67 results on '"Guillermo J. Pérez"'
Search Results
2. Generation of four induced pluripotent stem cell lines from a family harboring a single nucleotide variant in SCN5A
- Author
-
Rebecca Martínez-Moreno, David Carreras, Begoña Aran, Bernd Kuebler, Georgia Sarquella-Brugada, Ramon Brugada, Guillermo J. Pérez, Fabiana S. Scornik, and Elisabet Selga
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Patient-derived induced pluripotent stem cells (iPSC) are a valuable approach to model cardiovascular diseases. We nucleofected non-integrating episomal vectors in skin fibroblasts of three family members carrying a single nucleotide variant (SNV) in SCN5A, which encodes the cardiac-type sodium channel, and of a related healthy control. The SNV SCN5A_c.4573G > A had been previously identified in a Brugada Syndrome patient. The resulting iPS cell lines differentiate into cells of the 3 germ layers, display normal karyotypes and express pluripotency surface markers and genes. Thus, they are a reliable source to study the effect of the identified mutation in a physiologically relevant environment.
- Published
- 2022
- Full Text
- View/download PDF
3. Generation of an induced pluripotent stem cell line from a healthy Caucasian male
- Author
-
Rebecca Martínez-Moreno, Alexandra Pérez-Serra, David Carreras, Begoña Aran, Bernd Kuebler, Ramon Brugada, Fabiana S. Scornik, Guillermo J. Pérez, and Elisabet Selga
- Subjects
Biology (General) ,QH301-705.5 - Abstract
The effects of genetic mutations on protein function can be studied in a physiologically relevant environment using tissue-specific cells differentiated from patient-derived induced pluripotent stem cells (iPSC). However, it is crucial to use iPSC derived from healthy individuals as control. We generated an iPS cell line from skin fibroblasts of a healthy Caucasian male by nucleofection of non-integrating episomal vectors. This cell line has normal karyotype, expresses pluripotency surface markers and pluripotency genes, and successfully differentiates into cells of the 3 germ layers. Therefore, it can be used as control for any disease of interest that is modelled using iPSC.
- Published
- 2022
- Full Text
- View/download PDF
4. An SCN1B Variant Affects Both Cardiac-Type (NaV1.5) and Brain-Type (NaV1.1) Sodium Currents and Contributes to Complex Concomitant Brain and Cardiac Disorders
- Author
-
Rebecca Martinez-Moreno, Elisabet Selga, Helena Riuró, David Carreras, Mered Parnes, Chandra Srinivasan, Michael F. Wangler, Guillermo J. Pérez, Fabiana S. Scornik, and Ramon Brugada
- Subjects
NaV1.5 ,NaV1.1 ,NaVβ1 ,NaVβ1b ,cardiac arrhythmia ,brain hyperexcitability ,Biology (General) ,QH301-705.5 - Abstract
Voltage-gated sodium (NaV) channels are transmembrane proteins that initiate and propagate neuronal and cardiac action potentials. NaV channel β subunits have been widely studied due to their modulatory role. Mice null for Scn1b, which encodes NaV β1 and β1b subunits, have defects in neuronal development and excitability, spontaneous generalized seizures, cardiac arrhythmias, and early mortality. A mutation in exon 3 of SCN1B, c.308A>T leading to β1_p.D103V and β1b_p.D103V, was previously found in a patient with a history of proarrhythmic conditions with progressive atrial standstill as well as cognitive and motor deficits accompanying structural brain abnormalities. We investigated whether β1 or β1b subunits carrying this mutation affect NaV1.5 and/or NaV1.1 currents using a whole cell patch-clamp technique in tsA201 cells. We observed a decrease in sodium current density in cells co-expressing NaV1.5 or NaV1.1 and β1D103V compared to β1WT. Interestingly, β1bD103V did not affect NaV1.1 sodium current density but induced a positive shift in the voltage dependence of inactivation and a faster recovery from inactivation compared to β1bWT. The β1bD103V isoform did not affect NaV1.5 current properties. Although the SCN1B_c.308A>T mutation may not be the sole cause of the patient’s symptoms, we observed a clear loss of function in both cardiac and brain sodium channels. Our results suggest that the mutant β1 and β1b subunits play a fundamental role in the observed electrical dysfunction.
- Published
- 2020
- Full Text
- View/download PDF
5. FORNER MUÑOZ, Salvador (ed.), ¿El reencuentro europeo? A los veinticinco años de la caída del Muro de Berlín, Valencia, Tirant Humanidades, 2015, 316 pp.
- Author
-
Guillermo J. Pérez Casanova
- Subjects
Europa ,Transición política ,Socialismo ,Comunismo ,Reseñas bibliográficas ,History (General) and history of Europe - Published
- 2015
- Full Text
- View/download PDF
6. FUENTES CODERA, Maximiliano, España en la Primera Guerra Mundial. Una movilización cultural, Madrid, Akal, 2014, 238 pp.
- Author
-
Guillermo J. Pérez Casanova
- Subjects
Primera Guerra Mundial ,España ,Reseñas bibliográficas ,History (General) and history of Europe - Published
- 2014
- Full Text
- View/download PDF
7. Englund, Peter, La belleza y el dolor de la batalla, Barcelona, Roca Editorial, 2011, 761 pp.
- Author
-
Guillermo J. Pérez Casanova
- Subjects
Primera Guerra Mundial ,Reseñas bibliográficas ,History (General) and history of Europe - Published
- 2011
- Full Text
- View/download PDF
8. Fuentes Codera, Maximiliano, El campo de fuerzas europeo en Cataluña. Eugeni d’Ors en los primeros años de la Gran Guerra, Lleida, Pagès Editors, 2009, 308 pp.
- Author
-
Guillermo J. Pérez Casanova
- Subjects
Primera Guerra Mundial ,Cataluña ,Eugeni d’Ors ,Reseñas bibliográficas ,History (General) and history of Europe - Published
- 2010
- Full Text
- View/download PDF
9. Beneyto, José M.a (dir.), Maillo González-Orús, Jerónimo y Becerril Atienza, Belén (coords.), Desarrollo histórico y caracteres básicos de la Unión Europea. Naturaleza, valores, principios y competencias, Pamplona, Aranzadi, 2009, 633 pp.
- Author
-
Guillermo J. Pérez Casanova
- Subjects
Unión Europea ,Evolución política ,Naturaleza jurídica ,Reseñas bibliográficas ,History (General) and history of Europe - Published
- 2010
- Full Text
- View/download PDF
10. Alterations in Calcium Handling Are a Common Feature in an Arrhythmogenic Cardiomyopathy Cell Model Triggered by Desmosome Genes Loss
- Author
-
Marta Vallverdú-Prats, David Carreras, Guillermo J. Pérez, Oscar Campuzano, Ramon Brugada, and Mireia Alcalde
- Subjects
Inorganic Chemistry ,HL1 ,Organic Chemistry ,calcium handling ,desmosomes ,General Medicine ,Physical and Theoretical Chemistry ,Molecular Biology ,Arrhythmogenic Cardiomyopathy ,CRISPR/Cas9 ,Spectroscopy ,Catalysis ,Computer Science Applications - Abstract
Arrhythmogenic cardiomyopathy (ACM) is an inherited cardiac disease characterized by fibrofatty replacement of the myocardium. Deleterious variants in desmosomal genes are the main cause of ACM and lead to common and gene-specific molecular alterations, which are not yet fully understood. This article presents the first systematic in vitro study describing gene and protein expression alterations in desmosomes, electrical conduction-related genes, and genes involved in fibrosis and adipogenesis. Moreover, molecular and functional alterations in calcium handling were also characterized. This study was performed d with HL1 cells with homozygous knockouts of three of the most frequently mutated desmosomal genes in ACM: PKP2, DSG2, and DSC2 (generated by CRISPR/Cas9). Moreover, knockout and N-truncated clones of DSP were also included. Our results showed functional alterations in calcium handling, a slower calcium re-uptake was observed in the absence of PKP2, DSG2, and DSC2, and the DSP knockout clone showed a more rapid re-uptake. We propose that the described functional alterations of the calcium handling genes may be explained by mRNA expression levels of ANK2, CASQ2, ATP2A2, RYR2, and PLN. In conclusion, the loss of desmosomal genes provokes alterations in calcium handling, potentially contributing to the development of arrhythmogenic events in ACM.
- Published
- 2023
- Full Text
- View/download PDF
11. Epigenetic Changes Governing Scn5a Expression in Denervated Skeletal Muscle
- Author
-
Manel M. Santafé, Mel·lina Pinsach-Abuin, Rebecca Martinez-Moreno, Guillermo J. Pérez, Sara Pagans, Ramon Brugada, David Carreras, Pol Gomà, and Fabiana S. Scornik
- Subjects
Male ,epigenetic mechanisms ,Scn5a ,Epigenesis, Genetic ,NAV1.5 Voltage-Gated Sodium Channel ,lcsh:Chemistry ,Rats, Sprague-Dawley ,Transcriptome ,Transcriptional regulation ,transcriptional regulation ,RNA-Seq ,lcsh:QH301-705.5 ,Spectroscopy ,Denervation ,histone modifications ,GATA4 ,Cardiac action potential ,General Medicine ,Muscle Denervation ,Computer Science Applications ,Cell biology ,medicine.anatomical_structure ,cardiac arrhythmias ,cardiovascular system ,Epigenetics ,Arrhythmia ,Gene isoform ,congenital, hereditary, and neonatal diseases and abnormalities ,Arrítmia ,Biology ,Response Elements ,Article ,Catalysis ,Inorganic Chemistry ,Cor -- Malalties ,Cor -- Malalties -- Aspectes genètics ,skeletal muscle denervation ,medicine ,Animals ,cardiovascular diseases ,Physical and Theoretical Chemistry ,Muscle, Skeletal ,Molecular Biology ,Transcription factor ,Organic Chemistry ,Skeletal muscle ,Heart -- Diseases -- Genetic aspects ,Heart -- Diseases ,Epigenètica ,GATA4 Transcription Factor ,Rats ,lcsh:Biology (General) ,lcsh:QD1-999 ,H3K27 acetylation - Abstract
The SCN5A gene encodes the α-subunit of the voltage-gated cardiac sodium channel (NaV1.5), a key player in cardiac action potential depolarization. Genetic variants in protein-coding regions of the human SCN5A have been largely associated with inherited cardiac arrhythmias. Increasing evidence also suggests that aberrant expression of the SCN5A gene could increase susceptibility to arrhythmogenic diseases, but the mechanisms governing SCN5A expression are not yet well understood. To gain insights into the molecular basis of SCN5A gene regulation, we used rat gastrocnemius muscle four days following denervation, a process well known to stimulate Scn5a expression. Our results show that denervation of rat skeletal muscle induces the expression of the adult cardiac Scn5a isoform. RNA-seq experiments reveal that denervation leads to significant changes in the transcriptome, with Scn5a amongst the fifty top upregulated genes. Consistent with this increase in expression, ChIP-qPCR assays show enrichment of H3K27ac and H3K4me3 and binding of the transcription factor Gata4 near the Scn5a promoter region. Also, Gata4 mRNA levels are significantly induced upon denervation. Genome-wide analysis of H3K27ac by ChIP-seq suggest that a super enhancer recently described to regulate Scn5a in cardiac tissue is activated in response to denervation. Altogether, our experiments reveal that similar mechanisms regulate the expression of Scn5a in denervated muscle and cardiac tissue, suggesting a conserved pathway for SCN5A expression among striated muscles This research was funded by the University of Girona, grant number MPCUdG2016/039 and Obra Social La Caixa. D.C. acknowledges a predoctoral fellowship from the Agència de Gesitó d’Ajusts Universitaris I de Recerca-Generalitat de Catalunya (2018FI_B00969) and European Social Fund (ESF)
- Published
- 2021
12. B-PO04-026 NON-SELECTIVE VERSUS Β1-SELECTIVE BETA-BLOCKERS IN THE TREATMENT OF SYMPTOMATIC CHILDREN WITH CATECHOLAMINERGIC POLYMORPHIC VENTRICULAR TACHYCARDIA
- Author
-
Dania Kallas, Prince J. Kannankeril, Vincent Probst, Jonathan R. Skinner, Michael W.T. Tanck, Andrew D. Krahn, Guillermo J. Pérez, Jacob Tfelt-Hansen, Habib Khan, Vibeke M. Almaas, Kristina H. Haugaa, Sing-Chien Yap, Georgia Sarquella Brugada, Andrew M. Davis, Wataru Shimizu, Ferran Rosés I. Noguer, Martin J. LaPage, Arthur A.M. Wilde, Boris Rudic, Elijah R. Behr, Janneke A.E. Kammeraad, Thomas M. Roston, Esther Zorio Grima, Ramon Brugada, Maarten P. van den Berg, Michael J. Ackerman, Frederic Sacher, Heikki Swan, Minoru Horie, Christopher Semsarian, Christian van der Werf, Shubhayan Sanatani, Peter J. Schwartz, Antoine Leenhardt, Nico A. Blom, Alice Maltret, Jason D. Roberts, Puck Peltenburg, Krystien V.V. Lieve, John R. Giudicessi, Jan Till, Sonia Franciosi, Anwar Baban, Katrina B. Sorensen, Isabelle Denjoy, Yuko Wada, Johan M Bos, Terezia Tavacova, Sally-Ann B. Clur, and Tomas Robyns
- Subjects
medicine.medical_specialty ,business.industry ,Physiology (medical) ,Internal medicine ,medicine ,Cardiology ,Cardiology and Cardiovascular Medicine ,business ,Catecholaminergic polymorphic ventricular tachycardia ,medicine.disease ,Beta (finance) - Published
- 2021
- Full Text
- View/download PDF
13. Molecular heterogeneity of large-conductance calcium-activated potassium channels in canine intracardiac ganglia
- Author
-
Alexandra Pérez-Serra, Guillermo J. Pérez, Elisabet Selga, Fabiana S. Scornik, Ramon Brugada, Alba Moreno-Asso, Mayurika Desai, Seth Anderson, and Kristen Thomas
- Subjects
Nervous system ,BK channel ,Protein subunit ,Molecular Sequence Data ,Biophysics ,Cardiology ,chemistry.chemical_element ,β subunits ,Calcium ,Biochemistry ,Cardiologia ,chemistry.chemical_compound ,alternative splicing ,Dogs ,Cor -- Malalties ,medicine ,Animals ,Amino Acid Sequence ,Large-Conductance Calcium-Activated Potassium Channels ,Paxilline ,Neurons ,biology ,autonomic ,intracardiac neurons ,Myocardium ,Alternative splicing ,Heart -- Diseases ,Calcium-activated potassium channel ,Cell biology ,Article Addendum ,Protein Subunits ,medicine.anatomical_structure ,chemistry ,biology.protein ,Excitatory postsynaptic potential ,Ganglia ,BK channels ,Neuroscience - Abstract
Large conductance calcium-activated potassium (BK) channels are widely expressed in the nervous system. We have recently shown that principal neurons from canine intracardiac ganglia (ICG) express a paxilline- and TEA-sensitive BK current, which increases neuronal excitability. In the present work, we further explore the molecular constituents of the BK current in canine ICG. We found that the β1 and β4 regulatory subunits are expressed in ICG. Single channel voltage-dependence at different calcium concentrations suggested that association of the BKα with a particular β subunit was not enough to explain the channel activity in this tissue. Indeed, we detected the presence of several splice variants of the BKα subunit. In conclusion, BK channels in canine ICG may result from the arrangement of different BKα splice variants, plus accessory β subunits. The particular combinations expressed in canine IC neurons likely rule the excitatory role of BK current in this tissue This work was supported in part by a National Institute of Health grant HL073161 to GJP, an American Heart Association grant 0335446T to FSS, a grant from the Obra Social “La Caixa” to RB and a Sara Borrell postdoctoral fellowship CD11/00063 from the Instituto de Salud Carlos III to ES
- Published
- 2020
14. La flecainida reduce las arritmias ventriculares en pacientes con taquicardia ventricular polimórfica catecolaminérgica genotipo RyR2 positivo
- Author
-
Eduardo Caballero Dorta, María del Val Groba Marco, Guillermo J. Pérez, Josep Brugada Terradellas, Oscar Campuzano Larrea, Julio Salvador Hernández Afonso, Ramón Brugada Terradellas, Fernando Wangüemert Pérez, and Luis Álvarez Acosta
- Subjects
03 medical and health sciences ,0302 clinical medicine ,business.industry ,Medicine ,030212 general & internal medicine ,030204 cardiovascular system & hematology ,Cardiology and Cardiovascular Medicine ,business ,Humanities - Abstract
Resumen Introduccion y objetivos La taquicardia ventricular polimorfica catecolaminergica (TVPC) es una enfermedad hereditaria caracterizada por arritmias ventriculares (AV) polimorficas o bidireccionales desencadenadas por estres fisico o emocional en jovenes con corazon estructuralmente normal. El pilar del tratamiento son los bloqueadores beta y recientemente se ha incorporado la flecainida al arsenal terapeutico. El objetivo de este trabajo es exponer nuestra experiencia con su uso. Metodos De un total de 174 pacientes pertenecientes a 7 familias afectadas de TVPC con genotipo positivo, se analizo a los que tomaron flecainida. Se valoraron la indicacion, los efectos secundarios, la dosificacion, los eventos clinicos, las AV y la ventana arritmica en las pruebas de esfuerzo y las descargas del desfibrilador automatico (DAI). Resultados Recibieron flecainida 18 pacientes (10,4%); 17 en combinacion con bloqueadores beta y 1 como tratamiento unico por intolerancia al bloqueador beta. Ningun paciente sufrio efectos secundarios. La indicacion fue la persistencia de AV complejas en la prueba de esfuerzo en 13 pacientes (72,2%) y descargas frecuentes del DAI en los otros 5 (27,8%). En el 66,7% de la familia 1, la puntuacion cuantitativa de AV en la prueba de esfuerzo se redujo mas de un 50% (32,76 ± 84,06 frente a 74,38 ± 153,86 lpm; p = 0,018). La ventana arritmica fue menor (5,8 ± 11,9 frente a 19,69 ± 21,27 lpm; p = 0,007) y 4 de los 5 pacientes con descargas apropiadas del DAI no sufrieron mas descargas. Conclusiones En pacientes con TVPC, la flecainida reduce los eventos clinicos, las AV, la ventana arritmica y las descargas del DAI y los pacientes la toleran bien.
- Published
- 2018
- Full Text
- View/download PDF
15. Flecainide Reduces Ventricular Arrhythmias in Patients With Genotype RyR2-positive Catecholaminergic Polymorphic Ventricular Tachycardia
- Author
-
María del Val Groba Marco, Josep Brugada Terradellas, Julio Salvador Hernández Afonso, Fernando Wangüemert Pérez, Eduardo Caballero Dorta, Oscar Campuzano Larrea, Luis Álvarez Acosta, Guillermo J. Pérez, and Ramón Brugada Terradellas
- Subjects
Adult ,Male ,medicine.medical_specialty ,Genotype ,DNA Mutational Analysis ,030204 cardiovascular system & hematology ,Catecholaminergic polymorphic ventricular tachycardia ,Ryanodine receptor 2 ,Electrocardiography ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Humans ,Medicine ,In patient ,030212 general & internal medicine ,Adverse effect ,Flecainide ,business.industry ,Ryanodine Receptor Calcium Release Channel ,General Medicine ,medicine.disease ,Death, Sudden, Cardiac ,Anesthesia ,Mutation ,Cohort ,Tachycardia, Ventricular ,Cardiology ,Female ,Inherited disease ,business ,Anti-Arrhythmia Agents ,Follow-Up Studies ,medicine.drug - Abstract
Introduction and objectives Catecholaminergic polymorphic ventricular tachycardia (CPVT) is an inherited disease characterized by polymorphic or bidirectional ventricular arrhythmias (VA) triggered by physical or emotional stress in young people with a structurally normal heart. Beta-blockers are the cornerstone of treatment, while flecainide has recently been incorporated into the therapeutic arsenal. The aim of this study was to report our experience with this drug. Methods The cohort included 174 genotype-positive CPVT-patients from 7 families. We collected data from patients who were receiving flecainide and analyzed the indications, adverse effects and dosage, clinical events, VA and arrhythmic window during exercise testing, and implantable cardioverter-defibrillator (ICD) shocks during follow-up. Results Eighteen patients (10.4%) received flecainide; 17 patients in combination with beta-blockers, and 1 patient as monotherapy due to beta-blocker intolerance. None of the patients presented side effects. In 13 patients (72.2%) the indication was the persistence of exercise-induced VA and in 5 patients (27.7%) persistent ICD-shocks, despite on beta-blockers. After flecainide initiation, the exercise-induced VA quantitative score was reduced by more than 50% in 66.7% of the members of family 1 (32.76 ± 84.06 vs 74.38 ± 153.86; P = .018). The arrhythmic window was reduced (5.8 ± 11.9 bpm vs 19.69 ± 21.27 bpm; P = .007), and 4 of 5 patients with appropriate ICD shocks experienced no further shocks in the follow-up. Conclusions In CPVT-patients flecainide reduces clinical events, exercise-induced VA, the arrhythmic window, and ICD shocks, with good tolerance.
- Published
- 2018
- Full Text
- View/download PDF
16. Extra Virgin Olive Oil Contains a Phenolic Inhibitor of the Histone Demethylase LSD1/KDM1A
- Author
-
Fabiana S. Scornik, Laura Llorach-Parés, Antonio Segura-Carretero, David Carreras, Elisabet Cuyàs, Elisabet Selga, Javier A. Menendez, Juan Gumuzio, Melchor Sanchez-Martinez, Ramon Brugada, Guillermo J. Pérez, Jesús Lozano-Sánchez, Joaquim Bosch-Barrera, José Antonio Encinar, Angel G. Martin, and Sara Verdura
- Subjects
0301 basic medicine ,Models, Molecular ,cancer stem cells ,neurological disorders ,SOX2 ,Gene Expression ,Histones ,0302 clinical medicine ,Càncer ,Cancer ,Histone Demethylases ,Nutrition and Dietetics ,biology ,Molecular Structure ,Chemistry ,Cancer stem cells ,Communication ,Secoiridoids ,Biofenol ,Recombinant Proteins ,Chromatin ,Molecular Docking Simulation ,Histone ,Biochemistry ,030220 oncology & carcinogenesis ,MCF-7 Cells ,Neoplastic Stem Cells ,Obesitat ,Phenolics ,lcsh:Nutrition. Foods and food supply ,Co-Repressor Proteins ,animal structures ,phenolics ,secoiridoids ,lcsh:TX341-641 ,Breast Neoplasms ,03 medical and health sciences ,Histone H3 ,Phenols ,Cell Line, Tumor ,Humans ,cancer ,Obesity ,Epigenetics ,Binding site ,Nervous system -- Diseases ,Transcription factor ,Olive Oil ,Aldehydes ,Binding Sites ,SOXB1 Transcription Factors ,KDM1A ,Sistema nerviós -- Malalties ,Biophenol ,030104 developmental biology ,biology.protein ,Demethylase ,metabolism ,Neurological disorders ,Food Science - Abstract
The lysine-specific histone demethylase 1A (LSD1) also known as lysine (K)-specific demethylase 1A (KDM1A) is a central epigenetic regulator of metabolic reprogramming in obesity-associated diseases, neurological disorders, and cancer. Here, we evaluated the ability of oleacein, a biophenol secoiridoid naturally present in extra virgin olive oil (EVOO), to target LSD1. Molecular docking and dynamic simulation approaches revealed that oleacein could target the binding site of the LSD1 cofactor flavin adenosine dinucleotide with high affinity and at low concentrations. At higher concentrations, oleacein was predicted to target the interaction of LSD1 with histone H3 and the LSD1 co-repressor (RCOR1/CoREST), likely disturbing the anchorage of LSD1 to chromatin. AlphaScreen-based in vitro assays confirmed the ability of oleacein to act as a direct inhibitor of recombinant LSD1, with an IC50 as low as 2.5 umol/L. Further, oleacein fully suppressed the expression of the transcription factor SOX2 (SEX determining Region Y-box 2) in cancer stem-like and induced pluripotent stem (iPS) cells, which specifically occurs under the control of an LSD1-targeted distal enhancer. Conversely, oleacein failed to modify ectopic SOX2 overexpression driven by a constitutive promoter. Overall, our findings provide the first evidence that EVOO contains a naturally occurring phenolic inhibitor of LSD1, and support the use of oleacein as a template to design new secoiridoid-based LSD1 inhibitors., Work in the Menendez laboratory is supported by the Spanish Ministry of Science and Innovation (Grant SAF2016-80639-P, Plan Nacional de l+D+I, founded by the European Regional Development Fund, Spain) and by an unrestricted research grant from the Fundació Oncolliga Girona (Lliga catalana d’ajuda al malalt de càncer, Girona). The Spanish Ministry of Economy and Competitiveness (MINECO, Project RTI2018-096724-B-C21) and the Generalitat Valenciana (PROMETEO/2016/006) supports work in the Encinar laboratory
- Published
- 2019
17. Sudden infant death as the most severe phenotype caused by genetic modulation in a family with atrial fibrillation
- Author
-
Óscar Díaz-Castro, Ramon Brugada, Alejandro Blanco-Verea, Bernardo López-Abel, Montserrat Santori, Helena Riuró, Guillermo J. Pérez, Fabiana S. Scornik, Maria Brion, Angel Carracedo, and Rocio Gil
- Subjects
0301 basic medicine ,Male ,ERG1 Potassium Channel ,Heterozygote ,hERG ,Mutation, Missense ,Context (language use) ,Bioinformatics ,Pathology and Forensic Medicine ,Sudden cardiac death ,03 medical and health sciences ,0302 clinical medicine ,Genotype ,Atrial Fibrillation ,Genetics ,Medicine ,Missense mutation ,Humans ,030216 legal & forensic medicine ,Genetic testing ,Polymorphism, Genetic ,medicine.diagnostic_test ,biology ,business.industry ,Homozygote ,Infant ,Atrial fibrillation ,Sudden infant death syndrome ,Middle Aged ,medicine.disease ,Pedigree ,030104 developmental biology ,Phenotype ,biology.protein ,business ,Sudden Infant Death - Abstract
Aims To assess the functional impact of two combined KCNH2 variants involved in atrial fibrillation, syncope and sudden infant death syndrome. Methods and results Genetic testing of a 4-month old SIDS victim identified a rare missense heterozygous in KCNH2 variant (V483I) and a missense homozygous polymorphism (K897T) which is often described as a genetic modifier. Electrophysiological characterisation of heterologous HERG channels representing two different KCNH2 genotypes within the family, showed significant differences in both voltage and time dependence of activation and inactivation with a global gain-of-function effect of mutant versus wild type channels and, also, differences between both types of recombinant channels. Conclusions The rare variant V483I in combination with K897T produces a gain-of-function effect that represents a pathological substrate for atrial fibrillation, syncope and sudden infant death syndrome events in this family. Ascertaining the genotype-phenotype correlation of genetic variants is imperative for the correct assessment of genetic testing and counselling. Translational perspective According to the current guidelines for clinical interpretation of sequence variants, functional studies are an essential tool for the ascertainment of variant pathogenicity. They are especially relevant in the context of sudden infant death syndrome and sudden cardiac death, where individuals cannot be clinically evaluated. The patch-clamp technique is a gold-standard for analysis of the biophysical mechanisms of ion channels.
- Published
- 2019
18. Experimental models of Brugada syndrome
- Author
-
Fabiana S. Scornik, Guillermo J. Pérez, Elisabet Selga, Nicholas L. Mills, Ramon Brugada, and Franziska Sendfeld
- Subjects
0301 basic medicine ,Review ,Disease ,Stem cells ,030204 cardiovascular system & hematology ,NAV1.5 Voltage-Gated Sodium Channel ,Sudden cardiac death ,Animals, Genetically Modified ,model systems ,lcsh:Chemistry ,Mice ,0302 clinical medicine ,ST segment ,Myocytes, Cardiac ,Induced pluripotent stem cell ,lcsh:QH301-705.5 ,SCN5A ,Spectroscopy ,Brugada syndrome ,Brugada, Síndrome de ,Cell Differentiation ,General Medicine ,Pathophysiology ,Computer Science Applications ,Cardiology ,cardiovascular system ,Disease Susceptibility ,Stem cell ,Cèl·lules mare ,medicine.medical_specialty ,congenital, hereditary, and neonatal diseases and abnormalities ,Induced Pluripotent Stem Cells ,Catalysis ,Inorganic Chemistry ,03 medical and health sciences ,Dogs ,Internal medicine ,medicine ,Animals ,Humans ,Genetic Predisposition to Disease ,cardiovascular diseases ,Physical and Theoretical Chemistry ,Molecular Biology ,induced pluripotent stem cells (iPS) ,business.industry ,Organic Chemistry ,medicine.disease ,Disease Models, Animal ,030104 developmental biology ,lcsh:Biology (General) ,lcsh:QD1-999 ,Mutation ,Ventricular fibrillation ,business ,Biomarkers - Abstract
Brugada syndrome is an inherited, rare cardiac arrhythmogenic disease, associated with sudden cardiac death. It accounts for up to 20% of sudden deaths in patients without structural cardiac abnormalities. The majority of mutations involve the cardiac sodium channel gene SCN5A and give rise to classical abnormal electrocardiogram with ST segment elevation in the right precordial leads V1 to V3 and a predisposition to ventricular fibrillation. The pathophysiological mechanisms of Brugada syndrome have been investigated using model systems including transgenic mice, canine heart preparations, and expression systems to study different SCN5A mutations. These models have a number of limitations. The recent development of pluripotent stem cell technology creates an opportunity to study cardiomyocytes derived from patients and healthy individuals. To date, only a few studies have been done using Brugada syndrome patient-specific iPS-CM, which have provided novel insights into the mechanisms and pathophysiology of Brugada syndrome. This review provides an evaluation of the strengths and limitations of each of these model systems and summarizes the key mechanisms that have been identified to date. British Heart Foundation Intermediate Clinical Research Fellowship (NLM: FS/10/024/28266), UK Cardiovascular Regenerative Medicine Centre Award (NLM: RM/13/2/30158), Obra social “la Caixa”, Centro Nacional de Investigaciones Cardiovasculares (RB: CNIC-03-2008), Instituto de Salud Carlos III (RB: FIS-PI08/1800 and Fondo Europeo de Desarrollo Regional), Fundació La Marató de TV3 [FSS: 20153910] and Universitat de Girona (GJP: MPCUdG2016). ES, RB, GJP and FSS are members of the CIBERCV, an initiative of the Instituto de Salud Carlos III, Spanish Ministry of Economy and Competitiveness
- Published
- 2019
- Full Text
- View/download PDF
19. Comparative Study of the Effects of an SCN5A Mutation within a Family Diagnosed with Brugada Syndrome using iPS-CM
- Author
-
Guillermo J. Pérez, Georgia Sarquella-Brugada, Ramon Brugada, David Carreras, Elisabet Selga, Rebecca Martinez-Moreno, and Fabiana S. Scornik
- Subjects
Pediatrics ,medicine.medical_specialty ,Scn5a gene ,business.industry ,Biophysics ,medicine ,medicine.disease ,business ,Brugada syndrome - Published
- 2020
- Full Text
- View/download PDF
20. CPVT-Associated Mutation P.G357S-RYR2 Promotes a Gain of Function in Patient-Specific Induced Pluripotent Stem Cell-Derived Cardiomyocytes (iPS-CM)
- Author
-
Fabiana S. Scornik, Rebecca Martinez-Moreno, Elisabet Selga, David Carreras, Guillermo J. Pérez, and Ramon Brugada
- Subjects
Gain of function ,Mutation (genetic algorithm) ,Biophysics ,Cancer research ,In patient ,Biology ,Induced pluripotent stem cell ,Ryanodine receptor 2 - Published
- 2020
- Full Text
- View/download PDF
21. Clinical and molecular characterization of a cardiac ryanodine receptor founder mutation causing catecholaminergic polymorphic ventricular tachycardia
- Author
-
Pablo M. Ruiz Hernandez, Oscar Campuzano, Carmelo Pérez, Ramon Brugada, Cristina Bosch Calero, Guillermo J. Pérez, Anna Iglesias, Pedro Beltran-Alvarez, Fabiana S. Scornik, Josep Brugada, Catarina Allegue, Paola Berne, and Fernando Wanguemert
- Subjects
Adult ,Male ,medicine.medical_specialty ,Adolescent ,medicine.medical_treatment ,Adrenergic beta-Antagonists ,Catecholaminergic polymorphic ventricular tachycardia ,medicine.disease_cause ,Risk Assessment ,Ryanodine receptor 2 ,Sudden death ,Sudden cardiac death ,Physiology (medical) ,Internal medicine ,medicine ,Humans ,Genetic Predisposition to Disease ,Genetic Testing ,Child ,Genetic testing ,Mutation ,medicine.diagnostic_test ,Ryanodine receptor ,business.industry ,Mort sobtada ,Ryanodine Receptor Calcium Release Channel ,Middle Aged ,medicine.disease ,Implantable cardioverter-defibrillator ,Defibrillators, Implantable ,Pedigree ,Death, Sudden, Cardiac ,Treatment Outcome ,Spain ,Arítmia ,Electrocardiography, Ambulatory ,Exercise Test ,Tachycardia, Ventricular ,Cardiology ,Female ,Cardiology and Cardiovascular Medicine ,business ,Arrhythmia - Abstract
Background Catecholaminergic polymorphic ventricular tachycardia (CPVT) is a difficult-to-diagnose cause of sudden cardiac death (SCD). We identified a family of 1400 individuals with multiple cases of CPVT, including 36 SCDs during youth. Objectives We sought to identify the genetic cause of CPVT in this family, to preventively treat and clinically characterize the mutation-positive individuals, and to functionally characterize the pathogenic mechanisms of the mutation. Methods Genetic testing was performed for 1404 relatives. Mutation-positive individuals were preventively treated with β-blockers and clinically characterized with a serial exercise treadmill test (ETT) and Holter monitoring. In vitro functional studies included caffeine sensitivity and store overload–induced calcium release activity of the mutant channel in HEK293 cells. Results We identified the p.G357S_RyR2 mutation, in the cardiac ryanodine receptor, in 179 family members and in 6 SCD cases. No SCD was observed among treated mutation-positive individuals over a median follow-up of 37 months; however, 3 relatives who had refused genetic testing (confirmed mutation-positive individuals) experienced SCD. Holter monitoring did not provide relevant information for CPVT diagnosis. One single ETT was unable to detect complex cardiac arrhythmias in 72% of mutation-positive individuals, though the serial ETT improved the accuracy. Functional studies showed that the G357S mutation increased caffeine sensitivity and store overload–induced calcium release activity under conditions that mimic catecholaminergic stress. Conclusion Our study supports the use of genetic testing to identify individuals at risk of SCD to undertake prophylactic interventions. We also show that the pathogenic mechanisms of p.G357S_RyR2 appear to depend on β-adrenergic stimulation.
- Published
- 2018
22. Cardiac Sodium Current is Severely Impaired in Induced Pluripotent Stem Cell-Derived Cardiomyocytes from Brugada Syndrome Patients
- Author
-
Rebecca Martinez-Moreno, Guillermo J. Pérez, Fabiana S. Scornik, Ramon Brugada, Elisabet Selga, and Georgia Sarquella-Brugada
- Subjects
business.industry ,Biophysics ,Cancer research ,Medicine ,business ,Induced pluripotent stem cell ,medicine.disease ,Sodium current ,Brugada syndrome - Published
- 2019
- Full Text
- View/download PDF
23. A missense mutation in the sodium channel β1b subunit reveals SCN1B as a susceptibility gene underlying long QT syndrome
- Author
-
Anna Iglesias, Guillermo J. Pérez, Helena Riuró, Fabiana S. Scornik, Josep Brugada, Oscar Campuzano, Ramon Brugada, Félix Pérez-Villa, Montserrat Batlle, and Elena Arbelo
- Subjects
Adult ,Male ,medicine.medical_specialty ,Patch-Clamp Techniques ,Long QT syndrome ,Cell Culture Techniques ,Mutation, Missense ,Biology ,QT interval ,Sodium Channels ,Electrocardiography ,Young Adult ,SCN3B ,SCN1B ,Physiology (medical) ,Internal medicine ,medicine ,Humans ,Missense mutation ,Genetic Predisposition to Disease ,Genetic Testing ,Child ,Brugada syndrome ,Sodium channel ,Middle Aged ,Voltage-Gated Sodium Channel beta-1 Subunit ,Sudden infant death syndrome ,medicine.disease ,Molecular biology ,Long QT Syndrome ,Endocrinology ,Female ,Electrophysiologic Techniques, Cardiac ,Cardiology and Cardiovascular Medicine - Abstract
BACKGROUND Long QT syndrome (LQTS) is associated with sudden cardiac death and the prolongation of the QT interval on the electrocardiogram. A comprehensive screening of all genes previously associated with this disease leaves 30% of the patients without a genetic diagnosis. Pathogenic mutations in the sodium channel β subunits have been associated with cardiac channelopathies, including SCN4B mutations in LQTS. OBJECTIVE To evaluate the role of mutations in the sodium channel β subunits in LQTS. METHODS We screened for mutations in the genes encoding the 5 sodium β subunits (SCN1B isoforms a and b, SCN2B, SCN3B, and SCN4B) from 30 nonrelated patients who were clinically diagnosed with LQTS without mutations in common LQTS-related genes. We used the patch-clamp technique to study the properties of sodium currents and the action potential duration in human embryonic kidney and HL-1 cells, respectively, in the presence of β1b subunits. RESULTS The genetic screening revealed a novel mutation in the SCN1Bb gene (β1bP213T) in an 8-year-old boy. Our electrophysiological analysis revealed that β1bP213T increases late sodium current. In addition, β1bP213T subtly altered Nav1.5 function by shifting the window current, accelerating recovery from inactivation, and decreasing the slow inactivation rate. Moreover, experiments using HL-1 cells revealed that the action potential duration significantly increases when the mutant β1b was overexpressed compared with β1bWT. CONCLUSION These data revealed SCN1Bb as a susceptibility gene responsible for LQTS, highlighting the importance of continuing the search for new genes and mechanisms to decrease the percentage of patients with LQTS remaining without genetic diagnosis.
- Published
- 2014
- Full Text
- View/download PDF
24. β-Adrenergic Pathway is Enhanced by Hormone-Induced Maturation of Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes (iPS-CM)
- Author
-
Rebecca Martinez-Moreno, Elisabet Selga, David Carreras, Fabiana S. Scornik, Guillermo J. Pérez, and Ramon Brugada
- Subjects
Chemistry ,Biophysics ,β adrenergic ,Induced pluripotent stem cell ,Cell biology ,Hormone - Published
- 2019
- Full Text
- View/download PDF
25. DiBAC4(3) hits a 'sweet spot' for the activation of arterial large-conductance Ca2+-activated potassium channels independently of the β1-subunit
- Author
-
Elisabet Selga, Fabiana S. Scornik, Ronald S. Bucciero, Ramon Brugada, Cristina Bosch Calero, Guillermo J. Pérez, and Yuesheng Wu
- Subjects
Membrane potential ,BK channel ,biology ,Physiology ,Chemistry ,Activator (genetics) ,Sodium channel ,Conductance ,Potassium channel ,Biochemistry ,Physiology (medical) ,biology.protein ,Biophysics ,Patch clamp ,Cardiology and Cardiovascular Medicine ,Selectivity - Abstract
The voltage-sensitive dye bis-(1,3-dibutylbarbituric acid)trimethine oxonol [DiBAC4(3)] has been reported as a novel large-conductance Ca2+-activated K+ (BK) channel activator with selectivity for its β1- or β4-subunits. In arterial smooth muscle, BK channels are formed by a pore-forming α-subunit and a smooth muscle-abundant regulatory β1-subunit. This tissue specificity has driven extensive pharmacological research aimed at regulating arterial tone. Using animals with a disruption of the gene for the β1-subunit, we explored the effects of DiBAC4(3) in native channels from arterial smooth muscle. We tested the hypothesis that, in native BK channels, activation by DiBAC4(3) relies mostly on its α-subunit. We studied BK channels from wild-type and transgenic β1-knockout mice in excised patches. BK channels from brain arteries, with or without the β1-subunit, were similarly activated by DiBAC4(3). In addition, we found that saturating concentrations of DiBAC4(3) (∼30 μM) promote an unprecedented persistent activation of the channel that negatively shifts its voltage dependence by as much as −300 mV. This “sweet spot” for persistent activation is independent of Ca2+ and/or the β1–4-subunits and is fully achieved when DiBAC4(3) is applied to the intracellular side of the channel. Arterial BK channel response to DiBAC4(3) varies across species and/or vascular beds. DiBAC4(3) unique effects can reveal details of BK channel gating mechanisms and help in the rational design of BK channel activators.
- Published
- 2013
- Full Text
- View/download PDF
26. A Missense Mutation in the Sodium Channel β2 Subunit RevealsSCN2Bas a New Candidate Gene for Brugada Syndrome
- Author
-
Helena Riuró, Elisabet Selga, Pedro Brugada, Guillermo J. Pérez, Francisco M. Vázquez, Anna Tarradas, Fabiana S. Scornik, Anna Iglesias, Marcel Verges, Pedro Beltran-Alvarez, Oscar Campuzano, Ramon Brugada, Josep Brugada, and Sara Pagans
- Subjects
Candidate gene ,Sodium ,Mutant ,Mutation, Missense ,chemistry.chemical_element ,Biology ,medicine.disease_cause ,Sodium Channels ,Sudden cardiac death ,Genetics ,medicine ,Humans ,Missense mutation ,Genetic Predisposition to Disease ,Genetics (clinical) ,Brugada Syndrome ,Brugada syndrome ,Mutation ,Voltage-Gated Sodium Channel beta-2 Subunit ,Sodium channel ,Middle Aged ,medicine.disease ,Death, Sudden, Cardiac ,chemistry ,Female - Abstract
Brugada Syndrome (BrS) is a familial disease associated with sudden cardiac death. A 20%-25% of BrS patients carry genetic defects that cause loss-of-function of the voltage-gated cardiac sodium channel. Thus, 70%-75% of patients remain without a genetic diagnosis. In this work, we identified a novel missense mutation (p.Asp211Gly) in the sodium β2 subunit encoded by SCN2B, in a woman diagnosed with BrS. We studied the sodium current (INa ) from cells coexpressing Nav 1.5 and wild-type (β2WT) or mutant (β2D211G) β2 subunits. Our electrophysiological analysis showed a 39.4% reduction in INa density when Nav 1.5 was coexpressed with the β2D211G. Single channel analysis showed that the mutation did not affect the Nav 1.5 unitary channel conductance. Instead, protein membrane detection experiments suggested that β2D211G decreases Nav 1.5 cell surface expression. The effect of the mutant β2 subunit on the INa strongly suggests that SCN2B is a new candidate gene associated with BrS.
- Published
- 2013
- Full Text
- View/download PDF
27. Large-conductance calcium-activated potassium current modulates excitability in isolated canine intracardiac neurons
- Author
-
Seth Anderson, Mayurika Desai, Guillermo J. Pérez, and Fabiana S. Scornik
- Subjects
medicine.medical_specialty ,BK channel ,Indoles ,Patch-Clamp Techniques ,Physiology ,Potassium ,Action Potentials ,chemistry.chemical_element ,Intracardiac injection ,Membrane Potentials ,Dogs ,Internal medicine ,Potassium Channel Blockers ,medicine ,Animals ,Large-Conductance Calcium-Activated Potassium Channels ,Patch clamp ,Neurons ,Membrane potential ,biology ,T-type calcium channel ,Tetraethylammonium ,Heart ,Ryanodine Receptor Calcium Release Channel ,Potassium channel blocker ,Articles ,Cell Biology ,Calcium-activated potassium channel ,Endocrinology ,chemistry ,Biophysics ,biology.protein ,Calcium ,Ganglia ,medicine.drug - Abstract
We studied principal neurons from canine intracardiac (IC) ganglia to determine whether large-conductance calcium-activated potassium (BK) channels play a role in their excitability. We performed whole cell recordings in voltage- and current-clamp modes to measure ion currents and changes in membrane potential from isolated canine IC neurons. Whole cell currents from these neurons showed fast- and slow-activated outward components. Both current components decreased in the absence of calcium and following 1–2 mM tetraethylammonium (TEA) or paxilline. These results suggest that BK channels underlie these current components. Single-channel analysis showed that BK channels from IC neurons do not inactivate in a time-dependent manner, suggesting that the dynamic of the decay of the fast current component is akin to that of intracellular calcium. Immunohistochemical studies showed that BK channels and type 2 ryanodine receptors are coexpressed in IC principal neurons. We tested whether BK current activation in these neurons occurred via a calcium-induced calcium release mechanism. We found that the outward currents of these neurons were not affected by the calcium depletion of intracellular stores with 10 mM caffeine and 10 μM cyclopiazonic acid. Thus, in canine intracardiac neurons, BK currents are directly activated by calcium influx. Membrane potential changes elicited by long (400 ms) current injections showed a tonic firing response that was decreased by TEA or paxilline. These data strongly suggest that the BK current present in canine intracardiac neurons regulates action potential activity and could increase these neurons excitability.
- Published
- 2013
- Full Text
- View/download PDF
28. Sodium channel current loss of function in induced pluripotent stem cell-derived cardiomyocytes from a Brugada syndrome patient
- Author
-
Elisabet, Selga, Franziska, Sendfeld, Rebecca, Martinez-Moreno, Claire N, Medine, Olga, Tura-Ceide, Sir Ian, Wilmut, Guillermo J, Pérez, Fabiana S, Scornik, Ramon, Brugada, and Nicholas L, Mills
- Subjects
Induced Pluripotent Stem Cells ,EB, embryoid body ,Article ,cTnI, cardiac troponin I ,NAV1.5 Voltage-Gated Sodium Channel ,Pluripotent stem cells ,INa, sodium current ,cTnT, cardiac troponin T ,Humans ,Biotinylation ,Myocytes, Cardiac ,Brugada syndrome ,TEM, transmission electron microscopy ,Cell Shape ,tsA201 cells, immortalized HEK293 cells ,Cardiomyocytes ,AFP, alpha-fetoprotein ,Base Sequence ,Cell Membrane ,iPS, induced pluripotent stem cells ,SNP, single nucleotide polymorphism ,LQT, long QT syndrome ,HEK, human embryonic kidney cells ,Electrophysiology ,HEK293 Cells ,CPVT, catecholaminergic polymorphic ventricular tachycardia ,Mutant Proteins ,iPS-CM, iPS cell-derived cardiomyocytes ,Sodium current ,Ion Channel Gating ,Biomarkers - Abstract
Brugada syndrome predisposes to sudden death due to disruption of normal cardiac ion channel function, yet our understanding of the underlying cellular mechanisms is incomplete. Commonly used heterologous expression models lack many characteristics of native cardiomyocytes and, in particular, the individual genetic background of a patient. Patient-specific induced pluripotent stem (iPS) cell-derived cardiomyocytes (iPS-CM) may uncover cellular phenotypical characteristics not observed in heterologous models. Our objective was to determine the properties of the sodium current in iPS-CM with a mutation in SCN5A associated with Brugada syndrome. Dermal fibroblasts from a Brugada syndrome patient with a mutation in SCN5A (c.1100G > A, leading to Nav1.5_p.R367H) were reprogrammed to iPS cells. Clones were characterized and differentiated to form beating clusters and sheets. Patient and control iPS-CM were structurally indistinguishable. Sodium current properties of patient and control iPS-CM were compared. These results were contrasted with those obtained in tsA201 cells heterologously expressing sodium channels with the same mutation. Patient-derived iPS-CM showed a 33.1–45.5% reduction in INa density, a shift in both activation and inactivation voltage-dependence curves, and faster recovery from inactivation. Co-expression of wild-type and mutant channels in tsA201 cells did not compromise channel trafficking to the membrane, but resulted in a reduction of 49.8% in sodium current density without affecting any other parameters. Cardiomyocytes derived from iPS cells from a Brugada syndrome patient with a mutation in SCN5A recapitulate the loss of function of sodium channel current associated with this syndrome; including pro-arrhythmic changes in channel function not detected using conventional heterologous expression systems., Highlights • iPS-CM were generated from a Brugada Syndrome patient who carries an SNV in SCN5A. • Patient-specific iPS-CM show a loss of function of the sodium current. • Use of iPS-CM uncovers changes in INa properties not apparent in tsA201 cells.
- Published
- 2016
29. A Common Single Nucleotide Polymorphism Can Exacerbate Long-QT Type 2 Syndrome Leading to Sudden Infant Death
- Author
-
Charles Antzelevitch, Moshe Gunsburg, Guillermo J. Pérez, Kirstine Calloe, Fabiana S. Scornik, Elena Burashnikov, Jonathan M. Cordeiro, Gabriel Caceres, Eyal Nof, and Barry Love
- Subjects
ERG1 Potassium Channel ,Heterozygote ,Modern medicine ,Long QT syndrome ,Nonsense mutation ,Physiology ,Genetic Counseling ,Single-nucleotide polymorphism ,CHO Cells ,Polymorphism, Single Nucleotide ,QT interval ,Asymptomatic ,Article ,Sudden cardiac death ,Young Adult ,Cricetulus ,Pregnancy ,Cricetinae ,Genetics ,medicine ,Animals ,Humans ,Genetics (clinical) ,biology ,Infant ,KCNE2 ,medicine.disease ,Ether-A-Go-Go Potassium Channels ,Pedigree ,Electrophysiology ,Long QT Syndrome ,Death, Sudden, Cardiac ,Codon, Nonsense ,biology.protein ,Female ,medicine.symptom ,Cardiology and Cardiovascular Medicine - Abstract
Background— Identification of infants at risk for sudden arrhythmic death remains one of the leading challenges of modern medicine. We present a family in which a common polymorphism (single nucleotide polymorphism) inherited from the father, combined with a stop codon mutation inherited from the mother (both asymptomatic), led to 2 cases of sudden infant death. Methods and Results— KCNQ1, KCNH2, SCN5A, KCNE1, KCNE2, CACNA1c, CACNB2b , and KCNJ2 genes were amplified and analyzed by direct sequencing. Functional electrophysiological studies were performed with the single nucleotide polymorphism and mutation expressed singly and in combination in Chinese ovary (CHO-K1) and COS-1 cells. An asymptomatic woman presenting after the death of her 2-day-old infant and spontaneous abortion of a second baby in the first trimester was referred for genetic analysis. The newborn infant had nearly incessant ventricular tachycardia while in utero and a prolonged QTc (560 ms). The mother was asymptomatic but displayed a prolonged QTc. Genetic screening of the mother revealed a heterozygous nonsense mutation (P926AfsX14) in KCNH2 , predicting a stop codon. The father was asymptomatic with a normal QTc but had a heterozygous polymorphism (K897T) in KCNH2 . The baby who died at 2 days of age and the aborted fetus inherited both K897T and P926AfsX14. Heterologous coexpression of K897T and P926AfsX14 led to loss of function of HERG current much greater than expression of K897T or P926AfsX14 alone. Conclusions— Our data suggest that a common polymorphism (K897T) can markedly accentuate the loss of function of mildly defective HERG channels, leading to long-QT syndrome–mediated arrhythmias and sudden infant death.
- Published
- 2010
- Full Text
- View/download PDF
30. An SCN1B Variant Found in a Child Diagnosed with Epilepsy and Brugada Syndrome Modifies Brain-Type (NaV1.1) and Cardiac-Type (NaV1.5) Sodium Currents
- Author
-
Guillermo J. Pérez, Helena Riuró, Fabiana S. Scornik, Ramon Brugada, Michael F. Wangler, Rebecca Martinez-Moreno, and Elisabeth Selga
- Subjects
medicine.medical_specialty ,Epilepsy ,Endocrinology ,business.industry ,SCN1B ,Internal medicine ,Biophysics ,medicine ,medicine.disease ,business ,Sodium current ,Brugada syndrome - Published
- 2018
- Full Text
- View/download PDF
31. High sensitivity of the sheep pulmonary vein antrum to acetylcholine stimulation
- Author
-
Guillermo J. Pérez, Yangyang Zhang, Qin Pan, Pedro Brugada, Ramon Brugada, Yi Liu, Li Li, Jun Li, Cui-Mei Zhao, Huaizhi Chen, Lei Liang, Yi-Han Chen, Jie Qi, Kui Hong, Luying Peng, Cardio-vascular diseases, and Internal Medicine Specializations
- Subjects
medicine.medical_specialty ,Refractory Period, Electrophysiological ,Physiology ,Vasodilator Agents ,Action Potentials ,Fluorescent Antibody Technique ,Cholinergic Agonists ,In Vitro Techniques ,Muscarinic Agonists ,Muscle, Smooth, Vascular ,Pulmonary vein ,Dose-Response Relationship ,Physiology (medical) ,Internal medicine ,Muscarinic acetylcholine receptor ,medicine ,Animals ,Myocytes, Cardiac ,Heart Atria ,Antrum ,Acetylcholine receptor ,Receptor, Muscarinic M2 ,Sheep ,Dose-Response Relationship, Drug ,business.industry ,Effective refractory period ,Atrial fibrillation ,Anatomy ,medicine.disease ,Acetylcholine ,Electrophysiology ,Pulmonary Veins ,drug effects ,Circulatory system ,cardiovascular system ,Cardiology ,Carbachol ,business ,Microelectrodes ,medicine.drug - Abstract
Isolation of the pulmonary vein antrum can terminate atrial fibrillation, but the rationale has not been elucidated. In the present study, we show that sheep atrial effective refractory period (ERP) was heterogeneously shortened by acetylcholine administration. After perfusion with 15 μM acetylcholine, the shortest ERP occurred in the pulmonary vein antrum, which was recorded with the standard intracellular microelectrode technique (the ERP results in the pulmonary vein antrum, left atrial posterior wall, roof, free wall and appendage, and right atrial free wall were 52.0 ± 1.6, 75.1 ± 2.0, 77.2 ± 1.7, 85.6 ± 1.7, 64.3 ± 2.1, and 90.5 ± 1.3 ms, respectively; P < 0.05). Immunofluorescent staining revealed that muscarinic type 2 receptors (M2R) were also distributed heterogeneously in the atrial myocardium, with the highest density in the antrum (the relative fluorescent intensity results of the M2R in the pulmonary vein antrum, left atrial posterior wall, roof, free wall and appendage, and right atrial free wall were 62.64 ± 2.56, 53.12 ± 2.76, 51.83 ± 2.45, 47.90 ± 2.33, 55.27 ± 2.08, and 45.53 ± 2.02, respectively; P < 0.05), which was in accordance with the heterogeneity of ERP distribution. Thus the pulmonary vein antrum is a unique electrophysiological region with high sensitivity to acetylcholine, and its intensive response to acetylcholine is most likely associated with the dense M2R distribution of this region. Such an acetylcholine-induced ERP heterogeneity is possibly a substrate for atrial fibrillation and hence one of the potential electrophysiological bases for the isolation therapy.
- Published
- 2008
- Full Text
- View/download PDF
32. Contribution of Cardiac Sodium Channel beta-Subunit Variants to Brugada Syndrome
- Author
-
Helena Riuró, Guillermo J. Pérez, Sonia Van Dooren, Evrim Komurcu-Bayrak, Maryse Bonduelle, Dorien Daneels, Ramon Brugada, Gudrun Pappaert, Fabiana S. Scornik, Karine Breckpot, Sara Pagans, Uschi Peeters, Sophie Van Malderen, Pedro Brugada, Anna Tarradas, Psychiatry, Clinical sciences, Reproduction and Genetics, Internal Medicine Specializations, Heartrhythmmanagement, Laboratory of Molecullar and Cellular Therapy, Basic (bio-) Medical Sciences, and Cardio-vascular diseases
- Subjects
Adult ,Male ,medicine.medical_specialty ,In silico ,Population ,Sudden cardiac death ,NAV1.5 Voltage-Gated Sodium Channel ,SCN1-4B ,Internal medicine ,genomics ,Medicine ,Humans ,SCN1B ,SCN3B ,variant identification ,education ,Gene ,Brugada syndrome ,Aged ,Brugada Syndrome ,Medicine(all) ,Genetics ,education.field_of_study ,business.industry ,Brugada ,Sodium channel ,fungi ,General Medicine ,Middle Aged ,medicine.disease ,Penetrance ,Phenotype ,HEK293 Cells ,Mutation ,Cardiology ,Voltage-Gated Sodium Channel beta Subunits ,Female ,SCN2B ,SCN4B ,Cardiology and Cardiovascular Medicine ,business - Abstract
Background: Brugada syndrome (BrS) is an inheritable cardiac disease associated with syncope, malignant ventricular arrhythmias and sudden cardiac death. The largest proportion of mutations in BrS is found in the SCN5A gene encoding the α-subunit of cardiac sodium channels (Nav1.5). Causal SCN5A mutations are present in 18–30% of BrS patients. The additional genetic diagnostic yield of variants in cardiac sodium channel β-subunits in BrS patients was explored and functional studies on 3 novel candidate variants were performed. Methods and Results: The SCN1B-SCN4B genes were screened, which encode the 5 sodium channel β-subunits, in a SCN5A negative BrS population (n=74). Five novel variants were detected; in silico pathogenicity prediction classified 4 variants as possibly disease causing. Three variants were selected for functional study. These variants caused only limited alterations of Nav1.5 function. Next generation sequencing of a panel of 88 arrhythmia genes could not identify other major causal mutations. Conclusions: It was hypothesized that the studied variants are not the primary cause of BrS in these patients. However, because small functional effects of these β-subunit variants can be discriminated, they might contribute to the BrS phenotype and be considered a risk factor. The existence of these risk factors can give an explanation to the reduced penetrance and variable expressivity seen in this syndrome. We therefore recommend including the SCN1-4B genes in a next generation sequencing-based gene panel.
- Published
- 2015
33. Functional expression of 'cardiac-type' Nav1.5 sodium channel in canine intracardiac ganglia
- Author
-
Charles Antzelevitch, Ramon Brugada, Guido D. Pollevick, Guillermo J. Pérez, Alejandra Guerchicoff, Fabiana S. Scornik, and Mayurika Desai
- Subjects
medicine.medical_specialty ,Patch-Clamp Techniques ,Voltage clamp ,Action Potentials ,Gene Expression ,Muscle Proteins ,Tetrodotoxin ,In Vitro Techniques ,Nav1.5 ,NAV1.5 Voltage-Gated Sodium Channel ,Article ,Sodium Channels ,Intracardiac injection ,chemistry.chemical_compound ,Dogs ,Physiology (medical) ,Internal medicine ,medicine ,Animals ,Patch clamp ,Ganglia, Autonomic ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,musculoskeletal, neural, and ocular physiology ,Sodium channel ,Heart ,Immunohistochemistry ,Cell biology ,Autonomic nervous system ,Endocrinology ,nervous system ,chemistry ,biology.protein ,RNA ,Cardiology and Cardiovascular Medicine ,Signal Transduction - Abstract
Background The autonomic nervous system has been implicated in several arrhythmogenic diseases, including long QT syndrome type 3 (LQT3) and Brugada syndrome. Scarce information on the cellular components of the intrinsic cardiac ganglia from higher mammals has limited our understanding of the role of the autonomic nervous system in such diseases. Objectives The purpose of this study was to isolate and characterize the electrophysiologic properties of canine intracardiac neurons. Methods Action potentials (APs) and ionic currents were studied in enzymatically dissociated canine intracardiac neurons under current and voltage clamp conditions. Immunohistochemical and reverse transcription-polymerase chain reaction analysis was performed using freshly isolated intracardiac ganglia. Results APs recorded from intracardiac neurons displayed a tetrodotoxin-resistant (TTX-R) component. TTX-R APs were abolished in the absence of sodium but persisted in the absence of external calcium. Immunohistochemical studies showed the presence of TTX-R sodium channels in these ganglia. Sodium currents were characterized by two components with different affinities for TTX: a tetrodotoxin-sensitive (TTX-S) component and a TTX-R component. TTX-S current inactivation was characteristic of neuronal sodium currents, whereas TTX-R current inactivation time constants were similar to those previously reported for Na v 1.5 channels. TTX sensitivity (IC 50 = 1.17 μM) of the TTX-R component was in the range reported for Na v 1.5 channels. Expression of Na v 1.5 channels in intracardiac ganglia was confirmed by PCR analysis and sequencing. Conclusion Our results suggest that canine intracardiac neurons functionally express Na v 1.5 channels. These findings open an exciting new door to our understanding of autonomically modulated arrhythmogenic diseases linked to mutations in Na v 1.5 channels, including Brugada syndrome and LQT3.
- Published
- 2006
- Full Text
- View/download PDF
34. Dual Effect of Tamoxifen on Arterial KCa Channels Does Not Depend on the Presence of the β1 Subunit
- Author
-
Guillermo J. Pérez
- Subjects
Male ,medicine.medical_specialty ,Potassium Channels ,Time Factors ,Antineoplastic Agents, Hormonal ,Genotype ,Myocytes, Smooth Muscle ,Cerebral arteries ,Polymerase Chain Reaction ,Biochemistry ,Inhibitory Concentration 50 ,Mice ,Internal medicine ,β1 subunit ,medicine ,Animals ,Myocyte ,Molecular Biology ,IC50 ,Mice, Knockout ,Binding Sites ,Dose-Response Relationship, Drug ,Chemistry ,Cell Membrane ,Dual effect ,Conductance ,Estrogens ,Muscle, Smooth ,Arteries ,Cell Biology ,Cerebral Arteries ,Protein Structure, Tertiary ,Mice, Inbred C57BL ,Kinetics ,Tamoxifen ,Endocrinology ,Linear Models ,Calcium ,Female ,Ca2 channels ,Software ,Protein Binding ,medicine.drug - Abstract
Tamoxifen has been reported to directly activate large conductance calcium-activated potassium (KCa) channels through the KCa beta1 subunit, suggesting a cardio-protective role of this compound. The present study using knock-out (KO) mice for the KCa channel beta1 subunit was aimed at understanding the molecular mechanisms of the effects of tamoxifen on arterial smooth muscle KCa channels. Single channel studies were conducted in excised patches from cerebral artery myocytes from both wild-type and KO animals. The present data demonstrated that tamoxifen can inhibit arterial KCa channels due to a major decrease in channel open probability (P(o)), a mechanism different from the reduction in single channel amplitude reported previously and also observed in the present work. A tamoxifen-induced decrease in P(o) was present in arterial KCa channels from both wild-type and beta1 KO animals. This inhibition was concentration-dependent and partially reversible with a half-maximal concentration constant IC(50) of 2.6 microm. The effect of tamoxifen was actually dual Single channel kinetic analysis showed that tamoxifen shortens both mean closed time and mean open time; the latter is probably due to an intermediate duration voltage-independent blocking mechanism. Thus, tamoxifen block would predominate when KCa channel P(o) is0.1-0.2, limiting the maximum P(o), whereas a leftward shift in voltage or Ca(2+) activation curves can be observed for P(o) values lower than those values. This dual effect of tamoxifen appears to be independent of the beta1 subunit. The molecular specificity of tamoxifen, or eventually other xenoestrogen derivatives, for the KCa channel beta1 subunit is uncertain.
- Published
- 2005
- Full Text
- View/download PDF
35. Electrophysiologic Properties and Antiarrhythmic Actions of a Novel Antianginal Agent
- Author
-
Wu L, Andrew C. Zygmunt, Fabiana S. Scornik, Robert J. Goodrow, Guillermo J. Pérez, Jonathan M. Cordeiro, Di Diego Jm, Luiz Belardinelli, Alexander Burashnikov, Jeffrey M. Fish, Fraser H, and Charles Antzelevitch
- Subjects
medicine.medical_specialty ,Action Potentials ,Ranolazine ,030204 cardiovascular system & hematology ,Pharmacology ,030226 pharmacology & pharmacy ,QT interval ,Ion Channels ,Piperazines ,Angina Pectoris ,03 medical and health sciences ,0302 clinical medicine ,Torsades de Pointes ,Internal medicine ,Heart rate ,Animals ,Humans ,Medicine ,Repolarization ,Pharmacology (medical) ,business.industry ,Prolongation ,Stereoisomerism ,Long QT Syndrome ,Blood pressure ,Plasma concentration ,cardiovascular system ,Cardiology ,Action potential duration ,Acetanilides ,Electrophysiologic Techniques, Cardiac ,Cardiology and Cardiovascular Medicine ,business ,medicine.drug - Abstract
Ranolazine is a novel antianginal agent capable of producing anti-ischemic effects at plasma concentrations of 2 to 6 microM without a significant reduction of heart rate or blood pressure. This review summarizes the electrophysiologic properties of ranolazine. Ranolazine significantly blocks I(Kr) (IC(50) = 12 microM), late I(Na), late I(Ca), peak I(Ca), I(Na-Ca) (IC(50) = 5.9, 50, 296, and 91 microM, respectively) and I(Ks) (17% at 30 microM), but causes little or no inhibition of I(to) or I(K1). In left ventricular tissue and wedge preparations, ranolazine produces a concentration-dependent prolongation of action potential duration (APD) in epicardium, but abbreviation of APD of M cells, leading to either no change or a reduction in transmural dispersion of repolarization (TDR). The result is a modest prolongation of the QT interval. Prolongation of APD and QT by ranolazine is fundamentally different from that of other drugs that block I(Kr) and induce torsade de pointes in that APD prolongation is rate-independent (ie, does not display reverse rate-dependent prolongation of APD) and is not associated with early after depolarizations, triggered activity, increased spatial dispersion of repolarization, or polymorphic ventricular tachycardia. Torsade de pointes arrhythmias were not observed spontaneously nor could they be induced with programmed electrical stimulation in the presence of ranolazine at concentrations as high as 100 microM. Indeed, ranolazine was found to possess significant antiarrhythmic activity, acting to suppress the arrhythmogenic effects of other QT-prolonging drugs. Ranolazine produces ion channel effects similar to those observed after chronic exposure to amiodarone (reduced late I(Na), I(Kr), I(Ks), and I(Ca)). Ranolazine's actions to reduce TDR and suppress early after depolarization suggest that in addition to its anti-anginal actions, the drug possesses antiarrhythmic activity.
- Published
- 2004
- Full Text
- View/download PDF
36. Ionic and Cellular Basis for the Predominance of the Brugada Syndrome Phenotype in Males
- Author
-
Jeffrey M. Fish, José M. Di Diego, Andrew C. Zygmunt, Fabiana S. Scornik, Robert J. Goodrow, Jonathan M. Cordeiro, Charles Antzelevitch, and Guillermo J. Pérez
- Subjects
Male ,medicine.medical_specialty ,Heart disease ,Heart block ,Action Potentials ,Biology ,Dogs ,Sex Factors ,Culture Techniques ,Physiology (medical) ,Internal medicine ,medicine ,Animals ,Myocyte ,Cells, Cultured ,Brugada syndrome ,Bundle branch block ,Pinacidil ,Arrhythmias, Cardiac ,Heart ,Syndrome ,medicine.disease ,Phenotype ,Penetrance ,Kinetics ,Electrophysiology ,Endocrinology ,Female ,Terfenadine ,Cardiology and Cardiovascular Medicine ,Pericardium - Abstract
Background— The Brugada syndrome displays an autosomal dominant mode of transmission with low penetrance. Despite equal genetic transmission of the disease, the clinical phenotype is 8 to 10 times more prevalent in males than in females. The basis for this intriguing sex-related distinction is unknown. The present study tests the hypothesis that the disparity in expression of the Brugada phenotype is a result of a more prominent I to -mediated action potential notch in the right ventricular (RV) epicardium of males versus females. Methods and Results— We studied epicardial tissue slices, arterially perfused wedge preparations, and dissociated epicardial myocytes isolated from male and female canine hearts. RV epicardium action potential phase 1 amplitude was 64.8±2.0% of that of phase 2 in males compared with 73.8±4.4% in females ( P I to density was 26% smaller and time constant for inactivation 17% smaller at +40 mV in female versus male RV epicardial cells ( P I to , including the voltage dependence of inactivation and time course of reactivation, were no different between the sexes. Pinacidil caused loss of action potential dome in male, but not female, RV epicardial tissue slices. Terfenadine (5 μmol/L) induced phase 2 reentry in 6 of 7 male but only 2 of 7 female arterially perfused wedge preparations. Two of 6 male and 1 of 2 female preparations developed polymorphic ventricular tachycardia/ventricular fibrillation. Conclusions— Our results suggest that the predominance of the Brugada phenotype in males is a result of the presence of a more prominent I to in males versus females.
- Published
- 2002
- Full Text
- View/download PDF
37. Ca 2+ Sparks and Their Function in Human Cerebral Arteries
- Author
-
Adrian D. Bonev, Christine M. Saundry, Paul L. Penar, George C. Wellman, Mark T. Nelson, Guillermo J. Pérez, Bruce I. Tranmer, and David J. Nathan
- Subjects
Adult ,Male ,BK channel ,Patch-Clamp Techniques ,Cerebral arteries ,In Vitro Techniques ,Muscle, Smooth, Vascular ,Potassium Channels, Calcium-Activated ,Isometric Contraction ,Potassium Channel Blockers ,Humans ,Myocyte ,Medicine ,Calcium Signaling ,Large-Conductance Calcium-Activated Potassium Channels ,Patch clamp ,Aged ,Fluorescent Dyes ,Aged, 80 and over ,Advanced and Specialized Nursing ,Microscopy, Confocal ,biology ,Ryanodine ,Ryanodine receptor ,business.industry ,Anatomy ,Cerebral Arteries ,Middle Aged ,Iberiotoxin ,Potassium channel ,Electrophysiology ,Vasoconstriction ,Circulatory system ,Potassium ,Biophysics ,biology.protein ,Female ,Neurology (clinical) ,Cardiology and Cardiovascular Medicine ,business - Abstract
Background and Purpose — Local Ca 2+ release events (Ca 2+ sparks) caused by the opening of ryanodine-sensitive Ca 2+ channels in the sarcoplasmic reticulum have been suggested to oppose constriction in cerebral arteries through the activation of large-conductance Ca 2+ -activated K + (BK) channels. We report the first identification and characterization of Ca 2+ sparks and associated BK channel currents in smooth muscle cells isolated from human cerebral arteries. Methods — Membrane currents and intracellular Ca 2+ were measured with the use of the patch-clamp technique and laser scanning confocal microscopy. Results — Ca 2+ sparks with a peak fractional fluorescence change (F/F 0 ) of 2.02±0.04 and size of 8.2±0.5 μm 2 (n=108) occurred at a frequency of approximately 1 Hz in freshly isolated, cerebral artery myocytes from humans. At a holding potential of −40 mV, the majority of, but not all, Ca 2+ sparks (61 of 85 sparks) were associated with transient BK currents. Consistent with a role for Ca 2+ sparks in the control of cerebral artery diameter, agents that block Ca 2+ sparks (ryanodine) or BK channels (iberiotoxin) were found to contract human cerebral arteries. Conclusions — This study provides evidence for local Ca 2+ signaling in human arterial myocytes and suggests that these events may play an important role in control of cerebral artery diameter in humans.
- Published
- 2002
- Full Text
- View/download PDF
38. EFICACY AND SAFETY OF FLECAINIDE TO REDUCE EXERCISE-INDUCED VENTRICULAR ARRHYTHMIAS AND THE ARRHYTHMIC WINDOW IN PATIENTS WITH CATECHOLAMINERGIC POLYMORPHIC VENTRICULAR TACHYCARDIA
- Author
-
María del Val Groba Marco, Josep Brugada, Eduardo Caballero Dorta, Luis Álvarez Acosta, Fernando Wangüemert Pérez, Guillermo J. Pérez, Ramon Brugada-Terradellas, Oscar Campuzano Larrea, and Julio Salvador Hernández Afonso
- Subjects
medicine.medical_specialty ,business.industry ,Emotional stress ,Catecholaminergic polymorphic ventricular tachycardia ,medicine.disease ,Internal medicine ,cardiovascular system ,medicine ,Cardiology ,In patient ,cardiovascular diseases ,Inherited disease ,Cardiology and Cardiovascular Medicine ,business ,Beta (finance) ,Flecainide ,Normal heart ,medicine.drug - Abstract
Background: Catecholaminergic polymorphic ventricular tachycardia (CPVT) is an inherited disease characterized by ventricular arrhythmias (VA) triggered in a situation of physical or emotional stress in young people with structurally normal heart. The cornerstone of the treatment are the beta
- Published
- 2017
- Full Text
- View/download PDF
39. Micromolar Ca2+ from sparks activates Ca2+-sensitive K+ channels in rat cerebral artery smooth muscle
- Author
-
Guillermo J. Pérez, Mark T. Nelson, and Adrian D. Bonev
- Subjects
Male ,BK channel ,medicine.medical_specialty ,Patch-Clamp Techniques ,Potassium Channels ,Physiology ,Cerebral arteries ,Muscle, Smooth, Vascular ,Rats, Sprague-Dawley ,Potassium Channels, Calcium-Activated ,Internal medicine ,medicine ,Animals ,Myocyte ,Calcium Signaling ,Large-Conductance Calcium-Activated Potassium Channels ,Patch clamp ,Egtazic Acid ,Ion transporter ,Chelating Agents ,Membrane potential ,Microscopy, Confocal ,biology ,Ryanodine receptor ,Chemistry ,Cell Biology ,Cerebral Arteries ,Rats ,Endocrinology ,medicine.anatomical_structure ,biology.protein ,Biophysics ,Calcium ,Female ,Blood vessel - Abstract
The goal of the present study was to test the hypothesis that local Ca2+ release events (Ca2+ sparks) deliver high local Ca2+concentration to activate nearby Ca2+-sensitive K+ (BK) channels in the cell membrane of arterial smooth muscle cells. Ca2+ sparks and BK channels were examined in isolated myocytes from rat cerebral arteries with laser scanning confocal microscopy and patch-clamp techniques. BK channels had an apparent dissociation constant for Ca2+ of 19 μM and a Hill coefficient of 2.9 at −40 mV. At near-physiological intracellular Ca2+ concentration ([Ca2+]i; 100 nM) and membrane potential (−40 mV), the open probability of a single BK channel was low (1.2 × 10−6). A Ca2+spark increased BK channel activity to 18. Assuming that 1–100% of the BK channels are activated by a single Ca2+ spark, BK channel activity increases 6 × 105-fold to 6 × 103-fold, which corresponds to ∼30 μM to 4 μM spark Ca2+ concentration. 1,2-bis(2-aminophenoxy)ethane- N,N,N′,N′-tetraacetic acid acetoxymethyl ester caused the disappearance of all Ca2+sparks while leaving the transient BK currents unchanged. Our results support the idea that Ca2+ spark sites are in close proximity to the BK channels and that local [Ca2+]i reaches micromolar levels to activate BK channels.
- Published
- 2001
- Full Text
- View/download PDF
40. Vasoregulation by the β1 subunit of the calcium-activated potassium channel
- Author
-
Jon C. Kosek, Delrae M. Eckman, Mark T. Nelson, Adrian D. Bonev, Andrew J. Patterson, Richard W. Aldrich, Robert Brenner, Steven W. Wiler, and Guillermo J. Pérez
- Subjects
Male ,BK channel ,medicine.medical_specialty ,Potassium Channels ,P-type calcium channel ,chemistry.chemical_element ,Blood Pressure ,N-type calcium channel ,Calcium ,Muscle, Smooth, Vascular ,Mice ,Potassium Channels, Calcium-Activated ,Internal medicine ,medicine ,Animals ,Calcium Signaling ,Large-Conductance Calcium-Activated Potassium Channels ,RNA, Messenger ,Mice, Knockout ,Multidisciplinary ,biology ,Voltage-dependent calcium channel ,T-type calcium channel ,Cerebral Arteries ,Calcium-activated potassium channel ,Mice, Inbred C57BL ,Vasodilation ,R-type calcium channel ,Endocrinology ,chemistry ,Vasoconstriction ,Gene Targeting ,biology.protein ,Biophysics ,Female - Abstract
Small arteries exhibit tone, a partially contracted state that is an important determinant of blood pressure. In arterial smooth muscle cells, intracellular calcium paradoxically controls both contraction and relaxation. The mechanisms by which calcium can differentially regulate diverse physiological responses within a single cell remain unresolved. Calcium-dependent relaxation is mediated by local calcium release from the sarcoplasmic reticulum. These 'calcium sparks' activate calcium-dependent potassium (BK) channels comprised of alpha and beta1 subunits. Here we show that targeted deletion of the gene for the beta1 subunit leads to a decrease in the calcium sensitivity of BK channels, a reduction in functional coupling of calcium sparks to BK channel activation, and increases in arterial tone and blood pressure. The beta1 subunit of the BK channel, by tuning the channel's calcium sensitivity, is a key molecular component in translating calcium signals to the central physiological function of vasoregulation.
- Published
- 2000
- Full Text
- View/download PDF
41. Ca2+channels, ryanodine receptors and Ca2+-activated K+channels: a functional unit for regulating arterial tone
- Author
-
Thomas Kleppisch, Adrian D. Bonev, Thomas J. Heppner, Andra S. Stevenson, Michael Rubart, Harm J. Knot, Mark T. Nelson, Valerie A. Porter, George C. Wellman, W. J. Lederer, Jonathan H. Jaggar, Maik Gollasch, and Guillermo J. Pérez
- Subjects
Membrane potential ,medicine.medical_specialty ,Vascular smooth muscle ,Physiology ,Ryanodine receptor ,Chemistry ,Depolarization ,Cardiac action potential ,Membrane hyperpolarization ,Hyperpolarization (biology) ,musculoskeletal system ,Endocrinology ,Internal medicine ,cardiovascular system ,medicine ,Biophysics ,Repolarization - Abstract
Local calcium transients ('Ca2+ sparks') are thought to be elementary Ca2+ signals in heart, skeletal and smooth muscle cells. Ca2+ sparks result from the opening of a single, or the coordinated opening of many, tightly clustered ryanodine receptor (RyR) channels in the sarcoplasmic reticulum (SR). In arterial smooth muscle, Ca2+ sparks appear to be involved in opposing the tonic contraction of the blood vessel. Intravascular pressure causes a graded membrane potential depolarization to approximately -40 mV, an elevation of arterial wall [Ca2+]i and contraction ('myogenic tone') of arteries. Ca2+ sparks activate calcium-sensitive K+ (KCa) channels in the sarcolemmal membrane to cause membrane hyperpolarization, which opposes the pressure induced depolarization. Thus, inhibition of Ca2+ sparks by ryanodine, or of KCa channels by iberiotoxin, leads to membrane depolarization, activation of L-type voltage-gated Ca2+ channels, and vasoconstriction. Conversely, activation of Ca2+ sparks can lead to vasodilation through activation of KCa channels. Our recent work is aimed at studying the properties and roles of Ca2+ sparks in the regulation of arterial smooth muscle function. The modulation of Ca2+ spark frequency and amplitude by membrane potential, cyclic nucleotides and protein kinase C will be explored. The role of local Ca2+ entry through voltage-dependent Ca2+ channels in the regulation of Ca2+ spark properties will also be examined. Finally, using functional evidence from cardiac myocytes, and histological evidence from smooth muscle, we shall explore whether Ca2+ channels, RyR channels, and KCa channels function as a coupled unit, through Ca2+ and voltage, to regulate arterial smooth muscle membrane potential and vascular tone.
- Published
- 1998
- Full Text
- View/download PDF
42. The smooth muscle-type β1 subunit potentiates activation by DiBAC4(3) in recombinant BK channels
- Author
-
Guillermo J. Pérez, Elisabet Selga, Ramon Brugada, Cristina Bosch Calero, and Fabiana S. Scornik
- Subjects
BK channel ,Protein subunit ,Biophysics ,Cardiovascular pharmacology ,Biochemistry ,law.invention ,Cell Line ,Mice ,Smooth muscle ,law ,β1 subunit ,Animals ,Humans ,Large-Conductance Calcium-Activated Potassium Channels ,biology ,Chemistry ,Muscle, Smooth ,Isoxazoles ,Deactivation kinetics ,Recombinant Proteins ,Article Addendum ,Protein Subunits ,Cell culture ,Barbiturates ,biology.protein ,Recombinant DNA - Abstract
Large-conductance Ca(2+)-activated (BK) channels, expressed in a variety of tissues, play a fundamental role in regulating and maintaining arterial tone. We recently demonstrated that the slow voltage indicator DiBAC4(3) does not depend, as initially proposed, on the β 1 or β 4 subunits to activate native arterial smooth muscle BK channels. Using recombinant mslo BK channels, we now show that the β 1 subunit is not essential to this activation but exerts a large potentiating effect. DiBAC4(3) promotes concentration-dependent activation of BK channels and slows deactivation kinetics, changes that are independent of Ca(2+). Kd values for BK channel activation by DiBAC4(3) in 0 mM Ca(2+) are approximately 20 μM (α) and 5 μM (α+β 1), and G-V curves shift up to -40 mV and -110 mV, respectively. β1 to β2 mutations R11A and C18E do not interfere with the potentiating effect of the subunit. Our findings should help refine the role of the β 1 subunit in cardiovascular pharmacology.
- Published
- 2013
43. Protein arginine methyl transferases-3 and -5 increase cell surface expression of cardiac sodium channel
- Author
-
Ralf Schmauder, Pedro Beltran-Alvarez, Alexsandra Espejo, Félix Pérez-Villa, Carlos Beltrán, Fabiana S. Scornik, Klaus Benndorf, Ramon Brugada, Thomas Linke, Ralf Mrowka, Montserrat Batlle, Thomas Zimmer, Sara Pagans, and Guillermo J. Pérez
- Subjects
Protein-Arginine N-Methyltransferases ,Methyltransferase ,Patch-Clamp Techniques ,Arginine ,Sodium ,Biophysics ,chemistry.chemical_element ,Nav1.5 ,Biochemistry ,NAV1.5 Voltage-Gated Sodium Channel ,Transient receptor potential channel ,Arginine methylation ,Structural Biology ,Genetics ,Fluorescence Resonance Energy Transfer ,Humans ,Molecular Biology ,Cells, Cultured ,biology ,Sodium channel ,Chemistry ,Myocardium ,HEK 293 cells ,Cell Membrane ,Cell Biology ,Methylation ,biology.protein ,Post-translational modification ,Ion channel - Abstract
The α-subunit of the cardiac voltage-gated sodium channel (NaV1.5) plays a central role in cardiomyocyte excitability. We have recently reported that NaV1.5 is post-translationally modified by arginine methylation. Here, we aimed to identify the enzymes that methylate NaV1.5, and to describe the role of arginine methylation on NaV1.5 function. Our results show that protein arginine methyl transferase (PRMT)-3 and -5 methylate NaV1.5 in vitro, interact with NaV1.5 in human embryonic kidney (HEK) cells, and increase NaV1.5 current density by enhancing NaV1.5 cell surface expression. Our observations are the first evidence of regulation of a voltage-gated ion channel, including calcium, potassium, sodium and TRP channels, by arginine methylation.
- Published
- 2013
44. DiBAC₄(3) hits a 'sweet spot' for the activation of arterial large-conductance Ca²⁺-activated potassium channels independently of the β₁-subunit
- Author
-
Fabiana S, Scornik, Ronald S, Bucciero, Yuesheng, Wu, Elisabet, Selga, Cristina, Bosch Calero, Ramon, Brugada, and Guillermo J, Pérez
- Subjects
Mice, Knockout ,Patch-Clamp Techniques ,Large-Conductance Calcium-Activated Potassium Channel beta Subunits ,Vascular Biology and Microcirculation ,Arteries ,Isoxazoles ,Real-Time Polymerase Chain Reaction ,Muscle, Smooth, Vascular ,Sodium Channels ,Membrane Potentials ,Mice, Inbred C57BL ,Kinetics ,Mice ,Dogs ,Data Interpretation, Statistical ,Barbiturates ,Animals ,Large-Conductance Calcium-Activated Potassium Channels - Abstract
The voltage-sensitive dye bis-(1,3-dibutylbarbituric acid)trimethine oxonol [DiBAC4(3)] has been reported as a novel large-conductance Ca2+-activated K+ (BK) channel activator with selectivity for its β1- or β4-subunits. In arterial smooth muscle, BK channels are formed by a pore-forming α-subunit and a smooth muscle-abundant regulatory β1-subunit. This tissue specificity has driven extensive pharmacological research aimed at regulating arterial tone. Using animals with a disruption of the gene for the β1-subunit, we explored the effects of DiBAC4(3) in native channels from arterial smooth muscle. We tested the hypothesis that, in native BK channels, activation by DiBAC4(3) relies mostly on its α-subunit. We studied BK channels from wild-type and transgenic β1-knockout mice in excised patches. BK channels from brain arteries, with or without the β1-subunit, were similarly activated by DiBAC4(3). In addition, we found that saturating concentrations of DiBAC4(3) (∼30 μM) promote an unprecedented persistent activation of the channel that negatively shifts its voltage dependence by as much as −300 mV. This “sweet spot” for persistent activation is independent of Ca2+ and/or the β1–4-subunits and is fully achieved when DiBAC4(3) is applied to the intracellular side of the channel. Arterial BK channel response to DiBAC4(3) varies across species and/or vascular beds. DiBAC4(3) unique effects can reveal details of BK channel gating mechanisms and help in the rational design of BK channel activators.
- Published
- 2013
45. Sodium Current Measurements in HEK293 Cells
- Author
-
Guillermo J. Pérez, Elisabet Selga, Helena Riuró, Anna Tarradas, Ramon Brugada, and Fabiana S. Scornik
- Subjects
chemistry ,Strategy and Management ,Mechanical Engineering ,Sodium ,HEK 293 cells ,Metals and Alloys ,Biophysics ,chemistry.chemical_element ,Patch clamp ,Whole cell current ,Industrial and Manufacturing Engineering ,Ion channel ,Sodium current - Published
- 2013
- Full Text
- View/download PDF
46. IMPLANTABLE CARDIOVERTER DEFIBRILLATOR IN CATECHOLAMINERGIC POLYMORPHIC VENTRICULAR TACHYCARDIA
- Author
-
Pablo M. Ruiz Hernandez, Josep Brugada, Fernando Wangüemert Pérez, Ramon Brugada, Oscar Campuzano Larrea, Guillermo J. Pérez, and Julio Hernandez
- Subjects
medicine.medical_specialty ,business.industry ,Incidence (epidemiology) ,medicine.medical_treatment ,Genetic disorder ,Disease ,medicine.disease ,Implantable cardioverter-defibrillator ,Catecholaminergic polymorphic ventricular tachycardia ,Sudden cardiac death ,Internal medicine ,cardiovascular system ,Cardiology ,Medicine ,cardiovascular diseases ,High incidence ,Cardiology and Cardiovascular Medicine ,business - Abstract
Catecholaminergic polymorphic ventricular tachycardia (CPVT) is a genetic disorder with high incidence of sudden cardiac death (SCD). It has been published a rate of implantable cardioverter defibrillator (ICD) implantation in this disease around 30% of patients, with an incidence of appropriate
- Published
- 2016
- Full Text
- View/download PDF
47. Overlapping LQT1 and LQT2 phenotype in a patient with long QT syndrome associated with loss-of-function variations in KCNQ1 and KCNH2
- Author
-
Charles Antzelevitch, Rainer Schimpf, Ryan Pfeiffer, Vladislav V. Nesterenko, Nicole Schmitt, Guillermo J. Pérez, Christian Wolpert, Christian Veltmann, Martin Borggrefe, Elena Burashnikov, and Jonathan M. Cordeiro
- Subjects
Proband ,Adult ,medicine.medical_specialty ,congenital, hereditary, and neonatal diseases and abnormalities ,ERG1 Potassium Channel ,Physiology ,Long QT syndrome ,hERG ,Molecular Sequence Data ,CHO Cells ,Biology ,medicine.disease_cause ,QT interval ,Article ,Electrocardiography ,Cricetulus ,Polymorphism (computer science) ,Physiology (medical) ,Internal medicine ,Cricetinae ,medicine ,Missense mutation ,Animals ,Humans ,Amino Acid Sequence ,Cells, Cultured ,Pharmacology ,Genetics ,Mutation ,Polymorphism, Genetic ,Base Sequence ,Genetic Variation ,General Medicine ,medicine.disease ,Phenotype ,Ether-A-Go-Go Potassium Channels ,Long QT Syndrome ,Endocrinology ,KCNQ1 Potassium Channel ,biology.protein ,Female - Abstract
Long QT syndrome (LQTS) is an inherited disorder characterized by prolonged QT intervals and potentially life-threatening arrhythmias. Mutations in 12 different genes have been associated with LQTS. Here we describe a patient with LQTS who has a mutation in KCNQ1 as well as a polymorphism in KCNH2. The proband (MMRL0362), a 32-year-old female, exhibited multiple ventricular extrasystoles and one syncope. Her ECG (QT interval corrected for heart rate (QTc) = 518ms) showed an LQT2 morphology in leads V4–V6 and LQT1 morphology in leads V1–V2. Genomic DNA was isolated from lymphocytes. All exons and intron borders of 7 LQTS susceptibility genes were amplified and sequenced. Variations were detected predicting a novel missense mutation (V110I) in KCNQ1, as well as a common polymorphism in KCNH2 (K897T). We expressed wild-type (WT) or V110I Kv7.1 channels in CHO-K1 cells cotransfected with KCNE1 and performed patch-clamp analysis. In addition, WT or K897T Kv11.1 were also studied by patch clamp. Current–voltage (I-V) relations for V110I showed a significant reduction in both developing and tail current densities compared with WT at potentials >+20 mV (p < 0.05; n = 8 cells, each group), suggesting a reduction in IKs currents. K897T- Kv11.1 channels displayed a significantly reduced tail current density compared with WT-Kv11.1 at potentials >+10 mV. Interestingly, channel availability assessed using a triple-pulse protocol was slightly greater for K897T compared with WT (V0.5 = –53.1 ± 1.13 mV and –60.7 ± 1.15 mV for K897T and WT, respectively; p < 0.05). Comparison of the fully activated I-V revealed no difference in the rectification properties between WT and K897T channels. We report a patient with a loss-of-function mutation in KCNQ1 and a loss-of-function polymorphism in KCNH2. Our results suggest that a reduction of both IKr and IKs underlies the combined LQT1 and LQT2 phenotype observed in this patient.
- Published
- 2010
48. Genetics and cardiac channelopathies
- Author
-
Ramon Brugada, Oscar Campuzano, Pedro Beltran-Alvarez, Fabiana S. Scornik, Guillermo J. Pérez, and Anna Iglesias
- Subjects
Proband ,Genetics ,medicine.diagnostic_test ,Heart Diseases ,business.industry ,Autopsy ,Arrhythmias, Cardiac ,Disease ,medicine.disease ,Sudden death ,Sudden cardiac death ,Death, Sudden ,Breast cancer ,Death, Sudden, Cardiac ,Molecular Diagnostic Techniques ,medicine ,Humans ,Channelopathies ,Genetic Testing ,business ,Stroke ,Genetics (clinical) ,Genetic testing - Abstract
Sudden cardiac death is a major contributor to mortality in industrialized nations; in fact, it is the cause of more deaths than acquired immune deficiency syndrome, lung and breast cancer, and stroke together. Frequently, the autopsy becomes the principal diagnostic tool because macroscopic and microscopic analyses reveal the underlying cause of death. However, a significant number of sudden cardiac deaths remain unexplained. These cases are referred to as "natural" or arrhythmogenic. In the young, in up to 50% of sudden cardiac death cases, sudden death is the first and only clinical manifestation of an inherited cardiac disease that had remained undetected by conventional clinical investigations. To improve diagnosis, genetic testing has recently been added to these clinical tools. During the last two decades, there has been considerable progress in the understanding about genetics of sudden cardiac death. With that new information, the probands and their family members can make an informed decision regarding their care and know whether and to what extent they are at risk of suffering from the disease. Thus, genetic technology and expertise have become essential for the diagnosis of some forms of inherited cardiac diseases and to provide a basis for subsequent prevention strategies. This review focuses on recent advances in the understanding of cardiopathies owing to genetic investigations.
- Published
- 2010
49. Twinkle Twinkle Little Spark: Out of Tune Potassium Channels
- Author
-
Guillermo J. Pérez
- Subjects
Cardiac transient outward potassium current ,Optics ,Chemistry ,business.industry ,Spark (mathematics) ,business ,Potassium channel - Published
- 2005
- Full Text
- View/download PDF
50. How Do We Measure Repolarization Inside the Heart?
- Author
-
Fabiana S. Scornik, Andrew C. Zygmunt, Jeffrey M. Fish, Guillermo J. Pérez, and Charles Antzelevitch
- Subjects
business.industry ,Body surface mapping ,Endocardial cell ,Repolarization ,Medicine ,Voltage gradient ,Action potential duration ,business ,Cardiac repolarization ,Neuroscience - Abstract
Cardiac repolarization can be measured using a variety of electrophysiologic methodologies. Most studies make use of intracellular, extracellular, or optical recording techniques to record action potentials, monophasic action potentials or activation recovery intervals (ARI) from the epicardial or endocardial surfaces of the heart. Electrocardiographic techniques, including body surface mapping, delineate cardiac repolarization characteristics noninvasively, but the extent to which these can be used to assess local heterogeneities of repolarization remains an issue of debate. More recent studies have endeavored to quantitate repolarization characteristics beyond the surfaces of the heart, peering inside the walls of the heart. This chapter will focus on the latter. Our primary goal is to catalog, evaluate, and illustrate the capabilities of methodologies available for the quantitation of transmural heterogeneities of repolarization and probe the ionic basis for these distinctions.
- Published
- 2003
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.