Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 164
Filtrar
1.
Front Immunol ; 12: 748519, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34777360

RESUMEN

Inherited defects that abrogate the function of the adenosine deaminase (ADA) enzyme and consequently lead to the accumulation of toxic purine metabolites cause profound lymphopenia and severe combined immune deficiency. Additionally, neutropenia and impaired neutrophil function have been reported among ADA-deficient patients. However, due to the rarity of the disorder, the neutrophil developmental abnormalities and the mechanisms contributing to them have not been characterized. Induced pluripotent stem cells (iPSC) generated from two unrelated ADA-deficient patients and from healthy controls were differentiated through embryoid bodies into neutrophils. ADA deficiency led to a significant reduction in the number of all early multipotent hematopoietic progenitors. At later stages of differentiation, ADA deficiency impeded the formation of granulocyte colonies in methylcellulose cultures, leading to a significant decrease in the number of neutrophils generated from ADA-deficient iPSCs. The viability and apoptosis of ADA-deficient neutrophils isolated from methylcellulose cultures were unaffected, suggesting that the abnormal purine homeostasis in this condition interferes with differentiation or proliferation. Additionally, there was a significant increase in the percentage of hyperlobular ADA-deficient neutrophils, and these neutrophils demonstrated significantly reduced ability to phagocytize fluorescent microspheres. Supplementing iPSCs and methylcellulose cultures with exogenous ADA, which can correct adenosine metabolism, reversed all abnormalities, cementing the critical role of ADA in neutrophil development. Moreover, chemical inhibition of the ribonucleotide reductase (RNR) enzyme, using hydroxyurea or a combination of nicotinamide and trichostatin A in iPSCs from healthy controls, led to abnormal neutrophil differentiation similar to that observed in ADA deficiency, implicating RNR inhibition as a potential mechanism for the neutrophil abnormalities. In conclusion, the findings presented here demonstrate the important role of ADA in the development and function of neutrophils while clarifying the mechanisms responsible for the neutrophil abnormalities in ADA-deficient patients.


Asunto(s)
Adenosina Desaminasa/fisiología , Agammaglobulinemia/inmunología , Células Madre Pluripotentes Inducidas/citología , Neutrófilos/citología , Inmunodeficiencia Combinada Grave/inmunología , Adenosina Desaminasa/genética , Células Cultivadas , Cuerpos Embrioides/citología , Fibroblastos/enzimología , Granulocitos/citología , Humanos , Ácidos Hidroxámicos/farmacología , Hidroxiurea/farmacología , Lactante , Masculino , Mutación Missense , Mielopoyesis , Niacinamida/farmacología , Mutación Puntual , Ribonucleótido Reductasas/antagonistas & inhibidores
2.
Int J Mol Sci ; 22(21)2021 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-34768866

RESUMEN

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.


Asunto(s)
Adenosina Desaminasa/metabolismo , Adenosina Desaminasa/fisiología , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/fisiología , Adenosina , Animales , ADN de Forma Z/metabolismo , ADN de Forma Z/fisiología , Humanos , Inosina , Interferón Tipo I/metabolismo , Helicasa Inducida por Interferón IFIH1/genética , Ratones , Isoformas de Proteínas/metabolismo , Edición de ARN/fisiología , ARN Bicatenario
3.
Autoimmun Rev ; 20(1): 102709, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33197575

RESUMEN

Autoimmune diseases patients are characterized by the autoimmune disorders, whose immune system can't distinguish between auto- and foreign- antigens. Thus, Immune homeostasis disorder is the key factor for autoimmune diseases development. Adenosine deaminase (ADA) is the degrading enzyme for an immunosuppressive signal - adenosine, and play an important role in immune homeostasis regulation. Increasing evidences have shown that ADA is involved in various autoimmune diseases. ADA activity were changed in multiple autoimmune diseases patients and could be served as a biomarker for clinical diagnosis. In this study, we analyze the change of ADA activity in patients with autoimmune diseases, and we underline its potential diagnostic value for autoimmune diseases patients.


Asunto(s)
Adenosina Desaminasa , Enfermedades Autoinmunes , Adenosina Desaminasa/fisiología , Enfermedades Autoinmunes/diagnóstico , Enfermedades Autoinmunes/metabolismo , Biomarcadores , Humanos
4.
Methods Mol Biol ; 2181: 331-349, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32729089

RESUMEN

RNA editing activity can be exploited for the restoration of disease-causing nonsense and missense mutations and as a tool to manipulate the transcriptome in a simple and programmable way. The general concept is called site-directed RNA editing and has high potential for translation into the clinics. Due to its different mode of action RNA editing may well complement gene editing and other gene therapy options. In this method chapter, we particularly highlight RNA editing strategies that harness endogenous ADARs. Such strategies circumvent the delivery and expression of engineered editases and are notably precise and simple. This is particularly true if endogenous ADARs are recruited with chemically modified antisense oligonucleotides, an approach we call RESTORE (recruiting endogenous ADAR to specific transcripts for oligonucleotide-mediated RNA editing). To foster the research and development of RESTORE we now report a detailed protocol for the procedure of editing reactions, and a protocol for the generation of partly chemically modified RESTORE ASOs with a combination of in vitro transcription and ligation.


Asunto(s)
Adenosina Desaminasa/fisiología , Mutagénesis Sitio-Dirigida/métodos , Edición de ARN/fisiología , Proteínas de Unión al ARN/fisiología , Células A549 , Adenosina Desaminasa/genética , Células Cultivadas , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Mutagénesis Sitio-Dirigida/tendencias , Proteínas de Unión al ARN/genética
5.
J Biol Chem ; 295(52): 18199-18212, 2020 12 25.
Artículo en Inglés | MEDLINE | ID: mdl-33100268

RESUMEN

Post-transcriptional modifications of pre-mRNAs expand the diversity of proteomes in higher eukaryotes. In the brain, these modifications diversify the functional output of many critical neuronal signal molecules. In this study, we identified a brain-specific A-to-I RNA editing that changed glutamine to arginine (Q/R) at exon 20 and an alternative splicing of exon 4 in Tmem63b, which encodes a ubiquitously expressed osmosensitive cation channel. The channel isoforms lacking exon 4 occurred in ∼80% of Tmem63b mRNAs in the brain but were not detected in other tissues, suggesting a brain-specific splicing. We found that the Q/R editing was catalyzed by Adar2 (Adarb1) and required an editing site complementary sequence located in the proximal 5' end of intron 20. Moreover, the Q/R editing was almost exclusively identified in the splicing isoform lacking exon 4, indicating a coupling between the editing and the splicing. Elimination of the Q/R editing in brain-specific Adar2 knockout mice did not affect the splicing efficiency of exon 4. Furthermore, transfection with the splicing isoform containing exon 4 suppressed the Q/R editing in primary cultured cerebellar granule neurons. Thus, our study revealed a coupling between an RNA editing and a distant alternative splicing in the Tmem63b pre-mRNA, in which the splicing plays a dominant role. Finally, physiological analysis showed that the splicing and the editing coordinately regulate Ca2+ permeability and osmosensitivity of channel proteins, which may contribute to their functions in the brain.


Asunto(s)
Adenosina Desaminasa/fisiología , Empalme Alternativo , Encéfalo/metabolismo , Canales de Calcio/genética , Exones , Edición de ARN , Precursores del ARN/genética , Proteínas de Unión al ARN/fisiología , Animales , Canales de Calcio/metabolismo , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
6.
Genet Sel Evol ; 52(1): 35, 2020 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-32611306

RESUMEN

Base editing has the potential to improve important economic traits in agriculture and can precisely convert single nucleotides in DNA or RNA sequences into minimal double-strand DNA breaks (DSB). Adenine base editors (ABE) have recently emerged as a base editing tool for the conversion of targeted A:T to G:C, but have not yet been used in sheep. ABEmax is one of the latest versions of ABE, which consists of a catalytically-impaired nuclease and a laboratory-evolved DNA-adenosine deaminase. The Booroola fecundity (FecBB) mutation (g.A746G, p.Q249R) in the bone morphogenetic protein receptor 1B (BMPR1B) gene influences fecundity in many sheep breeds. In this study, by using ABEmax we successfully obtained lambs with defined point mutations that result in an amino acid substitution (p.Gln249Arg). The efficiency of the defined point mutations was 75% in newborn lambs, since six lambs were heterozygous at the FecBB mutation site (g.A746G, p.Q249R), and two lambs were wild-type. We did not detect off-target mutations in the eight edited lambs. Here, we report the validation of the first gene-edited sheep generated by ABE and highlight its potential to improve economically important traits in livestock.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Fertilidad/genética , Edición Génica/métodos , Adenina/metabolismo , Adenosina Desaminasa/metabolismo , Adenosina Desaminasa/fisiología , Animales , Cruzamiento , Femenino , Ingeniería Genética/métodos , Genotipo , Heterocigoto , Tamaño de la Camada/genética , Masculino , Mutación , Fenotipo , Polimorfismo de Nucleótido Simple , Embarazo , Ovinos/genética
7.
Nat Neurosci ; 23(6): 718-729, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32367065

RESUMEN

DNA forms conformational states beyond the right-handed double helix; however, the functional relevance of these noncanonical structures in the brain remains unknown. Here we show that, in the prefrontal cortex of mice, the formation of one such structure, Z-DNA, is involved in the regulation of extinction memory. Z-DNA is formed during fear learning and reduced during extinction learning, which is mediated, in part, by a direct interaction between Z-DNA and the RNA-editing enzyme Adar1. Adar1 binds to Z-DNA during fear extinction learning, which leads to a reduction in Z-DNA at sites where Adar1 is recruited. Knockdown of Adar1 leads to an inability to modify a previously acquired fear memory and blocks activity-dependent changes in DNA structure and RNA state-effects that are fully rescued by the introduction of full-length Adar1. These findings suggest a new mechanism of learning-induced gene regulation that is dependent on proteins that recognize alternate DNA structure states, which are required for memory flexibility.


Asunto(s)
Adenosina Desaminasa/metabolismo , Adenosina Desaminasa/fisiología , ADN de Forma Z/fisiología , Extinción Psicológica/fisiología , Edición de ARN/fisiología , Animales , ADN de Forma Z/metabolismo , Miedo , Aprendizaje/fisiología , Ratones , Corteza Prefrontal/metabolismo , ARN Interferente Pequeño/farmacología
9.
Brain Res ; 1735: 146760, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32142720

RESUMEN

Ca2+-permeable AMPA receptors (AMPAR) which crucially modify maturational programs of the developing brain are involved in seizure-induced glutamate excitotoxicity and apoptosis. Regulatory effects on AMPAR subunit composition and RNA-editing in the developing brain and their significance as therapeutic targets are not well understood. Here, we analyzed acute effects of recurrent pilocarpine-induced neonatal seizures on age- and region-specific expression of AMPAR subunits and adenosine deaminases (ADAR) in the developing mouse brain (P10). After recurrent seizure activity and regeneration periods of 6-72 h cerebral mRNA levels of GluR (glutamate receptor subunit) 1, GluR2, GluR3, and GluR4 were unaffected compared to controls. However, ratio of GluR2 and GluR4 to pooled GluR1-4 mRNA concentration significantly decreased in seizure-exposed brains in comparison to controls. After a regeneration period of 24-72 h ADAR1 and ADAR2 mRNA expression was significantly lower in seizure-exposed brains than in those of controls. This was confirmed at the protein level in the hippocampal CA3 region. We observed a regionally increased apoptosis (TUNEL+ and CC3+ cells) in the hippocampus, parietal cortex and subventricular zone of seizure-exposed brains in comparison to controls. Together, present in vivo data demonstrate the maturational age-specific, functional role of RNA-edited GluR2 in seizure-induced excitotoxicity in the developing mouse brain. In response to recurrent seizure activity, we observed reduced expression of GluR2 and the GluR2 mRNA-editing enzymes ADAR1 and ADAR2 accompanied by increased apoptosis in a region-specific manner. Thus, AMPA receptor subtype-specific mRNA editing is assessed as a promising target of novel neuroprotective treatment strategies in consideration of age-related developmental mechanisms.


Asunto(s)
Receptores AMPA/metabolismo , Convulsiones/fisiopatología , Adenosina Desaminasa/metabolismo , Adenosina Desaminasa/fisiología , Factores de Edad , Animales , Apoptosis/genética , Apoptosis/fisiología , Encéfalo/metabolismo , Femenino , Expresión Génica/genética , Ácido Glutámico/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Neuronas/fisiología , ARN/metabolismo , ARN Mensajero/metabolismo , Receptores AMPA/fisiología , Transcriptoma/genética
10.
Brain ; 142(3): 586-605, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30698736

RESUMEN

As clinical evidence supports a negative impact of dysfunctional energy metabolism on the disease progression in amyotrophic lateral sclerosis, it is vital to understand how the energy metabolic pathways are altered and whether they can be restored to slow disease progression. Possible approaches include increasing or rerouting catabolism of alternative fuel sources to supplement the glycolytic and mitochondrial pathways such as glycogen, ketone bodies and nucleosides. To analyse the basis of the catabolic defect in amyotrophic lateral sclerosis we used a novel phenotypic metabolic array. We profiled fibroblasts and induced neuronal progenitor-derived human induced astrocytes from C9orf72 amyotrophic lateral sclerosis patients compared to normal controls, measuring the rates of production of reduced nicotinamide adenine dinucleotides from 91 potential energy substrates. This approach shows for the first time that C9orf72 human induced astrocytes and fibroblasts have an adenosine to inosine deamination defect caused by reduction of adenosine deaminase, which is also observed in induced astrocytes from sporadic patients. Patient-derived induced astrocyte lines were more susceptible to adenosine-induced toxicity, which could be mimicked by inhibiting adenosine deaminase in control lines. Furthermore, adenosine deaminase inhibition in control induced astrocytes led to increased motor neuron toxicity in co-cultures, similar to the levels observed with patient derived induced astrocytes. Bypassing metabolically the adenosine deaminase defect by inosine supplementation was beneficial bioenergetically in vitro, increasing glycolytic energy output and leading to an increase in motor neuron survival in co-cultures with induced astrocytes. Inosine supplementation, in combination with modulation of the level of adenosine deaminase may represent a beneficial therapeutic approach to evaluate in patients with amyotrophic lateral sclerosis.


Asunto(s)
Adenosina Desaminasa/metabolismo , Esclerosis Amiotrófica Lateral/metabolismo , Neuronas Motoras/metabolismo , Adenosina Desaminasa/fisiología , Adulto , Esclerosis Amiotrófica Lateral/fisiopatología , Animales , Astrocitos/metabolismo , Proteína C9orf72/metabolismo , Muerte Celular , Supervivencia Celular , Células Cultivadas , Técnicas de Cocultivo , Progresión de la Enfermedad , Metabolismo Energético/fisiología , Femenino , Fibroblastos/metabolismo , Humanos , Inosina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Ratas , Ratas Sprague-Dawley , Células Madre/metabolismo
11.
Oncol Res ; 27(2): 237-251, 2019 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-29673423

RESUMEN

The aim of this study was to investigate the inhibition effects of cordycepin and its derivatives on endometrial cancer cell growth. Cytotoxicity MTT assays, clonogenic assays, and flow cytometry were used to observe the effects on apoptosis and regulation of the cell cycle of Ishikawa cells under various concentrations of cordycepin, cisplatin, and combinations of the two. Validated in silico docking simulations were performed on 31 cordycepin derivatives against adenosine deaminase (ADA) to predict their binding affinities and hence their potential tendency to be metabolized by ADA. Cordycepin has a significant dose-dependent inhibitory effect on cell proliferation. The combination of cordycepin and cisplatin produced greater inhibition effects than did cordycepin alone. Apoptosis investigations confirmed the ability of cordycepin to induce the apoptosis of Ishikawa cells. The in silico results indicate that compound MRS5698 is least metabolized by ADA and has acceptable drug likeness and safety profiles. This is the first study to confirm the cytotoxic effects of cordycepin on endometrial cancer cells. This study also identified cordycepin derivatives with promising pharmacological and pharmacokinetic properties for further investigation in the development of new treatments for endometrial cancer.


Asunto(s)
Adenosina/análogos & derivados , Desoxiadenosinas/farmacología , Neoplasias Endometriales/tratamiento farmacológico , Adenosina/farmacología , Adenosina Desaminasa/fisiología , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Cisplatino/farmacología , Desoxiadenosinas/metabolismo , Desoxiadenosinas/uso terapéutico , Neoplasias Endometriales/patología , Femenino , Humanos , Simulación del Acoplamiento Molecular
12.
PLoS Biol ; 16(11): e2006577, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30496178

RESUMEN

The interferon (IFN)-mediated innate immune response is the first line of defense against viruses. However, an IFN-stimulated gene, the adenosine deaminase acting on RNA 1 (ADAR1), favors the replication of several viruses. ADAR1 binds double-stranded RNA and converts adenosine to inosine by deamination. This form of editing makes duplex RNA unstable, thereby preventing IFN induction. To better understand how ADAR1 works at the cellular level, we generated cell lines that express exclusively either the IFN-inducible, cytoplasmic isoform ADAR1p150, the constitutively expressed nuclear isoform ADAR1p110, or no isoform. By comparing the transcriptome of these cell lines, we identified more than 150 polymerase II transcripts that are extensively edited, and we attributed most editing events to ADAR1p150. Editing is focused on inverted transposable elements, located mainly within introns and untranslated regions, and predicted to form duplex RNA structures. Editing of these elements occurs also in primary human samples, and there is evidence for cross-species evolutionary conservation of editing patterns in primates and, to a lesser extent, in rodents. Whereas ADAR1p150 rarely edits tightly encapsidated standard measles virus (MeV) genomes, it efficiently edits genomes with inverted repeats accidentally generated by a mutant MeV. We also show that immune activation occurs in fully ADAR1-deficient (ADAR1KO) cells, restricting virus growth, and that complementation of these cells with ADAR1p150 rescues virus growth and suppresses innate immunity activation. Finally, by knocking out either protein kinase R (PKR) or mitochondrial antiviral signaling protein (MAVS)-another protein controlling the response to duplex RNA-in ADAR1KO cells, we show that PKR activation elicits a stronger antiviral response. Thus, ADAR1 prevents innate immunity activation by cellular transcripts that include extensive duplex RNA structures. The trade-off is that viruses take advantage of ADAR1 to elude innate immunity control.


Asunto(s)
Adenosina Desaminasa/fisiología , Virus ARN/genética , Proteínas de Unión al ARN/fisiología , Adenosina/metabolismo , Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo , Células HeLa , Humanos , Inmunidad Innata/fisiología , Interferones/metabolismo , Isoformas de Proteínas , Provirus/genética , Provirus/inmunología , Virus ARN/metabolismo , ARN Bicatenario/fisiología , ARN Viral/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Transcriptoma/genética , Virión/genética
13.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 43(8): 904-911, 2018 Aug 28.
Artículo en Chino | MEDLINE | ID: mdl-30197321

RESUMEN

RNA editing, especially A-to-I RNA editing, is a common post-transcriptional modification in mammals. Adenosine deaminase acting on RNA (ADAR) is a key protein for A-to-I editing, which converts the adenosine group of a double-stranded RNA to creatinine group by deaminating it, resulting in a change of nucleotide sequence. There are 3 types of ADARs (ADAR1, ADAR2, ADAR3) that have been found in recent years. The abnormalities of ADARs are closely related to many human diseases such as viral infections, metabolic diseases, nervous system diseases, and tumors.


Asunto(s)
Adenosina Desaminasa/fisiología , Enfermedad/etiología , Edición de ARN/fisiología , Proteínas de Unión al ARN/fisiología , Adenosina/metabolismo , Secuencia de Bases , Creatinina/metabolismo , Desaminación , Humanos , ARN Bicatenario
14.
RNA Biol ; 15(7): 863-867, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30071181

RESUMEN

DNA harbors the blueprint for life. However, the instructions stored in the DNA could be altered at the RNA level before they are executed. One of these processes is RNA editing, which was shown to modify RNA sequences in many organisms. The most abundant modification is the deamination of adenosine (A) into inosine (I). In turn, inosine can be identified as a guanosine (G) by the ribosome and other cellular machineries such as reverse transcriptase. In multicellular organisms, enzymes from the ADAR (adenosine deaminase acting on RNA) family mediate RNA editing in mRNA, whereas enzymes from the ADAT family mediate A-to-I editing on tRNAs. In bacteria however, until recently, only one editing site was described, in tRNAArg, but never in mRNA. The tRNA site was shown to be modified by tadA (tRNA specific adenosine deaminase) which is believed to be the ancestral enzyme for the RNA editing family of enzymes. In our recent work, we have shown for the first time, editing on multiple sites in bacterial mRNAs and identified tadA as the enzyme responsible for this editing activity. Focusing on one of the identified targets - the self-killing toxin hokB, we found that editing is physiologically regulated and that it increases protein activity. Here we discuss possible modes of regulation on hokB editing, potential roles of RNA editing in bacteria, possible implications, and future research directions.


Asunto(s)
Adenosina Desaminasa/fisiología , Klebsiella pneumoniae/enzimología , Edición de ARN/fisiología , ARN Mensajero/metabolismo , Yersinia enterocolitica/enzimología , Adenosina/genética , Toxinas Bacterianas/metabolismo , Desaminación/fisiología , Farmacorresistencia Bacteriana/fisiología , Inosina/genética , ARN de Transferencia/metabolismo , Sistemas Toxina-Antitoxina/fisiología
16.
Bioessays ; 40(2)2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29280160

RESUMEN

RNA editing is a major post-transcriptional mechanism that changes specific nucleotides at the RNA level. The most common RNA editing type in humans is adenosine (A) to inosine (I) editing, which is mediated by ADAR enzymes. RNA editing events can not only change amino acids in proteins, but also affect the functions of non-coding RNAs such as miRNAs. Recent studies have characterized thousands of miRNA RNA editing events across different cancer types. Importantly, individual cases of miRNA editing have been reported to play a role in cancer development. In this review, we summarize the current knowledge of miRNA editing in cancer, and discuss the mechanisms on how miRNA-related editing events modulate the initiation and progression of human cancer. Finally, we discuss the challenges and future directions of studying miRNA editing in cancer.


Asunto(s)
MicroARNs/genética , Neoplasias/genética , Edición de ARN/genética , Adenosina/genética , Adenosina Desaminasa/genética , Adenosina Desaminasa/fisiología , Animales , Humanos , Inosina/genética , MicroARNs/fisiología , Proteínas de Unión al ARN/fisiología
17.
Pharmacol Ther ; 181: 13-21, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28716651

RESUMEN

RNA editing is a post-transcriptional process that alters the nucleotide sequence of RNA transcripts to generate transcriptome diversity. Among the various types of RNA editing, adenosine-to-inosine (A-to-I) RNA editing is the most frequent type of RNA editing in mammals. Adenosine deaminases acting on RNA (ADAR) enzymes, ADAR1 and ADAR2, convert adenosines in double-stranded RNA structures into inosines by hydrolytic deamination. Inosine forms a base pair with cytidine as if it were guanosine; therefore, the conversion may affect the amino acid sequence, splicing, microRNA targeting, and miRNA maturation. It became apparent that disrupted RNA editing or abnormal ADAR expression is associated with several diseases including cancer, neurological disorders, metabolic diseases, viral infections, and autoimmune disorders. The biological significance of RNA editing in pharmacokinetics/pharmacodynamics (PK/PD)-related genes is starting to be demonstrated. The authors conducted pioneering studies to reveal that RNA editing modulates drug metabolism potencies in the human liver, as well as the response of cancer cells to chemotherapy agents. Awareness of the importance of RNA editing in drug therapy is growing. This review summarizes the current knowledge on the RNA editing that affects the expression and function of drug response-related genes. Continuing studies on the RNA editing that regulates pharmacokinetics/pharmacodynamics would provide new beneficial information for personalized medicine.


Asunto(s)
Adenosina Desaminasa/fisiología , Adenosina/genética , Inactivación Metabólica/genética , Inactivación Metabólica/fisiología , Inosina/genética , Edición de ARN/fisiología , Animales , Resistencia a Medicamentos/fisiología , Humanos , Receptores de Hidrocarburo de Aril/biosíntesis , Tetrahidrofolato Deshidrogenasa/biosíntesis
18.
Genome Biol ; 18(1): 205, 2017 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-29084589

RESUMEN

One of the most prevalent forms of post-transcritpional RNA modification is the conversion of adenosine nucleosides to inosine (A-to-I), mediated by the ADAR family of enzymes. The functional requirement and regulatory landscape for the majority of A-to-I editing events are, at present, uncertain. Recent studies have identified key in vivo functions of ADAR enzymes, informing our understanding of the biological importance of A-to-I editing. Large-scale studies have revealed how editing is regulated both in cis and in trans. This review will explore these recent studies and how they broaden our understanding of the functions and regulation of ADAR-mediated RNA editing.


Asunto(s)
Adenosina Desaminasa/fisiología , Adenosina/metabolismo , Inosina/metabolismo , Edición de ARN , Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo , Animales , Enfermedad/genética , Humanos , Transcriptoma
19.
DNA Cell Biol ; 36(5): 367-376, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28278381

RESUMEN

A-to-I RNA editing, carried out by adenosine deaminase acting on RNA (ADAR) enzymes, is an epigenetic phenomenon of posttranscriptional modifications on pre-mRNA. RNA editing in intronic sequences may influence alternative splicing of flanking exons. We have previously shown that conditions that induce editing result in elevated expression of signal transducer and activator of transcription 3 (STAT3), preferentially the alternatively-spliced STAT3ß isoform. Mechanisms regulating alternative splicing of STAT3 have not been elucidated. STAT3 undergoes A-to-I RNA editing in an intron residing in proximity to the alternatively spliced exon. We hypothesized that RNA editing plays a role in regulating alternative splicing toward STAT3ß. In this study we extend our observation connecting RNA editing to the preferential induction of STAT3ß expression. We study the involvement of ADAR1 in STAT3 editing and reveal the connection between editing and alternative splicing of STAT3. Deferoaxamine treatment caused the induction in STAT3 RNA editing and STAT3ß expression. Silencing ADAR1 caused a decrease in STAT3 editing and expression with a preferential decrease in STAT3ß. Cells transfected with a mutated minigene showed preferential splicing toward the STAT3ß transcript. Editing in the STAT3 intron is performed by ADAR1 and affects STAT3 alternative splicing. These results suggest that RNA editing is one of the molecular mechanisms regulating the expression of STAT3ß.


Asunto(s)
Adenosina Desaminasa/fisiología , Empalme Alternativo , Edición de ARN/fisiología , Proteínas de Unión al ARN/fisiología , Factor de Transcripción STAT3/genética , Empalme Alternativo/genética , Elementos Alu , Secuencia de Bases , Línea Celular Transformada , Regulación Neoplásica de la Expresión Génica , Humanos , Células MCF-7 , Masculino , ARN Bicatenario/genética , ARN Bicatenario/metabolismo , Factor de Transcripción STAT3/metabolismo
20.
Oncotarget ; 7(52): 86161-86173, 2016 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-27863387

RESUMEN

ADAR1, one of adenosine deaminases acting on RNA, modulates RNA transcripts through converting adenosine (A) to inosine (I) by deamination. Emerging evidence has implicated that ADAR1 plays an important role in a few of human cancers, however, its expression and physiological significance in gastric cancer remain undefined. In the present study, we demonstrated that ADAR1 was frequently overexpressed in gastric cancer samples by quantitative real-time PCR analysis. In a gastric cancer tissue microarray, ADAR1 staining was closely correlated with tumor stage (P < 0.001) and N classification (P < 0.001). Functional analysis indicated that ADAR1 overexpression promoted cell proliferation and migration in vitro, whereas ADAR1 knockdown resulted in an opposite phenotypes. Furthermore, ADAR1 knockdown also inhibited tumorigenicity and lung metastasis potential of gastric cancer cells in nude mice models. Mechanistically, ADAR1 expression had a significant effect on phosphorylation level of mTOR, p70S kinase, and S6 ribosomal protein, implying its involvement in the regulation of mTOR signaling pathway. We conclude that ADAR1 contributes to gastric cancer development and progression via activating mTOR/p70S6K/S6 ribosomal protein signaling axis. Our findings suggest that ADAR1 may be a valuable biomarker for GC diagnosis and prognosis and may represent a new novel therapeutic opportunities.


Asunto(s)
Adenosina Desaminasa/fisiología , Proteínas de Unión al ARN/fisiología , Neoplasias Gástricas/etiología , Serina-Treonina Quinasas TOR/fisiología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Progresión de la Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , Proteínas Quinasas S6 Ribosómicas 70-kDa/fisiología , Transducción de Señal/fisiología , Neoplasias Gástricas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA