Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cell Rep ; 42(7): 112688, 2023 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-37355987

RESUMO

The methyltransferase-like 3 (METTL3)-/METTL14-containing complex predominantly catalyzes N6-methyladenosine (m6A) modification, which affects mRNA stability. Although the METTL14 R298P mutation is found in multiple cancer types, its biological effects are not completely understood. Here, we show that the heterozygous R298P mutation promotes cancer cell proliferation, whereas the homozygous mutation reduces proliferation. Methylated RNA immunoprecipitation sequencing analysis indicates that the R298P mutation reduces m6A modification at canonical motifs. Furthermore, this mutation induces m6A modification at aberrant motifs, which is evident only in cell lines harboring the homozygous mutation. The aberrant recognition of m6A modification sites alters the methylation efficiency at surrounding canonical motifs. One example is c-MET mRNA, which is highly methylated at canonical motifs close to the aberrantly methylated sites. Consequently, c-MET mRNA is severely destabilized, reducing c-Myc expression and suppressing cell proliferation. These data suggest that the METTL14 R298P mutation affects target recognition for m6A modification, perturbing gene expression patterns and cell growth.


Assuntos
Metiltransferases , Neoplasias , Humanos , Metiltransferases/genética , Neoplasias/genética , Ciclo Celular , Linhagem Celular , Mutação/genética
2.
J Biol Chem ; 299(7): 104840, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37209819

RESUMO

Adenosine-to-inosine RNA editing is catalyzed by nuclear adenosine deaminase acting on RNA 1 (ADAR1) p110 and ADAR2, and cytoplasmic ADAR1 p150 in mammals, all of which recognize dsRNAs as targets. RNA editing occurs in some coding regions, which alters protein functions by exchanging amino acid sequences, and is therefore physiologically significant. In general, such coding sites are edited by ADAR1 p110 and ADAR2 before splicing, given that the corresponding exon forms a dsRNA structure with an adjacent intron. We previously found that RNA editing at two coding sites of antizyme inhibitor 1 (AZIN1) is sustained in Adar1 p110/Aadr2 double KO mice. However, the molecular mechanisms underlying RNA editing of AZIN1 remain unknown. Here, we showed that Azin1 editing levels were increased upon type I interferon treatment, which activated Adar1 p150 transcription, in mouse Raw 264.7 cells. Azin1 RNA editing was observed in mature mRNA but not precursor mRNA. Furthermore, we revealed that the two coding sites were editable only by ADAR1 p150 in both mouse Raw 264.7 and human embryonic kidney 293T cells. This unique editing was achieved by forming a dsRNA structure with a downstream exon after splicing, and the intervening intron suppressed RNA editing. Therefore, deletion of a nuclear export signal from ADAR1 p150, shifting its localization to the nucleus, decreased Azin1 editing levels. Finally, we demonstrated that Azin1 RNA editing was completely absent in Adar1 p150 KO mice. Thus, these findings indicate that RNA editing of AZIN1 coding sites is exceptionally catalyzed by ADAR1 p150 after splicing.


Assuntos
Adenosina Desaminase , Proteínas de Transporte , Edição de RNA , Animais , Humanos , Camundongos , Adenosina Desaminase/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Catálise , Edição de RNA/efeitos dos fármacos , Edição de RNA/genética , RNA de Cadeia Dupla/genética , RNA Mensageiro/metabolismo , Células HEK293 , Camundongos Knockout , Células RAW 264.7 , Interferons/farmacologia , Transporte Proteico
3.
Int Immunol ; 35(3): 123-133, 2023 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-36469491

RESUMO

Adenosine deaminase acting on RNA 1 (ADAR1) is an RNA-editing enzyme that catalyzes adenosine-to-inosine conversions in double-stranded RNAs (dsRNAs). In mammals, ADAR1 is composed of two isoforms: a nuclear short p110 isoform and a cytoplasmic long p150 isoform. Whereas both isoforms contain right-handed dsRNA-binding and deaminase domains, ADAR1 p150 harbors a Zα domain that binds to left-handed dsRNAs, termed Z-RNAs. Myeloma differentiation-associated gene 5 (MDA5) sensing of endogenous dsRNAs as non-self leads to the induction of type I interferon (IFN)-stimulated genes, but recent studies revealed that ADAR1 p150-mediated RNA editing, but not ADAR1 p110, prevents this MDA5-mediated sensing. ADAR1 p150-specific RNA-editing sites are present and at least a Zα domain-Z-RNA interaction is required for this specificity. Mutations in the ADAR1 gene cause Aicardi-Goutières syndrome (AGS), an infant encephalopathy with type I IFN overproduction. Insertion of a point mutation in the Zα domain of the Adar1 gene induces AGS-like encephalopathy in mice, which is rescued by concurrent deletion of MDA5. This finding indicates that impaired ADAR1 p150-mediated RNA-editing is a mechanism underlying AGS caused by an ADAR1 mutation. ADAR1 p150 also prevents ZBP1 sensing of endogenous Z-RNA, which leads to programmed cell death, via the Zα domain and its RNA-editing activity. Furthermore, ADAR1 prevents protein kinase R (PKR) sensing of endogenous right-handed dsRNAs, which leads to translational shutdown and growth arrest. Thus, ADAR1 acts as a regulatory hub that blocks sensing of endogenous dsRNAs as non-self by multiple sensor proteins, both in RNA editing-dependent and -independent manners, and is a potential therapeutic target for diseases, especially cancer.


Assuntos
Edição de RNA , RNA de Cadeia Dupla , Camundongos , Animais , Adenosina Desaminase/genética , Adenosina Desaminase/metabolismo , Isoformas de Proteínas/genética , Helicase IFIH1 Induzida por Interferon/genética , Helicase IFIH1 Induzida por Interferon/metabolismo , Apoptose , Mamíferos/genética , Mamíferos/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo
4.
Nat Commun ; 13(1): 6435, 2022 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-36307435

RESUMO

During erythroid differentiation, the maintenance of genome integrity is key for the success of multiple rounds of cell division. However, molecular mechanisms coordinating the expression of DNA repair machinery in erythroid progenitors are poorly understood. Here, we discover that an RNA N6-methyladenosine (m6A) methyltransferase, METTL16, plays an essential role in proper erythropoiesis by safeguarding genome integrity via the control of DNA-repair-related genes. METTL16-deficient erythroblasts exhibit defective differentiation capacity, DNA damage and activation of the apoptotic program. Mechanistically, METTL16 controls m6A deposition at the structured motifs in DNA-repair-related transcripts including Brca2 and Fancm mRNAs, thereby upregulating their expression. Furthermore, a pairwise CRISPRi screen revealed that the MTR4-nuclear RNA exosome complex is involved in the regulation of METTL16 substrate mRNAs in erythroblasts. Collectively, our study uncovers that METTL16 and the MTR4-nuclear RNA exosome act as essential regulatory machinery to maintain genome integrity and erythropoiesis.


Assuntos
Eritropoese , Metiltransferases , Metiltransferases/metabolismo , Metilação , Eritropoese/genética , Adenosina/metabolismo , RNA Mensageiro/metabolismo , Eritroblastos/metabolismo , DNA/metabolismo
5.
J Immunol ; 207(12): 3016-3027, 2021 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-34772697

RESUMO

Aicardi-Goutières syndrome (AGS) is a congenital inflammatory disorder accompanied by overactivated type I IFN signaling and encephalopathy with leukodystrophy and intracranial calcification. To date, none of the mouse models carrying an AGS-causative mutation has mimicked such brain pathology. Here, we established a mutant mouse model carrying a K948N point mutation, corresponding to an AGS-causative K999N mutation, located in a deaminase domain of the Adar1 gene that encodes an RNA editing enzyme. Adar1K948N/K948N mice displayed postnatal growth retardation. Hyperplasia of splenic white pulps with germinal centers and hepatic focal inflammation were observed from 2 mo of age. Inflammation developed in the lungs and heart with lymphocyte infiltration in an age-dependent manner. Furthermore, white matter abnormalities with astrocytosis and microgliosis were detected at 1 y of age. The increased expression of IFN-stimulated genes was detected in multiple organs, including the brain, from birth. In addition, single-nucleus RNA sequencing revealed that this elevated expression of IFN-stimulated genes was commonly observed in all neuronal subtypes, including neurons, oligodendrocytes, and astrocytes. We further showed that a K948N point mutation reduced the RNA editing activity of ADAR1 in vivo. The pathological abnormalities found in Adar1K948N/K948N mice were ameliorated by either the concurrent deletion of MDA5, a cytosolic sensor of unedited transcripts, or the sole expression of active ADAR1 p150, an isoform of ADAR1. Collectively, such data suggest that although the degree is mild, Adar1K948N/K948N mice mimic multiple AGS phenotypes, including encephalopathy, which is caused by reduced RNA editing activity of the ADAR1 p150 isoform.


Assuntos
Adenosina Desaminase , Encefalopatias , Adenosina Desaminase/genética , Adenosina Desaminase/metabolismo , Animais , Doenças Autoimunes do Sistema Nervoso , Inflamação/genética , Inflamação/metabolismo , Camundongos , Mutação , Malformações do Sistema Nervoso , Mutação Puntual , Isoformas de Proteínas/genética , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo
6.
Int J Mol Sci ; 22(21)2021 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-34768866

RESUMO

Adenosine deaminase acting on RNA 1 (ADAR1) is an enzyme responsible for double-stranded RNA (dsRNA)-specific adenosine-to-inosine RNA editing, which is estimated to occur at over 100 million sites in humans. ADAR1 is composed of two isoforms transcribed from different promoters: p150 and N-terminal truncated p110. Deletion of ADAR1 p150 in mice activates melanoma differentiation-associated protein 5 (MDA5)-sensing pathway, which recognizes endogenous unedited RNA as non-self. In contrast, we have recently demonstrated that ADAR1 p110-mediated RNA editing does not contribute to this function, implying that a unique Z-DNA/RNA-binding domain α (Zα) in the N terminus of ADAR1 p150 provides specific RNA editing, which is critical for preventing MDA5 activation. In addition, a mutation in the Zα domain is identified in patients with Aicardi-Goutières syndrome (AGS), an inherited encephalopathy characterized by overproduction of type I interferon. Accordingly, we and other groups have recently demonstrated that Adar1 Zα-mutated mice show MDA5-dependent type I interferon responses. Furthermore, one such mutant mouse carrying a W197A point mutation in the Zα domain, which inhibits Z-RNA binding, manifests AGS-like encephalopathy. These findings collectively suggest that Z-RNA binding by ADAR1 p150 is essential for proper RNA editing at certain sites, preventing aberrant MDA5 activation.


Assuntos
Adenosina Desaminase/metabolismo , Adenosina Desaminase/fisiologia , Proteínas de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/fisiologia , Adenosina , Animais , DNA Forma Z/metabolismo , DNA Forma Z/fisiologia , Humanos , Inosina , Interferon Tipo I/metabolismo , Helicase IFIH1 Induzida por Interferon/genética , Camundongos , Isoformas de Proteínas/metabolismo , Edição de RNA/fisiologia , RNA de Cadeia Dupla
7.
Immunity ; 54(9): 1976-1988.e7, 2021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34525338

RESUMO

Mutations in the adenosine-to-inosine RNA-editing enzyme ADAR1 p150, including point mutations in the Z-RNA recognition domain Zα, are associated with Aicardi-Goutières syndrome (AGS). Here, we examined the in vivo relevance of ADAR1 binding of Z-RNA. Mutation of W197 in Zα, which abolished Z-RNA binding, reduced RNA editing. Adar1W197A/W197A mice displayed severe growth retardation after birth, broad expression of interferon-stimulated genes (ISGs), and abnormal development of multiple organs. Notably, malformation of the brain was accompanied by white matter vacuolation and gliosis, reminiscent of AGS-associated encephalopathy. Concurrent deletion of the double-stranded RNA sensor MDA5 ameliorated these abnormalities. ADAR1 (W197A) expression increased in a feedback manner downstream of type I interferons, resulting in increased RNA editing at a subset of, but not all, ADAR1 target sites. This increased expression did not ameliorate inflammation in Adar1W197A/W197A mice. Thus, editing of select endogenous RNAs by ADAR1 is essential for preventing inappropriate MDA5-mediated inflammation, with relevance to the pathogenesis of AGS.


Assuntos
Adenosina Desaminase/genética , Doenças Autoimunes do Sistema Nervoso/genética , Malformações do Sistema Nervoso/genética , Edição de RNA/genética , RNA de Cadeia Dupla/genética , Adenosina Desaminase/metabolismo , Animais , Doenças Autoimunes do Sistema Nervoso/fisiopatologia , Modelos Animais de Doenças , Helicase IFIH1 Induzida por Interferon/metabolismo , Camundongos , Mutação , Malformações do Sistema Nervoso/fisiopatologia , RNA de Cadeia Dupla/metabolismo
8.
PLoS Genet ; 17(5): e1009516, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33983932

RESUMO

Adenosine deaminase acting on RNA 1 (ADAR1), an enzyme responsible for adenosine-to-inosine RNA editing, is composed of two isoforms: nuclear p110 and cytoplasmic p150. Deletion of Adar1 or Adar1 p150 genes in mice results in embryonic lethality with overexpression of interferon-stimulating genes (ISGs), caused by the aberrant recognition of unedited endogenous transcripts by melanoma differentiation-associated protein 5 (MDA5). However, among numerous RNA editing sites, how many RNA sites require editing, especially by ADAR1 p150, to avoid MDA5 activation and whether ADAR1 p110 contributes to this function remains elusive. In particular, ADAR1 p110 is abundant in the mouse brain where a subtle amount of ADAR1 p150 is expressed, whereas ADAR1 mutations cause Aicardi-Goutières syndrome, in which the brain is one of the most affected organs accompanied by the elevated expression of ISGs. Therefore, understanding RNA editing-mediated prevention of MDA5 activation in the brain is especially important. Here, we established Adar1 p110-specific knockout mice, in which the upregulated expression of ISGs was not observed. This result suggests that ADAR1 p150-mediated RNA editing is enough to suppress MDA5 activation. Therefore, we further created Adar1 p110/Adar2 double knockout mice to identify ADAR1 p150-mediated editing sites. This analysis demonstrated that although the elevated expression of ISGs was not observed, only less than 2% of editing sites were preserved in the brains of Adar1 p110/Adar2 double knockout mice. Of note, we found that some sites were highly edited, which was comparable to those found in wild-type mice, indicating the presence of ADAR1 p150-specific sites. These data suggest that RNA editing at a very limited sites, which is mediated by a subtle amount of ADAR1 p150, is sufficient to prevents MDA5 activation, at least in the mouse brain.


Assuntos
Adenosina Desaminase/metabolismo , Encéfalo/metabolismo , Helicase IFIH1 Induzida por Interferon/metabolismo , Edição de RNA , Regiões 3' não Traduzidas/genética , Adenosina Desaminase/deficiência , Adenosina Desaminase/genética , Animais , Animais Recém-Nascidos , Feminino , Íntrons/genética , Isoenzimas/metabolismo , Camundongos , Camundongos Knockout , Mutação , Especificidade de Órgãos , Proteínas de Ligação a RNA/genética , Taxa de Sobrevida
9.
Methods Mol Biol ; 2181: 309-330, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32729088

RESUMO

The brain is one of the organs that are preferentially targeted by adenosine-to-inosine (A-to-I) RNA editing, a posttranscriptional modification. This chemical modification affects neuronal development and functions at multiple levels, leading to normal brain homeostasis by increasing the complexity of the transcriptome. This includes modulation of the properties of ion channel and neurotransmitter receptors by recoding, redirection of miRNA targets by changing sequence complementarity, and suppression of immune response by altering RNA structure. Therefore, from another perspective, it appears that the brain is highly vulnerable to dysregulation of A-to-I RNA editing. Here, we focus on how aberrant A-to-I RNA editing is involved in neurological and neurodegenerative diseases of humans including epilepsy, amyotrophic lateral sclerosis, psychiatric disorders, developmental disorders, brain tumors, and encephalopathy caused by autoimmunity. In addition, we provide information regarding animal models to better understand the mechanisms behind disease phenotype.


Assuntos
Doenças do Sistema Nervoso/genética , Doenças Neurodegenerativas/genética , Edição de RNA/fisiologia , Adenosina/química , Adenosina/genética , Esclerose Lateral Amiotrófica/genética , Animais , Epilepsia/genética , Humanos , Inosina/química , Inosina/genética , Fenótipo
10.
J Immunol ; 204(8): 2156-2168, 2020 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-32169840

RESUMO

ADAR1 is an RNA-editing enzyme that is abundant in the thymus. We have previously reported that ADAR1 is required for establishing central tolerance during the late stage of thymocyte development by preventing MDA5 sensing of endogenous dsRNA as nonself. However, the role of ADAR1 during the early developmental stage remains unknown. In this study, we demonstrate that early thymocyte-specific deletion of ADAR1 in mice caused severe thymic atrophy with excessive apoptosis and impaired transition to a late stage of development accompanied by the loss of TCR expression. Concurrent MDA5 deletion ameliorated apoptosis but did not restore impaired transition and TCR expression. In addition, forced TCR expression was insufficient to restore the transition. However, simultaneous TCR expression and MDA5 deletion efficiently ameliorated the impaired transition of ADAR1-deficient thymocytes to the late stage. These findings indicate that RNA-editing-dependent and -independent functions of ADAR1 synergistically regulate early thymocyte development.


Assuntos
Adenosina Desaminase/metabolismo , Helicase IFIH1 Induzida por Interferon/metabolismo , Timócitos/imunologia , Adenosina Desaminase/deficiência , Adenosina Desaminase/genética , Animais , Apoptose/genética , Apoptose/imunologia , Helicase IFIH1 Induzida por Interferon/deficiência , Helicase IFIH1 Induzida por Interferon/genética , Camundongos , Camundongos Knockout , Camundongos Mutantes , Edição de RNA/genética , Edição de RNA/imunologia , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia
11.
RNA ; 26(4): 454-469, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31941663

RESUMO

Adenosine-to-inosine RNA editing is an essential post-transcriptional modification catalyzed by adenosine deaminase acting on RNA (ADAR)1 and ADAR2 in mammals. For numerous sites in coding sequences (CDS) and microRNAs, editing is highly conserved and has significant biological consequences, for example, by altering amino acid residues and target recognition. However, no comprehensive and quantitative studies have been undertaken to determine how specific ADARs contribute to conserved sites in vivo. Here, we amplified each RNA region with editing site(s) separately and combined these for deep sequencing. Then, we compared the editing ratios of all sites that were conserved in CDS and microRNAs in the cerebral cortex and spleen of wild-type mice, Adar1E861A/E861AIfih-/- mice expressing inactive ADAR1 (Adar1 KI) and Adar2-/-Gria2R/R (Adar2 KO) mice. We found that most of the sites showed a preference for one ADAR. In contrast, some sites, such as miR-3099-3p, showed no ADAR preference. In addition, we found that the editing ratio for several sites, such as DACT3 R/G, was up-regulated in either Adar mutant mouse strain, whereas a coordinated interplay between ADAR1 and ADAR2 was required for the efficient editing of specific sites, such as the 5-HT2CR B site. We further created double mutant Adar1 KI Adar2 KO mice and observed viable and fertile animals with the complete absence of editing, demonstrating that ADAR1 and ADAR2 are the sole enzymes responsible for all editing sites in vivo. Collectively, these findings indicate that editing is regulated in a site-specific manner by the different interplay between ADAR1 and ADAR2.


Assuntos
Adenosina Desaminase/metabolismo , MicroRNAs/metabolismo , Edição de RNA , Proteínas de Ligação a RNA/metabolismo , Adenosina Desaminase/genética , Animais , Feminino , Masculino , Camundongos , MicroRNAs/genética , Mutação , Motivos de Nucleotídeos , Proteínas de Ligação a RNA/genética
12.
Cell Mol Life Sci ; 77(15): 2931-2948, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31996954

RESUMO

Our body expresses sensors to detect pathogens through the recognition of expressed molecules, including nucleic acids, lipids, and proteins, while immune tolerance prevents an overreaction with self and the development of autoimmune disease. Adenosine (A)-to-inosine (I) RNA editing, catalyzed by adenosine deaminases acting on RNA (ADARs), is a post-transcriptional modification that can potentially occur at over 100 million sites in the human genome, mainly in Alu repetitive elements that preferentially form a double-stranded RNA (dsRNA) structure. A-to-I conversion within dsRNA, which may induce a structural change, is required to escape from the host immune system, given that endogenous dsRNAs transcribed from Alu repetitive elements are potentially recognized by melanoma differentiation-associated protein 5 (MDA5) as non-self. Of note, loss-of-function mutations in the ADAR1 gene cause Aicardi-Goutières syndrome, a congenital autoimmune disease characterized by encephalopathy and a type I interferon (IFN) signature. However, the loss of ADAR1 in cancer cells with an IFN signature induces lethality via the activation of protein kinase R in addition to MDA5. This makes cells more sensitive to immunotherapy, highlighting the opposing immune status of autoimmune diseases (overreaction) and cancer (tolerance). In this review, we provide an overview of insights into two opposing aspects of RNA editing that functions as a modulator of the immune system in autoimmune diseases and cancer.


Assuntos
Adenosina/metabolismo , Sistema Imunitário/metabolismo , Inosina/metabolismo , Edição de RNA , Adenosina Desaminase/genética , Adenosina Desaminase/metabolismo , Animais , Doenças Autoimunes/genética , Doenças Autoimunes/patologia , Doenças Autoimunes do Sistema Nervoso/genética , Doenças Autoimunes do Sistema Nervoso/patologia , Humanos , Neoplasias/genética , Neoplasias/patologia , Malformações do Sistema Nervoso/genética , Malformações do Sistema Nervoso/patologia
13.
EMBO Rep ; 19(12)2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30361393

RESUMO

T cells play a crucial role in the adaptive immune system, and their maturation process is tightly regulated. Adenosine deaminase acting on RNA 1 (ADAR1) is the enzyme responsible for adenosine-to-inosine RNA editing in dsRNAs, and loss of ADAR1 activates the innate immune sensing response via melanoma differentiation-associated protein 5 (MDA5), which interprets unedited dsRNA as non-self. Although ADAR1 is highly expressed in the thymus, its role in the adaptive immune system, especially in T cells, remains elusive. Here, we demonstrate that T cell-specific deletion of Adar1 in mice causes abnormal thymic T cell maturation including impaired negative selection and autoimmunity such as spontaneous colitis. This is caused by excessive expression of interferon-stimulated genes, which reduces T cell receptor (TCR) signal transduction, due to a failure of RNA editing in ADAR1-deficient thymocytes. Intriguingly, concurrent deletion of MDA5 restores thymocyte maturation and prevents colitis. These findings suggest that prevention of MDA5 sensing of endogenous dsRNA by ADAR1-mediated RNA editing is required for preventing both innate immune responses and T cell-mediated autoimmunity.


Assuntos
Adenosina Desaminase/metabolismo , Autoimunidade , Edição de RNA , Tolerância a Antígenos Próprios , Timo/metabolismo , Adenosina Desaminase/deficiência , Animais , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/metabolismo , Diferenciação Celular/genética , Colite/imunologia , Colite/patologia , Deleção de Genes , Inflamação/imunologia , Inflamação/patologia , Helicase IFIH1 Induzida por Interferon/metabolismo , Interferons/metabolismo , Ativação Linfocitária/imunologia , Camundongos Knockout , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais , Timócitos/metabolismo , Regulação para Cima/genética
14.
Chem Commun (Camb) ; 54(69): 9627-9630, 2018 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-30095851

RESUMO

We found that insertion of artificial nucleic acid analogs, such as bridged nucleic acid (BNA), into DNA probes increases the difference in melting temperature between N6-methyladenosine (m6A)-containing RNA and unmethylated RNA. By applying this principle, we quantified methylation efficiency at m6A sites in E. coli 23S rRNA with high accuracy.


Assuntos
Adenosina/análogos & derivados , Sondas de DNA/genética , RNA Ribossômico 23S/metabolismo , Adenosina/química , Escherichia coli/metabolismo , Metilação , Hibridização de Ácido Nucleico , RNA Ribossômico 23S/química , RNA Ribossômico 23S/genética , Temperatura de Transição
15.
Genes Cells ; 22(9): 785-798, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28695676

RESUMO

Matrin3 is an RNA-binding protein that is localized in the nuclear matrix. Although various roles in RNA metabolism have been reported for Matrin3, in vivo target RNAs to which Matrin3 binds directly have not been investigated comprehensively so far. Here, we show that Matrin3 binds predominantly to intronic regions of pre-mRNAs. Photoactivatable Ribonucleoside-Enhanced Cross-linking and Immunoprecipitation (PAR-CLIP) analysis using human neuronal cells showed that Matrin3 recognized pyrimidine-rich sequences as binding motifs, including the polypyrimidine tract, a splicing regulatory element. Splicing-sensitive microarray analysis showed that depletion of Matrin3 preferentially increased the inclusion of cassette exons that were adjacent to introns that contained Matrin3-binding sites. We further found that although most of the genes targeted by polypyrimidine tract binding protein 1 (PTBP1) were also bound by Matrin3, Matrin3 could control alternative splicing in a PTBP1-independent manner, at least in part. These findings suggest that Matrin3 is a splicing regulator that targets intronic pyrimidine-rich sequences.


Assuntos
Processamento Alternativo , Íntrons , Proteínas Associadas à Matriz Nuclear/metabolismo , Proteínas de Ligação a RNA/metabolismo , Sítios de Ligação , Linhagem Celular Tumoral , Ribonucleoproteínas Nucleares Heterogêneas/química , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Humanos , Proteínas Associadas à Matriz Nuclear/genética , Motivos de Nucleotídeos , Proteína de Ligação a Regiões Ricas em Polipirimidinas/química , Proteína de Ligação a Regiões Ricas em Polipirimidinas/metabolismo , Ligação Proteica , Pirimidinas/química , RNA Mensageiro/química , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética
16.
Cell Rep ; 17(8): 2004-2014, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27851964

RESUMO

Calcium-dependent activator protein for secretion 1 (CAPS1) plays a distinct role in the priming step of dense core vesicle (DCV) exocytosis. CAPS1 pre-mRNA is known to undergo adenosine-to-inosine RNA editing in its coding region, which results in a glutamate-to-glycine conversion at a site in its C-terminal region. However, the physiological significance of CAPS1 RNA editing remains elusive. Here, we created mutant mice in which edited CAPS1 was solely expressed. These mice were lean due to increased energy expenditure caused by physical hyperactivity. Electrophysiological and biochemical analyses demonstrated that the exocytosis of DCVs was upregulated in the chromaffin cells and neurons of these mice. Furthermore, we showed that edited CAPS1 bound preferentially to the activated form of syntaxin-1A, a component of the exocytotic fusion complex. These findings suggest that RNA editing regulates DCV exocytosis in vivo, affecting physical activity.


Assuntos
Proteínas de Ligação ao Cálcio/genética , Exocitose , Proteínas do Tecido Nervoso/genética , Edição de RNA/genética , Vesículas Secretórias/metabolismo , Adenosina Desaminase/metabolismo , Animais , Biocatálise , Peso Corporal , Proteínas de Ligação ao Cálcio/metabolismo , Catecolaminas/metabolismo , Metabolismo Energético , Masculino , Camundongos , Camundongos Mutantes , Proteínas do Tecido Nervoso/metabolismo , Conformação de Ácido Nucleico , Células PC12 , Condicionamento Físico Animal , Ligação Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Ratos , Sintaxina 1/metabolismo
17.
Nat Commun ; 6: 6183, 2015 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-25630387

RESUMO

TAR DNA-binding protein of 43 kDa (TDP-43) and its C-terminal fragment of 25 kDa (CTF25) play critical roles in amyotrophic lateral sclerosis and frontotemporal lobar degeneration. Although the overexpression of TDP-43 in cultured cells and animals results in the production of CTF25, the cleavage site that generates CTF25 and biological significance of the cleavage remain undetermined. Here we identify Asp174 as a cleavage site for CTF25. TDP-43 is cleaved initially after Asp174, which activates caspase-3/7 to accelerate TDP-43 fragmentation. Consequently, blockage of this cleavage results in a severe delay in TDP-43 clearance and prolonged necrotic cell death. We further show that the endoplasmic reticulum membrane-bound caspase-4 is the enzyme responsible for the cleavage after Asp174 and inhibition of caspase-4 activity slows TDP-43 fragmentation and reduces cell viability. These findings suggest that caspase-4-mediated cleavage after Asp174 is an initiator of TDP-43 clearance, which is required to avoid cell death induced by overexpressed TDP-43.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/toxicidade , Sequência de Aminoácidos , Ácido Aspártico/metabolismo , Caspases Iniciadoras/metabolismo , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/química , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Humanos , Dados de Sequência Molecular , Proteínas Mutantes/metabolismo , Mutação/genética , Peptídeos/metabolismo , Fosforilação/efeitos dos fármacos , Agregados Proteicos/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Solubilidade
18.
Mol Cell ; 55(2): 186-98, 2014 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-24954906

RESUMO

It has been proposed that Ataxin-2, a member of the like-Sm (LSm) protein family, participates in the regulation of RNA metabolism through interaction with PABPC1. However, the exact biological mechanism and in vivo targets remain unknown. Here, we report that Ataxin-2 binds directly to RNAs in a PABPC1-independent manner. High-throughput sequencing of Ataxin-2-bound RNAs prepared by PAR-CLIP revealed that Ataxin-2 binds predominantly to uridine-rich elements, including well-characterized cis-regulatory AU-rich elements, in the 3' UTRs of target mRNAs. Gene expression analysis after Ataxin-2 depletion or overexpression revealed that Ataxin-2 stabilizes target mRNAs and increases the abundance of the corresponding proteins. A tethering assay demonstrated that Ataxin-2 elicits this effect by direct interaction with mRNAs. We also found that disease-associated polyglutamine expansion downregulates the physiological activity of Ataxin-2. These findings suggest that Ataxin-2 is an RNA-binding protein that targets cis-regulatory elements in 3' UTRs to stabilize a subset of mRNAs and increase protein expression.


Assuntos
Regiões 3' não Traduzidas , Proteínas do Tecido Nervoso/metabolismo , Estabilidade de RNA , RNA Mensageiro/metabolismo , Ataxinas , Sítios de Ligação , Proteínas ELAV/metabolismo , Regulação da Expressão Gênica , Ontologia Genética , Células HEK293 , Humanos , Mutação , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Peptídeos/genética , Ligação Proteica , Biossíntese de Proteínas , Estrutura Terciária de Proteína , RNA Mensageiro/genética
19.
Congenit Anom (Kyoto) ; 54(1): 12-21, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24330020

RESUMO

The human genome harbors approximately 2000 genes that encode microRNAs (miRNAs), small non-coding RNAs of approximately 20-22 nt that mediate post-transcriptional gene silencing. MiRNAs are generated from long transcripts through stepwise processing by the Drosha/DGCR8, Exportin-5/RanGTP and Dicer/TRBP complexes. Given that the expression of each individual miRNA is tightly regulated, the altered expression of certain miRNAs plays a pivotal role in human diseases. For instance, germline and somatic mutations in the genes encoding the miRNA processing machinery have been reported in different cancers. Furthermore, certain miRNA genes are encoded within regions that are deleted or duplicated in individuals with chromosomal abnormalities, and the fact that the knockout of these miRNAs in animal models results in lethality or the abnormal development of certain tissues indicates that these miRNA genes contribute to the disease phenotypes. It has also been reported that mutations in miRNA genes or in miRNA-binding sites, which result in the impairment of tight regulation of target mRNA expression, cause human genetic diseases, although these cases are rare. This is in contrast to the aberrant expression of certain miRNAs that results from the impairment of transcriptional or post-transcriptional regulation, which has been reported frequently in various human diseases. The present review focuses on human diseases caused by mutations in genes encoding miRNAs and the miRNA processing machinery as well as in miRNA-binding sites. Furthermore, human diseases caused by chromosomal abnormalities that involve the deletion or duplication of regions harboring genes that encode miRNAs or the miRNA processing machinery are also introduced.


Assuntos
Doenças Genéticas Inatas/genética , Células Germinativas/patologia , MicroRNAs/genética , Animais , Aberrações Cromossômicas , Modelos Animais de Doenças , Regulação da Expressão Gênica , Doenças Genéticas Inatas/etiologia , Doenças Genéticas Inatas/patologia , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA