37 results on '"Selga E"'
Search Results
2. AFI Innovative Regulatory Approaches Toolkit
- Author
-
Zetzsche, Dirk Andreas, Arner, Douglas, Buckley, Ross, Selga, E., Mohammad, G., and Consiglio, Roberta
- Subjects
Droit judiciaire [E06] [Droit, criminologie & sciences politiques] ,Judicial law [E06] [Law, criminology & political science] - Published
- 2021
3. P335Sudden unexplained death in Catalonia: comprehensive genetic analysis in post-mortem samples
- Author
-
Sanchez-Molero, OE, Campuzano, O, Allegue, C, Selga, E, Mademont, I, Coll, M, Mates, J, Iglesias, A, Medallo, J, and Brugada, R
- Published
- 2014
- Full Text
- View/download PDF
4. Genetic variants associated with cardiorespiratory symptoms of long COVID. The GINA-COVID study
- Author
-
Polo-Alonso, S, Camps-Vilaro, A, Degano, I R, Marti-Lluch, R, Pinsach-Abuin, M, Selga, E, Puigdecanet, E, Pedrerol-Alvarez, M, Puigmule, M, Perez, A, Subirana, I, Elosua, R, Brugada, R, Ramos, R, and Marrugat, J
- Published
- 2024
- Full Text
- View/download PDF
5. Large Genomic Imbalances in Brugada Syndrome
- Author
-
Mademont-Soler I, Pinsach-Abuin ML, Riuró H, Mates J, Pérez-Serra A, Coll M, Porres JM, Del Olmo B, Iglesias A, Selga E, Picó F, Pagans S, Ferrer-Costa C, Sarquella-Brugada G, Arbelo E, Cesar S, Brugada-Terradellas J, Campuzano Ó, and Brugada R
- Published
- 2016
6. Comprehensive Genetic Characterization of a Spanish Brugada Syndrome Cohort
- Author
-
Selga E, Campuzano O, Pinsach-Abuin ML, Pérez-Serra A, Mademont-Soler I, Riuró H, Picó F, Coll M, Iglesias A, Pagans S, Sarquella-Brugada G, Berne P, Benito B, Brugada-Terradellas J, Porres JM, López Zea M, Castro-Urda V, Fernández-Lozano I, and Brugada R
- Published
- 2015
7. A Missense Mutation in the Sodium Channel β2 Subunit Reveals SCN2B as a New Candidate Gene for Brugada Syndrome
- Author
-
Riuro, H., Beltran-Alvarez, P., Tarradas, A., Selga, E., Campuzano, O., Verges, M., Pagans, S., Iglesias, A., Brugada, J., Brugada, Pedro, Vazquez, F., Perez, G.j., Scornik, F., Brugada, R., Internal Medicine Specializations, and Cardio-vascular diseases
- Subjects
Brugada syndrome ,SCN2B ,sudden cardiac death ,SCN5A - 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 ?2 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 (?2WT) or mutant (?2D211G) ?2 subunits. Our electrophysiological analysis showed a 39.4% reduction in INa density when Nav 1.5 was coexpressed with the ?2D211G. Single channel analysis showed that the mutation did not affect the Nav 1.5 unitary channel conductance. Instead, protein membrane detection experiments suggested that ?2D211G decreases Nav 1.5 cell surface expression. The effect of the mutant ?2 subunit on the INa strongly suggests that SCN2B is a new candidate gene associated with BrS.
- Published
- 2013
8. Caffeic acid and instant coffee modulate the expression of transcription factors STAT5B and ATF2 in human cancer cells
- Author
-
Oleaga, C., primary, Selga, E., additional, Ciudad, C., additional, Izquierdo-Pulido, M., additional, and Noé, V., additional
- Published
- 2010
- Full Text
- View/download PDF
9. Gene expression profiles in rat mesenteric lymph nodes upon supplementation with conjugated linoleic acid during gestation and suckling
- Author
-
Selga, E., primary, Pérez-Cano, F. J., additional, Franch, A., additional, Castell, M., additional, Rivero, M., additional, Ciudad, C. J., additional, Castellote, C., additional, and Noé, V., additional
- Published
- 2010
- Full Text
- View/download PDF
10. Post-mortem genetic analysis in juvenile cases of sudden cardiac death
- Author
-
Campuzano O, Sanchez-Molero O, Allegue C, Coll M, Irene Mademont-Soler, Selga E, Ferrer-Costa C, Mates J, Iglesias A, Sarquella-Brugada G, Cesar S, Brugada J, Castellà J, Medallo J, and Brugada R
- Subjects
Adolescent, Forensics, Genetics, Next Generation Sequencing, Paediatric, Sudden cardiac death - Abstract
The reason behind a sudden death of a young individual remains unknown in up to 50% of postmortem cases. Pathogenic mutations in genes encoding heart proteins are known to cause sudden cardiac death.
11. Gene expression profiles in rat mesenteric lymph nodes upon supplementation with Conjugated Linoleic Acid during gestation and suckling
- Author
-
Rivero Montserrat, Ramírez-Santana Carolina, Franch Àngels, Pérez-Cano Francisco J, Selga Elisabet, Ciudad Carlos J, Castellote Cristina, and Noé Véronique
- Subjects
Biotechnology ,TP248.13-248.65 ,Genetics ,QH426-470 - Abstract
Abstract Background Diet plays a role on the development of the immune system, and polyunsaturated fatty acids can modulate the expression of a variety of genes. Human milk contains conjugated linoleic acid (CLA), a fatty acid that seems to contribute to immune development. Indeed, recent studies carried out in our group in suckling animals have shown that the immune function is enhanced after feeding them with an 80:20 isomer mix composed of c9,t11 and t10,c12 CLA. However, little work has been done on the effects of CLA on gene expression, and even less regarding immune system development in early life. Results The expression profile of mesenteric lymph nodes from animals supplemented with CLA during gestation and suckling through dam's milk (Group A) or by oral gavage (Group B), supplemented just during suckling (Group C) and control animals (Group D) was determined with the aid of the specific GeneChip® Rat Genome 230 2.0 (Affymettrix). Bioinformatics analyses were performed using the GeneSpring GX software package v10.0.2 and lead to the identification of 89 genes differentially expressed in all three dietary approaches. Generation of a biological association network evidenced several genes, such as connective tissue growth factor (Ctgf), tissue inhibitor of metalloproteinase 1 (Timp1), galanin (Gal), synaptotagmin 1 (Syt1), growth factor receptor bound protein 2 (Grb2), actin gamma 2 (Actg2) and smooth muscle alpha actin (Acta2), as highly interconnected nodes of the resulting network. Gene underexpression was confirmed by Real-Time RT-PCR. Conclusions Ctgf, Timp1, Gal and Syt1, among others, are genes modulated by CLA supplementation that may have a role on mucosal immune responses in early life.
- Published
- 2011
- Full Text
- View/download PDF
12. Overexpression of S100A4 in human cancer cell lines resistant to methotrexate
- Author
-
Hernández Jose L, Adan Jaume, Ramirez Sara, Villalobos Xenia, de Almagro M Cristina, Rico Isabel, Selga Elisabet, Mencía Nuria, Noé Véronique, and Ciudad Carlos J
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Methotrexate is a chemotherapeutic drug that is used in therapy of a wide variety of cancers. The efficiency of treatment with this drug is compromised by the appearance of resistance. Combination treatments of MTX with other drugs that could modulate the expression of genes involved in MTX resistance would be an adequate strategy to prevent the development of this resistance. Methods The differential expression pattern between sensitive and MTX-resistant cells was determined by whole human genome microarrays and analyzed with the GeneSpring GX software package. A global comparison of all the studied cell lines was performed in order to find out differentially expressed genes in the majority of the MTX-resistant cells. S100A4 mRNA and protein levels were determined by RT-Real-Time PCR and Western blot, respectively. Functional validations of S100A4 were performed either by transfection of an expression vector for S100A4 or a siRNA against S100A4. Transfection of an expression vector encoding for β-catenin was used to inquire for the possible transcriptional regulation of S100A4 through the Wnt pathway. Results S100A4 is overexpressed in five out of the seven MTX-resistant cell lines studied. Ectopic overexpression of this gene in HT29 sensitive cells augmented both the intracellular and extracellular S100A4 protein levels and caused desensitization toward MTX. siRNA against S100A4 decreased the levels of this protein and caused a chemosensitization in combined treatments with MTX. β-catenin overexpression experiments support a possible involvement of the Wnt signaling pathway in S100A4 transcriptional regulation in HT29 cells. Conclusions S100A4 is overexpressed in many MTX-resistant cells. S100A4 overexpression decreases the sensitivity of HT29 colon cancer human cells to MTX, whereas its knockdown causes chemosensitization toward MTX. Both approaches highlight a role for S100A4 in MTX resistance.
- Published
- 2010
- Full Text
- View/download PDF
13. Role of Caveolin 1, E-Cadherin, Enolase 2 and PKCalpha on resistance to methotrexate in human HT29 colon cancer cells
- Author
-
Noé Véronique, Morales Cristina, Selga Elisabet, Peinado Miguel A, and Ciudad Carlos J
- Subjects
Internal medicine ,RC31-1245 ,Genetics ,QH426-470 - Abstract
Abstract Background Methotrexate is one of the earliest cytotoxic drugs used in cancer therapy, and despite the isolation of multiple other folate antagonists, methotrexate maintains its significant role as a treatment for different types of cancer and other disorders. The usefulness of treatment with methotrexate is limited by the development of drug resistance, which may be acquired through different ways. To get insights into the mechanisms associated with drug resistance and sensitization we performed a functional analysis of genes deregulated in methotrexate resistant cells, either due to its co-amplification with the dhfr gene or as a result of a transcriptome screening using microarrays. Methods Gene expression levels were compared between triplicate samples from either HT29 sensitive cells and resistant to 10-5 M MTX by hybridization to the GeneChip® HG U133 PLUS 2.0 from Affymetrix. After normalization, a list of 3-fold differentially expressed genes with a p-value < 0.05 including multiple testing correction (Benjamini and Hochberg false discovery rate) was generated. RT-Real-time PCR was used to validate the expression levels of selected genes and copy-number was determined by qPCR. Functional validations were performed either by siRNAs or by transfection of an expression plasmid. Results Genes adjacent to the dhfr locus and included in the 5q14 amplicon were overexpressed in HT29 MTX-resistant cells. Treatment with siRNAs against those genes caused a slight reduction in cell viability in both HT29 sensitive and resistant cells. On the other hand, microarray analysis of HT29 and HT29 MTX resistant cells unveiled overexpression of caveolin 1, enolase 2 and PKCα genes in resistant cells without concomitant copy number gain. siRNAs against these three genes effectively reduced cell viability and caused a decreased MTX resistance capacity. Moreover, overexpression of E-cadherin, which was found underexpressed in MTX-resistant cells, also sensitized the cells toward the chemotherapeutic agent. Combined treatments targeting siRNA inhibition of caveolin 1 and overexpression of E-cadherin markedly reduced cell viability in both sensitive and MTX-resistant HT29 cells. Conclusion We provide functional evidences indicating that caveolin 1 and E-cadherin, deregulated in MTX resistant cells, may play a critical role in cell survival and may constitute potential targets for coadjuvant therapy.
- Published
- 2008
- Full Text
- View/download PDF
14. P335 Sudden unexplained death in Catalonia: comprehensive genetic analysis in post-mortem samples.
- Author
-
Sanchez-Molero, OE, Campuzano, O, Allegue, C, Selga, E, Mademont, I, Coll, M, Mates, J, Iglesias, A, Medallo, J, and Brugada, R
- Subjects
CARDIAC arrest ,AUTOPSY ,CARDIOMYOPATHIES ,GENETIC mutation ,DNA - Abstract
Purpose: Cardiovascular pathologies are the main cause of sudden death in developed countries. In population older than 50 years old, most deaths are due to coronary disease alterations, in the young deaths may be caused by "monogenetic disorders", channelopathies, or cardiomyopathies. Genetic analysis is not routinely performed in autopsy protocol when a cause of death is not identified in the young. In recent years the development of next generation sequencing (NGS) technology allows the investigation of high number of genes in a short period of time at low cost. We launched the MOSCAT project, a prospective post-mortem study that aims to identify the genetic cause of death in cases that remains without a conclusive diagnosis after a complete autopsy. Methods: Our cohort included a total of 88 cases: <50 years old, and no-conclusive cause of death after complete autopsy investigation. We divided the cohort into two groups, depending on DNA quality for NGS analysis. The first group included 59 samples with low quality. The main seven arrhythmogenic genes (KCNQ1, KCNH2, KCNE1, KCNE2, KCNE3, RYR2 and SCN5A) were analyzed using Sanger method. The second group included 43 samples analyzed by NGS, a total of 55 genes associated with SCD were analyzed. Potentially pathogenic role was determined when minor allele frequency (MAF) was less than 1% and at least one in silico prediction. A total of 14 samples were analyzed using both methods as internal control. Results: Our cohort of 88 samples showed a gender ratio of 2:1 (men: women respectively). In group 1 (sanger, 59 samples) we identified 30% arrythmogenic mutation carriers. In group 2 (NGS, 43 samples) we identified 15% arrythmogenic mutations and 85% associated with structural cardiomyopathies. Acording to age in group 1 we identified 27% mutations in patients younger than 35 and 34% in older than 35. By NGS we identified 50% arrythmogenic mutations and 50% structural mutations in less than 35 and 40% arrythmogenic mutations and 60% structural mutations in older than 35. Conclusions: In patients younger than 50, we identified arrythmogenic mutations in 25% and structural mutations in 70% of cases studied. Nearly 30% of post-mortem cases minor than 50 years with a no-conclusive cause of death carry a potential pathogenic variation that could explain the death. NGS technology adds additional information and may be useful in the identification of genetic causality. We believe that genetic analysis should be included in routine forensic guidelines when a no-conclusive cause of death arises, especially in cases under 35 years old. [ABSTRACT FROM PUBLISHER]
- Published
- 2014
- Full Text
- View/download PDF
15. Loss of sodium current caused by a Brugada syndrome-associated variant is determined by patient-specific genetic background.
- Author
-
Martínez-Moreno R, Carreras D, Sarquella-Brugada G, Pérez GJ, Selga E, Scornik FS, and Brugada R
- Subjects
- Humans, Sodium metabolism, Arrhythmias, Cardiac metabolism, Cardiac Conduction System Disease metabolism, Myocytes, Cardiac metabolism, NAV1.5 Voltage-Gated Sodium Channel genetics, NAV1.5 Voltage-Gated Sodium Channel metabolism, Brugada Syndrome
- Abstract
Background: Brugada syndrome (BrS) is an inherited cardiac arrhythmogenic disease that predisposes patients to sudden cardiac death. It is associated with mutations in SCN5A, which encodes the cardiac sodium channel alpha subunit (Na
V 1.5). BrS-related mutations have incomplete penetrance and variable expressivity within families., Objective: The purpose of this study was to determine the role of patient-specific genetic background on the cellular and clinical phenotype among carriers of NaV 1.5_p.V1525M., Methods: We studied sodium currents from patient-specific human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) and heterologously transfected human embryonic kidney (HEK) tsA201 cells using the whole-cell patch-clamp technique. We determined gene and protein expression by quantitative polymerase chain reaction, RNA sequencing, and western blot and performed a genetic panel for arrhythmogenic diseases., Results: Our results showed a large reduction in INa density in hiPSC-CM derived from 2 V1525M single nucleotide variant (SNV) carriers compared with hiPSC-CM derived from a noncarrier, suggesting a dominant-negative effect of the NaV 1.5_p.V1525M channel. INa was not affected in hiPSC-CMs derived from a V1525M SNV carrier who also carries the NaV 1.5_p.H558R polymorphism. Heterozygous expression of V1525M in HEK-293T cells produced a loss of INa function, not observed when this variant was expressed together with H558R. In addition, the antiarrhythmic drug mexiletine rescued INa function in hiPSC-CM. SCN5A expression was increased in the V1525M carrier who also expresses NaV 1.5_p.H558R., Conclusion: Our results in patient-specific hiPSC-CM point to a dominant-negative effect of NaV 1.5_p.V1525M, which can be reverted by the presence of NaV 1.5_p.H558R. Overall, our data points to a role of patient-specific genetic background as a determinant for incomplete penetrance in BrS., (Copyright © 2023 Heart Rhythm Society. Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
16. Generation of the induced pluripotent stem cell line ESi108-A from a familial atrial fibrillation patient.
- Author
-
Martínez-Moreno R, Pérez-Serra A, Selga E, Carreras D, Aran B, Kuebler B, Scornik FS, Pérez GJ, and Brugada R
- Subjects
- Humans, Cell Differentiation, Cell Line, Plasmids, Induced Pluripotent Stem Cells metabolism, Atrial Fibrillation genetics, Atrial Fibrillation metabolism
- Abstract
Tissue-specific cells differentiated from patient-derived human induced pluripotent stem cells (hiPSC) are a relevant cellular model to study several diseases. We obtained a hiPSC line from skin fibroblasts of a patient affected by familial atrial fibrillation by nucleofection of non-integrating episomal vectors. The resulting hiPSC line displays a normal karyotype, expresses pluripotency surface markers and pluripotency genes, and differentiates into cells of the 3 germ layers. Therefore, it represents a reliable model to study the disease in a physiologically relevant cellular environment., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
17. Generation of an induced pluripotent stem cell line from a healthy Caucasian male.
- Author
-
Martínez-Moreno R, Pérez-Serra A, Carreras D, Aran B, Kuebler B, Brugada R, Scornik FS, Pérez GJ, and Selga E
- Subjects
- Cell Differentiation, Cell Line, Fibroblasts, Germ Layers, Humans, Male, Induced Pluripotent Stem Cells metabolism
- 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., (Copyright © 2022 The Author(s). Published by Elsevier B.V. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
18. An SCN1B Variant Affects Both Cardiac-Type (Na V 1.5) and Brain-Type (Na V 1.1) Sodium Currents and Contributes to Complex Concomitant Brain and Cardiac Disorders.
- Author
-
Martinez-Moreno R, Selga E, Riuró H, Carreras D, Parnes M, Srinivasan C, Wangler MF, Pérez GJ, Scornik FS, and Brugada R
- Abstract
Voltage-gated sodium (Na
V ) 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 NaV 1.5 and/or NaV 1.1 currents using a whole cell patch-clamp technique in tsA201 cells. We observed a decrease in sodium current density in cells co-expressing NaV 1.5 or NaV 1.1 and β1D103V compared to β1WT . Interestingly, β1bD103V did not affect NaV 1.1 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 NaV 1.5 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., (Copyright © 2020 Martinez-Moreno, Selga, Riuró, Carreras, Parnes, Srinivasan, Wangler, Pérez, Scornik and Brugada.)- Published
- 2020
- Full Text
- View/download PDF
19. Extra Virgin Olive Oil Contains a Phenolic Inhibitor of the Histone Demethylase LSD1/KDM1A.
- Author
-
Cuyàs E, Gumuzio J, Lozano-Sánchez J, Carreras D, Verdura S, Llorach-Parés L, Sanchez-Martinez M, Selga E, Pérez GJ, Scornik FS, Brugada R, Bosch-Barrera J, Segura-Carretero A, Martin ÁG, Encinar JA, and Menendez JA
- Subjects
- Aldehydes analysis, Binding Sites drug effects, Breast Neoplasms, Cell Line, Tumor, Co-Repressor Proteins drug effects, Gene Expression drug effects, Histone Demethylases chemistry, Histone Demethylases metabolism, Histones metabolism, Humans, MCF-7 Cells, Models, Molecular, Molecular Docking Simulation, Molecular Structure, Neoplastic Stem Cells metabolism, Phenols analysis, Recombinant Proteins drug effects, SOXB1 Transcription Factors antagonists & inhibitors, SOXB1 Transcription Factors genetics, Aldehydes pharmacology, Histone Demethylases antagonists & inhibitors, Olive Oil chemistry, Phenols pharmacology
- 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 IC
50 as low as 2.5 μmol/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.- Published
- 2019
- Full Text
- View/download PDF
20. Experimental Models of Brugada syndrome.
- Author
-
Sendfeld F, Selga E, Scornik FS, Pérez GJ, Mills NL, and Brugada R
- Subjects
- Animals, Animals, Genetically Modified, Biomarkers, Brugada Syndrome diagnosis, Brugada Syndrome therapy, Cell Differentiation, Disease Susceptibility, Dogs, Genetic Predisposition to Disease, Humans, Induced Pluripotent Stem Cells cytology, Induced Pluripotent Stem Cells metabolism, Mice, Mutation, Myocytes, Cardiac cytology, Myocytes, Cardiac metabolism, NAV1.5 Voltage-Gated Sodium Channel genetics, Brugada Syndrome etiology, Brugada Syndrome physiopathology, Disease Models, Animal
- 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., Competing Interests: The authors declare no conflict of interest.
- Published
- 2019
- Full Text
- View/download PDF
21. Sodium channel current loss of function in induced pluripotent stem cell-derived cardiomyocytes from a Brugada syndrome patient.
- Author
-
Selga E, Sendfeld F, Martinez-Moreno R, Medine CN, Tura-Ceide O, Wilmut SI, Pérez GJ, Scornik FS, Brugada R, and Mills NL
- Subjects
- Base Sequence, Biomarkers metabolism, Biotinylation, Cell Membrane metabolism, Cell Shape, HEK293 Cells, Humans, Ion Channel Gating, Mutant Proteins metabolism, Brugada Syndrome metabolism, Brugada Syndrome pathology, Induced Pluripotent Stem Cells pathology, Myocytes, Cardiac metabolism, NAV1.5 Voltage-Gated Sodium Channel metabolism
- 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 Na
v 1.5_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., (Copyright © 2017 The Author(s). Published by Elsevier Ltd.. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
22. Post-mortem genetic analysis in juvenile cases of sudden cardiac death.
- Author
-
Campuzano O, Sanchez-Molero O, Allegue C, Coll M, Mademont-Soler I, Selga E, Ferrer-Costa C, Mates J, Iglesias A, Sarquella-Brugada G, Cesar S, Brugada J, Castellà J, Medallo J, and Brugada R
- Subjects
- Child, Child, Preschool, DNA genetics, Female, Forensic Genetics, High-Throughput Nucleotide Sequencing, Humans, Infant, Infant, Newborn, Male, Sequence Analysis, DNA, Death, Sudden, Cardiac etiology, Genetic Testing, Genetic Variation, Heart Diseases genetics
- Abstract
Background: The reason behind a sudden death of a young individual remains unknown in up to 50% of postmortem cases. Pathogenic mutations in genes encoding heart proteins are known to cause sudden cardiac death., Objective: The aim of our study was to ascertain whether genetic alterations could provide an explanation for sudden cardiac death in a juvenile cohort with no-conclusive cause of death after comprehensive autopsy., Methods: Twenty-nine cases <15 years showing no-conclusive cause of death after a complete autopsy were studied. Genetic analysis of 7 main genes associated with sudden cardiac death was performed using Sanger technology in low quality DNA cases, while in good quality cases the analysis of 55 genes associated with sudden cardiac death was performed using Next Generation Sequencing technology., Results: Thirty-five genetic variants were identified in 12 cases (41.37%). Ten genetic/variants in genes encoding cardiac ion channels were identified in 8 cases (27.58%). We also identified 9 cases (31.03%) carrying 25 genetic variants in genes encoding structural cardiac proteins. Nine cases carried more than one genetic variation, 5 of them combining structural and non-structural genes., Conclusions: Our study supports the inclusion of molecular autopsy in forensic routine protocols when no conclusive cause of death is identified. Around 40% of sudden cardiac death young cases carry a genetic variant that could provide an explanation for the cause of death. Because relatives could be at risk of sudden cardiac death, our data reinforce their need of clinical assessment and, if indicated, of genetic analysis., (Copyright © 2014 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
23. The smooth muscle-type β1 subunit potentiates activation by DiBAC4(3) in recombinant BK channels.
- Author
-
Bosch Calero C, Selga E, Brugada R, Scornik FS, and Pérez GJ
- Subjects
- Animals, Cell Line, Humans, Mice, Muscle, Smooth physiology, Recombinant Proteins, Barbiturates pharmacology, Isoxazoles pharmacology, Large-Conductance Calcium-Activated Potassium Channels physiology, Protein Subunits physiology
- 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
- 2014
- Full Text
- View/download PDF
24. Brugada syndrome and p.E61X_RANGRF.
- Author
-
Campuzano O, Berne P, Selga E, Allegue C, Iglesias A, Brugada J, and Brugada R
- Subjects
- Adult, Aged, Anti-Arrhythmia Agents, Brugada Syndrome diagnosis, Child, DNA Mutational Analysis, Electrocardiography, Female, Flecainide, Gene Frequency, Genetic Association Studies, Genetic Predisposition to Disease, Heredity, Humans, Male, Phenotype, Predictive Value of Tests, Spain, Voltage-Gated Sodium Channel Blockers, Brugada Syndrome genetics, Codon, Nonsense, ran GTP-Binding Protein genetics
- Abstract
Background: Brugada syndrome is an inherited cardiac condition transmitted with an autosomal dominant pattern which can lead to sudden cardiac death from malignant ventricular arrhythmias. The RANGRF gene has recently been proposed to be associated with Brugada syndrome. This gene encodes the MOG1 protein, a co-factor required for the full functioning of the cardiac sodium channel Nav1.5. The nonsense p.E61X genetic variation in the RANGRF gene has been postulated as responsible for Brugada syndrome although no clear association has been established., Methods: We clinically and genetically evaluated a Spanish family diagnosed with Brugada syndrome. A comprehensive genetic analysis of all genes to date responsible for Brugada syndrome was performed in the index case., Results: The index case was clinically diagnosed with Brugada syndrome after flecainide test. We identified a nonsense variation (p.E61X) in the index case and in other five family members. All of them showed a normal electrocardiogram in basal conditions. Flecainide test unmasked a type 1 Brugada syndrome electrocardiogram only in two of the relatives., Conclusions: We suggest that p.E61X_RANGRF is a rare genetic variation with an uncertain role in Brugada Syndrome. Further studies must be performed to elucidate the potential pathogenic role of p.E61X_RANGRF in Brugada Syndrome.
- Published
- 2014
- Full Text
- View/download PDF
25. A missense mutation in the sodium channel β2 subunit reveals SCN2B as a new candidate gene for Brugada syndrome.
- Author
-
Riuró H, Beltran-Alvarez P, Tarradas A, Selga E, Campuzano O, Vergés M, Pagans S, Iglesias A, Brugada J, Brugada P, Vázquez FM, Pérez GJ, Scornik FS, and Brugada R
- Subjects
- Death, Sudden, Cardiac etiology, Female, Humans, Middle Aged, Sodium Channels genetics, Sodium Channels metabolism, Voltage-Gated Sodium Channel beta-2 Subunit metabolism, Brugada Syndrome genetics, Genetic Predisposition to Disease, Mutation, Missense, Voltage-Gated Sodium Channel beta-2 Subunit genetics
- 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., (© 2013 WILEY PERIODICALS, INC.)
- Published
- 2013
- Full Text
- View/download PDF
26. Molecular heterogeneity of large-conductance calcium-activated potassium channels in canine intracardiac ganglia.
- Author
-
Selga E, Pérez-Serra A, Moreno-Asso A, Anderson S, Thomas K, Desai M, Brugada R, Pérez GJ, and Scornik FS
- Subjects
- Amino Acid Sequence, Animals, Calcium metabolism, Dogs, Large-Conductance Calcium-Activated Potassium Channels chemistry, Molecular Sequence Data, Neurons metabolism, Protein Subunits chemistry, Protein Subunits metabolism, Ganglia metabolism, Large-Conductance Calcium-Activated Potassium Channels metabolism, Myocardium cytology
- 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.
- Published
- 2013
- Full Text
- View/download PDF
27. DiBAC₄(3) hits a "sweet spot" for the activation of arterial large-conductance Ca²⁺-activated potassium channels independently of the β₁-subunit.
- Author
-
Scornik FS, Bucciero RS, Wu Y, Selga E, Bosch Calero C, Brugada R, and Pérez GJ
- Subjects
- Animals, Arteries drug effects, Arteries metabolism, Barbiturates metabolism, Data Interpretation, Statistical, Dogs, Isoxazoles metabolism, Kinetics, Large-Conductance Calcium-Activated Potassium Channel beta Subunits genetics, Large-Conductance Calcium-Activated Potassium Channels genetics, Membrane Potentials physiology, Mice, Mice, Inbred C57BL, Mice, Knockout, Muscle, Smooth, Vascular drug effects, Muscle, Smooth, Vascular metabolism, Patch-Clamp Techniques, Real-Time Polymerase Chain Reaction, Sodium Channels drug effects, Barbiturates pharmacology, Isoxazoles pharmacology, Large-Conductance Calcium-Activated Potassium Channel beta Subunits drug effects, Large-Conductance Calcium-Activated Potassium Channels drug effects
- 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
28. A novel missense mutation, I890T, in the pore region of cardiac sodium channel causes Brugada syndrome.
- Author
-
Tarradas A, Selga E, Beltran-Alvarez P, Pérez-Serra A, Riuró H, Picó F, Iglesias A, Campuzano O, Castro-Urda V, Fernández-Lozano I, Pérez GJ, Scornik FS, and Brugada R
- Subjects
- Adult, Amino Acid Sequence, Child, Female, HEK293 Cells, Humans, Male, Models, Molecular, Molecular Sequence Data, NAV1.5 Voltage-Gated Sodium Channel chemistry, Pedigree, Brugada Syndrome genetics, Mutation, Missense, NAV1.5 Voltage-Gated Sodium Channel genetics
- Abstract
Brugada syndrome (BrS) is a life-threatening, inherited arrhythmogenic syndrome associated with autosomal dominant mutations in SCN5A, the gene encoding the cardiac Na(+) channel alpha subunit (Na(v)1.5). The aim of this work was to characterize the functional alterations caused by a novel SCN5A mutation, I890T, and thus establish whether this mutation is associated with BrS. The mutation was identified by direct sequencing of SCN5A from the proband's DNA. Wild-type (WT) or I890T Na(v)1.5 channels were heterologously expressed in human embryonic kidney cells. Sodium currents were studied using standard whole cell patch-clamp protocols and immunodetection experiments were performed using an antibody against human Na(v)1.5 channel. A marked decrease in current density was observed in cells expressing the I890T channel (from -52.0 ± 6.5 pA/pF, n = 15 to -35.9 ± 3.4 pA/pF, n = 22, at -20 mV, WT and I890T, respectively). Moreover, a positive shift of the activation curve was identified (V(1/2) = -32.0 ± 0.3 mV, n = 18, and -27.3 ± 0.3 mV, n = 22, WT and I890T, respectively). No changes between WT and I890T currents were observed in steady-state inactivation, time course of inactivation, slow inactivation or recovery from inactivation parameters. Cell surface protein biotinylation analyses confirmed that Na(v)1.5 channel membrane expression levels were similar in WT and I890T cells. In summary, our data reveal that the I890T mutation, located within the pore of Na(v)1.5, causes an evident loss-of-function of the channel. Thus, the BrS phenotype observed in the proband is most likely due to this mutation.
- Published
- 2013
- Full Text
- View/download PDF
29. Identification of novel Sp1 targets involved in proliferation and cancer by functional genomics.
- Author
-
Oleaga C, Welten S, Belloc A, Solé A, Rodriguez L, Mencia N, Selga E, Tapias A, Noé V, and Ciudad CJ
- Subjects
- Base Sequence, Blotting, Western, Chromatin Immunoprecipitation, DNA Primers, HeLa Cells, Humans, Oligonucleotide Array Sequence Analysis, Promoter Regions, Genetic, Real-Time Polymerase Chain Reaction, Sp1 Transcription Factor metabolism, Cell Proliferation, Genomics, Neoplasms pathology, Sp1 Transcription Factor genetics
- Abstract
Sp1 is a transcription factor regulating many genes through its DNA binding domain, containing three zinc fingers. We were interested in identifying target genes regulated by Sp1, particularly those involved in proliferation and cancer. Our approach was to treat HeLa cells with a siRNA directed against Sp1 mRNA to decrease the expression of Sp1 and, in turn, the genes activated by this transcription factor. Sp1-siRNA treatment led to a great number of differentially expressed genes as determined by whole genome cDNA microarray analysis. Underexpressed genes were selected since they represent putative genes activated by Sp1 and classified in six Gene Onthology categories, namely proliferation and cancer, mRNA processing, lipid metabolism, glucidic metabolism, transcription and translation. Putative Sp1 binding sites were found in the promoters of the selected genes using the Match™ software. After literature mining, 11 genes were selected for further validation. Underexpression by qRT-PCR was confirmed for the 11 genes plus Sp1 in HeLa cells after Sp1-siRNA treatment. EMSA and ChIP assays were performed to test for binding of Sp1 to the promoters of these genes. We observed binding of Sp1 to the promoters of RAB20, FGF21, IHPK2, ARHGAP18, NPM3, SRSF7, CALM3, PGD and Sp1 itself. Furthermore, the mRNA levels of RAB20, FGF21 and IHPK2 and luciferase activity for these three genes related to proliferation and cancer, were determined after overexpression of Sp1 in HeLa cells, to confirm their regulation by Sp1. Involvement of these three genes in proliferation was validated by gene silencing using polypurine reverse hoogsteen hairpins., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
30. Transcriptional profiling of striatal neurons in response to single or concurrent activation of dopamine D2, adenosine A(2A) and metabotropic glutamate type 5 receptors: focus on beta-synuclein expression.
- Author
-
Canela L, Selga E, García-Martínez JM, Amaral OB, Fernández-Dueñas V, Alberch J, Canela EI, Franco R, Noé V, Lluís C, Ciudad CJ, and Ciruela F
- Subjects
- Animals, Blotting, Western, Cells, Cultured, Corpus Striatum cytology, Female, Neurons cytology, Oligonucleotide Array Sequence Analysis, RNA, Messenger genetics, Rats, Rats, Sprague-Dawley, Real-Time Polymerase Chain Reaction, Receptor, Metabotropic Glutamate 5, Reverse Transcriptase Polymerase Chain Reaction, alpha-Synuclein genetics, alpha-Synuclein metabolism, beta-Synuclein genetics, Biomarkers metabolism, Corpus Striatum metabolism, Gene Expression Profiling, Neurons metabolism, Receptor, Adenosine A2A metabolism, Receptors, Dopamine D2 metabolism, Receptors, Metabotropic Glutamate metabolism, beta-Synuclein metabolism
- Abstract
G protein-coupled receptor oligomerization is a concept which is changing the understanding of classical pharmacology. Both, oligomerization and functional interaction between adenosine A(2A,) dopamine D(2) and metabotropic glutamate type 5 receptors have been demonstrated in the striatum. However, the transcriptional consequences of receptors co-activation are still unexplored. We aim here to determine the changes in gene expression of striatal primary cultured neurons upon isolated or simultaneous receptor activation. Interestingly, we found that 95 genes of the total analyzed (15,866 transcripts and variants) changed their expression in response to simultaneous stimulation of all three receptors. Among these genes, we focused on the β-synuclein (β-Syn) gene (SCNB). Quantitative PCR verified the magnitude and direction of change in expression of SCNB. Since β-Syn belongs to the homologous synuclein family and may be considered a natural regulator of α-synuclein (α-Syn), it has been proposed that β-Syn might act protectively against α-Syn neuropathology., (Copyright © 2012 Elsevier B.V. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
31. Underexpression of miR-224 in methotrexate resistant human colon cancer cells.
- Author
-
Mencia N, Selga E, Noé V, and Ciudad CJ
- Subjects
- 3' Untranslated Regions, Caco-2 Cells, Cell Line, Tumor, Colonic Neoplasms, Humans, K562 Cells, Lectins metabolism, MicroRNAs antagonists & inhibitors, Oligonucleotide Array Sequence Analysis, Reverse Transcriptase Polymerase Chain Reaction, Sodium-Bicarbonate Symporters genetics, Sodium-Bicarbonate Symporters metabolism, Antimetabolites, Antineoplastic pharmacology, Drug Resistance, Neoplasm, Methotrexate pharmacology, MicroRNAs metabolism
- Abstract
MicroRNAs (miRNAs) are small non-coding RNAs involved in RNA silencing that play a role in many biological processes. They are involved in the development of many diseases, including cancer. Extensive experimental data show that they play a role in the pathogenesis of cancer as well as the development of drug resistance during treatments. The aim of this work was to detect differentially expressed miRNAs in MTX-resistant cells. Thus, miRNA microarrays of sensitive and MTX-resistant HT29 colon cancer cells were performed. The results were analyzed using the GeneSpring GX11.5 software. Differentially expressed miRNAs in resistant cells were identified and miR-224, which was one of the most differentially expressed miRNAs and with high raw signal values, was selected for further studies. The underexpression of miR-224 was also observed in CaCo-2 and K562 cells resistant to MTX. Putative targets were predicted using TargetScan 5.1 software and integrated with the data from expression microarrays previously performed. This approach allowed us to identify miR-224 targets that were differentially expressed more than 2-fold in resistant cells. Among them CDS2, DCP2, HSPC159, MYST3 and SLC4A4 were validated at the mRNA level by qRT-PCR. Functional assays using an anti-miR against miR-224 desensitized the cells towards MTX, mimicking the resistant phenotype. On the other hand, siRNA treatment against SLC4A4 or incubation of Poly Purine Reverse Hoogsteen (PPRH) hairpins against CDS2 or HSPC159 increased sensitivity to MTX. These results revealed a role for miR-224 and its targets in MTX resistance in HT29 colon cancer cells., (Copyright © 2011 Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
32. Gene expression profiles in rat mesenteric lymph nodes upon supplementation with conjugated linoleic acid during gestation and suckling.
- Author
-
Selga E, Pérez-Cano FJ, Franch A, Ramírez-Santana C, Rivero M, Ciudad CJ, Castellote C, and Noé V
- Subjects
- Actins genetics, Actins metabolism, Animals, Animals, Newborn, Animals, Suckling, Connective Tissue Growth Factor genetics, Connective Tissue Growth Factor metabolism, Dietary Supplements, Female, GRB2 Adaptor Protein genetics, GRB2 Adaptor Protein metabolism, Galanin genetics, Galanin metabolism, Gene Regulatory Networks, Lymph Nodes growth & development, Lymph Nodes immunology, Mesentery, Pregnancy, RNA, Messenger metabolism, Rats, Rats, Wistar, Synaptotagmin I genetics, Synaptotagmin I metabolism, Tissue Inhibitor of Metalloproteinase-1 genetics, Tissue Inhibitor of Metalloproteinase-1 metabolism, Gene Expression Profiling, Linoleic Acids, Conjugated pharmacology, Lymph Nodes metabolism
- Abstract
Background: Diet plays a role on the development of the immune system, and polyunsaturated fatty acids can modulate the expression of a variety of genes. Human milk contains conjugated linoleic acid (CLA), a fatty acid that seems to contribute to immune development. Indeed, recent studies carried out in our group in suckling animals have shown that the immune function is enhanced after feeding them with an 80:20 isomer mix composed of c9,t11 and t10,c12 CLA. However, little work has been done on the effects of CLA on gene expression, and even less regarding immune system development in early life., Results: The expression profile of mesenteric lymph nodes from animals supplemented with CLA during gestation and suckling through dam's milk (Group A) or by oral gavage (Group B), supplemented just during suckling (Group C) and control animals (Group D) was determined with the aid of the specific GeneChip(®) Rat Genome 230 2.0 (Affymettrix). Bioinformatics analyses were performed using the GeneSpring GX software package v10.0.2 and lead to the identification of 89 genes differentially expressed in all three dietary approaches. Generation of a biological association network evidenced several genes, such as connective tissue growth factor (Ctgf), tissue inhibitor of metalloproteinase 1 (Timp1), galanin (Gal), synaptotagmin 1 (Syt1), growth factor receptor bound protein 2 (Grb2), actin gamma 2 (Actg2) and smooth muscle alpha actin (Acta2), as highly interconnected nodes of the resulting network. Gene underexpression was confirmed by Real-Time RT-PCR., Conclusions: Ctgf, Timp1, Gal and Syt1, among others, are genes modulated by CLA supplementation that may have a role on mucosal immune responses in early life.
- Published
- 2011
- Full Text
- View/download PDF
33. UDP-glucuronosyltransferase 1A6 overexpression in breast cancer cells resistant to methotrexate.
- Author
-
de Almagro MC, Selga E, Thibaut R, Porte C, Noé V, and Ciudad CJ
- Subjects
- Cell Line, Tumor, Dose-Response Relationship, Drug, Female, Glucuronosyltransferase genetics, Humans, Methotrexate administration & dosage, RNA, Messenger genetics, RNA, Messenger metabolism, Tamoxifen administration & dosage, Tamoxifen pharmacology, Transcription Factors genetics, Transcription Factors metabolism, Antineoplastic Agents pharmacology, Breast Neoplasms drug therapy, Drug Resistance, Neoplasm, Gene Expression Regulation, Neoplastic drug effects, Glucuronosyltransferase metabolism, Methotrexate pharmacology
- Abstract
Methotrexate is a chemotherapeutic agent used in breast cancer treatment, but the occurrence of resistance limits its therapeutic use. A microarrays analysis between sensitive and methotrexate resistant MCF7 and MDA-MB-468 breast cancer cells pointed out the UDP-glucuronosyltransferase 1A (UGT1A) family as a common deregulated node in both cell lines. This family of genes is involved in Phase II metabolism. UGT1A6 was the main isoform responsible for UGT1A family overexpression in these cells. Its overexpression was not due to gene amplification. Transfection of a vector encoding for UGT1A6 in sensitive cells counteracted the cytotoxicity caused by methotrexate. Methotrexate increased the transcriptional activity from a luciferase reporter driven by the UGT1A6 promoter and induced UGT1A6 mRNA and enzymatic activity. Promoter analysis suggested that UGT1A6 induction by methotrexate could be driven by the transcription factors ARNT (HIF-1) and AhR/ARNT. Cells incubated with anticancer drugs susceptible to glucuronidation, such as tamoxifen or irinotecan, together with methotrexate, showed a lesser degree of cytotoxicity, due to UGT1A6 induction. The pharmacological effect of this induction should be taken into account when combining methotrexate with other drugs that are glucuronidated., (Copyright © 2010 Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
34. Overexpression of S100A4 in human cancer cell lines resistant to methotrexate.
- Author
-
Mencía N, Selga E, Rico I, de Almagro MC, Villalobos X, Ramirez S, Adan J, Hernández JL, Noé V, and Ciudad CJ
- Subjects
- Blotting, Western, Caco-2 Cells, Cell Survival drug effects, Colonic Neoplasms genetics, Colonic Neoplasms pathology, Gene Expression Profiling methods, Gene Expression Regulation, Neoplastic, HT29 Cells, Humans, Oligonucleotide Array Sequence Analysis, RNA Interference, RNA, Messenger metabolism, Reproducibility of Results, Reverse Transcriptase Polymerase Chain Reaction, S100 Calcium-Binding Protein A4, S100 Proteins genetics, Transcription, Genetic, Transfection, Up-Regulation, beta Catenin genetics, beta Catenin metabolism, Antimetabolites, Antineoplastic pharmacology, Colonic Neoplasms metabolism, Drug Resistance, Neoplasm genetics, Methotrexate pharmacology, S100 Proteins metabolism
- Abstract
Background: Methotrexate is a chemotherapeutic drug that is used in therapy of a wide variety of cancers. The efficiency of treatment with this drug is compromised by the appearance of resistance. Combination treatments of MTX with other drugs that could modulate the expression of genes involved in MTX resistance would be an adequate strategy to prevent the development of this resistance., Methods: The differential expression pattern between sensitive and MTX-resistant cells was determined by whole human genome microarrays and analyzed with the GeneSpring GX software package. A global comparison of all the studied cell lines was performed in order to find out differentially expressed genes in the majority of the MTX-resistant cells. S100A4 mRNA and protein levels were determined by RT-Real-Time PCR and Western blot, respectively. Functional validations of S100A4 were performed either by transfection of an expression vector for S100A4 or a siRNA against S100A4. Transfection of an expression vector encoding for beta-catenin was used to inquire for the possible transcriptional regulation of S100A4 through the Wnt pathway., Results: S100A4 is overexpressed in five out of the seven MTX-resistant cell lines studied. Ectopic overexpression of this gene in HT29 sensitive cells augmented both the intracellular and extracellular S100A4 protein levels and caused desensitization toward MTX. siRNA against S100A4 decreased the levels of this protein and caused a chemosensitization in combined treatments with MTX. beta-catenin overexpression experiments support a possible involvement of the Wnt signaling pathway in S100A4 transcriptional regulation in HT29 cells., Conclusions: S100A4 is overexpressed in many MTX-resistant cells. S100A4 overexpression decreases the sensitivity of HT29 colon cancer human cells to MTX, whereas its knockdown causes chemosensitization toward MTX. Both approaches highlight a role for S100A4 in MTX resistance.
- Published
- 2010
- Full Text
- View/download PDF
35. Networking of differentially expressed genes in human cancer cells resistant to methotrexate.
- Author
-
Selga E, Oleaga C, Ramírez S, de Almagro MC, Noé V, and Ciudad CJ
- Abstract
Background: The need for an integrated view of data obtained from high-throughput technologies gave rise to network analyses. These are especially useful to rationalize how external perturbations propagate through the expression of genes. To address this issue in the case of drug resistance, we constructed biological association networks of genes differentially expressed in cell lines resistant to methotrexate (MTX)., Methods: Seven cell lines representative of different types of cancer, including colon cancer (HT29 and Caco2), breast cancer (MCF-7 and MDA-MB-468), pancreatic cancer (MIA PaCa-2), erythroblastic leukemia (K562) and osteosarcoma (Saos-2), were used. The differential expression pattern between sensitive and MTX-resistant cells was determined by whole human genome microarrays and analyzed with the GeneSpring GX software package. Genes deregulated in common between the different cancer cell lines served to generate biological association networks using the Pathway Architect software., Results: Dikkopf homolog-1 (DKK1) is a highly interconnected node in the network generated with genes in common between the two colon cancer cell lines, and functional validations of this target using small interfering RNAs (siRNAs) showed a chemosensitization toward MTX. Members of the UDP-glucuronosyltransferase 1A (UGT1A) family formed a network of genes differentially expressed in the two breast cancer cell lines. siRNA treatment against UGT1A also showed an increase in MTX sensitivity. Eukaryotic translation elongation factor 1 alpha 1 (EEF1A1) was overexpressed among the pancreatic cancer, leukemia and osteosarcoma cell lines, and siRNA treatment against EEF1A1 produced a chemosensitization toward MTX., Conclusions: Biological association networks identified DKK1, UGT1As and EEF1A1 as important gene nodes in MTX-resistance. Treatments using siRNA technology against these three genes showed chemosensitization toward MTX.
- Published
- 2009
- Full Text
- View/download PDF
36. Role of caveolin 1, E-cadherin, Enolase 2 and PKCalpha on resistance to methotrexate in human HT29 colon cancer cells.
- Author
-
Selga E, Morales C, Noé V, Peinado MA, and Ciudad CJ
- Abstract
Background: Methotrexate is one of the earliest cytotoxic drugs used in cancer therapy, and despite the isolation of multiple other folate antagonists, methotrexate maintains its significant role as a treatment for different types of cancer and other disorders. The usefulness of treatment with methotrexate is limited by the development of drug resistance, which may be acquired through different ways. To get insights into the mechanisms associated with drug resistance and sensitization we performed a functional analysis of genes deregulated in methotrexate resistant cells, either due to its co-amplification with the dhfr gene or as a result of a transcriptome screening using microarrays., Methods: Gene expression levels were compared between triplicate samples from either HT29 sensitive cells and resistant to 10-5 M MTX by hybridization to the GeneChip(R) HG U133 PLUS 2.0 from Affymetrix. After normalization, a list of 3-fold differentially expressed genes with a p-value < 0.05 including multiple testing correction (Benjamini and Hochberg false discovery rate) was generated. RT-Real-time PCR was used to validate the expression levels of selected genes and copy-number was determined by qPCR. Functional validations were performed either by siRNAs or by transfection of an expression plasmid., Results: Genes adjacent to the dhfr locus and included in the 5q14 amplicon were overexpressed in HT29 MTX-resistant cells. Treatment with siRNAs against those genes caused a slight reduction in cell viability in both HT29 sensitive and resistant cells. On the other hand, microarray analysis of HT29 and HT29 MTX resistant cells unveiled overexpression of caveolin 1, enolase 2 and PKCalpha genes in resistant cells without concomitant copy number gain. siRNAs against these three genes effectively reduced cell viability and caused a decreased MTX resistance capacity. Moreover, overexpression of E-cadherin, which was found underexpressed in MTX-resistant cells, also sensitized the cells toward the chemotherapeutic agent. Combined treatments targeting siRNA inhibition of caveolin 1 and overexpression of E-cadherin markedly reduced cell viability in both sensitive and MTX-resistant HT29 cells., Conclusion: We provide functional evidences indicating that caveolin 1 and E-cadherin, deregulated in MTX resistant cells, may play a critical role in cell survival and may constitute potential targets for coadjuvant therapy.
- Published
- 2008
- Full Text
- View/download PDF
37. Transcriptional regulation of aldo-keto reductase 1C1 in HT29 human colon cancer cells resistant to methotrexate: role in the cell cycle and apoptosis.
- Author
-
Selga E, Noé V, and Ciudad CJ
- Subjects
- 20-Hydroxysteroid Dehydrogenases physiology, Base Sequence, Drug Resistance, Neoplasm, HT29 Cells, Humans, Molecular Sequence Data, Promoter Regions, Genetic, RNA, Small Interfering pharmacology, Sp1 Transcription Factor metabolism, Tetrahydrofolate Dehydrogenase genetics, Transcription, Genetic, 20-Hydroxysteroid Dehydrogenases genetics, Antimetabolites, Antineoplastic pharmacology, Apoptosis drug effects, Cell Cycle drug effects, Gene Expression Regulation, Neoplastic, Methotrexate pharmacology
- Abstract
While studying differentially expressed genes between sensitive and 10(-5)M Methotrexate (MTX) resistant HT29 human colon cancer cells, we identified some members of the aldo-keto reductase (AKR) superfamily. The study was followed with the member AKR1C1 (EC 1.1.1.213), validating its increase in mRNA and protein levels in MTX resistant cells. The genomic content for AKR1C1 remained unchanged between sensitive and resistant cells, thereby excluding a mechanism of AKR1C1 gene amplification. Thus, we cloned the AKR1C1 human promoter and performed luciferase experiments that revealed a transcriptional regulation of the gene in the resistant cells. Computational studies showed a putative binding site for the transcription factor Sp1. The co-transfection of Sp1 or Sp3 with different constructs of AKR1C1 promoter deletions, including and excluding the proximal GC-box, demonstrated a key role for these factors in regulating AKR1C1 transcriptional activity. Gel-shift assays revealed an increase in Sp1 and Sp3 binding in resistant compared to sensitive cells, without differences in Sp1 protein levels. Dephosphorylation of the extracts coincided with a decrease in Sp1 binding, which is consistent with a process of regulation of Sp1 by phosphorylation. We also investigated the possible relationship between AKR1C1 expression and MTX action. Overexpression of AKR1C1 counteracted the S-phase accumulation of cells and apoptosis caused by MTX treatment. This suggests a role of AKR1C1 in cell proliferation. Finally, overexpression of AKR1C1 in MTX sensitive HT29 cells conferred resistance to the chemotherapeutic agent and silencing of AKR1C1 by means of iRNA technology sensitized the cells to MTX.
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.