Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 171(4): 836-848.e13, 2017 Nov 02.
Article in English | MEDLINE | ID: mdl-28988768

ABSTRACT

Adrenergic stimulation promotes lipid mobilization and oxidation in brown and beige adipocytes, where the harnessed energy is dissipated as heat in a process known as adaptive thermogenesis. The signaling cascades and energy-dissipating pathways that facilitate thermogenesis have been extensively described, yet little is known about the counterbalancing negative regulatory mechanisms. Here, we identify a two-pore-domain potassium channel, KCNK3, as a built-in rheostat negatively regulating thermogenesis. Kcnk3 is transcriptionally wired into the thermogenic program by PRDM16, a master regulator of thermogenesis. KCNK3 antagonizes norepinephrine-induced membrane depolarization by promoting potassium efflux in brown adipocytes. This limits calcium influx through voltage-dependent calcium channels and dampens adrenergic signaling, thereby attenuating lipolysis and thermogenic respiration. Adipose-specific Kcnk3 knockout mice display increased energy expenditure and are resistant to hypothermia and obesity. These findings uncover a critical K+-Ca2+-adrenergic signaling axis that acts to dampen thermogenesis, maintain tissue homeostasis, and reveal an electrophysiological regulatory mechanism of adipocyte function.


Subject(s)
Adipose Tissue/metabolism , Nerve Tissue Proteins/metabolism , Obesity/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Receptors, Adrenergic/metabolism , Signal Transduction , Thermogenesis , Adipocytes, Brown/metabolism , Adipose Tissue/pathology , Animals , Cell Separation , Cells, Cultured , Electrophysiological Phenomena , Female , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Obesity/pathology , Potassium Channels, Tandem Pore Domain/genetics
2.
Int J Mol Sci ; 23(16)2022 Aug 11.
Article in English | MEDLINE | ID: mdl-36012204

ABSTRACT

Proteins interacting with CFTR and its mutants have been intensively studied using different experimental approaches. These studies provided information on the cellular processes leading to proper protein folding, routing to the plasma membrane, recycling, activation and degradation. Recently, new approaches have been developed based on the proximity labeling of protein partners or proteins in close vicinity and their subsequent identification by mass spectrometry. In this study, we evaluated TurboID- and APEX2-based proximity labeling of WT CFTR and compared the obtained data to those reported in databases. The CFTR-WT interactome was then compared to that of two CFTR (G551D and W1282X) mutants and the structurally unrelated potassium channel KCNK3. The two proximity labeling approaches identified both known and additional CFTR protein partners, including multiple SLC transporters. Proximity labeling approaches provided a more comprehensive picture of the CFTR interactome and improved our knowledge of the CFTR environment.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator , Protein Folding , Cell Membrane/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Mass Spectrometry , Mutation
3.
Exp Brain Res ; 239(12): 3601-3613, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34591125

ABSTRACT

The present study aimed to explore the potential mechanism of the effect of hyperbaric oxygenation (HBO) preconditioning on cerebral ischemia and reperfusion injury (CIRI). GSE23160 dataset was used to identify differentially expressed genes (DEGs) from striatum between the middle cerebral artery occlusion (MCAO)/reperfusion and sham rats. The gene clusters with continuous increase and decrease were identified by soft clustering analysis in Mfuzz, and functional enrichment analysis of these genes was performed using clusterProfiler package. The intersection set of the genes with significantly altered expression at post-reperfusion 2, 8, and 24 h were screened in comparison to 0 h (sham group), and the expression of these genes was detected in the MCAO/reperfusion model and HBO preconditioning groups by real-time PCR (RT-PCR) and western blotting. A total of 41 upregulated DEGs, and 7 downregulated DEGs were detected, among which the expression of Gpr84 and Ggta1 was significantly upregulated at each reperfusion phase as compared to the sham group, while the expression of Kcnk3 was significantly downregulated except in the postreperfusion 8 h in the striatum group. RT-PCR and western blotting analyses showed that the expression of Ggta1, Gpr84, and Kcnk3 genes between the MCAO/reperfusion and sham rats were consistent with the bioinformatics analysis. In addition, the HBO preconditioning reduced the expression of Ggta1 and Gpr84 and increased the expression of Kcnk3 in MCAO/reperfusion rats. Kcnk3, Ggta1, and Gpr84 may play a major role in HBO-mediated protection of the brain against CIRI.


Subject(s)
Brain Ischemia , Hyperbaric Oxygenation , Reperfusion Injury , Animals , Infarction, Middle Cerebral Artery , Rats , Rats, Sprague-Dawley , Reperfusion Injury/genetics , Reperfusion Injury/prevention & control
4.
Genomics ; 112(3): 2213-2222, 2020 05.
Article in English | MEDLINE | ID: mdl-31881264

ABSTRACT

As one important member of the two-pore-domain potassium channel (K2P) family, potassium channel subfamily K member 3 (KCNK3) has been reported for thermogenesis regulation, energy homeostasis, membrane potential conduction, and pulmonary hypertension in mammals. However, its roles in fishes are far less examined and published. In the present study, we identified two kcnk3 genes (kcnk3a and kcnk3b) in an euryhaline fish, Nile tilapia (Oreochromis niloticus), by molecular cloning, genomic survey and laboratory experiments to investigate their potential roles for osmoregulation. We obtained full-length coding sequences of the kcnk3a and kcnk3b genes (1209 and 1173 bp), which encode 402 and 390 amino acids, respectively. Subsequent multiple sequence alignments, putative 3D-structure model prediction, genomic survey and phylogenetic analysis confirmed that two kcnk3 paralogs are widely presented in fish genomes. Interestingly, a DNA fragment inversion of a kcnk3a cluster was found in Cypriniforme in comparison with other fishes. Quantitative real-time PCRs demonstrated that both the tilapia kcnk3 genes were detected in all the examined tissues with a similar distribution pattern, and the highest transcriptions were observed in the heart. Meanwhile, both kcnk3 genes in the gill were proved to have a similar transcriptional change pattern in response to various salinity of seawater, implying that they might be involved in osmoregulation. Furthermore, three predicted transcription factors (arid3a, arid3b, and arid5a) of both kcnk3 genes also showed a similar pattern as their target genes in response to the various salinity, suggesting their potential positive regulatory roles. In summary, we for the first time characterized the two kcnk3 genes in Nile tilapia, and demonstrated their potential involvement in osmoregulation for this economically important fish.


Subject(s)
Fish Proteins/genetics , Nerve Tissue Proteins/genetics , Potassium Channels, Tandem Pore Domain/genetics , Tilapia/genetics , Animals , Cloning, Molecular , Fish Proteins/chemistry , Fish Proteins/classification , Fish Proteins/metabolism , Genome , Models, Molecular , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/classification , Nerve Tissue Proteins/metabolism , Phylogeny , Potassium Channels, Tandem Pore Domain/chemistry , Potassium Channels, Tandem Pore Domain/classification , Potassium Channels, Tandem Pore Domain/metabolism , Protein Conformation , Salinity , Seawater , Sequence Alignment , Sequence Analysis, Protein , Tilapia/metabolism , Tissue Distribution , Transcription Factors/genetics , Transcription, Genetic
5.
Int J Mol Sci ; 22(9)2021 May 09.
Article in English | MEDLINE | ID: mdl-34065088

ABSTRACT

Loss of function KCNK3 mutation is one of the gene variants driving hereditary pulmonary arterial hypertension (PAH). KCNK3 is expressed in several cell and tissue types on both membrane and endoplasmic reticulum and potentially plays a role in multiple pathological process associated with PAH. However, the role of various stressors driving the susceptibility of KCNK3 mutation to PAH is unknown. Hence, we exposed kcnk3fl/fl animals to hypoxia, metabolic diet and low dose lipopolysaccharide (LPS) and performed molecular characterization of their tissue. We also used tissue samples from KCNK3 patients (skin fibroblast derived inducible pluripotent stem cells, blood, lungs, peripheral blood mononuclear cells) and performed microarray, immunohistochemistry (IHC) and mass cytometry time of flight (CyTOF) experiments. Although a hypoxic insult did not alter vascular tone in kcnk3fl/fl mice, RNASeq study of these lungs implied that inflammatory and metabolic factors were altered, and the follow-up diet study demonstrated a dysregulation of bone marrow cells in kcnk3fl/fl mice. Finally, a low dose LPS study clearly showed that inflammation could be a possible second hit driving PAH in kcnk3fl/fl mice. Multiplex, IHC and CyTOF immunophenotyping studies on human samples confirmed the mouse data and strongly indicated that cell mediated, and innate immune responses may drive PAH susceptibility in these patients. In conclusion, loss of function KCNK3 mutation alters various physiological processes from vascular tone to metabolic diet through inflammation. Our data suggests that altered circulating immune cells may drive PAH susceptibility in patients with KCNK3 mutation.


Subject(s)
Immunomodulation/genetics , Mutation , Nerve Tissue Proteins/genetics , Potassium Channels, Tandem Pore Domain/genetics , Pulmonary Arterial Hypertension/genetics , Pulmonary Arterial Hypertension/immunology , Animals , Biomarkers , Case-Control Studies , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility , Gene Expression Profiling , Genetic Predisposition to Disease , Humans , Hypoxia/genetics , Hypoxia/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Mice , Mice, Knockout , Models, Biological , Monocytes/immunology , Monocytes/metabolism , Nerve Tissue Proteins/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Pulmonary Arterial Hypertension/complications , Pulmonary Arterial Hypertension/physiopathology , Transcriptome
6.
Respir Res ; 21(1): 186, 2020 Jul 16.
Article in English | MEDLINE | ID: mdl-32678044

ABSTRACT

BACKGROUND: The pathogenesis of pulmonary arterial hypertension (PAH) involves many signalling pathways. MicroRNAs are potential candidates involved in simultaneously coordinating multiple genes under such multifactorial conditions. METHODS AND RESULTS: MiR-138-5p is overexpressed in pulmonary arterial smooth muscle cells (PASMCs) from PAH patients and in lungs from rats with monocrotaline-induced pulmonary hypertension (MCT-PH). MiR-138-5p is predicted to regulate the expression of the potassium channel KCNK3, whose loss is associated with the development and progression of PAH. We hypothesized that, in vivo, miR-138-5p inhibition would restore KCNK3 lung expression and subsequently alleviate PAH. Nebulization-based delivery of anti-miR-138-5p to rats with established MCT-PH significantly reduced the right ventricular systolic pressure and significantly improved the pulmonary arterial acceleration time (PAAT). These haemodynamic improvements were related to decrease pulmonary vascular remodelling, lung inflammation and pulmonary vascular cell proliferation in situ. In vivo inhibition of miR-138-5p restored KCNK3 mRNA expression and SLC45A3 protein expression in the lungs. CONCLUSIONS: We confirmed that in vivo inhibition of miR-138-5p reduces the development of PH in experimental MCT-PH. The possible curative mechanisms involve at least the normalization of lung KCNK3 as well as SLC45A3 expression.


Subject(s)
Antagomirs/administration & dosage , Arterial Pressure , MicroRNAs/antagonists & inhibitors , Monosaccharide Transport Proteins/metabolism , Nerve Tissue Proteins/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Pulmonary Arterial Hypertension/prevention & control , Pulmonary Artery/metabolism , Administration, Inhalation , Animals , Antagomirs/genetics , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation , Humans , Male , MicroRNAs/genetics , MicroRNAs/metabolism , Monocrotaline , Monosaccharide Transport Proteins/genetics , Nerve Tissue Proteins/genetics , Potassium Channels, Tandem Pore Domain/genetics , Pulmonary Arterial Hypertension/genetics , Pulmonary Arterial Hypertension/metabolism , Pulmonary Arterial Hypertension/physiopathology , Pulmonary Artery/physiopathology , Rats, Wistar , Signal Transduction , Vascular Remodeling
7.
Gen Comp Endocrinol ; 296: 113546, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32653428

ABSTRACT

KCNK3 is a two-pore-domain (K2P) potassium channel involved in maintaining ion homeostasis, mediating thermogenesis, controlling breath and modulating electrical membrane potential. Although the functions of this channel have been widely described in mammals, its roles in fishes are still rarely known. Here, we identified two kcnk3 genes from the euryhaline rabbitfish (Siganus canaliculatus), and their roles related to fatty acids metabolism and osmoregulation were investigated. The open reading frames of kcnk3a and kcnk3b were 1203 and 1176 bp in length, encoding 400 and 391 amino acids respectively. Multiple sequences alignment and phylogenetic analysis revealed that the two isotypes of kcnk3 were extensively presented in fishes. Quantitative real-time PCRs indicated that both genes were widely distributed in examined tissues but showed different patterns. kcnk3a primary distributed in adipose, eye, heart, and spleen tissues, while kcnk3b was mainly detectable in heart, kidney, muscle and spleen tissues. In vivo experiments showed that fish fed diets with fish oil as dietary lipid (rich in long chain polyunsaturated fatty acids, LC-PUFA) induced higher mRNA expression levels of kcnk3 genes in comparison with fish fed with plant oil diet at two different salinity environments (32 and 15‰). Meanwhile, the expression levels of kcnk3 genes were higher in seawater (32‰) than that in brackish water (15‰) when fishes were fed with both types of feeds. In vitro experiments with rabbitfish hepatocytes showed that LC-PUFA significantly improved hepatic kcnk3a expression level compared with treatment of linolenic acid. These results suggest that two kcnk3 genes are widely existed and they might be functionally related to fatty acids metabolism and osmoregulation in the rabbitfish.


Subject(s)
Fatty Acids/metabolism , Fishes/genetics , Osmoregulation , Potassium Channels/genetics , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Cloning, Molecular , DNA, Complementary/genetics , Fishes/metabolism , Gene Expression Regulation/drug effects , Hepatocytes/metabolism , Lipid Metabolism/genetics , Liver/metabolism , Phylogeny , Potassium Channels/chemistry , Potassium Channels/metabolism , Salinity , Tissue Distribution
8.
J Physiol ; 597(4): 1087-1101, 2019 02.
Article in English | MEDLINE | ID: mdl-30365877

ABSTRACT

KEY POINTS: The TASK-1 channel gene (KCNK3) has been identified as a possible disease-causing gene in heritable pulmonary arterial hypertension (PAH). In the present study, we show that novel mutated TASK-1 channels, seen in PAH patients, have a substantially reduced current compared to wild-type TASK-1 channels. These mutated TASK-1 channels are located at the plasma membrane to the same degree as wild-type TASK-1 channels. ONO-RS-082 and alkaline pH 8.4 both activate TASK-1 channels but do not recover current through mutant TASK-1 channels. We show that the guanylate cyclase activator, riociguat, a novel treatment for PAH, enhances current through TASK-1 channels but does not recover current through mutant TASK-1 channels. ABSTRACT: Pulmonary arterial hypertension (PAH) affects ∼15-50 people per million. KCNK3, the gene that encodes the two pore domain potassium channel TASK-1 (K2P3.1), has been identified as a possible disease-causing gene in heritable PAH. Recently, two new mutations have been identified in KCNK3 in PAH patients: G106R and L214R. The present study aimed to characterize the functional properties and regulation of wild-type (WT) and mutated TASK-1 channels and determine how these might contribute to PAH and its treatment. Currents through WT and mutated human TASK-1 channels transiently expressed in tsA201 cells were measured using whole-cell patch clamp electrophysiology. Localization of fluorescence-tagged channels was visualized using confocal microscopy and quantified with in-cell and on-cell westerns. G106R or L214R mutated channels were located at the plasma membrane to the same degree as WT channels; however, their current was markedly reduced compared to WT TASK-1 channels. Functional current through these mutated channels could not be restored using activators of WT TASK-1 channels (pH 8.4, ONO-RS-082). The guanylate cyclase activator, riociguat, enhanced current through WT TASK-1 channels; however, similar to the other activators investigated, riociguat did not have any effect on current through mutated TASK-1 channels. Thus, novel mutations in TASK-1 seen in PAH substantially alter the functional properties of these channels. Current through these channels could not be restored by activators of TASK-1 channels. Riociguat enhancement of current through TASK-1 channels could contribute to its therapeutic benefit in the treatment of PAH.


Subject(s)
Action Potentials , Hypertension, Pulmonary/genetics , Mutation, Missense , Nerve Tissue Proteins/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Membrane/physiology , Enzyme Activators/pharmacology , HEK293 Cells , Humans , Nerve Tissue Proteins/genetics , Potassium Channels, Tandem Pore Domain/genetics , Pyrazoles/pharmacology , Pyrimidines/pharmacology
9.
Cell Physiol Biochem ; 52(5): 1223-1235, 2019.
Article in English | MEDLINE | ID: mdl-31001961

ABSTRACT

BACKGROUND/AIMS: The two-pore-domain potassium channel TASK-1 regulates atrial action potential duration. Due to the atrium-specific expression of TASK-1 in the human heart and the functional upregulation of TASK-1 currents in atrial fibrillation (AF), TASK-1 represents a promising target for the treatment of AF. Therefore, detailed knowledge of the molecular determinants of TASK-1 inhibition may help to identify new drugs for the future therapy of AF. In the current study, the molecular determinants of TASK-1 inhibition by the potent and antiarrhythmic compound A293 (AVE1231) were studied in detail. METHODS: Alanine-scanning mutagenesis together with two-electrode voltage-clamp recordings were combined with in silico docking experiments. RESULTS: Here, we have identified Q126 located in the M2 segment together with L239 and N240 of the M4 segment as amino acids essential for the A293-mediated inhibition of TASK-1. These data indicate a binding site which is different to that of A1899 for which also residues of the pore signature sequence and the late M4 segments are essential. Using in silico docking experiments, we propose a binding site at the lower end of the cytosolic pore, located at the entry to lateral side fenestrations of TASK-1. Strikingly, TASK-1 inhibition by the low affinity antiarrhythmic TASK-1 blockers propafenone, amiodarone and carvedilol was also strongly diminished by mutations at this novel binding site. CONCLUSION: We have identified the A293 binding site in the central cavity of TASK-1 and propose that this site might represent a conserved site of action for many low affinity antiarrhythmic TASK-1 blockers.


Subject(s)
Anti-Arrhythmia Agents/chemistry , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/chemistry , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Potassium Channels, Tandem Pore Domain/chemistry , Amino Acid Substitution , Animals , Binding Sites , Humans , Mutagenesis, Site-Directed , Mutation, Missense , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Potassium Channels, Tandem Pore Domain/genetics , Potassium Channels, Tandem Pore Domain/metabolism , Xenopus laevis
10.
Gen Comp Endocrinol ; 281: 49-57, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31121162

ABSTRACT

Potassium channel subfamily K member 3 (KCNK3) has been reported to play important roles in membrane potential conduction, pulmonary hypertension and thermogenesis regulation in mammals. However, its roles remain largely unknown and scarce reports were seen in fish. In the present study, we for the first time identified two kcnk3 genes (kcnk3a and kcnk3b) from the carnivorous Northern snakehead (Channa argus) by molecular cloning and a genomic survey. Subsequently, their transcription changes in response to different feeding status were investigated. Full-length coding sequences of the kcnk3a and kcnk3b genes are 1203 and 1176 bp, encoding 400 and 391 amino acids, respectively. Multiple alignments, 3D-structure prediction and phylogenetic analysis further suggested that these kcnk3 genes may be highly conserved in vertebrates. Tissue distribution analysis by real-time PCR demonstrated that both the snakehead kcnk3s were widely transcribed in majority of the examined tissues but with different distribution patterns. In a short-term (24-h) fasting experiment, we observed that brain kcnk3a and kcnk3b genes showed totally opposite transcription patterns. In a long-term (2-week) fasting and refeeding experiment, we also observed differential change patterns for the brain kcnk3 genes. In summary, our findings suggest that the two kcnk3 genes are close while present different transcription responses to fasting and refeeding. They therefore can be potentially selected as novel target genes for improvement of production and quality of this economically important fish.


Subject(s)
Fasting/physiology , Feeding Behavior , Fishes/genetics , Potassium Channels, Tandem Pore Domain/genetics , Transcription, Genetic , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA, Complementary/genetics , Genome , Phylogeny , Potassium Channels, Tandem Pore Domain/chemistry , Potassium Channels, Tandem Pore Domain/metabolism , Synteny/genetics , Tissue Distribution , Zebrafish/genetics
11.
J Pathol ; 241(2): 273-280, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27770446

ABSTRACT

Group 1 pulmonary hypertension or pulmonary arterial hypertension (PAH) is a rare disease characterized by proliferation and occlusion of small pulmonary arterioles, leading to progressive elevation of pulmonary artery pressure and pulmonary vascular resistance, and right ventricular failure. Historically, it has been associated with a high mortality rate, although, over the last decade, treatment has improved survival. PAH includes idiopathic PAH (IPAH), heritable PAH (HPAH), and PAH associated with certain medical conditions. The aetiology of PAH is heterogeneous, and genetics play an important role in some cases. Mutations in BMPR2, encoding bone morphogenetic protein receptor 2, a member of the transforming growth factor-ß superfamily of receptors, have been identified in 70% of cases of HPAH, and in 10-40% of cases of IPAH. Other genetic causes of PAH include mutations in the genes encoding activin receptor-like type 1, endoglin, SMAD9, caveolin 1, and potassium two-pore-domain channel subfamily K member 3. Mutations in the gene encoding T-box 4 have been identified in 10-30% of paediatric PAH patients, but rarely in adults with PAH. PAH in children is much more heterogeneous than in adults, and can be associated with several genetic syndromes, congenital heart disease, pulmonary disease, and vascular disease. In addition to rare mutations as a monogenic cause of HPAH, common variants in the gene encoding cerebellin 2 increase the risk of PAH by approximately two-fold. A PAH panel of genes is available for clinical testing, and should be considered for use in clinical management, especially for patients with a family history of PAH. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Genetic Predisposition to Disease , Heart Defects, Congenital/genetics , Hypertension, Pulmonary/genetics , Vascular Resistance/genetics , Bone Morphogenetic Protein Receptors, Type II/metabolism , Humans , Mutation/genetics
12.
Int J Mol Sci ; 19(10)2018 Oct 14.
Article in English | MEDLINE | ID: mdl-30322215

ABSTRACT

Pulmonary arterial hypertension (PAH) is a multifactorial and severe disease without curative therapies. PAH pathobiology involves altered pulmonary arterial tone, endothelial dysfunction, distal pulmonary vessel remodeling, and inflammation, which could all depend on ion channel activities (K⁺, Ca2+, Na⁺ and Cl-). This review focuses on ion channels in the pulmonary vasculature and discusses their pathophysiological contribution to PAH as well as their therapeutic potential in PAH.


Subject(s)
Hypertension, Pulmonary/metabolism , Ion Channels/metabolism , Animals , Antihypertensive Agents/pharmacology , Antihypertensive Agents/therapeutic use , Gene Expression Regulation/drug effects , Humans , Hypertension, Pulmonary/drug therapy
13.
Clin Genet ; 91(3): 453-457, 2017 03.
Article in English | MEDLINE | ID: mdl-27649371

ABSTRACT

Pulmonary arterial hypertension (PAH) is a rare devastating disease characterized by a high genetic heterogeneity with several related genes recently described, including BMPR2,TBX4 and KCNK3. The association between KCNK3 and PAH has been recently identified, but the prognosis and phenotype associated with these mutations have been poorly described. We studied a series of 136 idiopathic and hereditary PAH Spanish patients for BMPR2, TBX4 and KCNK3 mutations. We report the results of KCNK3 in which we were able to describe two new mutations (p.Gly106Arg and p.Leu214Arg) in three patients. The first one was found in a patient belonging to a consanguineous Romani family, who carried a homozygous mutation in KCNK3 and developed a severe and early form of the disease. To the best of our knowledge, this is the first time that a homozygous mutation in KCNK3 is reported in a PAH patient. The second one was found in a patient who presented at the young adult age a severe form of the disease. The present report supports the contribution of KCNK3 mutations to the genetic etiology of PAH and strongly suggests that mutations in KCNK3 follow incomplete dominance with worsening of the clinical features in homozygous patients.


Subject(s)
Familial Primary Pulmonary Hypertension/genetics , Genetic Predisposition to Disease , Mutation , Nerve Tissue Proteins/genetics , Potassium Channels, Tandem Pore Domain/genetics , Adult , Child , Child, Preschool , Familial Primary Pulmonary Hypertension/physiopathology , Female , Homozygote , Humans , Male , Pedigree , Phenotype
14.
FASEB J ; 30(2): 909-22, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26527067

ABSTRACT

Brown adipose tissue (BAT) is essential for adaptive thermogenesis and dissipation of caloric excess through the activity of uncoupling protein (UCP)-1. BAT in humans is of great interest for the treatment of obesity and related diseases. In this study, the expression of Twik-related acid-sensitive K(+) channel (TASK)-1 [a pH-sensitive potassium channel encoded by the potassium channel, 2-pore domain, subfamily K, member 3 (Kcnk3) gene] correlated highly with Ucp1 expression in obese and cold-exposed mice. In addition, Task1-null mice, compared with their controls, became overweight, mainly because of an increase in white adipose tissue mass and BAT whitening. Task1(-/-)-mouse-derived brown adipocytes, compared with wild-type mouse-derived brown adipocytes, displayed an impaired ß3-adrenergic receptor response that was characterized by a decrease in oxygen consumption, Ucp1 expression, and lipolysis. This phenotype was thought to be caused by an exacerbation of mineralocorticoid receptor (MR) signaling, given that it was mimicked by corticoids and reversed by an MR inhibitor. We concluded that the K(+) channel TASK1 controls the thermogenic activity in brown adipocytes through modulation of ß-adrenergic receptor signaling.


Subject(s)
Adipocytes, Brown/metabolism , Adipose Tissue, Brown/metabolism , Nerve Tissue Proteins/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Receptors, Adrenergic, beta-3/metabolism , Receptors, Mineralocorticoid/metabolism , Signal Transduction/physiology , Adipocytes, Brown/cytology , Adipose Tissue, Brown/cytology , Animals , Female , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Oxygen Consumption/physiology , Potassium Channels, Tandem Pore Domain/genetics , Receptors, Mineralocorticoid/genetics , Thermogenesis/physiology
15.
Hum Mutat ; 36(12): 1113-27, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26387786

ABSTRACT

Pulmonary arterial hypertension (PAH) is an often fatal disorder resulting from several causes including heterogeneous genetic defects. While mutations in the bone morphogenetic protein receptor type II (BMPR2) gene are the single most common causal factor for hereditary cases, pathogenic mutations have been observed in approximately 25% of idiopathic PAH patients without a prior family history of disease. Additional defects of the transforming growth factor beta pathway have been implicated in disease pathogenesis. Specifically, studies have confirmed activin A receptor type II-like 1 (ACVRL1), endoglin (ENG), and members of the SMAD family as contributing to PAH both with and without associated clinical phenotypes. Most recently, next-generation sequencing has identified novel, rare genetic variation implicated in the PAH disease spectrum. Of importance, several identified genetic factors converge on related pathways and provide significant insight into the development, maintenance, and pathogenetic transformation of the pulmonary vascular bed. Together, these analyses represent the largest comprehensive compilation of BMPR2 and associated genetic risk factors for PAH, comprising known and novel variation. Additionally, with the inclusion of an allelic series of locus-specific variation in BMPR2, these data provide a key resource in data interpretation and development of contemporary therapeutic and diagnostic tools.


Subject(s)
Hypertension, Pulmonary/genetics , Animals , Bone Morphogenetic Protein Receptors, Type II/chemistry , Bone Morphogenetic Protein Receptors, Type II/genetics , Bone Morphogenetic Protein Receptors, Type II/metabolism , Disease Models, Animal , Genetic Association Studies , Genetic Counseling , Genetic Predisposition to Disease , Genetic Variation , High-Throughput Nucleotide Sequencing , Humans , Hypertension, Pulmonary/diagnosis , Hypertension, Pulmonary/metabolism , Multigene Family , Mutation , Signal Transduction , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
16.
Am J Physiol Gastrointest Liver Physiol ; 308(7): G579-90, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25634809

ABSTRACT

Various colonic motor activities are thought to mediate propulsion and mixing/absorption of colonic content. The Japanese traditional medicine daikenchuto (TU-100), which is widely used for postoperative ileus in Japan, accelerates colonic emptying in healthy humans. Hydroxy-α sanshool (HAS), a readily absorbable active ingredient of TU-100 and a KCNK3/KCNK9/KCNK18 blocker as well as TRPV1/TRPA1 agonist, has been investigated for its effects on colonic motility. Motility was evaluated by intraluminal pressure and video imaging of rat proximal colons in an organ bath. Distribution of KCNKs was investigated by RT-PCR, in situ hybridization, and immunohistochemistry. Current and membrane potential were evaluated with use of recombinant KCNK3- or KCNK9-expressing Xenopus oocytes and Chinese hamster ovary cells. Defecation frequency in rats was measured. HAS dose dependently induced strong propulsive "squeezing" motility, presumably as long-distance contraction (LDC). TRPV1/TRPA1 agonists induced different motility patterns. The effect of HAS was unaltered by TRPV1/TRPA1 antagonists and desensitization. Lidocaine (a nonselective KCNK blocker) and hydroxy-ß sanshool (a geometrical isomer of HAS and KCNK3 blocker) also induced colonic motility as a rhythmic propagating ripple (RPR) and a LDC-like motion, respectively. HAS-induced "LDC," but not lidocaine-induced "RPR," was abrogated by a neuroleptic agent tetrodotoxin. KCNK3 and KCNK9 were located mainly in longitudinal smooth muscle cells and in neural cells in the myenteric plexus, respectively. Administration of HAS or TU-100 increased defecation frequency in normal and laparotomy rats. HAS may evoke strong LDC possibly via blockage of the neural KCNK9 channel in the colonic myenteric plexus.


Subject(s)
Colon/innervation , Fatty Acids, Unsaturated/pharmacology , Gastrointestinal Agents/pharmacology , Gastrointestinal Motility/drug effects , Muscle, Smooth/innervation , Myenteric Plexus/drug effects , Polyunsaturated Alkamides/pharmacology , Potassium Channel Blockers/pharmacology , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Animals , CHO Cells , Cricetulus , Defecation/drug effects , Dose-Response Relationship, Drug , In Vitro Techniques , Male , Membrane Potentials , Myenteric Plexus/metabolism , Oocytes , Panax , Plant Extracts/pharmacology , Potassium Channels, Tandem Pore Domain/genetics , Potassium Channels, Tandem Pore Domain/metabolism , Pressure , Rats, Sprague-Dawley , Time Factors , Transfection , Video Recording , Xenopus , Zanthoxylum , Zingiberaceae
17.
Bioorg Med Chem Lett ; 24(16): 3968-73, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25017033

ABSTRACT

TASK-1 is a two-pore domain potassium channel that is important to modulating cell excitability, most notably in the context of neuronal pathways. In order to leverage TASK-1 for therapeutic benefit, its physiological role needs better characterization; however, designing selective inhibitors that avoid the closely related TASK-3 channel has been challenging. In this study, a series of bis-amide derived compounds were found to demonstrate improved TASK-1 selectivity over TASK-3 compared to reported inhibitors. Optimization of a marginally selective hit led to analog 35 which displays a TASK-1 IC50=16 nM with 62-fold selectivity over TASK-3 in an orthogonal electrophysiology assay.


Subject(s)
Amides/pharmacology , Nerve Tissue Proteins/antagonists & inhibitors , Potassium Channel Blockers/pharmacology , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Amides/chemical synthesis , Amides/chemistry , Dose-Response Relationship, Drug , Humans , Molecular Structure , Nerve Tissue Proteins/metabolism , Potassium Channel Blockers/chemical synthesis , Potassium Channel Blockers/chemistry , Potassium Channels, Tandem Pore Domain/metabolism , Structure-Activity Relationship
18.
Food Chem (Oxf) ; 6: 100155, 2023 Jul 30.
Article in English | MEDLINE | ID: mdl-36582744

ABSTRACT

Chronic Non-Communicable Diseases (NCDs) have been considered a global health problem, characterized as diseases of multiple factors, which are developed throughout life, and regardless of genetics as a risk factor of important relevance, the increase in mortality attributed to the disease to environmental factors and the lifestyle one leads. Although the reactive species (ROS/RNS) are necessary for several physiological processes, their overproduction is directly related to the pathogenesis and aggravation of NCDs. In contrast, dietary polyphenols have been widely associated with minimizing oxidative stress and inflammation. In addition to their antioxidant power, polyphenols have also drawn attention for being able to modulate both gene expression and modify epigenetic alterations, suggesting an essential involvement in the prevention and/or development of some pathologies. Therefore, this review briefly explained the mechanisms in the development of some NCDs, followed by a summary of some evidence related to the interaction of polyphenols in oxidative stress, as well as the modulation of epigenetic mechanisms involved in the management of NCDs.

19.
Biomolecules ; 12(10)2022 Sep 22.
Article in English | MEDLINE | ID: mdl-36291551

ABSTRACT

Pulmonary arterial hypertension (PAH) is a devastating disease with high morbidity and mortality. Deleterious remodeling in the pulmonary arterial system leads to irreversible arterial constriction and elevated pulmonary arterial pressures, right heart failure, and eventually death. The difficulty in treating PAH stems in part from the complex nature of disease pathogenesis, with several signaling compounds known to be involved (e.g., endothelin-1, prostacyclins) which are indeed targets of PAH therapy. Over the last decade, potassium channelopathies were established as novel causes of PAH. More specifically, loss-of-function mutations in the KCNK3 gene that encodes the two-pore-domain potassium channel KCNK3 (or TASK-1) and loss-of-function mutations in the ABCC8 gene that encodes a key subunit, SUR1, of the ATP-sensitive potassium channel (KATP) were established as the first two potassium channelopathies in human cohorts with pulmonary arterial hypertension. Moreover, voltage-gated potassium channels (Kv) represent a third family of potassium channels with genetic changes observed in association with PAH. While other ion channel genes have since been reported in association with PAH, this review focuses on KCNK3, KATP, and Kv potassium channels as promising therapeutic targets in PAH, with recent experimental pharmacologic discoveries significantly advancing the field.


Subject(s)
Channelopathies , Hypertension, Pulmonary , Potassium Channels, Tandem Pore Domain , Potassium Channels, Voltage-Gated , Pulmonary Arterial Hypertension , Humans , Potassium Channels, Tandem Pore Domain/genetics , Channelopathies/drug therapy , Channelopathies/genetics , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/pathology , Endothelin-1 , Nerve Tissue Proteins/metabolism , Familial Primary Pulmonary Hypertension/genetics , Prostaglandins I , Potassium , KATP Channels/genetics
20.
J Am Heart Assoc ; 11(7): e023472, 2022 04 05.
Article in English | MEDLINE | ID: mdl-35301863

ABSTRACT

Background Atrial fibrillation (AF) is the most common sustained cardiac arrhythmia. However, underlying molecular mechanisms are insufficiently understood. Previous studies suggested that microRNA (miRNA) dependent gene regulation plays an important role in the initiation and maintenance of AF. The 2-pore-domain potassium channel TASK-1 (tandem of P domains in a weak inward rectifying K+ channel-related acid sensitive K+ channel 1) is an atrial-specific ion channel that is upregulated in AF. Inhibition of TASK-1 current prolongs the atrial action potential duration to similar levels as in patients with sinus rhythm. Here, we hypothesize that miRNAs might be responsible for the regulation of KCNK3 that encodes for TASK-1. Methods and Results We selected miRNAs potentially regulating KCNK3 and studied their expression in atrial tissue samples obtained from patients with sinus rhythm, paroxysmal AF, or permanent/chronic AF. MiRNAs differentially expressed in AF were further investigated for their ability to regulate KCNK3 mRNA and TASK-1 protein expression in human induced pluripotent stem cells, transfected with miRNA mimics or inhibitors. Thereby, we observed that miR-34a increases TASK-1 expression and current and further decreases the resting membrane potential of Xenopus laevis oocytes, heterologously expressing hTASK-1. Finally, we investigated associations between miRNA expression in atrial tissues and clinical parameters of our patient cohort. A cluster containing AF stage, left ventricular end-diastolic diameter, left ventricular end-systolic diameter, left atrial diameter, atrial COL1A2 (collagen alpha-2(I) chain), and TASK-1 protein level was associated with increased expression of miR-25, miR-21, miR-34a, miR-23a, miR-124, miR-1, and miR-29b as well as decreased expression of miR-9 and miR-485. Conclusions These results suggest an important pathophysiological involvement of miRNAs in the regulation of atrial expression of the TASK-1 potassium channel in patients with atrial cardiomyopathy.


Subject(s)
Atrial Fibrillation , Induced Pluripotent Stem Cells , MicroRNAs , Nerve Tissue Proteins , Potassium Channels, Tandem Pore Domain , Dilatation , Heart Atria , Humans , Induced Pluripotent Stem Cells/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Nerve Tissue Proteins/metabolism , Potassium Channels, Tandem Pore Domain/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL