Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Nat Ecol Evol ; 8(6): 1140-1153, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38622362

ABSTRACT

Regulation of gene expression is arguably the main mechanism underlying the phenotypic diversity of tissues within and between species. Here we assembled an extensive transcriptomic dataset covering 8 tissues across 20 bilaterian species and performed analyses using a symmetric phylogeny that allowed the combined and parallel investigation of gene expression evolution between vertebrates and insects. We specifically focused on widely conserved ancestral genes, identifying strong cores of pan-bilaterian tissue-specific genes and even larger groups that diverged to define vertebrate and insect tissues. Systematic inferences of tissue-specificity gains and losses show that nearly half of all ancestral genes have been recruited into tissue-specific transcriptomes. This occurred during both ancient and, especially, recent bilaterian evolution, with several gains being associated with the emergence of unique phenotypes (for example, novel cell types). Such pervasive evolution of tissue specificity was linked to gene duplication coupled with expression specialization of one of the copies, revealing an unappreciated prolonged effect of whole-genome duplications on recent vertebrate evolution.


Subject(s)
Evolution, Molecular , Insecta , Vertebrates , Animals , Insecta/genetics , Vertebrates/genetics , Organ Specificity , Transcriptome , Phylogeny
2.
Nucleic Acids Res ; 51(10): e56, 2023 06 09.
Article in English | MEDLINE | ID: mdl-37026474

ABSTRACT

Although splicing occurs largely co-transcriptionally, the order by which introns are removed does not necessarily follow the order in which they are transcribed. Whereas several genomic features are known to influence whether or not an intron is spliced before its downstream neighbor, multiple questions related to adjacent introns' splicing order (AISO) remain unanswered. Here, we present Insplico, the first standalone software for quantifying AISO that works with both short and long read sequencing technologies. We first demonstrate its applicability and effectiveness using simulated reads and by recapitulating previously reported AISO patterns, which unveiled overlooked biases associated with long read sequencing. We next show that AISO around individual exons is remarkably constant across cell and tissue types and even upon major spliceosomal disruption, and it is evolutionarily conserved between human and mouse brains. We also establish a set of universal features associated with AISO patterns across various animal and plant species. Finally, we used Insplico to investigate AISO in the context of tissue-specific exons, particularly focusing on SRRM4-dependent microexons. We found that the majority of such microexons have non-canonical AISO, in which the downstream intron is spliced first, and we suggest two potential modes of SRRM4 regulation of microexons related to their AISO and various splicing-related features. Insplico is available on gitlab.com/aghr/insplico.


Subject(s)
Genome , RNA Splicing , Animals , Mice , Humans , Introns/genetics , RNA-Seq , RNA Splicing/genetics , Spliceosomes/genetics , Alternative Splicing , Nerve Tissue Proteins/genetics
3.
Sci Adv ; 8(4): eabk0445, 2022 01 28.
Article in English | MEDLINE | ID: mdl-35089784

ABSTRACT

Alternative splicing increases neuronal transcriptomic complexity throughout animal phylogeny. To delve into the mechanisms controlling the assembly and evolution of this regulatory layer, we characterized the neuronal microexon program in Drosophila and compared it with that of mammals. In nonvertebrate bilaterians, this splicing program is restricted to neurons by the posttranscriptional processing of the enhancer of microexons (eMIC) domain in Srrm234. In Drosophila, this processing is dependent on regulation by Elav/Fne. eMIC deficiency or misexpression leads to widespread neurological alterations largely emerging from impaired neuronal activity, as revealed by a combination of neuronal imaging experiments and cell type-specific rescues. These defects are associated with the genome-wide skipping of short neural exons, which are strongly enriched in ion channels. We found no overlap of eMIC-regulated exons between flies and mice, illustrating how ancient posttranscriptional programs can evolve independently in different phyla to affect distinct cellular modules while maintaining cell-type specificity.


Subject(s)
Drosophila Proteins , RNA Splicing , Alternative Splicing , Animals , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Mammals/genetics , Mammals/metabolism , Mice , Nerve Tissue Proteins/genetics , Neurons/metabolism , RNA-Binding Proteins
4.
PLoS Biol ; 19(2): e3001138, 2021 02.
Article in English | MEDLINE | ID: mdl-33621242

ABSTRACT

RNA splicing is widely dysregulated in cancer, frequently due to altered expression or activity of splicing factors (SFs). Microexons are extremely small exons (3-27 nucleotides long) that are highly evolutionarily conserved and play critical roles in promoting neuronal differentiation and development. Inclusion of microexons in mRNA transcripts is mediated by the SF Serine/Arginine Repetitive Matrix 4 (SRRM4), whose expression is largely restricted to neural tissues. However, microexons have been largely overlooked in prior analyses of splicing in cancer, as their small size necessitates specialized computational approaches for their detection. Here, we demonstrate that despite having low expression in normal nonneural tissues, SRRM4 is further silenced in tumors, resulting in the suppression of normal microexon inclusion. Remarkably, SRRM4 is the most consistently silenced SF across all tumor types analyzed, implying a general advantage of microexon down-regulation in cancer independent of its tissue of origin. We show that this silencing is favorable for tumor growth, as decreased SRRM4 expression in tumors is correlated with an increase in mitotic gene expression, and up-regulation of SRRM4 in cancer cell lines dose-dependently inhibits proliferation in vitro and in a mouse xenograft model. Further, this proliferation inhibition is accompanied by induction of neural-like expression and splicing patterns in cancer cells, suggesting that SRRM4 expression shifts the cell state away from proliferation and toward differentiation. We therefore conclude that SRRM4 acts as a proliferation brake, and tumors gain a selective advantage by cutting off this brake.


Subject(s)
Exons/physiology , Neoplasms/metabolism , Nerve Tissue Proteins/metabolism , RNA Splicing , Alternative Splicing , Animals , Cell Line , Female , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Male , Mice , Neoplasms/genetics , Nerve Tissue Proteins/genetics
5.
Mol Biol Cell ; 30(21): 2695-2708, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31483748

ABSTRACT

Although the function of microtubules (MTs) in chromosomal segregation during mitosis is well characterized, much less is known about the role of MTs in chromosomal functions during interphase. In the fission yeast Schizosaccharomyces pombe, dynamic cytoplasmic MT bundles move chromosomes in an oscillatory manner during interphase via linkages through the nuclear envelope (NE) at the spindle pole body (SPB) and other sites. Mto1 is a cytoplasmic factor that mediates the nucleation and attachment of cytoplasmic MTs to the nucleus. Here, we test the function of these cytoplasmic MTs and Mto1 on DNA repair and recombination during interphase. We find that mto1Δ cells exhibit defects in DNA repair and homologous recombination (HR) and abnormal DNA repair factory dynamics. In these cells, sister chromatids are not properly paired, and binding of Rad21 cohesin subunit along chromosomal arms is reduced. Our findings suggest a model in which cytoplasmic MTs and Mto1 facilitate efficient DNA repair and HR by promoting dynamic chromosomal organization and cohesion in the nucleus.


Subject(s)
Carrier Proteins/metabolism , Chromatids/metabolism , Chromosome Segregation , DNA Repair , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/metabolism , Sister Chromatid Exchange , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Nucleus/metabolism , Homologous Recombination , Interphase/genetics , Microtubules/metabolism , Nuclear Envelope/metabolism , Nuclear Proteins/metabolism , Schizosaccharomyces/genetics , Schizosaccharomyces pombe Proteins/genetics , Spindle Apparatus/metabolism , Spindle Pole Bodies/metabolism
6.
RNA ; 25(10): 1353-1365, 2019 10.
Article in English | MEDLINE | ID: mdl-31292260

ABSTRACT

Alternative splicing of pre-mRNA is a major mechanism to diversify protein functionality in metazoans from a limited number of genes. The Drosophila melanogaster Down syndrome cell adhesion molecule (Dscam) gene, which is important for neuronal wiring and phagocytosis of bacteria, can generate up to 38,016 isoforms by mutually exclusive alternative splicing in four clusters of variable exons. However, it is not understood how a specific exon is chosen from the many variables and how variable exons are prevented from being spliced together. A main role in the regulation of Dscam alternative splicing has been attributed to RNA binding proteins (RBPs), but how they impact on exon selection is not well understood. Serine-arginine rich (SR) proteins and hnRNP proteins are the two main types of RBPs with major roles in exon definition and splice site selection. Here, we analyzed the role of SR and hnRNP proteins in Dscam exon 9 alternative splicing in mutant Drosophila melanogaster embryos because of their essential function for development. Strikingly, loss or overexpression of canonical SR and hnRNP proteins even when multiple proteins are depleted together, does not affect Dscam alternative exon selection very dramatically. Conversely, noncanonical SR protein Serine-arginine repetitive matrix 2/3/4 (Srrm234) is a main determinant of exon inclusion in the Dscam exon 9 cluster. Since long-range base-pairings are absent in the exon 9 cluster, our data argue for a small complement of regulatory factors as main determinants of exon inclusion in the Dscam exon 9 cluster.


Subject(s)
Cell Adhesion Molecules/metabolism , Drosophila Proteins/metabolism , Exons , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , RNA-Binding Proteins/metabolism , Alternative Splicing , Animals , Drosophila melanogaster
7.
Nat Ecol Evol ; 3(4): 691-701, 2019 04.
Article in English | MEDLINE | ID: mdl-30833759

ABSTRACT

The mechanisms by which entire programmes of gene regulation emerged during evolution are poorly understood. Neuronal microexons represent the most conserved class of alternative splicing in vertebrates, and are critical for proper brain development and function. Here, we discover neural microexon programmes in non-vertebrate species and trace their origin to bilaterian ancestors through the emergence of a previously uncharacterized 'enhancer of microexons' (eMIC) protein domain. The eMIC domain originated as an alternative, neural-enriched splice isoform of the pan-eukaryotic Srrm2/SRm300 splicing factor gene, and subsequently became fixed in the vertebrate and neuronal-specific splicing regulator Srrm4/nSR100 and its paralogue Srrm3. Remarkably, the eMIC domain is necessary and sufficient for microexon splicing, and functions by interacting with the earliest components required for exon recognition. The emergence of a novel domain with restricted expression in the nervous system thus resulted in the evolution of splicing programmes that qualitatively expanded the neuronal molecular complexity in bilaterians.


Subject(s)
Exons , Neurons , RNA Splicing Factors/genetics , Alternative Splicing , Animals , Arthropods , Drosophila melanogaster , Evolution, Molecular , Humans , Lancelets , Mice , Protein Domains , Zebrafish
8.
ACS Synth Biol ; 7(4): 1056-1066, 2018 04 20.
Article in English | MEDLINE | ID: mdl-29544054

ABSTRACT

Cyanobacteria are important for fundamental studies of photosynthesis and have great biotechnological potential. In order to better study and fully exploit these organisms, the limited repertoire of genetic tools and parts must be expanded. A small number of inducible promoters have been used in cyanobacteria, allowing dynamic external control of gene expression through the addition of specific inducer molecules. However, the inducible promoters used to date suffer from various drawbacks including toxicity of inducers, leaky expression in the absence of inducer and inducer photolability, the latter being particularly relevant to cyanobacteria, which, as photoautotrophs, are grown under light. Here we introduce the rhamnose-inducible rhaBAD promoter of Escherichia coli into the model freshwater cyanobacterium Synechocystis sp. PCC 6803 and demonstrate it has superior properties to previously reported cyanobacterial inducible promoter systems, such as a non-toxic, photostable, non-metabolizable inducer, a linear response to inducer concentration and crucially no basal transcription in the absence of inducer.


Subject(s)
Escherichia coli/genetics , Gene Expression Regulation, Bacterial , Promoter Regions, Genetic , Rhamnose/metabolism , Synechocystis/genetics , Binding Sites , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Genes, Reporter , Light , Microorganisms, Genetically-Modified , Plasmids , Rhamnose/pharmacology , Sigma Factor/genetics , Sigma Factor/metabolism , Synechocystis/drug effects , Synechocystis/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
9.
Nat Commun ; 8(1): 1799, 2017 11 27.
Article in English | MEDLINE | ID: mdl-29180615

ABSTRACT

Epithelial-mesenchymal interactions are crucial for the development of numerous animal structures. Thus, unraveling how molecular tools are recruited in different lineages to control interplays between these tissues is key to understanding morphogenetic evolution. Here, we study Esrp genes, which regulate extensive splicing programs and are essential for mammalian organogenesis. We find that Esrp homologs have been independently recruited for the development of multiple structures across deuterostomes. Although Esrp is involved in a wide variety of ontogenetic processes, our results suggest ancient roles in non-neural ectoderm and regulating specific mesenchymal-to-epithelial transitions in deuterostome ancestors. However, consistent with the extensive rewiring of Esrp-dependent splicing programs between phyla, most developmental defects observed in vertebrate mutants are related to other types of morphogenetic processes. This is likely connected to the origin of an event in Fgfr, which was recruited as an Esrp target in stem chordates and subsequently co-opted into the development of many novel traits in vertebrates.


Subject(s)
Embryonic Development/genetics , Epithelial-Mesenchymal Transition/physiology , RNA Splicing/physiology , RNA-Binding Proteins/physiology , Animals , Biological Evolution , CRISPR-Cas Systems , Exons/physiology , Female , Gene Expression Regulation, Developmental/physiology , Gene Knockdown Techniques , Lancelets , Male , Mutation , RNA-Binding Proteins/genetics , Sequence Homology, Amino Acid , Signal Transduction/genetics , Strongylocentrotus purpuratus , Urochordata , Zebrafish
10.
Genetics ; 206(4): 1763-1778, 2017 08.
Article in English | MEDLINE | ID: mdl-28646043

ABSTRACT

Site-specific recombinases are potent tools to regulate gene expression. In particular, the Cre (cyclization recombination) and FLP (flipase) enzymes are widely used to either activate or inactivate genes in a precise spatiotemporal manner. Both recombinases work efficiently in the popular model organism Caenorhabditis elegans, but their use in this nematode is still only sporadic. To increase the utility of the FLP system in C. elegans, we have generated a series of single-copy transgenic strains that stably express an optimized version of FLP in specific tissues or by heat induction. We show that recombination efficiencies reach 100% in several cell types, such as muscles, intestine, and serotonin-producing neurons. Moreover, we demonstrate that most promoters drive recombination exclusively in the expected tissues. As examples of the potentials of the FLP lines, we describe novel tools for induced cell ablation by expression of the PEEL-1 toxin and a versatile FLP-out cassette for generation of GFP-tagged conditional knockout alleles. Together with other recombinase-based reagents created by the C. elegans community, this toolkit increases the possibilities for detailed analyses of specific biological processes at developmental stages inside intact animals.


Subject(s)
Caenorhabditis elegans/genetics , Gene Expression Regulation, Developmental , Genetic Engineering/methods , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , DNA Nucleotidyltransferases/genetics , DNA Nucleotidyltransferases/metabolism , Genes, Reporter , Integrases/genetics , Integrases/metabolism , Intestinal Mucosa/metabolism , Muscle, Skeletal/metabolism , Neurons/metabolism , Recombination, Genetic , Transgenes
11.
Cell Rep ; 18(12): 2868-2879, 2017 03 21.
Article in English | MEDLINE | ID: mdl-28329680

ABSTRACT

We have previously found that UV irradiation promotes RNA polymerase II (RNAPII) hyperphosphorylation and subsequent changes in alternative splicing (AS). We show now that UV-induced DNA damage is not only necessary but sufficient to trigger the AS response and that photolyase-mediated removal of the most abundant class of pyrimidine dimers (PDs) abrogates the global response to UV. We demonstrate that, in keratinocytes, RNAPII is the target, but not a sensor, of the signaling cascade initiated by PDs. The UV effect is enhanced by inhibition of gap-filling DNA synthesis, the last step in the nucleotide excision repair pathway (NER), and reduced by the absence of XPE, the main NER sensor of PDs. The mechanism involves activation of the protein kinase ATR that mediates the UV-induced RNAPII hyperphosphorylation. Our results define the sequence UV-PDs-NER-ATR-RNAPII-AS as a pathway linking DNA damage repair to the control of both RNAPII phosphorylation and AS regulation.


Subject(s)
Alternative Splicing/genetics , DNA Repair , Pyrimidine Dimers/metabolism , Ultraviolet Rays , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , DNA/metabolism , DNA Repair/genetics , Humans , Keratinocytes/metabolism , Keratinocytes/radiation effects , Phosphorylation/radiation effects , RNA Polymerase II/metabolism , Skin/cytology , Skin/radiation effects , Transcription, Genetic/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL
...