Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Mol Cell ; 80(1): 140-155.e6, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-33007254

RESUMO

The tissue-specific deployment of highly extended neural 3' UTR isoforms, generated by alternative polyadenylation (APA), is a broad and conserved feature of metazoan genomes. However, the factors and mechanisms that control neural APA isoforms are not well understood. Here, we show that three ELAV/Hu RNA binding proteins (Elav, Rbp9, and Fne) have similar capacities to induce a lengthened 3' UTR landscape in an ectopic setting. These factors promote accumulation of chromatin-associated, 3' UTR-extended, nascent transcripts, through inhibition of proximal polyadenylation site (PAS) usage. Notably, Elav represses an unannotated splice isoform of fne, switching the normally cytoplasmic Fne toward the nucleus in elav mutants. We use genomic profiling to reveal strong and broad loss of neural APA in elav/fne double mutant CNS, the first genetic background to largely abrogate this distinct APA signature. Overall, we demonstrate how regulatory interplay and functionally overlapping activities of neural ELAV/Hu RBPs drives the neural APA landscape.


Assuntos
Regiões 3' não Traduzidas/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Proteínas ELAV/metabolismo , Neurônios/metabolismo , Processamento Alternativo/genética , Motivos de Aminoácidos , Animais , Linhagem Celular , Núcleo Celular/metabolismo , Proteínas ELAV/química , Larva/metabolismo , Mutação/genética , Poli A/metabolismo , Isoformas de Proteínas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
2.
Genes Dev ; 32(5-6): 415-429, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29535189

RESUMO

N6-methyladenosine (m6A) is the most abundant mRNA modification in eukaryotes, playing crucial roles in multiple biological processes. m6A is catalyzed by the activity of methyltransferase-like 3 (Mettl3), which depends on additional proteins whose precise functions remain poorly understood. Here we identified Zc3h13 (zinc finger CCCH domain-containing protein 13)/Flacc [Fl(2)d-associated complex component] as a novel interactor of m6A methyltransferase complex components in Drosophila and mice. Like other components of this complex, Flacc controls m6A levels and is involved in sex determination in Drosophila We demonstrate that Flacc promotes m6A deposition by bridging Fl(2)d to the mRNA-binding factor Nito. Altogether, our work advances the molecular understanding of conservation and regulation of the m6A machinery.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Ligação a DNA/metabolismo , Drosophila melanogaster/fisiologia , Metiltransferases/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Ligação a RNA/metabolismo , Adenosina/metabolismo , Animais , Proteínas de Ciclo Celular , Linhagem Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/enzimologia , Regulação da Expressão Gênica no Desenvolvimento , Metilação , Camundongos , Células-Tronco Embrionárias Murinas , Transporte Proteico , Precursores de RNA/genética , Splicing de RNA , Fatores de Processamento de RNA , Processos de Determinação Sexual/genética
3.
Bioessays ; 45(3): e2200198, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36529693

RESUMO

Animal, protist and viral messenger RNAs (mRNAs) are most prominently modified at the beginning by methylation of cap-adjacent nucleotides at the 2'-O-position of the ribose (cOMe) by dedicated cap methyltransferases (CMTrs). If the first nucleotide of an mRNA is an adenosine, PCIF1 can methylate at the N6 -position (m6 A), while internally the Mettl3/14 writer complex can methylate. These modifications are introduced co-transcriptionally to affect many aspects of gene expression including localisation to synapses and local translation. Of particular interest, transcription start sites of many genes are heterogeneous leading to sequence diversity at the beginning of mRNAs, which together with cOMe and m6 Am could constitute an extensive novel layer of gene expression control. Given the role of cOMe and m6 A in local gene expression at synapses and higher brain functions including learning and memory, such code could be implemented at the transcriptional level for lasting memories through local gene expression at synapses.


Assuntos
Metiltransferases , Nucleotídeos , Animais , RNA Mensageiro/metabolismo , Metiltransferases/genética , Metilação , Nucleotídeos/genética , Nucleotídeos/metabolismo , Adenosina/genética , Eucariotos/genética
4.
RNA ; 28(10): 1377-1390, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35970556

RESUMO

Cap methyltransferases (CMTrs) O methylate the 2' position of the ribose (cOMe) of cap-adjacent nucleotides of animal, protist, and viral mRNAs. Animals generally have two CMTrs, whereas trypanosomes have three, and many viruses encode one in their genome. In the splice leader of mRNAs in trypanosomes, the first four nucleotides contain cOMe, but little is known about the status of cOMe in animals. Here, we show that cOMe is prominently present on the first two cap-adjacent nucleotides with species- and tissue-specific variations in Caenorhabditis elegans, honeybees, zebrafish, mouse, and human cell lines. In contrast, Drosophila contains cOMe primarily on the first cap-adjacent nucleotide. De novo RoseTTA modeling of CMTrs reveals close similarities of the overall structure and near identity for the catalytic tetrad, and for cap and cofactor binding for human, Drosophila and C. elegans CMTrs. Although viral CMTrs maintain the overall structure and catalytic tetrad, they have diverged in cap and cofactor binding. Consistent with the structural similarity, both CMTrs from Drosophila and humans methylate the first cap-adjacent nucleotide of an AGU consensus start. Because the second nucleotide is also methylated upon heat stress in Drosophila, these findings argue for regulated cOMe important for gene expression regulation.


Assuntos
Capuzes de RNA , Ribose , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Drosophila/genética , Drosophila/metabolismo , Humanos , Metilação , Metiltransferases/metabolismo , Camundongos , Nucleotídeos/genética , Nucleotídeos/metabolismo , Capuzes de RNA/química , RNA Mensageiro/genética , Ribose/metabolismo , Peixe-Zebra/genética
5.
Nature ; 540(7632): 301-304, 2016 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-27919081

RESUMO

N6-methyladenosine (m6A) is the most common internal modification of eukaryotic messenger RNA (mRNA) and is decoded by YTH domain proteins. The mammalian mRNA m6A methylosome is a complex of nuclear proteins that includes METTL3 (methyltransferase-like 3), METTL14, WTAP (Wilms tumour 1-associated protein) and KIAA1429. Drosophila has corresponding homologues named Ime4 and KAR4 (Inducer of meiosis 4 and Karyogamy protein 4), and Female-lethal (2)d (Fl(2)d) and Virilizer (Vir). In Drosophila, fl(2)d and vir are required for sex-dependent regulation of alternative splicing of the sex determination factor Sex lethal (Sxl). However, the functions of m6A in introns in the regulation of alternative splicing remain uncertain. Here we show that m6A is absent in the mRNA of Drosophila lacking Ime4. In contrast to mouse and plant knockout models, Drosophila Ime4-null mutants remain viable, though flightless, and show a sex bias towards maleness. This is because m6A is required for female-specific alternative splicing of Sxl, which determines female physiognomy, but also translationally represses male-specific lethal 2 (msl-2) to prevent dosage compensation in females. We further show that the m6A reader protein YT521-B decodes m6A in the sex-specifically spliced intron of Sxl, as its absence phenocopies Ime4 mutants. Loss of m6A also affects alternative splicing of additional genes, predominantly in the 5' untranslated region, and has global effects on the expression of metabolic genes. The requirement of m6A and its reader YT521-B for female-specific Sxl alternative splicing reveals that this hitherto enigmatic mRNA modification constitutes an ancient and specific mechanism to adjust levels of gene expression.


Assuntos
Adenosina/análogos & derivados , Processamento Alternativo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Precursores de RNA/metabolismo , Proteínas de Ligação a RNA/genética , Caracteres Sexuais , Processos de Determinação Sexual/genética , Regiões 5' não Traduzidas/genética , Adenosina/metabolismo , Animais , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Mecanismo Genético de Compensação de Dose , Proteínas de Drosophila/biossíntese , Proteínas de Drosophila/deficiência , Proteínas de Drosophila/metabolismo , Feminino , Íntrons/genética , Masculino , Metiltransferases/deficiência , Metiltransferases/genética , Metiltransferases/metabolismo , Neurônios/metabolismo , Proteínas Nucleares/biossíntese , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Precursores de RNA/química , Precursores de RNA/genética , RNA Mensageiro/química , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição/biossíntese , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Transcrição Gênica
6.
BMC Biol ; 19(1): 226, 2021 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-34666772

RESUMO

BACKGROUND: Female reproductive behaviors and physiology change profoundly after mating. The control of pregnancy-associated changes in physiology and behaviors are largely hard-wired into the brain to guarantee reproductive success, yet the gene expression programs that direct neuronal differentiation and circuit wiring at the end of the sex determination pathway in response to mating are largely unknown. In Drosophila, the post-mating response induced by male-derived sex-peptide in females is a well-established model to elucidate how complex innate behaviors are hard-wired into the brain. Here, we use a genetic approach to further characterize the molecular and cellular architecture of the sex-peptide response in Drosophila females. RESULTS: Screening for mutations that affect the sensitivity to sex-peptide, we identified the channel nuclear pore protein Nup54 gene as an essential component for mediating the sex-peptide response, with viable mutant alleles leading to the inability of laying eggs and reducing receptivity upon sex-peptide exposure. Nup54 directs correct wiring of eight adult brain neurons that express pickpocket and are required for egg-laying, while additional channel Nups also mediate sexual differentiation. Consistent with links of Nups to speciation, the Nup54 promoter is a hot spot for rapid evolution and promoter variants alter nucleo-cytoplasmic shuttling. CONCLUSIONS: These results implicate nuclear pore functionality to neuronal wiring underlying the sex-peptide response and sexual differentiation as a response to sexual conflict arising from male-derived sex-peptide to direct the female post-mating response.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Drosophila/genética , Proteínas de Drosophila/genética , Feminino , Masculino , Neurônios , Poro Nuclear , Peptídeos , Diferenciação Sexual/genética , Comportamento Sexual Animal
7.
RNA ; 25(10): 1353-1365, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31292260

RESUMO

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.


Assuntos
Moléculas de Adesão Celular/metabolismo , Proteínas de Drosophila/metabolismo , Éxons , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Proteínas de Ligação a RNA/metabolismo , Processamento Alternativo , Animais , Drosophila melanogaster
8.
Nucleic Acids Res ; 47(3): 1389-1403, 2019 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-30541104

RESUMO

Alternative splicing is a key feature of human genes, yet studying its regulation is often complicated by large introns. The Down Syndrome Cell Adhesion Molecule (Dscam) gene from Drosophila is one of the most complex genes generating vast molecular diversity by mutually exclusive alternative splicing. To resolve how alternative splicing in Dscam is regulated, we first developed plasmid-based UAS reporter genes for the Dscam variable exon 4 cluster and show that its alternative splicing is recapitulated by GAL4-mediated expression in neurons. We then developed gap-repair recombineering to very efficiently manipulate these large reporter plasmids in Escherichia coli using restriction enzymes or sgRNA/Cas9 DNA scission to capitalize on the many benefits of plasmids in phiC31 integrase-mediated transgenesis. Using these novel tools, we show that inclusion of Dscam exon 4 variables differs little in development and individual flies, and is robustly determined by sequences harbored in variable exons. We further show that introns drive selection of both proximal and distal variable exons. Since exon 4 cluster introns lack conserved sequences that could mediate robust long-range base-pairing to bring exons into proximity for splicing, our data argue for a central role of introns in mutually exclusive alternative splicing of Dscam exon 4 cluster.


Assuntos
Processamento Alternativo/genética , Moléculas de Adesão Celular/genética , Proteínas de Drosophila/genética , Fatores de Transcrição/genética , Animais , Sequência Conservada , Síndrome de Down/genética , Drosophila melanogaster/genética , Escherichia coli/genética , Éxons/genética , Expressão Gênica/genética , Técnicas de Transferência de Genes , Humanos , Íntrons/genética , Neurônios/metabolismo , Neurônios/patologia , Splicing de RNA/genética
9.
Trends Genet ; 33(6): 380-390, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28499622

RESUMO

Modifications in mRNA constitute ancient mechanisms to regulate gene expression post-transcriptionally. N6-methyladenosine (m6A) is the most prominent mRNA modification, and is installed by a large methyltransferase complex (the m6A 'writer'), not only specifically bound by RNA-binding proteins (the m6A 'readers'), but also removed by demethylases (the m6A 'erasers'). m6A mRNA modifications have been linked to regulation at multiple steps in mRNA processing. In analogy to the regulation of gene expression by miRNAs, we propose that the main function of m6A is post-transcriptional fine-tuning of gene expression. In contrast to miRNA regulation, which mostly reduces gene expression, we argue that m6A provides a fast mean to post-transcriptionally maximize gene expression. Additionally, m6A appears to have a second function during developmental transitions by targeting m6A-marked transcripts for degradation.


Assuntos
Metilação de DNA/genética , Epigênese Genética , Regulação da Expressão Gênica/genética , RNA Mensageiro/genética , Adenosina/análogos & derivados , Adenosina/genética , Adenosina/metabolismo , Humanos , Processamento Pós-Transcricional do RNA/genética , Proteínas de Ligação a RNA/genética
10.
Biochemistry ; 58(5): 363-378, 2019 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-30557013

RESUMO

The central dogma of molecular biology introduced by Crick describes a linear flow of information from DNA to mRNA to protein. Since then it has become evident that RNA undergoes several maturation steps such as capping, splicing, 3'-end processing, and editing. Likewise, nucleotide modifications are common in mRNA and are present in all organisms impacting on the regulation of gene expression. The most abundant modification found in mRNA is N6-methyladenosine (m6A). Deposition of m6A is a nuclear process and is performed by a megadalton writer complex primarily on mRNAs, but also on microRNAs and lncRNAs. The m6A methylosome is composed of the enzymatic core components METTL3 and METTL14, and several auxiliary proteins necessary for its correct positioning and functioning, which are WTAP, VIRMA, FLACC, RBM15, and HAKAI. The m6A epimark is decoded by YTH domain-containing reader proteins YTHDC and YTHDF, but METTLs can act as "readers" as well. Eraser proteins, such as FTO and ALKBH5, can remove the methyl group. Here we review recent progress on the role of m6A in regulating gene expression in light of Crick's central dogma of molecular biology. In particular, we address the complexity of the writer complex from an evolutionary perspective to obtain insights into the mechanism of ancient m6A methylation and its regulation.


Assuntos
Adenosina/análogos & derivados , Regulação da Expressão Gênica , Metiltransferases/metabolismo , Processamento Pós-Transcricional do RNA , RNA Mensageiro/química , Adenosina/química , Adenosina/genética , Humanos , Metilação , RNA Mensageiro/genética
11.
Brain ; 141(10): 2908-2924, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30239641

RESUMO

Accumulation and aggregation of TDP-43 is a major pathological hallmark of amyotrophic lateral sclerosis and frontotemporal dementia. TDP-43 inclusions also characterize patients with GGGGCC (G4C2) hexanucleotide repeat expansion in C9orf72 that causes the most common genetic form of amyotrophic lateral sclerosis and frontotemporal dementia (C9ALS/FTD). Functional studies in cell and animal models have identified pathogenic mechanisms including repeat-induced RNA toxicity and accumulation of G4C2-derived dipeptide-repeat proteins. The role of TDP-43 dysfunction in C9ALS/FTD, however, remains elusive. We found G4C2-derived dipeptide-repeat protein but not G4C2-RNA accumulation caused TDP-43 proteinopathy that triggered onset and progression of disease in Drosophila models of C9ALS/FTD. Timing and extent of TDP-43 dysfunction was dependent on levels and identity of dipeptide-repeat proteins produced, with poly-GR causing early and poly-GA/poly-GP causing late onset of disease. Accumulating cytosolic, but not insoluble aggregated TDP-43 caused karyopherin-α2/4 (KPNA2/4) pathology, increased levels of dipeptide-repeat proteins and enhanced G4C2-related toxicity. Comparable KPNA4 pathology was observed in both sporadic frontotemporal dementia and C9ALS/FTD patient brains characterized by its nuclear depletion and cytosolic accumulation, irrespective of TDP-43 or dipeptide-repeat protein aggregates. These findings identify a vicious feedback cycle for dipeptide-repeat protein-mediated TDP-43 and subsequent KPNA pathology, which becomes self-sufficient of the initiating trigger and causes C9-related neurodegeneration.


Assuntos
Esclerose Lateral Amiotrófica/patologia , Proteína C9orf72/metabolismo , Proteínas de Ligação a DNA/metabolismo , Demência Frontotemporal/patologia , Degeneração Neural/metabolismo , alfa Carioferinas/metabolismo , Esclerose Lateral Amiotrófica/metabolismo , Animais , Expansão das Repetições de DNA , Drosophila , Proteínas de Drosophila/metabolismo , Retroalimentação Fisiológica , Demência Frontotemporal/metabolismo , Humanos , Degeneração Neural/patologia
12.
RNA Biol ; 14(9): 1232-1240, 2017 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-28353398

RESUMO

RNA modifications are an emerging layer of posttranscriptional gene regulation in eukaryotes. N6-methyladenosine (m6A) is among the most abundant modifications in mRNAs (mRNAs) that was shown to influence many physiological processes from yeast to mammals. Like DNA methylation, m6A in mRNA is dynamically regulated. A conserved methyltransferase complex catalyzes the deposition of the methyl group on adenosine, which can be removed by specific classes of demethylases. Furthermore, YTH-domain containing proteins can recognize this modification to mediate m6A-dependent activities. Here we review the functions and mechanisms of the main m6A players with a particular focus on Drosophila melanogaster.


Assuntos
Adenosina/análogos & derivados , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Adenosina Trifosfatases/química , Adenosina Trifosfatases/metabolismo , Animais , Regulação da Expressão Gênica , Metilação , Splicing de RNA
13.
Biochem Soc Trans ; 44(5): 1385-1393, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27911721

RESUMO

Modified nucleotides in messenger RNA (mRNA) have been discovered over 40 years ago, but until recently little was known about which transcripts contain them and what their function is. High-throughput sequencing approaches revealed a dynamic landscape of the 'Epitranscriptome' for many mRNA modifications in various organisms from yeast to humans. Meanwhile, also many genes encoding mRNA-modifying enzymes and auxiliary proteins have been identified yielding functional insights by reverse genetics into their role in development and disease.


Assuntos
Doença/genética , Nucleotídeos/genética , Edição de RNA , RNA Mensageiro/genética , Animais , Arabidopsis/genética , Caenorhabditis elegans/genética , Drosophila melanogaster/genética , Predisposição Genética para Doença/genética , Humanos , Mutação , Saccharomyces cerevisiae/genética
14.
Biochem Soc Trans ; 42(4): 1147-51, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25110017

RESUMO

ELAV (embryonic lethal/abnormal visual system)/Hu proteins comprise a family of highly related neuronal RBPs (RNA-binding proteins) involved in many aspects of mRNA processing. Although they bind to highly similar short sequence motifs, they have acquired diverse functions suggesting that cellular signalling is important for their functional diversification. Indeed, ELAV/Hu proteins harbour many phosphorylatable amino acids. In the present article, we review our current knowledge about phosphorylation of ELAV/Hu proteins and how phosphorylation affects cellular localization of ELAV/Hu proteins and their binding to RNA.


Assuntos
Proteínas ELAV/metabolismo , Processamento Alternativo/fisiologia , Animais , Proteínas ELAV/genética , Humanos , Fosforilação
16.
Proc Biol Sci ; 280(1771): 20131938, 2013 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-24089336

RESUMO

Male-derived sex-peptide (SP) induces profound changes in the behaviour of Drosophila females, resulting in decreased receptivity to further mating and increased egg laying. SP can mediate the switch in female reproductive behaviours via a G protein-coupled receptor, SPR, in neurons expressing fruitless, doublesex and pickpocket. Whether SPR is the sole receptor and whether SP induces the postmating switch in a single pathway has not, to our knowledge been tested. Here we report that the SP response can be induced in the absence of SPR when SP is ectopically expressed in neurons or when SP, transferred by mating, can access neurons through a leaky blood brain barrier. Membrane-tethered SP can induce oviposition via doublesex, but not fruitless and pickpocket neurons in SPR mutant females. Although pickpocket and doublesex neurons rely on G(o) signalling to reduce receptivity and induce oviposition, G(o) signalling in fruitless neurons is required only to induce oviposition, but not to reduce receptivity. Our results show that SP's action in reducing receptivity and inducing oviposition can be separated in fruitless and doublesex neurons. Hence, the SP-induced postmating switch incorporates shared, but also distinct circuitry of fruitless, doublesex and pickpocket neurons and additional receptors.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/fisiologia , Neurônios/metabolismo , Oviposição/fisiologia , Peptídeos/metabolismo , Comportamento Sexual Animal/fisiologia , Transdução de Sinais/fisiologia , Animais , Cruzamentos Genéticos , Proteínas de Ligação a DNA/metabolismo , Doxiciclina , Drosophila/metabolismo , Feminino , Peptídeos e Proteínas de Sinalização Intercelular , Proteínas do Tecido Nervoso/metabolismo , Receptores de Peptídeos , Fatores Sexuais , Canais de Sódio/metabolismo , Fatores de Transcrição/metabolismo
17.
Sci Rep ; 13(1): 8035, 2023 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-37198214

RESUMO

Nuclear pore proteins (Nups) prominently are among the few genes linked to speciation from hybrid incompatibility in Drosophila. These studies have focused on coding sequence evolution of Nup96 and Nup160 and shown evidence of positive selection driving nucleoporin evolution. Intriguingly, channel Nup54 functionality is required for neuronal wiring underlying the female post-mating response induced by male-derived sex-peptide. A region of rapid evolution in the core promoter of Nup54 suggests a critical role for general transcriptional regulatory elements at the onset of speciation, but whether this is a general feature of Nup genes has not been determined. Consistent with findings for Nup54, additional channel Nup58 and Nup62 promoters also rapidly accumulate insertions/deletions (indels). Comprehensive examination of Nup upstream regions reveals that core Nup complex gene promoters accumulate indels rapidly. Since changes in promoters can drive changes in expression, these results indicate an evolutionary mechanism driven by indel accumulation in core Nup promoters. Compensation of such gene expression changes could lead to altered neuronal wiring, rapid fixation of traits caused by promoter changes and subsequently the rise of new species. Hence, the nuclear pore complex may act as a nexus for species-specific changes via nucleo-cytoplasmic transport regulated gene expression.


Assuntos
Complexo de Proteínas Formadoras de Poros Nucleares , Poro Nuclear , Animais , Masculino , Feminino , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Poro Nuclear/genética , Poro Nuclear/metabolismo , Transporte Ativo do Núcleo Celular/genética , Drosophila/genética , Drosophila/metabolismo , Mutação INDEL
18.
Front Mol Neurosci ; 16: 1322808, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38264345

RESUMO

Down syndrome cell adhesion molecule (Dscam) gene encodes a cell adhesion molecule required for neuronal wiring. A remarkable feature of arthropod Dscam is massive alternative splicing generating thousands of different isoforms from three variable clusters of alternative exons. Dscam expression and diversity arising from alternative splicing have been studied during development, but whether they exert functions in adult brains has not been determined. Here, using honey bees, we find that Dscam expression is critically linked to memory retention as reducing expression by RNAi enhances memory after reward learning in adult worker honey bees. Moreover, alternative splicing of Dscam is altered in all three variable clusters after learning. Since identical Dscam isoforms engage in homophilic interactions, these results suggest a mechanism to alter inclusion of variable exons during memory consolidation to modify neuronal connections for memory retention.

19.
Biochem Soc Trans ; 40(4): 804-9, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22817738

RESUMO

Alternative splicing of pre-mRNA is a major mechanism to increase protein diversity in higher eukaryotes. Dscam, the Drosophila homologue of human DSCAM (Down's syndrome cell adhesion molecule), generates up to 38016 isoforms through mutually exclusive splicing in four variable exon clusters. This enormous molecular diversity is functionally important for wiring of the nervous system and phagocytosis of invading pathogens. Current models explaining this complex splicing regulation include a default repressed state of the variable exon clusters to prevent the splicing together of adjacent exons, the presence of RNA secondary structures important for the release of one specific variable exon from the repressed state and combinatorial interaction of RNA-binding proteins for choosing a specific exon.


Assuntos
Moléculas de Adesão Celular/genética , Proteínas de Drosophila/genética , Proteínas de Ligação a RNA/genética , Processamento Alternativo/genética , Animais , Drosophila , Conformação de Ácido Nucleico
20.
Nat Commun ; 13(1): 1209, 2022 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-35260552

RESUMO

Cap-adjacent nucleotides of animal, protist and viral mRNAs can be O-methylated at the 2' position of the ribose (cOMe). The functions of cOMe in animals, however, remain largely unknown. Here we show that the two cap methyltransferases (CMTr1 and CMTr2) of Drosophila can methylate the ribose of the first nucleotide in mRNA. Double-mutant flies lack cOMe but are viable. Consistent with prominent neuronal expression, they have a reward learning defect that can be rescued by conditional expression in mushroom body neurons before training. Among CMTr targets are cell adhesion and signaling molecules. Many are relevant for learning, and are also targets of Fragile X Mental Retardation Protein (FMRP). Like FMRP, cOMe is required for localization of untranslated mRNAs to synapses and enhances binding of the cap binding complex in the nucleus. Hence, our study reveals a mechanism to co-transcriptionally prime mRNAs by cOMe for localized protein synthesis at synapses.


Assuntos
Síndrome do Cromossomo X Frágil , Metiltransferases , Animais , Proteína do X Frágil da Deficiência Intelectual/genética , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Síndrome do Cromossomo X Frágil/genética , Metiltransferases/genética , Metiltransferases/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Recompensa , Ribose/metabolismo , Sinapses/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA