Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
EMBO J ; 39(11): e101573, 2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32323871

RESUMEN

High expression of 2',5'-oligoadenylate synthetase 1 (OAS1), which adds AMP residues in 2',5' linkage to a variety of substrates, is observed in many cancers as a part of the interferon-related DNA damage resistance signature (IRDS). Poly(ADP-ribose) (PAR) is rapidly synthesized from NAD+ at sites of DNA damage to facilitate repair, but excessive PAR synthesis due to extensive DNA damage results in cell death by energy depletion and/or activation of PAR-dependent programmed cell death pathways. We find that OAS1 adds AMP residues in 2',5' linkage to PAR, inhibiting its synthesis in vitro and reducing its accumulation in cells. Increased OAS1 expression substantially improves cell viability following DNA-damaging treatments that stimulate PAR synthesis during DNA repair. We conclude that high expression of OAS1 in cancer cells promotes their ability to survive DNA damage by attenuating PAR synthesis and thus preventing cell death.


Asunto(s)
2',5'-Oligoadenilato Sintetasa/biosíntesis , Daño del ADN , Regulación Enzimológica de la Expresión Génica , Poli ADP Ribosilación , 2',5'-Oligoadenilato Sintetasa/genética , Adenosina Monofosfato/genética , Adenosina Monofosfato/metabolismo , Muerte Celular , Línea Celular Transformada , Humanos
2.
Proc Natl Acad Sci U S A ; 117(40): 24802-24812, 2020 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-32958664

RESUMEN

The oligoadenylate synthetase (OAS)-RNase L system is an IFN-inducible antiviral pathway activated by viral infection. Viral double-stranded (ds) RNA activates OAS isoforms that synthesize the second messenger 2-5A, which binds and activates the pseudokinase-endoribonuclease RNase L. In cells, OAS activation is tamped down by ADAR1, an adenosine deaminase that destabilizes dsRNA. Mutation of ADAR1 is one cause of Aicardi-Goutières syndrome (AGS), an interferonopathy in children. ADAR1 deficiency in human cells can lead to RNase L activation and subsequent cell death. To evaluate RNase L as a possible therapeutic target for AGS, we sought to identify small-molecule inhibitors of RNase L. A 500-compound library of protein kinase inhibitors was screened for modulators of RNase L activity in vitro. We identified ellagic acid (EA) as a hit with 10-fold higher selectivity against RNase L compared with its nearest paralog, IRE1. SAR analysis identified valoneic acid dilactone (VAL) as a superior inhibitor of RNase L, with 100-fold selectivity over IRE1. Mechanism-of-action analysis indicated that EA and VAL do not bind to the pseudokinase domain of RNase L despite acting as ATP competitive inhibitors of the protein kinase CK2. VAL is nontoxic and functional in cells, although with a 1,000-fold decrease in potency, as measured by RNA cleavage activity in response to treatment with dsRNA activator or by rescue of cell lethality resulting from self dsRNA induced by ADAR1 deficiency. These studies lay the foundation for understanding novel modes of regulating RNase L function using small-molecule inhibitors and avenues of therapeutic potential.


Asunto(s)
Adenosina Desaminasa/deficiencia , Enfermedades Autoinmunes del Sistema Nervioso/enzimología , Endorribonucleasas/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Malformaciones del Sistema Nervioso/enzimología , Fenol/farmacología , 2',5'-Oligoadenilato Sintetasa/genética , 2',5'-Oligoadenilato Sintetasa/metabolismo , Nucleótidos de Adenina/metabolismo , Adenosina Desaminasa/genética , Enfermedades Autoinmunes del Sistema Nervioso/genética , Enfermedades Autoinmunes del Sistema Nervioso/fisiopatología , Muerte Celular/efectos de los fármacos , Endorribonucleasas/genética , Endorribonucleasas/metabolismo , Inhibidores Enzimáticos/química , Humanos , Malformaciones del Sistema Nervioso/genética , Malformaciones del Sistema Nervioso/fisiopatología , Oligorribonucleótidos/metabolismo , Fenol/química , Proteínas de Unión al ARN/genética
3.
Mol Cell ; 53(2): 221-34, 2014 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-24462203

RESUMEN

RNase L is an ankyrin repeat domain-containing dual endoribonuclease-pseudokinase that is activated by unusual 2,'5'-oligoadenylate (2-5A) second messengers and which impedes viral infections in higher vertebrates. Despite its importance in interferon-regulated antiviral innate immunity, relatively little is known about its precise mechanism of action. Here we present a functional characterization of 2.5 Å and 3.25 Å X-ray crystal and small-angle X-ray scattering structures of RNase L bound to a natural 2-5A activator with and without ADP or the nonhydrolysable ATP mimetic AMP-PNP. These studies reveal how recognition of 2-5A through interactions with the ankyrin repeat domain and the pseudokinase domain, together with nucleotide binding, imposes a rigid intertwined dimer configuration that is essential for RNase catalytic and antiviral functions. The involvement of the pseudokinase domain of RNase L in 2-5A sensing, nucleotide binding, dimerization, and ribonuclease functions highlights the evolutionary adaptability of the eukaryotic protein kinase fold.


Asunto(s)
Nucleótidos de Adenina/química , Endorribonucleasas/química , Oligorribonucleótidos/química , Adenosina Difosfato/química , Adenilil Imidodifosfato/química , Animales , Repetición de Anquirina , Sitios de Unión , Cristalografía por Rayos X , Dimerización , Virus de la Encefalomiocarditis , Endorribonucleasas/genética , Endorribonucleasas/fisiología , Células HeLa , Humanos , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Picornaviridae , Estructura Terciaria de Proteína , Dispersión de Radiación , Relación Estructura-Actividad , Sus scrofa
4.
Proc Natl Acad Sci U S A ; 116(11): 5071-5076, 2019 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-30814222

RESUMEN

Drugs that reverse epigenetic silencing, such as the DNA methyltransferase inhibitor (DNMTi) 5-azacytidine (AZA), have profound effects on transcription and tumor cell survival. AZA is an approved drug for myelodysplastic syndromes and acute myeloid leukemia, and is under investigation for different solid malignant tumors. AZA treatment generates self, double-stranded RNA (dsRNA), transcribed from hypomethylated repetitive elements. Self dsRNA accumulation in DNMTi-treated cells leads to type I IFN production and IFN-stimulated gene expression. Here we report that cell death in response to AZA treatment occurs through the 2',5'-oligoadenylate synthetase (OAS)-RNase L pathway. OASs are IFN-induced enzymes that synthesize the RNase L activator 2-5A in response to dsRNA. Cells deficient in RNase L or OAS1 to 3 are highly resistant to AZA, as are wild-type cells treated with a small-molecule inhibitor of RNase L. A small-molecule inhibitor of c-Jun NH2-terminal kinases (JNKs) also antagonizes RNase L-dependent cell death in response to AZA, consistent with a role for JNK in RNase L-induced apoptosis. In contrast, the rates of AZA-induced and RNase L-dependent cell death were increased by transfection of 2-5A, by deficiencies in ADAR1 (which edits and destabilizes dsRNA), PDE12 or AKAP7 (which degrade 2-5A), or by ionizing radiation (which induces IFN-dependent signaling). Finally, OAS1 expression correlates with AZA sensitivity in the NCI-60 set of tumor cell lines, suggesting that the level of OAS1 can be a biomarker for predicting AZA sensitivity of tumor cells. These studies may eventually lead to pharmacologic strategies for regulating the antitumor activity and toxicity of AZA and related drugs.


Asunto(s)
2',5'-Oligoadenilato Sintetasa/metabolismo , Azacitidina/farmacología , Desmetilación del ADN , Endorribonucleasas/metabolismo , Inmunidad Innata , Células A549 , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Muerte Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Isoenzimas/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Hidrolasas Diéster Fosfóricas/metabolismo , Radiación Ionizante , Bibliotecas de Moléculas Pequeñas/farmacología
6.
PLoS Pathog ; 13(2): e1006195, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28158275

RESUMEN

Coronaviruses are of veterinary and medical importance and include highly pathogenic zoonotic viruses, such as SARS-CoV and MERS-CoV. They are known to efficiently evade early innate immune responses, manifesting in almost negligible expression of type-I interferons (IFN-I). This evasion strategy suggests an evolutionary conserved viral function that has evolved to prevent RNA-based sensing of infection in vertebrate hosts. Here we show that the coronavirus endonuclease (EndoU) activity is key to prevent early induction of double-stranded RNA (dsRNA) host cell responses. Replication of EndoU-deficient coronaviruses is greatly attenuated in vivo and severely restricted in primary cells even during the early phase of the infection. In macrophages we found immediate induction of IFN-I expression and RNase L-mediated breakdown of ribosomal RNA. Accordingly, EndoU-deficient viruses can retain replication only in cells that are deficient in IFN-I expression or sensing, and in cells lacking both RNase L and PKR. Collectively our results demonstrate that the coronavirus EndoU efficiently prevents simultaneous activation of host cell dsRNA sensors, such as Mda5, OAS and PKR. The localization of the EndoU activity at the site of viral RNA synthesis-within the replicase complex-suggests that coronaviruses have evolved a viral RNA decay pathway to evade early innate and intrinsic antiviral host cell responses.


Asunto(s)
Coronaviridae/enzimología , Infecciones por Coronavirus/inmunología , Endonucleasas/inmunología , Evasión Inmune/fisiología , Proteínas Virales/inmunología , Animales , Coronaviridae/inmunología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata/inmunología , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena en Tiempo Real de la Polimerasa
7.
Proc Natl Acad Sci U S A ; 113(8): 2241-6, 2016 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-26858407

RESUMEN

The 2',5'-oligoadenylate (2-5A) synthetase (OAS)-RNase L system is an IFN-induced antiviral pathway. RNase L activity depends on 2-5A, synthesized by OAS. Although all three enzymatically active OAS proteins in humans--OAS1, OAS2, and OAS3--synthesize 2-5A upon binding dsRNA, it is unclear which are responsible for RNase L activation during viral infection. We used clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated protein-9 nuclease (Cas9) technology to engineer human A549-derived cell lines in which each of the OAS genes or RNase L is knocked out. Upon transfection with poly(rI):poly(rC), a synthetic surrogate for viral dsRNA, or infection with each of four viruses from different groups (West Nile virus, Sindbis virus, influenza virus, or vaccinia virus), OAS1-KO and OAS2-KO cells synthesized amounts of 2-5A similar to those synthesized in parental wild-type cells, causing RNase L activation as assessed by rRNA degradation. In contrast, OAS3-KO cells synthesized minimal 2-5A, and rRNA remained intact, similar to infected RNase L-KO cells. All four viruses replicated to higher titers in OAS3-KO or RNase L-KO A549 cells than in parental, OAS1-KO, or OAS2-KO cells, demonstrating the antiviral effects of OAS3. OAS3 displayed a higher affinity for dsRNA in intact cells than either OAS1 or OAS2, consistent with its dominant role in RNase L activation. Finally, the requirement for OAS3 as the major OAS isoform responsible for RNase L activation was not restricted to A549 cells, because OAS3-KO cells derived from two other human cell lines also were deficient in RNase L activation.


Asunto(s)
2',5'-Oligoadenilato Sintetasa/metabolismo , Endorribonucleasas/metabolismo , Virosis/metabolismo , 2',5'-Oligoadenilato Sintetasa/antagonistas & inhibidores , 2',5'-Oligoadenilato Sintetasa/genética , Infecciones por Alphavirus/genética , Infecciones por Alphavirus/metabolismo , Sistemas CRISPR-Cas , Línea Celular , Endorribonucleasas/antagonistas & inhibidores , Endorribonucleasas/genética , Activación Enzimática , Técnicas de Inactivación de Genes , Humanos , Gripe Humana/genética , Gripe Humana/metabolismo , Modelos Biológicos , ARN Ribosómico/genética , ARN Ribosómico/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Virus Sindbis , Vaccinia/genética , Vaccinia/metabolismo , Virosis/genética , Fiebre del Nilo Occidental/genética , Fiebre del Nilo Occidental/metabolismo
8.
Cell Rep ; 43(4): 113998, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38551960

RESUMEN

RNase L is an endoribonuclease of higher vertebrates that functions in antiviral innate immunity. Interferons induce oligoadenylate synthetase enzymes that sense double-stranded RNA of viral origin leading to the synthesis of 2',5'-oligoadenylate (2-5A) activators of RNase L. However, it is unknown precisely how RNase L remodels the host cell transcriptome. To isolate effects of RNase L from other effects of double-stranded RNA or virus, 2-5A is directly introduced into cells. Here, we report that RNase L activation by 2-5A causes a ribotoxic stress response involving the MAP kinase kinase kinase (MAP3K) ZAKα, MAP2Ks, and the stress-activated protein kinases JNK and p38α. RNase L activation profoundly alters the transcriptome by widespread depletion of mRNAs associated with different cellular functions but also by JNK/p38α-stimulated induction of inflammatory genes. These results show that the 2-5A/RNase L system triggers a protein kinase cascade leading to proinflammatory signaling and apoptosis.


Asunto(s)
Endorribonucleasas , Inmunidad Innata , Endorribonucleasas/metabolismo , Endorribonucleasas/genética , Humanos , Nucleótidos de Adenina/metabolismo , Oligorribonucleótidos/metabolismo , Animales , Estrés Fisiológico , Transcriptoma/genética , ARN Bicatenario/metabolismo
9.
J Virol ; 86(20): 11311-21, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22875977

RESUMEN

Autophagy is a programmed homeostatic response to diverse types of cellular stress that disposes of long-lived proteins, organelles, and invading microbes within double-membraned structures called autophagosomes. The 2',5'-oligoadenylate/RNase L system is a virus-activated host RNase pathway that disposes of or processes viral and cellular single-stranded RNAs. Here we report that activation of RNase L during viral infections induces autophagy. Accordingly, infections with encephalomyocarditis virus or vesicular stomatitis virus led to higher levels of autophagy in wild-type mouse embryonic fibroblasts (MEF) than in RNase L-null MEF. Similarly, direct activation of RNase L with a 2',5'-oligoadenylate resulted in p62(SQSTM1) degradation, LC3BI/LC3BII conversion, and appearance of autophagosomes. To determine the effect of RNase L-mediated autophagy on viral replication, we compared viral yields in wild-type and RNase L-null MEF in the absence or presence of either chemical inhibitors of autophagy (bafilomycin A1 or 3-methyladenine) or small interfering RNA (siRNA) against ATG5 or beclin-1. At a low multiplicity of infection, induction of autophagy by RNase L during the initial cycle of virus growth contributed to the suppression of virus replication. However, in subsequent rounds of infection, autophagy promoted viral replication, reducing the antiviral effect of RNase L. Our results indicate a novel function of RNase L as an inducer of autophagy that affects viral yields.


Asunto(s)
Autofagia , Virus de la Encefalomiocarditis/fisiología , Endorribonucleasas/metabolismo , Virus de la Estomatitis Vesicular Indiana/fisiología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adenina/análogos & derivados , Adenina/farmacología , Animales , Proteínas Reguladoras de la Apoptosis/biosíntesis , Proteínas Reguladoras de la Apoptosis/genética , Autofagia/efectos de los fármacos , Proteína 5 Relacionada con la Autofagia , Beclina-1 , Células Cultivadas , Chlorocebus aethiops , Células HeLa , Proteínas de Choque Térmico/metabolismo , Humanos , Macrólidos/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/biosíntesis , Proteínas Asociadas a Microtúbulos/genética , Interferencia de ARN , ARN Interferente Pequeño , Proteína Sequestosoma-1 , Replicación Viral
10.
bioRxiv ; 2023 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-37873202

RESUMEN

RNase L is a regulated endoribonuclease in higher vertebrates that functions in antiviral innate immunity. Interferons induce OAS enzymes that sense double-stranded RNA of viral origin leading to synthesis of 2',5'-oligoadenylate (2-5A) activators of RNase L. However, it is unknown precisely how RNase L inhibits viral infections. To isolate effects of RNase L from other effects of double-stranded RNA or virus, 2-5A was directly introduced into cells. Here we report that RNase L activation by 2-5A causes a ribotoxic stress response that requires the ribosome-associated MAP3K, ZAKα. Subsequently, the stress-activated protein kinases (SAPK) JNK and p38α are phosphorylated. RNase L activation profoundly altered the transcriptome by widespread depletion of mRNAs associated with different cellular functions, but also by SAPK-dependent induction of inflammatory genes. Our findings show that 2-5A is a ribotoxic stressor that causes RNA damage through RNase L triggering a ZAKα kinase cascade leading to proinflammatory signaling and apoptosis.

11.
Viruses ; 15(7)2023 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-37515225

RESUMEN

Viral replication often depends on RNA maturation and degradation processes catalyzed by viral ribonucleases, which are therefore candidate targets for antiviral drugs. Here, we synthesized and studied the antiviral properties of a novel nitrocatechol compound (1c) and other analogs that are structurally related to the catechol derivative dynasore. Interestingly, compound 1c strongly inhibited two DEDD box viral ribonucleases, HIV-1 RNase H and SARS-CoV-2 nsp14 3'-to-5' exoribonuclease (ExoN). While 1c inhibited SARS-CoV-2 ExoN activity, it did not interfere with the mRNA methyltransferase activity of nsp14. In silico molecular docking placed compound 1c in the catalytic pocket of the ExoN domain of nsp14. Finally, 1c inhibited SARS-CoV-2 replication but had no toxicity to human lung adenocarcinoma cells. Given its simple chemical synthesis from easily available starting materials, these results suggest that 1c might be a lead compound for the design of new antiviral compounds that target coronavirus nsp14 ExoN and other viral ribonucleases.


Asunto(s)
COVID-19 , VIH-1 , Humanos , SARS-CoV-2/genética , Exorribonucleasas/genética , VIH-1/genética , Simulación del Acoplamiento Molecular , Antivirales/farmacología , Replicación Viral , Catecoles/farmacología , Ribonucleasa H/farmacología , Proteínas no Estructurales Virales/genética , ARN Viral/genética
12.
Science ; 379(6632): eabo3627, 2023 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-36538032

RESUMEN

Multisystem inflammatory syndrome in children (MIS-C) is a rare and severe condition that follows benign COVID-19. We report autosomal recessive deficiencies of OAS1, OAS2, or RNASEL in five unrelated children with MIS-C. The cytosolic double-stranded RNA (dsRNA)-sensing OAS1 and OAS2 generate 2'-5'-linked oligoadenylates (2-5A) that activate the single-stranded RNA-degrading ribonuclease L (RNase L). Monocytic cell lines and primary myeloid cells with OAS1, OAS2, or RNase L deficiencies produce excessive amounts of inflammatory cytokines upon dsRNA or severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) stimulation. Exogenous 2-5A suppresses cytokine production in OAS1-deficient but not RNase L-deficient cells. Cytokine production in RNase L-deficient cells is impaired by MDA5 or RIG-I deficiency and abolished by mitochondrial antiviral-signaling protein (MAVS) deficiency. Recessive OAS-RNase L deficiencies in these patients unleash the production of SARS-CoV-2-triggered, MAVS-mediated inflammatory cytokines by mononuclear phagocytes, thereby underlying MIS-C.


Asunto(s)
COVID-19 , Citocinas , Endorribonucleasas , SARS-CoV-2 , Síndrome de Respuesta Inflamatoria Sistémica , Niño , Humanos , COVID-19/inmunología , Citocinas/genética , Citocinas/inmunología , Endorribonucleasas/genética , Endorribonucleasas/metabolismo , ARN Bicatenario , SARS-CoV-2/genética , Síndrome de Respuesta Inflamatoria Sistémica/genética
13.
J Virol ; 85(9): 4547-57, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21325416

RESUMEN

Xenotropic murine leukemia-related virus (XMRV) was identified in association with human prostate cancer and chronic fatigue syndrome. To examine the infection potential, kinetics, and tissue distribution of XMRV in an animal model, we inoculated five macaques with XMRV intravenously. XMRV established a persistent, chronic disseminated infection, with low transient viremia and provirus in blood lymphocytes during acute infection. Although undetectable in blood after about a month, XMRV viremia was reactivated at 9 months, confirming the chronicity of the infection. Furthermore, XMRV Gag was detected in tissues throughout, with wide dissemination throughout the period of monitoring. Surprisingly, XMRV infection showed organ-specific cell tropism, infecting CD4 T cells in lymphoid organs including the gastrointestinal lamina propria, alveolar macrophages in lung, and epithelial/interstitial cells in other organs, including the reproductive tract. Of note, in spite of the intravenous inoculation, extensive XMRV replication was noted in prostate during acute but not chronic infection even though infected cells were still detectable by fluorescence in situ hybridization (FISH) in prostate at 5 and 9 months postinfection. Marked lymphocyte activation occurred immediately postinfection, but antigen-specific cellular responses were undetectable. Antibody responses were elicited and boosted upon reexposure, but titers decreased rapidly, suggesting low antigen stimulation over time. Our findings establish a nonhuman primate model to study XMRV replication/dissemination, transmission, pathogenesis, immune responses, and potential future therapies.


Asunto(s)
Anticuerpos Antivirales/sangre , Modelos Animales de Enfermedad , Macaca mulatta/virología , Enfermedades de los Primates/virología , Infecciones por Retroviridae/virología , Virus Relacionado con el Virus Xenotrópico de la Leucemia Murina/inmunología , Virus Relacionado con el Virus Xenotrópico de la Leucemia Murina/patogenicidad , Animales , Linfocitos T CD4-Positivos/virología , Enfermedad Crónica , Células Epiteliales/virología , Humanos , Linfocitos/virología , Macrófagos/virología , Masculino , Enfermedades de los Primates/inmunología , Enfermedades de los Primates/patología , Provirus/aislamiento & purificación , Infecciones por Retroviridae/inmunología , Infecciones por Retroviridae/patología , Tropismo Viral , Viremia , Activación Viral , Latencia del Virus
14.
J Med Chem ; 65(2): 1445-1457, 2022 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-34841869

RESUMEN

The pseudokinase-endoribonuclease RNase L plays important roles in antiviral innate immunity and is also implicated in many other cellular activities. The inhibition of RNase L showed therapeutic potential for Aicardi-Goutières syndrome (AGS). Thus, RNase L is a promising drug target. In this study, using an enzyme assay and NMR screening, we discovered 13 inhibitory fragments against RNase L. Cocrystal structures of RNase L separately complexed with two different fragments were determined in which both fragments bound to the ATP-binding pocket of the pseudokinase domain. Myricetin, vitexin, and hyperoside, three natural products sharing similar scaffolds with the fragment AC40357, demonstrated a potent inhibitory activity in vitro. In addition, myricetin has a promising cellular inhibitory activity. A cocrystal structure of RNase L with myricetin provided a structural basis for inhibitor design by allosterically modulating the ribonuclease activity. Our findings demonstrate that fragment screening can lead to the discovery of natural product inhibitors of RNase L.


Asunto(s)
Productos Biológicos/farmacología , Descubrimiento de Drogas , Endorribonucleasas/antagonistas & inhibidores , Ensayos Analíticos de Alto Rendimiento/métodos , Bibliotecas de Moléculas Pequeñas/farmacología , Humanos
15.
mBio ; 12(4): e0178121, 2021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34372695

RESUMEN

The 2',5'-oligoadenylate (2-5A)-dependent endoribonuclease, RNase L, is a principal mediator of the interferon (IFN) antiviral response. Therefore, the regulation of cellular levels of 2-5A is a key point of control in antiviral innate immunity. Cellular 2-5A levels are determined by IFN-inducible 2',5'-oligoadenylate synthetases (OASs) and by enzymes that degrade 2-5A. Importantly, many coronaviruses (CoVs) and rotaviruses encode 2-5A-degrading enzymes, thereby antagonizing RNase L and its antiviral effects. A-kinase-anchoring protein 7 (AKAP7), a mammalian counterpart, could possibly limit tissue damage from excessive or prolonged RNase L activation during viral infections or from self-double-stranded RNAs that activate OAS. We show that these enzymes, members of the two-histidine phosphoesterase (2H-PE) superfamily, constitute a subfamily referred here as 2',5'-PEs. 2',5'-PEs from the mouse CoV mouse hepatitis virus (MHV) (NS2), Middle East respiratory syndrome coronavirus (MERS-CoV) (NS4b), group A rotavirus (VP3), and mouse (AKAP7) were investigated for their evolutionary relationships and activities. While there was no activity against 3',5'-oligoribonucleotides, they all cleaved 2',5'-oligoadenylates efficiently but with variable activity against other 2',5'-oligonucleotides. The 2',5'-PEs are shown to be metal ion-independent enzymes that cleave trimer 2-5A (2',5'-p3A3) producing mono- or diadenylates with 2',3'-cyclic phosphate termini. Our results suggest that the elimination of 2-5A might be the sole function of viral 2',5'-PEs, thereby promoting viral escape from innate immunity by preventing or limiting the activation of RNase L. IMPORTANCE Viruses often encode accessory proteins that antagonize the host antiviral immune response. Here, we probed the evolutionary relationships and biochemical activities of two-histidine phosphoesterases (2H-PEs) that allow some coronaviruses and rotaviruses to counteract antiviral innate immunity. In addition, we investigated the mammalian enzyme AKAP7, which has homology and shared activities with the viral enzymes and might reduce self-injury. These viral and host enzymes, which we refer to as 2',5'-PEs, specifically degrade 2',5'-oligoadenylate activators of the antiviral enzyme RNase L. We show that the host and viral enzymes are metal ion independent and exclusively cleave 2',5'- and not 3',5'-phosphodiester bonds, producing cleavage products with cyclic 2',3'-phosphate termini. Our study defines 2',5'-PEs as enzymes that share characteristic conserved features with the 2H-PE superfamily but have specific and distinct biochemical cleavage activities. These findings may eventually lead to pharmacological strategies for developing antiviral drugs against coronaviruses, rotaviruses, and other viruses.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Nucleótidos de Adenina/metabolismo , Endorribonucleasas/metabolismo , Coronavirus del Síndrome Respiratorio de Oriente Medio/enzimología , Virus de la Hepatitis Murina/enzimología , Oligorribonucleótidos/metabolismo , Rotavirus/enzimología , Animales , Humanos , Inmunidad Innata/inmunología , Interferones/inmunología , Ratones
16.
J Virol ; 83(14): 6995-7003, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19403677

RESUMEN

The xenotropic murine leukemia virus-related virus (XMRV) has recently been detected in prostate cancer tissues and may play a role in tumorigenesis. It is currently unclear how this virus is transmitted and which factors promote its spread in the prostate. We show that amyloidogenic fragments known as semen-derived enhancer of virus infection (SEVI) originating from prostatic acid phosphatase greatly increase XMRV infections of primary prostatic epithelial and stromal cells. Hybrid simian/human immunodeficiency chimeric virus particles pseudotyped with XMRV envelope protein were used to demonstrate that the enhancing effect of SEVI, or of human semen itself, was at the level of viral attachment and entry. SEVI enhanced XMRV infectivity but did not bypass the requirement for the xenotropic and polytropic retrovirus receptor 1. Furthermore, XMRV RNA was detected in prostatic secretions of some men with prostate cancer. The fact that the precursor of SEVI is produced in abundance by the prostate indicates that XMRV replication occurs in an environment that provides a natural enhancer of viral infection, and this may play a role in the spread of this virus in the human population.


Asunto(s)
Gammaretrovirus/fisiología , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/virología , Proteínas Tirosina Fosfatasas/metabolismo , Infecciones por Retroviridae/enzimología , Infecciones por Retroviridae/virología , Fosfatasa Ácida , Línea Celular Tumoral , Células Cultivadas , Fibroblastos/enzimología , Fibroblastos/virología , Gammaretrovirus/genética , Humanos , Masculino , Proteínas Tirosina Fosfatasas/genética
17.
J Virol ; 82(20): 9964-77, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18684813

RESUMEN

Xenotropic murine leukemia virus-related virus (XMRV) is a new human gammaretrovirus identified in prostate cancer tissue from patients homozygous for a reduced-activity variant of the antiviral enzyme RNase L. Neither a casual relationship between XMRV infection and prostate cancer nor a mechanism of tumorigenesis has been established. To determine the integration site preferences of XMRV and the potential risk of proviral insertional mutagenesis, we carried out a genome-wide analysis of viral integration sites in the prostate cell line DU145 after an acute XMRV infection and compared the integration site pattern of XMRV with those found for murine leukemia virus and two human retroviruses, human immunodeficiency virus type 1 and human T-cell leukemia virus type 1. Among all retroviruses analyzed, XMRV has the strongest preference for transcription start sites, CpG islands, DNase-hypersensitive sites, and gene-dense regions; all are features frequently associated with structurally open transcription regulatory regions of a chromosome. Analyses of XMRV integration sites in tissues from prostate cancer patients found a similar preference for the aforementioned chromosomal features. Additionally, XMRV integration sites in cancer tissues were associated with cancer breakpoints, common fragile sites, microRNA, and cancer-related genes, suggesting a selection process that favors certain chromosomal integration sites. In both acutely infected cells and cancer tissues, no common integration site was detected within or near proto-oncogenes or tumor suppressor genes. These results are consistent with a model in which XMRV may contribute to tumorigenicity via a paracrine mechanism.


Asunto(s)
Gammaretrovirus/metabolismo , Virus de la Leucemia Murina/metabolismo , Neoplasias de la Próstata/virología , Integración Viral , Animales , Línea Celular , Mapeo Cromosómico , Islas de CpG , Endorribonucleasas/genética , Endorribonucleasas/metabolismo , Gammaretrovirus/genética , Humanos , Virus de la Leucemia Murina/genética , Masculino , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Datos de Secuencia Molecular , Neoplasias de la Próstata/genética , Secuencias Reguladoras de Ácidos Nucleicos , Infecciones por Retroviridae/metabolismo , Transcripción Genética
18.
FASEB J ; 21(1): 156-66, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17135368

RESUMEN

Maintenance of normal blood pressure is critical for preserving the integrity of the cardiovascular system. Angiotensin 1-converting enzyme (ACE) regulates normal blood pressure and fluid homeostasis through its action in the renin-angiotensin-aldosterone system (RAAS) and the renal tubuloglomerular feedback response. Although the two structurally related isozymic forms of ACE both generate the vasoactive octapeptide angiotensin II (Ang II) with equal efficiency, both are expressed in a nonoverlapping tissue-restricted fashion. To discriminate the precise physiological role of each ACE in its requisite tissue in vivo, we expressed one ACE isoform exclusively in a single cell type of an Ace null mouse. Previously, we demonstrated that vascular endothelial cell-specific expression of transgenic somatic ACE (sACE) could restore normal blood pressure of Ace-null mice. In this current study, we expressed germinal ACE (gACE) in the vascular endothelial cells of the Ace null mouse. These mice exhibited correct renal structure, renal function, and normal growth rates. Although the mice had elevated levels of gACE bound to vascular endothelial cells and high levels of gACE and Ang II in the circulating serum, blood pressure was restored only partially. This study demonstrated that gACE, even when expressed in the vasculature, could not functionally substitute for sACE.


Asunto(s)
Presión Sanguínea/genética , Peptidil-Dipeptidasa A/fisiología , Angiotensina II/sangre , Animales , Femenino , Perfilación de la Expresión Génica , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Peptidil-Dipeptidasa A/genética
19.
Elife ; 62017 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-28362255

RESUMEN

ADAR1 isoforms are adenosine deaminases that edit and destabilize double-stranded RNA reducing its immunostimulatory activities. Mutation of ADAR1 leads to a severe neurodevelopmental and inflammatory disease of children, Aicardi-Goutiéres syndrome. In mice, Adar1 mutations are embryonic lethal but are rescued by mutation of the Mda5 or Mavs genes, which function in IFN induction. However, the specific IFN regulated proteins responsible for the pathogenic effects of ADAR1 mutation are unknown. We show that the cell-lethal phenotype of ADAR1 deletion in human lung adenocarcinoma A549 cells is rescued by CRISPR/Cas9 mutagenesis of the RNASEL gene or by expression of the RNase L antagonist, murine coronavirus NS2 accessory protein. Our result demonstrate that ablation of RNase L activity promotes survival of ADAR1 deficient cells even in the presence of MDA5 and MAVS, suggesting that the RNase L system is the primary sensor pathway for endogenous dsRNA that leads to cell death.


Asunto(s)
Adenosina Desaminasa/deficiencia , Muerte Celular , Endorribonucleasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Línea Celular Tumoral , Células Epiteliales/fisiología , Humanos , Helicasa Inducida por Interferón IFIH1/metabolismo , Proteínas de Unión al ARN
20.
Respir Care ; 51(2): 133-9, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16441957

RESUMEN

PURPOSE: To determine if the measurement of minute ventilation recovery time (V (E)RT), a recently proposed predictor of extubation outcome, can be reproduced using a more practical, simpler method. METHODS: A case series with convenience sampling was performed in the surgical intensive care unit of a tertiary-care hospital. Nineteen patients were enrolled during weaning from mechanical ventilation, prior to the initial extubation attempt. Within-subject comparisons of V (E)RT were performed, using 2 alternative methods for measuring baseline V (E) and one alternative method for determining the threshold for recovery of V (E) during the final spontaneous breathing trial prior to extubation. Comparison methods for baseline V (E) included an 8-hour average and the last V (E) measurement prior to the spontaneous breathing trial. The alternative threshold for defining recovery of V (E) was 100% of the baseline value (vs 110% in the original method). RESULTS: The study subjects were primarily cardiac surgery patients (63%) and were ventilated for a median of 5 days prior to extubation. V (E)RT calculated using the 8-hour average or the last V (E) measurement prior to the spontaneous breathing trial as baseline, and a threshold of 100% of baseline V (E) to define recovery most closely approximated V (E)RT obtained by the original method and similarly classified patients at high risk for reintubation (kappa statistic = 0.78 +/- 0.2). CONCLUSIONS: V (E)RT can be determined using a simpler method for measuring both baseline V (E) and the recovery threshold. These methodological modifications may increase the feasibility of measuring V (E)RT, while reproducing the results obtained by the original method.


Asunto(s)
Intubación Intratraqueal , Ventilación Pulmonar , Desconexión del Ventilador/métodos , Adulto , Anciano , Remoción de Dispositivos/métodos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Recuperación de la Función , Mecánica Respiratoria , Volumen de Ventilación Pulmonar , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA