78 results on '"Morte B"'
Search Results
2. CIBERER: Spanish national network for research on rare diseases: A highly productive collaborative initiative
- Author
-
Luque J, Mendes I, Gómez B, Morte B, de Heredia ML, Herreras E, Corrochano V, Bueren J, Gallano P, Artuch-Iriberri R, Fillat C, Pérez-Jurado LA, Montoliu L, Carracedo Á, Millán JM, Webb SM, Palau F, CIBERER Network, and Lapunzina P
- Subjects
research network ,new therapeutic approaches ,rare diseases ,genetics ,novel genes - Abstract
CIBER (Center for Biomedical Network Research; Centro de Investigacion Biomedica En Red) is a public national consortium created in 2006 under the umbrella of the Spanish National Institute of Health Carlos III (ISCIII). This innovative research structure comprises 11 different specific areas dedicated to the main public health priorities in the National Health System. CIBERER, the thematic area of CIBER focused on rare diseases (RDs) currently consists of 75 research groups belonging to universities, research centers, and hospitals of the entire country. CIBERER's mission is to be a center prioritizing and favoring collaboration and cooperation between biomedical and clinical research groups, with special emphasis on the aspects of genetic, molecular, biochemical, and cellular research of RDs. This research is the basis for providing new tools for the diagnosis and therapy of low-prevalence diseases, in line with the International Rare Diseases Research Consortium (IRDiRC) objectives, thus favoring translational research between the scientific environment of the laboratory and the clinical setting of health centers. In this article, we intend to review CIBERER's 15-year journey and summarize the main results obtained in terms of internationalization, scientific production, contributions toward the discovery of new therapies and novel genes associated to diseases, cooperation with patients' associations and many other topics related to RD research.
- Published
- 2022
3. Late Maternal Hypothyroidism Alters the Expression of Camk4 in Neocortical Subplate Neurons: A Comparison with Nurr1 Labeling
- Author
-
Navarro, D., Alvarado, M., Morte, B., Berbel, D., Sesma, J., Pacheco, P., Morreale de Escobar, G., Bernal, J., and Berbel, P.
- Published
- 2014
- Full Text
- View/download PDF
4. EVIDENCE FOR A ROLE OF UNLIGANDED THYROID HORMONE RECEPTORS ON CEREBROCORTICAL NEURONS IN PRIMARY CULTURE: OP27
- Author
-
Gil-Ibañez, P, Morte, B, and Bernal, J
- Published
- 2013
5. Thyroid hormone action in the brain of mice with inactivated Slc16a2 and Slc7a8 genes: P11-18
- Author
-
Fernandez, B. N., Nunes, V., Palacin, M., Morte, B., and Bernal, J.
- Published
- 2012
6. Src kinases catalytic activity regulates proliferation, migration and invasiveness of MDA-MB-231 breast cancer cells: P06-63
- Author
-
Sánchez-Bailón, Ma. P., Calcabrini, A., Gómez-Domínguez, D., Morte, B., Martín-Forero, E., Gómez-López, G., and Martín-Pérez, J.
- Published
- 2012
7. A temporary compendium of thyroid hormone target genes in brain
- Author
-
Chatonnet, F., primary, Flamant, F., additional, and Morte, B., additional
- Published
- 2015
- Full Text
- View/download PDF
8. Late Maternal Hypothyroidism Alters the Expression of Camk4 in Neocortical Subplate Neurons: A Comparison with Nurr1 Labeling
- Author
-
Navarro, D., primary, Alvarado, M., additional, Morte, B., additional, Berbel, D., additional, Sesma, J., additional, Pacheco, P., additional, Morreale de Escobar, G., additional, Bernal, J., additional, and Berbel, P., additional
- Published
- 2013
- Full Text
- View/download PDF
9. Identification of Novel GH-regulated Genes in C2C12 Cells
- Author
-
Resmini, E., additional, Morte, B., additional, Sorianello, E., additional, Gallardo, E., additional, de Luna, N., additional, Illa, I., additional, Zorzano, A., additional, Bernal, J., additional, and Webb, S., additional
- Published
- 2011
- Full Text
- View/download PDF
10. Cell-specific effects of thyroid hormone on RC3/neurogranin expression in rat brain.
- Author
-
Iniguez, M A, primary, De Lecea, L, additional, Guadano-Ferraz, A, additional, Morte, B, additional, Gerendasy, D, additional, Sutcliffe, J G, additional, and Bernal, J, additional
- Published
- 1996
- Full Text
- View/download PDF
11. Thyroid hormone regulation of rhes, a novel Ras homolog gene expressed in the striatum
- Author
-
Vargiu, P., Morte, B., Manzano, J., Perez, J., Abajo, R. de, Sutcliffe, J. Gregor, and Bernal, J.
- Published
- 2001
- Full Text
- View/download PDF
12. Identification of a cis-acting element that interferes with thyroid hormone induction of the neurogranin (NRGN) gene
- Author
-
Morte, B., Arrieta, C. Martnez de, Manzano, J., Coloma, A., and Bernal, J.
- Published
- 1999
- Full Text
- View/download PDF
13. Transcriptional induction of RC3/neurogranin by thyroid hormone: differential neuronal sensitivity is not correlated with thyroid hormone receptor distribution in the brain
- Author
-
o-Ferraz, A. Guada, Escamez, M. Jose, Morte, B., Vargiu, P., and Bernal, J.
- Published
- 1997
- Full Text
- View/download PDF
14. Vita et transitus S. Hieronymi, siue Epistolae de eodem in unum collectae
- Author
-
Pseudo-Agustí. De laudibus B. Hieronymi, aut, Pseudo-Ciril de Jerusalem. De miraculis B. Hieronymi, aut, Pseudo-Eusebi de Cremona. Epistola de morte B. Hieronymi, aut, and Posa, Pere, ed.imp.lib
- Subjects
Jeroni, Sant Obres anteriors a 1800 ,Jeroni, sant, ca. 342-420 Obres anteriors al 1800 ,DIG-BH - Abstract
Sign.: [2], a-t8. - L'obra de Pseudo-Ciril de Jerusalem en català Epistola de morte B. Hieronymi / Pseudo-Eusebi de Cremona. De laudibus B. Hieronymi / Pseudo-Agustí. De miraculis B. Hyeronimi / Pseudo-Ciril de Jerusalem L. gòt. - 2 mides. - 27 lín. - Capll. grav. - Filigr.: corona
- Published
- 1492
15. Analysis of the thyroid hormone (T3)-dependent transcriptome in mouse cerebral cortex
- Author
-
Morte, B, primary
- Full Text
- View/download PDF
16. Characterization of the promoter region and flanking sequences of the neuron-specific gene RC3 (neurogranin)
- Author
-
iguez, M. A. I, Morte, B., a, A. Rodriguez-Pe, and oz, A. Mu
- Published
- 1994
- Full Text
- View/download PDF
17. Schuurs–Hoeijmakers Syndrome (PACS1 Neurodevelopmental Disorder): Seven Novel Patients and a Review
- Author
-
Beatriz Olivia Camarena Gómez, Angel Carracedo, Beatriz Morte, María Palomares-Bralo, Patricia Arias, Carmen Ayuso, Marta Pacio-Míguez, Fernando Santos-Simarro, Jair Tenorio-Castaño, Alma Kuechler, Pedro Arias, Feliciano J. Ramos, Eduardo F Tizzano, Sergio Ramos, Fermina López-Grondona, Luis A. Pérez-Jurado, María Pilar Méndez Perez, Julián Nevado, Berta Almoguera, Francisco Barros, Enrique Galán-Gómez, Sixto García-Miñaur, Alba Alcochea, Irene Valenzuela, Victor Martinez-Glez, Frank J. Kaiser, Ivon Cuscó, I. Lorda-Sánchez, Juan Pié, Pablo Lapunzina, Juan Carrión, UAM. Departamento de Medicina, Institut Català de la Salut, [Tenorio-Castaño J] CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, 28029 Madrid, Spain. Overgrowth Syndromes Laboratory, INGEMM, Instituto de Genética Médica y Molecular, IdiPAZ, Hospital Universitario la Paz, Universidad Autónoma de Madrid (UAM), 28046 Madrid, Spain. The SIDE Consortium: Spanish Intellectual Disability Exome Consortium, 28046 Madrid, Spain. Ithaca, European Reference Network, Hospital Universitario La Paz, 28046 Madrid, Spain. [Morte B] CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, 28029 Madrid, Spain. The SIDE Consortium: Spanish Intellectual Disability Exome Consortium, 28046 Madrid, Spain. [Nevado J] CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, 28029 Madrid, Spain. The SIDE Consortium: Spanish Intellectual Disability Exome Consortium, 28046 Madrid, Spain. Ithaca, European Reference Network, Hospital Universitario La Paz, 28046 Madrid, Spain. Structural and Functional Genomics—INGEMM, Instituto de Genética Médica y Molecular, IdiPAZ, Hospital Universitario la Paz, Universidad Autónoma de Madrid (UAM), 28046 Madrid, Spain. [Martinez-Glez V] CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, 28029 Madrid, Spain. Ithaca, European Reference Network, Hospital Universitario La Paz, 28046 Madrid, Spain. Structural and Functional Genomics—INGEMM, Instituto de Genética Médica y Molecular, IdiPAZ, Hospital Universitario la Paz, Universidad Autónoma de Madrid (UAM), 28046 Madrid, Spain. Clinical Genetics—INGEMM, Instituto de Genética Médica y Molecular, IdiPAZ, Hospital Universitario la Paz, Universidad Autónoma de Madrid (UAM), 28046 Madrid, Spain. [Santos-Simarro F] CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, 28029 Madrid, Spain. The SIDE Consortium: Spanish Intellectual Disability Exome Consortium, 28046 Madrid, Spain. Ithaca, European Reference Network, Hospital Universitario La Paz, 28046 Madrid, Spain. Clinical Genetics—INGEMM, Instituto de Genética Médica y Molecular, IdiPAZ, Hospital Universitario la Paz, Universidad Autónoma de Madrid (UAM), 28046 Madrid, Spain. [García-Miñaúr S] CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, 28029 Madrid, Spain. Ithaca, European Reference Network, Hospital Universitario La Paz, 28046 Madrid, Spain. Clinical Genetics—INGEMM, Instituto de Genética Médica y Molecular, IdiPAZ, Hospital Universitario la Paz, Universidad Autónoma de Madrid (UAM), 28046 Madrid, Spain. [Valenzuela I, Tizzano E] Ithaca, European Reference Network, Hospital Universitario La Paz, 28046 Madrid, Spain. Àrea de Genètica Clínica i Molecular, Vall d’Hebron Hospital Universitari, Barcelona, Spain. Grup de Genètica de la Medicina, Vall d’Hebron Institut de Recerca (VHIR), Barcelona, Spain. [Cuscó I] Àrea de Genètica Clínica i Molecular, Vall d’Hebron Hospital Universitari, Barcelona, Spain. Grup de Genètica de la Medicina, Vall d’Hebron Institut de Recerca (VHIR), Barcelona, Spain, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
0301 basic medicine ,Pediatrics ,medicine.medical_specialty ,PACS1 Gene ,Medicina ,Medizin ,Intellectual disability ,Disease ,030105 genetics & heredity ,+T%22">Pathogenic variant c.607C > T ,QH426-470 ,phosphofurin acidic cluster sorting protein 1 ,pathogenic variant c.607C > ,Trastorns neuroconductuals - Aspectes genètics ,Very frequent ,03 medical and health sciences ,Neurodevelopmental disorder ,Other subheadings::Other subheadings::/genetics [Other subheadings] ,Genetics ,Medicine ,Craniofacial ,education ,rare disorders ,PACS1 ,Genetics (clinical) ,education.field_of_study ,business.industry ,Otros calificadores::Otros calificadores::/genética [Otros calificadores] ,Deficiència mental - Aspectes genètics ,trastornos mentales::trastornos del desarrollo neurológico [PSIQUIATRÍA Y PSICOLOGÍA] ,afecciones patológicas, signos y síntomas::signos y síntomas::manifestaciones neurológicas::manifestaciones neuroconductuales::discapacidad intelectual [ENFERMEDADES] ,Congenital malformations ,Schuurs-Hoeijmakers syndrome ,medicine.disease ,+T%22">pathogenic variant c.607C > T ,030104 developmental biology ,intellectual disability ,Phosphofurin acidic cluster sorting protein 1 ,Schuurs–Hoeijmakers syndrome ,Rare disorders ,Phospho-furin acidic cluster sorting protein 1 ,business ,Mental Disorders::Neurodevelopmental Disorders [PSYCHIATRY AND PSYCHOLOGY] ,Pathological Conditions, Signs and Symptoms::Signs and Symptoms::Neurologic Manifestations::Neurobehavioral Manifestations::Intellectual Disability [DISEASES] ,+phosphofurin+acidic+cluster+sorting+protein+1%22">Pathogenic variant c.607C > phosphofurin acidic cluster sorting protein 1 - Abstract
