74 results on '"Rook MB"'
Search Results
2. Structure-Affinity Relationships (SARs) and Structure-Kinetics Relationships (SKRs) of Kv11.1 Blockers
- Author
-
Yu, Zhiyi, van Veldhoven, Jacobus P D, Louvel, Julien, 't Hart, Ingrid M E, Rook, MB, van der Heyden, MAG, Heitman, Laura H, IJzerman, Adriaan P, Yu, Zhiyi, van Veldhoven, Jacobus P D, Louvel, Julien, 't Hart, Ingrid M E, Rook, MB, van der Heyden, MAG, Heitman, Laura H, and IJzerman, Adriaan P
- Published
- 2015
3. Structure-Affinity Relationships (SARs) and Structure-Kinetics Relationships (SKRs) of Kv11.1 Blockers
- Author
-
Medische Fysiologie, Circulatory Health, TN groep Adan, Yu, Zhiyi, van Veldhoven, Jacobus P D, Louvel, Julien, 't Hart, Ingrid M E, Rook, MB, van der Heyden, MAG, Heitman, Laura H, IJzerman, Adriaan P, Medische Fysiologie, Circulatory Health, TN groep Adan, Yu, Zhiyi, van Veldhoven, Jacobus P D, Louvel, Julien, 't Hart, Ingrid M E, Rook, MB, van der Heyden, MAG, Heitman, Laura H, and IJzerman, Adriaan P
- Published
- 2015
4. The anti-protozoal drug pentamidine blocks KIR2.x-mediated inward rectifier current by entering the cytoplasmic pore region of the channel
- Author
-
de Boer, TP, Nalos, L, Stary, A, Kok, B, Houtman, MJC, Antoons, G, van Veen, TAB, Beekman, JDM, de Groot, BL, Opthof, T, Rook, MB, Vos, MA, and van der Heyden, MAG
- Subjects
Cytoplasm ,Dogs ,Patch-Clamp Techniques ,Blotting, Western ,Mutation ,Antiprotozoal Agents ,Animals ,Humans ,Potassium Channels, Inwardly Rectifying ,Research Papers ,Pentamidine ,Cell Line - Abstract
Pentamidine is a drug used in treatment of protozoal infections. Pentamidine treatment may cause sudden cardiac death by provoking cardiac arrhythmias associated with QTc prolongation and U-wave alterations. This proarrhythmic effect was linked to inhibition of hERG trafficking, but not to acute block of ion channels contributing to the action potential. Because the U-wave has been linked to the cardiac inward rectifier current (I(K1)), we examined the action and mechanism of pentamidine-mediated I(K1) block.Patch clamp measurements of I(K1) were made on cultured adult canine ventricular cardiomyocytes, K(IR)2.1-HEK293 cells and K(IR)2.x inside-out patches. Pentamidine binding to cytoplasmic amino acid residues of K(IR)2.1 channels was studied by molecular modelling.Pentamidine application (24 h) decreased I(K1) in cultured canine cardiomyocytes and K(IR)2.1-HEK293 cells under whole cell clamp conditions. Pentamidine inhibited I(K1) in K(IR)2.1-HEK293 cells 10 min after application. When applied to the cytoplasmic side under inside-out patch clamp conditions, pentamidine block of I(K1) was acute (IC(50)= 0.17 microM). Molecular modelling predicted pentamidine-channel interactions in the cytoplasmic pore region of K(IR)2.1 at amino acids E224, D259 and E299. Mutation of these conserved residues to alanine reduced pentamidine block of I(K1). Block was independent of the presence of spermine. K(IR)2.2, and K(IR)2.3 based I(K1) was also sensitive to pentamidine blockade.Pentamidine inhibits cardiac I(K1) by interacting with three negatively charged amino acids in the cytoplasmic pore region. Our findings may provide new insights for development of specific I(K1) blocking compounds.
- Published
- 2010
5. An electrophysiological characterizaton of Kv7.2 and Nav1.1 channel mutations in genetic epilepsy
- Author
-
Lindhout, D., Koeleman, BPC, Rook, MB, Volkers, L., Lindhout, D., Koeleman, BPC, Rook, MB, and Volkers, L.
- Published
- 2012
6. An electrophysiological characterizaton of Kv7.2 and Nav1.1 channel mutations in genetic epilepsy
- Author
-
Brain, Circulatory Health, Genetica, Lindhout, D., Koeleman, BPC, Rook, MB, Volkers, L., Brain, Circulatory Health, Genetica, Lindhout, D., Koeleman, BPC, Rook, MB, and Volkers, L.
- Published
- 2012
7. Probing cardiac repolarization reserve in drug safety assessment
- Author
-
Vos, MA, van der Heyden, MAG, Rook, MB, Nalos, L., Vos, MA, van der Heyden, MAG, Rook, MB, and Nalos, L.
- Published
- 2011
8. Probing cardiac repolarization reserve in drug safety assessment
- Author
-
Circulatory Health, Medische Fysiologie, Vos, MA, van der Heyden, Marcel, Rook, MB, Nalos, L., Circulatory Health, Medische Fysiologie, Vos, MA, van der Heyden, Marcel, Rook, MB, and Nalos, L.
- Published
- 2011
9. Comparison of the IKr blockers moxifloxacin, dofetilide and E-4031 in five screening models of pro-arrhythmia reveals lack of specificity of isolated cardiomyocytes
- Author
-
Nalos, L, primary, Varkevisser, R, additional, Jonsson, MKB, additional, Houtman, MJC, additional, Beekman, JD, additional, van der Nagel, R, additional, Thomsen, MB, additional, Duker, G, additional, Sartipy, P, additional, de Boer, TP, additional, Peschar, M, additional, Rook, MB, additional, van Veen, TAB, additional, van der Heyden, MAG, additional, and Vos, MA, additional
- Published
- 2011
- Full Text
- View/download PDF
10. The anti-protozoal drug pentamidine blocks KIR2.x-mediated inward rectifier current by entering the cytoplasmic pore region of the channel
- Author
-
De Boer, TP, primary, Nalos, L, additional, Stary, A, additional, Kok, B, additional, Houtman, MJC, additional, Antoons, G, additional, Van Veen, TAB, additional, Beekman, JDM, additional, De Groot, BL, additional, Opthof, T, additional, Rook, MB, additional, Vos, MA, additional, and Van Der Heyden, MAG, additional
- Published
- 2010
- Full Text
- View/download PDF
11. Comparison of the IKr blockers moxifloxacin, dofetilide and E-4031 in five screening models of pro-arrhythmia reveals lack of specificity of isolated cardiomyocytes.
- Author
-
Nalos, L, Varkevisser, R, Jonsson, MKB, Houtman, MJC, Beekman, JD, van der Nagel, R, Thomsen, MB, Duker, G, Sartipy, P, de Boer, TP, Peschar, M, Rook, MB, van Veen, TAB, van der Heyden, MAG, and Vos, MA
- Subjects
MOXIFLOXACIN ,DOFETILIDE ,ARRHYTHMIA ,HEART cells ,DRUG development ,DRUG side effects ,COMPARATIVE studies - Abstract
BACKGROUND AND PURPOSE Drug development requires the testing of new chemical entities for adverse effects. For cardiac safety screening, improved assays are urgently needed. Isolated adult cardiomyocytes (CM) and human embryonic stem cell-derived cardiomyocytes (hESC-CM) could be used to identify pro-arrhythmic compounds. In the present study, five assays were employed to investigate their sensitivity and specificity for evaluating the pro-arrhythmic properties of I
Kr blockers, using moxifloxacin (safe compound) and dofetilide or E-4031 (unsafe compounds). EXPERIMENTAL APPROACH Assays included the anaesthetized remodelled chronic complete AV block (CAVB) dog, the anaesthetized methoxamine-sensitized unremodelled rabbit, multi-cellular hESC-CM clusters, isolated CM obtained from CAVB dogs and isolated CM obtained from the normal rabbit. Arrhythmic outcome was defined as Torsade de Pointes (TdP) in the animal models and early afterdepolarizations (EADs) in the cell models. KEY RESULTS At clinically relevant concentrations (5-12 µM), moxifloxacin was free of pro-arrhythmic properties in all assays with the exception of the isolated CM, in which 10 µM induced EADs in 35% of the CAVB CM and in 23% of the rabbit CM. At supra-therapeutic concentrations (≥100 µM), moxifloxacin was pro-arrhythmic in the isolated rabbit CM (33%), in the hESC-CM clusters (18%), and in the methoxamine rabbit (17%). Dofetilide and E-4031 induced EADs or TdP in all assays (50-83%), and the induction correlated with a significant increase in beat-to-beat variability of repolarization. CONCLUSION AND IMPLICATIONS Isolated cardiomyocytes lack specificity to discriminate between TdP liability of the IKr blocking drugs moxifloxacin and dofetilide or E4031. [ABSTRACT FROM AUTHOR]- Published
- 2012
- Full Text
- View/download PDF
12. An electrophysiological characterizaton of Kv7.2 and Nav1.1 channel mutations in genetic epilepsy
- Author
-
Volkers, L., Lindhout, D., Koeleman, BPC, Rook, MB, and University Utrecht
- Subjects
KCNQ2 ,GEFS+ ,Econometric and Statistical Methods: General ,Epilepsy ,potassium ,Medical sciences ,General [Econometric and Statistical Methods] ,Dravet syndrome ,BFNE ,ion channel ,Bescherming en bevordering van de menselijke gezondheid ,SCN1A ,Geneeskunde(GENK) ,sodium - Abstract
Most genetically determined epilepsies are caused by gene mutations in genes that encode for ion channels and receptors. Functional analysis of these protein mutants shows changes in voltage dependency and/or kinetics or a complete loss of channel function via different mechanisms. Nevertheless, the severity of the epileptic seizures is not only determined by a specific gene mutation, but genetic background, modifying genes and genetic compensatory factors are playing a crucial role in the phenotypic outcome of patients.
- Published
- 2012
13. Probing cardiac repolarization reserve in drug safety assessment
- Author
-
Nalos, L., Vos, MA, van der Heyden, MAG, Rook, MB, and University Utrecht
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,KIR2.1 ,drug safety assessment ,hERG ,cardiovascular diseases ,cardiac repolarization reserve ,QT prolongation - Abstract
Excessive prolongation of cardiac repolarization, manifested as QT prolongation on ECG, is common unwanted side effect of many drugs and drug candidates. Prolongation of QT interval may lead to life threatening cardiac arrhythmia – Torsade de Point (TdP). Number of drugs was withdrawn from the market and development of many new drug candidates is terminated because of this side effect. Block of the hERG channel is believed to be the principal reason of QT prolongation and thus current safety testing assays are based on determining the hERG block potency of new drug candidates. Here we demonstrate that focus on hERG block is counterproductive. Drugs affecting other repolarizing currents might be proarrhythmic as well. In the same time, potent hERG blocker with effect on depolarizing currents is not always proarrhythmic. We also show, that TdP can occur after chronic treatment due to effect on protein trafficking. This chronic effect is not tested in cardiac safety assessment at all. We have designed and synthesized 40 analogues of potent hERG blocker dofetilide to analyze molecular substructures required for hERG block. This approach may help design new drugs with low affinity to hERG channel. We identify pentamidine as a direct blocker of another important repolarizing current - IK1. The role of this current in cardiac arrhythmias is poorly studied, mainly because of absence of a specific IK1 blocker. We have tested 7 pentamidine analogues to find a specific and effective IK1 blocker applicable in vivo. Based on our findings, to improve current cardiac safety assessment, we employed five test assays and compare their sensitivity and specificity using hERG blockers with known proarrhythmic potential. We demonstrate that isolated cardiomyocytes lack sufficient specificity to correctly identify safe hERG blocker moxifloxacin. On the other hand, hESC-CM seems to be a promising alternative to current assays.
- Published
- 2011
14. NaV1.1 and NaV1.6 selective compounds reduce the behavior phenotype and epileptiform activity in a novel zebrafish model for Dravet Syndrome.
- Author
-
Weuring WJ, Singh S, Volkers L, Rook MB, van 't Slot RH, Bosma M, Inserra M, Vetter I, Verhoeven-Duif NM, Braun KPJ, Rivara M, and Koeleman BPC
- Subjects
- Animals, Anticonvulsants pharmacology, Disease Models, Animal, Epilepsies, Myoclonic metabolism, Humans, Locomotion drug effects, Morpholinos metabolism, Mutagenesis, NAV1.1 Voltage-Gated Sodium Channel chemistry, NAV1.1 Voltage-Gated Sodium Channel genetics, NAV1.6 Voltage-Gated Sodium Channel chemistry, NAV1.6 Voltage-Gated Sodium Channel genetics, Neurons drug effects, Neurons metabolism, Pentylenetetrazole pharmacology, Phenotype, RNA, Guide, CRISPR-Cas Systems metabolism, RNA, Messenger metabolism, Voltage-Gated Sodium Channel Agonists pharmacology, Voltage-Gated Sodium Channel Blockers pharmacology, Zebrafish, Zebrafish Proteins chemistry, Zebrafish Proteins genetics, gamma-Aminobutyric Acid metabolism, Epilepsies, Myoclonic pathology, NAV1.1 Voltage-Gated Sodium Channel metabolism, NAV1.6 Voltage-Gated Sodium Channel metabolism, Zebrafish Proteins metabolism
- Abstract
Dravet syndrome is caused by dominant loss-of-function mutations in SCN1A which cause reduced activity of Nav1.1 leading to lack of neuronal inhibition. On the other hand, gain-of-function mutations in SCN8A can lead to a severe epileptic encephalopathy subtype by over activating NaV1.6 channels. These observations suggest that Nav1.1 and Nav1.6 represent two opposing sides of the neuronal balance between inhibition and activation. Here, we hypothesize that Dravet syndrome may be treated by either enhancing Nav1.1 or reducing Nav1.6 activity. To test this hypothesis we generated and characterized a novel DS zebrafish model and tested new compounds that selectively activate or inhibit the human NaV1.1 or NaV1.6 channel respectively. We used CRISPR/Cas9 to generate two separate Scn1Lab knockout lines as an alternative to previous zebrafish models generated by random mutagenesis or morpholino oligomers. Using an optimized locomotor assay, spontaneous burst movements were detected that were unique to Scn1Lab knockouts and disappear when introducing human SCN1A mRNA. Besides the behavioral phenotype, Scn1Lab knockouts show sudden, electrical discharges in the brain that indicate epileptic seizures in zebrafish. Scn1Lab knockouts showed increased sensitivity to the GABA antagonist pentylenetetrazole and a reduction in whole organism GABA levels. Drug screenings further validated a Dravet syndrome phenotype. We tested the NaV1.1 activator AA43279 and two novel NaV1.6 inhibitors MV1369 and MV1312 in the Scn1Lab knockouts. Both type of compounds significantly reduced the number of spontaneous burst movements and seizure activity. Our results show that selective inhibition of NaV1.6 could be just as efficient as selective activation of NaV1.1 and these approaches could prove to be novel potential treatment strategies for Dravet syndrome and other (genetic) epilepsies. Compounds tested in zebrafish however, should always be further validated in other model systems for efficacy in mammals and to screen for potential side effects., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2020
- Full Text
- View/download PDF
15. Structure-Affinity Relationships (SARs) and Structure-Kinetics Relationships (SKRs) of Kv11.1 Blockers.
- Author
-
Yu Z, van Veldhoven JP, Louvel J, 't Hart IM, Rook MB, van der Heyden MA, Heitman LH, and IJzerman AP
- Subjects
- ERG1 Potassium Channel, HEK293 Cells, Humans, Kinetics, Structure-Activity Relationship, Ether-A-Go-Go Potassium Channels drug effects, Potassium Channel Blockers chemistry, Potassium Channel Blockers pharmacology
- Abstract
Kv11.1 (hERG) blockers with comparable potencies but different binding kinetics might display divergent pro-arrhythmic risks. In the present study, we explored structure-kinetics relationships in four series of Kv11.1 blockers next to their structure-affinity relationships. We learned that despite dramatic differences in affinities and association rates, there were hardly any variations in the dissociation rate constants of these molecules with residence times (RTs) of a few minutes only. Hence, we synthesized 16 novel molecules, in particular in the pyridinium class of compounds, to further address this peculiar phenomenon. We found molecules with very short RTs (e.g., 0.34 min for 37) and much longer RTs (e.g., 105 min for 38). This enabled us to construct a k on-k off-KD kinetic map for all compounds and subsequently divide the map into four provisional quadrants, providing a possible framework for a further and more precise categorization of Kv11.1 blockers. Additionally, two representative compounds (21 and 38) were tested in patch clamp assays, and their RTs were linked to their functional IC50 values. Our findings strongly suggest the importance of the simultaneous study of ligand affinities and kinetic parameters, which may help to explain and predict Kv11.1-mediated cardiotoxicity.
- Published
- 2015
- Full Text
- View/download PDF
16. Febrile temperatures unmask biophysical defects in Nav1.1 epilepsy mutations supportive of seizure initiation.
- Author
-
Volkers L, Kahlig KM, Das JH, van Kempen MJ, Lindhout D, Koeleman BP, and Rook MB
- Subjects
- Action Potentials, Cell Line, Epilepsy genetics, Humans, NAV1.1 Voltage-Gated Sodium Channel genetics, Hot Temperature, Ion Channel Gating, Mutation, NAV1.1 Voltage-Gated Sodium Channel metabolism
- Abstract
Generalized epilepsy with febrile seizures plus (GEFS+) is an early onset febrile epileptic syndrome with therapeutic responsive (a)febrile seizures continuing later in life. Dravet syndrome (DS) or severe myoclonic epilepsy of infancy has a complex phenotype including febrile generalized or hemiclonic convulsions before the age of 1, followed by intractable myoclonic, complex partial, or absence seizures. Both diseases can result from mutations in the Nav1.1 sodium channel, and initially, seizures are typically triggered by fever. We previously characterized two Nav1.1 mutants-R859H (GEFS+) and R865G (DS)-at room temperature and reported a mixture of biophysical gating defects that could not easily predict the phenotype presentation as either GEFS+ or DS. In this study, we extend the characterization of Nav1.1 wild-type, R859H, and R865G channels to physiological (37°C) and febrile (40°C) temperatures. At physiological temperature, a variety of biophysical defects were detected in both mutants, including a hyperpolarized shift in the voltage dependence of activation and a delayed recovery from fast and slow inactivation. Interestingly, at 40°C we also detected additional gating defects for both R859H and R865G mutants. The GEFS+ mutant R859H showed a loss of function in the voltage dependence of inactivation and an increased channel use-dependency at 40°C with no reduction in peak current density. The DS mutant R865G exhibited reduced peak sodium currents, enhanced entry into slow inactivation, and increased use-dependency at 40°C. Our results suggest that fever-induced temperatures exacerbate the gating defects of R859H or R865G mutants and may predispose mutation carriers to febrile seizures.
- Published
- 2013
- Full Text
- View/download PDF
17. Efficient and specific cardiac IK₁ inhibition by a new pentamidine analogue.
- Author
-
Takanari H, Nalos L, Stary-Weinzinger A, de Git KC, Varkevisser R, Linder T, Houtman MJ, Peschar M, de Boer TP, Tidwell RR, Rook MB, Vos MA, and van der Heyden MA
- Subjects
- Action Potentials, Animals, Dogs, Dose-Response Relationship, Drug, HEK293 Cells, Humans, Kinetics, Mice, Molecular Docking Simulation, Molecular Structure, Myocytes, Cardiac metabolism, Pentamidine analogs & derivatives, Pentamidine chemistry, Potassium metabolism, Potassium Channel Blockers chemistry, Potassium Channels, Inwardly Rectifying genetics, Potassium Channels, Inwardly Rectifying metabolism, Protein Transport, Structure-Activity Relationship, Transfection, Myocytes, Cardiac drug effects, Pentamidine pharmacology, Potassium Channel Blockers pharmacology, Potassium Channels, Inwardly Rectifying antagonists & inhibitors
- Abstract
Aims: In excitable cells, KIR2.x ion-channel-carried inward rectifier current (IK₁) is thought to set the negative and stable resting membrane potential, and contributes to action potential repolarization. Loss- or gain-of-function mutations correlate with cardiac arrhythmias and pathological remodelling affects normal KIR2.x protein levels. No specific IK1 inhibitor is currently available for in vivo use, which severely hampers studies on the precise role of IK1 in normal cardiac physiology and pathophysiology. The diamine antiprotozoal drug pentamidine (P) acutely inhibits IK₁ by plugging the cytoplasmic pore region of the channel. We aim to develop more efficient and specific IK₁ inhibitors based on the P structure., Methods and Results: We analysed seven pentamidine analogues (PA-1 to PA-7) for IK₁ blocking potency at 200 nM using inside-out patches from KIR2.1 expressing HEK-293 cells. PA-6 showed the highest potency and was tested further. PA-6 blocked KIR2.x currents of human and mouse with low IC₅₀ values (12-15 nM). Modelling indicated that PA-6 had less electrostatic but more lipophilic interactions with the cytoplasmic channel pore than P, resulting in a higher channel affinity for PA-6 (ΔG -44.1 kJ/Mol) than for P (ΔG -31.7 kJ/Mol). The involvement of acidic amino acid residues E224 and E299 in drug-channel interaction was confirmed experimentally. PA-6 did not affect INav1.5, ICa-L, IKv4.3, IKv11.1, and IKv7.1/minK currents at 200 nM. PA-6 inhibited the inward (50 nM 40%; 100 nM 59%; 200 nM 77%) and outward (50 nM 40%; 100 nM 76%; 200 nM 100%) components of IK₁ in isolated canine adult-ventricular cardiomyocytes (CMs). PA-6 prolonged action potential duration of CMs by 8 (n = 9), 26 (n = 5), and 34% (n = 11) at 50, 100, and 200 nM, respectively. Unlike P, PA-6 had no effect on KIR2.1 channel expression at concentrations from 0.1 to 3 μM. However, PA-6 at 10 μM increased KIR2.1 expression levels. Also, PA-6 did not affect the maturation of hERG, except when applied at 10 μM., Conclusion: PA-6 has higher efficiency and specificity to KIR2.x-mediated current than P, lengthens action potential duration, and does not affect channel trafficking at concentrations relevant for complete IK₁ block.
- Published
- 2013
- Full Text
- View/download PDF
18. Dominant missense mutations in ABCC9 cause Cantú syndrome.
- Author
-
Harakalova M, van Harssel JJ, Terhal PA, van Lieshout S, Duran K, Renkens I, Amor DJ, Wilson LC, Kirk EP, Turner CL, Shears D, Garcia-Minaur S, Lees MM, Ross A, Venselaar H, Vriend G, Takanari H, Rook MB, van der Heyden MA, Asselbergs FW, Breur HM, Swinkels ME, Scurr IJ, Smithson SF, Knoers NV, van der Smagt JJ, Nijman IJ, Kloosterman WP, van Haelst MM, van Haaften G, and Cuppen E
- Subjects
- Adult, Cell Line, Transformed, Child, Child, Preschool, Exome, Female, Genetic Predisposition to Disease, HEK293 Cells, Humans, Infant, Infant, Newborn, KATP Channels genetics, Male, Protein Structure, Tertiary genetics, Sulfonylurea Receptors, Young Adult, ATP-Binding Cassette Transporters genetics, Cardiomegaly genetics, Genetic Diseases, X-Linked genetics, Hypertrichosis genetics, Mutation, Missense, Osteochondrodysplasias genetics, Potassium Channels, Inwardly Rectifying genetics, Receptors, Drug genetics
- Abstract
Cantú syndrome is characterized by congenital hypertrichosis, distinctive facial features, osteochondrodysplasia and cardiac defects. By using family-based exome sequencing, we identified a de novo mutation in ABCC9. Subsequently, we discovered novel dominant missense mutations in ABCC9 in 14 of the 16 individuals with Cantú syndrome examined. The ABCC9 protein is part of an ATP-dependent potassium (K(ATP)) channel that couples the metabolic state of a cell with its electrical activity. All mutations altered amino acids in or close to the transmembrane domains of ABCC9. Using electrophysiological measurements, we show that mutations in ABCC9 reduce the ATP-mediated potassium channel inhibition, resulting in channel opening. Moreover, similarities between the phenotype of individuals with Cantú syndrome and side effects from the K(ATP) channel agonist minoxidil indicate that the mutations in ABCC9 result in channel opening. Given the availability of ABCC9 antagonists, our findings may have direct implications for the treatment of individuals with Cantú syndrome.
- Published
- 2012
- Full Text
- View/download PDF
19. Biology of cardiac sodium channel Nav1.5 expression.
- Author
-
Rook MB, Evers MM, Vos MA, and Bierhuizen MF
- Subjects
- Arrhythmias, Cardiac genetics, Arrhythmias, Cardiac metabolism, Calcium metabolism, Electrophysiological Phenomena, GTP-Binding Protein alpha Subunits, Gs metabolism, Gene Expression Regulation, Genetic Variation, Humans, Models, Molecular, Mutation, NAV1.5 Voltage-Gated Sodium Channel, Phosphorylation, Protein Processing, Post-Translational, Protein Subunits, Sodium Channels chemistry, Myocardium metabolism, Sodium Channels genetics, Sodium Channels metabolism
- Abstract
Na(v)1.5, the pore forming α-subunit of the voltage-dependent cardiac Na(+) channel, is an integral membrane protein involved in the initiation and conduction of action potentials. Mutations in the gene-encoding Na(v)1.5, SCN5A, have been associated with a variety of arrhythmic disorders, including long QT, Brugada, and sick sinus syndromes as well as progressive cardiac conduction defect and atrial standstill. Moreover, alterations in the Na(v)1.5 expression level and/or sodium current density have been frequently noticed in acquired cardiac disorders, such as heart failure. The molecular mechanisms underlying these alterations are poorly understood, but are considered essential for conception of arrhythmogenesis and the development of therapeutic strategies for prevention or treatment of arrhythmias. The unravelling of such mechanisms requires critical molecular insight into the biology of Na(v)1.5 expression and function. Therefore, the aim of this review is to provide an up-to-date account of molecular determinants of normal Na(v)1.5 expression and function. The parts of the Na(v)1.5 life cycle that are discussed include (i) regulatory aspects of the SCN5A gene and transcript structure, (ii) the nature, molecular determinants, and functional consequences of Na(v)1.5 post-translational modifications, and (iii) the role of Na(v)1.5 interacting proteins in cellular trafficking. The reviewed studies have provided valuable information on how the Na(v)1.5 expression level, localization, and biophysical properties are regulated, but also revealed that our understanding of the underlying mechanisms is still limited.
- Published
- 2012
- Full Text
- View/download PDF
20. Nav 1.1 dysfunction in genetic epilepsy with febrile seizures-plus or Dravet syndrome.
- Author
-
Volkers L, Kahlig KM, Verbeek NE, Das JH, van Kempen MJ, Stroink H, Augustijn P, van Nieuwenhuizen O, Lindhout D, George AL Jr, Koeleman BP, and Rook MB
- Subjects
- Adult, Child, Child, Preschool, Female, Humans, Infant, Ion Channel Gating genetics, Male, Mutation, Missense, NAV1.1 Voltage-Gated Sodium Channel, Nerve Tissue Proteins chemistry, Patch-Clamp Techniques, Sodium Channels chemistry, Syndrome, Epilepsy genetics, Epilepsy physiopathology, Nerve Tissue Proteins genetics, Nerve Tissue Proteins metabolism, Seizures, Febrile genetics, Seizures, Febrile physiopathology, Sodium Channels genetics, Sodium Channels metabolism
- Abstract
Relatively few SCN1A mutations associated with genetic epilepsy with febrile seizures-plus (GEFS+) and Dravet syndrome (DS) have been functionally characterized. In contrast to GEFS+, many mutations detected in DS patients are predicted to have complete loss of function. However, functional consequences are not immediately apparent for DS missense mutations. Therefore, we performed a biophysical analysis of three SCN1A missense mutations (R865G, R946C and R946H) we detected in six patients with DS. Furthermore, we compared the functionality of the R865G DS mutation with that of a R859H mutation detected in a GEFS+ patient; the two mutations reside in the same voltage sensor domain of Na(v) 1.1. The four mutations were co-expressed with β1 and β2 subunits in tsA201 cells, and characterized using the whole-cell patch clamp technique. The two DS mutations, R946C and R946H, were nonfunctional. However, the novel voltage sensor mutants R859H (GEFS+) and R865G (DS) produced sodium current densities similar to those in wild-type channels. Both mutants had negative shifts in the voltage dependence of activation, slower recovery from inactivation, and increased persistent current. Only the GEFS+ mutant exhibited a loss of function in voltage-dependent channel availability. Our results suggest that the R859H mutation causes GEFS+ by a mixture of biophysical defects in Na(v) 1.1 gating. Interestingly, while loss of Na(v) 1.1 function is common in DS, the R865G mutation may cause DS by overall gain-of-function defects., (© 2011 The Authors. European Journal of Neuroscience © 2011 Federation of European Neuroscience Societies and Blackwell Publishing Ltd.)
- Published
- 2011
- Full Text
- View/download PDF
21. Inhibition of lysosomal degradation rescues pentamidine-mediated decreases of K(IR)2.1 ion channel expression but not that of K(v)11.1.
- Author
-
Nalos L, de Boer TP, Houtman MJ, Rook MB, Vos MA, and van der Heyden MA
- Subjects
- Animals, Blotting, Western, Cells, Cultured, Dogs, ERG1 Potassium Channel, Ether-A-Go-Go Potassium Channels genetics, Gene Expression Regulation drug effects, HEK293 Cells, Humans, Membrane Potentials drug effects, Microscopy, Confocal, Patch-Clamp Techniques, Potassium Channels, Inwardly Rectifying genetics, RNA, Messenger metabolism, Reverse Transcriptase Polymerase Chain Reaction, Antiprotozoal Agents pharmacology, Ether-A-Go-Go Potassium Channels metabolism, Lysosomes metabolism, Pentamidine pharmacology, Potassium Channels, Inwardly Rectifying metabolism
- Abstract
The antiprotozoal drug pentamidine inhibits two types of cardiac rectifier potassium currents, which can precipitate life-threatening arrhythmias. Here, we use pentamidine as a tool to investigate whether a single drug affects trafficking of two structurally different potassium channels by identical or different mechanisms, and whether the adverse drug effect can be suppressed in a channel specific fashion. Whole cell patch clamp, Western blot, real time PCR, and confocal laser scanning microscopy were used to determine potassium current density, ion channel protein levels, mRNA expression levels, and subcellular localization, respectively. We demonstrate that pentamidine inhibits delayed (I(Kr)) and inward (I(K1)) rectifier currents in cultured adult canine cardiomyocytes. In HEK293 cells, pentamidine inhibits functional K(v)11.1 channels, responsible for I(Kr), by interfering at the level of full glycosylation, yielding less mature form of K(v)11.1 at the plasma membrane. In contrast, total K(IR)2.1 expression levels, underlying I(K1), are strongly decreased, which cannot be explained from mRNA expression levels. No changes in molecular size of K(IR)2.1 protein were observed, excluding interference in overt glycosylation. Remaining K(IR)2.1 protein is mainly expressed at the plasma membrane. Inhibition of lysosomal protein degradation is able to partially rescue K(IR)2.1 levels, but not those of K(v)11.1. We conclude that 1) a single drug can interfere in cardiac potassium channel trafficking in a subtype specific mode and 2) adverse drug effects can be corrected in a channel specific manner., (2010 Elsevier B.V. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
22. Functional analysis of novel KCNQ2 mutations found in patients with Benign Familial Neonatal Convulsions.
- Author
-
Volkers L, Rook MB, Das JH, Verbeek NE, Groenewegen WA, van Kempen MJ, Lindhout D, and Koeleman BP
- Subjects
- Cell Line, Cell Membrane physiology, Endoplasmic Reticulum metabolism, Family, Female, Fluorescent Antibody Technique, Green Fluorescent Proteins, Humans, KCNQ3 Potassium Channel metabolism, Membrane Potentials physiology, Microscopy, Confocal, Microscopy, Fluorescence, Mutation, Mutation, Missense, Patch-Clamp Techniques, Potassium metabolism, Time Factors, Transfection, Epilepsy, Benign Neonatal genetics, KCNQ2 Potassium Channel genetics, KCNQ2 Potassium Channel metabolism
- Abstract
Benign Familial Neonatal Convulsions (BFNC) are a rare epilepsy disorder with an autosomal-dominant inheritance. It is linked to mutations in the potassium channel genes KCNQ2 and KCNQ3. These encode for Kv7.2 and Kv7.3 potassium ion channels, which produce an M-current that regulates the potential firing action in neurons through modulation of the membrane potential. We report on the biophysical and biochemical properties of V589X, T359K and P410fs12X mutant-KCNQ2 ion channels that were detected in three BFNC families. Mutant KCNQ2 cDNAs were co-expressed with WT-KCNQ2 and KCNQ3 cDNAs in HEK293 cells to mimic heterozygous expression of the KCNQ2 mutations in BFNC patients. The resulting potassium currents were measured using patch-clamp techniques and showed an approximately 75% reduction in current and a depolarized shift in the voltage dependence of activation. Furthermore, the time-constant of activation of M-currents in cells expressing T359K and P410fs12X was slower compared to cells expressing only wild-type proteins. Immunofluorescent labeling of HEK293 cells stably expressing GFP-tagged KCNQ2-WT or mutant alpha-subunits indicated cell surface expression of WT, V589X and T359K mutants, suggesting a loss-of-function, while P410fs12X was predominantly retained in the ER and sub-cellular compartments outside the ER suggesting an effectively haplo-insufficient effect.
- Published
- 2009
- Full Text
- View/download PDF
23. Exploring chemical substructures essential for HERG k(+) channel blockade by synthesis and biological evaluation of dofetilide analogues.
- Author
-
Guo D, Klaasse E, de Vries H, Brussee J, Nalos L, Rook MB, Vos MA, van der Heyden MA, and Ijzerman AP
- Subjects
- Cell Line, ERG1 Potassium Channel, Humans, Molecular Structure, Phenethylamines chemical synthesis, Phenethylamines pharmacology, Potassium Channel Blockers chemical synthesis, Potassium Channel Blockers pharmacology, Structure-Activity Relationship, Sulfonamides chemical synthesis, Sulfonamides pharmacology, Drug Design, Ether-A-Go-Go Potassium Channels antagonists & inhibitors, Phenethylamines chemistry, Potassium Channel Blockers chemistry, Sulfonamides chemistry
- Abstract
In this study we followed a new approach to analyze molecular substructures required for hERG channel blockade. We designed and synthesized 40 analogues of dofetilide (1), a potent hERG potassium channel blocker, and established structure-activity relationships (SAR) for their interaction with this important cardiotoxicity-related off-target. Structural modifications to dofetilide were made by diversifying the substituents on the phenyl rings and the protonated nitrogen and by varying the carbon chain length. The analogues were evaluated in a radioligand binding assay and SAR data were derived with the aim to specify structural features that give rise to hERG toxicity.
- Published
- 2009
- Full Text
- View/download PDF
24. Fever-induced QTc prolongation and ventricular arrhythmias in individuals with type 2 congenital long QT syndrome.
- Author
-
Amin AS, Herfst LJ, Delisle BP, Klemens CA, Rook MB, Bezzina CR, Underkofler HA, Holzem KM, Ruijter JM, Tan HL, January CT, and Wilde AA
- Subjects
- Adult, Aged, Arrhythmias, Cardiac etiology, Body Temperature, Cell Line, ERG1 Potassium Channel, Electrocardiography, Electrophysiology, Ether-A-Go-Go Potassium Channels metabolism, Heterozygote, Humans, Ion Channel Gating physiology, Long QT Syndrome etiology, Long QT Syndrome genetics, Male, Mutation, Missense, Protein Transport, Romano-Ward Syndrome diagnosis, Romano-Ward Syndrome physiopathology, Temperature, Transfection, Arrhythmias, Cardiac physiopathology, Ether-A-Go-Go Potassium Channels genetics, Fever complications, Long QT Syndrome physiopathology
- Abstract
Type 2 congenital long QT syndrome (LQT-2) is linked to mutations in the human ether a-go-go-related gene (HERG) and is characterized by rate-corrected QT interval (QTc) prolongation, ventricular arrhythmias, syncope, and sudden death. Recognized triggers of these cardiac events include emotional and acoustic stimuli. Here we investigated the repeated occurrence of fever-induced polymorphic ventricular tachycardia and ventricular fibrillation in 2 LQT-2 patients with A558P missense mutation in HERG. ECG analysis showed increased QTc with fever in both patients. WT, A558P, and WT+A558P HERG were expressed heterologously in HEK293 cells and were studied using biochemical and electrophysiological techniques. A558P proteins showed a trafficking-deficient phenotype. WT+A558P coexpression caused a dominant-negative effect, selectively accelerated the rate of channel inactivation, and reduced the temperature-dependent increase in the WT current. Thus, the WT+A558P current did not increase to the same extent as the WT current, leading to larger current density differences at higher temperatures. A similar temperature-dependent phenotype was seen for coexpression of the trafficking-deficient LQT-2 F640V mutation. We postulate that the weak increase in the HERG current density in WT-mutant coassembled channels contributes to the development of QTc prolongation and arrhythmias at febrile temperatures and suggest that fever is a potential trigger of life-threatening arrhythmias in LQT-2 patients.
- Published
- 2008
- Full Text
- View/download PDF
25. Physiologic function of I(K1) requires a combination of Kir2 isoforms.
- Author
-
Rook MB
- Subjects
- Animals, Electrophysiology, Heart Ventricles cytology, Hydrogen-Ion Concentration, Protein Isoforms, Myocytes, Cardiac physiology, Potassium Channels, Inwardly Rectifying physiology
- Published
- 2007
- Full Text
- View/download PDF
26. Connexin43 repression following epithelium-to-mesenchyme transition in embryonal carcinoma cells requires Snail1 transcription factor.
- Author
-
de Boer TP, van Veen TA, Bierhuizen MF, Kok B, Rook MB, Boonen KJ, Vos MA, Doevendans PA, de Bakker JM, and van der Heyden MA
- Subjects
- Animals, Carcinoma, Embryonal, Cell Line, Tumor, Connexin 43 metabolism, Down-Regulation, Embryonic Stem Cells cytology, Fluorescent Antibody Technique, Gene Silencing, Mesoderm cytology, Mice, Promoter Regions, Genetic, RNA, Messenger metabolism, RNA, Small Interfering metabolism, Snail Family Transcription Factors, Transcription Factors genetics, Transfection, Connexin 43 genetics, Embryonic Stem Cells metabolism, Epithelium metabolism, Mesoderm metabolism, Transcription Factors metabolism
- Abstract
Embryonic stem (ES) cells and embryonal carcinoma (EC) cells express high amounts of functional connexin43 (Cx43). During mesoderm formation and subsequent cardiac differentiation, Cx43 is initially down-regulated but is up-regulated again as the emerging cardiomyocytes mature. In this study, we investigated the regulation of Cx43 expression during early phases of differentiation in F9 and P19 EC cells. We found a striking inverse correlation between the expression of Cx43 and that of the transcriptional repressor Snail1. No clear relationship was found with Smad-interacting-protein1 (SIP1), another transcription factor inducing epithelium-to-mesenchyme transition (EMT). Promoter-reporter assays indicated Cx43 repression at the promoter level by ectopically expressed Snail1. To establish whether the Cx43 down-regulation depends on endogenous Snail1, MES-1 cells, differentiated derivatives of P19 EC, were stably transfected by an siRNA construct silencing Snail1 expression. This resulted in a mesenchyme-to-epithelium transition, which was accompanied by increased levels of Cx43 mRNA and protein and enhanced metabolic and electrical coupling. We conclude that Snail1-mediated EMT results in a Cx43 repression.
- Published
- 2007
- Full Text
- View/download PDF
27. Cloning, embryonic expression, and functional characterization of two novel connexins from Xenopus laevis.
- Author
-
de Boer TP, Kok B, Roël G, van Veen TA, Destrée OH, Rook MB, Vos MA, de Bakker JM, and van der Heyden MA
- Subjects
- Amino Acid Sequence, Animals, COS Cells, Chlorocebus aethiops, Cloning, Molecular, Connexin 26, Connexin 30, Connexins genetics, Connexins metabolism, Gap Junctions, Gene Expression Regulation, Developmental, Humans, Mesoderm metabolism, Molecular Sequence Data, Sequence Homology, Amino Acid, Xenopus Proteins metabolism, Gap Junction beta-1 Protein, Connexins chemistry, Xenopus Proteins chemistry, Xenopus Proteins genetics, Xenopus laevis metabolism
- Abstract
Vertebrate gap junctions are constituted of connexin (Cx) proteins. In Xenopus laevis, only seven different Cxs have been described so far. Here, we identify two new Cxs from X. laevis. Cx28.6 displays > 60% amino acid identity with human Cx25, Cx29 displays strong homology with mouse Cx26 and Cx30. Cx29 is expressed throughout embryonic development. Cx28.6 mRNA is only transiently found from stage 22 to 26 of development. While no Cx28.6 expression could be detected by whole mount in situ hybridization, expression of Cx29 was found in the developing endoderm, lateral mesoderm, liver anlage, pronephros, and proctodeum. Ectopic expression of Cx28.6 failed to produce functional gap-junctions. In contrast, ectopic expression of full-length Cx29 in HEK293 and COS-7 cells resulted in the formation of gap junction-like structures at the cell-cell interfaces. Ectopic expression of Cx29 in communication deficient N2A cell pairs led to functional electrical coupling.
- Published
- 2006
- Full Text
- View/download PDF
28. Dual role of calbindin-D28K in vesicular catecholamine release from mouse chromaffin cells.
- Author
-
Westerink RH, Rook MB, Beekwilder JP, and Wadman WJ
- Subjects
- Adrenal Medulla ultrastructure, Animals, Calbindin 1, Calbindins, Calcium metabolism, Cells, Cultured, Chromaffin Cells ultrastructure, Cytoplasmic Vesicles ultrastructure, Exocytosis genetics, Female, Immunohistochemistry, Intracellular Fluid metabolism, Male, Membrane Fusion genetics, Membrane Potentials genetics, Mice, Mice, Inbred C57BL, Mice, Knockout, Microscopy, Confocal, S100 Calcium Binding Protein G genetics, Synaptic Transmission genetics, Synaptic Vesicles metabolism, Synaptic Vesicles ultrastructure, Adrenal Medulla metabolism, Calcium Signaling physiology, Catecholamines metabolism, Chromaffin Cells metabolism, Cytoplasmic Vesicles metabolism, S100 Calcium Binding Protein G physiology
- Abstract
Calbindin-D(28K) is suggested to play a postsynaptic role in neurotransmission and in the regulation of the intracellular Ca(2+) concentration. However, it is still unclear whether calbindin-D(28K) has a role in the regulation of exocytosis, either as Ca(2+) buffer or as Ca(2+) sensor. Amperometric recordings of catecholamine exocytosis from wild-type and calbindin-D(28K) knockout mouse chromaffin cells reveal a strong reduction in the number of released vesicles, as well as in the amount of neurotransmitter released per fusion event in knockout cells. However, Ca(2+) current recordings and Ca(2+) imaging experiments, including video-rate confocal laser scanning microscopy, revealed that the intracellular Ca(2+) dynamics are remarkably similar in wild-type and knockout cells. The combined results demonstrate that calbindin-D(28K) plays an important and dual role in exocytosis, affecting both release frequency and quantal size, apparently without strong effects on intracellular Ca(2+) dynamics. Consequently, the possibility that calbindin-D(28K) functions not only as a Ca(2+) buffer but also as a modulator of vesicular catecholamine release is discussed.
- Published
- 2006
- Full Text
- View/download PDF
29. Pro-arrhythmogenic potential of immature cardiomyocytes is triggered by low coupling and cluster size.
- Author
-
de Boer TP, van der Heyden MA, Rook MB, Wilders R, Broekstra R, Kok B, Vos MA, de Bakker JM, and van Veen TA
- Subjects
- Actinin metabolism, Animals, Cadherins metabolism, Cell Communication, Cell Transplantation, Dogs, Electrophysiology, Heart Ventricles, Immunohistochemistry, Rats, Reverse Transcriptase Polymerase Chain Reaction, Arrhythmias, Cardiac physiopathology, Computer Simulation, Models, Cardiovascular, Myocytes, Cardiac physiology
- Abstract
Objective: Cell transplantation strategies to regenerate compromised myocardium take advance of in vitro generated cardiomyocytes. Common in those immature myocytes is spontaneous impulse formation and a restricted ability to establish proper electrical interaction. Spontaneous impulse formation and impaired cell-to-cell coupling have been shown to be arrhythmogenic. To investigate whether these features harbour a pro-arrhyhmogenic potential for cell transplantation, a co-culture of spontaneously active neonatal rat cardiomyocytes (NRC) and quiescent adult dog cardiomyocytes (ADC) was used., Methods: ADCs and NRCs were isolated and cultured on laminin-coated substrates. Connexin43, N-cadherin and alpha-actinin expression was evaluated with immunohistochemistry. Intercellular coupling was measured in cell pairs using the dual voltage clamp technique and fluorescent dye injection., Results: One day after isolation, NRCs were beating spontaneously, while ADCs remained quiescent in monoculture. ADC resting membrane potential was -80.3+/-0.2 mV (mean+/-SEM, N=24) and did not change significantly over time. NRCs had a maximal diastolic potential of -65.0+/-2.8 mV (N=4). After one day of co-culture, pseudopodia-like extensions developed at the former intercalated discs of ADCs, contacting the NRCs. Only ADCs that contacted three or more NRCs started to beat in synchrony. Expression of connexin43 and N-cadherin indicated presence of electrical and mechanical junctions at the interface between the two cell-types. Transfer of Lucifer Yellow demonstrated junctional permeability between ADCs and NRCs. Junctional conductance between ADC-ADC (31.9+/-5.1 nS, N=10) and NRC-NRC (35.0+/-9.6 nS, N=6) pairs was significantly higher compared to ADC-NRC pairs (9.7+/-2.9 nS, N=8). Gap-junctional blockade with halothane reversibly abolished NRC-triggered beating of ADCs. Computer simulations demonstrated that within a delicate 'window' of gap junctional conductance small clusters of spontaneously active cells are able to induce triggered activity in quiescent mature myocytes but also in a two-dimensional sheet of ventricular cells., Conclusion: Spontaneously active immature cardiomyocytes are able to trigger mature cardiomyocytes depending on their level of electrical coupling and the amount of coupled immature myocytes.
- Published
- 2006
- Full Text
- View/download PDF
30. Cloning and functional characterization of a novel connexin expressed in somites of Xenopus laevis.
- Author
-
De Boer TP, Kok B, Neuteboom KI, Spieker N, De Graaf J, Destrée OH, Rook MB, Van Veen TA, Jongsma HJ, Vos MA, De Bakker JM, and Van Der Heyden MA
- Subjects
- Aging physiology, Amino Acid Sequence, Animals, Cloning, Molecular, Computational Biology, Connexins chemistry, Connexins genetics, Electrophysiology, Molecular Sequence Data, Patch-Clamp Techniques, Phylogeny, Sequence Alignment, Somites chemistry, Xenopus Proteins chemistry, Xenopus Proteins genetics, Xenopus laevis genetics, Xenopus laevis growth & development, Connexins metabolism, Gene Expression Regulation, Developmental, Somites metabolism, Xenopus Proteins metabolism, Xenopus laevis embryology, Xenopus laevis metabolism
- Abstract
Connexin-containing gap junctions play an essential role in vertebrate development. More than 20 connexin isoforms have been identified in mammals. However, the number identified in Xenopus trails with only six isoforms described. Here, identification of a new connexin isoform from Xenopus laevis is described. Connexin40.4 was found by screening expressed sequence tag databases and carrying out polymerase chain reaction on genomic DNA. This new connexin has limited amino acid identity with mammalian (<50%) connexins, but conservation is higher (approximately 62%) with fish. During Xenopus laevis development, connexin40.4 was first expressed after the mid-blastula transition. There was prominent expression in the presomitic paraxial mesoderm and later in the developing somites. In adult frogs, expression was detected in kidney and stomach as well as in brain, heart, and skeletal muscle. Ectopic expression of connexin40.4 in HEK293 cells, resulted in formation of gap junction like structures at the cell interfaces. Similar ectopic expression in neural N2A cells resulted in functional electrical coupling, displaying mild, asymmetric voltage dependence. We thus cloned a novel connexin from Xenopus laevis, strongly expressed in developing somites, with no apparent orthologue in mammals.
- Published
- 2005
- Full Text
- View/download PDF
31. Trafficking and functional expression of cardiac Na+ channels.
- Author
-
Herfst LJ, Rook MB, and Jongsma HJ
- Subjects
- Action Potentials physiology, Animals, Heart Conduction System physiology, Heart Diseases genetics, Humans, Membrane Proteins metabolism, Mutation, NAV1.5 Voltage-Gated Sodium Channel, Protein Transport, RNA, Messenger genetics, Sodium Channels genetics, Heart physiology, Sodium Channels physiology
- Abstract
Voltage-gated Na(+)channels are essential for the amplitude and upstroke velocity of the cardiac action potential, which are important determinants for impulse propagation and impulse conduction velocity of throughout the working myocardium. Mutations in the major cardiac Na(+)channel gene SCN5A have been implicated in rare, familial forms of cardiac arrhythmias, namely LQT3, Brugada syndrome, progressive cardiac conduction disorder and sudden infant death syndrome. It is increasingly recognized that such mutations--apart from changing channel gating characteristics--may also be related to changes in channel protein trafficking and expression. Regulation of ion channel protein expression depends on a fine-tuned balance among various processes, such as gene transcription, RNA processing, protein synthesis, assembly and post-translational modification, the transport to the cell surface, the anchoring to the cytoskeleton, and regulation of endocytosis and controlled degradation of the protein. During the last decade, interest in factors that control the expression level of ion channels and mechanisms that are involved in targeting of channel proteins to specific sub-cellular and membrane domains is increasing. This review focuses on the current knowledge of mechanisms of cardiac Na(+) channel protein trafficking and expression in cardiomyocytes and its relation to Na(+)-channelopathies.
- Published
- 2004
- Full Text
- View/download PDF
32. A P19Cl6 GFP reporter line to quantify cardiomyocyte differentiation of stem cells.
- Author
-
Moore JC, Spijker R, Martens AC, de Boer T, Rook MB, van der Heyden MA, Tertoolen LG, and Mummery CL
- Subjects
- Animals, Biomarkers analysis, Cell Line, Clone Cells metabolism, Electrophysiology, Green Fluorescent Proteins, Humans, Luminescent Proteins genetics, Molecular Sequence Data, Myocytes, Cardiac metabolism, Promoter Regions, Genetic genetics, Rats, Stem Cells metabolism, Tropomyosin analysis, Cell Differentiation, Genes, Reporter genetics, Luminescent Proteins metabolism, Myocytes, Cardiac cytology, Stem Cells cytology
- Abstract
The clinical application of stem cell therapies is still limited by the ability to produce defined, differentiated cell populations in large numbers in culture. High throughput screens to identify factors which enhance differentiation to particular lineages and promote expansion of precursors in culture are dependent on the development of sensitive and reproducible assays for screening. Here we describe a bioassay to identify factors with cardiomyogenic activity which enhance the yield of cardiomyocytes from undifferentiated stem cells. The assay is based on a Green Fluorescent Protein (GFP) reporter under the transcriptional control of the 250 bp MLC-2v promoter expressed in pluripotent P19 embryonal carcinoma cells. We show that reporter expression is limited to developing cardiomyocytes and can be used to determine quantitatively the number of ventricular cardiomyocytes formed in cultures under inducing or non-inducing conditions. This assay differs from all others described previously in that it has an easily quantifiable readout, there is negligible background differentiation in the absence of exogenous cardiogenic factors and it is carried out feeder cell-free. Thus, it is entirely independent of competing differentiation inhibitory factors, such as leukemia inhibitory factor. Patch clamp electrophysiology of the GFP-positive cells confirmed their functional ventricular phenotype and indicated that selection on the basis of GFP would provide cells suitable for transplantation.
- Published
- 2004
- Full Text
- View/download PDF
33. Arabidopsis sterol endocytosis involves actin-mediated trafficking via ARA6-positive early endosomes.
- Author
-
Grebe M, Xu J, Möbius W, Ueda T, Nakano A, Geuze HJ, Rook MB, and Scheres B
- Subjects
- Arabidopsis drug effects, Arabidopsis genetics, Arabidopsis Proteins genetics, Base Sequence, Biological Transport, Active drug effects, Brefeldin A pharmacology, DNA, Plant genetics, Endocytosis drug effects, Endosomes metabolism, GTP Phosphohydrolases genetics, Green Fluorescent Proteins, Luminescent Proteins genetics, Luminescent Proteins metabolism, Microscopy, Electron, Models, Biological, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Subcellular Fractions metabolism, rab GTP-Binding Proteins genetics, Actins metabolism, Arabidopsis metabolism, Arabidopsis Proteins metabolism, GTP Phosphohydrolases metabolism, Sterols metabolism, rab GTP-Binding Proteins metabolism
- Abstract
Background: In contrast to the intense attention devoted to research on intracellular sterol trafficking in animal cells, knowledge about sterol transport in plant cells remains limited, and virtually nothing is known about plant endocytic sterol trafficking. Similar to animals, biosynthetic sterol transport occurs from the endoplasmic reticulum (ER) via the Golgi apparatus to the plasma membrane. The vesicle trafficking inhibitor brefeldin A (BFA) has been suggested to disrupt biosynthetic sterol transport at the Golgi level., Results: Here, we report on early endocytic sterol trafficking in Arabidopsis root epidermal cells by introducing filipin as a tool for fluorescent sterol detection. Sterols can be internalized from the plasma membrane and localize to endosomes positive for the early endosomal Rab5 GTPase homolog ARA6 fused to green fluorescent protein (GFP) (ARA6-GFP). Early endocytic sterol transport is actin dependent and highly BFA sensitive. BFA causes coaccumulation of sterols, endocytic markers like ARA6-GFP, and PIN2, a polarly localized presumptive auxin transport protein, in early endosome agglomerations that can be distinguished from ER and Golgi. Sterol accumulation in such aggregates is enhanced in actin2 mutants, and the actin-depolymerizing drug cytochalasin D inhibits sterol redistribution from endosome aggregations., Conclusions: Early endocytic sterol trafficking involves transport via ARA6-positive early endosomes that, in contrast to animal cells, is actin dependent. Our results reveal sterol-enriched early endosomes as targets for BFA interference in plants. Early endocytic sterol trafficking and recycling of polar PIN2 protein share a common pathway, suggesting a connection between plant endocytic sterol transport and polar sorting events.
- Published
- 2003
- Full Text
- View/download PDF
34. P19 embryonal carcinoma cells: a suitable model system for cardiac electrophysiological differentiation at the molecular and functional level.
- Author
-
van der Heyden MA, van Kempen MJ, Tsuji Y, Rook MB, Jongsma HJ, and Opthof T
- Subjects
- Actinin analysis, Action Potentials physiology, Animals, Calcium Channels, L-Type genetics, Cell Culture Techniques methods, Cell Differentiation physiology, Connexin 43 analysis, Embryonal Carcinoma Stem Cells, Ion Channels physiology, KCNQ Potassium Channels, Mice, Models, Animal, NAV1.5 Voltage-Gated Sodium Channel, Potassium Channels genetics, Reverse Transcriptase Polymerase Chain Reaction, Sodium Channels genetics, Troponin T analysis, Tubulin genetics, Tumor Cells, Cultured, Carcinoma, Embryonal pathology, Myocytes, Cardiac physiology, Neoplastic Stem Cells pathology, Potassium Channels, Voltage-Gated
- Abstract
Objective: Murine P19 embryonal carcinoma (EC) cells can differentiate into spontaneously beating cardiomyocytes in vitro and have revealed important insight into the early molecular processes of cardiomyocyte differentiation. We assessed the suitability of the P19 cell model for studying cardiac ion channel regulation at the molecular and functional level., Methods: P19 cells were induced to differentiate towards cardiomyocytes. mRNAs for cardiac markers and ion channels were determined by RT-PCR at six timepoints during the differentiation process. Action potentials and individual ion currents were measured by whole cell patch clamp., Results: Ion channel mRNA expression of several channels is temporally regulated during differentiation, while others show little or no regulation. L-type calcium and transient outward channels are expressed from very early on, while sodium and delayed and inward rectifier channels are upregulated at somewhat later stages during differentiation, which mirrors the in vivo murine cardiomyocyte differentiation during embryogenesis. Spontaneous cardiomyocyte action potentials exhibit a low upstroke velocity, which often can be enhanced by hyperpolarizing the cells, hence activating thusfar dormant ion channels to contribute to the action potential upstroke. Action potential duration decreases considerably during the differentiation of spontaneously beating cells. In late stages, non-beating myocytes can be found which only generate action potentials upon electrical stimulation. Their shape is comparable to neonatal/juvenile ventricular mouse myocytes in culture. Finally, we show that P19-derived cardiomyocytes display a very complete set of functional ion channels., Conclusion: P19 cells represent a powerful model to study the regulation of myocardial electrophysiological differentiation at the molecular and functional level.
- Published
- 2003
- Full Text
- View/download PDF
35. Na+ channel mutation leading to loss of function and non-progressive cardiac conduction defects.
- Author
-
Herfst LJ, Potet F, Bezzina CR, Groenewegen WA, Le Marec H, Hoorntje TM, Demolombe S, Baró I, Escande D, Jongsma HJ, Wilde AA, and Rook MB
- Subjects
- Animals, COS Cells, Electrocardiography, Female, Humans, Male, Patch-Clamp Techniques, Pedigree, Point Mutation, Protein Transport genetics, Sodium Channels metabolism, Electrophysiologic Techniques, Cardiac, Myocardium metabolism, Sequence Deletion, Sodium Channels genetics
- Abstract
Background: We previously described a Dutch family in which congenital cardiac conduction disorder has clinically been identified. The ECG of the index patient showed a first-degree AV block associated with extensive ventricular conduction delay. Sequencing of the SCN5A locus coding for the human cardiac Na+ channel revealed a single nucleotide deletion at position 5280, resulting in a frame-shift in the sequence coding for the pore region of domain IV and a premature stop codon at the C-terminus., Methods and Results: Wild type and mutant Na+ channel proteins were expressed in Xenopus laevis oocytes and in mammalian cells. Voltage clamp experiments demonstrated the presence of fast activating and inactivating inward currents in cells expressing the wild type channel alone or in combination with the beta1 subinut (SCN1B). In contrast, cells expressing the mutant channels did not show any activation of inward current with or without the beta1 subunit. Culturing transfected cells at 25 degrees C did not restore the Na+ channel activity of the mutant protein. Transient expression of WT and mutant Na+ channels in the form of GFP fusion proteins in COS-7 cells indicated protein expression in the cytosol. But in contrast to WT channels were not associated with the plasma membrane., Conclusions: The SCN5A/5280delG mutation results in the translation into non-function channel proteins that do not reach the plasma membrane. This could explain the cardiac conduction defects in patients carrying the mutation.
- Published
- 2003
- Full Text
- View/download PDF
36. A novel LQT3 mutation implicates the human cardiac sodium channel domain IVS6 in inactivation kinetics.
- Author
-
Groenewegen WA, Bezzina CR, van Tintelen JP, Hoorntje TM, Mannens MM, Wilde AA, Jongsma HJ, and Rook MB
- Subjects
- Adolescent, Adult, Child, DNA Mutational Analysis, Death, Sudden, Cardiac etiology, Electrocardiography, Female, Humans, Long QT Syndrome physiopathology, Male, NAV1.5 Voltage-Gated Sodium Channel, Pedigree, Sodium Channels physiology, Long QT Syndrome genetics, Mutation, Sodium Channels genetics
- Abstract
Unlabelled: The Long QT3 syndrome is associated with mutations in the cardiac sodium channel gene SCN5A., Objective: The aim of the present study was the identification and functional characterization of a mutation in a family with the long QT3 syndrome., Methods: The human cardiac sodium channel gene SCN5A was screened for mutations by single-stranded conformation polymorphism. The functional consequences of mutant sodium channels were characterized after expressing mutant and wild-type cRNAs in Xenopus oocytes by two-electrode voltage clamp measurements., Results: SCN5A screening revealed an A-->G substitution at codon 1768, close to the C-terminal end of domain IVS6, which changes an isoleucine to a valine. Functional expression of mutant I1768V-channels in Xenopus oocytes showed that the voltage-dependence and slope factors of activation and inactivation were unchanged compared to wild-type channels. No difference in persistent TTX-sensitive current could be detected between wild-type and I1768V channels, a channel feature often increased in LQT3 mutants. However, I1768V mutant channels recovered faster from inactivation (2.4 times) than wild-type channels and displayed less slow inactivation., Conclusions: We postulate that severe destabilization of the inactivated state leads to increased arrhythmogenesis and QT prolongation in I1768V mutation carriers in the absence of a persistent inward sodium current.
- Published
- 2003
- Full Text
- View/download PDF
37. Compound heterozygosity for mutations (W156X and R225W) in SCN5A associated with severe cardiac conduction disturbances and degenerative changes in the conduction system.
- Author
-
Bezzina CR, Rook MB, Groenewegen WA, Herfst LJ, van der Wal AC, Lam J, Jongsma HJ, Wilde AA, and Mannens MM
- Subjects
- Amino Acid Substitution, Animals, Arrhythmias, Cardiac genetics, Cell Line, Child, DNA Mutational Analysis, Electrocardiography, Fatal Outcome, Female, Haplotypes, Heterozygote, Humans, Infant, Infant, Newborn, Infant, Newborn, Diseases physiopathology, Male, Microinjections, NAV1.5 Voltage-Gated Sodium Channel, Oocytes metabolism, Patch-Clamp Techniques, Pedigree, Polymorphism, Genetic, Sodium Channels metabolism, Tachycardia diagnosis, Tachycardia genetics, Tachycardia physiopathology, Xenopus, Arrhythmias, Cardiac diagnosis, Arrhythmias, Cardiac physiopathology, Heart Conduction System physiopathology, Mutation, Sodium Channels genetics
- Abstract
Cardiac conduction defects associate with mutations in SCN5A, the gene encoding the cardiac Na+ channel. In the present study, we characterized a family in which the proband was born in severe distress with irregular wide complex tachycardia. His older sister died at 1 year of age from severe conduction disease with similarly widened QRS-complexes. Mutational analysis of SCN5A in the proband demonstrated compound heterozygosity for a nonsense mutation (W156X), inherited from the father, and a missense mutation (R225W), inherited from the mother. Genotyping on DNA extracted from tissue from the deceased sibling revealed the same SCN5A genotype. Injection of cRNA encoding the W156X mutation in Xenopus oocytes did not produce any current. The R225W substitution neutralizes the third Arg residue within the voltage-sensing segment of domain I. Expression studies showed that this mutation leads to a severe reduction in I(Na) and is also associated with gating changes. Histological examination of the heart from the deceased sibling revealed changes consistent with a dilated type of cardiomyopathy and severe degenerative abnormalities of the specialized conduction system. The occurrence of compound heterozygosity for these two mutations implies that the proband carries solely severely dysfunctional cardiac Na+ channels. This explains his severe phenotype and that of his deceased sister who had been a carrier of the same genotype. The morphological changes within the heart of the deceased sibling may have occurred secondary to the Na+ channel abnormality and contributed to the severity of the disorder in this individual.
- Published
- 2003
- Full Text
- View/download PDF
38. A cardiac sodium channel mutation cosegregates with a rare connexin40 genotype in familial atrial standstill.
- Author
-
Groenewegen WA, Firouzi M, Bezzina CR, Vliex S, van Langen IM, Sandkuijl L, Smits JP, Hulsbeek M, Rook MB, Jongsma HJ, and Wilde AA
- Subjects
- Adolescent, Adult, Aged, Amino Acid Substitution, Animals, Arrhythmias, Cardiac diagnosis, Arrhythmias, Cardiac physiopathology, DNA Mutational Analysis, Dizziness etiology, Electrocardiography, Female, Genotype, Humans, Male, NAV1.5 Voltage-Gated Sodium Channel, Netherlands, Oocytes metabolism, Patch-Clamp Techniques, Pedigree, Phenotype, Polymorphism, Genetic, Sodium Channels metabolism, Syncope etiology, Transfection, Xenopus laevis, Gap Junction alpha-5 Protein, Arrhythmias, Cardiac genetics, Atrial Function genetics, Connexins genetics, Mutation, Sodium Channels genetics
- Abstract
Atrial standstill (AS) is a rare arrhythmia that occasionally appears to be genetically determined. This study investigates the genetic background of this arrhythmogenic disorder in a large family. Forty-four family members were clinically evaluated. One deceased and three living relatives were unambiguously affected by AS. All other relatives appeared unaffected. Candidate gene screening revealed a novel mutation in the cardiac sodium channel gene SCN5A (D1275N) in all three affected living relatives and in five unaffected relatives, and the deceased relative was an obligate carrier. In addition, two closely linked polymorphisms were detected within regulatory regions of the gene for the atrial-specific gap junction protein connexin40 (Cx40) at nucleotides -44 (G-->A) and +71 (A-->G). Eight relatives were homozygous for both polymorphisms, which occurred in only approximately 7% of control subjects, and three of these relatives were affected by AS. The three living AS patients exclusively coinherited both the rare Cx40 genotype and the SCN5A-D1275N mutation. SCN5A-D1275N channels showed a small depolarizing shift in activation compared with wild-type channels. Rare Cx40 genotype reporter gene analysis showed a reduction in reporter gene expression compared with the more common Cx40 genotype. In this study, familial AS was associated with the concurrence of a cardiac sodium channel mutation and rare polymorphisms in the atrial-specific Cx40 gene. We propose that, although the functional effect of each genetic change is relatively benign, the combined effect of genetic changes eventually progresses to total AS.
- Published
- 2003
- Full Text
- View/download PDF
39. Irregular spiking in free calcium concentration in single, human platelets. Regulation by modulation of the inositol trisphosphate receptors.
- Author
-
van Gorp RM, Feijge MA, Vuist WM, Rook MB, and Heemskerk JW
- Subjects
- Alprostadil pharmacology, Cyclic AMP biosynthesis, Humans, Inositol 1,4,5-Trisphosphate biosynthesis, Inositol 1,4,5-Trisphosphate Receptors, Thimerosal pharmacology, Blood Platelets metabolism, Calcium metabolism, Calcium Channels physiology, Receptors, Cytoplasmic and Nuclear physiology
- Abstract
Fluorescence ratio imaging indicates that immobilized, aspirin-treated platelets, loaded with Fura-2, respond to inositol 1,4,5-trisphosphate- (InsP3)-generating agonists such as thrombin by high-frequency, irregular rises in cytosolic [Ca2+]i with spikes that vary in peak level and peak-to-peak interval. This differs from the regular [Ca2+]i oscillations observed in other, larger cells. We found that the thiol-reactive compounds thimerosal (10 microm) and U73122 (10 microm) evoked similar irregular Ca2+ responses in platelets, but in this case in the absence of InsP3 generation. Thrombin-induced spiking was acutely abolished by inhibiting phospholipase C or elevating intracellular cAMP levels, while spiking with sulfhydryl reagents was only partially blocked by cAMP elevation. Confocal laser scanning microscopy using fluo-3-loaded platelets indicated that, with all agonists or conditions, the irregular spikes were almost instantaneously raised in various regions within a single platelet. When using saponin-permeabilized platelets, we found that InsP3-induced Ca2+ release from stores was stimulated by modest Ca2+ concentrations, pointing to a mechanism of InsP3-dependent Ca2+-induced Ca2+ release (CICR). This process was completely inhibitable by heparin. The Ca2+ release by InsP3, but not the CICR sensor, was negatively regulated by cAMP elevation. Thimerosal treatment did not release Ca2+ from intracellular stores, but markedly potentiated the stimulatory effect of InsP3. In contrast, U73122 caused a heparin/cAMP-insensitive Ca2+ leak from stores that differed from those used by InsP3. Taken together, these results demonstrate that InsP3 receptor channels play a crucial role in the irregular, spiking Ca2+ signal of intact platelets, even when induced by agents such as thimerosal or U73122 which do not stimulate InsP3 formation. The irregular Ca2+ release events appear to be subjected to extensive regulation by: (a) InsP3 level, (b) the potentiating effect of elevated Ca2+ on InsP3 action via CICR, (c) InsP3 channel sensitization by sulfhydryl (thimerosal) modification, (d) InsP3 channel-independent Ca2+ leak with U73122, and (e) down-regulation via cAMP elevation. The observation that individual Ca2+ peaks were generated in various parts of a platelet at similar intervals and amplitudes points to effective cooperation of the various stores in the Ca2+-release process.
- Published
- 2002
- Full Text
- View/download PDF
40. Ragged spiking of free calcium in ADP-stimulated human platelets: regulation of puff-like calcium signals in vitro and ex vivo.
- Author
-
Heemskerk JW, Willems GM, Rook MB, and Sage SO
- Subjects
- Blood Platelets drug effects, Buffers, Calibration, Culture Media, GTP-Binding Proteins metabolism, HeLa Cells, Humans, In Vitro Techniques, Microscopy, Confocal, Microscopy, Fluorescence, Microscopy, Video, Stimulation, Chemical, Adenosine Diphosphate pharmacology, Blood Platelets metabolism, Calcium blood, Calcium Signaling drug effects
- Abstract
1. Human platelets respond to agonists of G protein (G(q))-coupled receptors by generating an irregular pattern of spiking changes in cytosolic Ca2+ ([Ca2+]i). We have investigated the ADP-induced Ca2+ responses of single, Fluo-3-loaded platelets in the presence or absence of autologous plasma or whole blood under flow conditions. 2. In plasma-free platelets, incubated in buffer medium, baseline separated [Ca2+]i peaks always consisted of a rapid rising phase (median time 0.8 s) which was abruptly followed by a slower, mono-exponential decay phase. The decay constant differed from platelet to platelet, ranging from 0.23 +/- 0.02 to 0.63 +/- 0.03 s(-1) (mean +/- S.E.M., n = 3-5), and was used to identify individual Ca2+ release events and to determine the Ca2+ fluxes of the events. 3. Confocal, high-frequency measurements of adherent, spread platelets (diameter 3-5 microm) indicated that different optical regions had simultaneous patterns of both low- and high-amplitude Ca2+ release events. 4. With or without plasma or flowing blood, the ADP-induced Ca2+ signals in platelets had the characteristics of irregular Ca2+ puffs as well as more regular Ca2+ oscillations. Individual [Ca2+]i peaks varied in amplitude and peak-to-peak interval, as observed for separated Ca2+ puffs within larger cells. On the other hand, the peaks appeared to group into periods of ragged, shorter-interval Ca2+ release events with little integration, which were alternated with longer-interval events. 5. We conclude that the spiking Ca2+ signal generated in these small cells has the characteristics of a 'poor' oscillator with an irregular frequency being reactivated from period to period. This platelet signal appears to be similar in an environment of non-physiological buffer medium and in flowing, whole blood.
- Published
- 2001
- Full Text
- View/download PDF
41. Gap junctions in the rabbit sinoatrial node.
- Author
-
Verheule S, van Kempen MJ, Postma S, Rook MB, and Jongsma HJ
- Subjects
- Animals, Atrial Function, Connexins analysis, Gap Junctions chemistry, Heart Atria cytology, Immunohistochemistry, Male, Membrane Potentials physiology, Muscle Fibers, Skeletal physiology, Myocardial Contraction physiology, Myocardium cytology, Patch-Clamp Techniques, Rabbits, Gap Junction alpha-5 Protein, Gap Junctions physiology, Sinoatrial Node physiology, Sinoatrial Node ultrastructure
- Abstract
In comparison to the cellular basis of pacemaking, the electrical interactions mediating synchronization and conduction in the sinoatrial node are poorly understood. Therefore, we have taken a combined immunohistochemical and electrophysiological approach to characterize gap junctions in the nodal area. We report that the pacemaker myocytes in the center of the rabbit sinoatrial node express the gap junction proteins connexin (Cx)40 and Cx46. In the periphery of the node, strands of pacemaker myocytes expressing Cx43 intermingle with strands expressing Cx40 and Cx46. Biophysical properties of gap junctions in isolated pairs of pacemaker myocytes were recorded under dual voltage clamp with the use of the perforated-patch method. Macroscopic junctional conductance ranged between 0.6 and 25 nS with a mean value of 7.5 nS. The junctional conductance did not show a pronounced sensitivity to the transjunctional potential difference. Single-channel recordings from pairs of pacemaker myocytes revealed populations of single-channel conductances at 133, 202, and 241 pS. With these single-channel conductances, the observed average macroscopic junctional conductance, 7.5 nS, would require only 30-60 open gap junction channels.
- Published
- 2001
- Full Text
- View/download PDF
42. Cardiac sodium channel and inherited arrhythmia syndromes.
- Author
-
Bezzina CR, Rook MB, and Wilde AA
- Subjects
- Arrhythmias, Cardiac metabolism, Arrhythmias, Cardiac therapy, Cardiac Pacing, Artificial, Death, Sudden, Cardiac etiology, Heart Conduction System physiopathology, Humans, Long QT Syndrome genetics, Long QT Syndrome metabolism, Long QT Syndrome therapy, Mutation, Sodium Channels metabolism, Arrhythmias, Cardiac genetics, Myocardium metabolism, Sodium Channels genetics
- Published
- 2001
- Full Text
- View/download PDF
43. Dual patch clamp.
- Author
-
Van Rijen HV, Wilders R, Rook MB, and Jongsma HJ
- Subjects
- Animals, Cytoplasm metabolism, Electric Conductivity, In Vitro Techniques, Indicators and Reagents, Kinetics, Membrane Potentials, Patch-Clamp Techniques instrumentation, Rats, Solutions, Gap Junctions metabolism, Patch-Clamp Techniques methods
- Published
- 2001
- Full Text
- View/download PDF
44. The hibernators heart. Nature's response to arrhythmogenesis?
- Author
-
Opthof T and Rook MB
- Subjects
- Animals, Arrhythmias, Cardiac physiopathology, Cricetinae, Electrophysiology, Gap Junctions physiology, Heart Conduction System physiology, Connexin 43 metabolism, Hibernation physiology, Myocardium metabolism
- Published
- 2000
- Full Text
- View/download PDF
45. Gap junctional remodeling in relation to stabilization of atrial fibrillation in the goat.
- Author
-
van der Velden HM, Ausma J, Rook MB, Hellemons AJ, van Veen TA, Allessie MA, and Jongsma HJ
- Subjects
- Animals, Atrial Appendage chemistry, Blotting, Western, Cardiac Pacing, Artificial, Connexin 43 analysis, Connexin 43 metabolism, Connexins analysis, Female, Goats, Immunohistochemistry, Microscopy, Confocal, Reverse Transcriptase Polymerase Chain Reaction, Time Factors, Gap Junction alpha-5 Protein, Atrial Appendage metabolism, Atrial Fibrillation metabolism, Connexins metabolism
- Abstract
Objective: It has been postulated that high atrial rate induced changes at the level of the gap junctions ('gap junctional remodeling', i.e. changes in distribution, intercellular orientation and expression of gap junction proteins), could be part of the vicious circle of electrophysiologic and structural changes leading to sustained atrial fibrillation (AF). To obtain experimental evidence in favour of such a postulate the timing of this remodeling process was studied in relation to the development of sustained AF in a goat model., Methods and Results: Thin sections from the left (LAA) and right atrial appendage (RAA) from goats in sinus rhythm (SR) or AF, induced through programmed endocardial burst pacing for time periods between 0 and 16 weeks, were immunolabeled with antibodies against connexin(Cx)40 and Cx43 and analysed by immunofluorescence and confocal laser scanning microscopy. During SR the distribution pattern for Cx43 was completely homogeneous (LAA and RAA) and for Cx40 mostly homogeneous (LAA: all five goats, RAA: three out of five goats). The distribution pattern for Cx43 remained stable during AF, while the Cx40 distribution pattern became increasingly heterogeneous, both in the LAA and RAA, with increasing duration of pacing. This increase in heterogeneity in Cx40 distribution correlated (Spearman rank order) with an increase in stability of AF and the occurrence of structural changes (myolysis) in atrial myocytes. The Cx40/Cx43 immunofluorescence signal ratio in both the LAA and RAA appeared to be significantly lower in AF (1-16 weeks) as compared to SR (0 weeks); going from 0 to 16 weeks average ratios decreased 54.5% (n=5; P=0.026) in the LAA and 35.8 (n=5; P=0.034) in the RAA. Western blot analyses revealed similar decreases in the total Cx40/Cx43 protein ratio, on average 50.0% (n=5; P=0.008) and 47.8% (n=5; P=0.02) in the LAA and RAA, respectively. No changes were measured in the levels of Cx40 or Cx43 mRNA, as was assessed through RT-PCR., Conclusion: The time course of changes in the distribution and content of Cx40 gap junctions as observed during endocardial burst pacing of the goat atrium suggests that Cx40 gap junctional remodeling might be involved in the pathogenesis of sustained atrial fibrillation.
- Published
- 2000
- Full Text
- View/download PDF
46. A single Na(+) channel mutation causing both long-QT and Brugada syndromes.
- Author
-
Bezzina C, Veldkamp MW, van Den Berg MP, Postma AV, Rook MB, Viersma JW, van Langen IM, Tan-Sindhunata G, Bink-Boelkens MT, van Der Hout AH, Mannens MM, and Wilde AA
- Subjects
- Adult, Electrocardiography, Female, Humans, Male, Pedigree, Death, Sudden, Cardiac etiology, Long QT Syndrome etiology, Long QT Syndrome genetics, Mutation, Sodium Channels genetics
- Abstract
Mutations in SCN5A, the gene encoding the cardiac Na(+) channel, have been identified in 2 distinct diseases associated with sudden death: one form of the long-QT syndrome (LQT(3)) and the Brugada syndrome. We have screened SCN5A in a large 8-generation kindred characterized by a high incidence of nocturnal sudden death, and QT-interval prolongation and the "Brugada ECG" occurring in the same subjects. An insertion of 3 nucleotides (TGA) at position 5537, predicted to cause an insertion of aspartic acid (1795insD) in the C-terminal domain of the protein, was linked to the phenotype and was identified in all electrocardiographically affected family members. ECGs were obtained from 79 adults with a defined genetic status (carriers, n=43; noncarriers, n=36). In affected individuals, PR and QRS durations and QT intervals are prolonged (P<0.0001 for all parameters). ST segment elevation in the right precordial leads is present as well (P<0.0001). Twenty-five family members died suddenly, 16 of them during the night. Expression of wild-type and mutant Na(+) channels in Xenopus oocytes revealed that the 1795insD mutation gives rise to a 7.3-mV negative shift of the steady-state inactivation curve and an 8.1-mV positive shift of the steady-state activation curve. The functional consequence of both shifts is likely to be a reduced Na(+) current during the upstroke of the action potential. LQT(3) and Brugada syndrome are allelic disorders but may also share a common genotype.
- Published
- 1999
- Full Text
- View/download PDF
47. Human SCN5A gene mutations alter cardiac sodium channel kinetics and are associated with the Brugada syndrome.
- Author
-
Rook MB, Bezzina Alshinawi C, Groenewegen WA, van Gelder IC, van Ginneken AC, Jongsma HJ, Mannens MM, and Wilde AA
- Subjects
- Action Potentials genetics, Animals, Bundle-Branch Block metabolism, Bundle-Branch Block physiopathology, Electrocardiography, Gene Expression, Heart Arrest metabolism, Heart Arrest physiopathology, Humans, Ion Channel Gating genetics, NAV1.5 Voltage-Gated Sodium Channel, Oocytes, Polymorphism, Single-Stranded Conformational, Sequence Analysis, DNA, Sodium Channels metabolism, Syndrome, Xenopus, Bundle-Branch Block genetics, Heart Arrest genetics, Mutation, Missense, Myocardium metabolism, Sodium Channels genetics
- Abstract
Background: Primary dysrhythmias other than those associated with the long QT syndrome, are increasingly recognized. One of these are represented by patients with a history of resuscitation from cardiac arrest but without any structural heart disease. These patients exhibit a distinct electrocardiographic (ECG) pattern consisting of a persistent ST-segment elevation in the right precordial leads often but not always accompanied by a right bundle branch block (Brugada syndrome). This syndrome is associated with a high mortality rate and has been shown to display familial occurrence., Methods and Results: Pharmacological sodium channel blockade elicits or worsens the electrocardiographic features associated with this syndrome. Hence, a candidate gene approach directed towards SCN5A, the gene encoding the alpha-subunit of the cardiac sodium channel, was followed in six affected individuals. In two patients missense mutations were identified in the coding region of the gene: R1512W in the DIII-DIV cytoplasmic linker and A1924T in the C-terminal cytoplasmic domain. In two other patients mutations were detected near intron/exon junctions. To assess the functional consequences of the R1512W and A1924T mutations, wild-type and mutant sodium channel proteins were expressed in Xenopus oocytes. Both missense mutations affected channel function, most notably a 4-5 mV negative voltage shift of the steady-state activation and inactivation curves in R1512W and a 9 mV negative voltage shift of the steady-state activation curve in A1924T, measured at 22 degrees C. Recovery from inactivation was slightly prolonged for R1512W channels. The time dependent kinetics of activation and inactivation at -20 mV were not significantly affected by either mutation., Conclusions: Two SCN5A mutations associated with the Brugada syndrome, significantly affect cardiac sodium channel characteristics. The alterations seem to be associated with an increase in inward sodium current during the action potential upstroke.
- Published
- 1999
- Full Text
- View/download PDF
48. Identification of connexin43 as a functional target for Wnt signalling.
- Author
-
van der Heyden MA, Rook MB, Hermans MM, Rijksen G, Boonstra J, Defize LH, and Destrée OH
- Subjects
- Animals, Carcinoma, Embryonal, Cell Communication physiology, Connexin 26, Connexin 43 biosynthesis, Connexins, Gap Junctions physiology, Humans, Mice, PC12 Cells, Promoter Regions, Genetic drug effects, Promoter Regions, Genetic genetics, Promoter Regions, Genetic physiology, Proto-Oncogene Proteins biosynthesis, Rats, Tumor Cells, Cultured, Wnt Proteins, Wnt1 Protein, Xenopus, Xenopus Proteins, Connexin 43 physiology, Proto-Oncogene Proteins physiology, Signal Transduction physiology, Zebrafish Proteins
- Abstract
Wnt mediated signal transduction is considered to regulate activity of target genes. In Xenopus embryos, ectopic Wnt1 and Wnt8 expression induces gap-junctional communication. During murine brain formation, Wnt1 and the gap-junctional protein connexin43 (Cx43) are co-expressed at the mid/hindbrain border, while interference with Wnt1 or Cx43 expression during embryogenesis leads to severe brain defects in the mid/hindbrain region. In PC12 cells, Wnt1 expression leads to an apparent increase in cell-cell adhesion. We investigated the effects of Wnt1 overexpression on gap-junctional communication in PC12 cells. Wnt1 expressing clones displayed an increased electrical and chemical coupling. This coincides with an increased expression of Cx43 mRNA and protein, while other connexins, Cx26, Cx32, Cx37, Cx40 and Cx45, were not up-regulated. Also, induction of Wnt1 expression in a mammary epithelial cell line leads to an increase in gap-junctional communication and Cx43 protein expression. In transient transactivation assays in P19 EC cells we found that Wnt1 and Li+, an ion that mimics Wnt signalling, increased transcription from the rat Cx43 promoter, potentially via TCF/LEF binding elements, in a pathway separate from cAMP-induced Cx43 transactivation. The results demonstrate that Cx43 acts as a functional target of Wnt1 signalling, and Cx43 expression can be regulated by Wnt1 at the transcriptional level. Our data suggest that Wnt1-induced cell fate determination is likely to involve regulation of gap-junctional communication.
- Published
- 1998
- Full Text
- View/download PDF
49. Gap junctions in human umbilical cord endothelial cells contain multiple connexins.
- Author
-
Van Rijen H, van Kempen MJ, Analbers LJ, Rook MB, van Ginneken AC, Gros D, and Jongsma HJ
- Subjects
- Cells, Cultured, Connexin 43 metabolism, Electrophysiology, Endothelium, Vascular cytology, Humans, Immunohistochemistry, Isoquinolines, Patch-Clamp Techniques, Umbilical Arteries cytology, Umbilical Veins cytology, Gap Junction alpha-5 Protein, Gap Junction alpha-4 Protein, Connexins metabolism, Endothelium, Vascular metabolism, Gap Junctions metabolism, Umbilical Arteries metabolism, Umbilical Veins metabolism
- Abstract
We investigated the expression pattern of gap junctional proteins (connexins, Cx) in situ and in vitro and their functional characteristics in cultured human umbilical vein endothelial cells (HUVEC) and cultured human umbilical artery endothelial cells (HUAEC). In both arteries and veins, Cx37, Cx40, and Cx43 could be detected in situ and in vitro (passages 2-4). Distribution patterns of Cx40 and Cx43 were homogeneous in situ but more heterogeneous in vitro. Cx37 is heterogeneously expressed both in situ and in vitro. Among most cells, no Cx37 staining could be detected; when present, it was found as bright spots between some clusters of cells. Cx40 was more abundant in cultured arterial endothelium than in cultured venous endothelium. Dye-coupling experiments with Lucifer yellow CH revealed extensive dye spread in HUVEC (15.2 +/- 0.4, mean +/- SE, n = 110) but was significantly restricted in HUAEC (9.8 +/- 0.3, n = 110). Electrophysiological gap junctional characteristics were determined in cultured HUVEC and HUAEC pairs by use of the dual voltage-clamp technique. In contrast to the dye-coupling experiments, mean macroscopic electrical conductance was significantly larger for HUAEC pairs (31.4 +/- 6.0 nS, n = 12) than for HUVEC pairs (16.6 +/- 2.8, n = 18). In HUVEC, we measured multiple single gap junctional channel conductances in the range of 19-75 pS. Interestingly, additional conductances of 80-200 pS were measured in HUAEC, possibly partially reflecting activity of channels formed of Cx40, which are more abundant in the cultured arterial endothelial cells.
- Published
- 1997
- Full Text
- View/download PDF
50. pH sensitivity of the cardiac gap junction proteins, connexin 45 and 43.
- Author
-
Hermans MM, Kortekaas P, Jongsma HJ, and Rook MB
- Subjects
- Animals, Humans, Hydrogen-Ion Concentration, Patch-Clamp Techniques, Rats, Tumor Cells, Cultured, Connexin 43 metabolism, Connexins metabolism, Gap Junctions metabolism, Myocardium metabolism
- Abstract
. Intercellular communication through gap junction channels can be regulated by changes in intracellular pH (pHi). This regulation may play an important role in ischemic heart tissue. Using the dual voltage-clamp technique, we compared the pHi sensitivity of gap junction channels composed of connexin 43 (Cx43) and Cx45, two of the gap junction proteins that are expressed in heart. We made use of SKHep1 cells, endogenously expressing low levels of Cx45 and SKHep1 cells stably transfected with rat Cx43. To manipulate the pHi we applied the NH3/NH+4 pH-clamp method. At pHi 6.7 the gj of Cx45 channels was reduced to approximately 20% of control values (pHi 7.0) and at pHi 6.3 all channels closed. The gj of Cx43 channels was approximately 70% of control values at pHi 6.7 and approximately 40% at pHi 6.3. Cx43 channels closed at pHi 5.8. Single channel conductances were 17.8 pS for Cx45 and 40.8 pS for Cx43 at pHi 7.0 and did not change significantly at lower pHi. This suggests that the decrease in macroscopic conductance observed at low pHi results from the decrease in open probability of gap junctional channels rather than from a decrease in single channel conductance. Our results demonstrate that gap junction channels built of Cx45 are far more pH sensitive than channels built of Cx43.
- Published
- 1995
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.