Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Nucleic Acids Res ; 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-39217468

RESUMO

ADAR3 is a catalytically inactive member of the family of adenosine deaminases acting on RNA (ADARs). Here we have investigated its function in the context of the developing mouse brain. The expression of ADAR3 gradually increases throughout embryogenesis and drops after birth. Using primary cortical neurons, we show that ADAR3 is only expressed in a subpopulation of in vitro differentiated neurons, which suggests specific functions rather than being a general regulator of ADAR editing in the brain. The analysis of the ADAR3 interactome suggested a role in mRNA stability and translation, and we show that expression of ADAR3 in a neuronal cell line that is otherwise ADAR3-negative changes the expression and stability of a large number of mRNAs. Notably, we show that ADAR3 associates with polysomes and inhibits translation. We propose that ADAR3 binds to target mRNAs and stabilizes them in non-productive polysome complexes. Interestingly, the expression of ADAR3 downregulates genes related to neuronal differentiation and inhibits neurofilament outgrowth in vitro. In summary, we propose that ADAR3 negatively regulates neuronal differentiation, and that it does so by regulating mRNA stability and translation in an editing-independent manner.

2.
J Biol Chem ; 298(3): 101682, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35124003

RESUMO

miRNAs are short noncoding RNA molecules that regulate gene expression by inhibiting translation or inducing degradation of target mRNAs. miRNAs are often expressed as polycistronic transcripts, so-called miRNA clusters, containing several miRNA precursors. The largest mammalian miRNA cluster, the miR-379-410 cluster, is expressed primarily during embryonic development and in the adult brain; however, downstream regulation of this cluster is not well understood. Here, we investigated adenosine deamination to inosine (RNA editing) in the miR-379-410 cluster by adenosine deaminase acting on RNA (ADAR) enzymes as a possible mechanism modulating the expression and activity of these miRNAs in a brain-specific manner. We show that the levels of editing in the majority of mature miRNAs are lower than the editing levels of the corresponding site in primary miRNA precursors. However, for one miRNA, miR-376b-3p, editing was significantly higher in the mature form than in the primary precursor. We found miR-376b-3p maturation is negatively regulated by ADAR2 in an editing activity-independent manner, whereas ADAR1-mediated and ADAR2-mediated editing were observed to be competitive. In addition, the edited miR-376b-3p targets a different set of mRNAs than unedited miR-376b-3p, including 4-aminobutyrate aminotransferase, encoding the enzyme responsible for the catabolism of the neurotransmitter gamma aminobutyric acid (GABA). Expression of edited miR-376b-3p led to increased intracellular GABA levels as well as increased cell surface presentation of GABA type A receptors. Our results indicate that both editing and editing-independent effects modulate the expression of miR-376b-3p, with the potential to regulate GABAergic signaling in the brain.


Assuntos
MicroRNAs , Proteínas de Ligação a RNA , Ácido gama-Aminobutírico , Adenosina Desaminase/genética , Adenosina Desaminase/metabolismo , Animais , Mamíferos/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Neurotransmissores , Edição de RNA , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Ácido gama-Aminobutírico/metabolismo
3.
BMC Biol ; 18(1): 6, 2020 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-31937309

RESUMO

BACKGROUND: Adenosine-to-inosine (A-to-I) RNA editing is a process that contributes to the diversification of proteins that has been shown to be essential for neurotransmission and other neuronal functions. However, the spatiotemporal and diversification properties of RNA editing in the brain are largely unknown. Here, we applied in situ sequencing to distinguish between edited and unedited transcripts in distinct regions of the mouse brain at four developmental stages, and investigate the diversity of the RNA landscape. RESULTS: We analyzed RNA editing at codon-altering sites using in situ sequencing at single-cell resolution, in combination with the detection of individual ADAR enzymes and specific cell type marker transcripts. This approach revealed cell-type-specific regulation of RNA editing of a set of transcripts, and developmental and regional variation in editing levels for many of the targeted sites. We found increasing editing diversity throughout development, which arises through regional- and cell type-specific regulation of ADAR enzymes and target transcripts. CONCLUSIONS: Our single-cell in situ sequencing method has proved useful to study the complex landscape of RNA editing and our results indicate that this complexity arises due to distinct mechanisms of regulating individual RNA editing sites, acting both regionally and in specific cell types.


Assuntos
Encéfalo/metabolismo , Edição de RNA , Adenosina/metabolismo , Animais , Encéfalo/crescimento & desenvolvimento , Inosina/metabolismo , Camundongos , Análise de Sequência de RNA , Análise Espaço-Temporal
4.
Nucleic Acids Res ; 47(4): e22, 2019 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-30590609

RESUMO

Adenosine to inosine editing is common in the human transcriptome and changes of this essential activity is associated with disease. Children with ADAR1 mutations develop fatal Aicardi-Goutières syndrome characterized by aberrant interferon expression. In contrast, ADAR1 overexpression is associated with increased malignancy of breast, lung and liver cancer. ADAR1 silencing in breast cancer cells leads to increased apoptosis, suggesting an anti-apoptotic function that promotes cancer progression. Yet, suitable high-throughput editing assays are needed to efficiently screen chemical libraries for modifiers of ADAR1 activity. We describe the development of a bioluminescent reporter system that facilitates rapid and accurate determination of endogenous editing activity. The system is based on the highly sensitive and quantitative Nanoluciferase that is conditionally expressed upon reporter-transcript editing. Stably introduced into cancer cell lines, the system reports on elevated endogenous ADAR1 editing activity induced by interferon as well as knockdown of ADAR1 and ADAR2. In a single-well setup we used the reporter in HeLa cells to screen a small molecule library of 33 000 compounds. This yielded a primary hit rate of 0.9% at 70% inhibition of editing. Thus, we provide a key tool for high-throughput identification of modifiers of A-to-I editing activity in cancer cells.


Assuntos
Adenosina Desaminase/genética , Ensaios de Triagem em Larga Escala , Neoplasias/genética , Proteínas de Ligação a RNA/genética , Adenosina/genética , Apoptose/genética , Doenças Autoimunes do Sistema Nervoso/genética , Doenças Autoimunes do Sistema Nervoso/patologia , Edição de Genes/métodos , Genes Reporter/genética , Células HeLa , Humanos , Inosina/genética , Interferons/genética , Luciferases/genética , Medições Luminescentes/métodos , Nanopartículas/química , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Malformações do Sistema Nervoso/genética , Malformações do Sistema Nervoso/patologia , Bibliotecas de Moléculas Pequenas/química , Transcriptoma/genética
5.
Genes (Basel) ; 10(1)2018 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-30585209

RESUMO

It is well established that somatic mutations and escape of immune disruption are two essential factors in cancer initiation and progression. With an increasing number of second-generation sequencing data, transcriptomic modifications, so called RNA mutations, are emerging as significant forces that drive the transition from normal cell to malignant tumor, as well as providing tumor diversity to escape an immune attack. Editing of adenosine to inosine (A-to-I) in double-stranded RNA, catalyzed by adenosine deaminases acting on RNA (ADARs), is one dynamic modification that in a combinatorial manner can give rise to a very diverse transcriptome. Since the cell interprets inosine as guanosine (G), A-to-I editing can result in non-synonymous codon changes in transcripts as well as yield alternative splicing, but also affect targeting and disrupt maturation of microRNAs. ADAR-mediated RNA editing is essential for survival in mammals, however, its dysregulation causes aberrant editing of its targets that may lead to cancer. ADAR1 is commonly overexpressed, for instance in breast, lung, liver and esophageal cancer as well as in chronic myelogenous leukemia, where it promotes cancer progression. It is well known that ADAR1 regulates type I interferon (IFN) and its induced gene signature, which are known to operate as a significant barrier to tumor formation and progression. Adding to the complexity, ADAR1 expression is also regulated by IFN. In this review, we discussed the regulatory mechanisms of ADAR1 during tumorigenesis through aberrant editing of specific substrates. Additionally, we hypothesized that elevated ADAR1 levels play a role in suppressing an innate immunity response in cancer cells.

6.
RNA Biol ; 15(6): 829-831, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29671387

RESUMO

The genetic alphabet consists of the four letters: C, A, G, and T in DNA and C,A,G, and U in RNA. Triplets of these four letters jointly encode 20 different amino acids out of which proteins of all organisms are built. This system is universal and is found in all kingdoms of life. However, bases in DNA and RNA can be chemically modified. In DNA, around 10 different modifications are known, and those have been studied intensively over the past 20 years. Scientific studies on DNA modifications and proteins that recognize them gave rise to the large field of epigenetic and epigenomic research. The outcome of this intense research field is the discovery that development, ageing, and stem-cell dependent regeneration but also several diseases including cancer are largely controlled by the epigenetic state of cells. Consequently, this research has already led to the first FDA approved drugs that exploit the gained knowledge to combat disease. In recent years, the ~150 modifications found in RNA have come to the focus of intense research. Here we provide a perspective on necessary and expected developments in the fast expanding area of RNA modifications, termed epitranscriptomics.


Assuntos
DNA de Neoplasias , Epigênese Genética , Epigenômica/normas , Perfilação da Expressão Gênica/normas , Regulação Neoplásica da Expressão Gênica , Neoplasias , RNA Neoplásico , Transcriptoma , DNA de Neoplasias/genética , DNA de Neoplasias/metabolismo , Europa (Continente) , Perfilação da Expressão Gênica/métodos , Humanos , Neoplasias/genética , Neoplasias/metabolismo , RNA Neoplásico/genética , RNA Neoplásico/metabolismo
7.
Nat Struct Mol Biol ; 25(1): 83-89, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29323280

RESUMO

Protein quality control depends on the tight regulation of interactions between molecular chaperones and polypeptide substrates. Substrate release from the chaperone Hsp70 is triggered by nucleotide-exchange factors (NEFs) that control folding and degradation fates via poorly understood mechanisms. We found that the armadillo-type NEFs budding yeast Fes1 and its human homolog HspBP1 employ flexible N-terminal release domains (RDs) with substrate-mimicking properties to ensure the efficient release of persistent substrates from Hsp70. The RD contacts the substrate-binding domain of the chaperone, competes with peptide substrate for binding and is essential for proper function in yeast and mammalian cells. Thus, the armadillo domain engages Hsp70 to trigger nucleotide exchange, whereas the RD safeguards the release of substrates. Our findings provide fundamental mechanistic insight into the functional specialization of Hsp70 NEFs and have implications for the understanding of proteostasis-related disorders, including Marinesco-Sjögren syndrome.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas de Choque Térmico HSP70/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/química , Proteínas de Saccharomyces cerevisiae/química , Trifosfato de Adenosina/química , Proteínas de Ligação a DNA/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Chaperonas Moleculares/metabolismo , Nucleotídeos/metabolismo , Fenótipo , Desnaturação Proteica , Dobramento de Proteína , Saccharomyces cerevisiae/metabolismo , Fatores de Transcrição/metabolismo
8.
Semin Cell Dev Biol ; 79: 123-130, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29146145

RESUMO

Cancer arises when pathways that control cell functions such as proliferation and migration are dysregulated to such an extent that cells start to divide uncontrollably and eventually spread throughout the body, ultimately endangering the survival of an affected individual. It is well established that somatic mutations are important in cancer initiation and progression as well as in creation of tumor diversity. Now also modifications of the transcriptome are emerging as a significant force during the transition from normal cell to malignant tumor. Editing of adenosine (A) to inosine (I) in double-stranded RNA, catalyzed by adenosine deaminases acting on RNA (ADARs), is one dynamic modification that in a combinatorial manner can give rise to a very diverse transcriptome. Since the cell interprets inosine as guanosine (G), editing can result in non-synonymous codon changes in transcripts as well as yield alternative splicing, but also affect targeting and disrupt maturation of microRNA. ADAR editing is essential for survival in mammals but its dysregulation can lead to cancer. ADAR1 is for instance overexpressed in, e.g., lung cancer, liver cancer, esophageal cancer and chronic myoelogenous leukemia, which with few exceptions promotes cancer progression. In contrast, ADAR2 is lowly expressed in e.g. glioblastoma, where the lower levels of ADAR2 editing leads to malignant phenotypes. Altogether, RNA editing by the ADAR enzymes is a powerful regulatory mechanism during tumorigenesis. Depending on the cell type, cancer progression seems to mainly be induced by ADAR1 upregulation or ADAR2 downregulation, although in a few cases ADAR1 is instead downregulated. In this review, we discuss how aberrant editing of specific substrates contributes to malignancy.


Assuntos
Adenosina Desaminase/metabolismo , Neoplasias/genética , Edição de RNA , RNA de Cadeia Dupla/genética , Proteínas de Ligação a RNA/metabolismo , Animais , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias/metabolismo , Neoplasias/patologia , Isoformas de RNA/genética , Isoformas de RNA/metabolismo , RNA de Cadeia Dupla/metabolismo
9.
Genome Biol ; 18(1): 195, 2017 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-29061182

RESUMO

BACKGROUND: Adenosine to inosine (A-to-I) RNA editing has been shown to be an essential event that plays a significant role in neuronal function, as well as innate immunity, in mammals. It requires a structure that is largely double-stranded for catalysis but little is known about what determines editing efficiency and specificity in vivo. We have previously shown that some editing sites require adjacent long stem loop structures acting as editing inducer elements (EIEs) for efficient editing. RESULTS: The glutamate receptor subunit A2 is edited at the Q/R site in almost 100% of all transcripts. We show that efficient editing at the Q/R site requires an EIE in the downstream intron, separated by an internal loop. Also, other efficiently edited sites are flanked by conserved, highly structured EIEs and we propose that this is a general requisite for efficient editing, while sites with low levels of editing lack EIEs. This phenomenon is not limited to mRNA, as non-coding primary miRNAs also use EIEs to recruit ADAR to specific sites. CONCLUSIONS: We propose a model where two regions of dsRNA are required for efficient editing: first, an RNA stem that recruits ADAR and increases the local concentration of the enzyme, then a shorter, less stable duplex that is ideal for efficient and specific catalysis. This discovery changes the way we define and determine a substrate for A-to-I editing. This will be important in the discovery of novel editing sites, as well as explaining cases of altered editing in relation to disease.


Assuntos
Adenosina/metabolismo , Inosina/metabolismo , Edição de RNA , Sequências Reguladoras de Ácido Ribonucleico , Adenosina Desaminase/metabolismo , Animais , Células HEK293 , Células HeLa , Humanos , Canal de Potássio Kv1.1/genética , Canal de Potássio Kv1.1/metabolismo , Camundongos , MicroRNAs/química , MicroRNAs/metabolismo , Conformação de Ácido Nucleico , Receptores de AMPA/genética , Receptores de AMPA/metabolismo , Receptores de Ácido Caínico/genética , Receptores de Ácido Caínico/metabolismo , Transcriptoma , Receptor de GluK2 Cainato
10.
Nucleic Acids Res ; 45(7): 4189-4201, 2017 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-28053121

RESUMO

Adenosine deaminases acting on RNA (ADARs) catalyze the editing of adenosine residues to inosine (A-to-I) within RNA sequences, mostly in the introns and UTRs (un-translated regions). The significance of editing within non-coding regions of RNA is poorly understood. Here, we demonstrate that association of ADAR2 with RNA stabilizes a subset of transcripts. ADAR2 interacts with and edits the 3΄UTR of nuclear-retained Cat2 transcribed nuclear RNA (Ctn RNA). In absence of ADAR2, the abundance and half-life of Ctn RNA are significantly reduced. Furthermore, ADAR2-mediated stabilization of Ctn RNA occurred in an editing-independent manner. Unedited Ctn RNA shows enhanced interaction with the RNA-binding proteins HuR and PARN [Poly(A) specific ribonuclease deadenylase]. HuR and PARN destabilize Ctn RNA in absence of ADAR2, indicating that ADAR2 stabilizes Ctn RNA by antagonizing its degradation by PARN and HuR. Transcriptomic analysis identified other RNAs that are regulated by a similar mechanism. In summary, we identify a regulatory mechanism whereby ADAR2 enhances target RNA stability by limiting the interaction of RNA-destabilizing proteins with their cognate substrates.


Assuntos
Adenosina Desaminase/metabolismo , Proteína Semelhante a ELAV 1/metabolismo , Exorribonucleases/metabolismo , Estabilidade de RNA , Proteínas de Ligação a RNA/metabolismo , Regiões 3' não Traduzidas , Animais , Transportador 2 de Aminoácidos Catiônicos/genética , Transportador 2 de Aminoácidos Catiônicos/metabolismo , Camundongos , Edição de RNA , RNA Longo não Codificante/metabolismo
11.
J Cell Sci ; 130(4): 745-753, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-28082424

RESUMO

Adenosine to inosine (A-to-I) RNA editing is important for a functional brain, and most known sites that are subject to selective RNA editing have been found to result in diversified protein isoforms that are involved in neurotransmission. In the absence of the active editing enzymes ADAR1 or ADAR2 (also known as ADAR and ADARB1, respectively), mice fail to survive until adulthood. Nuclear A-to-I editing of neuronal transcripts is regulated during brain development, with low levels of editing in the embryo and a dramatic increase after birth. Yet, little is known about the mechanisms that regulate editing during development. Here, we demonstrate lower levels of ADAR2 in the nucleus of immature neurons than in mature neurons. We show that importin-α4 (encoded by Kpna3), which increases during neuronal maturation, interacts with ADAR2 and contributes to the editing efficiency by bringing it into the nucleus. Moreover, we detect an increased number of interactions between ADAR2 and the nuclear isomerase Pin1 as neurons mature, which contribute to ADAR2 protein stability. Together, these findings explain how the nuclear editing of substrates that are important for neuronal function can increase as the brain develops.


Assuntos
Adenosina Desaminase/metabolismo , Adenosina/metabolismo , Núcleo Celular/metabolismo , Inosina/metabolismo , Neurogênese/genética , Neurônios/metabolismo , Edição de RNA , Adenosina Desaminase/química , Animais , Diferenciação Celular/genética , Células Cultivadas , Córtex Cerebral/citologia , Células HEK293 , Humanos , Camundongos , Modelos Biológicos , Peptidilprolil Isomerase de Interação com NIMA/metabolismo , Sinais de Localização Nuclear/metabolismo , Ligação Proteica , Ratos , alfa Carioferinas
12.
Trends Genet ; 32(3): 165-175, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26803450

RESUMO

Post-transcriptional RNA modification by adenosine to inosine (A-to-I) editing expands the functional output of many important neuronally expressed genes. The mechanism provides flexibility in the proteome by expanding the variety of isoforms, and is a requisite for neuronal function. Indeed, targets for editing include key mediators of synaptic transmission with an overall significant effect on neuronal signaling. In addition, editing influences splice-site choice and miRNA targeting capacity, and thereby regulates neuronal gene expression. Editing efficiency at most of these sites increases during neuronal differentiation and brain maturation in a spatiotemporal manner. This editing-induced dynamics in the transcriptome is essential for normal brain development, and we are only beginning to understand its role in neuronal function. In this review we discuss the impact of RNA editing in the brain, with special emphasis on the physiological consequences for neuronal development and plasticity.


Assuntos
Encéfalo/citologia , Neurônios/citologia , Edição de RNA , Animais , Mamíferos , Splicing de RNA , Transcrição Gênica
13.
Biochimie ; 117: 22-7, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26051678

RESUMO

It has for a long time been known that repetitive elements, particularly Alu sequences in human, are edited by the adenosine deaminases acting on RNA, ADAR, family. The functional interpretation of these events has been even more difficult than that of editing events in coding sequences, but today there is an emerging understanding of their downstream effects. A surprisingly large fraction of the human transcriptome contains inverted Alu repeats, often forming long double stranded structures in RNA transcripts, typically occurring in introns and UTRs of protein coding genes. Alu repeats are also common in other primates, and similar inverted repeats can frequently be found in non-primates, although the latter are less prone to duplex formation. In human, as many as 700,000 Alu elements have been identified as substrates for RNA editing, of which many are edited at several sites. In fact, recent advancements in transcriptome sequencing techniques and bioinformatics have revealed that the human editome comprises at least a hundred million adenosine to inosine (A-to-I) editing sites in Alu sequences. Although substantial additional efforts are required in order to map the editome, already present knowledge provides an excellent starting point for studying cis-regulation of editing. In this review, we will focus on editing of long stem loop structures in the human transcriptome and how it can effect gene expression.


Assuntos
Elementos Alu/genética , Regulação da Expressão Gênica , Edição de RNA , RNA não Traduzido/genética , Transcriptoma/genética , Animais , Humanos , Íntrons/genética , Modelos Genéticos , Primatas
14.
Cell Rep ; 9(4): 1482-94, 2014 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-25456137

RESUMO

The ADAR RNA-editing enzymes deaminate adenosine bases to inosines in cellular RNAs. Aberrant interferon expression occurs in patients in whom ADAR1 mutations cause Aicardi-Goutières syndrome (AGS) or dystonia arising from striatal neurodegeneration. Adar1 mutant mouse embryos show aberrant interferon induction and die by embryonic day E12.5. We demonstrate that Adar1 embryonic lethality is rescued to live birth in Adar1; Mavs double mutants in which the antiviral interferon induction response to cytoplasmic double-stranded RNA (dsRNA) is prevented. Aberrant immune responses in Adar1 mutant mouse embryo fibroblasts are dramatically reduced by restoring the expression of editing-active cytoplasmic ADARs. We propose that inosine in cellular RNA inhibits antiviral inflammatory and interferon responses by altering RLR interactions. Transfecting dsRNA oligonucleotides containing inosine-uracil base pairs into Adar1 mutant mouse embryo fibroblasts reduces the aberrant innate immune response. ADAR1 mutations causing AGS affect the activity of the interferon-inducible cytoplasmic isoform more severely than the nuclear isoform.


Assuntos
Adenosina Desaminase/metabolismo , Imunidade Inata , Edição de RNA , RNA de Cadeia Dupla/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Adenosina Desaminase/genética , Animais , Doenças Autoimunes do Sistema Nervoso/genética , Doenças Autoimunes do Sistema Nervoso/imunologia , Cruzamentos Genéticos , Citocinas/metabolismo , Perda do Embrião/patologia , Embrião de Mamíferos/patologia , Feminino , Fibroblastos/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Inosina/metabolismo , Fígado/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Mutação/genética , Malformações do Sistema Nervoso/genética , Malformações do Sistema Nervoso/imunologia , Fenótipo , Proteínas de Ligação a RNA/genética , Receptores de Interferon/metabolismo , Análise de Sobrevida , Transcrição Gênica , Proteína Supressora de Tumor p53/metabolismo , Uracila/metabolismo
15.
Genome Biol ; 15(2): R28, 2014 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-24485196

RESUMO

BACKGROUND: RNA editing by adenosine to inosine deamination is a widespread phenomenon, particularly frequent in the human transcriptome, largely due to the presence of inverted Alu repeats and their ability to form double-stranded structures--a requisite for ADAR editing. While several hundred thousand editing sites have been identified within these primate-specific repeats, the function of Alu-editing has yet to be elucidated. RESULTS: We show that inverted Alu repeats, expressed in the primate brain, can induce site-selective editing in cis on sites located several hundred nucleotides from the Alu elements. Furthermore, a computational analysis, based on available RNA-seq data, finds that site-selective editing occurs significantly closer to edited Alu elements than expected. These targets are poorly edited upon deletion of the editing inducers, as well as in homologous transcripts from organisms lacking Alus. Sequences surrounding sites near edited Alus in UTRs, have been subjected to a lesser extent of evolutionary selection than those far from edited Alus, indicating that their editing generally depends on cis-acting Alus. Interestingly, we find an enrichment of primate-specific editing within encoded sequence or the UTRs of zinc finger-containing transcription factors. CONCLUSIONS: We propose a model whereby primate-specific editing is induced by adjacent Alu elements that function as recruitment elements for the ADAR editing enzymes. The enrichment of site-selective editing with potentially functional consequences on the expression of transcription factors indicates that editing contributes more profoundly to the transcriptomic regulation and repertoire in primates than previously thought.


Assuntos
Elementos Alu/genética , Regulação da Expressão Gênica , Primatas/genética , Transcriptoma/genética , Adenosina/genética , Animais , Desaminação/genética , Genoma Humano , Humanos , Inosina/genética , Edição de RNA , Regiões não Traduzidas/genética
16.
Stem Cells Dev ; 22(16): 2254-67, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23534823

RESUMO

Urodele amphibians possess an amazing regenerative capacity that requires the activation of cellular plasticity in differentiated cells and progenitor/stem cells. Many aspects of regeneration in Urodele amphibians recapitulate development, making it unlikely that gene regulatory pathways which are essential for development are mutually exclusive from those necessary for regeneration. One such post-transcriptional gene regulatory pathway, which has been previously shown to be essential for functional metazoan development, is RNA editing. RNA editing catalyses discrete nucleotide changes in RNA transcripts, creating a molecular diversity that could create an enticing connection to the activated cellular plasticity found in newts during regeneration. To assess whether RNA editing occurs during regeneration, we demonstrated that GABRA3 and ADAR2 mRNA transcripts are edited in uninjured and regenerating tissues. Full open-reading frame sequences for ADAR1 and ADAR2, two enzymes responsible for adenosine-to-inosine RNA editing, were cloned from newt brain cDNA and exhibited a strong resemblance to ADAR (adenosine deaminase, RNA-specific) enzymes discovered in mammals. We demonstrated that ADAR1 and ADAR2 mRNA expression levels are differentially expressed during different phases of regeneration in multiple tissues, whereas protein expression levels remain unaltered. In addition, we have characterized a fascinating nucleocytoplasmic shuttling of ADAR1 in a variety of different cell types during regeneration, which could provide a mechanism for controlling RNA editing, without altering translational output of the editing enzyme. The link between RNA editing and regeneration provides further insights into how lower organisms, such as the newt, can activate essential molecular pathways via the discrete alteration of RNA sequences.


Assuntos
Adenosina Desaminase/genética , Regulação da Expressão Gênica , Regeneração Nervosa/fisiologia , Notophthalmus viridescens/genética , Edição de RNA , Regeneração/fisiologia , Adenosina/metabolismo , Adenosina Desaminase/metabolismo , Animais , Sequência de Bases , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Diferenciação Celular , Ativação Enzimática , Extremidades/lesões , Extremidades/fisiologia , Inosina/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Dados de Sequência Molecular , Miocárdio/citologia , Miocárdio/metabolismo , Notophthalmus viridescens/metabolismo , Proteínas de Ligação a RNA , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Transdução de Sinais
17.
RNA Biol ; 9(8): 1054-65, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22858680

RESUMO

Editing by ADAR enzymes is essential for mammalian life. Still, knowledge of the spatio-temporal editing patterns in mammals is limited. By use of 454 amplicon sequencing we examined the editing status of 12 regionally extracted mRNAs from porcine developing brain encompassing a total of 64 putative ADAR editing sites. In total 24 brain tissues, dissected from up to five regions from embryonic gestation day 23, 42, 60, 80, 100 and 115, were examined for editing.   Generally, editing increased during embryonic development concomitantly with an increase in ADAR2 mRNA level. Notably, the Gria2 (GluR-B) Q/R site, reported to be ~100% edited in previous studies, is only 54% edited at embryonic day 23. Transcripts with multiple editing sites in close proximity to each other exhibit coupled editing and an extraordinary incidence of long-range coupling of editing events more than 32 kb apart is observed for the kainate glutamate receptor 2 transcript, Grik2. Our study reveals complex spatio-temporal ADAR editing patterns of coordinated editing events that may play important roles in the development of the mammalian brain.


Assuntos
Adenosina Desaminase/genética , Adenosina Desaminase/metabolismo , Encéfalo/embriologia , Encéfalo/metabolismo , Edição de RNA , Sus scrofa/embriologia , Animais , Humanos , Camundongos , Análise de Sequência de DNA , Sus scrofa/metabolismo
18.
Nucleic Acids Res ; 40(19): 9876-86, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22848101

RESUMO

Transcripts have been found to be site selectively edited from adenosine-to-inosine (A-to-I) in the mammalian brain, mostly in genes involved in neurotransmission. While A-to-I editing occurs at double-stranded structures, other structural requirements are largely unknown. We have investigated the requirements for editing at the I/M site in the Gabra-3 transcript of the GABA(A) receptor. We identify an evolutionarily conserved intronic duplex, 150 nt downstream of the exonic hairpin where the I/M site resides, which is required for its editing. This is the first time a distant RNA structure has been shown to be important for A-to-I editing. We demonstrate that the element also can induce editing in related but normally not edited RNA sequences. In human, thousands of genes are edited in duplexes formed by inverted repeats in non-coding regions. It is likely that numerous such duplexes can induce editing of coding regions throughout the transcriptome.


Assuntos
Íntrons , Edição de RNA , Sequências Reguladoras de Ácido Ribonucleico , Animais , Encéfalo/metabolismo , Células HEK293 , Células HeLa , Humanos , Camundongos , Conformação de Ácido Nucleico , Splicing de RNA , Ratos , Receptores de GABA-A/genética , Suínos
19.
Genome Res ; 22(8): 1477-87, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22645261

RESUMO

Adenosine-to-inosine (A-to-I) RNA editing targets double-stranded RNA stem-loop structures in the mammalian brain. It has previously been shown that miRNAs are substrates for A-to-I editing. For the first time, we show that for several definitions of edited miRNA, the level of editing increases with development, thereby indicating a regulatory role for editing during brain maturation. We use high-throughput RNA sequencing to determine editing levels in mature miRNA, from the mouse transcriptome, and compare these with the levels of editing in pri-miRNA. We show that increased editing during development gradually changes the proportions of the two miR-376a isoforms, which previously have been shown to have different targets. Several other miRNAs that also are edited in the seed sequence show an increased level of editing through development. By comparing editing of pri-miRNA with editing and expression of the corresponding mature miRNA, we also show an editing-induced developmental regulation of miRNA expression. Taken together, our results imply that RNA editing influences the miRNA repertoire during brain maturation.


Assuntos
Adenosina/metabolismo , Encéfalo/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Inosina/metabolismo , MicroRNAs/metabolismo , Edição de RNA , Adenosina/genética , Animais , Sequência de Bases , Encéfalo/embriologia , Encéfalo/crescimento & desenvolvimento , Biologia Computacional , Dendritos/genética , Dendritos/metabolismo , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário/genética , Sequenciamento de Nucleotídeos em Larga Escala , Inosina/genética , Camundongos , MicroRNAs/genética , Isoformas de RNA/genética , Isoformas de RNA/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Transcriptoma
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA