Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.184
Filtrar
1.
Cell Biol Toxicol ; 40(1): 57, 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39060787

RESUMEN

It is well established that sevoflurane exposure leads to widespread neuronal cell death in the developing brain. Adenosine deaminase acting on RNA-1 (ADAR1) dependent adenosine-to-inosine (A-to-I) RNA editing is dynamically regulated throughout brain development. The current investigation is designed to interrogate the contributed role of ADAR1 in developmental sevoflurane neurotoxicity. Herein, we provide evidence to show that developmental sevoflurane priming triggers neuronal pyroptosis, apoptosis and necroptosis (PANoptosis), and elicits the release of inflammatory factors including IL-1ß, IL-18, TNF-α and IFN-γ. Additionally, ADAR1-P150, but not ADAR1-P110, depresses cellular PANoptosis and inflammatory response by competing with Z-DNA/RNA binding protein 1 (ZBP1) for binding to Z-RNA in the presence of sevoflurane. Further investigation demonstrates that ADAR1-dependent A-to-I RNA editing mitigates developmental sevoflurane-induced neuronal PANoptosis. To restore RNA editing, we utilize adeno-associated virus (AAV) to deliver engineered circular ADAR-recruiting guide RNAs (cadRNAs) into cells, which is capable of recruiting endogenous adenosine deaminases to promote cellular A-to-I RNA editing. As anticipated, AAV-cadRNAs diminishes sevoflurane-induced cellular Z-RNA production and PANoptosis, which could be abolished by ADAR1-P150 shRNA transfection. Moreover, AAV-cadRNAs delivery ameliorates developmental sevoflurane-induced spatial and emotional cognitive deficits without influence on locomotor activity. Taken together, these results illustrate that ADAR1-P150 exhibits a prominent role in preventing ZBP1-dependent PANoptosis through A-to-I RNA editing in developmental sevoflurane neurotoxicity. Application of engineered cadRNAs to rectify the compromised ADAR1-dependent A-to-I RNA editing provides an inspiring direction for possible clinical preventions and therapeutics.


Asunto(s)
Adenosina Desaminasa , Adenosina , Edición de ARN , Proteínas de Unión al ARN , Sevoflurano , Animales , Adenosina/metabolismo , Adenosina Desaminasa/metabolismo , Adenosina Desaminasa/genética , Apoptosis/efectos de los fármacos , Inosina/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Síndromes de Neurotoxicidad/genética , Síndromes de Neurotoxicidad/metabolismo , Piroptosis/efectos de los fármacos , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética
2.
Bioorg Med Chem ; 110: 117837, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39013280

RESUMEN

RNA molecules contain diverse modified nucleobases that play pivotal roles in numerous biological processes. Adenosine-to-inosine (A-to-I) RNA editing, one of the most prevalent RNA modifications in mammalian cells, is linked to a multitude of human diseases. To unveil the functions of A-to-I RNA editing, accurate quantification of inosine at specific sites is essential. In this study, we developed an endonuclease-mediated cleavage and real-time fluorescence quantitative PCR method for A-to-I RNA editing (EM-qPCR) to quantitatively analyze A-to-I RNA editing at a single site. By employing this method, we successfully quantified the levels of A-to-I RNA editing on various transfer RNA (tRNA) molecules at position 34 (I34) in mammalian cells with precision. Subsequently, this method was applied to tissues from sleep-deprived mice, revealing a notable alteration in the levels of I34 between sleep-deprived and control mice. The proposed method sets a precedent for the quantitative analysis of A-to-I RNA editing at specific sites, facilitating a deeper understanding of the biological implications of A-to-I RNA editing.


Asunto(s)
Adenosina , Inosina , Edición de ARN , Inosina/metabolismo , Inosina/química , Adenosina/metabolismo , Adenosina/química , Adenosina/análisis , Animales , Ratones , Humanos , Endonucleasas/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
3.
Int J Mol Sci ; 25(13)2024 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-39000531

RESUMEN

Epitranscriptomics is a field that delves into post-transcriptional changes. Among these modifications, the conversion of adenosine to inosine, traduced as guanosine (A>I(G)), is one of the known RNA-editing mechanisms, catalyzed by ADARs. This type of RNA editing is the most common type of editing in mammals and contributes to biological diversity. Disruption in the A>I(G) RNA-editing balance has been linked to diseases, including several types of cancer. Drug resistance in patients with cancer represents a significant public health concern, contributing to increased mortality rates resulting from therapy non-responsiveness and disease progression, representing the greatest challenge for researchers in this field. The A>I(G) RNA editing is involved in several mechanisms over the immunotherapy and genotoxic drug response and drug resistance. This review investigates the relationship between ADAR1 and specific A>I(G) RNA-edited sites, focusing particularly on breast cancer, and the impact of these sites on DNA damage repair and the immune response over anti-cancer therapy. We address the underlying mechanisms, bioinformatics, and in vitro strategies for the identification and validation of A>I(G) RNA-edited sites. We gathered databases related to A>I(G) RNA editing and cancer and discussed the potential clinical and research implications of understanding A>I(G) RNA-editing patterns. Understanding the intricate role of ADAR1-mediated A>I(G) RNA editing in breast cancer holds significant promise for the development of personalized treatment approaches tailored to individual patients' A>I(G) RNA-editing profiles.


Asunto(s)
Adenosina Desaminasa , Neoplasias de la Mama , Edición de ARN , Proteínas de Unión al ARN , Humanos , Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/tratamiento farmacológico , Femenino , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Adenosina/metabolismo , Resistencia a Antineoplásicos/genética , Inosina/metabolismo , Inosina/genética , Animales , Guanosina/metabolismo , Daño del ADN
4.
Genome Biol ; 25(1): 173, 2024 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-38956576

RESUMEN

BACKGROUND: RNA-seq has brought forth significant discoveries regarding aberrations in RNA processing, implicating these RNA variants in a variety of diseases. Aberrant splicing and single nucleotide variants (SNVs) in RNA have been demonstrated to alter transcript stability, localization, and function. In particular, the upregulation of ADAR, an enzyme that mediates adenosine-to-inosine editing, has been previously linked to an increase in the invasiveness of lung adenocarcinoma cells and associated with splicing regulation. Despite the functional importance of studying splicing and SNVs, the use of short-read RNA-seq has limited the community's ability to interrogate both forms of RNA variation simultaneously. RESULTS: We employ long-read sequencing technology to obtain full-length transcript sequences, elucidating cis-effects of variants on splicing changes at a single molecule level. We develop a computational workflow that augments FLAIR, a tool that calls isoform models expressed in long-read data, to integrate RNA variant calls with the associated isoforms that bear them. We generate nanopore data with high sequence accuracy from H1975 lung adenocarcinoma cells with and without knockdown of ADAR. We apply our workflow to identify key inosine isoform associations to help clarify the prominence of ADAR in tumorigenesis. CONCLUSIONS: Ultimately, we find that a long-read approach provides valuable insight toward characterizing the relationship between RNA variants and splicing patterns.


Asunto(s)
Haplotipos , Humanos , Línea Celular Tumoral , Polimorfismo de Nucleótido Simple , Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Neoplasias Pulmonares/genética , Empalme del ARN , Inosina/metabolismo , Inosina/genética , Análisis de Secuencia de ARN , Adenocarcinoma del Pulmón/genética , Edición de ARN , Programas Informáticos
5.
Genes (Basel) ; 15(7)2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-39062677

RESUMEN

Adenosine-to-inosine (A-to-I) RNA editing is an important post-transcriptional modification mediated by the adenosine deaminases acting on RNA (ADAR) family of enzymes, expanding the transcriptome by altering selected nucleotides A to I in RNA molecules. Recently, A-to-I editing has been explored for correcting disease-causing mutations in RNA using therapeutic guide oligonucleotides to direct ADAR editing at specific sites. Humans have two active ADARs whose preferences and specificities are not well understood. To investigate their substrate specificity, we introduced hADAR1 and hADAR2, respectively, into Schizosaccharomyces pombe (S. pombe), which lacks endogenous ADARs, and evaluated their editing activities in vivo. Using transcriptome sequencing of S. pombe cultured at optimal growth temperature (30 °C), we identified 483 A-to-I high-confident editing sites for hADAR1 and 404 for hADAR2, compared with the non-editing wild-type control strain. However, these sites were mostly divergent between hADAR1 and hADAR2-expressing strains, sharing 33 common sites that are less than 9% for each strain. Their differential specificity for substrates was attributed to their differential preference for neighboring sequences of editing sites. We found that at the -3-position relative to the editing site, hADAR1 exhibits a tendency toward T, whereas hADAR2 leans toward A. Additionally, when varying the growth temperature for hADAR1- and hADAR2-expressing strains, we observed increased editing sites for them at both 20 and 35 °C, compared with them growing at 30 °C. However, we did not observe a significant shift in hADAR1 and hADAR2's preference for neighboring sequences across three temperatures. The vast changes in RNA editing sites at lower and higher temperatures were also observed for hADAR2 previously in budding yeast, which was likely due to the influence of RNA folding at these different temperatures, among many other factors. We noticed examples of longer lengths of dsRNA around the editing sites that induced editing at 20 or 35 °C but were absent at the other two temperature conditions. We found genes' functions can be greatly affected by editing of their transcripts, for which over 50% of RNA editing sites for both hADAR1 and hADAR2 in S. pombe were in coding sequences (CDS), with more than 60% of them resulting in amino acid changes in protein products. This study revealed the extensive differences in substrate selectivity between the two active human ADARS, i.e., ADAR1 and ADAR2, and provided novel insight when utilizing the two different enzymes for in vivo treatment of human genetic diseases using the RNA editing approach.


Asunto(s)
Adenosina Desaminasa , Edición de ARN , Proteínas de Unión al ARN , Schizosaccharomyces , Schizosaccharomyces/genética , Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo , Edición de ARN/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Humanos , Especificidad por Sustrato , Proteínas de Schizosaccharomyces pombe/genética , Proteínas de Schizosaccharomyces pombe/metabolismo , Adenosina/metabolismo , Adenosina/genética , Inosina/genética , Inosina/metabolismo
6.
Sci Rep ; 14(1): 14973, 2024 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-38951658

RESUMEN

Deamination of bases is a form of DNA damage that occurs spontaneously via the hydrolysis and nitrosation of living cells, generating hypoxanthine from adenine. E. coli endonuclease V (eEndoV) cleaves hypoxanthine-containing double-stranded DNA, whereas human endonuclease V (hEndoV) cleaves hypoxanthine-containing RNA; however, hEndoV in vivo function remains unclear. To date, hEndoV has only been examined using hypoxanthine, because it binds closely to the base located at the cleavage site. Here, we examined whether hEndoV cleaves other lesions (e.g., AP site, 6-methyladenine, xanthine) to reveal its function and whether 2'-nucleoside modification affects its cleavage activity. We observed that hEndoV is hypoxanthine-specific; its activity was the highest with 2'-OH modification in ribose. The cleavage activity of hEndoV was compared based on its base sequence. We observed that it has specificity for adenine located on the 3'-end of hypoxanthine at the cleavage site, both before and after cleavage. These data suggest that hEndoV recognizes and cleaves the inosine generated on the poly A tail to maintain RNA quality. Our results provide mechanistic insight into the role of hEndoV in vivo.


Asunto(s)
Inosina , Inosina/metabolismo , Humanos , Poli A/metabolismo , Especificidad por Sustrato , Hipoxantina/metabolismo , Hipoxantina/química , Endodesoxirribonucleasas/metabolismo , Endodesoxirribonucleasas/química
7.
Nucleic Acids Res ; 52(12): 6733-6747, 2024 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-38828787

RESUMEN

Adenosine Deaminases Acting on RNA (ADARs) are enzymes that catalyze the conversion of adenosine to inosine in RNA duplexes. These enzymes can be harnessed to correct disease-causing G-to-A mutations in the transcriptome because inosine is translated as guanosine. Guide RNAs (gRNAs) can be used to direct the ADAR reaction to specific sites. Chemical modification of ADAR guide strands is required to facilitate delivery, increase metabolic stability, and increase the efficiency and selectivity of the editing reaction. Here, we show the ADAR reaction is highly sensitive to ribose modifications (e.g. 4'-C-methylation and Locked Nucleic Acid (LNA) substitution) at specific positions within the guide strand. Our studies were enabled by the synthesis of RNA containing a new, ribose-modified nucleoside analog (4'-C-methyladenosine). Importantly, the ADAR reaction is potently inhibited by LNA or 4'-C-methylation at different positions in the ADAR guide. While LNA at guide strand positions -1 and -2 block the ADAR reaction, 4'-C-methylation only inhibits at the -2 position. These effects are rationalized using high-resolution structures of ADAR-RNA complexes. This work sheds additional light on the mechanism of ADAR deamination and aids in the design of highly selective ADAR guide strands for therapeutic editing using chemically modified RNA.


Asunto(s)
Adenosina Desaminasa , Edición de ARN , Ribosa , Adenosina Desaminasa/metabolismo , Adenosina Desaminasa/genética , Adenosina Desaminasa/química , Ribosa/química , Ribosa/metabolismo , Humanos , Oligonucleótidos/química , Oligonucleótidos/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/química , Metilación , Adenosina/análogos & derivados , Adenosina/metabolismo , Adenosina/química , Nucleósidos/química , Nucleósidos/metabolismo , ARN/metabolismo , ARN/química , Inosina/metabolismo , Inosina/química
8.
Cell ; 187(14): 3602-3618.e20, 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38823389

RESUMEN

Purine nucleotides are vital for RNA and DNA synthesis, signaling, metabolism, and energy homeostasis. To synthesize purines, cells use two principal routes: the de novo and salvage pathways. Traditionally, it is believed that proliferating cells predominantly rely on de novo synthesis, whereas differentiated tissues favor the salvage pathway. Unexpectedly, we find that adenine and inosine are the most effective circulating precursors for supplying purine nucleotides to tissues and tumors, while hypoxanthine is rapidly catabolized and poorly salvaged in vivo. Quantitative metabolic analysis demonstrates comparative contribution from de novo synthesis and salvage pathways in maintaining purine nucleotide pools in tumors. Notably, feeding mice nucleotides accelerates tumor growth, while inhibiting purine salvage slows down tumor progression, revealing a crucial role of the salvage pathway in tumor metabolism. These findings provide fundamental insights into how normal tissues and tumors maintain purine nucleotides and highlight the significance of purine salvage in cancer.


Asunto(s)
Neoplasias , Nucleótidos de Purina , Purinas , Animales , Ratones , Purinas/metabolismo , Purinas/biosíntesis , Neoplasias/metabolismo , Neoplasias/patología , Nucleótidos de Purina/metabolismo , Humanos , Inosina/metabolismo , Hipoxantina/metabolismo , Ratones Endogámicos C57BL , Adenina/metabolismo , Línea Celular Tumoral , Femenino
9.
Nat Commun ; 15(1): 5366, 2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38926387

RESUMEN

Adenosine-to-inosine (A-to-I) editing is a prevalent post-transcriptional RNA modification within the brain. Yet, most research has relied on postmortem samples, assuming it is an accurate representation of RNA biology in the living brain. We challenge this assumption by comparing A-to-I editing between postmortem and living prefrontal cortical tissues. Major differences were found, with over 70,000 A-to-I sites showing higher editing levels in postmortem tissues. Increased A-to-I editing in postmortem tissues is linked to higher ADAR and ADARB1 expression, is more pronounced in non-neuronal cells, and indicative of postmortem activation of inflammation and hypoxia. Higher A-to-I editing in living tissues marks sites that are evolutionarily preserved, synaptic, developmentally timed, and disrupted in neurological conditions. Common genetic variants were also found to differentially affect A-to-I editing levels in living versus postmortem tissues. Collectively, these discoveries offer more nuanced and accurate insights into the regulatory mechanisms of RNA editing in the human brain.


Asunto(s)
Adenosina Desaminasa , Adenosina , Autopsia , Encéfalo , Inosina , Edición de ARN , Proteínas de Unión al ARN , Humanos , Adenosina/metabolismo , Adenosina Desaminasa/metabolismo , Adenosina Desaminasa/genética , Encéfalo/metabolismo , Inosina/metabolismo , Inosina/genética , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Corteza Prefrontal/metabolismo , Cambios Post Mortem , Masculino
10.
PLoS Pathog ; 20(6): e1012238, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38843141

RESUMEN

Although lack of ADAR (adenosine deaminase acting on RNA) orthologs, genome-wide A-to-I editing occurs specifically during sexual reproduction in a number of filamentous ascomycetes, including Fusarium graminearum and Neurospora crassa. Unlike ADAR-mediated editing in animals, fungal A-to-I editing has a strong preference for hairpin loops and U at -1 position, which leads to frequent editing of UAG and UAA stop codons. Majority of RNA editing events in fungi are in the coding region and cause amino acid changes. Some of these editing events have been experimentally characterized for providing heterozygote and adaptive advantages in F. graminearum. Recent studies showed that FgTad2 and FgTad3, 2 ADAT (adenosine deaminase acting on tRNA) enzymes that normally catalyze the editing of A34 in the anticodon of tRNA during vegetative growth mediate A-to-I mRNA editing during sexual reproduction. Stage specificity of RNA editing is conferred by stage-specific expression of short transcript isoforms of FgTAD2 and FgTAD3 as well as cofactors such as AME1 and FIP5 that facilitate the editing of mRNA in perithecia. Taken together, fungal A-to-I RNA editing during sexual reproduction is catalyzed by ADATs and it has the same sequence and structural preferences with editing of A34 in tRNA.


Asunto(s)
Adenosina Desaminasa , Edición de ARN , Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Ascomicetos/genética , ARN de Hongos/genética , ARN de Hongos/metabolismo , Adenosina/metabolismo , Adenosina/genética , Inosina/metabolismo , Inosina/genética , Fusarium/genética , Neurospora crassa/genética
11.
Viruses ; 16(6)2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38932237

RESUMEN

The genomes of positive-sense (+) single-stranded RNA (ssRNA) viruses are believed to be subjected to a wide range of RNA modifications. In this study, we focused on the chikungunya virus (CHIKV) as a model (+) ssRNA virus to study the landscape of viral RNA modification in infected human cells. Among the 32 distinct RNA modifications analysed by mass spectrometry, inosine was found enriched in the genomic CHIKV RNA. However, orthogonal validation by Illumina RNA-seq analyses did not identify any inosine modification along the CHIKV RNA genome. Moreover, CHIKV infection did not alter the expression of ADAR1 isoforms, the enzymes that catalyse the adenosine to inosine conversion. Together, this study highlights the importance of a multidisciplinary approach to assess the presence of RNA modifications in viral RNA genomes.


Asunto(s)
Virus Chikungunya , Genoma Viral , Procesamiento Postranscripcional del ARN , ARN Viral , Transcriptoma , Virus Chikungunya/genética , Humanos , ARN Viral/genética , ARN Viral/metabolismo , Fiebre Chikungunya/virología , Inosina/metabolismo , Inosina/genética , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Adenosina/metabolismo , Adenosina Desaminasa
12.
Fly (Austin) ; 18(1): 2367359, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38889318

RESUMEN

Adenosine-to-inosine (A-to-I) RNA editing recodes the genome and confers flexibility for the organisms to adapt to the environment. It is believed that RNA recoding sites are well suited for facilitating adaptive evolution by increasing the proteomic diversity in a temporal-spatial manner. The function and essentiality of a few conserved recoding sites are recognized. However, the experimentally discovered functional sites only make up a small corner of the total sites, and there is still the need to expand the repertoire of such functional sites with bioinformatic approaches. In this study, we define a new category of RNA editing sites termed 'conserved editing with non-conserved recoding' and systematically identify such sites in Drosophila editomes, figuring out their selection pressure and signals of adaptation at inter-species and intra-species levels. Surprisingly, conserved editing sites with non-conserved recoding are not suppressed and are even slightly overrepresented in Drosophila. DNA mutations leading to such cases are also favoured during evolution, suggesting that the function of those recoding events in different species might be diverged, specialized, and maintained. Finally, structural prediction suggests that such recoding in potassium channel Shab might increase ion permeability and compensate the effect of low temperature. In conclusion, conserved editing with non-conserved recoding might be functional as well. Our study provides novel aspects in considering the adaptive evolution of RNA editing sites and meanwhile expands the candidates of functional recoding sites for future validation.


Asunto(s)
Adenosina , Drosophila , Inosina , Edición de ARN , Animales , Inosina/metabolismo , Inosina/genética , Drosophila/genética , Drosophila/metabolismo , Adenosina/metabolismo , Adenosina/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Evolución Molecular , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo
13.
Virus Res ; 346: 199413, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38848818

RESUMEN

The conversion of Adenosine (A) to Inosine (I), by Adenosine Deaminases Acting on RNA or ADARs, is an essential post-transcriptional modification that contributes to proteome diversity and regulation in metazoans including humans. In addition to its transcriptome-regulating role, ADARs also play a major part in immune response to viral infection, where an interferon response activates interferon-stimulated genes, such as ADARp150, in turn dynamically regulating host-virus interactions. A previous report has shown that infection from reoviruses, despite strong activation of ADARp150, does not influence the editing of some of the major known editing targets, while likely editing others, suggesting a potentially nuanced editing pattern that may depend on different factors. However, the results were based on a handful of selected editing sites and did not cover the entire transcriptome. Thus, to determine whether and how reovirus infection specifically affects host ADAR editing patterns, we analyzed a publicly available deep-sequenced RNA-seq dataset, from murine fibroblasts infected with wild-type and mutant reovirus strains that allowed us to examine changes in editing patterns on a transcriptome-wide scale. To the best of our knowledge, this is the first transcriptome-wide report on host editing changes after reovirus infection. Our results demonstrate that reovirus infection induces unique nuanced editing changes in the host, including introducing sites uniquely edited in infected samples. Genes with edited sites are overrepresented in pathways related to immune regulation, cellular signaling, metabolism, and growth. Moreover, a shift in editing targets has also been observed, where the same genes are edited in infection and control conditions but at different sites, or where the editing rate is increased for some and decreased for other differential targets, supporting the hypothesis of dynamic and condition-specific editing by ADARs.


Asunto(s)
Adenosina Desaminasa , Fibroblastos , Inosina , Edición de ARN , Transcriptoma , Animales , Ratones , Fibroblastos/virología , Fibroblastos/metabolismo , Inosina/metabolismo , Inosina/genética , Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo , Adenosina/metabolismo , Adenosina/genética , Infecciones por Reoviridae/virología , Infecciones por Reoviridae/genética , Interacciones Huésped-Patógeno , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Reoviridae/genética , Reoviridae/fisiología
14.
Anal Chem ; 96(21): 8730-8739, 2024 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-38743814

RESUMEN

Adenosine-to-inosine (A-to-I) editing and N6-methyladenosine (m6A) modifications are pivotal RNA modifications with widespread functional significance in physiological and pathological processes. Although significant effort has been dedicated to developing methodologies for identifying and quantifying these modifications, traditional approaches have often focused on each modification independently, neglecting the potential co-occurrence of A-to-I editing and m6A modifications at the same adenosine residues. This limitation has constrained our understanding of the intricate regulatory mechanisms governing RNA function and the interplay between different types of RNA modifications. To address this gap, we introduced an innovative technique called deamination-assisted reverse transcription stalling (DARTS), specifically designed for the simultaneous quantification of A-to-I editing and m6A at the same RNA sites. DARTS leverages the selective deamination activity of the engineered TadA-TadA8e protein, which converts adenosine residues to inosine, in combination with the unique property of Bst 2.0 DNA polymerase, which stalls when encountering inosine during reverse transcription. This approach enables the accurate quantification of A-to-I editing, m6A, and unmodified adenosine at identical RNA sites. The DARTS method is remarkable for its ability to directly quantify two distinct types of RNA modifications simultaneously, a capability that has remained largely unexplored in the field of RNA biology. By facilitating a comprehensive analysis of the co-occurrence and interaction between A-to-I editing and m6A modifications, DARTS opens new avenues for exploring the complex regulatory networks modulated by different RNA modifications.


Asunto(s)
Adenosina , Inosina , Edición de ARN , Adenosina/análogos & derivados , Adenosina/análisis , Adenosina/metabolismo , Inosina/metabolismo , Inosina/análogos & derivados , Inosina/química , Desaminación , ARN/metabolismo , ARN/genética , ARN/análisis , Transcripción Reversa , Humanos
15.
Food Res Int ; 183: 114208, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38760138

RESUMEN

To explore the underlying mechanisms by which superchilling (SC, -3 °C within 5 h of slaughter) improves beef tenderness, an untargeted metabolomics strategy was employed. M. Longissimus lumborum (LL) muscles from twelve beef carcasses were assigned to either SC or very fast chilling (VFC, 0 °C within 5 h of slaughter) treatments, with conventional chilling (CC, 0 âˆ¼ 4 °C until 24 h post-mortem) serving as the control (6 per group). Biochemical properties and metabolites were investigated during the early post-mortem period. The results showed that the degradation of µ-calpain and caspase 3 occurred earlier in SC treated sample, which might be attributed to the accelerated accumulation of free Ca2+. The metabolomic profiles of samples from the SC and CC treatments were clearly distinguished based on partial least squares-discriminant analysis (PLS-DA) at each time point. It is noteworthy that more IMP and 4-hydroxyproline were found in the comparison between SC and CC treatments. According to the results of metabolic pathways analysis and the correlation analysis between traits related to tenderness and metabolites with significant differences (SC vs. CC), it can be suggested that the tenderization effect of the SC treatment may be related to the alteration of arginine and proline metabolism, and purine metabolism in the early post-mortem phase.


Asunto(s)
Metabolómica , Músculo Esquelético , Carne Roja , Animales , Bovinos , Calpaína/metabolismo , Caspasa 3/metabolismo , Frío , Análisis Discriminante , Manipulación de Alimentos/métodos , Inosina/metabolismo , Inosina/análisis , Análisis de los Mínimos Cuadrados , Cromatografía Líquida con Espectrometría de Masas , Metabolómica/métodos , Músculo Esquelético/metabolismo , Músculo Esquelético/química , Cambios Post Mortem , Prolina/metabolismo , Carne Roja/análisis
16.
Neuron ; 112(15): 2558-2580.e13, 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-38761794

RESUMEN

Neurodegenerative diseases are commonly classified as proteinopathies that are defined by the aggregation of a specific protein. Parkinson's disease (PD) and dementia with Lewy bodies (DLB) are classified as synucleinopathies since α-synuclein (α-syn)-containing inclusions histopathologically define these diseases. Unbiased biochemical analysis of PD and DLB patient material unexpectedly revealed novel pathological inclusions in the nucleus comprising adenosine-to-inosine (A-to-I)-edited mRNAs and NONO and SFPQ proteins. These inclusions showed no colocalization with Lewy bodies and accumulated at levels comparable to α-syn. NONO and SFPQ aggregates reduced the expression of the editing inhibitor ADAR3, increasing A-to-I editing mainly within human-specific, Alu-repeat regions of axon, synaptic, and mitochondrial transcripts. Inosine-containing transcripts aberrantly accumulated in the nucleus, bound tighter to recombinant purified SFPQ in vitro, and potentiated SFPQ aggregation in human dopamine neurons, resulting in a self-propagating pathological state. Our data offer new insight into the inclusion composition and pathophysiology of PD and DLB.


Asunto(s)
Enfermedad por Cuerpos de Lewy , Factor de Empalme Asociado a PTB , Enfermedad de Parkinson , Edición de ARN , Humanos , Enfermedad por Cuerpos de Lewy/metabolismo , Enfermedad por Cuerpos de Lewy/patología , Enfermedad por Cuerpos de Lewy/genética , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Factor de Empalme Asociado a PTB/metabolismo , Factor de Empalme Asociado a PTB/genética , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Inosina/metabolismo , Adenosina/metabolismo , Núcleo Celular/metabolismo , Masculino , Anciano , Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo , Femenino , ARN Mensajero/metabolismo , alfa-Sinucleína/metabolismo , alfa-Sinucleína/genética , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Anciano de 80 o más Años
17.
J Anim Sci ; 1022024 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-38738625

RESUMEN

Inosine monphosphate (IMP) is one of the important indicators for evaluating meat flavor, and long noncoding RNAs (lncRNAs) play an important role in its transcription and post-transcriptional regulation. Currently, there is little information about how lncRNA regulates the specific deposition of IMP in chicken muscle. In this study, we used transcriptome sequencing to analyze the lncRNAs of the breast and leg muscles of the Jingyuan chicken and identified a total of 357 differentially expressed lncRNAs (DELs), of which 158 were up-regulated and 199 were down-regulated. There were 2,203 and 7,377 cis- and trans-regulated target genes of lncRNAs, respectively, and we identified the lncRNA target genes that are involved in NEGF signaling pathway, glycolysis/glucoseogenesis, and biosynthesis of amino acids pathways. Meanwhile, 621 pairs of lncRNA-miRNA-mRNA interaction networks were constructed with target genes involved in purine metabolism, fatty acid metabolism, and biosynthesis of amino acids. Next, three interacting meso-networks gga-miR-1603-LNC_000324-PGM1, gga-miR-1768-LNC_000324-PGM1, and gga-miR-21-LNC_011339-AMPD1 were identified as closely associated with IMP-specific deposition. Both differentially expressed genes (DEGs) PGM1 and AMPD1 were significantly enriched in IMP synthesis and metabolism-related pathways, and participated in the anabolic process of IMP in the form of organic matter synthesis and energy metabolism. This study obtained lncRNAs and target genes affecting IMP-specific deposition in Jingyuan chickens based on transcriptome analysis, which deepened our insight into the role of lncRNAs in chicken meat quality.


Jingyuan chicken is an excellent local chicken breed listed in the Catalogue of Livestock and Poultry Genetic Resources of China. Its unique growing environment has enabled Jingyuan chicken to develop the characteristics of compact meat, unique flavor, and high nutritional value, which makes it the first choice for chicken food. Inosine monophosphate (IMP) is widely recognized as an important indicator for evaluating the flavor of livestock and poultry meat. To mine potential long noncoding RNAs (lncRNAs) and their regulatory IMP-specific deposition interaction networks, we used transcriptome sequencing to identify 357 lncRNAs that were differentially expressed in breast and leg muscles of 180-d-old Jingyuan hens. We screened the key lncRNAs affecting IMP and three lncRNA-miRNA-mRNA regulatory networks by bioinformatics methods. This provides a new approach to studying IMP-specific deposition, improvement of chicken meat flavor, and breed improvement in Jingyuan chickens.


Asunto(s)
Pollos , Perfilación de la Expresión Génica , Inosina Monofosfato , ARN Largo no Codificante , Animales , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Pollos/genética , Pollos/metabolismo , Inosina Monofosfato/metabolismo , Transcriptoma , MicroARNs/genética , MicroARNs/metabolismo , Carne/análisis , Inosina/metabolismo , Inosina/genética , Músculo Esquelético/metabolismo , Regulación de la Expresión Génica
18.
BMC Genomics ; 25(1): 431, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38693480

RESUMEN

Ophthalmic manifestations have recently been observed in acute and post-acute complications of COVID-19 caused by SARS-CoV-2 infection. Our precious study has shown that host RNA editing is linked to RNA viral infection, yet ocular adenosine to inosine (A-to-I) RNA editing during SARS-CoV-2 infection remains uninvestigated in COVID-19. Herein we used an epitranscriptomic pipeline to analyze 37 samples and investigate A-to-I editing associated with SARS-CoV-2 infection, in five ocular tissue types including the conjunctiva, limbus, cornea, sclera, and retinal organoids. Our results revealed dramatically altered A-to-I RNA editing across the five ocular tissues. Notably, the transcriptome-wide average level of RNA editing was increased in the cornea but generally decreased in the other four ocular tissues. Functional enrichment analysis showed that differential RNA editing (DRE) was mainly in genes related to ubiquitin-dependent protein catabolic process, transcriptional regulation, and RNA splicing. In addition to tissue-specific RNA editing found in each tissue, common RNA editing was observed across different tissues, especially in the innate antiviral immune gene MAVS and the E3 ubiquitin-protein ligase MDM2. Analysis in retinal organoids further revealed highly dynamic RNA editing alterations over time during SARS-CoV-2 infection. Our study thus suggested the potential role played by RNA editing in ophthalmic manifestations of COVID-19, and highlighted its potential transcriptome impact, especially on innate immunity.


Asunto(s)
COVID-19 , Edición de ARN , SARS-CoV-2 , Humanos , COVID-19/genética , COVID-19/virología , SARS-CoV-2/genética , Adenosina/metabolismo , Inosina/metabolismo , Inosina/genética , Transcriptoma , Ojo/metabolismo , Ojo/virología
19.
BMC Biol ; 22(1): 106, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38715001

RESUMEN

BACKGROUND: The significance of A-to-I RNA editing in nervous system development is widely recognized; however, its influence on retina development remains to be thoroughly understood. RESULTS: In this study, we performed RNA sequencing and ribosome profiling experiments on developing mouse retinas to characterize the temporal landscape of A-to-I editing. Our findings revealed temporal changes in A-to-I editing, with distinct editing patterns observed across different developmental stages. Further analysis showed the interplay between A-to-I editing and alternative splicing, with A-to-I editing influencing splicing efficiency and the quantity of splicing events. A-to-I editing held the potential to enhance translation diversity, but this came at the expense of reduced translational efficiency. When coupled with splicing, it could produce a coordinated effect on gene translation. CONCLUSIONS: Overall, this study presents a temporally resolved atlas of A-to-I editing, connecting its changes with the impact on alternative splicing and gene translation in retina development.


Asunto(s)
Biosíntesis de Proteínas , Edición de ARN , Retina , Animales , Ratones , Retina/metabolismo , Retina/embriología , Empalme Alternativo , Inosina/metabolismo , Inosina/genética , Adenosina/metabolismo
20.
Nat Commun ; 15(1): 4049, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38744925

RESUMEN

Nanopore direct RNA sequencing (DRS) has emerged as a powerful tool for RNA modification identification. However, concurrently detecting multiple types of modifications in a single DRS sample remains a challenge. Here, we develop TandemMod, a transferable deep learning framework capable of detecting multiple types of RNA modifications in single DRS data. To train high-performance TandemMod models, we generate in vitro epitranscriptome datasets from cDNA libraries, containing thousands of transcripts labeled with various types of RNA modifications. We validate the performance of TandemMod on both in vitro transcripts and in vivo human cell lines, confirming its high accuracy for profiling m6A and m5C modification sites. Furthermore, we perform transfer learning for identifying other modifications such as m7G, Ψ, and inosine, significantly reducing training data size and running time without compromising performance. Finally, we apply TandemMod to identify 3 types of RNA modifications in rice grown in different environments, demonstrating its applicability across species and conditions. In summary, we provide a resource with ground-truth labels that can serve as benchmark datasets for nanopore-based modification identification methods, and TandemMod for identifying diverse RNA modifications using a single DRS sample.


Asunto(s)
Oryza , Análisis de Secuencia de ARN , Humanos , Análisis de Secuencia de ARN/métodos , Oryza/genética , Procesamiento Postranscripcional del ARN , Nanoporos , ARN/genética , ARN/metabolismo , Secuenciación de Nanoporos/métodos , Aprendizaje Profundo , Inosina/metabolismo , Inosina/genética , Transcriptoma/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA