Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Int J Mol Sci ; 23(4)2022 Feb 13.
Artículo en Inglés | MEDLINE | ID: covidwho-1686819

RESUMEN

The COVID-19 pandemic has evidenced the urgent need for the discovery of broad-spectrum antiviral therapies that could be deployed in the case of future emergence of novel viral threats, as well as to back up current therapeutic options in the case of drug resistance development. Most current antivirals are directed to inhibit specific viruses since these therapeutic molecules are designed to act on a specific viral target with the objective of interfering with a precise step in the replication cycle. Therefore, antimicrobial peptides (AMPs) have been identified as promising antiviral agents that could help to overcome this limitation and provide compounds able to act on more than a single viral family. We evaluated the antiviral activity of an amphibian peptide known for its strong antimicrobial activity against both Gram-positive and Gram-negative bacteria, namely Temporin L (TL). Previous studies have revealed that TL is endowed with widespread antimicrobial activity and possesses marked haemolytic activity. Therefore, we analyzed TL and a previously identified TL derivative (Pro3, DLeu9 TL, where glutamine at position 3 is replaced with proline, and the D-Leucine enantiomer is present at position 9) as well as its analogs, for their activity against a wide panel of viruses comprising enveloped, naked, DNA and RNA viruses. We report significant inhibition activity against herpesviruses, paramyxoviruses, influenza virus and coronaviruses, including SARS-CoV-2. Moreover, we further modified our best candidate by lipidation and demonstrated a highly reduced cytotoxicity with improved antiviral effect. Our results show a potent and selective antiviral activity of TL peptides, indicating that the novel lipidated temporin-based antiviral agents could prove to be useful additions to current drugs in combatting rising drug resistance and epidemic/pandemic emergencies.


Asunto(s)
Proteínas Anfibias/farmacología , Anfibios/metabolismo , Péptidos Catiónicos Antimicrobianos/farmacología , Antivirales/química , Virus ADN/efectos de los fármacos , Virus ARN/efectos de los fármacos , Secuencia de Aminoácidos , Proteínas Anfibias/química , Proteínas Anfibias/metabolismo , Animales , Péptidos Catiónicos Antimicrobianos/química , Péptidos Catiónicos Antimicrobianos/metabolismo , Antivirales/farmacología , Supervivencia Celular/efectos de los fármacos , Chlorocebus aethiops , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Grampositivas/efectos de los fármacos , Humanos , Lípidos/química , SARS-CoV-2/efectos de los fármacos , Células Vero
3.
Int J Mol Sci ; 23(2)2022 Jan 14.
Artículo en Inglés | MEDLINE | ID: covidwho-1625084

RESUMEN

Viral infections represent a serious threat to the world population and are becoming more frequent. The search and identification of broad-spectrum antiviral molecules is necessary to ensure new therapeutic options, since there is a limited availability of effective antiviral drugs able to eradicate viral infections, and consequently due to the increase of strains that are resistant to the most used drugs. Recently, several studies on antimicrobial peptides identified them as promising antiviral agents. In detail, amphibian skin secretions serve as a rich source of natural antimicrobial peptides. Their antibacterial and antifungal activities have been widely reported, but their exploitation as potential antiviral agents have yet to be fully investigated. In the present study, the antiviral activity of the peptide derived from the secretion of Rana tagoi, named AR-23, was evaluated against both DNA and RNA viruses, with or without envelope. Different assays were performed to identify in which step of the infectious cycle the peptide could act. AR-23 exhibited a greater inhibitory activity in the early stages of infection against both DNA (HSV-1) and RNA (MeV, HPIV-2, HCoV-229E, and SARS-CoV-2) enveloped viruses and, on the contrary, it was inactive against naked viruses (PV-1). Altogether, the results indicated AR-23 as a peptide with potential therapeutic effects against a wide variety of human viruses.


Asunto(s)
Proteínas Anfibias/farmacología , Péptidos Antimicrobianos/farmacología , Antivirales/farmacología , Ranidae/metabolismo , Animales , Péptidos Catiónicos Antimicrobianos/farmacología , Supervivencia Celular/efectos de los fármacos , Chlorocebus aethiops , Virus ADN/efectos de los fármacos , Virus ARN/efectos de los fármacos , SARS-CoV-2/efectos de los fármacos , Células Vero , Envoltura Viral/efectos de los fármacos , Ensayo de Placa Viral , Virosis/tratamiento farmacológico
4.
J Biol Chem ; 298(2): 101529, 2022 02.
Artículo en Inglés | MEDLINE | ID: covidwho-1587355

RESUMEN

Remdesivir (RDV) is a direct-acting antiviral agent that is approved in several countries for the treatment of coronavirus disease 2019 caused by the severe acute respiratory syndrome coronavirus 2. RDV exhibits broad-spectrum antiviral activity against positive-sense RNA viruses, for example, severe acute respiratory syndrome coronavirus and hepatitis C virus, and nonsegmented negative-sense RNA viruses, for example, Nipah virus, whereas segmented negative-sense RNA viruses such as influenza virus or Crimean-Congo hemorrhagic fever virus are not sensitive to the drug. The reasons for this apparent efficacy pattern are unknown. Here, we expressed and purified representative RNA-dependent RNA polymerases and studied three biochemical parameters that have been associated with the inhibitory effects of RDV-triphosphate (TP): (i) selective incorporation of the nucleotide substrate RDV-TP, (ii) the effect of the incorporated RDV-monophosphate (MP) on primer extension, and (iii) the effect of RDV-MP in the template during incorporation of the complementary UTP. We found a strong correlation between antiviral effects and efficient incorporation of RDV-TP. Inhibition in primer extension reactions was heterogeneous and usually inefficient at higher NTP concentrations. In contrast, template-dependent inhibition of UTP incorporation opposite the embedded RDV-MP was seen with all polymerases. Molecular modeling suggests a steric conflict between the 1'-cyano group of the inhibitor and residues of the structurally conserved RNA-dependent RNA polymerase motif F. We conclude that future efforts in the development of nucleotide analogs with a broader spectrum of antiviral activities should focus on improving rates of incorporation while capitalizing on the inhibitory effects of a bulky 1'-modification.


Asunto(s)
Adenosina Monofosfato/análogos & derivados , Alanina/análogos & derivados , Modelos Moleculares , Virus ARN/enzimología , ARN Polimerasa Dependiente del ARN/antagonistas & inhibidores , Adenosina Monofosfato/química , Adenosina Monofosfato/farmacología , Alanina/química , Alanina/farmacología , Antivirales/farmacología , Hepacivirus/efectos de los fármacos , Hepacivirus/enzimología , Virus ARN de Sentido Negativo/efectos de los fármacos , Virus ARN de Sentido Negativo/enzimología , Virus Nipah/efectos de los fármacos , Virus Nipah/enzimología , Virus ARN Monocatenarios Positivos/efectos de los fármacos , Virus ARN Monocatenarios Positivos/enzimología , Virus ARN/efectos de los fármacos , ARN Viral/metabolismo , ARN Polimerasa Dependiente del ARN/química , ARN Polimerasa Dependiente del ARN/metabolismo , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/enzimología , Replicación Viral/efectos de los fármacos
5.
Viruses ; 13(12)2021 12 14.
Artículo en Inglés | MEDLINE | ID: covidwho-1572668

RESUMEN

Broad-spectrum antiviral therapies hold promise as a first-line defense against emerging viruses by blunting illness severity and spread until vaccines and virus-specific antivirals are developed. The nucleobase favipiravir, often discussed as a broad-spectrum inhibitor, was not effective in recent clinical trials involving patients infected with Ebola virus or SARS-CoV-2. A drawback of favipiravir use is its rapid clearance before conversion to its active nucleoside-5'-triphosphate form. In this work, we report a synergistic reduction of flavivirus (dengue, Zika), orthomyxovirus (influenza A), and coronavirus (HCoV-OC43 and SARS-CoV-2) replication when the nucleobases favipiravir or T-1105 were combined with the antimetabolite 6-methylmercaptopurine riboside (6MMPr). The 6MMPr/T-1105 combination increased the C-U and G-A mutation frequency compared to treatment with T-1105 or 6MMPr alone. A further analysis revealed that the 6MMPr/T-1105 co-treatment reduced cellular purine nucleotide triphosphate synthesis and increased conversion of the antiviral nucleobase to its nucleoside-5'-monophosphate, -diphosphate, and -triphosphate forms. The 6MMPr co-treatment specifically increased production of the active antiviral form of the nucleobases (but not corresponding nucleosides) while also reducing levels of competing cellular NTPs to produce the synergistic effect. This in-depth work establishes a foundation for development of small molecules as possible co-treatments with nucleobases like favipiravir in response to emerging RNA virus infections.


Asunto(s)
Antimetabolitos/farmacología , Antivirales/farmacología , Virus ARN/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Amidas/farmacología , Animales , Línea Celular , Sinergismo Farmacológico , Guanosina Trifosfato/metabolismo , Humanos , Metiltioinosina/farmacología , Mutación/efectos de los fármacos , Fosforribosil Pirofosfato/metabolismo , Pirazinas/farmacología , Virus ARN/clasificación , Virus ARN/genética , ARN Viral/efectos de los fármacos , ARN Viral/genética , Replicación Viral/efectos de los fármacos
6.
Mol Aspects Med ; 81: 101005, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: covidwho-1562128

RESUMEN

Viruses with positive-sense single stranded RNA (+ssRNA) genomes are responsible for different diseases and represent a global health problem. In addition to developing new vaccines that protect against severe illness on infection, it is imperative to identify new antiviral molecules to treat infected patients. The genome of these RNA viruses generally codes for an enzyme with RNA dependent RNA polymerase (RdRP) activity. This molecule is centrally involved in the duplication of the RNA genome. Inhibition of this enzyme by small molecules will prevent duplication of the RNA genome and thus reduce the viral titer. An overview of the different therapeutic strategies used to inhibit RdRPs from +ssRNA viruses is provided, along with an analysis of these enzymes to highlight new binding sites for inhibitors.


Asunto(s)
Antivirales , Virus ARN , ARN Polimerasa Dependiente del ARN , Antivirales/uso terapéutico , Humanos , Virus ARN/efectos de los fármacos , Virus ARN/genética , ARN Polimerasa Dependiente del ARN/antagonistas & inhibidores , ARN Polimerasa Dependiente del ARN/genética
7.
PLoS Pathog ; 17(10): e1009726, 2021 10.
Artículo en Inglés | MEDLINE | ID: covidwho-1484867

RESUMEN

The zinc finger antiviral protein (ZAP) is a broad inhibitor of virus replication. Its best-characterized function is to bind CpG dinucleotides present in viral RNAs and, through the recruitment of TRIM25, KHNYN and other cofactors, target them for degradation or prevent their translation. The long and short isoforms of ZAP (ZAP-L and ZAP-S) have different intracellular localization and it is unclear how this regulates their antiviral activity against viruses with different sites of replication. Using ZAP-sensitive and ZAP-insensitive human immunodeficiency virus type I (HIV-1), which transcribe the viral RNA in the nucleus and assemble virions at the plasma membrane, we show that the catalytically inactive poly-ADP-ribose polymerase (PARP) domain in ZAP-L is essential for CpG-specific viral restriction. Mutation of a crucial cysteine in the C-terminal CaaX box that mediates S-farnesylation and, to a lesser extent, the residues in place of the catalytic site triad within the PARP domain, disrupted the activity of ZAP-L. Addition of the CaaX box to ZAP-S partly restored antiviral activity, explaining why ZAP-S lacks antiviral activity for CpG-enriched HIV-1 despite conservation of the RNA-binding domain. Confocal microscopy confirmed the CaaX motif mediated localization of ZAP-L to vesicular structures and enhanced physical association with intracellular membranes. Importantly, the PARP domain and CaaX box together jointly modulate the interaction between ZAP-L and its cofactors TRIM25 and KHNYN, implying that its proper subcellular localisation is required to establish an antiviral complex. The essential contribution of the PARP domain and CaaX box to ZAP-L antiviral activity was further confirmed by inhibition of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) replication, which replicates in double-membrane vesicles derived from the endoplasmic reticulum. Thus, compartmentalization of ZAP-L on intracellular membranes provides an essential effector function in ZAP-L-mediated antiviral activity against divergent viruses with different subcellular replication sites.


Asunto(s)
Prenilación/fisiología , Virus ARN/efectos de los fármacos , Proteínas de Unión al ARN/farmacología , Replicación Viral/fisiología , Islas de CpG/fisiología , Células HEK293 , VIH-1/fisiología , Células HeLa , Humanos , Virus ARN/fisiología , ARN Viral/química , ARN Viral/metabolismo , Motivos de Unión al ARN/fisiología , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/metabolismo , SARS-CoV-2/fisiología , Transfección , Replicación Viral/efectos de los fármacos
8.
Curr Res Transl Med ; 69(4): 103309, 2021 10.
Artículo en Inglés | MEDLINE | ID: covidwho-1459004

RESUMEN

PURPOSE OF THE STUDY: Currently no treatment has been proven to be efficacious for patients with early symptoms of COVID-19. Although most patients present mild or moderate symptoms, up to 5-10% may have a poor disease progression, so there is an urgent need for effective drugs, which can be administered even before the onset of severe symptoms, i.e. when the course of the disease is modifiable. Recently, promising results of several studies on oral ivermectin have been published, which has prompted us to conduct the present review of the scientific literature. METHODS: A narrative review has been carried out, focusing on the following four main topics: a) short-term efficacy in the treatment of the disease, b) long-term efficacy in the treatment of patients with post-acute symptoms of COVID-19, c) efficacy in the prophylaxis of the disease, and c) safety of ivermectin. RESULTS: The reviewed literature suggests that there seems to be sufficient evidence about the safety of oral ivermectin, as well as the efficacy of the drug in the early-treatment and the prophylaxis of COVID-19. CONCLUSIONS: In the view of the available evidence, the Frontline COVID-19 Critical Care Alliance (FLCCC) recommends the use of oral ivermectin for both prophylaxis and early-treatment of COVID-19. Further well-designed studies should be conducted in order to explore the efficacy and safety of invermectin at low and high doses, following different dosing schedules, in both, the short and long-term treatment.


Asunto(s)
Antivirales/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Reposicionamiento de Medicamentos , Ivermectina/uso terapéutico , SARS-CoV-2/efectos de los fármacos , Antivirales/efectos adversos , COVID-19/prevención & control , Estudios de Casos y Controles , Relación Dosis-Respuesta a Droga , Humanos , Ivermectina/administración & dosificación , Ivermectina/efectos adversos , Ivermectina/farmacología , Metaanálisis como Asunto , Estudios Multicéntricos como Asunto , Guías de Práctica Clínica como Asunto , Transporte de Proteínas/efectos de los fármacos , Virus ARN/efectos de los fármacos , Ensayos Clínicos Controlados Aleatorios como Asunto , Factores de Tiempo , Resultado del Tratamiento
9.
Virol J ; 17(1): 136, 2020 09 09.
Artículo en Inglés | MEDLINE | ID: covidwho-1435256

RESUMEN

BACKGROUND: Coronaviruses (CoVs) were long thought to only cause mild respiratory and gastrointestinal symptoms in humans but outbreaks of Middle East Respiratory Syndrome (MERS)-CoV, Severe Acute Respiratory Syndrome (SARS)-CoV-1, and the recently identified SARS-CoV-2 have cemented their zoonotic potential and their capacity to cause serious morbidity and mortality, with case fatality rates ranging from 4 to 35%. Currently, no specific prophylaxis or treatment is available for CoV infections. Therefore we investigated the virucidal and antiviral potential of Echinacea purpurea (Echinaforce®) against human coronavirus (HCoV) 229E, highly pathogenic MERS- and SARS-CoVs, as well as the newly identified SARS-CoV-2, in vitro. METHODS: To evaluate the antiviral potential of the extract, we pre-treated virus particles and cells and evaluated remaining infectivity by limited dilution. Furthermore, we exposed cells to the extract after infection to further evaluate its potential as a prophylaxis and treatment against coronaviruses. We also determined the protective effect of Echinaforce® in re-constituted nasal epithelium. RESULTS: In the current study, we found that HCoV-229E was irreversibly inactivated when exposed to Echinaforce® at 3.2 µg/ml IC50. Pre-treatment of cell lines, however, did not inhibit infection with HCoV-229E and post-infection treatment had only a marginal effect on virus propagation at 50 µg/ml. However, we did observe a protective effect in an organotypic respiratory cell culture system by exposing pre-treated respiratory epithelium to droplets of HCoV-229E, imitating a natural infection. The observed virucidal activity of Echinaforce® was not restricted to common cold coronaviruses, as both SARS-CoV-1 and MERS-CoVs were inactivated at comparable concentrations. Finally, the causative agent of COVID-19, SARS-CoV-2 was also inactivated upon treatment with 50µg/ml Echinaforce®. CONCLUSIONS: These results show that Echinaforce® is virucidal against HCoV-229E, upon direct contact and in an organotypic cell culture model. Furthermore, MERS-CoV and both SARS-CoV-1 and SARS-CoV-2 were inactivated at similar concentrations of the extract. Therefore we hypothesize that Echinacea purpurea preparations, such as Echinaforce®, could be effective as prophylactic treatment for all CoVs due to their structural similarities.


Asunto(s)
Antivirales/farmacología , Betacoronavirus/efectos de los fármacos , Coronavirus Humano 229E/efectos de los fármacos , Infecciones por Coronavirus/tratamiento farmacológico , Coronavirus/efectos de los fármacos , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Animales , COVID-19 , Línea Celular , Chlorocebus aethiops , Resfriado Común/tratamiento farmacológico , Resfriado Común/virología , Infecciones por Coronavirus/virología , Humanos , Coronavirus del Síndrome Respiratorio de Oriente Medio/efectos de los fármacos , Pandemias , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/virología , Virus ARN/efectos de los fármacos , Ensayos Clínicos Controlados Aleatorios como Asunto , SARS-CoV-2 , Síndrome Respiratorio Agudo Grave/tratamiento farmacológico , Síndrome Respiratorio Agudo Grave/virología , Células Vero
10.
mBio ; 12(2)2021 04 13.
Artículo en Inglés | MEDLINE | ID: covidwho-1388457

RESUMEN

Mammalian cells detect microbial molecules known as pathogen-associated molecular patterns (PAMPs) as indicators of potential infection. Upon PAMP detection, diverse defensive responses are induced by the host, including those that promote inflammation and cell-intrinsic antimicrobial activities. Host-encoded molecules released from dying or damaged cells, known as damage-associated molecular patterns (DAMPs), also induce defensive responses. Both DAMPs and PAMPs are recognized for their inflammatory potential, but only the latter are well established to stimulate cell-intrinsic host defense. Here, we report a class of DAMPs that engender an antiviral state in human epithelial cells. These DAMPs include oxPAPC (oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine), PGPC (1-palmitoyl-2-glutaryl phosphatidylcholine), and POVPC [1-palmitoyl-2-(5-oxovaleroyl)-sn-glycero-3-phosphatidylcholine], oxidized lipids that are naturally released from dead or dying cells. Exposing cells to these DAMPs prior to vesicular stomatitis virus (VSV) infection limits viral replication. Mechanistically, these DAMPs prevent viral entry, thereby limiting the percentage of cells that are productively infected and consequently restricting viral load. We found that the antiviral actions of oxidized lipids are distinct from those mediated by the PAMP Poly I:C, in that the former induces a more rapid antiviral response without the induction of the interferon response. These data support a model whereby interferon-independent defensive activities can be induced by DAMPs, which may limit viral replication before PAMP-mediated interferon responses are induced. This antiviral activity may impact viruses that disrupt interferon responses in the oxygenated environment of the lung, such as influenza virus and SARS-CoV-2.IMPORTANCE In this work, we explored how a class of oxidized lipids, spontaneously created during tissue damage and unprogrammed cell lysis, block the earliest events in RNA virus infection in the human epithelium. This gives us novel insight into the ways that we view infection models, unveiling a built-in mechanism to slow viral growth that neither engages the interferon response nor is subject to known viral antagonism. These oxidized phospholipids act prior to infection, allowing time for other, better-known innate immune mechanisms to take effect. This discovery broadens our understanding of host defenses, introducing a soluble factor that alters the cellular environment to protect from RNA virus infection.


Asunto(s)
Alarminas/farmacología , Antivirales/farmacología , Virus ARN/efectos de los fármacos , Internalización del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Células A549 , Muerte Celular/efectos de los fármacos , Humanos , Inmunidad Innata , Interferones/genética , Interferones/metabolismo , Cinética , Moléculas de Patrón Molecular Asociado a Patógenos/farmacología , Fosfatidilcolinas/farmacología , Virus ARN/fisiología , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/fisiología , Vesiculovirus/efectos de los fármacos , Vesiculovirus/fisiología , Carga Viral
11.
Viruses ; 12(6)2020 05 30.
Artículo en Inglés | MEDLINE | ID: covidwho-1389514

RESUMEN

Single-stranded positive RNA ((+) ssRNA) viruses include several important human pathogens. Some members are responsible for large outbreaks, such as Zika virus, West Nile virus, SARS-CoV, and SARS-CoV-2, while others are endemic, causing an enormous global health burden. Since vaccines or specific treatments are not available for most viral infections, the discovery of direct-acting antivirals (DAA) is an urgent need. Still, the low-throughput nature of and biosafety concerns related to traditional antiviral assays hinders the discovery of new inhibitors. With the advances of reverse genetics, reporter replicon systems have become an alternative tool for the screening of DAAs. Herein, we review decades of the use of (+) ssRNA viruses replicon systems for the discovery of antiviral agents. We summarize different strategies used to develop those systems, as well as highlight some of the most promising inhibitors identified by the method. Despite the genetic alterations introduced, reporter replicons have been shown to be reliable systems for screening and identification of viral replication inhibitors and, therefore, an important tool for the discovery of new DAAs.


Asunto(s)
Antivirales/farmacología , Descubrimiento de Drogas/métodos , Genes Reporteros/fisiología , Virus ARN/efectos de los fármacos , Replicón/fisiología , Animales , Antivirales/química , Línea Celular , Chlorocebus aethiops , Cricetinae , Humanos , Virus ARN/genética , Transfección , Células Vero
12.
Viruses ; 13(7)2021 06 27.
Artículo en Inglés | MEDLINE | ID: covidwho-1289026

RESUMEN

Many viruses, especially RNA viruses, utilize programmed ribosomal frameshifting and/or stop codon readthrough in their expression, and in the decoding of a few a UGA is dynamically redefined to specify selenocysteine. This recoding can effectively increase viral coding capacity and generate a set ratio of products with the same N-terminal domain(s) but different C-terminal domains. Recoding can also be regulatory or generate a product with the non-universal 21st directly encoded amino acid. Selection for translation speed in the expression of many viruses at the expense of fidelity creates host immune defensive opportunities. In contrast to host opportunism, certain viruses, including some persistent viruses, utilize recoding or adventitious frameshifting as part of their strategy to evade an immune response or specific drugs. Several instances of recoding in small intensively studied viruses escaped detection for many years and their identification resolved dilemmas. The fundamental importance of ribosome ratcheting is consistent with the initial strong view of invariant triplet decoding which however did not foresee the possibility of transitory anticodon:codon dissociation. Deep level dynamics and structural understanding of recoding is underway, and a high level structure relevant to the frameshifting required for expression of the SARS CoV-2 genome has just been determined.


Asunto(s)
Virus ADN/genética , Virus ADN/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Evasión Inmune , Virus ARN/genética , Antivirales/farmacología , Codón de Terminación , Virus ADN/efectos de los fármacos , Sistema de Lectura Ribosómico , Antígenos de Histocompatibilidad Clase I/genética , Conformación de Ácido Nucleico , Péptidos/inmunología , Biosíntesis de Proteínas , Virus ARN/efectos de los fármacos , Virus ARN/inmunología
13.
Sci China Life Sci ; 65(2): 341-361, 2022 02.
Artículo en Inglés | MEDLINE | ID: covidwho-1245727

RESUMEN

Viruses utilize cellular lipids and manipulate host lipid metabolism to ensure their replication and spread. Therefore, the identification of lipids and metabolic pathways that are suitable targets for antiviral development is crucial. Using a library of compounds targeting host lipid metabolic factors and testing them for their ability to block pseudorabies virus (PRV) and vesicular stomatitis virus (VSV) infection, we found that U18666A, a specific inhibitor of Niemann-Pick C1 (NPC1), is highly potent in suppressing the entry of diverse viruses including pseudotyped severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). NPC1 deficiency markedly attenuates viral growth by decreasing cholesterol abundance in the plasma membrane, thereby inhibiting the dynamics of clathrin-coated pits (CCPs), which are indispensable for clathrin-mediated endocytosis. Significantly, exogenous cholesterol can complement the dynamics of CCPs, leading to efficient viral entry and infectivity. Administration of U18666A improves the survival and pathology of PRV- and influenza A virus-infected mice. Thus, our studies demonstrate a unique mechanism by which NPC1 inhibition achieves broad antiviral activity, indicating a potential new therapeutic strategy against SARS-CoV-2, as well as other emerging viruses.


Asunto(s)
Androstenos/farmacología , Clatrina/fisiología , Invaginaciones Cubiertas de la Membrana Celular/fisiología , Virus ADN/efectos de los fármacos , Proteína Niemann-Pick C1/fisiología , Virus ARN/efectos de los fármacos , Internalización del Virus/efectos de los fármacos , Virus ADN/fisiología , Proteína Niemann-Pick C1/antagonistas & inhibidores , Virus ARN/fisiología
14.
Int J Mol Sci ; 22(8)2021 Apr 17.
Artículo en Inglés | MEDLINE | ID: covidwho-1206368

RESUMEN

Viral infections cause a host of fatal diseases and seriously affect every form of life from bacteria to humans. Although most viral infections can receive appropriate treatment thereby limiting damage to life and livelihood with modern medicine and early diagnosis, new types of viral infections are continuously emerging that need to be properly and timely treated. As time is the most important factor in the progress of many deadly viral diseases, early detection becomes of paramount importance for effective treatment. Aptamers are small oligonucleotide molecules made by the systematic evolution of ligands by exponential enrichment (SELEX). Aptamers are characterized by being able to specifically bind to a target, much like antibodies. However, unlike antibodies, aptamers are easily synthesized, modified, and are able to target a wider range of substances, including proteins and carbohydrates. With these advantages in mind, many studies on aptamer-based viral diagnosis and treatments are currently in progress. The use of aptamers for viral diagnosis requires a system that recognizes the binding of viral molecules to aptamers in samples of blood, serum, plasma, or in virus-infected cells. From a therapeutic perspective, aptamers target viral particles or host cell receptors to prevent the interaction between the virus and host cells or target intracellular viral proteins to interrupt the life cycle of the virus within infected cells. In this paper, we review recent attempts to use aptamers for the diagnosis and treatment of various viral infections.


Asunto(s)
Antivirales/uso terapéutico , Aptámeros de Nucleótidos/uso terapéutico , Virosis/diagnóstico , Virosis/tratamiento farmacológico , Animales , Virus ADN/efectos de los fármacos , Humanos , Virus ARN/efectos de los fármacos , Proteínas Virales/efectos de los fármacos , Virión/efectos de los fármacos
15.
FEBS Open Bio ; 11(5): 1452-1464, 2021 05.
Artículo en Inglés | MEDLINE | ID: covidwho-1168813

RESUMEN

Human pathogenic RNA viruses are threats to public health because they are prone to escaping the human immune system through mutations of genomic RNA, thereby causing local outbreaks and global pandemics of emerging or re-emerging viral diseases. While specific therapeutics and vaccines are being developed, a broad-spectrum therapeutic agent for RNA viruses would be beneficial for targeting newly emerging and mutated RNA viruses. In this study, we conducted a screen of repurposed drugs using Sendai virus (an RNA virus of the family Paramyxoviridae), with human-induced pluripotent stem cells (iPSCs) to explore existing drugs that may present anti-RNA viral activity. Selected hit compounds were evaluated for their efficacy against two important human pathogens: Ebola virus (EBOV) using Huh7 cells and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) using Vero E6 cells. Selective estrogen receptor modulators (SERMs), including raloxifene, exhibited antiviral activities against EBOV and SARS-CoV-2. Pioglitazone, a PPARγ agonist, also exhibited antiviral activities against SARS-CoV-2, and both raloxifene and pioglitazone presented a synergistic antiviral effect. Finally, we demonstrated that SERMs blocked entry steps of SARS-CoV-2 into host cells. These findings suggest that the identified FDA-approved drugs can modulate host cell susceptibility against RNA viruses.


Asunto(s)
Antivirales/farmacología , Reposicionamiento de Medicamentos , Virus ARN/efectos de los fármacos , ARN Viral/antagonistas & inhibidores , SARS-CoV-2/efectos de los fármacos , Animales , Línea Celular , Chlorocebus aethiops , Reposicionamiento de Medicamentos/métodos , Ebolavirus/efectos de los fármacos , Ebolavirus/fisiología , Humanos , Células Madre Pluripotentes Inducidas/virología , Pruebas de Sensibilidad Microbiana/métodos , Pioglitazona/farmacología , Virus ARN/fisiología , Clorhidrato de Raloxifeno/farmacología , SARS-CoV-2/fisiología , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Virus Sendai/efectos de los fármacos , Virus Sendai/fisiología , Células Vero , Tratamiento Farmacológico de COVID-19
16.
Virus Res ; 297: 198384, 2021 05.
Artículo en Inglés | MEDLINE | ID: covidwho-1127061

RESUMEN

Bovine respiratory disease (BRD) complex is an important viral infection that causes huge economic losses in cattle herds worldwide. However, there is no directly effective antiviral drug application against respiratory viral pathogens; generally, the metaphylactic antibacterial drug applications are used for BRD. Ivermectin (IVM) is currently used as a broad-spectrum anti-parasitic agent both for veterinary and human medicine on some occasions. Moreover, since it is identified as an inhibitor for importin α/ß-mediated nuclear localization signal (NLS), IVM is also reported to have antiviral potential against several RNA and DNA viruses. Since therapeutic use of IVM in COVID-19 cases has recently been postulated, the potential antiviral activity of IVM against bovine respiratory viruses including BRSV, BPIV-3, BoHV-1, BCoV and BVDV are evaluated in this study. For these purposes, virus titration assay was used to evaluate titers in viral harvest from infected cells treated with non-cytotoxic IVM concentrations (1, 2.5 and 5 µM) and compared to titers from non-treated infected cells. This study indicated that IVM inhibits the replication of BCoV, BVDV, BRSV, BPIV-3 and BoHV-1 in a dose-dependent manner in vitro as well as number of extracellular infectious virions. In addition, it was demonstrated that IVM has no clear effect on the attachment and penetration steps of the replication of the studied viruses. Finally, this study shows for the first time that IVM can inhibit infection of BRD-related viral agents namely BCoV, BPIV-3, BVDV, BRSV and BoHV-1 at the concentrations of 2.5 and 5 µM. Consequently, IVM, which is licensed for antiparasitic indications, also deserves to be evaluated as a broad-spectrum antiviral in BRD cases caused by viral pathogens.


Asunto(s)
Antivirales/farmacología , Ivermectina/farmacología , Virus ARN/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Animales , Complejo Respiratorio Bovino/tratamiento farmacológico , Bovinos , Perros , Evaluación Preclínica de Medicamentos , Células de Riñón Canino Madin Darby , Virus ARN/fisiología , Acoplamiento Viral/efectos de los fármacos
17.
ACS Infect Dis ; 7(2): 471-478, 2021 02 12.
Artículo en Inglés | MEDLINE | ID: covidwho-1006383

RESUMEN

A series of 7-deazaadenine ribonucleosides bearing alkyl, alkenyl, alkynyl, aryl, or hetaryl groups at position 7 as well as their 5'-O-triphosphates and two types of monophosphate prodrugs (phosphoramidates and S-acylthioethanol esters) were prepared and tested for antiviral activity against selected RNA viruses (Dengue, Zika, tick-borne encephalitis, West Nile, and SARS-CoV-2). The modified triphosphates inhibited the viral RNA-dependent RNA polymerases at micromolar concentrations through the incorporation of the modified nucleotide and stopping a further extension of the RNA chain. 7-Deazaadenosine nucleosides bearing ethynyl or small hetaryl groups at position 7 showed (sub)micromolar antiviral activities but significant cytotoxicity, whereas the nucleosides bearing bulkier heterocycles were still active but less toxic. Unexpectedly, the monophosphate prodrugs were similarly or less active than the corresponding nucleosides in the in vitro antiviral assays, although the bis(S-acylthioethanol) prodrug 14h was transported to the Huh7 cells and efficiently released the nucleoside monophosphate.


Asunto(s)
Antivirales/farmacología , Profármacos/farmacología , Purinas/farmacología , Virus ARN/efectos de los fármacos , Ribonucleósidos/farmacología , COVID-19/virología , Línea Celular Tumoral , Virus del Dengue/efectos de los fármacos , Virus de la Encefalitis Transmitidos por Garrapatas/efectos de los fármacos , Humanos , Fosfatos/farmacología , Nucleósidos de Purina , ARN Polimerasa Dependiente del ARN/metabolismo , SARS-CoV-2/efectos de los fármacos , Virus del Nilo Occidental/efectos de los fármacos , Virus Zika/efectos de los fármacos , Tratamiento Farmacológico de COVID-19
18.
J Nat Prod ; 84(1): 161-182, 2021 01 22.
Artículo en Inglés | MEDLINE | ID: covidwho-989652

RESUMEN

Three families of RNA viruses, the Coronaviridae, Flaviviridae, and Filoviridae, collectively have great potential to cause epidemic disease in human populations. The current SARS-CoV-2 (Coronaviridae) responsible for the COVID-19 pandemic underscores the lack of effective medications currently available to treat these classes of viral pathogens. Similarly, the Flaviviridae, which includes such viruses as Dengue, West Nile, and Zika, and the Filoviridae, with the Ebola-type viruses, as examples, all lack effective therapeutics. In this review, we present fundamental information concerning the biology of these three virus families, including their genomic makeup, mode of infection of human cells, and key proteins that may offer targeted therapies. Further, we present the natural products and their derivatives that have documented activities to these viral and host proteins, offering hope for future mechanism-based antiviral therapeutics. By arranging these potential protein targets and their natural product inhibitors by target type across these three families of virus, new insights are developed, and crossover treatment strategies are suggested. Hence, natural products, as is the case for other therapeutic areas, continue to be a promising source of structurally diverse new anti-RNA virus therapeutics.


Asunto(s)
Antivirales/uso terapéutico , Productos Biológicos/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Infecciones por Virus ARN/tratamiento farmacológico , Animales , Desarrollo de Medicamentos , Genoma Viral , Humanos , Virus ARN/efectos de los fármacos , Virus ARN/enzimología , Virus ARN/fisiología , Replicación Viral
19.
Viruses ; 12(12)2020 12 10.
Artículo en Inglés | MEDLINE | ID: covidwho-969583

RESUMEN

Recent RNA virus outbreaks such as Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and Ebola virus (EBOV) have caused worldwide health emergencies highlighting the urgent need for new antiviral strategies. Targeting host cell pathways supporting viral replication is an attractive approach for development of antiviral compounds, especially with new, unexplored viruses where knowledge of virus biology is limited. Here, we present a strategy to identify host-targeted small molecule inhibitors using an image-based phenotypic antiviral screening assay followed by extensive target identification efforts revealing altered cellular pathways upon antiviral compound treatment. The newly discovered antiviral compounds showed broad-range antiviral activity against pathogenic RNA viruses such as SARS-CoV-2, EBOV and Crimean-Congo hemorrhagic fever virus (CCHFV). Target identification of the antiviral compounds by thermal protein profiling revealed major effects on proteostasis pathways and disturbance in interactions between cellular HSP70 complex and viral proteins, illustrating the supportive role of HSP70 on many RNA viruses across virus families. Collectively, this strategy identifies new small molecule inhibitors with broad antiviral activity against pathogenic RNA viruses, but also uncovers novel virus biology urgently needed for design of new antiviral therapies.


Asunto(s)
Antivirales/farmacología , Interacciones Huésped-Patógeno/efectos de los fármacos , Virus ARN/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Animales , Línea Celular , Ebolavirus/efectos de los fármacos , Ebolavirus/fisiología , Proteínas HSP70 de Choque Térmico/metabolismo , Virus de la Fiebre Hemorrágica de Crimea-Congo/efectos de los fármacos , Virus de la Fiebre Hemorrágica de Crimea-Congo/fisiología , Humanos , Unión Proteica/efectos de los fármacos , Estabilidad Proteica , Proteoma/efectos de los fármacos , Proteostasis/efectos de los fármacos , Infecciones por Virus ARN/metabolismo , Infecciones por Virus ARN/virología , Virus ARN/fisiología , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/fisiología , Bibliotecas de Moléculas Pequeñas/farmacología , Proteínas Virales/metabolismo
20.
Molecules ; 25(23)2020 Dec 03.
Artículo en Inglés | MEDLINE | ID: covidwho-963646

RESUMEN

The RNA-dependent RNA polymerase (RdRp) is an essential enzyme for the viral replication process, catalyzing the viral RNA synthesis using a metal ion-dependent mechanism. In recent years, RdRp has emerged as an optimal target for the development of antiviral drugs, as demonstrated by recent approvals of sofosbuvir and remdesivir against Hepatitis C virus (HCV) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), respectively. In this work, we overview the main sequence and structural features of the RdRp of emerging RNA viruses such as Coronaviruses, Flaviviruses, and HCV, as well as inhibition strategies implemented so far. While analyzing the structural information available on the RdRp of emerging RNA viruses, we provide examples of success stories such as for HCV and SARS-CoV-2. In contrast, Flaviviruses' story has raised attention about how the lack of structural details on catalytically-competent or ligand-bound RdRp strongly hampers the application of structure-based drug design, either in repurposing and conventional approaches.


Asunto(s)
Antivirales/química , Antivirales/farmacología , Virus ARN/enzimología , ARN Polimerasa Dependiente del ARN/química , Amidas/química , Amidas/farmacología , Coronavirus/efectos de los fármacos , Coronavirus/enzimología , Coronavirus/genética , Diseño de Fármacos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Flavivirus/efectos de los fármacos , Flavivirus/enzimología , Flavivirus/genética , Hepacivirus/efectos de los fármacos , Hepacivirus/enzimología , Hepacivirus/genética , Humanos , Pirazinas/química , Pirazinas/farmacología , Infecciones por Virus ARN/epidemiología , Virus ARN/efectos de los fármacos , ARN Polimerasa Dependiente del ARN/antagonistas & inhibidores , ARN Polimerasa Dependiente del ARN/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA