Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Wiley Interdiscip Rev RNA ; 13(4): e1703, 2022 07.
Artículo en Inglés | MEDLINE | ID: covidwho-2277282

RESUMEN

The COVID-19 crisis and the development of the first approved mRNA vaccine have highlighted the power of RNA-based therapeutic strategies for the development of new medicines. Aside from RNA-vaccines, antisense oligonucleotides (ASOs) represent a new and very promising class of RNA-targeted therapy. Few drugs have already received approval from the Food and Drug Administration. Here, we underscored why and how ASOs hold the potential to change the therapeutic landscape to beat SARS-CoV-2 viral infections. This article is categorized under: RNA Interactions with Proteins and Other Molecules > Small Molecule-RNA Interactions.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Oligonucleótidos Antisentido , Humanos , Oligonucleótidos , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/farmacología , ARN , SARS-CoV-2 , Estados Unidos , Vacunas Sintéticas , Vacunas de ARNm
2.
PLoS One ; 18(2): e0281281, 2023.
Artículo en Inglés | MEDLINE | ID: covidwho-2234628

RESUMEN

Although the COVID-19 pandemic began over three years ago, the virus responsible for the disease, SARS-CoV-2, continues to infect people across the globe. As such, there remains a critical need for development of novel therapeutics against SARS-CoV-2. One technology that has remained relatively unexplored in COVID-19 is the use of antisense oligonucleotides (ASOs)-short single-stranded nucleic acids that bind to target RNA transcripts to modulate their expression. In this study, ASOs targeted against the SARS-CoV-2 genome and host entry factors, ACE2 and TMPRSS2, were designed and tested for their ability to inhibit cellular infection by SARS-CoV-2. Using our previously developed SARS-CoV-2 bioassay platform, we screened 180 total ASOs targeting various regions of the SARS-CoV-2 genome and validated several ASOs that potently blocked SARS-CoV-2 infection in vitro. Notably, select ASOs retained activity against both the WA1 and B.1.1.7 (commonly known as alpha) variants. Screening of ACE2 and TMPRSS2 ASOs showed that targeting of ACE2 also potently prevented infection by the WA1 and B.1.1.7 SARS-CoV-2 viruses in the tested cell lines. Combined with the demonstrated success of ASOs in other disease indications, these results support further research into the development of ASOs targeting SARS-CoV-2 and host entry factors as potential COVID-19 therapeutics.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , SARS-CoV-2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , Oligonucleótidos Antisentido/farmacología , Oligonucleótidos Antisentido/uso terapéutico , Pandemias , Peptidil-Dipeptidasa A/metabolismo , Internalización del Virus
3.
Nat Commun ; 13(1): 4503, 2022 08 03.
Artículo en Inglés | MEDLINE | ID: covidwho-1972603

RESUMEN

The COVID-19 pandemic is exacting an increasing toll worldwide, with new SARS-CoV-2 variants emerging that exhibit higher infectivity rates and that may partially evade vaccine and antibody immunity. Rapid deployment of non-invasive therapeutic avenues capable of preventing infection by all SARS-CoV-2 variants could complement current vaccination efforts and help turn the tide on the COVID-19 pandemic. Here, we describe a novel therapeutic strategy targeting the SARS-CoV-2 RNA using locked nucleic acid antisense oligonucleotides (LNA ASOs). We identify an LNA ASO binding to the 5' leader sequence of SARS-CoV-2 that disrupts a highly conserved stem-loop structure with nanomolar efficacy in preventing viral replication in human cells. Daily intranasal administration of this LNA ASO in the COVID-19 mouse model potently suppresses viral replication (>80-fold) in the lungs of infected mice. We find that the LNA ASO is efficacious in countering all SARS-CoV-2 "variants of concern" tested both in vitro and in vivo. Hence, inhaled LNA ASOs targeting SARS-CoV-2 represents a promising therapeutic approach to reduce or prevent transmission and decrease severity of COVID-19 in infected individuals. LNA ASOs are chemically stable and can be flexibly modified to target different viral RNA sequences and could be stockpiled for future coronavirus pandemics.


Asunto(s)
COVID-19 , SARS-CoV-2 , Administración Intranasal , Animales , Humanos , Ratones , Oligonucleótidos Antisentido/farmacología , Oligonucleótidos Antisentido/uso terapéutico , Pandemias/prevención & control , ARN Viral/genética
4.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Artículo en Inglés | MEDLINE | ID: covidwho-1684241

RESUMEN

SARS-CoV-2 is a highly pathogenic virus that evades antiviral immunity by interfering with host protein synthesis, mRNA stability, and protein trafficking. The SARS-CoV-2 nonstructural protein 1 (Nsp1) uses its C-terminal domain to block the messenger RNA (mRNA) entry channel of the 40S ribosome to inhibit host protein synthesis. However, how SARS-CoV-2 circumvents Nsp1-mediated suppression for viral protein synthesis and if the mechanism can be targeted therapeutically remain unclear. Here, we show that N- and C-terminal domains of Nsp1 coordinate to drive a tuned ratio of viral to host translation, likely to maintain a certain level of host fitness while maximizing replication. We reveal that the stem-loop 1 (SL1) region of the SARS-CoV-2 5' untranslated region (5' UTR) is necessary and sufficient to evade Nsp1-mediated translational suppression. Targeting SL1 with locked nucleic acid antisense oligonucleotides inhibits viral translation and makes SARS-CoV-2 5' UTR vulnerable to Nsp1 suppression, hindering viral replication in vitro at a nanomolar concentration, as well as providing protection against SARS-CoV-2-induced lethality in transgenic mice expressing human ACE2. Thus, SL1 allows Nsp1 to switch infected cells from host to SARS-CoV-2 translation, presenting a therapeutic target against COVID-19 that is conserved among immune-evasive variants. This unique strategy of unleashing a virus' own virulence mechanism against itself could force a critical trade-off between drug resistance and pathogenicity.


Asunto(s)
Regiones no Traducidas 5'/genética , Evasión Inmune/genética , Biosíntesis de Proteínas , SARS-CoV-2/genética , Proteínas no Estructurales Virales/genética , Animales , Secuencia de Bases , Chlorocebus aethiops , Células HEK293 , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/genética , Humanos , Evasión Inmune/efectos de los fármacos , Ratones Transgénicos , Modelos Biológicos , Oligonucleótidos Antisentido/farmacología , Biosíntesis de Proteínas/efectos de los fármacos , SARS-CoV-2/efectos de los fármacos , Células Vero , Replicación Viral/efectos de los fármacos
5.
Molecules ; 27(2)2022 Jan 15.
Artículo en Inglés | MEDLINE | ID: covidwho-1625662

RESUMEN

Antisense oligonucleotides (ASOs) are an increasingly represented class of drugs. These small sequences of nucleotides are designed to precisely target other oligonucleotides, usually RNA species, and are modified to protect them from degradation by nucleases. Their specificity is due to their sequence, so it is possible to target any RNA sequence that is already known. These molecules are very versatile and adaptable given that their sequence and chemistry can be custom manufactured. Based on the chemistry being used, their activity may significantly change and their effects on cell function and phenotypes can differ dramatically. While some will cause the target RNA to decay, others will only bind to the target and act as a steric blocker. Their incredible versatility is the key to manipulating several aspects of nucleic acid function as well as their process, and alter the transcriptome profile of a specific cell type or tissue. For example, they can be used to modify splicing or mask specific sites on a target. The entire design rather than just the sequence is essential to ensuring the specificity of the ASO to its target. Thus, it is vitally important to ensure that the complete process of drug design and testing is taken into account. ASOs' adaptability is a considerable advantage, and over the past decades has allowed multiple new drugs to be approved. This, in turn, has had a significant and positive impact on patient lives. Given current challenges presented by the COVID-19 pandemic, it is necessary to find new therapeutic strategies that would complement the vaccination efforts being used across the globe. ASOs may be a very powerful tool that can be used to target the virus RNA and provide a therapeutic paradigm. The proof of the efficacy of ASOs as an anti-viral agent is long-standing, yet no molecule currently has FDA approval. The emergence and widespread use of RNA vaccines during this health crisis might provide an ideal opportunity to develop the first anti-viral ASOs on the market. In this review, we describe the story of ASOs, the different characteristics of their chemistry, and how their characteristics translate into research and as a clinical tool.


Asunto(s)
Desarrollo de Medicamentos/métodos , Oligonucleótidos Antisentido/química , Oligonucleótidos Antisentido/farmacología , Animales , COVID-19/terapia , Aprobación de Drogas , Diseño de Fármacos , Humanos , Oligonucleótidos Antisentido/uso terapéutico , SARS-CoV-2/efectos de los fármacos , Estados Unidos , United States Food and Drug Administration
6.
Nat Commun ; 12(1): 4396, 2021 07 20.
Artículo en Inglés | MEDLINE | ID: covidwho-1387353

RESUMEN

Rapid development of antisense therapies can enable on-demand responses to new viral pathogens and make personalized medicine for genetic diseases practical. Antisense phosphorodiamidate morpholino oligomers (PMOs) are promising candidates to fill such a role, but their challenging synthesis limits their widespread application. To rapidly prototype potential PMO drug candidates, we report a fully automated flow-based oligonucleotide synthesizer. Our optimized synthesis platform reduces coupling times by up to 22-fold compared to previously reported methods. We demonstrate the power of our automated technology with the synthesis of milligram quantities of three candidate therapeutic PMO sequences for an unserved class of Duchenne muscular dystrophy (DMD). To further test our platform, we synthesize a PMO that targets the genomic mRNA of SARS-CoV-2 and demonstrate its antiviral effects. This platform could find broad application not only in designing new SARS-CoV-2 and DMD antisense therapeutics, but also for rapid development of PMO candidates to treat new and emerging diseases.


Asunto(s)
Técnicas de Química Sintética/instrumentación , Química Farmacéutica/instrumentación , Ensayos Analíticos de Alto Rendimiento/instrumentación , Morfolinos/síntesis química , Oligonucleótidos Antisentido/síntesis química , Animales , COVID-19/virología , Chlorocebus aethiops , Enfermedades Transmisibles Emergentes/tratamiento farmacológico , Enfermedades Transmisibles Emergentes/microbiología , Modelos Animales de Enfermedad , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Morfolinos/farmacología , Morfolinos/uso terapéutico , Distrofia Muscular de Duchenne/tratamiento farmacológico , Distrofia Muscular de Duchenne/genética , Oligonucleótidos Antisentido/farmacología , Oligonucleótidos Antisentido/uso terapéutico , Medicina de Precisión/métodos , ARN Mensajero/antagonistas & inhibidores , ARN Viral/antagonistas & inhibidores , SARS-CoV-2/genética , Factores de Tiempo , Células Vero , Tratamiento Farmacológico de COVID-19
7.
Nat Struct Mol Biol ; 28(9): 747-754, 2021 09.
Artículo en Inglés | MEDLINE | ID: covidwho-1370728

RESUMEN

Drug discovery campaigns against COVID-19 are beginning to target the SARS-CoV-2 RNA genome. The highly conserved frameshift stimulation element (FSE), required for balanced expression of viral proteins, is a particularly attractive SARS-CoV-2 RNA target. Here we present a 6.9 Å resolution cryo-EM structure of the FSE (88 nucleotides, ~28 kDa), validated through an RNA nanostructure tagging method. The tertiary structure presents a topologically complex fold in which the 5' end is threaded through a ring formed inside a three-stem pseudoknot. Guided by this structure, we develop antisense oligonucleotides that impair FSE function in frameshifting assays and knock down SARS-CoV-2 virus replication in A549-ACE2 cells at 100 nM concentration.


Asunto(s)
COVID-19/prevención & control , Microscopía por Crioelectrón/métodos , Mutación del Sistema de Lectura/genética , Oligonucleótidos Antisentido/genética , ARN Viral/genética , Elementos de Respuesta/genética , SARS-CoV-2/genética , Células A549 , Animales , Secuencia de Bases , COVID-19/virología , Línea Celular Tumoral , Chlorocebus aethiops , Genoma Viral/genética , Humanos , Modelos Moleculares , Conformación de Ácido Nucleico , Oligonucleótidos Antisentido/farmacología , ARN Viral/química , ARN Viral/ultraestructura , SARS-CoV-2/fisiología , SARS-CoV-2/ultraestructura , Células Vero , Replicación Viral/efectos de los fármacos , Replicación Viral/genética
8.
Angew Chem Int Ed Engl ; 60(40): 21662-21667, 2021 09 27.
Artículo en Inglés | MEDLINE | ID: covidwho-1363645

RESUMEN

There is an urgent need to develop antiviral drugs and alleviate the current COVID-19 pandemic. Herein we report the design and construction of chimeric oligonucleotides comprising a 2'-OMe-modified antisense oligonucleotide and a 5'-phosphorylated 2'-5' poly(A)4 (4A2-5 ) to degrade envelope and spike RNAs of SARS-CoV-2. The oligonucleotide was used for searching and recognizing target viral RNA sequence, and the conjugated 4A2-5 was used for guided RNase L activation to sequence-specifically degrade viral RNAs. Since RNase L can potently cleave single-stranded RNA during innate antiviral response, degradation efficiencies with these chimeras were twice as much as those with only antisense oligonucleotides for both SARS-CoV-2 RNA targets. In pseudovirus infection models, chimera-S4 achieved potent and broad-spectrum inhibition of SARS-CoV-2 and its N501Y and/or ΔH69/ΔV70 mutants, indicating a promising antiviral agent based on the nucleic acid-hydrolysis targeting chimera (NATAC) strategy.


Asunto(s)
Antivirales/farmacología , Endorribonucleasas/metabolismo , Activación Enzimática/efectos de los fármacos , Oligonucleótidos Antisentido/farmacología , SARS-CoV-2/efectos de los fármacos , Animales , Chlorocebus aethiops , Proteínas de la Envoltura de Coronavirus/genética , Diseño de Fármacos , Células HEK293 , Humanos , Hidrólisis/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Mutación , ARN Viral/metabolismo , Glicoproteína de la Espiga del Coronavirus/genética , Células Vero
9.
Pharmacol Rev ; 72(4): 862-898, 2020 10.
Artículo en Inglés | MEDLINE | ID: covidwho-767668

RESUMEN

RNA-based therapies, including RNA molecules as drugs and RNA-targeted small molecules, offer unique opportunities to expand the range of therapeutic targets. Various forms of RNAs may be used to selectively act on proteins, transcripts, and genes that cannot be targeted by conventional small molecules or proteins. Although development of RNA drugs faces unparalleled challenges, many strategies have been developed to improve RNA metabolic stability and intracellular delivery. A number of RNA drugs have been approved for medical use, including aptamers (e.g., pegaptanib) that mechanistically act on protein target and small interfering RNAs (e.g., patisiran and givosiran) and antisense oligonucleotides (e.g., inotersen and golodirsen) that directly interfere with RNA targets. Furthermore, guide RNAs are essential components of novel gene editing modalities, and mRNA therapeutics are under development for protein replacement therapy or vaccination, including those against unprecedented severe acute respiratory syndrome coronavirus pandemic. Moreover, functional RNAs or RNA motifs are highly structured to form binding pockets or clefts that are accessible by small molecules. Many natural, semisynthetic, or synthetic antibiotics (e.g., aminoglycosides, tetracyclines, macrolides, oxazolidinones, and phenicols) can directly bind to ribosomal RNAs to achieve the inhibition of bacterial infections. Therefore, there is growing interest in developing RNA-targeted small-molecule drugs amenable to oral administration, and some (e.g., risdiplam and branaplam) have entered clinical trials. Here, we review the pharmacology of novel RNA drugs and RNA-targeted small-molecule medications, with a focus on recent progresses and strategies. Challenges in the development of novel druggable RNA entities and identification of viable RNA targets and selective small-molecule binders are discussed. SIGNIFICANCE STATEMENT: With the understanding of RNA functions and critical roles in diseases, as well as the development of RNA-related technologies, there is growing interest in developing novel RNA-based therapeutics. This comprehensive review presents pharmacology of both RNA drugs and RNA-targeted small-molecule medications, focusing on novel mechanisms of action, the most recent progress, and existing challenges.


Asunto(s)
ARN/efectos de los fármacos , ARN/farmacología , Aptámeros de Nucleótidos/farmacología , Aptámeros de Nucleótidos/uso terapéutico , Betacoronavirus , COVID-19 , Técnicas de Química Analítica/métodos , Técnicas de Química Analítica/normas , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Infecciones por Coronavirus/tratamiento farmacológico , Sistemas de Liberación de Medicamentos/métodos , Desarrollo de Medicamentos/organización & administración , Descubrimiento de Drogas , Humanos , MicroARNs/farmacología , MicroARNs/uso terapéutico , Oligonucleótidos Antisentido/farmacología , Oligonucleótidos Antisentido/uso terapéutico , Pandemias , Neumonía Viral/tratamiento farmacológico , ARN/efectos adversos , ARN sin Sentido/farmacología , ARN sin Sentido/uso terapéutico , ARN Guía de Kinetoplastida/farmacología , ARN Guía de Kinetoplastida/uso terapéutico , ARN Mensajero/efectos de los fármacos , ARN Mensajero/farmacología , ARN Ribosómico/efectos de los fármacos , ARN Ribosómico/farmacología , ARN Interferente Pequeño/farmacología , ARN Interferente Pequeño/uso terapéutico , ARN Viral/efectos de los fármacos , Ribonucleasas/metabolismo , Riboswitch/efectos de los fármacos , SARS-CoV-2
10.
Clin Sci (Lond) ; 134(10): 1143-1150, 2020 05 29.
Artículo en Inglés | MEDLINE | ID: covidwho-342905

RESUMEN

Angiotensin-converting enzyme 2 (ACE2) plays an essential role in maintaining the balance of the renin-angiotensin system and also serves as a receptor for the SARS-CoV-2, SARS-CoV, and HCoV-NL63. Following the recent outbreak of SARS-CoV-2 infection, there has been an urgent need to develop therapeutic interventions. ACE2 is a potential target for many treatment approaches for the SARS-CoV-2. With the help of bioinformatics, we have predicted several novel exons of the human ACE2 gene. The inclusion of novel exons located in the 5'UTR/intronic region in the mature transcript may remove the critical ACE2 residues responsible for the interaction with the receptor-binding domain (RBD) of SARS-CoV-2, thus preventing their binding and entry into the cell. Additionally, inclusion of a novel predicted exons located in the 3'UTR by alternative splicing may remove the C-terminal transmembrane domain of ACE2 and generate soluble ACE2 isoforms. Splice-switching antisense oligonucleotides (SSOs) have been employed effectively as a therapeutic strategy in several disease conditions. Alternative splicing of the ACE2 gene could similarly be modulated using SSOs to exclude critical domains required for the entry of SARS-CoV-2. Strategies can also be designed to deliver these SSOs directly to the lungs in order to minimize the damage caused by SARS-CoV-2 pathogenesis.


Asunto(s)
Empalme Alternativo , Infecciones por Coronavirus/genética , Oligonucleótidos Antisentido/farmacología , Peptidil-Dipeptidasa A/genética , Neumonía Viral/genética , Internalización del Virus , Enzima Convertidora de Angiotensina 2 , Betacoronavirus/fisiología , COVID-19 , Biología Computacional , Infecciones por Coronavirus/terapia , Exones , Humanos , Modelos Moleculares , Pandemias , Neumonía Viral/terapia , Dominios Proteicos , Receptores Virales/genética , SARS-CoV-2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA