28 results on '"Gomez-Skarmeta JL"'
Search Results
2. Conservative route to genome compaction in a miniature annelid (November, 10.1038/s41559-020-01327-6, 2020)
- Author
-
Martin-Duran, JM, Vellutini, BC, Marletaz, F, Cetrangolo, V, Cvetesic, N, Thiel, D, Henriet, S, Grau-Bove, X, Carrillo-Baltodano, AM, Gu, W, Kerbl, A, Marquez, Y, Bekkouche, N, Chourrout, D, Gomez-Skarmeta, JL, Irimia, M, Lenhard, B, Worsaae, K, Hejnol, A, and Wellcome Trust
- Subjects
Evolutionary Biology ,Science & Technology ,Ecology ,Environmental Sciences & Ecology ,Life Sciences & Biomedicine - Published
- 2020
3. 3D genomics across the tree of life reveals condensin II as a determinant of architecture type
- Author
-
Hoencamp, C, Dudchenko, O, Elbatsh, AMO, Brahmachari, S, Raaijmakers, JA, van Schaik, T, Cacciatore, AS, Contessoto, VG, van Heesbeen, RGHP, van den Broek, B, Mhaskar, AN, Teunissen, H, St Hilaire, BG, Weisz, D, Omer, AD, Pham, M, Colaric, Z, Yang, Z, Rao, SSP, Mitra, N, Lui, C, Yao, W, Khan, R, Moroz, LL, Kohn, A, St Leger, J, Mena, A, Holcroft, K, Gambetta, MC, Lim, F, Farley, E, Stein, N, Haddad, A, Chauss, D, Mutlu, AS, Wang, MC, Young, ND, Hildebrandt, E, Cheng, HH, Knight, CJ, Burnham, TLU, Hovel, KA, Beel, AJ, Mattei, P-J, Kornberg, RD, Warren, WC, Cary, G, Gomez-Skarmeta, JL, Hinman, V, Lindblad-Toh, K, Di Palma, F, Maeshima, K, Multani, AS, Sen, P, Nel-Themaat, L, Behringer, RR, Kaur, P, Medema, RH, van Steensel, B, de Wit, E, Onuchic, JN, Di Pierro, M, Aiden, EL, Rowland, BD, Hoencamp, C, Dudchenko, O, Elbatsh, AMO, Brahmachari, S, Raaijmakers, JA, van Schaik, T, Cacciatore, AS, Contessoto, VG, van Heesbeen, RGHP, van den Broek, B, Mhaskar, AN, Teunissen, H, St Hilaire, BG, Weisz, D, Omer, AD, Pham, M, Colaric, Z, Yang, Z, Rao, SSP, Mitra, N, Lui, C, Yao, W, Khan, R, Moroz, LL, Kohn, A, St Leger, J, Mena, A, Holcroft, K, Gambetta, MC, Lim, F, Farley, E, Stein, N, Haddad, A, Chauss, D, Mutlu, AS, Wang, MC, Young, ND, Hildebrandt, E, Cheng, HH, Knight, CJ, Burnham, TLU, Hovel, KA, Beel, AJ, Mattei, P-J, Kornberg, RD, Warren, WC, Cary, G, Gomez-Skarmeta, JL, Hinman, V, Lindblad-Toh, K, Di Palma, F, Maeshima, K, Multani, AS, Sen, P, Nel-Themaat, L, Behringer, RR, Kaur, P, Medema, RH, van Steensel, B, de Wit, E, Onuchic, JN, Di Pierro, M, Aiden, EL, and Rowland, BD
- Abstract
We investigated genome folding across the eukaryotic tree of life. We find two types of three-dimensional (3D) genome architectures at the chromosome scale. Each type appears and disappears repeatedly during eukaryotic evolution. The type of genome architecture that an organism exhibits correlates with the absence of condensin II subunits. Moreover, condensin II depletion converts the architecture of the human genome to a state resembling that seen in organisms such as fungi or mosquitoes. In this state, centromeres cluster together at nucleoli, and heterochromatin domains merge. We propose a physical model in which lengthwise compaction of chromosomes by condensin II during mitosis determines chromosome-scale genome architecture, with effects that are retained during the subsequent interphase. This mechanism likely has been conserved since the last common ancestor of all eukaryotes.
- Published
- 2021
4. CTCF counter-regulates cardiomyocyte development and maturation programs in the embryonic heart
- Author
-
Gomez-Velazquez, M, Badia-Careaga, C, Lechuga-Vieco, AV, Nieto-Arellano, R, Tena, JJ, Rollan, I, Alvarez, A, Torroja, C, Caceres, EF, Roy, A, Galjart, Niels, Delgado-Olguin, P, Sanchez-Cabo, F, Enriquez, JA, Gomez-Skarmeta, JL, Manzanares, M, Gomez-Velazquez, M, Badia-Careaga, C, Lechuga-Vieco, AV, Nieto-Arellano, R, Tena, JJ, Rollan, I, Alvarez, A, Torroja, C, Caceres, EF, Roy, A, Galjart, Niels, Delgado-Olguin, P, Sanchez-Cabo, F, Enriquez, JA, Gomez-Skarmeta, JL, and Manzanares, M
- Published
- 2017
5. Xiro-1 controls mesoderm patterning by repressing bmp-4 expression in the spemann organizer
- Author
-
Glavic, A, Gomez-Skarmeta, JL, Mayor, R, and Fundación Andes
- Subjects
Mesoderm ,animal structures ,Xiro-1 ,embryonic structures ,BMP-4 ,Spemann organizer - Abstract
The Iroquois genes code for homeodomain proteins that have been implicated in the neural development of Drosophila and vertebrates. We show here for the first time that Xiro‐1, one of the Xenopus Iroquois genes, is expressed in the Spemann organizer from the start of gastrulation and that its overexpression induces a secondary axis as well as the ectopic expression of several organizer genes, such as chordin, goosecoid, and Xlim‐1. Our results also indicate that Xiro‐1 normally functions as a transcriptional repressor in the mesoderm. Overexpression of Xiro‐1 or a chimeric form fused to the repressor domain of Engrailed cause similar phenotypes while overexpression of functional derivatives of Xiro‐1 fused with transactivation domains (VP16 or E1A) produce the opposite effects. Finally, we show that Xiro‐1 works as a repressor of bmp‐4 transcription and that its effect on organizer development is dependent on BMP‐4 activity. We propose that the previously observed down regulation of bmp‐4 in the dorsal mesoderm during gastrulation can be explained by the repressor activity of Xiro‐1 described here. Thus, Xiro‐1 seems to have at least two different functions: control of neural plate and organizer development, both of which could be mediated by repression of bmp‐4 transcription., A.G is a fellow from Fundación Andes.
- Published
- 2001
6. An amphioxus neurula stage cell atlas supports a complex scenario for the emergence of vertebrate head mesoderm.
- Author
-
Grau-Bové X, Subirana L, Meister L, Soubigou A, Neto A, Elek A, Naranjo S, Fornas O, Gomez-Skarmeta JL, Tena JJ, Irimia M, Bertrand S, Sebé-Pedrós A, and Escriva H
- Subjects
- Animals, Somites embryology, Somites cytology, Somites metabolism, Biological Evolution, Transcriptome, Mesoderm cytology, Mesoderm embryology, Lancelets embryology, Lancelets genetics, Head embryology, Gene Expression Regulation, Developmental, Vertebrates embryology, Vertebrates genetics
- Abstract
The emergence of new structures can often be linked to the evolution of novel cell types that follows the rewiring of developmental gene regulatory subnetworks. Vertebrates are characterized by a complex body plan compared to the other chordate clades and the question remains of whether and how the emergence of vertebrate morphological innovations can be related to the appearance of new embryonic cell populations. We previously proposed, by studying mesoderm development in the cephalochordate amphioxus, a scenario for the evolution of the vertebrate head mesoderm. To further test this scenario at the cell population level, we used scRNA-seq to construct a cell atlas of the amphioxus neurula, stage at which the main mesodermal compartments are specified. Our data allowed us to validate the presence of a prechordal-plate like territory in amphioxus. Additionally, the transcriptomic profile of somite cell populations supports the homology between specific territories of amphioxus somites and vertebrate cranial/pharyngeal and lateral plate mesoderm. Finally, our work provides evidence that the appearance of the specific mesodermal structures of the vertebrate head was associated to both segregation of pre-existing cell populations, and co-option of new genes for the control of myogenesis., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
7. Pluripotency factors regulate the onset of Hox cluster activation in the early embryo.
- Author
-
Tiana M, Lopez-Jimenez E, de Aja JS, Barral A, Victorino J, Badia-Careaga C, Rollan I, Rouco R, Santos E, Sanchez-Iranzo H, Acemel RD, Torroja C, Adan J, Andres-Leon E, Gomez-Skarmeta JL, Giovinazzo G, Sanchez-Cabo F, and Manzanares M
- Abstract
Pluripotent cells are a transient population of the mammalian embryo dependent on transcription factors, such as OCT4 and NANOG, which maintain pluripotency while suppressing lineage specification. However, these factors are also expressed during early phases of differentiation, and their role in the transition from pluripotency to lineage specification is largely unknown. We found that pluripotency factors play a dual role in regulating key lineage specifiers, initially repressing their expression and later being required for their proper activation. We show that Oct4 is necessary for activation of HoxB genes during differentiation of embryonic stem cells and in the embryo. In addition, we show that the HoxB cluster is coordinately regulated by OCT4 binding sites located at the 3' end of the cluster. Our results show that core pluripotency factors are not limited to maintaining the precommitted epiblast but are also necessary for the proper deployment of subsequent developmental programs.
- Published
- 2022
- Full Text
- View/download PDF
8. Gene Regulatory Networks of Epidermal and Neural Fate Choice in a Chordate.
- Author
-
Leon A, Subirana L, Magre K, Cases I, Tena JJ, Irimia M, Gomez-Skarmeta JL, Escriva H, and Bertrand S
- Subjects
- Animals, Epidermis metabolism, Gene Expression Regulation, Developmental, Nervous System metabolism, Transcription Factors genetics, Transcription Factors metabolism, Gene Regulatory Networks, Lancelets
- Abstract
Neurons are a highly specialized cell type only found in metazoans. They can be scattered throughout the body or grouped together, forming ganglia or nerve cords. During embryogenesis, centralized nervous systems develop from the ectoderm, which also forms the epidermis. How pluripotent ectodermal cells are directed toward neural or epidermal fates, and to which extent this process is shared among different animal lineages, are still open questions. Here, by using micromere explants, we were able to define in silico the putative gene regulatory networks (GRNs) underlying the first steps of the epidermis and the central nervous system formation in the cephalochordate amphioxus. We propose that although the signal triggering neural induction in amphioxus (i.e., Nodal) is different from vertebrates, the main transcription factors implicated in this process are conserved. Moreover, our data reveal that transcription factors of the neural program seem to not only activate neural genes but also to potentially have direct inputs into the epidermal GRN, suggesting that the Nodal signal might also contribute to neural fate commitment by repressing the epidermal program. Our functional data on whole embryos support this result and highlight the complex interactions among the transcription factors activated by the signaling pathways that drive ectodermal cell fate choice in chordates., (© The Author(s) 2022. Published by Oxford University Press on behalf of Society for Molecular Biology and Evolution.)
- Published
- 2022
- Full Text
- View/download PDF
9. Asthma-associated genetic variants induce IL33 differential expression through an enhancer-blocking regulatory region.
- Author
-
Aneas I, Decker DC, Howard CL, Sobreira DR, Sakabe NJ, Blaine KM, Stein MM, Hrusch CL, Montefiori LE, Tena J, Magnaye KM, Clay SM, Gern JE, Jackson DJ, Altman MC, Naureckas ET, Hogarth DK, White SR, Gomez-Skarmeta JL, Schoetler N, Ober C, Sperling AI, and Nóbrega MA
- Subjects
- Alleles, Animals, Asthma metabolism, Chromatin genetics, Chromatin metabolism, Female, Genetic Predisposition to Disease, Humans, Interleukin-33 metabolism, Male, Mice, Transgenic, Octamer Transcription Factor-1 genetics, Octamer Transcription Factor-1 metabolism, Polymorphism, Single Nucleotide, Promoter Regions, Genetic, Zebrafish, Asthma genetics, Enhancer Elements, Genetic, Interleukin-33 genetics
- Abstract
Genome-wide association studies (GWAS) have implicated the IL33 locus in asthma, but the underlying mechanisms remain unclear. Here, we identify a 5 kb region within the GWAS-defined segment that acts as an enhancer-blocking element in vivo and in vitro. Chromatin conformation capture showed that this 5 kb region loops to the IL33 promoter, potentially regulating its expression. We show that the asthma-associated single nucleotide polymorphism (SNP) rs1888909, located within the 5 kb region, is associated with IL33 gene expression in human airway epithelial cells and IL-33 protein expression in human plasma, potentially through differential binding of OCT-1 (POU2F1) to the asthma-risk allele. Our data demonstrate that asthma-associated variants at the IL33 locus mediate allele-specific regulatory activity and IL33 expression, providing a mechanism through which a regulatory SNP contributes to genetic risk of asthma., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
10. Straightforward protocol for allele-specific chromatin conformation capture.
- Author
-
Acemel RD, Tena JJ, Gomez-Skarmeta JL, Fibla J, and Royo JL
- Subjects
- Alleles, Animals, DNA genetics, DNA Copy Number Variations genetics, Databases, Genetic, Enhancer Elements, Genetic genetics, Humans, Mice, Nucleic Acid Conformation, Polymorphism, Single Nucleotide genetics, Promoter Regions, Genetic genetics, Quantitative Trait Loci genetics, Receptors, Cell Surface genetics, Chromatin metabolism, Chromatin physiology, Polymerase Chain Reaction methods
- Abstract
A key advance in our understanding of gene regulation came with the finding that the genome undergoes three-dimensional nuclear folding in a genetically determined process. This 3D conformation directly influences the association between enhancers and their target promoters. This complex interplay has been proven to be essential for gene regulation, and genetic variants affecting this process have been associated to human diseases. The development of new technologies that quantify these DNA interactions represented a revolution in the field. High throughput techniques like HiC provide a general picture of chromatin topology. However, they often lack resolution to evidence subtle effects that single nucleotide polymorphisms exert over the contacts between cis-regulatory regions and target promoters. Here we propose a cost-efficient approach to perform allele-specific chromatin conformation analysis. As a proof of concept, we analyzed the impact of a common deletion mapping between SIRPB1 promoter and one of its downstream enhancers., (Copyright © 2020. Published by Elsevier B.V.)
- Published
- 2021
- Full Text
- View/download PDF
11. Boundary sequences flanking the mouse tyrosinase locus ensure faithful pattern of gene expression.
- Author
-
Seruggia D, Fernández A, Cantero M, Fernández-Miñán A, Gomez-Skarmeta JL, Pelczar P, and Montoliu L
- Subjects
- Animals, CRISPR-Associated Protein 9, CRISPR-Cas Systems, Cell Line, Chromatin metabolism, Chromatin ultrastructure, Enhancer Elements, Genetic genetics, Gene Editing, HEK293 Cells, Humans, Mice, Monophenol Monooxygenase metabolism, Promoter Regions, Genetic genetics, Zebrafish embryology, Zebrafish genetics, Gene Expression Regulation genetics, Genetic Loci genetics, Monophenol Monooxygenase genetics
- Abstract
Control of gene expression is dictated by cell-type specific regulatory sequences that physically organize the structure of chromatin, including promoters, enhancers and insulators. While promoters and enhancers convey cell-type specific activating signals, insulators prevent the cross-talk of regulatory elements within adjacent loci and safeguard the specificity of action of promoters and enhancers towards their targets in a tissue specific manner. Using the mouse tyrosinase (Tyr) locus as an experimental model, a gene whose mutations are associated with albinism, we described the chromatin structure in cells at two distinct transcriptional states. Guided by chromatin structure, through the use of Chromosome Conformation Capture (3C), we identified sequences at the 5' and 3' boundaries of this mammalian gene that function as enhancers and insulators. By CRISPR/Cas9-mediated chromosomal deletion, we dissected the functions of these two regulatory elements in vivo in the mouse, at the endogenous chromosomal context, and proved their mechanistic role as genomic insulators, shielding the Tyr locus from the expression patterns of adjacent genes.
- Published
- 2020
- Full Text
- View/download PDF
12. Dynamic changes in the epigenomic landscape regulate human organogenesis and link to developmental disorders.
- Author
-
Gerrard DT, Berry AA, Jennings RE, Birket MJ, Zarrineh P, Garstang MG, Withey SL, Short P, Jiménez-Gancedo S, Firbas PN, Donaldson I, Sharrocks AD, Hanley KP, Hurles ME, Gomez-Skarmeta JL, Bobola N, and Hanley NA
- Subjects
- Animals, Animals, Genetically Modified, Databases, Genetic, Enhancer Elements, Genetic, Gene Expression Regulation, Developmental, Histone Code genetics, Humans, Models, Genetic, Mutation, Organogenesis physiology, Promoter Regions, Genetic, Tissue Distribution, Transcription Factors metabolism, Zebrafish embryology, Zebrafish genetics, Developmental Disabilities genetics, Epigenesis, Genetic, Organogenesis genetics
- Abstract
How the genome activates or silences transcriptional programmes governs organ formation. Little is known in human embryos undermining our ability to benchmark the fidelity of stem cell differentiation or cell programming, or interpret the pathogenicity of noncoding variation. Here, we study histone modifications across thirteen tissues during human organogenesis. We integrate the data with transcription to build an overview of how the human genome differentially regulates alternative organ fates including by repression. Promoters from nearly 20,000 genes partition into discrete states. Key developmental gene sets are actively repressed outside of the appropriate organ without obvious bivalency. Candidate enhancers, functional in zebrafish, allow imputation of tissue-specific and shared patterns of transcription factor binding. Overlaying more than 700 noncoding mutations from patients with developmental disorders allows correlation to unanticipated target genes. Taken together, the data provide a comprehensive genomic framework for investigating normal and abnormal human development.
- Published
- 2020
- Full Text
- View/download PDF
13. Genetic regulation of amphioxus somitogenesis informs the evolution of the vertebrate head mesoderm.
- Author
-
Aldea D, Subirana L, Keime C, Meister L, Maeso I, Marcellini S, Gomez-Skarmeta JL, Bertrand S, and Escriva H
- Subjects
- Animals, Gene Expression Regulation, Developmental, Mesoderm, Somites, Vertebrates, Lancelets
- Abstract
The evolution of vertebrates from an ancestral chordate was accompanied by the acquisition of a predatory lifestyle closely associated to the origin of a novel anterior structure, the highly specialized head. While the vertebrate head mesoderm is unsegmented, the paraxial mesoderm of the earliest divergent chordate clade, the cephalochordates (amphioxus), is fully segmented in somites. We have previously shown that fibroblast growth factor signalling controls the formation of the most anterior somites in amphioxus; therefore, unravelling the fibroblast growth factor signalling downstream effectors is of crucial importance to shed light on the evolutionary origin of vertebrate head muscles. By using a comparative RNA sequencing approach and genetic functional analyses, we show that several transcription factors, such as Six1/2, Pax3/7 and Zic, act in combination to ensure the formation of three different somite populations. Interestingly, these proteins are orthologous to key regulators of trunk, and not head, muscle formation in vertebrates. Contrary to prevailing thinking, our results suggest that the vertebrate head mesoderm is of visceral and not paraxial origin and support a multistep evolutionary scenario for the appearance of the unsegmented mesoderm of the vertebrates new 'head'.
- Published
- 2019
- Full Text
- View/download PDF
14. Evolution of embryonic cis-regulatory landscapes between divergent Phallusia and Ciona ascidians.
- Author
-
Madgwick A, Magri MS, Dantec C, Gailly D, Fiuza UM, Guignard L, Hettinger S, Gomez-Skarmeta JL, and Lemaire P
- Subjects
- Animals, Base Sequence, Body Patterning genetics, Chromatin genetics, Conserved Sequence genetics, Embryonic Development genetics, Enhancer Elements, Genetic, Gastrula embryology, Gene Expression Regulation, Developmental, Species Specificity, Time Factors, Ciona embryology, Ciona genetics, Embryo, Nonmammalian metabolism, Evolution, Molecular, Genetic Variation, Regulatory Sequences, Nucleic Acid genetics, Urochordata embryology, Urochordata genetics
- Abstract
Ascidian species of the Phallusia and Ciona genera are distantly related, their last common ancestor dating several hundred million years ago. Although their genome sequences have extensively diverged since this radiation, Phallusia and Ciona species share almost identical early morphogenesis and stereotyped cell lineages. Here, we explored the evolution of transcriptional control between P. mammillata and C. robusta. We combined genome-wide mapping of open chromatin regions in both species with a comparative analysis of the regulatory sequences of a test set of 10 pairs of orthologous early regulatory genes with conserved expression patterns. We find that ascidian chromatin accessibility landscapes obey similar rules as in other metazoa. Open-chromatin regions are short, highly conserved within each genus and cluster around regulatory genes. The dynamics of chromatin accessibility and closest-gene expression are strongly correlated during early embryogenesis. Open-chromatin regions are highly enriched in cis-regulatory elements: 73% of 49 open chromatin regions around our test genes behaved as either distal enhancers or proximal enhancer/promoters following electroporation in Phallusia eggs. Analysis of this datasets suggests a pervasive use in ascidians of "shadow" enhancers with partially overlapping activities. Cross-species electroporations point to a deep conservation of both the trans-regulatory logic between these distantly-related ascidians and the cis-regulatory activities of individual enhancers. Finally, we found that the relative order and approximate distance to the transcription start site of open chromatin regions can be conserved between Ciona and Phallusia species despite extensive sequence divergence, a property that can be used to identify orthologous enhancers, whose regulatory activity can partially diverge., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
15. Evolutionary emergence of the rac3b / rfng / sgca regulatory cluster refined mechanisms for hindbrain boundaries formation.
- Author
-
Letelier J, Terriente J, Belzunce I, Voltes A, Undurraga CA, Polvillo R, Devos L, Tena JJ, Maeso I, Retaux S, Gomez-Skarmeta JL, Martínez-Morales JR, and Pujades C
- Subjects
- Animals, Body Patterning genetics, CRISPR-Cas Systems, Cell Movement, Characidae genetics, Characidae physiology, Chromatin genetics, Chromatin ultrastructure, Evolution, Molecular, Fishes classification, Fishes genetics, Morphogenesis, Mutagenesis, Site-Directed, Neurogenesis, Phylogeny, Sarcoglycans genetics, Species Specificity, Zebrafish genetics, Zebrafish Proteins genetics, rac GTP-Binding Proteins genetics, Actomyosin physiology, Gene Expression Regulation, Developmental, Rhombencephalon embryology, Sarcoglycans physiology, Zebrafish embryology, Zebrafish Proteins physiology, rac GTP-Binding Proteins physiology
- Abstract
Developmental programs often rely on parallel morphogenetic mechanisms that guarantee precise tissue architecture. While redundancy constitutes an obvious selective advantage, little is known on how novel morphogenetic mechanisms emerge during evolution. In zebrafish, rhombomeric boundaries behave as an elastic barrier, preventing cell intermingling between adjacent compartments. Here, we identify the fundamental role of the small-GTPase Rac3b in actomyosin cable assembly at hindbrain boundaries. We show that the novel rac3b / rfng / sgca regulatory cluster, which is specifically expressed at the boundaries, emerged in the Ostariophysi superorder by chromosomal rearrangement that generated new cis -regulatory interactions. By combining 4C-seq, ATAC-seq, transgenesis, and CRISPR-induced deletions, we characterized this regulatory domain, identifying hindbrain boundary-specific cis -regulatory elements. Our results suggest that the capacity of boundaries to act as an elastic mesh for segregating rhombomeric cells evolved by cooption of critical genes to a novel regulatory block, refining the mechanisms for hindbrain segmentation., Competing Interests: The authors declare no conflict of interest.
- Published
- 2018
- Full Text
- View/download PDF
16. A common copy-number variant within SIRPB1 correlates with human Out-of-Africa migration after genetic drift correction.
- Author
-
Royo JL, Valls J, Acemel RD, Gómez-Marin C, Pascual-Pons M, Lupiañez A, Gomez-Skarmeta JL, and Fibla J
- Subjects
- Africa, Animals, Animals, Genetically Modified, Central Nervous System metabolism, Chromatin metabolism, Epigenesis, Genetic, Gene Expression, Gene Frequency, Genetic Association Studies, Haplotypes, Humans, Promoter Regions, Genetic, Racial Groups genetics, Receptors, Cell Surface metabolism, Zebrafish, DNA Copy Number Variations, Genetic Drift, Human Migration, Receptors, Cell Surface genetics
- Abstract
Previous reports have proposed that personality may have played a role on human Out-Of-Africa migration, pinpointing some genetic variants that were positively selected in the migrating populations. In this work, we discuss the role of a common copy-number variant within the SIRPB1 gene, recently associated with impulsive behavior, in the human Out-Of-Africa migration. With the analysis of the variant distribution across forty-two different populations, we found that the SIRPB1 haplotype containing duplicated allele significantly correlated with human migratory distance, being one of the few examples of positively selected loci found across the human world colonization. Circular Chromosome Conformation Capture (4C-seq) experiments from the SIRPB1 promoter revealed important 3D modifications in the locus depending on the presence or absence of the duplication variant. In addition, a 3' enhancer showed neural activity in transgenic models, suggesting that the presence of the CNV may compromise the expression of SIRPB1 in the central nervous system, paving the way to construct a molecular explanation of the SIRPB1 variants role in human migration.
- Published
- 2018
- Full Text
- View/download PDF
17. CTCF counter-regulates cardiomyocyte development and maturation programs in the embryonic heart.
- Author
-
Gomez-Velazquez M, Badia-Careaga C, Lechuga-Vieco AV, Nieto-Arellano R, Tena JJ, Rollan I, Alvarez A, Torroja C, Caceres EF, Roy AR, Galjart N, Delgado-Olguin P, Sanchez-Cabo F, Enriquez JA, Gomez-Skarmeta JL, and Manzanares M
- Subjects
- Animals, CCCTC-Binding Factor, Cell Differentiation genetics, Enhancer Elements, Genetic, Gene Expression Regulation, Developmental, Heart embryology, Heart Ventricles embryology, Mice, Mitochondria genetics, Mitochondria metabolism, Organogenesis genetics, Promoter Regions, Genetic, Protein Binding, Transcriptional Activation genetics, Chromatin genetics, Embryonic Development genetics, Heart growth & development, Heart Ventricles growth & development, Repressor Proteins genetics
- Abstract
Cardiac progenitors are specified early in development and progressively differentiate and mature into fully functional cardiomyocytes. This process is controlled by an extensively studied transcriptional program. However, the regulatory events coordinating the progression of such program from development to maturation are largely unknown. Here, we show that the genome organizer CTCF is essential for cardiogenesis and that it mediates genomic interactions to coordinate cardiomyocyte differentiation and maturation in the developing heart. Inactivation of Ctcf in cardiac progenitor cells and their derivatives in vivo during development caused severe cardiac defects and death at embryonic day 12.5. Genome wide expression analysis in Ctcf mutant hearts revealed that genes controlling mitochondrial function and protein production, required for cardiomyocyte maturation, were upregulated. However, mitochondria from mutant cardiomyocytes do not mature properly. In contrast, multiple development regulatory genes near predicted heart enhancers, including genes in the IrxA cluster, were downregulated in Ctcf mutants, suggesting that CTCF promotes cardiomyocyte differentiation by facilitating enhancer-promoter interactions. Accordingly, loss of CTCF disrupts gene expression and chromatin interactions as shown by chromatin conformation capture followed by deep sequencing. Furthermore, CRISPR-mediated deletion of an intergenic CTCF site within the IrxA cluster alters gene expression in the developing heart. Thus, CTCF mediates local regulatory interactions to coordinate transcriptional programs controlling transitions in morphology and function during heart development.
- Published
- 2017
- Full Text
- View/download PDF
18. Meis1 coordinates a network of genes implicated in eye development and microphthalmia.
- Author
-
Marcos S, González-Lázaro M, Beccari L, Carramolino L, Martin-Bermejo MJ, Amarie O, Mateos-San Martín D, Torroja C, Bogdanović O, Doohan R, Puk O, Hrabě de Angelis M, Graw J, Gomez-Skarmeta JL, Casares F, Torres M, and Bovolenta P
- Subjects
- Aging pathology, Animals, Apoptosis genetics, Base Sequence, Binding Sites, Blood Vessels metabolism, Blood Vessels pathology, Chromatin Immunoprecipitation, Embryo, Mammalian metabolism, Embryo, Mammalian pathology, Enhancer Elements, Genetic genetics, Haploinsufficiency genetics, Hematopoiesis genetics, Homeodomain Proteins genetics, Humans, Mice, Molecular Sequence Data, Myeloid Ecotropic Viral Integration Site 1 Protein, Neoplasm Proteins deficiency, Neoplasm Proteins genetics, Neurogenesis genetics, Protein Binding, Receptors, Notch metabolism, Signal Transduction genetics, Eye embryology, Eye metabolism, Gene Regulatory Networks, Homeodomain Proteins metabolism, Microphthalmos embryology, Microphthalmos genetics, Neoplasm Proteins metabolism
- Abstract
Microphthalmos is a rare congenital anomaly characterized by reduced eye size and visual deficits of variable degree. Sporadic and hereditary microphthalmos have been associated with heterozygous mutations in genes fundamental for eye development. Yet, many cases are idiopathic or await the identification of molecular causes. Here we show that haploinsufficiency of Meis1, which encodes a transcription factor with evolutionarily conserved expression in the embryonic trunk, brain and sensory organs, including the eye, causes microphthalmic traits and visual impairment in adult mice. By combining analysis of Meis1 loss-of-function and conditional Meis1 functional rescue with ChIP-seq and RNA-seq approaches we show that, in contrast to its preferential association with Hox-Pbx BSs in the trunk, Meis1 binds to Hox/Pbx-independent sites during optic cup development. In the eye primordium, Meis1 coordinates, in a dose-dependent manner, retinal proliferation and differentiation by regulating genes responsible for human microphthalmia and components of the Notch signaling pathway. In addition, Meis1 is required for eye patterning by controlling a set of eye territory-specific transcription factors, so that in Meis1(-/-) embryos boundaries among the different eye territories are shifted or blurred. We propose that Meis1 is at the core of a genetic network implicated in eye patterning/microphthalmia, and represents an additional candidate for syndromic cases of these ocular malformations., (© 2015. Published by The Company of Biologists Ltd.)
- Published
- 2015
- Full Text
- View/download PDF
19. TEAD and YAP regulate the enhancer network of human embryonic pancreatic progenitors.
- Author
-
Cebola I, Rodríguez-Seguí SA, Cho CH, Bessa J, Rovira M, Luengo M, Chhatriwala M, Berry A, Ponsa-Cobas J, Maestro MA, Jennings RE, Pasquali L, Morán I, Castro N, Hanley NA, Gomez-Skarmeta JL, Vallier L, and Ferrer J
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Animals, Animals, Genetically Modified, Cell Differentiation, Cell Lineage, Cell Proliferation, Cells, Cultured, Computational Biology, DNA-Binding Proteins genetics, Databases, Genetic, Gene Expression Regulation, Developmental, Gene Regulatory Networks, Humans, Mice, Inbred C57BL, Nuclear Proteins genetics, Organogenesis, Pancreas embryology, Phenotype, Phosphoproteins genetics, RNA, Messenger metabolism, TEA Domain Transcription Factors, Time Factors, Transcription Factors genetics, YAP-Signaling Proteins, Zebrafish embryology, Zebrafish genetics, Zebrafish metabolism, Adaptor Proteins, Signal Transducing metabolism, DNA-Binding Proteins metabolism, Embryonic Stem Cells metabolism, Multipotent Stem Cells metabolism, Nuclear Proteins metabolism, Pancreas metabolism, Phosphoproteins metabolism, Signal Transduction, Transcription Factors metabolism
- Abstract
The genomic regulatory programmes that underlie human organogenesis are poorly understood. Pancreas development, in particular, has pivotal implications for pancreatic regeneration, cancer and diabetes. We have now characterized the regulatory landscape of embryonic multipotent progenitor cells that give rise to all pancreatic epithelial lineages. Using human embryonic pancreas and embryonic-stem-cell-derived progenitors we identify stage-specific transcripts and associated enhancers, many of which are co-occupied by transcription factors that are essential for pancreas development. We further show that TEAD1, a Hippo signalling effector, is an integral component of the transcription factor combinatorial code of pancreatic progenitor enhancers. TEAD and its coactivator YAP activate key pancreatic signalling mediators and transcription factors, and regulate the expansion of pancreatic progenitors. This work therefore uncovers a central role for TEAD and YAP as signal-responsive regulators of multipotent pancreatic progenitors, and provides a resource for the study of embryonic development of the human pancreas.
- Published
- 2015
- Full Text
- View/download PDF
20. SIRPB1 copy-number polymorphism as candidate quantitative trait locus for impulsive-disinhibited personality.
- Author
-
Laplana M, Royo JL, García LF, Aluja A, Gomez-Skarmeta JL, and Fibla J
- Subjects
- Adult, Animals, Case-Control Studies, Criminals, Haplotypes, Humans, Insulator Elements, Male, RNA, Messenger genetics, RNA, Messenger metabolism, Receptors, Cell Surface metabolism, Zebrafish, DNA Copy Number Variations, Impulsive Behavior, Inhibition, Psychological, Quantitative Trait Loci, Receptors, Cell Surface genetics
- Abstract
Impulsive-disinhibited personality (IDP) is a behavioral trait mainly characterized by seeking immediate gratification at the expense of more enduring or long-term gains. This trait has a major role in the development of several disinhibitory behaviors and syndromes, including psychopathy, attention-deficit and hyperactivity disorder, cluster-B personality disorders, criminality and alcoholism. Available data consistently support a strong heritable component, accounting for 30-60% of the observed variance in personality traits. A genome-wide analysis of copy-number variants was designed to identify novel genetic pathways associated with the IDP trait, using a series of 261 male participants with maximized opposite IDP scores. Quantitative trait locus analysis of candidate copy-number variants (CNVs) was conducted across the entire IDP continuum. Functional effects of associated variants were evaluated in zebrafish embryos. A common CNV mapping to the immune-related gene SIRPB1 was significantly associated with IDP scores in a dose-dependent manner (β=-0.172, P<0.017). Expression quantitative trait locus analysis of the critical region revealed higher SIRPB1 mRNA levels associated with the haplotype containing the deleted allele (P<0.0007). Epigenetic marks highlighted the presence of two potential insulators within the deleted region, confirmed by functional assays in zebrafish embryos, which suggests that SIRPB1 expression rates are affected by the presence/absence of the insulator regions. Upregulation of SIRPB1 has been described in prefrontal cortex of patients with schizophrenia, providing a link between SIRPB1 and diseases involving disinhibition and failure to control impulsivity. We propose SIRPB1 as a novel candidate gene to account for phenotypic differences observed in the IDP trait., (© 2014 John Wiley & Sons Ltd and International Behavioural and Neural Genetics Society.)
- Published
- 2014
- Full Text
- View/download PDF
21. A polymorphic enhancer near GREM1 influences bowel cancer risk through differential CDX2 and TCF7L2 binding.
- Author
-
Lewis A, Freeman-Mills L, de la Calle-Mustienes E, Giráldez-Pérez RM, Davis H, Jaeger E, Becker M, Hubner NC, Nguyen LN, Zeron-Medina J, Bond G, Stunnenberg HG, Carvajal JJ, Gomez-Skarmeta JL, Leedham S, and Tomlinson I
- Subjects
- Animals, Base Sequence, CDX2 Transcription Factor, Cell Line, Tumor, Enhancer Elements, Genetic, Gene Expression Regulation, Neoplastic, Genetic Association Studies, Genetic Predisposition to Disease, Humans, Mice, Transgenic, Organ Specificity, Polymorphism, Single Nucleotide, Risk, Colonic Neoplasms genetics, Homeodomain Proteins metabolism, Intercellular Signaling Peptides and Proteins genetics, Transcription Factor 7-Like 2 Protein metabolism
- Abstract
A rare germline duplication upstream of the bone morphogenetic protein antagonist GREM1 causes a Mendelian-dominant predisposition to colorectal cancer (CRC). The underlying disease mechanism is strong, ectopic GREM1 overexpression in the intestinal epithelium. Here, we confirm that a common GREM1 polymorphism, rs16969681, is also associated with CRC susceptibility, conferring ∼20% differential risk in the general population. We hypothesized the underlying cause to be moderate differences in GREM1 expression. We showed that rs16969681 lies in a region of active chromatin with allele- and tissue-specific enhancer activity. The CRC high-risk allele was associated with stronger gene expression, and higher Grem1 mRNA levels increased the intestinal tumor burden in Apc(Min) mice. The intestine-specific transcription factor CDX2 and Wnt effector TCF7L2 bound near rs16969681, with significantly higher affinity for the risk allele, and CDX2 overexpression in CDX2/GREM1-negative cells caused re-expression of GREM1. rs16969681 influences CRC risk through effects on Wnt-driven GREM1 expression in colorectal tumors., (Copyright © 2014 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
22. Obesity-associated variants within FTO form long-range functional connections with IRX3.
- Author
-
Smemo S, Tena JJ, Kim KH, Gamazon ER, Sakabe NJ, Gómez-Marín C, Aneas I, Credidio FL, Sobreira DR, Wasserman NF, Lee JH, Puviindran V, Tam D, Shen M, Son JE, Vakili NA, Sung HK, Naranjo S, Acemel RD, Manzanares M, Nagy A, Cox NJ, Hui CC, Gomez-Skarmeta JL, and Nóbrega MA
- Subjects
- Adipose Tissue metabolism, Alpha-Ketoglutarate-Dependent Dioxygenase FTO, Animals, Basal Metabolism genetics, Body Mass Index, Body Weight genetics, Brain metabolism, Diabetes Mellitus, Type 2 genetics, Diet, Genes, Dominant genetics, Homeodomain Proteins metabolism, Humans, Hypothalamus metabolism, Male, Mice, Phenotype, Polymorphism, Single Nucleotide genetics, Promoter Regions, Genetic genetics, Thinness genetics, Transcription Factors deficiency, Transcription Factors metabolism, Zebrafish embryology, Zebrafish genetics, Homeodomain Proteins genetics, Introns genetics, Mixed Function Oxygenases genetics, Obesity genetics, Oxo-Acid-Lyases genetics, Proteins genetics, Transcription Factors genetics
- Abstract
Genome-wide association studies (GWAS) have reproducibly associated variants within introns of FTO with increased risk for obesity and type 2 diabetes (T2D). Although the molecular mechanisms linking these noncoding variants with obesity are not immediately obvious, subsequent studies in mice demonstrated that FTO expression levels influence body mass and composition phenotypes. However, no direct connection between the obesity-associated variants and FTO expression or function has been made. Here we show that the obesity-associated noncoding sequences within FTO are functionally connected, at megabase distances, with the homeobox gene IRX3. The obesity-associated FTO region directly interacts with the promoters of IRX3 as well as FTO in the human, mouse and zebrafish genomes. Furthermore, long-range enhancers within this region recapitulate aspects of IRX3 expression, suggesting that the obesity-associated interval belongs to the regulatory landscape of IRX3. Consistent with this, obesity-associated single nucleotide polymorphisms are associated with expression of IRX3, but not FTO, in human brains. A direct link between IRX3 expression and regulation of body mass and composition is demonstrated by a reduction in body weight of 25 to 30% in Irx3-deficient mice, primarily through the loss of fat mass and increase in basal metabolic rate with browning of white adipose tissue. Finally, hypothalamic expression of a dominant-negative form of Irx3 reproduces the metabolic phenotypes of Irx3-deficient mice. Our data suggest that IRX3 is a functional long-range target of obesity-associated variants within FTO and represents a novel determinant of body mass and composition.
- Published
- 2014
- Full Text
- View/download PDF
23. The genome sequencing of an albino Western lowland gorilla reveals inbreeding in the wild.
- Author
-
Prado-Martinez J, Hernando-Herraez I, Lorente-Galdos B, Dabad M, Ramirez O, Baeza-Delgado C, Morcillo-Suarez C, Alkan C, Hormozdiari F, Raineri E, Estellé J, Fernandez-Callejo M, Valles M, Ritscher L, Schöneberg T, de la Calle-Mustienes E, Casillas S, Rubio-Acero R, Melé M, Engelken J, Caceres M, Gomez-Skarmeta JL, Gut M, Bertranpetit J, Gut IG, Abello T, Eichler EE, Mingarro I, Lalueza-Fox C, Navarro A, and Marques-Bonet T
- Subjects
- Amino Acid Sequence, Animals, Female, Heterozygote, Male, Membrane Transport Proteins chemistry, Membrane Transport Proteins genetics, Microsatellite Repeats genetics, Molecular Sequence Data, Mutation, Sequence Analysis, DNA, Genomics, Gorilla gorilla genetics, High-Throughput Nucleotide Sequencing, Inbreeding
- Abstract
Background: The only known albino gorilla, named Snowflake, was a male wild born individual from Equatorial Guinea who lived at the Barcelona Zoo for almost 40 years. He was diagnosed with non-syndromic oculocutaneous albinism, i.e. white hair, light eyes, pink skin, photophobia and reduced visual acuity. Despite previous efforts to explain the genetic cause, this is still unknown. Here, we study the genetic cause of his albinism and making use of whole genome sequencing data we find a higher inbreeding coefficient compared to other gorillas., Results: We successfully identified the causal genetic variant for Snowflake's albinism, a non-synonymous single nucleotide variant located in a transmembrane region of SLC45A2. This transporter is known to be involved in oculocutaneous albinism type 4 (OCA4) in humans. We provide experimental evidence that shows that this amino acid replacement alters the membrane spanning capability of this transmembrane region. Finally, we provide a comprehensive study of genome-wide patterns of autozygogosity revealing that Snowflake's parents were related, being this the first report of inbreeding in a wild born Western lowland gorilla., Conclusions: In this study we demonstrate how the use of whole genome sequencing can be extended to link genotype and phenotype in non-model organisms and it can be a powerful tool in conservation genetics (e.g., inbreeding and genetic diversity) with the expected decrease in sequencing cost.
- Published
- 2013
- Full Text
- View/download PDF
24. Non-coding regulatory regions in genomes. Editorial.
- Author
-
Gomez Skarmeta JL
- Subjects
- Animals, Conserved Sequence, Drosophila melanogaster genetics, Evolution, Molecular, Gene Expression Regulation, Developmental, Humans, DNA, Intergenic genetics, Genome genetics, Regulatory Sequences, Nucleic Acid genetics
- Published
- 2009
- Full Text
- View/download PDF
25. Coenzyme Q10 and alpha-tocopherol protect against amitriptyline toxicity.
- Author
-
Cordero MD, Moreno-Fernández AM, Gomez-Skarmeta JL, de Miguel M, Garrido-Maraver J, Oropesa-Avila M, Rodríguez-Hernández A, Navas P, and Sánchez-Alcázar JA
- Subjects
- Animals, Cell Death drug effects, Cell Death physiology, Cell Proliferation drug effects, Cells, Cultured, Embryo, Nonmammalian cytology, Embryo, Nonmammalian drug effects, Embryo, Nonmammalian metabolism, Fibroblasts cytology, Fibroblasts drug effects, Fibroblasts metabolism, Fibroblasts pathology, Humans, Oxidative Stress drug effects, Oxidative Stress physiology, Reactive Oxygen Species metabolism, Ubiquinone pharmacology, Zebrafish embryology, Amitriptyline antagonists & inhibitors, Amitriptyline toxicity, Ubiquinone analogs & derivatives, alpha-Tocopherol pharmacology
- Abstract
Since amitriptyline is a very frequently prescribed antidepressant drug, it is not surprising that amitriptyline toxicity is relatively common. Amitriptyline toxic systemic effects include cardiovascular, autonomous nervous, and central nervous systems. To understand the mechanisms of amitriptyline toxicity we studied the cytotoxic effects of amitriptyline treatment on cultured primary human fibroblasts and zebrafish embryos, and the protective role of coenzyme Q(10) and alpha-tocopherol, two membrane antioxidants. We found that amitriptyline treatment induced oxidative stress and mitochondrial dysfunction in primary human fibroblasts. Mitochondrial dysfunction in amitriptyline treatment was characterized by reduced expression levels of mitochondrial proteins and coenzyme Q(10), decreased NADH:cytochrome c reductase activity, and a drop in mitochondrial membrane potential. Moreover, and as a consequence of these toxic effects, amitriptyline treatment induced a significant increase in apoptotic cell death activating mitochondrial permeability transition. Coenzyme Q(10) and alpha-tocopherol supplementation attenuated ROS production, lipid peroxidation, mitochondrial dysfunction, and cell death, suggesting that oxidative stress affecting cell membrane components is involved in amitriptyline cytotoxicity. Furthermore, amitriptyline-dependent toxicity and antioxidant protection were also evaluated in zebrafish embryos, a well established vertebrate model to study developmental toxicity. Amitriptyline significantly increased embryonic cell death and apoptosis rate, and both antioxidants provided a significant protection against amitriptyline embryotoxicity.
- Published
- 2009
- Full Text
- View/download PDF
26. Silencing of Smed-betacatenin1 generates radial-like hypercephalized planarians.
- Author
-
Iglesias M, Gomez-Skarmeta JL, Saló E, and Adell T
- Subjects
- Animals, Body Patterning, Cloning, Molecular, Embryo, Nonmammalian metabolism, Gene Expression Regulation, Developmental, Head, Helminth Proteins chemistry, Helminth Proteins genetics, In Situ Hybridization, Molecular Sequence Data, Planarians embryology, Planarians genetics, Planarians metabolism, RNA Interference, RNA, Messenger metabolism, Wnt Proteins genetics, Wnt Proteins physiology, Xenopus embryology, beta Catenin chemistry, beta Catenin genetics, Helminth Proteins physiology, Planarians physiology, Regeneration, beta Catenin physiology
- Abstract
Little is known about the molecular mechanisms responsible for axis establishment during non-embryonic processes such as regeneration and homeostasis. To address this issue, we set out to analyze the role of the canonical Wnt pathway in planarians, flatworms renowned for their extraordinary morphological plasticity. Canonical Wnt signalling is an evolutionarily conserved mechanism to confer polarity during embryonic development, specifying the anteroposterior (AP) axis in most bilaterians and the dorsoventral (DV) axis in early vertebrate embryos. beta-Catenin is a key element in this pathway, although it is a bifunctional protein that is also involved in cell-cell adhesion. Here, we report the characterization of two beta-catenin homologs from Schmidtea mediterranea (Smed-betacatenin1/2). Loss of function of Smed-betacatenin1, but not Smed-betacatenin2, in both regenerating and intact planarians, generates radial-like hypercephalized planarians in which the AP axis disappears but the DV axis remains unaffected, representing a unique example of a striking body symmetry transformation. The radial-like hypercephalized phenotype demonstrates the requirement for Smed-betacatenin1 in AP axis re-establishment and maintenance, and supports a conserved role for canonical Wnt signalling in AP axis specification, whereas the role of beta-catenin in DV axis establishment would be a vertebrate innovation. When considered alongside the protein domains present in each S. mediterranea beta-catenin and the results of functional assays in Xenopus embryos demonstrating nuclear accumulation and axis induction with Smed-betacatenin1, but not Smed-betacatenin2, these data suggest that S. mediterranea beta-catenins could be functionally specialized and that only Smed-betacatenin1 is involved in Wnt signalling.
- Published
- 2008
- Full Text
- View/download PDF
27. A novel function for the Xslug gene: control of dorsal mesendoderm development by repressing BMP-4.
- Author
-
Mayor R, Guerrero N, Young RM, Gomez-Skarmeta JL, and Cuellar C
- Subjects
- Animals, Biomarkers, Bone Morphogenetic Protein 4, Cell Nucleus metabolism, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Ectoderm metabolism, Ectoderm physiology, Homeodomain Proteins genetics, Mesoderm metabolism, Neural Crest metabolism, Organizers, Embryonic metabolism, PAX3 Transcription Factor, Paired Box Transcription Factors, Proteins genetics, Proto-Oncogene Proteins genetics, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Recombinant Fusion Proteins physiology, Repressor Proteins genetics, Repressor Proteins metabolism, Snail Family Transcription Factors, Transcription Factors genetics, Transcription Factors metabolism, Wnt Proteins, Xenopus laevis embryology, Zinc Fingers, Bone Morphogenetic Proteins genetics, Drosophila Proteins, Gene Expression Regulation, Developmental, Glycoproteins, Intercellular Signaling Peptides and Proteins, Mesoderm physiology, Repressor Proteins physiology, Transcription Factors physiology, Xenopus Proteins, Zebrafish Proteins
- Abstract
The Snail family of genes comprise a group of transcription factors with characteristic zinc finger motifs. One of the members of this family is the Slug gene. Slug has been implicated in the development of neural crest in chick and Xenopus by antisense loss of function experiments. Here, we have generated functional derivatives of Xslug by constructing cDNAs that encode the Xslug protein fused with the transactivation domain of the virus-derived VP16 activator or with the repressor domain of the Drosophila Engrailed protein. Our results suggest that Xslug normally functions as a transcriptional repressor and that Xslug-VP16 behaves as a dominant negative of Xslug. In the present work, we confirm and extend previous results that suggest that Xslug has an important function in neural crest development, by controlling its own transcription. In addition we have uncovered a new function for Xslug. We show that Xslug is expressed in the dorsal mesendoderm at the beginning of gastrulation, where is it able to upregulate the expression of dorsal genes. On the other hand when Xslug is expressed outside of the organizer it represses the expression of ventral genes. Our results indicate that this effect on mesodermal patterning depends on BMP activity, showing that Xslug can directly control the transcription of BMP-4.
- Published
- 2000
- Full Text
- View/download PDF
28. Araucan and caupolican, two members of the novel iroquois complex, encode homeoproteins that control proneural and vein-forming genes.
- Author
-
Gomez-Skarmeta JL, Diez del Corral R, de la Calle-Mustienes E, Ferré-Marcó D, and Modolell J
- Subjects
- Alleles, Animals, Base Sequence, Cloning, Molecular, Enhancer Elements, Genetic physiology, Gene Expression Regulation, Developmental genetics, Homeodomain Proteins metabolism, Molecular Sequence Data, Mutation genetics, Nervous System growth & development, Sequence Homology, Amino Acid, Transcription, Genetic genetics, Veins growth & development, Wings, Animal growth & development, Drosophila genetics, Drosophila Proteins, Genes, Insect genetics, Homeodomain Proteins genetics, Transcription Factors
- Abstract
In Drosophila imaginal wing discs, the achaete-scute (ac-sc) proneural genes and rhomboid (veinlet) are expressed in highly resolved patterns that prefigure the positions of sensory organs and wing veins, respectively. It is thought that these patterns are generated by a combination of factors (a prepattern) regulating these genes. We provide evidence for the existence of this prepattern by identifying two of its factors, Araucan and Caupolican. They are members of a novel family of homeoproteins, with homologs in vertebrates. Araucan and Caupolican, present in domains of the imaginal discs larger than those expressing ac-sc and rhomboid, are necessary for expression of these genes in the overlapping domains. Araucan and Caupolican appear to be positive, direct regulators of ac-sc.
- Published
- 1996
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.