Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 553
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Cell ; 186(12): 2544-2555.e13, 2023 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-37295402

RESUMEN

In poikilotherms, temperature changes challenge the integration of physiological function. Within the complex nervous systems of the behaviorally sophisticated coleoid cephalopods, these problems are substantial. RNA editing by adenosine deamination is a well-positioned mechanism for environmental acclimation. We report that the neural proteome of Octopus bimaculoides undergoes massive reconfigurations via RNA editing following a temperature challenge. Over 13,000 codons are affected, and many alter proteins that are vital for neural processes. For two highly temperature-sensitive examples, recoding tunes protein function. For synaptotagmin, a key component of Ca2+-dependent neurotransmitter release, crystal structures and supporting experiments show that editing alters Ca2+ binding. For kinesin-1, a motor protein driving axonal transport, editing regulates transport velocity down microtubules. Seasonal sampling of wild-caught specimens indicates that temperature-dependent editing occurs in the field as well. These data show that A-to-I editing tunes neurophysiological function in response to temperature in octopus and most likely other coleoids.


Asunto(s)
Octopodiformes , Proteoma , Animales , Proteoma/metabolismo , Octopodiformes/genética , Edición de ARN , Temperatura , Sistema Nervioso/metabolismo , Adenosina Desaminasa/metabolismo , ARN/metabolismo
2.
Cell ; 172(4): 811-824.e14, 2018 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-29395325

RESUMEN

Type I interferon (IFN) is produced when host sensors detect foreign nucleic acids, but how sensors differentiate self from nonself nucleic acids, such as double-stranded RNA (dsRNA), is incompletely understood. Mutations in ADAR1, an adenosine-to-inosine editing enzyme of dsRNA, cause Aicardi-Goutières syndrome, an autoinflammatory disorder associated with spontaneous interferon production and neurologic sequelae. We generated ADAR1 knockout human cells to explore ADAR1 substrates and function. ADAR1 primarily edited Alu elements in RNA polymerase II (pol II)-transcribed mRNAs, but not putative pol III-transcribed Alus. During the IFN response, ADAR1 blocked translational shutdown by inhibiting hyperactivation of PKR, a dsRNA sensor. ADAR1 dsRNA binding and catalytic activities were required to fully prevent endogenous RNA from activating PKR. Remarkably, ADAR1 knockout neuronal progenitor cells exhibited MDA5 (dsRNA sensor)-dependent spontaneous interferon production, PKR activation, and cell death. Thus, human ADAR1 regulates sensing of self versus nonself RNA, allowing pathogen detection while avoiding autoinflammation.


Asunto(s)
Adenosina Desaminasa/metabolismo , Elementos Alu , Enfermedades Autoinmunes del Sistema Nervioso/metabolismo , Malformaciones del Sistema Nervioso/metabolismo , Células-Madre Neurales/metabolismo , Biosíntesis de Proteínas , ARN Bicatenario/metabolismo , Proteínas de Unión al ARN/metabolismo , Adenosina Desaminasa/genética , Adenosina Desaminasa/inmunología , Enfermedades Autoinmunes del Sistema Nervioso/genética , Enfermedades Autoinmunes del Sistema Nervioso/inmunología , Muerte Celular/genética , Muerte Celular/inmunología , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Helicasa Inducida por Interferón IFIH1/genética , Helicasa Inducida por Interferón IFIH1/inmunología , Helicasa Inducida por Interferón IFIH1/metabolismo , Malformaciones del Sistema Nervioso/genética , Malformaciones del Sistema Nervioso/inmunología , Células-Madre Neurales/citología , Células-Madre Neurales/inmunología , Células-Madre Neurales/patología , ARN Polimerasa II/genética , ARN Polimerasa II/inmunología , ARN Polimerasa II/metabolismo , ARN Bicatenario/genética , ARN Bicatenario/inmunología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/inmunología , eIF-2 Quinasa/genética , eIF-2 Quinasa/inmunología , eIF-2 Quinasa/metabolismo
3.
Immunity ; 56(5): 979-997.e11, 2023 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-37100060

RESUMEN

Immune cell trafficking constitutes a fundamental component of immunological response to tissue injury, but the contribution of intrinsic RNA nucleotide modifications to this response remains elusive. We report that RNA editor ADAR2 exerts a tissue- and stress-specific regulation of endothelial responses to interleukin-6 (IL-6), which tightly controls leukocyte trafficking in IL-6-inflamed and ischemic tissues. Genetic ablation of ADAR2 from vascular endothelial cells diminished myeloid cell rolling and adhesion on vascular walls and reduced immune cell infiltration within ischemic tissues. ADAR2 was required in the endothelium for the expression of the IL-6 receptor subunit, IL-6 signal transducer (IL6ST; gp130), and subsequently, for IL-6 trans-signaling responses. ADAR2-induced adenosine-to-inosine RNA editing suppressed the Drosha-dependent primary microRNA processing, thereby overwriting the default endothelial transcriptional program to safeguard gp130 expression. This work demonstrates a role for ADAR2 epitranscriptional activity as a checkpoint in IL-6 trans-signaling and immune cell trafficking to sites of tissue injury.


Asunto(s)
Interleucina-6 , ARN , Células Endoteliales/metabolismo , Receptor gp130 de Citocinas , Endotelio/metabolismo , Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo
4.
Cell ; 169(2): 191-202.e11, 2017 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-28388405

RESUMEN

RNA editing, a post-transcriptional process, allows the diversification of proteomes beyond the genomic blueprint; however it is infrequently used among animals for this purpose. Recent reports suggesting increased levels of RNA editing in squids thus raise the question of the nature and effects of these events. We here show that RNA editing is particularly common in behaviorally sophisticated coleoid cephalopods, with tens of thousands of evolutionarily conserved sites. Editing is enriched in the nervous system, affecting molecules pertinent for excitability and neuronal morphology. The genomic sequence flanking editing sites is highly conserved, suggesting that the process confers a selective advantage. Due to the large number of sites, the surrounding conservation greatly reduces the number of mutations and genomic polymorphisms in protein-coding regions. This trade-off between genome evolution and transcriptome plasticity highlights the importance of RNA recoding as a strategy for diversifying proteins, particularly those associated with neural function. PAPERCLIP.


Asunto(s)
Evolución Biológica , Cefalópodos/genética , Edición de ARN , Transcriptoma , Adenosina Desaminasa/metabolismo , Secuencia de Aminoácidos , Animales , Cefalópodos/clasificación , Cefalópodos/metabolismo , Sistema Nervioso/metabolismo , Canales de Potasio con Entrada de Voltaje/química , Canales de Potasio con Entrada de Voltaje/genética , Alineación de Secuencia
5.
Mol Cell ; 84(1): 107-119, 2024 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-38118451

RESUMEN

The ability to sense and respond to infection is essential for life. Viral infection produces double-stranded RNAs (dsRNAs) that are sensed by proteins that recognize the structure of dsRNA. This structure-based recognition of viral dsRNA allows dsRNA sensors to recognize infection by many viruses, but it comes at a cost-the dsRNA sensors cannot always distinguish between "self" and "nonself" dsRNAs. "Self" RNAs often contain dsRNA regions, and not surprisingly, mechanisms have evolved to prevent aberrant activation of dsRNA sensors by "self" RNA. Here, we review current knowledge about the life of endogenous dsRNAs in mammals-the biosynthesis and processing of dsRNAs, the proteins they encounter, and their ultimate degradation. We highlight mechanisms that evolved to prevent aberrant dsRNA sensor activation and the importance of competition in the regulation of dsRNA sensors and other dsRNA-binding proteins.


Asunto(s)
ARN Bicatenario , Virosis , Animales , ARN Bicatenario/genética , ARN Helicasas DEAD-box/metabolismo , Inmunidad Innata , Mamíferos/metabolismo
6.
Mol Cell ; 83(21): 3869-3884.e7, 2023 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-37797622

RESUMEN

Effective immunity requires the innate immune system to distinguish foreign nucleic acids from cellular ones. Cellular double-stranded RNAs (dsRNAs) are edited by the RNA-editing enzyme ADAR1 to evade being recognized as viral dsRNA by cytoplasmic dsRNA sensors, including MDA5 and PKR. The loss of ADAR1-mediated RNA editing of cellular dsRNA activates MDA5. Additional RNA-editing-independent functions of ADAR1 have been proposed, but a specific mechanism has not been delineated. We now demonstrate that the loss of ADAR1-mediated RNA editing specifically activates MDA5, whereas loss of the cytoplasmic ADAR1p150 isoform or its dsRNA-binding activity enabled PKR activation. Deleting both MDA5 and PKR resulted in complete rescue of the embryonic lethality of Adar1p150-/- mice to adulthood, contrasting with the limited or no rescue by removing MDA5 or PKR alone. Our findings demonstrate that MDA5 and PKR are the primary in vivo effectors of fatal autoinflammation following the loss of ADAR1p150.


Asunto(s)
Inmunidad Innata , ARN Bicatenario , Animales , Ratones , Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo , Citoplasma/metabolismo , Inmunidad Innata/genética , ARN Bicatenario/genética
7.
Mol Cell ; 83(7): 1180-1196.e8, 2023 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-37028415

RESUMEN

Proper defense against microbial infection depends on the controlled activation of the immune system. This is particularly important for the RIG-I-like receptors (RLRs), which recognize viral dsRNA and initiate antiviral innate immune responses with the potential of triggering systemic inflammation and immunopathology. Here, we show that stress granules (SGs), molecular condensates that form in response to various stresses including viral dsRNA, play key roles in the controlled activation of RLR signaling. Without the SG nucleators G3BP1/2 and UBAP2L, dsRNA triggers excessive inflammation and immune-mediated apoptosis. In addition to exogenous dsRNA, host-derived dsRNA generated in response to ADAR1 deficiency is also controlled by SG biology. Intriguingly, SGs can function beyond immune control by suppressing viral replication independently of the RLR pathway. These observations thus highlight the multi-functional nature of SGs as cellular "shock absorbers" that converge on protecting cell homeostasis by dampening both toxic immune response and viral replication.


Asunto(s)
ADN Helicasas , ARN Helicasas , Humanos , ADN Helicasas/metabolismo , ARN Helicasas/genética , ARN Helicasas/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Gránulos de Estrés , Proteínas con Motivos de Reconocimiento de ARN/metabolismo , Inmunidad Innata , Inflamación/metabolismo , Gránulos Citoplasmáticos/metabolismo , Proteínas Portadoras/metabolismo
8.
Immunity ; 54(9): 1976-1988.e7, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34525338

RESUMEN

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.


Asunto(s)
Adenosina Desaminasa/genética , Enfermedades Autoinmunes del Sistema Nervioso/genética , Malformaciones del Sistema Nervioso/genética , Edición de ARN/genética , ARN Bicatenario/genética , Adenosina Desaminasa/metabolismo , Animales , Enfermedades Autoinmunes del Sistema Nervioso/fisiopatología , Modelos Animales de Enfermedad , Helicasa Inducida por Interferón IFIH1/metabolismo , Ratones , Mutación , Malformaciones del Sistema Nervioso/fisiopatología , ARN Bicatenario/metabolismo
9.
Immunity ; 54(9): 1948-1960.e5, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34343497

RESUMEN

The RNA deaminase ADAR1 is an essential negative regulator of the RNA sensor MDA5, and loss of ADAR1 function triggers inappropriate activation of MDA5 by self-RNAs. Mutations in ADAR, the gene that encodes ADAR1, cause human immune diseases, including Aicardi-Goutières syndrome (AGS). However, the mechanisms of MDA5-dependent disease pathogenesis in vivo remain unknown. Here we generated mice with a single amino acid change in ADAR1 that models the most common human ADAR AGS mutation. These Adar mutant mice developed lethal disease that required MDA5, the RIG-I-like receptor LGP2, type I interferons, and the eIF2α kinase PKR. A small-molecule inhibitor of the integrated stress response (ISR) that acts downstream of eIF2α phosphorylation prevented immunopathology and rescued the mice from mortality. These findings place PKR and the ISR as central components of immunopathology in vivo and identify therapeutic targets for treatment of human diseases associated with the ADAR1-MDA5 axis.


Asunto(s)
Adenosina Desaminasa/metabolismo , Enfermedades Autoinmunes del Sistema Nervioso/patología , Malformaciones del Sistema Nervioso/patología , Estrés Fisiológico/fisiología , eIF-2 Quinasa/metabolismo , Células A549 , Animales , Enfermedades Autoinmunes del Sistema Nervioso/genética , Enfermedades Autoinmunes del Sistema Nervioso/metabolismo , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Ratones , Ratones Mutantes , Mutación , Malformaciones del Sistema Nervioso/genética , Malformaciones del Sistema Nervioso/metabolismo
10.
Immunity ; 54(9): 1961-1975.e5, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34525337

RESUMEN

Nucleic acids are powerful triggers of innate immunity and can adopt the Z-conformation, an unusual left-handed double helix. Here, we studied the biological function(s) of Z-RNA recognition by the adenosine deaminase ADAR1, mutations in which cause Aicardi-Goutières syndrome. Adar1mZα/mZα mice, bearing two point mutations in the Z-nucleic acid binding (Zα) domain that abolish Z-RNA binding, displayed spontaneous induction of type I interferons (IFNs) in multiple organs, including in the lung, where both stromal and hematopoietic cells showed IFN-stimulated gene (ISG) induction. Lung neutrophils expressed ISGs induced by the transcription factor IRF3, indicating an initiating role for neutrophils in this IFN response. The IFN response in Adar1mZα/mZα mice required the adaptor MAVS, implicating cytosolic RNA sensing. Adenosine-to-inosine changes were enriched in transposable elements and revealed a specific requirement of ADAR1's Zα domain in editing of a subset of RNAs. Thus, endogenous RNAs in Z-conformation have immunostimulatory potential curtailed by ADAR1, with relevance to autoinflammatory disease in humans.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Adenosina Desaminasa/genética , Interferón Tipo I/inmunología , ARN Bicatenario/genética , Adenosina/genética , Adenosina/metabolismo , Animales , Enfermedades Autoinmunes del Sistema Nervioso/genética , Enfermedades Autoinmunes del Sistema Nervioso/inmunología , Inosina/genética , Inosina/metabolismo , Interferón Tipo I/genética , Ratones , Mutación , Malformaciones del Sistema Nervioso/genética , Malformaciones del Sistema Nervioso/inmunología , Edición de ARN/genética , ARN Bicatenario/metabolismo
11.
Mol Cell ; 81(11): 2374-2387.e3, 2021 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-33905683

RESUMEN

Adenosine-to-inosine editing is catalyzed by ADAR1 at thousands of sites transcriptome-wide. Despite intense interest in ADAR1 from physiological, bioengineering, and therapeutic perspectives, the rules of ADAR1 substrate selection are poorly understood. Here, we used large-scale systematic probing of ∼2,000 synthetic constructs to explore the structure and sequence context determining editability. We uncover two structural layers determining the formation and propagation of A-to-I editing, independent of sequence. First, editing is robustly induced at fixed intervals of 35 bp upstream and 30 bp downstream of structural disruptions. Second, editing is symmetrically introduced on opposite sites on a double-stranded structure. Our findings suggest a recursive model for RNA editing, whereby the structural alteration induced by the editing at one site iteratively gives rise to the formation of an additional editing site at a fixed periodicity, serving as a basis for the propagation of editing along and across both strands of double-stranded RNA structures.


Asunto(s)
Adenosina Desaminasa/genética , Adenosina/metabolismo , Inosina/metabolismo , Edición de ARN , ARN Bicatenario/genética , Proteínas de Unión al ARN/genética , Células A549 , Adenosina/genética , Adenosina Desaminasa/metabolismo , Animales , Emparejamiento Base , Células HEK293 , Humanos , Inosina/genética , Células MCF-7 , Ratones , Células 3T3 NIH , Conformación de Ácido Nucleico , ARN Bicatenario/química , ARN Bicatenario/metabolismo , Proteínas de Unión al ARN/metabolismo
12.
Trends Genet ; 40(3): 250-259, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38160061

RESUMEN

Recent studies have underscored the pivotal role of adenosine-to-inosine RNA editing, catalyzed by ADAR1, in suppressing innate immune interferon responses triggered by cellular double-stranded RNA (dsRNA). However, the specific ADAR1 editing targets crucial for this regulatory function remain elusive. We review analyses of transcriptome-wide ADAR1 editing patterns and their evolutionary dynamics, which offer valuable insights into this unresolved query. The growing appreciation of the significance of immunogenic dsRNAs and their editing in inflammatory and autoimmune diseases and cancer calls for a more comprehensive understanding of dsRNA immunogenicity, which may promote our understanding of these diseases and open doors to therapeutic avenues.


Asunto(s)
Enfermedades Autoinmunes , ARN Bicatenario , Humanos , ARN Bicatenario/genética , Inmunidad Innata/genética , Transcriptoma/genética
13.
Development ; 150(2)2023 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-36458554

RESUMEN

Adenosine deaminase acting on RNA 1 (ADAR1) is an RNA-binding protein that deaminates adenosine (A) to inosine (I). A-to-I editing alters post-transcriptional RNA processing, making ADAR1 a crucial regulator of gene expression. Consequently, Adar1 has been implicated in organogenesis. To determine the role of Adar1 in pancreatic development and homeostasis, we conditionally deleted Adar1 from the murine pancreas (Ptf1aCre/+; Adar1Fl/Fl). The resulting mice had stunted growth, likely due to malabsorption associated with exocrine pancreatic insufficiency. Analyses of pancreata revealed ductal cell expansion, heightened interferon-stimulated gene expression and an increased influx of immune cells. Concurrent deletion of Adar1 and Mavs, a signaling protein implicated in the innate immune pathway, rescued the degenerative phenotype and resulted in normal pancreatic development. Taken together, our work suggests that the primary function of Adar1 in the pancreas is to prevent aberrant activation of the Mavs-mediated innate immune pathway, thereby maintaining pancreatic homeostasis.


Asunto(s)
Páncreas Exocrino , Animales , Ratones , Páncreas Exocrino/metabolismo , Interferones/genética , Interferones/metabolismo , Fenotipo , Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo
14.
RNA ; 30(9): 1164-1183, 2024 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-38844344

RESUMEN

In recent years, numerous evidence has been accumulated about the extent of A-to-I editing in human RNAs and the key role ADAR1 plays in the cellular editing machinery. It has been shown that A-to-I editing occurrence and frequency are tissue-specific and essential for some tissue development, such as the liver. To study the effect of ADAR1 function in hepatocytes, we have created Huh7.5 ADAR1 KO cell lines. Upon IFN treatment, the Huh7.5 ADAR1 KO cells show rapid arrest of growth and translation, from which they do not recover. We analyzed translatome changes by using a method based on sequencing of separate polysome profile RNA fractions. We found significant changes in the transcriptome and translatome of the Huh7.5 ADAR1 KO cells. The most prominent changes include negatively affected transcription by RNA polymerase III and the deregulation of snoRNA and Y RNA levels. Furthermore, we observed that ADAR1 KO polysomes are enriched in mRNAs coding for proteins pivotal in a wide range of biological processes such as RNA localization and RNA processing, whereas the unbound fraction is enriched mainly in mRNAs coding for ribosomal proteins and translational factors. This indicates that ADAR1 plays a more relevant role in small RNA metabolism and ribosome biogenesis.


Asunto(s)
Adenosina Desaminasa , Hepatocitos , Edición de ARN , Proteínas de Unión al ARN , Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo , Humanos , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Hepatocitos/metabolismo , Polirribosomas/metabolismo , Polirribosomas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Biosíntesis de Proteínas , Transcriptoma , Técnicas de Inactivación de Genes , Línea Celular
15.
RNA ; 30(5): 521-529, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38531651

RESUMEN

In this article, I recount my memories of key experiments that led to my entry into the RNA editing/modification field. I highlight initial observations made by the pioneers in the ADAR field, and how they fit into our current understanding of this family of enzymes. I discuss early mysteries that have now been solved, as well as those that still linger. Finally, I discuss important, outstanding questions and acknowledge my hope for the future of the RNA editing/modification field.


Asunto(s)
Adenosina Desaminasa , ARN , ARN/genética , Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo , Edición de ARN , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Inosina/metabolismo , ARN Bicatenario
16.
RNA ; 30(5): 500-511, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38531645

RESUMEN

Innate immunity must be tightly regulated to enable sensitive pathogen detection while averting autoimmunity triggered by pathogen-like host molecules. A hallmark of viral infection, double-stranded RNAs (dsRNAs) are also abundantly encoded in mammalian genomes, necessitating surveillance mechanisms to distinguish "self" from "nonself." ADAR1, an RNA editing enzyme, has emerged as an essential safeguard against dsRNA-induced autoimmunity. By converting adenosines to inosines (A-to-I) in long dsRNAs, ADAR1 covalently marks endogenous dsRNAs, thereby blocking the activation of the cytoplasmic dsRNA sensor MDA5. Moreover, beyond its editing function, ADAR1 binding to dsRNA impedes the activation of innate immune sensors PKR and ZBP1. Recent landmark studies underscore the utility of silencing ADAR1 for cancer immunotherapy, by exploiting the ADAR1-dependence developed by certain tumors to unleash an antitumor immune response. In this perspective, we summarize the genetic and mechanistic evidence for ADAR1's multipronged role in suppressing dsRNA-mediated autoimmunity and explore the evolving roles of ADAR1 as an immuno-oncology target.


Asunto(s)
Adenosina Desaminasa , Edición de ARN , Animales , Adenosina Desaminasa/metabolismo , Inmunidad Innata/genética , Helicasa Inducida por Interferón IFIH1/genética , Mamíferos/genética , ARN Bicatenario/genética , Humanos
17.
Trends Immunol ; 44(3): 201-216, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36710220

RESUMEN

ADAR1 and ZBP1 are the only two mammalian proteins that contain Zα domains, which are thought to bind to nucleic acids in the Z-conformation. These two molecules are crucial in regulating diverse biological processes. While ADAR1-mediated RNA editing supports host survival and development, ZBP1-mediated immune responses provide host defense against infection and disease. Recent studies have expanded our understanding of the functions of ADAR1 and ZBP1 beyond their classical roles and established their fundamental regulation of innate immune responses, including NLRP3 inflammasome activation, inflammation, and cell death. Their roles in these processes have physiological impacts across development, infectious and inflammatory diseases, and cancer. In this review, we discuss the functions of ADAR1 and ZBP1 in regulating innate immune responses in development and disease.


Asunto(s)
Inmunidad Innata , Ácidos Nucleicos , Animales , Humanos , Muerte Celular , Inflamación/metabolismo , Mamíferos
18.
Mol Cell ; 69(1): 126-135.e6, 2018 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-29304330

RESUMEN

N6-methyladenosine (m6A) and adenosine-to-inosine (A-to-I) editing are two of the most abundant RNA modifications, both at adenosines. Yet, the interaction of these two types of adenosine modifications is largely unknown. Here we show a global A-to-I difference between m6A-positive and m6A-negative RNA populations. Both the presence and extent of A-to-I sites in m6A-negative RNA transcripts suggest a negative correlation between m6A and A-to-I. Suppression of m6A-catalyzing enzymes results in global A-to-I RNA editing changes. Further depletion of m6A modification increases the association of m6A-depleted transcripts with adenosine deaminase acting on RNA (ADAR) enzymes, resulting in upregulated A-to-I editing on the same m6A-depleted transcripts. Collectively, the effect of m6A on A-to-I suggests a previously underappreciated interplay between two distinct and abundant RNA modifications, highlighting a complex epitranscriptomic landscape.


Asunto(s)
Adenosina/análogos & derivados , Adenosina/química , Inosina/química , Edición de ARN/genética , ARN/genética , Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo , Línea Celular Tumoral , Regulación de la Expresión Génica/genética , Células HEK293 , Células HeLa , Humanos , Metiltransferasas/genética , Metiltransferasas/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
19.
J Biol Chem ; : 107789, 2024 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-39303916

RESUMEN

Esophageal cancer is mainly divided into esophageal adenocarcinoma (EADC) and esophageal squamous cell carcinoma (ESCC). China is one of the high-incidence areas of esophageal cancer, of which about 90% are ESCC. The deubiquitinase USP38 has been reported to play significant roles in several biological processes, including inflammatory responses, antiviral infection, cell proliferation, migration, invasion, DNA damage repair, and chemotherapy resistance. However, the role and mechanisms of USP38 in ESCC development remain still unclear. Furthermore, although many substrates of USP38 have been identified, few upstream regulatory factors of USP38 have been identified. In this study, we found that USP38 was significantly upregulated in esophageal cancer tissues. Knockdown of USP38 inhibited ESCC growth. USP38 stabilized itself through auto-deubiquitylation. In addition, we demonstrate that ADAR could enhance the stability of USP38 protein and facilitate USP38 auto-deubiquitylation by interacting with USP38 in an RNA editing-independent manner. ADAR inhibition of ESCC cell proliferation depended on USP38. In summary, these results highlight that the potential of targeting the ADAR-USP38 axis for ESCC treatment.

20.
J Biol Chem ; 300(8): 107504, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38944123

RESUMEN

Z-nucleic acid structures play vital roles in cellular processes and have implications in innate immunity due to their recognition by Zα domains containing proteins (Z-DNA/Z-RNA binding proteins, ZBPs). Although Zα domains have been identified in six proteins, including viral E3L, ORF112, and I73R, as well as, cellular ADAR1, ZBP1, and PKZ, their prevalence across living organisms remains largely unexplored. In this study, we introduce a computational approach to predict Zα domains, leading to the revelation of previously unidentified Zα domain-containing proteins in eukaryotic organisms, including non-metazoan species. Our findings encompass the discovery of new ZBPs in previously unexplored giant viruses, members of the Nucleocytoviricota phylum. Through experimental validation, we confirm the Zα functionality of select proteins, establishing their capability to induce the B-to-Z conversion. Additionally, we identify Zα-like domains within bacterial proteins. While these domains share certain features with Zα domains, they lack the ability to bind to Z-nucleic acids or facilitate the B-to-Z DNA conversion. Our findings significantly expand the ZBP family across a wide spectrum of organisms and raise intriguing questions about the evolutionary origins of Zα-containing proteins. Moreover, our study offers fresh perspectives on the functional significance of Zα domains in virus sensing and innate immunity and opens avenues for exploring hitherto undiscovered functions of ZBPs.


Asunto(s)
ADN de Forma Z , Proteínas de Unión al ARN , ADN de Forma Z/química , ADN de Forma Z/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/inmunología , Virus Gigantes/metabolismo , Virus Gigantes/genética , Virus Gigantes/química , Dominios Proteicos , Proteínas Virales/química , Proteínas Virales/metabolismo , Proteínas Virales/inmunología , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Inmunidad Innata , Humanos , Unión Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA