Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2023 Nov 02.
Article in English | MEDLINE | ID: mdl-37693611

ABSTRACT

The polygenic contribution to heart development and function along the health-disease continuum remains unresolved. To gain insight into the genetic basis of quantitative cardiac phenotypes, we utilize highly inbred Japanese rice fish models, Oryzias latipes, and Oryzias sakaizumii. Employing automated quantification of embryonic heart rates as core metric, we profiled phenotype variability across five inbred strains. We observed maximal phenotypic contrast between individuals of the HO5 and the HdrR strain. HO5 showed elevated heart rates associated with embryonic ventricular hypoplasia and impaired adult cardiac function. This contrast served as the basis for genome-wide mapping. In a segregation population of 1192 HO5 x HdrR F2 embryos, we mapped 59 loci (173 genes) associated with heart rate. Experimental validation of the top 12 candidate genes in loss-of-function models revealed their causal and distinct impact on heart rate, development, ventricle size, and arrhythmia. Our study uncovers new diagnostic and therapeutic targets for developmental and electrophysiological cardiac diseases and provides a novel scalable approach to investigate the intricate genetic architecture of the vertebrate heart.

2.
Dis Model Mech ; 16(8)2023 08 01.
Article in English | MEDLINE | ID: mdl-37584388

ABSTRACT

High-throughput DNA sequencing studies increasingly associate DNA variants with congenital heart disease (CHD). However, functional modeling is a crucial prerequisite for translating genomic data into clinical care. We used CRISPR-Cas9-mediated targeting of 12 candidate genes in the vertebrate model medaka (Oryzias latipes), five of which displayed a novel cardiovascular phenotype spectrum in F0 (crispants): mapre2, smg7, cdc42bpab, ankrd11 and myrf, encoding a transcription factor recently linked to cardiac-urogenital syndrome. Our myrf mutant line showed particularly prominent embryonic cardiac defects recapitulating phenotypes of pediatric patients, including hypoplastic ventricle. Mimicking human mutations, we edited three sites to generate specific myrf single-nucleotide variants via cytosine and adenine base editors. The Glu749Lys missense mutation in the conserved intramolecular chaperon autocleavage domain fully recapitulated the characteristic myrf mutant phenotype with high penetrance, underlining the crucial function of this protein domain. The efficiency and scalability of base editing to model specific point mutations accelerate gene validation studies and the generation of human-relevant disease models.


Subject(s)
Gene Editing , Heart Defects, Congenital , Humans , Child , Mutation/genetics , Point Mutation , Transcription Factors/metabolism , Heart Defects, Congenital/genetics , CRISPR-Cas Systems/genetics
3.
Elife ; 112022 04 04.
Article in English | MEDLINE | ID: mdl-35373735

ABSTRACT

Single nucleotide variants (SNVs) are prevalent genetic factors shaping individual trait profiles and disease susceptibility. The recent development and optimizations of base editors, rubber and pencil genome editing tools now promise to enable direct functional assessment of SNVs in model organisms. However, the lack of bioinformatic tools aiding target prediction limits the application of base editing in vivo. Here, we provide a framework for adenine and cytosine base editing in medaka (Oryzias latipes) and zebrafish (Danio rerio), ideal for scalable validation studies. We developed an online base editing tool ACEofBASEs (a careful evaluation of base-edits), to facilitate decision-making by streamlining sgRNA design and performing off-target evaluation. We used state-of-the-art adenine (ABE) and cytosine base editors (CBE) in medaka and zebrafish to edit eye pigmentation genes and transgenic GFP function with high efficiencies. Base editing in the genes encoding troponin T and the potassium channel ERG faithfully recreated known cardiac phenotypes. Deep-sequencing of alleles revealed the abundance of intended edits in comparison to low levels of insertion or deletion (indel) events for ABE8e and evoBE4max. We finally validated missense mutations in novel candidate genes of congenital heart disease (CHD) dapk3, ube2b, usp44, and ptpn11 in F0 and F1 for a subset of these target genes with genotype-phenotype correlation. This base editing framework applies to a wide range of SNV-susceptible traits accessible in fish, facilitating straight-forward candidate validation and prioritization for detailed mechanistic downstream studies.


DNA contains sequences of four different molecules known as bases that represent our genetic code. In a mutation called a single nucleotide variant (or SNV for short), a single base in the sequence is swapped for another base. This can lead the individual carrying this SNV to produce a slightly different version of a protein to that found in other people. This slightly different protein may not work properly, or may perform a different task. In recent years, researchers have identified thousands of SNVs in humans linked with congenital heart diseases, but the roles of many of these SNVs remain unclear. Tools known as base editors allow researchers to efficiently modify single bases in DNA. Base editors use molecules known as short guide RNAs (or sgRNAs for short) to direct enzymes to specific positions in the DNA to swap, delete or insert a base. The sgRNAs need to be carefully designed to target the correct bases, however, which is a time consuming process. Furthermore, base editors were developed in cells grown in laboratories and so far only a few studies have demonstrated how they could be used in living animals. To overcome these limitations, Cornean, Gierten, Welz et al. developed a framework for base editing in two species of fish that are often used as models in research, namely medaka and zebrafish. The framework uses existing base editors that swap individual target bases and a new online tool ­ referred to as ACEofBASEs ­ to help design the required sgRNAs. The team were able to use the framework to characterize the medaka equivalents of four SNVs that have been previously associated with congenital heart disease in humans. The new framework developed here will help researchers to investigate the roles of SNVs in fish and other animals and validate human disease candidates. This approach could also be used to study the various ways that cells modify proteins by changing the specific bases involved in such modifications.


Subject(s)
Gene Editing , Zebrafish , Adenine , Animals , CRISPR-Cas Systems , Cytosine , DNA , Mutation , Zebrafish/genetics
4.
Genome Biol ; 23(1): 59, 2022 02 21.
Article in English | MEDLINE | ID: mdl-35189950

ABSTRACT

BACKGROUND: Unraveling the relationship between genetic variation and phenotypic traits remains a fundamental challenge in biology. Mapping variants underlying complex traits while controlling for confounding environmental factors is often problematic. To address this, we establish a vertebrate genetic resource specifically to allow for robust genotype-to-phenotype investigations. The teleost medaka (Oryzias latipes) is an established genetic model system with a long history of genetic research and a high tolerance to inbreeding from the wild. RESULTS: Here we present the Medaka Inbred Kiyosu-Karlsruhe (MIKK) panel: the first near-isogenic panel of 80 inbred lines in a vertebrate model derived from a wild founder population. Inbred lines provide fixed genomes that are a prerequisite for the replication of studies, studies which vary both the genetics and environment in a controlled manner, and functional testing. The MIKK panel will therefore enable phenotype-to-genotype association studies of complex genetic traits while allowing for careful control of interacting factors, with numerous applications in genetic research, human health, drug development, and fundamental biology. CONCLUSIONS: Here we present a detailed characterization of the genetic variation across the MIKK panel, which provides a rich and unique genetic resource to the community by enabling large-scale experiments for mapping complex traits.


Subject(s)
Oryzias , Animals , Genome , Inbreeding , Oryzias/genetics , Phenotype
5.
Genome Biol ; 23(1): 58, 2022 02 21.
Article in English | MEDLINE | ID: mdl-35189951

ABSTRACT

BACKGROUND: The teleost medaka (Oryzias latipes) is a well-established vertebrate model system, with a long history of genetic research, and multiple high-quality reference genomes available for several inbred strains. Medaka has a high tolerance to inbreeding from the wild, thus allowing one to establish inbred lines from wild founder individuals. RESULTS: We exploit this feature to create an inbred panel resource: the Medaka Inbred Kiyosu-Karlsruhe (MIKK) panel. This panel of 80 near-isogenic inbred lines contains a large amount of genetic variation inherited from the original wild population. We use Oxford Nanopore Technologies (ONT) long read data to further investigate the genomic and epigenomic landscapes of a subset of the MIKK panel. Nanopore sequencing allows us to identify a large variety of high-quality structural variants, and we present results and methods using a pan-genome graph representation of 12 individual medaka lines. This graph-based reference MIKK panel genome reveals novel differences between the MIKK panel lines and standard linear reference genomes. We find additional MIKK panel-specific genomic content that would be missing from linear reference alignment approaches. We are also able to identify and quantify the presence of repeat elements in each of the lines. Finally, we investigate line-specific CpG methylation and performed differential DNA methylation analysis across these 12 lines. CONCLUSIONS: We present a detailed analysis of the MIKK panel genomes using long and short read sequence technologies, creating a MIKK panel-specific pan genome reference dataset allowing for investigation of novel variation types that would be elusive using standard approaches.


Subject(s)
Oryzias , Animals , Epigenomics , Genome , Genomics/methods , Humans , Oryzias/genetics
6.
PLoS One ; 16(12): e0261572, 2021.
Article in English | MEDLINE | ID: mdl-34919578

ABSTRACT

Genetics crucially contributes to cardiovascular diseases (CVDs), the global leading cause of death. Since the majority of CVDs can be prevented by early intervention there is a high demand for the identification of predictive causative genes. While genome wide association studies (GWAS) correlate genes and CVDs after diagnosis and provide a valuable resource for such causative candidate genes, often preferentially those with previously known or suspected function are addressed further. To tackle the unaddressed blind spot of understudied genes, we particularly focused on the validation of human heart phenotype-associated GWAS candidates with little or no apparent connection to cardiac function. Building on the conservation of basic heart function and underlying genetics from fish to human we combined CRISPR/Cas9 genome editing of the orthologs of human GWAS candidates in isogenic medaka with automated high-throughput heart rate analysis. Our functional analyses of understudied human candidates uncovered a prominent fraction of heart rate associated genes from adult human patients impacting on the heart rate in embryonic medaka already in the injected generation. Following this pipeline, we identified 16 GWAS candidates with potential diagnostic and predictive power for human CVDs.


Subject(s)
Cardiovascular Diseases/genetics , Heart Rate/genetics , Myosin Light Chains/genetics , Oryzias/genetics , Animals , Animals, Genetically Modified , CRISPR-Cas Systems/genetics , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/pathology , Gene Editing , Genome-Wide Association Study , Humans , Promoter Regions, Genetic/genetics
7.
Nat Methods ; 18(5): 557-563, 2021 05.
Article in English | MEDLINE | ID: mdl-33963344

ABSTRACT

Visualizing dynamic processes over large, three-dimensional fields of view at high speed is essential for many applications in the life sciences. Light-field microscopy (LFM) has emerged as a tool for fast volumetric image acquisition, but its effective throughput and widespread use in biology has been hampered by a computationally demanding and artifact-prone image reconstruction process. Here, we present a framework for artificial intelligence-enhanced microscopy, integrating a hybrid light-field light-sheet microscope and deep learning-based volume reconstruction. In our approach, concomitantly acquired, high-resolution two-dimensional light-sheet images continuously serve as training data and validation for the convolutional neural network reconstructing the raw LFM data during extended volumetric time-lapse imaging experiments. Our network delivers high-quality three-dimensional reconstructions at video-rate throughput, which can be further refined based on the high-resolution light-sheet images. We demonstrate the capabilities of our approach by imaging medaka heart dynamics and zebrafish neural activity with volumetric imaging rates up to 100 Hz.


Subject(s)
Deep Learning , Heart/physiology , Image Processing, Computer-Assisted/methods , Microscopy/methods , Animals , Biomechanical Phenomena , Calcium/chemistry , Larva/physiology , Oryzias/physiology , Reproducibility of Results , Zebrafish/physiology
8.
Nature ; 584(7822): 589-594, 2020 08.
Article in English | MEDLINE | ID: mdl-32814899

ABSTRACT

The inner surfaces of the human heart are covered by a complex network of muscular strands that is thought to be a remnant of embryonic development1,2. The function of these trabeculae in adults and their genetic architecture are unknown. Here we performed a genome-wide association study to investigate image-derived phenotypes of trabeculae using the fractal analysis of trabecular morphology in 18,096 participants of the UK Biobank. We identified 16 significant loci that contain genes associated with haemodynamic phenotypes and regulation of cytoskeletal arborization3,4. Using biomechanical simulations and observational data from human participants, we demonstrate that trabecular morphology is an important determinant of cardiac performance. Through genetic association studies with cardiac disease phenotypes and Mendelian randomization, we find a causal relationship between trabecular morphology and risk of cardiovascular disease. These findings suggest a previously unknown role for myocardial trabeculae in the function of the adult heart, identify conserved pathways that regulate structural complexity and reveal the influence of the myocardial trabeculae on susceptibility to cardiovascular disease.


Subject(s)
Cardiovascular Diseases/genetics , Fractals , Genetic Predisposition to Disease , Heart/anatomy & histology , Heart/physiology , Myocardium/metabolism , Adult , Aged , Animals , Cardiovascular Diseases/physiopathology , Cytoskeleton/genetics , Cytoskeleton/physiology , Gene Knockout Techniques , Genetic Loci/genetics , Genome-Wide Association Study , Heart/embryology , Hemodynamics , Humans , Middle Aged , Myocardium/cytology , Oryzias/embryology , Oryzias/genetics , Phenotype
9.
Sci Rep ; 10(1): 2046, 2020 02 06.
Article in English | MEDLINE | ID: mdl-32029752

ABSTRACT

Accurate quantification of heartbeats in fish models is an important readout to study cardiovascular biology, disease states and pharmacology. However, dependence on anaesthesia, laborious sample orientation or requirement for fluorescent reporters have hampered the use of high-throughput heartbeat analysis. To overcome these limitations, we established an efficient screening assay employing automated label-free heart rate determination of randomly oriented, non-anesthetized medaka (Oryzias latipes) and zebrafish (Danio rerio) embryos in microtiter plates. Automatically acquired bright-field data feeds into an easy-to-use HeartBeat software with graphical user interface for automated quantification of heart rate and rhythm. Sensitivity of the assay was demonstrated by profiling heart rates during entire embryonic development. Our analysis revealed rapid adaption of heart rates to temperature changes, which has implications for standardization of experimental layout. The assay allows scoring of multiple embryos per well enabling a throughput of >500 embryos per 96-well plate. In a proof of principle screen for compound testing, we captured concentration-dependent effects of nifedipine and terfenadine over time. Our novel assay permits large-scale applications ranging from phenotypic screening, interrogation of gene functions to cardiovascular drug development.


Subject(s)
Heart Rate/physiology , High-Throughput Screening Assays , Monitoring, Physiologic/methods , Oryzias/physiology , Zebrafish/physiology , Animals , Drug Evaluation, Preclinical/methods , Embryo, Nonmammalian , Heart Rate/drug effects , Models, Animal , Nifedipine/pharmacology , Proof of Concept Study , Software , Terfenadine/pharmacology
10.
Zebrafish ; 16(6): 542-545, 2019 12.
Article in English | MEDLINE | ID: mdl-31536467

ABSTRACT

Medaka (Oryzias latipes) and zebrafish (Danio rerio) contribute substantially to our understanding of the genetic and molecular etiology of human cardiovascular diseases. In this context, the quantification of important cardiac functional parameters is fundamental. We have developed a framework that segments the ventricle of a medaka hatchling from image sequences and subsequently quantifies ventricular dimensions.


Subject(s)
Heart Ventricles/anatomy & histology , Machine Learning , Oryzias/anatomy & histology , Animals
11.
Nat Methods ; 16(6): 497-500, 2019 06.
Article in English | MEDLINE | ID: mdl-31036959

ABSTRACT

To capture highly dynamic biological processes at cellular resolution is a recurring challenge in biology. Here we show that combining selective-volume illumination with simultaneous acquisition of orthogonal light fields yields three-dimensional images with high, isotropic spatial resolution and a significant reduction of reconstruction artefacts, thereby overcoming current limitations of light-field microscopy implementations. We demonstrate medaka heart and blood flow imaging at single-cell resolution and free of motion artefacts at volume rates of up to 200 Hz.


Subject(s)
Heart/diagnostic imaging , Heart/physiology , Image Processing, Computer-Assisted/methods , Microscopy/methods , Single-Cell Analysis/methods , Algorithms , Animals , Animals, Genetically Modified , Artifacts , Blood Flow Velocity , Humans , Imaging, Three-Dimensional/methods , Oryzias
12.
J Mol Cell Cardiol ; 126: 96-104, 2019 01.
Article in English | MEDLINE | ID: mdl-30472253

ABSTRACT

Two-pore-domain potassium (K2P) channels conduct background potassium currents in the heart and other tissues. K2P currents are involved in the repolarization of action potentials and stabilize the resting membrane potential. Human K2P13.1 (THIK-1) channels are expressed in the heart and have recently been implicated in atrial fibrillation. The in vivo significance of K2P13.1 currents in cardiac electrophysiology is not known. We hypothesized that Danio rerio (zebrafish) may serve as model to elucidate the functional role of cardiac K2P13.1 channels. This work was designed to characterize zebrafish orthologs of K2P13.1. Two zkcnk13 coding sequences were identified by DNA database searches and amplified from zebrafish cDNA. Human and zebrafish K2P13.1 proteins exhibit 70% (K2P13.1a) and 66% (K2P13.1b) identity. Kcnk13 expression in zebrafish was studied using polymerase chain reaction. Zebrafish kcnk13a and zkcnk13b mRNAs were detected in brain and heart. Human and zebrafish K2P13.1 currents were analyzed in the Xenopus oocyte expression system by voltage clamp electrophysiology. Zebrafish K2P13.1a polypeptides were non-functional, while zK2P13.1b channels exhibited K+ selective, outwardly rectifying currents. Zebrafish and human K2P13.1 currents were similarly activated by arachidonic acid and reduced by barium, mexiletine, lidocaine, and inhibition of phospholipase C. In conclusion, zebrafish K2P13.1b channels and their human orthologs exhibit structural and regulatory similarities. Zebrafish may be used as in vivo model for the assessment of physiology and therapeutic significance of K2P13.1.


Subject(s)
Potassium Channels, Tandem Pore Domain/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Cloning, Molecular , Humans , Hydrogen-Ion Concentration , Peptides/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Zebrafish Proteins/chemistry , Zebrafish Proteins/metabolism
13.
Eur J Pharmacol ; 831: 94-102, 2018 Jul 15.
Article in English | MEDLINE | ID: mdl-29753045

ABSTRACT

Human K2P17.1 (TASK-4, TALK-2) two-pore-domain potassium (K2P) channels have recently been implicated in heart rhythm disorders including atrial fibrillation and conduction disease. The functional in vivo significance of K2P17.1 currents in cardiac electrophysiology remains incompletely understood. Danio rerio (zebrafish) may be utilized to elucidate the role of cardiac K2P channels in vivo. The aim of this work was to identify and characterize the zebrafish ortholog of K2P17.1 in comparison to its human counterpart. The zkcnk17 coding sequence was amplified from zebrafish cDNA. Zebrafish kcnk17 mRNA expression was assessed by polymerase chain reaction. Human and zebrafish K2P17.1 currents were analyzed using two-electrode voltage clamp electrophysiology and the Xenopus oocyte expression system. Kcnk17 mRNA was detected in zebrafish brain. Human and zebrafish K2P17.1 proteins exhibited 33.4% identity. Zebrafish K2P17.1 channels conducted K+ selective currents with open rectification properties. Both human and zebrafish K2P17.1 were inhibited by barium. In contrast to human K2P17.1, zK2P17.1 currents were not sensitive to extracellular alkalization, likely due to the lack of a lysine residue involved in pH sensing of hK2P17.1. In conclusion, zebrafish and human K2P17.1 channels display similar structural and regulatory properties. Zebrafish may serve as an in vivo model to study neuronal K2P17.1 function but does not appear appropriate for cardiac electrophysiology studies. Differences in pH sensitivity of zK2P17.1 currents need to be considered when zebrafish data are extrapolated to human physiology.


Subject(s)
Potassium Channels, Tandem Pore Domain/metabolism , Zebrafish Proteins/metabolism , Animals , Barium Compounds/pharmacology , Female , Gene Expression Regulation , Humans , Hydrogen-Ion Concentration , Membrane Potentials , Oocytes , Potassium Channel Blockers/pharmacology , Potassium Channels, Tandem Pore Domain/chemistry , Potassium Channels, Tandem Pore Domain/drug effects , Potassium Channels, Tandem Pore Domain/genetics , Protein Conformation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Structure-Activity Relationship , Xenopus laevis/genetics , Xenopus laevis/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics
14.
Naunyn Schmiedebergs Arch Pharmacol ; 387(3): 291-300, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24311292

ABSTRACT

The human KCNK18 gene is predominantly expressed in brain, spinal cord, and dorsal root ganglion neurons. Encoded K2P18.1K(+) channels are functionally implicated in migraine, pain and anesthesia. Data delineating the in vivo significance of K2P18.1 are still limited owing to a lack of model systems allowing for rapid, whole organism phenotypic analyses. We hypothesized that zebrafish (Danio rerio) might close this scientific gap. This work was designed to characterize the zebrafish ortholog of K2P18.1 in comparison to human K2P18.1 channels. The complete coding sequence of zKCNK18 was amplified from zebrafish cDNA. Zebrafish KCNK18 expression was assessed by in situ hybridization. Human and zebrafish K2P18.1 currents were functionally analyzed using two-electrode voltage clamp electrophysiology and the Xenopus oocyte expression system. KCNK18 mRNA is expressed in zebrafish brain and eyes. Human and zebrafish K2P18.1 proteins share 32 % identity. Zebrafish K2P18.1 channels mediate K(+)-selective background currents that stabilize the negative resting membrane potential. Functional similarities between human and zK2P18.1 currents include open rectification properties, inhibition by barium, and regulation by signaling molecules protein kinase (PK)C, PKA, and phospholipase C. In contrast to the human ortholog, zK2P18.1 exhibited reduced sensitivity to elevation of intracellular calcium levels by ionomycin and was virtually insensitive to inhibition by quinidine. Zebrafish and human K2P18.1 channels share functional and regulatory properties, indicating that the zebrafish may serve as model to assess K2P18.1 function in vivo. However, distinct differences in K2P18.1 current regulation require careful consideration when zebrafish data are extrapolated to human physiology.


Subject(s)
Potassium Channels, Tandem Pore Domain/metabolism , Potassium Channels/metabolism , Animals , Calcium/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , DNA, Complementary/genetics , Female , Humans , In Situ Hybridization , Membrane Potentials/physiology , Oocytes , Patch-Clamp Techniques , Potassium Channels/genetics , Protein Kinase C/metabolism , Quinidine/pharmacology , Species Specificity , Type C Phospholipases/metabolism , Xenopus laevis , Zebrafish
15.
Biochim Biophys Acta ; 1818(1): 33-41, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21963410

ABSTRACT

Two-pore-domain potassium (K(2P)) channels mediate K(+) background currents that stabilize the resting membrane potential and contribute to repolarization of action potentials in excitable cells. The functional significance of K(2P) currents in cardiac electrophysiology remains poorly understood. Danio rerio (zebrafish) may be utilized to elucidate the role of cardiac K(2P) channels in vivo. The aim of this work was to identify and functionally characterize a zebrafish otholog of the human K(2P)10.1 channel. K(2P)10.1 orthologs in the D. rerio genome were identified by database analysis, and the full zK(2P)10.1 coding sequence was amplified from zebrafish cDNA. Human and zebrafish K(2P)10.1 proteins share 61% identity. High degrees of conservation were observed in protein domains relevant for structural integrity and regulation. K(2P)10.1 channels were heterologously expressed in Xenopus oocytes, and currents were recorded using two-electrode voltage clamp electrophysiology. Human and zebrafish channels mediated K(+) selective background currents leading to membrane hyperpolarization. Arachidonic acid, an activator of hK(2P)10.1, induced robust activation of zK(2P)10.1. Activity of both channels was reduced by protein kinase C. Similar to its human counterpart, zK(2P)10.1 was inhibited by the antiarrhythmic drug amiodarone. In summary, zebrafish harbor K(2P)10.1 two-pore-domain K(+) channels that exhibit structural and functional properties largely similar to human K(2P)10.1. We conclude that the zebrafish represents a valid model to study K(2P)10.1 function in vivo.


Subject(s)
Potassium Channels, Tandem Pore Domain/metabolism , Potassium/metabolism , Zebrafish Proteins/metabolism , Amiodarone/pharmacology , Animals , Anti-Arrhythmia Agents/pharmacology , Arachidonic Acid/pharmacology , Conserved Sequence , DNA, Complementary/biosynthesis , Electrophysiology , Gene Expression , Humans , Membrane Potentials/drug effects , Oocytes/cytology , Oocytes/metabolism , Patch-Clamp Techniques , Phylogeny , Plasmids , Potassium Channels, Tandem Pore Domain/chemistry , Potassium Channels, Tandem Pore Domain/genetics , Protein Kinase C/metabolism , Sequence Homology, Amino Acid , Transfection , Xenopus laevis , Zebrafish , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics
16.
Br J Pharmacol ; 166(2): 764-73, 2012 May.
Article in English | MEDLINE | ID: mdl-22168364

ABSTRACT

BACKGROUND AND PURPOSE: Two-pore-domain K(+) channels (K(2P) ) mediate K(+) background currents that modulate the membrane potential of excitable cells. K(2P) 18.1 (TWIK-related spinal cord K(+) channel) provides hyperpolarizing background currents in neurons. Recently, a dominant-negative loss-of-function mutation in K(2P) 18.1 has been implicated in migraine, and activation of K(2P) 18.1 channels was proposed as a therapeutic strategy. Here we elucidated the molecular mechanisms underlying PKC-dependent activation of K(2P) 18.1 currents. EXPERIMENTAL APPROACH: Human K(2P) 18.1 channels were heterologously expressed in Xenopus laevis oocytes, and currents were recorded with the two-electrode voltage clamp technique. KEY RESULTS: Stimulation of PKC using phorbol 12-myristate-13-acetate (PMA) activated the hK(2P) 18.1 current by 3.1-fold in a concentration-dependent fashion. The inactive analogue 4α-PMA had no effect on channel activity. The specific PKC inhibitors bisindolylmaleimide I, Ro-32-0432 and chelerythrine reduced PMA-induced channel activation indicating that PKC is involved in this effect of PMA. Selective activation of conventional PKC isoforms with thymeleatoxin (100 nM) did not reproduce K(2P) 18.1 channel activation. Current activation by PMA was not affected by pretreatment with CsA (calcineurin inhibitor) or KT 5720 (PKA inhibitor), ruling out a significant contribution of calcineurin or cross-talk with PKA to the PKC-dependent hK(2P) 18.1 activation. Finally, mutation of putative PKC phosphorylation sites did not prevent PMA-induced K(2P) 18.1 channel activation. CONCLUSIONS AND IMPLICATIONS: We demonstrated that activation of hK(2P) 18.1 (TRESK) by PMA is mediated by PKC stimulation. Hence, PKC-mediated activation of K(2P) 18.1 background currents may serve as a novel molecular target for migraine treatment.


Subject(s)
Potassium Channels, Tandem Pore Domain/physiology , Protein Kinase C/physiology , Animals , Humans , Oocytes/drug effects , Oocytes/physiology , Signal Transduction , Tetradecanoylphorbol Acetate/pharmacology , Xenopus laevis
17.
Naunyn Schmiedebergs Arch Pharmacol ; 381(3): 261-70, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19777211

ABSTRACT

Two-pore-domain (K(2P)) potassium channels mediate background potassium currents, stabilizing resting membrane potential and expediting action potential repolarization. In the heart, K(2P)3.1 (TASK-1) channels are implicated in the cardiac plateau current, I ( KP ). Class III antiarrhythmic drugs target cardiac K(+) currents, resulting in action potential prolongation and suppression of atrial and ventricular arrhythmias. The objective of this study was to investigate acute effects of the class III antiarrhythmic drug amiodarone on human K(2P)3.1 channels. Potassium currents were recorded from Xenopus oocytes using the two-microelectrode voltage clamp technique. Amiodarone produced concentration-dependent inhibition of hK(2P)3.1 currents (IC(50) = 0.40 microM) with maximum current reduction of 58.1%. Open rectification properties that are characteristic to hK(2P)3.1 currents were not altered by amiodarone. Channels were blocked in open and closed states in reverse frequency-dependent manner. hK(2P)3.1 channel inhibition was voltage-independent at voltages between -40 and +60 mV. Modulation of protein kinase C activity by amiodarone does not contribute to hK(2P)3.1 current reduction, as pre-treatment with the protein kinase C inhibitor, staurosporine, did not affect amiodarone block. Amiodarone is an inhibitor of cardiac hK(2P)3.1 background channels. Amiodarone blockade of hK(2P)3.1 may cause prolongation of cardiac repolarization and action potential duration in patients with high individual plasma concentrations, possibly contributing to the antiarrhythmic efficacy of the class III drug.


Subject(s)
Amiodarone/pharmacology , Anti-Arrhythmia Agents/pharmacology , Myocardium/metabolism , Nerve Tissue Proteins/antagonists & inhibitors , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Action Potentials/drug effects , Animals , Cloning, Molecular , Dose-Response Relationship, Drug , Female , Oocytes/cytology , Oocytes/metabolism , Patch-Clamp Techniques , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Staurosporine/pharmacology , Xenopus laevis
18.
Eur J Pharmacol ; 579(1-3): 98-103, 2008 Jan 28.
Article in English | MEDLINE | ID: mdl-18054910

ABSTRACT

The antihypertensive drug doxazosin has been associated with an increased risk for congestive heart failure and cardiomyocyte apoptosis. Human ether-a-go-go-related gene (hERG) K(+) channels, previously shown to be blocked by doxazosin at therapeutically relevant concentrations, represent plasma membrane receptors for the antihypertensive drug. To elucidate the molecular basis for doxazosin-associated pro-apoptotic effects, cell death was studied in human embryonic kidney cells using three independent apoptosis assays. Doxazosin specifically induced apoptosis in hERG-expressing HEK cells, while untransfected control groups were insensitive to treatment with the antihypertensive agent. An unexpected biological mechanism has emerged: binding of doxazosin to its novel membrane receptor, hERG, triggers apoptosis, possibly representing a broader pathophysiological mechanism in drug-induced heart failure.


Subject(s)
Antihypertensive Agents/pharmacology , Apoptosis/drug effects , Doxazosin/pharmacology , Ether-A-Go-Go Potassium Channels/drug effects , Adrenergic alpha-Antagonists/pharmacology , Cell Line , Electrophysiology , Ether-A-Go-Go Potassium Channels/metabolism , Flow Cytometry , Humans , In Situ Nick-End Labeling , Staining and Labeling
SELECTION OF CITATIONS
SEARCH DETAIL
...