Schuurs–Hoeijmakers syndrome (SHMS) or PACS1 Neurodevelopmental disorder is a rare disorder characterized by intellectual disability, abnormal craniofacial features and congenital malformations. SHMS is an autosomal dominant hereditary disease caused by pathogenic variants in the PACS1 gene. PACS1 is a trans-Golgi-membrane traffic regulator that directs protein cargo and several viral envelope proteins. It is upregulated during human embryonic brain development and has low expression after birth. So far, only 54 patients with SHMS have been reported. In this work, we report on seven new identified SHMS individuals with the classical c.607C > T: p.Arg206Trp PACS1 pathogenic variant and review clinical and molecular aspects of all the patients reported in the literature, providing a summary of clinical findings grouped as very frequent (≥75% of patients), frequent (50–74%), infrequent (26–49%) and rare (less than ≤25%), This work was possible thanks to the funding provided by the project “Proyecto Piloto para la mejora del diagnóstico genético en personas y familias afectadas o con sospecha de padecer enfermedades raras de base genética” of the Ministry of Health, under the grant BOCM-20181126-24 provided by the Consejería de Sanidad de la Comunidad de Madrid. Funding to J.P. and F.J.R. was partially provided by the group research grant DGA/FEDER B32_17R/B32_20R
- Published
- 2021
18. CSVS, a crowdsourcing database of the Spanish population genetic variability
- Author
-
Peña-Chilet, María, Roldán Gema, Perez-Florido, Javier, Ortuño, Francisco M., Carmona, Rosario, Aquino, Virginia, Lopez-Lopez, Daniel, Loucera, Carlos, Fernandez-Rueda, Jose L., Gallego, Asunción, García-García, Francisco, González-Neira, Anna, Pita, Guillermo, Núñez-Torres, Rocío, Santoyo-López, Javier, Ayuso, Carmen, Minguez, Pablo, Avila-Fernandez, Almudena, Corton, Marta, Moreno-Pelayo, Miguel Ángel, Morin, Matías, Gallego-Martinez, Alvaro, Lopez-Escamez, Jose A., Borrego, Salud, Antiñolo, Guillermo, Amigo, Jorge, Salgado-Garrido, Josefa, Pasalodos-Sanchez, Sara, Morte, Beatriz, The Spanish Exome Crowdsourcing Consortium, Carracedo Álvarez, Ángel, Alonso, Ángel, Dopazo, Joaquín, Grinberg Vaisman, Daniel Raúl, [Peña-Chilet,M, Roldán,G, Perez-Florido,J, Ortuño,FM, Carmona,R, Aquino,V, Lopez-Lopez,D, Loucera,C, Fernandez-Rueda,JL, Dopazo,J] Clinical Bioinformatics Area, Fundacion Progreso y Salud (FPS), Hospital Virgen del Rocío, Sevilla, Spain. [Peña-Chilet,M, Dopazo,J] Bioinformatics in Rare Diseases (BiER), Center for Biomedical Network Research on Rare Diseases (CIBERER), ISCIII, Sevilla, Spain. [Peña-Chilet,M, Dopazo,J] Computational Systems Medicine group, Institute of Biomedicine of Seville (IBIS) Hospital Virgen del Rocío, Sevilla, Spain. [Perez-Florido,J, Dopazo,J] Functional Genomics Node, FPS/ELIXIR-ES, Hospital Virgen del Rocío, Sevilla, Spain. [Gallego,A] Sistemas Genomicos, Paterna, Valencia, Spain. [García-Garcia,F] Unidad de Bioinformatica y Bioestadística, Centro de Investigacion Príncipe Felipe (CIPF), Valencia, Spain. [González-Neira,A, Pita,G, Núñez-Torres,R] Human Genotyping Unit–Centro Nacional de Genotipado (CEGEN), Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain. [Santoyo-López,J] Edinburgh Genomics, The University of Edinburgh, Edinburgh, UK. [Ayuso,C, Minguez,P, Avila-Fernandez,A, Corton,M] Department of Genetics, Instituto de Investigacion Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autonoma de Madrid (IIS-FJD, UAM), Madrid, Spain. [Minguez,P] Center for Biomedical Network Research on Rare Diseases (CIBERER), ISCIII, Madrid, Spain. [Moreno-Pelayo,MÁ, Morin,M] Servicio de Genetica, Ramón y Cajal Institute of Health Research (IRYCIS) and Biomedical Network Research Centre on Rare Diseases (CIBERER), Madrid, Spain, [Gallego-Martinez,A, Lopez-Escamez,JA] Otology & Neurotology Group CTS 495, Department of Genomic Medicine, Centre for Genomics and Oncological Research (GENYO), Pfizer University of Granada, Granada, Spain. [Gallego-Martinez,A, Lopez-Escamez,JA] Department of Otolaryngology, Instituto de Investigacion Biosanitaria, IBS. GRANADA, Hospital Universitario Virgen de las Nieves, Universidad de Granada, Granada, Spain. [Borrego,S, Antiñolo,G] Department of Maternofetal Medicine, Genetics and Reproduction, Institute of Biomedicine of Seville (IBIS), University Hospital Virgen del Rocío /CSIC/University of Seville, Seville, Spain. [Borrego,S, Antiñolo,G] Centre for Biomedical Network Research on Rare Diseases (CIBERER), Seville, Spain. [Amigo,J, Carracedo,Á] Fundacion Pública Galega de Medicina Xenómica, SERGAS, IDIS, Santiago de Compostela, Spain. [Salgado-Garrido,J, Pasalodos-Sanchez,S, Alonso,Á] Navarrabiomed-IdiSNA, Complejo Hospitalario de Navarra, Universidad Publica de Navarra (UPNA), IdiSNA (Navarra Institute for Health Research), Pamplona, Navarra, Spain. [Morte,B] Undiagnosed Rare Diseases Programme (ENoD). Center for Biomedical Research on Rare Diseases (CIBERER), ISCIII, Madrid, Spain. [Carracedo,Á] Grupo de Medicina Xenomica, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), CIMUS, Universidade de Santiago de Compostela, Santiago de Compostela, España., Spanish Ministry of Economy and Competitiveness [SAF2017-88908-R, PT17/0009/0006 to J.D., PI19/00321 and CIBERER ACCI-06/07/0036 to C.A., PI14-948, PI17-1659 and CIBERER ACCI-06/07/0036 to M.A.M.P.], Regional Government of Madrid, RAREGenomics CM [B2017/BMD-3721 to C.A. and B2017/BMD3721 to M.A.M.P.], all co-funded with European Regional Development Funds (ERDF) as well as EU H2020-INFRADEV-1-2015-1 ELIXIR-EXCELERATE [676559], University Chair UAM-IIS-FJD of Genomic Medicine and the Ramon Areces Foundation also supported this work. Funding for open access charge: Spanish Ministry of Economy and Competitiveness [SAF2017-88908-R]., Ministerio de Economía, Industria y Competitividad (España), Comunidad de Madrid, European Commission, Fundación Ramón Areces, Ministerio de Economía y Competitividad (España), Centro de Investigación Biomédica en Red Enfermedades Raras (España), and Universidad Autónoma de Madrid
- Subjects
Genetics, population ,Població ,AcademicSubjects/SCI00010 ,computer.software_genre ,Bases de dades ,Organisms::Eukaryota::Animals::Chordata::Vertebrates::Mammals::Primates::Haplorhini::Catarrhini::Hominidae::Humans [Medical Subject Headings] ,0302 clinical medicine ,Gene Frequency ,Genética de poblaciones ,Databases, Genetic ,Database Issue ,Exome ,Precision Medicine ,0303 health sciences ,education.field_of_study ,Database ,Disciplines and Occupations::Natural Science Disciplines::Biological Science Disciplines::Biology::Genetics::Genetics, Population [Medical Subject Headings] ,Chromosome Mapping ,Genomics ,Bases de datos genéticas ,Phenomena and Processes::Genetic Phenomena::Genetic Structures::Genome::Genome, Human [Medical Subject Headings] ,3. Good health ,Databases, genetic ,Information Science::Information Science::Data Collection::Crowdsourcing [Medical Subject Headings] ,Crowdsourcing ,Disciplines and Occupations::Natural Science Disciplines::Biological Science Disciplines::Biology::Genetics::Genomics [Medical Subject Headings] ,Analytical, Diagnostic and Therapeutic Techniques and Equipment::Investigative Techniques::Genetic Techniques::Chromosome Mapping::Physical Chromosome Mapping [Medical Subject Headings] ,Phenomena and Processes::Genetic Phenomena::Genetic Variation [Medical Subject Headings] ,Participación colectiva ,Population ,Proveïment participatiu ,Biology ,Variación genética ,Phenomena and Processes::Genetic Phenomena::Genetic Structures::Genome::Exome [Medical Subject Headings] ,Genètica de poblacions humanes ,03 medical and health sciences ,Databases ,Information Science::Information Science::Information Storage and Retrieval::Databases as Topic::Databases, Factual::Databases, Genetic [Medical Subject Headings] ,Genetic variation ,Genetics ,Humans ,Genetic variability ,Espanya ,education ,Allele frequency ,Alleles ,Phenomena and Processes::Genetic Phenomena::Genetic Structures::Genome::Genome Components::Genes::Alleles [Medical Subject Headings] ,030304 developmental biology ,Geographical Locations::Geographic Locations::Europe::Spain [Medical Subject Headings] ,Internet ,Genoma humà -- Espanya ,business.industry ,Genome, Human ,Genetic Variation ,Human population genetics ,Gene frequency ,Frecuencia génica ,Genetics, Population ,Spain ,Personalized medicine ,business ,Phenomena and Processes::Genetic Phenomena::Gene Frequency [Medical Subject Headings] ,computer ,030217 neurology & neurosurgery ,Imputation (genetics) ,Software - Abstract
The knowledge of the genetic variability of the local population is of utmost importance in personalized medicine and has been revealed as a critical factor for the discovery of new disease variants. Here, we present the Collaborative Spanish Variability Server (CSVS), which currently contains more than 2000 genomes and exomes of unrelated Spanish individuals. This database has been generated in a collaborative crowdsourcing effort collecting sequencing data produced by local genomic projects and for other purposes. Sequences have been grouped by ICD10 upper categories. A web interface allows querying the database removing one or more ICD10 categories. In this way, aggregated counts of allele frequencies of the pseudo-control Spanish population can be obtained for diseases belonging to the category removed. Interestingly, in addition to pseudo-control studies, some population studies can be made, as, for example, prevalence of pharmacogenomic variants, etc. In addition, this genomic data has been used to define the first Spanish Genome Reference Panel (SGRP1.0) for imputation. This is the first local repository of variability entirely produced by a crowdsourcing effort and constitutes an example for future initiatives to characterize local variabilityworldwide. CSVS is also part of the GA4GH Beacon network., Spanish Ministry of Economy and Competitiveness SAF2017-88908-R PT17/0009/0006 PI19/00321 CIBERER ACCI-06/07/0036 PI14-948 PI171659, Regional Government of Madrid, RAREGenomicsCM B2017/BMD3721 B2017/BMD-3721, European Union (EU), European Union (EU) 676559, University Chair UAM-IIS-FJD of Genomic Medicine, Ramon Areces Foundation
- Published
- 2020
19. Expanding the genetic and phenotypic spectrum of congenital myasthenic syndrome: new homozygous VAMP1 splicing variants in 2 novel individuals.
- Author
-
Cotrina-Vinagre FJ, Rodríguez-García ME, Del Pozo-Filíu L, Hernández-Laín A, Arteche-López A, Morte B, Sevilla M, Pérez-Jurado LA, Quijada-Fraile P, Camacho A, and Martínez-Azorín F
- Subjects
- Female, Humans, Male, Alternative Splicing genetics, Exome Sequencing, Mutation, Protein Isoforms genetics, RNA Splicing genetics, Infant, Child, Preschool, Homozygote, Myasthenic Syndromes, Congenital genetics, Myasthenic Syndromes, Congenital pathology, Phenotype, Vesicle-Associated Membrane Protein 1 genetics
- Abstract
We report the cases of two Spanish pediatric patients with hypotonia, muscle weakness and feeding difficulties at birth. Whole-exome sequencing (WES) uncovered two new homozygous VAMP1 (Vesicle Associated Membrane Protein 1) splicing variants, NM_014231.5:c.129+5 G > A in the boy patient (P1) and c.341-24_341-16delinsAGAAAA in the girl patient (P2). This gene encodes the vesicle-associated membrane protein 1 (VAMP1) that is a component of a protein complex involved in the fusion of synaptic vesicles with the presynaptic membrane. VAMP1 has a highly variable C-terminus generated by alternative splicing that gives rise to three main isoforms (A, B and D), being VAMP1A the only isoform expressed in the nervous system. In order to assess the pathogenicity of these variants, expression experiments of RNA for VAMP1 were carried out. The c.129+5 G > A and c.341-24_341-16delinsAGAAAA variants induced aberrant splicing events resulting in the deletion of exon 2 (r.5_131del; p.Ser2TrpfsTer7) in the three isoforms in the first case, and the retention of the last 14 nucleotides of the 3' of intron 4 (r.340_341ins341-14_341-1; p.Ile114AsnfsTer77) in the VAMP1A isoform in the second case. Pathogenic VAMP1 variants have been associated with autosomal dominant spastic ataxia 1 (SPAX1) and with autosomal recessive presynaptic congenital myasthenic syndrome (CMS). Our patients share the clinical manifestations of CMS patients with two important differences: they do not show the typical electrophysiological pattern that suggests pathology of pre-synaptic neuromuscular junction, and their muscular biopsies present hypertrophic fibers type 1. In conclusion, our data expand both genetic and phenotypic spectrum associated with VAMP1 variants., (© 2024. The Author(s), under exclusive licence to The Japan Society of Human Genetics.)
- Published
- 2024
- Full Text
- View/download PDF
20. Utility of exome sequencing for the diagnosis of pediatric-onset neuromuscular diseases beyond diagnostic yield: a narrative review.
- Author
-
Piñeros-Fernández MC, Morte B, and García-Giménez JL
- Subjects
- Child, Humans, Exome Sequencing, Genetic Testing, Patient Selection, Neuromuscular Diseases diagnosis, Neuromuscular Diseases genetics
- Abstract
Diagnosis of neuromuscular diseases (NMD) can be challenging because of the heterogeneity of this group of diseases. This review aimed to describe the diagnostic yield of whole exome sequencing (WES) for pediatric-onset neuromuscular disease diagnosis, as well as other benefits of this approach in patient management since WES can contribute to appropriate treatment selection in NMD patients. WES increases the possibility of reaching a conclusive genetic diagnosis when other technologies have failed and even exploring new genes not previously associated with a specific NMD. Moreover, this strategy can be useful when a dual diagnosis is suspected in complex congenital anomalies and undiagnosed cases., (© 2023. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
21. Thyroid hormone regulators in human cerebral cortex development.
- Author
-
Bernal J, Morte B, and Diez D
- Subjects
- Animals, Humans, Mice, Monocarboxylic Acid Transporters genetics, Monocarboxylic Acid Transporters metabolism, Thyroid Hormones metabolism, Muscle Hypotonia genetics, Muscular Atrophy metabolism, Cerebral Cortex metabolism, Symporters genetics, Symporters metabolism, Mental Retardation, X-Linked genetics
- Abstract
Brain development is critically dependent on the timely supply of thyroid hormones. The thyroid hormone transporters are central to the action of thyroid hormones in the brain, facilitating their passage through the blood-brain barrier. Mutations of the monocarboxylate transporter 8 (MCT8) cause the Allan-Herndon-Dudley syndrome, with altered thyroid hormone concentrations in the blood and profound neurological impairment and intellectual deficit. Mouse disease models have revealed interplay between transport, deiodination, and availability of T3 to receptors in specific cells. However, the mouse models are not satisfactory, given the fundamental differences between the mouse and human brains. The goal of the present work is to review human neocortex development in the context of thyroid pathophysiology. Recent developments in single-cell transcriptomic approaches aimed at the human brain make it possible to profile the expression of thyroid hormone regulators in single-cell RNA-Seq datasets of the developing human neocortex. The data provide novel insights into the specific cellular expression of thyroid hormone transporters, deiodinases, and receptors.
- Published
- 2022
- Full Text
- View/download PDF
22. Single-Cell Transcriptome Profiling of Thyroid Hormone Effectors in the Human Fetal Neocortex: Expression of SLCO1C1 , DIO2 , and THRB in Specific Cell Types.
- Author
-
Diez D, Morte B, and Bernal J
- Subjects
- Animals, Humans, Mice, Neocortex cytology, Iodothyronine Deiodinase Type II, Gene Expression genetics, Gene Expression Profiling methods, Iodide Peroxidase genetics, Iodide Peroxidase metabolism, Neocortex embryology, Neocortex metabolism, Organic Anion Transporters genetics, Organic Anion Transporters metabolism, Thyroid Hormone Receptors beta genetics, Thyroid Hormone Receptors beta metabolism, Thyroid Hormones genetics, Thyroid Hormones metabolism
- Abstract
Background: Thyroid hormones are crucial for brain development, acting through the thyroid hormone nuclear receptors (TR)α1 and β to control gene expression. Triiodothyronine (T3), the receptor-ligand, is transported into the brain from the blood by the monocarboxylate transporter 8 (MCT8). Another source of brain T3 is from the local deiodination of thyroxine (T4) by type 2 deiodinase (DIO2). While these mechanisms are very similar in mice and humans, important species-specific differences confound our understanding of disease using mouse models. To fill this knowledge gap on thyroid hormone action in the human fetal brain, we analyzed the expression of transporters, DIO2, and TRs, which we call thyroid hormone effectors, at single-cell resolution. Methods: We analyzed publicly available single-cell transcriptome data sets of isolated cerebral cortex neural cells from three different studies, with expression data from 393 to almost 40,000 cells. We generated Uniform Manifold Approximation and Projection scatterplots and cell clusters to identify differentially expressed genes between clusters, and correlated their gene signatures with the expression of thyroid effectors. Results: The radial glia, mainly the outer radial glia, and astrocytes coexpress SLCO1C1 and DIO2, indicating close cooperation between the T4 transporter OATP1C1 and DIO2 in local T3 formation. Strikingly, THRB was mainly present in two classes of interneurons: a majority expressing CALB2 /calretinin, from the caudal ganglionic eminence, and in somatostatin-expressing interneurons from the medial ganglionic eminence. By contrast, many cell types express SLC16A2 and THRA . Conclusions: SLCO1C1 and DIO2 coexpression in the outer radial glia, the universal stem cell of the cerebral cortex, highlights the likely importance of brain-generated T3 in neurogenesis. The unique expression of THRB in discrete subsets of interneurons is a novel finding whose pathophysiological meaning deserves further investigation.
- Published
- 2021
- Full Text
- View/download PDF
23. Brain Gene Expression in Systemic Hypothyroidism and Mouse Models of MCT8 Deficiency: The Mct8-Oatp1c1-Dio2 Triad.
- Author
-
Morte B, Gil-Ibañez P, Heuer H, and Bernal J
- Subjects
- Animals, Brain physiopathology, Cerebral Cortex metabolism, Gene Expression Profiling, Hypothyroidism metabolism, Hypothyroidism physiopathology, Iodide Peroxidase metabolism, Mental Retardation, X-Linked metabolism, Mental Retardation, X-Linked physiopathology, Mice, Mice, Knockout, Monocarboxylic Acid Transporters metabolism, Muscle Hypotonia metabolism, Muscle Hypotonia physiopathology, Muscular Atrophy metabolism, Muscular Atrophy physiopathology, Neostriatum metabolism, Organic Cation Transport Proteins metabolism, Symporters metabolism, Iodothyronine Deiodinase Type II, Brain metabolism, Gene Expression, Hypothyroidism genetics, Iodide Peroxidase genetics, Mental Retardation, X-Linked genetics, Monocarboxylic Acid Transporters genetics, Muscle Hypotonia genetics, Muscular Atrophy genetics, Organic Cation Transport Proteins genetics, Symporters genetics, Thyroxine metabolism, Triiodothyronine metabolism
- Abstract
Background: The monocarboxylate transporter 8 (Mct8) protein is a primary thyroxine (T4) and triiodothyronine (T3) (thyroid hormone [TH]) transporter. Mutations of the MCT8-encoding, SLC16A2 gene alter thyroid function and TH metabolism and severely impair neurodevelopment (Allan-Herndon-Dudley syndrome [AHDS]). Mct8-deficient mice manifest thyroid alterations but lack neurological signs. It is believed that Mct8 deficiency in mice is compensated by T4 transport through the Slco1c1 -encoded organic anion transporter polypeptide 1c1 (Oatp1c1). This allows local brain generation of sufficient T3 by the Dio2 -encoded type 2 deiodinase, thus preventing brain hypothyroidism. The Slc16a2 / Slco1c1 (MO) and Slc16a2 / Dio2 (MD) double knockout (KO) mice lacking T4 and T3 transport, or T3 transport and T4 deiodination, respectively, should be appropriate models of AHDS. Our goal was to compare the cerebral hypothyroidism of systemic hypothyroidism (SH) caused by thyroid gland blockade with that present in the double KO mice. Methods: We performed RNA sequencing by using RNA from the cerebral cortex and striatum of SH mice and the double KO mice on postnatal days 21-23. Real-time polymerase chain reaction was used to confirm RNA-Seq results in replicate biological samples. Cell type involvement was assessed from cell type-enriched genes. Functional genomic differences were analyzed by functional node activity based on a probabilistic graphical model. Results: Each of the three conditions gave a different pattern of gene expression, with partial overlaps. SH gave a wider and highest variation of gene expression than MD or MO. This was partially due to secondary gene responses to hypothyroidism. The set of primary transcriptional T3 targets showed a tighter overlap, but quantitative gene responses indicated that the gene responses in SH were more severe than in MD or MO. Examination of cell type-enriched genes indicated cellular differences between the three conditions. Conclusions: The results indicate that the neurological impairment of AHDS is too severe to be fully explained by TH deprivation only.
- Published
- 2021
- Full Text
- View/download PDF
24. Schuurs-Hoeijmakers Syndrome ( PACS1 Neurodevelopmental Disorder): Seven Novel Patients and a Review.
- Author
-
Tenorio-Castaño J, Morte B, Nevado J, Martinez-Glez V, Santos-Simarro F, García-Miñaúr S, Palomares-Bralo M, Pacio-Míguez M, Gómez B, Arias P, Alcochea A, Carrión J, Arias P, Almoguera B, López-Grondona F, Lorda-Sanchez I, Galán-Gómez E, Valenzuela I, Méndez Perez MP, Cuscó I, Barros F, Pié J, Ramos S, Ramos FJ, Kuechler A, Tizzano E, Ayuso C, Kaiser FJ, Pérez-Jurado LA, Carracedo Á, The ENoD-Ciberer Consortium, The Side Consortium, and Lapunzina P
- Subjects
- Abnormalities, Multiple genetics, Female, Humans, Intellectual Disability genetics, Male, Mutation genetics, Phenotype, Syndrome, Neurodevelopmental Disorders genetics, Vesicular Transport Proteins genetics
- Abstract
Schuurs-Hoeijmakers syndrome (SHMS) or PACS1 Neurodevelopmental disorder is a rare disorder characterized by intellectual disability, abnormal craniofacial features and congenital malformations. SHMS is an autosomal dominant hereditary disease caused by pathogenic variants in the PACS1 gene. PACS1 is a trans-Golgi-membrane traffic regulator that directs protein cargo and several viral envelope proteins. It is upregulated during human embryonic brain development and has low expression after birth. So far, only 54 patients with SHMS have been reported. In this work, we report on seven new identified SHMS individuals with the classical c.607C > T: p.Arg206Trp PACS1 pathogenic variant and review clinical and molecular aspects of all the patients reported in the literature, providing a summary of clinical findings grouped as very frequent (≥75% of patients), frequent (50-74%), infrequent (26-49%) and rare (less than ≤25%).
- Published
- 2021
- Full Text
- View/download PDF
25. CSVS, a crowdsourcing database of the Spanish population genetic variability.
- Author
-
Peña-Chilet M, Roldán G, Perez-Florido J, Ortuño FM, Carmona R, Aquino V, Lopez-Lopez D, Loucera C, Fernandez-Rueda JL, Gallego A, García-Garcia F, González-Neira A, Pita G, Núñez-Torres R, Santoyo-López J, Ayuso C, Minguez P, Avila-Fernandez A, Corton M, Moreno-Pelayo MÁ, Morin M, Gallego-Martinez A, Lopez-Escamez JA, Borrego S, Antiñolo G, Amigo J, Salgado-Garrido J, Pasalodos-Sanchez S, Morte B, Carracedo Á, Alonso Á, and Dopazo J
- Subjects
- Alleles, Chromosome Mapping, Exome, Gene Frequency, Genetic Variation, Genomics, Humans, Internet, Precision Medicine methods, Spain, Crowdsourcing, Databases, Genetic, Genetics, Population methods, Genome, Human, Software
- Abstract
The knowledge of the genetic variability of the local population is of utmost importance in personalized medicine and has been revealed as a critical factor for the discovery of new disease variants. Here, we present the Collaborative Spanish Variability Server (CSVS), which currently contains more than 2000 genomes and exomes of unrelated Spanish individuals. This database has been generated in a collaborative crowdsourcing effort collecting sequencing data produced by local genomic projects and for other purposes. Sequences have been grouped by ICD10 upper categories. A web interface allows querying the database removing one or more ICD10 categories. In this way, aggregated counts of allele frequencies of the pseudo-control Spanish population can be obtained for diseases belonging to the category removed. Interestingly, in addition to pseudo-control studies, some population studies can be made, as, for example, prevalence of pharmacogenomic variants, etc. In addition, this genomic data has been used to define the first Spanish Genome Reference Panel (SGRP1.0) for imputation. This is the first local repository of variability entirely produced by a crowdsourcing effort and constitutes an example for future initiatives to characterize local variability worldwide. CSVS is also part of the GA4GH Beacon network. CSVS can be accessed at: http://csvs.babelomics.org/., (© The Author(s) 2020. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2021
- Full Text
- View/download PDF
26. Regulation of Gene Expression by Thyroid Hormone in Primary Astrocytes: Factors Influencing the Genomic Response.
- Author
-
Morte B, Gil-Ibáñez P, and Bernal J
- Subjects
- Animals, Astrocytes metabolism, Cells, Cultured, Cerebral Cortex cytology, Cerebral Cortex metabolism, Cycloheximide pharmacology, Fetus cytology, Fetus metabolism, Gene Expression Regulation, Developmental drug effects, Genome drug effects, Genome genetics, Iodide Peroxidase drug effects, Iodide Peroxidase genetics, Membrane Transport Proteins drug effects, Membrane Transport Proteins genetics, Mice, Monocarboxylic Acid Transporters, Nuclear Receptor Co-Repressor 1 drug effects, Nuclear Receptor Co-Repressor 1 genetics, Nuclear Receptor Coactivator 1 drug effects, Nuclear Receptor Coactivator 1 genetics, Protein Synthesis Inhibitors pharmacology, Receptors, Notch drug effects, Receptors, Notch metabolism, Symporters, Thyroid Hormone Receptors alpha drug effects, Thyroid Hormone Receptors alpha genetics, Thyroid Hormone Receptors beta drug effects, Thyroid Hormone Receptors beta genetics, Thyroxine, Wnt Signaling Pathway drug effects, Iodothyronine Deiodinase Type II, Astrocytes drug effects, Gene Expression Regulation drug effects, Triiodothyronine pharmacology
- Abstract
Astrocytes mediate the action of thyroid hormone in the brain on other neural cells through the production of the active hormone triiodothyronine (T3) from its precursor thyroxine. T3 has also many effects on the astrocytes in vivo and in culture, but whether these actions are directly mediated by transcriptional regulation is not clear. In this work, we have analyzed the genomic response to T3 of cultured astrocytes isolated from the postnatal mouse cerebral cortex using RNA sequencing. Cultured astrocytes express relevant genes of thyroid hormone metabolism and action encoding type 2 deiodinase (Dio2), Mct8 transporter (Slc16a2), T3 receptors (Thra1 and Thrb), and nuclear corepressor (Ncor1) and coactivator (Ncoa1). T3 changed the expression of 668 genes (4.5% of expressed genes), of which 117 were responsive to T3 in the presence of cycloheximide. The Wnt and Notch pathways were downregulated at the posttranscriptional level. Comparison with the effect of T3 on astrocyte-enriched genes in mixed cerebrocortical cultures isolated from fetal cortex revealed that the response to T3 is influenced by the degree of astrocyte maturation and that, in agreement with its physiological effects, T3 promotes the transition between the fetal and adult patterns of gene expression.
- Published
- 2018
- Full Text
- View/download PDF
27. Expression Analysis of Genes Regulated by Thyroid Hormone in Neural Cells.
- Author
-
Bernal J and Morte B
- Subjects
- Animals, Cells, Cultured, Cerebral Cortex cytology, Computational Biology methods, High-Throughput Nucleotide Sequencing, Mice, Primary Cell Culture, Reproducibility of Results, Sequence Analysis, RNA, Transcriptome, Gene Expression Profiling methods, Gene Expression Regulation drug effects, Neurons drug effects, Neurons metabolism, Thyroid Hormones metabolism, Thyroid Hormones pharmacology
- Abstract
The actions of thyroid hormones on brain development and function are due primarily to regulation of gene expression. Identification of direct transcriptional responses requires cell culture approaches given the difficulty of in vivo studies. Here, we describe the use of primary cells in culture obtained from embryonic mouse cerebral cortex, to identify the set of genes regulated directly and indirectly by T3 using RNA-Seq.
- Published
- 2018
- Full Text
- View/download PDF
28. Is the Intrinsic Genomic Activity of Thyroxine Relevant In Vivo? Effects on Gene Expression in Primary Cerebrocortical and Neuroblastoma Cells.
- Author
-
Gil-Ibáñez P, Belinchón MM, Morte B, Obregón MJ, and Bernal J
- Subjects
- Animals, Astrocytes cytology, Astrocytes enzymology, Avian Proteins agonists, Avian Proteins genetics, Avian Proteins metabolism, Cell Line, Cells, Cultured, Cerebral Cortex cytology, Cerebral Cortex enzymology, Chickens, Embryo, Mammalian cytology, Gene Expression Regulation, Neoplastic, Iodide Peroxidase genetics, Iodide Peroxidase metabolism, Mice, Mice, Knockout, Neoplasm Proteins genetics, Neoplasm Proteins metabolism, Nerve Tissue Proteins genetics, Neuroblastoma enzymology, Neuroblastoma pathology, Neurons cytology, Neurons enzymology, Recombinant Proteins chemistry, Recombinant Proteins metabolism, Thyroid Hormone Receptors alpha agonists, Thyroid Hormone Receptors alpha genetics, Thyroid Hormone Receptors alpha metabolism, Triiodothyronine metabolism, Iodothyronine Deiodinase Type II, Astrocytes metabolism, Cerebral Cortex metabolism, Gene Expression Regulation, Developmental, Nerve Tissue Proteins metabolism, Neuroblastoma metabolism, Neurons metabolism, Thyroxine metabolism
- Abstract
Background: The possibility that the intrinsic genomic activity of thyroxine (T4) is of physiological relevance has been frequently hypothesized. It might explain gene expression patterns in the brain found in type 2-deiodinase (Dio2)-deficient mice. These mice display normal expression of most thyroid hormone-dependent genes, despite decreased brain triiodothyronine (T3)., Methods: The relative effects of T4 and T3 on gene expression were analyzed in mouse neuro-2a (N2a) cells stably expressing the thyroid hormone receptor α1, and in primary mouse cerebrocortical cells enriched in astrocytes or in neurons. Cortical cells were derived from Dio2-deficient mice to prevent conversion of T4 to T3. T4 and T3 were measured in the media at the beginning and end of incubation, and T4 and T3 antibodies were used to block T4 and T3 action., Results: In all cell types, T4 had intrinsic genomic activity. In N2a cells, T4 activity was higher on negative regulation (1/5th of T3 activity) than on positive regulation (1/40th of T3 activity). T4 activity on positive regulation was dependent on the cell context, and was higher in primary cells than in N2a cells., Conclusion: T4 has intrinsic genomic activity. Positive regulation depends on the cell context, and primary cells appear much more sensitive than neuroblastoma cells. In all cells, negative regulation is more sensitive to T4 than positive regulation. These properties may explain the mostly normal gene expression in the brain of Dio2-deficient mice.
- Published
- 2017
- Full Text
- View/download PDF
29. Global Transcriptome Analysis of Primary Cerebrocortical Cells: Identification of Genes Regulated by Triiodothyronine in Specific Cell Types.
- Author
-
Gil-Ibañez P, García-García F, Dopazo J, Bernal J, and Morte B
- Subjects
- Animals, Astrocytes cytology, Astrocytes metabolism, Cells, Cultured, Fluorescent Antibody Technique, Gene Expression Profiling, Mice, 129 Strain, Mice, Inbred BALB C, Mice, Inbred C57BL, Neurons cytology, Neurons metabolism, Piperazines metabolism, Transcriptome, Cerebral Cortex cytology, Cerebral Cortex metabolism, Triiodothyronine metabolism
- Abstract
Thyroid hormones, thyroxine, and triiodothyronine (T3) are crucial for cerebral cortex development acting through regulation of gene expression. To define the transcriptional program under T3 regulation, we have performed RNA-Seq of T3-treated and untreated primary mouse cerebrocortical cells. The expression of 1145 genes or 7.7% of expressed genes was changed upon T3 addition, of which 371 responded to T3 in the presence of cycloheximide indicating direct transcriptional regulation. The results were compared with available transcriptomic datasets of defined cellular types. In this way, we could identify targets of T3 within genes enriched in astrocytes and neurons, in specific layers including the subplate, and in specific neurons such as prepronociceptin, cholecystokinin, or cortistatin neurons. The subplate and the prepronociceptin neurons appear as potentially major targets of T3 action. T3 upregulates mostly genes related to cell membrane events, such as G-protein signaling, neurotransmission, and ion transport and downregulates genes involved in nuclear events associated with the M phase of cell cycle, such as chromosome organization and segregation. Remarkably, the transcriptomic changes induced by T3 sustain the transition from fetal to adult patterns of gene expression. The results allow defining in molecular terms the elusive role of thyroid hormones on neocortical development., (© The Author 2015. Published by Oxford University Press. All rights reserved. For Permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2017
- Full Text
- View/download PDF
30. MCT8 Deficiency in Male Mice Mitigates the Phenotypic Abnormalities Associated With the Absence of a Functional Type 3 Deiodinase.
- Author
-
Stohn JP, Martinez ME, Matoin K, Morte B, Bernal J, Galton VA, St Germain D, and Hernandez A
- Subjects
- Animals, Animals, Newborn, Fetal Growth Retardation genetics, Hypothalamus physiology, Hypothyroidism genetics, Male, Mice, Mice, Knockout, Monocarboxylic Acid Transporters, Phenotype, Symporters, Thyroid Gland physiology, Fetal Viability genetics, Growth and Development genetics, Iodide Peroxidase genetics, Membrane Transport Proteins genetics
- Abstract
Mice deficient in the type 3 deiodinase (D3KO mice) manifest impaired clearance of thyroid hormone (TH), leading to elevated levels of TH action during development. This alteration causes reduced neonatal viability, growth retardation, and central hypothyroidism. Here we examined how these phenotypes are affected by a deficiency in the monocarboxylate transporter 8 (MCT8), which is a major contributor to the transport of the active thyroid hormone, T3, into the cell. MCT8 deficiency eliminated the neonatal lethality of type 3 deiodinase (D3)-deficient mice and significantly ameliorated their growth retardation. Double-mutant newborn mice exhibited similar peripheral thyrotoxicosis and increased brain expression of T3-dependent genes as mice with D3 deficiency only. Later in neonatal life and adulthood, double-mutant mice manifested central and peripheral TH status similar to mice with single MCT8 deficiency, with low serum T4, elevated serum TSH and T3, and decreased T3-dependent gene expression in the hypothalamus. In double-mutant adult mice, both thyroid gland size and the hypothyroidism-induced rise in TSH were greater than those in mice with single D3 deficiency but less than those in mice with MCT8 deficiency alone. Our results demonstrate that the marked phenotypic abnormalities observed in the D3-deficient mouse, including perinatal mortality, growth retardation, and central hypothyroidism in adult animals, require expression of MCT8, confirming the interdependent relationship between the TH transport into cells and the deiodination processes.
- Published
- 2016
- Full Text
- View/download PDF
31. Effect of Triiodothyroacetic Acid Treatment in Mct8 Deficiency: A Word of Caution.
- Author
-
Bárez-López S, Obregon MJ, Martínez-de-Mena R, Bernal J, Guadaño-Ferraz A, and Morte B
- Subjects
- Animals, Cerebral Cortex drug effects, Cerebral Cortex metabolism, Corpus Striatum drug effects, Corpus Striatum metabolism, Iodide Peroxidase metabolism, Liver drug effects, Liver metabolism, Membrane Transport Proteins metabolism, Mice, Mice, Knockout, Monocarboxylic Acid Transporters, Symporters, Triiodothyronine pharmacology, Membrane Transport Proteins genetics, Thyroxine blood, Triiodothyronine analogs & derivatives, Triiodothyronine blood
- Abstract
Background: Monocarboxylate transporter 8 (MCT8) is a thyroid hormone-specific cell membrane transporter. Mutations in the MCT8 gene lead to profound psychomotor retardation and abnormal thyroid hormone serum levels with low thyroxine (T4) and high triiodothyronine (T3). Currently, therapeutic options for patients are limited. Triiodothyroacetic acid (TRIAC) has potential therapeutic value. The aim of this study was to evaluate the effects and efficacy of therapeutic doses of TRIAC on Mct8-deficient mice (Mct8KO)., Methods: Wild-type (Wt) and Mct8KO mice were treated with 30 ng TRIAC/g of body weight/day, given in drinking water, from postnatal day 21 to 30. TRIAC, T4 and T3 levels in plasma, as well as T3 and TRIAC content in the cerebral cortex and striatum were measured by specific radioimmunoassays. The activities of deiodinases 1 and 2 were measured in liver and cortex. The effect of TRIAC treatment in the expression of T3-dependent genes was measured in the heart, cerebral cortex, and striatum., Results: Plasma TRIAC concentration were the same in Wt and Mct8KO animals after treatment. TRIAC treatment greatly decreased plasma T4 in Wt and Mct8KO mice, and reduced T3 to normal levels in the Mct8KO mice. Deiodinase 1 activity and gene expression in the liver increased, while it did not have any effect on the expression of Serca2a in the heart. TRIAC treatment did not induce the expression of T3-dependent genes in the cerebral cortex or striatum, but further decreased expression of Flywch2 in the cortex and Aldh1a1 and Flywch2 in the striatum. Direct measurements of TRIAC and T3 content in the cortex and striatum revealed a decrease in T3 after treatment with no significant increase in the level of endogenous TRIAC., Conclusions: Therapeutic doses of TRIAC in Mct8KO mice restored plasma T3 levels but severely decreased T4 levels. TRIAC has a direct effect on deiodinase 1 in the liver and does not have an effect on gene expression in the heart. The increase in the plasma TRIAC levels after treatment is not sufficient to increase TRIAC levels in the brain and to promote the expression of T3-dependent genes in brain cells. Instead, it leads to a state of brain hypothyroidism with reduced T3 content.
- Published
- 2016
- Full Text
- View/download PDF
32. Thyroid hormone transporters-functions and clinical implications.
- Author
-
Bernal J, Guadaño-Ferraz A, and Morte B
- Published
- 2015
- Full Text
- View/download PDF
33. Nuclear DICKKOPF-1 as a biomarker of chemoresistance and poor clinical outcome in colorectal cancer.
- Author
-
Aguilera Ó, González-Sancho JM, Zazo S, Rincón R, Fernández AF, Tapia O, Canals F, Morte B, Calvanese V, Orgaz JL, Niell N, Aguilar S, Freije JM, Graña O, Pisano DG, Borrero A, Martínez-Useros J, Jiménez B, Fraga MF, García-Foncillas J, López-Otín C, Lafarga M, Rojo F, and Muñoz A
- Subjects
- Aldehyde Dehydrogenase biosynthesis, Aldehyde Dehydrogenase genetics, Aldehyde Dehydrogenase 1 Family, Biomarkers, Tumor biosynthesis, Biomarkers, Tumor genetics, Calcium-Binding Proteins, Cell Line, Tumor, Cell Nucleus metabolism, Colorectal Neoplasms genetics, Colorectal Neoplasms pathology, Drug Resistance, Neoplasm, Female, Gene Expression Regulation, Neoplastic, Humans, Intercellular Signaling Peptides and Proteins genetics, Intestinal Mucosa metabolism, Intracellular Signaling Peptides and Proteins biosynthesis, Intracellular Signaling Peptides and Proteins genetics, Male, Prognosis, Retinal Dehydrogenase, Signal Transduction, Biomarkers, Tumor metabolism, Colorectal Neoplasms drug therapy, Colorectal Neoplasms metabolism, Intercellular Signaling Peptides and Proteins metabolism
- Abstract
Sporadic colorectal cancer (CRC) insurgence and progression depend on the activation of Wnt/β-catenin signaling. Dickkopf (DKK)-1 is an extracellular inhibitor of Wnt/β-catenin signaling that also has undefined β-catenin-independent actions. Here we report for the first time that a proportion of DKK-1 locates within the nucleus of healthy small intestine and colon mucosa, and of CRC cells at specific chromatin sites of active transcription. Moreover, we show that DKK-1 regulates several cancer-related genes including the cancer stem cell marker aldehyde dehydrogenase 1A1 (ALDH1A1) and Ral-binding protein 1-associated Eps domain-containing 2 (REPS2), which are involved in detoxification of chemotherapeutic agents. Nuclear DKK-1 expression is lost along CRC progression; however, it remains high in a subset (15%) of CRC patients (n = 699) and associates with decreased progression-free survival (PFS) after chemotherapy administration and overall survival (OS) [adjusted HR, 1.65; 95% confidence interval (CI), 1.23-2.21; P = 0.002)]. Overexpression of ALDH1A1 and REPS2 associates with nuclear DKK-1 expression in tumors and correlates with decreased OS (P = 0.001 and 0.014) and PFS. In summary, our findings demonstrate a novel location of DKK-1 within the cell nucleus and support a role of nuclear DKK-1 as a predictive biomarker of chemoresistance in colorectal cancer.
- Published
- 2015
- Full Text
- View/download PDF
34. Hypotonic male infant and MCT8 deficiency - a diagnosis to think about.
- Author
-
Rodrigues F, Grenha J, Ortez C, Nascimento A, Morte B, M-Belinchón M, Armstrong J, and Colomer J
- Subjects
- Child, Preschool, Humans, Male, Mental Retardation, X-Linked genetics, Muscle Hypotonia genetics, Muscular Atrophy genetics, Rare Diseases, Symporters, Thyroxine blood, Triiodothyronine blood, Mental Retardation, X-Linked diagnosis, Monocarboxylic Acid Transporters genetics, Muscle Hypotonia diagnosis, Muscular Atrophy diagnosis, Mutation
- Abstract
Background: Thyroid hormone is crucial in the development of different organs, particularly the brain. MCT8 is a specific transporter of triiodothyronine (T3) hormone and MCT8 gene mutations cause a rare X-linked disorder named MCT8 deficiency, also known as Allan-Herndon-Dudley syndrome, characterized by psychomotor retardation and hypotonia. Typically, elevation of T3 and delayed myelination in cerebral magnetic resonance imaging are found., Case Presentation: We present a 24-month-old boy, born from non-consanguineous healthy parents, with severe motor and cognitive delay and global hypotonia, being unable to hold head upright or sit without support. Deep tendon reflexes were absent bilaterally at the ankles. T3 was elevated and thyroxine slightly decreased, consistent with MCT8 deficiency. Genetic studies confirmed the diagnosis., Conclusions: Although a rare disease (MCT8 mutations have been reported in about 50 families all around the world), we illustrate the importance of excluding Allan-Herndon-Dudley syndrome in the evaluation of floppy male infants with development delay, without history of perinatal asphyxia. The simple evaluation of thyroid status, including T3, T4 and TSH can guide the diagnosis, avoiding a number of useless, expensive and invasive investigations and allowing appropriate genetic counseling to the affected families.
- Published
- 2014
- Full Text
- View/download PDF
35. Thyroid hormone action: astrocyte-neuron communication.
- Author
-
Morte B and Bernal J
- Abstract
Thyroid hormone (TH) action is exerted mainly through regulation of gene expression by binding of T3 to the nuclear receptors. T4 plays an important role as a source of intracellular T3 in the central nervous system via the action of the type 2 deiodinase (D2), expressed in the astrocytes. A model of T3 availability to neural cells has been proposed and validated. The model contemplates that brain T3 has a double origin: a fraction is available directly from the circulation, and another is produced locally from T4 in the astrocytes by D2. The fetal brain depends almost entirely on the T3 generated locally. The contribution of systemic T3 increases subsequently during development to account for approximately 50% of total brain T3 in the late postnatal and adult stages. In this article, we review the experimental data in support of this model, and how the factors affecting T3 availability in the brain, such as deiodinases and transporters, play a decisive role in modulating local TH action during development.
- Published
- 2014
- Full Text
- View/download PDF
36. Cerebral cortex hyperthyroidism of newborn mct8-deficient mice transiently suppressed by lat2 inactivation.
- Author
-
Núñez B, Martínez de Mena R, Obregon MJ, Font-Llitjós M, Nunes V, Palacín M, Dumitrescu AM, Morte B, and Bernal J
- Subjects
- Amino Acid Transport System y+ genetics, Animals, Animals, Newborn, Female, Fusion Regulatory Protein 1, Light Chains genetics, Hyperthyroidism genetics, Male, Membrane Transport Proteins genetics, Mice, Monocarboxylic Acid Transporters, Symporters, Triiodothyronine metabolism, Amino Acid Transport System y+ metabolism, Cerebral Cortex metabolism, Fusion Regulatory Protein 1, Light Chains metabolism, Hyperthyroidism metabolism, Membrane Transport Proteins metabolism
- Abstract
Thyroid hormone entry into cells is facilitated by transmembrane transporters. Mutations of the specific thyroid hormone transporter, MCT8 (Monocarboxylate Transporter 8, SLC16A2) cause an X-linked syndrome of profound neurological impairment and altered thyroid function known as the Allan-Herndon-Dudley syndrome. MCT8 deficiency presumably results in failure of thyroid hormone to reach the neural target cells in adequate amounts to sustain normal brain development. However during the perinatal period the absence of Mct8 in mice induces a state of cerebral cortex hyperthyroidism, indicating increased brain access and/or retention of thyroid hormone. The contribution of other transporters to thyroid hormone metabolism and action, especially in the context of MCT8 deficiency is not clear. We have analyzed the role of the heterodimeric aminoacid transporter Lat2 (Slc7a8), in the presence or absence of Mct8, on thyroid hormone concentrations and on expression of thyroid hormone-dependent cerebral cortex genes. To this end we generated Lat2-/-, and Mct8-/yLat2-/- mice, to compare with wild type and Mct8-/y mice during postnatal development. As described previously the single Mct8 KO neonates had a transient increase of 3,5,3'-triiodothyronine concentration and expression of thyroid hormone target genes in the cerebral cortex. Strikingly the absence of Lat2 in the double Mct8Lat2 KO prevented the effect of Mct8 inactivation in newborns. The Lat2 effect was not observed from postnatal day 5 onwards. On postnatal day 21 the Mct8 KO displayed the typical pattern of thyroid hormone concentrations in plasma, decreased cortex 3,5,3'-triiodothyronine concentration and Hr expression, and concomitant Lat2 inactivation produced little to no modifications. As Lat2 is expressed in neurons and in the choroid plexus, the results support a role for Lat2 in the supply of thyroid hormone to the cerebral cortex during early postnatal development.
- Published
- 2014
- Full Text
- View/download PDF
37. Thyroid hormone regulation of gene expression in primary cerebrocortical cells: role of thyroid hormone receptor subtypes and interactions with retinoic acid and glucocorticoids.
- Author
-
Gil-Ibáñez P, Bernal J, and Morte B
- Subjects
- Animals, Cells, Cultured, Cerebral Cortex drug effects, Glucocorticoids pharmacology, Mice, Mice, Knockout, Models, Biological, Receptors, Thyroid Hormone genetics, Receptors, Thyroid Hormone metabolism, Thyroid Hormone Receptors alpha genetics, Thyroid Hormone Receptors alpha metabolism, Thyroid Hormone Receptors beta genetics, Thyroid Hormone Receptors beta metabolism, Thyroid Hormones pharmacology, Tretinoin pharmacology, Cerebral Cortex cytology, Cerebral Cortex metabolism, Gene Expression Regulation drug effects, Thyroid Hormones metabolism
- Abstract
The effects of thyroid hormone on brain development and function are largely mediated by the binding of 3,5,3'-triiodo-L-thyronine (T3) to its nuclear receptors (TR) to regulate positively or negatively gene expression. We have analyzed by quantitative polymerase chain reaction the effect of T3 on primary cultured cells from the embryonic mouse cerebral cortex, on the expression of Hr, Klf9, Shh, Dio3, Aldh1a1, and Aldh1a3. In particular we focused on T3 receptor specificity, and on the crosstalk between T3, retinoic acid and dexamethasone. To check for receptor subtype specificity we used cerebrocortical cells derived from wild type mice and from mice deficient in thyroid hormone receptor subtypes. Receptor subtype specificity was found for Dio3 and Aldh1a1, which were induced by T3 only in cells expressing the T3 receptor alpha 1 subtype. Interactions of T3 with retinoic acid signaling through the control of retinoic acid metabolism are likely to be important during development. T3 had opposing influences on retinoic acid synthesizing enzymes, increasing the expression of Aldh1a1, and decreasing Aldh1a3, while increasing the retinoic acid degrading enzyme Cyp26b1. Dexamethasone increased Klf9 and Aldh1a1 expression. The effects of T3 and dexamethasone on Aldh1a1 were highly synergistic, with mRNA increments of up to 20 fold. The results provide new data on thyroid hormone regulation of gene expression and underscore the importance of thyroid hormone interactions with retinoic acid and glucocorticoids during neural development.
- Published
- 2014
- Full Text
- View/download PDF
38. Increased oxidative metabolism and neurotransmitter cycling in the brain of mice lacking the thyroid hormone transporter SLC16A2 (MCT8).
- Author
-
Rodrigues TB, Ceballos A, Grijota-Martínez C, Nuñez B, Refetoff S, Cerdán S, Morte B, and Bernal J
- Subjects
- Animals, Enzyme Activation, Gene Knockout Techniques, Hyperthyroidism metabolism, Iodide Peroxidase metabolism, Male, Mice, Monocarboxylic Acid Transporters, Oxidation-Reduction, Symporters, Thyroid Hormones metabolism, Iodothyronine Deiodinase Type II, Brain metabolism, Membrane Transport Proteins deficiency, Membrane Transport Proteins genetics, Neurotransmitter Agents metabolism
- Abstract
Mutations of the monocarboxylate transporter 8 (MCT8) cause a severe X-linked intellectual deficit and neurological impairment. MCT8 is a specific thyroid hormone (T4 and T3) transporter and the patients also present unusual abnormalities in the serum profile of thyroid hormone concentrations due to altered secretion and metabolism of T4 and T3. Given the role of thyroid hormones in brain development, it is thought that the neurological impairment is due to restricted transport of thyroid hormones to the target neurons. In this work we have investigated cerebral metabolism in mice with Mct8 deficiency. Adult male mice were infused for 30 minutes with (1-(13)C) glucose and brain extracts prepared and analyzed by (13)C nuclear magnetic resonance spectroscopy. Genetic inactivation of Mct8 resulted in increased oxidative metabolism as reflected by increased glutamate C4 enrichment, and of glutamatergic and GABAergic neurotransmissions as observed by the increases in glutamine C4 and GABA C2 enrichments, respectively. These changes were distinct to those produced by hypothyroidism or hyperthyroidism. Similar increments in glutamate C4 enrichment and GABAergic neurotransmission were observed in the combined inactivation of Mct8 and D2, indicating that the increased neurotransmission and metabolic activity were not due to increased production of cerebral T3 by the D2-encoded type 2 deiodinase. In conclusion, Mct8 deficiency has important metabolic consequences in the brain that could not be correlated with deficiency or excess of thyroid hormone supply to the brain during adulthood.
- Published
- 2013
- Full Text
- View/download PDF
39. Thyroid hormone receptor activity in the absence of ligand: physiological and developmental implications.
- Author
-
Bernal J and Morte B
- Subjects
- Amphibians genetics, Amphibians metabolism, Animals, Gene Expression Regulation, Developmental, Humans, Ligands, Metamorphosis, Biological, Signal Transduction, Thyroid Gland growth & development, Thyroid Gland metabolism, Thyroid Gland physiology, Transcription, Genetic, Amphibians growth & development, Receptors, Thyroid Hormone genetics, Receptors, Thyroid Hormone metabolism, Thyroid Hormones genetics, Thyroid Hormones metabolism
- Abstract
Background: The transcriptional activity of the thyroid hormone receptors is modulated by the ligand, T3, but they have also activity as aporeceptors, in the unliganded state. Aporeceptor activity is thought to contribute to the severity of profound hypothyroidism. During development thyroid hormone receptors are expressed before onset of thyroid gland function and are present therefore in many tissues mainly as aporeceptors. The question we address is whether thyroid hormone aporeceptors are involved in physiological and/or developmental processes., Scope of Review: The scope of this article is to review the evidence for a role of thyroid hormone aporeceptors in physiology and development. Related to this topic is the activity of mutant receptors unable to bind hormone. These receptors usually have dominant negative activity. This review focuses on the wild type receptors, and does not discuss the properties of mutant receptors., Major Conclusions: Unliganded thyroid hormone receptors influence the timing and control certain aspects of amphibian pre-metamorphosis. In mammals they are likely to influence maturational processes in the brain and other organs before onset of thyroid gland function. Expression of types 2 and 3 deiodinases which control the local tissue concentration of T3 regulates the fractional receptor occupancy and therefore the relative proportion of aporeceptors. This article is part of a Special Issue entitled Thyroid hormone signalling., (Copyright © 2012 Elsevier B.V. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
40. Role of thyroid hormone receptor subtypes α and β on gene expression in the cerebral cortex and striatum of postnatal mice.
- Author
-
Gil-Ibañez P, Morte B, and Bernal J
- Subjects
- Animals, Animals, Newborn, Cerebral Cortex growth & development, Corpus Striatum growth & development, Female, Gene Expression Profiling, Male, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Transgenic, Microarray Analysis, Protein Isoforms genetics, Protein Isoforms metabolism, Protein Isoforms physiology, Thyroid Hormone Receptors alpha genetics, Thyroid Hormone Receptors alpha metabolism, Thyroid Hormone Receptors beta genetics, Thyroid Hormone Receptors beta metabolism, Cerebral Cortex metabolism, Corpus Striatum metabolism, Gene Expression genetics, Thyroid Hormone Receptors alpha physiology, Thyroid Hormone Receptors beta physiology
- Abstract
The effects of thyroid hormones (THs) on brain development and function are largely mediated by the control of gene expression. This is achieved by the binding of the genomically active T3 to transcriptionally active nuclear TH receptors (TRs). T3 and the TRs can either induce or repress transcription. In hypothyroidism, the reduction of T3 lowers the expression of a set of genes, the positively regulated genes, and increases the expression of negatively regulated genes. Two mechanisms may account for the effect of hypothyroidism on genes regulated directly by T3: first, the loss of T3 signaling and TR transactivation, and second, an intrinsic activity of the unliganded TRs directly responsible for repression of positive genes and enhancement of negative genes. To analyze the contribution of the TR subtypes α and β, we have measured by RT-PCR the expression of a set of positive and negative genes in the cerebral cortex and the striatum of TR-knockout male and female mice. The results indicate that TRα1 exerts a predominant but not exclusive role in the regulation of positive and negative genes. However, a fraction of the genes analyzed are not or only mildly affected by the total absence of TRs. Furthermore, hypothyroidism has a mild effect on these genes in the absence of TRα1, in agreement with a role of unliganded TRα1 in the effects of hypothyroidism.
- Published
- 2013
- Full Text
- View/download PDF
41. Src kinases catalytic activity regulates proliferation, migration and invasiveness of MDA-MB-231 breast cancer cells.
- Author
-
Sánchez-Bailón MP, Calcabrini A, Gómez-Domínguez D, Morte B, Martín-Forero E, Gómez-López G, Molinari A, Wagner KU, and Martín-Pérez J
- Subjects
- Breast Neoplasms drug therapy, Cell Line, Tumor, Cell Movement drug effects, Cell Proliferation drug effects, Dasatinib, Female, Humans, Indoles, Neoplasm Invasiveness prevention & control, Sulfonamides, src-Family Kinases antagonists & inhibitors, Breast Neoplasms enzymology, Breast Neoplasms pathology, Paxillin pharmacology, Protein Kinase Inhibitors pharmacology, Pyrimidines pharmacology, Thiazoles pharmacology, src-Family Kinases metabolism
- Abstract
SFKs are frequently deregulated in cancer where they control cellular proliferation, migration, survival and metastasis. Here we study the role of SFKs catalytic activity in triple-negative/basal-like and metastatic human breast cancer MDA-MB-231 cells employing three well-established inhibitors: Dasatinib, PP2 and SU6656. These compounds inhibited migration and invasion. Concomitantly, they reduced Fak, paxillin, p130CAS, caveolin-1 phosphorylation and altered cytoskeletal structures. They also inhibited cell proliferation, but in different manners. Dasatinib and PP2 increased p27(Kip1) expression and reduced c-Myc levels, restraining G1–S transition. In contrast, SU6656 did not modify p27(Kip1) expression, slightly altered c-Myc levels and generated polyploid multinucleated cells, indicating inhibition of cytokinesis. These later effects were also observed in SYF fibroblasts, suggesting a SFKs-independent action. ZM447439, an Aurora B kinase inhibitor, produced similar cell cycle and morphological alterations in MDA-MB-231 cells, indicating that SU6656 blocked Aurora B kinase. This was confirmed by inhibition of histone H3 phosphorylation, the canonical Aurora B kinase substrate. Furthermore, hierarchical clustering analysis of gene expression profiles showed that SU6656 defined a set of genes that differed from Dasatinib and PP2. Additionally, Gene Set Enrichment Analyses revealed that SU6656 significantly reduces the Src pathway. Together, these results show the importance of SFKs catalytic activity for MDA-MB-231 proliferation, migration and invasiveness. They also illustrate that SU6656 acts as dual SFKs and Aurora B kinase inhibitor, suggesting its possible use as a therapeutic agent in breast cancer.
- Published
- 2012
- Full Text
- View/download PDF
42. Critical role of types 2 and 3 deiodinases in the negative regulation of gene expression by T₃in the mouse cerebral cortex.
- Author
-
Hernandez A, Morte B, Belinchón MM, Ceballos A, and Bernal J
- Subjects
- Animals, Cell Line, Tumor, Cerebral Cortex metabolism, Female, Gene Expression Profiling, Hyperthyroidism genetics, Hypothyroidism genetics, Iodide Peroxidase genetics, Iodide Peroxidase metabolism, Male, Mice, Mice, 129 Strain, Mice, Inbred C57BL, Mice, Knockout, Oligonucleotide Array Sequence Analysis, Reverse Transcriptase Polymerase Chain Reaction, Thyroxine blood, Triiodothyronine blood, Cerebral Cortex drug effects, Gene Expression Regulation genetics, Iodide Peroxidase deficiency, Triiodothyronine pharmacology
- Abstract
Thyroid hormones regulate brain development and function through the control of gene expression, mediated by binding of T(3) to nuclear receptors. Brain T(3) concentration is tightly controlled by homeostatic mechanisms regulating transport and metabolism of T(4) and T(3). We have examined the role of the inactivating enzyme type 3 deiodinase (D3) in the regulation of 43 thyroid hormone-dependent genes in the cerebral cortex of 30-d-old mice. D3 inactivation increased slightly the expression of two of 22 positively regulated genes and significantly decreased the expression of seven of 21 negatively regulated genes. Administration of high doses of T(3) led to significant changes in the expression of 12 positive genes and three negative genes in wild-type mice. The response to T(3) treatment was enhanced in D3-deficient mice, both in the number of genes and in the amplitude of the response, demonstrating the role of D3 in modulating T(3) action. Comparison of the effects on gene expression observed in D3 deficiency with those in hypothyroidism, hyperthyroidism, and type 2 deiodinase (D2) deficiency revealed that the negative genes are more sensitive to D2 and D3 deficiencies than the positive genes. This observation indicates that, in normal physiological conditions, D2 and D3 play critical roles in maintaining local T(3) concentrations within a very narrow range. It also suggests that negatively and positively regulated genes do not have the same physiological significance or that their regulation by thyroid hormone obeys different paradigms at the molecular or cellular levels.
- Published
- 2012
- Full Text
- View/download PDF
43. Monocyte-mediated regulation of genes by the amyloid and prion peptides in SH-SY5Y neuroblastoma cells.
- Author
-
Morte B, Martínez T, Zambrano A, and Pascual A
- Subjects
- Amyloid beta-Peptides chemistry, Cell Line, Cell Line, Tumor, Culture Media, Conditioned, Humans, Neuroblastoma genetics, Prions chemistry, RNA Interference, Reverse Transcriptase Polymerase Chain Reaction, Amyloid beta-Peptides physiology, Gene Expression Profiling, Monocytes cytology, Neuroblastoma pathology, Peptide Fragments physiology, Prions physiology
- Abstract
Alzheimer's disease as well as prion-related encephalopathies are neurodegenerative disorders of the central nervous system, which cause mental deterioration and progressive dementia. Both pathologies appear to be primarily associated with the pathological accumulation and deposit of β-amyloid or prion peptides in the brain, and it has been even suggested that neurotoxicity induced by these peptides would be associated to essentially similar pathogenic mechanisms, in particular to those that follow the activation of microglial cells. To probe whether the neurotoxic effects induced by the β-amyloid and prion peptides are actually mediated by similar glial-associated mechanisms, we have examined the differential expression of genes in SH-SY5Y neuroblastoma cells incubated with conditioned media from β-amyloid or prion-stimulated THP-1 monocytic cells. According to microarray analysis, not many coincidences are observed and only four genes (Hint3, Psph, Daam1 and c-Jun) appear to be commonly upregulated by both peptides. Furthermore, c-Jun appears to be involved in the cell death mediated by both peptides., (Copyright © 2011 Elsevier Ltd. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
44. Lack of action of exogenously administered T3 on the fetal rat brain despite expression of the monocarboxylate transporter 8.
- Author
-
Grijota-Martínez C, Díez D, Morreale de Escobar G, Bernal J, and Morte B
- Subjects
- Amino Acid Transport Systems, Basic metabolism, Animals, Brain embryology, Calcium-Calmodulin-Dependent Protein Kinase Type 4 metabolism, Female, Fluorescent Antibody Technique, Intracellular Signaling Peptides and Proteins metabolism, Microscopy, Confocal, Monocarboxylic Acid Transporters genetics, Nerve Tissue Proteins metabolism, Organic Anion Transporters metabolism, Organic Cation Transport Proteins, Rats, Rats, Wistar, Thyroxine pharmacology, Brain drug effects, Brain metabolism, Monocarboxylic Acid Transporters metabolism, Triiodothyronine pharmacology
- Abstract
Mutations of the monocarboxylate transporter 8 gene (MCT8, SLC16A2) cause the Allan-Herndon-Dudley syndrome, an X-linked syndrome of severe intellectual deficit and neurological impairment. Mct8 transports thyroid hormones (T4 and T3), and the Allan-Herndon-Dudley syndrome is likely caused by lack of T3 transport to neurons during critical periods of fetal brain development. To evaluate the role of Mct8 in thyroid hormone action in the fetal brain we administered T4 or T3 to thyroidectomized pregnant dams treated with methyl-mercapto-imidazol to produce maternal and fetal hypothyroidism. Gene expression was then measured in the fetal cerebral cortex. T4 increased Camk4, Sema3c, and Slc7a3 expression, but T3 was without effect. To investigate the cause for the lack of T3 action we analyzed the expression of organic anion transport polypeptide (Oatp14, Slco1c1), a T4 transporter, and Mct8 (Slc16a2), a T4 and T3 transporter, by confocal microscopy. Both proteins were present in the brain capillaries forming the blood-brain barrier and in the epithelial cells of the choroid plexus forming the blood-cerebrospinal fluid barrier. It is concluded that T4 from the maternal compartment influences gene expression in the fetal cerebral cortex, possibly after transport via organic anion transporter polypeptide and/or Mct8, and conversion to T3 in the astrocytes. On the other hand, T3 does not reach the target neurons despite the presence of Mct8. The data indicate that T4, through local deiodination, provides most T3 in the fetal rat brain. The role of Mct8 as a T3 transporter in the fetal rat brain is therefore uncertain.
- Published
- 2011
- Full Text
- View/download PDF
45. In vivo activity of the thyroid hormone receptor beta- and α-selective agonists GC-24 and CO23 on rat liver, heart, and brain.
- Author
-
Grijota-Martínez C, Samarut E, Scanlan TS, Morte B, and Bernal J
- Subjects
- Animals, Brain drug effects, Gene Expression Regulation drug effects, Heart drug effects, Hypothyroidism drug therapy, Liver drug effects, Rats, Rats, Wistar, Triiodothyronine, Acetates pharmacology, Benzhydryl Compounds pharmacology, Hydantoins pharmacology, Thyroid Hormone Receptors alpha agonists, Thyroid Hormone Receptors beta agonists
- Abstract
Thyroid hormone analogs with selective actions through specific thyroid hormone receptor (TR) subtypes are of great interest. They might offer the possibility of mimicking physiological actions of thyroid hormone with receptor subtype or tissue specificity with therapeutic aims. They are also pharmacological tools to dissect biochemical pathways mediated by specific receptor subtypes, in a complementary way to mouse genetic modifications. In this work, we studied the in vivo activity in developing rats of two thyroid hormone agonists, the TRβ-selective GC-24 and the TRα-selective CO23. Our principal goal was to check whether these compounds were active in the rat brain. Analog activity was assessed by measuring the expression of thyroid hormone target genes in liver, heart, and brain, after administration to hypothyroid rats. GC-24 was very selective for TRβ and lacked activity on the brain. On the other hand, CO23 was active in liver, heart, and brain on genes regulated by either TRα or TRβ. This compound, previously shown to be TRα-selective in tadpoles, displayed no selectivity in the rat in vivo.
- Published
- 2011
- Full Text
- View/download PDF
46. Thyroid hormone-regulated mouse cerebral cortex genes are differentially dependent on the source of the hormone: a study in monocarboxylate transporter-8- and deiodinase-2-deficient mice.
- Author
-
Morte B, Ceballos A, Diez D, Grijota-Martínez C, Dumitrescu AM, Di Cosmo C, Galton VA, Refetoff S, and Bernal J
- Subjects
- Animals, Animals, Newborn, Antithyroid Agents administration & dosage, Cerebral Cortex embryology, Cerebral Cortex growth & development, Female, Gene Expression Profiling, Gene Expression Regulation, Developmental drug effects, Hypothyroidism genetics, Iodide Peroxidase genetics, Iodide Peroxidase metabolism, Male, Membrane Transport Proteins genetics, Membrane Transport Proteins metabolism, Methimazole administration & dosage, Mice, Mice, Inbred C57BL, Mice, Knockout, Monocarboxylic Acid Transporters, Oligonucleotide Array Sequence Analysis, Pregnancy, Reverse Transcriptase Polymerase Chain Reaction, Symporters, Thyroxine metabolism, Triiodothyronine metabolism, Iodothyronine Deiodinase Type II, Cerebral Cortex metabolism, Iodide Peroxidase deficiency, Membrane Transport Proteins deficiency, Thyroid Hormones metabolism
- Abstract
Thyroid hormones influence brain development through the control of gene expression. The concentration of the active hormone T(3) in the brain depends on T(3) transport through the blood-brain barrier, mediated in part by the monocarboxylate transporter 8 (Mct8/MCT8) and the activity of type 2 deiodinase (D2) generating T(3) from T(4). The relative roles of each of these pathways in the regulation of brain gene expression is not known. To shed light on this question, we analyzed thyroid hormone-dependent gene expression in the cerebral cortex of mice with inactivated Mct8 (Slc16a2) and Dio2 genes, alone or in combination. We used 34 target genes identified to be controlled by thyroid hormone in microarray comparisons of cerebral cortex from wild-type control and hypothyroid mice on postnatal d 21. Inactivation of the Mct8 gene (Mct8KO) was without effect on the expression of 31 of these genes. Normal gene expression in the absence of the transporter was mostly due to D2 activity because the combined disruption of Mct8 and Dio2 led to similar effects as hypothyroidism on the expression of 24 genes. Dio2 disruption alone did not affect the expression of positively regulated genes, but, as in hypothyroidism, it increased that of negatively regulated genes. We conclude that gene expression in the Mct8KO cerebral cortex is compensated in part by D2-dependent mechanisms. Intriguingly, positive or negative regulation of genes by thyroid hormone is sensitive to the source of T(3) because Dio2 inactivation selectively affects the expression of negatively regulated genes.
- Published
- 2010
- Full Text
- View/download PDF
47. Thyroid hormone regulation of gene expression in the developing rat fetal cerebral cortex: prominent role of the Ca2+/calmodulin-dependent protein kinase IV pathway.
- Author
-
Morte B, Díez D, Ausó E, Belinchón MM, Gil-Ibáñez P, Grijota-Martínez C, Navarro D, de Escobar GM, Berbel P, and Bernal J
- Subjects
- Animals, Cells, Cultured, Cerebral Cortex drug effects, Cerebral Cortex enzymology, DNA Primers, Female, Humans, Hypothyroidism embryology, Imidazoles pharmacology, Neurons cytology, Neurons drug effects, Neurons enzymology, Nucleic Acid Hybridization, Oligonucleotide Array Sequence Analysis, Polymerase Chain Reaction, RNA genetics, RNA isolation & purification, Rats, Rats, Wistar, Sex-Determining Region Y Protein genetics, Thyroidectomy, Thyrotropin blood, Triiodothyronine pharmacology, Calcium-Calmodulin-Dependent Protein Kinase Type 4 genetics, Cerebral Cortex embryology, Gene Expression Regulation drug effects, Hypothyroidism genetics
- Abstract
Thyroid hormones influence brain development through regulation of gene expression mediated by nuclear receptors. Nuclear receptor concentration increases rapidly in the human fetus during the second trimester, a period of high sensitivity of the brain to thyroid hormones. In the rat, the equivalent period is the last quarter of pregnancy. However, little is known about thyroid hormone action in the fetal brain, and in rodents, most thyroid hormone-regulated genes have been identified during the postnatal period. To identify potential targets of thyroid hormone in the fetal brain, we induced maternal and fetal hypothyroidism by maternal thyroidectomy followed by antithyroid drug (2-mercapto-1-methylimidazole) treatment. Microarray analysis identified differentially expressed genes in the cerebral cortex of hypothyroid fetuses on d 21 after conception. Gene function analysis revealed genes involved in the biogenesis of the cytoskeleton, neuronal migration and growth, and branching of neurites. Twenty percent of the differentially expressed genes were related to each other centered on the Ca(2+) and calmodulin-activated kinase (Camk4) pathway. Camk4 was regulated directly by T(3) in primary cultured neurons from fetal cortex, and the Camk4 protein was also induced by thyroid hormone. No differentially expressed genes were recovered when euthyroid fetuses from hypothyroid mothers were compared with fetuses from normal mothers. Although the results do not rule out a specific contribution from the mother, especially at earlier stages of pregnancy, they indicate that the main regulators of thyroid hormone-dependent, fetal brain gene expression near term are the fetal thyroid hormones.
- Published
- 2010
- Full Text
- View/download PDF
48. Importance of monocarboxylate transporter 8 for the blood-brain barrier-dependent availability of 3,5,3'-triiodo-L-thyronine.
- Author
-
Ceballos A, Belinchon MM, Sanchez-Mendoza E, Grijota-Martinez C, Dumitrescu AM, Refetoff S, Morte B, and Bernal J
- Subjects
- Animals, Animals, Newborn, Biological Transport genetics, Cells, Cultured, Female, Male, Membrane Transport Proteins genetics, Membrane Transport Proteins metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Monocarboxylic Acid Transporters, Neurogranin metabolism, Neurons metabolism, Symporters, Blood-Brain Barrier metabolism, Membrane Transport Proteins physiology, Triiodothyronine metabolism
- Abstract
Mutations of the gene expressing plasma membrane transporter for thyroid hormones MCT8 (SLC16A2) in humans lead to altered thyroid hormone levels and a severe neurodevelopmental disorder. Genetically engineered defect of the Mct8 gene in mice leads to similar thyroid hormone abnormalities but no obvious impairment of brain development or function. In this work we studied the relative role of the blood-brain barrier and the neuronal plasma cell membrane in the restricted access of T(3) to the target neurons. To this end we compared the effects of low doses of T(4) and T(3) on cerebellar structure and gene expression in wild-type (Wt) and Mct8 null male mice [Mct8-/y, knockout (KO)] made hypothyroid during the neonatal period. We found that compared with Wt animals, T(4) was considerably more potent than T(3) in the Mct8KO mice, indicating a restricted access of T(3), but not T(4), to neurons after systemic administration in vivo. In contrast, T(3) action in cultured cerebellar neurons was similar in Wt cells as in Mct8KO cells. The results suggest that the main restriction for T(3) entry into the neural target cells of the mouse deficient in Mct8 is at the blood-brain barrier.
- Published
- 2009
- Full Text
- View/download PDF
49. Thyroid hormone action in the adult brain: gene expression profiling of the effects of single and multiple doses of triiodo-L-thyronine in the rat striatum.
- Author
-
Diez D, Grijota-Martinez C, Agretti P, De Marco G, Tonacchera M, Pinchera A, de Escobar GM, Bernal J, and Morte B
- Subjects
- Age Factors, Animals, Brain metabolism, Cluster Analysis, Corpus Striatum metabolism, Dose-Response Relationship, Drug, Drug Administration Schedule, Gene Expression Regulation drug effects, Male, Models, Biological, Rats, Rats, Wistar, Triiodothyronine administration & dosage, Brain drug effects, Corpus Striatum drug effects, Gene Expression Profiling, Microarray Analysis, Triiodothyronine pharmacology
- Abstract
Thyroid hormones have profound effects on mood and behavior, but the molecular basis of thyroid hormone action in the adult brain is relatively unknown. In particular, few thyroid hormone-dependent genes have been identified in the adult brain despite extensive work carried out on the developing brain. In this work we performed global analysis of gene expression in the adult rat striatum in search for genomic changes taking place after administration of T(3) to hypothyroid rats. The hormone was administered in two different schedules: 1) a single, large dose of 25 microg per 100 g body weight (SD) or 2) 1.5 microg per 100 g body weight once daily for 5 d (RD). Twenty-four hours after the single or last of multiple doses, gene expression in the striatum was analyzed using Codelink microarrays. SD caused up-regulation of 149 genes and down-regulation of 88 genes. RD caused up-regulation of 18 genes and down-regulation of one gene. The results were confirmed by hybridization to Affymetrix microarrays and by TaqMan PCR. Among the genes identified are genes involved in circadian regulation and the regulation of signaling pathways in the striatum. These results suggest that thyroid hormone is involved in regulation of striatal physiology at multiple control points. In addition, they may explain the beneficial effects of large doses of thyroid hormone in bipolar disorders.
- Published
- 2008
- Full Text
- View/download PDF
50. Influence of thyroid hormone and thyroid hormone receptors in the generation of cerebellar gamma-aminobutyric acid-ergic interneurons from precursor cells.
- Author
-
Manzano J, Cuadrado M, Morte B, and Bernal J
- Subjects
- Acetates pharmacology, Age Factors, Animals, Blotting, Western, Cell Differentiation, Cell Proliferation drug effects, Cerebellum cytology, GABA Plasma Membrane Transport Proteins metabolism, GABA Plasma Membrane Transport Proteins physiology, Hypothyroidism metabolism, Immunohistochemistry, Interneurons cytology, Interneurons metabolism, Ki-67 Antigen metabolism, Mice, Mice, Inbred BALB C, PAX2 Transcription Factor metabolism, Phenols pharmacology, Rats, Rats, Wistar, Receptors, Thyroid Hormone physiology, Interneurons drug effects, Receptors, Thyroid Hormone antagonists & inhibitors, Thyroid Hormones pharmacology, gamma-Aminobutyric Acid metabolism
- Abstract
Thyroid hormones have important actions in the developing central nervous system. We describe here a novel action of thyroid hormone and its nuclear receptors on maturation of cerebellar gamma-aminobutyric acid (GABA)-ergic interneurons from their precursor cells. In rats, the density of GABAergic terminals in the cerebellum was decreased by hypothyroidism, as shown by immunohistochemistry for the GABA transporter GAT-1. This was due, at least partially, to a decreased number of GABAergic cells, because the number of Golgi II cells in the internal granular layer was decreased. GABAergic interneurons in the cerebellum differentiate from precursors expressing the Pax-2 transcription factor, generated in the subventricular zone of the embryonic fourth ventricle from where they migrate to the cerebellum. Hypothyroidism caused both decreased proliferation and delayed differentiation of precursors, with the net effect being an accumulation of immature cells during the neonatal period. The contribution of thyroid hormone receptors was studied by treating hypothyroid rats with T(3) or with the thyroid hormone receptor (TR) beta-selective agonist GC-1. Whereas treatment with T(3) reduced the number of precursors to control levels, GC-1 had only a partial effect, indicating that both TRalpha1 and TRbeta mediate the actions of T(3). Deletion of TRalpha1 in mice decreased cerebellar GAT-1 expression and Pax-2 precursor cell proliferation. It is concluded that thyroid hormone, acting through the nuclear receptors, has a major role in the proliferation and further differentiation of the Pax-2 precursors of cerebellar GABAergic cells.
- Published
- 2007
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.