here further support our previous findings that hyperoxia induces not only ventricular remodeling, but also atrial electrical remodeling.


Kv Channel-Interacting Proteins/genetics , Kv1.6 Potassium Channel/genetics , Lung Diseases/therapy , Oxygen/adverse effects , Shal Potassium Channels/genetics , Action Potentials/drug effects , Animals , Gene Expression Regulation , Heart Atria/physiopathology , Hospital Mortality , Humans , Hyperoxia/etiology , Hyperoxia/physiopathology , Intensive Care Units , Lung/metabolism , Lung/physiopathology , Lung Diseases/complications , Lung Diseases/mortality , Lung Diseases/physiopathology , Mice , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Patch-Clamp Techniques , Potassium/metabolism
8.
BMC Genomics ; 13: 635, 2012 Nov 17.
Article En | MEDLINE | ID: mdl-23157480

BACKGROUND: LEF1/TCF transcription factors and their activator ß-catenin are effectors of the canonical Wnt pathway. Although Wnt/ß-catenin signaling has been implicated in neurodegenerative and psychiatric disorders, its possible role in the adult brain remains enigmatic. To address this issue, we sought to identify the genetic program activated by ß-catenin in neurons. We recently showed that ß-catenin accumulates specifically in thalamic neurons where it activates Cacna1g gene expression. In the present study, we combined bioinformatics and experimental approaches to find new ß-catenin targets in the adult thalamus. RESULTS: We first selected the genes with at least two conserved LEF/TCF motifs within the regulatory elements. The resulting list of 428 putative LEF1/TCF targets was significantly enriched in known Wnt targets, validating our approach. Functional annotation of the presumed targets also revealed a group of 41 genes, heretofore not associated with Wnt pathway activity, that encode proteins involved in neuronal signal transmission. Using custom polymerase chain reaction arrays, we profiled the expression of these genes in the rat forebrain. We found that nine of the analyzed genes were highly expressed in the thalamus compared with the cortex and hippocampus. Removal of nuclear ß-catenin from thalamic neurons in vitro by introducing its negative regulator Axin2 reduced the expression of six of the nine genes. Immunoprecipitation of chromatin from the brain tissues confirmed the interaction between ß-catenin and some of the predicted LEF1/TCF motifs. The results of these experiments validated four genes as authentic and direct targets of ß-catenin: Gabra3 for the receptor of GABA neurotransmitter, Calb2 for the Ca(2+)-binding protein calretinin, and the Cacna1g and Kcna6 genes for voltage-gated ion channels. Two other genes from the latter cluster, Cacna2d2 and Kcnh8, appeared to be regulated by ß-catenin, although the binding of ß-catenin to the regulatory sequences of these genes could not be confirmed. CONCLUSIONS: In the thalamus, ß-catenin regulates the expression of a novel group of genes that encode proteins involved in neuronal excitation. This implies that the transcriptional activity of ß-catenin is necessary for the proper excitability of thalamic neurons, may influence activity in the thalamocortical circuit, and may contribute to thalamic pathologies.


Calcium Channels, T-Type/genetics , Kv1.6 Potassium Channel/genetics , Neurons/metabolism , Receptors, GABA-A/genetics , S100 Calcium Binding Protein G/genetics , Thalamus/metabolism , beta Catenin/genetics , Adaptor Proteins, Signal Transducing , Animals , Binding Sites , Calbindin 2 , Calcium Channels, T-Type/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Kv1.6 Potassium Channel/metabolism , Lymphoid Enhancer-Binding Factor 1/genetics , Lymphoid Enhancer-Binding Factor 1/metabolism , Male , Neurons/cytology , Neurotransmitter Agents , Primary Cell Culture , Promoter Regions, Genetic , Protein Binding , Rats , Receptors, GABA-A/metabolism , S100 Calcium Binding Protein G/metabolism , Signal Transduction , Thalamus/cytology , Transcriptional Activation , Wnt Proteins/genetics , Wnt Proteins/metabolism , beta Catenin/metabolism
9.
Neuromuscul Disord ; 22(6): 558-65, 2012 Jun.
Article En | MEDLINE | ID: mdl-22342001

KCNA1, KCNA2, KCNA6 and KCNQ2 are associated with peripheral nerve hyperexcitability in humans. In order to determine if these genes are also involved in Jack Russell Terriers with a similar syndrome characterized by myokymia and neuromyotonia, their predicted canine orthologs were first validated experimentally. They were found either incompletely or even incorrectly annotated, mainly due to gaps in the canine genomic sequence and insufficient transcript data. Canine KCNQ2 was found to contain 20 coding exons, of which three are not described in humans. It encodes for at least 14 different transcript variants in the frontal cortex of a single dog, of which only four are also described in humans. Mutation detection in Jack Russell Terriers diagnosed with peripheral nerve hyperexcitability revealed no pathogenetic relevant structural mutations. However, the four missense sequence variations and the 14 transcript variants of KCNQ2 will contribute to the study of the functional diversity of voltage-gated potassium channels.


Dog Diseases/genetics , KCNQ2 Potassium Channel/genetics , Kv1.1 Potassium Channel/genetics , Kv1.2 Potassium Channel/genetics , Kv1.6 Potassium Channel/genetics , Peripheral Nervous System Diseases/veterinary , Animals , Dogs , Genetic Association Studies , Isaacs Syndrome/genetics , Isaacs Syndrome/veterinary , Mutation , Myokymia/genetics , Myokymia/veterinary , Peripheral Nervous System Diseases/genetics
10.
J Neurosci ; 30(22): 7554-62, 2010 Jun 02.
Article En | MEDLINE | ID: mdl-20519529

Slo2 Na(+)-activated potassium channels are widely expressed in neurons and other cells, such as kidney, heart, and skeletal muscle. Although their important physiological roles continue to be appreciated, molecular determinants responsible for sensing intracellular Na(+) remain unknown. Here we report identification of an Na(+) regulatory site, similar to an Na(+) coordination motif described in Kir channels, localized in the RCK2 domain of Slo2.2 channels. Molecular simulations of the homology-modeled Slo2.2 RCK2 domain provided structural insights into the organization of this Na(+) coordination site. Furthermore, free energy calculations reproduced the experimentally derived monovalent cation selectivity. Our results suggest that Slo2.2 and Kir channels share a similar mechanism to coordinate Na(+). The localization of an Na(+) sensor within the RCK2 domain of Slo2.2 further supports the role of RCK (regulators of conductance of K(+)) domains of Slo channels in coupling ion sensing to channel gating.


Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Kv1.6 Potassium Channel/metabolism , Sodium/metabolism , Amino Acid Sequence , Amino Acids/genetics , Animals , Biophysical Phenomena/drug effects , Biophysical Phenomena/genetics , Biophysical Phenomena/physiology , Chlorides/pharmacology , Dose-Response Relationship, Drug , Electric Stimulation/methods , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Ion Channel Gating/drug effects , Ion Channel Gating/genetics , Kv1.6 Potassium Channel/genetics , Membrane Potentials/drug effects , Membrane Potentials/physiology , Microinjections/methods , Models, Molecular , Molecular Sequence Data , Mutation/genetics , Oocytes , Patch-Clamp Techniques , Sodium/pharmacology , Xenopus laevis
11.
Channels (Austin) ; 4(3): 179-91, 2010.
Article En | MEDLINE | ID: mdl-20372061

Membrane currents and resting potential of isolated primary mouse articular chondrocytes maintained in monolayer cell culture for 1-9 days were recorded using patch clamp methods. Quantitative RT-PCR showed that the most abundantly expressed transcript of voltage-gated K(+) channels was for K(V)1.6, and immunological methods confirmed the expression of K(V)1.6 α-subunit proteins. These chondrocytes expressed a large time- and potential-dependent, Ca(2+)-independent 'delayed rectifier' K(+) current. Steady-state activation was well-fit by a Boltzmann function with a threshold near -50 mV, and a half-activation potential of -34.5 mV. The current was 50% blocked by 1.48 mM tetraethylammonium, 0.66 mM 4-aminopyridine and 20.6 nM α-dendrotoxin. The current inactivated very slowly at membrane potentials in the range of the resting potential of the chondrocytes. Resting membrane potential of the chondrocytes at room temperature (19-21°C) and in 5 mM external K(+) was -46.4 ± 1.3 mV (mean ± s.e.m; n = 23), near the 'foot' of the activation curve of this K(+) current. Resting potential was depolarized by an average of 4.2 ± 0.8 mV by 25 mM TEA, which blocked about 95% of the K(+) current. At a membrane potential of -50 mV, the apparent time constant of inactivation (tau(in)) was 37.9 s, and the 'steady-state' current level was 19% of that at a holding potential of -90 mV; at -40 mV, tau(in) was 20.3 s, and 'steady-state' current was 5% of that at -90 mV. These results demonstrate that in these primary cultured, mouse articular chondrocytes steady-state activation of a voltage-gated K(+) current contributes to resting membrane potential. However, this current is also likely to have a significant physiological role in repolarizing the chondrocyte following depolarizing stimuli that might occur in conditions of membrane stretch. For example, activation of TRP('transient receptor potential') non-specific cation channels in these cells during cyclic loading and unloading of the joint cartilage, or in response to hypertonic challenge is expected to result in depolarization and Ca(2+) entry. Potassium currents are required to maintain the resting membrane potential.


Cartilage, Articular/cytology , Chondrocytes/physiology , Membrane Potentials , Potassium Channels, Voltage-Gated/physiology , Animals , Cells, Cultured , Kv1.6 Potassium Channel/genetics , Kv1.6 Potassium Channel/physiology , Mice , Patch-Clamp Techniques , Potassium/metabolism , Potassium/physiology , Potassium Channels, Voltage-Gated/genetics , RNA, Messenger/analysis
12.
Br J Pharmacol ; 150(8): 1003-10, 2007 Apr.
Article En | MEDLINE | ID: mdl-17339841

BACKGROUND AND PURPOSE: Methylamine is an endogenous aliphatic amine exhibiting anorexigenic properties in mice. The aim of this work was to show whether methylamine also modifies feeding behaviour in rats and, if so, to identify the mediator(s) responsible for such effects. EXPERIMENTAL APPROACH: Microdialysis experiments with the probe inserted in the periventricular hypothalamic nucleus were carried out in 12 h starved, freely moving rats. Collected perfusate samples following methylamine injection (i.c.v.) were analysed for nitric oxide by chemiluminescence and for dopamine and 5-hydroxytryptamine content by HPLC. Kv1.6 potassium channel expression was reduced by antisense strategy and this decrease quantified by semi-quantitative RT-PCR analysis. KEY RESULTS: Methylamine showed biphasic dose-related effects on rat feeding. At doses of 15-30 microg per rat, it was hyperphagic whereas higher doses (60-80 microg) were hypophagic. Methylamine stimulated central nitric oxide (+115% vs. basal) following hyperphagic and dopamine release (60% over basal values) at hypophagic doses, respectively. Treatment with L-N(G)-nitro-L-arginine-methyl ester (i.c.v. 2 microg 10 microl(-1)) or with alpha-methyl-p-tyrosine (i.p. 100 mg kg(-1)) before methylamine injection, reduced nitric oxide output and hyperphagia, or dopamine release and hypophagia respectively. Moreover, hypophagia and hyperphagia, as well as nitric oxide and dopamine release were significantly reduced by down-regulating brain Kv1.6 potassium channel expression. CONCLUSIONS AND IMPLICATIONS: The effects of methylamine on feeding depend on the hypothalamic release of nitric oxide and dopamine as a result of interaction at the Kv1.6 channels. The study of methylamine levels in the CNS may provide new perspectives on the physiopathology of alimentary behaviour.


Appetite Depressants/metabolism , Appetite Regulation , Dopamine/metabolism , Fasting , Hypothalamus/metabolism , Methylamines/metabolism , Nitric Oxide/metabolism , Animals , Appetite Depressants/administration & dosage , Appetite Regulation/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Hyperphagia/metabolism , Hyperphagia/physiopathology , Hypothalamus/drug effects , Injections, Intraventricular , Kv1.6 Potassium Channel/drug effects , Kv1.6 Potassium Channel/genetics , Kv1.6 Potassium Channel/metabolism , Male , Methylamines/administration & dosage , Microdialysis , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/metabolism , Rats , Rats, Wistar , Serotonin/metabolism , Time Factors , Tyrosine 3-Monooxygenase/antagonists & inhibitors , Tyrosine 3-Monooxygenase/metabolism , alpha-Methyltyrosine/pharmacology
13.
J Neurophysiol ; 96(4): 1887-901, 2006 Oct.
Article En | MEDLINE | ID: mdl-16624997

The axon initial segment plays important roles in spike initiation and invasion of axonal spikes into the soma. Among primary sensory neurons, those in the mesencephalic trigeminal nucleus (MTN) are exceptional in their ability to initiate soma spikes (S-spikes) in response to synaptic inputs, consequently displaying two kinds of S-spikes, one caused by invasion of an axonal spike arising from the sensory receptor and the other initiated by somatic inputs. We investigated where spikes are initiated in such MTN neurons and whether there are any differences between the two kinds of S-spikes. Simultaneous patch-clamp recordings from the soma and axon hillock revealed a spike-backpropagation from the spike-initiation site in the stem axon to the soma in response to 1-ms somatic current pulse, which disclosed the delayed emergence of S-spikes after the current-pulse offset. These initiated S-spikes were smaller in amplitude than S-spikes generated by stimulation of the stem axon; however, 4-AP (< or =0.5 mM) eliminated the amplitude difference. Furthermore, 4-AP dramatically shortened the delay in spike initiation without affecting the spike-backpropagation time in the stem axon, whereas it substantially prolonged the refractory period of S-spikes arising from axonal-spike invasion without significantly affecting that of presumed axonal spikes. These observations suggest that 4-AP-sensitive K(+) currents exert two opposing effects on S-spikes depending on their origins: suppression of spike initiation and facilitation of axonal-spike invasion at higher frequencies. Consistent with these findings, strong immunoreactivities for Kv1.1 and Kv1.6, among 4-AP-sensitive and low-voltage-activated Kv1 family examined, were detected in the soma but not in the stem axon of MTN neurons.


4-Aminopyridine/pharmacology , Action Potentials/physiology , Neurons, Afferent/physiology , Potassium Channel Blockers/pharmacology , Potassium Channels/physiology , Trigeminal Nuclei/physiology , Action Potentials/drug effects , Animals , Axons/physiology , Gene Expression Regulation , Kv1.1 Potassium Channel/genetics , Kv1.1 Potassium Channel/metabolism , Kv1.6 Potassium Channel/genetics , Kv1.6 Potassium Channel/metabolism , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mesencephalon/physiology , Neurons, Afferent/metabolism , Patch-Clamp Techniques , Potassium Channels/drug effects , Rats , Rats, Wistar , Trigeminal Nuclei/metabolism
...